1
|
Brown HJ, Shinde VD, Bosi L, Duggin IG. Evolution of the cytoskeleton: Emerging clues from the diversification and specialisation of archaeal cytoskeletal proteins. Curr Opin Cell Biol 2025; 95:102557. [PMID: 40513207 DOI: 10.1016/j.ceb.2025.102557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 05/02/2025] [Accepted: 05/19/2025] [Indexed: 06/16/2025]
Abstract
Recent research in archaeal cell biology has revealed a remarkable diversity of cytoskeletal proteins related to those found in bacteria and eukaryotes, such as the tubulin, actin, and ESCRT protein superfamilies, and archaea-specific proteins that self-assemble and have been implicated in cytoskeletal roles. Here, we outline an emerging view that the archaeal cytoskeleton has several conceptual ties to the sophisticated eukaryotic cytoskeleton. We highlight that duplication and specialisation of protein function is common among archaeal cytoskeletal systems, and that some paralogues show coordinated, opposing functions in the regulation of cell morphogenesis and structural homeostasis. Furthermore, the presence of homologues of eukaryotic cytoskeletal regulators in Asgard archaea, the closest known relatives of eukaryotes, underscores further linkages between eukaryotic and increasingly sophisticated archaeal cytoskeletal systems.
Collapse
Affiliation(s)
- Hannah J Brown
- The Australian Institute for Microbiology and Infection, University of Technology Sydney, NSW, 2007 Australia
| | - Vinaya D Shinde
- The Australian Institute for Microbiology and Infection, University of Technology Sydney, NSW, 2007 Australia
| | - Leonardo Bosi
- The Australian Institute for Microbiology and Infection, University of Technology Sydney, NSW, 2007 Australia
| | - Iain G Duggin
- The Australian Institute for Microbiology and Infection, University of Technology Sydney, NSW, 2007 Australia.
| |
Collapse
|
2
|
Liao Y, Shinde VD, Hu D, Xu Z, Söderström B, Michie KA, Duggin IG. Cell division protein CdpA organises and anchors the midcell ring in haloarchaea. Nat Commun 2025; 16:5076. [PMID: 40450033 DOI: 10.1038/s41467-025-60079-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 05/12/2025] [Indexed: 06/03/2025] Open
Abstract
Many archaea appear to divide through the coordinated activities of two FtsZ homologues (FtsZ1 and FtsZ2) and another bacterial cell division homologue (SepF), which are part of the midcell division ring. Here, we identify an additional protein (HVO_0739, renamed CdpA) that is involved in cell division in Haloferax volcanii, with homologues in other Haloarchaea. CdpA localises at the midcell division ring, and this requires the presence of the ring-assembly protein FtsZ1. The division constriction protein FtsZ2 also influences the proper midcell assembly and structure of CdpA. In the absence of CdpA, cells frequently fail to divide properly, and FtsZ1 formed poorly condensed pseudo-helical structures spanning across a broad region of the cell, whereas FtsZ2 showed mispositioned foci, nano-rings, and filaments. The rate of directional movement of FtsZ1 and FtsZ2 structures around the division ring appears minimally affected by loss of CdpA, which resulted in continual repositioning of the aberrant FtsZ structures in the cells. In contrast to the FtsZ proteins, CdpA formed relatively immobile foci around the ring. Protein domain function studies, pull-down assays, and multimer structure predictions suggest that CdpA is part of a membrane complex that tethers FtsZ2 and other division proteins to the midcell membrane. Our discovery of an archaeal FtsZ organisation and midcell anchor protein offers new insights into cell division mechanisms that are similar across the tree of life.
Collapse
Affiliation(s)
- Yan Liao
- Australian Institute for Microbiology and Infection, University of Technology Sydney, Sydney, NSW, Australia.
| | - Vinaya D Shinde
- Australian Institute for Microbiology and Infection, University of Technology Sydney, Sydney, NSW, Australia
| | - Dalong Hu
- Australian Institute for Microbiology and Infection, University of Technology Sydney, Sydney, NSW, Australia
- Saw SweeHock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Zhuang Xu
- School of Mathematics and Statistics, The University of New South Wales, Sydney, NSW, Australia
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Bill Söderström
- Australian Institute for Microbiology and Infection, University of Technology Sydney, Sydney, NSW, Australia
| | - Katharine A Michie
- Structural Biology Facility, Mark Wainwright Analytical Centre, The University of New South Wales, Sydney, NSW, Australia
| | - Iain G Duggin
- Australian Institute for Microbiology and Infection, University of Technology Sydney, Sydney, NSW, Australia.
| |
Collapse
|
3
|
Sun N, Deng X, Kong H, Zhi Z, Jiang G, Xiong J, Chen S, Li S, Yuan W, Wong WL. Magnolol as an Antibacterial Agent Against Drug-resistant Bacteria Targeting Filamentous Temperature-sensitive Mutant Z. Chem Biodivers 2025; 22:e202402800. [PMID: 39714990 DOI: 10.1002/cbdv.202402800] [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: 10/29/2024] [Revised: 11/25/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
The emergence of multiple drug-resistant bacteria poses critical health threats worldwide. It is urgently needed to develop potent and safe antibacterial agents with novel bactericidal mechanisms to treat these infections. In this study, magnolol was identified as a potential bacterial cell division inhibitor by a cell-based screening approach. This compound showed good antibacterial activity against a number of Gram-positive pathogens (minimum inhibitory concentration 8-16 µg/mL) including methicillin-resistant Staphylococcus aureus and vancomycin-resistant Enterococcus. Further results obtained from biochemical experiments demonstrated that magnolol could markedly disrupt GTPase activity and filamentous temperature-sensitive mutant Z (FtsZ) polymerization, consistent with the impediment to cell division in the bacteria tested. The in vivo antibacterial activity of magnolol was evaluated with a Galleria mellonella larvae model. The results showed that magnolol significantly increased the survival rate of larvae infected with methicillin-resistant S. aureus. The interaction pattern of magnolol with FtsZ was investigated through molecular docking. The finding may offer meaningful insights into the mechanism of action of the compound. The results point to magnolol as a promising antimicrobial compound that inhibits cell division by affecting FtsZ polymerization and has the potential to be developed into an effective antimicrobial drug by further structure modification.
Collapse
Affiliation(s)
- Ning Sun
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, P. R. China
| | - Xin Deng
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, P. R. China
| | - Hanqin Kong
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, P. R. China
| | - Ziling Zhi
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, P. R. China
| | - Guli Jiang
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, P. R. China
| | - Jing Xiong
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, P. R. China
| | - Sisi Chen
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, P. R. China
| | - Song Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, P. R. China
| | - Wenchang Yuan
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, P. R. China
| | - Wing-Leung Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, P. R. China
| |
Collapse
|
4
|
Ramos-León F, Ramamurthi KS. How do spherical bacteria regulate cell division? Biochem Soc Trans 2025; 53:BST20240956. [PMID: 40259574 DOI: 10.1042/bst20240956] [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: 12/01/2024] [Revised: 03/07/2025] [Accepted: 03/28/2025] [Indexed: 04/23/2025]
Abstract
Many bacteria divide by binary fission, producing two identical daughter cells, which requires proper placement of the division machinery at mid-cell. Spherical bacteria (cocci) face unique challenges due to their lack of natural polarity. In this review, we compile current knowledge on how cocci regulate cell division, how they select the proper division plane, and ensure accurate Z-ring positioning at mid-cell. While Streptococcus pneumoniae and Staphylococcus aureus are the most well-studied models for cell division in cocci, we also cover other less-characterized cocci across different bacterial groups and discuss the conservation of known Z-ring positioning mechanisms in these understudied bacteria.
Collapse
Affiliation(s)
- Félix Ramos-León
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, U.S.A
| | - Kumaran S Ramamurthi
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, U.S.A
| |
Collapse
|
5
|
Dutta S, Poddar S, Chakraborty J, Srinivasan R, Gayathri P. Membrane Binding and Cholesterol Sensing Motif in Mycoplasma genitalium FtsZ: A Novel Mode of Membrane Recruitment for Bacterial FtsZ. Biochemistry 2025; 64:1864-1877. [PMID: 40184362 DOI: 10.1021/acs.biochem.4c00543] [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: 04/06/2025]
Abstract
Cell division in bacteria is initiated by constriction of the Z-ring comprising two essential proteins, FtsZ and FtsA. Though the essential function of the Z-ring in bacterial division has been established, the precise roles of FtsZ and FtsA in the constriction process remain elusive. Due to the minimal number of components, FtsZ/FtsA in cell wall-less bacteria is an ideal model system for obtaining mechanistic insights into Z-ring constriction in the absence of a cell wall synthesis machinery. In this study, we undertook a comparative analysis of FtsZ and FtsA protein sequences from 113 mycoplasma species and the corresponding sequences in cell-walled bacteria. We report a phylogenetically distinct group of 12 species that possess a putative membrane binding amphipathic helix at either the N- or C-terminal extensions of the globular FtsZ domain. Importantly, these FtsZs lack conservation of the conserved C-terminal peptide sequence. We experimentally prove that the proposed C-terminal amphipathic helix in Mycoplasma genitalium (M. genitalium) FtsZ exhibits membrane binding. Additionally, we identify a potential cholesterol recognition motif within the C-terminal amphipathic helix region of M. genitalium FtsZ. Our study catalogues the functional variations of membrane attachment by the FtsZ and FtsA system in cell wall-less mycoplasmas and provides a new perspective to dissect the role of FtsZ and FtsA in cell division.
Collapse
Affiliation(s)
- Soumyajit Dutta
- Biology Division, Indian Institute of Science Education and Research, Pune 411008, India
| | - Sakshi Poddar
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India
- Homi Bhabha National Institute (HBNI), Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Joyeeta Chakraborty
- Biology Division, Indian Institute of Science Education and Research, Pune 411008, India
| | - Ramanujam Srinivasan
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India
- Homi Bhabha National Institute (HBNI), Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Pananghat Gayathri
- Biology Division, Indian Institute of Science Education and Research, Pune 411008, India
| |
Collapse
|
6
|
Ding L, Wang X, Wang J, Wang H, Yu L, Liu J, Yu J, Xue T, Yang X, Xue L. Fluorogenic Probes for Real-Time Tracking of Bacterial Cell Wall Dynamics with Nanoscopy. ACS NANO 2025; 19:14389-14403. [PMID: 40173278 DOI: 10.1021/acsnano.5c01930] [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: 04/04/2025]
Abstract
The bacterial cell wall, an essential structure for maintaining cell morphology and protecting against environmental hazards, is predominantly composed of peptidoglycan (PG). This intricate macromolecule undergoes dynamic synthesis and remodeling throughout the cell cycle. Despite its importance, monitoring PG dynamics in live cells, particularly with detailed spatial distribution, poses significant challenges. To this end, we present a series of rhodamine-based fluorogenic probes specifically optimized for real-time and super-resolution imaging of PG synthesis. By fine-tuning the self-aggregation of the probes through the incorporation of hydrophobic linkers, we achieved a substantial reduction in background fluorescence and significant fluorogenicity after labeling. These advancements have enabled us to attain wash-free labeling across a diverse array of bacterial species. Our approach facilitates the direct visualization of PG synthesis patterns, enabling the quantification of septal PG (sPG) synthesis rates in living bacterial cells. Furthermore, it allows for simultaneous imaging of cell division machinery in living cells via both two-dimensional (2D) and three-dimensional (3D) STED microscopy. This study provides a powerful toolkit for investigating the architecture and dynamics of the bacterial cell wall, paving new paths for research on PG-related cellular processes.
Collapse
Affiliation(s)
- Lihao Ding
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Xinci Wang
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Jiajia Wang
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Hui Wang
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Anhui Basic Discipline Research Center of Artificial Intelligence Biotechnology and Synthetic Biology, Hefei 230027, China
| | - Le Yu
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Jiang Liu
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Jiangliu Yu
- College of Life Science, Anhui Agricultural University, Hefei 230036, China
| | - Ting Xue
- College of Life Science, Anhui Agricultural University, Hefei 230036, China
| | - Xinxing Yang
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Anhui Basic Discipline Research Center of Artificial Intelligence Biotechnology and Synthetic Biology, Hefei 230027, China
| | - Lin Xue
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Anhui Basic Discipline Research Center of Artificial Intelligence Biotechnology and Synthetic Biology, Hefei 230027, China
| |
Collapse
|
7
|
Dingding H, Muhammad S, Manzoor I, Ghaffar SA, Alodaini HA, Moubayed NMS, Hatamleh AA, Songxiao X. Subtractive proteomics and reverse-vaccinology approaches for novel drug targets and designing a chimeric vaccine against Ruminococcus gnavus strain RJX1120. Front Immunol 2025; 16:1555741. [PMID: 40297578 PMCID: PMC12034673 DOI: 10.3389/fimmu.2025.1555741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Mediterraneibacter gnavus, also known as Ruminococcus gnavus, is a Gram-positive anaerobic bacterium that resides in the human gut microbiota. Notably, this bacterium plays dual roles in health and disease. On one side it supports nutrient metabolism essential for bodily functions and on the other it contributes to the development of Inflammatory Bowel Disease (IBD) and other gastrointestinal disorders. R. gnavus strain RJX1120 is an encapsulated strain and has been linked to develop IBD. Despite the advances made on its role in gut homeostasis, limited information is available on strain-specific virulence factors, metabolic pathways, and regulatory mechanisms. The study of such aspects is crucial to make microbiota-targeted therapy and understand its implications in host health. A multi-epitope vaccine against R. gnavus strain RJX1120 was designed using reverse vaccinology-based subtractive proteomics approach. Among the 3,219 proteins identified in the R. gnavus strain RJX1120, two critical virulent and antigenic proteins, a Single-stranded DNA-binding protein SSB (A0A2N5PT08) and Cell division ATP-binding protein FtsE (A0A2N5NK05) were screened and identified as potential targets. The predicted B-cell and T-cell epitopes from these proteins were screened for essential immunological properties such as antigenicity, allergenicity, solubility, MHC binding affinity, and toxicity. Epitopes chosen were cross-linked using suitable spacers and an adjuvant to develop a multi-epitope vaccine. Structural refinement of the construct revealed that 95.7% of the amino acid residues were located in favored regions, indicating a high-quality structural model. Molecular docking analysis demonstrated a robust interaction between the vaccine construct and the human Toll-like receptor 4 (TLR4), with a binding energy of -1277.0 kcal/mol. The results of molecular dynamics simulations further confirmed the stability of the vaccine-receptor complex under physiological conditions. In silico cloning of the vaccine construct yielded a GC content of 48% and a Codon Adaptation Index (CAI) value of 1.0, indicating optimal expression in the host system. These results indicate the possibility of the designed vaccine construct as a candidate for the prevention of R. gnavus-associated diseases. However, experimental validation is required to confirm its immunogenicity and protective efficacy.
Collapse
Affiliation(s)
- Hou Dingding
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
| | - Sher Muhammad
- Faculty of Agriculture and Veterinary Sciences, Superior University Lahore, Lahore, Pakistan
| | - Irfan Manzoor
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad (GCUF), Faisalabad, Pakistan
| | - Sana Abdul Ghaffar
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad (GCUF), Faisalabad, Pakistan
| | | | - Nadine MS. Moubayed
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ashraf Atef Hatamleh
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Xu Songxiao
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|
8
|
Tinajero-Trejo M, Aindow M, Pasquina-Lemonche L, Lafage L, Adedeji-Olulana AF, Sutton JAF, Wacnik K, Jia Y, Bilyk B, Yu W, Hobbs JK, Foster SJ. Control of morphogenesis during the Staphylococcus aureus cell cycle. SCIENCE ADVANCES 2025; 11:eadr5011. [PMID: 40215301 PMCID: PMC11988411 DOI: 10.1126/sciadv.adr5011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 03/06/2025] [Indexed: 04/14/2025]
Abstract
Bacterial cell division is a complex, multistage process requiring septum development while maintaining cell wall integrity. A dynamic, macromolecular protein complex, the divisome, tightly controls morphogenesis both spatially and temporally, but the mechanisms that tune septal progression are largely unknown. By studying conditional mutants of genes encoding DivIB, DivIC, and FtsL, an essential trimeric complex central to cell division in bacteria, we demonstrate that FtsL and DivIB play independent, hierarchical roles coordinating peptidoglycan synthesis across specific septal developmental checkpoints. They are required for the localization of downstream divisome components and the redistribution of peptidoglycan synthesis from the cell periphery to the septum. This is achieved by positive regulation of septum production and negative regulation of peripheral cell wall synthesis. Our analysis has led to a model for the coordination of cell division in Staphylococcus aureus, forming a framework for understanding how protein localization and function are integrated with cell wall structural dynamics across the bacteria.
Collapse
Affiliation(s)
- Mariana Tinajero-Trejo
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
| | - Matthew Aindow
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
| | - Laia Pasquina-Lemonche
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
| | - Lucia Lafage
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
| | - Abimbola Feyisara Adedeji-Olulana
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
- Department of Physics and Astronomy, University of Sheffield, Sheffield, UK
| | - Joshua A. F. Sutton
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
| | - Katarzyna Wacnik
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
| | - Yaosheng Jia
- Department of Molecular Biosciences, University of South Florida, Tampa, FL, USA
| | - Bohdan Bilyk
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
| | - Wenqi Yu
- Department of Molecular Biosciences, University of South Florida, Tampa, FL, USA
| | - Jamie K. Hobbs
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
- Department of Physics and Astronomy, University of Sheffield, Sheffield, UK
| | - Simon J. Foster
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
| |
Collapse
|
9
|
Harpring M, Lee J, Zhong G, Ouellette SP, Cox JV. FtsK is critical for the assembly of the unique divisome complex of the FtsZ-less Chlamydia trachomatis. eLife 2025; 13:RP104199. [PMID: 40193186 PMCID: PMC11975371 DOI: 10.7554/elife.104199] [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] [Indexed: 04/09/2025] Open
Abstract
Chlamydia trachomatis serovar L2 (Ct), an obligate intracellular bacterium that does not encode FtsZ, divides by a polarized budding process. In the absence of FtsZ, we show that FtsK, a chromosomal translocase, is critical for divisome assembly in Ct. Chlamydial FtsK forms discrete foci at the septum and at the base of the progenitor mother cell, and our data indicate that FtsK foci at the base of the mother cell mark the location of nascent divisome complexes that form at the site where a daughter cell will emerge in the next round of division. The divisome in Ct has a hybrid composition, containing elements of the divisome and elongasome from other bacteria, and FtsK is recruited to nascent divisomes prior to the other chlamydial divisome proteins assayed, including the PBP2 and PBP3 transpeptidases, and MreB and MreC. Knocking down FtsK prevents divisome assembly in Ct and inhibits cell division and septal peptidoglycan synthesis. We further show that MreB does not function like FtsZ and serve as a scaffold for the assembly of the Ct divisome. Rather, MreB is one of the last proteins recruited to the chlamydial divisome, and it is necessary for the formation of septal peptidoglycan rings. Our studies illustrate the critical role of chlamydial FtsK in coordinating divisome assembly and peptidoglycan synthesis in this obligate intracellular bacterial pathogen.
Collapse
Affiliation(s)
- McKenna Harpring
- Department of Microbiology, Immunology, and Biochemistry. University of Tennessee Health Science CenterMemphisUnited States
| | - Junghoon Lee
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical CenterOmahaUnited States
| | - Guangming Zhong
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health San AntonioSan AntonioUnited States
| | - Scot P Ouellette
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical CenterOmahaUnited States
| | - John V Cox
- Department of Microbiology, Immunology, and Biochemistry. University of Tennessee Health Science CenterMemphisUnited States
| |
Collapse
|
10
|
Navarro PP, Vettiger A, Hajdu R, Ananda VY, López-Tavares A, Schmid EW, Walter JC, Loose M, Chao LH, Bernhardt TG. The aPBP-type cell wall synthase PBP1b plays a specialized role in fortifying the Escherichia coli division site against osmotic rupture. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.02.646830. [PMID: 40236067 PMCID: PMC11996507 DOI: 10.1101/2025.04.02.646830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
A multi-protein system called the divisome promotes bacterial division. This apparatus synthesizes the peptidoglycan (PG) cell wall layer that forms the daughter cell poles and protects them from osmotic lysis. In the model Gram-negative bacterium Escherichia coli , PG synthases called class A penicillin-binding proteins (aPBPs) have been proposed to play crucial roles in division. However, there is limited experimental support for aPBPs playing a specialized role in division that is distinct from their general function in the expansion and fortification of the PG matrix. Here, we present in situ cryogenic electron tomography data indicating that the aPBP-type enzyme PBP1b is required to produce a wedge-like density of PG at the division site. Furthermore, atomic force and live cell microscopy showed that loss of this structure weakens the division site and renders it susceptible to lysis. Surprisingly, we found that the lipoprotein activator LpoB needed to promote the general function of PBP1b was not required for normal division site architecture or its integrity. Additionally, we show that of the two PBP1b isoforms produced in cells, it is the one with an extended cytoplasmic N-terminus that functions in division, likely via recruitment by the FtsA component of the divisome. Altogether, our results demonstrate that PBP1b plays a specialized, LpoB-independent role in E. coli cell division involving the biogenesis of a PG structure that prevents osmotic rupture. The conservation of aPBPs with extended cytoplasmic N-termini suggests that other Gram-negative bacteria may use similar mechanisms to reinforce their division site.
Collapse
|
11
|
Li B, Fu CL, Sun ZY. Shaping membrane vesicles by tuning the activity of confined active polymer chains. J Chem Phys 2025; 162:094901. [PMID: 40029089 DOI: 10.1063/5.0244184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/13/2025] [Indexed: 03/05/2025] Open
Abstract
Semi-flexible polymers, such as actin filaments, can deform the shape of membrane when confined in a membrane vesicle, playing an important role in biological processes. Here, we use dynamic Monte Carlo simulations to study an active polymer chain confined in a membrane vesicle. For flexible polymer chains, the membrane shape is governed by the competition between membrane bending rigidity and polymer activity. Stiff membrane is unaffected by small active forces, but moderate forces cause the polymer to alternate between stretched and disordered configurations, increasing the asphericity of both the polymer and the vesicle. For semi-flexible polymer chains, their stiffness can significantly impact both the vesicle and polymer shapes. We identify distinct classes of configurations that emerge as a function of polymer stiffness, membrane bending rigidity, and polymer activity. A weak polymer activity can cause the polymer to align along its contour, effectively increasing its stiffness. However, a moderate polymer activity softens the polymer chain. For membranes with low bending rigidities κ, large-scale deformations, such as wormlike or tadpole-shaped vesicles, appear at a weak polymer activity and high polymer stiffness. In the wormlike configuration, the polymer chain adopts a hairpin configuration to minimize the polymer bending energy. As the polymer stiffness increases, a tadpole-like vesicle forms, with part of the polymer deforming the membrane into a protrusion while the rest remaining confined in a bud-like structure. For stiffer membranes, we observe oblate vesicles containing toroidal polymer chains, resulting from the high cost of membrane bending energy. A moderate polymer activity causes the softening of the polymer chain, leading to a nearly spherical vesicle with slight shape fluctuation. We further characterize the order parameter of toroidal polymer chains in oblate vesicles and reveal that a slight increase in polymer activity leads to a more ordered helical structure of polymer chains.
Collapse
Affiliation(s)
- Bing Li
- State Key Laboratory of Polymer Physics and Chemistry and Key Laboratory of Polymer Science and Technology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022 Changchun, China
| | - Cui-Liu Fu
- State Key Laboratory of Polymer Physics and Chemistry and Key Laboratory of Polymer Science and Technology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022 Changchun, China
| | - Zhao-Yan Sun
- State Key Laboratory of Polymer Physics and Chemistry and Key Laboratory of Polymer Science and Technology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022 Changchun, China
- University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
12
|
Zhao K, Du S, Tian L, Wang S, Shi R, Sun H, Zhou Y, Huang C, Sun Y, Wang S, Chen Y. Bacteriophage P1 protein Icd inhibits bacterial division by targeting FtsZ. Front Microbiol 2025; 16:1533694. [PMID: 40078545 PMCID: PMC11897509 DOI: 10.3389/fmicb.2025.1533694] [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: 11/24/2024] [Accepted: 02/14/2025] [Indexed: 03/14/2025] Open
Abstract
The study of bacteriophage (phage) gene products and their effects on the host helps to better understand the phage-host relationship and provides clues for the development of new antimicrobial proteins. In this study, we focused on a small protein named Icd with 73 amino acids from phage P1. It inhibits the growth of Escherichia coli and rapidly blocks the formation of Z-ring. The results of bacterial two-hybrid and pull-down experiments showed that Icd directly targets FtsZ, a key protein in bacterial division. Furthermore, we identified the core region of Icd as amino acids 12-51; this 40-amino acid protein had similar antibacterial activity to the full-length Icd, inhibiting bacterial growth and division.
Collapse
Affiliation(s)
- Kairui Zhao
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, China
| | - Shuheng Du
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, China
| | - Linlin Tian
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, China
| | - Shenping Wang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, China
| | - Runqin Shi
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, China
| | - Haiyu Sun
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, China
| | - Yao Zhou
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, China
| | - Chenhao Huang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, China
| | - Yanmei Sun
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, China
- Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi’an, China
| | - Shiwei Wang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, China
- Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi’an, China
| | - Yaodong Chen
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, China
- Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi’an, China
| |
Collapse
|
13
|
Hernandez-Rocamora V, Martorana AM, Belloso A, Ballesteros D, Zaccaria M, Perez AJ, Iorga BI, Abia D, Gray J, Breukink E, Xiao J, Pazos M, Polissi A, Vollmer W. A novel peptidoglycan deacetylase modulates daughter cell separation in E. coli. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638797. [PMID: 40027703 PMCID: PMC11870482 DOI: 10.1101/2025.02.18.638797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Peptidoglycan hydrolases facilitate bacterial cell wall growth by creating space for insertion of new material and allowing physical separation of daughter cells. In Escherichia coli, three peptidoglycan amidases, AmiA, AmiB and AmiC, cleave septal peptidoglycan during cell division. The LytM-domain proteins EnvC and NlpD activate these amidases either from inside the cell or the outer membrane: EnvC binds to the cytoplasmic membrane-anchored divisome components FtsEX, and NlpD and ActS are outer membrane lipoproteins. Here we report the identification of a novel periplasmic deacetylase called SddA that removes acetyl groups from denuded peptidoglycan glycan strands, the products of amidases. SddA is a substrate for the periplasmic protease Prc, suggesting regulation via protein degradation. The sddA gene is co-expressed with the gene encoding EnvC, linking SddA function to amidase activation. Consistent with this link, the deletion of sddA alleviates phenotypes associated with lack of amidase activation, while overexpression of sddA alleviates phenotypes related to a defective Tol-Pal system and causes cell chaining due to reduced septum peptidoglycan cleavage unless envC is co-expressed. We present a model according to which SddA modulates the activation of the septum-splitting amidases during cell division.
Collapse
Affiliation(s)
- Victor Hernandez-Rocamora
- Centre for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Alessandra M Martorana
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milano, Milano, Italy
| | - Aitana Belloso
- Centro de Biología Molecular Severo Ochoa (CBM), CSIC - Universidad Autónoma de Madrid, Madrid, Spain
| | - Daniel Ballesteros
- Centro de Biología Molecular Severo Ochoa (CBM), CSIC - Universidad Autónoma de Madrid, Madrid, Spain
| | - Marta Zaccaria
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milano, Milano, Italy
| | - Amilcar J Perez
- Department of Biophysics & Biophysical Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bogdan I Iorga
- Université Paris-Saclay, CNRS, Institut de Chimie des Substances Naturelles, Gif-sur-Yvette, France
| | - David Abia
- Centro de Biología Molecular Severo Ochoa (CBM), CSIC - Universidad Autónoma de Madrid, Madrid, Spain
| | - Joe Gray
- Centre for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Eefjan Breukink
- Membrane Biochemistry and Biophysics, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Utrecht University, Utrecht, The Netherlands
| | - Jie Xiao
- Department of Biophysics & Biophysical Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Manuel Pazos
- Centro de Biología Molecular Severo Ochoa (CBM), CSIC - Universidad Autónoma de Madrid, Madrid, Spain
- Instituto Universitario de Biología Molecular (IUBM) y Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alessandra Polissi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milano, Milano, Italy
| | - Waldemar Vollmer
- Centre for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| |
Collapse
|
14
|
Zhidkov ME, Smirnova PA, Grammatikova NE, Isakova EB, Shchekotikhin AE, Styshova ON, Klimovich AA, Popov AM. Comparative Evaluation of the Antibacterial and Antitumor Activities of Marine Alkaloid 3,10-Dibromofascaplysin. Mar Drugs 2025; 23:68. [PMID: 39997192 PMCID: PMC11857626 DOI: 10.3390/md23020068] [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: 12/30/2024] [Revised: 01/30/2025] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
Fascaplysins form a group of marine natural products with unique cationic five-ring coplanar backbone. Native fascaplysin exhibits a broad spectrum of bioactivities, among which the cytotoxic activity has been the most investigated. Several fascaplysin derivatives have more selective biological effects and are promising as lead compounds. Thus, the introduction of a substituent at C-9 of fascaplysin leads to a strong increase in its antimicrobial properties. Here, a comparative assessment of the antimicrobial activity of synthetic analogs of the marine alkaloids 3-bromofascaplysin, 10-bromofascaplysin, and 3,10-dibromofascaplysin, along with some of their isomers and analogs, was carried out against a panel of Gram-positive bacteria in vitro. For the first time, a significant increase in the antimicrobial activity of fascaplysin was observed when a substituent was introduced at C-3. The introduction of two bromine atoms at C-2 and C-9 enhances the antimicrobial properties by 4 to 16 times, depending on the tested strain. Evaluation of the antimicrobial potential in vivo showed that fascaplysin and 3,10-dibromofascaplysin had comparable efficacy in the mouse staphylococcal sepsis model. Additionally, 3,10-dibromofascaplysin demonstrated a strong and reliable antitumor effect in vivo on the Ehrlich carcinoma inoculated subcutaneously, with a value of tumor growth inhibition by 49.2% 20 days after treatment. However, further studies on alternative chemical modifications of fascaplysin are needed to improve its chemotherapeutic properties.
Collapse
Affiliation(s)
- Maxim E. Zhidkov
- Department of Chemistry and Materials, Institute of High Technologies and Advanced Materials, FEFU Campus, Far Eastern Federal University, Ajax Bay 10, Russky Island, Vladivostok 690922, Russia;
| | - Polina A. Smirnova
- Department of Chemistry and Materials, Institute of High Technologies and Advanced Materials, FEFU Campus, Far Eastern Federal University, Ajax Bay 10, Russky Island, Vladivostok 690922, Russia;
| | - Natalia E. Grammatikova
- Laboratory of Chemical Transformation of Antibiotics, Gause Institute of New Antibiotics, Moscow 119021, Russia; (N.E.G.); (E.B.I.); (A.E.S.)
| | - Elena B. Isakova
- Laboratory of Chemical Transformation of Antibiotics, Gause Institute of New Antibiotics, Moscow 119021, Russia; (N.E.G.); (E.B.I.); (A.E.S.)
| | - Andrey E. Shchekotikhin
- Laboratory of Chemical Transformation of Antibiotics, Gause Institute of New Antibiotics, Moscow 119021, Russia; (N.E.G.); (E.B.I.); (A.E.S.)
| | - Olga N. Styshova
- Departments of Biotechnology and Marine Natural Compounds Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of The Russian Academy of Sciences, Vladivostok 690922, Russia; (O.N.S.); (A.A.K.); (A.M.P.)
| | - Anna A. Klimovich
- Departments of Biotechnology and Marine Natural Compounds Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of The Russian Academy of Sciences, Vladivostok 690922, Russia; (O.N.S.); (A.A.K.); (A.M.P.)
| | - Aleksandr M. Popov
- Departments of Biotechnology and Marine Natural Compounds Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of The Russian Academy of Sciences, Vladivostok 690922, Russia; (O.N.S.); (A.A.K.); (A.M.P.)
| |
Collapse
|
15
|
Liu J, Lelek M, Yang Y, Salles A, Zimmer C, Shen Y, Krupovic M. A relay race of ESCRT-III paralogs drives cell division in a hyperthermophilic archaeon. mBio 2025; 16:e0099124. [PMID: 39699168 PMCID: PMC11796394 DOI: 10.1128/mbio.00991-24] [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: 04/02/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024] Open
Abstract
Cell division is a fundamental process ensuring the perpetuation of all cellular life forms. Archaea of the order Sulfolobales divide using a simpler version of the eukaryotic endosomal sorting complexes required for transport (ESCRT) machinery, composed of three ESCRT-III homologs (ESCRT-III, -III-1, and -III-2), AAA+ ATPase Vps4 and an archaea-specific component CdvA. Here, we clarify how these components act sequentially to drive the division of the hyperthermophilic archaeon Saccharolobus islandicus. Our data suggest that ESCRT-III plays an active role during the early stage of membrane constriction during cytokinesis, whereas ESCRT-III-1 and ESCRT-III-2 are indispensable for the "pre-late" and "late" stages of cytokinesis, respectively. In the escrt-III-1 deletion strain, the division is blocked when the mid-cell constriction reaches ~30% of the initial cell diameter ("pre-late" stage), yielding "chain-like" cellular aggregates. Depletion of ESCRT-III-2 leads to the accumulation of cells connected through narrow membrane bridges ("late" stage), consistent with the key role of this protein in the final membrane abscission. We used 3D-single molecule localization microscopy to image ESCRT-III rings of different diameters and show that the decrease in the ESCRT-III ring diameter and membrane constriction are inconsistent with a mechanism exclusively based on spiraling of the ESCRT-III filaments. By contrast, the cone-shaped assemblies of ESCRT-III-1 and ESCRT-III-2 are consistent with spiral formation, highlighting the distinct roles of the three ESCRT-III proteins during the cytokinesis. We propose the "relay race" model, whereby the cytokinesis is achieved through a sequential and concerted action of different ESCRT machinery components. IMPORTANCE Two major cytokinesis mechanisms, rooted in contractile FtsZ and endosomal sorting complexes required for transport (ESCRT) rings, respectively, have emerged in the course of evolution. Whereas bacteria rely on the FtsZ-based mechanism, different lineages of archaea use either of the two systems, and eukaryotes have inherited the ESCRT-based cell division machinery from their archaeal ancestors. The mechanism of ESCRT-based cell division in archaea remains poorly understood and mechanistic studies on different archaeal model systems are essential to unravel the natural history of the ESCRT machinery. Here we investigate the interplay between three major ESCRT-III homologs during the division of a hyperthermophilic archaeon Saccharolobus islandicus and propose the "relay race" model of cytokinesis.
Collapse
Affiliation(s)
- Junfeng Liu
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Archaeal Virology Unit, Paris, France
- CRISPR and Archaea Biology Research Centre, Microbial Technology Institute and State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Mickaël Lelek
- Imaging and Modeling Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Yunfeng Yang
- CRISPR and Archaea Biology Research Centre, Microbial Technology Institute and State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Audrey Salles
- Institut Pasteur, Université Paris Cité, Unit of Technology and Service Photonic BioImaging (UTechS PBI), C2RT, Paris, France
| | - Christophe Zimmer
- Imaging and Modeling Unit, Institut Pasteur, Université Paris Cité, Paris, France
- Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Yulong Shen
- CRISPR and Archaea Biology Research Centre, Microbial Technology Institute and State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Mart Krupovic
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Archaeal Virology Unit, Paris, France
| |
Collapse
|
16
|
Milferstaedt SWL, Joest M, Bohlender LL, Hoernstein SNW, Özdemir B, Decker EL, van der Does C, Reski R. Differential GTP-dependent in-vitro polymerization of recombinant Physcomitrella FtsZ proteins. Sci Rep 2025; 15:3095. [PMID: 39856123 PMCID: PMC11760385 DOI: 10.1038/s41598-024-85077-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
Bacterial cell division and plant chloroplast division require selfassembling Filamentous temperature-sensitive Z (FtsZ) proteins. FtsZ proteins are GTPases sharing structural and biochemical similarities with eukaryotic tubulin. In the moss Physcomitrella, the morphology of the FtsZ polymer networks varies between the different FtsZ isoforms. The underlying mechanism and foundation of the distinct networks is unknown. Here, we investigated the interaction of Physcomitrella FtsZ2-1 with FtsZ1 isoforms via co-immunoprecipitation and mass spectrometry, and found protein-protein interaction in vivo. We tagged FtsZ1-2 and FtsZ2-1 with different fluorophores and expressed both in E. coli, which led to the formation of defined structures within the cells and to an influence on bacterial cell division and morphology. Furthermore, we have optimized the purification protocols for FtsZ1-2 and FtsZ2-1 expressed in E. coli and characterized their GTPase activity and polymerization in vitro. Both FtsZ isoforms showed GTPase activity. Stoichiometric mixing of both proteins led to a significantly increased GTPase activity, indicating a synergistic interaction between them. In light scattering assays, we observed GTP-dependent assembly of FtsZ1-2 and of FtsZ2-1 in a protein concentration dependent manner. Stoichiometric mixing of both proteins resulted in significantly faster polymerization, again indicating a synergistic interaction between them. Under the same conditions used for GTPase and light scattering assays both FtsZ isoforms formed filaments in a GTP-dependent manner as visualized by transmission electron microscopy (TEM). Taken together, our results reveal that Physcomitrella FtsZ1-2 and FtsZ2-1 are functionally different, can synergistically interact in vivo and in vitro, and differ in their properties from FtsZ proteins from bacteria, archaea and vascular plants.
Collapse
Affiliation(s)
- Stella W L Milferstaedt
- Plant Biotechnology, Faculty of Biology, University of Freiburg, Schaenzlestr. 1, 79104, Freiburg, Germany
- Cluster of Excellence livMatS @ FIT - Freiburg Centre for Interactive Materials and Bioinspired Technologies, University of Freiburg, Georges-Köhler-Allee 105, 79110, Freiburg, Germany
| | - Marie Joest
- Molecular Biology of Archaea, Faculty of Biology, University of Freiburg, Schaenzlestr. 1, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine SGBM, University of Freiburg, Albertstraße 19A, 79104, Freiburg, Germany
| | - Lennard L Bohlender
- Plant Biotechnology, Faculty of Biology, University of Freiburg, Schaenzlestr. 1, 79104, Freiburg, Germany
| | - Sebastian N W Hoernstein
- Plant Biotechnology, Faculty of Biology, University of Freiburg, Schaenzlestr. 1, 79104, Freiburg, Germany
| | - Buğra Özdemir
- Plant Biotechnology, Faculty of Biology, University of Freiburg, Schaenzlestr. 1, 79104, Freiburg, Germany
- , Euro-BioImaging Bio-Hub, EMBL, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Eva L Decker
- Plant Biotechnology, Faculty of Biology, University of Freiburg, Schaenzlestr. 1, 79104, Freiburg, Germany
| | - Chris van der Does
- Molecular Biology of Archaea, Faculty of Biology, University of Freiburg, Schaenzlestr. 1, 79104, Freiburg, Germany
| | - Ralf Reski
- Plant Biotechnology, Faculty of Biology, University of Freiburg, Schaenzlestr. 1, 79104, Freiburg, Germany.
- Cluster of Excellence livMatS @ FIT - Freiburg Centre for Interactive Materials and Bioinspired Technologies, University of Freiburg, Georges-Köhler-Allee 105, 79110, Freiburg, Germany.
- Spemann Graduate School of Biology and Medicine SGBM, University of Freiburg, Albertstraße 19A, 79104, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, Schaenzlestr. 18, 79104, Freiburg, Germany.
| |
Collapse
|
17
|
López-Menéndez H, Luque-Rioja C, Kharbedia M, Herráez-Aguilar D, Santiago JA, Monroy F. Multiscale modelling of active hydrogel elasticity driven by living polymers: softening by bacterial motor protein FtsZ. SOFT MATTER 2025; 21:670-686. [PMID: 39760521 DOI: 10.1039/d4sm00839a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
We present a neo-Hookean elasticity theory for hybrid mechano-active hydrogels, integrating motor proteins into polymer meshes to create composite materials with active softening due to modulable chain overlaps. Focusing on polyacrylamide (PA) hydrogels embedded with FtsZ, a bacterial cytokinetic protein powered by GTP, we develop a multiscale model using microscopic Flory theory of rubbery meshes through mesoscopic De Gennes' scaling concepts for meshwork dynamics and phenomenological Landau's formalism for second-order phase transitions. Our theoretical multiscale model explains the active softening observed in hybrid FtsZ-PA hydrogels by incorporating modulable meshwork dynamics, such as overlapping functionality and reptation dynamics, into an active mean-field of unbinding interactions. The novel FtsZ-based metamaterial and companion multiscale theory offer insights for designing, predicting, and controlling complex active hydrogels, with potential applications in technology and biomedicine.
Collapse
Affiliation(s)
- Horacio López-Menéndez
- Department of Physical Chemistry, Complutense University of Madrid, Av. Complutense s/n, 28040 Madrid, Spain.
| | - Clara Luque-Rioja
- Department of Physical Chemistry, Complutense University of Madrid, Av. Complutense s/n, 28040 Madrid, Spain.
- Unit of Translational Biophysics, IIS Hospital Doce de Octubre (Imas12), Av. Andalucía s/n, 28041 Madrid, Spain
| | - Mikheil Kharbedia
- Department of Physical Chemistry, Complutense University of Madrid, Av. Complutense s/n, 28040 Madrid, Spain.
| | - Diego Herráez-Aguilar
- Instituto de Investigaciones Biosanitarias, Universidad Francisco de Vitoria, Ctra. Pozuelo-Majadahonda, 28223 Pozuelo de Alarcón, Spain
| | - José A Santiago
- Department of Physical Chemistry, Complutense University of Madrid, Av. Complutense s/n, 28040 Madrid, Spain.
- Departamento de Matemáticas Aplicadas y Sistemas, Universidad Autónoma Metropolitana Cuajimalpa, Vasco de Quiroga 4871, 05348 Ciudad de México, Mexico
| | - Francisco Monroy
- Department of Physical Chemistry, Complutense University of Madrid, Av. Complutense s/n, 28040 Madrid, Spain.
- Unit of Translational Biophysics, IIS Hospital Doce de Octubre (Imas12), Av. Andalucía s/n, 28041 Madrid, Spain
| |
Collapse
|
18
|
Harpring M, Lee J, Zhong G, Ouellette SP, Cox JV. FtsK is Critical for the Assembly of the Unique Divisome Complex of the FtsZ-less Chlamydia trachomatis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.24.620021. [PMID: 39484550 PMCID: PMC11527202 DOI: 10.1101/2024.10.24.620021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Chlamydia trachomatis serovar L2 (Ct), an obligate intracellular bacterium that does not encode FtsZ, divides by a polarized budding process. In the absence of FtsZ, we show that FtsK, a chromosomal translocase, is critical for divisome assembly in Ct. Chlamydial FtsK forms discrete foci at the septum and at the base of the progenitor mother cell, and our data indicate that FtsK foci at the base of the mother cell mark the location of nascent divisome complexes that form at the site where a daughter cell will emerge in the next round of division. The divisome in Ct has a hybrid composition, containing elements of the divisome and elongasome from other bacteria, and FtsK is recruited to nascent divisomes prior to the other chlamydial divisome proteins assayed, including the PBP2 and PBP3 transpeptidases, and MreB and MreC. Knocking down FtsK prevents divisome assembly in Ct and inhibits cell division and septal peptidoglycan synthesis. We further show that MreB does not function like FtsZ and serve as a scaffold for the assembly of the Ct divisome. Rather, MreB is one of the last proteins recruited to the chlamydial divisome, and it is necessary for the formation of septal peptidoglycan rings. Our studies illustrate the critical role of chlamydial FtsK in coordinating divisome assembly and peptidoglycan synthesis in this obligate intracellular bacterial pathogen.
Collapse
Affiliation(s)
- McKenna Harpring
- Department of Microbiology, Immunology, and Biochemistry. University of Tennessee Health Science Center. Memphis, TN, USA
| | - Junghoon Lee
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Guangming Zhong
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health San Antonio, TX, USA
| | - Scot P. Ouellette
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | - John V. Cox
- Department of Microbiology, Immunology, and Biochemistry. University of Tennessee Health Science Center. Memphis, TN, USA
| |
Collapse
|
19
|
Liu J, Ha T. Connecting single-molecule and superresolution microscopies to cell biology through theoretical modeling. Biophys J 2025; 124:15-24. [PMID: 39600094 PMCID: PMC11739872 DOI: 10.1016/j.bpj.2024.11.3308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 11/12/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024] Open
Abstract
Recent developments of single-molecule and superresolution microscopies reveal novel spatial-temporal features of various cellular processes with unprecedented details, and greatly facilitate the development of theoretical models. In this review, we synthesize our view of how to meaningfully integrate these experimental approaches with theoretical modeling to obtain deeper understanding of the physical mechanisms of cell biology.
Collapse
Affiliation(s)
- Jian Liu
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Taekjip Ha
- Howard Hughes Medical Institute and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
20
|
Gulsoy IC, Saaki TNV, Wenzel M, Syvertsson S, Morimoto T, Siersma TK, Hamoen LW. Minimization of the Bacillus subtilis divisome suggests FtsZ and SepF can form an active Z-ring, and reveals the amino acid transporter BraB as a new cell division influencing factor. PLoS Genet 2025; 21:e1011567. [PMID: 39869651 PMCID: PMC11790237 DOI: 10.1371/journal.pgen.1011567] [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: 08/02/2024] [Revised: 02/03/2025] [Accepted: 01/06/2025] [Indexed: 01/29/2025] Open
Abstract
Bacterial cytokinesis begins with polymerization of the tubulin homologue FtsZ into a ring-like structure at midcell, the Z-ring, which recruits the late cell division proteins that synthesize the division septum. Assembly of FtsZ is carefully regulated and supported by a dozen conserved cell division proteins. Generally, these proteins are not essential, but removing more than one is in many cases lethal. Therefore, it is still not fully clear how the different protein components contribute to cell division, and whether there is a minimal set of proteins that can execute cell division. In this study, we tried to find the minimal set of proteins that is required to establish an active Z-ring in the model bacterium Bacillus subtilis. By making use of known suppressor mutations we were able to find a gene deletion route that eventually enabled us the remove eight conserved cell division proteins: ZapA, MinC, MinJ, UgtP, ClpX, Noc, EzrA and FtsA. Only FtsZ and its membrane anchor SepF appeared to be required for Z-ring formation. Interestingly, SepF is also the FtsZ anchor in archaea, and both proteins date back to the Last Universal Common Ancestor (LUCA). Viability of the multiple deletion mutant was not greatly affected, although the frequency of cell division was considerably reduced. Whole genome sequencing suggested that the construction of this minimal divisome strain was also possible due to the accumulation of suppressor mutations. After extensive phenotypic testing of these mutations, we found an unexpected cell division regulation function for the branched chain amino acid transporter BraB, which may be related to a change in fatty acid composition. The implications of these findings for the role of SepF, and the construction of a minimal cell division machinery are discussed.
Collapse
Affiliation(s)
- Ilkay Celik Gulsoy
- Centre for Bacterial Cell Biology, Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle, United Kingdom
| | - Terrens N. V. Saaki
- Bacterial Cell Biology and Physiology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Michaela Wenzel
- Bacterial Cell Biology and Physiology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Simon Syvertsson
- Centre for Bacterial Cell Biology, Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle, United Kingdom
| | - Taku Morimoto
- Graduate School of Information Science, Nara Institute of Science and Technology, Ikoma, Nara, Japan
| | - Tjalling K. Siersma
- Bacterial Cell Biology and Physiology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Leendert W. Hamoen
- Centre for Bacterial Cell Biology, Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle, United Kingdom
- Bacterial Cell Biology and Physiology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Wang Y, Zhou Z, Liu X. Auxin promotes chloroplast division by increasing the expression of chloroplast division genes. PLANT CELL REPORTS 2024; 44:20. [PMID: 39741196 DOI: 10.1007/s00299-024-03415-4] [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: 10/21/2024] [Accepted: 12/21/2024] [Indexed: 01/02/2025]
Abstract
KEY MESSAGE Auxin stimulates chloroplast division by upregulating the expression of genes involved in chloroplast division and influencing the positioning of chloroplast division rings. Chloroplasts divide by binary fission, forming a ring complex at the division site. Auxin, particularly indole acetic acid (IAA), significantly influences various aspects of plant growth. However, the impact of auxin on chloroplast division remains unclear. In this study, different concentrations of exogenous IAA were applied to wild Arabidopsis thaliana. The results showed that the number and size differences of chloroplasts in the cells of Arabidopsis thaliana treated with high concentrations of IAA increased compared to untreated plants. Further investigation revealed that high concentrations of IAA affected the expression of chloroplast division genes and the formation of division rings. In chloroplast division mutants, the effect of IAA on promoting chloroplast division is impaired. Defects of IAA synthetic gene also lead to a reduced effect of IAA on chloroplast division. Our findings demonstrate that auxin influences chloroplast division by regulating the expressions of chloroplast division genes and affecting the localization of division rings. These results are significant for further exploring the relationship between auxin and chloroplast division.
Collapse
Affiliation(s)
- Yixuan Wang
- State Key Laboratory of Tree Genetics and Breeding, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083, China
| | - Zhongyang Zhou
- State Key Laboratory of Tree Genetics and Breeding, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083, China
| | - Xiaomin Liu
- State Key Laboratory of Tree Genetics and Breeding, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 100083, China.
| |
Collapse
|
22
|
Choudhury J, Chaudhuri BN. Tubules, Rods, and Spirals: Diverse Modes of SepF-FtsZ Assembling. Cytoskeleton (Hoboken) 2024. [PMID: 39703081 DOI: 10.1002/cm.21975] [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: 08/07/2024] [Revised: 11/08/2024] [Accepted: 11/17/2024] [Indexed: 12/21/2024]
Abstract
Z-ring formation by FtsZ, the master assembler of the divisome, is a key step in bacterial cell division. Membrane anchoring of the Z-ring requires the assistance of dedicated Z-ring binding proteins, such as SepF and FtsA. SepF participates in bundling and membrane anchoring of FtsZ in gram-positive bacteria. We report in vitro biophysical studies of the interactions between FtsZ and a cytoplasmic component of cognate SepF from three different bacteria: Mycobacterium tuberculosis, Staphylococcus aureus, and Enterococcus gallinarum. While the cytosolic domain of SepF from M. tuberculosis is primarily a dimer, those from S. aureus and E. gallinarum polymerize to form ring-like structures. Mycobacterial SepF helps in the bundling of FtsZ filaments to form thick filaments and large spirals. On the other hand, ring-forming SepF from the Firmicutes bundle FtsZ into tubules. Our results suggest that the oligomeric form of SepF directs how it bundles FtsZ filaments.
Collapse
Affiliation(s)
- Jagrity Choudhury
- GN Ramachandran Protein Center, CSIR Institute of Microbial Technology, Chandigarh, India
| | - Barnali N Chaudhuri
- GN Ramachandran Protein Center, CSIR Institute of Microbial Technology, Chandigarh, India
- Academy of Scientific and Innovative Research (AcSIR), AcSIR Headquarters CSIR-HRDC Campus, Postal Staff College Area, Ghaziabad, India
| |
Collapse
|
23
|
Shi H, Nguyen J, Gitai Z, Shaevitz J, Bratton BP, Gopinathan A, Grason G, Huang KC. Sensing the shape of a surface by intracellular filaments. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.18.624198. [PMID: 39605553 PMCID: PMC11601562 DOI: 10.1101/2024.11.18.624198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Understanding the mechanisms that dictate the localization of cytoskeletal filaments is crucial for elucidating cell shape regulation in prokaryotes. The actin homolog MreB plays a pivotal role in maintaining the shape of many rod-shaped bacteria such as Escherichia coli by directing cell-wall synthesis according to local curvature cues. However, the basis of MreB's curvature-dependent localization has remained elusive. Here, we develop a biophysical model for the energetics of filament binding to a surface that integrates the complex interplay between filament twist and bending and the two-dimensional surface geometry. Our model predicts that the spatial localization of a filament like MreB with substantial intrinsic twist is governed by both the mean and Gaussian curvatures of the cell envelope, which strongly covary in rod-shaped cells. Using molecular dynamics simulations to estimate the mechanical properties of MreB filaments, we show that their thermodynamic preference for regions with lower mean and Gaussian curvatures matches experimental observations for physiologically relevant filament lengths of ∼50 nm. We find that the experimentally measured statistical curvature preference is maintained in the absence of filament motion and after a cycle of depolymerization, repolymerization, and membrane rebinding, indicating that equilibrium energetics can explain MreB localization. These findings provide critical insights into the physical principles underlying cytoskeletal filament localization, and suggest new design principles for synthetic shape sensing nanomaterials. Significance statement The protein MreB, a homolog of eukaryotic actin, regulates the shape of bacteria like Escherichia coli by guiding new cell-wall insertion based on local curvature cues. However, the mechanism by which a nanometer-scale MreB filament "senses" the micron-scale curvature of the cell wall has remained a mystery. We introduce a biophysical model of the energetics of twisted and bent filaments bound to curved surfaces, which predicts that localization of filaments like MreB is sensitive to both mean and Gaussian curvature. The model captures experimentally measured curvature enrichment patterns and explains how MreB naturally localizes to saddle-shaped regions without energy-consuming processes. Beyond cell shape regulation, our work suggests design principles for synthetic systems that can sense and respond to surface shape.
Collapse
|
24
|
Männik J, Kar P, Amarasinghe C, Amir A, Männik J. Determining the rate-limiting processes for cell division in Escherichia coli. Nat Commun 2024; 15:9948. [PMID: 39550358 PMCID: PMC11569214 DOI: 10.1038/s41467-024-54242-w] [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: 03/01/2024] [Accepted: 11/01/2024] [Indexed: 11/18/2024] Open
Abstract
A critical cell cycle checkpoint for most bacteria is the onset of constriction when the septal peptidoglycan synthesis starts. According to the current understanding, the arrival of FtsN to midcell triggers this checkpoint in Escherichia coli. Recent structural and in vitro data suggests that recruitment of FtsN to the Z-ring leads to a conformational switch in actin-like FtsA, which links FtsZ protofilaments to the cell membrane and acts as a hub for the late divisome proteins. Here, we investigate this putative pathway using in vivo measurements and stochastic cell cycle modeling at moderately fast growth rates. Quantitatively upregulating protein concentrations and determining the resulting division timings shows that FtsN and FtsA numbers are not rate-limiting for the division in E. coli. However, at higher overexpression levels, they affect divisions: FtsN by accelerating and FtsA by inhibiting them. At the same time, we find that the FtsZ numbers in the cell are one of the rate-limiting factors for cell divisions in E. coli. Altogether, these findings suggest that instead of FtsN, accumulation of FtsZ in the Z-ring is one of the main drivers of the onset of constriction in E. coli at faster growth rates.
Collapse
Affiliation(s)
- Jaana Männik
- Department of Physics and Astronomy, University of Tennessee, Knoxville, TN, 37996, USA
| | - Prathitha Kar
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02134, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02134, USA
| | | | - Ariel Amir
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot, Israel
| | - Jaan Männik
- Department of Physics and Astronomy, University of Tennessee, Knoxville, TN, 37996, USA.
| |
Collapse
|
25
|
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
|
26
|
Rani R, Marinho Righetto G, Schäfer AB, Wenzel M. The Diverse Activities and Mechanisms of the Acylphloroglucinol Antibiotic Rhodomyrtone: Antibacterial Activity and Beyond. Antibiotics (Basel) 2024; 13:936. [PMID: 39452203 PMCID: PMC11504083 DOI: 10.3390/antibiotics13100936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/17/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024] Open
Abstract
Background/Objectives: The rose myrtle Rhodomyrtus tomentosa is a medicinal plant used in traditional Asian medicine. The active compound in R. tomentosa leaf extracts is rhodomyrtone, a chiral acylphloroglucinol. Rhodomyrtone exhibits an impressive breadth of activities, including antibacterial, antiviral, antiplasmodial, immunomodulatory, and anticancer properties. Its antibacterial properties have been extensively studied. Methods: We performed a comprehensive literature review on rhodomyrtone and summarized the current knowledge about this promising acylphloroglucinol antibiotic and its diverse functions in this review. Results: Rhodomyrtone shows nano to micromolar activities against a broad range of Gram-positive pathogens, including multidrug-resistant clinical isolates, and possesses a unique mechanism of action. It increases membrane fluidity and creates hyperfluid domains that attract membrane proteins prior to forming large membrane vesicles, effectively acting as a membrane protein trap. This mechanism affects a multitude of cellular processes, including cell division and cell wall synthesis. Additionally, rhodomyrtone reduces the expression of inflammatory cytokines, such as TNF-α, IL-17A, IL1β, and IL8. Generally showing low toxicity against mammalian cells, rhodomyrtone does inhibit the proliferation of cancer cell lines, such as epidermal carcinoma cells. The primary mechanism behind this activity appears to be the downregulation of adhesion kinases and growth factors. Furthermore, rhodomyrtone has shown antioxidant activity and displays cognitive effects, such as decreasing depressive symptoms in mice. Conclusions: Rhodomyrtone shows great promise as therapeutic agent, mostly for antibacterial but also for diverse other applications. Yet, bottlenecks such as resistance development and a better understanding of mammalian cell toxictiy demand careful assessment.
Collapse
Affiliation(s)
- Rupa Rani
- Division of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden
- Centre for Antibiotic Resistance Research in Gothenburg (CARe), 413 45 Gothenburg, Sweden
| | - Gabriela Marinho Righetto
- Division of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden
- Centre for Antibiotic Resistance Research in Gothenburg (CARe), 413 45 Gothenburg, Sweden
| | - Ann-Britt Schäfer
- Division of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden
- Centre for Antibiotic Resistance Research in Gothenburg (CARe), 413 45 Gothenburg, Sweden
| | - Michaela Wenzel
- Division of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden
- Centre for Antibiotic Resistance Research in Gothenburg (CARe), 413 45 Gothenburg, Sweden
| |
Collapse
|
27
|
Knapp BD, Shi H, Huang KC. Complex state transitions of the bacterial cell division protein FtsZ. Mol Biol Cell 2024; 35:ar130. [PMID: 39083352 PMCID: PMC11481701 DOI: 10.1091/mbc.e23-11-0446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
The key bacterial cell division protein FtsZ can adopt multiple conformations, and prevailing models suggest that transitions of FtsZ subunits from the closed to open state are necessary for filament formation and stability. Using all-atom molecular dynamics simulations, we analyzed state transitions of Staphylococcus aureus FtsZ as a monomer, dimer, and hexamer. We found that monomers can adopt intermediate states but preferentially adopt a closed state that is robust to forced reopening. Dimer subunits transitioned between open and closed states, and dimers with both subunits in the closed state remained highly stable, suggesting that open-state conformations are not necessary for filament formation. Mg2+ strongly stabilized the conformation of GTP-bound subunits and the dimer filament interface. Our hexamer simulations indicate that the plus end subunit preferentially closes and that other subunits can transition between states without affecting inter-subunit stability. We found that rather than being correlated with subunit opening, inter-subunit stability was strongly correlated with catalytic site interactions. By leveraging deep-learning models, we identified key intrasubunit interactions governing state transitions. Our findings suggest a greater range of possible monomer and filament states than previously considered and offer new insights into the nuanced interplay between subunit states and the critical role of nucleotide hydrolysis and Mg2+ in FtsZ filament dynamics.
Collapse
Affiliation(s)
| | - Handuo Shi
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
- Department of Bioengineering, Stanford University, Stanford, CA 94305
| | - Kerwyn Casey Huang
- Biophysics Program, Stanford University, Stanford, CA 94305
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
- Department of Bioengineering, Stanford University, Stanford, CA 94305
- Chan Zuckerberg Biohub, San Francisco, CA 94158
| |
Collapse
|
28
|
Pelech P, Navarro PP, Vettiger A, Chao LH, Allolio C. Stress-mediated growth determines E. coli division site morphogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612282. [PMID: 39314472 PMCID: PMC11419054 DOI: 10.1101/2024.09.11.612282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
In order to proliferate, bacteria must remodel their cell wall at the division site. The division process is driven by the enzymatic activity of peptidoglycan (PG) synthases and hydrolases around the constricting Z-ring. PG remodelling is reg-ulated by de-and re-crosslinking enzymes, and the directing constrictive force of the Z-ring. We introduce a model that is able to reproduce correctly the shape of the division site during the constriction and septation phase of E. coli . The model represents mechanochemical coupling within the mathematical framework of morphoelasticity. It contains only two parameters, associated with volumet-ric growth and PG remodelling, that are coupled to the mechanical stress in the bacterial wall. Different morphologies, corresponding either to mutant or wild type cells were recovered as a function of the remodeling parameter. In addition, a plausible range for the cell stiffness and turgor pressure was determined by comparing numerical simulations with bacterial cell lysis data.
Collapse
|
29
|
Ludwig K, Puls JS, Matos de Opitz CL, Innocenti P, Daniel JM, Bornikoel J, Arts M, Krannich S, Straetener J, Brajtenbach D, Henrichfreise B, Sass P, Mueller A, Martin NI, Brötz-Oesterhelt H, Kubitscheck U, Grein F, Schneider T. The Dual Mode of Antibacterial Action of the Synthetic Small Molecule DCAP Involves Lipid II Binding. J Am Chem Soc 2024; 146:24855-24862. [PMID: 39197836 PMCID: PMC11403595 DOI: 10.1021/jacs.4c05138] [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: 04/16/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024]
Abstract
The synthetic small molecule DCAP is a chemically well-characterized compound with antibiotic activity against Gram-positive and Gram-negative bacteria, including drug-resistant pathogens. Until now, its mechanism of action was proposed to rely exclusively on targeting the bacterial membrane, thereby causing membrane depolarization, and increasing membrane permeability (Eun et al. 2012, J. Am. Chem. Soc. 134 (28), 11322-11325; Hurley et al. 2015, ACS Med. Chem. Lett. 6, 466-471). Here, we show that the antibiotic activity of DCAP results from a dual mode of action that is more targeted and multifaceted than previously anticipated. Using microbiological and biochemical assays in combination with fluorescence microscopy, we provide evidence that DCAP interacts with undecaprenyl pyrophosphate-coupled cell envelope precursors, thereby blocking peptidoglycan biosynthesis and impairing cell division site organization. Our work discloses a concise model for the mode of action of DCAP which involves the binding to a specific target molecule to exert pleiotropic effects on cell wall biosynthetic and divisome machineries.
Collapse
Affiliation(s)
- Kevin
C. Ludwig
- Institute
for Pharmaceutical Microbiology, University of Bonn, University Hospital Bonn, Meckenheimer Allee 168, 53115 Bonn, Germany
- German
Center for Infection Research (DZIF), Partner
Site Bonn-Cologne, 53115 Bonn, Germany
| | - Jan-Samuel Puls
- Institute
for Pharmaceutical Microbiology, University of Bonn, University Hospital Bonn, Meckenheimer Allee 168, 53115 Bonn, Germany
| | - Cruz L. Matos de Opitz
- Department
of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology
& Infection Medicine, University of
Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany
| | - Paolo Innocenti
- Biological
Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, the Netherlands
| | - Jan-Martin Daniel
- Institute
for Pharmaceutical Microbiology, University of Bonn, University Hospital Bonn, Meckenheimer Allee 168, 53115 Bonn, Germany
- German
Center for Infection Research (DZIF), Partner
Site Bonn-Cologne, 53115 Bonn, Germany
| | - Jan Bornikoel
- Department
of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology
& Infection Medicine, University of
Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany
| | - Melina Arts
- Institute
for Pharmaceutical Microbiology, University of Bonn, University Hospital Bonn, Meckenheimer Allee 168, 53115 Bonn, Germany
| | - Sebastian Krannich
- Institute
for Pharmaceutical Microbiology, University of Bonn, University Hospital Bonn, Meckenheimer Allee 168, 53115 Bonn, Germany
| | - Jan Straetener
- Department
of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology
& Infection Medicine, University of
Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany
| | - Dominik Brajtenbach
- Clausius-Institute
for Physical and Theoretical Chemistry, University of Bonn, Wegelerstraße 12, 53115 Bonn, Germany
| | - Beate Henrichfreise
- Institute
for Pharmaceutical Microbiology, University of Bonn, University Hospital Bonn, Meckenheimer Allee 168, 53115 Bonn, Germany
| | - Peter Sass
- Department
of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology
& Infection Medicine, University of
Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany
- German Center
for Infection Research (DZIF), Partner Site
Tübingen, 72076 Tübingen, Germany
| | - Anna Mueller
- Institute
for Pharmaceutical Microbiology, University of Bonn, University Hospital Bonn, Meckenheimer Allee 168, 53115 Bonn, Germany
| | - Nathaniel I. Martin
- Biological
Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, the Netherlands
| | - Heike Brötz-Oesterhelt
- Department
of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology
& Infection Medicine, University of
Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany
- German Center
for Infection Research (DZIF), Partner Site
Tübingen, 72076 Tübingen, Germany
- Cluster
of Excellence “Controlling Microbes to Fight Infections”, University of Tübingen, 72076 Tübingen, Germany
| | - Ulrich Kubitscheck
- Clausius-Institute
for Physical and Theoretical Chemistry, University of Bonn, Wegelerstraße 12, 53115 Bonn, Germany
| | - Fabian Grein
- Institute
for Pharmaceutical Microbiology, University of Bonn, University Hospital Bonn, Meckenheimer Allee 168, 53115 Bonn, Germany
- German
Center for Infection Research (DZIF), Partner
Site Bonn-Cologne, 53115 Bonn, Germany
| | - Tanja Schneider
- Institute
for Pharmaceutical Microbiology, University of Bonn, University Hospital Bonn, Meckenheimer Allee 168, 53115 Bonn, Germany
- German
Center for Infection Research (DZIF), Partner
Site Bonn-Cologne, 53115 Bonn, Germany
| |
Collapse
|
30
|
Marrin ME, Foster MR, Santana CM, Choi Y, Jassal AS, Rancic SJ, Greenwald CR, Drucker MN, Feldman DT, Thrall ES. The translesion polymerase Pol Y1 is a constitutive component of the B. subtilis replication machinery. Nucleic Acids Res 2024; 52:9613-9629. [PMID: 39051562 PMCID: PMC11381352 DOI: 10.1093/nar/gkae637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024] Open
Abstract
Unrepaired DNA damage encountered by the cellular replication machinery can stall DNA replication, ultimately leading to cell death. In the DNA damage tolerance pathway translesion synthesis (TLS), replication stalling is alleviated by the recruitment of specialized polymerases to synthesize short stretches of DNA near a lesion. Although TLS promotes cell survival, most TLS polymerases are low-fidelity and must be tightly regulated to avoid harmful mutagenesis. The gram-negative bacterium Escherichia coli has served as the model organism for studies of the molecular mechanisms of bacterial TLS. However, it is poorly understood whether these same mechanisms apply to other bacteria. Here, we use in vivo single-molecule fluorescence microscopy to investigate the TLS polymerase Pol Y1 in the model gram-positive bacterium Bacillus subtilis. We find significant differences in the localization and dynamics of Pol Y1 in comparison to its E. coli homolog, Pol IV. Notably, Pol Y1 is constitutively enriched at or near sites of replication in the absence of DNA damage through interactions with the DnaN clamp; in contrast, Pol IV has been shown to be selectively enriched only upon replication stalling. These results suggest key differences in the roles and mechanisms of regulation of TLS polymerases across different bacterial species.
Collapse
Affiliation(s)
- McKayla E Marrin
- Department of Chemistry and Biochemistry, Fordham University, Bronx, NY 10458, USA
| | - Michael R Foster
- Department of Chemistry and Biochemistry, Fordham University, Bronx, NY 10458, USA
| | - Chloe M Santana
- Department of Chemistry and Biochemistry, Fordham University, Bronx, NY 10458, USA
| | - Yoonhee Choi
- Department of Chemistry and Biochemistry, Fordham University, Bronx, NY 10458, USA
| | - Avtar S Jassal
- Department of Chemistry and Biochemistry, Fordham University, Bronx, NY 10458, USA
| | - Sarah J Rancic
- Department of Chemistry and Biochemistry, Fordham University, Bronx, NY 10458, USA
| | - Carolyn R Greenwald
- Department of Chemistry and Biochemistry, Fordham University, Bronx, NY 10458, USA
| | - Madeline N Drucker
- Department of Chemistry and Biochemistry, Fordham University, Bronx, NY 10458, USA
| | - Denholm T Feldman
- Department of Chemistry and Biochemistry, Fordham University, Bronx, NY 10458, USA
| | - Elizabeth S Thrall
- Department of Chemistry and Biochemistry, Fordham University, Bronx, NY 10458, USA
| |
Collapse
|
31
|
Poddar SM, Chakraborty J, Gayathri P, Srinivasan R. Disruption of salt bridge interactions in the inter-domain cleft of the tubulin-like protein FtsZ of Escherichia coli makes cells sensitive to the cell division inhibitor PC190723. Cytoskeleton (Hoboken) 2024. [PMID: 39230425 DOI: 10.1002/cm.21924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 08/01/2024] [Accepted: 08/22/2024] [Indexed: 09/05/2024]
Abstract
FtsZ forms a ring-like assembly at the site of division in bacteria. It is the first protein involved in the formation of the divisome complex to split the cell into two halves, indicating its importance in bacterial cell division. FtsZ is an attractive target for developing new anti-microbial drugs to overcome the challenges of antibiotic resistance. The most potent inhibitor against FtsZ is PC190723, which is effective against all strains and species of Staphylococcus, including the methicillin- and multi-drug-resistant Staphylococcus aureus and strains of Bacillus. However, FtsZs from bacteria such as E. coli, Streptococcus, and Enterococcus were shown to be resistant to this inhibitor. In this study, we provide further evidence that the three pairwise bridging interactions, between residues S227 and G191, R307 and E198 and D299 and R202, between S7, S9, S10 β-strands and the H7 helix occlude the inhibitor from binding to E. coli FtsZ. We generated single, double and triple mutations to disrupt those bridges and tested the effectiveness of PC190723 directly on Z-ring assembly in vivo. Our results show that the disruption of S227-G191 and R307-E198 bridges render EcFtsZ highly sensitive to PC190723 for Z-ring assembly. Ectopic expression of the double mutants, FtsZ S227I R307V results in hypersensitivity of the susceptible E. coli imp4213 strain to PC190723. Our studies could further predict the effectiveness of PC190723 or its derivatives towards FtsZs of other bacterial genera.
Collapse
Affiliation(s)
- Sakshi Mahesh Poddar
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India
- Homi Bhabha National Institutes (HBNI), Training School Complex, Mumbai, India
| | | | - Pananghat Gayathri
- Biology, Indian Institute of Science Education and Research, Pune, India
| | - Ramanujam Srinivasan
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India
- Homi Bhabha National Institutes (HBNI), Training School Complex, Mumbai, India
| |
Collapse
|
32
|
Cui XH, Wei YC, Li XG, Qi XQ, Wu LF, Zhang WJ. N-terminus GTPase domain of the cytoskeleton protein FtsZ plays a critical role in its adaptation to high hydrostatic pressure. Front Microbiol 2024; 15:1441398. [PMID: 39220037 PMCID: PMC11362102 DOI: 10.3389/fmicb.2024.1441398] [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: 05/31/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
Studies in model microorganisms showed that cell division is highly vulnerable to high hydrostatic pressure (HHP). Disassembly of FtsZ filaments induced by HHP results in the failure of cell division and formation of filamentous cells in E. coli. The specific characteristics of FtsZ that allow for functional cell division in the deep-sea environments, especially in obligate piezophiles that grow exclusively under HHP condition, remain enigmatic. In this study, by using a self-developed HHP in-situ fixation apparatus, we investigated the effect of HHP on FtsZ by examining the subcellular localization of GFP-tagged FtsZ in vivo and the stability of FtsZ filament in vitro. We compared the pressure tolerance of FtsZ proteins from pressure-sensitive strain Shewanella oneidensis MR-1 (FtsZSo) and obligately piezophilic strain Shewanella benthica DB21MT-2 (FtsZSb). Our findings showed that, unlike FtsZSo, HHP hardly affected the Z-ring formation of FtsZSb, and filaments composed of FtsZSb were more stable after incubation under 50 MPa. By constructing chimeric and single amino acid mutated FtsZ proteins, we identified five residues in the N-terminal GTPase domain of FtsZSb whose mutation would impair the Z-ring formation under HHP conditions. Overall, these results demonstrate that FtsZ from the obligately piezophilic strain exhibits superior pressure tolerance than its homologue from shallow water species, both in vivo and in vitro. Differences in pressure tolerance of FtsZ are largely attributed to the N-terminal GTPase domain. This represents the first in-depth study of the adaptation of microbial cytoskeleton protein FtsZ to high hydrostatic pressure, which may provide insights into understanding the complex bioprocess of cell division under extreme environments.
Collapse
Affiliation(s)
- Xue-Hua Cui
- Laboratory of Deep-Sea Microbial Cell Biology, Institute of Deep-Sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
- College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yu-Chen Wei
- Laboratory of Deep-Sea Microbial Cell Biology, Institute of Deep-Sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
| | - Xue-Gong Li
- Laboratory of Deep-Sea Microbial Cell Biology, Institute of Deep-Sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
- Institution of Deep-Sea Life Sciences, IDSSE-BGI, Sanya, China
| | - Xiao-Qing Qi
- Laboratory of Deep-Sea Microbial Cell Biology, Institute of Deep-Sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
- Institution of Deep-Sea Life Sciences, IDSSE-BGI, Sanya, China
| | - Long-Fei Wu
- Laboratory of Deep-Sea Microbial Cell Biology, Institute of Deep-Sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
- Institution of Deep-Sea Life Sciences, IDSSE-BGI, Sanya, China
- Aix Marseille University, CNRS, LCB, Marseille, France
| | - Wei-Jia Zhang
- Laboratory of Deep-Sea Microbial Cell Biology, Institute of Deep-Sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
- Institution of Deep-Sea Life Sciences, IDSSE-BGI, Sanya, China
| |
Collapse
|
33
|
Vanhille-Campos C, Whitley KD, Radler P, Loose M, Holden S, Šarić A. Self-organization of mortal filaments and its role in bacterial division ring formation. NATURE PHYSICS 2024; 20:1670-1678. [PMID: 39416851 PMCID: PMC11473364 DOI: 10.1038/s41567-024-02597-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 06/27/2024] [Indexed: 10/19/2024]
Abstract
Filaments in the cell commonly treadmill. Driven by energy consumption, they grow on one end while shrinking on the other, causing filaments to appear motile even though individual proteins remain static. This process is characteristic of cytoskeletal filaments and leads to collective filament self-organization. Here we show that treadmilling drives filament nematic ordering by dissolving misaligned filaments. Taking the bacterial FtsZ protein involved in cell division as an example, we show that this mechanism aligns FtsZ filaments in vitro and drives the organization of the division ring in living Bacillus subtilis cells. We find that ordering via local dissolution also allows the system to quickly respond to chemical and geometrical biases in the cell, enabling us to quantitatively explain the ring formation dynamics in vivo. Beyond FtsZ and other cytoskeletal filaments, our study identifies a mechanism for self-organization via constant birth and death of energy-consuming filaments.
Collapse
Affiliation(s)
- Christian Vanhille-Campos
- Institute of Science and Technology Austria, Klosterneuburg, Austria
- Department of Physics and Astronomy, Institute for the Physics of Living Systems, University College London, London, UK
| | - Kevin D. Whitley
- Centre for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Philipp Radler
- Institute of Science and Technology Austria, Klosterneuburg, Austria
- Archaea Biology and Ecogenomics Unit, Department of Functional and Evolutionary Ecology, University of Vienna, Vienna, Austria
| | - Martin Loose
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Séamus Holden
- School of Life Sciences, The University of Warwick, Coventry, UK
| | - Anđela Šarić
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| |
Collapse
|
34
|
Joshi K, Wright CS, Biswas RR, Iyer-Biswas S. Architectural underpinnings of stochastic intergenerational homeostasis. Phys Rev E 2024; 110:024405. [PMID: 39295040 DOI: 10.1103/physreve.110.024405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 07/24/2024] [Indexed: 09/21/2024]
Abstract
Living systems are naturally complex and adaptive and offer unique insights into the strategies for achieving and sustaining stochastic homeostasis in different conditions. Here we focus on homeostasis in the context of stochastic growth and division of individual bacterial cells. We take advantage of high-precision long-term dynamical data that have recently been used to extract emergent simplicities and to articulate empirical intra- and intergenerational scaling laws governing these stochastic dynamics. From these data, we identify the core motif in the mechanistic coupling between division and growth, which naturally yields these precise rules, thus also bridging the intra- and intergenerational phenomenologies. By developing and utilizing techniques for solving a broad class of first-passage processes, we derive the exact analytic necessary and sufficient condition for sustaining stochastic intergenerational cell-size homeostasis within this framework. Furthermore, we provide predictions for the precision kinematics of cell-size homeostasis and the shape of the interdivision time distribution, which are compellingly borne out by the high-precision data. Taken together, these results provide insights into the functional architecture of control systems that yield robust yet flexible stochastic homeostasis.
Collapse
|
35
|
Velle KB, Swafford AJM, Garner E, Fritz-Laylin LK. Actin network evolution as a key driver of eukaryotic diversification. J Cell Sci 2024; 137:jcs261660. [PMID: 39120594 PMCID: PMC12050087 DOI: 10.1242/jcs.261660] [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] [Indexed: 08/10/2024] Open
Abstract
Eukaryotic cells have been evolving for billions of years, giving rise to wildly diverse cell forms and functions. Despite their variability, all eukaryotic cells share key hallmarks, including membrane-bound organelles, heavily regulated cytoskeletal networks and complex signaling cascades. Because the actin cytoskeleton interfaces with each of these features, understanding how it evolved and diversified across eukaryotic phyla is essential to understanding the evolution and diversification of eukaryotic cells themselves. Here, we discuss what we know about the origin and diversity of actin networks in terms of their compositions, structures and regulation, and how actin evolution contributes to the diversity of eukaryotic form and function.
Collapse
Affiliation(s)
- Katrina B. Velle
- Department of Biology, University of Massachusetts Dartmouth, Dartmouth, MA 02747, USA
| | | | - Ethan Garner
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | | |
Collapse
|
36
|
Sun N, Zhi Z, Xiao T, Deng X, He T, Dong W, Feng S, Chen S, Wong WL, Yuan W. The study of honokiol as a natural product-based antimicrobial agent and its potential interaction with FtsZ protein. Front Microbiol 2024; 15:1361508. [PMID: 39104591 PMCID: PMC11298477 DOI: 10.3389/fmicb.2024.1361508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 07/10/2024] [Indexed: 08/07/2024] Open
Abstract
Multidrug resistant bacteria have been a global health threat currently and frontline clinical treatments for these infections are very limited. To develop potent antibacterial agents with new bactericidal mechanisms is thus needed urgently to address this critical antibiotic resistance challenge. Natural products are a treasure of small molecules with high bioactive and low toxicity. In the present study, we demonstrated that a natural compound, honokiol, showed potent antibacterial activity against a number of Gram-positive bacteria including MRSA and VRE. Moreover, honokiol in combination with clinically used β-lactam antibiotics exhibits strong synergistic antimicrobial effects against drug-resistant S. aureus strains. Biochemical studies further reveal that honokiol may disrupt the GTPase activity, FtsZ polymerization, cell division. These biological impacts induced by honokiol may ultimately cause bacterial cell death. The in vivo antibacterial activity of honokiol against S. aureus infection was also verified with a biological model of G. mellonella larvae. The in vivo results support that honokiol is low toxic against the larvae and effectively increases the survival rate of the larvae infected with S. aureus. These findings demonstrate the potential of honokiol for further structural advancement as a new class of antibacterial agents with high potency against multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Ning Sun
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, China
| | - Ziling Zhi
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| | - Ting Xiao
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, China
| | - Xin Deng
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, China
| | - Tenghui He
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, China
| | - Wanyang Dong
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| | - Shuyi Feng
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, China
| | - Sisi Chen
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| | - Wing-Leung Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Wenchang Yuan
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
37
|
Takasawa T, Matsui T, Watanabe G, Kodera Y. Molecular dynamics simulations reveal differences in the conformational stability of FtsZs derived from Staphylococcus aureus and Bacillus subtilis. Sci Rep 2024; 14:16043. [PMID: 38992051 PMCID: PMC11239868 DOI: 10.1038/s41598-024-66763-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 07/03/2024] [Indexed: 07/13/2024] Open
Abstract
FtsZ is highly conserved among bacteria and plays an essential role in bacterial cell division. The tense conformation of FtsZ bound to GTP assembles into a straight filament via head-to-tail associations, and then the upper subunit of FtsZ hydrolyzes GTP bound to the lower FtsZ subunit. The subunit with GDP bound disassembles accompanied by a conformational change in the subunit from the tense to relaxed conformation. Although crystal structures of FtsZ derived from several bacterial species have been determined, the conformational change from the relaxed to tense conformation has only been observed in Staphylococcus aureus FtsZ (SaFtsZ). Recent cryo-electron microscopy analyses revealed the three-dimensional reconstruction of the protofilament, in which tense molecules assemble via head-to-tail associations. However, the lower resolution of the protofilament suggested that the flexibility of the FtsZ protomers between the relaxed and tense conformations caused them to form in less-strict alignments. Furthermore, this flexibility may also prevent FtsZs other than SaFtsZ from crystalizing in the tense conformation, suggesting that the flexibility of bacterial FtsZs differs. In this study, molecular dynamics simulations were performed using SaFtsZ and Bacillus subtilis FtsZ in several situations, which suggested that different features of the FtsZs affect their conformational stability.
Collapse
Affiliation(s)
- Taichi Takasawa
- Department of Physics, School of Science, Kitasato University, 1-15-1 Kitasato, Minami-Ku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Takashi Matsui
- Department of Physics, School of Science, Kitasato University, 1-15-1 Kitasato, Minami-Ku, Sagamihara, Kanagawa, 252-0373, Japan.
- Center for Disease Proteomics, School of Science, Kitasato University, 1-15-1 Kitasato, Minami-Ku, Sagamihara, Kanagawa, 252-0373, Japan.
| | - Go Watanabe
- Department of Data Science, School of Frontier Engineering, Kitasato University, 1-15-1 Kitasato, Minami-Ku, Sagamihara, Kanagawa, 252-0373, Japan.
- Kanagawa Institute of Industrial Science and Technology (KISTEC), 705-1 Shimoimaizumi, Ebina, Kanagawa, 243-0435, Japan.
| | - Yoshio Kodera
- Department of Physics, School of Science, Kitasato University, 1-15-1 Kitasato, Minami-Ku, Sagamihara, Kanagawa, 252-0373, Japan
- Center for Disease Proteomics, School of Science, Kitasato University, 1-15-1 Kitasato, Minami-Ku, Sagamihara, Kanagawa, 252-0373, Japan
| |
Collapse
|
38
|
Barrows JM, Talavera-Figueroa BK, Payne IP, Smith EL, Goley ED. GTPase activity regulates FtsZ ring positioning in Caulobacter crescentus. Mol Biol Cell 2024; 35:ar97. [PMID: 38758654 PMCID: PMC11244171 DOI: 10.1091/mbc.e23-09-0365] [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: 09/13/2023] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/19/2024] Open
Abstract
Bacterial cell division is crucial for replication and requires careful coordination via proteins collectively called the divisome. The tubulin-like GTPase FtsZ is the master regulator of this process and serves to recruit downstream divisome proteins and regulate their activities. Upon assembling at mid-cell, FtsZ exhibits treadmilling motion driven by GTP binding and hydrolysis. Treadmilling is proposed to play roles in Z-ring condensation and in distribution and regulation of peptidoglycan (PG) cell wall enzymes. FtsZ polymer superstructure and dynamics are central to its function, yet their regulation is incompletely understood. We addressed these gaps in knowledge by evaluating the contribution of GTPase activity to FtsZ's function in vitro and in Caulobacter crescentus cells. We observed that a lethal mutation that abrogates FtsZ GTP hydrolysis impacts FtsZ dynamics and Z-ring positioning, but not constriction. Aberrant Z-ring positioning was due to insensitivity to the FtsZ regulator MipZ when GTPase activity is reduced. Z-ring mislocalization resulted in DNA damage, likely due to constriction over the nucleoid. Collectively, our results indicate that GTP hydrolysis serves primarily to position the Z-ring at mid-cell in Caulobacter. Proper Z-ring localization is required for effective coordination with chromosome segregation to prevent DNA damage and ensure successful cell division.
Collapse
Affiliation(s)
- Jordan M. Barrows
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | | | - Isaac P. Payne
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Erika L. Smith
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Erin D. Goley
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
39
|
Foster AJ, van den Noort M, Poolman B. Bacterial cell volume regulation and the importance of cyclic di-AMP. Microbiol Mol Biol Rev 2024; 88:e0018123. [PMID: 38856222 PMCID: PMC11332354 DOI: 10.1128/mmbr.00181-23] [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] [Indexed: 06/11/2024] Open
Abstract
SUMMARYNucleotide-derived second messengers are present in all domains of life. In prokaryotes, most of their functionality is associated with general lifestyle and metabolic adaptations, often in response to environmental fluctuations of physical parameters. In the last two decades, cyclic di-AMP has emerged as an important signaling nucleotide in many prokaryotic lineages, including Firmicutes, Actinobacteria, and Cyanobacteria. Its importance is highlighted by the fact that both the lack and overproduction of cyclic di-AMP affect viability of prokaryotes that utilize cyclic di-AMP, and that it generates a strong innate immune response in eukaryotes. In bacteria that produce the second messenger, most molecular targets of cyclic di-AMP are associated with cell volume control. Besides, other evidence links the second messenger to cell wall remodeling, DNA damage repair, sporulation, central metabolism, and the regulation of glycogen turnover. In this review, we take a biochemical, quantitative approach to address the main cellular processes that are directly regulated by cyclic di-AMP and show that these processes are very connected and require regulation of a similar set of proteins to which cyclic di-AMP binds. Altogether, we argue that cyclic di-AMP is a master regulator of cell volume and that other cellular processes can be connected with cyclic di-AMP through this core function. We further highlight important directions in which the cyclic di-AMP field has to develop to gain a full understanding of the cyclic di-AMP signaling network and why some processes are regulated, while others are not.
Collapse
Affiliation(s)
- Alexander J. Foster
- Department of Biochemistry, Groningen Biomolecular Science and Biotechnology Institute, University of Groningen, Groningen, the Netherlands
| | - Marco van den Noort
- Department of Biochemistry, Groningen Biomolecular Science and Biotechnology Institute, University of Groningen, Groningen, the Netherlands
| | - Bert Poolman
- Department of Biochemistry, Groningen Biomolecular Science and Biotechnology Institute, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
40
|
Perez AJ, Lamanna MM, Bruce KE, Touraev MA, Page JE, Shaw SL, Tsui HCT, Winkler ME. Elongasome core proteins and class A PBP1a display zonal, processive movement at the midcell of Streptococcus pneumoniae. Proc Natl Acad Sci U S A 2024; 121:e2401831121. [PMID: 38875147 PMCID: PMC11194595 DOI: 10.1073/pnas.2401831121] [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: 02/07/2024] [Accepted: 05/02/2024] [Indexed: 06/16/2024] Open
Abstract
Ovoid-shaped bacteria, such as Streptococcus pneumoniae (pneumococcus), have two spatially separated peptidoglycan (PG) synthase nanomachines that locate zonally to the midcell of dividing cells. The septal PG synthase bPBP2x:FtsW closes the septum of dividing pneumococcal cells, whereas the elongasome located on the outer edge of the septal annulus synthesizes peripheral PG outward. We showed previously by sm-TIRFm that the septal PG synthase moves circumferentially at midcell, driven by PG synthesis and not by FtsZ treadmilling. The pneumococcal elongasome consists of the PG synthase bPBP2b:RodA, regulators MreC, MreD, and RodZ, but not MreB, and genetically associated proteins Class A aPBP1a and muramidase MpgA. Given its zonal location separate from FtsZ, it was of considerable interest to determine the dynamics of proteins in the pneumococcal elongasome. We found that bPBP2b, RodA, and MreC move circumferentially with the same velocities and durations at midcell, driven by PG synthesis. However, outside of the midcell zone, the majority of these elongasome proteins move diffusively over the entire surface of cells. Depletion of MreC resulted in loss of circumferential movement of bPBP2b, and bPBP2b and RodA require each other for localization and circumferential movement. Notably, a fraction of aPBP1a molecules also moved circumferentially at midcell with velocities similar to those of components of the core elongasome, but for shorter durations. Other aPBP1a molecules were static at midcell or diffusing over cell bodies. Last, MpgA displayed nonprocessive, subdiffusive motion that was largely confined to the midcell region and less frequently detected over the cell body.
Collapse
Affiliation(s)
- Amilcar J. Perez
- Department of Biology, Indiana University Bloomington, Bloomington, IN47405
| | - Melissa M. Lamanna
- Department of Biology, Indiana University Bloomington, Bloomington, IN47405
| | - Kevin E. Bruce
- Department of Biology, Indiana University Bloomington, Bloomington, IN47405
| | - Marc A. Touraev
- Department of Biology, Indiana University Bloomington, Bloomington, IN47405
| | - Julia E. Page
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Sidney L. Shaw
- Department of Biology, Indiana University Bloomington, Bloomington, IN47405
| | | | - Malcolm E. Winkler
- Department of Biology, Indiana University Bloomington, Bloomington, IN47405
| |
Collapse
|
41
|
Zhao J, Han X. Investigation of artificial cells containing the Par system for bacterial plasmid segregation and inheritance mimicry. Nat Commun 2024; 15:4956. [PMID: 38858376 PMCID: PMC11164925 DOI: 10.1038/s41467-024-49412-9] [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: 08/15/2023] [Accepted: 06/05/2024] [Indexed: 06/12/2024] Open
Abstract
A crucial step in life processes is the transfer of accurate and correct genetic material to offspring. During the construction of autonomous artificial cells, a very important step is the inheritance of genetic information in divided artificial cells. The ParMRC system, as one of the most representative systems for DNA segregation in bacteria, can be purified and reconstituted into GUVs to form artificial cells. In this study, we demonstrate that the eGFP gene is segregated into two poles by a ParM filament with ParR as the intermediate linker to bind ParM and parC-eGFP DNA in artificial cells. After the ParM filament splits, the cells are externally induced to divide into two daughter cells that contain parC-eGFP DNA by osmotic pressure and laser irradiation. Using a PURE system, we translate eGFP DNA into enhanced green fluorescent proteins in daughter cells, and bacterial plasmid segregation and inheritance are successfully mimicked in artificial cells. Our results could lead to the construction of more sophisticated artificial cells that can reproduce with genetic information.
Collapse
Affiliation(s)
- Jingjing Zhao
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Xiaojun Han
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China.
| |
Collapse
|
42
|
Li A, Zhang Y, Wan L, Peng R, Zhang X, Guo Q, Xu S, Qiao D, Zheng P, Li N, Zhu W, Pan Q. Coordination-Driven Self-Assembly of Metal Ion-Antisense Oligonucleotide Nanohybrids for Chronic Bacterial Infection Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:28041-28055. [PMID: 38767982 DOI: 10.1021/acsami.4c01453] [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: 05/22/2024]
Abstract
Bacterial infection poses a significant challenge to wound healing and skin regeneration, leading to substantial economic burdens on patients and society. Therefore, it is crucial to promptly explore and develop effective methodologies for bacterial infections. Herein, we propose a novel approach for synthesizing nanostructures based on antisense oligonucleotides (ASOs) through the coordination-driven self-assembly of Zn2+ with ASO molecules. This approach aims to provide effective synergistic therapy for chronic wound infections caused by Staphylococcus aureus (S. aureus). The resulting hybrid nanoparticles successfully preserve the structural integrity and biological functionalities of ASOs, demonstrating excellent ASO encapsulation efficiency and bioaccessibility. In vitro antibacterial experiments reveal that Zn-ASO NPs exhibit antimicrobial properties against Escherichia coli, Staphylococcus aureus, and Bacillus subtilis. This antibacterial ability is attributed to the high concentration of metal zinc ions and the generation of high levels of reactive oxygen species. Additionally, the ftsZ-ASO effectively inhibits the expression of the ftsZ gene, further enhancing the antimicrobial effect. In vivo antibacterial assays demonstrate that the Zn-ASO NPs promote optimal skin wound healing and exhibit favorable biocompatibility against S. aureus infections, resulting in a residual infected area of less than 8%. This combined antibacterial strategy, which integrates antisense gene therapy and metal-coordination-directed self-assembly, not only achieves synergistic and augmented antibacterial outcomes but also expands the horizons of ASO coordination chemistry. Moreover, it addresses the gap in the antimicrobial application of metal-coordination ASO self-assembly, thereby advancing the field of ASO-based therapeutic approaches.
Collapse
Affiliation(s)
- Anqi Li
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, Jiangxi, China
| | - Yan Zhang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, Jiangxi, China
| | - Li Wan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, Jiangxi, China
| | - Rujue Peng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, Jiangxi, China
| | - Xuan Zhang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, Jiangxi, China
| | - Qiuyan Guo
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, Jiangxi, China
| | - Shan Xu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, Jiangxi, China
| | - Dan Qiao
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, Jiangxi, China
| | - Pengwu Zheng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, Jiangxi, China
| | - Na Li
- Key Laboratory for Research and Utilization of Characteristic Biological Resources in Southern Yunnan, College of Biological and Agricultural Sciences, Honghe University, Mengzi 661199, China
| | - Wufu Zhu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, Jiangxi, China
| | - Qingshan Pan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, Jiangxi, China
| |
Collapse
|
43
|
Perez AJ, Xiao J. Stay on track - revelations of bacterial cell wall synthesis enzymes and things that go by single-molecule imaging. Curr Opin Microbiol 2024; 79:102490. [PMID: 38821027 PMCID: PMC11162910 DOI: 10.1016/j.mib.2024.102490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 06/02/2024]
Abstract
In this review, we explore the regulation of septal peptidoglycan (sPG) synthesis in bacterial cell division, a critical process for cell viability and proper morphology. Recent single-molecule imaging studies have revealed the processive movement of the FtsW:bPBP synthase complex along the septum, shedding light on the spatiotemporal dynamics of sPG synthases and their regulators. In diderm bacteria (E. coli and C. crescentus), the movement occurs at two distinct speeds, reflecting active synthesis or inactivity driven by FtsZ-treadmilling. In monoderm bacteria (B. subtilis, S. pneumoniae, and S. aureus), however, these enzymes exhibit only the active sPG-track-coupled processive movement. By comparing the dynamics of sPG synthases in these organisms and that of class-A penicillin-binding proteins in vivo and in vitro, we propose a unifying model for septal cell wall synthesis regulation across species, highlighting the roles of the sPG- and Z-tracks in orchestrating a robust bacterial cell wall constriction process.
Collapse
Affiliation(s)
- Amilcar J Perez
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jie Xiao
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
44
|
Chakraborty J, Poddar S, Dutta S, Bahulekar V, Harne S, Srinivasan R, Gayathri P. Dynamics of interdomain rotation facilitates FtsZ filament assembly. J Biol Chem 2024; 300:107336. [PMID: 38718863 PMCID: PMC11157280 DOI: 10.1016/j.jbc.2024.107336] [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: 03/09/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/31/2024] Open
Abstract
FtsZ, the tubulin homolog essential for bacterial cell division, assembles as the Z-ring at the division site, and directs peptidoglycan synthesis by treadmilling. It is unclear how FtsZ achieves kinetic polarity that drives treadmilling. To obtain insights into fundamental features of FtsZ assembly dynamics independent of peptidoglycan synthesis, we carried out structural and biochemical characterization of FtsZ from the cell wall-less bacteria, Spiroplasma melliferum (SmFtsZ). Interestingly the structures of SmFtsZ, bound to GDP and GMPPNP respectively, were captured as domain swapped dimers. SmFtsZ was found to be a slower GTPase with a higher critical concentration (CC) compared to Escherichia coli FtsZ (EcFtsZ). In FtsZs, a conformational switch from R-state (close) to T-state (open) favors polymerization. We identified that Phe224, located at the interdomain cleft of SmFtsZ, is crucial for R- to T-state transition. SmFtsZF224M exhibited higher GTPase activity and lower CC, whereas the corresponding EcFtsZM225F resulted in cell division defects in E. coli. Our results demonstrate that relative rotation of the domains is a rate-limiting step of polymerization. Our structural analysis suggests that the rotation is plausibly triggered upon addition of a GTP-bound monomer to the filament through interaction of the preformed N-terminal domain (NTD). Hence, addition of monomers to the NTD-exposed end of filament is slower in comparison to the C-terminal domain (CTD) end, thus explaining kinetic polarity. In summary, the study highlights the importance of interdomain interactions and conformational changes in regulating FtsZ assembly dynamics.
Collapse
Affiliation(s)
- Joyeeta Chakraborty
- Biology Division, Indian Institute of Science Education and Research, Pune, India
| | - Sakshi Poddar
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India; Homi Bhabha National Institutes (HBNI), Training School Complex, Mumbai, India
| | - Soumyajit Dutta
- Biology Division, Indian Institute of Science Education and Research, Pune, India
| | - Vaishnavi Bahulekar
- Biology Division, Indian Institute of Science Education and Research, Pune, India
| | - Shrikant Harne
- Biology Division, Indian Institute of Science Education and Research, Pune, India
| | - Ramanujam Srinivasan
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India; Homi Bhabha National Institutes (HBNI), Training School Complex, Mumbai, India
| | - Pananghat Gayathri
- Biology Division, Indian Institute of Science Education and Research, Pune, India.
| |
Collapse
|
45
|
Hale VL, Hooker J, Russo CJ, Löwe J. Honeycomb gold specimen supports enabling orthogonal focussed ion beam-milling of elongated cells for cryo-ET. J Struct Biol 2024; 216:108097. [PMID: 38772448 PMCID: PMC7616276 DOI: 10.1016/j.jsb.2024.108097] [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] [Revised: 05/11/2024] [Accepted: 05/18/2024] [Indexed: 05/23/2024]
Abstract
Cryo-focussed ion beam (FIB)-milling is a powerful technique that opens up thick, cellular specimens to high-resolution structural analysis by electron cryotomography (cryo-ET). FIB-milled lamellae can be produced from cells on grids, or cut from thicker, high-pressure frozen specimens. However, these approaches can put geometrical constraints on the specimen that may be unhelpful, particularly when imaging structures within the cell that have a very defined orientation. For example, plunge frozen rod-shaped bacteria orient parallel to the plane of the grid, yet the Z-ring, a filamentous structure of the tubulin-like protein FtsZ and the key organiser of bacterial division, runs around the circumference of the cell such that it is perpendicular to the imaging plane. It is therefore difficult or impractical to image many complete rings with current technologies. To circumvent this problem, we have fabricated monolithic gold specimen supports with a regular array of cylindrical wells in a honeycomb geometry, which trap bacteria in a vertical orientation. These supports, which we call "honeycomb gold discs", replace standard EM grids and when combined with FIB-milling enable the production of lamellae containing cross-sections through cells. The resulting lamellae are more stable and resistant to breakage and charging than conventional lamellae. The design of the honeycomb discs can be modified according to need and so will also enable cryo-ET and cryo-EM imaging of other specimens in otherwise difficult to obtain orientations.
Collapse
Affiliation(s)
| | - James Hooker
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | | | - Jan Löwe
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK.
| |
Collapse
|
46
|
Perez AJ, Lamanna MM, Bruce KE, Touraev MA, Page JE, Shaw SL, Tsui HCT, Winkler ME. Elongasome core proteins and class A PBP1a display zonal, processive movement at the midcell of Streptococcus pneumoniae. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.575112. [PMID: 38328058 PMCID: PMC10849506 DOI: 10.1101/2024.01.10.575112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Ovoid-shaped bacteria, such as Streptococcus pneumoniae (pneumococcus), have two spatially separated peptidoglycan (PG) synthase nanomachines that locate zonally to the midcell of dividing cells. The septal PG synthase bPBP2x:FtsW closes the septum of dividing pneumococcal cells, whereas the elongasome located on the outer edge of the septal annulus synthesizes peripheral PG outward. We showed previously by sm-TIRFm that the septal PG synthase moves circumferentially at midcell, driven by PG synthesis and not by FtsZ treadmilling. The pneumococcal elongasome consists of the PG synthase bPBP2b:RodA, regulators MreC, MreD, and RodZ, but not MreB, and genetically associated proteins Class A aPBP1a and muramidase MpgA. Given its zonal location separate from FtsZ, it was of considerable interest to determine the dynamics of proteins in the pneumococcal elongasome. We found that bPBP2b, RodA, and MreC move circumferentially with the same velocities and durations at midcell, driven by PG synthesis. However, outside of the midcell zone, the majority of these elongasome proteins move diffusively over the entire surface of cells. Depletion of MreC resulted in loss of circumferential movement of bPBP2b, and bPBP2b and RodA require each other for localization and circumferential movement. Notably, a fraction of aPBP1a molecules also moved circumferentially at midcell with velocities similar to those of components of the core elongasome, but for shorter durations. Other aPBP1a molecules were static at midcell or diffusing over cell bodies. Last, MpgA displayed non-processive, subdiffusive motion that was largely confined to the midcell region and less frequently detected over the cell body.
Collapse
|
47
|
Zheng Y, Zhu X, Jiang M, Cao F, You Q, Chen X. Development and Applications of D-Amino Acid Derivatives-based Metabolic Labeling of Bacterial Peptidoglycan. Angew Chem Int Ed Engl 2024; 63:e202319400. [PMID: 38284300 DOI: 10.1002/anie.202319400] [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: 12/18/2023] [Revised: 01/28/2024] [Accepted: 01/29/2024] [Indexed: 01/30/2024]
Abstract
Peptidoglycan, an essential component within the cell walls of virtually all bacteria, is composed of glycan strands linked by stem peptides that contain D-amino acids. The peptidoglycan biosynthesis machinery exhibits high tolerance to various D-amino acid derivatives. D-amino acid derivatives with different functionalities can thus be specifically incorporated into and label the peptidoglycan of bacteria, but not the host mammalian cells. This metabolic labeling strategy is highly selective, highly biocompatible, and broadly applicable, which has been utilized in various fields. This review introduces the metabolic labeling strategies of peptidoglycan by using D-amino acid derivatives, including one-step and two-step strategies. In addition, we emphasize the various applications of D-amino acid derivative-based metabolic labeling, including bacterial peptidoglycan visualization (existence, biosynthesis, and dynamics, etc.), bacterial visualization (including bacterial imaging and visualization of growth and division, metabolic activity, antibiotic susceptibility, etc.), pathogenic bacteria-targeted diagnostics and treatment (positron emission tomography (PET) imaging, photodynamic therapy, photothermal therapy, gas therapy, immunotherapy, etc.), and live bacteria-based therapy. Finally, a summary of this metabolic labeling and an outlook is provided.
Collapse
Affiliation(s)
- Yongfang Zheng
- Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Provincial Key Laboratory of Polymer Materials, College of Chemistry and Materials Science, Fujian Normal University, 32 Shangsan Road, Fuzhou, 350007, P.R. China
| | - Xinyu Zhu
- Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Provincial Key Laboratory of Polymer Materials, College of Chemistry and Materials Science, Fujian Normal University, 32 Shangsan Road, Fuzhou, 350007, P.R. China
| | - Mingyi Jiang
- Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Provincial Key Laboratory of Polymer Materials, College of Chemistry and Materials Science, Fujian Normal University, 32 Shangsan Road, Fuzhou, 350007, P.R. China
| | - Fangfang Cao
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Qing You
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| |
Collapse
|
48
|
Ocius KL, Kolli SH, Ahmad SS, Dressler JM, Chordia MD, Jutras BL, Rutkowski MR, Pires MM. Noninvasive Analysis of Peptidoglycan from Living Animals. Bioconjug Chem 2024; 35:489-498. [PMID: 38591251 PMCID: PMC11036361 DOI: 10.1021/acs.bioconjchem.4c00007] [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: 01/09/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/10/2024]
Abstract
The role of the intestinal microbiota in host health is increasingly revealed in its contributions to disease states. The host-microbiome interaction is multifactorial and dynamic. One of the factors that has recently been strongly associated with host physiological responses is peptidoglycan from bacterial cell walls. Peptidoglycan from gut commensal bacteria activates peptidoglycan sensors in human cells, including the nucleotide-binding oligomerization domain-containing protein 2. When present in the gastrointestinal tract, both the polymeric form (sacculi) and depolymerized fragments can modulate host physiology, including checkpoint anticancer therapy efficacy, body temperature and appetite, and postnatal growth. To utilize this growing area of biology toward therapeutic prescriptions, it will be critical to directly analyze a key feature of the host-microbiome interaction from living hosts in a reproducible and noninvasive way. Here we show that metabolically labeled peptidoglycan/sacculi can be readily isolated from fecal samples collected from both mice and humans. Analysis of fecal samples provided a noninvasive route to probe the gut commensal community including the metabolic synchronicity with the host circadian clock. Together, these results pave the way for noninvasive diagnostic tools to interrogate the causal nature of peptidoglycan in host health and disease.
Collapse
Affiliation(s)
- Karl L. Ocius
- Department
of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Sree H. Kolli
- Department
of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Saadman S. Ahmad
- Department
of Biochemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
- Fralin
Life Sciences Institute, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Jules M. Dressler
- Department
of Biochemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
- Fralin
Life Sciences Institute, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Mahendra D. Chordia
- Department
of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Brandon L. Jutras
- Department
of Biochemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
- Fralin
Life Sciences Institute, Virginia Tech, Blacksburg, Virginia 24061, United States
- Center
for Emerging, Zoonotic and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Melanie R. Rutkowski
- Department
of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Marcos M. Pires
- Department
of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| |
Collapse
|
49
|
Zhao X, Cao X, Qiu H, Liang W, Jiang Y, Wang Q, Wang W, Li C, Li Y, Han B, Tang K, Zhao L, Zhang X, Wang X, Liang H. Rational molecular design converting fascaplysin derivatives to potent broad-spectrum inhibitors against bacterial pathogens via targeting FtsZ. Eur J Med Chem 2024; 270:116347. [PMID: 38552428 DOI: 10.1016/j.ejmech.2024.116347] [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: 02/05/2024] [Revised: 03/06/2024] [Accepted: 03/17/2024] [Indexed: 04/21/2024]
Abstract
The filamentous temperature-sensitive mutant Z protein (FtsZ), a key player in bacterial cell division machinery, emerges as an attractive target to tackle the plight posed by the ever growing antibiotic resistance over the world. Therefore in this regard, agents with scaffold diversities and broad-spectrum antibacterial activity against Gram-positive and Gram-negative pathogens are highly needed. In this study, a new class of marine-derived fascaplysin derivatives has been designed and synthesized by Suzuki-Miyaura cross-coupling. Some compounds exhibited potent bactericidal activities against a panel of Gram-positive (MIC = 0.024-6.25 μg/mL) and Gram-negative (MIC = 1.56-12.5 μg/mL) bacteria including methicillin-resistant S. aureus (MRSA). They exerted their effects by dual action mechanism via disrupting the integrity of the bacterial cell membrane and targeting FtsZ protein. These compounds stimulated polymerization of FtsZ monomers and bundling of the polymers, and stabilized the resulting polymer network, thus leading to the dysfunction of FtsZ in cell division. In addition, these agents showed negligible hemolytic activity and low cytotoxicity to mammalian cells. The studies on docking and molecular dynamics simulations suggest that these inhibitors bind to the hydrophilic inter-domain cleft of FtsZ protein and the insights obtained in this study would facilitate the development of potential drugs with broad-spectrum bioactivities.
Collapse
Affiliation(s)
- Xing Zhao
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China; Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Xuanyu Cao
- Health Science Center, Ningbo University, Ningbo, 315211, China; Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315211, China
| | - Hongda Qiu
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Weida Liang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Yinli Jiang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Qiang Wang
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Weile Wang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Chengxi Li
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Yang Li
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315211, China
| | - Bowen Han
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315211, China
| | - Keqi Tang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Lingling Zhao
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Xuan Zhang
- Health Science Center, Ningbo University, Ningbo, 315211, China; Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315211, China.
| | - Xiao Wang
- Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Hongze Liang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
50
|
Cossart P, Hacker J, Holden DH, Normark S, Vogel J. Meeting report 'Microbiology 2023: from single cell to microbiome and host', an international interacademy conference in Würzburg. MICROLIFE 2024; 5:uqae008. [PMID: 38665235 PMCID: PMC11044969 DOI: 10.1093/femsml/uqae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024]
Abstract
On September 20-22 September 2023, the international conference 'Microbiology 2023: from single cell to microbiome and host' convened microbiologists from across the globe for a very successful symposium, showcasing cutting-edge research in the field. Invited lecturers delivered exceptional presentations covering a wide range of topics, with a major emphasis on phages and microbiomes, on the relevant bacteria within these ecosystems, and their multifaceted roles in diverse environments. Discussions also spanned the intricate analysis of fundamental bacterial processes, such as cell division, stress resistance, and interactions with phages. Organized by four renowned Academies, the German Leopoldina, the French Académie des sciences, the Royal Society UK, and the Royal Swedish Academy of Sciences, the symposium provided a dynamic platform for experts to share insights and discoveries, leaving participants inspired and eager to integrate new knowledge into their respective projects. The success of Microbiology 2023 prompted the decision to host the next quadrennial academic meeting in Sweden. This choice underscores the commitment to fostering international collaboration and advancing the frontiers of microbiological knowledge. The transition to Sweden promises to be an exciting step in the ongoing global dialogue and specific collaborations on microbiology, a field where researchers will continue to push the boundaries of knowledge, understanding, and innovation not only in health and disease but also in ecology.
Collapse
Affiliation(s)
| | - Jörg Hacker
- German National Academy of Science Leopoldina, Jägerberg 1, D-06108 Halle, Germany
| | - David H Holden
- Department of Infectious Disease, Centre for Bacterial Resistance Biology, Flowers Building, South Kensington Campus, Exhibition Road, Imperial College London, London SW7 2AZ, United Kingdom
| | - Staffan Normark
- Karolinska Institute, Tumor-och-cellbiologi, C1 Microbial Pathogenesis, 17177 Stockholm, Sweden
| | - Jörg Vogel
- Faculty of Medicine, Institute for Molecular Infection Biology (IMIB), University of Würzburg, D-97080 Würzburg, Germany
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Josef-Schneider-Str2/Gebaude D15; É. D-97080 Würzburg, Germany
| |
Collapse
|