1
|
Gerschler S, Maaß S, Gerth P, Schulig L, Wildgrube T, Rockstroh J, Wurster M, Methling K, Becher D, Lalk M, Schulze C, Guenther S, Schultze N. Drosera rotundifolia L. as E. coli biofilm inhibitor: Insights into the mechanism of action using proteomics/metabolomics and toxicity studies. Biofilm 2025; 9:100268. [PMID: 40124935 PMCID: PMC11930149 DOI: 10.1016/j.bioflm.2025.100268] [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: 12/10/2024] [Revised: 02/25/2025] [Accepted: 02/28/2025] [Indexed: 03/25/2025] Open
Abstract
The successful sustainable cultivation of the well-known medicinal plant sundew on rewetted peatlands not only leads to the preservation of natural populations, but also provides a basis for the sustainable pharmaceutical use of the plant. The bioactive compounds of sundew, flavonoids and naphthoquinones, show biofilm-inhibiting properties against multidrug-resistant, ESBL-producing E. coli strains and open up new therapeutic possibilities. This study investigates the molecular mechanisms of these compounds in biofilm inhibition through proteomic analyses. Specific fractions of flavonoids and naphthoquinones, as well as individual substances like 7-methyljuglone and 2″-O-galloylhyperoside, are analyzed. Results show that naphthoquinones appear to act via central regulatory proteins such as OmpR and alter the stress response while flavonoids likely affect biofilm formation by creating an iron-poor environment through iron complexation and additionally influence polyamine balance, reducing intracellular spermidine levels. Further investigations including assays for iron complexation and analysis of polyamines confirmed the proteomic data. Safety evaluations through cytotoxicity tests in 3D cell cultures and the Galleria mellonella in vivo model confirm the safety of the extracts used. These findings highlight sundew as a promising candidate for new phytopharmaceuticals.
Collapse
Affiliation(s)
- Sandy Gerschler
- Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Straße 17, 17489, Greifswald, Germany
| | - Sandra Maaß
- Institute of Microbiology, University of Greifswald, Felix-Hausdorff-Straße 8, 17489, Greifswald, Germany
| | - Philip Gerth
- Institute of Microbiology, University of Greifswald, Felix-Hausdorff-Straße 8, 17489, Greifswald, Germany
| | - Lukas Schulig
- Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Straße 17, 17489, Greifswald, Germany
| | - Toni Wildgrube
- Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Straße 17, 17489, Greifswald, Germany
| | - Jan Rockstroh
- Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Straße 4, 17489, Greifswald, Germany
| | - Martina Wurster
- Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Straße 4, 17489, Greifswald, Germany
| | - Karen Methling
- Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Straße 4, 17489, Greifswald, Germany
| | - Dörte Becher
- Institute of Microbiology, University of Greifswald, Felix-Hausdorff-Straße 8, 17489, Greifswald, Germany
| | - Michael Lalk
- Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Straße 4, 17489, Greifswald, Germany
| | - Christian Schulze
- Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Straße 17, 17489, Greifswald, Germany
| | - Sebastian Guenther
- Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Straße 17, 17489, Greifswald, Germany
- Partner in the Greifswald Mire Centre, Soldmannstr. 15, 17487 Greifswald, Germany
| | - Nadin Schultze
- Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Straße 17, 17489, Greifswald, Germany
| |
Collapse
|
2
|
Lamb ER, Criss AK. Terminal complement complexes with or without C9 potentiate antimicrobial activity against Neisseria gonorrhoeae. mBio 2025; 16:e0014125. [PMID: 40162779 DOI: 10.1128/mbio.00141-25] [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: 01/10/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
The complement cascade is a front-line defense against pathogens. Complement activation generates the membrane attack complex (MAC), a 10-11 nm diameter pore formed by complement proteins C5b through C8 and polymerized C9. The MAC embeds within the outer membrane of Gram-negative bacteria and displays bactericidal activity. In the absence of C9, C5b-C8 complexes can form 2-4 nm pores on membranes, but their relevance to microbial control is poorly understood. Deficiencies in terminal complement components uniquely predispose individuals to infections by pathogenic Neisseria, including N. gonorrhoeae (Gc). Increasing antibiotic resistance in Gc makes new therapeutic strategies a priority. Here, we demonstrate that MAC formed by complement activity in human serum disrupts the Gc outer and inner membranes, potentiating the activity of antimicrobials against Gc and re-sensitizing multidrug-resistant Gc to antibiotics. C9-depleted serum also exerts bactericidal activity against Gc and, unlike other Gram-negative bacteria, disrupts both the outer and inner membranes. C5b-C8 complex formation potentiates Gc sensitivity to azithromycin and ceftriaxone, but not lysozyme or nisin. These findings expand our mechanistic understanding of complement lytic activity, suggest a size limitation for terminal complement-mediated enhancement of antimicrobials against Gc, and suggest that complement manipulation can be used to combat drug-resistant gonorrhea. IMPORTANCE The complement cascade is a front-line arm of the innate immune system against pathogens. Complement activation results in membrane attack complex (MAC) pores forming on the outer membrane of Gram-negative bacteria, resulting in bacterial death. Individuals who cannot generate MAC are specifically susceptible to infection by pathogenic Neisseria species including N. gonorrhoeae (Gc). High rates of gonorrhea, its complications like infertility, and high-frequency resistance to multiple antibiotics make it important to identify new approaches to combat Gc. Beyond direct anti-Gc activity, we found that the MAC increases the ability of antibiotics and antimicrobial proteins to kill Gc and re-sensitizes multidrug-resistant bacteria to antibiotics. The most terminal component, C9, is needed to potentiate the anti-Gc activity of lysozyme and nisin, but azithromycin and ceftriaxone activity is potentiated regardless of C9. These findings highlight the unique effects of MAC on Gc and suggest novel translational avenues to combat drug-resistant gonorrhea.
Collapse
Affiliation(s)
- Evan R Lamb
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Alison K Criss
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
3
|
Shi H, Bai H, Deng H, Xia Y. Revealing Acyl Chain Selectivity of Cis-to-Trans Isomerase through Profiling of C═C Geometry and Location Isomers of Bacterial Lipids. Anal Chem 2025. [PMID: 40331359 DOI: 10.1021/acs.analchem.5c00675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Bacteria adapt to environmental stress by modifying their membrane lipid structures, including the C═C geometry. Profiling of bacterial lipids with accurate C═C geometry assignment is challenging due to the lack of standards and interference from C═C location isomers. By leveraging two radical reactions: thiyl radical-catalyzed C═C isomerization and the Paternò-Büchi (PB) reaction, we developed an analytical workflow to profile C═C geometric and location isomers in bacterial lipidomes. The high yield (∼80%) of cis (Z)-to-trans (E) C═C isomerization catalyzed by thiyl radicals allows for on-demand synthesis of commercially unavailable lipid C═C geometric isomers. By comparing the retention behavior of Z vs E isomers from reversed-phase liquid chromatography-mass spectrometry (RPLC-MS), we can determine C═C geometry at sub-nM levels. The location of C═C can be further obtained by conducting an online acetone PB reaction after RPLC separation. Applying this workflow to Pseudomonas putida, we profiled 60 lipid species across six subclasses, including the rarely reported glucosaminyl phosphatidylglycerol. We found that both Z and E isomers were present in bacterial lipids, however, with an increase in E isomers after toluene exposure, which correlated with an upregulation of cis-to-trans isomerase (Cti). Our workflow further revealed the chain selectivity of Cti, with a preference for C16:1(n-7Z) > C18:1(n-7Z) > C18:1(n-9Z). This finding provides valuable insights into the dynamics of lipid metabolism during bacterial stress responses.
Collapse
Affiliation(s)
- Hengxue Shi
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 10084, China
| | - Huijiao Bai
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing 100730, China
| | - Yu Xia
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 10084, China
| |
Collapse
|
4
|
Wang L, Xie J, Gong T, Wu H, Tu Y, Peng X, Shang S, Jia X, Ma H, Zou J, Xu S, Zheng X, Zhang D, Liu Y, Zhang C, Luo Y, Huang Z, Shao B, Ying B, Cheng Y, Guo Y, Lai Y, Huang D, Liu J, Wei Y, Sun S, Zhou X, Su Z. Cryo-EM reveals mechanisms of natural RNA multivalency. Science 2025; 388:545-550. [PMID: 40080543 DOI: 10.1126/science.adv3451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
Homo-oligomerization of biological macromolecules leads to functional assemblies that are critical to understanding various cellular processes. However, RNA quaternary structures have rarely been reported. Comparative genomics analysis has identified RNA families containing hundreds of sequences that adopt conserved secondary structures and likely fold into complex three-dimensional structures. In this study, we used cryo-electron microscopy (cryo-EM) to determine structures from four RNA families, including ARRPOF and OLE forming dimers and ROOL and GOLLD forming hexameric, octameric, and dodecameric nanostructures, at 2.6- to 4.6-angstrom resolutions. These homo-oligomeric assemblies reveal a plethora of structural motifs that contribute to RNA multivalency, including kissing-loop, palindromic base-pairing, A-stacking, metal ion coordination, pseudoknot, and minor-groove interactions. These results provide the molecular basis of intermolecular interactions driving RNA multivalency with potential functional relevance.
Collapse
Affiliation(s)
- Liu Wang
- The State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital; The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | | | - Tao Gong
- The State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital; The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hao Wu
- Research Institute of Intelligent Complex Systems, Fudan University, Shanghai, China
- Shanghai Artificial Intelligence Laboratory, Shanghai, China
| | - Yifan Tu
- The Key Laboratory for Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Xin Peng
- The Key Laboratory for Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Sitong Shang
- The Key Laboratory for Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Xinyu Jia
- The State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital; The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Haiyun Ma
- The State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital; The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jian Zou
- The State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital; The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Sheng Xu
- Research Institute of Intelligent Complex Systems, Fudan University, Shanghai, China
- Shanghai Artificial Intelligence Laboratory, Shanghai, China
| | - Xin Zheng
- The State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital; The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dong Zhang
- The Key Laboratory for Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yang Liu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Chong Zhang
- The State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital; The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yongbo Luo
- The State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital; The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zirui Huang
- The State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital; The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bin Shao
- The State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital; The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Cheng
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Yingqiang Guo
- Cardiovascular Surgery Research Laboratory, Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Lai
- The State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital; The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dingming Huang
- The State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital; The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jianquan Liu
- The Key Laboratory for Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yuquan Wei
- The State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital; The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Siqi Sun
- Research Institute of Intelligent Complex Systems, Fudan University, Shanghai, China
- Shanghai Artificial Intelligence Laboratory, Shanghai, China
| | - Xuedong Zhou
- The State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital; The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhaoming Su
- The State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital; The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Venkatraman K, Lipp NF, Budin I. Origin and evolution of mitochondrial inner membrane composition. J Cell Sci 2025; 138:jcs263780. [PMID: 40265338 DOI: 10.1242/jcs.263780] [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: 04/24/2025] Open
Abstract
Unique membrane architectures and lipid building blocks underlie the metabolic and non-metabolic functions of mitochondria. During eukaryogenesis, mitochondria likely arose from an alphaproteobacterial symbiont of an Asgard archaea-related host cell. Subsequently, mitochondria evolved inner membrane folds known as cristae alongside a specialized lipid composition supported by metabolic and transport machinery. Advancements in phylogenetic methods and genomic and metagenomic data have suggested potential origins for cristae-shaping protein complexes, such as the mitochondrial contact site and cristae-organizing system (MICOS). MICOS protein homologs function in the formation of cristae-like intracytoplasmic membranes (ICMs) in diverse extant alphaproteobacteria. The machinery responsible for synthesizing key mitochondrial phospholipids - which cooperate with cristae-shaping proteins to establish inner membrane architecture - could have also evolved from a bacterial ancestor, but its origins have been less explored. In this Review, we examine the current understanding of mitochondrial membrane evolution, highlighting distinctions between prokaryotic and eukaryotic mitochondrial-specific proteins and lipids and their differing roles in shaping cristae and ICM architecture, and propose a model explaining the concurrent specialization of the mitochondrial lipidome and inner membrane structure in eukaryogenesis. We discuss how advancements across a range of disciplines are shedding light on how multiple membrane components co-evolved to support the central functions of eukaryotic mitochondria.
Collapse
Affiliation(s)
- Kailash Venkatraman
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - Nicolas-Frédéric Lipp
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - Itay Budin
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
6
|
Helms A, Brodbelt JS. Progress in Characterization of Lipopolysaccharides and Lipid A by Mass Spectrometry. MASS SPECTROMETRY REVIEWS 2025. [PMID: 40302133 DOI: 10.1002/mas.21934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 04/11/2025] [Accepted: 04/13/2025] [Indexed: 05/01/2025]
Abstract
Lipopolysaccharides (LPS) are complex molecules embedded in the outer membrane of Gram-negative bacteria. LPS are highly heterogeneous across species and strains, posing a significant analytical challenge. This review article explores recent advances in the identification and characterization of LPS, with a particular focus on the role of mass spectrometry (MS) techniques. The review highlights how MS, in conjunction with various separation methods and spanning different ionization techniques and dissociation modes, has enabled more precise and sensitive determination of LPS composition, with a focus on lipid A structure. Finally, emerging trends in MS applications for LPS research are discussed and its potential to provide deeper insights into the development of antibiotic resistance among pathogenic bacteria.
Collapse
Affiliation(s)
- Amanda Helms
- Department of Chemistry, University of Texas at Austin, Austin, Texas, USA
| | | |
Collapse
|
7
|
Chiang MC, Clarke BR, Tew GN, Schiffman JD. Antifouling Activity of Bottlebrush Network Hydrogels. ACS APPLIED BIO MATERIALS 2025. [PMID: 40270321 DOI: 10.1021/acsabm.5c00291] [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: 04/25/2025]
Abstract
Mitigating the attachment of microorganisms to polymer biomaterials is critical for preventing hospital-acquired infections. Two chemical strategies to mitigate fouling include fabricating fouling-resistant surfaces, which typically present hydrophilic polymers, such as polyethylene glycol (PEG), or creating fouling-release surfaces, which are generally hydrophobic featuring polydimethylsiloxane (PDMS). Despite the demonstrated promise of employing PEG or PDMS, amphiphilic PEG/PDMS copolymer materials remain understudied. Here, for the first time, we investigated if phase-separated amphiphilic copolymers confounded microbial adhesion. We used bottlebrush amphiphilic PEG/PDMS co-networks and homopolymer networks to study bacterial adhesion across a library of gels (ϕPEG = 0.00, 0.21, 0.40, 0.55, 0.80, and 1.00). Hydrated atomic force microscopy measurements revealed that most of the gels had low surface roughness, less than 5 nm, and an elastic modulus of ∼80 kPa. Interestingly, the surface roughness and elastic modulus of the ϕPEG = 0.40 gel were twice as high as those of the other gels due to the presence of crystalline domains, as confirmed using polarized optical microscopy on the hydrated gel. The interactions of these six well-characterized gels with bacteria were determined using Escherichia coli K12 MG1655 and Staphylococcus aureus SH1000. The attachment of both microbes decreased by at least 60% on all polymer gels versus the glass controls. S. aureus adhesion peaked on the ϕPEG = 0.40, likely due to its increased elastic modulus, consistent with previous literature demonstrating that modulus impacts microbial adhesion. These findings suggest that hydrophilic, hydrophobic, and amphiphilic biomaterials effectively resist the early attachment of Gram-negative and Gram-positive microorganisms, providing guidance for the design of next-generation antifouling surfaces.
Collapse
Affiliation(s)
- Meng-Chen Chiang
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003-9303, United States
| | - Brandon R Clarke
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Gregory N Tew
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Jessica D Schiffman
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003-9303, United States
- Materials Science and Engineering Graduate Program, University of Massachusetts Amherst, Amherst, Massachusetts 01003-9303, United States
| |
Collapse
|
8
|
Ganusova EE, Banerjee I, Seats T, Alexandre G. Indole-3-acetic acid (IAA) protects Azospirillum brasilense from indole-induced stress. Appl Environ Microbiol 2025; 91:e0238424. [PMID: 40130845 PMCID: PMC12016523 DOI: 10.1128/aem.02384-24] [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: 12/02/2024] [Accepted: 02/20/2025] [Indexed: 03/26/2025] Open
Abstract
Azospirillum brasilense is plant-growth promoting rhizobacteria that produces the phytohormone indole-3-acetic acid (IAA) to induce changes in plant root architecture. The major pathway for IAA biosynthesis in A. brasilense converts tryptophan into indole-3-pyruvic acid (I3P) and then, through the rate-limiting enzyme, indole-3-pyruvate decarboxylase (IpdC), into IAA. Here, we characterize the potential role for IAA biosynthesis in the physiology of these bacteria by characterizing the expression pattern of the ipdC promoter, analyzing an A. brasilense ipdC mutant using multiple physiological assays and characterizing the effect of I3P, which likely accumulates in the absence of ipdC and affects bacterial physiology. We found that the ipdC mutant derivative has a reduced growth rate and an altered physiology, including reduced translation activity as well as a more depolarized membrane potential compared to the parent strain. Similar effects could be recapitulated in the parent strain by exposing these cells to increasing concentrations of I3P, as well as other indole intermediates of IAA biosynthesis. Our results also indicate a protective role for IAA against the harmful effects of indole derivatives, with exogenous IAA restoring the membrane potential of cells exposed to indole derivatives for prolonged periods. These protective effects appeared to restore cell physiology, including in the wheat rhizosphere. Together, our data suggest that the IAA biosynthesis pathway plays a major role in A. brasilense physiology by maintaining membrane potential homeostasis and regulating translation, likely to mitigate the potential membrane-damaging effects of indoles that accumulate during growth under stressful conditions.IMPORTANCEIAA is widely synthesized in bacteria, particularly in soil and rhizosphere bacteria, where it functions as a phytohormone to modulate plant root architecture. IAA as a secondary metabolite has been shown to serve as a signaling molecule in several bacterial species, but the role of IAA biosynthesis in the physiology of the producing bacterium remains seldom explored. Results obtained here suggest that IAA serves to protect A. brasilense from the toxic effect of indoles, including metabolite biosynthetic precursors of IAA, on membrane potential homeostasis. Given the widespread production of IAA in soil bacteria, this protective effect of IAA may be conserved in diverse soil bacteria.
Collapse
Affiliation(s)
- Elena E. Ganusova
- Biochemistry and Cellular and Molecular Biology Department, University of Tennessee, Knoxville, Tennessee, USA
| | - Ishita Banerjee
- Biochemistry and Cellular and Molecular Biology Department, University of Tennessee, Knoxville, Tennessee, USA
| | - Trey Seats
- Biochemistry and Cellular and Molecular Biology Department, University of Tennessee, Knoxville, Tennessee, USA
| | - Gladys Alexandre
- Biochemistry and Cellular and Molecular Biology Department, University of Tennessee, Knoxville, Tennessee, USA
| |
Collapse
|
9
|
Karimullina E, Guo Y, Khan HM, Emde T, Quade B, Di Leo R, Otwinowski Z, Tieleman DP, Borek D, Savchenko A. Structural architecture of TolQ-TolR inner membrane protein complex from opportunistic pathogen Acinetobacter baumannii. SCIENCE ADVANCES 2025; 11:eadq9845. [PMID: 40184442 PMCID: PMC11970459 DOI: 10.1126/sciadv.adq9845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 02/28/2025] [Indexed: 04/06/2025]
Abstract
Gram-negative bacteria harness the proton motive force (PMF) within their inner membrane (IM) to uphold cell envelope integrity, an indispensable aspect for both division and survival. The IM TolQ-TolR complex is the essential part of the Tol-Pal system, serving as a conduit for PMF energy transfer to the outer membrane. Here we present cryo-electron microscopy reconstructions of Acinetobacter baumannii TolQ in apo and TolR-bound forms at atomic resolution. The apo TolQ configuration manifests as a symmetric pentameric pore, featuring a transmembrane funnel leading toward a cytoplasmic chamber. In contrast, the TolQ-TolR complex assumes a proton nonpermeable stance, characterized by the TolQ pentamer's flexure to accommodate the TolR dimer, where two protomers undergo a translation-based relationship. Our structure-guided analysis and simulations support the rotor-stator mechanism of action, wherein the rotation of the TolQ pentamer harmonizes with the TolR protomers' interplay. These findings broaden our mechanistic comprehension of molecular stator units empowering critical functions within the Gram-negative bacterial cell envelope.
Collapse
Affiliation(s)
- Elina Karimullina
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta T2N 4N1, Canada
- Center for Structural Biology of Infectious Diseases (CSBID), Chicago, IL 60611, USA
| | - Yirui Guo
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Ligo Analytics, 2207 Chunk Ct., Dallas, TX 75206, USA
| | - Hanif M. Khan
- Department of Biological Sciences and Centre for Molecular Simulation, University of Calgary, Calgary T2N 1N4, Canada
| | - Tabitha Emde
- Center for Structural Biology of Infectious Diseases (CSBID), Chicago, IL 60611, USA
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bradley Quade
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rosa Di Leo
- Center for Structural Biology of Infectious Diseases (CSBID), Chicago, IL 60611, USA
- Department of Chemical Engineering and Applied Sciences, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Zbyszek Otwinowski
- Center for Structural Biology of Infectious Diseases (CSBID), Chicago, IL 60611, USA
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - D. Peter Tieleman
- Department of Biological Sciences and Centre for Molecular Simulation, University of Calgary, Calgary T2N 1N4, Canada
| | - Dominika Borek
- Center for Structural Biology of Infectious Diseases (CSBID), Chicago, IL 60611, USA
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Alexei Savchenko
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta T2N 4N1, Canada
- Center for Structural Biology of Infectious Diseases (CSBID), Chicago, IL 60611, USA
| |
Collapse
|
10
|
Gentilhomme AS, Dhakar K, Timmins-Schiffman E, Chaw M, Firth E, Junge K, Nunn BL. Proteomic Insights into Psychrophile Growth in Perchlorate-Amended Subzero Conditions: Implications for Martian Life Detection. ASTROBIOLOGY 2025; 25:177-188. [PMID: 39960803 DOI: 10.1089/ast.2024.0065] [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: 03/21/2025]
Abstract
Since the discovery of perchlorates in martian soils, astrobiologists have been curious if and how life could survive in these low-water, high-salt environments. Perchlorates induce chaotropic and oxidative stress but can also confer increased cold tolerance in some extremophiles. Though bacterial survival has been demonstrated at subzero temperatures and in perchlorate solution, proteomic analysis of cells growing in an environment like martian regolith brines-perchlorate with subzero temperatures-has yet to be demonstrated. By defining biosignatures of survival and growth in perchlorate-amended media at subzero conditions, we move closer to understanding the mechanisms that underlie the feasibility of life on Mars. Colwellia psychrerythraea str. 34H (Cp34H), a marine psychrophile, was exposed to perchlorate ions in the form of a diluted Phoenix Mars Lander Wet Chemistry Laboratory solution at -1°C and -5°C. At both temperatures in perchlorate-amended media, Cp34H grew at reduced rates. Mass spectrometry-based proteomics analyses revealed that proteins responsible for mitigating effects of oxidative and chaotropic stress increased, while cellular transport proteins decreased. Cumulative protein signatures suggested modifications to cell-cell or cell-surface adhesion properties. These physical and biochemical traits could serve as putative identifiable biosignatures for life detection in martian environments.
Collapse
Affiliation(s)
- Anais S Gentilhomme
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Kusum Dhakar
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | | | - Matthew Chaw
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Erin Firth
- Polar Science Center, Applied Physics Laboratory, University of Washington, Seattle, Washington, USA
| | - Karen Junge
- Polar Science Center, Applied Physics Laboratory, University of Washington, Seattle, Washington, USA
| | - Brook L Nunn
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| |
Collapse
|
11
|
Van Wyk R, Serem JC, Oosthuizen CB, Semenya D, Serian M, Lorenz CD, Mason AJ, Bester MJ, Gaspar ARM. Carboxy-Amidated AamAP1-Lys has Superior Conformational Flexibility and Accelerated Killing of Gram-Negative Bacteria. Biochemistry 2025; 64:841-859. [PMID: 39873636 PMCID: PMC11840929 DOI: 10.1021/acs.biochem.4c00580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 01/30/2025]
Abstract
C-terminal amidation of antimicrobial peptides (AMPs) is a frequent minor modification used to improve antibacterial potency, commonly ascribed to increased positive charge, protection from proteases, and a stabilized secondary structure. Although the activity of AMPs is primarily associated with the ability to penetrate bacterial membranes, hitherto the effect of amidation on this interaction has not been understood in detail. Here, we show that amidation of the scorpion-derived membranolytic peptide AamAP1-Lys produces a potent analog with faster bactericidal activity, increased membrane permeabilization, and greater Gram-negative membrane penetration associated with greater conformational flexibility. AamAP1-lys-NH2 has improved antibiofilm activity against Acinetobacter baumannii and Escherichia coli, benefits from a two- to 3-fold selectivity improvement, and provides protection against A. baumannii infection in a Galleria mellonella burn wound model. Circular dichroism spectroscopy shows both peptides adopt α-helix conformations in the steady state. However, molecular dynamics (MD) simulations reveal that, during initial binding, AamAP1-Lys-NH2 has greater conformation heterogeneity, with substantial polyproline-II conformation detected alongside α-helix, and penetrates the bilayer more readily than AamAP1-Lys. AamAP1-Lys-NH2 induced membrane permeabilization of A. baumannii occurs only above a critical concentration with slow and weak permeabilization and slow killing occurring at its lower MIC but causes greater and faster permeabilization than AamAP1-Lys, and kills more rapidly, when applied at equal concentrations. Therefore, while the increased potency of AamAP1-Lys-NH2 is associated with slow bactericidal killing, amidation, and the conformational flexibility it induces, affords an improvement in the AMP pharmacodynamic profile and may need to be considered to achieve improved therapeutic performance.
Collapse
Affiliation(s)
- Rosalind
J. Van Wyk
- Department
of Biochemistry, Genetics and Microbiology, Faculty of Natural and
Agricultural Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - June C. Serem
- Department
of Anatomy, Faculty of Health Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Carel B. Oosthuizen
- Drug
Discovery and Development Centre (H3D), University of Cape Town, Cape Town 7701, South Africa
| | - Dorothy Semenya
- Institute
of Pharmaceutical Science, School of Cancer & Pharmaceutical Sciences, King’s College London, London SE1 9NH, United Kingdom
| | - Miruna Serian
- Department
of Physics, Faculty of Natural, Mathematical and Engineering Sciences, King’s College London, London WC2R 2LS, United Kingdom.
| | - Christian D. Lorenz
- Department
of Engineering, Faculty of Natural, Mathematical and Engineering Sciences, King’s College London, London WC2R 2LS, United Kingdom
| | - A. James Mason
- Institute
of Pharmaceutical Science, School of Cancer & Pharmaceutical Sciences, King’s College London, London SE1 9NH, United Kingdom
| | - Megan J. Bester
- Department
of Anatomy, Faculty of Health Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Anabella R. M. Gaspar
- Department
of Biochemistry, Genetics and Microbiology, Faculty of Natural and
Agricultural Sciences, University of Pretoria, Pretoria 0002, South Africa
| |
Collapse
|
12
|
Jose A, Chathangad SN, Sivadas P, Barik D, Kannan K, Das SL, Sadhukhan S, Porel M. Dithiocarbamate-Based Sequence-Defined Oligomers as Promising Membrane-Disrupting Antibacterial Agents: Design, Activity, and Mechanism. ACS APPLIED BIO MATERIALS 2025; 8:1547-1558. [PMID: 39882634 DOI: 10.1021/acsabm.4c01732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
The emerging prevalence of antimicrobial resistance demands cutting-edge therapeutic agents to treat bacterial infections. We present a synthetic strategy to construct sequence-defined oligomers (SDOs) by using dithiocarbamate (DTC). The antibacterial activity of the synthesized library of SDOs was studied using a Gram-positive B. subtilis and a Gram-negative E. coli. Among SDOs, Dec (with C10 aliphatic chain) was found to be the most promising antibacterial agent exhibiting a minimum inhibitory concentration (MIC) of 3 μg/mL against B. subtilis. Structure-activity relationship studies led to a 400-fold improvement in the MIC within the SDO library. The mode of action of the SDOs was elucidated on a model system, where bacterial membranes mimicking giant unilamellar vesicles (GUVs) were exposed to the SDOs. Membrane disruption and pore formation were found to be the key mechanisms through which SDOs act. In addition, scanning electron microscopy (SEM) and confocal laser scanning microscopy analysis of Dec-treated bacteria confirmed the loss of cell membrane integrity. Finally, the hemolysis assay with SDOs revealed their excellent selectivity toward bacterial cells. Taken together, we developed a modular platform for the synthesis of SDOs having promising antibacterial activity and superior selectivity toward bacteria, with the membrane disruption mode of action confirmed via studies on the model GUV system and SEM analysis.
Collapse
Affiliation(s)
- Anna Jose
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad, Kerala 678623, India
| | - Shabin N Chathangad
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad, Kerala 678623, India
- Physical and Chemical Biology Laboratory, Indian Institute of Technology Palakkad, Palakkad, Kerala 678623, India
| | - Palliyil Sivadas
- Physical and Chemical Biology Laboratory, Indian Institute of Technology Palakkad, Palakkad, Kerala 678623, India
- Department of Physics, Indian Institute of Technology Palakkad, Palakkad, Kerala 678623, India
| | - Debashis Barik
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad, Kerala 678623, India
| | - Karthika Kannan
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad, Kerala 678623, India
- Physical and Chemical Biology Laboratory, Indian Institute of Technology Palakkad, Palakkad, Kerala 678623, India
| | - Sovan Lal Das
- Physical and Chemical Biology Laboratory, Indian Institute of Technology Palakkad, Palakkad, Kerala 678623, India
- Department of Mechanical Engineering, Indian Institute of Technology Palakkad, Palakkad, Kerala 678623, India
| | - Sushabhan Sadhukhan
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad, Kerala 678623, India
- Physical and Chemical Biology Laboratory, Indian Institute of Technology Palakkad, Palakkad, Kerala 678623, India
- Department of Biological Sciences and Engineering, Indian Institute of Technology Palakkad, Palakkad, Kerala 678623, India
| | - Mintu Porel
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad, Kerala 678623, India
- Environmental Sciences and Sustainable Engineering Centre, Indian Institute of Technology Palakkad, Palakkad, Kerala 678623, India
| |
Collapse
|
13
|
Pavia N, Potenza A, Hornos F, Poveda JA, Gonorazky G, Neira JL, Giudici AM, Beligni MV. The Diacylglycerol Acyltransferase 3 of Chlamydomonas reinhardtii Is a Disordered Protein Capable of Binding to Lipids Derived from Chloroplasts. Biomolecules 2025; 15:245. [PMID: 40001548 PMCID: PMC11852920 DOI: 10.3390/biom15020245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Understanding triacylglycerol (TAG) metabolism is crucial for developing algae as a source of biodiesel. TAGs are the main reservoir of energy in most eukaryotes. The final, rate-limiting step in the formation of TAGs is catalyzed by 1,2-diacylglycerol acyltransferases (DGATs). In the green alga Chlamydomonas reinhardtii, DGAT3 is phylogenetically related to plant DGAT3 but unrelated to other DGATs from eukaryotes, such as DGAT1 and DGAT2. In this study, we described the conformational preferences and the lipid-binding features of the DGAT3 from C. reinhardtii. To characterize its conformational stability and structural features, we used several biophysical probes, namely, fluorescence, circular dichroism (CD), and differential scanning calorimetry (DSC). Our results showed that the protein was mainly disordered, containing a small population of folded conformations in a narrow pH range (pH 8 to 10). The conformational stability of the folded structure of DGAT3 was very low, as shown by urea or guanidinium denaturations. Thermal denaturation, followed by fluorescence or CD, as well as calorimetric denaturation, followed by DSC, did not yield any transition in the pH range where DGAT3 acquired a "native-like" conformation. Furthermore, we used two approaches to demonstrate the interaction of DGAT3 with lipid membranes at the pH at which it had acquired a "native-like" conformation. The first involved the measurement of anisotropy and fluorescence quenching of the protein. The second approach focused on examining possible modifications of the biophysical properties of lipids due to their interaction with DGAT3, through anisotropy measurements and leakage assays. Both methods produced consistent results, suggesting that DGAT3 preferentially interacted with negatively charged membranes. These results will allow the design of a more efficient and stable DGAT3, as well as an in-depth understanding of how the metabolism of TAGs is accomplished in C. reinhardtii.
Collapse
Affiliation(s)
- Natalia Pavia
- Instituto de Investigaciones Biológicas (IIB-CONICET-UNMdP), Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata, Mar del Plata 7600, Argentina; (N.P.); (G.G.)
| | - Alberto Potenza
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, 03202 Elche, Spain; (A.P.); (F.H.); (J.A.P.); (J.L.N.)
| | - Felipe Hornos
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, 03202 Elche, Spain; (A.P.); (F.H.); (J.A.P.); (J.L.N.)
| | - José A. Poveda
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, 03202 Elche, Spain; (A.P.); (F.H.); (J.A.P.); (J.L.N.)
| | - Gabriela Gonorazky
- Instituto de Investigaciones Biológicas (IIB-CONICET-UNMdP), Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata, Mar del Plata 7600, Argentina; (N.P.); (G.G.)
| | - José L. Neira
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, 03202 Elche, Spain; (A.P.); (F.H.); (J.A.P.); (J.L.N.)
- Institute of Biocomputation and Physics of Complex Systems (BIFI), Universidad de Zaragoza, 50018 Zaragoza, Spain
| | - Ana M. Giudici
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, 03202 Elche, Spain; (A.P.); (F.H.); (J.A.P.); (J.L.N.)
| | - María Verónica Beligni
- Instituto de Investigaciones Biológicas (IIB-CONICET-UNMdP), Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata, Mar del Plata 7600, Argentina; (N.P.); (G.G.)
| |
Collapse
|
14
|
Basak P, Ekka M, Pandiyan A, Tandon S, Gowrishankar J. The membrane-targeting-sequence motif is required for exhibition of recessive resurrection in Escherichia coli RNase E. Nucleic Acids Res 2025; 53:gkaf055. [PMID: 39898549 PMCID: PMC11788932 DOI: 10.1093/nar/gkaf055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 01/13/2025] [Accepted: 01/21/2025] [Indexed: 02/04/2025] Open
Abstract
The essential homotetrameric endoribonuclease RNase E of Escherichia coli participates in global RNA turnover as well as stable RNA maturation. The protomer's N-terminal half (residues 1-529) bears the catalytic, allosteric, and tetramerization domains, including the active site residues D303 and D346. The C-terminal half (CTH, residues 530-1061) is dispensable for viability. We have previously described a phenomenon of recessive resurrection in RNase E that requires the CTH, wherein the wild-type homotetramer apparently displays nearly identical activity in vivo as a heterotetramer comprising three catalytically dead subunits (with D303A or D346A substitutions) and one wild-type subunit. Here, we show that recessive resurrection is exhibited even in dimeric RNase E with the CTH, and that it is largely dependent on the presence of a membrane-targeting-sequence motif (residues 565-582). A single F575E substitution also impaired recessive resurrection, whereas other CTH motifs (such as those for binding of RNA or of partner proteins) were dispensable. The phenomenon was independent of RNA 5'-monophosphate sensing by the enzyme. We propose that membrane-anchoring of RNase E renders it processive for endoribonucleolytic action, and that recessive resurrection and dominant negativity associated with mutant protomers are mutually exclusive manifestations of, respectively, processive and distributive catalytic mechanisms in a homo-oligomeric enzyme.
Collapse
Affiliation(s)
- Papri Basak
- Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar 140306 Punjab, India
| | - Manjula Ekka
- Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar 140306 Punjab, India
| | - Apuratha Pandiyan
- Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar 140306 Punjab, India
| | - Smriti Tandon
- Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar 140306 Punjab, India
| | - Jayaraman Gowrishankar
- Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar 140306 Punjab, India
- Department of Systems and Computational Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, Telangana, India
| |
Collapse
|
15
|
Sawasato K, Dowhan W, Bogdanov M. Its own architect: Flipping cardiolipin synthase. SCIENCE ADVANCES 2025; 11:eads0244. [PMID: 39752486 PMCID: PMC11698083 DOI: 10.1126/sciadv.ads0244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 12/02/2024] [Indexed: 01/06/2025]
Abstract
Current dogma assumes that lipid asymmetry in biological membranes is actively maintained and dispensable for cell viability. The inner (cytoplasmic) membrane (IM) of Escherichia coli is asymmetric. However, the molecular mechanism that maintains this uneven distribution is unknown. We engineered a conditionally lethal phosphatidylethanolamine (PE)-deficient mutant in which the presence of cardiolipin (CL) on the periplasmic leaflet of the IM is essential for viability, revealing a mechanism that provides CL on the desired leaflet of the IM. CL synthase (ClsA) flips its catalytic cytoplasmic domain upon depletion of PE to supply nonbilayer-prone CL in the periplasmic leaflet of the IM for cell viability. In the presence of a physiological amount of PE, osmotic down-shock induces a topological inversion of ClsA, establishing the biological relevance of membrane protein reorientations in wild-type cells. These findings support a flippase-less mechanism for maintaining membrane lipid asymmetry in biogenic membranes by self-organization of a lipid-synthesizing enzyme.
Collapse
Affiliation(s)
- Katsuhiro Sawasato
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - William Dowhan
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Mikhail Bogdanov
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
16
|
Guan X, Li Y, Yang Y, Liu Z, Shi R, Xu Y. Root exudates regulate soil antibiotic resistance genes via rhizosphere microbes under long-term fertilization. ENVIRONMENT INTERNATIONAL 2025; 195:109180. [PMID: 39700687 DOI: 10.1016/j.envint.2024.109180] [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: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Abstract
Organic fertilizer application promotes the prevalence of antibiotic resistance genes (ARGs), yet the factors driving temporal differences in ARG abundance under long-term organic fertilizer application remain unclear. This study investigated the temporal dynamics of ARG diversity and abundance in both bulk and rhizosphere soils over 17 years (2003-2019), and explored microbial evolution strategies, ARG hosts succession and the influence of root exudates on ARGs regulation. The results showed that the ARGs abundance in rhizosphere soil was lower than that in bulk soil under long-term fertilization, and ARGs abundance exhibited a decrease and then remained stable in rhizosphere soil over time. There was a strong association between host bacteria and dominant ARGs (p < 0.05). Structural equations demonstrated that bacterial community had a most pronounced influence on ARGs (p < 0.05), and metabolites exhibited an important mediation effect on bacterial community (p < 0.05), thereby impacting ARGs. The metabolome analysis evidenced that significant correlations were found between defensive root exudates and most ARGs abundance (p < 0.05), like, luteolin-7-glucoside was negatively correlated with tetA(58). These findings provide deeper insights into the dynamics of soil ARGs under long-term fertilization, and identify critical factors that influence ARGs colonization in soils, providing support for controlling the spread of ARGs in agriculture soils.
Collapse
Affiliation(s)
- Xiujing Guan
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin 300191, China
| | - Yuhui Li
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin 300191, China
| | - Yanying Yang
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin 300191, China
| | - Zihua Liu
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin 300191, China
| | - Rongguang Shi
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin 300191, China.
| | - Yan Xu
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin 300191, China.
| |
Collapse
|
17
|
Qiao W, Shen C, Chen Y, Chang S, Wang X, Yang L, Pang J, Luo Q, Zhang Z, Xiang Y, Zhao C, Lu G, Ding BS, Ying B, Tang X, Dong H. Deciphering the molecular basis of lipoprotein recognition and transport by LolCDE. Signal Transduct Target Ther 2024; 9:354. [PMID: 39725716 DOI: 10.1038/s41392-024-02067-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 12/28/2024] Open
Abstract
Outer membrane (OM) lipoproteins serve vital roles in Gram-negative bacteria, contributing to their pathogenicity and drug resistance. For these lipoproteins to function, they must be transported from the inner membrane (IM), where they are assembled, to the OM by the ABC transporter LolCDE. We have previously captured structural snapshots of LolCDE in multiple states, revealing its dynamic conformational changes. However, the exact mechanism by which LolCDE recognizes and transfers lipoprotein between domains remains unclear. Here, we characterized the E. coli LolCDE complex bound with endogenous lipoprotein or ATP to explore the molecular features governing its substrate binding and transport functions. We found that the N-terminal unstructured linker of lipoprotein is critical for efficient binding by LolCDE; it must be sufficiently long to keep the lipoprotein's main body outside the complex while allowing the triacyl chains to bind within the central cavity. Mutagenic assays identified key residues that mediate allosteric communication between the cytoplasmic and transmembrane domains and in the periplasmic domain to form a lipoprotein transport pathway at the LolC-LolE interface. This study provides insights into the OM lipoprotein relocation process mediated by LolCDE, with significant implications for antimicrobial drug development.
Collapse
Affiliation(s)
- Wen Qiao
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Chongrong Shen
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Yujiao Chen
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Shenghai Chang
- Center of Cryo Electron Microscopy, Zhejiang University, Hangzhou, China
| | - Xin Wang
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Lili Yang
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Jie Pang
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Qinghua Luo
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Zhibo Zhang
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Yingxin Xiang
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Chao Zhao
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Guangwen Lu
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Bi-Sen Ding
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Binwu Ying
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China.
| | - Xiaodi Tang
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China.
| | - Haohao Dong
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
18
|
Reynoso EC, Delgado-Suárez EJ, Hernández-Pérez CF, Chavarin-Pineda Y, Godoy-Lozano EE, Fierros-Zárate G, Aguilar-Vera OA, Castillo-Ramírez S, Gómez-Pedroso LDCS, Sánchez-Zamorano LM. Geography, Antimicrobial Resistance, and Genomics of Salmonella enterica (Serotypes Newport and Anatum) from Meat in Mexico (2021-2023). Microorganisms 2024; 12:2485. [PMID: 39770688 PMCID: PMC11727726 DOI: 10.3390/microorganisms12122485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Salmonella enterica non-typhoidal is a major contributor to diarrheal diseases, with over 2600 serovars identified across diverse environments. In Mexico, serovars Newport and Anatum have shown a marked increase, especially in foodborne disease, posing a public health problem. We conducted a cross-sectional study from 2021 to 2023 using active epidemiological surveillance to assess contamination in ground beef and pork at butcher shops nationwide. It involved isolation, phenotypic antimicrobial resistance, comparative genomics, spatial distribution, antimicrobial-resistance genes, and pangenome analysis. A total of 402 non-typhoidal S. enterica strains were isolated, including 59 Newport and 50 Anatum. After curating for redundancy, 45 Newport and 32 Anatum strains remained. We found that 75% of Newport strains exhibited multidrug resistance (MDR), compared to 25% of Anatum strains. Salmonella Newport also showed a broader distribution and stronger antibiotic-resistance capacity, particularly due to genes such as mphA and ramA. Our pangenome analysis showed a predominance of cell maintenance and survival-process genes in the accessory genome of both serotypes. Considering unique genes, Salmonella Anatum and Newport showed a notorious abundance of genes with functions related to replication, recombination, and repair. The substantial rise of Anatum and Newport strains in meat samples for human consumption presents an epidemiological alert, highlighting the critical need for stringent surveillance programs to mitigate human and ecosystem health risks.
Collapse
Affiliation(s)
- Eduardo Canek Reynoso
- Centro de Investigación en Salud Poblacional, Instituto Nacional de Salud Pública (INSP), Morelos 62100, Mexico; (E.C.R.); (G.F.-Z.)
| | - Enrique Jesús Delgado-Suárez
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico; (E.J.D.-S.); (L.d.C.S.G.-P.)
| | - Cindy Fabiola Hernández-Pérez
- Centro Nacional de Referencia de Inocuidad y Bioseguridad Agroalimentaria, Servicio Nacional de Sanidad, Inocuidad y Calidad Agroalimentaria (SENASICA), Tecámac 55740, Mexico;
| | - Yaselda Chavarin-Pineda
- Centro de Investigación en Ciencias Agrícolas, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla 72570, Mexico
| | | | - Geny Fierros-Zárate
- Centro de Investigación en Salud Poblacional, Instituto Nacional de Salud Pública (INSP), Morelos 62100, Mexico; (E.C.R.); (G.F.-Z.)
| | - Omar Alejandro Aguilar-Vera
- Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México (UNAM), Morelos 62210, Mexico; (O.A.A.-V.); (S.C.-R.)
| | - Santiago Castillo-Ramírez
- Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México (UNAM), Morelos 62210, Mexico; (O.A.A.-V.); (S.C.-R.)
| | - Luz del Carmen Sierra Gómez-Pedroso
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico; (E.J.D.-S.); (L.d.C.S.G.-P.)
| | - Luisa María Sánchez-Zamorano
- Centro de Investigación en Salud Poblacional, Instituto Nacional de Salud Pública (INSP), Morelos 62100, Mexico; (E.C.R.); (G.F.-Z.)
| |
Collapse
|
19
|
Zhao Y, Wu R, Wu X, Zhou N, Ren J, Liu W, Yu R, Zhang S, Yang J, Li H, Liu H. Modulation of physiological functions and metabolome of Vibrio alginolyticus by quorum-regulatory sRNA, Qrr1. Lett Appl Microbiol 2024; 77:ovae126. [PMID: 39657312 DOI: 10.1093/lambio/ovae126] [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: 07/26/2024] [Revised: 11/05/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024]
Abstract
Vibrio alginolyticus, the causative agent of aquatic vertebrates and invertebrates, can cause severe infections (e.g. septicemia, gill necrosis, and surface ulcers) and high mortality in aquatic organisms, leading to serious economic losses in global aquaculture. Small non-coding RNAs (sRNAs), emerging modulators of gene expression, played vital regulatory roles in virulence, pathogenicity, and physiological metabolism of bacteria. In this work, the modulation of physiological functions and metabolome of V. alginolyticus by the quorum-regulatory sRNA, Qrr1, was figured out. We found that the deletion of qrr1 induced significant cell shape elongation. Meanwhile, Qrr1 could inhibit the production of alkaline serine protease by weakening the expression of main regulator LuxR in the quorum sensing (QS) system. Moreover, the untargeted metabolomics and lipidomics approaches showed that most of nucleotides, organic acids, carbohydrates, and lipidome (both lipid content and category) were significantly altered in response to the qrr1 deletion. Spearman correlation analysis demonstrated that most of the intermediates involved in glutamate metabolism, sphingolipid metabolism, and glycerolipid metabolism displayed high correlations with cell virulence factors. These findings illuminate the mechanism of bacterial virulence regulation and further exploit potential therapeutic targets for virulence prevention in V. alginolyticus.
Collapse
Affiliation(s)
- Yanni Zhao
- School of Food Science and Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
- Shaanxi Research Institute of Agricultural Products Processing Technology, Xi'an 710021, China
| | - Ruobing Wu
- School of Food Science and Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Xuan Wu
- School of Food Science and Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Ningning Zhou
- School of Food Science and Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Jiamin Ren
- School of Food Science and Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Wang Liu
- School of Food Science and Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Rui Yu
- School of Food Science and Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Senhu Zhang
- School of Food Science and Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Jinfang Yang
- School of Food Science and Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Hua Li
- SUSTech Core Research Facilities, Southern University of Science and Technology, Shenzhen 518055, China
| | - Huan Liu
- School of Food Science and Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
- Shaanxi Research Institute of Agricultural Products Processing Technology, Xi'an 710021, China
| |
Collapse
|
20
|
Hwang JH, Choi TR, Kim S, Lee Y, Shin Y, Choi S, Oh J, Kim SH, Park JH, Bhatia SK, Yang YH. Evaluation of simplified ester-linked fatty acid analysis (ELFA) for phospholipid fatty acid (PLFA) analysis of bacterial population. Anal Biochem 2024; 695:115638. [PMID: 39127328 DOI: 10.1016/j.ab.2024.115638] [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: 06/28/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Phospholipid fatty acid (PLFA) analysis is used for characterizing microbial communities based on their lipid profiles. This method avoids biases from PCR or culture, allowing data collection in a natural state. However, PLFA is labor-intensive due to lipid fractionation. Simplified ester-linked fatty acid analysis (ELFA), which skips lipid fractionation, offers an alternative. It utilizes base-catalyzed methylation to derivatize only lipids, not free fatty acids, and found glycolipid and neutral lipid fractions are scarcely present in most bacteria, allowing lipid fractionation to be skipped. ELFA method showed a high correlation to PLFA data (r = 0.99) and higher sensitivity than the PLFA method by 1.5-2.57-fold, mainly due to the higher recovery of lipids, which was 1.5-1.9 times higher than with PLFA. The theoretical limit of detection (LOD) and limit of quantification (LOQ) for the ELFA method indicated that 1.54-fold less sample was needed for analysis than with the PLFA method. Our analysis of three bacterial cultures and a simulated consortium revealed the effectiveness of the ELFA method by its simple procedure and enhanced sensitivity for detecting strain-specific markers, which were not detected in PLFA analysis. Overall, this method could be easily used for the population analysis of synthetic consortia.
Collapse
Affiliation(s)
- Jeong Hyeon Hwang
- Department of Biological Engineering, College of Engineering, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Tae-Rim Choi
- Department of Biological Engineering, College of Engineering, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Suwon Kim
- Department of Biological Engineering, College of Engineering, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Yeda Lee
- Department of Biological Engineering, College of Engineering, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Yuni Shin
- Department of Biological Engineering, College of Engineering, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Suhye Choi
- Department of Biological Engineering, College of Engineering, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Jinok Oh
- Department of Biological Engineering, College of Engineering, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Sang-Hyoun Kim
- Department of Civil and Environmental Engineering, Yonsei University, 50, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jeong-Hoon Park
- Sustainable Technology and Wellness R&D Group, Korea Institute of Industrial Technology (KITECH), Jeju-si, 63243, Republic of Korea
| | - Shashi Kant Bhatia
- Department of Biological Engineering, College of Engineering, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea; Institute for Ubiquitous Information Technology and Application, Konkuk University, Seoul, 05029, Republic of Korea
| | - Yung-Hun Yang
- Department of Biological Engineering, College of Engineering, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea; Institute for Ubiquitous Information Technology and Application, Konkuk University, Seoul, 05029, Republic of Korea.
| |
Collapse
|
21
|
Hu W, Huo X, Ma T, Li Z, Yang T, Yang H, Feng S. Insights into the role of cyclopropane fatty acid synthase (CfaS) from extreme acidophile in bacterial defense against environmental acid stress. Extremophiles 2024; 29:1. [PMID: 39549088 DOI: 10.1007/s00792-024-01368-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: 05/15/2024] [Accepted: 10/26/2024] [Indexed: 11/18/2024]
Abstract
The cell membrane remodeling mediated by cyclopropane fatty acid synthase (CfaS) plays a crucial role in microbial physiological processes resisting various environmental stressors, including acid. Herein, we found a relatively high proportion (24.8%-28.3%) of cyclopropane fatty acid (CFA) Cy-19:0 in the cell membrane of a newly isolated extreme acidophile, Acidithiobacillus caldus CCTCC AB 2019256, under extreme acid stress. Overexpression of the CfaS encoding gene cfaS2 in Escherichia coli conferred enhanced acid resistance. GC-MS analysis revealed a 3.52-fold increase in the relative proportion of Cy-19:0 in the cell membrane of the overexpression strain compared to the control. Correspondingly, membrane fluidity, permeability and cell surface hydrophobicity were reduced to varying degrees. Additionally, HPLC analysis indicated that the overexpression strain had 1.54-, 1.42-, 1.85-, 1.20- and 1.05-fold higher levels of intracellular glutamic acid, arginine, aspartic acid, methionine and alanine, respectively, compared to the control. Overall, our findings shed light on the role of CfaS derived from extreme acidophile in bacterial defense against environmental acid stress, potentially facilitating its application in the design and development of industrial microbial chassis cells for organic acid production.
Collapse
Affiliation(s)
- Wenbo Hu
- School of Life Sciences, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - Xingyu Huo
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, People's Republic of China
| | - Tengfei Ma
- School of Life Sciences, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - Zhigang Li
- School of Life Sciences, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - Tianyou Yang
- School of Life Sciences, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - Hailin Yang
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, People's Republic of China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, 1800 Lihu Road, Wuxi, People's Republic of China
| | - Shoushuai Feng
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, People's Republic of China.
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, 1800 Lihu Road, Wuxi, People's Republic of China.
| |
Collapse
|
22
|
Hussein M, Kang Z, Neville SL, Allobawi R, Thrombare V, Koh AJJ, Wilksch J, Crawford S, Mohammed MK, McDevitt CA, Baker M, Rao GG, Li J, Velkov T. Metabolic profiling unveils enhanced antibacterial synergy of polymyxin B and teixobactin against multi-drug resistant Acinetobacter baumannii. Sci Rep 2024; 14:27145. [PMID: 39511424 PMCID: PMC11543821 DOI: 10.1038/s41598-024-78769-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024] Open
Abstract
This untargeted metabolomics study investigated the synergistic antibacterial activity of polymyxin B and Leu10-teixobactin, a depsipeptide inhibitor of cell wall biosynthesis. Checkerboard microdilution assays revealed a significant synergy against polymyxin-susceptible and -resistant A. baumannii, excluding lipopolysaccharide-deficient variants. Time-kill assays confirmed bactericidal synergy, reducing bacterial burden by approximately 4-6-log10CFU/mL. The combination (2xMIC polymyxin B and 0.5xMIC Leu10-teixobactin) prevented bacterial regrowth after 24 h, indicating sustained efficacy against the emergence of resistant mutants. The analysis of A. baumannii ATCC™ 19606 metabolome demonstrated that the polymyxin B-Leu10-teixobactin combination produced more pronounced perturbation compared to the individual antibiotics across all time points (1, 3 and 6 h). Pathway analysis revealed that lipid metabolism, cell envelope biogenesis, and cellular respiration were predominantly impacted by the combination, and to a lesser extent by polymyxin B monotherapy. Leu10-teixobactin treatment alone had only a minor impact on the metabolome, primarily at the 6 h time point. Peptidoglycan assays confirmed the combination's concerted deleterious effects on bacterial cell envelope integrity. Electron microscopy further substantiated these findings, revealing pronounced cell envelope damage, membrane blebbing, and vacuole formation. These findings highlight the potential of the polymyxin B-Leu10-teixobactin combination as an effective treatment in preventing resistance in A. baumannii.
Collapse
Affiliation(s)
- Maytham Hussein
- Monash Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia.
| | - Zhisen Kang
- Monash Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Stephanie L Neville
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Rafah Allobawi
- Monash Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Varsha Thrombare
- Monash Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Augustine Jing Jie Koh
- Monash Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jonathan Wilksch
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Simon Crawford
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, VIC, 3800, Australia
| | | | - Christopher A McDevitt
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Mark Baker
- Discipline of Biological Sciences, Priority Research Centre in Reproductive Biology, Faculty of Science and IT, University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - Gauri G Rao
- Titus Family, Department of Clinical Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA, 90089-9121, USA.
| | - Jian Li
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, VIC, 3800, Australia.
| | - Tony Velkov
- Monash Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia.
| |
Collapse
|
23
|
van Hoogstraten SWG, Kuik C, Arts JJC, Cillero-Pastor B. Molecular imaging of bacterial biofilms-a systematic review. Crit Rev Microbiol 2024; 50:971-992. [PMID: 37452571 PMCID: PMC11523921 DOI: 10.1080/1040841x.2023.2223704] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/16/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023]
Abstract
The formation of bacterial biofilms in the human body and on medical devices is a serious human health concern. Infections related to bacterial biofilms are often chronic and difficult to treat. Detailed information on biofilm formation and composition over time is essential for a fundamental understanding of the underlying mechanisms of biofilm formation and its response to anti-biofilm therapy. However, information on the chemical composition, structural components of biofilms, and molecular interactions regarding metabolism- and communication pathways within the biofilm, such as uptake of administered drugs or inter-bacteria communication, remains elusive. Imaging these molecules and their distribution in the biofilm increases insight into biofilm development, growth, and response to environmental factors or drugs. This systematic review provides an overview of molecular imaging techniques used for bacterial biofilm imaging. The techniques included mass spectrometry-based techniques, fluorescence-labelling techniques, spectroscopic techniques, nuclear magnetic resonance spectroscopy (NMR), micro-computed tomography (µCT), and several multimodal approaches. Many molecules were imaged, such as proteins, lipids, metabolites, and quorum-sensing (QS) molecules, which are crucial in intercellular communication pathways. Advantages and disadvantages of each technique, including multimodal approaches, to study molecular processes in bacterial biofilms are discussed, and recommendations on which technique best suits specific research aims are provided.
Collapse
Affiliation(s)
- S. W. G. van Hoogstraten
- Laboratory for Experimental Orthopaedics, Department of Orthopaedic Surgery, CAPHRI, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - C. Kuik
- Maastricht MultiModal Molecular Imaging Institute (M4I), Maastricht University, Maastricht, the Netherlands
| | - J. J. C. Arts
- Laboratory for Experimental Orthopaedics, Department of Orthopaedic Surgery, CAPHRI, Maastricht University Medical Centre, Maastricht, the Netherlands
- Department of Biomedical Engineering, Orthopaedic Biomechanics, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - B. Cillero-Pastor
- Maastricht MultiModal Molecular Imaging Institute (M4I), Maastricht University, Maastricht, the Netherlands
- Department of Cell Biology-Inspired Tissue Engineering, The MERLN Institute for Technology-Inspired Regenerative Medicine, University of Maastricht, Maastricht, the Netherlands
| |
Collapse
|
24
|
Jimenez M, L'Heureux J, Kolaya E, Liu GW, Martin KB, Ellis H, Dao A, Yang M, Villaverde Z, Khazi-Syed A, Cao Q, Fabian N, Jenkins J, Fitzgerald N, Karavasili C, Muller B, Byrne JD, Traverso G. Synthetic extremophiles via species-specific formulations improve microbial therapeutics. NATURE MATERIALS 2024; 23:1436-1443. [PMID: 38969782 PMCID: PMC11840811 DOI: 10.1038/s41563-024-01937-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/31/2024] [Indexed: 07/07/2024]
Abstract
Microorganisms typically used to produce food and pharmaceuticals are now being explored as medicines and agricultural supplements. However, maintaining high viability from manufacturing until use remains an important challenge, requiring sophisticated cold chains and packaging. Here we report synthetic extremophiles of industrially relevant gram-negative bacteria (Escherichia coli Nissle 1917, Ensifer meliloti), gram-positive bacteria (Lactobacillus plantarum) and yeast (Saccharomyces boulardii). We develop a high-throughput pipeline to define species-specific materials that enable survival through drying, elevated temperatures, organic solvents and ionizing radiation. Using this pipeline, we enhance the stability of E. coli Nissle 1917 by more than four orders of magnitude over commercial formulations and demonstrate its capacity to remain viable while undergoing tableting and pharmaceutical processing. We further show, in live animals and plants, that synthetic extremophiles remain functional against enteric pathogens and as nitrogen-fixing plant supplements even after exposure to elevated temperatures. This synthetic, material-based stabilization enhances our capacity to apply microorganisms in extreme environments on Earth and potentially during exploratory space travel.
Collapse
Affiliation(s)
- Miguel Jimenez
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Johanna L'Heureux
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emily Kolaya
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gary W Liu
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kyle B Martin
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Husna Ellis
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alfred Dao
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Margaret Yang
- Department of Chemistry and Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Zachary Villaverde
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Afeefah Khazi-Syed
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Qinhao Cao
- Department of Chemistry and Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Niora Fabian
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Joshua Jenkins
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nina Fitzgerald
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Christina Karavasili
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Benjamin Muller
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - James D Byrne
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Giovanni Traverso
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
25
|
Piccinini A, Kohlbrecher J, Moussaoui D, Winter A, Prévost S. Effect of cardiolipin on the lamellarity and elongation of liposomes hydrated in PBS. J Colloid Interface Sci 2024; 669:844-855. [PMID: 38749223 DOI: 10.1016/j.jcis.2024.04.211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/25/2024] [Accepted: 04/29/2024] [Indexed: 05/27/2024]
Abstract
Lamellarity and shape are important factors in the formation of vesicles and determine their role in biological systems and pharmaceutical applications. Cardiolipin (CL) is a major lipid in many biological membranes and exerts a great influence on their structural organization due to its particular structure and physico-chemical properties. Here, we used small-angle X-ray and neutron scattering to study the effects of CL with different acyl chain lengths and saturations (CL14:0, CL18:1, CL18:2) on vesicle morphology and lamellarity in membrane models containing mixtures of phosphatidylcholine and phosphatidylethanolamine with different acyl chain lengths and saturations (C14:0 and C 18:1). Measurements were performed in the presence of Phosphate Buffer Saline (PBS), at 37°C, to better reflect physiological conditions, which resulted in strong effects on vesicle morphology, depending on the type and amount of CL used. The presence of small quantities of CL (from 2.5%) reduced inter-membrane correlations and increased perturbation of the membrane, an effect which is enhanced in the presence of matched shorter saturated acyl chains, and mainly unilamellar vesicles (ULV) are formed. In extruded vesicles, employed for SANS experiments, flattened vesicles are observed partly due to the hypertonic effect of PBS, but also influenced by the type of CL added. Our experimental data from SAXS and SANS revealed a strong dependence on CL content in shaping the membrane microstructure, with an apparent optimum in the PC:CL mixture in terms of promoting reduced correlations, preferred curvature and elongation. However, the use of PBS caused distinct differences from previously published studies in water in terms of vesicle shape, and highlights the need to investigate vesicle formation under physiological conditions in order to be able to draw conclusions about membrane formation in biological systems.
Collapse
Affiliation(s)
- Alice Piccinini
- Institut Laue-Langevin - The European Neutron Source, 38042 Grenoble, France; School of Life Sciences, Keele University, ST55BG Staffordshire, United Kingdom; Dept. of Medical Biotechnology and Translational Medicine Università degli Studi di Milano, 20054 Milan, Italy
| | | | | | - Anja Winter
- School of Life Sciences, Keele University, ST55BG Staffordshire, United Kingdom.
| | - Sylvain Prévost
- Institut Laue-Langevin - The European Neutron Source, 38042 Grenoble, France
| |
Collapse
|
26
|
Bianco CM, Caballero-Rothar NN, Ma X, Farley KR, Vanderpool CK. Transcriptional and post-transcriptional mechanisms modulate cyclopropane fatty acid synthase through small RNAs in Escherichia coli. J Bacteriol 2024; 206:e0004924. [PMID: 38980083 PMCID: PMC11340327 DOI: 10.1128/jb.00049-24] [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: 02/10/2024] [Accepted: 06/12/2024] [Indexed: 07/10/2024] Open
Abstract
The small RNA (sRNA) RydC strongly activates cfa, which encodes the cyclopropane fatty acid synthase. Previous work demonstrated that RydC activation of cfa increases the conversion of unsaturated fatty acids to cyclopropanated fatty acids in membrane lipids and changes the biophysical properties of membranes, making cells more resistant to acid stress. The regulators that control RydC synthesis had not previously been identified. In this study, we identify a GntR-family transcription factor, YieP, that represses rydC transcription. YieP positively autoregulates its own transcription and indirectly regulates cfa through RydC. We further identify additional sRNA regulatory inputs that contribute to the control of RydC and cfa. The translation of yieP is repressed by the Fnr-dependent sRNA, FnrS, making FnrS an indirect activator of rydC and cfa. Conversely, RydC activity on cfa is antagonized by the OmpR-dependent sRNA OmrB. Altogether, this work illuminates a complex regulatory network involving transcriptional and post-transcriptional inputs that link the control of membrane biophysical properties to multiple environmental signals. IMPORTANCE Bacteria experience many environmental stresses that challenge their membrane integrity. To withstand these challenges, bacteria sense what stress is occurring and mount a response that protects membranes. Previous work documented the important roles of small RNA (sRNA) regulators in membrane stress responses. One sRNA, RydC, helps cells cope with membrane-disrupting stresses by promoting changes in the types of lipids incorporated into membranes. In this study, we identified a regulator, YieP, that controls when RydC is produced and additional sRNA regulators that modulate YieP levels and RydC activity. These findings illuminate a complex regulatory network that helps bacteria sense and respond to membrane stress.
Collapse
Affiliation(s)
- Colleen M. Bianco
- Department of Microbiology, University of Illinois, Urbana, Illinois, USA
| | | | - Xiangqian Ma
- Department of Microbiology, University of Illinois, Urbana, Illinois, USA
| | - Kristen R. Farley
- Department of Microbiology, University of Illinois, Urbana, Illinois, USA
| | | |
Collapse
|
27
|
Sanders G, Borbat PP, Georgieva ER. Conformations of influenza A M2 protein in DOPC/DOPS and E. coli native lipids and proteins. Biophys J 2024; 123:2584-2593. [PMID: 38932458 PMCID: PMC11365223 DOI: 10.1016/j.bpj.2024.06.025] [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: 01/08/2024] [Revised: 04/21/2024] [Accepted: 06/24/2024] [Indexed: 06/28/2024] Open
Abstract
We compared the conformations of the transmembrane domain (TMD) of influenza A M2 (IM2) protein reconstituted in 1,2-dioleoyl-sn-glycero-3-phosphocholine/1,2-dioleoyl-sn-glycero-3-phospho-L-serine (DOPC/DOPS) bilayers to those in isolated Escherichia coli (E. coli) membranes, having preserved its native proteins and lipids. IM2 is a single-pass transmembrane protein known to assemble into a homo-tetrameric proton channel. To represent this channel, we made a construct containing the IM2's TMD region flanked by the juxtamembrane residues. The single cysteine substitution, L43C, of leucine located in the bilayer polar region was paramagnetically tagged with a methanethiosulfonate nitroxide label for the electron spin resonance (ESR) study. For this particular residue, we probed the conformations of the spin-labeled IM2 reconstituted in DOPC/DOPS and isolated E. coli membranes using continuous-wave ESR and double electron-electron resonance (DEER) spectroscopy. The total protein-to-lipid molar ratio spanned the range from 1:230 to 1:10,400. The continuous-wave ESR spectra corresponded to very slow spin-label motion in both environments. In all cases, the DEER data were reconstructed into distance distributions with well-resolved peaks at 1.68 and 2.37 nm in distance and amplitude ratios of 1.41 ± 0.2 and 2:1, respectively. This suggests four nitroxide spin labels located at the corners of a square, indicative of an axially symmetric tetramer. The distance modeling of DEER data with molecular modeling software applied to the NMR molecular structures (PDB: 2L0J) confirmed the symmetry and closed state of the C-terminal exit pore of the IM2 TMD tetramer in agreement with the model. Thus, we can conclude that, under conditions of pH 7.4 used in this study, IM2 TMD has similar conformations in model lipid bilayers and membranes made of native E. coli lipids and proteins of comparable thickness and fluidity, notwithstanding the complexity of the E. coli membranes caused by their lipid diversity and the abundance of integral and peripheral membrane proteins.
Collapse
Affiliation(s)
- Griffin Sanders
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas
| | - Peter P Borbat
- Department of Chemistry and Chemical Biology, ACERT, Cornell University, Ithaca, New York
| | - Elka R Georgieva
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas.
| |
Collapse
|
28
|
Ogrinc N, Barka EA, Clément C, Salzet M, Sanchez L, Fournier I. In Vivo and Real-Time Metabolic Profiling of Plant-Microbe Interactions in Leaves, Stems, and Roots of Bacterially Inoculated Chardonnay Plantlets using SpiderMass. Anal Chem 2024. [PMID: 39155838 DOI: 10.1021/acs.analchem.4c01470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
There is growing interest in limiting the use of fungicides and implementing innovative, environmentally friendly strategies, such as the use of beneficial bacteria-triggered immunity, to protect grapevines from natural pathogens. Therefore, we need rapid and innovative ways to translate the knowledge of the molecular mechanisms underlying the activation of grapevine defenses against pathogens to induced resistance. Here, we have implemented an in vivo minimally invasive approach to study the interaction between plants and beneficial bacteria based on metabolic signatures. Paraburkholderia phytofirmans strain PsJN and PsJN-grapevine were used as bacterial and plant-bacterium interaction models, respectively. Using an innovative tool, SpiderMass, based on water-assisted laser desorption ionization with an IR microsampling probe, we simultaneously detect metabolic and lipidomic species. A metabolomic spectrum was thus generated, which was used to build a library and identify the most variable and discriminative peaks between the two conditions. We then showed that caftaric acid (m/z 311.04), caftaric acid dimer (m/z 623.09), derived caftaric acid (m/z 653.15), and quercetin-O-glucuronide tended to accumulate in grapevine leaves after root bacterization with PsJN. In addition, together with these phenolic messengers, we identified lipid biomarkers such as palmitic acid, linoleic acid, and α-linoleic acid as important messengers of enhanced defense mechanisms in Chardonnay plantlets. Taken together, SpiderMass is the next-generation methodology for studying plant-microorganism metabolic interactions with the prospect of in vivo real-time analysis in viticulture.
Collapse
Affiliation(s)
- Nina Ogrinc
- Inserm, CHU Lille, U1192-Protéomique Réponse Inflammatoire Spectrométrie de Masse (PRISM), Université de Lille, F-59000 Lille, France
| | - Essaïd Ait Barka
- Unité de Recherche RIBP (Résistance Induite et Bioprotec-tion des Plantes), USC INRAE 1488, Université de Reims Champagne-Ardenne Moulin de la Housse BP 1039, 51687 Reim Cedex 2, France
| | - Christophe Clément
- Unité de Recherche RIBP (Résistance Induite et Bioprotec-tion des Plantes), USC INRAE 1488, Université de Reims Champagne-Ardenne Moulin de la Housse BP 1039, 51687 Reim Cedex 2, France
| | - Michel Salzet
- Inserm, CHU Lille, U1192-Protéomique Réponse Inflammatoire Spectrométrie de Masse (PRISM), Université de Lille, F-59000 Lille, France
| | - Lisa Sanchez
- Unité de Recherche RIBP (Résistance Induite et Bioprotec-tion des Plantes), USC INRAE 1488, Université de Reims Champagne-Ardenne Moulin de la Housse BP 1039, 51687 Reim Cedex 2, France
- Institut Universitaire de France (IUF), Paris, France, https://www.iufrance.fr/
| | - Isabelle Fournier
- Inserm, CHU Lille, U1192-Protéomique Réponse Inflammatoire Spectrométrie de Masse (PRISM), Université de Lille, F-59000 Lille, France
- Institut Universitaire de France (IUF), Paris, France, https://www.iufrance.fr/
| |
Collapse
|
29
|
Khakimzhan A, Izri Z, Thompson S, Dmytrenko O, Fischer P, Beisel C, Noireaux V. Cell-free expression with a quartz crystal microbalance enables rapid, dynamic, and label-free characterization of membrane-interacting proteins. Commun Biol 2024; 7:1005. [PMID: 39152195 PMCID: PMC11329788 DOI: 10.1038/s42003-024-06690-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 08/06/2024] [Indexed: 08/19/2024] Open
Abstract
Integral and interacting membrane proteins (IIMPs) constitute a vast family of biomolecules that perform essential functions in all forms of life. However, characterizing their interactions with lipid bilayers remains limited due to challenges in purifying and reconstituting IIMPs in vitro or labeling IIMPs without disrupting their function in vivo. Here, we report cell-free transcription-translation in a quartz crystal microbalance with dissipation (TXTL-QCMD) to dynamically characterize interactions between diverse IIMPs and membranes without protein purification or labeling. As part of TXTL-QCMD, IIMPs are synthesized using cell-free transcription-translation (TXTL), and their interactions with supported lipid bilayers are measured using a quartz crystal microbalance with dissipation (QCMD). TXTL-QCMD reconstitutes known IIMP-membrane dependencies, including specific association with prokaryotic or eukaryotic membranes, and the multiple-IIMP dynamical pattern-forming association of the E. coli division-coordinating proteins MinCDE. Applying TXTL-QCMD to the recently discovered Zorya anti-phage system that is unamenable to labeling, we discovered that ZorA and ZorB integrate within the lipids found at the poles of bacteria while ZorE diffuses freely on the non-pole membrane. These efforts establish the potential of TXTL-QCMD to broadly characterize the large diversity of IIMPs.
Collapse
Affiliation(s)
- Aset Khakimzhan
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Ziane Izri
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Seth Thompson
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Oleg Dmytrenko
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Centre for Infection Research (HZI), 97080, Würzburg, Germany
| | - Patrick Fischer
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Centre for Infection Research (HZI), 97080, Würzburg, Germany
| | - Chase Beisel
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Centre for Infection Research (HZI), 97080, Würzburg, Germany
- Medical Faculty, University of Würzburg, 97080, Würzburg, Germany
| | - Vincent Noireaux
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
30
|
Dubey AK, Sardana D, Verma T, Alam P, Chattopadhyay A, Nandini SS, Khamari B, Bulagonda EP, Sen S, Nandi D. Quantifying Membrane Alterations with Tailored Fluorescent Dyes: A Rapid Antibiotic Resistance Profiling Methodology. ACS Infect Dis 2024; 10:2836-2859. [PMID: 39024306 DOI: 10.1021/acsinfecdis.4c00249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Accurate detection of bacterial antibiotic sensitivity is crucial for theranostics and the containment of antibiotic-resistant infections. However, the intricate task of detecting and quantifying the antibiotic-induced changes in the bacterial cytoplasmic membrane, and their correlation with other metabolic pathways leading to antibiotic resistance, poses significant challenges. Using a novel class of 4-aminophthalimide (4AP)-based fluorescent dyes with precisely tailored alkyl chains, namely 4AP-C9 and 4AP-C13, we quantify stress-mediated alterations in E. coli membranes. Leveraging the unique depth-dependent positioning and environment-sensitive fluorescence properties of these dyes, we detect antibiotic-induced membrane damage through single-cell imaging and monitoring the fluorescence peak maxima difference ratio (PMDR) of the dyes within the bacterial membrane, complemented by other methods. The correlation between the ROS-induced cytoplasmic membrane damage and the PMDR of dyes quantifies sensitivity against bactericidal antibiotics, which correlates to antibiotic-induced lipid peroxidation. Significantly, our findings largely extend to clinical isolates of E. coli and other ESKAPE pathogens like K. pneumoniae and Enterobacter subspecies. Our data reveal that 4AP-Cn probes can potentially act as precise scales to detect antibiotic-induced membrane damage ("thinning") occurring at a subnanometer scale through the quantification of dyes' PMDR, making them promising membrane dyes for rapid detection of bacterial antibiotic resistance, distinguishing sensitive and resistant infections with high specificity in a clinical setup.
Collapse
Affiliation(s)
- Ashim Kumar Dubey
- Undergraduate Programme, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Deepika Sardana
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Taru Verma
- Centre for BioSystems, Science and Engineering, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Parvez Alam
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Avik Chattopadhyay
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Santhi Sanil Nandini
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Balaram Khamari
- Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Puttaparthi 515134, Andhra Pradesh, India
| | - Eswarappa Pradeep Bulagonda
- Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Puttaparthi 515134, Andhra Pradesh, India
| | - Sobhan Sen
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Dipankar Nandi
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, Karnataka, India
| |
Collapse
|
31
|
Yadav R, Pandey V, Yadav SK, Khare P. Comparative evaluation of biodegradation of chlorpyrifos by various bacterial strains: Kinetics and pathway elucidation. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 203:105989. [PMID: 39084792 DOI: 10.1016/j.pestbp.2024.105989] [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: 01/09/2024] [Revised: 05/29/2024] [Accepted: 06/11/2024] [Indexed: 08/02/2024]
Abstract
The present study focused on the isolation and identification of CP and TCP bacteria degrading bacteria from the rhizospheric zone of aromatic grasses i.e. palmarosa (Cymbopogon martinii (Roxb. Wats), lemongrass (Cymbopogon flexuosus) and vetiver (Chrysopogon zizaniodes (L.) Nash.). So that these isolates alone or in combination with the vegetation of aromatic grasses will be used to clean up CP-contaminated soils. The study also explored enzymatic activities, CO2 release, dechlorination potential, and degradation pathways of bacterial strains. A total of 53 CP-tolerant bacteria were isolated on their physical characteristics and their ability to degrade CP. The ten highly CP-tolerant isolates were Pseudomonas aeruginosa Pa608, three strains of Pseudomonas hibiscicola R4-721 from different rhizosphere, Enterococcus lectis PP2a, Pseudomonas monteilii NBFPALD_RAS131, Enterobacter cloacae L3, Stenotrophomonas maltophilia PEG-390, Escherichia coli ABRL132, and Escherichia coli O104:H4 strain FWSEC0009. The CO2 emission and phosphatase activities of the isolates varied from 3.1 to 8.6 μmol mL-1 and 12.3 to 31 μmol PNP h-1, respectively in the CP medium. The degradation kinetics of CP by these isolates followed a one-phase decay model with a dissipation rate ranging from 0.048 to 0.41 d-1 and a half-life of 1.7-14.3 days. The growth data fitted in the SGompertz equation showed a growth rate (K) of 0.21 ± 0.28 to 0.91 ± 0.33 d-1. The P. monteilii strain had a faster growth rate while E. coli ABRL132 had slower growth among the isolates. The rate of TCP accumulation calculated by the SGompertz equation was 0.21 ± 0.02 to 1.18 ± 0.19 d-1. The Pseudomonas monteilii showed a lower accumulation rate of TCP. Among these, four highly effective isolates were Pseudomonas aeruginosa Pa608, Pseudomonas monteilii NBFPALD_RAS131, Stenotrophomonas maltophilia PEG-390, and Pseudomonas hibiscicola R4-721. Illustrations of the degradation pathways indicated that the difference in metabolic pathways of each isolate was associated with their growth rate, phosphatase, dehydrogenase, oxidase, and dechlorination activities.
Collapse
Affiliation(s)
- Ranu Yadav
- Crop Production and Protection Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Versha Pandey
- Crop Production and Protection Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Santosh Kumar Yadav
- Crop Production and Protection Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India
| | - Puja Khare
- Crop Production and Protection Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
32
|
Maciunas LJ, Rotsides P, D'Lauro EJ, Brady S, Beld J, Loll PJ. The VanS sensor histidine kinase from type-B VRE recognizes vancomycin directly. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.09.548278. [PMID: 37503228 PMCID: PMC10369886 DOI: 10.1101/2023.07.09.548278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
V ancomycin- r esistant e nterococci (VRE) are among the most common causes of nosocomial infections and have been prioritized as targets for new therapeutic development. Many genetically distinct types of VRE have been identified; however, they all share a common suite of resistance genes that function together to confer resistance to vancomycin. Expression of the resistance phenotype is controlled by the VanRS two-component system. This system senses the presence of the antibiotic, and responds by initiating transcription of resistance genes. VanS is a transmembrane sensor histidine kinase, and plays a fundamental role in antibiotic resistance by detecting vancomycin or its effects; it then transduces this signal to the VanR transcription factor, thereby alerting the organism to the presence of the antibiotic. Despite the critical role played by VanS, fundamental questions remain about its function, and in particular about how it senses vancomycin. Here, we focus on a purified VanRS system from one of the most clinically prevalent forms of VRE, type B. We show that in a native-like membrane environment, the autokinase activity of type-B VanS is strongly stimulated by vancomycin. We additionally demonstrate that this effect is mediated by a direct physical interaction between the antibiotic and the type-B VanS protein, and localize the interacting region to the protein's periplasmic domain. This represents the first time that a direct sensing mechanism has been confirmed for any VanS protein. Significance Statement When v ancomycin- r esistant e nterococci (VRE) sense the presence of vancomycin, they remodel their cell walls to block antibiotic binding. This resistance phenotype is controlled by the VanS protein, a histidine kinase that senses the antibiotic or its effects and signals for transcription of resistance genes. However, the mechanism by which VanS detects the antibiotic has remained unclear, with no consensus emerging as to whether the protein interacts directly with vancomycin, or instead detects some downstream consequence of vancomycin's action. Here, we show that for one of the most clinically relevant types of VRE, type B, VanS is activated by direct binding of the antibiotic. Such mechanistic insights will likely prove useful in circumventing vancomycin resistance.
Collapse
|
33
|
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
|
34
|
Karimullina E, Guo Y, Khan HM, Emde T, Quade B, Leo RD, Otwinowski Z, Tieleman Peter D, Borek D, Savchenko A. Structural architecture of TolQ-TolR inner membrane protein complex from opportunistic pathogen Acinetobacter baumannii. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.19.599759. [PMID: 38948712 PMCID: PMC11212960 DOI: 10.1101/2024.06.19.599759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Gram-negative bacteria harness the proton motive force (PMF) within their inner membrane (IM) to uphold the integrity of their cell envelope, an indispensable aspect for both division and survival. The IM TolQ-TolR complex is the essential part of the Tol-Pal system, serving as a conduit for PMF energy transfer to the outer membrane. Here we present cryo-EM reconstructions of Acinetobacter baumannii TolQ in apo and TolR- bound forms at atomic resolution. The apo TolQ configuration manifests as a symmetric pentameric pore, featuring a trans-membrane funnel leading towards a cytoplasmic chamber. In contrast, the TolQ-TolR complex assumes a proton non-permeable stance, characterized by the TolQ pentamer's flexure to accommodate the TolR dimer, where two protomers undergo a translation-based relationship. Our structure-guided analysis and simulations support the rotor-stator mechanism of action, wherein the rotation of the TolQ pentamer harmonizes with the TolR protomers' interplay. These findings broaden our mechanistic comprehension of molecular stator units empowering critical functions within the Gram-negative bacterial cell envelope. Teaser Apo TolQ and TolQ-TolR structures depict structural rearrangements required for cell envelope organization in bacterial cell division.
Collapse
|
35
|
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
|
36
|
Rai AK, Sawasato K, Bennett HC, Kozlova A, Sparagna GC, Bogdanov M, Mitchell AM. Genetic evidence for functional diversification of gram-negative intermembrane phospholipid transporters. PLoS Genet 2024; 20:e1011335. [PMID: 38913742 PMCID: PMC11226057 DOI: 10.1371/journal.pgen.1011335] [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: 03/19/2024] [Revised: 07/05/2024] [Accepted: 06/07/2024] [Indexed: 06/26/2024] Open
Abstract
The outer membrane of gram-negative bacteria is a barrier to chemical and physical stress. Phospholipid transport between the inner and outer membranes has been an area of intense investigation and, in E. coli K-12, it has recently been shown to be mediated by YhdP, TamB, and YdbH, which are suggested to provide hydrophobic channels for phospholipid diffusion, with YhdP and TamB playing the major roles. However, YhdP and TamB have different phenotypes suggesting distinct functions. It remains unclear whether these functions are related to phospholipid metabolism. We investigated a synthetic cold sensitivity caused by deletion of fadR, a transcriptional regulator controlling fatty acid degradation and unsaturated fatty acid production, and yhdP, but not by ΔtamB ΔfadR or ΔydbH ΔfadR. Deletion of tamB recuses the ΔyhdP ΔfadR cold sensitivity further demonstrating the phenotype is related to functional diversification between these genes. The ΔyhdP ΔfadR strain shows a greater increase in cardiolipin upon transfer to the non-permissive temperature and genetically lowering cardiolipin levels can suppress cold sensitivity. These data also reveal a qualitative difference between cardiolipin synthases in E. coli, as deletion of clsA and clsC suppresses cold sensitivity but deletion of clsB does not. Moreover, increased fatty acid saturation is necessary for cold sensitivity and lowering this level genetically or through supplementation of oleic acid suppresses the cold sensitivity of the ΔyhdP ΔfadR strain. Together, our data clearly demonstrate that the diversification of function between YhdP and TamB is related to phospholipid metabolism. Although indirect regulatory effects are possible, we favor the parsimonious hypothesis that YhdP and TamB have differential phospholipid-substrate transport preferences. Thus, our data provide a potential mechanism for independent control of the phospholipid composition of the inner and outer membranes in response to changing conditions based on regulation of abundance or activity of YhdP and TamB.
Collapse
Affiliation(s)
- Ashutosh K. Rai
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| | - Katsuhiro Sawasato
- Department of Biochemistry and Molecular Biology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Haley C. Bennett
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| | - Anastasiia Kozlova
- Department of Biochemistry and Molecular Biology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Genevieve C. Sparagna
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Mikhail Bogdanov
- Department of Biochemistry and Molecular Biology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Angela M. Mitchell
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
37
|
Liao G, Wang T, Li X, Gu J, Jia Q, Wang Z, Li H, Qian Y, Qiu J. Comparison of the Lipid Composition of Milk Fat Globules in Goat ( Capra hircus) Milk during Different Lactations and Human Milk. Foods 2024; 13:1618. [PMID: 38890847 PMCID: PMC11171730 DOI: 10.3390/foods13111618] [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: 04/24/2024] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
Goat milk is considered the optimal substitute for human milk and is characterized by variations in the lipid composition of its fat globules across lactation phases. Therefore, the objective of this study was to thoroughly analyze the differences between goat milk during different lactations and human milk, aiming to offer scientific guidance for the production of functional dairy products. Compared with transitional and mature milk, the findings indicated that the total membrane protein content in goat colostrum exhibited greater similarity to that found in human milk. Additionally, goat milk exhibited higher milk fat globule size, as well as a higher total lipid and protein content than human milk. A total of 1461 lipid molecules across 61 subclasses were identified in goat milk and human milk. The contents of glycerides and glycerophospholipids were higher in goat colostrum, whereas sphingolipids and fatty acids were more abundant in human milk. Meanwhile, the compositions of lipid subclasses were inconsistent. There were 584 differentially expressed lipids identified between human and goat milk, including 47 subclasses that were primarily involved in the metabolism of glycerophospholipids, sphingolipids, and triglycerides. In summary, for both the membrane protein and the lipid composition, there were differences between the milk of different goat lactations and human milk.
Collapse
Affiliation(s)
- Guangqin Liao
- Key Laboratory of Agri-Food Quality and Safety, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Ministry of Agriculture and Rural Affairs, Beijing 100081, China; (G.L.); (T.W.); (X.L.); (J.G.); (Q.J.); (Z.W.); (H.L.); (Y.Q.)
| | - Tiancai Wang
- Key Laboratory of Agri-Food Quality and Safety, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Ministry of Agriculture and Rural Affairs, Beijing 100081, China; (G.L.); (T.W.); (X.L.); (J.G.); (Q.J.); (Z.W.); (H.L.); (Y.Q.)
| | - Xiabing Li
- Key Laboratory of Agri-Food Quality and Safety, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Ministry of Agriculture and Rural Affairs, Beijing 100081, China; (G.L.); (T.W.); (X.L.); (J.G.); (Q.J.); (Z.W.); (H.L.); (Y.Q.)
| | - Jingyi Gu
- Key Laboratory of Agri-Food Quality and Safety, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Ministry of Agriculture and Rural Affairs, Beijing 100081, China; (G.L.); (T.W.); (X.L.); (J.G.); (Q.J.); (Z.W.); (H.L.); (Y.Q.)
| | - Qi Jia
- Key Laboratory of Agri-Food Quality and Safety, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Ministry of Agriculture and Rural Affairs, Beijing 100081, China; (G.L.); (T.W.); (X.L.); (J.G.); (Q.J.); (Z.W.); (H.L.); (Y.Q.)
| | - Zishuang Wang
- Key Laboratory of Agri-Food Quality and Safety, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Ministry of Agriculture and Rural Affairs, Beijing 100081, China; (G.L.); (T.W.); (X.L.); (J.G.); (Q.J.); (Z.W.); (H.L.); (Y.Q.)
| | - Houru Li
- Key Laboratory of Agri-Food Quality and Safety, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Ministry of Agriculture and Rural Affairs, Beijing 100081, China; (G.L.); (T.W.); (X.L.); (J.G.); (Q.J.); (Z.W.); (H.L.); (Y.Q.)
- College of Food and Biological Engineering, Chengdu University, Chengdu 610065, China
| | - Yongzhong Qian
- Key Laboratory of Agri-Food Quality and Safety, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Ministry of Agriculture and Rural Affairs, Beijing 100081, China; (G.L.); (T.W.); (X.L.); (J.G.); (Q.J.); (Z.W.); (H.L.); (Y.Q.)
| | - Jing Qiu
- Key Laboratory of Agri-Food Quality and Safety, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Ministry of Agriculture and Rural Affairs, Beijing 100081, China; (G.L.); (T.W.); (X.L.); (J.G.); (Q.J.); (Z.W.); (H.L.); (Y.Q.)
| |
Collapse
|
38
|
Rai AK, Sawasato K, Bennett HC, Kozlova A, Sparagna GC, Bogdanov M, Mitchell AM. Genetic evidence for functional diversification of gram-negative intermembrane phospholipid transporters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.21.545913. [PMID: 37745482 PMCID: PMC10515749 DOI: 10.1101/2023.06.21.545913] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
The outer membrane of Gram-negative bacteria is a barrier to chemical and physical stress. Phospholipid transport between the inner and outer membranes has been an area of intense investigation and, in E. coli K-12, it has recently been shown to be mediated by YhdP, TamB, and YdbH, which are suggested to provide hydrophobic channels for phospholipid diffusion, with YhdP and TamB playing the major roles. However, YhdP and TamB have different phenotypes suggesting distinct functions. We investigated these functions using synthetic cold sensitivity (at 30 °C) caused by deletion of yhdP and fadR, a transcriptional regulator controlling fatty acid degradation and unsaturated fatty acid production, but not by ΔtamB ΔfadR or ΔydbH ΔfadR,. Deletion of tamB suppresses the ΔyhdP ΔfadR cold sensitivity suggesting this phenotype is related to phospholipid transport. The ΔyhdP ΔfadR strain shows a greater increase in cardiolipin upon transfer to the non-permissive temperature and genetically lowering cardiolipin levels can suppress cold sensitivity. These data also reveal a qualitative difference between cardiolipin synthases in E. coli, as deletion of clsA and clsC suppresses cold sensitivity but deletion of clsB does not despite lower cardiolipin levels. In addition to increased cardiolipin, increased fatty acid saturation is necessary for cold sensitivity and lowering this level genetically or through supplementation of oleic acid suppresses the cold sensitivity of the ΔyhdP ΔfadR strain. Although indirect effects are possible, we favor the parsimonious hypothesis that YhdP and TamB have differential substrate transport preferences, most likely with YhdP preferentially transporting more saturated phospholipids and TamB preferentially transporting more unsaturated phospholipids. We envision cardiolipin contributing to this transport preference by sterically clogging TamB-mediated transport of saturated phospholipids. Thus, our data provide a potential mechanism for independent control of the phospholipid composition of the inner and outer membranes in response to changing conditions.
Collapse
Affiliation(s)
- Ashutosh K. Rai
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Katsuhiro Sawasato
- Department of Biochemistry and Molecular Biology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Haley C. Bennett
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Anastasiia Kozlova
- Department of Biochemistry and Molecular Biology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Genevieve C. Sparagna
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mikhail Bogdanov
- Department of Biochemistry and Molecular Biology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Angela M. Mitchell
- Department of Biology, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
39
|
Eisenhauer K, Weber W, Kemp P, Gebhardt C, Kaufmann M, Tewes N, Zhdanova H, Tietze A, Rauh O, Stein V. Scaling the Functional Nanopore (FuN) Screen: Systematic Evaluation of Self-Assembling Membrane Peptides and Extension with a K +-Responsive Fluorescent Protein Sensor. ACS Synth Biol 2024; 13:1382-1392. [PMID: 38598783 DOI: 10.1021/acssynbio.3c00671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
The functional analysis of protein nanopores is typically conducted in planar lipid bilayers or liposomes exploiting high-resolution but low-throughput electrical and optical read-outs. Yet, the reconstitution of protein nanopores in vitro still constitutes an empiric and low-throughput process. Addressing these limitations, nanopores can now be analyzed using the functional nanopore (FuN) screen exploiting genetically encoded fluorescent protein sensors that resolve distinct nanopore-dependent Ca2+ in- and efflux patterns across the inner membrane of Escherichia coli. With a primary proof-of-concept established for the S2168 holin, and thereof based recombinant nanopore assemblies, the question arises to what extent alternative nanopores can be analyzed with the FuN screen and to what extent alternative fluorescent protein sensors can be adapted. Focusing on self-assembling membrane peptides, three sets of 13 different nanopores are assessed for their capacity to form nanopores in the context of the FuN screen. Nanopores tested comprise both natural and computationally designed nanopores. Further, the FuN screen is extended to K+-specific fluorescent protein sensors and now provides a capacity to assess the specificity of a nanopore or ion channel. Finally, a comparison to high-resolution biophysical and electrophysiological studies in planar lipid bilayers provides an experimental benchmark for future studies.
Collapse
Affiliation(s)
- Klara Eisenhauer
- Department of Biology, TU Darmstadt, 64287 Darmstadt, Germany
- Centre for Synthetic Biology, TU Darmstadt, 64283 Darmstadt, Germany
| | - Wadim Weber
- Department of Biology, TU Darmstadt, 64287 Darmstadt, Germany
- Centre for Synthetic Biology, TU Darmstadt, 64283 Darmstadt, Germany
| | - Philipp Kemp
- Department of Biology, TU Darmstadt, 64287 Darmstadt, Germany
- Centre for Synthetic Biology, TU Darmstadt, 64283 Darmstadt, Germany
| | - Carolin Gebhardt
- Department of Biology, TU Darmstadt, 64287 Darmstadt, Germany
- Centre for Synthetic Biology, TU Darmstadt, 64283 Darmstadt, Germany
| | - Marwan Kaufmann
- Department of Biology, TU Darmstadt, 64287 Darmstadt, Germany
- Centre for Synthetic Biology, TU Darmstadt, 64283 Darmstadt, Germany
| | - Noel Tewes
- Department of Biology, TU Darmstadt, 64287 Darmstadt, Germany
| | - Hanna Zhdanova
- Department of Chemistry and Molecular Biology, Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 412 96 Göteborg, Sweden
| | - Alesia Tietze
- Department of Chemistry and Molecular Biology, Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 412 96 Göteborg, Sweden
| | - Oliver Rauh
- Department of Biology, TU Darmstadt, 64287 Darmstadt, Germany
| | - Viktor Stein
- Department of Biology, TU Darmstadt, 64287 Darmstadt, Germany
- Centre for Synthetic Biology, TU Darmstadt, 64283 Darmstadt, Germany
| |
Collapse
|
40
|
Strange N, Luu L, Ong V, Wee BA, Phillips MJA, McCaughey L, Steele JR, Barlow CK, Cranfield CG, Myers G, Mazraani R, Rock C, Timms P, Huston WM. HtrA, fatty acids, and membrane protein interplay in Chlamydia trachomatis to impact stress response and trigger early cellular exit. J Bacteriol 2024; 206:e0037123. [PMID: 38445896 PMCID: PMC11025325 DOI: 10.1128/jb.00371-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 02/15/2024] [Indexed: 03/07/2024] Open
Abstract
Chlamydia trachomatis is an intracellular bacterial pathogen that undergoes a biphasic developmental cycle, consisting of intracellular reticulate bodies and extracellular infectious elementary bodies. A conserved bacterial protease, HtrA, was shown previously to be essential for Chlamydia during the reticulate body phase, using a novel inhibitor (JO146). In this study, isolates selected for the survival of JO146 treatment were found to have polymorphisms in the acyl-acyl carrier protein synthetase gene (aasC). AasC encodes the enzyme responsible for activating fatty acids from the host cell or synthesis to be incorporated into lipid bilayers. The isolates had distinct lipidomes with varied fatty acid compositions. A reduction in the lipid compositions that HtrA prefers to bind to was detected, yet HtrA and MOMP (a key outer membrane protein) were present at higher levels in the variants. Reduced progeny production and an earlier cellular exit were observed. Transcriptome analysis identified that multiple genes were downregulated in the variants especially stress and DNA processing factors. Here, we have shown that the fatty acid composition of chlamydial lipids, HtrA, and membrane proteins interplay and, when disrupted, impact chlamydial stress response that could trigger early cellular exit. IMPORTANCE Chlamydia trachomatis is an important obligate intracellular pathogen that has a unique biphasic developmental cycle. HtrA is an essential stress or virulence protease in many bacteria, with many different functions. Previously, we demonstrated that HtrA is critical for Chlamydia using a novel inhibitor. In the present study, we characterized genetic variants of Chlamydia trachomatis with reduced susceptibility to the HtrA inhibitor. The variants were changed in membrane fatty acid composition, outer membrane proteins, and transcription of stress genes. Earlier and more synchronous cellular exit was observed. Combined, this links stress response to fatty acids, membrane proteins, and HtrA interplay with the outcome of disrupted timing of chlamydial cellular exit.
Collapse
Affiliation(s)
- Natalie Strange
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Laurence Luu
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Vanissa Ong
- Faculty of Health, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Bryan A. Wee
- Faculty of Health, Queensland University of Technology, Kelvin Grove, Queensland, Australia
- The Roslin Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Matthew J. A. Phillips
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Laura McCaughey
- Australian Institute for Microbiology and Infection, Faculty of Science, University of Technology Sydney, Ultimo, New South Wales, Australia
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Joel R. Steele
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, New South Wales, Australia
- Department of Biochemistry and Molecular Biology, Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Christopher K. Barlow
- Department of Biochemistry and Molecular Biology, Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Charles G. Cranfield
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Garry Myers
- Australian Institute for Microbiology and Infection, Faculty of Science, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Rami Mazraani
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Charles Rock
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Peter Timms
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore, Queensland, Australia
| | - Wilhelmina M. Huston
- Faculty of Science, University of Technology Sydney, Ultimo, New South Wales, Australia
| |
Collapse
|
41
|
Verma RK, Gondu P, Saha T, Chatterjee S. The Global Transcription Regulator XooClp Governs Type IV Pili System-Mediated Bacterial Virulence by Directly Binding to TFP-Chp Promoters to Coordinate Virulence Associated Functions. MOLECULAR PLANT-MICROBE INTERACTIONS : MPMI 2024; 37:357-369. [PMID: 38105438 DOI: 10.1094/mpmi-07-23-0100-r] [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: 12/19/2023]
Abstract
Type IV pili (TFP) play a crucial role in the sensing of the external environment for several bacteria. This surface sensing is essential for the lifestyle transitions of several bacteria and involvement in pathogenesis. However, the precise mechanisms underlying TFP's integration of environmental cues, particularly in regulating the TFP-Chp system and its effects on Xanthomonas physiology, social behavior, and virulence, remain poorly understood. In this study, we focused on investigating Clp, a global transcriptional regulator similar to CRP-like proteins, in Xanthomonas oryzae pv. oryzae, a plant pathogen. Our findings reveal that Clp integrates environmental cues detected through diffusible signaling factor (DSF) quorum sensing into the TFP-Chp regulatory system. It accomplishes this by directly binding to TFP-Chp promoters in conjunction with intracellular levels of cyclic-di-GMP, a ubiquitous bacterial second messenger, thereby controlling TFP expression. Moreover, Clp-mediated regulation is involved in regulating several cellular processes, including the production of virulence-associated functions. Collectively, these processes contribute to host colonization and disease initiation. Our study elucidates the intricate regulatory network encompassing Clp, environmental cues, and the TFP-Chp system, providing insights into the molecular mechanisms that drive bacterial virulence in Xanthomonas spp. These findings offer valuable knowledge regarding Xanthomonas pathogenicity and present new avenues for innovative strategies aimed at combating plant diseases caused by these bacteria. [Formula: see text] Copyright © 2024 The Author(s). This is an open access article distributed under the CC BY-NC-ND 4.0 International license.
Collapse
Affiliation(s)
- Raj Kumar Verma
- Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad 500039, India
| | - Parimala Gondu
- Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad 500039, India
| | - Tirthankar Saha
- Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad 500039, India
| | | |
Collapse
|
42
|
Salvati B, Flórez-Castillo JM, Santagapita PR, Barja BC, Perullini M. One-pot synthesis of alginate-antimicrobial peptide nanogel. Photochem Photobiol Sci 2024; 23:665-679. [PMID: 38443738 DOI: 10.1007/s43630-024-00542-5] [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: 10/09/2023] [Accepted: 01/23/2024] [Indexed: 03/07/2024]
Abstract
Nanosized alginate-based particles (NAPs) were obtained in a one-pot solvent-free synthesis procedure, achieving the design of a biocompatible nanocarrier for the encapsulation of IbM6 antimicrobial peptide (IbM6). IbM6 is integrated in the nascent nanosized hydrogel self-assembly guided by electrostatic interactions and by weak interactions, typical of soft matter. The formation of the nanogel is a dynamic and complex process, which presents an interesting temporal evolution. In this work, we optimized the synthesis conditions of IbM6-NAPs based on small-angle X-ray scattering (SAXS) measurements and evaluated its time evolution over several weeks by sensing the IbM6 environment in IbM6-NAPs from photochemical experiments. Fluorescence deactivation experiments revealed that the accessibility of different quenchers to the IbM6 peptide embedded in NAPs is dependent on the aging time of the alginate network. Lifetimes measurements indicate that the deactivation paths of the excited state of the IbM6 in the nanoaggregates are reduced when compared with those exhibited by the peptide in aqueous solution, and are also dependent on the aging time of the nanosized alginate network. Finally, the entrapment of IbM6 in NAPs hinders the degradation of the peptide by trypsin, increasing its antimicrobial activity against Escherichia coli K-12 in simulated operation conditions.
Collapse
Affiliation(s)
- Brianne Salvati
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Inorgánica, Analítica y Química Física (DQIAQF), Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química de Materiales medio Ambiente y Energía (INQUIMAE), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Johanna Marcela Flórez-Castillo
- Universidad de Magdalena, Santa Marta, Colombia
- Universidad de Santander UDES, Grupo de Investigación en Ciencias Básicas y Aplicadas para la Sostenibilidad-CIBAS, Santander, Colombia
| | - Patricio Román Santagapita
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Orgánica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Centro de Investigaciones en Hidratos de Carbono (CIHIDECAR), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Beatriz C Barja
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Inorgánica, Analítica y Química Física (DQIAQF), Universidad de Buenos Aires, Buenos Aires, Argentina.
- Instituto de Química de Materiales medio Ambiente y Energía (INQUIMAE), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Mercedes Perullini
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Inorgánica, Analítica y Química Física (DQIAQF), Universidad de Buenos Aires, Buenos Aires, Argentina.
- Instituto de Química de Materiales medio Ambiente y Energía (INQUIMAE), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
43
|
Shobhna, Dutta A, Kumari P, Kashyap HK. Stability of Cytoplasmic Membrane of Escherichia coli Bacteria in Aqueous and Ethanolic Environment. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:2893-2906. [PMID: 38311936 DOI: 10.1021/acs.langmuir.3c02780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
The mechanism of action of any antibacterial agent or disinfectant depends largely on their interaction with the bacterial membrane. Herein, we use the SPICA (surface property fitting coarse graining) force-field and develop a coarse-grained (CG) model for the structure of the cytoplasmic membrane of Escherichia coli (E. coli) and its interaction with water and ethanol. We elucidate the impact of different concentrations of ethanol on the cytoplasmic membrane bilayers and vesicles of E. coli using the CG molecular dynamics (CG MD) simulations. Our modeling approach first focuses on the parametrization of the required force-field for POPG lipid and its interaction with water, ethanol, and POPE lipid. Subsequently, the structural stability of the E. coli bacterial membrane in the presence of high and low concentrations of ethanol is delineated. Both flat bilayers as well as vesicles of E. coli membrane were considered for the CG MD. Our results reveal that, at low ethanol concentrations (<30 mol %), the size of the E. coli vesicles increases with discernible deformations in their shapes. Because of ethanol-induced interdigitation, thinning of the E. coli vesicular membrane is also observed. However, at higher ethanol concentrations (>30 mol %), the integrity of the vesicles is lost because of deteriorating invasion of ethanol molecules into the vesicle bilayer and significant weakening of lipid-lipid interactions. At higher ethanol concentrations (40 and 70 mol %), both the multivesicle and single-vesicle bacterial membranes exhibit a similar rupturing pattern wherein the extraction of lipids from the membrane and formation of aggregates of the component lipids are observed. These aggregates consist of polar head groups of 3-5 POPE/POPG lipids with intertwined nonpolar tails.
Collapse
Affiliation(s)
- Shobhna
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Ayishwarya Dutta
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Pratibha Kumari
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Hemant K Kashyap
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| |
Collapse
|
44
|
Zhang Y, Cui X, Lin S, Lu T, Li H, Lu Y, Cao M, Lin X, Ling X. Knockout of a PLD gene in Schizochytrium limacinum SR21 enhances docosahexaenoic acid accumulation by modulation of the phospholipid profile. BIOTECHNOLOGY FOR BIOFUELS AND BIOPRODUCTS 2024; 17:16. [PMID: 38291531 PMCID: PMC10826259 DOI: 10.1186/s13068-024-02465-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 01/18/2024] [Indexed: 02/01/2024]
Abstract
BACKGROUND The hydrolysis and transphosphatidylation of phospholipase D (PLD) play important roles in the interconversion of phospholipids (PLs), which has been shown to profoundly impact lipid metabolism in plants. In this study, the effect of the PLD1 gene of Schizochytrium limacinum SR21 (S. limacinum SR21) on lipid metabolism was investigated. RESULTS PLD1 knockout had little impact on cell growth and lipid production, but it significantly improved the percentage of polyunsaturated fatty acids in lipids, of which docosahexaenoic acid (DHA) content increased by 13.3% compared to the wild-type strain. Phospholipomics and real-time quantitative PCR analysis revealed the knockout of PLD1 reduced the interexchange and increased de novo synthesis of PLs, which altered the composition of PLs, accompanied by a final decrease in phosphatidylcholine (PC) and an increase in phosphatidylinositol, lysophosphatidylcholine, and phosphatidic acid levels. PLD1 knockout also increased DHA content in triglycerides (TAGs) and decreased it in PLs. CONCLUSIONS These results indicate that PLD1 mainly performs the transphosphatidylation activity in S. limacinum SR21, and its knockout promotes the migration of DHA from PLs to TAGs, which is conducive to DHA accumulation and storage in TAGs via an acyl CoA-independent pathway. This study provides a novel approach for identifying the mechanism of DHA accumulation and metabolic regulation strategies for DHA production in S. limacinum SR21.
Collapse
Affiliation(s)
- Yiting Zhang
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Xiaowen Cui
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Shuizhi Lin
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Tao Lu
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Hao Li
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
| | - Yinghua Lu
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
- Xiamen Key Laboratory of Synthetic Biotechnology, Xiamen University, Xiamen, People's Republic of China
- The Key Laboratory for Chemical Biology of Fujian Province (Xiamen University), Xiamen, People's Republic of China
| | - Mingfeng Cao
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China
- Xiamen Key Laboratory of Synthetic Biotechnology, Xiamen University, Xiamen, People's Republic of China
| | - Xihuang Lin
- Analysis and Test Center, Ministry of Natural Resources, Third Institute of Oceanography, Xiamen, 361005, People's Republic of China.
| | - Xueping Ling
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, People's Republic of China.
- Xiamen Key Laboratory of Synthetic Biotechnology, Xiamen University, Xiamen, People's Republic of China.
| |
Collapse
|
45
|
Britt M, Sawasato K, Moller E, Kidd G, Bogdanov M, Sukharev S. On the lipid dependence of bacterial mechanosensitive channel gating in situ. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.576706. [PMID: 38328048 PMCID: PMC10849563 DOI: 10.1101/2024.01.22.576706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
For bacterial mechanosensitive channels acting as turgor-adjusting osmolyte release valves, membrane tension is the primary stimulus driving opening transitions. Because tension is transmitted through the surrounding lipid bilayer, it is possible that the presence or absence of different lipid species may influence the function of these channels. In this work, we characterize the lipid dependence of chromosome-encoded MscS and MscL in E. coli strains with genetically altered lipid composition. We use two previously generated strains that lack one or two major lipid species (PE, PG, or CL) and engineer a third strain that is highly enriched in CL due to the presence of hyperactive cardiolipin synthase ClsA. We characterize the functional behavior of these channels using patch-clamp and quantify the relative tension midpoints, closing rates, inactivation depth, and the rate of recovery back to the closed state. We also measure the osmotic survival of lipid-deficient strains, which characterizes the functional consequences of lipid-mediated channel function at the cell level. We find that the opening and closing behavior of MscS and MscL tolerate the absence of specific lipid species remarkably well. The lack of cardiolipin (CL), however, reduces the active MscS population relative to MscL and decreases the closing rate, slightly increasing the propensity of MscS toward inactivation and slowing the recovery process. The data points to the robustness of the osmolyte release system and the importance of cardiolipin for the adaptive behavior of MscS.
Collapse
|
46
|
Sanders G, Borbat PP, Georgieva ER. A comparative study of influenza A M2 protein conformations in DOPC/DOPS liposomes and in native E. coli membranes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.08.574681. [PMID: 38260371 PMCID: PMC10802500 DOI: 10.1101/2024.01.08.574681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
We compared the conformations of the transmembrane domain (TMD) of influenza A M2 (IAM2) protein reconstituted at pH 7.4 in DOPC/DOPS bilayers to those in isolated E. coli membranes, having preserved its native proteins and lipids. IAM2 is a single-pass transmembrane protein known to assemble into homo-tetrameric proton channel. To represent this channel, we made a construct containing the IAM2's TMD region flanked by the juxtamembrane residues. The single cysteine substitute, L43C, of leucine located in the bilayer polar region was paramagnetically tagged with a methanethiosulfonate nitroxide label for the ESR (electron spin resonance) study. We compared the conformations of the spin-labeled IAM2 residing in DOPC/DOPS and native E. coli membranes using continuous-wave (CW) ESR and double electron-electron resonance (DEER) spectroscopy. The total protein-to-lipid molar ratio spanned the range from 1:230 to 1:10,400⩦ The CW ESR spectra corresponded to a nearly rigid limit spin label dynamics in both environments. In all cases, the DEER data were reconstructed into the distance distributions showing well-resolved peaks at 1.68 nm and 2.37 nm. The peak distance ratio was 1.41±0.2 and the amplitude ratio was 2:1. This is what one expects from four nitroxide spin-labels located at the corners of a square, indicative of an axially symmetric tetramer. Distance modeling of DEER data with molecular modeling software applied to the NMR molecular structures (PDB: 2L0J) confirmed the symmetry and closed state of the C-terminal exit pore of the IAM2 tetramer in agreement with the NMR model. Thus, we can conclude that IAM2 TMD has similar conformations in model and native E. coli membranes of comparable thickness and fluidity, notwithstanding the complexity of the E. coli membranes caused by their lipid diversity and the abundance of integral and peripheral membrane proteins.
Collapse
Affiliation(s)
- Griffin Sanders
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, 79409
| | - Peter P. Borbat
- Department of Chemistry and Chemical Biology and ACERT, Cornell University, Ithaca NY 14853
| | - Elka R. Georgieva
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, 79409
| |
Collapse
|
47
|
Venkatraman K, Lee CT, Garcia GC, Mahapatra A, Milshteyn D, Perkins G, Kim K, Pasolli HA, Phan S, Lippincott‐Schwartz J, Ellisman MH, Rangamani P, Budin I. Cristae formation is a mechanical buckling event controlled by the inner mitochondrial membrane lipidome. EMBO J 2023; 42:e114054. [PMID: 37933600 PMCID: PMC10711667 DOI: 10.15252/embj.2023114054] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 11/08/2023] Open
Abstract
Cristae are high-curvature structures in the inner mitochondrial membrane (IMM) that are crucial for ATP production. While cristae-shaping proteins have been defined, analogous lipid-based mechanisms have yet to be elucidated. Here, we combine experimental lipidome dissection with multi-scale modeling to investigate how lipid interactions dictate IMM morphology and ATP generation. When modulating phospholipid (PL) saturation in engineered yeast strains, we observed a surprisingly abrupt breakpoint in IMM topology driven by a continuous loss of ATP synthase organization at cristae ridges. We found that cardiolipin (CL) specifically buffers the inner mitochondrial membrane against curvature loss, an effect that is independent of ATP synthase dimerization. To explain this interaction, we developed a continuum model for cristae tubule formation that integrates both lipid and protein-mediated curvatures. This model highlighted a snapthrough instability, which drives IMM collapse upon small changes in membrane properties. We also showed that cardiolipin is essential in low-oxygen conditions that promote PL saturation. These results demonstrate that the mechanical function of cardiolipin is dependent on the surrounding lipid and protein components of the IMM.
Collapse
Affiliation(s)
- Kailash Venkatraman
- Department of Chemistry and BiochemistryUniversity of California San DiegoLa JollaCAUSA
| | - Christopher T Lee
- Department of Mechanical and Aerospace EngineeringUniversity of California San DiegoLa JollaCAUSA
| | - Guadalupe C Garcia
- Computational Neurobiology LaboratorySalk Institute for Biological StudiesLa JollaCAUSA
| | - Arijit Mahapatra
- Department of Mechanical and Aerospace EngineeringUniversity of California San DiegoLa JollaCAUSA
- Present address:
Applied Physical SciencesUniversity of North Carolina Chapel HillChapel HillNCUSA
| | - Daniel Milshteyn
- Department of Chemistry and BiochemistryUniversity of California San DiegoLa JollaCAUSA
| | - Guy Perkins
- National Center for Microscopy and Imaging Research, Center for Research in Biological SystemsUniversity of California San DiegoLa JollaCAUSA
| | - Keun‐Young Kim
- National Center for Microscopy and Imaging Research, Center for Research in Biological SystemsUniversity of California San DiegoLa JollaCAUSA
| | - H Amalia Pasolli
- Howard Hughes Medical InstituteAshburnVAUSA
- Present address:
Electron Microscopy Resource CenterThe Rockefeller UniversityNew YorkNYUSA
| | - Sebastien Phan
- National Center for Microscopy and Imaging Research, Center for Research in Biological SystemsUniversity of California San DiegoLa JollaCAUSA
| | | | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, Center for Research in Biological SystemsUniversity of California San DiegoLa JollaCAUSA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace EngineeringUniversity of California San DiegoLa JollaCAUSA
| | - Itay Budin
- Department of Chemistry and BiochemistryUniversity of California San DiegoLa JollaCAUSA
| |
Collapse
|
48
|
Schnoor SB, Neubauer P, Gimpel M. Recent insights into the world of dual-function bacterial sRNAs. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023:e1824. [PMID: 38039556 DOI: 10.1002/wrna.1824] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 12/03/2023]
Abstract
Dual-function sRNAs refer to a small subgroup of small regulatory RNAs that merges base-pairing properties of antisense RNAs with peptide-encoding properties of mRNA. Both functions can be part of either same or in another metabolic pathway. Here, we want to update the knowledge of to the already known dual-function sRNAs and review the six new sRNAs found since 2017 regarding their structure, functional mechanisms, evolutionary conservation, and role in the regulation of distinct biological/physiological processes. The increasing identification of dual-function sRNAs through bioinformatics approaches, RNomics and RNA-sequencing and the associated increase in regulatory understanding will likely continue to increase at the same rate in the future. This may improve our understanding of the physiology, virulence and resistance of bacteria, as well as enable their use in technical applications. This article is categorized under: Regulatory RNAs/RNAi/Riboswitches > Regulatory RNAs.
Collapse
Affiliation(s)
| | - Peter Neubauer
- Department of Bioprocess Engineering, Technische Universitat Berlin, Berlin, Germany
| | - Matthias Gimpel
- Department of Bioprocess Engineering, Technische Universitat Berlin, Berlin, Germany
| |
Collapse
|
49
|
Rempfert KR, Kraus EA, Nothaft DB, Dildar N, Spear JR, Sepúlveda J, Templeton AS. Intact polar lipidome and membrane adaptations of microbial communities inhabiting serpentinite-hosted fluids. Front Microbiol 2023; 14:1198786. [PMID: 38029177 PMCID: PMC10667739 DOI: 10.3389/fmicb.2023.1198786] [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: 04/02/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
The generation of hydrogen and reduced carbon compounds during serpentinization provides sustained energy for microorganisms on Earth, and possibly on other extraterrestrial bodies (e.g., Mars, icy satellites). However, the geochemical conditions that arise from water-rock reaction also challenge the known limits of microbial physiology, such as hyperalkaline pH, limited electron acceptors and inorganic carbon. Because cell membranes act as a primary barrier between a cell and its environment, lipids are a vital component in microbial acclimation to challenging physicochemical conditions. To probe the diversity of cell membrane lipids produced in serpentinizing settings and identify membrane adaptations to this environment, we conducted the first comprehensive intact polar lipid (IPL) biomarker survey of microbial communities inhabiting the subsurface at a terrestrial site of serpentinization. We used an expansive, custom environmental lipid database that expands the application of targeted and untargeted lipodomics in the study of microbial and biogeochemical processes. IPLs extracted from serpentinite-hosted fluid communities were comprised of >90% isoprenoidal and non-isoprenoidal diether glycolipids likely produced by archaeal methanogens and sulfate-reducing bacteria. Phospholipids only constituted ~1% of the intact polar lipidome. In addition to abundant diether glycolipids, betaine and trimethylated-ornithine aminolipids and glycosphingolipids were also detected, indicating pervasive membrane modifications in response to phosphate limitation. The carbon oxidation state of IPL backbones was positively correlated with the reduction potential of fluids, which may signify an energy conservation strategy for lipid synthesis. Together, these data suggest microorganisms inhabiting serpentinites possess a unique combination of membrane adaptations that allow for their survival in polyextreme environments. The persistence of IPLs in fluids beyond the presence of their source organisms, as indicated by 16S rRNA genes and transcripts, is promising for the detection of extinct life in serpentinizing settings through lipid biomarker signatures. These data contribute new insights into the complexity of lipid structures generated in actively serpentinizing environments and provide valuable context to aid in the reconstruction of past microbial activity from fossil lipid records of terrestrial serpentinites and the search for biosignatures elsewhere in our solar system.
Collapse
Affiliation(s)
- Kaitlin R. Rempfert
- Department of Geological Sciences, University of Colorado, Boulder, CO, United States
| | - Emily A. Kraus
- Department of Civil and Environmental Engineering, Colorado School of Mines, Golden, CO, United States
| | - Daniel B. Nothaft
- Department of Geological Sciences, University of Colorado, Boulder, CO, United States
| | - Nadia Dildar
- Department of Geological Sciences, University of Colorado, Boulder, CO, United States
| | - John R. Spear
- Department of Civil and Environmental Engineering, Colorado School of Mines, Golden, CO, United States
- Department of Quantitative Biosciences and Engineering, Colorado School of Mines, Golden, CO, United States
| | - Julio Sepúlveda
- Department of Geological Sciences, University of Colorado, Boulder, CO, United States
| | - Alexis S. Templeton
- Department of Geological Sciences, University of Colorado, Boulder, CO, United States
| |
Collapse
|
50
|
Walczak-Skierska J, Monedeiro F, Maślak E, Złoch M. Lipidomics Characterization of the Microbiome in People with Diabetic Foot Infection Using MALDI-TOF MS. Anal Chem 2023; 95:16251-16262. [PMID: 37877781 PMCID: PMC10633811 DOI: 10.1021/acs.analchem.3c03071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/11/2023] [Accepted: 10/09/2023] [Indexed: 10/26/2023]
Abstract
Lipidomic profiling has emerged as a powerful tool for the comprehensive characterization of bacterial species, particularly in the context of clinical diagnostics. Utilizing matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS), this study aims to elucidate the lipidomic landscapes of bacterial strains isolated from diabetic foot infections (DFI). Our analysis successfully identified a diverse array of lipids in the cellular membranes of both Gram-positive and Gram-negative bacteria, revealing a total of 108 unique fatty acid combinations. Specifically, we identified 26 LPG, 33 LPE, 43 PE, 114 PG, 89 TAG, and 120 CLP in Gram-positive bacteria and 10 LPG, 14 LPE, 124 PE, 37 PG, 13 TAG, and 22 CLP in Gram-negative strains. Key fatty acids, such as palmitic acid, palmitoleic acid, stearic acid, and oleic acid, were prominently featured. Univariate analysis further highlighted distinct lipidomic signatures among the bacterial strains, revealing elevated levels of phosphatidylethanolamine (PE) and phosphatidylglycerol (PG) in Gram-negative bacteria associated with DFI. In contrast, Gram-positive strains demonstrated increased or uniquely fluctuating levels of triglyceride (TAG) and cardiolipin (CLP). These findings not only underscore the utility of MALDI-TOF MS in bacterial lipidomics but also provide valuable insights into the lipidomic adaptations of bacteria in diabetic foot infections, thereby laying the groundwork for future studies aimed at constructing microbial lipid libraries for enhanced bacterial identification.
Collapse
Affiliation(s)
- Justyna Walczak-Skierska
- Centre
for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Wileńska 4 Str., 87-100 Toruń, Poland
- Chair
of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus a Copernicus University in Toruń, Gagarina 7 Str., 87-100 Toruń, Poland
| | - Fernanda Monedeiro
- Centre
for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Wileńska 4 Str., 87-100 Toruń, Poland
| | - Ewelina Maślak
- Centre
for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Wileńska 4 Str., 87-100 Toruń, Poland
| | - Michał Złoch
- Centre
for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Wileńska 4 Str., 87-100 Toruń, Poland
- Chair
of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus a Copernicus University in Toruń, Gagarina 7 Str., 87-100 Toruń, Poland
| |
Collapse
|