1
|
Liu H, Yu Y, Wang C, Wang Y, Wu R, Zhang Z, Liu D, Liao M, Rong X, Li B, Luo Z, Zhang Z. The molecular mechanism of gspD gene in regulating the biological characteristics, pathogenicity and virulence gene expression of Photobacterium damselae subsp. damselae. Int J Biol Macromol 2025; 306:141559. [PMID: 40020817 DOI: 10.1016/j.ijbiomac.2025.141559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/14/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
The pathogenic bacterium Photobacterium damselae subsp. damselae (PDD) has the capacity to infect various mariculture fish species. The Type II secretion system is a component of PDD, facilitating the secretion of extracellular products. The gspD gene encodes the outer membrane secretory channel protein of T2SS. To investigate the role of the gspD gene in T2SS during PDD infection, we generated a gspD gene deletion mutant of PDD (ΔgspD-PDD) using a suicide plasmid-mediated homologous recombination technique and compared the biological characteristics, virulence gene expression, and pathogenicity of ΔgspD-PDD with those of the wild-type strain (WT-PDD). Our results indicated that the hemolytic activity and phospholipase activity of ΔgspD-PDD were significantly diminished compared to WT-PDD, and the complementation strain was restored to levels similar to those of the WT-PDD. The expression levels of T2SS-related genes and the virulence genes were significantly down-regulated, while the outer membrane-related gene and flagella-related genes exhibited significant up-regulation. The LD50 values of ΔgspD-PDD and its ECP in Sebastes schlegelii were 62.70-fold and 18.76-fold higher than those of WT-PDD, respectively. In summary, the mutation of the gspD gene may lead to the down-regulation of T2SS-related genes, resulting in aberrant secretion of ECPs in PDD and subsequently diminishing its pathogenicity.
Collapse
Affiliation(s)
- Haozhe Liu
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academic of Fishery Sciences, Qingdao, Shandong 266071, China; College of Fisheries, Tianjin Agricultural University, Tianjin 300392, China
| | - Yongxiang Yu
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academic of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory Qingdao, Shandong 266237, China
| | - Chunyuan Wang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academic of Fishery Sciences, Qingdao, Shandong 266071, China
| | - Yingeng Wang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academic of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory Qingdao, Shandong 266237, China
| | - Ronghua Wu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Aquaculture Engineering Technology Research Center, College of Fisheries, Southwest University, Chongqing 400715, China
| | - Zhiqi Zhang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academic of Fishery Sciences, Qingdao, Shandong 266071, China
| | - Dingyuan Liu
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academic of Fishery Sciences, Qingdao, Shandong 266071, China
| | - Meijie Liao
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academic of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory Qingdao, Shandong 266237, China
| | - Xiaojun Rong
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academic of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory Qingdao, Shandong 266237, China
| | - Bin Li
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academic of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory Qingdao, Shandong 266237, China
| | - Zhang Luo
- College of Fisheries, Tianjin Agricultural University, Tianjin 300392, China
| | - Zheng Zhang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academic of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory Qingdao, Shandong 266237, China.
| |
Collapse
|
2
|
Kaufmann H, Salvador C, Salazar VW, Cruz N, Dias GM, Tschoeke D, Campos L, Sawabe T, Miyazaki M, Maruyama F, Thompson F, Thompson C. Genomic Repertoire of Twenty-Two Novel Vibrionaceae Species Isolated from Marine Sediments. MICROBIAL ECOLOGY 2025; 88:36. [PMID: 40301151 PMCID: PMC12041005 DOI: 10.1007/s00248-025-02533-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 04/10/2025] [Indexed: 05/01/2025]
Abstract
The genomic repertoire of vibrios has been extensively studied, particularly regarding their metabolic plasticity, symbiotic interactions, and resistance mechanisms to environmental stressors. However, little is known about the genomic diversity and adaptations of vibrios inhabiting deep-sea marine sediments. In this study, we investigated the genomic diversity of vibrios isolated from deep-sea core sediments collected using a manned submersible off Japan. A total of 50 vibrio isolates were obtained and characterized phenotypically, and by genome sequencing. From this total, we disclosed 22 novel species examining genome-to-genome distance, average amino acid identity, and phenotypes (Alivibrio: 1; Enterovibrio: 1; Photobacterium: 8; Vibrio: 12). The novel species have fallen within known clades (e.g., Fisheri, Enterovibrio, Profundum, and Splendidus) and novel clades (JAMM0721, JAMM0388, JAMM0395). The 28 remainder isolates were identified as known species: Aliivibrio sifiae (2), A. salmonicida (1), Enterovibrio baiacu (1), E. norvegicus (1), Photobacterium profundum (3), P. angustum (1), P. chitiniliticum (1), P. frigidiphilum (1), Photobacterium indicum (1), P. sanguinicancri (1). P. swingsii (2), Vibrio alginolyticus (3), V. anguillarum (1), V. campbellii (1), V. fluvialis (1), V. gigantis (1), V. lentus (1), V. splendidus (4), and V. tasmaniensis (1). Genomic analyses revealed that all 50 vibrios harbored genes associated with high-pressure adaptation, including sensor kinases, chaperones, autoinducer-2 (AI-2) signaling, oxidative damage repair, polyunsaturated fatty acid biosynthesis, and stress response mechanisms related to periplasmic and outer membrane protein misfolding under heat shock and osmotic stress. Additionally, alternative sigma factors, trimethylamine oxide (TMAO) respiration, and osmoprotectant acquisition pathways were identified, further supporting their ability to thrive in deep-sea environments. Notably, the genomes exhibited a high prevalence of antibiotic resistance genes, with antibiotic efflux pumps being the most abundant group. The ugd gene expanded in number in some novel species (Photobacterium satsumensis sp. nov. JAMM1754: 4 copies; Vibrio makurazakiensis sp. nov. JAMM1826: 3 copies). This gene may confer antibiotic (polymyxin) resistance to these vibrios.
Collapse
Affiliation(s)
- Hannah Kaufmann
- Laboratory of Microbiology, Institute of Biology, UFRJ, Rio de Janeiro, Brazil
| | - Carolina Salvador
- Laboratory of Microbiology, Institute of Biology, UFRJ, Rio de Janeiro, Brazil
| | - Vinicius W Salazar
- Laboratory of Microbiology, Institute of Biology, UFRJ, Rio de Janeiro, Brazil
- Faculty of Medicine, Dentistry, and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Natália Cruz
- Laboratory of Microbiology, Institute of Biology, UFRJ, Rio de Janeiro, Brazil
| | - Graciela Maria Dias
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Diogo Tschoeke
- Laboratory of Microbiology, Institute of Biology, UFRJ, Rio de Janeiro, Brazil
- Instituto Alberto Luiz Coimbra de Pós-Graduação E Pesquisa de Engenharia (COPPE), Universidade Federal Do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Lucia Campos
- Laboratory of Microbiology, Institute of Biology, UFRJ, Rio de Janeiro, Brazil
| | - Tomoo Sawabe
- Laboratory of Microbiology, Fisheries Sciences School, Hokkaido University, Hakodate, Japan
| | - Masayuki Miyazaki
- Institute for Extra-Cutting-Edge Science and Technology Avant-Garde Research (X-Star), Yokosuka, Japan
- Agency for Marine-Earth Science and Technology (JAMSTEC), Yokosuka, Japan
| | - Fumito Maruyama
- Microbial Genomics and Ecology Laboratory, Hiroshima University, Hiroshima, Japan
| | - Fabiano Thompson
- Laboratory of Microbiology, Institute of Biology, UFRJ, Rio de Janeiro, Brazil.
| | - Cristiane Thompson
- Laboratory of Microbiology, Institute of Biology, UFRJ, Rio de Janeiro, Brazil.
| |
Collapse
|
3
|
Tavakoli H, Najaflou M, Yarikhosroushahi A. Biomaterial-based chitosan nanohydrogel films: combination of Bistorta officinalis and Ca-doped carbon dots for improved blood clotting. J Biol Eng 2025; 19:31. [PMID: 40211334 PMCID: PMC11987453 DOI: 10.1186/s13036-025-00498-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 04/01/2025] [Indexed: 04/13/2025] Open
Abstract
BACKGROUND Bleeding and traumatic injuries are still a major issue necessitating the development of advanced hemostatic materials that are economical, biocompatible, and effective. Chitosan's (CS) haemostatic and biocompatible properties make it a promising wound-healing material, however, effective cross-linking is essential for appropriate physiochemical properties. In this study, calcium-doped carbon dots (CDs) produced from coriander leaves were used as cross-linking agents to improve the functional performance and structural integrity of nanohydrogel films. Furthermore, extract of the medicinal plant Bistorta officinalis (BEX), a traditional medicinal plant with strong hemostatic and antibacterial qualities, was incorporated into the hydrogel matrix. RESULTS Analysis and characterization of the synthesized CDs thoroughly confirmed that they have monodispersed spherical shape, negative zeta potential, and active functional groups which effectively cross-linked the chitosan matrix and increased the mechanical strength and stability of the film. Cytotoxicity and antibacterial results of the final films showed the desired cytocompatibility against Human skin fibroblast (HFF-1 cells) with over 80% viability at the highest concentration and effective antibacterial activity against gram-positive and gram-negative bacteria (further improved by cross-linking with CDs and incorporating BEX), respectively. The incorporation of BEX and CDs in hydrogel films significantly enhanced the film's blood-clotting ability with negligible hemolysis due to blood clotting index and hemolysis tests. CONCLUSIONS The findings of this study highlight the potential of biomaterial-based nano hydrogel film, composed of CS cross-linked with CDs and containing BEX, as a promising wound dressing with outstanding biocompatibility, minimal cytotoxicity, enhanced hemostatic efficacy, and strong antibacterial properties.
Collapse
Affiliation(s)
- Hassan Tavakoli
- Department of Chemistry, Faculty of Basic Sciences, Imam Ali University, Imam Khomeini Street, P.O. Box 1317893471, Tehran, 1317893471, Iran.
| | - Meysam Najaflou
- Drug Applied Research Center, Tabriz University of Medical Sciences, Golgasht Street, Tabriz, 516615731, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Golgasht Street, Tabriz, 516615731, Iran
| | - Ahmad Yarikhosroushahi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Golgasht Street, Tabriz, 516615731, Iran.
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Golgasht Street, Tabriz, 516615731, Iran.
| |
Collapse
|
4
|
De Smet T, Baland E, Giovannercole F, Mignon J, Lizen L, Dugauquier R, Lauber F, Dieu M, Lima-Mendez G, Michaux C, Devos D, Renzi F. LolA and LolB are conserved in Bacteroidota and are crucial for gliding motility and Type IX secretion. Commun Biol 2025; 8:376. [PMID: 40050408 PMCID: PMC11885536 DOI: 10.1038/s42003-025-07817-2] [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: 11/18/2024] [Accepted: 02/25/2025] [Indexed: 03/09/2025] Open
Abstract
Lipoproteins are key outer membrane (OM) components in Gram-negative bacteria, essential for functions like membrane biogenesis and virulence. Bacteroidota, a diverse and widespread phylum, produce numerous OM lipoproteins that play vital roles in nutrient acquisition, Type IX secretion system (T9SS), and gliding motility. In Escherichia coli, lipoprotein transport to the OM is mediated by the Lol system, where LolA shuttles lipoproteins to LolB, which anchors them in the OM. However, LolB homologs were previously thought to be limited to γ- and β-proteobacteria. This study uncovers the presence of LolB in Bacteroidota and demonstrates that multiple LolA and LolB proteins co-exist in various species. Specifically, in Flavobacterium johnsoniae, LolA1 and LolB1 transport gliding motility and T9SS lipoproteins to the OM. Notably, these proteins are not interchangeable with their E. coli counterparts, indicating functional specialization. Some lipoproteins still localize to the OM in the absence of LolA and LolB, suggesting the existence of alternative transport pathways in Bacteroidota. This points to a more complex lipoprotein transport system in Bacteroidota compared to other Gram-negative bacteria. These findings reveal previously unrecognized lipoprotein transport mechanisms in Bacteroidota and suggest that this phylum has evolved unique strategies to manage the essential task of lipoprotein localization.
Collapse
Affiliation(s)
- Tom De Smet
- Research Unit in Biology of Microorganisms (URBM), Namur Research Institute for Life Sciences (Narilis), University of Namur, Namur, Belgium
| | - Elisabeth Baland
- Research Unit in Biology of Microorganisms (URBM), Namur Research Institute for Life Sciences (Narilis), University of Namur, Namur, Belgium
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - Fabio Giovannercole
- Research Unit in Biology of Microorganisms (URBM), Namur Research Institute for Life Sciences (Narilis), University of Namur, Namur, Belgium
| | - Julien Mignon
- Laboratoire de Chimie Physique des Biomolécules, UCPTS, Namur Institute of Structured Matter (NISM), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Laura Lizen
- Research Unit in Biology of Microorganisms (URBM), Namur Research Institute for Life Sciences (Narilis), University of Namur, Namur, Belgium
- Laboratoire de Chimie Bactérienne (LCB) CNRS-Aix-Marseille University, Marseille, France
| | - Rémy Dugauquier
- Research Unit in Biology of Microorganisms (URBM), Namur Research Institute for Life Sciences (Narilis), University of Namur, Namur, Belgium
| | - Frédéric Lauber
- Research Unit in Biology of Microorganisms (URBM), Namur Research Institute for Life Sciences (Narilis), University of Namur, Namur, Belgium
- De Duve Institute, UCLouvain, Brussels, Belgium
| | - Marc Dieu
- Technological Platform Mass Spectrometry Service (MaSUN), Namur Research Institute for Life Sciences (Narilis), University of Namur, Namur, Belgium
| | - Gipsi Lima-Mendez
- Research Unit in Biology of Microorganisms (URBM), Namur Research Institute for Life Sciences (Narilis), University of Namur, Namur, Belgium
| | - Catherine Michaux
- Laboratoire de Chimie Physique des Biomolécules, UCPTS, Namur Institute of Structured Matter (NISM), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Damien Devos
- Centro Andaluz de Biología del Desarrollo (CSIC), Universidad Pablo de Olavide, Sevilla, Spain
- Center for Infection and Immunity of Lille, Pasteur Institute, Lille, France
| | - Francesco Renzi
- Research Unit in Biology of Microorganisms (URBM), Namur Research Institute for Life Sciences (Narilis), University of Namur, Namur, Belgium.
| |
Collapse
|
5
|
Zhang M, Li J, Xue J, Xu H, Li M, Xia Y, Qi C, Zhang P, Liu Y, Liu J. Development and evaluation of optimized PCR and indirect ELISA for the detection of Morganella morganii in dairy cows. Front Vet Sci 2025; 12:1532600. [PMID: 39886025 PMCID: PMC11774872 DOI: 10.3389/fvets.2025.1532600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025] Open
Abstract
Introduction Morganella morganii (M. morganii) is a Gram-negative opportunistic pathogen, whose increasing virulence and antibiotic resistance negatively impact dairy cow health and productivity, raising concerns in livestock health management. To mitigate this risk, rapid and reliable diagnostic methods for detection are essential. Currently, detection methods for M. morganii are underdeveloped, prompting us to develop both pathogenic and serological detection methods, including an optimized PCR technique and an indirect enzyme-linked immunosorbent assay (I-ELISA). Methods The optimized PCR method utilized bacterial suspensions directly as templates, bypassing the need for DNA extraction and thereby allowing the direct detection of M. morganii in fecal samples. Primer concentrations and annealing temperatures were optimized to minimize primer dimer formation, ensuring high specificity. Clinical evaluation was conducted using 771 fecal and nasal fluid samples collected from dairy farms in five regions. The I-ELISA method was developed using M. morganii lipoprotein (LPP) antigen. Parameters such as antigen coating, blocking conditions, and antibody dilution were optimized to improve specificity. Stability and reproducibility were validated through intra- and inter-assay tests. A total of 476 serum samples from dairy cows were tested to assess the method's clinical applicability. Results The optimized PCR method demonstrated high sensitivity and specificity, achieving a detection threshold of 0.2 CFU/μL. Clinical testing revealed a positivity rate of 1.4% among 771 fecal and nasal fluid samples. The I-ELISA method showed excellent stability and reproducibility, confirmed through intra- and inter-assay consistency. In testing 476 dairy cow serum samples, the positivity rate for M. morganii was 5.9%. These results indicate the utility of I-ELISA as a reliable serological diagnostic tool. Discussion The PCR and I-ELISA methods collectively offer practical solutions for the early clinical diagnosis of M. morganii infections in dairy cows. The PCR technique's efficiency and sensitivity make it ideal for pathogen detection in fecal samples, while the I-ELISA method provides a robust platform for serological analysis. Together, these tools enable timely intervention, contributing to improved livestock health management and mitigating the negative impacts of M. morganii on dairy cow productivity. Future research may focus on further refining these techniques and exploring their applications in broader livestock management contexts.
Collapse
Affiliation(s)
- Meihua Zhang
- College of Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Jiayi Li
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, Tai’an, China
| | - Jianfeng Xue
- The Affiliated Tai’an City Central Hospital of Qingdao University, Tai’an, China
| | - Huiling Xu
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, Tai’an, China
| | - Muzi Li
- College of Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Yibo Xia
- College of Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Changxi Qi
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, Tai’an, China
| | - Pu Zhang
- The Affiliated Tai’an City Central Hospital of Qingdao University, Tai’an, China
| | - Yongxia Liu
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, Tai’an, China
| | - Jianzhu Liu
- College of Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| |
Collapse
|
6
|
Robertson P, Allan DS, Garduño RA. The Passage of Chaperonins to Extracellular Locations in Legionella pneumophila Requires a Functional Dot/Icm System. Biomolecules 2025; 15:91. [PMID: 39858485 PMCID: PMC11763710 DOI: 10.3390/biom15010091] [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: 12/05/2024] [Revised: 12/31/2024] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
HtpB, the chaperonin of the bacterial pathogen L. pneumophila, is found in extracellular locations, even the cytoplasm of host cells. Although chaperonins have an essential cytoplasmic function in protein folding, HtpB exits the cytoplasm to perform extracellular virulence-related functions that support L. pneumophila's lifestyle. The mechanism by which HtpB reaches extracellular locations is not currently understood. To address this experimental gap, immunoelectron microscopy, trypsin-accessibility assays, and cell fractionation were used to localize HtpB in various L. pneumophila secretion mutants. Dot/Icm type IV secretion mutants displayed less surface-exposed HtpB and more periplasmic HtpB than parent strains. The analysis of periplasmic extracts and outer membrane vesicles of these mutants, where HtpB co-localized with bona fide periplasmic proteins, confirmed the elevated levels of periplasmic HtpB. Genetic complementation of the mutants recovered parent strain levels of surface-exposed and periplasmic HtpB. The export of GSK-tagged HtpB into the cytoplasm of infected cells was also Dot/Icm-dependent. The translocating role of the Dot/Icm system was not specific for HtpB because GroEL, the chaperonin of Escherichia coli, was found at the cell surface and accumulated in the periplasm of Dot mutants when expressed in L. pneumophila. These findings establish that a functional Dot/Icm system is required for HtpB to reach extracellular locations, but the mechanism by which cytoplasmic HtpB reaches the periplasm remains partially unidentified.
Collapse
Affiliation(s)
- Peter Robertson
- Department of Microbiology-Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; (P.R.); (D.S.A.)
| | - David S. Allan
- Department of Microbiology-Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; (P.R.); (D.S.A.)
| | - Rafael A. Garduño
- Department of Microbiology-Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; (P.R.); (D.S.A.)
- Department of Medicine, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 2Y9, Canada
| |
Collapse
|
7
|
Kalia M, Sauer K. Distinct transcriptome and traits of freshly dispersed Pseudomonas aeruginosa cells. mSphere 2024; 9:e0088424. [PMID: 39601567 DOI: 10.1128/msphere.00884-24] [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: 10/17/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Bacteria assume two distinct lifestyles: the planktonic and biofilm modes of growth. Additionally, dispersion has emerged as a third phenotype, accompanied by the distinct phenotypes and the unique expression of >600 genes. Here, we asked whether the distinct phenotype of dispersed cells is already apparent within minutes of egressing from the biofilm. We used RNA-seq to show that the physiology of freshly dispersed cells from Pseudomonas aeruginosa biofilms is highly different from those of planktonic and biofilm cells, apparent by dispersed cells uniquely expressing 194 genes. Unique and differentially expressed genes relative to planktonic or biofilm cells include genes associated with type IV pili, pyoverdine, type III and type VI secretion systems, and antibiotic resistance that are downregulated in dispersed cells, whereas the transcript abundance of genes involved in swimming motility, Hxc type II secretion system and various other virulence factors, and metabolic and energy-generating pathways are increased, indicative of dispersion coinciding with an awakening and re-energizing of dispersed cells, and a switch in virulence, further apparent by freshly dispersed cells significantly subverting engulfment by macrophages. The findings suggest that dispersed cells display a distinct phenotype within minutes of egressing from the biofilm, with freshly dispersed cells already capable of efficiently evading phagocytosis. IMPORTANCE Dispersion is considered a transitionary phenotype, enabling bacteria to switch between the communal, biofilm lifestyle, where cells share resources and are protected from harmful conditions to the planktonic state. Here, we demonstrate that within minutes of leaving the biofilm, dispersed cells express genes and display phenotypic traits that are distinct from biofilms and planktonic cells. Our findings suggest that dispersed cells quickly adapt to a less structured and protected but more nutrient-rich environment, with this trade-off in environment coinciding with an awakening and a switch in virulence, specifically a switch from directly intoxicating host cells and potential competitors toward more broadly active virulence factors and strategies of evasion. To our knowledge, this is the first report of dispersed cells' distinct (trade-off) phenotype and their enhanced resilience so soon after egressing from the biofilm.
Collapse
Affiliation(s)
- Manmohit Kalia
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| | - Karin Sauer
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| |
Collapse
|
8
|
Hori S, Okazaki F. Identification, heterologous expression, and characterisation of β-1,3-xylanase BcXyn26B from human gut bacterium Bacteroides cellulosilyticus WH2. Biotechnol Lett 2024; 47:10. [PMID: 39621179 PMCID: PMC11611983 DOI: 10.1007/s10529-024-03547-3] [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: 08/02/2024] [Revised: 09/26/2024] [Accepted: 10/19/2024] [Indexed: 12/06/2024]
Abstract
The cell walls of red and green algae contain β-1,3-xylan, which is hydrolysed by the endo-type enzyme β-1,3-xylanase. Notably, only marine-bacteria-derived β-1,3-xylanases have been functionally characterised to date. In this study, we characterised the enzymatic properties of a potential β-1,3-xylanase (BcXyn26B) derived from the human gut bacterium, Bacteroides cellulosilyticus WH2. The codon optimized BcXyn26B gene was synthesised and expressed in Escherichia coli BL21(DE3). The recombinant protein was purified by a two-step purification process using Ni-affinity chromatography followed by anion exchange chromatography, and its enzymatic properties were characterised. The recombinant BcXyn26B exhibited specific hydrolytic activity against β-1,3-xylan and released various β-1,3-xylooligosaccharides, with β-1,3-xylobiose as the primary product. The optimum reaction temperature was 50 °C, higher than that for other enzymes derived from marine bacteria. This study represents the first report on the identification, heterologous expression, and characterisation of β-1,3-xylanase from human gut microbes. Notably, the substrate specificity of BcXyn26B indicates that human gut Bacteroides species possess an unknown β-1,3-xylan utilisation system.
Collapse
Affiliation(s)
- Sanae Hori
- Department of Life Sciences, Graduate School of Bioresources, Mie University, 1577 Kurimamachiya, Tsu, Mie, 514-8507, Japan
| | - Fumiyoshi Okazaki
- Department of Life Sciences, Graduate School of Bioresources, Mie University, 1577 Kurimamachiya, Tsu, Mie, 514-8507, Japan.
| |
Collapse
|
9
|
Perva IT, Simina IE, Bende R, Motofelea AC, Chirita Emandi A, Andreescu N, Sima A, Vlad A, Sporea I, Zimbru C, Tutac PC, Puiu M, Niculescu MD. Use of a Micronutrient Cocktail to Improve Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) in Adults with Obesity: A Randomized, Double-Blinded Pilot Clinical Trial. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1366. [PMID: 39202647 PMCID: PMC11356300 DOI: 10.3390/medicina60081366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/06/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024]
Abstract
Background and Objectives: The goal of this study was to assess the impact of supplementation with a combination of nutrients on metabolic-dysfunction-associated steatotic liver disease (MASLD)-related liver parameters, and other parameters related to metabolic syndrome in adults with obesity. These measurements included anthropometric and lipid profiling, and FibroScan technology (controlled attenuation parameter (CAP) and transient elastography (TE) values). Materials and Methods: A double-blind, placebo-controlled pilot clinical trial was conducted over a three-month treatment period. Adults with metabolic syndrome and obesity were allocated to receive either a cocktail of nutrients with defined daily dosages (5-MTHF, betaine, alpha-linolenic acid, eicosapentaenoic acid, choline bitartrate, docosahexaenoic acid, and vitamin B12) or a placebo. The participants were evaluated at the start and the end of the three-month treatment period. Results: A total of 155 participants entered the study, comprising 84 in the treatment group and 71 in the placebo group. The administration of the nutritional supplement resulted in a notable reduction in both CAP and TE scores when compared to the placebo group. The treatment group exhibited a mean reduction in CAP of 4% (p < 0.05) and a mean reduction in TE of 7.8% (p < 0.05), indicative of a decline in liver fat content and fibrosis. Conclusions: The supplementation over a period of three months led to a significant amelioration of liver fibrosis and steatosis parameters in adults with metabolic syndrome and obesity. These findings suggest that this supplementation regimen could be a beneficial adjunct therapy for improving liver health in adults with obesity-induced MASLD.
Collapse
Affiliation(s)
- Iulia Teodora Perva
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (I.T.P.); (A.C.E.); (N.A.); (M.P.); (M.D.N.)
- Regional Center of Medical Genetics Timiș, Clinical Emergency Hospital for Children “Louis Țurcanu”, Iosif Nemoianu Street N°2, 300011 Timisoara, Romania
- Department of Medical Genetics, Asociatia Oncohelp, 300239 Timișoara, Romania
| | - Iulia Elena Simina
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (I.T.P.); (A.C.E.); (N.A.); (M.P.); (M.D.N.)
- Department of Medical Genetics, Asociatia Oncohelp, 300239 Timișoara, Romania
| | - Renata Bende
- Department of Gastroenterology and Hepatology, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (R.B.); (I.S.)
- Center of Advanced Research in Gastroenterology and Hepatology, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Alexandru Cătălin Motofelea
- Department of Internal Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania;
| | - Adela Chirita Emandi
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (I.T.P.); (A.C.E.); (N.A.); (M.P.); (M.D.N.)
- Regional Center of Medical Genetics Timiș, Clinical Emergency Hospital for Children “Louis Țurcanu”, Iosif Nemoianu Street N°2, 300011 Timisoara, Romania
| | - Nicoleta Andreescu
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (I.T.P.); (A.C.E.); (N.A.); (M.P.); (M.D.N.)
- Regional Center of Medical Genetics Timiș, Clinical Emergency Hospital for Children “Louis Țurcanu”, Iosif Nemoianu Street N°2, 300011 Timisoara, Romania
| | - Alexandra Sima
- Department of Internal Medicine II, Division of Diabetes, Nutrition and Metabolic Diseases, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (A.S.); (A.V.)
- Center for Research in Preventive Medicine, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Adrian Vlad
- Department of Internal Medicine II, Division of Diabetes, Nutrition and Metabolic Diseases, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (A.S.); (A.V.)
- Center for Molecular Research in Nephrology and Vascular Disease, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Ioan Sporea
- Department of Gastroenterology and Hepatology, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (R.B.); (I.S.)
- Center of Advanced Research in Gastroenterology and Hepatology, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Cristian Zimbru
- Department of Automation and Applied Informatics, Politehnica University Timișoara, 300223 Timișoara, Romania;
| | - Paul Calin Tutac
- Toxicology and Molecular Biology Department, “Pius Brinzeu” Clinical Emergency County Hospital, 300723 Timisoara, Romania;
| | - Maria Puiu
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (I.T.P.); (A.C.E.); (N.A.); (M.P.); (M.D.N.)
- Regional Center of Medical Genetics Timiș, Clinical Emergency Hospital for Children “Louis Țurcanu”, Iosif Nemoianu Street N°2, 300011 Timisoara, Romania
| | - Mihai Dinu Niculescu
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (I.T.P.); (A.C.E.); (N.A.); (M.P.); (M.D.N.)
- Advanced Nutrigenomics LLC, Durham, NC 27703, USA
| |
Collapse
|
10
|
Cianciotto NP. The type II secretion system as an underappreciated and understudied mediator of interbacterial antagonism. Infect Immun 2024; 92:e0020724. [PMID: 38980047 PMCID: PMC11320942 DOI: 10.1128/iai.00207-24] [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] [Indexed: 07/10/2024] Open
Abstract
Interbacterial antagonism involves all major phyla, occurs across the full range of ecological niches, and has great significance for the environment, clinical arena, and agricultural and industrial sectors. Though the earliest insight into interbacterial antagonism traces back to the discovery of antibiotics, a paradigm shift happened when it was learned that protein secretion systems (e.g., types VI and IV secretion systems) deliver toxic "effectors" against competitors. However, a link between interbacterial antagonism and the Gram-negative type II secretion system (T2SS), which exists in many pathogens and environmental species, is not evident in prior reviews on bacterial competition or T2SS function. A current examination of the literature revealed four examples of a T2SS or one of its known substrates having a bactericidal activity against a Gram-positive target or another Gram-negative. When further studied, the T2SS effectors proved to be peptidases that target the peptidoglycan of the competitor. There are also reports of various bacteriolytic enzymes occurring in the culture supernatants of some other Gram-negative species, and a link between these bactericidal activities and T2SS is suggested. Thus, a T2SS can be a mediator of interbacterial antagonism, and it is possible that many T2SSs have antibacterial outputs. Yet, at present, the T2SS remains relatively understudied for its role in interbacterial competition. Arguably, there is a need to analyze the T2SSs of a broader range of species for their role in interbacterial antagonism. Such investigation offers, among other things, a possible pathway toward developing new antimicrobials for treating disease.
Collapse
Affiliation(s)
- Nicholas P. Cianciotto
- Department of Microbiology-Immunology, Northwestern University School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
11
|
Kataoka N. Ketogluconate production by Gluconobacter strains: enzymes and biotechnological applications. Biosci Biotechnol Biochem 2024; 88:499-508. [PMID: 38323387 DOI: 10.1093/bbb/zbae013] [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: 12/26/2023] [Accepted: 02/01/2024] [Indexed: 02/08/2024]
Abstract
Gluconobacter strains perform incomplete oxidation of various sugars and alcohols, employing regio- and stereoselective membrane-bound dehydrogenases oriented toward the periplasmic space. This oxidative fermentation process is utilized industrially. The ketogluconate production pathway, characteristic of these strains, begins with the conversion of d-glucose to d-gluconate, which then diverges and splits into 2 pathways producing 5-keto-d-gluconate and 2-keto-d-gluconate and subsequently 2,5-diketo-d-gluconate. These transformations are facilitated by membrane-bound d-glucose dehydrogenase, glycerol dehydrogenase, d-gluconate dehydrogenase, and 2-keto-d-gluconate dehydrogenase. The variance in end products across Gluconobacter strains stems from the diversity of enzymes and their activities. This review synthesizes biochemical and genetic knowledge with biotechnological applications, highlighting recent advances in metabolic engineering and the development of an efficient production process focusing on enzymes relevant to the ketogluconate production pathway in Gluconobacter strains.
Collapse
Affiliation(s)
- Naoya Kataoka
- Organization for Research Initiatives, Yamaguchi University, Yamaguchi, Japan
- Research Center for Thermotolerant Microbial Resources, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
12
|
Mintz KP, Danforth DR, Ruiz T. The Trimeric Autotransporter Adhesin EmaA and Infective Endocarditis. Pathogens 2024; 13:99. [PMID: 38392837 PMCID: PMC10892112 DOI: 10.3390/pathogens13020099] [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: 12/11/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Infective endocarditis (IE), a disease of the endocardial surface of the heart, is usually of bacterial origin and disproportionally affects individuals with underlying structural heart disease. Although IE is typically associated with Gram-positive bacteria, a minority of cases are caused by a group of Gram-negative species referred to as the HACEK group. These species, classically associated with the oral cavity, consist of bacteria from the genera Haemophilus (excluding Haemophilus influenzae), Aggregatibacter, Cardiobacterium, Eikenella, and Kingella. Aggregatibacter actinomycetemcomitans, a bacterium of the Pasteurellaceae family, is classically associated with Aggressive Periodontitis and is also concomitant with the chronic form of the disease. Bacterial colonization of the oral cavity serves as a reservoir for infection at distal body sites via hematological spreading. A. actinomycetemcomitans adheres to and causes disease at multiple physiologic niches using a diverse array of bacterial cell surface structures, which include both fimbrial and nonfimbrial adhesins. The nonfimbrial adhesin EmaA (extracellular matrix binding protein adhesin A), which displays sequence heterogeneity dependent on the serotype of the bacterium, has been identified as a virulence determinant in the initiation of IE. In this chapter, we will discuss the known biochemical, molecular, and structural aspects of this protein, including its interactions with extracellular matrix components and how this multifunctional adhesin may contribute to the pathogenicity of A. actinomycetemcomitans.
Collapse
Affiliation(s)
- Keith P. Mintz
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405, USA;
| | - David R. Danforth
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405, USA;
| | - Teresa Ruiz
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT 05405, USA;
| |
Collapse
|
13
|
Liu C, Angius F, Pol A, Mesman RA, Versantvoort W, Op den Camp HJM. Identification and characterization of an abundant lipoprotein from Methylacidiphilum fumariolicum SolV. Arch Microbiol 2023; 205:261. [PMID: 37306788 DOI: 10.1007/s00203-023-03603-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/17/2023] [Accepted: 05/30/2023] [Indexed: 06/13/2023]
Abstract
Bacterial lipoproteins are characterized by the presence of a conserved N-terminal lipid-modified cysteine residue that allows the hydrophilic protein to anchor into bacterial cell membranes. These lipoproteins play essential roles in a wide variety of physiological processes. Based on transcriptome analysis of the verrucomicrobial methanotroph Methylacidiphilum fumariolicum SolV, we identified a highly expressed lipoprotein, WP_009060351 (139 amino acids), in its genome. The first 86 amino acids are specific for the methanotrophic genera Methylacidiphilum and Methylacidmicrobium, while the last 53 amino acids are present only in lipoproteins of members from the phylum Verrucomicrobiota (Hedlund). Heterologous expression of WP_009060351 in Escherichia coli revealed a 25-kDa dimeric protein and a 60-kDa tetrameric protein. Immunoblotting showed that WP_009060351 was present in the total membrane protein and peptidoglycan fractions of M. fumariolicum SolV. The results suggest an involvement of lipoprotein WP_009060351 in the linkage between the outer membrane and the peptidoglycan.
Collapse
Affiliation(s)
- Changqing Liu
- Faculty of Science, Department of Microbiology, Radboud Institute for Biological and Environmental Sciences, Radboud University, Nijmegen, The Netherlands
| | - Federica Angius
- Faculty of Science, Department of Microbiology, Radboud Institute for Biological and Environmental Sciences, Radboud University, Nijmegen, The Netherlands
| | - Arjan Pol
- Faculty of Science, Department of Microbiology, Radboud Institute for Biological and Environmental Sciences, Radboud University, Nijmegen, The Netherlands
| | - Rob A Mesman
- Faculty of Science, Department of Microbiology, Radboud Institute for Biological and Environmental Sciences, Radboud University, Nijmegen, The Netherlands
| | - Wouter Versantvoort
- Faculty of Science, Department of Microbiology, Radboud Institute for Biological and Environmental Sciences, Radboud University, Nijmegen, The Netherlands
| | - Huub J M Op den Camp
- Faculty of Science, Department of Microbiology, Radboud Institute for Biological and Environmental Sciences, Radboud University, Nijmegen, The Netherlands.
| |
Collapse
|
14
|
Bilsing FL, Anlauf MT, Hachani E, Khosa S, Schmitt L. ABC Transporters in Bacterial Nanomachineries. Int J Mol Sci 2023; 24:ijms24076227. [PMID: 37047196 PMCID: PMC10094684 DOI: 10.3390/ijms24076227] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/18/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Members of the superfamily of ABC transporters are found in all domains of life. Most of these primary active transporters act as isolated entities and export or import their substrates in an ATP-dependent manner across biological membranes. However, some ABC transporters are also part of larger protein complexes, so-called nanomachineries that catalyze the vectorial transport of their substrates. Here, we will focus on four bacterial examples of such nanomachineries: the Mac system providing drug resistance, the Lpt system catalyzing vectorial LPS transport, the Mla system responsible for phospholipid transport, and the Lol system, which is required for lipoprotein transport to the outer membrane of Gram-negative bacteria. For all four systems, we tried to summarize the existing data and provide a structure-function analysis highlighting the mechanistical aspect of the coupling of ATP hydrolysis to substrate translocation.
Collapse
|
15
|
Pelicic V. Mechanism of assembly of type 4 filaments: everything you always wanted to know (but were afraid to ask). MICROBIOLOGY (READING, ENGLAND) 2023; 169. [PMID: 36947586 DOI: 10.1099/mic.0.001311] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
Type 4 filaments (T4F) are a superfamily of filamentous nanomachines - virtually ubiquitous in prokaryotes and functionally versatile - of which type 4 pili (T4P) are the defining member. T4F are polymers of type 4 pilins, assembled by conserved multi-protein machineries. They have long been an important topic for research because they are key virulence factors in numerous bacterial pathogens. Our poor understanding of the molecular mechanisms of T4F assembly is a serious hindrance to the design of anti-T4F therapeutics. This review attempts to shed light on the fundamental mechanistic principles at play in T4F assembly by focusing on similarities rather than differences between several (mostly bacterial) T4F. This holistic approach, complemented by the revolutionary ability of artificial intelligence to predict protein structures, led to an intriguing mechanistic model of T4F assembly.
Collapse
Affiliation(s)
- Vladimir Pelicic
- Laboratoire de Chimie Bactérienne, UMR 7283 CNRS/Aix-Marseille Université, Institut de Microbiologie de la Méditerranée, Marseille, France
| |
Collapse
|
16
|
Dazzoni R, Li Y, López-Castilla A, Brier S, Mechaly A, Cordier F, Haouz A, Nilges M, Francetic O, Bardiaux B, Izadi-Pruneyre N. Structure and dynamic association of an assembly platform subcomplex of the bacterial type II secretion system. Structure 2023; 31:152-165.e7. [PMID: 36586404 DOI: 10.1016/j.str.2022.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/20/2022] [Accepted: 11/29/2022] [Indexed: 12/31/2022]
Abstract
Type II secretion systems (T2SSs) allow diderm bacteria to secrete hydrolytic enzymes, adhesins, or toxins important for growth and virulence. To promote secretion of folded proteins, T2SSs assemble periplasmic filaments called pseudopili or endopili at an inner membrane subcomplex, the assembly platform (AP). Here, we combined biophysical approaches, nuclear magnetic resonance (NMR) and X-ray crystallography, to study the Klebsiella AP components PulL and PulM. We determined the structure and associations of their periplasmic domains and describe the structure of the heterodimer formed by their ferredoxin-like domains. We show how structural complementarity and plasticity favor their association during the secretion process. Cysteine scanning and crosslinking data provided additional constraints to build a structural model of the PulL-PulM assembly in the cellular context. Our structural and functional insights, together with the relative cellular abundance of its components, support the role of AP as a dynamic hub that orchestrates pilus polymerization.
Collapse
Affiliation(s)
- Régine Dazzoni
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Structural Bioinformatics Unit, 75015 Paris, France
| | - Yuanyuan Li
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Biochemistry of Macromolecular Interactions Unit, 75015 Paris, France
| | - Aracelys López-Castilla
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Structural Bioinformatics Unit, 75015 Paris, France
| | - Sébastien Brier
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Biological NMR and HDX-MS Technological Platform, 75015 Paris, France
| | - Ariel Mechaly
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Crystallography Platform, 75015 Paris, France
| | - Florence Cordier
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Structural Bioinformatics Unit, 75015 Paris, France; Institut Pasteur, Université Paris Cité, CNRS UMR3528, Biological NMR and HDX-MS Technological Platform, 75015 Paris, France
| | - Ahmed Haouz
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Crystallography Platform, 75015 Paris, France
| | - Michael Nilges
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Structural Bioinformatics Unit, 75015 Paris, France
| | - Olivera Francetic
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Biochemistry of Macromolecular Interactions Unit, 75015 Paris, France
| | - Benjamin Bardiaux
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Structural Bioinformatics Unit, 75015 Paris, France; Institut Pasteur, Université Paris Cité, CNRS UMR3528, Bacterial Transmembrane Systems Unit, 75015 Paris.
| | - Nadia Izadi-Pruneyre
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Structural Bioinformatics Unit, 75015 Paris, France; Institut Pasteur, Université Paris Cité, CNRS UMR3528, Bacterial Transmembrane Systems Unit, 75015 Paris.
| |
Collapse
|
17
|
Secretome of Paenibacillus sp. S-12 provides an insight about its survival and possible pathogenicity. Folia Microbiol (Praha) 2023:10.1007/s12223-023-01032-4. [PMID: 36642775 DOI: 10.1007/s12223-023-01032-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 01/01/2023] [Indexed: 01/17/2023]
Abstract
Our aim in this study was to characterize and investigate the secretome of Paenibacillus sp. S-12 by nanoLC-MS/MS tool-based analysis of trypsin digested culture supernatant proteins. Using a bioinformatics and combined approach of mass spectrometry, we identified 657 proteins in the secretome. Bioinformatic tools such as PREDLIPO, SecretomeP 2.0, SignalP 4.1, and PSORTb were used for the subcellular localization and categorization of secretome on basis of signal peptides. Among the identified proteins, more than 25% of the secretome proteins were associated with virulence proteins including flagellar, adherence, and immune modulators. Gene ontology analysis using Blast2GO tools categorized 60 proteins of the secretome into biological processes, cellular components, and molecular functions. KEGG pathway analysis identified the enzymes or proteins involved in various biosynthesis and degradation pathways. Functional analysis of secretomes reveals a large number of proteins involved in the uptake and exchange of nutrients, colonization, and chemotaxis. A good number of proteins were involved in survival and defense mechanism against oxidative stress, the production of toxins and antimicrobial compounds. The present study is the first report of the in-depth protein profiling of Paenibacillus bacterium. In summary, the current findings of Paenibacillus sp. S-12 secretome provide basic information to understand its survival and the possible pathogenic mechanism.
Collapse
|
18
|
Yan J, Guo X, Li J, Li Y, Sun H, Li A, Cao B. RpoN is required for the motility and contributes to the killing ability of Plesiomonas shigelloides. BMC Microbiol 2022; 22:299. [PMID: 36510135 PMCID: PMC9743648 DOI: 10.1186/s12866-022-02722-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND RpoN, also known as σ54, first reported in Escherichia coli, is a subunit of RNA polymerase that strictly controls the expression of different genes by identifying specific promoter elements. RpoN has an important regulatory function in carbon and nitrogen metabolism and participates in the regulation of flagellar synthesis, bacterial motility and virulence. However, little is known about the effect of RpoN in Plesiomonas shigelloides. RESULTS To identify pathways controlled by RpoN, RNA sequencing (RNA-Seq) of the WT and the rpoN deletion strain was carried out for comparison. The RNA-seq results showed that RpoN regulates ~ 13.2% of the P. shigelloides transcriptome, involves amino acid transport and metabolism, glycerophospholipid metabolism, pantothenate and CoA biosynthesis, ribosome biosynthesis, flagellar assembly and bacterial secretion system. Furthermore, we verified the results of RNA-seq using quantitative real-time reverse transcription PCR, which indicated that the absence of rpoN caused downregulation of more than half of the polar and lateral flagella genes in P. shigelloides, and the ΔrpoN mutant was also non-motile and lacked flagella. In the present study, the ability of the ΔrpoN mutant to kill E. coli MG1655 was reduced by 54.6% compared with that of the WT, which was consistent with results in RNA-seq, which showed that the type II secretion system (T2SS-2) genes and the type VI secretion system (T6SS) genes were repressed. By contrast, the expression of type III secretion system genes was largely unchanged in the ΔrpoN mutant transcriptome and the ability of the ΔrpoN mutant to infect Caco-2 cells was also not significantly different compared with the WT. CONCLUSIONS We showed that RpoN is required for the motility and contributes to the killing ability of P. shigelloides and positively regulates the T6SS and T2SS-2 genes.
Collapse
Affiliation(s)
- Junxiang Yan
- grid.216938.70000 0000 9878 7032TEDA Institute of Biological Sciences and Biotechnology, Nankai University, No. 23, Hongda StreetTianjin Economic and Technological Development Area, Tianjin, 300457 China ,grid.216938.70000 0000 9878 7032Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Nankai University, No.23, Hongda StreetTianjin Economic and Technological Development Area, Tianjin, 300457 China ,grid.216938.70000 0000 9878 7032Tianjin Key Laboratory of Microbial Functional Genomics, TEDA College, Nankai University, No.23, Hongda StreetTianjin Economic and Technological Development Area, Tianjin, 300457 China
| | - Xueqian Guo
- grid.216938.70000 0000 9878 7032TEDA Institute of Biological Sciences and Biotechnology, Nankai University, No. 23, Hongda StreetTianjin Economic and Technological Development Area, Tianjin, 300457 China ,grid.216938.70000 0000 9878 7032Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Nankai University, No.23, Hongda StreetTianjin Economic and Technological Development Area, Tianjin, 300457 China ,grid.216938.70000 0000 9878 7032Tianjin Key Laboratory of Microbial Functional Genomics, TEDA College, Nankai University, No.23, Hongda StreetTianjin Economic and Technological Development Area, Tianjin, 300457 China
| | - Jinghao Li
- grid.216938.70000 0000 9878 7032TEDA Institute of Biological Sciences and Biotechnology, Nankai University, No. 23, Hongda StreetTianjin Economic and Technological Development Area, Tianjin, 300457 China ,grid.216938.70000 0000 9878 7032Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Nankai University, No.23, Hongda StreetTianjin Economic and Technological Development Area, Tianjin, 300457 China ,grid.216938.70000 0000 9878 7032Tianjin Key Laboratory of Microbial Functional Genomics, TEDA College, Nankai University, No.23, Hongda StreetTianjin Economic and Technological Development Area, Tianjin, 300457 China
| | - Yuehua Li
- grid.216938.70000 0000 9878 7032TEDA Institute of Biological Sciences and Biotechnology, Nankai University, No. 23, Hongda StreetTianjin Economic and Technological Development Area, Tianjin, 300457 China ,grid.216938.70000 0000 9878 7032Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Nankai University, No.23, Hongda StreetTianjin Economic and Technological Development Area, Tianjin, 300457 China ,grid.216938.70000 0000 9878 7032Tianjin Key Laboratory of Microbial Functional Genomics, TEDA College, Nankai University, No.23, Hongda StreetTianjin Economic and Technological Development Area, Tianjin, 300457 China
| | - Hongmin Sun
- grid.216938.70000 0000 9878 7032TEDA Institute of Biological Sciences and Biotechnology, Nankai University, No. 23, Hongda StreetTianjin Economic and Technological Development Area, Tianjin, 300457 China ,grid.216938.70000 0000 9878 7032Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Nankai University, No.23, Hongda StreetTianjin Economic and Technological Development Area, Tianjin, 300457 China ,grid.216938.70000 0000 9878 7032Tianjin Key Laboratory of Microbial Functional Genomics, TEDA College, Nankai University, No.23, Hongda StreetTianjin Economic and Technological Development Area, Tianjin, 300457 China
| | - Ang Li
- grid.216938.70000 0000 9878 7032State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353 China
| | - Boyang Cao
- grid.216938.70000 0000 9878 7032TEDA Institute of Biological Sciences and Biotechnology, Nankai University, No.23, Hongda StreetTianjin Economic and Technological Development Area, Tianjin, 300457 China ,grid.216938.70000 0000 9878 7032Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Nankai University, No. 23, Hongda StreetTianjin Economic and Technological Development Area, Tianjin, 300457 China ,grid.216938.70000 0000 9878 7032Tianjin Key Laboratory of Microbial Functional Genomics, TEDA College, Nankai University, No. 23, Hongda StreetTianjin Economic and Technological Development Area, Tianjin, 300457 China
| |
Collapse
|
19
|
Ngashangva N, Mukherjee PK, Sharma C, Kalita MC, Sarangthem I. Integrated genomics and proteomics analysis of Paenibacillus peoriae IBSD35 and insights into its antimicrobial characteristics. Sci Rep 2022; 12:18861. [PMID: 36344671 PMCID: PMC9640621 DOI: 10.1038/s41598-022-23613-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Antimicrobial resistance has been developing fast and incurring a loss of human life, and there is a need for new antimicrobial agents. Naturally occurring antimicrobial peptides offer the characteristics to counter AMR because the resistance development is low or no resistance. Antimicrobial peptides from Paenibacillus peoriae IBSD35 cell-free supernatant were salted out and purified using chromatography and characterized with liquid chromatography-tandem-mass spectrometry. The extract has shown a high and broad spectrum of antimicrobial activity. Combining the strain IBSD35 genome sequence with its proteomic data enabled the prediction of biosynthetic gene clusters by connecting the peptide from LC-MS/MS data to the gene that encode. Antimicrobial peptide databases offered a platform for the effective search, prediction, and design of AMPs and expanded the studies on their isolation, structure elucidation, biological evaluation, and pathway engineering. The genome-based taxonomy and comparisons have shown that P. peoriae IBSD35 is closely related to Paenibacillus peoriae FSL J3-0120. P. peoriae IBSD35 harbored endophytic trait genes and nonribosomal peptide synthases biosynthetic gene clusters. The comparative genomics revealed evolutionary insights and facilitated the discovery of novel SMs using proteomics from the extract of P. peoriae IBSD35. It will increase the potential to find novel bio-molecules to counter AMR.
Collapse
Affiliation(s)
- Ng Ngashangva
- grid.464584.f0000 0004 0640 0101A National Institute of Department of Biotechnology, Institute of Bioresources and Sustainable Development (IBSD), Govt. of India, Takyelpat, Imphal, Manipur 795001 India
| | - Pulok K. Mukherjee
- grid.464584.f0000 0004 0640 0101A National Institute of Department of Biotechnology, Institute of Bioresources and Sustainable Development (IBSD), Govt. of India, Takyelpat, Imphal, Manipur 795001 India
| | - Chandradev Sharma
- grid.464584.f0000 0004 0640 0101A National Institute of Department of Biotechnology, Institute of Bioresources and Sustainable Development (IBSD), Govt. of India, Takyelpat, Imphal, Manipur 795001 India
| | - Mohan C. Kalita
- grid.411779.d0000 0001 2109 4622Department of Biotechnology, Gauhati University, Jalukbari, Guwahati, Assam 781014 India
| | - Indira Sarangthem
- grid.464584.f0000 0004 0640 0101A National Institute of Department of Biotechnology, Institute of Bioresources and Sustainable Development (IBSD), Govt. of India, Takyelpat, Imphal, Manipur 795001 India
| |
Collapse
|
20
|
Characterization of ampicillin-resistant genes in Vibrio parahaemolyticus. Microb Pathog 2022; 168:105573. [PMID: 35588966 DOI: 10.1016/j.micpath.2022.105573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/28/2022] [Accepted: 05/07/2022] [Indexed: 12/29/2022]
Abstract
Vibrio parahaemolyticus is strongly resistant to ampicillin (AMP). In this study, AMP-resistant genes in V. parahaemolyticus ATCC33846 were characterized. Transcriptomic analysis of V. parahaemolyticus exposed to AMP revealed 4608 differentially transcribed genes, including 670 significantly up-regulated genes and 655 significantly down-regulated genes. Based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses, significantly modulated genes in ATCC33846 under AMP stimulation were observed in the following categories: microbial metabolism in diverse environments, metabolic pathways, bacterial secretion system, citrate cycle, biofilm formation, oxidative phosphorylation, ribosome, citrate cycle, pyruvate metabolism, carbon metabolism, nitrogen metabolism, fatty acid metabolism and tryptophan metabolism. The genes VPA0510, VPA0252, VPA0699, VPA0768, VPA0320, VP0636, VPA1096, VPA0947 and VP1775 were significantly up-regulated at the similar level to blaA in V. parahaemolyticus under AMP stimulation, and their overexpression in V. parahaemolyticus could increase its resistance to AMP. These results indicate that AMP has a global influence on V. parahaemolyticus cells. The findings would provide new insights into the resistant mechanism of V. parahaemolyticus to AMP, which would be helpful for developing novel drugs for treating V. parahaemolyticus infection.
Collapse
|
21
|
Filloux A. Bacterial protein secretion systems: Game of types. MICROBIOLOGY (READING, ENGLAND) 2022; 168. [PMID: 35536734 DOI: 10.1099/mic.0.001193] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Protein trafficking across the bacterial envelope is a process that contributes to the organisation and integrity of the cell. It is the foundation for establishing contact and exchange between the environment and the cytosol. It helps cells to communicate with one another, whether they establish symbiotic or competitive behaviours. It is instrumental for pathogenesis and for bacteria to subvert the host immune response. Understanding the formation of envelope conduits and the manifold strategies employed for moving macromolecules across these channels is a fascinating playground. The diversity of the nanomachines involved in this process logically resulted in an attempt to classify them, which is where the protein secretion system types emerged. As our knowledge grew, so did the number of types, and their rightful nomenclature started to be questioned. While this may seem a semantic or philosophical issue, it also reflects scientific rigour when it comes to assimilating findings into textbooks and science history. Here I give an overview on bacterial protein secretion systems, their history, their nomenclature and why it can be misleading for newcomers in the field. Note that I do not try to suggest a new nomenclature. Instead, I explore the reasons why naming could have escaped our control and I try to reiterate basic concepts that underlie protein trafficking cross membranes.
Collapse
Affiliation(s)
- Alain Filloux
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
22
|
Kataoka N, Saichana N, Matsutani M, Toyama H, Matsushita K, Yakushi T. Characterization of 3 phylogenetically distinct membrane-bound d-gluconate dehydrogenases of Gluconobacter spp. and their biotechnological application for efficient 2-keto-d-gluconate production. Biosci Biotechnol Biochem 2022; 86:681-690. [PMID: 35150230 DOI: 10.1093/bbb/zbac024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 02/07/2022] [Indexed: 11/13/2022]
Abstract
We identified a novel flavoprotein-cytochrome c complex d-gluconate dehydrogenase (GADH) encoded by gndXYZ of Gluconobacter oxydans NBRC 3293, which is phylogenetically distinct from previously reported GADHs encoded by gndFGH and gndSLC of Gluconobacter spp. To analyze the biochemical properties of respective GADHs, Gluconobacter japonicus NBRC 3271 mutant strain lacking membranous d-gluconate dehydrogenase activity was constructed. All GADHs (GndFGH, GndSLC, and GndXYZ) were successfully overexpressed in the constructed strain. The optimal pH and KM value at that pH of GndFGH, GndSLC, and GndXYZ were 5, 6, and 4, and 8.82 ± 1.15, 22.9 ± 5.0, and 11.3 ± 1.5 m m, respectively. When the mutants overexpressing respective GADHs were cultured in d-glucose-containing medium, all of them produced 2-keto-d-gluconate, revealing that GndXYZ converts d-gluconate to 2-keto-d-gluconate as well as other GADHs. Among the recombinants, the gndXYZ-overexpressing strain accumulated the highest level of 2-keto-d-gluconate, suggesting its potential for 2-keto-d-gluconate production.
Collapse
Affiliation(s)
- Naoya Kataoka
- Division of Agricultural Sciences, Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi, Japan.,Department of Biological Science, Faculty of Agriculture, Yamaguchi University, Yamaguchi, Japan.,Research Center for Thermotolerant Microbial Resources, Yamaguchi University, Yamaguchi, Japan
| | - Natsaran Saichana
- School of Science, Mae Fah Luang University, Chiang Rai, Thailand.,Microbial Products and Innovation Research Group, Mae Fah Luang University, Chiang Rai, Thailand
| | | | - Hirohide Toyama
- Department of Bioscience and Biotechnology, Faculty of Agriculture, University of the Ryukyus, Okinawa, Japan
| | - Kazunobu Matsushita
- Division of Agricultural Sciences, Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi, Japan.,Department of Biological Science, Faculty of Agriculture, Yamaguchi University, Yamaguchi, Japan.,Research Center for Thermotolerant Microbial Resources, Yamaguchi University, Yamaguchi, Japan
| | - Toshiharu Yakushi
- Division of Agricultural Sciences, Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi, Japan.,Department of Biological Science, Faculty of Agriculture, Yamaguchi University, Yamaguchi, Japan.,Research Center for Thermotolerant Microbial Resources, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
23
|
Li H, Wu G, Zhao L, Zhang M. Suppressed inflammation in obese children induced by a high-fiber diet is associated with the attenuation of gut microbial virulence factor genes. Virulence 2021; 12:1754-1770. [PMID: 34233588 PMCID: PMC8274444 DOI: 10.1080/21505594.2021.1948252] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 06/04/2021] [Accepted: 06/18/2021] [Indexed: 12/15/2022] Open
Abstract
In our previous study, a gut microbiota-targeted dietary intervention with a high-fiber diet improved the immune status of both genetically obese (Prader-Willi Syndrome, PWS) and simple obese (SO) children. However, PWS children had higher inflammation levels than SO children throughout the trial, the gut microbiota of the two cohorts was similar. As some virulence factors (VFs) produced by the gut microbiota play a role in triggering host inflammation, this study compared the characteristics and changes of gut microbial VF genes of the two cohorts before and after the intervention using a fecal metagenomic dataset. We found that in both cohorts, the high-fiber diet reduced the abundance of VF, and particularly pathogen-specific, genes. The composition of VF genes was also modulated, especially for offensive and defensive VF genes. Furthermore, genes belonging to invasion, T3SS (type III secretion system), and adherence classes were suppressed. Co-occurrence network analysis detected VF gene clusters closely related to host inflammation in each cohort. Though these cohort-specific clusters varied in VF gene combinations and cascade reactions affecting inflammation, they mainly contained VFs belonging to iron uptake, T3SS, and invasion classes. The PWS group had a lower abundance of VF genes before the trial, which suggested that other factors could also be responsible for the increased inflammation in this cohort. This study provides insight into the modulation of VF gene structure in the gut microbiota by a high-fiber diet, with respect to reduced inflammation in obese children, and differences in VF genes between these two cohorts.
Collapse
Affiliation(s)
- Hui Li
- State Key Laboratory of Microbial Metabolism and Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Guojun Wu
- State Key Laboratory of Microbial Metabolism and Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Liping Zhao
- State Key Laboratory of Microbial Metabolism and Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, P. R. China
- Ministry of Education Key Laboratory for Systems Biomedicine, Shanghai Centre for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Biochemistry and Microbiology and New Jersey Institute for Food, Nutrition and Health, School of Environmental and Biological Sciences, Rutgers University, NJ, USA
| | - Menghui Zhang
- State Key Laboratory of Microbial Metabolism and Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, P. R. China
| |
Collapse
|
24
|
Rauch J, Barton J, Kwiatkowski M, Wunderlich M, Steffen P, Moderzynski K, Papp S, Höhn K, Schwanke H, Witt S, Richardt U, Mehlhoop U, Schlüter H, Pianka V, Fleischer B, Tappe D, Osterloh A. GroEL is an immunodominant surface-exposed antigen of Rickettsia typhi. PLoS One 2021; 16:e0253084. [PMID: 34111210 PMCID: PMC8191997 DOI: 10.1371/journal.pone.0253084] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/28/2021] [Indexed: 11/26/2022] Open
Abstract
Rickettsioses are neglected and emerging potentially fatal febrile diseases that are caused by obligate intracellular bacteria, rickettsiae. Rickettsia (R.) typhi and R. prowazekii constitute the typhus group (TG) of rickettsiae and are the causative agents of endemic and epidemic typhus, respectively. We recently generated a monoclonal antibody (BNI52) against R. typhi. Characterization of BNI52 revealed that it specifically recognizes TG rickettsiae but not the members of the spotted fever group (SFG) rickettsiae. We further show that BNI52 binds to protein fragments of ±30 kDa that are exposed on the bacterial surface and also present in the periplasmic space. These protein fragments apparently derive from the cytosolic GroEL protein of R. typhi and are also recognized by antibodies in the sera from patients and infected mice. Furthermore, BNI52 opsonizes the bacteria for the uptake by antigen presenting cells (APC), indicating a contribution of GroEL-specific antibodies to protective immunity. Finally, it is interesting that the GroEL protein belongs to 32 proteins that are differentially downregulated by R. typhi after passage through immunodeficient BALB/c CB17 SCID mice. This could be a hint that the rickettsia GroEL protein may have immunomodulatory properties as shown for the homologous protein from several other bacteria, too. Overall, the results of this study provide evidence that GroEL represents an immunodominant antigen of TG rickettsiae that is recognized by the humoral immune response against these pathogens and that may be interesting as a vaccine candidate. Apart from that, the BNI52 antibody represents a new tool for specific detection of TG rickettsiae in various diagnostic and experimental setups.
Collapse
Affiliation(s)
- Jessica Rauch
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Jessica Barton
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | - Malte Wunderlich
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Pascal Steffen
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Stefanie Papp
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Katharina Höhn
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Hella Schwanke
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Susanne Witt
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Ulricke Richardt
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Ute Mehlhoop
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | - Verena Pianka
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | - Dennis Tappe
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Anke Osterloh
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| |
Collapse
|
25
|
Jiang C, Wynne M, Huber D. How Quality Control Systems AID Sec-Dependent Protein Translocation. Front Mol Biosci 2021; 8:669376. [PMID: 33928127 PMCID: PMC8076867 DOI: 10.3389/fmolb.2021.669376] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/17/2021] [Indexed: 02/01/2023] Open
Abstract
The evolutionarily conserved Sec machinery is responsible for transporting proteins across the cytoplasmic membrane. Protein substrates of the Sec machinery must be in an unfolded conformation in order to be translocated across (or inserted into) the cytoplasmic membrane. In bacteria, the requirement for unfolded proteins is strict: substrate proteins that fold (or misfold) prematurely in the cytoplasm prior to translocation become irreversibly trapped in the cytoplasm. Partially folded Sec substrate proteins and stalled ribosomes containing nascent Sec substrates can also inhibit translocation by blocking (i.e., “jamming”) the membrane-embedded Sec machinery. To avoid these issues, bacteria have evolved a complex network of quality control systems to ensure that Sec substrate proteins do not fold in the cytoplasm. This quality control network can be broken into three branches, for which we have defined the acronym “AID”: (i) avoidance of cytoplasmic intermediates through cotranslationally channeling newly synthesized Sec substrates to the Sec machinery; (ii) inhibition of folding Sec substrate proteins that transiently reside in the cytoplasm by molecular chaperones and the requirement for posttranslational modifications; (iii) destruction of products that could potentially inhibit translocation. In addition, several stress response pathways help to restore protein-folding homeostasis when environmental conditions that inhibit translocation overcome the AID quality control systems.
Collapse
Affiliation(s)
- Chen Jiang
- School of Biosciences and the Institute for Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Max Wynne
- School of Biosciences and the Institute for Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Damon Huber
- School of Biosciences and the Institute for Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
26
|
Weaver SJ, Ortega DR, Sazinsky MH, Dalia TN, Dalia AB, Jensen GJ. CryoEM structure of the type IVa pilus secretin required for natural competence in Vibrio cholerae. Nat Commun 2020; 11:5080. [PMID: 33033258 PMCID: PMC7545093 DOI: 10.1038/s41467-020-18866-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023] Open
Abstract
Natural transformation is the process by which bacteria take up genetic material from their environment and integrate it into their genome by homologous recombination. It represents one mode of horizontal gene transfer and contributes to the spread of traits like antibiotic resistance. In Vibrio cholerae, a type IVa pilus (T4aP) is thought to facilitate natural transformation by extending from the cell surface, binding to exogenous DNA, and retracting to thread this DNA through the outer membrane secretin, PilQ. Here, we use a functional tagged allele of VcPilQ purified from native V. cholerae cells to determine the cryoEM structure of the VcPilQ secretin in amphipol to ~2.7 Å. We use bioinformatics to examine the domain architecture and gene neighborhood of T4aP secretins in Proteobacteria in comparison with VcPilQ. This structure highlights differences in the architecture of the T4aP secretin from the type II and type III secretion system secretins. Based on our cryoEM structure, we design a series of mutants to reversibly regulate VcPilQ gate dynamics. These experiments support the idea of VcPilQ as a potential druggable target and provide insight into the channel that DNA likely traverses to promote the spread of antibiotic resistance via horizontal gene transfer by natural transformation.
Collapse
Affiliation(s)
- Sara J Weaver
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 E. California Blvd, Pasadena, CA, 91125, USA.,Howard Hughes Medical Institute, David Geffen School of Medicine, Departments of Biological Chemistry and Physiology, University of California Los Angeles, 615 Charles E Young Drive South, Los Angeles, CA, 90095, USA
| | - Davi R Ortega
- Division of Biology and Biological Engineering and Howard Hughes Medical Institute, California Institute of Technology, 1200 E. California Blvd, Pasadena, CA, 91125, USA
| | - Matthew H Sazinsky
- Department of Chemistry, Pomona College, 333N. College Way, Claremont, CA, 91711, USA
| | - Triana N Dalia
- Department of Biology, Indiana University, 107S. Indiana Avenue, Bloomington, IN, 47405, USA
| | - Ankur B Dalia
- Department of Biology, Indiana University, 107S. Indiana Avenue, Bloomington, IN, 47405, USA
| | - Grant J Jensen
- Division of Biology and Biological Engineering and Howard Hughes Medical Institute, California Institute of Technology, 1200 E. California Blvd, Pasadena, CA, 91125, USA.
| |
Collapse
|
27
|
Rivas ZP, Talbot KM, Merselis LC, McCormack RM, Adkins B, Munson GP. CexE Is a Coat Protein and Virulence Factor of Diarrheagenic Pathogens. Front Microbiol 2020; 11:1374. [PMID: 32714302 PMCID: PMC7344145 DOI: 10.3389/fmicb.2020.01374] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/28/2020] [Indexed: 12/27/2022] Open
Abstract
CexE is a 12 kDa protein that was originally reported to be present in just three strains of enterotoxigenic Escherichia coli (ETEC); a frequent cause of diarrheal illnesses worldwide. However, an examination of sequenced genomes has revealed that CexE is actually present in a majority of ETEC strains. In addition, homologs of CexE are present in enteroaggregative E. coli (EAEC), Yersinia enterocolitica, Providencia alcalifaciens, and Citrobacter rodentium. Although it has been hypothesized that CexE and its homologs are virulence factors, this has yet to be tested. Thus the primary aim of this study was to determine if these proteins contribute to pathogenicity. Our secondary aim was determine if they are secreted coat proteins. Here we report that all neonatal mice infected with a wild-type strain of C. rodentium perished. In contrast a cexE mutant was significantly attenuated with 45% neonate survival. In adult mice the wild-type strain reached significantly higher loads in the large intestines and were shed in greater numbers than cexE mutants. Secretion of the CexE homolog in EAEC is dependent upon an atypical Type I secretion system that accepts its client from the periplasm rather than the cytoplasm. Insertion mutants of cexC, the putative ATPase of the CexE secretion system, were attenuated in our murine model. In vitro we found that CexC is required for the secretion of CexE to the outer membranes of both ETEC and C. rodentium. Secretion is not constitutive because CexE accumulates in the periplasm when the two pathogens are cultured under noninducing conditions. Although secretion conditions differ between ETEC and C. rodentium, secreted CexE remains predominantly associated with the outer membranes of both species. In aggregate these findings demonstrate that CexE is a secreted coat protein and virulence factor that promotes colonization of host intestinal tissues by enteric pathogens.
Collapse
Affiliation(s)
| | | | | | | | | | - George P. Munson
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
28
|
Chen JZ, Fowler DM, Tokuriki N. Comprehensive exploration of the translocation, stability and substrate recognition requirements in VIM-2 lactamase. eLife 2020; 9:e56707. [PMID: 32510322 PMCID: PMC7308095 DOI: 10.7554/elife.56707] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/06/2020] [Indexed: 12/12/2022] Open
Abstract
Metallo-β-lactamases (MBLs) degrade a broad spectrum of β-lactam antibiotics, and are a major disseminating source for multidrug resistant bacteria. Despite many biochemical studies in diverse MBLs, molecular understanding of the roles of residues in the enzyme's stability and function, and especially substrate specificity, is lacking. Here, we employ deep mutational scanning (DMS) to generate comprehensive single amino acid variant data on a major clinical MBL, VIM-2, by measuring the effect of thousands of VIM-2 mutants on the degradation of three representative classes of β-lactams (ampicillin, cefotaxime, and meropenem) and at two different temperatures (25°C and 37°C). We revealed residues responsible for expression and translocation, and mutations that increase resistance and/or alter substrate specificity. The distribution of specificity-altering mutations unveiled distinct molecular recognition of the three substrates. Moreover, these function-altering mutations are frequently observed among naturally occurring variants, suggesting that the enzymes have continuously evolved to become more potent resistance genes.
Collapse
Affiliation(s)
- John Z Chen
- Michael Smith Laboratories, University of British ColumbiaVancouverCanada
| | - Douglas M Fowler
- Department of Genome Sciences, University of WashingtonSeattleUnited States
- Department of Bioengineering, University of WashingtonSeattleUnited States
| | - Nobuhiko Tokuriki
- Michael Smith Laboratories, University of British ColumbiaVancouverCanada
| |
Collapse
|
29
|
Honarmand Ebrahimi K. SARS-CoV-2 spike glycoprotein-binding proteins expressed by upper respiratory tract bacteria may prevent severe viral infection. FEBS Lett 2020; 594:1651-1660. [PMID: 32449939 PMCID: PMC7280584 DOI: 10.1002/1873-3468.13845] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/18/2020] [Accepted: 05/18/2020] [Indexed: 12/26/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a major global challenge. The virus infects host cells using its spike glycoprotein (S-protein) and has significantly higher infectivity and mortality rates among the aged population. Here, based on bioinformatic analysis, I provide evidence that some members of the upper respiratory tract (URT) commensal bacteria express viral S-protein -binding proteins. Based on this analysis and available data showing a decline in the population of these bacteria in the elderly, I propose that some URT commensal bacteria hamper SARS-CoV-2 infectivity and that a decline in the population of these bacteria contributes to the severity of infection. Further studies should provide a better understanding of the interaction of URT bacteria and SARS-CoV-2, which may lead to new therapeutic approaches.
Collapse
|
30
|
Acuña L, Corbalán N, Quintela-Baluja M, Barros-Velázquez J, Bellomio A. Expression of the hybrid bacteriocin Ent35-MccV in Lactococcus lactis and its use for controlling Listeria monocytogenes and Escherichia coli in milk. Int Dairy J 2020. [DOI: 10.1016/j.idairyj.2020.104650] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
31
|
Naha A, Mandal RS, Samanta P, Saha RN, Shaw S, Ghosh A, Chatterjee NS, Dutta P, Okamoto K, Dutta S, Mukhopadhyay AK. Deciphering the possible role of ctxB7 allele on higher production of cholera toxin by Haitian variant Vibrio cholerae O1. PLoS Negl Trop Dis 2020; 14:e0008128. [PMID: 32236098 PMCID: PMC7112172 DOI: 10.1371/journal.pntd.0008128] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 02/10/2020] [Indexed: 12/16/2022] Open
Abstract
Cholera continues to be an important public health concern in developing countries where proper hygiene and sanitation are compromised. This severe diarrheal disease is caused by the Gram-negative pathogen Vibrio cholerae belonging to serogroups O1 and O139. Cholera toxin (CT) is the prime virulence factor and is directly responsible for the disease manifestation. The ctxB gene encodes cholera toxin B subunit (CTB) whereas the A subunit (CTA) is the product of ctxA gene. Enzymatic action of CT depends on binding of B pentamers to the lipid-based receptor ganglioside GM1. In recent years, emergence of V. cholerae Haitian variant strains with ctxB7 allele and their rapid spread throughout the globe has been linked to various cholera outbreaks in Africa and Asia. These strains produce classical type (WT) CTB except for an additional mutation in the signal sequence region where an asparagine (N) residue replaces a histidine (H) at the 20th amino acid position (H20N) of CTB precursor (pre-CTB). Here we report that Haitian variant V. cholerae O1 strains isolated in Kolkata produced higher amount of CT compared to contemporary O1 El Tor variant strains under in vitro virulence inducing conditions. We observed that the ctxB7 allele, itself plays a pivotal role in higher CT production. Based on our in silico analysis, we hypothesized that higher accumulation of toxin subunits from ctxB7 allele might be attributed to the structural alteration at the CTB signal peptide region of pre-H20N CTB. Overall, this study provides plausible explanation regarding the hypertoxigenic phenotype of the Haitian variant strains which have spread globally, possibly through positive selection for increased pathogenic traits.
Collapse
Affiliation(s)
- Arindam Naha
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Rahul Shubhra Mandal
- Biomedical Informatics Center, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Prosenjit Samanta
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Rudra Narayan Saha
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Sreeja Shaw
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Amit Ghosh
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | | | - Pujarini Dutta
- Division of Clinical Medicine, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Keinosuke Okamoto
- Collaborative Research Center of Okayama University for Infectious Diseases at NICED, Kolkata, India
| | - Shanta Dutta
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Asish Kumar Mukhopadhyay
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
- * E-mail:
| |
Collapse
|
32
|
Sah GP, Cao P, Wall D. MYXO-CTERM sorting tag directs proteins to the cell surface via the type II secretion system. Mol Microbiol 2020; 113:1038-1051. [PMID: 31975447 DOI: 10.1111/mmi.14473] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/16/2020] [Accepted: 01/18/2020] [Indexed: 11/30/2022]
Abstract
Cells interact with their surrounding environment through surface proteins. However, knowledge gaps remain in understanding how these important types of proteins are transported and anchored on the cell surface. In the Gram-negative social bacterium, Myxococcus xanthus, a putative C-terminal sorting tag (MYXO-CTERM) is predicted to help direct 34 different proteins onto the cell surface. Here we investigate the sorting pathway for MYXO-CTERM proteins by using the TraA cell surface receptor as a paradigm. Deleting this motif from TraA abolishes the cell surface anchoring and results in extracellular secretion. Our findings indicate that conserved cysteines within the MYXO-CTERM are posttranslationally modified and are required for TraA cell surface localization and function. A region immediately upstream of these residues is predicted to be disordered and removing this motif caused a secretion defect and blocked cell surface anchoring. We further show that the type II secretion system is required for translocation across the outer membrane and that a cysteine-rich region directs TraA to the T2SS. Similar results were found with another MYXO-CTERM protein indicating our findings can be generalized. Further, we show the universal distribution of MXYO-CTERM motif across the Myxococcales order and provide a working model for sorting of these proteins.
Collapse
Affiliation(s)
- Govind Prasad Sah
- Department of Molecular Biology, University of Wyoming, Laramie, WY, USA
| | - Pengbo Cao
- Department of Molecular Biology, University of Wyoming, Laramie, WY, USA
| | - Daniel Wall
- Department of Molecular Biology, University of Wyoming, Laramie, WY, USA
| |
Collapse
|
33
|
Takahashi S, Morooka Y, Kumakura T, Abe K, Kera Y. Enzymatic characterization and regulation of gene expression of PhoK alkaline phosphatase in Sphingobium sp. strain TCM1. Appl Microbiol Biotechnol 2019; 104:1125-1134. [DOI: 10.1007/s00253-019-10291-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 11/13/2019] [Accepted: 11/28/2019] [Indexed: 11/30/2022]
|
34
|
Cranford-Smith T, Huber D. The way is the goal: how SecA transports proteins across the cytoplasmic membrane in bacteria. FEMS Microbiol Lett 2019; 365:4969678. [PMID: 29790985 PMCID: PMC5963308 DOI: 10.1093/femsle/fny093] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/10/2018] [Indexed: 02/06/2023] Open
Abstract
In bacteria, translocation of most soluble secreted proteins (and outer membrane proteins in Gram-negative bacteria) across the cytoplasmic membrane by the Sec machinery is mediated by the essential ATPase SecA. At its core, this machinery consists of SecA and the integral membrane proteins SecYEG, which form a protein conducting channel in the membrane. Proteins are recognised by the Sec machinery by virtue of an internally encoded targeting signal, which usually takes the form of an N-terminal signal sequence. In addition, substrate proteins must be maintained in an unfolded conformation in the cytoplasm, prior to translocation, in order to be competent for translocation through SecYEG. Recognition of substrate proteins occurs via SecA—either through direct recognition by SecA or through secondary recognition by a molecular chaperone that delivers proteins to SecA. Substrate proteins are then screened for the presence of a functional signal sequence by SecYEG. Proteins with functional signal sequences are translocated across the membrane in an ATP-dependent fashion. The current research investigating each of these steps is reviewed here.
Collapse
Affiliation(s)
- Tamar Cranford-Smith
- Institute for Microbiology and Infection School of Biosciences University of Birmingham Edgbaston Birmingham B15 2TT, UK
| | - Damon Huber
- Institute for Microbiology and Infection School of Biosciences University of Birmingham Edgbaston Birmingham B15 2TT, UK
| |
Collapse
|
35
|
Structural Basis for the Differential Regulatory Roles of the PDZ Domain in C-Terminal Processing Proteases. mBio 2019; 10:mBio.01129-19. [PMID: 31387902 PMCID: PMC6686036 DOI: 10.1128/mbio.01129-19] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Prc, also known previously as Tsp, is the founding member of the carboxyl-terminal processing protease (CTP) family of PDZ domain-containing proteases that include CtpA and CtpB. The substrate-binding PDZ domain is responsible for regulating the protease activity of CTP proteases; however, the regulatory role of PDZ domain is stimulatory in Prc but inhibitory in CtpA/B. By determining a series of crystal structures of Prc in the unliganded resting state, this study presents the structural basis for PDZ-dependent activation of Prc, the results of which explain the contrasting roles of the PDZ domain in the regulation of the protease activity of CTPs. Carboxyl (C)-terminal processing proteases (CTPs) participate in protective and regulatory proteolysis in bacteria. The PDZ domain is central to the activity of CTPs but plays inherently different regulatory roles. For example, the PDZ domain inhibits the activity of the signaling protease CtpB by blocking the active site but is required for the activation of Prc (or Tsp), a tail-specific protease that degrades SsrA-tagged proteins. Here, by structural and functional analyses, we show that in the unliganded resting state of Prc, the PDZ domain is docked inside the bowl-shaped scaffold without contacting the active site, which is kept in a default misaligned conformation. In Prc, a hydrophobic substrate sensor distinct from CtpB engages substrate binding to the PDZ domain and triggers a structural remodeling to align the active-site residues. Therefore, this work reveals the structural basis for understanding the contrasting roles of the PDZ domain in the regulation of CTPs.
Collapse
|
36
|
Bertucci M, Calusinska M, Goux X, Rouland-Lefèvre C, Untereiner B, Ferrer P, Gerin PA, Delfosse P. Carbohydrate Hydrolytic Potential and Redundancy of an Anaerobic Digestion Microbiome Exposed to Acidosis, as Uncovered by Metagenomics. Appl Environ Microbiol 2019; 85:e00895-19. [PMID: 31152018 PMCID: PMC6643232 DOI: 10.1128/aem.00895-19] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 05/26/2019] [Indexed: 12/22/2022] Open
Abstract
Increased hydrolysis of easily digestible biomass may lead to acidosis of anaerobic reactors and decreased methane production. Previously, it was shown that the structure of microbial communities changed during acidosis; however, once the conditions are back to optimal, biogas (initially CO2) production quickly restarts. This suggests the retention of the community functional redundancy during the process failure. In this study, with the use of metagenomics and downstream bioinformatics analyses, we characterize the carbohydrate hydrolytic potential of the microbial community, with a special focus on acidosis. To that purpose, carbohydrate-active enzymes were identified, and to further link the community hydrolytic potential with key microbes, bacterial genomes were reconstructed. In addition, we characterized biochemically the specificity and activity of selected enzymes, thus verifying the accuracy of the in silico predictions. The results confirm the retention of the community hydrolytic potential during acidosis and indicate Bacteroidetes to be largely involved in biomass degradation. Bacteroidetes showed higher diversity and genomic content of carbohydrate hydrolytic enzymes that might favor the dominance of this phylum over other bacteria in some anaerobic reactors. The combination of bioinformatic analyses and activity tests enabled us to propose a model of acetylated glucomannan degradation by BacteroidetesIMPORTANCE The enzymatic hydrolysis of lignocellulosic biomass is mainly driven by the action of carbohydrate-active enzymes. By characterizing the gene profiles at the different stages of the anaerobic digestion experiment, we showed that the microbiome retains its hydrolytic functional redundancy even during severe acidosis, despite significant changes in taxonomic composition. By analyzing reconstructed bacterial genomes, we demonstrate that Bacteroidetes hydrolytic gene diversity likely favors the abundance of this phylum in some anaerobic digestion systems. Further, we observe genetic redundancy within the Bacteroidetes group, which accounts for the preserved hydrolytic potential during acidosis. This work also uncovers new polysaccharide utilization loci involved in the deconstruction of various biomasses and proposes the model of acetylated glucomannan degradation by Bacteroidetes Acetylated glucomannan-enriched biomass is a common substrate for many industries, including pulp and paper production. Using naturally evolved cocktails of enzymes for biomass pretreatment could be an interesting alternative to the commonly used chemical pretreatments.
Collapse
Affiliation(s)
- Marie Bertucci
- Environmental Research and Innovation Department, Luxembourg Institute of Science and Technology, Belvaux, Luxembourg
- Laboratory of Bioengineering, Earth and Life Institute, Applied Microbiology, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Magdalena Calusinska
- Environmental Research and Innovation Department, Luxembourg Institute of Science and Technology, Belvaux, Luxembourg
| | - Xavier Goux
- Environmental Research and Innovation Department, Luxembourg Institute of Science and Technology, Belvaux, Luxembourg
| | - Corinne Rouland-Lefèvre
- Institute of Ecology and Environmental Sciences, Research Institute Development, Sorbonne Universités, Bondy, France
| | - Boris Untereiner
- Environmental Research and Innovation Department, Luxembourg Institute of Science and Technology, Belvaux, Luxembourg
| | - Pau Ferrer
- Environmental Research and Innovation Department, Luxembourg Institute of Science and Technology, Belvaux, Luxembourg
| | - Patrick A Gerin
- Laboratory of Bioengineering, Earth and Life Institute, Applied Microbiology, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Philippe Delfosse
- Environmental Research and Innovation Department, Luxembourg Institute of Science and Technology, Belvaux, Luxembourg
| |
Collapse
|
37
|
Kaushik H, Deshmukh SK, Solanki AK, Bhatia B, Tiwari A, Garg LC. Immunization with recombinant fusion of LTB and linear epitope (40-62) of epsilon toxin elicits protective immune response against the epsilon toxin of Clostridium perfringens type D. AMB Express 2019; 9:105. [PMID: 31300915 PMCID: PMC6626085 DOI: 10.1186/s13568-019-0824-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/26/2019] [Indexed: 11/10/2022] Open
Abstract
Epsilon toxin (Etx) produced by Clostridium perfringens types B and D, a major causative agent of enterotoxaemia causes significant economic losses to animal industry. Conventional vaccines against these pathogens generally employ formalin-inactivated culture supernatants. However, immunization with the culture supernatant and full length toxin subjects the animal to antigenic load and often have adverse effect due to incomplete inactivation of the toxins. In the present study, an epitope-based vaccine against Clostridium perfringens Etx, comprising 40-62 amino acid residues of the toxin in translational fusion with heat labile enterotoxin B subunit (LTB) of E. coli, was evaluated for its protective potential. The ability of the fusion protein rLTB.Etx40-62 to form pentamers and biologically active holotoxin with LTA of E. coli indicated that the LTB present in the fusion protein retained its biological activity. Antigenicity of both the components in the fusion protein was retained as anti-fusion protein antisera detected both the wild type Etx and LTB in Western blot analysis. Immunization of BALB/c mice with the fusion protein resulted in a significant increase in all isotypes, predominantly IgG1, IgG2a and IgG2b. Anti-fusion protein antisera neutralized the cytotoxicity of epsilon toxin both in vitro and in vivo. Thus, the results demonstrate the potential of rLTB.Etx40-62 as a candidate vaccine against C. perfringens.
Collapse
|
38
|
Denise R, Abby SS, Rocha EPC. Diversification of the type IV filament superfamily into machines for adhesion, protein secretion, DNA uptake, and motility. PLoS Biol 2019; 17:e3000390. [PMID: 31323028 PMCID: PMC6668835 DOI: 10.1371/journal.pbio.3000390] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 07/31/2019] [Accepted: 07/03/2019] [Indexed: 12/21/2022] Open
Abstract
Processes of molecular innovation require tinkering and shifting in the function of existing genes. How this occurs in terms of molecular evolution at long evolutionary scales remains poorly understood. Here, we analyse the natural history of a vast group of membrane-associated molecular systems in Bacteria and Archaea-the type IV filament (TFF) superfamily-that diversified in systems involved in flagellar or twitching motility, adhesion, protein secretion, and DNA uptake. The phylogeny of the thousands of detected systems suggests they may have been present in the last universal common ancestor. From there, two lineages-a bacterial and an archaeal-diversified by multiple gene duplications, gene fissions and deletions, and accretion of novel components. Surprisingly, we find that the 'tight adherence' (Tad) systems originated from the interkingdom transfer from Archaea to Bacteria of a system resembling the 'EppA-dependent' (Epd) pilus and were associated with the acquisition of a secretin. The phylogeny and content of ancestral systems suggest that initial bacterial pili were engaged in cell motility and/or DNA uptake. In contrast, specialised protein secretion systems arose several times independently and much later in natural history. The functional diversification of the TFF superfamily was accompanied by genetic rearrangements with implications for genetic regulation and horizontal gene transfer: systems encoded in fewer loci were more frequently exchanged between taxa. This may have contributed to their rapid evolution and spread across Bacteria and Archaea. Hence, the evolutionary history of the superfamily reveals an impressive catalogue of molecular evolution mechanisms that resulted in remarkable functional innovation and specialisation from a relatively small set of components.
Collapse
Affiliation(s)
- Rémi Denise
- Microbial Evolutionary Genomics, Institut Pasteur, CNRS, UMR3525, Paris, France
- Sorbonne Université, Collège doctoral, Paris, France
| | - Sophie S. Abby
- Université Grenoble Alpes, CNRS, Grenoble INP, TIMC-IMAG, Grenoble, France
| | - Eduardo P. C. Rocha
- Microbial Evolutionary Genomics, Institut Pasteur, CNRS, UMR3525, Paris, France
| |
Collapse
|
39
|
Keogh RA, Zapf RL, Trzeciak E, Null GG, Wiemels RE, Carroll RK. Novel Regulation of Alpha-Toxin and the Phenol-Soluble Modulins by Peptidyl-Prolyl cis/trans Isomerase Enzymes in Staphylococcus aureus. Toxins (Basel) 2019; 11:toxins11060343. [PMID: 31208155 PMCID: PMC6628628 DOI: 10.3390/toxins11060343] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/10/2019] [Accepted: 06/12/2019] [Indexed: 02/07/2023] Open
Abstract
Peptidyl-prolyl cis/trans isomerases (PPIases) are enzymes that catalyze the cis-to-trans isomerization around proline bonds, allowing proteins to fold into their correct confirmation. Previously, we identified two PPIase enzymes in Staphylococcus aureus (PpiB and PrsA) that are involved in the regulation of virulence determinants and have shown that PpiB contributes to S. aureus virulence in a murine abscess model of infection. Here, we further examine the role of these PPIases in S. aureus virulence and, in particular, their regulation of hemolytic toxins. Using murine abscess and systemic models of infection, we show that a ppiB mutant in a USA300 background is attenuated for virulence but that a prsA mutant is not. Deletion of the ppiB gene leads to decreased bacterial survival in macrophages and nasal epithelial cells, while there is no significant difference when prsA is deleted. Analysis of culture supernatants reveals that a ppiB mutant strain has reduced levels of the phenol-soluble modulins and that both ppiB and prsA mutants have reduced alpha-toxin activity. Finally, we perform immunoprecipitation to identify cellular targets of PpiB and PrsA. Results suggest a novel role for PpiB in S. aureus protein secretion. Collectively, our results demonstrate that PpiB and PrsA influence S. aureus toxins via distinct mechanisms, and that PpiB but not PrsA contributes to disease.
Collapse
Affiliation(s)
- Rebecca A Keogh
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.
| | - Rachel L Zapf
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.
| | - Emily Trzeciak
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.
| | - Gillian G Null
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.
| | - Richard E Wiemels
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.
| | - Ronan K Carroll
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.
- The Infectious and Tropical Disease Institute, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
40
|
White RC, Cianciotto NP. Assessing the impact, genomics and evolution of type II secretion across a large, medically important genus: the Legionella type II secretion paradigm. Microb Genom 2019; 5. [PMID: 31166887 PMCID: PMC6617341 DOI: 10.1099/mgen.0.000273] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The type II secretion system (T2SS) plays a major role in promoting bacterial survival in the environment and in human hosts. One of the best characterized T2SS is that of Legionella pneumophila, the agent of Legionnaires’ disease. Secreting at least 25 proteins, including degradative enzymes, eukaryotic-like proteins and novel effectors, this T2SS contributes to the ability of L. pneumophila to grow at low temperatures, infect amoebal and macrophage hosts, damage lung tissue, evade the immune system, and undergo sliding motility. The genes encoding the T2SS are conserved across the genus Legionella, which includes 62 species and >30 pathogens in addition to L. pneumophila. The vast majority of effectors associated with L. pneumophila are shared by a large number of Legionella species, hinting at a critical role for them in the ecology of Legionella as a whole. However, no other species has the same repertoire as L. pneumophila, with, as a general rule, phylogenetically more closely related species sharing similar sets of effectors. T2SS effectors that are involved in infection of a eukaryotic host(s) are more prevalent throughout Legionella, indicating that they are under stronger selective pressure. The Legionella T2SS apparatus is closest to that of Aquicella (another parasite of amoebae), and a significant number of L. pneumophila effectors have their closest homologues in Aquicella. Thus, the T2SS of L. pneumophila probably originated within the order Legionellales, with some of its effectors having arisen within that Aquicella-like progenitor, while other effectors derived from the amoebal host, mimiviruses, fungi and less closely related bacteria.
Collapse
Affiliation(s)
- Richard C White
- 1 Department of Microbiology and Immunology, Northwestern University Medical School, Chicago, IL 60611, USA
| | - Nicholas P Cianciotto
- 1 Department of Microbiology and Immunology, Northwestern University Medical School, Chicago, IL 60611, USA
| |
Collapse
|
41
|
Cholera Toxin Encapsulated within Several Vibrio cholerae O1 Serotype Inaba Outer Membrane Vesicles Lacks a Functional B-Subunit. Toxins (Basel) 2019; 11:toxins11040207. [PMID: 30959895 PMCID: PMC6521164 DOI: 10.3390/toxins11040207] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 03/29/2019] [Accepted: 04/04/2019] [Indexed: 01/01/2023] Open
Abstract
Cholera toxin (CT), the major virulence factor of Vibrio cholerae, is an AB5 toxin secreted through the type II secretion system (T2SS). Upon secretion, the toxin initiates endocytosis through the interaction of the B pentamer with the GM1 ganglioside receptor on small intestinal cells. In addition to the release of CT in the free form, the bacteria secrete CT in association with outer membrane vesicles (OMVs). Previously, we demonstrated that strain 569B releases OMVs that encapsulate CT and which interact with host cells in a GM1-independent mechanism. Here, we have demonstrated that OMV-encapsulated CT, while biologically active, does not exist in an AB5 form; rather, the OMVs encapsulate two enzymatic A-subunit (CTA) polypeptides. We further investigated the assembly and secretion of the periplasmic CT and found that a major fraction of periplasmic CTA does not participate in the CT assembly process and instead is continuously encapsulated within the OMVs. Additionally, we found that the encapsulation of CTA fragments in OMVs is conserved among several Inaba O1 strains. We further found that under conditions in which the amount of extracellularly secreted CT increases, the concentration of OMV-encapsulated likewise CTA increases. These results point to a secondary mechanism for the secretion of biologically active CT that does not depend on the CTB-GM1 interaction for endocytosis.
Collapse
|
42
|
Yang H, Wang F, Wang H, Lu X, Shen W, Chen X. Deleting mrdA and mrcB to significantly improve extracellular recombinant protein production in Escherichia coli. Biochem Eng J 2019. [DOI: 10.1016/j.bej.2019.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
43
|
Veith PD, Glew MD, Gorasia DG, Chen D, O’Brien-Simpson NM, Reynolds EC. Localization of Outer Membrane Proteins in Treponema denticola by Quantitative Proteome Analyses of Outer Membrane Vesicles and Cellular Fractions. J Proteome Res 2019; 18:1567-1581. [DOI: 10.1021/acs.jproteome.8b00860] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Paul D. Veith
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Michelle D. Glew
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Dhana G. Gorasia
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Dina Chen
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Neil M. O’Brien-Simpson
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Eric C. Reynolds
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
44
|
Heinrich J, Drewniok C, Neugebauer E, Kellner H, Wiegert T. The YoaW signal peptide directs efficient secretion of different heterologous proteins fused to a StrepII-SUMO tag in Bacillus subtilis. Microb Cell Fact 2019; 18:31. [PMID: 30732606 PMCID: PMC6366066 DOI: 10.1186/s12934-019-1078-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/29/2019] [Indexed: 01/05/2023] Open
Abstract
Background Heterologous gene expression is well established for various prokaryotic model systems. However, low yield, incorrect folding and instability still impede the production of soluble, bioactive proteins. To improve protein production with the Gram-positive host Bacillus subtilis, a secretory expression system was designed that enhances translocation, folding and stability of heterologous proteins, and simplifies purification. Based on the theta-replication plasmid pHT01, a B. subtilis secretory expression vector was constructed that encodes a fusion protein consisting of a signal peptide and a StrepII-tag linked to a SUMO-tag serving as a folding catalyst. The gene of a protein of interest can be translationally fused to the SUMO cassette and an additional 6xHis-tag encoding region. In order to maximize secretory expression of the construct by fitting the signal peptide to the StrepII-SUMO part of the fusion protein, a B. subtilis signal-peptide library was screened with the Escherichia coli alkaline phosphatase PhoA as a reporter. Results The YoaW signal peptide-encoding region (SPyoaW) was identified with highest secretory expression capacity in context with the StrepII-SUMO-tag fusion in a B. subtilis eightfold extracellular protease deletion strain. PhoA activity and fusion protein production was elevated by a factor of approximately five when compared to an α-amylase (AmyQ) signal peptide construct. Replacement of PhoA with a single-chain variable fragment antibody specific for GFP or the B. amyloliquefaciens RNase barnase, respectively, resulted in a similar enhancement of secretory expression, demonstrating universality of the YoaW signal peptide-StrepII-SUMO encoding cassette for secretory expression in B. subtilis. Optimisation of codon usage and culture conditions further increased GFP-specific scFv fusion-protein production, and a simple affinity purification strategy from culture supernatant with removal of the StrepII-SUMO-tag by SenP-processing yielded 4 mg of pure, soluble and active GFP-specific scFv from 1 l of culture under standard laboratory conditions. Conclusions The new expression system employing a YoaW signal peptide-StrepII-SUMO fusion will simplify secretory protein production and purification with B. subtilis. It can obviate the need for time consuming individual signal-peptide fitting to maximize yield for many different heterologous proteins of interest. Electronic supplementary material The online version of this article (10.1186/s12934-019-1078-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Janine Heinrich
- Department of Microbiology, Faculty of Natural and Environmental Sciences, University of Applied Sciences Zittau/Görlitz, Theodor-Körner-Allee 16, 02763, Zittau, Germany
| | - Chris Drewniok
- Department of Microbiology, Faculty of Natural and Environmental Sciences, University of Applied Sciences Zittau/Görlitz, Theodor-Körner-Allee 16, 02763, Zittau, Germany
| | - Eva Neugebauer
- EUROIMMUN AG, Im Kreppel 1, 02747, Herrnhut/Rennersdorf, Germany
| | - Harald Kellner
- Department of Bio- and Environmental Sciences, International Institute Zittau, Technical University of Dresden, Markt 23, 02763, Zittau, Germany
| | - Thomas Wiegert
- Department of Microbiology, Faculty of Natural and Environmental Sciences, University of Applied Sciences Zittau/Görlitz, Theodor-Körner-Allee 16, 02763, Zittau, Germany.
| |
Collapse
|
45
|
Alfaro-Saldaña E, Hernández-Sánchez A, Patrón-Soberano OA, Astello-García M, Méndez-Cabañas JA, García-Meza JV. Sequence analysis and confirmation of the type IV pili-associated proteins PilY1, PilW and PilV in Acidithiobacillus thiooxidans. PLoS One 2019; 14:e0199854. [PMID: 30615628 PMCID: PMC6322766 DOI: 10.1371/journal.pone.0199854] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 11/24/2018] [Indexed: 11/19/2022] Open
Abstract
Acidithiobacillus thiooxidans is an acidophilic chemolithoautotrophic bacterium widely used in the mining industry due to its metabolic sulfur-oxidizing capability. The biooxidation of sulfide minerals is enhanced through the attachment of At. thiooxidans cells to the mineral surface. The Type IV pili (TfP) of At. thiooxidans may play an important role in the bacteria attachment since TfP play a key adhesive role in the attachment and colonization of different surfaces. In this work, we report for the first time the mRNA sequence of three TfP proteins from At. thiooxidans, the adhesin protein PilY1 and the TfP pilins PilW and PilV. The nucleotide sequences of these TfP proteins show changes in some nucleotide positions with respect to the corresponding annotated sequences. The bioinformatic analyses and 3D-modeling of protein structures sustain their classification as TfP proteins, as structural homologs of the corresponding proteins of Ps. aeruginosa, results that sustain the role of PilY1, PilW and PilV in pili assembly. Also, that PilY1 comprises the conserved Neisseria-PilC (superfamily) domain of the tip-associated adhesin, while PilW of the superfamily of putative TfP assembly proteins and PilV belongs to the superfamily of TfP assembly protein. In addition, the analyses suggested the presence of specific functional domains involved in adhesion, energy transduction and signaling functions. The phylogenetic analysis indicated that the PilY1 of Acidithiobacillus genus forms a cohesive group linked with iron- and/or sulfur-oxidizing microorganisms from acid mine drainage or mine tailings.
Collapse
Affiliation(s)
- Elvia Alfaro-Saldaña
- Geomicrobiología, Metalurgia, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
- Biofísica Molecular, Instituto de Física, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - Araceli Hernández-Sánchez
- Biofísica Molecular, Instituto de Física, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - O. Araceli Patrón-Soberano
- División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica, San Luis Potosí, México
| | - Marizel Astello-García
- Geomicrobiología, Metalurgia, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - J. Alfredo Méndez-Cabañas
- Biofísica Molecular, Instituto de Física, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - J. Viridiana García-Meza
- Geomicrobiología, Metalurgia, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
- * E-mail:
| |
Collapse
|
46
|
Durgeau A, Virk Y, Gros G, Voilin E, Corgnac S, Djenidi F, Salmon J, Adam J, de Montpréville V, Validire P, Ferrone S, Chouaib S, Eggermont A, Soria JC, Lemonnier F, Tartour E, Chaput N, Besse B, Mami-Chouaib F. Human preprocalcitonin self-antigen generates TAP-dependent and -independent epitopes triggering optimised T-cell responses toward immune-escaped tumours. Nat Commun 2018; 9:5097. [PMID: 30504837 PMCID: PMC6269466 DOI: 10.1038/s41467-018-07603-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 10/30/2018] [Indexed: 12/25/2022] Open
Abstract
Tumours often evade CD8 T-cell immunity by downregulating TAP. T-cell epitopes associated with impaired peptide processing are immunogenic non-mutated neoantigens that emerge during tumour immune evasion. The preprocalcitonin (ppCT)16-25 neoepitope belongs to this category of antigens. Here we show that most human lung tumours display altered expression of TAP and frequently express ppCT self-antigen. We also show that ppCT includes HLA-A2-restricted epitopes that are processed by TAP-independent and -dependent pathways. Processing occurs in either the endoplasmic reticulum, by signal peptidase and signal peptide peptidase, or in the cytosol after release of a signal peptide precursor or retrotranslocation of a procalcitonin substrate by endoplasmic-reticulum-associated degradation. Remarkably, ppCT peptide-based immunotherapy induces efficient T-cell responses toward antigen processing and presenting machinery-impaired tumours transplanted into HLA-A*0201-transgenic mice and in NOD-scid-Il2rγnull mice adoptively transferred with human PBMC. Thus, ppCT-specific T lymphocytes are promising effectors for treatment of tumours that have escaped immune recognition.
Collapse
Affiliation(s)
- Aurélie Durgeau
- INSERM UMR 1186, Integrative Tumour Immunology and Genetic Oncology, Gustave Roussy, EPHE, PSL, Fac. de Médecine, Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France.,ElyssaMed, Paris Biotech Santé, 75014, Paris, France
| | - Yasemin Virk
- INSERM UMR 1186, Integrative Tumour Immunology and Genetic Oncology, Gustave Roussy, EPHE, PSL, Fac. de Médecine, Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France
| | - Gwendoline Gros
- INSERM UMR 1186, Integrative Tumour Immunology and Genetic Oncology, Gustave Roussy, EPHE, PSL, Fac. de Médecine, Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France
| | - Elodie Voilin
- INSERM UMR 1186, Integrative Tumour Immunology and Genetic Oncology, Gustave Roussy, EPHE, PSL, Fac. de Médecine, Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France
| | - Stéphanie Corgnac
- INSERM UMR 1186, Integrative Tumour Immunology and Genetic Oncology, Gustave Roussy, EPHE, PSL, Fac. de Médecine, Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France
| | - Fayçal Djenidi
- INSERM UMR 1186, Integrative Tumour Immunology and Genetic Oncology, Gustave Roussy, EPHE, PSL, Fac. de Médecine, Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France
| | - Jérôme Salmon
- CNRS (Centre National de la Recherche Scientifique) UMR 8122, Gustave Roussy, Faculté de Médecine, Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France
| | - Julien Adam
- INSERM U 981, Gustave Roussy, Faculté de Médecine, Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France
| | - Vincent de Montpréville
- INSERM UMR 1186, Integrative Tumour Immunology and Genetic Oncology, Gustave Roussy, EPHE, PSL, Fac. de Médecine, Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France.,Service d'Anatomie Pathologique, Centre Chirurgical Marie-Lannelongue, 92350, Le-Plessis-Robinson, France
| | - Pierre Validire
- Service d'Anatomie Pathologique, Institut Mutualiste Montsouris, 75014, Paris, France
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Salem Chouaib
- INSERM UMR 1186, Integrative Tumour Immunology and Genetic Oncology, Gustave Roussy, EPHE, PSL, Fac. de Médecine, Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France.,Thumbay Institute for Precision Medicine, Gulf Medical University, Ajman, 4184, UAE
| | - Alexander Eggermont
- Cancer Institute, Gustave Roussy Cancer Campus, Grand Paris, 94805, Villejuif, France
| | - Jean-Charles Soria
- Department of Drug Development (DITEP), Gustave Roussy, 94805, Villejuif, France
| | - François Lemonnier
- Département Endocrinologie, Métabolisme et Diabète, Equipe Immunologie des Diabètes, INSERM U1016, 75014, Paris, France
| | - Eric Tartour
- INSERM U970, Paris Cardiovascular Research Centre, Université Paris-Descartes, Sorbonne Paris Cité, Equipe Labellisée Ligue Contre le Cancer, Hôpital Européen Georges Pompidou, Service d'Immunologie Biologique, 75015, Paris, France
| | - Nathalie Chaput
- Laboratory of Immunomonitoring in Oncology, and CNRS-UMS 3655 and INSERM-US23, Gustave Roussy Cancer Campus, Villejuif, France.,Faculté de Pharmacie, University Paris-Sud, F-92296, Chatenay-Malabry, France
| | - Benjamin Besse
- Département de Médecine, Gustave Roussy, 94805, Villejuif, France
| | - Fathia Mami-Chouaib
- INSERM UMR 1186, Integrative Tumour Immunology and Genetic Oncology, Gustave Roussy, EPHE, PSL, Fac. de Médecine, Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France.
| |
Collapse
|
47
|
Padmanabhan S, Biswal MR, Manjithaya R, Prakash MK. Exploring the context of diacidic motif DE as a signal for unconventional protein secretion in eukaryotic proteins. Wellcome Open Res 2018; 3:148. [PMID: 30607372 PMCID: PMC6305234 DOI: 10.12688/wellcomeopenres.14914.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2018] [Indexed: 12/18/2022] Open
Abstract
Unconventional protein secretion (UPS) is an important phenomenon with fundamental implications to cargo export. How eukaryotic proteins transported by UPS are recognized without a conventional signal peptide has been an open question. It was recently observed that a diacidic amino acid motif (ASP-GLU or DE) is necessary for the secretion of superoxide dismutase 1 (SOD1) from yeast under nutrient starvation. Taking cue from this discovery, we explore the hypothesis of whether the diacidic motif DE, which can occur fairly ubiquitously, along with its context, can be a generic signal for unconventional secretion of proteins. Four different contexts were evaluated: a physical context encompassing the structural order and charge signature in the neighbourhood of DE, two signalling contexts reflecting the presence of either a phosphorylatable amino acid ('X' in XDE, DXE, DEX) or an LC3 interacting region (LIR) which can trigger autophagy and a co-evolutionary constraint relative to other amino acids in the protein interpreted by examining sequences across different species. Among the 100 proteins we curated from different physiological or pathological conditions, we observe a pattern in the unconventional secretion of heat shock proteins in the cancer secretome, where DE in an ordered structural region has higher odds of being a UPS signal.
Collapse
Affiliation(s)
- Sreedevi Padmanabhan
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre For Advanced Scientific Research, Bangalore, Karnataka, 560064, India
| | - Malay Ranjan Biswal
- Computational Biophysics Group, Theoretical Sciences Unit, Jawaharlal Nehru Centre For Advanced Scientific Research, Bangalore, Karnataka, 560064, India
| | - Ravi Manjithaya
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre For Advanced Scientific Research, Bangalore, Karnataka, 560064, India
| | - Meher K Prakash
- Computational Biophysics Group, Theoretical Sciences Unit, Jawaharlal Nehru Centre For Advanced Scientific Research, Bangalore, Karnataka, 560064, India
| |
Collapse
|
48
|
Ghosh R, Steiert M, Hardmeyer A, Wang YF, Rosenbusch JP. Overexpression of outer membrane porins in E. coli using pBluescript-derived vectors. Gene Expr 2018; 7:149-61. [PMID: 9840808 PMCID: PMC6151949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The genes coding for four major outer membrane porins of Escherichia coli, ompF, ompC, phoE, and lamB, have been cloned into pBluescript-derived vectors and overexpressed to very high level (approximately 80% of the total membrane protein) in widely used host strains lacking one or more porins. For OmpF, OmpC, and PhoE porins it is shown that, contrary to current dogma, the genes can be overexpressed without undue deleterious effects upon cell growth and are stable, even under conditions of continuous expression. In contrast, overexpression of LamB is toxic to cell growth, but can be performed using tightly regulated lac promotor-driven expression. The vectors described allow overexpression, sequencing, and mutagenesis to be performed using a single system, without the necessity of subcloning, thus simplifying genetic manipulation. A particular advantage of these new vectors (with the exception of the vector for LamB) is that they do not require a particular regime for inducing the recombinant protein. To our knowledge, this study is the only comparative study of widely used membrane porin expression systems and the first to show that several porins can be stably expressed individually and maintained on high copy number vectors.
Collapse
Affiliation(s)
- R Ghosh
- Department of Microbiology, Biozentrum of the University of Basel, Switzerland.
| | | | | | | | | |
Collapse
|
49
|
Rapisarda C, Tassinari M, Gubellini F, Fronzes R. Using Cryo-EM to Investigate Bacterial Secretion Systems. Annu Rev Microbiol 2018; 72:231-254. [PMID: 30004822 DOI: 10.1146/annurev-micro-090817-062702] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Bacterial secretion systems are responsible for releasing macromolecules to the extracellular milieu or directly into other cells. These membrane complexes are associated with pathogenicity and bacterial fitness. Understanding of these large assemblies has exponentially increased in the last few years thanks to electron microscopy. In fact, a revolution in this field has led to breakthroughs in characterizing the structures of secretion systems and other macromolecular machineries so as to obtain high-resolution images of complexes that could not be crystallized. In this review, we give a brief overview of structural advancements in the understanding of secretion systems, focusing in particular on cryo-electron microscopy, whether tomography or single-particle analysis. We describe how such techniques have contributed to knowledge of the mechanism of macromolecule secretion in bacteria and the impact they will have in the future.
Collapse
Affiliation(s)
- Chiara Rapisarda
- Structure et Fonction des Nanomachines Bactériennes, Institut Européen de Chimie et Biologie, 33607 Pessac, France; , .,CNRS UMR5234, Université de Bordeaux, 33076 Bordeaux, France
| | - Matteo Tassinari
- Institut Pasteur, Unité de Microbiologie Structurale, 75724 Paris, France; .,CNRS UMR3528, Institut Pasteur, 75015 Paris, France
| | - Francesca Gubellini
- Institut Pasteur, Unité de Microbiologie Structurale, 75724 Paris, France; .,CNRS UMR3528, Institut Pasteur, 75015 Paris, France
| | - Rémi Fronzes
- Structure et Fonction des Nanomachines Bactériennes, Institut Européen de Chimie et Biologie, 33607 Pessac, France; , .,CNRS UMR5234, Université de Bordeaux, 33076 Bordeaux, France
| |
Collapse
|
50
|
Veith PD, Luong C, Tan KH, Dashper SG, Reynolds EC. Outer Membrane Vesicle Proteome of Porphyromonas gingivalis Is Differentially Modulated Relative to the Outer Membrane in Response to Heme Availability. J Proteome Res 2018; 17:2377-2389. [PMID: 29766714 DOI: 10.1021/acs.jproteome.8b00153] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Porphyromonas gingivalis is an anaerobic, Gram-negative oral pathogen associated with chronic periodontitis. P. gingivalis has an obligate requirement for heme, which it obtains from the host. Heme availability has been linked to disease initiation and progression. In this study we used continuous culture of the bacterium to determine the effect of heme limitation and excess on the P. gingivalis proteome. Four biological replicates of whole cell lysate (WCL) and outer membrane vesicle (OMV) samples were digested with trypsin and analyzed by tandem mass spectrometry and MaxQuant label-free quantification. In total, 1211 proteins were quantified, with 108 and 49 proteins significantly changing in abundance more than 1.5-fold ( p < 0.05) in the WCLs and OMVs, respectively. The proteins most upregulated in response to heme limitation were those involved in binding and transporting heme, whereas the four proteins most upregulated under the heme-excess condition constitute a putative heme efflux system. In general, the protein abundance ratios obtained for OMVs and WCLs agreed, indicating that changes to the OM protein composition are passed onto OMVs; however, 16 proteins were preferentially packaged into OMVs under one condition more than the other. In particular, moonlighting cytoplasmic proteins were preferentially associated with OMVs under heme excess.
Collapse
Affiliation(s)
- Paul D Veith
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute , University of Melbourne , Victoria , Victoria 3010 , Australia
| | - Caroline Luong
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute , University of Melbourne , Victoria , Victoria 3010 , Australia
| | - Kheng H Tan
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute , University of Melbourne , Victoria , Victoria 3010 , Australia
| | - Stuart G Dashper
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute , University of Melbourne , Victoria , Victoria 3010 , Australia
| | - Eric C Reynolds
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute , University of Melbourne , Victoria , Victoria 3010 , Australia
| |
Collapse
|