1
|
Zhang P, Yan H, Liang Z, Zhang P, Li XH, Yuan XZ, Yu G, Wang W, Cai C. Synthesis of Fucoidan-Biomimetic Glycopolymers with Flexible Skeletons for Enhanced Anti-Herpes Virus Efficacy. ACS NANO 2025; 19:15411-15424. [PMID: 40243454 DOI: 10.1021/acsnano.4c15060] [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: 04/18/2025]
Abstract
Synthetic glycopolymers can be designed to mimic the structure and biological function of natural polysaccharides, offering a wide range of potential applications in the pharmaceutical and medicine. Nevertheless, amphiphilic synthetic glycopolymers commonly form biologically inert nanomicelle structures in aqueous solutions through spontaneous self-assembly. Envisioning that preventing self-assembly is pivotal to the full realization of the biological activities of the glycopolymers, we design and prepare a class of norbornene-derived hydrophilic glycopolymers containing sulfated fucose amenable to skeleton modification through ring-opening metathesis polymerization (ROMP). The skeleton of the fucoidan glycopolymers was chemically modified with hydrogen reduction, dihydroxylation, and oxidation following subsequent sulfation. We conducted physicochemical property characterization of the skeleton-modified glycopolymers to demonstrate that the hydrophilic glycopolymers have a more flexible structure compared to conventional polymers, and the sulfated fucoidan glycopolymers form a non-assembly morphology similar to the natural polysaccharides. Furthermore, the non-assembly glycopolymers exhibit significantly enhanced anti-HSV-1 activities. Our findings underscore the significance of the rational design of polymer skeletons in the development of structural and functional mimics of natural polysaccharides.
Collapse
Affiliation(s)
- Ping Zhang
- Key Laboratory of Marine Drugs, Ministry of Education & Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Han Yan
- Key Laboratory of Marine Drugs, Ministry of Education & Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Zhihe Liang
- Key Laboratory of Marine Drugs, Ministry of Education & Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Peng Zhang
- Key Laboratory of Marine Drugs, Ministry of Education & Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Xiao-Hua Li
- Shandong Key Laboratory of Water Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Shandong Universit, Qingdao 266237, China
| | - Xian-Zheng Yuan
- Shandong Key Laboratory of Water Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Shandong Universit, Qingdao 266237, China
| | - Guangli Yu
- Key Laboratory of Marine Drugs, Ministry of Education & Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266003, China
| | - Wei Wang
- Key Laboratory of Marine Drugs, Ministry of Education & Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266003, China
| | - Chao Cai
- Key Laboratory of Marine Drugs, Ministry of Education & Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266003, China
| |
Collapse
|
2
|
Rodriguez-Valverde D, Leon-Montes N, Belmont-Monroy L, Ruiz-Perez F, Santiago AE. Lipoprotein Lpp and L, D-transpeptidases regulate the master regulator of virulence AggR in EAEC. Sci Rep 2025; 15:13988. [PMID: 40263412 PMCID: PMC12015436 DOI: 10.1038/s41598-025-96373-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/27/2025] [Indexed: 04/24/2025] Open
Abstract
Enteroaggregative Escherichia coli (EAEC) is a diarrheagenic pathotype associated with traveler's diarrhea, foodborne outbreaks, and sporadic diarrhea in industrialized and developing countries. Regulation of virulence factors in EAEC is mediated by the master regulator AggR, an AraC/XylS family member controlling the expression of more than 44 genes associated with metabolism and virulence. Although the AggR regulon is well-characterized, the mechanism and upstream signaling cascades that regulate its activation are poorly understood. This study demonstrates that Lpp (Braun's lipoprotein) and L, D-transpeptidases are required for AggR activation. We found that deletion lpp in EAEC resulted in the downregulation of more than 100 genes involved in transport, metabolism, and virulence. Among the genes, fourteen transcriptional factors, including AggR, were differentially expressed in 042Δlpp. Our findings also showed that Lpp anchoring to the peptidoglycan is a requisite for AggR-activation. Hence, chemical inhibition or genetic deletion of L, D-transpeptidases encoding genes involved in the crosslink of Lpp to the peptidoglycan abolished AggR activation. Moreover, the 042Δlpp mutant exhibited reduced biofilm formation on abiotic surfaces and reduced colonization of human intestinal colonoids. This is the first study to demonstrate the tight regulation of the AraC/XylS transcriptional regulator AggR, essential in EAEC virulence and intestinal colonization by components of the bacterial cell envelope.
Collapse
Affiliation(s)
- Diana Rodriguez-Valverde
- University of Virginia, School of Medicine, Department of Pediatrics, Child Health Research Center, 409 Lane Road, MR-4 Building, P.O Box 801326, Charlottesville, VA, 22908, USA
| | - Nancy Leon-Montes
- University of Virginia, School of Medicine, Department of Pediatrics, Child Health Research Center, 409 Lane Road, MR-4 Building, P.O Box 801326, Charlottesville, VA, 22908, USA
| | - Laura Belmont-Monroy
- Laboratorio de Microbiología Molecular, Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Fernando Ruiz-Perez
- University of Virginia, School of Medicine, Department of Pediatrics, Child Health Research Center, 409 Lane Road, MR-4 Building, P.O Box 801326, Charlottesville, VA, 22908, USA
| | - Araceli E Santiago
- University of Virginia, School of Medicine, Department of Pediatrics, Child Health Research Center, 409 Lane Road, MR-4 Building, P.O Box 801326, Charlottesville, VA, 22908, USA.
| |
Collapse
|
3
|
Kato R, Miyazawa K, Imura T, Minamikawa T. Toward nanoscale structural and chemical analysis of microbial surfaces. Biosci Biotechnol Biochem 2025; 89:489-495. [PMID: 39577857 DOI: 10.1093/bbb/zbae176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
Microbial surfaces play a critical role in various biological processes, including cell adhesion and biofilm formation. Understanding these surfaces at the nanoscale is essential for both fundamental and applied microbiology. This review explores recent advancements in nanoscale structural and chemical analyses of microbial surfaces, with a focus on vibrational spectroscopy, such as Raman spectroscopy, infrared spectroscopy, and atomic force microscopy. The review also discusses current challenges of these techniques, including variability in sample preparation and the reproducibility of data, and highlights future directions in nanoscale analysis that could lead to new insights in microbial physiology, antimicrobial resistance, and biofilm research.
Collapse
Affiliation(s)
- Ryo Kato
- Department of Systems Innovation, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| | - Keisuke Miyazawa
- Faculty of Frontier Engineering, Kanazawa University, Kanazawa, Japan
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Takumi Imura
- Department of Systems Innovation, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| | - Takeo Minamikawa
- Department of Systems Innovation, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| |
Collapse
|
4
|
Ye G, Chen G, Avellán-Llaguno RD, Cao Y, Huang Q. Distinctive gut antibiotic resistome, potential health risks and underlying pathways upon cerebral ischemia-reperfusion injury. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 367:125614. [PMID: 39743194 DOI: 10.1016/j.envpol.2024.125614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/10/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Antibiotic resistance genes (ARGs) as emerging pollutants pose health risks to humans and the environment. Gut microbiota is an important reservoir for ARGs and hotspot for ARG acquisition and dissemination. Non-antibiotic factors (such as disease pathophysiology) affect ARG emergence and dissemination. Cerebral ischemia-reperfusion injury (I/R) commonly occurs in stroke patients. However, effects of I/R on ARG emergence and dissemination are unknown. Therefore, metagenomics was used to unveil selective collection of gut antibiotic resistome and its health risks, key ARG hosts and underlying pathways upon I/R. Changes in gut antibiotic resistome upon I/R were characterized by tetracycline ARG accumulation and decreases in aminoglycoside and glycopeptide ARGs. Besides, changes in gut antibiotic resistome were corrected with those in gut microbiota from phylum to species, serum lipid accumulation and glucose depletion upon I/R. Additionally, health risks of gut microbial multidrug ARGs (such as abem, adek and TolC), macA, aph(3')-I and carO, co-localized with mobile gene elements, were increased upon I/R. Moreover, phyla Firmicutes (especially order Eubacteriales, class Clostridia) and Bacteroidota were key ARG hosts in gut microbiota of I/R gerbils. Furthermore, suppression of vancomycin resistance, and lantibiotic biosynthesis and immunity, disturbances in peptidoglycan biosynthesis and hydrolysis, activation of antimicrobial peptide resistance, lipopolysaccharide biosynthesis, teichoic acid biosynthesis, arabinogalactan biosynthesis, aromatic compound degradation, oxidative phosphorylation, the tricarboxylic acid cycle and its anaplerotic pathways were observed in upon I/R. This study provides novel insights and intervention targets related to selective collection of gut antibiotic resistome and its potential health risks upon I/R.
Collapse
Affiliation(s)
- Guozhu Ye
- Xiamen Key Laboratory of Indoor Air and Health, Center for Excellence in Regional Atmospheric Environment, Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, 361021, China.
| | - Guoyou Chen
- College of Basic Medical Sciences, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Ricardo David Avellán-Llaguno
- Xiamen Key Laboratory of Indoor Air and Health, Center for Excellence in Regional Atmospheric Environment, Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, 361021, China
| | - Yonggang Cao
- College of Basic Medical Sciences, Harbin Medical University-Daqing, Daqing, 163319, China.
| | - Qiansheng Huang
- Xiamen Key Laboratory of Indoor Air and Health, Center for Excellence in Regional Atmospheric Environment, Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, 361021, China
| |
Collapse
|
5
|
Ganser C, Nishiguchi S, Chan FY, Uchihashi T. A look beyond topography: Transient phenomena of Escherichia coli cell division captured with high-speed in-line force mapping. SCIENCE ADVANCES 2025; 11:eads3010. [PMID: 39879298 PMCID: PMC11777186 DOI: 10.1126/sciadv.ads3010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025]
Abstract
Life on the nanoscale has been made accessible in recent decades by the development of fast and noninvasive techniques. High-speed atomic force microscopy (HS-AFM) is one such technique that shed light on single protein dynamics. Extending HS-AFM to effortlessly incorporate mechanical property mapping while maintaining fast imaging speed allows a look deeper than topography and reveal details of nanoscale mechanisms that govern life. Here, we present high-speed in-line force mapping (HS-iFM) to record mechanical properties and topography maps with high spatiotemporal resolution. Using HS-iFM, a comprehensive study of the nanoscale mechanical properties of living Escherichia coli revealed localized stiffening and details during cell division, formation and diffusion of pores in the membrane, and the impact of depressurization of a cell. The frame time was as low as 15 seconds with a spatial resolution of 5.5 nanometers per pixel in topography and 22 nanometers per pixel in force maps, allowing the capture of transient phenomena on bacterial surfaces in striking detail.
Collapse
Affiliation(s)
- Christian Ganser
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
| | - Shigetaka Nishiguchi
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
| | | | - Takayuki Uchihashi
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
- Department of Physics, Nagoya University, Nagoya, Japan
| |
Collapse
|
6
|
Cohen EJ, Drobnič T, Ribardo DA, Yoshioka A, Umrekar T, Guo X, Fernandez JJ, Brock EE, Wilson L, Nakane D, Hendrixson DR, Beeby M. Evolution of a large periplasmic disk in Campylobacterota flagella enables both efficient motility and autoagglutination. Dev Cell 2024; 59:3306-3321.e5. [PMID: 39362219 PMCID: PMC11652260 DOI: 10.1016/j.devcel.2024.09.008] [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: 01/07/2024] [Revised: 06/10/2024] [Accepted: 09/09/2024] [Indexed: 10/05/2024]
Abstract
The flagellar motors of Campylobacter jejuni (C. jejuni) and related Campylobacterota (previously epsilonproteobacteria) feature 100-nm-wide periplasmic "basal disks" that have been implicated in scaffolding a wider ring of additional motor proteins to increase torque, but the size of these disks is excessive for a role solely in scaffolding motor proteins. Here, we show that the basal disk is a flange that braces the flagellar motor during disentanglement of its flagellar filament from interactions with the cell body and other filaments. We show that motor output is unaffected when we shrink or displace the basal disk, and suppressor mutations of debilitated motors occur in flagellar-filament or cell-surface glycosylation pathways, thus sidestepping the need for a flange to overcome the interactions between two flagellar filaments and between flagellar filaments and the cell body. Our results identify unanticipated co-dependencies in the evolution of flagellar motor structure and cell-surface properties in the Campylobacterota.
Collapse
Affiliation(s)
- Eli J Cohen
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK.
| | - Tina Drobnič
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Deborah A Ribardo
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Aoba Yoshioka
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, Tokyo, Japan
| | - Trishant Umrekar
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Xuefei Guo
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Jose-Jesus Fernandez
- Spanish National Research Council (CINN-CSIC), Health Research Institute of Asturias (ISPA), Av Hospital Universitario s/n, Oviedo 33011, Spain
| | - Emma E Brock
- Department of Physics, School of Physics, Engineering and Technology, University of York, York YO10 5DD, UK
| | - Laurence Wilson
- Department of Physics, School of Physics, Engineering and Technology, University of York, York YO10 5DD, UK
| | - Daisuke Nakane
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, Tokyo, Japan
| | - David R Hendrixson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Morgan Beeby
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
7
|
Nguyen D, Bhattacharyya S, Richman H, Yu Y, Li Y. Targeting the Weak Spot: Preferential Disruption of Bacterial Poles by Janus Nanoparticles. NANO LETTERS 2024; 24:15886-15895. [PMID: 39584791 DOI: 10.1021/acs.nanolett.4c04946] [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: 11/26/2024]
Abstract
The interaction between nanoparticles (NPs) and bacterial cell envelopes is crucial for designing effective antibacterial materials against multi-drug-resistant pathogens. However, the current understanding assumes a uniform bacterial cell wall. This study challenges that assumption by investigating how bacterial cell wall curvature impacts antibacterial NP action. Focusing on Janus NPs, which feature segregated hydrophobic and polycationic ligands and previously demonstrated high efficacy against diverse bacteria, we found that these NPs preferentially target and disrupt bacterial poles. Experimental and computational approaches reveal that curvature at E. coli poles induces conformational changes in lipopolysaccharide (LPS) polymers on the outer membrane, exposing underlying lipids for NP-mediated disruption. We establish that curvature-induced targeting by Janus NPs depends on the outer membrane composition and is most pronounced at physiologically relevant LPS densities. This work demonstrates that high-curvature regions of bacterial cell walls are "weak spots" for Janus NPs, thereby aiding the development of more effective targeted therapies.
Collapse
Affiliation(s)
- Danh Nguyen
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Swagata Bhattacharyya
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Hunter Richman
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Yan Yu
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Ying Li
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
8
|
Benn G, Borrelli C, Prakaash D, Johnson ANT, Fideli VA, Starr T, Fitzmaurice D, Combs AN, Wühr M, Rojas ER, Khalid S, Hoogenboom BW, Silhavy TJ. OmpA controls order in the outer membrane and shares the mechanical load. Proc Natl Acad Sci U S A 2024; 121:e2416426121. [PMID: 39630873 DOI: 10.1073/pnas.2416426121] [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: 08/13/2024] [Accepted: 10/23/2024] [Indexed: 12/07/2024] Open
Abstract
OmpA, a predominant outer membrane (OM) protein in Escherichia coli, affects virulence, adhesion, and bacterial OM integrity. However, despite more than 50 y of research, the molecular basis for the role of OmpA has remained elusive. In this study, we demonstrate that OmpA organizes the OM protein lattice and mechanically connects it to the cell wall (CW). Using gene fusions, atomic force microscopy, simulations, and microfluidics, we show that the β-barrel domain of OmpA is critical for maintaining the permeability barrier, but both the β-barrel and CW-binding domains are necessary to enhance the cell envelope's strength. OmpA integrates the compressive properties of the OM protein lattice with the tensile strength of the CW, forming a mechanically robust composite that increases overall integrity. This coupling likely underpins the ability of the entire envelope to function as a cohesive, resilient structure, critical for the survival of bacteria.
Collapse
Affiliation(s)
- Georgina Benn
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540
| | - Carolina Borrelli
- London Centre for Nanotechnology, University College London, London WC1H 0AH, United Kingdom
- Department of Physics & Astronomy, University College London, London WC1E 6BT, United Kingdom
- Centre for Bacterial Resistance Biology, Imperial College London, London SW7 2AZ, United Kingdom
| | - Dheeraj Prakaash
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Alex N T Johnson
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08540
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08540
| | - Vincent A Fideli
- London Centre for Nanotechnology, University College London, London WC1H 0AH, United Kingdom
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom
| | - Tahj Starr
- Department of Biology, New York University, New York, NY 10003
| | | | - Ashton N Combs
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540
| | - Martin Wühr
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08540
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08540
| | - Enrique R Rojas
- Department of Biology, New York University, New York, NY 10003
| | - Syma Khalid
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Bart W Hoogenboom
- London Centre for Nanotechnology, University College London, London WC1H 0AH, United Kingdom
- Department of Physics & Astronomy, University College London, London WC1E 6BT, United Kingdom
| | - Thomas J Silhavy
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540
| |
Collapse
|
9
|
Szczepaniak J, Webby MN. The Tol Pal system integrates maintenance of the three layered cell envelope. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:46. [PMID: 39843782 PMCID: PMC11721397 DOI: 10.1038/s44259-024-00065-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/19/2024] [Indexed: 01/24/2025]
Abstract
The rapid emergence of antibiotic-resistant superbugs poses a significant global health threat. Gram-negative bacteria are the primary culprits due to their robust, tripartite cell envelope. This review explores the emerging role of the trans-envelope Tol-Pal system in maintaining envelope integrity, by connecting envelope layers and serving as a protein interaction hub. Targeting the Tol-Pal system offers a promising approach for the development of novel envelope-disrupting antimicrobials.
Collapse
Affiliation(s)
- Joanna Szczepaniak
- Department of Biochemistry, South Parks Road, University of Oxford, Oxford, OX1 3QU, UK
| | - Melissa N Webby
- Department of Biochemistry, South Parks Road, University of Oxford, Oxford, OX1 3QU, UK.
| |
Collapse
|
10
|
González MJ, Navarro N, Cruz E, Sánchez S, Morales JO, Zunino P, Robino L, Lima A, Scavone P. First report on the physicochemical and proteomic characterization of Proteus mirabilis outer membrane vesicles under urine-mimicking growth conditions: comparative analysis with Escherichia coli. Front Microbiol 2024; 15:1493859. [PMID: 39568990 PMCID: PMC11578119 DOI: 10.3389/fmicb.2024.1493859] [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: 09/09/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024] Open
Abstract
Introduction Uropathogenic bacteria employ multiple strategies to colonize the urinary tract, including biofilm formation, invasion of urothelial cells, and the production of adhesins, toxins, and siderophores. Among the most prevalent pathogens causing urinary tract infections (UTIs) are Uropathogenic Escherichia coli and Proteus mirabilis. A notable feature of Gram-negative bacteria is their ability to produce outer membrane vesicles (OMVs), which play critical roles in bacterial survival, virulence, and host-pathogen interactions, including UTIs. Methods In this study, OMVs were isolated and characterized from two clinical strains, E. coli U144 and P. mirabilis 2,921, cultured in both Luria-Bertani broth and artificial urine. Result and discussion The OMVs ranged in size from 85 to 260 nm, with the largest vesicles observed in artificial urine. Proteomic analysis allowed the identification of 282 proteins in OMVs from E. coli and 353 proteins from P. mirabilis when cultured LB medium, while 215 were identified from E. coli and 103 from P. mirabilis when cultured in artificial urine. The majority of these proteins originated from the bacterial envelope, while others were linked to motility and adhesion. Notably, the protein composition of OMVs varied depending on the growth medium, and proteins associated with zinc and iron uptake being more prominent in artificial urine, suggesting their importance in the urinary environment. Crucially, this is the first report to characterize P. mirabilis OMVs under different culture conditions, offering novel insights into the role of OMVs in UTI pathogenesis. These findings provide a deeper understanding of the molecular mechanisms by which OMVs contribute to bacterial virulence, establishing the foundation for potential therapeutic interventions targeting OMV-mediated processes in UTIs.
Collapse
Affiliation(s)
- María José González
- Laboratorio de Biofilms Microbianos, Departamento de Microbiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Nicolás Navarro
- Laboratorio de Biofilms Microbianos, Departamento de Microbiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Erlen Cruz
- Laboratorio de Biofilms Microbianos, Departamento de Microbiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Sofía Sánchez
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Center of New Drugs for Hypertension and Heart Failure (CENDHY), Santiago, Chile
| | - Javier O Morales
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Center of New Drugs for Hypertension and Heart Failure (CENDHY), Santiago, Chile
| | - Pablo Zunino
- Departamento de Microbiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Luciana Robino
- Unidad Académica de Bacteriología y Virología, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Analía Lima
- Unidad de Bioquímica y Proteómica Analíticas, Institut Pasteur de Montevideo & Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Paola Scavone
- Laboratorio de Biofilms Microbianos, Departamento de Microbiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| |
Collapse
|
11
|
Kaur M, Mozaheb N, Paiva TO, Herent MF, Goormaghtigh F, Paquot A, Terrasi R, Mignolet E, Décout JL, Lorent JH, Larondelle Y, Muccioli GG, Quetin-Leclercq J, Dufrêne YF, Mingeot-Leclercq MP. Insight into the outer membrane asymmetry of P. aeruginosa and the role of MlaA in modulating the lipidic composition, mechanical, biophysical, and functional membrane properties of the cell envelope. Microbiol Spectr 2024; 12:e0148424. [PMID: 39373473 PMCID: PMC11537012 DOI: 10.1128/spectrum.01484-24] [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: 07/10/2024] [Accepted: 08/14/2024] [Indexed: 10/08/2024] Open
Abstract
In Gram-negative bacteria, the outer membrane (OM) is asymmetric, with lipopolysaccharides (LPS) in the outer leaflet and glycerophospholipids (GPLs) in the inner leaflet. The asymmetry is maintained by the Mla system (MlaA-MlaBCDEF), which contributes to lipid homeostasis by removing mislocalized GPLs from the outer leaflet of the OM. Here, we ascribed how Pseudomonas aeruginosa ATCC 27853 coordinately regulates pathways to provide defense against the threats posed by the deletion of mlaA. Especially, we explored (i) the effects on membrane lipid composition including LPS, GPLs, and lysophospholipids, (ii) the biophysical properties of the OM such as stiffness and fluidity, and (iii) the impact of these changes on permeability, antibiotic susceptibility, and membrane vesicles (MVs) generation. Deletion of mlaA induced an increase in total GPLs and a decrease in LPS level while also triggering alterations in lipid A structures (arabinosylation and palmitoylation), likely to be induced by a two-component system (PhoPQ-PmrAB). Altered lipid composition may serve a physiological purpose in regulating the mechanobiological and functional properties of P. aeruginosa. We demonstrated an increase in cell stiffness without alteration of turgor pressure and inner membrane (IM) fluidity in ∆mlaA. In addition, membrane vesiculation increased without any change in OM/IM permeability. An amphiphilic aminoglycoside derivative (3',6-dinonyl neamine) that targets P. aeruginosa membranes induced an opposite effect on ∆mlaA strain with a trend toward a return to the situation observed for the WT strain. Efforts dedicated to understanding the crosstalk between the OM lipid composition, and the mechanical behavior of bacterial envelope, is one needed step for designing new targets or new drugs to fight P. aeruginosa infections.IMPORTANCEPseudomonas aeruginosa is a Gram-negative bacterium responsible for severe hospital-acquired infections. The outer membrane (OM) of Gram-negative bacteria acts as an effective barrier against toxic compounds, and therefore, compromising this structure could increase sensitivity to antibiotics. The OM is asymmetric with the highly packed lipopolysaccharide monolayer at the outer leaflet and glycerophospholipids at the inner leaflet. OM asymmetry is maintained by the Mla pathway resulting in the retrograde transport of glycerophospholipids from the OM to the inner membrane. In this study, we show that deleting mlaA, the membrane component of Mla system located at the OM, affects the mechanical and functional properties of P. aeruginosa cell envelope. Our results provide insights into the role of MlaA, involved in the Mla transport pathway in P. aeruginosa.
Collapse
Affiliation(s)
- M. Kaur
- UCLouvain, Louvain Drug Research Institute, Cellular & Molecular Pharmacology, Brussels, Belgium
| | - N. Mozaheb
- UCLouvain, Louvain Drug Research Institute, Cellular & Molecular Pharmacology, Brussels, Belgium
| | - T. O. Paiva
- UCLouvain, Louvain Institute of Biomolecular Science and Technology, nanoBiophysics, Louvain-la-Neuve, Belgium
| | - M.-F. Herent
- UCLouvain, Louvain Drug Research Institute, Pharmacognosy, Brussels, Belgium
| | - F. Goormaghtigh
- UCLouvain, Louvain Drug Research Institute, Cellular & Molecular Pharmacology, Brussels, Belgium
| | - A. Paquot
- UCLouvain, Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Brussels, Belgium
| | - R. Terrasi
- UCLouvain, Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Brussels, Belgium
| | - E. Mignolet
- UCLouvain, Louvain Institute of Biomolecular Science and Technology, Biochemistry of Nutrition and Environmental Toxicology Louvain-la-Neuve, Brussels, Belgium
| | - J.-L. Décout
- Université Grenoble Alpes, CNRS, DPM, Grenoble, France
| | - J. H. Lorent
- UCLouvain, Louvain Drug Research Institute, Cellular & Molecular Pharmacology, Brussels, Belgium
| | - Y. Larondelle
- UCLouvain, Louvain Institute of Biomolecular Science and Technology, Biochemistry of Nutrition and Environmental Toxicology Louvain-la-Neuve, Brussels, Belgium
| | - G. G. Muccioli
- UCLouvain, Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Brussels, Belgium
| | - J. Quetin-Leclercq
- UCLouvain, Louvain Drug Research Institute, Pharmacognosy, Brussels, Belgium
| | - Y. F. Dufrêne
- UCLouvain, Louvain Institute of Biomolecular Science and Technology, nanoBiophysics, Louvain-la-Neuve, Belgium
| | - M.-P. Mingeot-Leclercq
- UCLouvain, Louvain Drug Research Institute, Cellular & Molecular Pharmacology, Brussels, Belgium
| |
Collapse
|
12
|
Rachwalski K, Madden SJ, Ritchie N, French S, Bhando T, Girgis-Gabardo A, Tu M, Gordzevich R, Ives R, Guo AB, Johnson JW, Xu Y, Kapadia SB, Magolan J, Brown ED. A screen for cell envelope stress uncovers an inhibitor of prolipoprotein diacylglyceryl transferase, Lgt, in Escherichia coli. iScience 2024; 27:110894. [PMID: 39376497 PMCID: PMC11456916 DOI: 10.1016/j.isci.2024.110894] [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: 02/20/2024] [Revised: 07/25/2024] [Accepted: 09/03/2024] [Indexed: 10/09/2024] Open
Abstract
The increasing prevalence of antibiotic resistance demands the discovery of antibacterial chemical scaffolds with unique mechanisms of action. Phenotypic screening approaches, such as the use of reporters for bacterial cell stress, offer promise to identify compounds while providing strong hypotheses for follow-on mechanism of action studies. From a collection of ∼1,800 Escherichia coli GFP transcriptional reporter strains, we identified a reporter that is highly induced by cell envelope stress-pProm rcsA -GFP. After characterizing pProm rcsA -GFP induction, we assessed a collection of bioactive small molecules for reporter induction, identifying 24 compounds of interest. Spontaneous suppressors to one compound in particular, MAC-0452936, mapped to the gene encoding the essential prolipoprotein diacylglyceryl transferase, lgt. Lgt inhibition by MAC-0452936 inhibition was confirmed through genetic, phenotypic, and biochemical approaches. The oxime ester, MAC-0452936, represents a useful small molecule inhibitor of Lgt and highlights the potential of using pProm rcsA -GFP as a phenotypic screening tool.
Collapse
Affiliation(s)
- Kenneth Rachwalski
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Sean J. Madden
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Nicole Ritchie
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Shawn French
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Timsy Bhando
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Adele Girgis-Gabardo
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Megan Tu
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Rodion Gordzevich
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Rowan Ives
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Amelia B.Y. Guo
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Jarrod W. Johnson
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Yiming Xu
- Department of Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA, USA
| | | | - Jakob Magolan
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Eric D. Brown
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
13
|
Liang Y, Hugonnet JE, Rusconi F, Arthur M. Peptidoglycan-tethered and free forms of the Braun lipoprotein are in dynamic equilibrium in Escherichia coli. eLife 2024; 12:RP91598. [PMID: 39360705 PMCID: PMC11449479 DOI: 10.7554/elife.91598] [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: 10/04/2024] Open
Abstract
Peptidoglycan (PG) is a giant macromolecule that completely surrounds bacterial cells and prevents lysis in hypo-osmotic environments. This net-like macromolecule is made of glycan strands linked to each other by two types of transpeptidases that form either 4→3 (PBPs) or 3→3 (LDTs) cross-links. Previously, we devised a heavy isotope-based PG full labeling method coupled to mass spectrometry to determine the mode of insertion of new subunits into the expanding PG network (Atze et al., 2022). We showed that PG polymerization operates according to different modes for the formation of the septum and of the lateral cell walls, as well as for bacterial growth in the presence or absence of β-lactams in engineered strains that can exclusively rely on LDTs for PG cross-linking when drugs are present. Here, we apply our method to the resolution of the kinetics of the reactions leading to the covalent tethering of the Braun lipoprotein (Lpp) to PG and the subsequent hydrolysis of that same covalent link. We find that Lpp and disaccharide-peptide subunits are independently incorporated into the expanding lateral cell walls. Newly synthesized septum PG appears to contain small amounts of tethered Lpp. LDTs did mediate intense shuffling of Lpp between PG stems leading to a dynamic equilibrium between the PG-tethered and free forms of Lpp.
Collapse
Affiliation(s)
- Yucheng Liang
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, Paris, France
| | - Jean-Emmanuel Hugonnet
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, Paris, France
| | - Filippo Rusconi
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, Paris, France
- GQE-Le Moulon/PA, Université Paris-Saclay, INRAE, CNRS, AgroParisTech, IDEEV, Gif-sur-Yvette, France
| | - Michel Arthur
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, Paris, France
| |
Collapse
|
14
|
Zhu M, Kawamoto J, Imai T, Ogawa T, Kurihara T. Enhancing extracellular membrane vesicle productivity of Shewanella vesiculosa HM13, a prospective host for vesiculation-mediated protein secretion, by weakening outer membrane-peptidoglycan linkage. J Biosci Bioeng 2024; 138:137-143. [PMID: 38796341 DOI: 10.1016/j.jbiosc.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/19/2024] [Accepted: 05/09/2024] [Indexed: 05/28/2024]
Abstract
Shewanella vesiculosa HM13, a psychrotrophic gram-negative bacterium isolated from the intestinal contents of horse mackerel, produces abundant extracellular membrane vesicles (EMVs) by budding the outer membrane. The EMVs of this bacterium carry a single major cargo protein, P49, of unknown function, which may be useful as a carrier for the secretory production of heterologous proteins as cargoes of EMVs. In this study, to increase the utility of S. vesiculosa HM13 as a host for EMV-mediated protein production, we improved its EMV productivity by weakening the linkage between the outer membrane and underlying peptidoglycan layer. In gram-negative bacteria, the outer membrane is connected to peptidoglycans predominantly through Braun's lipoprotein (Lpp), and the formation of this linkage is catalyzed by an l,d-transpeptidase (Ldt). We constructed gene-disrupted mutants of Lpp and Ldt and assessed their EMV productivity. The EMVs of the lpp- and ldt-disrupted mutants grown at 18 °C were evaluated using nanoparticle tracking analysis, and their morphologies were observed using transmission electron microscopy. As a result, an approximately 2.5-fold increase in EMV production was achieved, whereas the morphology of the EMVs of these mutants remained almost identical to that of the parent strain. In accordance with the increase in EMV production, the mutants secreted approximately 2-fold higher amounts of P49 than the parent strain into the culture broth as the EMV cargo. These findings will contribute to the development of an EMV-based secretory production system for heterologous proteins using S. vesiculosa HM13 as a host.
Collapse
Affiliation(s)
- Mengshan Zhu
- Institute for Chemical Research, Kyoto University, Gokasho, Uji, Kyoto 611-0011, Japan
| | - Jun Kawamoto
- Institute for Chemical Research, Kyoto University, Gokasho, Uji, Kyoto 611-0011, Japan
| | - Tomoya Imai
- Research Institute for Sustainable Humanosphere, Kyoto University, Gokasho, Uji, Kyoto 611-0011, Japan
| | - Takuya Ogawa
- Institute for Chemical Research, Kyoto University, Gokasho, Uji, Kyoto 611-0011, Japan
| | - Tatsuo Kurihara
- Institute for Chemical Research, Kyoto University, Gokasho, Uji, Kyoto 611-0011, Japan.
| |
Collapse
|
15
|
Pontejo SM, Martinez S, Zhao A, Barnes K, de Anda J, Alimohamadi H, Lee EY, Dishman AF, Volkman BF, Wong GC, Garboczi DN, Ballesteros A, Murphy PM. Chemokines Kill Bacteria by Binding Anionic Phospholipids without Triggering Antimicrobial Resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.25.604863. [PMID: 39091850 PMCID: PMC11291121 DOI: 10.1101/2024.07.25.604863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Classically, chemokines coordinate leukocyte trafficking during immune responses; however, many chemokines have also been reported to possess direct antibacterial activity in vitro. Yet, the bacterial killing mechanism of chemokines and the biochemical properties that define which members of the chemokine superfamily are antimicrobial remain poorly understood. Here we report that the antimicrobial activity of chemokines is defined by their ability to bind phosphatidylglycerol and cardiolipin, two anionic phospholipids commonly found in the bacterial plasma membrane. We show that only chemokines able to bind these two phospholipids kill Escherichia coli and Staphylococcus aureus and that they exert rapid bacteriostatic and bactericidal effects against E. coli with a higher potency than the antimicrobial peptide beta-defensin 3. Furthermore, our data support that bacterial membrane cardiolipin facilitates the antimicrobial action of chemokines. Both biochemical and genetic interference with the chemokine-cardiolipin interaction impaired microbial growth arrest, bacterial killing, and membrane disruption by chemokines. Moreover, unlike conventional antibiotics, E. coli failed to develop resistance when placed under increasing antimicrobial chemokine pressure in vitro. Thus, we have identified cardiolipin and phosphatidylglycerol as novel binding partners for chemokines responsible for chemokine antimicrobial action. Our results provide proof of principle for developing chemokines as novel antibiotics resistant to bacterial antimicrobial resistance mechanisms.
Collapse
Affiliation(s)
- Sergio M. Pontejo
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sophia Martinez
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Allison Zhao
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kevin Barnes
- Structural Biology Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jaime de Anda
- Department of Bioengineering, University of California Los Angeles, Los Angeles, California, USA
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, CA, USA
- Department of Microbiology, Immunology & Molecular Genetics, University of California Los Angeles, Los Angeles, CA, USA
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Haleh Alimohamadi
- Department of Bioengineering, University of California Los Angeles, Los Angeles, California, USA
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, CA, USA
- Department of Microbiology, Immunology & Molecular Genetics, University of California Los Angeles, Los Angeles, CA, USA
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Ernest Y. Lee
- Department of Bioengineering, University of California Los Angeles, Los Angeles, California, USA
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, CA, USA
- Department of Microbiology, Immunology & Molecular Genetics, University of California Los Angeles, Los Angeles, CA, USA
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Acacia F. Dishman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Brian F. Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Gerard C.L. Wong
- Department of Bioengineering, University of California Los Angeles, Los Angeles, California, USA
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, CA, USA
- Department of Microbiology, Immunology & Molecular Genetics, University of California Los Angeles, Los Angeles, CA, USA
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - David N. Garboczi
- Structural Biology Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Angela Ballesteros
- Section of Sensory Physiology and Biophysics, National Institute of Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Philip M. Murphy
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
16
|
Dorrazehi GM, Winkle M, Desmet M, Stroobant V, Tanriver G, Degand H, Evrard D, Desguin B, Morsomme P, Biboy J, Gray J, Mitusińska K, Góra A, Vollmer W, Soumillion P. PBP-A, a cyanobacterial DD-peptidase with high specificity for amidated muropeptides, exhibits pH-dependent promiscuous activity harmful to Escherichia coli. Sci Rep 2024; 14:13999. [PMID: 38890528 PMCID: PMC11189452 DOI: 10.1038/s41598-024-64806-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: 02/22/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024] Open
Abstract
Penicillin binding proteins (PBPs) are involved in biosynthesis, remodeling and recycling of peptidoglycan (PG) in bacteria. PBP-A from Thermosynechococcus elongatus belongs to a cyanobacterial family of enzymes sharing close structural and phylogenetic proximity to class A β-lactamases. With the long-term aim of converting PBP-A into a β-lactamase by directed evolution, we simulated what may happen when an organism like Escherichia coli acquires such a new PBP and observed growth defect associated with the enzyme activity. To further explore the molecular origins of this harmful effect, we decided to characterize deeper the activity of PBP-A both in vitro and in vivo. We found that PBP-A is an enzyme endowed with DD-carboxypeptidase and DD-endopeptidase activities, featuring high specificity towards muropeptides amidated on the D-iso-glutamyl residue. We also show that a low promiscuous activity on non-amidated peptidoglycan deteriorates E. coli's envelope, which is much higher under acidic conditions where substrate discrimination is mitigated. Besides expanding our knowledge of the biochemical activity of PBP-A, this work also highlights that promiscuity may depend on environmental conditions and how it may hinder rather than promote enzyme evolution in nature or in the laboratory.
Collapse
Affiliation(s)
- Gol Mohammad Dorrazehi
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Place Croix du Sud 4-5, 1348, Louvain-la-Neuve, Belgium
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Matthias Winkle
- Centre for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Richardson Road, Newcastle upon Tyne, NE2 4AX, UK
- Benchmark Animal Health Ltd, 1 Pioneer Building, Edinburgh Technopole, Milton Bridge, Penicuik, EH26 0GB, UK
| | - Martin Desmet
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Place Croix du Sud 4-5, 1348, Louvain-la-Neuve, Belgium
| | - Vincent Stroobant
- Ludwig Institute for Cancer Research, Brussels, Belgium
- de Duve Institute, UCLouvain, Brussels, Belgium
| | - Gamze Tanriver
- Tunneling Group, Biotechnology Centre, Silesian University of Technology, 44-100, Gliwice, Poland
| | - Hervé Degand
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Place Croix du Sud 4-5, 1348, Louvain-la-Neuve, Belgium
| | - Damien Evrard
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Place Croix du Sud 4-5, 1348, Louvain-la-Neuve, Belgium
| | - Benoît Desguin
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Place Croix du Sud 4-5, 1348, Louvain-la-Neuve, Belgium
| | - Pierre Morsomme
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Place Croix du Sud 4-5, 1348, Louvain-la-Neuve, Belgium
| | - Jacob Biboy
- Centre for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Richardson Road, Newcastle upon Tyne, NE2 4AX, UK
| | - Joe Gray
- Biosciences Institute, Newcastle University, Richardson Road, Newcastle upon Tyne, NE2 4AX, UK
| | - Karolina Mitusińska
- Tunneling Group, Biotechnology Centre, Silesian University of Technology, 44-100, Gliwice, Poland
| | - Artur Góra
- Tunneling Group, Biotechnology Centre, Silesian University of Technology, 44-100, Gliwice, Poland
| | - Waldemar Vollmer
- Centre for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Richardson Road, Newcastle upon Tyne, NE2 4AX, UK
| | - Patrice Soumillion
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Place Croix du Sud 4-5, 1348, Louvain-la-Neuve, Belgium.
| |
Collapse
|
17
|
Ryoo D, Hwang H, Gumbart JC. Thicket and Mesh: How the Outer Membrane Can Resist Tension Imposed by the Cell Wall. J Phys Chem B 2024; 128:5371-5377. [PMID: 38787347 PMCID: PMC11163421 DOI: 10.1021/acs.jpcb.3c08510] [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: 01/02/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
The cell envelope of Gram-negative bacteria is composed of an outer membrane (OM) and an inner membrane (IM) and a peptidoglycan cell wall (CW) between them. Combined with Braun's lipoprotein (Lpp), which connects the OM and the CW, and numerous membrane proteins that exist in both OM and IM, the cell envelope creates a mechanically stable environment that resists various physical and chemical perturbations to the cell, including turgor pressure caused by the solute concentration difference between the cytoplasm of the cell and the extracellular environment. Previous computational studies have explored how individual components (OM, IM, and CW) can resist turgor pressure although combinations of them have been less well studied. To that end, we constructed multiple OM-CW systems, including the Lpp connections with the CW under increasing degrees of strain. The results show that the OM can effectively resist the tension imposed by the CW, shrinking by only 3-5% in area even when the CW is stretched to 2.5× its relaxed area. The area expansion modulus of the system increases with increasing CW strain, although the OM remains a significant contributor to the envelope's mechanical stability. Additionally, we find that when the protein TolC is embedded in the OM, its stiffness increases.
Collapse
Affiliation(s)
- David Ryoo
- Interdisciplinary
Bioengineering Graduate Program, Georgia
Institute of Technology, Atlanta, Georgia 30332, United States
| | - Hyea Hwang
- School
of Materials Science and Engineering, Georgia
Institute of Technology, Atlanta, Georgia 30332, United States
| | - James C. Gumbart
- School
of Physics, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
18
|
Zhu S, Alexander MK, Paiva TO, Rachwalski K, Miu A, Xu Y, Verma V, Reichelt M, Dufrêne YF, Brown ED, Cox G. The inactivation of tolC sensitizes Escherichia coli to perturbations in lipopolysaccharide transport. iScience 2024; 27:109592. [PMID: 38628966 PMCID: PMC11019271 DOI: 10.1016/j.isci.2024.109592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/02/2024] [Accepted: 03/25/2024] [Indexed: 04/19/2024] Open
Abstract
The Escherichia coli outer membrane channel TolC complexes with several inner membrane efflux pumps to export compounds across the cell envelope. All components of these complexes are essential for robust efflux activity, yet E. coli is more sensitive to antimicrobial compounds when tolC is inactivated compared to the inactivation of genes encoding the inner membrane drug efflux pumps. While investigating these susceptibility differences, we identified a distinct class of inhibitors targeting the core-lipopolysaccharide translocase, MsbA. We show that tolC null mutants are sensitized to structurally unrelated MsbA inhibitors and msbA knockdown, highlighting a synthetic-sick interaction. Phenotypic profiling revealed that tolC inactivation induced cell envelope softening and increased outer membrane permeability. Overall, this work identified a chemical probe of MsbA, revealed that tolC is associated with cell envelope mechanics and integrity, and highlighted that these findings should be considered when using tolC null mutants to study efflux deficiency.
Collapse
Affiliation(s)
- Shawna Zhu
- College of Biological Sciences, Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Road E, Guelph, ON N1G 2W1, Canada
| | | | - Telmo O. Paiva
- Institute of Life Sciences, UCLouvain, Croix du Sud, 4-5, bte L7.07.06, B-1348 Louvain-la-Neuve, Belgium
| | - Kenneth Rachwalski
- Biochemistry and Biomedical Sciences and Degroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Anh Miu
- Genentech Inc, Biochemical and Cellular Pharmacology, South San Francisco, CA, USA
| | - Yiming Xu
- Genentech Inc, Infectious Diseases, South San Francisco, CA, USA
| | - Vishal Verma
- Genentech Inc, Discovery Chemistry, South San Francisco, CA, USA
| | - Mike Reichelt
- Genentech Inc, Pathology, South San Francisco, CA, USA
| | - Yves F. Dufrêne
- Institute of Life Sciences, UCLouvain, Croix du Sud, 4-5, bte L7.07.06, B-1348 Louvain-la-Neuve, Belgium
| | - Eric D. Brown
- Biochemistry and Biomedical Sciences and Degroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Georgina Cox
- College of Biological Sciences, Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Road E, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
19
|
Muñoz-Echeverri LM, Benavides-López S, Geiger O, Trujillo-Roldán MA, Valdez-Cruz NA. Bacterial extracellular vesicles: biotechnological perspective for enhanced productivity. World J Microbiol Biotechnol 2024; 40:174. [PMID: 38642254 PMCID: PMC11032300 DOI: 10.1007/s11274-024-03963-7] [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: 01/17/2024] [Accepted: 03/19/2024] [Indexed: 04/22/2024]
Abstract
Bacterial extracellular vesicles (BEVs) are non-replicative nanostructures released by Gram-negative and Gram-positive bacteria as a survival mechanism and inter- and intraspecific communication mechanism. Due to BEVs physical, biochemical, and biofunctional characteristics, there is interest in producing and using them in developing new therapeutics, vaccines, or delivery systems. However, BEV release is typically low, limiting their application. Here, we provide a biotechnological perspective to enhance BEV production, highlighting current strategies. The strategies include the production of hypervesiculating strains through gene modification, bacteria culture under stress conditions, and artificial vesicles production. We discussed the effect of these production strategies on BEVs types, morphology, composition, and activity. Furthermore, we summarized general aspects of BEV biogenesis, functional capabilities, and applications, framing their current importance and the need to produce them in abundance. This review will expand the knowledge about the range of strategies associated with BEV bioprocesses to increase their productivity and extend their application possibilities.
Collapse
Affiliation(s)
- Laura M Muñoz-Echeverri
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México AP. 70228, Ciudad de México, C.P. 04510, México
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Unidad de Posgrado, Edificio D, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán CDMX, C.P. 04510, México
| | - Santiago Benavides-López
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México AP. 70228, Ciudad de México, C.P. 04510, México
- Posgrado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Unidad de Posgrado, Edificio B, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán CDMX, C.P. 04510, México
| | - Otto Geiger
- Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Av. Universidad s/n, Cuernavaca, Morelos, CP 62210, México
| | - Mauricio A Trujillo-Roldán
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México AP. 70228, Ciudad de México, C.P. 04510, México
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 Carretera, Tijuana-Ensenada, Baja California, 22860, México
| | - Norma A Valdez-Cruz
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México AP. 70228, Ciudad de México, C.P. 04510, México.
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 Carretera, Tijuana-Ensenada, Baja California, 22860, México.
| |
Collapse
|
20
|
Rais A, Sharma S, Mishra P, Khan LA, Prasad T. Biocompatible carbon quantum dots as versatile imaging nanotrackers of fungal pathogen - Candida albicans. Nanomedicine (Lond) 2024; 19:671-688. [PMID: 38426561 DOI: 10.2217/nnm-2023-0292] [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: 03/02/2024] Open
Abstract
Aim: The development of carbon quantum dots (C-QDs) as nanotrackers to understand drug-pathogen interactions, virulence and multidrug resistance. Methods: Microwave synthesis of C-QDs was performed using citric acid and polyethylene glycol. Further, in vitro toxicity was evaluated and imaging applications were demonstrated in Candida albicans isolates. Results: Well-dispersed, ultra small C-QDs exhibited no cyto/microbial/reactive oxygen species-mediated toxicity and internalized effectively in Candida yeast and hyphal cells. C-QDs were employed for confocal imaging of drug-sensitive and -resistant cells, and a study of the yeast-to-hyphal transition using atomic force microscopy in Candida was conducted for the first time. Conclusion: These biocompatible C-QDs have promising potential as next-generation nanotrackers for in vitro and in vivo targeted cellular and live imaging, after functionalization with biomolecules and drugs.
Collapse
Affiliation(s)
- Anam Rais
- Special Centre for Nano Science & AIRF, Jawaharlal Nehru University, New Delhi, 110067, India
- Department of Biosciences, Jamia Millia Islamia, New Delhi, 110025, India
| | - Shubham Sharma
- Department of Biochemical Engineering & Biotechnology, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Prashant Mishra
- Department of Biochemical Engineering & Biotechnology, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Luqman Ahmad Khan
- Department of Biosciences, Jamia Millia Islamia, New Delhi, 110025, India
| | - Tulika Prasad
- Special Centre for Nano Science & AIRF, Jawaharlal Nehru University, New Delhi, 110067, India
| |
Collapse
|
21
|
Bai Y, Zhou Y, Chang R, Hu X, Zhou Y, Chen J, Zhang Z, Yao J. Transcription profiles and phenotype reveal global response of Staphylococcus aureus exposed to ultrasound and ultraviolet stressors. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:169146. [PMID: 38061661 DOI: 10.1016/j.scitotenv.2023.169146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/05/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
Ultrasound and ultraviolet light have good inactivation performance against pathogens in sewage. In this study, the inactivation mechanisms of 60 kHz ultrasound and ultraviolet radiation against Staphylococcus aureus (S. aureus) were studied from the perspectives of cell phenotype and transcriptome for the first time. The results showed that both ultrasound and ultraviolet treatments had adverse impacts on the cellular morphology of S. aureus to varying degrees at cellular level. The transcriptomic analysis revealed that there were 225 and 1077 differentially expressed genes (DEGs) in the ultrasound and ultraviolet treatments, respectively. The result revealed that both ultrasound and ultraviolet could interfere with the expression of the genes involved in ABC transporters, amino acid and fatty acid metabolism to influence the membrane permeability. Besides the membrane permeability, ultraviolet also could disturb the ATP synthesis, DNA replication and cell division through restraining the expression of several genes related to carbohydrate metabolism, peptidoglycan synthesis, DNA-binding/repair protein synthesis. Compared with the single inactivation pathway of ultrasound, ultraviolet inactivation of S. aureus is multi-target and multi-pathway. We believe that the bactericidal mechanisms of ultrasound and ultraviolet radiation presented by this study could provide theoretical guidance for the synergistic inactivation of pathogens in sewage by ultrasound and ultraviolet radiation in the future.
Collapse
Affiliation(s)
- Yun Bai
- Key Laboratory of Three Gorges Reservoir Region's Eco-Environment, Ministry of Education, Chongqing University, Chongqing 400045, China; College of Environment and Ecology, Chongqing University, Chongqing 400045, China
| | - Yuanhang Zhou
- Key Laboratory of Three Gorges Reservoir Region's Eco-Environment, Ministry of Education, Chongqing University, Chongqing 400045, China; College of Environment and Ecology, Chongqing University, Chongqing 400045, China
| | - Ruiting Chang
- Key Laboratory of Three Gorges Reservoir Region's Eco-Environment, Ministry of Education, Chongqing University, Chongqing 400045, China; College of Environment and Ecology, Chongqing University, Chongqing 400045, China
| | - Xueli Hu
- Key Laboratory of Three Gorges Reservoir Region's Eco-Environment, Ministry of Education, Chongqing University, Chongqing 400045, China; College of Environment and Ecology, Chongqing University, Chongqing 400045, China
| | - Yingying Zhou
- Key Laboratory of Three Gorges Reservoir Region's Eco-Environment, Ministry of Education, Chongqing University, Chongqing 400045, China; College of Environment and Ecology, Chongqing University, Chongqing 400045, China
| | - Jiabo Chen
- Key Laboratory of Three Gorges Reservoir Region's Eco-Environment, Ministry of Education, Chongqing University, Chongqing 400045, China; College of Environment and Ecology, Chongqing University, Chongqing 400045, China
| | - Zhi Zhang
- Key Laboratory of Three Gorges Reservoir Region's Eco-Environment, Ministry of Education, Chongqing University, Chongqing 400045, China; College of Environment and Ecology, Chongqing University, Chongqing 400045, China.
| | - Juanjuan Yao
- Key Laboratory of Three Gorges Reservoir Region's Eco-Environment, Ministry of Education, Chongqing University, Chongqing 400045, China; College of Environment and Ecology, Chongqing University, Chongqing 400045, China.
| |
Collapse
|
22
|
Wellner SM, Alobaidallah MSA, Fei X, Herrero-Fresno A, Olsen JE. Genome-wide identification of fitness-genes in aminoglycoside-resistant Escherichia coli during antibiotic stress. Sci Rep 2024; 14:4163. [PMID: 38378700 PMCID: PMC10879529 DOI: 10.1038/s41598-024-54169-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/09/2024] [Indexed: 02/22/2024] Open
Abstract
Resistance against aminoglycosides is widespread in bacteria. This study aimed to identify genes that are important for growth of E. coli during aminoglycoside exposure, since such genes may be targeted to re-sensitize resistant E. coli to treatment. We constructed three transposon mutant libraries each containing > 230.000 mutants in E. coli MG1655 strains harboring streptomycin (aph(3″)-Ib/aph(6)-Id), gentamicin (aac(3)-IV), or neomycin (aph(3″)-Ia) resistance gene(s). Transposon Directed Insertion-site Sequencing (TraDIS), a combination of transposon mutagenesis and high-throughput sequencing, identified 56 genes which were deemed important for growth during streptomycin, 39 during gentamicin and 32 during neomycin exposure. Most of these fitness-genes were membrane-located (n = 55) and involved in either cell division, ATP-synthesis or stress response in the streptomycin and gentamicin exposed libraries, and enterobacterial common antigen biosynthesis or magnesium sensing/transport in the neomycin exposed library. For validation, eight selected fitness-genes/gene-clusters were deleted (minCDE, hflCK, clsA and cpxR associated with streptomycin and gentamicin resistance, and phoPQ, wecA, lpp and pal associated with neomycin resistance), and all mutants were shown to be growth attenuated upon exposure to the corresponding antibiotics. In summary, we identified genes that are advantageous in aminoglycoside-resistant E. coli during antibiotic stress. In addition, we increased the understanding of how aminoglycoside-resistant E. coli respond to antibiotic exposure.
Collapse
Affiliation(s)
- Sandra Marina Wellner
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark
| | - Mosaed Saleh A Alobaidallah
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, 21423, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, 22384, Jeddah, Saudi Arabia
| | - Xiao Fei
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark
| | - Ana Herrero-Fresno
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, Universidade da Santiago de Compostela (USC), Campus Terra, 27002, Lugo, Spain.
| | - John Elmerdahl Olsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark.
| |
Collapse
|
23
|
Janssens A, Nguyen VS, Cecil AJ, Van der Verren SE, Timmerman E, Deghelt M, Pak AJ, Collet JF, Impens F, Remaut H. SlyB encapsulates outer membrane proteins in stress-induced lipid nanodomains. Nature 2024; 626:617-625. [PMID: 38081298 DOI: 10.1038/s41586-023-06925-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 12/01/2023] [Indexed: 01/19/2024]
Abstract
The outer membrane in Gram-negative bacteria consists of an asymmetric phospholipid-lipopolysaccharide bilayer that is densely packed with outer-membrane β-barrel proteins (OMPs) and lipoproteins1. The architecture and composition of this bilayer is closely monitored and is essential to cell integrity and survival2-4. Here we find that SlyB, a lipoprotein in the PhoPQ stress regulon, forms stable stress-induced complexes with the outer-membrane proteome. SlyB comprises a 10 kDa periplasmic β-sandwich domain and a glycine zipper domain that forms a transmembrane α-helical hairpin with discrete phospholipid- and lipopolysaccharide-binding sites. After loss in lipid asymmetry, SlyB oligomerizes into ring-shaped transmembrane complexes that encapsulate β-barrel proteins into lipid nanodomains of variable size. We find that the formation of SlyB nanodomains is essential during lipopolysaccharide destabilization by antimicrobial peptides or acute cation shortage, conditions that result in a loss of OMPs and compromised outer-membrane barrier function in the absence of a functional SlyB. Our data reveal that SlyB is a compartmentalizing transmembrane guard protein that is involved in cell-envelope proteostasis and integrity, and suggest that SlyB represents a larger family of broadly conserved lipoproteins with 2TM glycine zipper domains with the ability to form lipid nanodomains.
Collapse
Affiliation(s)
- Arne Janssens
- Structural and Molecular Microbiology, VIB-VUB Center for Structural Biology, VIB, Brussels, Belgium
- Department of Bioengineering Sciences, Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Van Son Nguyen
- Structural and Molecular Microbiology, VIB-VUB Center for Structural Biology, VIB, Brussels, Belgium
- Department of Bioengineering Sciences, Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Adam J Cecil
- Chemical and Biological Engineering, Colorado School of Mines, Golden, CO, USA
| | - Sander E Van der Verren
- Structural and Molecular Microbiology, VIB-VUB Center for Structural Biology, VIB, Brussels, Belgium
- Department of Bioengineering Sciences, Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Evy Timmerman
- VIB Proteomics Core, Ghent, Belgium
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Michaël Deghelt
- Walloon Excellence in Life Sciences and Biotechnology, WELBIO, Brussels, Belgium
- De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Alexander J Pak
- Chemical and Biological Engineering, Colorado School of Mines, Golden, CO, USA
- Quantitative Biosciences and Engineering, Colorado School of Mines, Golden, CO, USA
| | - Jean-François Collet
- Walloon Excellence in Life Sciences and Biotechnology, WELBIO, Brussels, Belgium
- De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Francis Impens
- VIB Proteomics Core, Ghent, Belgium
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Han Remaut
- Structural and Molecular Microbiology, VIB-VUB Center for Structural Biology, VIB, Brussels, Belgium.
- Department of Bioengineering Sciences, Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
24
|
Rachwalski K, Tu MM, Madden SJ, French S, Hansen DM, Brown ED. A mobile CRISPRi collection enables genetic interaction studies for the essential genes of Escherichia coli. CELL REPORTS METHODS 2024; 4:100693. [PMID: 38262349 PMCID: PMC10832289 DOI: 10.1016/j.crmeth.2023.100693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 10/27/2023] [Accepted: 12/22/2023] [Indexed: 01/25/2024]
Abstract
Advances in gene editing, in particular CRISPR interference (CRISPRi), have enabled depletion of essential cellular machinery to study the downstream effects on bacterial physiology. Here, we describe the construction of an ordered E. coli CRISPRi collection, designed to knock down the expression of 356 essential genes with the induction of a catalytically inactive Cas9, harbored on the conjugative plasmid pFD152. This mobile CRISPRi library can be conjugated into other ordered genetic libraries to assess combined effects of essential gene knockdowns with non-essential gene deletions. As proof of concept, we probed cell envelope synthesis with two complementary crosses: (1) an Lpp deletion into every CRISPRi knockdown strain and (2) the lolA knockdown plasmid into the Keio collection. These experiments revealed a number of notable genetic interactions for the essential phenotype probed and, in particular, showed suppressing interactions for the loci in question.
Collapse
Affiliation(s)
- Kenneth Rachwalski
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada; Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Megan M Tu
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada; Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Sean J Madden
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada; Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Shawn French
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada; Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Drew M Hansen
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada; Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Eric D Brown
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada; Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada.
| |
Collapse
|
25
|
Vazulka S, Schiavinato M, Tauer C, Wagenknecht M, Cserjan-Puschmann M, Striedner G. RNA-seq reveals multifaceted gene expression response to Fab production in Escherichia coli fed-batch processes with particular focus on ribosome stalling. Microb Cell Fact 2024; 23:14. [PMID: 38183013 PMCID: PMC10768439 DOI: 10.1186/s12934-023-02278-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/18/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND Escherichia coli is a cost-effective expression system for production of antibody fragments like Fabs. Various yield improvement strategies have been applied, however, Fabs remain challenging to produce. This study aimed to characterize the gene expression response of commonly used E. coli strains BL21(DE3) and HMS174(DE3) to periplasmic Fab expression using RNA sequencing (RNA-seq). Two Fabs, Fabx and FTN2, fused to a post-translational translocation signal sequence, were produced in carbon-limited fed-batch cultivations. RESULTS Production of Fabx impeded cell growth substantially stronger than FTN2 and yields of both Fabs differed considerably. The most noticeable, common changes in Fab-producing cells suggested by our RNA-seq data concern the cell envelope. The Cpx and Psp stress responses, both connected to inner membrane integrity, were activated, presumably by recombinant protein aggregation and impairment of the Sec translocon. The data additionally suggest changes in lipopolysaccharide synthesis, adjustment of membrane permeability, and peptidoglycan maturation and remodeling. Moreover, all Fab-producing strains showed depletion of Mg2+, indicated by activation of the PhoQP two-component signal transduction system during the early stage and sulfur and phosphate starvation during the later stage of the process. Furthermore, our data revealed ribosome stalling, caused by the Fabx amino acid sequence, as a contributor to low Fabx yields. Increased Fabx yields were obtained by a site-specific amino acid exchange replacing the stalling sequence. Contrary to expectations, cell growth was not impacted by presence or removal of the stalling sequence. Considering ribosome rescue is a conserved mechanism, the substantial differences observed in gene expression between BL21(DE3) and HMS174(DE3) in response to ribosome stalling on the recombinant mRNA were surprising. CONCLUSIONS Through characterization of the gene expression response to Fab production under industrially relevant cultivation conditions, we identified potential cell engineering targets. Thereby, we hope to enable rational approaches to improve cell fitness and Fab yields. Furthermore, we highlight ribosome stalling caused by the amino acid sequence of the recombinant protein as a possible challenge during recombinant protein production.
Collapse
Affiliation(s)
- Sophie Vazulka
- Christian Doppler Laboratory for Production of Next-Level Biopharmaceuticals in E. Coli, Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190, Vienna, Austria
| | - Matteo Schiavinato
- Department of Biotechnology, Institute of Computational Biology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190, Vienna, Austria
| | - Christopher Tauer
- Christian Doppler Laboratory for Production of Next-Level Biopharmaceuticals in E. Coli, Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190, Vienna, Austria
| | - Martin Wagenknecht
- Boehringer Ingelheim RCV, GmbH & Co KG, Dr.-Boehringer-Gasse 5-11, A-1120, Vienna, Austria
| | - Monika Cserjan-Puschmann
- Christian Doppler Laboratory for Production of Next-Level Biopharmaceuticals in E. Coli, Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190, Vienna, Austria.
| | - Gerald Striedner
- Christian Doppler Laboratory for Production of Next-Level Biopharmaceuticals in E. Coli, Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190, Vienna, Austria
| |
Collapse
|
26
|
Orsini Delgado ML, Gamelas Magalhaes J, Morra R, Cultrone A. Muropeptides and muropeptide transporters impact on host immune response. Gut Microbes 2024; 16:2418412. [PMID: 39439228 PMCID: PMC11509177 DOI: 10.1080/19490976.2024.2418412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024] Open
Abstract
In bacteria, the cell envelope is the key element surrounding and protecting the bacterial content from mechanical or osmotic damages. It allows the selective interchanges of solutes, ions, cellular debris, and drugs between the cellular compartments and the external environment, thanks to the presence of transmembrane proteins called transporters. The major component of the cell envelope is the peptidoglycan, consisting of long linear glycan strands cross-linked by short peptide stems. During cell growth or under stress conditions, peptidoglycan fragments, the muropeptides, are released by bacteria and recognized by the host Pattern Recognition Receptor, promoting the activation of their innate defense mechanisms. The review sums up the salient aspects of microbiota-host interaction with a focus on the NOD-dependent immune response to bacterial peptidoglycan and on the accountability of muropeptide transporters in the crosstalk with the host and in antibiotic resistance. Furthermore, it retraces the discoveries and applications of microorganisms-derived components such as vaccines or vaccine adjuvants.
Collapse
|
27
|
Davies-Jones J, Davies PR, Graf A, Hewes D, Hill KE, Pascoe M. Photoinduced force microscopy as a novel method for the study of microbial nanostructures. NANOSCALE 2023; 16:223-236. [PMID: 38053416 DOI: 10.1039/d3nr03499b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
A detailed comparison of the capabilities of electron microscopy and nano-infrared (IR) microscopy for imaging microbial nanostructures has been carried out for the first time. The surface sensitivity, chemical specificity, and non-destructive nature of spectroscopic mapping is shown to offer significant advantages over transmission electron microscopy (TEM) for the study of biological samples. As well as yielding important topographical information, the distribution of amides, lipids, and carbohydrates across cross-sections of bacterial (Escherichia coli, Staphylococcus aureus) and fungal (Candida albicans) cells was demonstrated using PiFM. The unique information derived from this new mode of spectroscopic mapping of the surface chemistry and biology of microbial cell walls and membranes, may provide new insights into fungal/bacterial cell function as well as having potential use in determining mechanisms of antimicrobial resistance, especially those targeting the cell wall.
Collapse
Affiliation(s)
- Josh Davies-Jones
- Cardiff Catalysis Institute, Cardiff School of Chemistry, Cardiff University, Cardiff, CF10 3A, UK.
| | - Philip R Davies
- Cardiff Catalysis Institute, Cardiff School of Chemistry, Cardiff University, Cardiff, CF10 3A, UK.
| | - Arthur Graf
- Cardiff Catalysis Institute, Cardiff School of Chemistry, Cardiff University, Cardiff, CF10 3A, UK.
| | - Dan Hewes
- Cardiff Catalysis Institute, Cardiff School of Chemistry, Cardiff University, Cardiff, CF10 3A, UK.
| | - Katja E Hill
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff, CF14 4XY, UK.
| | - Michael Pascoe
- Cardiff Catalysis Institute, Cardiff School of Chemistry, Cardiff University, Cardiff, CF10 3A, UK.
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, CF10 3BN, UK.
| |
Collapse
|
28
|
Wykes H, Le VVH, Olivera C, Rakonjac J. When less is more: shortening the Lpp protein leads to increased vancomycin resistance in Escherichia coli. J Antibiot (Tokyo) 2023; 76:746-750. [PMID: 37749219 PMCID: PMC10678296 DOI: 10.1038/s41429-023-00658-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 09/01/2023] [Accepted: 09/11/2023] [Indexed: 09/27/2023]
Abstract
Vancomycin is a naturally occurring cell-wall-targeting glycopeptide antibiotic. Due to the low potency of this antibiotic against Gram-negative pathogens, such as Escherichia coli, there is a limited knowledge about interactions between vancomycin and this group of bacteria. Here, we show that an in-frame 63 bp deletion of the lpp gene caused a fourfold increase in vancomycin resistance in E. coli. The resulting protein, LppΔ21, is 21 amino acids shorter than the wild-type Lpp, a helical structural lipoprotein that controls the width of the periplasmic space through its length. The mutant remains susceptible to synergistic growth inhibition by combination of furazolidone and vancomycin; with furazolidone decreasing the vancomycin MIC by eightfold. These findings have clinical relevance, given that the vancomycin concentration required to select the lpp mutation is reachable during typical vancomycin oral administration for treating Clostridioides difficile infections. Combination therapy with furazolidone, however, is likely to prevent emergence and outgrowth of the lpp-mutated Gram-negative coliforms, avoiding exacerbation of the patient's condition during the treatment.
Collapse
Affiliation(s)
- Hannah Wykes
- School of Natural Sciences, Massey University, Palmerston North, New Zealand
| | - Vuong Van Hung Le
- School of Natural Sciences, Massey University, Palmerston North, New Zealand.
- Section of Microbiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| | - Catrina Olivera
- School of Natural Sciences, Massey University, Palmerston North, New Zealand
| | - Jasna Rakonjac
- School of Natural Sciences, Massey University, Palmerston North, New Zealand.
| |
Collapse
|
29
|
Wang HJ, Hernández-Rocamora VM, Kuo CI, Hsieh KY, Lee SH, Ho MR, Tu Z, Vollmer W, Chang CI. Structural basis for the hydrolytic activity of the transpeptidase-like protein DpaA to detach Braun's lipoprotein from peptidoglycan. mBio 2023; 14:e0137923. [PMID: 37830798 PMCID: PMC10653827 DOI: 10.1128/mbio.01379-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/21/2023] [Indexed: 10/14/2023] Open
Abstract
IMPORTANCE Cross-linking reaction of Braun's lipoprotein (Lpp) to peptidoglycan (PG) is catalyzed by some members of the YkuD family of transpeptidases. However, the exact opposite reaction of cleaving the Lpp-PG cross-link is performed by DpaA, which is also a YkuD-like protein. In this work, we determined the crystal structure of DpaA to provide the molecular rationale for the ability of the transpeptidase-like protein to cleave, rather than form, the Lpp-PG linkage. Our findings also revealed the structural features that distinguish the different functional types of the YkuD family enzymes from one another.
Collapse
Affiliation(s)
- Hsiu-Jung Wang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Víctor M. Hernández-Rocamora
- Centre for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Chiao-I Kuo
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Kan-Yen Hsieh
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Szu-Hui Lee
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Meng-Ru Ho
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Zhijay Tu
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - 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, Queensland, Australia
| | - Chung-I Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
- College of Life Science, Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
30
|
Fordjour E, Bai Z, Li S, Li S, Sackey I, Yang Y, Liu CL. Improved Membrane Permeability via Hypervesiculation for In Situ Recovery of Lycopene in Escherichia coli. ACS Synth Biol 2023; 12:2725-2739. [PMID: 37607052 DOI: 10.1021/acssynbio.3c00306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Lycopene biosynthesis is frequently hampered by downstream processing hugely due to its inability to be secreted out from the producing chassis. Engineering cell factories can resolve this issue by secreting this hydrophobic compound. A highly permeable E. coli strain was developed for a better release rate of lycopene. Specifically, the heterologous mevalonate pathway and crtEBI genes from Corynebacterium glutamicum were overexpressed in Escherichia coli BL21 (DE3) for lycopene synthesis. To ensure in situ lycopene production, murein lipoprotein, lipoprotein NlpI, inner membrane permease protein, and membrane-anchored protein in TolA-TolQ-TolR were deleted for improved membrane permeability. The final strain, LYC-8, produced 438.44 ± 8.11 and 136.94 ± 1.94 mg/L of extracellular and intracellular lycopene in fed-batch fermentation. Both proteomics and lipidomics analyses of secreted outer membrane vesicles were perfect indicators of hypervesiculation. Changes in the ratio of saturated fatty acids, unsaturated fatty acids, and cyclopropane fatty acids coupled with the branching and acyl chain lengths altered the membrane fatty acid composition. This ensured membrane fluidity and permeability for in situ lycopene release. The combinatorial deletion of these genes altered the cellular morphology. The structural and morphological changes in cell shape, size, and length were associated with changes in the mechanical strength of the cell envelope. The enhanced lycopene production and secretion mediated by improved membrane permeability established a cell lysis-free system for an efficient releasing rate and downstream processing, demonstrating the importance of vesicle-associated membrane permeability in efficient lycopene production.
Collapse
Affiliation(s)
- Eric Fordjour
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center of Cereal Fermentation, and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
- Jiangsu Provincial Research Centre for Bioactive Product Processing Technology, Jiangnan University, Wuxi 214122, China
| | - Zhonghu Bai
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center of Cereal Fermentation, and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
- Jiangsu Provincial Research Centre for Bioactive Product Processing Technology, Jiangnan University, Wuxi 214122, China
| | - Sihan Li
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center of Cereal Fermentation, and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
- Jiangsu Provincial Research Centre for Bioactive Product Processing Technology, Jiangnan University, Wuxi 214122, China
| | - Shijie Li
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center of Cereal Fermentation, and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
- Jiangsu Provincial Research Centre for Bioactive Product Processing Technology, Jiangnan University, Wuxi 214122, China
| | - Isaac Sackey
- Department of Biological Sciences, Faculty of Biosciences, University for Development Studies, P.O. Box TL1350, NT-0272-1946 Tamale, Ghana
| | - Yankun Yang
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center of Cereal Fermentation, and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
- Jiangsu Provincial Research Centre for Bioactive Product Processing Technology, Jiangnan University, Wuxi 214122, China
| | - Chun-Li Liu
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center of Cereal Fermentation, and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China
- Jiangsu Provincial Research Centre for Bioactive Product Processing Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
31
|
Alberge F, Lakey BD, Schaub RE, Dohnalkova AC, Lemmer KC, Dillard JP, Noguera DR, Donohue TJ. A previously uncharacterized divisome-associated lipoprotein, DalA, is needed for normal cell division in Rhodobacterales. mBio 2023; 14:e0120323. [PMID: 37389444 PMCID: PMC10470522 DOI: 10.1128/mbio.01203-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 05/23/2023] [Indexed: 07/01/2023] Open
Abstract
The bacterial cell envelope is a key subcellular compartment with important roles in antibiotic resistance, nutrient acquisition, and cell morphology. We seek to gain a better understanding of proteins that contribute to the function of the cell envelope in Alphaproteobacteria. Using Rhodobacter sphaeroides, we show that a previously uncharacterized protein, RSP_1200, is an outer membrane (OM) lipoprotein that non-covalently binds peptidoglycan (PG). Using a fluorescently tagged version of this protein, we find that RSP_1200 undergoes a dynamic repositioning during the cell cycle and is enriched at the septum during cell division. We show that the position of RSP_1200 mirrors the location of FtsZ rings, leading us to propose that RSP_1200 is a newly identified component of the R. sphaeroides' divisome. Additional support for this hypothesis includes the co-precipitation of RSP_1200 with FtsZ, the Pal protein, and several predicted PG L,D-transpeptidases. We also find that a ∆RSP_1200 mutation leads to defects in cell division, sensitivity to PG-active antibiotics, and results in the formation of OM protrusions at the septum during cell division. Based on these results, we propose to name RSP_1200 DalA (for division-associated lipoprotein A) and postulate that DalA serves as a scaffold to position or modulate the activity of PG transpeptidases that are needed to form envelope invaginations during cell division. We find that DalA homologs are present in members of the Rhodobacterales order within Alphaproteobacteria. Therefore, we propose that further analysis of this and related proteins will increase our understanding of the macromolecular machinery and proteins that participate in cell division in Gram-negative bacteria. IMPORTANCE Multi-protein complexes of the bacterial cell envelope orchestrate key processes like growth, division, biofilm formation, antimicrobial resistance, and production of valuable compounds. The subunits of these protein complexes are well studied in some bacteria, and differences in their composition and function are linked to variations in cell envelope composition, shape, and proliferation. However, some envelope protein complex subunits have no known homologs across the bacterial phylogeny. We find that Rhodobacter sphaeroides RSP_1200 is a newly identified lipoprotein (DalA) and that loss of this protein causes defects in cell division and changes the sensitivity to compounds, affecting cell envelope synthesis and function. We find that DalA forms a complex with proteins needed for cell division, binds the cell envelope polymer peptidoglycan, and colocalizes with enzymes involved in the assembly of this macromolecule. The analysis of DalA provides new information on the cell division machinery in this and possibly other Alphaproteobacteria.
Collapse
Affiliation(s)
- François Alberge
- />Wisconsin Energy Institute, Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Bryan D. Lakey
- />Wisconsin Energy Institute, Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ryan E. Schaub
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Alice C. Dohnalkova
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington, USA
| | | | - Joseph P. Dillard
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Daniel R. Noguera
- />Wisconsin Energy Institute, Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Civil and Environmental Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Timothy J. Donohue
- />Wisconsin Energy Institute, Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
32
|
Guo M, Zhang X, Ismail BB, He Q, Yang Z, Xianyu Y, Liu W, Zhou J, Ye X, Liu D. Super Antibacterial Capacity and Cell Envelope-Disruptive Mechanism of Ultrasonically Grafted N-Halamine PBAT/PBF Films against Escherichia coli. ACS APPLIED MATERIALS & INTERFACES 2023; 15:38910-38929. [PMID: 37550824 DOI: 10.1021/acsami.3c05378] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
Antibacterial materials are urgently needed to combat bacterial contamination, growth, or attachment on contact surfaces, as bacterial infections remain a public health crisis worldwide. Here, a novel ultrasound-assisted method is utilized for the first time to fabricate oxidative chlorine-loaded AH@PBAT/PBF-Cl films with more superior grafting efficiency and rechargeable antibacterial effect than those from conventional techniques. The films remarkably inactivate 99.9999% Escherichia coli and Staphylococcus aureus cells, inducing noticeable cell deformations and mechanical instability. The specific antibacterial mechanism against E. coli used as a model organism is unveiled using several cell envelope structural and functional analyses combined with proteomics, peptidoglycomics, and fluorescence probe techniques. Film treatment partially neutralizes the bacterial surface charge, induces oxidative stress and cytoskeleton deformity, alters membrane properties, and disrupts the expression of key proteins involved in the synthesis and transport of the lipopolysaccharide and peptidoglycan, indicating the cell envelope as the primary target. The films specifically target lipopolysaccharides, resulting in structural impairment of the polysaccharide and lipid A components, and inhibit peptidoglycan precursor synthesis. Together, these lead to metabolic disorders, membrane dysfunction, structural collapse, and eventual death. Given the films' antibacterial effects via the disruption of key cell envelope components, they can potentially combat a wide range of bacteria. These findings lay a theoretical basis for developing efficient antibacterial materials for food safety or biomedical applications.
Collapse
Affiliation(s)
- Mingming Guo
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Integrated Research Base of Southern Fruit and Vegetable Preservation Technology, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China
- Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China
- Ningbo Research Institute, Zhejiang University, Ningbo 315100, China
| | - Xinhui Zhang
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Integrated Research Base of Southern Fruit and Vegetable Preservation Technology, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China
| | - Balarabe B Ismail
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Integrated Research Base of Southern Fruit and Vegetable Preservation Technology, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China
| | - Qiao He
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Integrated Research Base of Southern Fruit and Vegetable Preservation Technology, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China
| | - Zhehao Yang
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Integrated Research Base of Southern Fruit and Vegetable Preservation Technology, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China
| | - Yunlei Xianyu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Integrated Research Base of Southern Fruit and Vegetable Preservation Technology, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China
- Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China
- Ningbo Research Institute, Zhejiang University, Ningbo 315100, China
| | - Wentao Liu
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Jianwei Zhou
- School of Mechatronics and Energy Engineering, NingboTech University, Ningbo 315100, China
| | - Xingqian Ye
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Integrated Research Base of Southern Fruit and Vegetable Preservation Technology, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China
- Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China
- Ningbo Research Institute, Zhejiang University, Ningbo 315100, China
| | - Donghong Liu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Integrated Research Base of Southern Fruit and Vegetable Preservation Technology, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China
- Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China
- Ningbo Research Institute, Zhejiang University, Ningbo 315100, China
| |
Collapse
|
33
|
Kadeřábková N, Mahmood AJS, Furniss RCD, Mavridou DAI. Making a chink in their armor: Current and next-generation antimicrobial strategies against the bacterial cell envelope. Adv Microb Physiol 2023; 83:221-307. [PMID: 37507160 PMCID: PMC10517717 DOI: 10.1016/bs.ampbs.2023.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
Gram-negative bacteria are uniquely equipped to defeat antibiotics. Their outermost layer, the cell envelope, is a natural permeability barrier that contains an array of resistance proteins capable of neutralizing most existing antimicrobials. As a result, its presence creates a major obstacle for the treatment of resistant infections and for the development of new antibiotics. Despite this seemingly impenetrable armor, in-depth understanding of the cell envelope, including structural, functional and systems biology insights, has promoted efforts to target it that can ultimately lead to the generation of new antibacterial therapies. In this article, we broadly overview the biology of the cell envelope and highlight attempts and successes in generating inhibitors that impair its function or biogenesis. We argue that the very structure that has hampered antibiotic discovery for decades has untapped potential for the design of novel next-generation therapeutics against bacterial pathogens.
Collapse
Affiliation(s)
- Nikol Kadeřábková
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States
| | - Ayesha J S Mahmood
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States
| | - R Christopher D Furniss
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Despoina A I Mavridou
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States; John Ring LaMontagne Center for Infectious Diseases, The University of Texas at Austin, Austin, TX, United States.
| |
Collapse
|
34
|
Liu C, Angius F, Pol A, Mesman RA, Versantvoort W, Op den Camp HJM. Identification and characterization of an abundant lipoprotein from Methylacidiphilum fumariolicum SolV. Arch Microbiol 2023; 205:261. [PMID: 37306788 DOI: 10.1007/s00203-023-03603-y] [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: 04/05/2023] [Revised: 05/17/2023] [Accepted: 05/30/2023] [Indexed: 06/13/2023]
Abstract
Bacterial lipoproteins are characterized by the presence of a conserved N-terminal lipid-modified cysteine residue that allows the hydrophilic protein to anchor into bacterial cell membranes. These lipoproteins play essential roles in a wide variety of physiological processes. Based on transcriptome analysis of the verrucomicrobial methanotroph Methylacidiphilum fumariolicum SolV, we identified a highly expressed lipoprotein, WP_009060351 (139 amino acids), in its genome. The first 86 amino acids are specific for the methanotrophic genera Methylacidiphilum and Methylacidmicrobium, while the last 53 amino acids are present only in lipoproteins of members from the phylum Verrucomicrobiota (Hedlund). Heterologous expression of WP_009060351 in Escherichia coli revealed a 25-kDa dimeric protein and a 60-kDa tetrameric protein. Immunoblotting showed that WP_009060351 was present in the total membrane protein and peptidoglycan fractions of M. fumariolicum SolV. The results suggest an involvement of lipoprotein WP_009060351 in the linkage between the outer membrane and the peptidoglycan.
Collapse
Affiliation(s)
- Changqing Liu
- Faculty of Science, Department of Microbiology, Radboud Institute for Biological and Environmental Sciences, Radboud University, Nijmegen, The Netherlands
| | - Federica Angius
- Faculty of Science, Department of Microbiology, Radboud Institute for Biological and Environmental Sciences, Radboud University, Nijmegen, The Netherlands
| | - Arjan Pol
- Faculty of Science, Department of Microbiology, Radboud Institute for Biological and Environmental Sciences, Radboud University, Nijmegen, The Netherlands
| | - Rob A Mesman
- Faculty of Science, Department of Microbiology, Radboud Institute for Biological and Environmental Sciences, Radboud University, Nijmegen, The Netherlands
| | - Wouter Versantvoort
- Faculty of Science, Department of Microbiology, Radboud Institute for Biological and Environmental Sciences, Radboud University, Nijmegen, The Netherlands
| | - Huub J M Op den Camp
- Faculty of Science, Department of Microbiology, Radboud Institute for Biological and Environmental Sciences, Radboud University, Nijmegen, The Netherlands.
| |
Collapse
|
35
|
Bakhurin KI, Hughes RN, Jiang Q, Hossain M, Gutkin B, Fallon IP, Yin H. Force tuning explains changes in phasic dopamine signaling during stimulus-reward learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.23.537994. [PMID: 37162997 PMCID: PMC10168281 DOI: 10.1101/2023.04.23.537994] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
According to a popular hypothesis, phasic dopamine (DA) activity encodes a reward prediction error (RPE) necessary for reinforcement learning. However, recent work showed that DA neurons are necessary for performance rather than learning. One limitation of previous work on phasic DA signaling and RPE is the limited behavioral measures. Here, we measured subtle force exertion while recording and manipulating DA activity in the ventral tegmental area (VTA) during stimulus-reward learning. We found two major populations of DA neurons that increased firing before forward and backward force exertion. Force tuning is the same regardless of learning, reward predictability, or outcome valence. Changes in the pattern of force exertion can explain results traditionally used to support the RPE hypothesis, such as modulation by reward magnitude, probability, and unpredicted reward delivery or omission. Thus VTA DA neurons are not used to signal RPE but to regulate force exertion during motivated behavior.
Collapse
|
36
|
Trotta KL, Hayes BM, Schneider JP, Wang J, Todor H, Rockefeller Grimes P, Zhao Z, Hatleberg WL, Silvis MR, Kim R, Koo BM, Basler M, Chou S. Lipopolysaccharide transport regulates bacterial sensitivity to a cell wall-degrading intermicrobial toxin. PLoS Pathog 2023; 19:e1011454. [PMID: 37363922 PMCID: PMC10328246 DOI: 10.1371/journal.ppat.1011454] [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: 05/02/2023] [Revised: 07/07/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
Gram-negative bacteria can antagonize neighboring microbes using a type VI secretion system (T6SS) to deliver toxins that target different essential cellular features. Despite the conserved nature of these targets, T6SS potency can vary across recipient species. To understand the functional basis of intrinsic T6SS susceptibility, we screened for essential Escherichia coli (Eco) genes that affect its survival when antagonized by a cell wall-degrading T6SS toxin from Pseudomonas aeruginosa, Tae1. We revealed genes associated with both the cell wall and a separate layer of the cell envelope, lipopolysaccharide, that modulate Tae1 toxicity in vivo. Disruption of genes in early lipopolysaccharide biosynthesis provided Eco with novel resistance to Tae1, despite significant cell wall degradation. These data suggest that Tae1 toxicity is determined not only by direct substrate damage, but also by indirect cell envelope homeostasis activities. We also found that Tae1-resistant Eco exhibited reduced cell wall synthesis and overall slowed growth, suggesting that reactive cell envelope maintenance pathways could promote, not prevent, self-lysis. Together, our study reveals the complex functional underpinnings of susceptibility to Tae1 and T6SS which regulate the impact of toxin-substrate interactions in vivo.
Collapse
Affiliation(s)
- Kristine L. Trotta
- Department of Biochemistry & Biophysics, University of California–San Francisco, San Francisco, California, United States of America
| | - Beth M. Hayes
- Department of Biochemistry & Biophysics, University of California–San Francisco, San Francisco, California, United States of America
| | | | - Jing Wang
- Biozentrum, University of Basel, Basel, Switzerland
| | - Horia Todor
- Department of Cell and Tissue Biology, University of California–San Francisco, San Francisco, California, United States of America
| | - Patrick Rockefeller Grimes
- Department of Biochemistry & Biophysics, University of California–San Francisco, San Francisco, California, United States of America
| | - Ziyi Zhao
- Department of Biochemistry & Biophysics, University of California–San Francisco, San Francisco, California, United States of America
| | | | - Melanie R. Silvis
- Department of Cell and Tissue Biology, University of California–San Francisco, San Francisco, California, United States of America
| | - Rachel Kim
- Department of Biochemistry & Biophysics, University of California–San Francisco, San Francisco, California, United States of America
| | - Byoung Mo Koo
- Department of Cell and Tissue Biology, University of California–San Francisco, San Francisco, California, United States of America
| | - Marek Basler
- Biozentrum, University of Basel, Basel, Switzerland
| | - Seemay Chou
- Department of Biochemistry & Biophysics, University of California–San Francisco, San Francisco, California, United States of America
| |
Collapse
|
37
|
Trotta KL, Hayes BM, Schneider JP, Wang J, Todor H, Grimes PR, Zhao Z, Hatleberg WL, Silvis MR, Kim R, Koo BM, Basler M, Chou S. Lipopolysaccharide integrity primes bacterial sensitivity to a cell wall-degrading intermicrobial toxin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.20.524922. [PMID: 36747731 PMCID: PMC9900751 DOI: 10.1101/2023.01.20.524922] [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: 01/22/2023]
Abstract
Gram-negative bacteria can antagonize neighboring microbes using a type VI secretion system (T6SS) to deliver toxins that target different essential cellular features. Despite the conserved nature of these targets, T6SS potency can vary across recipient species. To understand the molecular basis of intrinsic T6SS susceptibility, we screened for essential Escherichia coli genes that affect its survival when antagonized by a cell wall-degrading T6SS toxin from Pseudomonas aeruginosa , Tae1. We revealed genes associated with both the cell wall and a separate layer of the cell envelope, surface lipopolysaccharide, that modulate Tae1 toxicity in vivo . Disruption of lipopolysaccharide synthesis provided Escherichia coli (Eco) with novel resistance to Tae1, despite significant cell wall degradation. These data suggest that Tae1 toxicity is determined not only by direct substrate damage, but also by indirect cell envelope homeostasis activities. We also found that Tae1-resistant Eco exhibited reduced cell wall synthesis and overall slowed growth, suggesting that reactive cell envelope maintenance pathways could promote, not prevent, self-lysis. Together, our study highlights the consequences of co-regulating essential pathways on recipient fitness during interbacterial competition, and how antibacterial toxins leverage cellular vulnerabilities that are both direct and indirect to their specific targets in vivo .
Collapse
Affiliation(s)
- Kristine L Trotta
- Department of Biochemistry & Biophysics, University of California – San Francisco, San Francisco, CA, USA
| | - Beth M Hayes
- Department of Biochemistry & Biophysics, University of California – San Francisco, San Francisco, CA, USA
| | - Johannes P Schneider
- Focal Area Infection Biology, Biozentrum, University of Basel, Klingelbergstrasse 50/70, CH - 4056 Basel, Switzerland
| | - Jing Wang
- Focal Area Infection Biology, Biozentrum, University of Basel, Klingelbergstrasse 50/70, CH - 4056 Basel, Switzerland
| | - Horia Todor
- Department of Cell and Tissue Biology, University of California – San Francisco, San Francisco, CA, USA
| | - Patrick Rockefeller Grimes
- Department of Biochemistry & Biophysics, University of California – San Francisco, San Francisco, CA, USA
| | - Ziyi Zhao
- Department of Biochemistry & Biophysics, University of California – San Francisco, San Francisco, CA, USA
| | | | - Melanie R Silvis
- Department of Cell and Tissue Biology, University of California – San Francisco, San Francisco, CA, USA
| | - Rachel Kim
- Department of Biochemistry & Biophysics, University of California – San Francisco, San Francisco, CA, USA
| | - Byoung Mo Koo
- Department of Cell and Tissue Biology, University of California – San Francisco, San Francisco, CA, USA
| | - Marek Basler
- Focal Area Infection Biology, Biozentrum, University of Basel, Klingelbergstrasse 50/70, CH - 4056 Basel, Switzerland
| | - Seemay Chou
- Department of Biochemistry & Biophysics, University of California – San Francisco, San Francisco, CA, USA
| |
Collapse
|
38
|
Volle C, Núñez ME, Spain EM, Hart BC, Wengen MB, Lane S, Criollo A, Mahoney CA, Ferguson MA. AFM Force Mapping Elucidates Pilus Deployment and Key Lifestyle-Dependent Surface Properties in Bdellovibrio bacteriovorus. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:4233-4244. [PMID: 36926913 PMCID: PMC10062353 DOI: 10.1021/acs.langmuir.2c03134] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/03/2023] [Indexed: 06/18/2023]
Abstract
Bdellovibrio bacteriovorus is known for predation of a wide variety of Gram-negative bacteria, making it of interest as an alternative or supplement to chemical antibiotics. However, a fraction of B. bacteriovorus follows a nonpredatory, "host-independent" (HI) life cycle. In this study, live predatory and HI B. bacteriovorus were captured on a surface and examined, in buffer, by collecting force maps using atomic force microscopy (AFM). The approach curves obtained on HI cells are similar to those on other Gram-negative cells, with a short nonlinear region followed by a linear region. In contrast, the approach curves obtained on predatory cells have a large nonlinear region, reflecting the unusual flexibility of the predatory cell. As the AFM tip is retracted, it shows virtually no adhesion to predatory B. bacteriovorus but has multiple adhesion events on HI cells and the 200-500+ nm region immediately surrounding them. Measured pull-off forces, pull-off distances, and effective spring constants are consistent with the multiple stretching events of Type IV pili, both on and especially adjacent to the cells. Exposure of the HI B. bacteriovorus to a pH-neutral 10% cranberry juice solution, which contains type A proanthocyanidins that are known to interfere with the adhesion of multiple types of pili, results in a substantial reduction in adhesion. Type IV pili are required for successful predation by B. bacteriovorus, but pili used in the predation process are located at the non-flagellated pole of the cell and can retract when not in use. Such pili are rarely observed under the conditions of this study, where the predator has not encountered a prey cell. In contrast, HI cells appear to have many pili distributed on and around the whole cell, presumably ready to be utilized for a variety of HI cell activities including attachment to surfaces.
Collapse
Affiliation(s)
- Catherine
B. Volle
- Departments
of Chemistry and Biology, Cornell College, Mount Vernon, Iowa 52314, United States
| | - Megan E. Núñez
- Department
of Chemistry, Wellesley College, Wellesley, Massachusetts 02481, United States
| | - Eileen M. Spain
- Department
of Chemistry, Occidental College, Los Angeles, California 90041, United States
| | - Bridget C. Hart
- Department
of Chemistry, State University of New York, New Paltz, New York 12561, United States
| | - Michael B. Wengen
- Department
of Chemistry, State University of New York, New Paltz, New York 12561, United States
| | - Sophia Lane
- Department
of Chemistry, State University of New York, New Paltz, New York 12561, United States
| | - Alexa Criollo
- Department
of Chemistry, State University of New York, New Paltz, New York 12561, United States
| | - Catherine A. Mahoney
- Department
of Chemistry, State University of New York, New Paltz, New York 12561, United States
| | - Megan A. Ferguson
- Department
of Chemistry, State University of New York, New Paltz, New York 12561, United States
| |
Collapse
|
39
|
Paesa M, Remirez de Ganuza C, Alejo T, Yus C, Irusta S, Arruebo M, Sebastian V, Mendoza G. Elucidating the mechanisms of action of antibiotic-like ionic gold and biogenic gold nanoparticles against bacteria. J Colloid Interface Sci 2023; 633:786-799. [PMID: 36493743 DOI: 10.1016/j.jcis.2022.11.138] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/17/2022] [Accepted: 11/27/2022] [Indexed: 11/30/2022]
Abstract
The antimicrobial action of gold depends on different factors including its oxidation state in the intra- and extracellular medium, the redox potential, its ability to produce reactive oxygen species (ROS), the medium components, the properties of the targeted bacteria wall, its penetration in the bacterial cytosol, the cell membrane potential, and its interaction with intracellular components. We demonstrate that different gold species are able to induce bacterial wall damage as a result of their electrostatic interaction with the cell membrane, the promotion of ROS generation, and the consequent DNA damage. In-depth genomic and proteomic studies on Escherichia coli confirmed the superior toxicity of Au (III) vs Au (I) based on the different molecular mechanisms analyzed including oxidative stress, bacterial energetic metabolism, biosynthetic processes, and cell transport. At equivalent bactericidal doses of Au (III) and Au (I) eukaryotic cells were not as affected as bacteria did, maintaining unaffected cell viability, morphology, and focal adhesions; however, increased ROS generation and disruption in the mitochondrial membrane potential were also observed. Herein, we shed light on the antimicrobial mechanisms of ionic and biogenic gold nanoparticles against bacteria. Under selected conditions antibiotic-like ionic gold can exert a strong antimicrobial activity while being harmless to human cells.
Collapse
Affiliation(s)
- Monica Paesa
- Department of Chemical Engineering, University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain; Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain
| | - Cristina Remirez de Ganuza
- Department of Chemical Engineering, University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain; Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain
| | - Teresa Alejo
- Department of Chemical Engineering, University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain; Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain; Aragon Health Research Institute (IIS Aragon), 50009-Zaragoza, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029-Madrid, Spain
| | - Cristina Yus
- Department of Chemical Engineering, University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain
| | - Silvia Irusta
- Department of Chemical Engineering, University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain; Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain; Aragon Health Research Institute (IIS Aragon), 50009-Zaragoza, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029-Madrid, Spain
| | - Manuel Arruebo
- Department of Chemical Engineering, University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain; Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain; Aragon Health Research Institute (IIS Aragon), 50009-Zaragoza, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029-Madrid, Spain.
| | - Víctor Sebastian
- Department of Chemical Engineering, University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain; Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain; Aragon Health Research Institute (IIS Aragon), 50009-Zaragoza, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029-Madrid, Spain.
| | - Gracia Mendoza
- Aragon Health Research Institute (IIS Aragon), 50009-Zaragoza, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029-Madrid, Spain
| |
Collapse
|
40
|
Sperandeo P, Martorana AM, Zaccaria M, Polissi A. Targeting the LPS export pathway for the development of novel therapeutics. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119406. [PMID: 36473551 DOI: 10.1016/j.bbamcr.2022.119406] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 10/14/2022] [Accepted: 11/01/2022] [Indexed: 12/12/2022]
Abstract
The rapid rise of multi-resistant bacteria is a global health threat. This is especially serious for Gram-negative bacteria in which the impermeable outer membrane (OM) acts as a shield against antibiotics. The development of new drugs with novel modes of actions to combat multi-drug resistant pathogens requires the selection of suitable processes to be targeted. The LPS export pathway is an excellent under exploited target for drug development. Indeed, LPS is the major determinant of the OM permeability barrier, and its biogenetic pathway is conserved in most Gram-negatives. Here we describe efforts to identify inhibitors of the multiprotein Lpt system that transports LPS to the cell surface. Despite none of these molecules has been approved for clinical use, they may represent valuable compounds for optimization. Finally, the recent discovery of a link between inhibition of LPS biogenesis and changes in peptidoglycan structure uncovers additional targets to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Paola Sperandeo
- Department of Pharmacological and Biomolecular Sciences, Via Balzaretti 9, 20133 Milano, Italy
| | - Alessandra M Martorana
- Department of Pharmacological and Biomolecular Sciences, Via Balzaretti 9, 20133 Milano, Italy
| | - Marta Zaccaria
- Department of Pharmacological and Biomolecular Sciences, Via Balzaretti 9, 20133 Milano, Italy
| | - Alessandra Polissi
- Department of Pharmacological and Biomolecular Sciences, Via Balzaretti 9, 20133 Milano, Italy.
| |
Collapse
|
41
|
Type III Secretion System Repressor RhpR Induces GrlP, a Glycine-Rich Outer Membrane Lipoprotein with Functions in Regulating the Periplasmic Space and Pleiotropic Responses. Appl Environ Microbiol 2023; 89:e0158722. [PMID: 36602318 PMCID: PMC9888284 DOI: 10.1128/aem.01587-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The two-component system RhpRS was initially identified as a regulator of genes encoding the type III secretion system (T3SS) in Pseudomonas syringae. Phosphorylated RhpR (P-RhpR) negatively regulates the T3SS genes by repressing the hrpR promoter, but directly activates the expression of a small gene named here as grlp. Here, we show that grlp is expressed higher in rich medium than in minimal medium in P. s. pv. tomato DC3000 and encodes a glycine rich lipoprotein (GrlP) located in the outer membrane (OM). The grlp gene has a pleiotropic effect on bacterial behaviors such as reductions in pathogenicity, swimming motility, biofilm formation, tolerance to various stresses and antibiotics, and long-term survival when overexpressed, but induces these responses when it is deleted in P. s. pv. tomato DC3000. Overexpression of grlp increases the size of periplasm while deletion of grlp decreases the periplasmic space. Further, GrlP interacts with OprI, the ortholog of E. coli OM lipoprotein Lpp, a key player in determining the size of periplasm and mechanic stiffness of the OM by tethering the OM to peptidoglycan (PG) in periplasm. As periplasmic space and OM mechanics play central roles in regulating bacterial physiology, we speculate that GrlP probably imposes its functions on bacterial physiology by regulating the periplasmic space and OM mechanics. These findings suggest that the T3SS gene regulation is closely coordinated with bacterial cell envelope properties by RhpRS in P. syringe. IMPORTANCE The OM of Gram-negative bacteria is the most front line in contact with extracellular milieu. OM is not only a protective layer, but also a structure that determines the envelope stiffness. Recent evidence indicated that components determining the periplasmic space and cross-links of lipopolysaccharide on the OM play key roles in regulating the mechanical properties of the OM. However, whether the OM composition and mechanical properties are coordinated with the expression of the T3SS genes is unknown. Here, we found that the two-component system (TCS) regulator P-RhpR, a direct repressor of the T3SS regulator hrpRS operon, directly activates the expression of the OM lipoprotein gene grlp bearing a function in regulating the periplasmic space. This finding suggests a coordination between the OM properties and the T3SS gene regulation and reveals a new target for control of the T3SS gene expression and bacterial pathogenicity.
Collapse
|
42
|
Weber A, Gibisch M, Tyrakowski D, Cserjan-Puschmann M, Toca-Herrera JL, Striedner G. Recombinant Peptide Production Softens Escherichia coli Cells and Increases Their Size during C-Limited Fed-Batch Cultivation. Int J Mol Sci 2023; 24:2641. [PMID: 36768962 PMCID: PMC9916741 DOI: 10.3390/ijms24032641] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Stress-associated changes in the mechanical properties at the single-cell level of Escherichia coli (E. coli) cultures in bioreactors are still poorly investigated. In our study, we compared peptide-producing and non-producing BL21(DE3) cells in a fed-batch cultivation with tightly controlled process parameters. The cell growth, peptide content, and cell lysis were analysed, and changes in the mechanical properties were investigated using atomic force microscopy. Recombinant-tagged somatostatin-28 was expressed as soluble up to 197 ± 11 mg g-1. The length of both cultivated strains increased throughout the cultivation by up to 17.6%, with nearly constant diameters. The peptide-producing cells were significantly softer than the non-producers throughout the cultivation, and respective Young's moduli decreased by up to 57% over time. A minimum Young's modulus of 1.6 MPa was observed after 23 h of the fed-batch. Furthermore, an analysis of the viscoelastic properties revealed that peptide-producing BL21(DE3) appeared more fluid-like and softer than the non-producing reference. For the first time, we provide evidence that the physical properties (i.e., the mechanical properties) on the single-cell level are significantly influenced by the metabolic burden imposed by the recombinant peptide expression and C-limitation in bioreactors.
Collapse
Affiliation(s)
- Andreas Weber
- Christian Doppler Laboratory for Production of Next-Level Biopharmaceuticals in E. coli, Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
- Institute of Biophysics, Department of Bionanosciences, University of Natural Resources and Life Sciences, Muthgasse 11, 1190 Vienna, Austria
| | - Martin Gibisch
- Christian Doppler Laboratory for Production of Next-Level Biopharmaceuticals in E. coli, Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| | - Daniel Tyrakowski
- Institute of Biophysics, Department of Bionanosciences, University of Natural Resources and Life Sciences, Muthgasse 11, 1190 Vienna, Austria
| | - Monika Cserjan-Puschmann
- Christian Doppler Laboratory for Production of Next-Level Biopharmaceuticals in E. coli, Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| | - José L. Toca-Herrera
- Institute of Biophysics, Department of Bionanosciences, University of Natural Resources and Life Sciences, Muthgasse 11, 1190 Vienna, Austria
| | - Gerald Striedner
- Christian Doppler Laboratory for Production of Next-Level Biopharmaceuticals in E. coli, Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| |
Collapse
|
43
|
Sheng Q, Zhang MY, Liu SM, Chen ZW, Yang PL, Zhang HS, Liu MY, Li K, Zhao LS, Liu NH, Liu LN, Chen XL, Hobbs JK, Foster SJ, Zhang YZ, Su HN. In situ visualization of Braun's lipoprotein on E. coli sacculi. SCIENCE ADVANCES 2023; 9:eadd8659. [PMID: 36662863 PMCID: PMC9858504 DOI: 10.1126/sciadv.add8659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 12/20/2022] [Indexed: 06/17/2023]
Abstract
Braun's lipoprotein (Lpp) plays a major role in stabilizing the integrity of the cell envelope in Escherichia coli, as it provides a covalent cross-link between the outer membrane and the peptidoglycan layer. An important challenge in elucidating the physiological role of Lpp lies in attaining a detailed understanding of its distribution on the peptidoglycan layer. Here, using atomic force microscopy, we visualized Lpp directly on peptidoglycan sacculi. Lpp is homogeneously distributed over the outer surface of the sacculus at a high density. However, it is absent at the constriction site during cell division, revealing its role in the cell division process with Pal, another cell envelope-associated protein. Collectively, we have established a framework to elucidate the distribution of Lpp and other peptidoglycan-bound proteins via a direct imaging modality.
Collapse
Affiliation(s)
- Qi Sheng
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
- College of Marine Life Sciences and Frontiers Science Center for Deep Ocean Multispheres and Earth System, Ocean University of China, Qingdao 266003, China
| | - Meng-Yao Zhang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Si-Min Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Zhuo-Wei Chen
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Pei-Ling Yang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Hong-Su Zhang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Meng-Yun Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Kang Li
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Long-Sheng Zhao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Ning-Hua Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Lu-Ning Liu
- College of Marine Life Sciences and Frontiers Science Center for Deep Ocean Multispheres and Earth System, Ocean University of China, Qingdao 266003, China
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Xiu-Lan Chen
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - 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
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, UK
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Yu-Zhong Zhang
- College of Marine Life Sciences and Frontiers Science Center for Deep Ocean Multispheres and Earth System, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao 266237, China
- Marine Biotechnology Research Center, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Hai-Nan Su
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao 266237, China
| |
Collapse
|
44
|
Voedts H, Kennedy SP, Sezonov G, Arthur M, Hugonnet JE. Genome-wide identification of genes required for alternative peptidoglycan cross-linking in Escherichia coli revealed unexpected impacts of β-lactams. Nat Commun 2022; 13:7962. [PMID: 36575173 PMCID: PMC9794725 DOI: 10.1038/s41467-022-35528-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 12/06/2022] [Indexed: 12/28/2022] Open
Abstract
The D,D-transpeptidase activity of penicillin-binding proteins (PBPs) is the well-known primary target of β-lactam antibiotics that block peptidoglycan polymerization. β-lactam-induced bacterial killing involves complex downstream responses whose causes and consequences are difficult to resolve. Here, we use the functional replacement of PBPs by a β-lactam-insensitive L,D-transpeptidase to identify genes essential to mitigate the effects of PBP inactivation by β-lactams in actively dividing bacteria. The functions of the 179 conditionally essential genes identified by this approach extend far beyond L,D-transpeptidase partners for peptidoglycan polymerization to include proteins involved in stress response and in the assembly of outer membrane polymers. The unsuspected effects of β-lactams include loss of the lipoprotein-mediated covalent bond that links the outer membrane to the peptidoglycan, destabilization of the cell envelope in spite of effective peptidoglycan cross-linking, and increased permeability of the outer membrane. The latter effect indicates that the mode of action of β-lactams involves self-promoted penetration through the outer membrane.
Collapse
Affiliation(s)
- Henri Voedts
- grid.417925.cCentre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France
| | - Sean P. Kennedy
- Institut Pasteur, Université Paris Cité, Département Biologie Computationnelle, F-75015 Paris, France
| | - Guennadi Sezonov
- grid.417925.cCentre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France
| | - Michel Arthur
- grid.417925.cCentre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France
| | - Jean-Emmanuel Hugonnet
- grid.417925.cCentre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France
| |
Collapse
|
45
|
Weber A, Tyrakowski D, Toca-Herrera JL. Power Laws Describe Bacterial Viscoelasticity. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:15552-15558. [PMID: 36484724 PMCID: PMC9776528 DOI: 10.1021/acs.langmuir.2c02172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/24/2022] [Indexed: 06/17/2023]
Abstract
Bacterial cells survive in a wide range of different environments and actively tune their mechanical properties for purposes of growth, movement, division, and nutrition. In Gram-negative bacteria, the cell envelope with its outer membrane and peptidoglycan are the main determinants of mechanical properties and are common targets for the use of antibiotics. The study of bacterial mechanical properties has shown promise in elucidating a structure-function relationship in bacteria, connecting, shape, mechanics, and biochemistry. In this work, we study frequency and time-dependent viscoelastic properties of E. coli cells by atomic force microscopy (AFM). We perform force cycles, oscillatory microrheology, stress relaxation, and creep experiments, and use power law rheology models to fit the experimental results. All data sets could be fitted with the models and provided power law exponents of 0.01 to 0.1 while showing moduli in the range of a few MPa. We provide evidence for the interchangeability of the properties derived from these four different measurement approaches.
Collapse
|
46
|
Rodrigues IC, Rodrigues SC, Duarte FV, da Costa PM, da Costa PM. The Role of Outer Membrane Proteins in UPEC Antimicrobial Resistance: A Systematic Review. MEMBRANES 2022; 12:981. [PMID: 36295740 PMCID: PMC9609314 DOI: 10.3390/membranes12100981] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 06/16/2023]
Abstract
Uropathogenic Escherichia coli (UPEC) are one of the most common agents of urinary tract infection. In the last decade, several UPEC strains have acquired antibiotic resistance mechanisms and some have become resistant to all classes of antibiotics. UPEC outer membrane proteins (OMPs) seem to have a decisive role not only in the processes of invasion and colonization of the bladder mucosa, but also in mechanisms of drug resistance, by which bacteria avoid killing by antimicrobial molecules. This systematic review was performed according to the PRISMA guidelines, aiming to characterize UPEC OMPs and identify their potential role in antimicrobial resistance. The search was limited to studies in English published during the last decade. Twenty-nine studies were included for revision and, among the 76 proteins identified, seven were associated with antibiotic resistance. Indeed, OmpC was associated with β-lactams resistance and OmpF with β-lactams and fluoroquinolone resistance. In turn, TolC, OmpX, YddB, TosA and murein lipoprotein (Lpp) were associated with fluoroquinolones, enrofloxacin, novobiocin, β-lactams and globomycin resistances, respectively. The clinical implications of UPEC resistance to antimicrobial agents in both veterinary and human medicine must propel the implementation of new strategies of administration of antimicrobial agents, while also promoting the development of improved antimicrobials, protective vaccines and specific inhibitors of virulence and resistance factors.
Collapse
Affiliation(s)
- Inês C. Rodrigues
- Laboratório de Microbiologia e Tecnologia Alimentar, Departamento de Produção Aquática, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Sílvia C. Rodrigues
- Pharmaissues, Consultoria, Lda, Rua da Esperança n° 101, Ribeira de Frades, 3045-420 Coimbra, Portugal
| | - Filipe V. Duarte
- Centro de Neurociências e Biologia Celular (CNC), Faculdade de Medicina, Pólo 1, Universidade de Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
| | - Paula M. da Costa
- Microbiology Department, Centro Hospitalar Universitário do Porto, Largo do Prof. Abel Salazar, 4099-001 Porto, Portugal
| | - Paulo M. da Costa
- Laboratório de Microbiologia e Tecnologia Alimentar, Departamento de Produção Aquática, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), Terminal de Cruzeiros do Porto, de Lexões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal
| |
Collapse
|
47
|
Thomson NM, Turner AK, Yasir M, Bastkowski S, Lott M, Webber MA, Charles IG. A whole-genome assay identifies four principal gene functions that confer tolerance of meropenem stress upon Escherichia coli. FRONTIERS IN ANTIBIOTICS 2022; 1:957942. [PMID: 39816415 PMCID: PMC11731830 DOI: 10.3389/frabi.2022.957942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/23/2022] [Indexed: 01/18/2025]
Abstract
We report here the identification of four gene functions of principal importance for the tolerance of meropenem stress in Escherichia coli: cell division, cell envelope synthesis and maintenance, ATP metabolism, and transcription regulation. The primary mechanism of β-lactam antibiotics such as meropenem is inhibition of penicillin binding proteins, thus interfering with peptidoglycan crosslinking, weakening the cell envelope, and promoting cell lysis. However, recent systems biology approaches have revealed numerous downstream effects that are triggered by cell envelope damage and involve diverse cell processes. Subpopulations of persister cells can also arise, which can survive elevated concentrations of meropenem despite the absence of a specific resistance factor. We used Transposon-Directed Insertion Sequencing with inducible gene expression to simultaneously assay the effects of upregulation, downregulation, and disruption of every gene in a model E. coli strain on survival of exposure to four concentrations of meropenem. Automated Gene Functional Classification and manual categorization highlighted the importance at all meropenem concentrations of genes involved in peptidoglycan remodeling during cell division, suggesting that cell division is the primary function affected by meropenem. Genes involved in cell envelope synthesis and maintenance, ATP metabolism, and transcriptional regulation were generally important at higher meropenem concentrations, suggesting that these three functions are therefore secondary or downstream targets. Our analysis revealed the importance of multiple two-component signal transduction mechanisms, suggesting an as-yet unexplored coordinated transcriptional response to meropenem stress. The inclusion of an inducible, transposon-encoded promoter allowed sensitive detection of genes involved in proton transport, ATP production and tRNA synthesis, for which modulation of expression affects survival in the presence of meropenem: a finding that would not be possible with other technologies. We were also able to suggest new targets for future antibiotic development or for synergistic effects between gene or protein inhibitors and existing antibiotics. Overall, in a single massively parallel assay we were able to recapitulate many of the findings from decades of research into β-lactam antibiotics, add to the list of genes known to be important for meropenem tolerance, and categorize the four principal gene functions involved.
Collapse
Affiliation(s)
- Nicholas M. Thomson
- Microbes in the Food Chain, Quadram Institute Bioscience, Norwich, United Kingdom
| | - A. Keith Turner
- Microbes in the Food Chain, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Muhammad Yasir
- Microbes in the Food Chain, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Sarah Bastkowski
- Microbes in the Food Chain, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Martin Lott
- Microbes in the Food Chain, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Mark A. Webber
- Microbes in the Food Chain, Quadram Institute Bioscience, Norwich, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Ian G. Charles
- Microbes in the Food Chain, Quadram Institute Bioscience, Norwich, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
48
|
A Defect in Lipoprotein Modification by Lgt Leads to Abnormal Morphology and Cell Death in Escherichia coli That Is Independent of Major Lipoprotein Lpp. J Bacteriol 2022; 204:e0016422. [PMID: 35938851 PMCID: PMC9487459 DOI: 10.1128/jb.00164-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Lgt is an essential enzyme in proteobacteria and therefore a potential target for novel antibiotics. The effect of Lgt depletion on growth, morphology, and viability was studied in Escherichia coli to assess whether absence of Lgt leads to cell death. Two Lgt depletion strains were used in which lgt was under the control of an arabinose-inducible promoter that allowed regulation of Lgt protein levels. Reduced levels of Lgt led to severe growth and morphological defects that could be restored by expressing lgt in trans, demonstrating that only Lgt is responsible for the distorted phenotypes. In the absence of major lipoprotein Lpp, growth defects were partially restored when low levels of Lgt were still present; however, lgt could not be deleted in the absence of Lpp. Our results demonstrate that Lpp is not the main cause of cell death under conditions of Lgt depletion and that other lipoproteins are important in cell envelope biogenesis and cell viability. Specific inhibitors of Lgt are thus promising for the development of novel antibiotics. IMPORTANCE Incomplete maturation and envelope mislocalization of lipoproteins, through inhibition or mutations in lipoprotein modification enzymes or transport to the outer membrane, are lethal in proteobacteria. Resistance to small-molecule inhibition or the appearance of suppressor mutations is often directly correlated with the presence of abundant outer membrane lipoprotein Lpp. Our results show that Lgt, the first enzyme of the lipoprotein modification pathway, is still required for growth and viability in the absence of Lpp and thus is necessary for the function of other essential lipoproteins in the cell envelope. This adds credence to the hypothesis that Lgt is essential in proteobacteria and an attractive target for the development of novel antibiotics.
Collapse
|
49
|
Lazenby JJ, Li ES, Whitchurch CB. Cell wall deficiency - an alternate bacterial lifestyle? MICROBIOLOGY (READING, ENGLAND) 2022; 168. [PMID: 35925044 DOI: 10.1099/mic.0.001218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Historically, many species of bacteria have been reported to produce viable, cell wall deficient (CWD) variants. A variety of terms have been used to refer to CWD bacteria and a plethora of methods described in which to induce, cultivate and propagate them. In this review, we will examine the long history of scientific research on CWD bacteria examining the methods by which CWD bacteria are generated; the requirements for survival in a CWD state; the replicative processes within a CWD state; and the reversion of CWD bacteria into a walled state, or lack thereof. In doing so, we will present evidence that not all CWD variants are alike and that, at least in some cases, CWD variants arise through an adaptive lifestyle switch that enables them to live and thrive without a cell wall, often to avoid antimicrobial activity. Finally, the implications of CWD bacteria in recurring infections, tolerance to antibiotic therapy and antimicrobial resistance will be examined to illustrate the importance of greater understanding of the CWD bacteria in human health and disease.
Collapse
Affiliation(s)
- James J Lazenby
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Erica S Li
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Cynthia B Whitchurch
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TK, UK
| |
Collapse
|
50
|
Kim M, Im E, Jin Ahn Y. Enhanced Acetate Tolerance and Recombinant Protein Accumulation in Escherichia coli by Transgenic Expression of a Heat Shock Protein from Carrot ( Daucus carota L.). IRANIAN JOURNAL OF BIOTECHNOLOGY 2022; 20:e3177. [PMID: 36381282 PMCID: PMC9618011 DOI: 10.30498/ijb.2022.309657.3177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND In Escherichia coli (E. coli) culture, acetate accumulates as an undesirable by-product of aerobic fermentation on glucose and inhibits cell growth and recombinant protein production. OBJECTIVES We examined whether the heterologous expression of a eukaryotic heat shock protein (Hsp) can confer tolerance to acetate in E. coli. MATERIALS AND METHODS Transgenic cell lines (TCLs) heterologously expressing a small heat shock protein (sHsp) from carrot (Daucus carota L.), DcHsp17.7, were exposed to heat, sodium acetate, and alkaline conditions. The cell growth and cell viability were examined by measuring O.D.600 and colony-forming units (CFU), respectively. The His-tagged recombinant alcohol dehydrogenase (ADH) gene cloned in a pET11a expression vector was introduced into TCL1 and expressed by isopropyl β-D-1-thiogalactopyranoside treatment. After purifying using Ni-NTA affinity chromatography, its accumulation levels were examined using SDS-PAGE in the presence of acetate. RESULTS TCLs constitutively expressing DcHsp17.7 showed improved growth, cell density, and cell viability under the stress conditions of heat, acetate, and alkaline compared to an empty vector control line. In acetate stress conditions, TCL1 accumulated more cellular proteins (approximately 130%) than the control. The recombinant ADH accumulated to a higher level in TCL1 (2.2-fold at 16 °C) than the control. The addition of acetate reduced the recombinant ADH level by 70% in the control when compared with the absence of acetate. In contrast, recombinant ADH accumulation was not affected by acetate in TCL1. In the presence of acetate, TCL1 accumulated 6.4-fold more recombinant ADH than did the control. Furthermore, recombinant ADH produced in TCL1 showed 1.5-fold higher enzyme activity than that produced in the control in the presence or absence of acetate. CONCLUSION Our study showed that heterologously expressed DcHsp17.7 from carrot can alleviate the negative effects of acetate on E. coli.
Collapse
Affiliation(s)
- Minhye Kim
- Department of Biotechnology, Sangmyung University, 20 Hongjimun 2-gil, Jongno-gu, Seoul 03016, Korea
| | - Eunju Im
- Department of Biotechnology, Sangmyung University, 20 Hongjimun 2-gil, Jongno-gu, Seoul 03016, Korea
| | - Yeh Jin Ahn
- Department of Biotechnology, Sangmyung University, 20 Hongjimun 2-gil, Jongno-gu, Seoul 03016, Korea
| |
Collapse
|