1
|
Paymal S, Barale SS, Supanekar SV, Sonawane KD, Pawar KD. Overexpression, Purification, and Biochemical Characterization of the vanC2 d-Ala-d-Ser Ligase from Enterococcus casseliflavus SSK and Its Inhibition by an Oxadiazole Derivative. ACS OMEGA 2025; 10:14390-14402. [PMID: 40256530 PMCID: PMC12004154 DOI: 10.1021/acsomega.5c00599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/22/2025]
Abstract
The bacterial cell wall and enzymes involved in peptidoglycan biosynthesis are prime targets for the discovery of novel antibacterial agents. Among these enzymes, d-alanine-d-alanine ligases (Ddl) are particularly significant due to their utilization of specific substrates (d-amino acids) essential for bacterial viability. Isozymes of Ddl that utilize alternative substrates such as d-lactate or d-serine are found in vancomycin-resistant Gram-positive bacteria, initially identified in Enterococcus species, and now represent a growing concern in clinical settings. In this study, we isolated and identified vancomycin-resistant Enterococcus casseliflavus (E. casseliflavus) strain SSK and used it for amplification, cloning, and purification of the vanC2 type of d-alanine-d-serine ligase (EcfDdls). Investigations of substrate specificity and enzyme kinetics provided insights into the enzyme's mechanistic action. Evaluation of the inhibitory potential of the previously virtually screened oxadiazole derivative 1-[(5-methyl-1,2-oxazol-3-yl)methyl]-4-{[3-(propan-2-yl)-1,2,4-oxadiazol-5-yl]methyl}piperazine (CID 45805715) was carried out using an inorganic phosphate detection assay, which demonstrated complete enzymatic inhibition of purified EcfDdls. When tested, CID 45805715 significantly inhibited activity of Ddl, with an IC50 of 76.7 μM, compared to 313 μM for the reference compound DCS. Moreover, this compound also exhibited antimicrobial activity against vancomycin-resistant E. casseliflavus strain SSK. Thus, these findings provide valuable insights into the activity and inhibition of vanC2 EcfDdls, offering a promising avenue for addressing vancomycin resistance in enterococci, particularly in nosocomial infections affecting immunocompromised patients.
Collapse
Affiliation(s)
- Sneha
B. Paymal
- Department
of Microbiology, Shivaji University, Vidyanagar, Kolhapur, Maharashtra 416004, India
- Rayat
Institute
of Research and Development (RIRD), Satara, Maharashtra 415001, India
| | - Sagar S. Barale
- Department
of Microbiology, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan 305817, India
| | | | - Kailas D. Sonawane
- Department
of Biochemistry, Shivaji University, Vidyanagar, Kolhapur, Maharashtra 416004, India
| | - Kiran D. Pawar
- School
of
Nanoscience and Biotechnology, Shivaji University, Vidyanagar, Kolhapur, Maharashtra 416004, India
| |
Collapse
|
2
|
Kogay R, Wolf YI, Koonin EV. Horizontal Transfer of Bacterial Operons into Eukaryote Genomes. Genome Biol Evol 2025; 17:evaf055. [PMID: 40111106 PMCID: PMC11965790 DOI: 10.1093/gbe/evaf055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025] Open
Abstract
In prokaryotes, functionally linked genes are typically clustered into operons, which are transcribed into a single mRNA, providing for the coregulation of the production of the respective proteins, whereas eukaryotes generally lack operons. We explored the possibility that some prokaryotic operons persist in eukaryotic genomes after horizontal gene transfer (HGT) from bacteria. Extensive comparative analysis of prokaryote and eukaryote genomes revealed 33 gene pairs originating from bacterial operons, mostly encoding enzymes of the same metabolic pathways, and represented in distinct clades of fungi or amoebozoa. This amount of HGT is about an order of magnitude less than that observed for the respective individual genes. These operon fragments appear to be relatively recent acquisitions as indicated by their narrow phylogenetic spread and low intron density. In 20 of the 33 horizontally acquired operonic gene pairs, the genes are fused in the respective group of eukaryotes so that the encoded proteins become domains of a multifunctional protein ensuring coregulation and correct stoichiometry. We hypothesize that bacterial operons acquired via HGT initially persist in eukaryotic genomes under a neutral evolution regime and subsequently are either disrupted by genome rearrangement or undergo gene fusion which is then maintained by selection.
Collapse
Affiliation(s)
- Roman Kogay
- Computational Biology Branch, Division of Intramural Research, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | - Yuri I Wolf
- Computational Biology Branch, Division of Intramural Research, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | - Eugene V Koonin
- Computational Biology Branch, Division of Intramural Research, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| |
Collapse
|
3
|
Pederick JL, Kumar A, Pukala TL, Bruning JB. Functional and structural characterization of Staphylococcus aureus N-acetylglucosamine 1-phosphate uridyltransferase (GlmU) reveals a redox-sensitive acetyltransferase activity. Protein Sci 2025; 34:e70111. [PMID: 40143772 PMCID: PMC11947611 DOI: 10.1002/pro.70111] [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: 12/02/2024] [Revised: 03/12/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025]
Abstract
The bifunctional enzyme N-acetylglucosamine 1-phosphate uridyltransferase (GlmU) is a promising antibiotic drug target, as it facilitates the biosynthesis of uridine 5'-diphospho-N-acetylglucosamine, an essential precursor of cell wall constituents. We identified that Staphylococcus aureus GlmU (SaGlmU), which was previously targeted for inhibitor development, possesses a dual-cysteine variation (C379/C404) within the acetyltransferase active site. Enzyme assays performed under reducing and non-reducing conditions revealed that the acetyltransferase activity of SaGlmU is redox-sensitive, displaying ~15-fold lower turnover and ~3-fold higher KM value for the acetyl CoA substrate under non-reducing conditions. This sensitivity was absent in a C379A SaGlmU mutant. Analysis of SaGlmU by mass spectrometry, x-ray crystallography, and in silico modeling support that C379 and C404 act as a reversible, redox-sensitive switch by forming a disulfide under non-reducing conditions that impedes acetyl CoA recognition and turnover. Therefore, we recommend that future in vitro screening and characterization of SaGlmU inhibitors consider both reducing and non-reducing conditions.
Collapse
Affiliation(s)
- Jordan L. Pederick
- Institute for Photonics and Advanced Sensing (IPAS), School of Biological SciencesThe University of AdelaideAdelaideSouth AustraliaAustralia
| | - Akhil Kumar
- Institute for Photonics and Advanced Sensing (IPAS), School of Biological SciencesThe University of AdelaideAdelaideSouth AustraliaAustralia
| | - Tara L. Pukala
- Department of ChemistrySchool of Physical Sciences, North Terrace Campus, The University of AdelaideAdelaideSouth AustraliaAustralia
| | - John B. Bruning
- Institute for Photonics and Advanced Sensing (IPAS), School of Biological SciencesThe University of AdelaideAdelaideSouth AustraliaAustralia
| |
Collapse
|
4
|
Kosho MXF, Ciurli A, Giera M, Neefjes J, Loos BG. Metabolomic Profiles of Oral Rinse Samples to Distinguish Severe Periodontitis Patients From Non-Periodontitis Controls. J Periodontal Res 2025. [PMID: 40083241 DOI: 10.1111/jre.13379] [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: 09/14/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 03/16/2025]
Abstract
AIMS To explore the potential of metabolomic profiles of oral rinse samples to distinguish between patients with severe periodontitis (stage III/IV) and non-periodontitis controls. This is coupled to an analysis of differences in metabolomic profiles between individuals without periodontitis, patients with localized periodontitis, and patients with generalized periodontitis. METHODS Periodontitis patients and controls were recruited, all aged ≥ 40 years. Study participants were asked to rinse vigorously for 30 s with 10 mL phosphate buffered saline. Metabolites were identified using a semi-targeted liquid chromatography tandem mass spectrometry (LC-MS/MS) platform. RESULTS In total, 38 periodontitis patients (18 localized, 20 generalized stage III/IV periodontitis patients) and 16 controls were included. Metabolomic profiles of oral rinse samples were able to distinguish patients with severe periodontitis (stage III/IV) from non-periodontitis controls. Among various variables for the severity of periodontitis, we found that the number of sites with deep pockets (PPD) ≥ 6 mm explained best the differences in metabolomic profiles between controls and patients with severe periodontitis. Subjects with a high number of sites with PPD ≥ 6 mm were characterized by a higher level of phosphorylated nucleotides, amino acids, peptides, and dicarboxylic acids. Metabolomic profiles were also significantly different between controls vs. generalized periodontitis and between localized periodontitis vs. generalized periodontitis (p < 0.05). CONCLUSION Our study demonstrates that simply collected oral rinse samples are suitable for LC-MS/MS based metabolomic analysis. We show that a metabolomic profile with a substantial number of metabolites can distinguish severe periodontitis patients from non-periodontitis controls. These observations can be a basis for further studies into screening to identify subjects with the risk of having severe periodontitis.
Collapse
Affiliation(s)
- Madeline X F Kosho
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Alessio Ciurli
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, the Netherlands
| | - Martin Giera
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, the Netherlands
| | - Jacques Neefjes
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Bruno G Loos
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
5
|
Süssmuth RD, Kulike-Koczula M, Gao P, Kosol S. Fighting Antimicrobial Resistance: Innovative Drugs in Antibacterial Research. Angew Chem Int Ed Engl 2025; 64:e202414325. [PMID: 39611429 DOI: 10.1002/anie.202414325] [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: 07/29/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 11/30/2024]
Abstract
In the fight against bacterial infections, particularly those caused by multi-resistant pathogens known as "superbugs", the need for new antibacterials is undoubted in scientific communities and is by now also widely perceived by the general population. However, the antibacterial research landscape has changed considerably over the past years. With few exceptions, the majority of big pharma companies has left the field and thus, the decline in R&D on antibacterials severely impacts the drug pipeline. In recent years, antibacterial research has increasingly relied on smaller companies or academic research institutions, which mostly have only limited financial resources, to carry a drug discovery and development process from the beginning and through to the beginning of clinical phases. This review formulates the requirements for an antibacterial in regard of targeted pathogens, resistance mechanisms and drug discovery. Strategies are shown for the discovery of new antibacterial structures originating from natural sources, by chemical synthesis and more recently from artificial intelligence approaches. This is complemented by principles for the computer-aided design of antibacterials and the refinement of a lead structure. The second part of the article comprises a compilation of antibacterial molecules classified according to bacterial target structures, e.g. cell wall synthesis, protein synthesis, as well as more recently emerging target classes, e.g. fatty acid synthesis, proteases and membrane proteins. Aspects of the origin, the antibacterial spectrum, resistance and the current development status of the presented drug molecules are highlighted.
Collapse
Affiliation(s)
- Roderich D Süssmuth
- Institut für Chemie, Technische Universität Berlin, Strasse des 17. Juni 124, TC2, 10629, Berlin, Germany
| | - Marcel Kulike-Koczula
- Institut für Chemie, Technische Universität Berlin, Strasse des 17. Juni 124, TC2, 10629, Berlin, Germany
| | - Peng Gao
- Institut für Chemie, Technische Universität Berlin, Strasse des 17. Juni 124, TC2, 10629, Berlin, Germany
| | - Simone Kosol
- Medical School Berlin, Department Human Medicine, Rüdesheimer Strasse 50, 14195, Berlin, Germany
| |
Collapse
|
6
|
Minjárez-Sáenz M, Rivero M, Correa-Pérez V, Boneta S, Suárez P, Polo V, Sadeghi SJ, Yruela I, Martínez-Júlvez M, Medina M. Structural and functional insights into UDP-N-acetylglucosamine-enolpyruvate reductase (MurB) from Brucella ovis. Arch Biochem Biophys 2025; 765:110288. [PMID: 39761724 DOI: 10.1016/j.abb.2025.110288] [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: 10/06/2024] [Revised: 12/27/2024] [Accepted: 01/03/2025] [Indexed: 01/14/2025]
Abstract
The peptidoglycan biosynthetic pathway involves a series of enzymatic reactions in which UDP-N-acetylglucosamine-enolpyruvate reductase (MurB) plays a crucial role in catalyzing the conversion of UDP-N-acetylglucosamine-enolpyruvate (UNAGEP) to UDP-N-acetylmuramic acid. This reaction relies on NADPH and FAD and, since MurB is not found in eukaryotes, it is an attractive target for the development of antimicrobials. MurB from Brucella ovis, the causative agent of brucellosis in sheep, is characterized here. The FAD cofactor in MurB of B. ovis is reduced to the hydroquinone state without semiquinone stabilization with an estimated Eox/hq of -260 mV. MurB from B. ovis catalyzes the oxidation of NADPH in a slow process that is positively influenced by the presence of the second product, UNAGEP. The crystallographic structure of the MurBox:UNAGEP complex confirms its folding into three domains and the binding of UNAGEP, positioning its enolpyruvyl group for hydride transfer from FAD. MurB shows a complex thermal unfolding pathway that is influenced by UNAGEP and NADP+, confirming its ability to bind both molecules. Molecular dynamics (MD) simulations predict that the nicotinamide of NADP+ is more stable at the active site than the enolpyruvyl of UNAGEP, and suggests that MurB can simultaneously accommodate NADPH and UNAGEP in the substrate channel, increasing overall protein-ligand flexibility. Sequence and evolutionary analyses show that MurB from B. ovis conserves all motifs predicted to be involved in catalysis within the Type IIa family.
Collapse
Affiliation(s)
- Martha Minjárez-Sáenz
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BIFI) Universidad de Zaragoza, GBsC (Unizar) Join Unit to CSIC, Zaragoza, Spain
| | - Maribel Rivero
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BIFI) Universidad de Zaragoza, GBsC (Unizar) Join Unit to CSIC, Zaragoza, Spain
| | - Víctor Correa-Pérez
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BIFI) Universidad de Zaragoza, GBsC (Unizar) Join Unit to CSIC, Zaragoza, Spain
| | - Sergio Boneta
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BIFI) Universidad de Zaragoza, GBsC (Unizar) Join Unit to CSIC, Zaragoza, Spain
| | - Paula Suárez
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BIFI) Universidad de Zaragoza, GBsC (Unizar) Join Unit to CSIC, Zaragoza, Spain
| | - Víctor Polo
- Instituto de Biocomputación y Física de Sistemas Complejos (BIFI) Universidad de Zaragoza, GBsC (Unizar) Join Unit to CSIC, Zaragoza, Spain; Departamento de Química Física, Universidad de Zaragoza, Zaragoza, Spain
| | - Sheila J Sadeghi
- Department of Life Sciences and Systems Biology, University of Torino, Italy
| | - Inmaculada Yruela
- Estación Experimental de Aula Dei (EEAD), CSIC and GBsC (Unizar) Join Unit to CSIC, Zaragoza, Spain
| | - Marta Martínez-Júlvez
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BIFI) Universidad de Zaragoza, GBsC (Unizar) Join Unit to CSIC, Zaragoza, Spain.
| | - Milagros Medina
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BIFI) Universidad de Zaragoza, GBsC (Unizar) Join Unit to CSIC, Zaragoza, Spain.
| |
Collapse
|
7
|
Gilmore MC, Cava F. Bacterial peptidoglycan recycling. Trends Microbiol 2025; 33:340-353. [PMID: 39613687 DOI: 10.1016/j.tim.2024.11.004] [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: 07/10/2024] [Revised: 10/31/2024] [Accepted: 11/06/2024] [Indexed: 12/01/2024]
Abstract
During growth and division, the bacterial cell wall is remodeled, resulting in the liberation of peptidoglycan (PG) fragments which are typically reinternalized and recycled. Recycling of PG has been studied in a few model species, but its importance and diversity are not yet well understood. Here, we review how bacteria transport and recycle the components of their PG, highlighting updates and new findings.
Collapse
Affiliation(s)
- Michael C Gilmore
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Department of Molecular Biology, SciLifeLab, Umeå University, 90187 Umeå, Sweden
| | - Felipe Cava
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Department of Molecular Biology, SciLifeLab, Umeå University, 90187 Umeå, Sweden.
| |
Collapse
|
8
|
Hu M, Xu Y, Wang Y, Huang Z, Wang L, Zeng F, Qiu B, Liu Z, Yuan P, Wan Y, Ge S, Zhong D, Xiao S, Luo R, He J, Sun M, Zhuang X, Guo N, Cui C, Gao J, Zhou H, He X. Gut microbial-derived N-acetylmuramic acid alleviates colorectal cancer via the AKT1 pathway. Gut 2025:gutjnl-2024-332891. [PMID: 40015949 DOI: 10.1136/gutjnl-2024-332891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 02/15/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Gut microbial metabolites are recognised as critical effector molecules that influence the development of colorectal cancer (CRC). Peptidoglycan fragments (PGFs) produced by microbiota play a crucial role in maintaining intestinal homeostasis, but their role in CRC remains unclear. OBJECTIVE Here, we aimed to explore the potential contribution of PGFs in intestinal tumourigenesis. DESIGN The relative abundance of peptidoglycan synthase and hydrolase genes was assessed by metagenomic analysis. Specific PGFs in the faeces and serum of CRC patients were quantified using targeted mass spectrometry. The effects of PGF on intestinal tumourigenesis were systematically evaluated using various murine models of CRC and organoids derived from CRC patients. Downstream molecular targets were screened and evaluated using proteome microarray, transcriptome sequencing and rescue assays. RESULTS Metagenomic analysis across seven independent cohorts (n=1121) revealed a comprehensive reduction in peptidoglycan synthase gene relative abundance in CRC patients. Targeted mass spectrometry identified significant depletion of a specific PGF, N-acetylmuramic acid (NAM) in CRC patients, which decreased as tumours progressed (p<0.001). NAM significantly inhibits intestinal tumourigenesis in various models, including Apc Min/+, AOM/DSS-treated and MC38 tumour-bearing mice. Additionally, NAM inhibits the growth of patient-derived CRC organoids in a concentration-dependent manner. Mechanistically, NAM inhibits the activation of AKT1 by directly binding to it and blocking its phosphorylation, which is a partial mediator of NAM's anticancer effects. CONCLUSION The PGF NAM protects against intestinal tumourigenesis by targeting the AKT1 signalling pathway. NAM may serve as a novel potential preventive and therapeutic biomarker against CRC.
Collapse
Affiliation(s)
- Mengyao Hu
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yi Xu
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuqing Wang
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhenhe Huang
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lei Wang
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Fanan Zeng
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Bowen Qiu
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zefeng Liu
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Peibo Yuan
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu Wan
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shuang Ge
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Dian Zhong
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Siyu Xiao
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Rongrong Luo
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiaqi He
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Meiling Sun
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoduan Zhuang
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Nannan Guo
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chunhui Cui
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jie Gao
- Department of Gastroenterology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
- State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaolong He
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Southern Medical University, Guangzhou, Guangdong, China
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
Critchlow JM, Barraza JP, Munneke MJ, Krystofiak E, Green ER, Skaar EP. The interplay between Acinetobacter baumannii ZigA and SltB promotes zinc homeostasis and cell envelope integrity. Infect Immun 2025; 93:e0042224. [PMID: 39846731 PMCID: PMC11834433 DOI: 10.1128/iai.00422-24] [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: 09/23/2024] [Accepted: 12/18/2024] [Indexed: 01/24/2025] Open
Abstract
Acinetobacter baumannii is an opportunistic human pathogen that acquires nutrient metals from the vertebrate host amid infection. During zinc (Zn) scarcity, A. baumannii upregulates the expression of the predicted Zn metallochaperone, zigA. Loss of zigA compromises fitness during Zn deficiency, highlighting its role in this condition. To assess the contribution of ZigA to Zn-deficient A. baumannii, a multiparallel transposon sequencing and genetic interaction mapping approach was used. Transposon insertions in A1S_3027, encoding a predicted soluble lytic transglycosylase that tailors the bacterial cell wall, were enriched in the Zn-starved ΔzigA transposon library. Based on previous studies as well as structural and sequence homology, we designated A1S_3027 as soluble lytic transglycosylase B (SltB). Further analyses revealed that inactivating sltB rescued ΔzigA fitness defects during Zn starvation. An A. baumannii ΔzigAΔsltB mutant demonstrated altered cell envelope structures and increased cellular permeability, highlighting the roles of ZigA and SltB in maintaining cell envelope integrity. Furthermore, these mutants exhibited heightened resistance to β-lactam antibiotics and other cell wall-targeting agents. Alterations in cell envelope integrity in the ΔzigAΔsltB mutant improved fitness in a murine pneumonia infection model, emphasizing the contribution of ZigA and SltB to A. baumannii pathogenesis. This study elucidates how functional interactions between ZigA and SltB modulate cell envelope integrity and pathogenesis of A. baumannii during Zn depletion. These findings reveal an interplay between metal homeostasis and cell envelope integrity, offering insights into how A. baumannii ZigA contributes to these critical cellular processes.
Collapse
Affiliation(s)
- Jeanette M. Critchlow
- Microbe-Host Interactions Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Juan P. Barraza
- Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Matthew J. Munneke
- Microbe-Host Interactions Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Evan Krystofiak
- Cell Imaging Shared Resource, Vanderbilt University, Nashville, Tennessee, USA
| | - Erin R. Green
- Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| | - Eric P. Skaar
- Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
10
|
Sarkar S, Kulshrestha M, Chaudhary A, Tiwari V. In-silico identification and experimental validation of possible inhibitor for the bifunctional protein GlmU of Acinetobacter baumannii. Int J Biol Macromol 2025; 305:141239. [PMID: 39971047 DOI: 10.1016/j.ijbiomac.2025.141239] [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: 12/06/2024] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 02/21/2025]
Abstract
Antimicrobial resistance renders numerous antibiotics ineffective, resulting in persistent infections and increased mortality rates. This makes identifying novel therapeutic targets imperative, necessitating the investigation of vital bacterial mechanisms. The bifunctional protein N-Acetyl-glucosamine-1-phosphate uridyltransferase (GlmU) is an essential enzyme in Acinetobacter baumannii. GlmU catalyzes the synthesis of an intermediate that enters the peptidoglycan and lipopolysaccharide synthesis, which is essential for bacterial survival. In this study, the GlmU monomer was modeled using Phyre2, refined with GalaxyWeb, and confirmed through PSVS, ensuring a reliable 3D structure. Binding sites on GlmU were identified using PUResNetV2.0, revealing two key sites corresponding to uridyltransferase and acetyltransferase activities. A GlmU trimer was constructed, and molecular docking of 55 potential inhibitors was performed against both the monomer and trimer. 3,3'-methylenebis-(4-hydroxy-coumarin) emerged as the most promising inhibitor, with strong interactions at both binding sites and the trimer. Molecular dynamics simulations confirmed the stability of the GlmU-3,3'-methylenebis-(4-hydroxy-coumarin) complex. Mutation analysis of key interacting residues further highlighted their importance in maintaining the protein's stability. Experimental validation of lead confirmed its effect on bacterial growth, peptidoglycan content, lipid accumulation, carbohydrate concentration, reactive oxygen species (ROS) production, protein carbonylation, and biofilm formation in A. baumannii. All these results suggest the therapeutic potential of this molecule against A. baumannii via targeting GlmU protein.
Collapse
Affiliation(s)
- Sayani Sarkar
- Department of Biochemistry, Central University of Rajasthan, Ajmer 305817, India
| | - Mukta Kulshrestha
- Department of Biochemistry, Central University of Rajasthan, Ajmer 305817, India
| | - Aryan Chaudhary
- Department of Biochemistry, Central University of Rajasthan, Ajmer 305817, India
| | - Vishvanath Tiwari
- Department of Biochemistry, Central University of Rajasthan, Ajmer 305817, India.
| |
Collapse
|
11
|
Shook JC, Genito CJ, Darwitz BP, Tyson KJ, Velez AZ, Bridwell SK, Parsons JB, Rowe SE, Marshall CW, Conlon BP, Thurlow LR. Diabetes potentiates the emergence and expansion of antibiotic resistance. SCIENCE ADVANCES 2025; 11:eads1591. [PMID: 39937900 PMCID: PMC11817934 DOI: 10.1126/sciadv.ads1591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/13/2025] [Indexed: 02/14/2025]
Abstract
Individuals with diabetes mellitus frequently develop severe skin and soft tissue infections (SSTIs) that are recalcitrant to antibiotic treatment. We examined how diabetes affects the emergence of antibiotic resistance in a Staphylococcus aureus SSTI. We determined that S. aureus evolves antibiotic resistance rapidly in diabetic mice, while resistance did not occur in nondiabetic mice over the course of infection. Diabetes-associated immune cell dysfunction plays a minor role in the emergence of resistance, while hyperglycemia plays a dominant role facilitating the expansion and takeover of resistant mutants in diabetic infections. Furthermore, vancomycin intermediate resistant isolates display a pronounced fitness defect in nondiabetic mice but not in diabetic mice. Together, these data suggest that the diabetic infection environment represents an ideal reservoir for the emergence and proliferation of antibiotic resistance. Controlling the blood sugar of diabetic mice with insulin resulted in significantly decreased incidence of antibiotic-resistant S. aureus.
Collapse
Affiliation(s)
- John C. Shook
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Christopher J. Genito
- Department of Biomedical Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Benjamin P. Darwitz
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kaleb J. Tyson
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Amanda Z. Velez
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sophia K. Bridwell
- Department of Biological Sciences, Marquette University, Milwaukee, WI 53233, USA
| | - Joshua B. Parsons
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Division of Infectious Diseases, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sarah E. Rowe
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Brian P. Conlon
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lance R. Thurlow
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biomedical Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
12
|
Lee KM, Kim KH. Improved Protective Efficacy in Rainbow Trout (Oncorhynchus mykiss) Against Vibrio anguillarum Through Immunization with a Combination of Formalin-Killed and Auxotrophic-Live V. anguillarum. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2025; 27:35. [PMID: 39869218 DOI: 10.1007/s10126-025-10414-3] [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: 12/10/2024] [Accepted: 01/15/2025] [Indexed: 01/28/2025]
Abstract
Vibriosis caused by Vibrio anguillarum has been an important bacterial disease in cultured rainbow trout (Oncorhynchus mykiss). In the present study, we evaluated the protective efficacy of a vaccine that consists of formalin-killed (FK) V. anguillarum and the alr genes knockout auxotrophic-live (AL) V. anguillarum (Δalr1Δalr2 V. anguillarum). Fish were immunized with a high dose of the FK V. anguillarum vaccine or four different combinations of FK and AL V. anguillarum. In the challenge test, fish immunized with 1 × 106 CFU of FK V. anguillarum plus 1 × 104 CFU of AL V. anguillarum (FK-106 + AL-104) showed complete protection (100% RPS) against V. anguillarum. In comparison, fish immunized with 1 × 107 CFU of FK V. anguillarum (FK-107) showed much lower survival rates. In the result of ELISA, the antibody titer of fish immunized with FK-106 + AL-104 was significantly higher than that of the PBS group, but the titer was not higher than FK-107, suggesting that the higher protection by the FK + AL combination vaccine might be mediated by not only humoral immunity but also other protective factors conferred by live bacteria in the combination vaccine. In conclusion, the present FK + AL combination vaccine efficiently protected rainbow trout with approximately 10 times lower doses of bacteria than FK-107, which could lessen the safety problem caused by a high-tittered live bacteria vaccine and the lower immunogenicity problem of killed bacteria vaccine.
Collapse
Affiliation(s)
- Kyung Min Lee
- Department of Aquatic Life Medicine, Pukyong National University, Busan, 48513, South Korea
| | - Ki Hong Kim
- Department of Aquatic Life Medicine, Pukyong National University, Busan, 48513, South Korea.
| |
Collapse
|
13
|
Diab A, Dickerson H, Al Musaimi O. Targeting the Heart of Mycobacterium: Advances in Anti-Tubercular Agents Disrupting Cell Wall Biosynthesis. Pharmaceuticals (Basel) 2025; 18:70. [PMID: 39861133 PMCID: PMC11768153 DOI: 10.3390/ph18010070] [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: 11/23/2024] [Revised: 12/12/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Mycobacterium tuberculosis infections continue to pose a significant global health challenge, particularly due to the rise of multidrug-resistant strains, random mycobacterial mutations, and the complications associated with short-term antibiotic regimens. Currently, five approved drugs target cell wall biosynthesis in Mycobacterium tuberculosis. This review provides a comprehensive analysis of these drugs and their molecular mechanisms. Isoniazid, thioamides, and delamanid primarily disrupt mycolic acid synthesis, with recent evidence indicating that delamanid also inhibits decaprenylphosphoryl-β-D-ribose-2-epimerase, thereby impairing arabinogalactan biosynthesis. Cycloserine remains the sole approved drug that inhibits peptidoglycan synthesis, the foundational layer of the mycobacterial cell wall. Furthermore, ethambutol interferes with arabinogalactan synthesis by targeting arabinosyl transferase enzymes, particularly embB- and embC-encoded variants. Beyond these, six promising molecules currently in Phase II clinical trials are designed to target arabinan synthesis pathways, sutezolid, TBA 7371, OPC-167832, SQ109, and both benzothiazinone derivatives BTZ043 and PBTZ169, highlighting advancements in the development of cell wall-targeting therapies.
Collapse
Affiliation(s)
- Ahmad Diab
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, UK;
| | - Henry Dickerson
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, UK;
| | - Othman Al Musaimi
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, UK;
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
14
|
Casimo G, Micca Longo G, Longo S. Beyond Homochirality: Computer Modeling Hints of Heterochiral Proteins in Early and Extraterrestrial Life. ASTROBIOLOGY 2025; 25:22-31. [PMID: 39786977 DOI: 10.1089/ast.2024.0072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Agent-based simulations are set to describe the early biotic selection of oligomers made of monomers of different chirality. The simulations consider the spatial distribution of agents and resources, the balance of biomass of different chirality, and the balance of chemical energy. Following the well-known Wald's hypothesis, a disadvantage is attributed to the change in chirality along the biochemical sequence. A racemic amino acid budget is considered, based on findings in meteorites and the results of Miller's experiments. It is also hypothesized that the very first life forms were heterotrophic. Given these assumptions, our simulations showed that biological sequences were not strictly homochiral and had few chirality changes. These results suggest that the current dominance of homochiral species may have been preceded by a more structurally varied biochemistry. This might be reflected in the few known heterochiral proteins, whose structures are based neither on alpha-helices nor on beta-sheets. Extraterrestrial life forms might be based on such heterochiral proteins.
Collapse
Affiliation(s)
- Gianluigi Casimo
- Dipartimento di Bioscienze, Biotecnologie e Ambientale, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Gaia Micca Longo
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Savino Longo
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Bari, Italy
- Istituto per la Scienza e Tecnologia dei Plasmi - Consiglio Nazionale delle Ricerche, Bari, Italy
| |
Collapse
|
15
|
Han L, Lin C, Lan Y, Hua Y, Wu J, Fan Z, Li Y. Metagenomic Analysis of Gut Microbiome of Persistent Pulmonary Hypertension of the Newborn. Cardiovasc Toxicol 2025; 25:135-147. [PMID: 39680298 DOI: 10.1007/s12012-024-09949-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 12/05/2024] [Indexed: 12/17/2024]
Abstract
Persistent pulmonary hypertension of the newborn (PPHN) is one of the most common diseases in the neonatal intensive care unit which severely affects neonatal survival. Gut microbes play an increasingly important role in human health, but there are rarely reported how gut microbiota contribute to PPHN. In our study, the metagenomic sequencing of feces from 12 PPHN's neonates and 8 controls were performed to expose the relation between neonatal gut microbes and PPHN disease. Firstly, we found that the abundance of Actinobacteria, Proteobacteria, Bacteroidetes were significantly increased in PPHN compared with controls, but the Firmicutes components was reduced. And some pathogenic strains (like Vibrio metschnikovii) were significantly enriched in the PPHN compared with controls. Secondly, functional annotation of genes found that PPHN up-regulated transmembrane transport, but down-regulated ribosome and ATP binding. Lastly, microbial metabolic pathway enrichment analysis indicated that some metabolic pathway in PPHN were conflicting and contradictory, showed that an abnormally increased metabolism, disturbed protein synthesis and genomic instability in the PPHN neonate. Our results contribute to understanding the changes in the species and function of gut microbiota in PPHN, thus providing a theoretical basis for the explanation and treatment of PPHN.
Collapse
Affiliation(s)
- Linli Han
- Department of Pediatrics, Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chuyang Lin
- Department of Pediatrics, Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yue Lan
- Key Laboratory of Bioresources and Ecoenvironment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Yimin Hua
- Department of Pediatrics, Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinlin Wu
- Department of Pediatrics, Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, 20 3rd Section, Renmin S. Rd., Chengdu, 610041, Sichuan, China.
| | - Zhenxin Fan
- Key Laboratory of Bioresources and Ecoenvironment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, Sichuan, China.
- Department of Pediatrics, Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, 24 1rd Section, Renmin S. Rd., Chengdu, 610065, Sichuan, China.
| | - Yifei Li
- Department of Pediatrics, Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, 20 3rd Section, Renmin S. Rd., Chengdu, 610041, Sichuan, China.
| |
Collapse
|
16
|
Chiang CY, West NP. The fall of the mycobacterial cell wall: interrogating peptidoglycan synthesis for novel anti-TB agents. PeerJ 2024; 12:e18404. [PMID: 39553715 PMCID: PMC11569785 DOI: 10.7717/peerj.18404] [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: 06/10/2024] [Accepted: 10/04/2024] [Indexed: 11/19/2024] Open
Abstract
Tuberculosis (TB) caused by Mycobacterium tuberculosis has been a threat to human health for thousands of years and still leads to millions of deaths each year. TB is a disease that is refractory to treatment, partially due to its capacity for in-host persistence. The cell wall of mycobacteria, rich in mycolic acid, is broadly associated with bacterial persistence together with antimicrobial and immunological resistance. Enzymes for the biosynthesis of bacterial peptidoglycan, an essential component of the cell wall, have been addressed and considered as appealing drug targets in pathogens. Significant effort has been dedicated to finding inhibitors that hinder peptidoglycan biosynthesis, many with demonstrated enzymatic inhibition in vitro being published. One family of critical biosynthetic enzymes are the Mur enzymes, with many enzyme specific inhibitors having been reported. However, a lesser developed strategy which may have positive clinical implications is to take advantage of the common structural and catalytic characteristics among Mur enzymes and to allow simultaneous, multiple Mur inhibition, and avert the development of drug resistance. M. tuberculosis relies on these essential Mur enzymes, with the best-known subset being Mur ligases, but also utilizes unique functions of atypical transpeptidases resulting in peptidoglycan peptide cross-linking beneficial to the bacteria's capacity for chronic persistence in humans. A systematic review is now needed, with an emphasis on M. tuberculosis. The urgent development of novel anti-TB agents to counter rapidly developing drug resistance requires a revisit of the literature, past successes and failures, in an attempt to reveal liabilities in critical cellular functions and drive innovation.
Collapse
Affiliation(s)
- Cheng-Yu Chiang
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Queensland, Australia
| | - Nicholas P. West
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
17
|
Pinho MG, Foster SJ. Cell Growth and Division of Staphylococcus aureus. Annu Rev Microbiol 2024; 78:293-310. [PMID: 39565951 DOI: 10.1146/annurev-micro-041222-125931] [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: 11/22/2024]
Abstract
Bacterial cell growth and division require temporal and spatial coordination of multiple processes to ensure viability and morphogenesis. These mechanisms both determine and are determined by dynamic cellular structures and components, from within the cytoplasm to the cell envelope. The characteristic morphological changes during the cell cycle are largely driven by the architecture and mechanics of the cell wall. A constellation of proteins governs growth and division in Staphylococcus aureus, with counterparts also found in other organisms, alluding to underlying conserved mechanisms. Here, we review the status of knowledge regarding the cell cycle of this important pathogen and describe how this informs our understanding of the action of antibiotics and the specter of antimicrobial resistance.
Collapse
Affiliation(s)
- Mariana G Pinho
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade NOVA de Lisboa, Oeiras, Portugal;
| | - Simon J Foster
- The Florey Institute, School of Biosciences, University of Sheffield, Sheffield, United Kingdom;
| |
Collapse
|
18
|
Oluwole AO, Kalmankar NV, Guida M, Bennett JL, Poce G, Bolla JR, Robinson CV. Lipopeptide antibiotics disrupt interactions of undecaprenyl phosphate with UptA. Proc Natl Acad Sci U S A 2024; 121:e2408315121. [PMID: 39361645 PMCID: PMC11474028 DOI: 10.1073/pnas.2408315121] [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: 04/26/2024] [Accepted: 08/19/2024] [Indexed: 10/05/2024] Open
Abstract
The peptidoglycan pathway represents one of the most successful antibacterial targets with the last critical step being the flipping of carrier lipid, undecaprenyl phosphate (C55-P), across the membrane to reenter the pathway. This translocation of C55-P is facilitated by DedA and DUF368 domain-containing family membrane proteins via unknown mechanisms. Here, we employ native mass spectrometry to investigate the interactions of UptA, a member of the DedA family of membrane protein from Bacillus subtilis, with C55-P, membrane phospholipids, and cell wall-targeting antibiotics. Our results show that UptA, expressed and purified in Escherichia coli, forms monomer-dimer equilibria, and binds to C55-P in a pH-dependent fashion. Specifically, we show that UptA interacts more favorably with C55-P over shorter-chain analogs and membrane phospholipids. Moreover, we demonstrate that lipopeptide antibiotics, amphomycin and aspartocin D, can directly inhibit UptA function by out-competing the substrate for the protein binding, in addition to their propensity to form complex with free C55-P. Overall, this study shows that UptA-mediated translocation of C55-P is potentially mediated by pH and anionic phospholipids and provides insights for future development of antibiotics targeting carrier lipid recycling.
Collapse
Affiliation(s)
- Abraham O. Oluwole
- Department of Chemistry, University of Oxford, OxfordOX1 3QZ, United Kingdom
- The Kavli Institute for Nanoscience Discovery, University of Oxford, OxfordOX1 3QU, United Kingdom
| | - Neha V. Kalmankar
- Department of Chemistry, University of Oxford, OxfordOX1 3QZ, United Kingdom
- The Kavli Institute for Nanoscience Discovery, University of Oxford, OxfordOX1 3QU, United Kingdom
| | - Michela Guida
- The Kavli Institute for Nanoscience Discovery, University of Oxford, OxfordOX1 3QU, United Kingdom
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, Rome00185, Italy
| | - Jack L. Bennett
- Department of Chemistry, University of Oxford, OxfordOX1 3QZ, United Kingdom
- The Kavli Institute for Nanoscience Discovery, University of Oxford, OxfordOX1 3QU, United Kingdom
| | - Giovanna Poce
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, Rome00185, Italy
| | - Jani R. Bolla
- The Kavli Institute for Nanoscience Discovery, University of Oxford, OxfordOX1 3QU, United Kingdom
- Department of Biology, University of Oxford, OxfordOX1 3RB, United Kingdom
| | - Carol V. Robinson
- Department of Chemistry, University of Oxford, OxfordOX1 3QZ, United Kingdom
- The Kavli Institute for Nanoscience Discovery, University of Oxford, OxfordOX1 3QU, United Kingdom
| |
Collapse
|
19
|
Zhuang H, Wang R, Qi Y, Liu Y, Xiong H, Yao J. Nanocoated bacteria with H 2S generation-triggered self-amplified photothermal and photodynamic effect for breast cancer therapy. J Control Release 2024; 373:507-519. [PMID: 39025267 DOI: 10.1016/j.jconrel.2024.07.036] [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: 02/08/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Phototherapy utilizing bacterial carriers has demonstrated efficacy in anti-tumor therapy, while the poor delivery of phototherapeutic agents and immunogenicity of microbial substances remain problematic. Herein, we develop a nanocoated bacterial delivery system (IF-S.T) that in situ forms the efficient photothermal agents via biomineralization and improves the intracellular oxygenation, thus triggering the self-enhanced photothermal therapy (PTT) and photodynamic therapy (PDT) on tumor. We densely coat self-assembled IF (ICG-Fe2+) nanocomplex onto the surface of LT2, weakly virulent strain of Salmonella typhimurium (S.T), by bioadaptive nanocoating techniques, masking bacterial virulence factors and reducing the potential immune adverse effects. Upon penetrating into the tumor environment, IF-S.T responds to H2O2 to trigger the removal of the IF coating, where S.T produces excess hydrogen sulfide (H2S). H2S reacts with Fe2+, yielding ferrous sulfide (FeS) for PTT, and inhibits mitochondrial respiration to enhance tumor cell oxygenation for PDT. Consequently, IF-S.T plus laser irradiation exhibits direct tumor cells killing and elicits robust antitumor immune responses, leading to the complete tumor elimination. Thus, IF-S.T represents a promising platform for effective tumor delivery of photoactive agents for improved PTT/PDT efficacy.
Collapse
Affiliation(s)
- He Zhuang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Rui Wang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Yao Qi
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Yufei Liu
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Hui Xiong
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China.
| | - Jing Yao
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China.
| |
Collapse
|
20
|
Chen J, Wang W, Hu X, Yue Y, Lu X, Wang C, Wei B, Zhang H, Wang H. Medium-sized peptides from microbial sources with potential for antibacterial drug development. Nat Prod Rep 2024; 41:1235-1263. [PMID: 38651516 DOI: 10.1039/d4np00002a] [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: 04/25/2024]
Abstract
Covering: 1993 to the end of 2022As the rapid development of antibiotic resistance shrinks the number of clinically available antibiotics, there is an urgent need for novel options to fill the existing antibiotic pipeline. In recent years, antimicrobial peptides have attracted increased interest due to their impressive broad-spectrum antimicrobial activity and low probability of antibiotic resistance. However, macromolecular antimicrobial peptides of plant and animal origin face obstacles in antibiotic development because of their extremely short elimination half-life and poor chemical stability. Herein, we focus on medium-sized antibacterial peptides (MAPs) of microbial origin with molecular weights below 2000 Da. The low molecular weight is not sufficient to form complex protein conformations and is also associated to a better chemical stability and easier modifications. Microbially-produced peptides are often composed of a variety of non-protein amino acids and terminal modifications, which contribute to improving the elimination half-life of compounds. Therefore, MAPs have great potential for drug discovery and are likely to become key players in the development of next-generation antibiotics. In this review, we provide a detailed exploration of the modes of action demonstrated by 45 MAPs and offer a concise summary of the structure-activity relationships observed in these MAPs.
Collapse
Affiliation(s)
- Jianwei Chen
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Wei Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xubin Hu
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yujie Yue
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xingyue Lu
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Chenjie Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Bin Wei
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Huawei Zhang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Hong Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
21
|
Monselise EBI, Vyazmensky M, Scherf T, Batushansky A, Fishov I. D-Glutamate production by stressed Escherichia coli gives a clue for the hypothetical induction mechanism of the ALS disease. Sci Rep 2024; 14:18247. [PMID: 39107374 PMCID: PMC11303787 DOI: 10.1038/s41598-024-68645-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
In the search for the origin of Amyotrophic Lateral Sclerosis disease (ALS), we hypothesized earlier (Monselise, 2019) that D-amino acids produced by stressed microbiome may serve as inducers of the disease development. Many examples of D-amino acid accumulation under various stress conditions were demonstrated in prokaryotic and eukaryotic cells. In this work, wild-type Escherichia coli, members of the digestive system, were subjected to carbon and nitrogen starvation stress. Using NMR and LC-MS techniques, we found for the first time that D-glutamate accumulated in the stressed bacteria but not in control cells. These results together with the existing knowledge, allow us to suggest a new insight into the pathway of ALS development: D-glutamate, produced by the stressed microbiome, induces neurobiochemical miscommunication setting on C1q of the complement system. Proving this insight may have great importance in preventive medicine of such MND modern-age diseases as ALS, Alzheimer, and Parkinson.
Collapse
Affiliation(s)
- Edna Ben-Izhak Monselise
- Department of Life Science, Bergman Campus, Ben-Gurion University of the Negev, 8441901, Beer-Sheva, Israel.
| | - Maria Vyazmensky
- Department of Life Science, Bergman Campus, Ben-Gurion University of the Negev, 8441901, Beer-Sheva, Israel
| | - Tali Scherf
- Department of Chemical Research Support, The Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Albert Batushansky
- Ilse Katz Institute for Nanoscale Science & Technology, Marcus Campus, Ben-Gurion University of the Negev, 8410501, Beer-Sheva, Israel
| | - Itzhak Fishov
- Department of Life Science, Bergman Campus, Ben-Gurion University of the Negev, 8441901, Beer-Sheva, Israel.
| |
Collapse
|
22
|
BELITSKY BORISR. Histidine kinase-mediated cross-regulation of the vancomycin-resistance operon in Clostridioides difficile. Mol Microbiol 2024; 121:1182-1199. [PMID: 38690761 PMCID: PMC11176017 DOI: 10.1111/mmi.15273] [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: 02/20/2024] [Revised: 04/03/2024] [Accepted: 04/17/2024] [Indexed: 05/03/2024]
Abstract
The dipeptide D-Ala-D-Ala is an essential component of peptidoglycan and the target of vancomycin. Most Clostridioides difficile strains possess the vanG operon responsible for the synthesis of D-Ala-D-Ser, which can replace D-Ala-D-Ala in peptidoglycan. The C. difficile vanG operon is regulated by a two-component system, VanRS, but is not induced sufficiently by vancomycin to confer resistance to this antibiotic. Surprisingly, in the absence of the VanS histidine kinase (HK), the vanG operon is still induced by vancomycin and also by another antibiotic, ramoplanin, in a VanR-dependent manner. This suggested the cross-regulation of VanR by another HK or kinases that are activated in the presence of certain lipid II-targeting antibiotics. We identified these HKs as CD35990 and CD22880. However, mutations in either or both HKs did not affect the regulation of the vanG operon in wild-type cells suggesting that intact VanS prevents the cross-activation of VanR by non-cognate HKs. Overproduction of VanR in the absence of VanS, CD35990, and CD22880 led to high expression of the vanG operon indicating that VanR can potentially utilize at least one more phosphate donor for its activation. Candidate targets of CD35990- and CD22880-mediated regulation in the presence of vancomycin or ramoplanin were identified by RNA-Seq.
Collapse
Affiliation(s)
- BORIS R. BELITSKY
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts 02111, USA
| |
Collapse
|
23
|
Rajguru V, Chatterjee S, Garde S, Reddy M. Crosslink cleaving enzymes: the smart autolysins that remodel the bacterial cell wall. Trends Microbiol 2024; 32:494-506. [PMID: 38072724 DOI: 10.1016/j.tim.2023.11.004] [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: 09/11/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 05/12/2024]
Abstract
Peptidoglycan (PG) is a protective mesh-like polymer in bacterial cell walls that enables their survival in almost every ecological niche. PG is formed by crosslinking of several glycan strands through short peptides, conferring a characteristic structure and elasticity, distinguishing it from other polymeric exoskeletons. The significance of PG crosslink formation has been known for decades, as some of the most widely used antibiotics, namely β-lactams, target the enzymes that catalyze this step. However, the importance of crosslink hydrolysis in PG biology remained largely underappreciated. Recent advances demonstrate the functions of crosslink cleavage in diverse physiological processes, including an indispensable role in PG expansion during the cell cycle, thereby making crosslink cleaving enzymes an untapped target for novel drugs. Here, we elaborate on the fundamental roles of crosslink-specific endopeptidases and their regulation across the bacterial kingdom.
Collapse
Affiliation(s)
- Vaidehi Rajguru
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Stuti Chatterjee
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shambhavi Garde
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, 500007, India
| | - Manjula Reddy
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
24
|
Hogan AM, Motnenko A, Rahman ASMZ, Cardona ST. Cell envelope structural and functional contributions to antibiotic resistance in Burkholderia cenocepacia. J Bacteriol 2024; 206:e0044123. [PMID: 38501654 PMCID: PMC11025338 DOI: 10.1128/jb.00441-23] [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: 12/29/2023] [Accepted: 03/05/2024] [Indexed: 03/20/2024] Open
Abstract
Antibiotic activity is limited by the physical construction of the Gram-negative cell envelope. Species of the Burkholderia cepacia complex (Bcc) are known as intrinsically multidrug-resistant opportunistic pathogens with low permeability cell envelopes. Here, we re-examined a previously performed chemical-genetic screen of barcoded transposon mutants in B. cenocepacia K56-2, focusing on cell envelope structural and functional processes. We identified structures mechanistically important for resistance to singular and multiple antibiotic classes. For example, susceptibility to novobiocin, avibactam, and the LpxC inhibitor, PF-04753299, was linked to the BpeAB-OprB efflux pump, suggesting these drugs are substrates for this pump in B. cenocepacia. Defects in peptidoglycan precursor synthesis specifically increased susceptibility to cycloserine and revealed a new putative amino acid racemase, while defects in divisome accessory proteins increased susceptibility to multiple β-lactams. Additionally, disruption of the periplasmic disulfide bond formation system caused pleiotropic defects on outer membrane integrity and β-lactamase activity. Our findings highlight the layering of resistance mechanisms in the structure and function of the cell envelope. Consequently, we point out processes that can be targeted for developing antibiotic potentiators.IMPORTANCEThe Gram-negative cell envelope is a double-layered physical barrier that protects cells from extracellular stressors, such as antibiotics. The Burkholderia cell envelope is known to contain additional modifications that reduce permeability. We investigated Burkholderia cell envelope factors contributing to antibiotic resistance from a genome-wide view by re-examining data from a transposon mutant library exposed to an antibiotic panel. We identified susceptible phenotypes for defects in structures and functions in the outer membrane, periplasm, and cytoplasm. Overall, we show that resistance linked to the cell envelope is multifaceted and provides new targets for the development of antibiotic potentiators.
Collapse
Affiliation(s)
- Andrew M. Hogan
- Department of Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Anna Motnenko
- Department of Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | - Silvia T. Cardona
- Department of Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
25
|
Hsu TW, Fang JM. Advances and prospects of analytic methods for bacterial transglycosylation and inhibitor discovery. Analyst 2024; 149:2204-2222. [PMID: 38517346 DOI: 10.1039/d3an01968c] [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: 03/23/2024]
Abstract
The cell wall is essential for bacteria to maintain structural rigidity and withstand external osmotic pressure. In bacteria, the cell wall is composed of peptidoglycan. Lipid II is the basic unit for constructing highly cross-linked peptidoglycan scaffolds. Transglycosylase (TGase) is the initiating enzyme in peptidoglycan synthesis that catalyzes the ligation of lipid II moieties into repeating GlcNAc-MurNAc polysaccharides, followed by transpeptidation to generate cross-linked structures. In addition to the transglycosylases in the class-A penicillin-binding proteins (aPBPs), SEDS (shape, elongation, division and sporulation) proteins are also present in most bacteria and play vital roles in cell wall renewal, elongation, and division. In this review, we focus on the latest analytical methods including the use of radioactive labeling, gel electrophoresis, mass spectrometry, fluorescence labeling, probing undecaprenyl pyrophosphate, fluorescence anisotropy, ligand-binding-induced tryptophan fluorescence quenching, and surface plasmon resonance to evaluate TGase activity in cell wall formation. This review also covers the discovery of TGase inhibitors as potential antibacterial agents. We hope that this review will give readers a better understanding of the chemistry and basic research for the development of novel antibiotics.
Collapse
Affiliation(s)
- Tse-Wei Hsu
- Department of Chemistry, National Taiwan University, Taipei 106, Taiwan.
| | - Jim-Min Fang
- Department of Chemistry, National Taiwan University, Taipei 106, Taiwan.
| |
Collapse
|
26
|
Roney IJ, Rudner DZ. Bacillus subtilis uses the SigM signaling pathway to prioritize the use of its lipid carrier for cell wall synthesis. PLoS Biol 2024; 22:e3002589. [PMID: 38683856 PMCID: PMC11081497 DOI: 10.1371/journal.pbio.3002589] [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: 08/25/2023] [Revised: 05/09/2024] [Accepted: 03/13/2024] [Indexed: 05/02/2024] Open
Abstract
Peptidoglycan (PG) and most surface glycopolymers and their modifications are built in the cytoplasm on the lipid carrier undecaprenyl phosphate (UndP). These lipid-linked precursors are then flipped across the membrane and polymerized or directly transferred to surface polymers, lipids, or proteins. Despite its essential role in envelope biogenesis, UndP is maintained at low levels in the cytoplasmic membrane. The mechanisms by which bacteria distribute this limited resource among competing pathways is currently unknown. Here, we report that the Bacillus subtilis transcription factor SigM and its membrane-anchored anti-sigma factor respond to UndP levels and prioritize its use for the synthesis of the only essential surface polymer, the cell wall. Antibiotics that target virtually every step in PG synthesis activate SigM-directed gene expression, confounding identification of the signal and the logic of this stress-response pathway. Through systematic analyses, we discovered 2 distinct responses to these antibiotics. Drugs that trap UndP, UndP-linked intermediates, or precursors trigger SigM release from the membrane in <2 min, rapidly activating transcription. By contrasts, antibiotics that inhibited cell wall synthesis without directly affecting UndP induce SigM more slowly. We show that activation in the latter case can be explained by the accumulation of UndP-linked wall teichoic acid precursors that cannot be transferred to the PG due to the block in its synthesis. Furthermore, we report that reduction in UndP synthesis rapidly induces SigM, while increasing UndP production can dampen the SigM response. Finally, we show that SigM becomes essential for viability when the availability of UndP is restricted. Altogether, our data support a model in which the SigM pathway functions to homeostatically control UndP usage. When UndP levels are sufficiently high, the anti-sigma factor complex holds SigM inactive. When levels of UndP are reduced, SigM activates genes that increase flux through the PG synthesis pathway, boost UndP recycling, and liberate the lipid carrier from nonessential surface polymer pathways. Analogous homeostatic pathways that prioritize UndP usage are likely to be common in bacteria.
Collapse
Affiliation(s)
- Ian J. Roney
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - David Z. Rudner
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
27
|
de Oliveira MVD, da Costa KS, Silva JRA, Lameira J, Lima AH. Role of UDP-N-acetylmuramic acid in the regulation of MurA activity revealed by molecular dynamics simulations. Protein Sci 2024; 33:e4969. [PMID: 38532715 PMCID: PMC10966352 DOI: 10.1002/pro.4969] [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: 10/14/2023] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024]
Abstract
The peptidoglycan biosynthesis pathway plays a vital role in bacterial cells, and facilitates peptidoglycan layer formation, a fundamental structural component of the bacterial cell wall. The enzymes in this pathway are candidates for antibiotic development, as most do not have mammalian homologues. The UDP-N-acetylglucosamine (UNAG) enolpyruvyl transferase enzyme (MurA) in the peptidoglycan pathway cytoplasmic step is responsible for the phosphoenolpyruvate (PEP)-UNAG catalytic reaction, forming UNAG enolpyruvate and inorganic phosphate. Reportedly, UDP-N-acetylmuramic acid (UNAM) binds tightly to MurA forming a dormant UNAM-PEP-MurA complex and acting as a MurA feedback inhibitor. MurA inhibitors are complex, owing to competitive binding interactions with PEP, UNAM, and UNAG at the MurA active site. We used computational methods to explore UNAM and UNAG binding. UNAM showed stronger hydrogen-bond interactions with the Arg120 and Arg91 residues, which help to stabilize the closed conformation of MurA, than UNAG. Binding free energy calculations using end-point computational methods showed that UNAM has a higher binding affinity than UNAG, when PEP is attached to Cys115. The unbinding process, simulated using τ-random acceleration molecular dynamics, showed that UNAM has a longer relative residence time than UNAG, which is related to several complex dissociation pathways, each with multiple intermediate metastable states. This prevents the loop from opening and exposing the Arg120 residue to accommodate UNAG and potential new ligands. Moreover, we demonstrate the importance of Cys115-linked PEP in closed-state loop stabilization. We provide a basis for evaluating novel UNAM analogues as potential MurA inhibitors. PUBLIC SIGNIFICANCE: MurA is a critical enzyme involved in bacterial cell wall biosynthesis and is involved in antibiotic resistance development. UNAM can remain in the target protein's active site for an extended time compared to its natural substrate, UNAG. The prolonged interaction of this highly stable complex known as the 'dormant complex' comprises UNAM-PEP-MurA and offers insights into antibiotic development, providing potential options against drug-resistant bacteria and advancing our understanding of microbial biology.
Collapse
Affiliation(s)
| | - Kauê S. da Costa
- Institute of BiodiversityFederal University of Western ParáSantarémParáBrazil
| | - José Rogério A. Silva
- Laboratório de Planejamento e Desenvolvimento de Fármacos, Instituto de Ciências Exatas e NaturaisUniversidade Federal do ParáBelémParáBrazil
- Catalysis and Peptide Research UnitUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - Jerônimo Lameira
- Laboratório de Planejamento e Desenvolvimento de Fármacos, Instituto de Ciências Exatas e NaturaisUniversidade Federal do ParáBelémParáBrazil
| | - Anderson H. Lima
- Laboratório de Planejamento e Desenvolvimento de Fármacos, Instituto de Ciências Exatas e NaturaisUniversidade Federal do ParáBelémParáBrazil
| |
Collapse
|
28
|
Shtaiwi A, Khan SU, Khedraoui M, Alaraj M, Samadi A, Chtita S. A comprehensive computational study to explore promising natural bioactive compounds targeting glycosyltransferase MurG in Escherichia coli for potential drug development. Sci Rep 2024; 14:7098. [PMID: 38532068 DOI: 10.1038/s41598-024-57702-x] [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: 11/24/2023] [Accepted: 03/21/2024] [Indexed: 03/28/2024] Open
Abstract
Peptidoglycan is a carbohydrate with a cross-linked structure that protects the cytoplasmic membrane of bacterial cells from damage. The mechanism of peptidoglycan biosynthesis involves the main synthesizing enzyme glycosyltransferase MurG, which is known as a potential target for antibiotic therapy. Many MurG inhibitors have been recognized as MurG targets, but high toxicity and drug-resistant Escherichia coli strains remain the most important problems for further development. In addition, the discovery of selective MurG inhibitors has been limited to the synthesis of peptidoglycan-mimicking compounds. The present study employed drug discovery, such as virtual screening using molecular docking, drug likeness ADMET proprieties predictions, and molecular dynamics (MD) simulation, to identify potential natural products (NPs) for Escherichia coli. We conducted a screening of 30,926 NPs from the NPASS database. Subsequently, 20 of these compounds successfully passed the potency, pharmacokinetic, ADMET screening assays, and their validation was further confirmed through molecular docking. The best three hits and the standard were chosen for further MD simulations up to 400 ns and energy calculations to investigate the stability of the NPs-MurG complexes. The analyses of MD simulations and total binding energies suggested the higher stability of NPC272174. The potential compounds can be further explored in vivo and in vitro for promising novel antibacterial drug discovery.
Collapse
Affiliation(s)
- Amneh Shtaiwi
- Faculty of Pharmacy, Middle East University, Queen Alia Airport Street, Amman, P.O. Box No. 11610, Jordan.
| | - Shafi Ullah Khan
- Interdisciplinary Research Unit for Cancer Prevention and Treatment, Baclesse Cancer Centre, Université de Caen Normandie Inserm Anticipe UMR 1086, Normandie Univ, Research Building, F‑14000 François 3 Avenue Général Harris, BP 45026, 14076, Cedex 05 Caen, France
- Centre François Baclesse, Avenue Général Harris, 14076, Caen Cedex, France
| | - Meriem Khedraoui
- Laboratory of Analytical and Molecular Chemistry, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, B. P 7955, Casablanca, Morocco
| | - Mohd Alaraj
- Faculty of Pharmacy, University of Jerash, Jerash, Jordan
| | - Abdelouahid Samadi
- Department of Chemistry, College of Science, UAEU, P.O. Box No. 15551, Al Ain, UAE.
| | - Samir Chtita
- Laboratory of Analytical and Molecular Chemistry, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, B. P 7955, Casablanca, Morocco
| |
Collapse
|
29
|
Li Q, Zhou W, Yu X, Cui F, Tan X, Sun T, Li J. Preparation and characterization of zein/gelatin electrospun film loaded with ε-polylysine and gallic acid as tuna packaging system. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:1942-1952. [PMID: 37886811 DOI: 10.1002/jsfa.13080] [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: 04/13/2023] [Revised: 08/29/2023] [Accepted: 10/27/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Composite nanofiber films loaded with ε-polylysine (PL) and gallic acid (GA) were prepared using a zein/gelatin (ZG) electrospinning method to develop effective active packaging films for tuna preservation. The morphology, structure, thermal stability, hydrophobicity, antibacterial, and antioxidant properties of the films, and their application for tuna during a period of storage of 4 °C were investigated. RESULTS PL reduced the average diameter of ZG fibers, whereas GA increased it. The PL/GA/ZG film possessed a well distributed fiber morphology with an average diameter of 810 ± 150 nm. Fourier-transform infrared spectroscopy and X-ray diffraction results showed the physical loading of PL and GA in ZG film with the main chemical bonds and crystal structure unchanged. The addition of both PL and GA reduced hydrophobicity of the ZG film while the PL/GA/ZG film was still hydrophobic. GA enhanced its thermal stability and contributed to its antioxidant activity. PL and GA synergetically enhanced the antibacterial activity of ZG film against Shewanella putrefaciens. PL combined with GA is more suitable for modifying ZG film than GA alone. The PL/GA/ZG film effectively inhibited total viable counts, total volatile base nitrogen, fat oxidation, and texture deterioration of tuna fillets at 4 °C storage, and could extend the shelf life by 3 days. CONCLUSIONS The PL/GA/ZG nanofiber film demonstrated promising potential for application in the preservation of aquatic products as a new antibacterial and antioxidant food packaging. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Qiuying Li
- College of Food Science and Engineering, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, Jinzhou, China
| | - Wenxuan Zhou
- College of Food Science and Engineering, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, Jinzhou, China
| | - Xinrui Yu
- College of Food Science and Engineering, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, Jinzhou, China
| | - Fangchao Cui
- College of Food Science and Engineering, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, Jinzhou, China
| | - Xiqian Tan
- College of Food Science and Engineering, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, Jinzhou, China
| | - Tong Sun
- College of Food Science and Engineering, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, Jinzhou, China
| | - Jianrong Li
- College of Food Science and Engineering, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, Jinzhou, China
| |
Collapse
|
30
|
Oluwole A, Hernández-Rocamora VM, Cao Y, Li X, Vollmer W, Robinson CV, Bolla JR. Real-Time Biosynthetic Reaction Monitoring Informs the Mechanism of Action of Antibiotics. J Am Chem Soc 2024; 146:7007-7017. [PMID: 38428018 PMCID: PMC10941186 DOI: 10.1021/jacs.4c00081] [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: 01/05/2024] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 03/03/2024]
Abstract
The rapid spread of drug-resistant pathogens and the declining discovery of new antibiotics have created a global health crisis and heightened interest in the search for novel antibiotics. Beyond their discovery, elucidating mechanisms of action has necessitated new approaches, especially for antibiotics that interact with lipidic substrates and membrane proteins. Here, we develop a methodology for real-time reaction monitoring of the activities of two bacterial membrane phosphatases, UppP and PgpB. We then show how we can inhibit their activities using existing and newly discovered antibiotics such as bacitracin and teixobactin. Additionally, we found that the UppP dimer is stabilized by phosphatidylethanolamine, which, unexpectedly, enhanced the speed of substrate processing. Overall, our results demonstrate the potential of native mass spectrometry for real-time biosynthetic reaction monitoring of membrane enzymes, as well as their in situ inhibition and cofactor binding, to inform the mode of action of emerging antibiotics.
Collapse
Affiliation(s)
- Abraham
O. Oluwole
- Department
of Chemistry, University of Oxford, South Parks Road, Oxford OX1 3QZ, U.K.
- The
Kavli Institute for Nanoscience Discovery, University of Oxford, South Parks Road, Oxford OX1 3QU, U.K.
| | - Víctor M. Hernández-Rocamora
- Centre
for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Richardson Road, Newcastle upon Tyne NE2 4AX, U.K.
| | - Yihui Cao
- Department
of Chemistry, State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR 999077, China
| | - Xuechen Li
- Department
of Chemistry, State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR 999077, China
| | - Waldemar Vollmer
- Centre
for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Richardson Road, Newcastle upon Tyne NE2 4AX, U.K.
- Institute
for Molecular Bioscience, University of
Queensland, Carmody Road, Brisbane, Queensland 4072, Australia
| | - Carol V. Robinson
- Department
of Chemistry, University of Oxford, South Parks Road, Oxford OX1 3QZ, U.K.
- The
Kavli Institute for Nanoscience Discovery, University of Oxford, South Parks Road, Oxford OX1 3QU, U.K.
| | - Jani R. Bolla
- The
Kavli Institute for Nanoscience Discovery, University of Oxford, South Parks Road, Oxford OX1 3QU, U.K.
- Department
of Biology, University of Oxford, South Parks Road, Oxford OX1 3RB, U.K.
| |
Collapse
|
31
|
Koatale P, Welling MM, Ndlovu H, Kgatle M, Mdanda S, Mdlophane A, Okem A, Takyi-Williams J, Sathekge MM, Ebenhan T. Insights into Peptidoglycan-Targeting Radiotracers for Imaging Bacterial Infections: Updates, Challenges, and Future Perspectives. ACS Infect Dis 2024; 10:270-286. [PMID: 38290525 PMCID: PMC10862554 DOI: 10.1021/acsinfecdis.3c00443] [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: 08/28/2023] [Revised: 12/16/2023] [Accepted: 12/18/2023] [Indexed: 02/01/2024]
Abstract
The unique structural architecture of the peptidoglycan allows for the stratification of bacteria as either Gram-negative or Gram-positive, which makes bacterial cells distinguishable from mammalian cells. This classification has received attention as a potential target for diagnostic and therapeutic purposes. Bacteria's ability to metabolically integrate peptidoglycan precursors during cell wall biosynthesis and recycling offers an opportunity to target and image pathogens in their biological state. This Review explores the peptidoglycan biosynthesis for bacteria-specific targeting for infection imaging. Current and potential radiolabeled peptidoglycan precursors for bacterial infection imaging, their development status, and their performance in vitro and/or in vivo are highlighted. We conclude by providing our thoughts on how to shape this area of research for future clinical translation.
Collapse
Affiliation(s)
- Palesa
C. Koatale
- Department
of Nuclear Medicine, University of Pretoria, 0001 Pretoria, South Africa
- Nuclear
Medicine Research Infrastructure (NuMeRI) NPC, 0001 Pretoria, South Africa
| | - Mick M. Welling
- Interventional
Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Honest Ndlovu
- Department
of Nuclear Medicine, University of Pretoria, 0001 Pretoria, South Africa
- Nuclear
Medicine Research Infrastructure (NuMeRI) NPC, 0001 Pretoria, South Africa
| | - Mankgopo Kgatle
- Department
of Nuclear Medicine, University of Pretoria, 0001 Pretoria, South Africa
- Nuclear
Medicine Research Infrastructure (NuMeRI) NPC, 0001 Pretoria, South Africa
| | - Sipho Mdanda
- Department
of Nuclear Medicine, University of Pretoria, 0001 Pretoria, South Africa
- Nuclear
Medicine Research Infrastructure (NuMeRI) NPC, 0001 Pretoria, South Africa
| | - Amanda Mdlophane
- Department
of Nuclear Medicine, University of Pretoria, 0001 Pretoria, South Africa
- Nuclear
Medicine Research Infrastructure (NuMeRI) NPC, 0001 Pretoria, South Africa
| | - Ambrose Okem
- Department
of Anaesthesia, School of Clinical Medicine, University of Witwatersrand, 2050 Johannesburg, South Africa
| | - John Takyi-Williams
- Pharmacokinetic
and Mass Spectrometry Core, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Mike M. Sathekge
- Department
of Nuclear Medicine, University of Pretoria, 0001 Pretoria, South Africa
- Nuclear
Medicine Research Infrastructure (NuMeRI) NPC, 0001 Pretoria, South Africa
| | - Thomas Ebenhan
- Department
of Nuclear Medicine, University of Pretoria, 0001 Pretoria, South Africa
- Nuclear
Medicine Research Infrastructure (NuMeRI) NPC, 0001 Pretoria, South Africa
- DSI/NWU Pre-clinical
Drug Development Platform, North West University, 2520 Potchefstroom, South Africa
| |
Collapse
|
32
|
Orsini Delgado ML, Gamelas Magalhaes J, Morra R, Cultrone A. Muropeptides and muropeptide transporters impact on host immune response. Gut Microbes 2024; 16:2418412. [PMID: 39439228 PMCID: PMC11509177 DOI: 10.1080/19490976.2024.2418412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024] Open
Abstract
In bacteria, the cell envelope is the key element surrounding and protecting the bacterial content from mechanical or osmotic damages. It allows the selective interchanges of solutes, ions, cellular debris, and drugs between the cellular compartments and the external environment, thanks to the presence of transmembrane proteins called transporters. The major component of the cell envelope is the peptidoglycan, consisting of long linear glycan strands cross-linked by short peptide stems. During cell growth or under stress conditions, peptidoglycan fragments, the muropeptides, are released by bacteria and recognized by the host Pattern Recognition Receptor, promoting the activation of their innate defense mechanisms. The review sums up the salient aspects of microbiota-host interaction with a focus on the NOD-dependent immune response to bacterial peptidoglycan and on the accountability of muropeptide transporters in the crosstalk with the host and in antibiotic resistance. Furthermore, it retraces the discoveries and applications of microorganisms-derived components such as vaccines or vaccine adjuvants.
Collapse
|
33
|
King KA, Benton AH, Caudill MT, Stoyanof ST, Kang L, Michalak P, Lahmers KK, Dunman PM, DeHart TG, Ahmad SS, Jutras BL, Poncin K, De Bolle X, Caswell CC. Post-transcriptional control of the essential enzyme MurF by a small regulatory RNA in Brucella abortus. Mol Microbiol 2024; 121:129-141. [PMID: 38082493 DOI: 10.1111/mmi.15207] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/22/2023] [Accepted: 11/25/2023] [Indexed: 01/15/2024]
Abstract
Brucella abortus is a facultative, intracellular, zoonotic pathogen that resides inside macrophages during infection. This is a specialized niche where B. abortus encounters various stresses as it navigates through the macrophage. In order to survive this harsh environment, B. abortus utilizes post-transcriptional regulation of gene expression through the use of small regulatory RNAs (sRNAs). Here, we characterize a Brucella sRNAs called MavR (for MurF- and virulence-regulating sRNA), and we demonstrate that MavR is required for the full virulence of B. abortus in macrophages and in a mouse model of chronic infection. Transcriptomic and proteomic studies revealed that a major regulatory target of MavR is MurF. MurF is an essential protein that catalyzes the final cytoplasmic step in peptidoglycan (PG) synthesis; however, we did not detect any differences in the amount or chemical composition of PG in the ΔmavR mutant. A 6-nucleotide regulatory seed region within MavR was identified, and mutation of this seed region resulted in dysregulation of MurF production, as well as significant attenuation of infection in a mouse model. Overall, the present study underscores the importance of sRNA regulation in the physiology and virulence of Brucella.
Collapse
Affiliation(s)
- Kellie A King
- Department of Biomedical Sciences and Pathobiology, Center for One Health Research, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Angela H Benton
- Department of Biomedical Sciences and Pathobiology, Center for One Health Research, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Mitchell T Caudill
- Department of Biomedical Sciences and Pathobiology, Center for One Health Research, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - S Tristan Stoyanof
- Department of Biomedical Sciences and Pathobiology, Center for One Health Research, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Lin Kang
- Department of Biomedical Sciences, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, USA
- College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
- Center for One Health Research, Edward Via College of Osteopathic Medicine, Blacksburg, Virginia, USA
| | - Pawel Michalak
- Department of Biomedical Sciences, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, USA
- Center for One Health Research, Edward Via College of Osteopathic Medicine, Blacksburg, Virginia, USA
- Institute for Evolution, University of Haifa, Haifa, Israel
| | - Kevin K Lahmers
- Department of Biomedical Sciences and Pathobiology, Center for One Health Research, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Paul M Dunman
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Tanner G DeHart
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia, USA
| | - Saadman S Ahmad
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia, USA
| | - Brandon L Jutras
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia, USA
| | - Katy Poncin
- URBM, Narilis, University of Namur, Namur, Belgium
| | | | - Clayton C Caswell
- Department of Biomedical Sciences and Pathobiology, Center for One Health Research, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
34
|
Hussein M, Allobawi R, Zhao J, Yu H, Neville SL, Wilksch J, Wong LJM, Baker M, McDevitt CA, Rao GG, Li J, Velkov T. Integrated Transcriptomic and Metabolomic Mapping Reveals the Mechanism of Action of Ceftazidime/Avibactam against Pan-Drug-Resistant Klebsiella pneumoniae. ACS Infect Dis 2023; 9:2409-2422. [PMID: 37878861 PMCID: PMC10714405 DOI: 10.1021/acsinfecdis.3c00264] [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: 06/05/2023] [Revised: 10/02/2023] [Accepted: 10/11/2023] [Indexed: 10/27/2023]
Abstract
Here, we employed an integrated metabolomics and transcriptomics approach to investigate the molecular mechanism(s) of action of ceftazidime/avibactam against a pan-drug-resistant K. pneumoniae clinical isolate from a patient with urinary tract infection. Ceftazidime/avibactam induced time-dependent perturbations in the metabolome and transcriptome of the bacterium, mainly at 6 h, with minimal effects at 1 and 3 h. Metabolomics analysis revealed a notable reduction in essential lipids involved in outer membrane glycerolipid biogenesis. This disruption effect extended to peptidoglycan and lipopolysaccharide biosynthetic pathways, including lipid A and O-antigen assembly. Importantly, ceftazidime/avibactam not only affected the final steps of peptidoglycan biosynthesis in the periplasm, a common mechanism of ceftazidime action, but also influenced the synthesis of lipid-linked intermediates and early stages of cytoplasmic peptidoglycan synthesis. Furthermore, ceftazidime/avibactam substantially inhibited central carbon metabolism (e.g., the pentose phosphate pathway and tricarboxylic acid cycle). Consistently, the dysregulation of genes governing these metabolic pathways aligned with the metabolomics findings. Certain metabolomics and transcriptomics signatures associated with ceftazidime resistance were also perturbed. Consistent with the primary target of antibiotic activity, biochemical assays also confirmed the direct impact of ceftazidime/avibactam on peptidoglycan production. This study explored the intricate interactions of ceftazidime and avibactam within bacterial cells, including their impact on cell envelope biogenesis and central carbon metabolism. Our findings revealed the complexities of how ceftazidime/avibactam operates, such as hindering peptidoglycan formation in different cellular compartments. In summary, this study confirms the existing hypotheses about the antibacterial and resistance mechanisms of ceftazidime/avibactam while uncovering novel insights, including its impact on lipopolysaccharide formation.
Collapse
Affiliation(s)
- Maytham Hussein
- Monash
Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| | - Rafah Allobawi
- Monash
Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| | - Jinxin Zhao
- Monash
Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| | - Heidi Yu
- Monash
Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| | - Stephanie L. Neville
- Department
of Microbiology and Immunology, The Peter Doherty Institute for Infection
and Immunity, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Jonathan Wilksch
- Department
of Microbiology and Immunology, The Peter Doherty Institute for Infection
and Immunity, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Labell J. M. Wong
- Monash
Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| | - Mark Baker
- Discipline
of Biological Sciences, Priority Research Centre in Reproductive Biology,
Faculty of Science and IT, University of
Newcastle, University
Drive, Callaghan, NSW 2308, Australia
| | - Christopher A. McDevitt
- Department
of Microbiology and Immunology, The Peter Doherty Institute for Infection
and Immunity, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Gauri G. Rao
- Division
of Pharmacotherapy and Experimental Therapeutics, Eshelman School
of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599-7355, United
States
| | - Jian Li
- Monash
Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| | - Tony Velkov
- Monash
Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
35
|
Wagdy RA, Abutaleb NS, Fathalla RK, Elgammal Y, Weck S, Pal R, Fischer PD, Ducho C, Abadi AH, N Seleem M, Engel M, Abdel-Halim M. Discovery of 1,2-diaryl-3-oxopyrazolidin-4-carboxamides as a new class of MurA enzyme inhibitors and characterization of their antibacterial activity. Eur J Med Chem 2023; 261:115789. [PMID: 37717380 DOI: 10.1016/j.ejmech.2023.115789] [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: 04/13/2023] [Revised: 08/29/2023] [Accepted: 09/01/2023] [Indexed: 09/19/2023]
Abstract
The cytoplasmic steps of peptidoglycan synthesis represent an important targeted pathway for development of new antibiotics. Herein, we report the synthesis of novel 3-oxopyrazolidin-4-carboxamide derivatives with variable amide side chains as potential antibacterial agents targeting MurA enzyme, the first committed enzyme in these cytosolic steps. Compounds 15 (isoindoline-1,3-dione-5-yl), 16 (4-(1H-pyrazol-4-yl)phenyl), 20 (5-cyanothiazol-2-yl), 21 and 31 (5-nitrothiazol-2-yl derivatives) exhibited the most potent MurA inhibition, with IC50 values of 9.8-12.2 μM. Compounds 15, 16 and 21 showed equipotent inhibition of the C115D MurA mutant developed by fosfomycin-resistant Escherichia coli. NMR binding studies revealed that some of the MurA residues targeted by 15 also interacted with fosfomycin, but not all, indicating an overlapping but not identical binding site. The antibacterial activity of the compounds against E. coli ΔtolC suggests that inhibition of MurA accounts for the observed effect on bacterial growth, considering that a few potent MurA inhibitors could not penetrate the bacterial outer membrane and were therefore inactive as proven by the bacterial cell uptake assay. The most promising compounds were also evaluated against a panel of Gram-positive bacteria. Remarkably, compounds 21 and 31 (MurA IC50 = 9.8 and 10.2 μM respectively) exhibited a potent activity against Clostridioides difficile strains with MIC values ranging from 0.125 to 1 μg/mL, and were also shown to be bactericidal with MBC values between 0.25 and 1 μg/mL. Furthermore, both compounds were shown to have a limited activity against human normal intestinal flora and showed high safety towards human colon cells (Caco-2) in vitro. The thiolactone derivative (compound 5) exhibited an interesting broad spectrum antibacterial activity despite its weak MurA inhibition. Altogether, the presented series provides a promising class of antibiotics that merits further investigation.
Collapse
Affiliation(s)
- Reem A Wagdy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, 11835, Egypt
| | - Nader S Abutaleb
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA; Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Reem K Fathalla
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, D-66123, Saarbrücken, Germany
| | - Yehia Elgammal
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Stefanie Weck
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, D-66123, Saarbrücken, Germany
| | - Rusha Pal
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Patrick D Fischer
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, D-66123, Saarbrücken, Germany; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Christian Ducho
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, D-66123, Saarbrücken, Germany
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, 11835, Egypt
| | - Mohamed N Seleem
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA; Center for One Health Research, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Matthias Engel
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, D-66123, Saarbrücken, Germany.
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, 11835, Egypt.
| |
Collapse
|
36
|
Rambaher MH, Zdovc I, Glavač NK, Gobec S, Frlan R. Mur ligase F as a new target for the flavonoids quercitrin, myricetin, and (-)-epicatechin. J Comput Aided Mol Des 2023; 37:721-733. [PMID: 37796382 PMCID: PMC10618370 DOI: 10.1007/s10822-023-00535-z] [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: 07/12/2023] [Accepted: 09/14/2023] [Indexed: 10/06/2023]
Abstract
MurC, D, E, and F are ATP-dependent ligases involved in the stepwise assembly of the tetrapeptide stem of forming peptidoglycan. As highly conserved targets found exclusively in bacterial cells, they are of significant interest for antibacterial drug discovery. In this study, we employed a computer-aided molecular design approach to identify potential inhibitors of MurF. A biochemical inhibition assay was conducted, screening twenty-four flavonoids and related compounds against MurC-F, resulting in the identification of quercitrin, myricetin, and (-)-epicatechin as MurF inhibitors with IC50 values of 143 µM, 139 µM, and 92 µM, respectively. Notably, (-)-epicatechin demonstrated mixed type inhibition with ATP and uncompetitive inhibition with D-Ala-D-Ala dipeptide and UM3DAP substrates. Furthermore, in silico analysis using Sitemap and subsequent docking analysis using Glide revealed two plausible binding sites for (-)-epicatechin. The study also investigated the crucial structural features required for activity, with a particular focus on the substitution pattern and hydroxyl group positions, which were found to be important for the activity. The study highlights the significance of computational approaches in targeting essential enzymes involved in bacterial peptidoglycan synthesis.
Collapse
Affiliation(s)
- Martina Hrast Rambaher
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Irena Zdovc
- Veterinary Faculty, Institute of Microbiology and Parasitology, University of Ljubljana, Gerbičeva ul. 60, Ljubljana, Slovenia
| | - Nina Kočevar Glavač
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Stanislav Gobec
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia.
| | - Rok Frlan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia.
| |
Collapse
|
37
|
Proj M, Hrast M, Bajc G, Frlan R, Meden A, Butala M, Gobec S. Discovery of a fragment hit compound targeting D-Ala:D-Ala ligase of bacterial peptidoglycan biosynthesis. J Enzyme Inhib Med Chem 2023; 38:387-397. [PMID: 36446617 PMCID: PMC9718554 DOI: 10.1080/14756366.2022.2149745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Bacterial resistance is an increasing threat to healthcare systems, highlighting the need for discovering new antibacterial agents. An established technique, fragment-based drug discovery, was used to target a bacterial enzyme Ddl involved in the biosynthesis of peptidoglycan. We assembled general and focused fragment libraries that were screened in a biochemical inhibition assay. Screening revealed a new fragment-hit inhibitor of DdlB with a Ki value of 20.7 ± 4.5 µM. Binding to the enzyme was confirmed by an orthogonal biophysical method, surface plasmon resonance, making the hit a promising starting point for fragment development.
Collapse
Affiliation(s)
- Matic Proj
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Ljubljana, Ljubljana, Slovenia
| | - Martina Hrast
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Ljubljana, Ljubljana, Slovenia
| | - Gregor Bajc
- Biotechnical Faculty, Department of Biology, University of Ljubljana, Ljubljana, Slovenia
| | - Rok Frlan
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Ljubljana, Ljubljana, Slovenia
| | - Anže Meden
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Ljubljana, Ljubljana, Slovenia
| | - Matej Butala
- Biotechnical Faculty, Department of Biology, University of Ljubljana, Ljubljana, Slovenia
| | - Stanislav Gobec
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Ljubljana, Ljubljana, Slovenia,CONTACT Stanislav Gobec Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Ljubljana, Askerceva 7, 1000Ljubljana, Slovenia
| |
Collapse
|
38
|
Mishra NR, Sharma AD, Gargvanshi S, Gutheil WG. Deconvolution of multichannel LC-MS/MS chromatograms of glucosamine-phosphates: Evidence of a GlmS regulatory difference between Staphylococcus aureus and Enterococcus faecium. TALANTA OPEN 2023; 8:100241. [PMID: 38187186 PMCID: PMC10769159 DOI: 10.1016/j.talo.2023.100241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024] Open
Abstract
Resolving isomeric analytes is challenging given their physical similarity - making chromatographic resolution difficult, and their identical masses - making simple mass resolution impossible. MS/MS data provides a means to resolve isomeric analytes if their MS/MS intensity profiles are sufficiently different. Glucosamine-6-phosphate (GlcN-6P) and glucosamine-1-phosphate (GlcN-1P) are early bacterial cell wall intermediates. These and other isomeric hexosamine-phosphates are highly polar and unretained on reverse-phase chromatography media. Three commercially available hexosamine-phosphate standards (GlcN-6P, GlcN-1P, and GalN-1P) were derivatized with octanoic anhydride, and chromatographic conditions were established to resolve these analytes on C18 columns. GlcN-1P and GalN-1P overlapped chromatographically under all tested chromatography conditions. Three MS/MS fragments (79, 97, and 199 m/z) were common to all three commercially available hexosamine-phosphates. Intensity ratios of the three MS/MS fragments from these three hexosamine-phosphate standards were used to deconvolute mixture chromatograms of these standards by non-negative linear regression. This approach allowed the complete resolution of these analytes. The chromatographically overlapping GlcN-1P and GalN-1P, which shared similar but modestly different MS/MS intensity profiles, were fully resolved with this non-negative deconvolution approach. This approach was then applied to MRSA, VSE, and VRE bacterial extracts before and after exposure to vancomycin. This demonstrated a substantial (3-fold) increase in GlcN-6P in vancomycin-treated MRSA samples but not in vancomycin-treated VSE or VRE samples. These observations appear to localize previously observed differences between MRSA and VRE/VSE peptidoglycan biosynthesis regulation to GlmS, which synthesizes GlcN-6P and is the product of a regulatory ribozyme sensitive to the levels of GlcN-6P.
Collapse
Affiliation(s)
| | | | - Shivani Gargvanshi
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, 64108, USA
| | - William G. Gutheil
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, 64108, USA
| |
Collapse
|
39
|
Ragab A, Ibrahim SA, Aboul-Magd DS, Baren MH. One-pot synthesis of pyrazolo[4,3- d]thiazole derivatives containing α-aminophosphonate as potential Mur A inhibitors against MDR pathogens with radiosterilization and molecular modeling simulation. RSC Adv 2023; 13:34756-34771. [PMID: 38035237 PMCID: PMC10685179 DOI: 10.1039/d3ra07040a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/14/2023] [Indexed: 12/02/2023] Open
Abstract
The present study involves the synthesis of a new series of α-aminophosphonate derivatives in good yields with a simple workup via the Kabachnik-Fields reaction using lithium perchlorate (LiClO4) as a catalyst to facilitate the reaction. All the newly synthesized compounds were confirmed using various physical, spectroscopic, and analytical data, and the obtained results correlated with the proposed molecular structure. The in vitro antimicrobial activities of each compound were evaluated against different clinical isolates. The results indicated that among these derivatives, two compounds (5a and 5b) were the most active and displayed potent activity with MICs in the range from 0.06 to 0.25 μg mL-1 compared with fosfomycin and fluconazole as standard antibiotics. Moreover, the synthesized phosphonates displayed a broad spectrum of bactericidal and fungicidal activities depending on MICs, MBCs/MFCs, and the time-kill kinetics. In addition, the checkerboard assay showed synergistic and partial synergistic activities between the active compounds combined with fosfomycin and fluconazole. Furthermore, the SEM images showed distinct ruptures of the OM integrity of the FOS-R E. coli at their MICs, which was further indicated by the increased EtBr accumulation within the bacterial cells. Moreover, active derivatives revealed MurA inhibitory activity with IC50 values of 3.8 ± 0.39 and 4.5 ± 0.23 μM compared with fosfomycin (IC50 = 12.7 ± 0.27 μM). To our surprise, exposing 5a and 5b compounds to different gamma radiation doses revealed that 7.0 kGy eradicated the microbial load completely. Finally, the results of quantum chemical study supported the binding mode obtained from the docking study performed inside the active site of MurA (PDB: 1UAE), suggesting that these phosphonates may be promising safe candidates for MDR infection therapy clinical trials with no toxic effects on the normal human cells.
Collapse
Affiliation(s)
- Ahmed Ragab
- Chemistry Department, Faculty of Science (Boys), Al-Azhar University Nasr City Cairo 11884 Egypt
| | - Seham A Ibrahim
- Chemistry Department, Faculty of Science, Tanta University Tanta 31527 Egypt
| | - Dina S Aboul-Magd
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority Egypt
| | - Mohamed H Baren
- Chemistry Department, Faculty of Science, Tanta University Tanta 31527 Egypt
| |
Collapse
|
40
|
Braun C, Wingen LM, Menche D. Strategies and tactics for the synthesis of lipid I and II and shortened analogues: functional building blocks of bacterial cell wall biosynthesis. Nat Prod Rep 2023; 40:1718-1734. [PMID: 37492928 DOI: 10.1039/d3np00018d] [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: 07/27/2023]
Abstract
Covering: the literature up to 2022This study discusses various synthetic strategies for the synthesis of lipid II, the pivotal bacterial cell wall precursor. In detail, it examines different solution phase approaches, reviews various solid phase sequences, and evaluates enzymatic ventures. The underlying rationale, scope, limitations, and perspectives of these strategies are discussed. The focus is on the tactics and strategies towards the authentic peptidoglycan compound, as well as analogues thereof with shortened side chains, which are increasingly recognized as more beneficial surrogates with more favorable physicochemical properties.
Collapse
Affiliation(s)
- Christina Braun
- Kekulé Institute of Organic Chemistry and Biochemistry, University of Bonn, D-53121 Bonn, Germany.
| | - Lukas Martin Wingen
- Kekulé Institute of Organic Chemistry and Biochemistry, University of Bonn, D-53121 Bonn, Germany.
| | - Dirk Menche
- Kekulé Institute of Organic Chemistry and Biochemistry, University of Bonn, D-53121 Bonn, Germany.
| |
Collapse
|
41
|
Liu Y, Zhang Y, Kang C, Tian D, Lu H, Xu B, Xia Y, Kashiwagi A, Westermann M, Hoischen C, Xu J, Yomo T. Comparative genomics hints at dispensability of multiple essential genes in two Escherichia coli L-form strains. Biosci Rep 2023; 43:BSR20231227. [PMID: 37819245 PMCID: PMC10600066 DOI: 10.1042/bsr20231227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 10/13/2023] Open
Abstract
Despite the critical role of bacterial cell walls in maintaining cell shapes, certain environmental stressors can induce the transition of many bacterial species into a wall-deficient state called L-form. Long-term induced Escherichia coli L-forms lose their rod shape and usually hold significant mutations that affect cell division and growth. Besides this, the genetic background of L-form bacteria is still poorly understood. In the present study, the genomes of two stable L-form strains of E. coli (NC-7 and LWF+) were sequenced and their gene mutation status was determined and compared with their parental strains. Comparative genomic analysis between two L-forms reveals both unique adaptions and common mutated genes, many of which belong to essential gene categories not involved in cell wall biosynthesis, indicating that L-form genetic adaptation impacts crucial metabolic pathways. Missense variants from L-forms and Lenski's long-term evolution experiment (LTEE) were analyzed in parallel using an optimized DeepSequence pipeline to investigate predicted mutation effects (α) on protein functions. We report that the two L-form strains analyzed display a frequency of 6-10% (0% for LTEE) in mutated essential genes where the missense variants have substantial impact on protein functions (α<0.5). This indicates the emergence of different survival strategies in L-forms through changes in essential genes during adaptions to cell wall deficiency. Collectively, our results shed light on the detailed genetic background of two E. coli L-forms and pave the way for further investigations of the gene functions in L-form bacterial models.
Collapse
Affiliation(s)
- Yunfei Liu
- Laboratory of Biology and Information Science, School of Life Sciences, East China Normal University, Shanghai 200062, PR China
| | - Yueyue Zhang
- Laboratory of Biology and Information Science, School of Life Sciences, East China Normal University, Shanghai 200062, PR China
| | - Chen Kang
- School of Software Engineering, East China Normal University, Shanghai 200062, PR China
| | - Di Tian
- Laboratory of Biology and Information Science, School of Life Sciences, East China Normal University, Shanghai 200062, PR China
| | - Hui Lu
- Laboratory of Biology and Information Science, School of Life Sciences, East China Normal University, Shanghai 200062, PR China
| | - Boying Xu
- Laboratory of Biology and Information Science, School of Life Sciences, East China Normal University, Shanghai 200062, PR China
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Yang Xia
- Laboratory of Biology and Information Science, School of Life Sciences, East China Normal University, Shanghai 200062, PR China
| | - Akiko Kashiwagi
- Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki 036-8561, Japan
| | - Martin Westermann
- Center for Electron Microscopy, Medical Faculty, Friedrich–Schiller–University Jena, Ziegelmühlenweg 1, D-07743 Jena, Germany
| | - Christian Hoischen
- CF Imaging, Leibniz Institute On Aging, Fritz–Lipmann–Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Jian Xu
- Laboratory of Biology and Information Science, School of Life Sciences, East China Normal University, Shanghai 200062, PR China
| | - Tetsuya Yomo
- Laboratory of Biology and Information Science, School of Life Sciences, East China Normal University, Shanghai 200062, PR China
| |
Collapse
|
42
|
Drouin P, da Silva ÉB, Tremblay J, Chevaux E, Apper E, Castex M. Inoculation with Lentilactobacillus buchneri alone or in combination with Lentilactobacillus hilgardii modifies gene expression, fermentation profile, and starch digestibility in high-moisture corn. Front Microbiol 2023; 14:1253588. [PMID: 37901805 PMCID: PMC10602787 DOI: 10.3389/fmicb.2023.1253588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023] Open
Abstract
Inoculants combining Lentilactobacillus buchneri and Lentilactobacillus hilgardii have been shown to improve the aerobic stability of high-moisture corn (HMC) and whole-plant corn silage, but the mode of action of this co-inoculation remains to be elucidated. This study used metatranscriptomics to evaluate the effects of inoculation with L. buchneri alone or combined with L. hilgardii on the bacterial community, gene expression, fermentation profile, and starch digestibility in HMC. High-moisture corn not inoculated (Control) or inoculated with L. buchneri NCIMB 40788 (LB) or L. buchneri NCIMB 40788 combined with L. hilgardii CNCM-I-4785 (Combo) was ensiled in mini silo bags for 30, 60, 120, and 180 days. The fermentation profile was evaluated at all time points. Metatranscriptomics was performed on samples collected on day 120. Combo had a greater alpha diversity richness index of contigs than LB and Control, and inoculation with Combo and LB modified the beta-diversity of contigs compared to Control. Out of 69 genes of interest, 20 were differentially expressed in LB compared to Control and 25 in Combo compared to Control. Of those differently expressed genes, 16 (10 of which were associated with carbohydrate metabolism and six with amino acid metabolism) were differently expressed in both LB and Combo compared to Control, and all those genes were upregulated in the inoculated silages. When we compared Combo and LB, we found seven genes expressed differently, four associated with carbohydrate metabolism and downregulated in Combo, and three associated with amino acid metabolism and upregulated in Combo. At day 120, the inoculated silages had more culturable lactic acid bacteria, higher Lactobacillus relative abundance, and lower Leuconostoc relative abundance than Control. The concentration of acetic acid remained low throughout ensiling in Control, but in LB and Combo, it increased up to day 60 and remained stable from day 60 to 180. The 1,2-propanediol was only detected in LB and Combo. Inoculation did not affect the concentration of starch, but starch digestibility was greater in Combo than in Control. Inoculation of HMC with Combo modified the gene expression and fermentation profile compared to Control and LB, improving starch digestibility compared to uninoculated HMC.
Collapse
Affiliation(s)
- Pascal Drouin
- Independent Researcher, Saint-Jean-sur-Richelieu, QC, Canada
| | | | - Julien Tremblay
- Energy, Mining, and Environment, National Research Council of Canada, Montréal, QC, Canada
| | | | | | | |
Collapse
|
43
|
Ambade SS, Gupta VK, Bhole RP, Khedekar PB, Chikhale RV. A Review on Five and Six-Membered Heterocyclic Compounds Targeting the Penicillin-Binding Protein 2 (PBP2A) of Methicillin-Resistant Staphylococcus aureus (MRSA). Molecules 2023; 28:7008. [PMID: 37894491 PMCID: PMC10609489 DOI: 10.3390/molecules28207008] [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: 09/08/2023] [Revised: 10/05/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Staphylococcus aureus is a common human pathogen. Methicillin-resistant Staphylococcus aureus (MRSA) infections pose significant and challenging therapeutic difficulties. MRSA often acquires the non-native gene PBP2a, which results in reduced susceptibility to β-lactam antibiotics, thus conferring resistance. PBP2a has a lower affinity for methicillin, allowing bacteria to maintain peptidoglycan biosynthesis, a core component of the bacterial cell wall. Consequently, even in the presence of methicillin or other antibiotics, bacteria can develop resistance. Due to genes responsible for resistance, S. aureus becomes MRSA. The fundamental premise of this resistance mechanism is well-understood. Given the therapeutic concerns posed by resistant microorganisms, there is a legitimate demand for novel antibiotics. This review primarily focuses on PBP2a scaffolds and the various screening approaches used to identify PBP2a inhibitors. The following classes of compounds and their biological activities are discussed: Penicillin, Cephalosporins, Pyrazole-Benzimidazole-based derivatives, Oxadiazole-containing derivatives, non-β-lactam allosteric inhibitors, 4-(3H)-Quinazolinones, Pyrrolylated chalcone, Bis-2-Oxoazetidinyl macrocycles (β-lactam antibiotics with 1,3-Bridges), Macrocycle-embedded β-lactams as novel inhibitors, Pyridine-Coupled Pyrimidinones, novel Naphthalimide corbelled aminothiazoximes, non-covalent inhibitors, Investigational-β-lactam antibiotics, Carbapenem, novel Benzoxazole derivatives, Pyrazolylpyridine analogues, and other miscellaneous classes of scaffolds for PBP2a. Additionally, we discuss the penicillin-binding protein, a crucial target in the MRSA cell wall. Various aspects of PBP2a, bacterial cell walls, peptidoglycans, different crystal structures of PBP2a, synthetic routes for PBP2a inhibitors, and future perspectives on MRSA inhibitors are also explored.
Collapse
Affiliation(s)
- Shraddha S. Ambade
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440033, MH, India (P.B.K.)
| | - Vivek Kumar Gupta
- Department of Biochemistry, National JALMA Institute for Leprosy & Other Mycobacterial Diseases (ICMR), Agra 282004, UP, India
| | - Ritesh P. Bhole
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune 411018, MH, India
- Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune 411018, MH, India
| | - Pramod B. Khedekar
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440033, MH, India (P.B.K.)
| | | |
Collapse
|
44
|
Moiketsi BN, Makale KPP, Rantong G, Rahube TO, Makhzoum A. Potential of Selected African Medicinal Plants as Alternative Therapeutics against Multi-Drug-Resistant Bacteria. Biomedicines 2023; 11:2605. [PMID: 37892979 PMCID: PMC10604549 DOI: 10.3390/biomedicines11102605] [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: 05/30/2023] [Revised: 09/04/2023] [Accepted: 09/18/2023] [Indexed: 10/29/2023] Open
Abstract
Antimicrobial resistance is considered a "One-Health" problem, impacting humans, animals, and the environment. The problem of the rapid development and spread of bacteria resistant to multiple antibiotics is a rising global health threat affecting both rich and poor nations. Low- and middle-income countries are at highest risk, in part due to the lack of innovative research on the surveillance and discovery of novel therapeutic options. Fast and effective drug discovery is crucial towards combatting antimicrobial resistance and reducing the burden of infectious diseases. African medicinal plants have been used for millennia in folk medicine to cure many diseases and ailments. Over 10% of the Southern African vegetation is applied in traditional medicine, with over 15 species being partially or fully commercialized. These include the genera Euclea, Ficus, Aloe, Lippia. And Artemisia, amongst many others. Bioactive compounds from indigenous medicinal plants, alone or in combination with existing antimicrobials, offer promising solutions towards overcoming multi-drug resistance. Secondary metabolites have different mechanisms and modes of action against bacteria, such as the inhibition and disruption of cell wall synthesis; inhibition of DNA replication and ATP synthesis; inhibition of quorum sensing; inhibition of AHL or oligopeptide signal generation, broadcasting, and reception; inhibition of the formation of biofilm; disruption of pathogenicity activities; and generation of reactive oxygen species. The aim of this review is to highlight some promising traditional medicinal plants found in Africa and provide insights into their secondary metabolites as alternative options in antibiotic therapy against multi-drug-resistant bacteria. Additionally, synergism between plant secondary metabolites and antibiotics has been discussed.
Collapse
Affiliation(s)
| | | | | | - Teddie O. Rahube
- Department of Biological Sciences and Biotechnology, Faculty of Science, Botswana International University of Science and Technology (BIUST), Private Bag 16, Palapye, Botswana; (B.N.M.); (K.P.P.M.); (G.R.)
| | - Abdullah Makhzoum
- Department of Biological Sciences and Biotechnology, Faculty of Science, Botswana International University of Science and Technology (BIUST), Private Bag 16, Palapye, Botswana; (B.N.M.); (K.P.P.M.); (G.R.)
| |
Collapse
|
45
|
Swain B, Campodonico VA, Curtiss R. Recombinant Attenuated Edwardsiella piscicida Vaccine Displaying Regulated Lysis to Confer Biological Containment and Protect Catfish against Edwardsiellosis. Vaccines (Basel) 2023; 11:1470. [PMID: 37766146 PMCID: PMC10534663 DOI: 10.3390/vaccines11091470] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
We implemented a unique strategy to construct a recombinant attenuated Edwardsiella vaccine (RAEV) with a biological containment phenotype that causes regulated bacterial cell wall lysis. This process ensures that the vaccine strain is not able to persist in the environment. The murA gene is responsible for the catalysis of one of the first steps in the biosynthesis of muramic acid, which is a crucial component of the bacterial cell wall. The regulated lysis phenotype was achieved by inserting the tightly regulated araC ParaBAD cassette in place of the chromosomal murA promoter. Strains with this mutation require growth media supplemented with arabinose in order to survive. Without arabinose, they are unable to synthesize the peptidoglycan cell wall. Following the colonization of fish lymphoid tissues, the murA protein is no longer synthesized due to the lack of arabinose. Lysis is subsequently achieved in vivo, thus preventing the generation of disease symptoms and the spread of the strain into the environment. Vaccine strain χ16016 with the genotype ΔPmurA180::TT araC ParaBADmurA is attenuated and shows a higher LD50 value than that of the wild-type strain. Studies have demonstrated that χ16016 induced TLR4, TLR5, TLR8, TLR9, NOD1 and NOD2-mediated NF-κB pathways and upregulated the gene expression of various cytokines, such as il-8, il-1β, tnf-a, il-6 and ifn-γ in catfish. We observed significant upregulation of the expression profiles of cd4, cd8 and mhc-II genes in different organs of vaccinated catfish. Vaccine strain χ16016 induced systemic and mucosal IgM titers and conferred significant protection to catfish against E. piscicida wild-type challenge. Our lysis RAEV is the first live attenuated vaccine candidate designed to be used in the aquaculture industry that displays this biological containment property.
Collapse
Affiliation(s)
- Banikalyan Swain
- Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| | | | | |
Collapse
|
46
|
Qun T, Zhou T, Hao J, Wang C, Zhang K, Xu J, Wang X, Zhou W. Antibacterial activities of anthraquinones: structure-activity relationships and action mechanisms. RSC Med Chem 2023; 14:1446-1471. [PMID: 37593578 PMCID: PMC10429894 DOI: 10.1039/d3md00116d] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/24/2023] [Indexed: 08/19/2023] Open
Abstract
With the increasing prevalence of untreatable infections caused by antibiotic-resistant bacteria, the discovery of new drugs from natural products has become a hot research topic. The antibacterial activity of anthraquinones widely distributed in traditional Chinese medicine has attracted much attention. Herein, the structure and activity relationships (SARs) of anthraquinones as bacteriostatic agents are reviewed and elucidated. The substituents of anthraquinone and its derivatives are closely related to their antibacterial activities. The stronger the polarity of anthraquinone substituents is, the more potent the antibacterial effects appear. The presence of hydroxyl groups is not necessary for the antibacterial activity of hydroxyanthraquinone derivatives. Substitution of di-isopentenyl groups can improve the antibacterial activity of anthraquinone derivatives. The rigid plane structure of anthraquinone lowers its water solubility and results in the reduced activity. Meanwhile, the antibacterial mechanisms of anthraquinone and its analogs are explored, mainly including biofilm formation inhibition, destruction of the cell wall, endotoxin inhibition, inhibition of nucleic acid and protein synthesis, and blockage of energy metabolism and other substances.
Collapse
Affiliation(s)
- Tang Qun
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
| | - Tiantian Zhou
- School of Chinese Materia Medica, Guangdong Pharmaceutical University 440113 Guangzhou China
| | - Jiongkai Hao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
| | - Chunmei Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences Shanghai 200241 China
| | - Keyu Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences Shanghai 200241 China
| | - Jing Xu
- Huanghua Agricultural and Rural Development Bureau Bohai New Area 061100 Hebei China
| | - Xiaoyang Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences Shanghai 200241 China
| | - Wen Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences Shanghai 200241 China
| |
Collapse
|
47
|
Soni V, Rosenn EH, Venkataraman R. Insights into the central role of N-acetyl-glucosamine-1-phosphate uridyltransferase (GlmU) in peptidoglycan metabolism and its potential as a therapeutic target. Biochem J 2023; 480:1147-1164. [PMID: 37498748 DOI: 10.1042/bcj20230173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/07/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023]
Abstract
Several decades after the discovery of the first antibiotic (penicillin) microbes have evolved novel mechanisms of resistance; endangering not only our abilities to combat future bacterial pandemics but many other clinical challenges such as acquired infections during surgeries. Antimicrobial resistance (AMR) is attributed to the mismanagement and overuse of these medications and is complicated by a slower rate of the discovery of novel drugs and targets. Bacterial peptidoglycan (PG), a three-dimensional mesh of glycan units, is the foundation of the cell wall that protects bacteria against environmental insults. A significant percentage of drugs target PG, however, these have been rendered ineffective due to growing drug resistance. Identifying novel druggable targets is, therefore, imperative. Uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) is one of the key building blocks in PG production, biosynthesized by the bifunctional enzyme N-acetyl-glucosamine-1-phosphate uridyltransferase (GlmU). UDP-GlcNAc metabolism has been studied in many organisms, but it holds some distinctive features in bacteria, especially regarding the bacterial GlmU enzyme. In this review, we provide an overview of different steps in PG biogenesis, discuss the biochemistry of GlmU, and summarize the characteristic structural elements of bacterial GlmU vital to its catalytic function. Finally, we will discuss various studies on the development of GlmU inhibitors and their significance in aiding future drug discoveries.
Collapse
Affiliation(s)
- Vijay Soni
- Division of Infectious Diseases, Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065, U.S.A
| | - Eric H Rosenn
- Tel Aviv University School of Medicine, Tel Aviv 6997801, Israel
| | - Ramya Venkataraman
- Laboratory of Innate Immunity, National Institute of Immunology, New Delhi 110067, India
| |
Collapse
|
48
|
Colquhoun JM, Farokhyfar M, Anderson AC, Bethel CR, Bonomo RA, Clarke AJ, Rather PN. Collateral Changes in Cell Physiology Associated with ADC-7 β-Lactamase Expression in Acinetobacter baumannii. Microbiol Spectr 2023; 11:e0464622. [PMID: 37074187 PMCID: PMC10269689 DOI: 10.1128/spectrum.04646-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/15/2023] [Indexed: 04/20/2023] Open
Abstract
The ADC (AmpC) β-lactamase is universally present in the Acinetobacter baumannii chromosome, suggesting it may have a yet-to-be-identified cellular function. Using peptidoglycan composition analysis, we show that overexpressing the ADC-7 β-lactamase in A. baumannii drives changes consistent with altered l,d-transpeptidase activity. Based on this, we tested whether cells overexpressing ADC-7 would exhibit new vulnerabilities. As proof of principle, a screen of transposon insertions revealed that an insertion in the distal 3' end of canB, encoding carbonic anhydrase, resulted in a significant loss of viability when the adc-7 gene was overexpressed. A canB deletion mutant exhibited a more pronounced loss of viability than the transposon insertion, and this became amplified when cells overexpressed ADC-7. Interestingly, overexpression of the OXA-23 or TEM-1 β-lactamases also led to a pronounced loss of viability in cells with reduced carbonic anhydrase activity. In addition, we demonstrate that reduced CanB activity led to increased sensitivity to peptidoglycan synthesis inhibitors and to the carbonic anhydrase inhibitor ethoxzolamide. Furthermore, this strain exhibited a synergistic interaction with the peptidoglycan inhibitor fosfomycin and ethoxzolamide. Our results highlight the impact of ADC-7 overexpression on cell physiology and reveal that the essential carbonic anhydrase CanB may represent a novel target for antimicrobial agents that would exhibit increased potency against β-lactamase-overexpressing A. baumannii. IMPORTANCE Acinetobacter baumannii has become resistant to all classes of antibiotics, with β-lactam resistance responsible for the majority of treatment failures. New classes of antimicrobials are needed to treat this high-priority pathogen. This study had uncovered a new genetic vulnerability in β-lactamase-expressing A. baumannii, where reduced carbonic anhydrase activity becomes lethal. Inhibitors of carbonic anhydrase could represent a new method for treating A. baumannii infections.
Collapse
Affiliation(s)
- Jennifer M. Colquhoun
- Department of Microbiology and Immunology, Emory University, Atlanta, Georgia, USA
- Emory Antibiotic Resistance Center, Emory University, Atlanta, Georgia, USA
| | | | - Alexander C. Anderson
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Christopher R. Bethel
- Research Service, Louis Stokes Cleveland Department of Veterans Affairs, Cleveland, Ohio, USA
| | - Robert A. Bonomo
- Research Service, Louis Stokes Cleveland Department of Veterans Affairs, Cleveland, Ohio, USA
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, Ohio, USA
- CWRU-Cleveland VAMC Center for Antimicrobial Resistance and Epidemiology (Case VA CARES), Case Western Reserve University, Cleveland, Ohio, USA
| | - Anthony J. Clarke
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
- Department of Chemistry & Biochemistry, Wilfrid Laurier University, Waterloo, Ontario, Canada
| | - Philip N. Rather
- Department of Microbiology and Immunology, Emory University, Atlanta, Georgia, USA
- Emory Antibiotic Resistance Center, Emory University, Atlanta, Georgia, USA
- Research Service, Atlanta VA Medical Center, Decatur, Georgia, USA
| |
Collapse
|
49
|
Boamah D, Gilmore M, Bourget S, Ghosh A, Hossain M, Vogel J, Cava F, O’Connor T. Peptidoglycan deacetylation controls type IV secretion and the intracellular survival of the bacterial pathogen Legionella pneumophila. Proc Natl Acad Sci U S A 2023; 120:e2119658120. [PMID: 37252954 PMCID: PMC10266036 DOI: 10.1073/pnas.2119658120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 04/18/2023] [Indexed: 06/01/2023] Open
Abstract
Peptidoglycan is a critical component of the bacteria cell envelope. Remodeling of the peptidoglycan is required for numerous essential cellular processes and has been linked to bacterial pathogenesis. Peptidoglycan deacetylases that remove the acetyl group of the N-acetylglucosamine (NAG) subunit protect bacterial pathogens from immune recognition and digestive enzymes secreted at the site of infection. However, the full extent of this modification on bacterial physiology and pathogenesis is not known. Here, we identify a polysaccharide deacetylase of the intracellular bacterial pathogen Legionella pneumophila and define a two-tiered role for this enzyme in Legionella pathogenesis. First, NAG deacetylation is important for the proper localization and function of the Type IVb secretion system, linking peptidoglycan editing to the modulation of host cellular processes through the action of secreted virulence factors. As a consequence, the Legionella vacuole mis-traffics along the endocytic pathway to the lysosome, preventing the formation of a replication permissive compartment. Second, within the lysosome, the inability to deacetylate the peptidoglycan renders the bacteria more sensitive to lysozyme-mediated degradation, resulting in increased bacterial death. Thus, the ability to deacetylate NAG is important for bacteria to persist within host cells and in turn, Legionella virulence. Collectively, these results expand the function of peptidoglycan deacetylases in bacteria, linking peptidoglycan editing, Type IV secretion, and the intracellular fate of a bacterial pathogen.
Collapse
Affiliation(s)
- David Boamah
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Michael C. Gilmore
- Department of Molecular Biology, Laboratory for Molecular Infection Medicine Sweden, Umeå Centre for Microbial Research, Umeå University, Umeå90187, Sweden
| | - Sarah Bourget
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Anushka Ghosh
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Mohammad J. Hossain
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Joseph P. Vogel
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO63110
| | - Felipe Cava
- Department of Molecular Biology, Laboratory for Molecular Infection Medicine Sweden, Umeå Centre for Microbial Research, Umeå University, Umeå90187, Sweden
| | - Tamara J. O’Connor
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD21205
| |
Collapse
|
50
|
Xu N, Du LH, Chen YC, Zhang JH, Zhu QF, Chen R, Peng GP, Wang QM, Yu HZ, Rao LQ. Lonicera japonica Thunb. as a promising antibacterial agent for Bacillus cereus ATCC14579 based on network pharmacology, metabolomics, and in vitro experiments. RSC Adv 2023; 13:15379-15390. [PMID: 37223411 PMCID: PMC10201548 DOI: 10.1039/d3ra00802a] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 05/15/2023] [Indexed: 05/25/2023] Open
Abstract
Lonicera japonica Thunb. has attracted much attention for its treatment of bacterial and viral infectious diseases, while its active ingredients and potential mechanisms of action have not been fully elucidated. Here, we combined metabolomics, and network pharmacology to explore the molecular mechanism of Bacillus cereus ATCC14579 inhibition by Lonicera japonica Thunb. In vitro inhibition experiments showed that the Lonicera japonica Thunb.'s water extracts, ethanolic extract, luteolin, quercetin, and kaempferol strongly inhibited Bacillus cereus ATCC14579. In contrast, chlorogenic acid and macranthoidin B had no inhibitory effect on Bacillus cereus ATCC14579. Meanwhile, the minimum inhibitory concentrations of luteolin, quercetin, and kaempferol against Bacillus cereus ATCC14579 were 15.625 μg mL-1, 31.25 μg mL-1, and 15.625 μg mL-1. Based on the previous experimental basis, the metabolomic analysis showed the presence of 16 active ingredients in Lonicera japonica Thunb.'s water extracts and ethanol extracts, with differences in the luteolin, quercetin, and kaempferol contents between the water extracts and ethanol extracts. Network pharmacology studies indicated that fabZ, tig, glmU, secA, deoD, nagB, pgi, rpmB, recA, and upp were potential key targets. Active ingredients of Lonicera japonica Thunb. may exert their inhibitory effects by inhibiting ribosome assembly, the peptidoglycan biosynthesis process, and the phospholipid biosynthesis process of Bacillus cereus ATCC14579. An alkaline phosphatase activity assay, peptidoglycan concentration assay, and protein concentration assay showed that luteolin, quercetin, and kaempferol disrupted the Bacillus cereus ATCC14579 cell wall and cell membrane integrity. Transmission electron microscopy results showed significant changes in the morphology and ultrastructure of the cell wall and cell membrane of Bacillus cereus ATCC14579, further confirming the disruption of the cell wall and cell membrane integrity of Bacillus cereus ATCC14579 by luteolin, quercetin, and kaempferol. In conclusion, Lonicera japonica Thunb. can be used as a potential antibacterial agent for Bacillus cereus ATCC14579, which may exert its antibacterial activity by destroying the integrity of the cell wall and membrane.
Collapse
Affiliation(s)
- Nan Xu
- Hunan Engineering Laboratory for Good Agricultural Practice and Comprehensive Utilization of Famous-Region Medicinal Plants, Hunan Agricultural University Changsha China
| | - Li-Hua Du
- Hunan Engineering Laboratory for Good Agricultural Practice and Comprehensive Utilization of Famous-Region Medicinal Plants, Hunan Agricultural University Changsha China
| | - Yan-Chao Chen
- Hunan Engineering Laboratory for Good Agricultural Practice and Comprehensive Utilization of Famous-Region Medicinal Plants, Hunan Agricultural University Changsha China
| | - Jin-Hao Zhang
- Hunan Engineering Laboratory for Good Agricultural Practice and Comprehensive Utilization of Famous-Region Medicinal Plants, Hunan Agricultural University Changsha China
| | - Qian-Feng Zhu
- Hunan Engineering Laboratory for Good Agricultural Practice and Comprehensive Utilization of Famous-Region Medicinal Plants, Hunan Agricultural University Changsha China
| | - Rong Chen
- Hunan Engineering Laboratory for Good Agricultural Practice and Comprehensive Utilization of Famous-Region Medicinal Plants, Hunan Agricultural University Changsha China
| | - Guo-Ping Peng
- Hunan Engineering Laboratory for Good Agricultural Practice and Comprehensive Utilization of Famous-Region Medicinal Plants, Hunan Agricultural University Changsha China
| | - Qi-Ming Wang
- Hunan Engineering Laboratory for Good Agricultural Practice and Comprehensive Utilization of Famous-Region Medicinal Plants, Hunan Agricultural University Changsha China
| | - Hua-Zhong Yu
- Key Laboratory of Hunan Forest Products and Chemical Industry Engineering, Jishou University Jishou China
| | - Li-Qun Rao
- Hunan Engineering Laboratory for Good Agricultural Practice and Comprehensive Utilization of Famous-Region Medicinal Plants, Hunan Agricultural University Changsha China
| |
Collapse
|