1
|
Saha KK, Mandal S, Barman A, Chatterjee S, Mandal NC. Deciphering the genomic and physiological basis of pH dependent siderophore production in Enterobacter sp. DRP3 and mitigation of lead stress in rice seedlings. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137587. [PMID: 39954437 DOI: 10.1016/j.jhazmat.2025.137587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/26/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Anthropogenic activities like heavy metal pollution exert the most devastating effect on agriculture. Siderophores are small peptides capable to chelate iron and different heavy metals; thereby reduce metal toxicity. However, very little information is available about their physiology (siderophore types, effect of temperature, pH, toxic metals), and especially of their gene expression patterns. Here, we have carried out a detailed study on siderophore production dynamics along with their gene expression pattern in Enterobacter sp. DRP3. DRP3 was able to produce two different types of siderophores hydroxamate type (19.81 µg ml-1) during early stages and catecholate type (59.52 µg ml-1) later stages of its growth, especially at pH-6.8. DRP3 was able to produce similar concentrations of siderophores even under high lead concentrations. Further whole genome analysis has revealed the presence of enterobactin and aerobactin gene clusters. Quantitative real-time PCR observed a 5.02-fold and 1.90-fold overexpression of the enterobactin biosynthesis genes entC and entF, respectively, and a 3.12-fold upregulation of the aerobactin biosynthesis gene iucC in the absence of exogenously added Fe3+ by DRP3. Our study also highlighted that following root colonization DRP3 is excellent in mitigating Pb(II) stress in rice seedlings while promoting iron content and reducing lead content in plant tissue.
Collapse
Affiliation(s)
- Kunal Kumar Saha
- Mycology and Plant Pathology Laboratory, Department of Botany, Visva Bharati, Santiniketan, West Bengal, India
| | - Subhrangshu Mandal
- Stress Physiology and Environmental Microbiology Laboratory, Department of Botany, Visva Bharati, Santiniketan, West Bengal, India.
| | - Anik Barman
- Department of Microbiology, Bose Institute, P-1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Sumit Chatterjee
- Department of Biological Sciences, Bose Institute, Kolkata 700091, India
| | - Narayan Chandra Mandal
- Mycology and Plant Pathology Laboratory, Department of Botany, Visva Bharati, Santiniketan, West Bengal, India.
| |
Collapse
|
2
|
Miao ZY, Lin J, Chen WM. Natural sideromycins and siderophore-conjugated natural products as inspiration for novel antimicrobial agents. Eur J Med Chem 2025; 287:117333. [PMID: 39892091 DOI: 10.1016/j.ejmech.2025.117333] [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: 12/10/2024] [Revised: 01/25/2025] [Accepted: 01/25/2025] [Indexed: 02/03/2025]
Abstract
The widespread emergence of multidrug-resistant (MDR) Gram-negative pathogens has posed a major challenge to clinical anti-infective therapy, and new effective treatments are urgently needed. A promising "Trojan horse" strategy involves conjugating antibiotics to siderophore molecules; the resulting siderophore-antibiotic conjugates (SACs) deliver antibiotics directly into cells by hijacking the sophisticated iron transport systems of Gram-negative bacteria, bypassing the outer membrane permeability barrier to enhance uptake and antibacterial efficacy. The clinical release of the first siderophore-antibiotic conjugate, cefiderocol, has aroused tremendous interest in the field among researchers and pharmaceutical companies. To date, most of the reported SACs have focused on the conjugation of siderophores to traditional antibacterial drugs. However, these antibacterial agents designed on the basis of the traditional antibiotic skeleton theoretically bear the risk of cross-resistance caused by shared molecular scaffolds. In this case, exploring novel natural product antibacterial conjugate scaffolds to circumvent the risk of early cross-resistance represents a presumably more sustainable approach for the development of SACs. In this review, we systematically summarize the research progress on siderophore-natural product conjugates as novel antimicrobial agents reported since 2010. Additionally, we propose challenges to be overcome and prospects for future development in this field.
Collapse
Affiliation(s)
- Zhi-Ying Miao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 511400, China
| | - Jing Lin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 511400, China.
| | - Wei-Min Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 511400, China.
| |
Collapse
|
3
|
Pantaleoni A, Marrocchi A, Russo P, Malucelli G, Altamura D, Nardelli F, Pizzanelli S, Freni A, Giannini C, Santarelli ML, Bavasso I, Bracciale MP, Sarasini F. Advanced flame-retardant biocomposites: Polylactic acid reinforced with green gallic acid‑iron‑phosphorus coated flax fibers. Int J Biol Macromol 2025; 300:140215. [PMID: 39855498 DOI: 10.1016/j.ijbiomac.2025.140215] [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: 10/21/2024] [Revised: 12/30/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
This study introduces a sustainable approach for enhancing the fire retardancy and smoke suppression of poly(lactic acid) (PLA) composites, contributing to addressing one of the major challenges in biocomposites that limits their application in various engineering fields, as automotive and construction sectors. Flax fibers (FF) were surface functionalized with a novel organic-inorganic hybrid flame retardant (FR), offering a sustainable bioinspired approach that mitigates potential mechanical properties impairment and FR leaching, which can cause environmental concerns and reduced composite durability. The process involves a three-step coating procedure. First, the flax fibers (FF) are pretreated with ozone to promote carboxylic group formation (FF-O3); subsequently, gallic acid (GA) units are covalently immobilized on the fiber surface (FF-GA); finally, the hybrid FR iron phenylphosphonate is complexed with the phenolic groups of GA units (FF-GA-FeP). Fourier transform infrared (FT-IR) analysis of FF-GA-FeP confirmed the presence of specific absorptions associated with the deposited FR coating. Scanning electron microscopy coupled with energy-dispersive X-ray spectroscopy (SEM-EDS) revealed changes in fiber morphology and confirmed the incorporation of iron and phosphorus. Solid-state nuclear magnetic resonance (SSNMR) spectroscopy and X-ray WAXS microscopy revealed that fibers' crystallinity was not significantly affected by derivatization. Microwave plasma atomic emission spectroscopy (MP-AES) detected a precise 0.1 wt% iron loading. Using FF-GA-FeP as reinforcement in PLA-based composites (PLA/FF-FeP) resulted in enhanced thermal stability and flame retardancy of the composites, with minimal coating application, as revealed by thermogravimetric analysis (TGA) and cone calorimetry tests (CCT). A decrease in peak of heat release rate (pHRR), total smoke release (TSR), specific extinction area (SEA), and Fire Propagating Index (FPI) of 5, 87, 68, and 9.5 %, respectively, was achieved for PLA/FF-FeP, compared to untreated flax fiber reinforced PLA (PLA/FF). Furthermore, preliminary tensile tests indicate minor changes in tensile strength and a slight increase in stiffness of the PLA/FF-FeP compared to PLA/FF. Hence, in the biocomposite, the immobilization of a minimal amount of iron phenylphosphonate directly on the flax fiber surface proved to be an effective strategy for smoke suppression while preserving the mechanical integrity of the composite.
Collapse
Affiliation(s)
- Alessia Pantaleoni
- Department of Chemical Engineering Materials Environment, Sapienza University of Rome, Via Eudossiana 18, 00184 Rome, Italy.
| | - Assunta Marrocchi
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy
| | - Pietro Russo
- Institute for Polymers, Composites and Biomaterials, National Research Council, Via Campi Flegrei 34, 80078 Pozzuoli, Na, Italy
| | - Giulio Malucelli
- Department of Applied Science and Technology, Politecnico di Torino, Viale Teresa Michel 5, 15121 Alessandria, Italy
| | - Davide Altamura
- Institute of Crystallography, National Research Council, v. Amendola 122/O, 70126 Bari, Italy
| | - Francesca Nardelli
- Institute for the Chemistry of OrganoMetallic Compounds, Italian National Research Council, CNR-ICCOM, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy; Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy
| | - Silvia Pizzanelli
- Institute for the Chemistry of OrganoMetallic Compounds, Italian National Research Council, CNR-ICCOM, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy; Centre for Instrument Sharing (CISUP), University of Pisa, Lungarno Pacinotti 43, 56126 Pisa, Italy
| | - Angelo Freni
- Institute for the Chemistry of OrganoMetallic Compounds, Italian National Research Council, CNR-ICCOM, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy
| | - Cinzia Giannini
- Institute of Crystallography, National Research Council, v. Amendola 122/O, 70126 Bari, Italy
| | - Maria Laura Santarelli
- Department of Chemical Engineering Materials Environment, Sapienza University of Rome, Via Eudossiana 18, 00184 Rome, Italy
| | - Irene Bavasso
- Department of Chemical Engineering Materials Environment, Sapienza University of Rome, Via Eudossiana 18, 00184 Rome, Italy
| | - Maria Paola Bracciale
- Department of Chemical Engineering Materials Environment, Sapienza University of Rome, Via Eudossiana 18, 00184 Rome, Italy
| | - Fabrizio Sarasini
- Department of Chemical Engineering Materials Environment, Sapienza University of Rome, Via Eudossiana 18, 00184 Rome, Italy
| |
Collapse
|
4
|
Will V, Moynié L, Si Ahmed Charrier E, Le Bas A, Kuhn L, Volck F, Chicher J, Aksoy H, Madec M, Antheaume C, Mislin GLA, Schalk IJ. Structure of the Outer Membrane Transporter FemA and Its Role in the Uptake of Ferric Dihydro-Aeruginoic Acid and Ferric Aeruginoic Acid in Pseudomonas aeruginosa. ACS Chem Biol 2025; 20:690-706. [PMID: 40035455 DOI: 10.1021/acschembio.4c00820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Iron is essential for bacterial growth, and Pseudomonas aeruginosa synthesizes the siderophores pyochelin (PCH) and pyoverdine to acquire it. PCH contains a thiazolidine ring that aids in iron chelation but is prone to hydrolysis, leading to the formation of 2-(2-hydroxylphenyl)-thiazole-4-carbaldehyde (IQS). Using mass spectrometry, we demonstrated that PCH undergoes hydrolysis and oxidation in solution, resulting in the formation of aeruginoic acid (AA). This study used proteomic analyses and fluorescent reporters to show that AA, dihydroaeruginoic acid (DHA), and PCH induce the expression of femA, a gene encoding the ferri-mycobactin outer membrane transporter in P. aeruginosa. Notably, the induction by AA and DHA was observed only in strains unable to produce pyoverdine, suggesting their weaker iron-chelating ability compared to that of pyoverdine. 55Fe uptake assays demonstrated that both AA-Fe and DHA-Fe complexes are transported via FemA; however, no uptake was observed for PCH-Fe through this transporter. Structural studies revealed that FemA is able to bind AA2-Fe or DHA2-Fe complexes. Key interactions are conserved between FemA and these two complexes, with specificity primarily driven by one of the two siderophore molecules. Interestingly, although no iron uptake was noted for PCH through FemA, the transporter also binds PCH-Fe in a similar manner. These findings show that under moderate iron deficiency, when only PCH is produced by P. aeruginosa, degradation products AA and DHA enhance iron uptake by inducing femA expression and facilitating iron transport through FemA. This provides new insights into the pathogen's strategies for iron homeostasis.
Collapse
Affiliation(s)
- Virginie Will
- CNRS, University of Strasbourg, UMR7242, UMR7242, ESBS, Bld Sébastien Brant, F-67412 Strasbourg, Illkirch, France
- University of Strasbourg, UMR7242, ESBS, Bld Sébastien Brant, F-67412 Strasbourg, Illkirch, France
| | - Lucile Moynié
- The Rosalind Franklin Institute, Harwell Campus, Oxfordshire OX11 0QS, United Kingdom
| | - Elise Si Ahmed Charrier
- CNRS, University of Strasbourg, UMR7242, UMR7242, ESBS, Bld Sébastien Brant, F-67412 Strasbourg, Illkirch, France
- University of Strasbourg, UMR7242, ESBS, Bld Sébastien Brant, F-67412 Strasbourg, Illkirch, France
| | - Audrey Le Bas
- The Rosalind Franklin Institute, Harwell Campus, Oxfordshire OX11 0QS, United Kingdom
| | - Lauriane Kuhn
- Plateforme Protéomique Strasbourg-Esplanade, CNRS, Université de Strasbourg, IBMC, 2 Allée Konrad Roentgen, F-67084 Strasbourg, France
| | - Florian Volck
- CNRS, University of Strasbourg, UMR7242, UMR7242, ESBS, Bld Sébastien Brant, F-67412 Strasbourg, Illkirch, France
- University of Strasbourg, UMR7242, ESBS, Bld Sébastien Brant, F-67412 Strasbourg, Illkirch, France
| | - Johana Chicher
- Plateforme Protéomique Strasbourg-Esplanade, CNRS, Université de Strasbourg, IBMC, 2 Allée Konrad Roentgen, F-67084 Strasbourg, France
| | - Hava Aksoy
- Université de Strasbourg, Institut des Sciences et de L'Ingénieurie Supramoleculaire, Plateforme d'analyses Chimiques 8 allée Gaspard Monge, F-67000 Strasbourg, France
| | - Morgan Madec
- ICube Laboratory, UMR 7357 (CNRS/University of Strasbourg), Bld Sébastien Brant, F-67412 Strasbourg, Illkirch, France
| | - Cyril Antheaume
- Université de Strasbourg, Institut des Sciences et de L'Ingénieurie Supramoleculaire, Plateforme d'analyses Chimiques 8 allée Gaspard Monge, F-67000 Strasbourg, France
| | - Gaëtan L A Mislin
- CNRS, University of Strasbourg, UMR7242, UMR7242, ESBS, Bld Sébastien Brant, F-67412 Strasbourg, Illkirch, France
- University of Strasbourg, UMR7242, ESBS, Bld Sébastien Brant, F-67412 Strasbourg, Illkirch, France
| | - Isabelle J Schalk
- CNRS, University of Strasbourg, UMR7242, UMR7242, ESBS, Bld Sébastien Brant, F-67412 Strasbourg, Illkirch, France
- University of Strasbourg, UMR7242, ESBS, Bld Sébastien Brant, F-67412 Strasbourg, Illkirch, France
| |
Collapse
|
5
|
Hohmann M, Iliasov D, Larralde M, Johannes W, Janßen KP, Zeller G, Mascher T, Gulder TAM. Heterologous Expression of a Cryptic BGC from Bilophila sp. Provides Access to a Novel Family of Antibacterial Thiazoles. ACS Synth Biol 2025; 14:967-978. [PMID: 39999339 PMCID: PMC11934131 DOI: 10.1021/acssynbio.5c00042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025]
Abstract
Human health is greatly influenced by the gut microbiota and microbiota imbalance can lead to the development of diseases. It is widely acknowledged that the interaction of bacteria within competitive ecosystems is influenced by their specialized metabolites, which act, e.g., as antibacterials or siderophores. However, our understanding of the occurrence and impact of such natural products in the human gut microbiome remains very limited. As arylthiazole siderophores are an emerging family of growth-promoting molecules in pathogenic bacteria, we analyzed a metagenomic data set from the human microbiome and thereby identified the bil-BGC, which originates from an uncultured Bilophila strain. Through gene synthesis and BGC assembly, heterologous expression and mutasynthetic experiments, we discovered the arylthiazole natural products bilothiazoles A-F. While established activities of related molecules indicate their involvement in metal-binding and -uptake, which could promote the growth of pathogenic strains, we also found antibiotic activity for some bilothiazoles. This is supported by biosensor-experiments, where bilothiazoles C and E show PrecA-suppressing activity, while bilothiazole F induces PblaZ, a biosensor characteristic for β-lactam antibiotics. These findings serve as a starting point for investigating the role of bilothiazoles in the pathogenicity of Bilophila species in the gut.
Collapse
Affiliation(s)
- Maximilian Hohmann
- Chair
of Technical Biochemistry, TUD Dresden University
of Technology, Bergstraße 66, 01069 Dresden, Germany
| | - Denis Iliasov
- General
Microbiology, TUD Dresden University of
Technology, Zellescher
Weg 20b, 01217 Dresden, Germany
| | - Martin Larralde
- Leiden
University Center for Infectious Diseases (LUCID), Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Widya Johannes
- Department
of Surgery, School of Medicine and Health, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Klaus-Peter Janßen
- Department
of Surgery, School of Medicine and Health, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Georg Zeller
- Leiden
University Center for Infectious Diseases (LUCID) and Center for Microbiome
Analyses and Therapeutics (CMAT), Leiden
University Medical Center, 2333 ZA Leiden, Netherlands
| | - Thorsten Mascher
- General
Microbiology, TUD Dresden University of
Technology, Zellescher
Weg 20b, 01217 Dresden, Germany
| | - Tobias A. M. Gulder
- Chair
of Technical Biochemistry, TUD Dresden University
of Technology, Bergstraße 66, 01069 Dresden, Germany
- Department
of Natural Product Biotechnology, Helmholtz Institute for Pharmaceutical
Research Saarland (HIPS), Helmholtz Centre for Infection Research
(HZI) and Department of Pharmacy, PharmaScienceHub (PSH), Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| |
Collapse
|
6
|
Davidov Y, Tejman-Yarden N, Robinson A, Rahav G, Nissan I. Enterobactin and salmochelin S4 inhibit the growth of Staphylococcus aureus. Front Cell Infect Microbiol 2025; 15:1456046. [PMID: 40110026 PMCID: PMC11919883 DOI: 10.3389/fcimb.2025.1456046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 02/12/2025] [Indexed: 03/22/2025] Open
Abstract
There is increasing demand for novel antimicrobial agents to tackle the antimicrobial resistance crisis. Here we report that two Enterobacteriaceae-produced siderophores, enterobactin and salmochelin S4, inhibit the growth of Staphylococcus aureus isolates, including methicillin-resistance S. aureus (MRSA) clinical isolates. The IC50 for different S. aureus isolates were 2-5 µM for salmochelin S4 and 5-10 µM for enterobactin. This inhibitory activity was partially repressed by adding Fe+3. These siderophores also inhibited the growth of Enterococcus strains, including vancomycin-resistant enterococci (VRE) clinical isolates, though less effectively than for S. aureus. The growth of various Gram-negative bacteria was barely affected by these siderophores. These results shed new light on the role of enterobactin and salmochelin in bacterial physiology and ecology and have potential for the development of novel strategies to combat the rapid rise of multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Yaacov Davidov
- Infectious Disease Unit, Sheba Medical Center, Tel Hashomer, Israel
| | - Noa Tejman-Yarden
- Department of Laboratories, Public Health Directorate, Ministry of Health, Jerusalem, Israel
| | - Ari Robinson
- Infectious Disease Unit, Sheba Medical Center, Tel Hashomer, Israel
| | - Galia Rahav
- Infectious Disease Unit, Sheba Medical Center, Tel Hashomer, Israel
- Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Israel Nissan
- Infectious Disease Unit, Sheba Medical Center, Tel Hashomer, Israel
- Department of Avian Diseases, Kimron Veterinary Institute, Beit Dagan, Israel
| |
Collapse
|
7
|
Yeh TY, Lu HF, Li LH, Lin YT, Yang TC. Contribution of fepA sm, fciABC, sbaA, sbaBCDEF, and feoB to ferri-stenobactin acquisition in Stenotrophomonas maltophilia KJ. BMC Microbiol 2025; 25:91. [PMID: 40000954 PMCID: PMC11852561 DOI: 10.1186/s12866-025-03792-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Stenotrophomonas maltophilia, an opportunistic pathogen, is ubiquitously distributed in the environment. In response to iron-depletion stress, S. maltophilia synthesizes the sole catecholate-type siderophore, stenobactin, for ferric iron acquisition. FepAsm, a TonB-dependent transporter (TBDT), is the sole known outer membrane receptor responsible for ferri-stenobactin uptake in S. maltophilia K279a. However, S. maltophilia KJ and its isogenic fepA mutant displayed comparable ability to utilize FeCl3 as the sole iron source for growth in iron-depleted conditions, suggesting the involvement of additional TBDT in ferri-stenobactin uptake in the KJ strain. Here, we aimed to determine additional TBDT required for ferri-stenobactin uptake and the post-TBDT ferri-stenobactin transport system in the KJ strain. METHODS AND RESULTS Twelve TBDTs, whose expression were significantly upregulated in 2,2'-dipyridyl-treated KJ strain, were selected as candidates for ferri-stenobactin uptake. The involvement of these selected candidates in ferri-stenobactin acquisition was investigated using deletion mutant construction and FeCl3 utilization assay. Among the 12 TBDTs tested, FepAsm, FciA, and SbaA were the TBDTs for ferri-stenobactin uptake in KJ strain. Because fciA is a member of fciTABC operon, the involvement of fciTABC operon in ferri-stenobactin uptake was also investigated. Of the fciTABC operon, fciA, fciB and fciC, but not fciT, contributed to ferri-stenobatin acquisition. SbaE is the homolog of FepD/FepG, the inner membrane transporters for ferri-enterobactin in E. coli; therefore, sbaBCDEF operon was selected as a candidate for the post-TBDT transport system of ferri-stenobactin. All proteins encoded by sbaBCDEF operon participated in ferri-stenobactin acquisition. Due to the contribution of the putative periplasmic esterase SbaB to ferri-stenobactin acquisition, FeoB, a ferrous iron inner membrane transporter, was included as a candidate and proved to be involved in ferri-stenobactin acquisition. Accordingly, contributions of feoB and sbaE to ferri-stenobactin acquisition illustrated that ferric and ferrous iron could be transported across the inner membrane via SbaE and FeoB, respectively. CONCLUSIONS FepAsm, fciABC, sbaA, sbaBCDEF, and feoB contribute to ferri-stenobatin acquisition in Stenotrophomonas maltophilia KJ.
Collapse
Affiliation(s)
- Ting-Yu Yeh
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
| | - Hsu-Feng Lu
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan, Republic of China
| | - Li-Hua Li
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - Yi-Tsung Lin
- Division of Infectious Diseases, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
| | - Tsuey-Ching Yang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China.
| |
Collapse
|
8
|
Piper KR, Souza SSR, Ikhimiukor OO, Workman AA, Martin IW, Andam CP. Lineage-specific variation in frequency and hotspots of recombination in invasive Escherichia coli. BMC Genomics 2025; 26:190. [PMID: 39994515 PMCID: PMC11853335 DOI: 10.1186/s12864-025-11367-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 02/14/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND The opportunistic bacterium Escherichia coli can invade normally sterile sites in the human body, potentially leading to life-threatening organ dysfunction and even death. However, our understanding of the evolutionary processes that shape its genetic diversity in this sterile environment remains limited. Here, we aim to quantify the frequency and characteristics of homologous recombination in E. coli from bloodstream infections. RESULTS Analysis of 557 short-read genome sequences revealed that the propensity to exchange DNA by homologous recombination varies within a distinct population (bloodstream) at narrow geographic (Dartmouth Hitchcock Medical Center, New Hampshire, USA) and temporal (years 2016 - 2022) scope. We identified the four largest monophyletic sequence clusters in the core genome phylogeny that are represented by prominent sequence types (ST): BAPS1 (mainly ST95), BAPS4 (mainly ST73), BAPS10 (mainly ST131), BAPS14 (mainly ST58). We show that the four dominant clusters vary in different characteristics of recombination: number of single nucleotide polymorphisms due to recombination, number of recombination blocks, cumulative bases in recombination blocks, ratio of probabilities that a given site was altered through recombination and mutation (r/m), and ratio of rates at which recombination and mutation occurred (ρ/θ). Each sequence cluster contains a unique set of antimicrobial resistance (AMR) and virulence genes that have experienced recombination. Common among the four sequence clusters were the recombined virulence genes with functions associated with the Curli secretion channel (csgG) and ferric enterobactin transport (entEF, fepEG). We did not identify any one recombined AMR gene that was present in all four sequence clusters. However, AMR genes mdtABC, baeSR, emrKY and tolC had experienced recombination in sequence clusters BAPS4, BAPS10, and BAPS14. These differences lie in part on the contributions of vertically inherited ancestral recombination and contemporary branch-specific recombination, with some genomes having relatively higher proportions of recombined DNA. CONCLUSIONS Our results highlight the variation in the propensity to exchange DNA via homologous recombination within a distinct population at narrow geographic and temporal ranges. Understanding the sources of the genetic variation in invasive E. coli will help inform the implementation of effective strategies to reduce the burden of disease and AMR.
Collapse
Affiliation(s)
- Kathryn R Piper
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, USA
| | - Stephanie S R Souza
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, USA
| | - Odion O Ikhimiukor
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, USA
| | - Adrienne A Workman
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center and Dartmouth College Geisel School of Medicine, Lebanon, NH, USA
| | - Isabella W Martin
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center and Dartmouth College Geisel School of Medicine, Lebanon, NH, USA.
| | - Cheryl P Andam
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, USA.
| |
Collapse
|
9
|
Fu Y, Yin M, Cao L, Lu Y, Li Y, Zhang L. Capsule mutations serve as a key strategy of phage resistance evolution of K54 hypervirulent Klebsiella pneumoniae. Commun Biol 2025; 8:257. [PMID: 39966630 PMCID: PMC11836320 DOI: 10.1038/s42003-025-07687-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
Phage therapy is a promising antibacterial strategy against the antibiotic resistance crisis. The evolved phage resistance could pose a big challenge to clinical phage therapy. Therefore, it is necessary to conduct a comprehensive analysis of phage resistance mechanisms during treatment. Here, we characterize 37 phage-resistant mutants of hypervirulent K. pneumoniae strain SCNJ1 under phage-imposed selection in both in vitro and in vivo experiments. We show that 97.3% (36/37) of phage-resistant clones possessed at least one mutation in genes related to the CPS biosynthesis. Notably, the wcaJ gene emerges as a mutation hotspot, as mutations in this gene are detected at a high frequency under both conditions. In contrast, mutations in wzc exhibit more association with in vivo samples. These CPS-related mutants all exhibit compromised bacterial fitness and attenuated virulence in mice. Strain CM8 is the only non-CPS-related mutant, which has a bglA mutation that confers phage resistance and retains full fitness and virulence. This study highlights that laboratory characterization of phage resistance evolution can give useful insights for clinical phage therapy.
Collapse
Affiliation(s)
- Yu Fu
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Ming Yin
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Li Cao
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Yanjun Lu
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Ying Li
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China.
| | - Luhua Zhang
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
10
|
Sewell AK, Cui M, Zhu M, Host MR, Han M. Enterobactin carries iron into Caenorhabditis elegans and mammalian intestinal cells by a mechanism independent of divalent metal transporter DMT1. J Biol Chem 2025; 301:108158. [PMID: 39761858 PMCID: PMC11815940 DOI: 10.1016/j.jbc.2025.108158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/17/2024] [Accepted: 12/28/2024] [Indexed: 01/15/2025] Open
Abstract
The diverse microbiota of the intestine is expected to benefit the host, yet the beneficial metabolites derived from the microbiota are still poorly understood. Enterobactin (Ent) is a well-known secreted iron-scavenging siderophore made by bacteria to fetch iron from the host or environment. Little was known about the positive role of Ent until a recent discovery in the nematode Caenorhabditis elegans indicated a beneficial role of Ent in promoting mitochondrial iron level in the animal intestine. To solidify this new paradigm, we further tested this role in C. elegans and multiple mammalian cell models and its relationship with the primary iron transporter DMT1/SMF-3 and several other iron-related genes. Here we show that ferric enterobactin (FeEnt) supplementation promotes whole organism development in C. elegans, increases iron uptake in caco-2 human intestinal epithelial cells, and supports iron-dependent differentiation of murine erythroid progenitor cells, indicating that the FeEnt complex can effectively enter these cells and be bioavailable. Our data in multiple models demonstrate that FeEnt-mediated iron transport is independent of all tested iron transporters. In addition, FeEnt supplementation robustly suppresses the developmental defects of a hif-1 mutant under low iron condition, suggesting the critical role in iron homeostasis for this well-known hypoxia regulator. These results suggest that FeEnt can effectively enter animal cells and their mitochondria through a previously unknown mechanism that may be leveraged as a therapeutic ferric iron carrier for the treatment of DMT1-or HIF-1-related iron deficiency and anemia.
Collapse
Affiliation(s)
- Aileen K Sewell
- Department of MCDB, University of Colorado Boulder, Boulder, Colorado, USA
| | - Mingxue Cui
- Department of MCDB, University of Colorado Boulder, Boulder, Colorado, USA
| | - Mengnan Zhu
- Department of MCDB, University of Colorado Boulder, Boulder, Colorado, USA
| | - Miranda R Host
- Department of MCDB, University of Colorado Boulder, Boulder, Colorado, USA
| | - Min Han
- Department of MCDB, University of Colorado Boulder, Boulder, Colorado, USA.
| |
Collapse
|
11
|
Keskey RC, Xiao J, Hyoju S, Lam A, Kim D, Sidebottom AM, Zaborin A, Dijkstra A, Meltzer R, Thakur A, Zhang K, Chen HJ, Beloborodova NV, Pautova AK, Wolfe K, Patel B, Thewissen R, Zaborina O, Alverdy JC. Enterobactin inhibits microbiota-dependent activation of AhR to promote bacterial sepsis in mice. Nat Microbiol 2025; 10:388-404. [PMID: 39779878 PMCID: PMC11905502 DOI: 10.1038/s41564-024-01882-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 11/13/2024] [Indexed: 01/11/2025]
Abstract
Sepsis is a major cause of morbidity and mortality, but our understanding of the mechanisms underlying survival or susceptibility is limited. Here, as pathogens often subvert host defence mechanisms, we hypothesized that this might influence the outcome of sepsis. We used microbiota analysis, faecal microbiota transplantation, antibiotic treatment and caecal metabolite analysis to show that gut-microbiota-derived tryptophan metabolites including indoles increased host survival in a mouse model of Serratia marcescens sepsis. Infection in macrophage-specific aryl hydrocarbon receptor (AhR) knockout mice revealed that AhR activation induced transcriptional reprogramming in macrophages and increased bacterial clearance and host survival. However, culture supernatants from multiple bacterial pathogens inhibited AhR activation in vitro. We showed that the secreted siderophore, enterobactin, inhibited AhR activation in vitro and increased sepsis mortality in vivo. By contrast, oral or systemic tryptophan supplementation increased survival. These findings show that sepsis survival depends upon the interplay between pathogen inhibition and the activation of AhR by a microbiota-derived metabolite.
Collapse
Affiliation(s)
- Robert C Keskey
- Section of General Surgery, Department of Surgery, University of Chicago, Chicago, IL, USA.
- Committee on Immunology, Biological Sciences Division, University of Chicago, Chicago, IL, USA.
| | - Jason Xiao
- Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Sanjiv Hyoju
- Section of General Surgery, Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Adam Lam
- Section of General Surgery, Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Daniel Kim
- Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Ashley M Sidebottom
- Host-Microbe Metabolomics Facility, Duchossois Family Institute, The University of Chicago, Chicago, IL, USA
| | - Alexander Zaborin
- Section of General Surgery, Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Anne Dijkstra
- Department of Surgery, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rebecca Meltzer
- Section of General Surgery, Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Abhimanyu Thakur
- Prtizker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Kui Zhang
- Prtizker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Huanhuan Joyce Chen
- Prtizker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Natalia V Beloborodova
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow, Russia
| | - Alisa K Pautova
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow, Russia
| | - Krysta Wolfe
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Bhakti Patel
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Renee Thewissen
- Host-Microbe Metabolomics Facility, Duchossois Family Institute, The University of Chicago, Chicago, IL, USA
| | - Olga Zaborina
- Section of General Surgery, Department of Surgery, University of Chicago, Chicago, IL, USA
| | - John C Alverdy
- Section of General Surgery, Department of Surgery, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
12
|
Chiou SL, Chang CY, Chu J. "Cofactors" for Natural Products. ChemMedChem 2025:e202400498. [PMID: 39822069 DOI: 10.1002/cmdc.202400498] [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: 07/01/2024] [Revised: 12/10/2024] [Accepted: 01/14/2025] [Indexed: 01/19/2025]
Abstract
Cofactors are non-protein entities necessary for proteins to operate. They provide "functional groups" beyond those of the 20 canonical amino acids and enable proteins to carry out more diverse functions. Such a viewpoint is rarely mentioned, if at all, when it comes to natural products and is the theme of this Concept. Even though the mechanisms of action (MOA) of only a few natural products are known to require cofactors, we believe that cofactor mediated MOA in natural products are far more prevalent than what we currently know. Bleomycin is a case in point. It binds iron cation to form a pseudoenzyme that generates reactive oxygen species. As another example, calcium cations induce laspartomycin to "fold" into the active conformation. Iron and calcium are bona fide cofactors for bleomycin and laspartomycin, respectively, as these natural products do not display their characteristic anticancer and antibacterial activities without Fe(II) and Ca(II). These types of cofactor mediated MOA in natural products were discovered mostly serendipitously, and being conscious of such a possibility is the first step toward identifying more novel chemistry that nature performs.
Collapse
Affiliation(s)
- Shao-Lun Chiou
- Department of Chemistry, National Taiwan University, 106319, Taipei City, Taiwan
| | - Chin-Yuan Chang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, 300193, Hsinchu City, Taiwan
| | - John Chu
- Department of Chemistry, National Taiwan University, 106319, Taipei City, Taiwan
| |
Collapse
|
13
|
Rózsa Á, Orosz L, Szemerédi N, Spengler G, Kecskeméti G, Vágó O, Sárvári KP, Szabó D, Szabó Z, Burián K, Virok DP. Bacteriophage Treatment Induces Phenotype Switching and Alters Antibiotic Resistance of ESBL Escherichia coli. Antibiotics (Basel) 2025; 14:76. [PMID: 39858362 PMCID: PMC11761592 DOI: 10.3390/antibiotics14010076] [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: 11/02/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Bacteriophage therapy represents a promising strategy to combat multidrug-resistant pathogens, such as Escherichia coli. In this study, we explored the effects of a bacteriophage infection on an Extended Spectrum Beta-Lactamase (ESBL) positive E. coli isolate. Methods: We used next generation sequencing, proteomics and phenotypic screens to investigate the effect of bacteriophage infections on E. coli metabolism and resistance phenotypes. Results: The bacteriophage infection led to notable alterations in colony morphology, indicating profound changes in bacterial metabolism. Proteomic analysis revealed significant shifts in protein expression, with 65 proteins upregulated and 246 downregulated post-infection. The downregulated proteins were involved in various metabolic pathways, including nucleic acid, protein and lipid metabolism, and iron acquisition. Bacteriophage treatment also led to increased bacterial membrane permeability. Altogether, these alterations in bacterial metabolism and membrane permeability may lead to a general reduction in antibiotic resistance. Indeed, the bacteriophage-infected E. coli exhibited increased sensitivity to various classes of antibiotics, including beta-lactams, fluoroquinolones, trimethoprim-sulfamethoxazole, and aminoglycosides. Conclusions: Our findings highlight the potential of bacteriophage therapy as an adjunct to existing antibiotics, enhancing their efficacy against resistant strains.
Collapse
Affiliation(s)
- Árpád Rózsa
- Pándy Kálmán County Hospital, Semmelweis Str. 1, H-5700 Gyula, Hungary; (Á.R.)
| | - László Orosz
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis Str. 6, H-6725 Szeged, Hungary (N.S.); (G.S.); (K.P.S.); (K.B.)
| | - Nikoletta Szemerédi
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis Str. 6, H-6725 Szeged, Hungary (N.S.); (G.S.); (K.P.S.); (K.B.)
| | - Gabriella Spengler
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis Str. 6, H-6725 Szeged, Hungary (N.S.); (G.S.); (K.P.S.); (K.B.)
| | - Gábor Kecskeméti
- Department of Medical Chemistry, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Dóm Sq. 8, H-6720 Szeged, Hungary; (G.K.); (Z.S.)
| | - Otília Vágó
- Pándy Kálmán County Hospital, Semmelweis Str. 1, H-5700 Gyula, Hungary; (Á.R.)
| | - Károly Péter Sárvári
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis Str. 6, H-6725 Szeged, Hungary (N.S.); (G.S.); (K.P.S.); (K.B.)
| | - Diana Szabó
- Department of Oto-Rhino-Laryngology and Head & Neck Surgery, University of Szeged, Tisza Lajos Str. 111, H-6724 Szeged, Hungary;
| | - Zoltán Szabó
- Department of Medical Chemistry, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Dóm Sq. 8, H-6720 Szeged, Hungary; (G.K.); (Z.S.)
| | - Katalin Burián
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis Str. 6, H-6725 Szeged, Hungary (N.S.); (G.S.); (K.P.S.); (K.B.)
| | - Dezső Péter Virok
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis Str. 6, H-6725 Szeged, Hungary (N.S.); (G.S.); (K.P.S.); (K.B.)
| |
Collapse
|
14
|
Motz RN, Anderson JK, Nolan EM. Re-evaluation of the C-Glucosyltransferase IroB Illuminates Its Ability to C-Glucosylate Non-native Triscatecholate Enterobactin Mimics. Biochemistry 2025; 64:224-237. [PMID: 39718537 DOI: 10.1021/acs.biochem.4c00581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
The pathogen-associated C-glucosyltransferase IroB is involved in the biosynthesis of salmochelins, C-glucosylated derivatives of enterobactin (Ent), which is a triscatecholate siderophore of enteric bacteria including Salmonella enterica and Escherichia coli. Here, we reassess the ability of IroB to C-glucosylate non-native triscatecholate mimics of Ent, which may have utility in the design and development of siderophore-based therapeutics and diagnostics. We establish TRENCAM (TC) and MECAM (MC), synthetic Ent analogs with tris(2-aminoethyl)amine- or mesitylene-derived backbones replacing the trilactone core of Ent, respectively, and their monoglucosylated congeners as substrates of IroB. Time course analyses and steady-state kinetic studies, which were performed under conditions that provide enhanced activity relative to prior studies, inform the substrate selectivity and catalytic efficiencies of this enzyme. We extend these findings to the preparation of a siderophore-antibiotic conjugate composed of monoglucosylated TC and ampicillin (MGT-Amp). Examination of its antibacterial activity and receptor specificity demonstrates that MGT-Amp targets pathogenicity because it shows specificty for the pathogen-associated outer membrane receptor IroN. Overall, our findings extend the biochemical characterization of IroB and its substrate scope and illustrate the ability to leverage a bacterial C-glucosyltransferase for non-native chemoenzymatic transformations along with potential applications of salmochelin mimics.
Collapse
Affiliation(s)
- Rachel N Motz
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Jaden K Anderson
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Elizabeth M Nolan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
15
|
Schalk IJ. Bacterial siderophores: diversity, uptake pathways and applications. Nat Rev Microbiol 2025; 23:24-40. [PMID: 39251840 DOI: 10.1038/s41579-024-01090-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2024] [Indexed: 09/11/2024]
Abstract
Iron is an essential nutrient for the growth, survival and virulence of almost all bacteria. To access iron, many bacteria produce siderophores, molecules with a high affinity for iron. Research has highlighted substantial diversity in the chemical structure of siderophores produced by bacteria, as well as remarkable variety in the molecular mechanisms involved in strategies for acquiring iron through these molecules. The metal-chelating properties of siderophores, characterized by their high affinity for iron and ability to chelate numerous other metals (albeit with lower affinity compared with iron), have also generated interest in diverse fields. Siderophores find applications in the environment, such as in bioremediation and agriculture, in which emerging and innovative strategies are being developed to address pollution and enhance nutrient availability for plants. Moreover, in medicine, siderophores could be used as a tool for novel antimicrobial therapies and medical imaging, as well as in haemochromatosis, thalassemia or cancer treatments. This Review offers insights into the diversity of siderophores, highlighting their potential applications in environmental and medical contexts.
Collapse
|
16
|
Zhou Z, Yu H, Wang G, Li M, Shi K. High antimony resistance strain Enterobacter sp. Z1 mediates biomineralization of antimony trioxide. ENVIRONMENT INTERNATIONAL 2025; 195:109237. [PMID: 39721567 DOI: 10.1016/j.envint.2024.109237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/03/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
The increasing antimony (Sb) contamination prevalence poses a concern owing to its toxicity and potential carcinogenic properties. However, mechanisms underlying the microbial conversion of soluble Sb into insoluble Sb minerals remain unclear. In the present study, Enterobacter sp. Z1 strain demonstrated remarkable resistance to antimony potassium tartrate [Sb(III)] (>250 mM) in R2A medium. Furthermore, Enterobacter sp. Z1 produced antimony trioxide (Sb2O3) via biomineralization during cultivation. Omics analysis revealed the upregulation of pyruvate metabolism and accumulation of DL-3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) in the presence of Sb(III).Using pyruvate as the sole carbon source in a chemically defined medium significantly enhanced Sb(III) biomineralization ratio from 20.8 % to 90.4 % compared with that using R2A medium. Additionally, reduced Sb(III) biomineralization and intracellular pH levels were observed following aceE gene knockout in Enterobacter sp. Z1. However, this impaired phenotype was rescued by complementing the aceE gene or introducing purified AceE into the bacterial lysates. Notably, AceE exhibited binding affinity for Sb(III). Our findings revealed the pyruvate-HMG-CoA pathway as the mechanism underlying Sb biomineralization, facilitating the release of Sb ions from tartrate and maintaining intracellular pH stability, thereby catalyzing Sb2O3 synthesis. This study provides insights into the Sb biogeochemical cycle.
Collapse
Affiliation(s)
- Zijie Zhou
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Hongbo Yu
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Gejiao Wang
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Mingshun Li
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Kaixiang Shi
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
17
|
Toribio-Celestino L, Calvo-Villamañán A, Herencias C, Alonso-Del Valle A, Sastre-Dominguez J, Quesada S, Mazel D, Rocha EPC, Fernández-Calvet A, San Millan A. A plasmid-chromosome crosstalk in multidrug resistant enterobacteria. Nat Commun 2024; 15:10859. [PMID: 39738078 DOI: 10.1038/s41467-024-55169-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 12/03/2024] [Indexed: 01/01/2025] Open
Abstract
Conjugative plasmids promote the dissemination and evolution of antimicrobial resistance in bacterial pathogens. However, plasmid acquisition can produce physiological alterations in the bacterial host, leading to potential fitness costs that determine the clinical success of bacteria-plasmid associations. In this study, we use a transcriptomic approach to characterize the interactions between a globally disseminated carbapenem resistance plasmid, pOXA-48, and a diverse collection of multidrug resistant (MDR) enterobacteria. Although pOXA-48 produces mostly strain-specific transcriptional alterations, it also leads to the common overexpression of a small chromosomal operon present in Klebsiella spp. and Citrobacter freundii strains. This operon includes two genes coding for a pirin and an isochorismatase family proteins (pfp and ifp), and shows evidence of horizontal mobilization across Proteobacteria species. Combining genetic engineering, transcriptomics, and CRISPRi gene silencing, we show that a pOXA-48-encoded LysR regulator is responsible for the plasmid-chromosome crosstalk. Crucially, the operon overexpression produces a fitness benefit in a pOXA-48-carrying MDR K. pneumoniae strain, suggesting that this crosstalk promotes the dissemination of carbapenem resistance in clinical settings.
Collapse
Affiliation(s)
| | | | - Cristina Herencias
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
- Centro de Investigación Biológica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | | | | | - Susana Quesada
- Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Didier Mazel
- Institut Pasteur, Université de Paris Cité, CNRS UMR3525, Bacterial Genome Plasticity, Paris, France
| | - Eduardo P C Rocha
- Institut Pasteur, Université de Paris Cité, CNRS UMR3525, Microbial Evolutionary Genomics, Paris, France
| | | | - Alvaro San Millan
- Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain.
- Centro de Investigación Biológica en Red de Epidemiología y Salud Pública, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
18
|
Sewell AK, Cui M, Zhu M, Host MR, Han M. Enterobactin carries iron into C. elegans and mammalian intestinal cells by a mechanism independent of divalent metal transporter DMT1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.20.629725. [PMID: 39763789 PMCID: PMC11702613 DOI: 10.1101/2024.12.20.629725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
The diverse microbiota of the intestine is expected to benefit the host, yet the beneficial metabolites derived from the microbiota are still poorly understood. Enterobactin (Ent) is a well-known secreted iron-scavenging siderophore made by bacteria to fetch iron from the host or environment. Little was known about a positive role of Ent until a recent discovery in the nematode C. elegans indicated a beneficial role of Ent in promoting mitochondrial iron level in the animal intestine. To solidify this new paradigm, we further tested this role in C. elegans and multiple mammalian cell models and its relationship with the primary iron transporter DMT1/SMF-3 and several other iron-related genes. Here we show that ferric enterobactin (FeEnt) supplementation promotes whole organism development in C. elegans, increases iron uptake in caco-2 human intestinal epithelial cells, and supports iron-dependent differentiation of murine erythroid progenitor cells, indicating that the FeEnt complex can effectively enter these cells and be bioavailable. Our data in multiple models demonstrate that FeEnt-mediated iron transport is independent of all tested iron transporters. In addition, FeEnt supplementation robustly suppresses the developmental defects of a hif-1 mutant under low iron condition, suggesting the critical role in iron homeostasis for this well-known hypoxia regulator. These results suggest that FeEnt can effectively enter animal cells and their mitochondria through a previously unknown mechanism that may be leveraged as a therapeutic ferric iron carrier for the treatment of DMT1- or HIF-1-related iron deficiency and anemia.
Collapse
Affiliation(s)
| | - Mingxue Cui
- Department of MCDB, University of Colorado Boulder
| | - Mengnan Zhu
- Department of MCDB, University of Colorado Boulder
| | | | - Min Han
- Department of MCDB, University of Colorado Boulder
| |
Collapse
|
19
|
Jiang J, Okuda S, Itoh H, Okamoto K, Nakanishi H, Suzuki M, Lu P, Nagata K. Structure-Guided Discovery of a Potent Inhibitor of the Ferric Citrate Binding Protein FecB in Vibrio Bacteria. Angew Chem Int Ed Engl 2024; 63:e202411688. [PMID: 39304960 DOI: 10.1002/anie.202411688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/22/2024]
Abstract
Infections caused by Gram-negative bacteria present a significant risk to human health worldwide. Novel strategies are needed to deal with the challenge caused by drug-resistant bacteria. Here, we report a new approach to combat infections by targeting iron-binding proteins to suppress bacterial growth. We investigated the function of the conserved periplasmic binding protein FecB from Vibrio alginolyticus. FecB was known to play a crucial role in the bacterial growth and to relate with biofilm formation. We then solved the crystal structures and elucidated the binding mechanism of FecB with ferric ion chelated by citrate. The results indicated that FecB binds weakly to one citrate molecule and strongly to the Fe3+-(citrate)2 complex. Based on these results, a structure-based virtual screening approach was conducted against FecB to identify small molecules that block ferric citrate uptake. Further evaluations in vivo and in vitro demonstrated that salvianolic acid C significantly suppressed bacterial growth, indicating that targeting bacterial nutrient absorption is a promising strategy for identifying potential antibacterial drugs.
Collapse
Affiliation(s)
- Jinyan Jiang
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Suguru Okuda
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Hideaki Itoh
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Ken Okamoto
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Hiromi Nakanishi
- Department of Global Agricultural Sciences, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Michio Suzuki
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Peng Lu
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
- Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, 828, Zhongxing Road, Xitang Town, Jiashan County, Jiaxing City, Zhejiang Province, 314100, China
| | - Koji Nagata
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
- Agricultural Bioinformatics Research Unit, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
- Research Center for Food Safety, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
20
|
Gräff ÁT, Barry SM. Siderophores as tools and treatments. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:47. [PMID: 39649077 PMCID: PMC11621027 DOI: 10.1038/s44259-024-00053-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/03/2024] [Indexed: 12/10/2024]
Abstract
In the search for iron, an essential element in many biochemical processes, microorganisms biosynthesise dedicated chelators, known as siderophores, to sequester iron from their environment and actively transport the siderophore complex into the cell. This process has been implicated in bacterial pathogenesis and exploited through siderophore-antibiotic conjugates as a method for selective antibiotic delivery. Here we review this Trojan-horse approach including design considerations and potential in diagnostics and infection imaging.
Collapse
Affiliation(s)
- Á. Tamás Gräff
- Department of Chemistry, Faculty of Natural, Mathematical and Engineering Sciences, King’s College London, Britannia House, London, SE1 1DB UK
| | - Sarah M. Barry
- Department of Chemistry, Faculty of Natural, Mathematical and Engineering Sciences, King’s College London, Britannia House, London, SE1 1DB UK
| |
Collapse
|
21
|
Yang C, Wang L, Lv J, Wen Y, Gao Q, Qian F, Tian X, Zhu J, Zhu Z, Chen L, Du H. Effects of different carbapenemase and siderophore production on cefiderocol susceptibility in Klebsiella pneumoniae. Antimicrob Agents Chemother 2024; 68:e0101924. [PMID: 39470196 PMCID: PMC11619314 DOI: 10.1128/aac.01019-24] [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: 07/09/2024] [Accepted: 09/23/2024] [Indexed: 10/30/2024] Open
Abstract
The resistance mechanism of Gram-negative bacteria to the siderophore antibiotic cefiderocol is primarily attributed to carbapenemase and siderophore uptake pathways; however, specific factors and their relationships remain to be fully elucidated. Here, we constructed cefiderocol-resistant Klebsiella pneumoniae (CRKP) strains carrying different carbapenemases and knocked out siderophore genes to investigate the roles of various carbapenemases and siderophores in the development of cefiderocol resistance. Antimicrobial susceptibility testing revealed that both blaNDM and blaKPC significantly increased the minimum inhibitory concentration (MIC) of Klebsiella pneumoniae (KP) to cefiderocol, while blaOXA-48 showed a modest increase. Notably, KP expressing NDM exhibited a higher cefiderocol MIC compared to KP expressing KPC, although expression of NDM alone did not induce cefiderocol resistance. Laboratory evolutionary experiments demonstrated that combining pNDM with mutations in the siderophore uptake receptor gene cirA and pKPC with a mutation in the two-component system gene envZ led to KP reaching a high level of cefiderocol resistance. Although combining pOXA with mutations in the two-component system gene baeS did not induce cefiderocol resistance, it significantly reduced susceptibility. Moreover, siderophores could influence the development of cefiderocol resistance. Strains deficient in enterobactin exhibited increased susceptibility to cefiderocol, while deficiencies in yersiniabactin and salmochelin showed no significant alterations. In conclusion, carbapenemase gene expression facilitates cefiderocol resistance, but its presence alone is insufficient. Cefiderocol resistance in CRKP typically involves abnormal expression of certain genes and other factors, such as mutations in siderophore uptake receptor genes and two-component system genes. The enterobactin siderophore synthesis gene entB may also contribute to resistance.
Collapse
Affiliation(s)
- Chengcheng Yang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jingnan Lv
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Key Laboratory of Alkene-Carbon Fibres-Based Technology and Application for Detection of Major Infectious Diseases, Suzhou, China
| | - Yicheng Wen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qizhao Gao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Feinan Qian
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiangxiang Tian
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jie Zhu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhichen Zhu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Chen
- Department of Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Hong Du
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Key Laboratory of Alkene-Carbon Fibres-Based Technology and Application for Detection of Major Infectious Diseases, Suzhou, China
| |
Collapse
|
22
|
Raviranga NGH, Ayinla M, Perera HA, Qi Y, Yan M, Ramström O. Antimicrobial Potency of Nor-Pyochelin Analogues and Their Cation Complexes against Multidrug-Resistant Pathogens. ACS Infect Dis 2024; 10:3842-3852. [PMID: 39469860 DOI: 10.1021/acsinfecdis.4c00421] [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] [Indexed: 10/30/2024]
Abstract
The opportunistic pathogen Pseudomonas aeruginosa develops increasing resistance toward even the most potent antibiotics. Like other bacteria, the pathogen produces a number of virulence factors including metallophores, which constitute an important group. Pseudomonads produce the iron-chelating metallophore (siderophore) pyochelin, which, in addition to its iron-scavenging ability, is an effector for the transcriptional regulator PchR in its FeIII-bound form (ferripyochelin). In the present study, docking studies predicted a major ferripyochelin binding site in PchR, which prompted the exploration of nor-pyochelin analogues to produce tight binding to PchR, and thereby upregulation of the pyochelin metabolism. In addition, we investigated the effects of using the analogues to bind the antimicrobial cations GaIII and InIII. Selected analogues of nor-pyochelin were synthesized, and their GaIII- and InIII-based complexes were assessed for antimicrobial activity. The results indicate that the GaIII complexes inhibit the pathogens under iron-limited conditions, while the InIII-based systems are more effective in iron-rich media. Several of the GaIII complexes were shown to be highly effective against a multidrug-resistant P. aeruginosa clinical isolate, with minimum inhibitory concentrations (MICs) of ≤1 μg/mL. Similarly, two of the InIII-based systems were particularly effective against the isolate, with an MIC of 8 μg/mL. These results show high promise in comparison with other, traditionally potent antibiotics, as the compounds generally indicated low cytotoxicity toward mammalian cells. Preliminary mechanistic investigations using pseudomonal transposon mutants suggested that the inhibitory effects of the InIII-based systems could be due to acute iron deficiency as a result of InIII-bound bacterioferritin.
Collapse
Affiliation(s)
- N G Hasitha Raviranga
- Department of Chemistry, University of Massachusetts Lowell, One University Avenue, Lowell, Massachusetts 01854, United States
| | - Mubarak Ayinla
- Department of Chemistry, University of Massachusetts Lowell, One University Avenue, Lowell, Massachusetts 01854, United States
| | - Harini A Perera
- Department of Chemistry, University of Massachusetts Lowell, One University Avenue, Lowell, Massachusetts 01854, United States
| | - Yunchuan Qi
- Department of Chemistry, University of Massachusetts Lowell, One University Avenue, Lowell, Massachusetts 01854, United States
| | - Mingdi Yan
- Department of Chemistry, University of Massachusetts Lowell, One University Avenue, Lowell, Massachusetts 01854, United States
| | - Olof Ramström
- Department of Chemistry, University of Massachusetts Lowell, One University Avenue, Lowell, Massachusetts 01854, United States
- Department of Chemistry and Biomedical Sciences, Linnaeus University, SE-39182 Kalmar, Sweden
| |
Collapse
|
23
|
Jiang Y, Shin HH, Park BS, Li Z. Potential siderophore-dependent mutualism in the harmful dinoflagellate Alexandrium pacificum (Group IV) and bacterium Photobacterium sp. TY1-4 under iron-limited conditions. HARMFUL ALGAE 2024; 139:102726. [PMID: 39567080 DOI: 10.1016/j.hal.2024.102726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/26/2024] [Accepted: 09/14/2024] [Indexed: 11/22/2024]
Abstract
Specific bacterial species induce algal blooms by producing growth-promoting substances, such as siderophores, under iron-limited conditions. However, the molecular mechanisms underlying these effects remain poorly understood. This study investigates the interactions between the harmful dinoflagellate Alexandrium pacificum (Group IV) and siderophore-producing bacteria, with a focus on iron acquisition facilitated by bacterial siderophores. During algal bloom seasons in the South Sea of Korea, Photobacterium sp. TY1-4 was isolated, which enhances A. pacificum cell density under iron-deficient conditions, TY1-4 can use the sterile exudates from A. pacificum as the sole source of carbon, suggesting a mutualistic relationship. Transcriptomic and genomic analyses revealed siderophore-mediated redox-based signaling and non-reductive pathways enhancing iron bioavailability. Photobacterium sp. TY1-4 initiates siderophore production through quorum sensing, whereas A. pacificum utilizes specific receptors and transporters for hydroxamate-type siderophores (ApFHUA and ApFHUC) to uptake iron. Three redox key iron-uptake genes were also identified in A. pacificum: membrane-bound ferroxidase ApFET3, high-affinity iron permease ApFTR1, and ferric-chelate reductases/oxidoreductases ApFRE1, with transcription levels inversely related to bioavailable iron. Increased iron bioavailability mediated by siderophores alleviates iron stress in A. pacificum, supporting its growth in iron-scarce environments. Additionally, A. pacificum co-cultured with Photobacterium sp. TY1-4 synthesized high-toxicity STXs, including GTX4, GTX2, and STX. These findings highlight the critical role of bacterial siderophores in iron binding and their potential impact on harmful algal bloom dynamics.
Collapse
Affiliation(s)
- Yue Jiang
- Biological Resource Center/Korean Collection for Type Cultures (KCTC), Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea; Department of Integrative Food Bioscience and Biotechnology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hyeon Ho Shin
- Division of Fisheries Life Science, Pukyong National University, Busan 48574, Republic of Korea
| | - Bum Soo Park
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Republic of Korea; Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul 04763, Republic of Korea; Research Institute for Convergence of Basic Science, Hanyang University, Seoul 04763, Republic of Korea; Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Republic of Korea.
| | - Zhun Li
- Biological Resource Center/Korean Collection for Type Cultures (KCTC), Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea.
| |
Collapse
|
24
|
Hershko Y, Slutzkin M, Barkan D, Adler A. Construction of core genome multi-locus sequence typing schemes for population structure analyses of Nocardia species. Res Microbiol 2024; 175:104246. [PMID: 39393617 DOI: 10.1016/j.resmic.2024.104246] [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: 07/21/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
Nocardia, a member of the Actinobacteria phylum, populates diverse habitats globally, with certain species being the cause of various clinical infections in humans. There is paucity of data regarding the population structure of this genus and of established genomic-based phylogenetic methods. We examined the whole genome sequences of 193 isolates spanning five major pathogenic Nocardia species sourced from public databases, encompassing diverse geographic regions. Using the chewBBACA pipeline, a species-specific core genome multilocus sequence typing (cgMLST) schema was created for N. cyriacigeorgica, N. farcinica, N. brasiliensis, N. wallacei, and N. abscessus. Additional genomic features that were examined included virulence factor (VF) profile, total length and open-reading frame count, the core genome length and core gene count, and GC content. Our findings indicated that: (i) N. brasiliensis diverges significantly from the other four species, underscoring its distinct evolutionary trajectory; (ii) the population structures of all species were polyclonal, with phylogenetic clustering occurring in the minority of isolates; (iii) clonal complexes were largely restricted to specific geographical locations, rather than demonstrating a global distribution, and (iv) initial evidence suggests no direct common-source transmission amongst the studied strains. Our study establishes a comprehensive genome-based phylogenetic methodology for population structure of Nocardia species.
Collapse
Affiliation(s)
- Yizhak Hershko
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Israel; Clinical Microbiology Laboratory, Tel Aviv Sourasky Medical Center, Israel.
| | - Matan Slutzkin
- Clinical Microbiology Laboratory, Tel Aviv Sourasky Medical Center, Israel
| | - Daniel Barkan
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Israel.
| | - Amos Adler
- Clinical Microbiology Laboratory, Tel Aviv Sourasky Medical Center, Israel; Department of Epidemiology and Preventive Medicine, School of Public Health, Faculty of Medicine, Tel Aviv University, Israel.
| |
Collapse
|
25
|
Peng Y, Moffat JG, DuPai C, Kofoed EM, Skippington E, Modrusan Z, Gloor SL, Clark K, Xu Y, Li S, Chen L, Liu X, Wu P, Harris SF, Wang S, Crawford TD, Li CS, Liu Z, Wai J, Tan MW. Differential effects of inosine monophosphate dehydrogenase (IMPDH/GuaB) inhibition in Acinetobacter baumannii and Escherichia coli. J Bacteriol 2024; 206:e0010224. [PMID: 39235234 PMCID: PMC11500612 DOI: 10.1128/jb.00102-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/25/2024] [Indexed: 09/06/2024] Open
Abstract
Inosine 5'-monophosphate dehydrogenase (IMPDH), known as GuaB in bacteria, catalyzes the rate-limiting step in de novo guanine biosynthesis and is conserved from humans to bacteria. We developed a series of potent inhibitors that selectively target GuaB over its human homolog. Here, we show that these GuaB inhibitors are bactericidal, generate phenotypic signatures that are distinct from other antibiotics, and elicit different time-kill kinetics and regulatory responses in two important Gram-negative pathogens: Acinetobacter baumannii and Escherichia coli. Specifically, the GuaB inhibitor G6 rapidly kills A. baumannii but only kills E. coli after 24 h. After exposure to G6, the expression of genes involved in purine biosynthesis and stress responses change in opposite directions while siderophore biosynthesis is downregulated in both species. Our results suggest that different species respond to GuaB inhibition using distinct regulatory programs and possibly explain the different bactericidal kinetics upon GuaB inhibition. The comparison highlights opportunities for developing GuaB inhibitors as novel antibiotics.IMPORTANCEA. baumannii is a priority bacterial pathogen for which development of new antibiotics is urgently needed due to the emergence of multidrug resistance. We recently developed a series of specific inhibitors against GuaB, a bacterial inosine 5'-monophosphate dehydrogenase, and achieved sub-micromolar minimum inhibitory concentrations against A. baumannii. GuaB catalyzes the rate-limiting step of de novo guanine biosynthesis and is highly conserved across bacterial pathogens. This study shows that inhibition of GuaB induced a bacterial morphological profile distinct from that of other classes of antibiotics, highlighting a novel mechanism of action. Moreover, our transcriptomic analysis showed that regulation of de novo purine biosynthesis and stress responses of A. baumannii upon GuaB inhibition differed significantly from that of E. coli.
Collapse
Affiliation(s)
- Yutian Peng
- Department of Infectious Diseases, Genentech Inc., South San Francisco, California, USA
| | - John G. Moffat
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Cory DuPai
- Department of Bioinformatics, Genentech Inc., South San Francisco, California, USA
| | - Eric M. Kofoed
- Department of Infectious Diseases, Genentech Inc., South San Francisco, California, USA
| | | | - Zora Modrusan
- Department of Proteomic and Genomic Technologies, Genentech Inc., South San Francisco, California, USA
| | - Susan L. Gloor
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Kevin Clark
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Yiming Xu
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Shuxuan Li
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California, USA
| | - Liuxi Chen
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California, USA
| | - Xingrong Liu
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California, USA
| | - Ping Wu
- Department of Structural Biology, Genentech Inc., South San Francisco, California, USA
| | - Seth F. Harris
- Department of Structural Biology, Genentech Inc., South San Francisco, California, USA
| | - Shumei Wang
- Department of Discovery Chemistry, Genentech Inc., South San Francisco, California, USA
| | - Terry D. Crawford
- Department of Discovery Chemistry, Genentech Inc., South San Francisco, California, USA
| | - Chun Sing Li
- WuXi AppTec Co., Ltd., Waigaoqiao Free Trade Zone, Shanghai, China
| | - Zhiguo Liu
- WuXi AppTec Co., Ltd., Waigaoqiao Free Trade Zone, Shanghai, China
| | - John Wai
- WuXi AppTec Co., Ltd., Waigaoqiao Free Trade Zone, Shanghai, China
| | - Man-Wah Tan
- Department of Infectious Diseases, Genentech Inc., South San Francisco, California, USA
| |
Collapse
|
26
|
Hamchand R, Wang K, Song D, Palm NW, Crawford JM. Mucosal sugars delineate pyrazine vs pyrazinone autoinducer signaling in Klebsiella oxytoca. Nat Commun 2024; 15:8902. [PMID: 39406708 PMCID: PMC11480411 DOI: 10.1038/s41467-024-53185-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024] Open
Abstract
Virulent Klebsiella oxytoca strains are associated with gut and lung pathologies, yet our understanding of the molecular signals governing pathogenesis remains limited. Here, we characterized a family of K. oxytoca pyrazine and pyrazinone autoinducers and explored their roles in microbial and host signaling. We identified the human mucin capping sugar Neu5Ac as a selective elicitor of leupeptin, a protease inhibitor prevalent in clinical lung isolates of K. oxytoca, and leupeptin-derived pyrazinone biosynthesis. Additionally, we uncovered a separate pyrazine pathway, regulated by general carbohydrate metabolism, derived from a broadly conserved PLP-dependent enzyme. While both pyrazine and pyrazinone signaling induce iron acquisition responses, including enterobactin biosynthesis, pyrazinone signaling enhances yersiniabactin virulence factor production and selectively activates the proinflammatory human histamine receptor H4 (HRH4). Our findings suggest that the availability of specific carbohydrates delineates distinct autoinducer pathways in K. oxytoca that may have differential effects on bacterial virulence and host immune responses.
Collapse
Affiliation(s)
- Randy Hamchand
- Department of Chemistry, Yale University, New Haven, CT, USA
- Institute of Biomolecular Design & Discovery, Yale University, West Haven, CT, USA
| | - Kevin Wang
- Institute of Biomolecular Design & Discovery, Yale University, West Haven, CT, USA
| | - Deguang Song
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Noah W Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Jason M Crawford
- Department of Chemistry, Yale University, New Haven, CT, USA.
- Institute of Biomolecular Design & Discovery, Yale University, West Haven, CT, USA.
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
27
|
Vollenweider V, Roncoroni F, Kümmerli R. Pyoverdine-antibiotic combination treatment: its efficacy and effects on resistance evolution in Escherichia coli. MICROLIFE 2024; 5:uqae021. [PMID: 39502382 PMCID: PMC11536758 DOI: 10.1093/femsml/uqae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/18/2024] [Accepted: 10/13/2024] [Indexed: 11/08/2024]
Abstract
Antibiotic resistance is a growing concern for global health, demanding innovative and effective strategies to combat pathogenic bacteria. Pyoverdines, iron-chelating siderophores produced by environmental Pseudomonas spp., present a novel class of promising compounds to induce growth arrest in pathogens through iron starvation. While we previously demonstrated the efficacy of pyoverdines as antibacterials, our understanding of how these molecules interact with antibiotics and impact resistance evolution remains unknown. Here, we investigated the propensity of three Escherichia coli strains to evolve resistance against pyoverdine, the cephalosporin antibiotic ceftazidime, and their combination. We used a naive E. coli wildtype strain and two isogenic variants carrying the bla TEM-1 β-lactamase gene on either the chromosome or a costly multicopy plasmid to explore the influence of genetic background on selection for resistance. We found that strong resistance against ceftazidime and weak resistance against pyoverdine evolved in all E. coli variants under single treatment. Ceftazidime resistance was linked to mutations in outer membrane porin genes (envZ and ompF), whereas pyoverdine resistance was associated with mutations in the oligopeptide permease (opp) operon. In contrast, ceftazidime resistance phenotypes were attenuated under combination treatment, especially for the E. coli variant carrying bla TEM-1 on the multicopy plasmid. Altogether, our results show that ceftazidime and pyoverdine interact neutrally and that pyoverdine as an antibacterial is particularly potent against plasmid-carrying E. coli strains, presumably because iron starvation compromises both cellular metabolism and plasmid replication.
Collapse
Affiliation(s)
- Vera Vollenweider
- Department of Quantitative Biomedicine, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Flavie Roncoroni
- Department of Quantitative Biomedicine, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Rolf Kümmerli
- Department of Quantitative Biomedicine, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
28
|
Han X, Yao J, He J, Liu H, Jiang Y, Zhao D, Shi Q, Zhou J, Hu H, Lan P, Zhou H, Li X. Clinical and laboratory insights into the threat of hypervirulent Klebsiella pneumoniae. Int J Antimicrob Agents 2024; 64:107275. [PMID: 39002700 DOI: 10.1016/j.ijantimicag.2024.107275] [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: 12/14/2023] [Revised: 06/15/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
Hypervirulent Klebsiella pneumoniae (hvKP) typically causes severe invasive infections affecting multiple sites in healthy individuals. In the past, hvKP was characterized by a hypermucoviscosity phenotype, susceptibility to antimicrobial agents, and its tendency to cause invasive infections in healthy individuals within the community. However, there has been an alarming increase in reports of multidrug-resistant hvKP, particularly carbapenem-resistant strains, causing nosocomial infections in critically ill or immunocompromised patients. This presents a significant challenge for clinical treatment. Early identification of hvKP is crucial for timely infection control. Notably, identifying hvKP has become confusing due to its prevalence in nosocomial settings and the limited predictive specificity of the hypermucoviscosity phenotype. Novel virulence predictors for hvKP have been discovered through animal models or machine learning algorithms, while standardization of identification criteria is still necessary. Timely source control and antibiotic therapy have been widely employed for the treatment of hvKP infections. Additionally, phage therapy is a promising alternative approach due to escalating antibiotic resistance. In summary, this narrative review highlights the latest research progress in the development, virulence factors, identification, epidemiology of hvKP, and treatment options available for hvKP infection.
Collapse
Affiliation(s)
- Xinhong Han
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Jiayao Yao
- Centre of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jintao He
- Department of Infectious Diseases, Sir Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, Zhejiang, China; Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haiyang Liu
- Centre of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yan Jiang
- Department of Infectious Diseases, Sir Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, Zhejiang, China; Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Dongdong Zhao
- Department of Infectious Diseases, Sir Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, Zhejiang, China; Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qiucheng Shi
- Department of Infectious Diseases, Sir Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, Zhejiang, China; Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Junxin Zhou
- Department of Infectious Diseases, Sir Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, Zhejiang, China; Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huangdu Hu
- Department of Infectious Diseases, Centre for General Practice Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Peng Lan
- Department of Infectious Diseases, Sir Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, Zhejiang, China; Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hua Zhou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Xi Li
- Centre of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
29
|
Nicotra D, Ghadamgahi F, Ghosh S, Anzalone A, Dimaria G, Mosca A, Massimino ME, Vetukuri RR, Catara V. Genomic insights and biocontrol potential of ten bacterial strains from the tomato core microbiome. FRONTIERS IN PLANT SCIENCE 2024; 15:1437947. [PMID: 39253574 PMCID: PMC11381245 DOI: 10.3389/fpls.2024.1437947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/05/2024] [Indexed: 09/11/2024]
Abstract
Introduction Despite their adverse environmental effects, modern agriculture relies heavily on agrochemicals to manage diseases and pests and enhance plant growth and productivity. Some of these functions could instead be fulfilled by endophytes from the plant microbiota, which have diverse activities beneficial for plant growth and health. Methods We therefore used a microbiome-guided top-down approach to select ten bacterial strains from different taxa in the core microbiome of tomato plants in the production chain for evaluation as potential bioinoculants. High-quality genomes for each strain were obtained using Oxford Nanopore long-read and Illumina short-read sequencing, enabling the dissection of their genetic makeup to identify phyto-beneficial traits. Results Bacterial strains included both taxa commonly used as biofertilizers and biocontrol agents (i.e. Pseudomonas and Bacillus) as well as the less studied genera Leclercia, Chryseobacterium, Glutamicibacter, and Paenarthorbacter. When inoculated in the tomato rhizosphere, these strains promoted plant growth and reduced the severity of Fusarium Crown and Root Rot and Bacterial Spot infections. Genome analysis yielded a comprehensive inventory of genes from each strain related to processes including colonization, biofertilization, phytohormones, and plant signaling. Traits directly relevant to fertilization including phosphate solubilization and acquisition of nitrogen and iron were also identified. Moreover, the strains carried several functional genes putatively involved in abiotic stress alleviation and biotic stress management, traits that indirectly foster plant health and growth. Discussion This study employs a top-down approach to identify new plant growth-promoting rhizobacteria (PGPRs), offering an alternative to the conventional bottom-up strategy. This method goes beyond the traditional screening of the strains and thus can expand the range of potential bioinoculants available for market application, paving the way to the use of new still underexplored genera.
Collapse
Affiliation(s)
- Daniele Nicotra
- Department of Agriculture, Food and Environment, University of Catania, Catania, Italy
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Farideh Ghadamgahi
- Department of Plant Breeding, Swedish University of Agricultural Sciences, Alnarp, Sweden
| | - Samrat Ghosh
- Department of Plant Breeding, Swedish University of Agricultural Sciences, Alnarp, Sweden
| | - Alice Anzalone
- Department of Agriculture, Food and Environment, University of Catania, Catania, Italy
| | - Giulio Dimaria
- Department of Agriculture, Food and Environment, University of Catania, Catania, Italy
| | - Alexandros Mosca
- Department of Agriculture, Food and Environment, University of Catania, Catania, Italy
| | - Maria Elena Massimino
- Department of Agriculture, Food and Environment, University of Catania, Catania, Italy
| | - Ramesh Raju Vetukuri
- Department of Plant Breeding, Swedish University of Agricultural Sciences, Alnarp, Sweden
| | - Vittoria Catara
- Department of Agriculture, Food and Environment, University of Catania, Catania, Italy
| |
Collapse
|
30
|
Liuu S, Damont A, Perret A, Firmesse O, Becher F, Lavison-Bompard G, Hueber A, Woods AS, Darii E, Fenaille F, Tabet JC. Origin and characterization of cyclodepsipeptides: Comprehensive structural approaches with focus on mass spectrometry analysis of alkali-cationized molecular species. MASS SPECTROMETRY REVIEWS 2024. [PMID: 39166474 DOI: 10.1002/mas.21904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 07/11/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024]
Abstract
Cyclodepsipeptides (CDPs) represent a huge family of chemically and structurally diverse molecules with a wide ability for molecular interactions. CDPs are cyclic peptide-related natural products made up of both proteinogenic and nonproteinogenic amino acids linked by amide and ester bonds. The combined use of different analytical methods is required to accurately determine their integral structures including stereochemistry, thus allowing deeper insights into their often-intriguing bioactivities and their possible usefulness. Our goal is to present the various methods developed to accurately characterize CDPs. Presently, Marfey's method and NMR (nuclear magnetic resonance) are still considered the best for characterizing CDP configuration. Nevertheless, electrospray-high resolution tandem mass spectrometry (ESI-HRMS/MS) is of great value for efficiently resolving CDP's composition and sequences. For instance, recent data shows that the fragmentation of cationized CDPs (e.g., [M + Li]+ and [M + Na]+) leads to selective cleavage of ester bonds and specific cationized product ions (b series) useful to get unprecedented sequence information. Thus, after a brief presentation of their structure, biological functions, and biosynthesis, we also provide a historic overview of these various analytical approaches as well as their advantages and limitations with a special emphasis on the emergence of methods based on HRMS/MS through recent fundamental works and applications.
Collapse
Affiliation(s)
- Sophie Liuu
- Staphylococcus, Bacillus & Clostridium (SBCL) unit, Laboratory for Food Safety, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), Université Paris-Est, Maisons-Alfort, France
| | - Annelaure Damont
- Université Paris-Saclay, CEA-INRAE, Laboratoire Innovations en Spectrométrie de Masse pour la Santé (LI-MS), DRF/Institut Joliot/DMTS/SPI, MetaboHUB, CEA Saclay, Gif sur Yvette, France
| | - Alain Perret
- Génomique métabolique, Genoscope, Institut François Jacob, CEA, CNRS, Univ Evry, Université Paris-Saclay, Evry, France
| | - Olivier Firmesse
- Staphylococcus, Bacillus & Clostridium (SBCL) unit, Laboratory for Food Safety, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), Université Paris-Est, Maisons-Alfort, France
| | - François Becher
- Université Paris-Saclay, CEA-INRAE, Laboratoire Innovations en Spectrométrie de Masse pour la Santé (LI-MS), DRF/Institut Joliot/DMTS/SPI, MetaboHUB, CEA Saclay, Gif sur Yvette, France
| | - Gwenaëlle Lavison-Bompard
- Pesticides and Marine Biotoxins (PBM) unit, Laboratory for Food Safety, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), Université Paris-Est, Maisons-Alfort, France
| | - Amandine Hueber
- Staphylococcus, Bacillus & Clostridium (SBCL) unit, Laboratory for Food Safety, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), Université Paris-Est, Maisons-Alfort, France
| | - Amina S Woods
- National Institute on Drug Abuse Intramural Research Program (NIDA IRP), National Institute of Health (NIH), Baltimore, Maryland, USA
- Johns Hopkins School of Medicine, Pharmacology and Molecular Sciences, Baltimore, Maryland, USA
| | - Ekaterina Darii
- Génomique métabolique, Genoscope, Institut François Jacob, CEA, CNRS, Univ Evry, Université Paris-Saclay, Evry, France
| | - François Fenaille
- Université Paris-Saclay, CEA-INRAE, Laboratoire Innovations en Spectrométrie de Masse pour la Santé (LI-MS), DRF/Institut Joliot/DMTS/SPI, MetaboHUB, CEA Saclay, Gif sur Yvette, France
| | - Jean-Claude Tabet
- Université Paris-Saclay, CEA-INRAE, Laboratoire Innovations en Spectrométrie de Masse pour la Santé (LI-MS), DRF/Institut Joliot/DMTS/SPI, MetaboHUB, CEA Saclay, Gif sur Yvette, France
- Faculté des Sciences et de l'Ingénierie, Institut Parisien de Chimie Moléculaire (IPCM), Sorbonne Université, Paris, France
| |
Collapse
|
31
|
Luo VC, Peczuh MW. Location, Location, Location: Establishing Design Principles for New Antibacterials from Ferric Siderophore Transport Systems. Molecules 2024; 29:3889. [PMID: 39202968 PMCID: PMC11357680 DOI: 10.3390/molecules29163889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/03/2024] Open
Abstract
This review strives to assemble a set of molecular design principles that enables the delivery of antibiotic warheads to Gram-negative bacterial targets (ESKAPE pathogens) using iron-chelating siderophores, known as the Trojan Horse strategy for antibiotic development. Principles are derived along two main lines. First, archetypical siderophores and their conjugates are used as case studies for native iron transport. They enable the consideration of the correspondence of iron transport and antibacterial target location. The second line of study charts the rationale behind the clinical antibiotic cefiderocol. It illustrates the potential versatility for the design of new Trojan Horse-based antibiotics. Themes such as matching the warhead to a location where the siderophore delivers its cargo (i.e., periplasm vs. cytoplasm), whether or not a cleavable linker is required, and the relevance of cheaters to the effectiveness and selectivity of new conjugates will be explored. The effort to articulate rules has identified gaps in the current understanding of iron transport pathways and suggests directions for new investigations.
Collapse
Affiliation(s)
| | - Mark W. Peczuh
- Department of Chemistry, University of Connecticut, 55 N. Eagleville Road, U3060, Storrs, CT 06269, USA;
| |
Collapse
|
32
|
Will V, Frey C, Normant V, Kuhn L, Chicher J, Volck F, Schalk IJ. The role of FoxA, FiuA, and FpvB in iron acquisition via hydroxamate-type siderophores in Pseudomonas aeruginosa. Sci Rep 2024; 14:18795. [PMID: 39138320 PMCID: PMC11322547 DOI: 10.1038/s41598-024-69152-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/01/2024] [Indexed: 08/15/2024] Open
Abstract
Siderophores are specialized molecules produced by bacteria and fungi to scavenge iron, a crucial nutrient for growth and metabolism. Catecholate-type siderophores are mainly produced by bacteria, while hydroxamates are mostly from fungi. This study investigates the capacity of nine hydroxamate-type siderophores from fungi and Streptomyces to facilitate iron acquisition by the human pathogen Pseudomonas aeruginosa. Growth assays under iron limitation and 55Fe incorporation tests showed that all nine siderophores promoted bacterial growth and iron transport. The study also aimed to identify the TonB-dependent transporters (TBDTs) involved in iron import by these siderophores. Using mutant strains lacking specific TBDT genes, it was found that iron is imported into P. aeruginosa cells by FpvB for coprogen, triacetylfusarinine, fusigen, ferrirhodin, and ferrirubin. Iron complexed by desferioxamine G is transported by FpvB and FoxA, ferricrocin-Fe and ferrichrycin-Fe by FpvB and FiuA, and rhodotoluric acid-Fe by FpvB, FiuA, and another unidentified TBDT. These findings highlight the effectiveness of hydroxamate-type siderophores in iron transport into P. aeruginosa and provide insights into the complex molecular mechanisms involved, which are important for understanding microbial interactions and ecological balance.
Collapse
Affiliation(s)
- Virginie Will
- CNRS, UMR7242, UMR7242, ESBS, University of Strasbourg, Bld Sébastien Brant, 67412, Illkirch, Strasbourg, France
- UMR7242, ESBS, University of Strasbourg, Bld Sébastien Brant, 67412, Illkirch, Strasbourg, France
| | - Chloé Frey
- CNRS, UMR7242, UMR7242, ESBS, University of Strasbourg, Bld Sébastien Brant, 67412, Illkirch, Strasbourg, France
- UMR7242, ESBS, University of Strasbourg, Bld Sébastien Brant, 67412, Illkirch, Strasbourg, France
| | - Vincent Normant
- CNRS, UMR7242, UMR7242, ESBS, University of Strasbourg, Bld Sébastien Brant, 67412, Illkirch, Strasbourg, France
- UMR7242, ESBS, University of Strasbourg, Bld Sébastien Brant, 67412, Illkirch, Strasbourg, France
| | - Lauriane Kuhn
- Institut de Biologie Moléculaire Et Cellulaire, CNRS, UAR1589, Plateforme Proteomique Strasbourg - Esplanade, 2 Allée Konrad Roentgen, 67084, Strasbourg Cedex, France
| | - Johana Chicher
- Institut de Biologie Moléculaire Et Cellulaire, CNRS, UAR1589, Plateforme Proteomique Strasbourg - Esplanade, 2 Allée Konrad Roentgen, 67084, Strasbourg Cedex, France
| | - Florian Volck
- CNRS, UMR7242, UMR7242, ESBS, University of Strasbourg, Bld Sébastien Brant, 67412, Illkirch, Strasbourg, France
- UMR7242, ESBS, University of Strasbourg, Bld Sébastien Brant, 67412, Illkirch, Strasbourg, France
| | - Isabelle J Schalk
- CNRS, UMR7242, UMR7242, ESBS, University of Strasbourg, Bld Sébastien Brant, 67412, Illkirch, Strasbourg, France.
- UMR7242, ESBS, University of Strasbourg, Bld Sébastien Brant, 67412, Illkirch, Strasbourg, France.
| |
Collapse
|
33
|
LeBlanc A, Wuest WM. Siderophores: A Case Study in Translational Chemical Biology. Biochemistry 2024; 63:1877-1891. [PMID: 39041827 PMCID: PMC11308372 DOI: 10.1021/acs.biochem.4c00276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024]
Abstract
Siderophores are metal-binding secondary metabolites that assist in iron homeostasis and have been of interest to the scientific community for the last half century. Foundational siderophore research has enabled several translational applications including siderophore-antibiotic and siderophore-peptide conjugates, identification of new antimicrobial targets, advances in disease imaging, and novel therapeutics. This review aims to connect the basic science research (biosynthesis, cellular uptake, gene regulation, and effects on homeostasis) of well-known siderophores with the successive translational application that results. Intertwined throughout are connections to the career of Christopher T. Walsh, his impact on the field of chemical biology, and the legacy of his trainees who continue to innovate.
Collapse
Affiliation(s)
- Andrew
R. LeBlanc
- Department of Chemistry, Emory
University, Atlanta, Georgia 30322, United States
| | - William M. Wuest
- Department of Chemistry, Emory
University, Atlanta, Georgia 30322, United States
| |
Collapse
|
34
|
Bin X, Pawelek PD. Evidence of isochorismate channeling between the Escherichia coli enterobactin biosynthetic enzymes EntC and EntB. Protein Sci 2024; 33:e5122. [PMID: 39031458 PMCID: PMC11258883 DOI: 10.1002/pro.5122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/21/2024] [Accepted: 07/10/2024] [Indexed: 07/22/2024]
Abstract
Enterobactin is a high-affinity iron chelator produced and secreted by Escherichia coli and Salmonella typhimurium to scavenge scarce extracellular Fe3+ as a micronutrient. EntC and EntB are the first two enzymes in the enterobactin biosynthetic pathway. Isochorismate, produced by EntC, is a substrate for EntB isochorismatase. By using a competing isochorismate-consuming enzyme (the E. coli SEPHCHC synthase MenD), we found in a coupled assay that residual EntB isochorismatase activity decreased as a function of increasing MenD concentration. In the presence of excess MenD, EntB isochorismatase activity was observed to decrease by 84%, indicative of partial EntC-EntB channeling (16%) of isochorismate. Furthermore, addition of glycerol to the assay resulted in an increase of residual EntB isochorismatase activity to approximately 25% while in the presence of excess MenD. These experimental outcomes supported the existence of a substrate channeling surface identified in a previously reported protein-docking model of the EntC-EntB complex. Two positively charged EntB residues (K21 and R196) that were predicted to electrostatically guide negatively charged isochorismate between the EntC and EntB active sites were mutagenized to determine their effects on substrate channeling. The EntB variants K21D and R196D exhibited a near complete loss of isochorismatase activity, likely due to electrostatic repulsion of the negatively charged isochorismate substrate. Variants K21A, R196A, and K21A/R196A retained partial EntB isochorismatase activity in the absence of EntC; in the presence of EntC, isochorismatase activity in all variants increased to near wild-type levels. The MenD competition assay of the variants revealed that while K21A channeled isochorismate as efficiently as wild-type EntB (~ 15%), the variants K21A/R196A and R196A exhibited an approximately 5-fold loss in observed channeling efficiency (~3%). Taken together, these results demonstrate that partial substrate channeling occurs between EntC and EntB via a leaky electrostatic tunnel formed upon dynamic EntC-EntB complex formation and that EntB R196 plays an essential role in isochorismate channeling.
Collapse
Affiliation(s)
- Xue Bin
- Department of Chemistry and BiochemistryConcordia UniversityMontrealQuebecCanada
| | - Peter D. Pawelek
- Department of Chemistry and BiochemistryConcordia UniversityMontrealQuebecCanada
| |
Collapse
|
35
|
Yazzie MT, Reitz ZL, Schmid R, Petras D, Aron AT. Native metabolomics for mass spectrometry-based siderophore discovery. Methods Enzymol 2024; 702:317-352. [PMID: 39155117 DOI: 10.1016/bs.mie.2024.07.001] [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] [Indexed: 08/20/2024]
Abstract
Microorganisms, plants, and animals alike have specialized acquisition pathways for obtaining metals, with microorganisms and plants biosynthesizing and secreting small molecule natural products called siderophores and metallophores with high affinities and specificities for iron or other non-iron metals, respectively. This chapter details a novel approach to discovering metal-binding molecules, including siderophores and metallophores, from complex samples ranging from microbial supernatants to biological tissue to environmental samples. This approach, called Native Metabolomics, is a mass spectrometry method in which pH adjustment and metal infusion post-liquid chromatography are interfaced with ion identity molecular networking (IIMN). This rule-based data analysis workflow that enables the identification of metal-binding species based on defined mass (m/z) offsets with the same chromatographic profiles and retention times. Ion identity molecular networking connects compounds that are structurally similar by their fragmentation pattern and species that are ion adducts of the same compound by chromatographic shape correlations. This approach has previously revealed new insights into metal binding metabolites, including that yersiniabactin can act as a biological zincophore (in addition to its known role as a siderophore), that the recently elucidated lepotchelin natural products are cyanobacterial metallophores, and that antioxidants in traditional medicine bind iron. Native metabolomics can be conducted on any liquid chromatography-mass spectrometry system to explore the binding of any metal or multiple metals simultaneously, underscoring the potential for this method to become an essential strategy for elucidating biological metal-binding molecules.
Collapse
Affiliation(s)
- Marquis T Yazzie
- Department of Chemistry and Biochemistry, University of Denver, Denver, CO, United States
| | - Zachary L Reitz
- Department of Ecology, Evolution and Marine Biology, University of California, Santa Barbara, CA, United States
| | - Robin Schmid
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czechia
| | - Daniel Petras
- Department of Biochemistry, University of California Riverside, Riverside, CA, United States; Interfaculty of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Allegra T Aron
- Department of Chemistry and Biochemistry, University of Denver, Denver, CO, United States.
| |
Collapse
|
36
|
Sun Y, Liu M, Sun W, Tang X, Zhou Y, Zhang J, Yang B. A Hemoglobin Bionics-Based System for Combating Antibiotic Resistance in Chronic Diabetic Wounds via Iron Homeostasis Regulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405002. [PMID: 38738270 DOI: 10.1002/adma.202405002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/09/2024] [Indexed: 05/14/2024]
Abstract
Owing to the increased tissue iron accumulation in patients with diabetes, microorganisms may activate high expression of iron-involved metabolic pathways, leading to the exacerbation of bacterial infections and disruption of systemic glucose metabolism. Therefore, an on-demand transdermal dosing approach that utilizes iron homeostasis regulation to combat antimicrobial resistance is a promising strategy to address the challenges associated with low administration bioavailability and high antibiotic resistance in treating infected diabetic wounds. Here, it is aimed to propose an effective therapy based on hemoglobin bionics to induce disturbances in bacterial iron homeostasis. The preferred "iron cargo" is synthesized by protoporphyrin IX chelated with dopamine and gallium (PDGa), and is delivered via a glucose/pH-responsive microneedle bandage (PDGa@GMB). The PDGa@GMB downregulates the expression levels of the iron uptake regulator (Fur) and the peroxide response regulator (perR) in Staphylococcus aureus, leading to iron nutrient starvation and oxidative stress, ultimately suppressing iron-dependent bacterial activities. Consequently, PDGa@GMB demonstrates insusceptibility to genetic resistance while maintaining sustainable antimicrobial effects (>90%) against resistant strains of both S. aureus and E. coli, and accelerates tissue recovery (<20 d). Overall, PDGa@GMB not only counteracts antibiotic resistance but also holds tremendous potential in mediating microbial-host crosstalk, synergistically attenuating pathogen virulence and pathogenicity.
Collapse
Affiliation(s)
- Yihan Sun
- Joint Laboratory of Opto, Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130021, P. R. China
- State Key Laboratory of Supramolecular Structure and Material, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
| | - Manxuan Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
| | - Weihong Sun
- Joint Laboratory of Opto, Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130021, P. R. China
- State Key Laboratory of Supramolecular Structure and Material, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
| | - Xiaoduo Tang
- Joint Laboratory of Opto, Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130021, P. R. China
- State Key Laboratory of Supramolecular Structure and Material, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
| | - Yanmin Zhou
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, P. R. China
| | - Junhu Zhang
- Joint Laboratory of Opto, Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130021, P. R. China
- State Key Laboratory of Supramolecular Structure and Material, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
| | - Bai Yang
- Joint Laboratory of Opto, Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130021, P. R. China
- State Key Laboratory of Supramolecular Structure and Material, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
| |
Collapse
|
37
|
Yan J, Zhou G, Ren R, Zhang X, Zhang N, Wang Z, Peng L, Yang Y. Siderophore-harboring gut bacteria and fecal siderophore genes for predicting the responsiveness of fecal microbiota transplantation for active ulcerative colitis. J Transl Med 2024; 22:589. [PMID: 38915068 PMCID: PMC11194913 DOI: 10.1186/s12967-024-05419-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/18/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Predictive markers for fecal microbiota transplantation (FMT) outcomes in patients with active ulcerative colitis (UC) are poorly defined. We aimed to investigate changes in gut microbiota pre- and post-FMT and to assess the potential value in determining the total copy number of fecal bacterial siderophore genes in predicting FMT responsiveness. METHODS Patients with active UC (Mayo score ≥ 3) who had undergone two FMT procedures were enrolled. Fecal samples were collected before and 8 weeks after each FMT session. Patients were classified into clinical response and non-response groups, based on their Mayo scores. The fecal microbiota profile was accessed using metagenomic sequencing, and the total siderophore genes copy number via quantitative real-time polymerase chain reaction. Additionally, we examined the association between the total siderophore genes copy number and FMT efficacy. RESULTS Seventy patients with UC had undergone FMT. The clinical response and remission rates were 50% and 10% after the first FMT procedure, increasing to 72.41% and 27.59% after the second FMT. The cumulative clinical response and clinical remission rates were 72.86% and 25.71%. Compared with baseline, the response group showed a significant increase in Faecalibacterium, and decrease in Enterobacteriaceae, consisted with the changes of the total bacterial siderophore genes copy number after the second FMT (1889.14 vs. 98.73 copies/ng, P < 0.01). Virulence factor analysis showed an enriched iron uptake system, especially bacterial siderophores, in the pre-FMT response group, with a greater contribution from Escherichia coli. The total baseline copy number was significantly higher in the response group than non-response group (1889.14 vs. 94.86 copies/ng, P < 0.01). A total baseline copy number cutoff value of 755.88 copies/ng showed 94.7% specificity and 72.5% sensitivity in predicting FMT responsiveness. CONCLUSIONS A significant increase in Faecalibacterium, and decrease in Enterobacteriaceae and the total fecal siderophore genes copy number were observed in responders after FMT. The siderophore genes and its encoding bacteria may be of predictive value for the clinical responsiveness of FMT to active ulcerative colitis.
Collapse
Affiliation(s)
- Jingshuang Yan
- School of Medicine, Nankai University, Tianjin, 300071, China
- Microbiota Laboratory and Microbiota Division, Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Guanzhou Zhou
- School of Medicine, Nankai University, Tianjin, 300071, China
- Microbiota Laboratory and Microbiota Division, Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Rongrong Ren
- Microbiota Laboratory and Microbiota Division, Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiaohan Zhang
- School of Medicine, Nankai University, Tianjin, 300071, China
- Microbiota Laboratory and Microbiota Division, Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Nana Zhang
- Microbiota Laboratory and Microbiota Division, Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zikai Wang
- Microbiota Laboratory and Microbiota Division, Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Lihua Peng
- Microbiota Laboratory and Microbiota Division, Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yunsheng Yang
- School of Medicine, Nankai University, Tianjin, 300071, China.
- Microbiota Laboratory and Microbiota Division, Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
- National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
38
|
Huang YJ, Yang MH, Lin LY, Liu J, Zang YP, Lin J, Chen WM. Exploring the Localization of Siderophore-Mediated Cargo Delivery in Gram-Negative Bacteria Using 3-Hydroxypyridin-4(1 H)-one-Fluorescein Probes. ACS Infect Dis 2024; 10:2303-2317. [PMID: 38725130 DOI: 10.1021/acsinfecdis.4c00287] [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] [Indexed: 06/15/2024]
Abstract
The design of siderophore-antibiotic conjugates is a promising strategy to overcome drug resistance in negative bacteria. However, accumulating studies have shown that only those antibiotics acting on the cell wall or cell membrane multiply their antibacterial effects when coupled with siderophores, while antibiotics acting on targets in the cytoplasm of bacteria do not show an obvious enhancement of their antibacterial effects when coupled with siderophores. To explore the causes of this phenomenon, we synthesized several conjugate probes using 3-hydroxypyridin-4(1H)-ones as siderophores and replacing the antibiotic cargo with 5-carboxyfluorescein (5-FAM) or malachite green (MG) cargo. By monitoring changes in the fluorescence intensity of FAM conjugate 20 in bacteria, the translocation of the conjugate across the outer membranes of Gram-negative pathogens was confirmed. Further, the use of the fluorogen activating protein(FAP)/MG system revealed that 3-hydroxypyridin-4(1H)-one-MG conjugate 26 was ultimately distributed mainly in the periplasm rather than being translocated into the cytosol of Escherichia coli and Pseudomonas aeruginosa PAO1. Additional mechanistic studies suggested that the uptake of the conjugate involved the siderophore-dependent iron transport pathway and the 3-hydroxypyridin-4(1H)-ones siderophore receptor-dependent mechanism. Meanwhile, we demonstrated that the conjugation of 3-hydroxypyridin-4(1H)-ones to the fluorescein 5-FAM can reduce the possibility of the conjugates crossing the membrane layers of mammalian Vero cells by passive diffusion, and the advantages of the mono-3-hydroxypyridin-4(1H)-ones as a delivery vehicle in the design of conjugates compared to the tri-3-hydroxypyridin-4(1H)-ones. Overall, this work reveals the localization rules of 3-hydroxypyridin-4(1H)-ones as siderophores to deliver the cargo into Gram-negative bacteria. It provides a theoretical basis for the subsequent design of siderophore-antibiotic conjugates, especially based on 3-hydroxypyridin-4(1H)-ones as siderophores.
Collapse
Affiliation(s)
- Yong-Jun Huang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| | - Ming-Han Yang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| | - Ling-Yin Lin
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| | - Jun Liu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| | - Yi-Peng Zang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| | - Jing Lin
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| | - Wei-Min Chen
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| |
Collapse
|
39
|
Jabborova D, Mamarasulov B, Davranov K, Enakiev Y, Bisht N, Singh S, Stoyanov S, Garg AP. Diversity and Plant Growth Properties of Rhizospheric Bacteria Associated with Medicinal Plants. Indian J Microbiol 2024; 64:409-417. [PMID: 39010983 PMCID: PMC11246357 DOI: 10.1007/s12088-024-01275-w] [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: 11/07/2023] [Accepted: 03/28/2024] [Indexed: 07/17/2024] Open
Abstract
Microbes in the rhizosphere play a significant role in the growth, development, and efficiency of plants and trees. The rhizospheric area's microbes are reliant on the soil's characteristics and the substances that the plants release. The majority of previous research on medicinal plants concentrated on their bioactive phytochemicals, but this is changing now that it is understood that a large proportion of phytotherapeutic substances are actually created by related microorganisms or through contact with their host. The roots of medicinal plants secrete a large number of secondary metabolites that determine the diversity of microbial communities in their rhizosphere. The dominant bacteria isolated from a variety of medicinal plants include various species of Bacillus, Rhizobium, Pseudomonas, Azotobacter, Burkholderia, Enterobacte, Microbacterium, Serratia, Burkholderia, and Beijerinckia. Actinobacteria also colonize the rhizosphere of medicinal plants that release low molecular weight organic solute that facilitate the solubilisation of inorganic phosphate. Root exudates of medicinal plants resist abiotic stress and accumulate in soil to produce autotoxic effects that exhibit strong obstacles to continuous cropping. Although having a vast bioresource that may be used in agriculture and modern medicine, medicinal plants' microbiomes are largely unknown. The purpose of this review is to (i) Present new insights into the plant microbiome with a focus on medicinal plants, (ii) Provide information about the components of medicinal plants derived from plants and microbes, and (iii) Discuss options for promoting plant growth and protecting plants for commercial cultivation of medicinal plants. The scientific community has paid a lot of attention to the use of rhizobacteria, particularly plant growth-promoting rhizobacteria (PGPR), as an alternative to chemical pesticides. By a variety of processes, these rhizobacteria support plant growth, manage plant pests, and foster resilience to a range of abiotic challenges. It also focuses on how PGPR inoculation affects plant growth and survival in stressful environments.
Collapse
Affiliation(s)
- Dilfuza Jabborova
- Institute of Genetics and Plant Experimental Biology, Uzbekistan Academy of Sciences, 111208 Qibray, Uzbekistan
- Faculty of Biology, National University of Uzbekistan, 100174 Tashkent, Uzbekistan
- School of Biological Engineering and Life Sciences, Shobhit Institute of Engineering and Technology (NAAC Accredited Grade 'A', Deemed to-be-University), NH-58, Modipuram, Meerut, 250110 India
| | - Bakhodir Mamarasulov
- Institute of Microbiology of the Academy of Sciences of Uzbekistan, 100128 Tashkent, Uzbekistan
| | - Kakhramon Davranov
- Institute of Microbiology of the Academy of Sciences of Uzbekistan, 100128 Tashkent, Uzbekistan
| | - Yuriy Enakiev
- Nikola Pushkarov Institute of Soil Science, Agrotechnologies and Plant Protection, Agricultural Academy, Sofia, Bulgaria
| | - Neha Bisht
- School of Biological Engineering and Life Sciences, Shobhit Institute of Engineering and Technology (NAAC Accredited Grade 'A', Deemed to-be-University), NH-58, Modipuram, Meerut, 250110 India
| | - Sachidanand Singh
- Department of Biotechnology School of Energy and Technology, Pandit Deendayal Energy University, Knowledge Corridor, Raisan Village, PDPU Rd, Gandhinagar, 382007 Gujarat India
| | - Svilen Stoyanov
- Dobrudzha College of Technology, Technical University of Varna, 9010 Varna, Bulgaria
| | - Amar P Garg
- Swami Vivekanand Subharti University, NH-58, Subhartipuram, Meerut, 250005 India
| |
Collapse
|
40
|
Sengupta S, Pabbaraja S, Mehta G. Natural products from the human microbiome: an emergent frontier in organic synthesis and drug discovery. Org Biomol Chem 2024; 22:4006-4030. [PMID: 38669195 DOI: 10.1039/d4ob00236a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Often referred to as the "second genome", the human microbiome is at the epicenter of complex inter-habitat biochemical networks like the "gut-brain axis", which has emerged as a significant determinant of cognition, overall health and well-being, as well as resistance to antibiotics and susceptibility to diseases. As part of a broader understanding of the nexus between the human microbiome, diseases and microbial interactions, whether encoded secondary metabolites (natural products) play crucial signalling roles has been the subject of intense scrutiny in the recent past. A major focus of these activities involves harvesting the genomic potential of the human microbiome via bioinformatics guided genome mining and culturomics. Through these efforts, an impressive number of structurally intriguing antibiotics, with enhanced chemical diversity vis-à-vis conventional antibiotics have been isolated from human commensal bacteria, thereby generating considerable interest in their total synthesis and expanding their therapeutic space for drug discovery. These developments augur well for the discovery of new drugs and antibiotics, particularly in the context of challenges posed by mycobacterial resistance and emerging new diseases. The current landscape of various synthetic campaigns and drug discovery initiatives on antibacterial natural products from the human microbiome is captured in this review with an intent to stimulate further activities in this interdisciplinary arena among the new generation.
Collapse
Affiliation(s)
- Saumitra Sengupta
- School of Chemistry, University of Hyderabad, Hyderabad-500046, India.
- Department of Organic Synthesis and Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad-500007, India
| | - Srihari Pabbaraja
- Department of Organic Synthesis and Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad-500007, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Goverdhan Mehta
- School of Chemistry, University of Hyderabad, Hyderabad-500046, India.
| |
Collapse
|
41
|
Xie B, Wei X, Wan C, Zhao W, Song R, Xin S, Song K. Exploring the Biological Pathways of Siderophores and Their Multidisciplinary Applications: A Comprehensive Review. Molecules 2024; 29:2318. [PMID: 38792179 PMCID: PMC11123847 DOI: 10.3390/molecules29102318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Siderophores are a class of small molecules renowned for their high iron binding capacity, essential for all life forms requiring iron. This article provides a detailed review of the diverse classifications, and biosynthetic pathways of siderophores, with a particular emphasis on siderophores synthesized via nonribosomal peptide synthetase (NRPS) and non-NRPS pathways. We further explore the secretion mechanisms of siderophores in microbes and plants, and their role in regulating bioavailable iron levels. Beyond biological functions, the applications of siderophores in medicine, agriculture, and environmental sciences are extensively discussed. These applications include biological pest control, disease treatment, ecological pollution remediation, and heavy metal ion removal. Through a comprehensive analysis of the chemical properties and biological activities of siderophores, this paper demonstrates their wide prospects in scientific research and practical applications, while also highlighting current research gaps and potential future directions.
Collapse
Affiliation(s)
| | | | | | | | | | - Shuquan Xin
- School of Life Science, Changchun Normal University, Changchun 130032, China; (B.X.); (X.W.); (C.W.); (W.Z.); (R.S.)
| | - Kai Song
- School of Life Science, Changchun Normal University, Changchun 130032, China; (B.X.); (X.W.); (C.W.); (W.Z.); (R.S.)
| |
Collapse
|
42
|
Jin T, Ren J, Bai B, Wu W, Cao Y, Meng J, Zhang L. Effects of Klebsiella michiganensis LDS17 on Codonopsis pilosula growth, rhizosphere soil enzyme activities, and microflora, and genome-wide analysis of plant growth-promoting genes. Microbiol Spectr 2024; 12:e0405623. [PMID: 38563743 PMCID: PMC11064500 DOI: 10.1128/spectrum.04056-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Codonopsis pilosula is a perennial herbaceous liana with medicinal value. It is critical to promote Codonopsis pilosula growth through effective and sustainable methods, and the use of plant growth-promoting bacteria (PGPB) is a promising candidate. In this study, we isolated a PGPB, Klebsiella michiganensis LDS17, that produced a highly active 1-aminocyclopropane-1-carboxylate deaminase from the Codonopsis pilosula rhizosphere. The strain exhibited multiple plant growth-promoting properties. The antagonistic activity of strain LDS17 against eight phytopathogenic fungi was investigated, and the results showed that strain LDS17 had obvious antagonistic effects on Rhizoctonia solani, Colletotrichum camelliae, Cytospora chrysosperma, and Phomopsis macrospore with growth inhibition rates of 54.22%, 49.41%, 48.89%, and 41.11%, respectively. Inoculation of strain LDS17 not only significantly increased the growth of Codonopsis pilosula seedlings but also increased the invertase and urease activities, the number of culturable bacteria, actinomycetes, and fungi, as well as the functional diversity of microbial communities in the rhizosphere soil of the seedlings. Heavy metal (HM) resistance tests showed that LDS17 is resistant to copper, zinc, and nickel. Whole-genome analysis of strain LDS17 revealed the genes involved in IAA production, siderophore synthesis, nitrogen fixation, P solubilization, and HM resistance. We further identified a gene (koyR) encoding a plant-responsive LuxR solo in the LDS17 genome. Klebsiella michiganensis LDS17 may therefore be useful in microbial fertilizers for Codonopsis pilosula. The identification of genes related to plant growth and HM resistance provides an important foundation for future analyses of the molecular mechanisms underlying the plant growth promotion and HM resistance of LDS17. IMPORTANCE We comprehensively evaluated the plant growth-promoting characteristics and heavy metal (HM) resistance ability of the LDS17 strain, as well as the effects of strain LDS17 inoculation on the Codonopsis pilosula seedling growth and the soil qualities in the Codonopsis pilosula rhizosphere. We conducted whole-genome analysis and identified lots of genes and gene clusters contributing to plant-beneficial functions and HM resistance, which is critical for further elucidating the plant growth-promoting mechanism of strain LDS17 and expanding its application in the development of plant growth-promoting agents used in the environment under HM stress.
Collapse
Affiliation(s)
- Tingting Jin
- Department of Life Sciences, Changzhi University, Changzhi, China
| | - Jiahong Ren
- Department of Life Sciences, Changzhi University, Changzhi, China
| | - Bianxia Bai
- Department of Life Sciences, Changzhi University, Changzhi, China
| | - Wei Wu
- Department of Life Sciences, Changzhi University, Changzhi, China
| | - Yongqing Cao
- Department of Life Sciences, Changzhi University, Changzhi, China
| | - Jing Meng
- Department of Life Sciences, Changzhi University, Changzhi, China
| | - Lihui Zhang
- Department of Life Sciences, Changzhi University, Changzhi, China
| |
Collapse
|
43
|
Pantaleoni A, Sarasini F, Russo P, Passaro J, Giorgini L, Bavasso I, Santarelli ML, Petrucci E, Valentini F, Bracciale MP, Marrocchi A. Facile and Bioinspired Approach from Gallic Acid for the Synthesis of Biobased Flame Retardant Coatings of Basalt Fibers. ACS OMEGA 2024; 9:19099-19107. [PMID: 38708227 PMCID: PMC11064428 DOI: 10.1021/acsomega.3c10129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/28/2024] [Accepted: 04/09/2024] [Indexed: 05/07/2024]
Abstract
A sustainable, bioinspired approach to functionalize basalt fibers with an innovative gallic acid (GA)-iron phenyl phosphonate complex (BF-GA-FeP), for the purpose of improving the flame retardancy in composite materials, is developed. BFs were at first pretreated with O3, obtaining surface free hydroxyl groups that allowed the subsequent covalent immobilization of biosourced GA units on the fiber through ester linkages. Phenolic -OH groups of the GA units were then exploited for the complexation of iron phenyl phosphonate, resulting in the target-complex-coated BF fiber (BF-GA-FeP). Microwave plasma atomic emission spectroscopy and scanning electron microscopy coupled with energy-dispersive X-ray spectroscopy analyses of BF-GA-FeP highlighted an increase in iron content, modification of fiber morphology, and occurrence of phosphorus, respectively. BFs, modified with a low amount of the developed complex, were used to reinforce a poly(lactic acid) (PLA) matrix in the production of a biocomposite (PLA/BF-FeP). PLA/BF-FeP showed a higher thermal stability than neat PLA and PLA reinforced with untreated BFs (PLA/BF), as confirmed by thermogravimetric analysis. The cone calorimeter test highlighted several advantages for PLA/BF-FeP, including a prolonged time to ignition, a reduced time to flame out, an 8% decrease in the peak heat release rate, and a 15% reduced fire propagating index compared to PLA/BF.
Collapse
Affiliation(s)
- Alessia Pantaleoni
- Department
of Chemical Engineering Materials Environment, Sapienza University of Rome, Via Eudossiana 18, Rome 00184, Italy
| | - Fabrizio Sarasini
- Department
of Chemical Engineering Materials Environment, Sapienza University of Rome, Via Eudossiana 18, Rome 00184, Italy
| | - Pietro Russo
- Institute
for Polymers, Composites and Biomaterials, National Research Council, Via Campi Flegrei 34, Pozzuoli, NA 80078, Italy
| | - Jessica Passaro
- Institute
for Polymers, Composites and Biomaterials, National Research Council, Via Campi Flegrei 34, Pozzuoli, NA 80078, Italy
| | - Loris Giorgini
- Department
of Industrial Chemistry “Toso Montanari”, University of Bologna, Viale Risorgimento 4, Bologna 40136, Italy
| | - Irene Bavasso
- Department
of Chemical Engineering Materials Environment, Sapienza University of Rome, Via Eudossiana 18, Rome 00184, Italy
| | - Maria Laura Santarelli
- Department
of Chemical Engineering Materials Environment, Sapienza University of Rome, Via Eudossiana 18, Rome 00184, Italy
| | - Elisabetta Petrucci
- Department
of Chemical Engineering Materials Environment, Sapienza University of Rome, Via Eudossiana 18, Rome 00184, Italy
| | - Federica Valentini
- Department
of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto 8, Perugia 06123, Italy
| | - Maria Paola Bracciale
- Department
of Chemical Engineering Materials Environment, Sapienza University of Rome, Via Eudossiana 18, Rome 00184, Italy
| | - Assunta Marrocchi
- Department
of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto 8, Perugia 06123, Italy
| |
Collapse
|
44
|
Liu F, Kou Q, Li H, Cao Y, Chen M, Meng X, Zhang Y, Wang T, Wang H, Zhang D, Yang Y. Discovery of YFJ-36: Design, Synthesis, and Antibacterial Activities of Catechol-Conjugated β-Lactams against Gram-Negative Bacteria. J Med Chem 2024; 67:6705-6725. [PMID: 38596897 DOI: 10.1021/acs.jmedchem.4c00265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Cefiderocol is the first approved catechol-conjugated cephalosporin against multidrug-resistant Gram-negative bacteria, while its application was limited by poor chemical stability associated with the pyrrolidinium linker, moderate potency against Klebsiella pneumoniae and Acinetobacter baumannii, intricate procedures for salt preparation, and potential hypersensitivity. To address these issues, a series of novel catechol-conjugated derivatives were designed, synthesized, and evaluated. Extensive structure-activity relationships and structure-metabolism relationships (SMR) were conducted, leading to the discovery of a promising compound 86b (Code no. YFJ-36) with a new thioether linker. 86b exhibited superior and broad-spectrum in vitro antibacterial activity, especially against A. baumannii and K. pneumoniae, compared with cefiderocol. Potent in vivo efficacy was observed in a murine systemic infection model. Furthermore, the physicochemical stability of 86b in fluid medium at pH 6-8 was enhanced. 86b also reduced potential the risk of allergy owing to the quaternary ammonium linker. The improved properties of 86b supported its further research and development.
Collapse
Affiliation(s)
- Fangjun Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Qunhuan Kou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Hongyuan Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Yangzhi Cao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Meng Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Xin Meng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yinyong Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Ting Wang
- Department of Microbiology, Sichuan Primed Bio-Tech Group Co., Ltd., Chengdu, Sichuan Province 610041, P. R. China
| | - Hui Wang
- China Pharmaceutical University, Jiangsu 211198, China
| | - Dan Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yushe Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| |
Collapse
|
45
|
Abstract
Covering: 1997 to 2023The shikimate pathway is the metabolic process responsible for the biosynthesis of the aromatic amino acids phenylalanine, tyrosine, and tryptophan. Seven metabolic steps convert phosphoenolpyruvate (PEP) and erythrose 4-phosphate (E4P) into shikimate and ultimately chorismate, which serves as the branch point for dedicated aromatic amino acid biosynthesis. Bacteria, fungi, algae, and plants (yet not animals) biosynthesize chorismate and exploit its intermediates in their specialized metabolism. This review highlights the metabolic diversity derived from intermediates of the shikimate pathway along the seven steps from PEP and E4P to chorismate, as well as additional sections on compounds derived from prephenate, anthranilate and the synonymous aminoshikimate pathway. We discuss the genomic basis and biochemical support leading to shikimate-derived antibiotics, lipids, pigments, cofactors, and other metabolites across the tree of life.
Collapse
Affiliation(s)
- Vikram V Shende
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Katherine D Bauman
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Bradley S Moore
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, 92093, USA.
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
46
|
Li Y, Zhang Y, Sun X, Wu Y, Yan Z, Ju X, Huang Y, Zhou H, Wang Z, Wang S, Zhang R, Li R. National genomic epidemiology investigation revealed the spread of carbapenem-resistant Escherichia coli in healthy populations and the impact on public health. Genome Med 2024; 16:57. [PMID: 38627827 PMCID: PMC11020349 DOI: 10.1186/s13073-024-01310-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 03/04/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Carbapenem-resistant Escherichia coli (CREC) has been considered as WHO priority pathogens, causing a great public health concern globally. While CREC from patients has been thoroughly investigated, the prevalence and underlying risks of CREC in healthy populations have been overlooked. Systematic research on the prevalence of CREC in healthy individuals was conducted here. We aimed to characterize CREC collected from healthy populations in China between 2020 and 2022 and to compare the genomes of CREC isolates isolated from healthy individuals and clinical patients. METHODS We present a nationwide investigation of CREC isolates among healthy populations in China, employing robust molecular and genomic analyses. Antimicrobial susceptibility testing, whole-genome sequencing, and bioinformatics were utilized to analyze a cohort of CREC isolates (n = 113) obtained from fecal samples of 5 064 healthy individuals. Representative plasmids were extracted for third-generation nanopore sequencing. We previously collected 113 non-duplicate CREC isolates (59 in 2018, 54 in 2020) collected from ICU patients in 15 provinces and municipalities in China, and these clinical isolates were used to compare with the isolates in this study. Furthermore, we employ comparative genomics approaches to elucidate molecular variations and potential correlations between clinical and non-clinical CREC isolates. RESULTS A total of 147 CREC isolates were identified from 5 064 samples collected across 11 provinces in China. These isolates were classified into 64 known sequence types (STs), but no dominant STs were observed. In total, seven carbapenemase genes were detected with blaNDM-5 (n = 116) being the most prevalent one. Genetic environments and plasmid backbones of blaNDM were conserved in CREC isolated from healthy individuals. Furthermore, we compared clinical and healthy human-originated CRECs, revealing noteworthy distinctions in 23 resistance genes, including blaNDM-1, blaNDM-5, and blaKPC (χ2 test, p < 0.05). Clinical isolates contained more virulence factors associated with iron uptake, adhesion, and invasion than those obtained from healthy individuals. Notably, CREC isolates generally found healthy people are detected in hospitalized patients. CONCLUSIONS Our findings underscore the significance of healthy populations-derived CRECs as a crucial reservoir of antibiotic resistance genes (ARGs). This highlights the need for ongoing monitoring of CREC isolates in healthy populations to accurately assess the potential risks posed by clinical CREC isolates.
Collapse
Affiliation(s)
- Yan Li
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China
| | - Yanyan Zhang
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, P. R. China
| | - Xinran Sun
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China
| | - Yuchen Wu
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, P. R. China
| | - Zelin Yan
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, P. R. China
| | - Xiaoyang Ju
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, P. R. China
| | - Yonglu Huang
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, P. R. China
| | - Hongwei Zhou
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, P. R. China
| | - Zhiqiang Wang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China
- Jiangsu Key Lab of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, P. R. China
| | - Shaolin Wang
- College of Veterinary Medicine, China Agricultural University, Beijing, P. R. China
| | - Rong Zhang
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, P. R. China.
| | - Ruichao Li
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China.
- Jiangsu Key Lab of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, P. R. China.
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu, P. R. China.
| |
Collapse
|
47
|
Guo C, Nolan EM. Exploring the Antibacterial Activity and Cellular Fates of Enterobactin-Drug Conjugates That Target Gram-Negative Bacterial Pathogens. Acc Chem Res 2024; 57:1046-1056. [PMID: 38483177 PMCID: PMC11258919 DOI: 10.1021/acs.accounts.3c00814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Siderophores are secondary metabolites utilized by bacteria to acquire iron (Fe), an essential transition metal nutrient. Fe levels in the host environment are tightly regulated and can be further restricted to starve invading bacterial pathogens in a host-defense process known as nutritional immunity. To survive and colonize the Fe-limited host environment, bacteria produce siderophores and express cognate siderophore transport machinery. These active transport pathways present an opportunity for selective and efficient drug delivery into bacterial cells, motivating decades of research on synthetic siderophore-antibiotic conjugates (SACs) as a Trojan-horse strategy for the development of targeted antibiotics.Enterobactin (Ent) is a triscatecholate siderophore produced and utilized by many Gram-negative bacteria, including all Escherichia coli and Salmonella species. Within these species, pathogenic strains cause a variety of human diseases including urinary tract infections, gastroenteritis, and sepsis. Infections caused by these Gram-negative pathogens can be difficult to treat because of the impermeability of the outer membrane (OM). This impermeability can be overcome by utilizing siderophores as drug delivery vectors for targeting Gram-negative pathogens. Ent is a promising delivery vector because it undergoes active transport across the OM mediated by the Ent uptake machinery after scavenging Fe(III) from the extracellular environment. Despite the well-elucidated chemistry and biology of Ent, its use for SAC development was hampered by the lack of an appropriate functional group for cargo attachment. Our laboratory addressed this need by designing and synthesizing monofunctionalized Ent scaffolds. Over the past decade, we have used these scaffolds to explore Ent-based SACs with a variety of drug warheads, including β-lactam and fluoroquinolone antibiotics, and Pt(IV) prodrugs. Investigations of the antibacterial activities of these conjugates and their cellular fates have informed our design principles and revealed approaches to achieving enhanced antibacterial potency and pathogen-targeted activity. Collectively, our studies of Ent-drug conjugates have provided discoveries, understanding, and invaluable insights for future design and evaluation of SACs.In this Account, we present the story of our work on Ent-drug conjugates that began about ten years ago with the development of monofunctionalized Ent scaffolds and the design and synthesis of various conjugates based on these scaffolds. We describe the antibacterial activity profiles and uptake pathways of Ent-drug conjugates harboring traditional antibiotics and repurposed platinum anticancer agents as well as studies that address cellular targets and fates. Finally, we discuss other applications of monofunctionalized Ent scaffolds, including a siderophore-based immunization strategy. We intend for this Account to inspire further investigations into the fundamental understanding and translational applications of siderophores and siderophore-drug conjugates.
Collapse
Affiliation(s)
- Chuchu Guo
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Elizabeth M. Nolan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
48
|
Sun L, David KT, Wolters JF, Karlen SD, Gonçalves C, Opulente DA, LaBella AL, Groenewald M, Zhou X, Shen XX, Rokas A, Hittinger CT. Functional and Evolutionary Integration of a Fungal Gene With a Bacterial Operon. Mol Biol Evol 2024; 41:msae045. [PMID: 38415839 PMCID: PMC11043216 DOI: 10.1093/molbev/msae045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 02/29/2024] Open
Abstract
Siderophores are crucial for iron-scavenging in microorganisms. While many yeasts can uptake siderophores produced by other organisms, they are typically unable to synthesize siderophores themselves. In contrast, Wickerhamiella/Starmerella (W/S) clade yeasts gained the capacity to make the siderophore enterobactin following the remarkable horizontal acquisition of a bacterial operon enabling enterobactin synthesis. Yet, how these yeasts absorb the iron bound by enterobactin remains unresolved. Here, we demonstrate that Enb1 is the key enterobactin importer in the W/S-clade species Starmerella bombicola. Through phylogenomic analyses, we show that ENB1 is present in all W/S clade yeast species that retained the enterobactin biosynthetic genes. Conversely, it is absent in species that lost the ent genes, except for Starmerella stellata, making this species the only cheater in the W/S clade that can utilize enterobactin without producing it. Through phylogenetic analyses, we infer that ENB1 is a fungal gene that likely existed in the W/S clade prior to the acquisition of the ent genes and subsequently experienced multiple gene losses and duplications. Through phylogenetic topology tests, we show that ENB1 likely underwent horizontal gene transfer from an ancient W/S clade yeast to the order Saccharomycetales, which includes the model yeast Saccharomyces cerevisiae, followed by extensive secondary losses. Taken together, these results suggest that the fungal ENB1 and bacterial ent genes were cooperatively integrated into a functional unit within the W/S clade that enabled adaptation to iron-limited environments. This integrated fungal-bacterial circuit and its dynamic evolution determine the extant distribution of yeast enterobactin producers and cheaters.
Collapse
Affiliation(s)
- Liang Sun
- DOE Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI 53726, USA
- Laboratory of Genetics, Center for Genomic Science Innovation, Wisconsin Energy Institute, J. F. Crow Institute for the Study of Evolution, University of Wisconsin-Madison, Madison, WI 53726, USA
| | - Kyle T David
- Evolutionary Studies Initiative and Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA
| | - John F Wolters
- DOE Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI 53726, USA
- Laboratory of Genetics, Center for Genomic Science Innovation, Wisconsin Energy Institute, J. F. Crow Institute for the Study of Evolution, University of Wisconsin-Madison, Madison, WI 53726, USA
| | - Steven D Karlen
- DOE Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI 53726, USA
| | - Carla Gonçalves
- Laboratory of Genetics, Center for Genomic Science Innovation, Wisconsin Energy Institute, J. F. Crow Institute for the Study of Evolution, University of Wisconsin-Madison, Madison, WI 53726, USA
- Evolutionary Studies Initiative and Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA
- UCIBIO, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Dana A Opulente
- DOE Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI 53726, USA
- Laboratory of Genetics, Center for Genomic Science Innovation, Wisconsin Energy Institute, J. F. Crow Institute for the Study of Evolution, University of Wisconsin-Madison, Madison, WI 53726, USA
- Biology Department, Villanova University, Villanova, PA 19085, USA
| | - Abigail Leavitt LaBella
- Evolutionary Studies Initiative and Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | | | - Xiaofan Zhou
- Evolutionary Studies Initiative and Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Center, South China Agricultural University, Guangzhou 510642, China
| | - Xing-Xing Shen
- Evolutionary Studies Initiative and Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA
- College of Agriculture and Biotechnology and Centre for Evolutionary & Organismal Biology, Zhejiang University, Hangzhou 310058, China
| | - Antonis Rokas
- Evolutionary Studies Initiative and Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA
| | - Chris Todd Hittinger
- DOE Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI 53726, USA
- Laboratory of Genetics, Center for Genomic Science Innovation, Wisconsin Energy Institute, J. F. Crow Institute for the Study of Evolution, University of Wisconsin-Madison, Madison, WI 53726, USA
| |
Collapse
|
49
|
Motz RN, Guo C, Sargun A, Walker GT, Sassone-Corsi M, Raffatellu M, Nolan EM. Conjugation to Native and Nonnative Triscatecholate Siderophores Enhances Delivery and Antibacterial Activity of a β-Lactam to Gram-Negative Bacterial Pathogens. J Am Chem Soc 2024; 146:7708-7722. [PMID: 38457782 PMCID: PMC11037102 DOI: 10.1021/jacs.3c14490] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2024]
Abstract
Developing new antibiotics and delivery strategies is of critical importance for treating infections caused by Gram-negative bacterial pathogens. Hijacking bacterial iron uptake machinery, such as that of the siderophore enterobactin (Ent), represents one promising approach toward these goals. Here, we report a novel Ent-inspired siderophore-antibiotic conjugate (SAC) employing an alternative siderophore moiety as the delivery vector and demonstrate the potency of our SACs harboring the β-lactam antibiotic ampicillin (Amp) against multiple pathogenic Gram-negative bacterial strains. We establish the ability of N,N',N''-(nitrilotris(ethane-2,1-diyl))tris(2,3-dihydroxybenzamide) (TRENCAM, hereafter TC), a synthetic mimic of Ent, to facilitate drug delivery across the outer membrane (OM) of Gram-negative pathogens. Conjugation of Amp to a new monofunctionalized TC scaffold affords TC-Amp, which displays markedly enhanced antibacterial activity against the gastrointestinal pathogen Salmonella enterica serovar Typhimurium (STm) compared with unmodified Amp. Bacterial uptake, antibiotic susceptibility, and microscopy studies with STm show that the TC moiety facilitates TC-Amp uptake by the OM receptors FepA and IroN and that the Amp warhead inhibits penicillin-binding proteins. Moreover, TC-Amp achieves targeted activity, selectively killing STm in the presence of a commensal lactobacillus. Remarkably, we uncover that TC-Amp and its Ent-based predecessor Ent-Amp achieve enhanced antibacterial activity against diverse Gram-negative ESKAPE pathogens that express Ent uptake machinery, including strains that possess intrinsic β-lactam resistance. TC-Amp and Ent-Amp exhibit potency comparable to that of the FDA-approved SAC cefiderocol against Gram-negative pathogens. These results demonstrate the effective application of native and appropriately designed nonnative siderophores as vectors for drug delivery across the OM of multiple Gram-negative bacterial pathogens.
Collapse
Affiliation(s)
- Rachel N. Motz
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Chuchu Guo
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Artur Sargun
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Gregory T. Walker
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Martina Sassone-Corsi
- Department of Microbiology & Molecular Genetics, University of California Irvine, Irvine, CA 92697, USA
| | - Manuela Raffatellu
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
- Department of Microbiology & Molecular Genetics, University of California Irvine, Irvine, CA 92697, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA 92093, USA
- Chiba University-UC San Diego Center for Mucosal Immunology, Allergy, and Vaccines, La Jolla, CA 92093, USA
| | - Elizabeth M. Nolan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
50
|
Ju HH, Park HK, Wu J, Nam YR, Kim E, Seo J, Lee H. Simultaneous coupling of metal removal and visual detection by nature-inspired polyphenol-amine surface chemistry. SOFT MATTER 2024; 20:2584-2591. [PMID: 38415992 DOI: 10.1039/d3sm01363d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
The interplay between polyphenols, amines, and metals has broad implications for surface chemistry, biomaterials, energy storage, and environmental science. Traditionally, polyphenol-amine combinations have been recognized for their ability to form adhesive, material-independent thin layers that offer a diverse range of surface functionalities. Herein, we demonstrate that a coating of tannic acid (TA) and polyethyleneimine (PEI) provides an efficient platform for capturing and monitoring metal ions in water. A unique feature of our PEI/TA-coated microbeads is the 'Detection-Capture' (Detec-Ture) mechanism. The galloyl groups in TA coordinate with Fe(III) ions (capture), initiating their oxidation to gallol-quinone. These oxidized groups subsequently react with PEI amines, leading to the formation of an Fe(II/III)-gallol-PEI network that produces a vivid purple color, thereby enabling visual detection. This mechanism couples metal capture directly with detection, distinguishing our approach from existing studies, which have either solely focused on metal removal or metal detection. The metal capturing capacity of our materials stands at 0.55 mg g-1, comparable to that of established materials like alginate and wollastonite. The detection sensitivity reaches down to 0.5 ppm. Our findings introduce a novel approach to the utility of metal-polyphenol-amine networks, presenting a new class of materials suited for simultaneous metal ion detection and capture in environmental applications.
Collapse
Affiliation(s)
- Helen H Ju
- Department of Chemistry, KAIST (Korea Advanced Institute of Science and Technology), Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Hong K Park
- Department of Chemistry, KAIST (Korea Advanced Institute of Science and Technology), Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Jingxian Wu
- Department of Chemistry, KAIST (Korea Advanced Institute of Science and Technology), Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Yu Ri Nam
- Department of Chemistry, KAIST (Korea Advanced Institute of Science and Technology), Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Eunu Kim
- Department of Chemistry, KAIST (Korea Advanced Institute of Science and Technology), Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Jeongin Seo
- Department of Chemistry, KAIST (Korea Advanced Institute of Science and Technology), Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Haeshin Lee
- Department of Chemistry, KAIST (Korea Advanced Institute of Science and Technology), Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| |
Collapse
|