1
|
Ni M, Pan Y, Gong J, Chen Z, Li D, Huang Y, Li L, Ding Y, Bi Z. Glycogen-accumulating organisms promote phosphate recovery from wastewater by pilot-scale biofilm sequencing batch reactor: Performance and mechanism. BIORESOURCE TECHNOLOGY 2025; 418:131910. [PMID: 39615760 DOI: 10.1016/j.biortech.2024.131910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/12/2024]
Abstract
A high phosphate (P) recovery concentration was achieved in pilot-scale biofilm sequencing batch reactor (BSBR) with a low carbon source (C) cost. Especially, a high-abundance glycogen-accumulating organisms (GAOs) (13.93-31.72%) was detected that was accompanied by a high P recovery concentration of BSBR. High-abundance GAOs obtain additional C through various C compensation pathways (split tricarboxylic acid cycle (TCA cycle), glyoxylate shunt and gluconeogenesis), thus reducing the need to compete with polyphosphate-accumulating organisms (PAOs) for C and weakening the adverse effects on P recovery by PAO cells. Under the action of N-acyl homoserine lactones (AHLs)-mediated quorum sensing (QS), GAOs promoted the secretion of a large amount of extracellular polymeric substances (EPS), which helped to realize the P recovery of EPS-dominated biofilms (68.02%-96.89%). This study provides a low-carbon technology for the recovery of high concentration P from municipal wastewater, and improves the ecological theory of P recovery in collaboration with GAOs and PAOs.
Collapse
Affiliation(s)
- Min Ni
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China; Jiangsu Key Laboratory of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Yang Pan
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China; National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, Suzhou University of Science and Technology, Suzhou 215009, China.
| | - Jiahui Gong
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Zhiqiang Chen
- Harbin Institute of Technology, Harbin 150006, China
| | - Dapeng Li
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China; National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Yong Huang
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China; National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Lu Li
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China; National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Yanyan Ding
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China; National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Zhen Bi
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China; National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, Suzhou University of Science and Technology, Suzhou 215009, China
| |
Collapse
|
2
|
Dong J, Chi Z, Lu S, Xie X, Gong P, Li H, Liu W. Bacterial exopolysaccharides: Characteristics and antioxidant mechanism. Int J Biol Macromol 2025; 289:138849. [PMID: 39701244 DOI: 10.1016/j.ijbiomac.2024.138849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/26/2024] [Accepted: 12/15/2024] [Indexed: 12/21/2024]
Abstract
Bacterial exopolysaccharides (EPS) are secondary metabolites of microorganisms which play important roles in adhesion, protection, biofilm formation, and as a source of nutrition. Compared with polysaccharides obtained from animal and plant species, bacterial polysaccharides have significant advantages in terms of production cost and large-scale production due to their abundant metabolic pathways and efficient polysaccharide production capacity. Most extracellular polysaccharides are water-soluble, and some are insoluble, such as bacterial cellulose. Some soluble bacterial EPS also have biological activities such as anticancer, antioxidant, antibacterial and immunomodulatory activities. These biological activities are mainly affected by the molecular weight, monosaccharide type, composition and structure of EPS. In recent years, bacterial EPS are considered as novel functional polysaccharides with important application prospects, especially in free radical scavenging and antioxidation. This review focuses on the characteristics of bacterial EPS, their ability to scavenge free radicals and their corresponding antioxidant mechanisms, and summarizes the relationship between different structures (such as monosaccharide composition, functional groups, molecular weight, etc.) and antioxidant activities. It provides a new idea for the development of more bioactive bacterial EPS antioxidants, points out a new direction for the commercial production of natural, safe and economical polysaccharide drugs and health products.
Collapse
Affiliation(s)
- Junqi Dong
- School of Marine Science and Technology, Harbin Institute of Technology at Weihai, Shandong 264209, PR China
| | - Zhenxing Chi
- School of Marine Science and Technology, Harbin Institute of Technology at Weihai, Shandong 264209, PR China
| | - Siqi Lu
- School of Marine Science and Technology, Harbin Institute of Technology at Weihai, Shandong 264209, PR China
| | - Xiaoqin Xie
- School of Marine Science and Technology, Harbin Institute of Technology at Weihai, Shandong 264209, PR China
| | - Pixian Gong
- School of Marine Science and Technology, Harbin Institute of Technology at Weihai, Shandong 264209, PR China.
| | - Huijing Li
- School of Marine Science and Technology, Harbin Institute of Technology at Weihai, Shandong 264209, PR China
| | - Wei Liu
- School of Marine Science and Technology, Harbin Institute of Technology at Weihai, Shandong 264209, PR China.
| |
Collapse
|
3
|
Elizondo-Reyna E, Martínez-Montoya H, Tamayo-Ordoñez Y, Cruz-Hernández MA, Carrillo-Tripp M, Tamayo-Ordoñez MC, Sosa-Santillán GDJ, Rodríguez-de la Garza JA, Hernández-Guzmán M, Bocanegra-García V, Acosta-Cruz E. Insights from a Genome-Wide Study of Pantoea agglomerans UADEC20: A Promising Strain for Phosphate Solubilization and Exopolysaccharides Production. Curr Issues Mol Biol 2025; 47:56. [PMID: 39852170 PMCID: PMC11763638 DOI: 10.3390/cimb47010056] [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: 12/13/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/26/2025] Open
Abstract
The genome sequence of Pantoea agglomerans UADEC20 is presented, which is a strain isolated from agricultural fields in northeast Mexico. The genome was assembled into 13 scaffolds, constituting a total chromosome size of 4.2 Mbp, with two of the scaffolds representing closed plasmids. The strain exhibits activity in phosphate solubilization and exopolysaccharide (EPS) production and secretion; therefore, we explored its biotechnological potential via its genome sequencing and annotation. Genomic analyses showed that a total of 57 and 58 coding sequences (CDSs) related to phosphate solubilization and EPS production were identified within its genome, in addition to a reduced number of CDSs related to drug resistance and phages. The comprehensive set of genes supporting phosphate solubilization, EPS synthesis, and secretion, along with its low virulence and antibiotic resistance levels, justify further research for its potential biotechnological application and possible use as a plant growth-promoting agent in the field. These findings suggest a unique genetic background in the P. agglomerans UADEC20 strain.
Collapse
Affiliation(s)
- Edith Elizondo-Reyna
- Departamento de Biotecnología, Facultad de Ciencias Químicas, Universidad Autónoma de Coahuila, Saltillo C.P. 25280, Mexico
| | - Humberto Martínez-Montoya
- Departamento de Microbiología, U.A.M. Reynosa Aztlán, Universidad Autónoma de Tamaulipas, Reynosa C.P. 88740, Mexico
| | - Yahaira Tamayo-Ordoñez
- Laboratorio Interacción Ambiente-Microorganismo, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa C.P. 88710, Mexico
| | - María Antonia Cruz-Hernández
- Laboratorio Interacción Ambiente-Microorganismo, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa C.P. 88710, Mexico
| | - Mauricio Carrillo-Tripp
- Biomolecular Diversity Laboratory, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Unidad Monterrey, Vía del Conocimiento 201, PIIT, Apodaca C.P. 66600, Mexico
| | | | | | | | - Mario Hernández-Guzmán
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada C.P. 22860, Mexico
| | - Virgilio Bocanegra-García
- Laboratorio Interacción Ambiente-Microorganismo, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa C.P. 88710, Mexico
| | - Erika Acosta-Cruz
- Departamento de Biotecnología, Facultad de Ciencias Químicas, Universidad Autónoma de Coahuila, Saltillo C.P. 25280, Mexico
| |
Collapse
|
4
|
Kumari J, Kumawat R, Prasanna R, Jothieswari D, Debnath R, Ikbal AMA, Palit P, Rawat R, Gopikrishna K, Tiwari ON. Microbial exopolysaccharides: Classification, biosynthetic pathway, industrial extraction and commercial production to unveil its bioprospection: A comprehensive review. Int J Biol Macromol 2025; 297:139917. [PMID: 39824430 DOI: 10.1016/j.ijbiomac.2025.139917] [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: 08/07/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/20/2025]
Abstract
Polysaccharides, found universally in all living-species, exhibit diverse biochemical structures and play crucial roles in microorganisms, animals, and plants to defend against pathogens, environmental stress and climate-changing. Microbial exopolysaccharides are essential for cell adhesion and stress resilience and using them has notable advantages over synthetic polysaccharides. Exopolysaccharides have versatile structures and physicochemical properties, used in food systems, therapeutics, cosmetics, agriculture, and polymer industries. Immense economic and infrastructural constraints hinder its widespread commercial use, necessitating a deeper understanding of metabolic-pathways amidst changing environmental climate that influences the biomass composition of EPS-producing wild-microbes. Green and sustainable extraction of EPS from microbes followed by commercial product development has still not been exploited comprehensively. Yield of EPS production vary from 0.1 to 3 g/g of cell weight, influenced by fermentation conditions. Economic barriers, including substrate and processing costs, limit commercial viability. Key biosynthetic pathways involve glycosyltransferases enzymes, whose regulatory network gaps and substrate specificity remain areas for optimization. Addressing these could enhance yields and lower production costs. Review illustrates various microbial-exopolysaccharides, influencing factors of production, and offer valuable insights on the bioplastic, biofuel, agri-bioproduct, and biomedicine. But their bioprospecting potential is yet to be exhaustively explored, along with their pros and cons nor documented comprehensively in scientific literature.
Collapse
Affiliation(s)
- Juhi Kumari
- Centre for Conservation and Utilization of Blue Green Algae, Division of Microbiology, ICAR-Indian Agricultural Research Institute, New Delhi 110012, India
| | - Roopam Kumawat
- Centre for Conservation and Utilization of Blue Green Algae, Division of Microbiology, ICAR-Indian Agricultural Research Institute, New Delhi 110012, India
| | - Radha Prasanna
- Centre for Conservation and Utilization of Blue Green Algae, Division of Microbiology, ICAR-Indian Agricultural Research Institute, New Delhi 110012, India
| | - D Jothieswari
- Sri Venkateswara College of Pharmacy, Chittoor 517 127, Andhra Pradesh, India
| | | | - Abu Md Ashif Ikbal
- Department of Pharmaceutical Sciences, Drug Discovery Research Laboratory, Assam University, Silchar 788011, India
| | - Partha Palit
- Department of Pharmaceutical Sciences, Drug Discovery Research Laboratory, Assam University, Silchar 788011, India
| | - Rajni Rawat
- DST, Science for Equity, Empowerment & Development (SEED) Division, New Delhi 110016, India
| | - K Gopikrishna
- DST, Science for Equity, Empowerment & Development (SEED) Division, New Delhi 110016, India
| | - Onkar Nath Tiwari
- Centre for Conservation and Utilization of Blue Green Algae, Division of Microbiology, ICAR-Indian Agricultural Research Institute, New Delhi 110012, India.
| |
Collapse
|
5
|
Chen X, Toth CRA, Guo S, Luo F, Howe J, Nesbo CL, Edwards EA. Visualization of Syntrophic Benzene-Fermenting Desulfobacterota ORM2 in a Methanogenic Enrichment Culture Using Fluorescence In Situ Hybridization. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:591-602. [PMID: 39752256 DOI: 10.1021/acs.est.4c08254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Benzene degradation under anoxic conditions was first reported more than 25 years ago; however, the activation mechanism in the absence of oxygen remains elusive. Progress has been hindered by the difficulty in cultivating anaerobic benzene-degrading enrichment cultures. Our laboratory has sustained a methanogenic enrichment culture harboring Desulfobacterota ORM2, a benzene fermenter distinct from any known genus but related to other known or predicted benzene degraders. ORM2's slow doubling time (∼30 days) and extended lag phase after inoculation complicate its study. We developed a fluorescent in situ hybridization (FISH) probe for ORM2, revealing rod-shaped cells of variable length that tend to cluster with other organisms, particularly methanogens. Microscopy and genomic evidence suggest that ORM2 may produce extracellular polymeric substances, facilitating cell aggregation and possibly consuming energy that contributes to the lag phase. Interestingly, higher benzene concentrations (90-120 mg/L) appeared to reduce cell aggregation. This study visualized the cells of Desulfobacterota ORM2 within a methanogenic community, offering insights into spatial organization and potential strategies to enhance its growth rate.
Collapse
Affiliation(s)
- Xu Chen
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Courtney R A Toth
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Shen Guo
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Fei Luo
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Jane Howe
- Department of Materials Science and Engineering, University of Toronto, 184 College Street, Toronto, Ontario M5S 3E4, Canada
| | - Camilla L Nesbo
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Elizabeth A Edwards
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| |
Collapse
|
6
|
Li Z, Wang Y, Zhao X, Meng Q, Ma G, Xie L, Jiang X, Liu Y, Huang D. Advances in bacterial glycoprotein engineering: A critical review of current technologies, emerging challenges, and future directions. Biotechnol Adv 2025; 79:108514. [PMID: 39755221 DOI: 10.1016/j.biotechadv.2024.108514] [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: 08/05/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/06/2025]
Abstract
Protein glycosylation, which involves the addition of carbohydrate chains to amino acid side chains, imparts essential properties to proteins, offering immense potential in synthetic biology applications. Despite its importance, natural glycosylation pathways present several limitations, highlighting the need for new tools to better understand glycan structures, recognition, metabolism, and biosynthesis, and to facilitate the production of biologically relevant glycoproteins. The field of bacterial glycoengineering has gained significant attention due to the ongoing discovery and study of bacterial glycosylation systems. By utilizing protein glycan coupling technology, a wide range of valuable glycoproteins for clinical and diagnostic purposes have been successfully engineered. This review outlines the recent advances in bacterial protein glycosylation from the perspective of synthetic biology and metabolic engineering, focusing on the development of new glycoprotein therapeutics and vaccines. We provide an overview of the production of high-value, customized glycoproteins using prokaryotic glycosylation platforms, with particular emphasis on four key elements: (i) glycosyltransferases, (ii) carrier proteins, (iii) glycosyl donors, and (iv) host bacteria. Optimization of these elements enables precise control over glycosylation patterns, thus enhancing the potential of the resulting products. Finally, we discuss the challenges and future prospects of leveraging synthetic biology technologies to develop microbial glyco-factories and cell-free systems for efficient glycoprotein production.
Collapse
Affiliation(s)
- Ziyu Li
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China; Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China; Nankai International Advanced Research Institute, Nankai University, Shenzhen, China
| | - Yujie Wang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China; Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China; Nankai International Advanced Research Institute, Nankai University, Shenzhen, China
| | - Xiaojing Zhao
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China; Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China; Nankai International Advanced Research Institute, Nankai University, Shenzhen, China
| | - Qing Meng
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China; Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China
| | - Guozhen Ma
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China; Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China
| | - Lijie Xie
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China; Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China
| | - Xiaolong Jiang
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China.
| | - Yutao Liu
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China; Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China.
| | - Di Huang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China; Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China; Nankai International Advanced Research Institute, Nankai University, Shenzhen, China.
| |
Collapse
|
7
|
Li FH, Liang ZH, Sun H, Tang Q, Yu HQ. Engineering Programmable Electroactive Living Materials for Highly Efficient Uranium Capture and Accumulation. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024. [PMID: 39688929 DOI: 10.1021/acs.est.4c07276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Uranium is the primary fuel for nuclear energy, critical for sustainable, carbon-neutral energy transitions. However, limited terrestrial resources and environmental risks from uranium contamination require innovative immobilization and recovery solutions. In this work, we present a novel uranium recovery method using programmable electroactive living materials (ELMs). Utilizing Shewanella oneidensis, this approach leverages the intrinsic extracellular electron transfer capability of exoelectrogenic species, combining their adaptability and programmability with the robustness of engineered multicellular systems. These exoelectrogenic cells were endowed to selectively capture and enhance U(VI) reduction by expressing uranyl-binding proteins, coupled with a reconfigured transmembrane Mtr electron nanoconduit. By incorporating biofilm-promoting circuits, we improved cell-to-cell interactions and biofilm formation, enabling the stable assembly of ELMs with robust structural integrity. The ELMs demonstrated superior electrogenic activity, achieving a 3.30-fold increase in current density and a 3.15-fold increase in voltage output compared to controls in microbial electrochemical and fuel cells. When applied for uranium recovery, the ELMs exhibited robust U(VI) capture, reduction, and accumulation capabilities, with a maximum capacity of 808.42 μmol/g. This work not only provides a versatile and environmentally friendly solution for uranium recovery, but also highlights the potential of ELMs in sustainable environmental and energy technologies.
Collapse
Affiliation(s)
- Feng-He Li
- CAS Key Laboratory of Urban Pollutant Conversion, Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei 230026, China
- School of Life Sciences, University of Science and Technology of China, Hefei 230026, China
- School of Pharmacy, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Zi-Han Liang
- CAS Key Laboratory of Urban Pollutant Conversion, Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Hong Sun
- CAS Key Laboratory of Urban Pollutant Conversion, Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Qiang Tang
- CAS Key Laboratory of Urban Pollutant Conversion, Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Han-Qing Yu
- CAS Key Laboratory of Urban Pollutant Conversion, Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
8
|
Vastel M, Pau-Roblot C, Ferré S, Tocqueville V, Ambroset C, Marois-Créhan C, Gautier-Bouchardon AV, Tardy F, Gaurivaud P. Capsular Polysaccharide Production in Bacteria of the Mycoplasma Genus: A Huge Diversity of Pathways and Synthases for So-Called Minimal Bacteria. Mol Microbiol 2024; 122:866-878. [PMID: 39473362 DOI: 10.1111/mmi.15325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/18/2024] [Accepted: 10/05/2024] [Indexed: 12/21/2024]
Abstract
Mycoplasmas are wall-less bacteria with many species spread across various animal hosts in which they can be pathogenic. Despite their reduced anabolic capacity, some mycoplasmas are known to secrete hetero- and homopolysaccharides, which play a role in host colonization through biofilm formation or immune evasion, for instance. This study explores how widespread the phenomenon of capsular homopolysaccharide secretion is within mycoplasmas, and investigates the diversity of both the molecules produced and the synthase-type glycosyltransferases responsible for their production. Fourteen strains representing 14 (sub)species from four types of hosts were tested in vitro for their polysaccharide secretion using both specific (immunodetection) and nonspecific (sugar dosage) assays. We evidenced a new, atypical homopolymer of β-(1 → 6)-glucofuranose (named glucofuranan) in the human pathogen Mycoplasma (M.) fermentans, as well as a β-(1 → 6)-glucopyranose polymer for the turkey pathogen M. iowae and galactan (β-(1 → 6)-galactofuranose) and β-(1 → 2)-glucopyranose for M. bovigenitalium infecting ruminants. Sequence and phylogenetic analyses revealed a huge diversity of synthases from varied Mycoplasma species. The clustering of these membrane-embedded glycosyltransferases into three main groups was only partially correlated to the structure of the produced homopolysaccharides.
Collapse
Affiliation(s)
- Manon Vastel
- ANSES-Laboratoire de Lyon, VetAgro Sup, UMR Mycoplasmoses Animales, Université de Lyon, Lyon, France
- ANSES-Laboratoire de Ploufragan-Plouzané-Niort, Unité Mycoplasmologie, Bactériologie et Antibiorésistance, Ploufragan, France
| | - Corinne Pau-Roblot
- UMRT INRAE 1158 BioEcoAgro - Biologie des Plantes et Innovation, Université de Picardie Jules Verne, UFR des Sciences, Amiens, France
| | - Séverine Ferré
- ANSES-Laboratoire de Ploufragan-Plouzané-Niort, Unité Mycoplasmologie, Bactériologie et Antibiorésistance, Ploufragan, France
| | - Véronique Tocqueville
- ANSES-Laboratoire de Ploufragan-Plouzané-Niort, Unité Mycoplasmologie, Bactériologie et Antibiorésistance, Ploufragan, France
| | - Chloé Ambroset
- ANSES-Laboratoire de Lyon, VetAgro Sup, UMR Mycoplasmoses Animales, Université de Lyon, Lyon, France
| | - Corinne Marois-Créhan
- ANSES-Laboratoire de Ploufragan-Plouzané-Niort, Unité Mycoplasmologie, Bactériologie et Antibiorésistance, Ploufragan, France
| | - Anne V Gautier-Bouchardon
- ANSES-Laboratoire de Ploufragan-Plouzané-Niort, Unité Mycoplasmologie, Bactériologie et Antibiorésistance, Ploufragan, France
| | - Florence Tardy
- ANSES-Laboratoire de Lyon, VetAgro Sup, UMR Mycoplasmoses Animales, Université de Lyon, Lyon, France
- ANSES-Laboratoire de Ploufragan-Plouzané-Niort, Unité Mycoplasmologie, Bactériologie et Antibiorésistance, Ploufragan, France
| | - Patrice Gaurivaud
- ANSES-Laboratoire de Lyon, VetAgro Sup, UMR Mycoplasmoses Animales, Université de Lyon, Lyon, France
| |
Collapse
|
9
|
Pandey S, Kannaujiya VK. Bacterial extracellular biopolymers: Eco-diversification, biosynthesis, technological development and commercial applications. Int J Biol Macromol 2024; 279:135261. [PMID: 39244116 DOI: 10.1016/j.ijbiomac.2024.135261] [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: 05/21/2024] [Revised: 08/22/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
Synthetic polymers have been widely thriving as mega industries at a commercial scale in various commercial sectors over the last few decades. The extensive use of synthetic polymers has caused several negative repercussions on the health of humans and the environment. Recently, biopolymers have gained more attention among scientists of different disciplines by their potential therapeutic and commercial applications. Biopolymers are chain-like repeating units of molecules isolated from green sources. They are self-degradable, biocompatible, and non-toxic in nature. Recently, eco-friendly biopolymers such as extracellular polymeric substances (EPSs) have received much attention for their wide applications in the fields of emulsification, flocculation, preservatives, wastewater treatment, nanomaterial functionalization, drug delivery, cosmetics, glycomics, medicinal chemistry, and purification technology. The dynamicity of applications has raised the industrial and consumer demands to cater to the needs of mankind. This review deals with current insights and highlights on database surveys, potential sources, classification, extremophilic EPSs, bioprospecting, patents, microenvironment stability, biosynthesis, and genetic advances for production of high valued ecofriendly polymers. The importance of high valued EPSs in commercial and industrial applications in the global market economy is also summarized. This review concludes with future perspectives and commercial applications for the well-being of humanity.
Collapse
Affiliation(s)
- Saumi Pandey
- Department of Botany, MMV, Banaras Hindu University, Varanasi 221005, India
| | - Vinod K Kannaujiya
- Department of Botany, MMV, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
10
|
Ghosh A, Sah D, Chakraborty M, Rai JPN. Mechanism and application of bacterial exopolysaccharides: An advanced approach for sustainable heavy metal abolition from soil. Carbohydr Res 2024; 544:109247. [PMID: 39180879 DOI: 10.1016/j.carres.2024.109247] [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: 05/07/2024] [Revised: 07/11/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
The escalation of heavy metal pollutants in soils and effluents, driven by industrialization and human activities, poses significant environmental and health risks. Conventional remediation methods are often costly and ineffective, prompting a shift towards sustainable alternatives such as biological treatments. Natural biosorbents, including microbial cells and their byproducts, have emerged as promising solutions. One such approach involves leveraging exopolysaccharides (EPS), complex high-molecular-weight biopolymers synthesized by microbes under environmental stress conditions. EPS are intricate organic macromolecules comprising proteins, polysaccharides, uronic acids, humic compounds, and lipids, either located within microbial cells or secreted into their surroundings. Their anionic functional groups enable efficient electrostatic binding of cationic heavy metals, making EPS effective biosorbents for soil remediation. This review thoroughly explores the pivotal role of bacterial EPS in the removal of heavy metals, focusing on EPS biosynthesis mechanisms, the dynamics of interaction with heavy metals, and case studies that illustrate their effectiveness in practical remediation strategies. By highlighting these aspects, the review underscores the innovation and practical implications of EPS-based bioremediation technologies, demonstrating their potential to address critical environmental challenges effectively while paving the way for sustainable environmental management practices. Key findings reveal that EPS exhibit robust metal-binding capacities, facilitated by their anionic functional groups, thereby offering a promising solution for mitigating metal pollution in diverse environmental matrices.
Collapse
Affiliation(s)
- Ankita Ghosh
- Department of Environmental Sciences, Govind Ballabh Pant University of Agriculture & Technology, Pantnagar, 263145, Uttarakhand, India.
| | - Diksha Sah
- Department of Environmental Sciences, Govind Ballabh Pant University of Agriculture & Technology, Pantnagar, 263145, Uttarakhand, India
| | - Moumita Chakraborty
- Department of Environmental Sciences, Govind Ballabh Pant University of Agriculture & Technology, Pantnagar, 263145, Uttarakhand, India
| | - J P N Rai
- Department of Environmental Sciences, Govind Ballabh Pant University of Agriculture & Technology, Pantnagar, 263145, Uttarakhand, India
| |
Collapse
|
11
|
Yu M, Guo W, Liang Y, Xiang H, Xia Y, Feng H. Towards rapid formation of electroactive biofilm: insights from thermodynamics and electric field manipulation. WATER RESEARCH 2024; 261:121992. [PMID: 38971076 DOI: 10.1016/j.watres.2024.121992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/08/2024]
Abstract
Electroactive biofilm (EAB) has garnered significant attention due to its effectiveness in pollutant remediation, electricity generation, and chemical synthesis. However, achieving precise control over the rapid formation of EAB presents challenges for the practical implementation of bioelectrochemical technology. In this study, we investigated the regulation of EAB formation by manipulating applied electric potential. We developed a modified XDLVO model for the applied electric field and quantitatively assessed the feasibility of existing rapid formation strategies for EAB. Our results revealed that electrostatic (EL) force significantly influenced EAB formation in the presence of the applied electric field, with the potential difference between the electrode and the microbial solution being the primary determinant of EL force. Compared to -0.2 V and 0 V vs.Ag/AgCl, EAB exhibited the highest electrochemical performance at 0.2 V vs.Ag/AgCl, with a maximum current density of 6.044 ± 0.10 A/m2, surpassing that at -0.2 V vs.Ag/AgCl and 0 V vs.Ag/AgCl by 1.73 times and 1.31 times, respectively. Furthermore, EAB demonstrated the highest biomass accumulation, measuring a thickness of 25 ± 2 μm at 0.2 V vs. Ag/AgCl, representing increases of 1.67 and 1.25 times compared to -0.2 V vs.Ag/AgCl and 0 V vs.Ag/AgCl, respectively. The strong electrostatic attraction under the anodic potential promoted the formation of a monolayer of biofilm. Additionally, the hydrophilicity and hydrophobicity of the biofilm were altered following inversion culture. The Lewis acid-base (AB) attraction offset the electrostatic repulsion caused by negative charges, it is beneficial for the formation of biofilms. This study, for the first time, elucidated the difference in the formation of cathode and anode biofilm from a thermodynamic perspective in the context of electric field introduction, laying the theoretical foundation for the directional regulation of the rapid formation of typical electroactive biofilms.
Collapse
Affiliation(s)
- Mengfan Yu
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, China
| | - Wei Guo
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, China
| | - Yuxiang Liang
- College of Environment and Resources, College of Carbon Neutral, Zhejiang A&F University, Hangzhou 311300, Zhejiang, China; Sino-Spain Joint Laboratory for Agricultural Environment Emerging Contaminants of Zhejiang Province, Zhejiang A&F University, Hangzhou 311300, Zhejiang, China
| | - Hai Xiang
- College of Environment and Resources, College of Carbon Neutral, Zhejiang A&F University, Hangzhou 311300, Zhejiang, China; Sino-Spain Joint Laboratory for Agricultural Environment Emerging Contaminants of Zhejiang Province, Zhejiang A&F University, Hangzhou 311300, Zhejiang, China
| | - Yijing Xia
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, China
| | - Huajun Feng
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, China; College of Environment and Resources, College of Carbon Neutral, Zhejiang A&F University, Hangzhou 311300, Zhejiang, China; Sino-Spain Joint Laboratory for Agricultural Environment Emerging Contaminants of Zhejiang Province, Zhejiang A&F University, Hangzhou 311300, Zhejiang, China.
| |
Collapse
|
12
|
Fernandes DC, Eto SF, Baldassi AC, Balbuena TS, Charlie-Silva I, de Andrade Belo MA, Pizauro JM. Meningitis caused by Aeromonas hydrophila in Oreochromis niloticus: Proteomics and druggability of virulence factors. FISH & SHELLFISH IMMUNOLOGY 2024; 151:109687. [PMID: 38866348 DOI: 10.1016/j.fsi.2024.109687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/30/2024] [Accepted: 06/08/2024] [Indexed: 06/14/2024]
Abstract
Meningitis caused by Gram-negative bacteria is a serious public health problem, causing morbidity and mortality in both children and adults. Here, we propose a novel experimental model using Nile tilapia (Oreochromis niloticus) to study neuroinflammation. The fish were infected with Aeromonas hydrophila, and the course of infection was monitored in the peripheral blood. Septicemia was obvious in the blood, while in the brain tissue, infection of the meninges was present. The histopathological examination showed suppurative meningitis, and the cellular immune response in the brain tissue during infection was mediated by microglia. These cells were morphologically characterized and phenotyped by MHC class II markers and CD68. The increased production of TNF-α, IL-1β and iNOS supported the infiltration of these cells during the neuroinflammatory process. In the proteomic analysis of A. hydrophila isolated from brain tissue, we found chemotactic and transport proteins, proteolytic enzymes and enzymes associated with the dismutation of nitric oxide (NO), as well as motor proteins and those responsible for cell division. After characterizing the most abundant proteins during the course of infection, we investigated the druggability index of these proteins and identified promising peptide sequences as molecular targets that are similar among bacteria. Thus, these findings deepened the understanding of the pathophysiology of meningitis caused by A. hydrophila. Moreover, through the proteomics analysis, important mechanisms and pathways used by the pathogen to subvert the host response were revealed, providing insights for the development of novel antibiotics and vaccines.
Collapse
Affiliation(s)
- Dayanne Carla Fernandes
- Institute of Chemistry, São Paulo State University (Unesp), Araraquara, Sao Paulo, SP, Brazil.
| | - Silas Fernandes Eto
- Laboratory Center of Excellence in New Target Discovery (CENTD) Special Laboratory, Butantan Institute, São Paulo, SP, Brazil
| | - Amanda Cristina Baldassi
- Department of Technology, School of Agrarian and Veterinary Sciences, São Paulo State University (Unesp), Jaboticabal, Sao Paulo, SP, Brazil
| | - Thiago Santana Balbuena
- Department of Technology, School of Agrarian and Veterinary Sciences, São Paulo State University (Unesp), Jaboticabal, Sao Paulo, SP, Brazil
| | - Ives Charlie-Silva
- Institute of Chemistry, São Paulo State University (Unesp), Araraquara, Sao Paulo, SP, Brazil
| | | | - João Martins Pizauro
- Department of Technology, School of Agrarian and Veterinary Sciences, São Paulo State University (Unesp), Jaboticabal, Sao Paulo, SP, Brazil
| |
Collapse
|
13
|
Lampugnani ER, Ford K, Ho YY, van de Meene A, Lahnstein J, Tan HT, Burton RA, Fincher GB, Shafee T, Bacic A, Zimmer J, Xing X, Bulone V, Doblin MS, Roberts EM. Glycosyl transferase GT2 genes mediate the biosynthesis of an unusual (1,3;1,4)-β-glucan exopolysaccharide in the bacterium Sarcina ventriculi. Mol Microbiol 2024; 121:1245-1261. [PMID: 38750617 DOI: 10.1111/mmi.15276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/10/2024] [Accepted: 04/23/2024] [Indexed: 06/14/2024]
Abstract
Linear, unbranched (1,3;1,4)-β-glucans (mixed-linkage glucans or MLGs) are commonly found in the cell walls of grasses, but have also been detected in basal land plants, algae, fungi and bacteria. Here we show that two family GT2 glycosyltransferases from the Gram-positive bacterium Sarcina ventriculi are capable of synthesizing MLGs. Immunotransmission electron microscopy demonstrates that MLG is secreted as an exopolysaccharide, where it may play a role in organizing individual cells into packets that are characteristic of Sarcina species. Heterologous expression of these two genes shows that they are capable of producing MLGs in planta, including an MLG that is chemically identical to the MLG secreted from S. ventriculi cells but which has regularly spaced (1,3)-β-linkages in a structure not reported previously for MLGs. The tandemly arranged, paralogous pair of genes are designated SvBmlgs1 and SvBmlgs2. The data indicate that MLG synthases have evolved different enzymic mechanisms for the incorporation of (1,3)-β- and (1,4)-β-glucosyl residues into a single polysaccharide chain. Amino acid variants associated with the evolutionary switch from (1,4)-β-glucan (cellulose) to MLG synthesis have been identified in the active site regions of the enzymes. The presence of MLG synthesis in bacteria could prove valuable for large-scale production of MLG for medical, food and beverage applications.
Collapse
Affiliation(s)
- Edwin R Lampugnani
- School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Kris Ford
- School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
- La Trobe Institute for Sustainable Agriculture and Food, La Trobe University, Bundoora, Victoria, Australia
| | - Yin Ying Ho
- School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
| | - Allison van de Meene
- School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
- Ian Holmes Imaging Centre, Bio21, The University of Melbourne, Parkville, Victoria, Australia
| | - Jelle Lahnstein
- School of Agriculture, Food and Wine, University of Adelaide, Glen Osmond, South Australia, Australia
| | - Hwei-Ting Tan
- School of Agriculture, Food and Wine, University of Adelaide, Glen Osmond, South Australia, Australia
| | - Rachel A Burton
- School of Agriculture, Food and Wine, University of Adelaide, Glen Osmond, South Australia, Australia
| | - Geoffrey B Fincher
- School of Agriculture, Food and Wine, University of Adelaide, Glen Osmond, South Australia, Australia
| | - Thomas Shafee
- La Trobe Institute for Sustainable Agriculture and Food, La Trobe University, Bundoora, Victoria, Australia
| | - Antony Bacic
- School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
- La Trobe Institute for Sustainable Agriculture and Food, La Trobe University, Bundoora, Victoria, Australia
| | - Jochen Zimmer
- Howard Hughes Medical Institute, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Xiaohui Xing
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, Royal Institute of Technology (KTH), AlbaNova University Centre, Stockholm, Sweden
| | - Vincent Bulone
- School of Agriculture, Food and Wine, University of Adelaide, Glen Osmond, South Australia, Australia
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, Royal Institute of Technology (KTH), AlbaNova University Centre, Stockholm, Sweden
| | - Monika S Doblin
- School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
- La Trobe Institute for Sustainable Agriculture and Food, La Trobe University, Bundoora, Victoria, Australia
| | - Eric M Roberts
- Department of Biology, Rhode Island College, Providence, Rhode Island, USA
| |
Collapse
|
14
|
França TC, Saïdi F, Ajamian A, Islam ST, LaPlante SR. Molecular Dynamics of Outer Membrane-Embedded Polysaccharide Secretion Porins Reveals Closed Resting-State Surface Gates Targetable by Virtual Fragment Screening for Drug Hotspot Identification. ACS OMEGA 2024; 9:13217-13226. [PMID: 38524450 PMCID: PMC10955716 DOI: 10.1021/acsomega.3c09970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/06/2024] [Accepted: 02/14/2024] [Indexed: 03/26/2024]
Abstract
Recent advances in iterative neural network analyses (e.g., AlphaFold2 and RoseTTA fold) have been revolutionary for protein 3D structure prediction, especially for difficult-to-manipulate α-helical/β-barrel integral membrane proteins. These model structures are calculated based on the coevolution of amino acids within the protein of interest and similarities to existing protein structures; the local effects of the membrane on folding and stability of the calculated model structures are not considered. We recently reported the discovery, 3D modeling, and characterization of 18-β-stranded outer-membrane (OM) WzpX, WzpS, and WzpB β-barrel secretion porins for the exopolysaccharide (EPS), major spore coat polysaccharide (MASC), and biosurfactant polysaccharide (BPS) pathways (respectively) in the Gram-negative social predatory bacterium Myxococcus xanthus DZ2. However, information was not obtained regarding the dynamic behavior of surface-gating WzpX/S/B loop domains or on potential treatments to inactivate these porins. Herein, we developed a molecular dynamics (MD) protocol to study the core stability and loop dynamism of neural network-based integral membrane protein structure models embedded in an asymmetric OM bilayer, using the M. xanthus WzpX, WzpS, and WzpB proteins as test candidates. This was accomplished through integration of the CHARMM-graphical user interface (GUI) and Molecular Operating Environment (MOE) workflows to allow for a rapid simulation system setup and facilitate data analysis. In addition to serving as a method of model structure validation, our molecular dynamics simulations revealed a minimal movement of extracellular WzpX/S/B loops in the absence of an external stimulus as well as druggable cavities between the loops. Virtual screening of a commercial fragment library against these cavities revealed putative fragment-binding hotspots on the cell-surface face of each β-barrel, along with key interacting residues, and identified promising hits for the design of potential binders capable of plugging the β-barrels and inhibiting polysaccharide secretion.
Collapse
Affiliation(s)
- Tanos
C. C. França
- Institut
National de la Recherche Scientifique (INRS), Centre Armand-Frappier
Santé Biotechnologie, Université
du Québec, Institut Pasteur International Network, Laval, QC H7V 1B7, Canada
- PROTEO,
the Quebec Network for Research on Protein Function, Engineering,
and Applications, Université Laval, Quebec, QC G1V 0A6, Canada
- Laboratory
of Molecular Modeling Applied to Chemical and Biological Defense, Military Institute of Engineering, Rio de Janeiro 22290-270, Brazil
- Department
of Chemistry, Faculty of Science, University
of Hradec Kralove, Rokitanskeho
62, 50003 Hradec
Kralove, Czech Republic
| | - Fares Saïdi
- Institut
National de la Recherche Scientifique (INRS), Centre Armand-Frappier
Santé Biotechnologie, Université
du Québec, Institut Pasteur International Network, Laval, QC H7V 1B7, Canada
- PROTEO,
the Quebec Network for Research on Protein Function, Engineering,
and Applications, Université Laval, Quebec, QC G1V 0A6, Canada
| | - Alain Ajamian
- Chemical
Computing Group, Montreal, Quebec H3A 2R7, Canada
| | - Salim T. Islam
- Institut
National de la Recherche Scientifique (INRS), Centre Armand-Frappier
Santé Biotechnologie, Université
du Québec, Institut Pasteur International Network, Laval, QC H7V 1B7, Canada
- PROTEO,
the Quebec Network for Research on Protein Function, Engineering,
and Applications, Université Laval, Quebec, QC G1V 0A6, Canada
| | - Steven R. LaPlante
- Institut
National de la Recherche Scientifique (INRS), Centre Armand-Frappier
Santé Biotechnologie, Université
du Québec, Institut Pasteur International Network, Laval, QC H7V 1B7, Canada
- PROTEO,
the Quebec Network for Research on Protein Function, Engineering,
and Applications, Université Laval, Quebec, QC G1V 0A6, Canada
| |
Collapse
|
15
|
Prinčič L, Orsi RH, Martin NH, Wiedmann M, Trmčić A. Phenotypic and genomic characterizations of Klebsiella pneumoniae ssp. pneumoniae and Rahnella inusitata strains reveal no clear association between genetic content and ropy phenotype. J Dairy Sci 2024; 107:1370-1385. [PMID: 37944807 DOI: 10.3168/jds.2023-23922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/25/2023] [Indexed: 11/12/2023]
Abstract
Ropy defect of pasteurized fluid milk is a type of spoilage which manifests itself by an increased viscosity, slimy body, and string-like flow during pouring. This defect has, among other causes, been attributed to the growth, proliferation and exopolysaccharide production by coliform bacteria, which are most commonly introduced in milk as post-pasteurization contaminants. As we identified both Klebsiella pneumoniae ssp. pneumoniae and Rahnella inusitata that were linked to a ropy defect, the goal of this study was to characterize 3 K. pneumoniae ssp. pneumoniae strains and 2 R. inusitata for (1) their ability to grow and cause ropy defect in milk at 6°C and 21°C and to (2) probe the genetic basis for observed ropy phenotype. Although all K. pneumoniae ssp. pneumoniae and R. inusitata strains showed net growth of >4 log10 over 48 h in UHT milk at 21°C, only R. inusitata strains displayed growth during 28-d incubation period at 6°C (>6 log10). Two out of 3 K. pneumoniae ssp. pneumoniae strains were capable of causing the ropy defect in milk at 21°C, as supported by an increase in the viscosity of milk and string-like flow during pouring; these 2 strains were originally isolated from raw milk. Only one R. inusitata strains was able to cause the ropy defect in milk; this strain was able to cause the defect at both 6°C and 21°C, and was originally isolated from a pasteurized milk. These findings suggest that the potential of K. pneumoniae ssp. pneumoniae and R. inusitata to cause ropy defect in milk is a strain-dependent characteristic. Comparative genomics provided no definitive answer on genetic basis for the ropy phenotype. However, for K. pneumoniae ssp. pneumoniae, genes rffG, rffH, rfbD, and rfbC involved in biosynthesis and secretion of enterobacterial common antigen (ECA) could only be found in the 2 strains that produced ropy defect, and for R. inusitata a set of 2 glycosyltransferase- and flippase genes involved in nucleotide sugar biosynthesis and export could only be identified in the ropy strain. Although these results provide some initial information for potential markers for strains that can cause ropy milk, the relationship between genetic content and ropiness in milk remains poorly understood and merits further investigation.
Collapse
Affiliation(s)
- Lucija Prinčič
- Institute of Food Science, Department of Food Science and Technology, University of Natural Resources and Life Sciences, Vienna, Austria 1190
| | - Renato H Orsi
- Department of Food Science, Cornell University, Ithaca, NY 14853
| | - Nicole H Martin
- Department of Food Science, Cornell University, Ithaca, NY 14853
| | - Martin Wiedmann
- Department of Food Science, Cornell University, Ithaca, NY 14853
| | - Aljoša Trmčić
- Department of Food Science, Cornell University, Ithaca, NY 14853.
| |
Collapse
|
16
|
Ponzio A, Rebecchi A, Zivoli R, Morelli L. Reuterin, Phenyllactic Acid, and Exopolysaccharides as Main Antifungal Molecules Produced by Lactic Acid Bacteria: A Scoping Review. Foods 2024; 13:752. [PMID: 38472865 PMCID: PMC10930965 DOI: 10.3390/foods13050752] [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: 02/02/2024] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
The primary goal of this scoping review is to collect, analyze, and critically describe information regarding the role of the main compounds (reuterin, phenyllactic acid, and exopolysaccharides) produced by LAB that possess antifungal properties and provide some suggestions for further research. The use of lactic acid bacteria (LAB) to mitigate spoilage and extend the shelf life of foodstuffs has a long history. Recently, there has been a growing interest in the unique properties of these additions to the foodstuffs in which they are applied. In recent studies regarding biopreservation, significant attention has been given to the role of these microorganisms and their metabolites. This fascinating recent discipline aims not only to replace traditional preservation systems, but also to improve the overall quality of the final product. The biologically active by-products produced by lactic acid bacteria are synthesized under certain conditions (time, temperature, aerobiosis, acidity, water activity, etc.), which can be enacted through one of the oldest approaches to food processing: fermentation (commonly used in the dairy and bakery sectors). This study also delves into the biosynthetic pathways through which they are synthesized, with a particular emphasis on what is known about the mechanisms of action against molds in relation to the type of food.
Collapse
Affiliation(s)
- Andrea Ponzio
- Department for Sustainable Food Process, Faculty of Agriculture, Food and Environmental Sciences, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy; (A.R.); (L.M.)
| | - Annalisa Rebecchi
- Department for Sustainable Food Process, Faculty of Agriculture, Food and Environmental Sciences, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy; (A.R.); (L.M.)
| | - Rosanna Zivoli
- Soremartec Italia S.r.l. (Ferrero Group), P.le P. Ferrero 1, 12051 Alba, Italy
| | - Lorenzo Morelli
- Department for Sustainable Food Process, Faculty of Agriculture, Food and Environmental Sciences, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy; (A.R.); (L.M.)
| |
Collapse
|
17
|
Eddenden A, Dooda MK, Morrison ZA, Subramanian AS, Howell PL, Troutman JM, Nitz M. Metabolic Usage and Glycan Destinations of GlcNAz in E. coli. ACS Chem Biol 2024; 19:69-80. [PMID: 38146215 PMCID: PMC11138243 DOI: 10.1021/acschembio.3c00501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Bacteria use a diverse range of carbohydrates to generate a profusion of glycans, with amino sugars, such as N-acetylglucosamine (GlcNAc), being prevalent in the cell wall and in many exopolysaccharides. The primary substrate for GlcNAc-containing glycans, UDP-GlcNAc, is the product of the bacterial hexosamine pathway and a key target for bacterial metabolic glycan engineering. Using the strategy of expressing NahK, to circumvent the hexosamine pathway, it is possible to directly feed the analogue of GlcNAc, N-azidoacetylglucosamine (GlcNAz), for metabolic labeling in Escherichia coli. The cytosolic production of UDP-GlcNAz was confirmed by using fluorescence-assisted polyacrylamide gel electrophoresis. The key question of where GlcNAz is incorporated was interrogated by analyzing potential sites including peptidoglycan (PGN), the biofilm-related exopolysaccharide poly-β-1,6-N-acetylglucosamine (PNAG), lipopolysaccharide (LPS), and the enterobacterial common antigen (ECA). The highest levels of incorporation were observed in PGN with lower levels in PNAG and no observable incorporation in LPS or ECA. The promiscuity of the PNAG synthase (PgaCD) toward UDP-GlcNAz in vitro and the lack of undecaprenyl-pyrophosphoryl-GlcNAz intermediates generated in vivo confirmed the incorporation preferences. The results of this work will guide the future development of carbohydrate-based probes and metabolic engineering strategies.
Collapse
Affiliation(s)
- Alexander Eddenden
- Department of Chemistry, University of Toronto, Toronto, Ontario, M5S 3H6, Canada
| | - Manoj K. Dooda
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, North Carolina, 28223-0001, United States
| | - Zachary A. Morrison
- Department of Chemistry, University of Toronto, Toronto, Ontario, M5S 3H6, Canada
| | - Adithya Shankara Subramanian
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, M5G 0A4, Canada
| | - P. Lynne Howell
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, M5G 0A4, Canada
| | - Jerry M. Troutman
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, North Carolina, 28223-0001, United States
| | - Mark Nitz
- Department of Chemistry, University of Toronto, Toronto, Ontario, M5S 3H6, Canada
| |
Collapse
|
18
|
Ramamoorthy S, Pena M, Ghosh P, Liao YY, Paret M, Jones JB, Potnis N. Transcriptome profiling of type VI secretion system core gene tssM mutant of Xanthomonas perforans highlights regulators controlling diverse functions ranging from virulence to metabolism. Microbiol Spectr 2024; 12:e0285223. [PMID: 38018859 PMCID: PMC10782981 DOI: 10.1128/spectrum.02852-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/20/2023] [Indexed: 11/30/2023] Open
Abstract
IMPORTANCE T6SS has received attention due to its significance in mediating interorganismal competition through contact-dependent release of effector molecules into prokaryotic and eukaryotic cells. Reverse-genetic studies have indicated the role of T6SS in virulence in a variety of plant pathogenic bacteria, including the one studied here, Xanthomonas. However, it is not clear whether such effect on virulence is merely due to a shift in the microbiome-mediated protection or if T6SS is involved in a complex virulence regulatory network. In this study, we conducted in vitro transcriptome profiling in minimal medium to decipher the signaling pathways regulated by tssM-i3* in X. perforans AL65. We show that TssM-i3* regulates the expression of a suite of genes associated with virulence and metabolism either directly or indirectly by altering the transcription of several regulators. These findings further expand our knowledge on the intricate molecular circuits regulated by T6SS in phytopathogenic bacteria.
Collapse
Affiliation(s)
- Sivakumar Ramamoorthy
- Department of Entomology and Plant Pathology, Auburn University, Auburn, Alabama, USA
| | - Michelle Pena
- Department of Entomology and Plant Pathology, Auburn University, Auburn, Alabama, USA
| | - Palash Ghosh
- Department of Entomology and Plant Pathology, Auburn University, Auburn, Alabama, USA
| | - Ying-Yu Liao
- Department of Plant Pathology, University of Florida, Gainesville, Florida, USA
| | - Mathews Paret
- Department of Plant Pathology, University of Florida, Gainesville, Florida, USA
| | - Jeffrey B. Jones
- Department of Plant Pathology, University of Florida, Gainesville, Florida, USA
| | - Neha Potnis
- Department of Entomology and Plant Pathology, Auburn University, Auburn, Alabama, USA
| |
Collapse
|
19
|
Zhao Y, Chen Z, Cai Y, Xue J, Zhang L, Wang L, Zhao M, Zheng Y, Xia T, Yu H, Jiang T, Sun Y. Aloe-emodin destroys the biofilm of Helicobacter pylori by targeting the outer membrane protein 6. Microbiol Res 2024; 278:127539. [PMID: 37956613 DOI: 10.1016/j.micres.2023.127539] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023]
Abstract
Biofilm formation is one of the most important factors causing drug resistance of Helicobacter pylori. Therefore, it is necessary to explore the mechanism underlying the biofilm formation and its eradication methods. The outer membrane proteins (OMPs) play important roles in the formation of bacterial biofilms and are considered the essential targets for new drug discovery. Natural products play significant roles in anti-bacterial and anti-biofilm functions. This study explored the key OMPs involved in the biofilm formation of H. pylori and the natural products that target these OMPs. Transcriptome sequencing, gene knockout, and electrophoretic mobility shift assay (EMSA) were performed to reveal that OMP6 was involved in the biofilm formation of H. pylori, which was regulated by non-phosphorylated ArsR. Molecular docking suggested that aloe-emodin (AE) could target OMP6 and destroy the biofilms of H. pylori. Further exploration of its mechanism found that AE could also inhibit the expression of omp6 mRNA by binding to its regulator ArsR. In summary, we have discovered a novel molecular mechanism regulating the biofilm formation of H. pylori and identified a natural product against H. pylori biofilms, providing potential clues for clinical treatment of H. pylori.
Collapse
Affiliation(s)
- Yican Zhao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Microbiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhenghong Chen
- Department of Microbiology, Key Laboratory of Medical Microbiology and Parasitology, Guizhou Medical University, Guiyang, China
| | - Yuying Cai
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Microbiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Junyuan Xue
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Microbiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lu Zhang
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Microbiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Liyuan Wang
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Microbiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Mingzhong Zhao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Microbiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yantong Zheng
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Microbiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Tian Xia
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Microbiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Han Yu
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Microbiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ting Jiang
- Jiangsu Luye Diagnostic Technology, Wuxi, China
| | - Yundong Sun
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Microbiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
20
|
Gheorghita AA, Wozniak DJ, Parsek MR, Howell PL. Pseudomonas aeruginosa biofilm exopolysaccharides: assembly, function, and degradation. FEMS Microbiol Rev 2023; 47:fuad060. [PMID: 37884397 PMCID: PMC10644985 DOI: 10.1093/femsre/fuad060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 10/04/2023] [Accepted: 10/25/2023] [Indexed: 10/28/2023] Open
Abstract
The biofilm matrix is a fortress; sheltering bacteria in a protective and nourishing barrier that allows for growth and adaptation to various surroundings. A variety of different components are found within the matrix including water, lipids, proteins, extracellular DNA, RNA, membrane vesicles, phages, and exopolysaccharides. As part of its biofilm matrix, Pseudomonas aeruginosa is genetically capable of producing three chemically distinct exopolysaccharides - alginate, Pel, and Psl - each of which has a distinct role in biofilm formation and immune evasion during infection. The polymers are produced by highly conserved mechanisms of secretion, involving many proteins that span both the inner and outer bacterial membranes. Experimentally determined structures, predictive modelling of proteins whose structures are yet to be solved, and structural homology comparisons give us insight into the molecular mechanisms of these secretion systems, from polymer synthesis to modification and export. Here, we review recent advances that enhance our understanding of P. aeruginosa multiprotein exopolysaccharide biosynthetic complexes, and how the glycoside hydrolases/lyases within these systems have been commandeered for antimicrobial applications.
Collapse
Affiliation(s)
- Andreea A Gheorghita
- Program in Molecular Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay St, Toronto, ON M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Medical Science Building, 1 King's College Cir, Toronto, ON M5S 1A8, Canada
| | - Daniel J Wozniak
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, 776 Biomedical Research Tower, 460 W 12th Ave, Columbus, OH 43210, United States
- Department of Microbiology, The Ohio State University College, Biological Sciences Bldg, 105, 484 W 12th Ave, Columbus, OH 43210, United States
| | - Matthew R Parsek
- Department of Microbiology, University of Washington, Health Sciences Bldg, 1705 NE Pacific St, Seattle, WA 98195-7735, United States
| | - P Lynne Howell
- Program in Molecular Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay St, Toronto, ON M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Medical Science Building, 1 King's College Cir, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
21
|
Jaramillo-Rodríguez JB, Vega-Alvarado L, Rodríguez-Torres LM, Huerta-Miranda GA, Hernández-Eligio A, Juarez K. Global transcriptional analysis of Geobacter sulfurreducens gsu1771 mutant biofilm grown on two different support structures. PLoS One 2023; 18:e0293359. [PMID: 37878651 PMCID: PMC10599522 DOI: 10.1371/journal.pone.0293359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 09/28/2023] [Indexed: 10/27/2023] Open
Abstract
Electroactive biofilms formation by the metal-reducing bacterium Geobacter sulfurreducens is a step crucial for bioelectricity generation and bioremediation. The transcriptional regulator GSU1771 controls the expression of essential genes involved in electron transfer and biofilm formation in G. sulfurreducens, with GSU1771-deficient producing thicker and more electroactive biofilms. Here, RNA-seq analyses were conducted to compare the global gene expression patterns of wild-type and Δgsu1771 mutant biofilms grown on non-conductive (glass) and conductive (graphite electrode) materials. The Δgsu1771 biofilm grown on the glass surface exhibited 467 differentially expressed (DE) genes (167 upregulated and 300 downregulated) versus the wild-type biofilm. In contrast, the Δgsu1771 biofilm grown on the graphite electrode exhibited 119 DE genes (79 upregulated and 40 downregulated) versus the wild-type biofilm. Among these DE genes, 67 were also differentially expressed in the Δgsu1771 biofilm grown on glass (56 with the same regulation and 11 exhibiting counter-regulation). Among the upregulated genes in the Δgsu1771 biofilms, we identified potential target genes involved in exopolysaccharide synthesis (gsu1961-63, gsu1959, gsu1972-73, gsu1976-77). RT-qPCR analyses were then conducted to confirm the differential expression of a selection of genes of interest. DNA-protein binding assays demonstrated the direct binding of the GSU1771 regulator to the promoter region of pgcA, pulF, relA, and gsu3356. Furthermore, heme-staining and western blotting revealed an increase in c-type cytochromes including OmcS and OmcZ in Δgsu1771 biofilms. Collectively, our findings demonstrated that GSU1771 is a global regulator that controls extracellular electron transfer and exopolysaccharide synthesis in G. sulfurreducens, which is crucial for electroconductive biofilm development.
Collapse
Affiliation(s)
- Juan B. Jaramillo-Rodríguez
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Leticia Vega-Alvarado
- Instituto de Ciencias Aplicadas y Tecnología, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, México
| | - Luis M. Rodríguez-Torres
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Guillermo A. Huerta-Miranda
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Alberto Hernández-Eligio
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
- Investigador por México, Consejo Nacional de Ciencia y Tecnología, Ciudad de México, México
| | - Katy Juarez
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| |
Collapse
|
22
|
Wei X, Chen Z, Liu A, Yang L, Xu Y, Cao M, He N. Advanced strategies for metabolic engineering of Bacillus to produce extracellular polymeric substances. Biotechnol Adv 2023; 67:108199. [PMID: 37330153 DOI: 10.1016/j.biotechadv.2023.108199] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/24/2023] [Accepted: 06/11/2023] [Indexed: 06/19/2023]
Abstract
Extracellular polymeric substances are mainly synthesized via a variety of biosynthetic pathways in bacteria. Bacilli-sourced extracellular polymeric substances, such as exopolysaccharides (EPS) and poly-γ-glutamic acid (γ-PGA), can serve as active ingredients and hydrogels, and have other important industrial applications. However, the functional diversity and widespread applications of these extracellular polymeric substances, are hampered by their low yields and high costs. Biosynthesis of extracellular polymeric substances is very complex in Bacillus, and there is no detailed elucidation of the reactions and regulations among various metabolic pathways. Therefore, a better understanding of the metabolic mechanisms is required to broaden the functions and increase the yield of extracellular polymeric substances. This review systematically summarizes the biosynthesis and metabolic mechanisms of extracellular polymeric substances in Bacillus, providing an in-depth understanding of the relationships between EPS and γ-PGA synthesis. This review provides a better clarification of Bacillus metabolic mechanisms during extracellular polymeric substance secretion and thus benefits their application and commercialization.
Collapse
Affiliation(s)
- Xiaoyu Wei
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; The Key Lab for Synthetic Biotechnology of Xiamen City, Xiamen University, Xiamen 361005, China
| | - Zhen Chen
- College of Life Science, Xinyang Normal University, Xinyang 464000, China.
| | - Ailing Liu
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; The Key Lab for Synthetic Biotechnology of Xiamen City, Xiamen University, Xiamen 361005, China
| | - Lijie Yang
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; The Key Lab for Synthetic Biotechnology of Xiamen City, Xiamen University, Xiamen 361005, China
| | - Yiyuan Xu
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; The Key Lab for Synthetic Biotechnology of Xiamen City, Xiamen University, Xiamen 361005, China
| | - Mingfeng Cao
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; The Key Lab for Synthetic Biotechnology of Xiamen City, Xiamen University, Xiamen 361005, China; Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen 361005, China.
| | - Ning He
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; The Key Lab for Synthetic Biotechnology of Xiamen City, Xiamen University, Xiamen 361005, China.
| |
Collapse
|
23
|
Cui Y, Dong S, Qu X. New progress in the identifying regulatory factors of exopolysaccharide synthesis in lactic acid bacteria. World J Microbiol Biotechnol 2023; 39:301. [PMID: 37688654 DOI: 10.1007/s11274-023-03756-4] [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: 08/17/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
The exopolysaccharides (EPSs) of lactic acid bacteria (LAB) have presented various bioactivities and beneficial characteristics, rendering their vast commercial value and attracting a broad interest of researchers. The diversity of EPS structures contributes to the changes of EPS functions. However, the low yield of EPS of LAB has severely limited these biopolymers' comprehensive studies and applications in different areas, such as functional food, health and medicine fields. The clarification of biosynthesis mechanism of EPS will accelerate the synthesis and reconstruction of EPS. In recent years, with the development of new genetic manipulation techniques, there has been significant progress in the EPS biosynthesis mechanisms in LAB. In this review, the structure of LAB-derived EPSs, the EPS biosynthesis basic pathways in LAB, the EPS biosynthetic gene cluster, and the regulation mechanism of EPS biosynthesis will be summarized. It will focus on the latest progress in EPS biosynthesis regulation of LAB and provide prospects for future related developments.
Collapse
Affiliation(s)
- Yanhua Cui
- Department of Food Nutrition and Health, School of Medicine and Health, Harbin Institute of Technology, Harbin, 150001, China.
| | - Shiyuan Dong
- Department of Food Nutrition and Health, School of Medicine and Health, Harbin Institute of Technology, Harbin, 150001, China
| | - Xiaojun Qu
- Institute of Microbiology, Heilongjiang Academy of Sciences, Harbin, 150010, China
| |
Collapse
|
24
|
Murali R, Yu H, Speth DR, Wu F, Metcalfe KS, Crémière A, Laso-Pèrez R, Malmstrom RR, Goudeau D, Woyke T, Hatzenpichler R, Chadwick GL, Connon SA, Orphan VJ. Physiological potential and evolutionary trajectories of syntrophic sulfate-reducing bacterial partners of anaerobic methanotrophic archaea. PLoS Biol 2023; 21:e3002292. [PMID: 37747940 PMCID: PMC10553843 DOI: 10.1371/journal.pbio.3002292] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 10/05/2023] [Accepted: 08/08/2023] [Indexed: 09/27/2023] Open
Abstract
Sulfate-coupled anaerobic oxidation of methane (AOM) is performed by multicellular consortia of anaerobic methanotrophic archaea (ANME) in obligate syntrophic partnership with sulfate-reducing bacteria (SRB). Diverse ANME and SRB clades co-associate but the physiological basis for their adaptation and diversification is not well understood. In this work, we used comparative metagenomics and phylogenetics to investigate the metabolic adaptation among the 4 main syntrophic SRB clades (HotSeep-1, Seep-SRB2, Seep-SRB1a, and Seep-SRB1g) and identified features associated with their syntrophic lifestyle that distinguish them from their non-syntrophic evolutionary neighbors in the phylum Desulfobacterota. We show that the protein complexes involved in direct interspecies electron transfer (DIET) from ANME to the SRB outer membrane are conserved between the syntrophic lineages. In contrast, the proteins involved in electron transfer within the SRB inner membrane differ between clades, indicative of convergent evolution in the adaptation to a syntrophic lifestyle. Our analysis suggests that in most cases, this adaptation likely occurred after the acquisition of the DIET complexes in an ancestral clade and involve horizontal gene transfers within pathways for electron transfer (CbcBA) and biofilm formation (Pel). We also provide evidence for unique adaptations within syntrophic SRB clades, which vary depending on the archaeal partner. Among the most widespread syntrophic SRB, Seep-SRB1a, subclades that specifically partner ANME-2a are missing the cobalamin synthesis pathway, suggestive of nutritional dependency on its partner, while closely related Seep-SRB1a partners of ANME-2c lack nutritional auxotrophies. Our work provides insight into the features associated with DIET-based syntrophy and the adaptation of SRB towards it.
Collapse
Affiliation(s)
- Ranjani Murali
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Hang Yu
- Division of Geological and Planetary Sciences, California Institute of Technology, Pasadena, California, Unites Stated of America
- Department of Physics and Astronomy, University of Southern California, Los Angeles, California, United States of America
| | - Daan R. Speth
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
- Division of Microbial Ecology, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - Fabai Wu
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
| | - Kyle S. Metcalfe
- Department of Plant and Molecular Biology, University of California, Berkeley. Berkeley, California, United States of America
| | - Antoine Crémière
- Division of Geological and Planetary Sciences, California Institute of Technology, Pasadena, California, Unites Stated of America
| | - Rafael Laso-Pèrez
- Systems Biology Department, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Rex R. Malmstrom
- DOE Joint Genome Institute, Department of Energy, Berkeley, California, United States of America
| | - Danielle Goudeau
- DOE Joint Genome Institute, Department of Energy, Berkeley, California, United States of America
| | - Tanja Woyke
- DOE Joint Genome Institute, Department of Energy, Berkeley, California, United States of America
| | - Roland Hatzenpichler
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, United States of America
| | - Grayson L. Chadwick
- Division of Geological and Planetary Sciences, California Institute of Technology, Pasadena, California, Unites Stated of America
- Department of Plant and Molecular Biology, University of California, Berkeley. Berkeley, California, United States of America
| | - Stephanie A. Connon
- Division of Geological and Planetary Sciences, California Institute of Technology, Pasadena, California, Unites Stated of America
| | - Victoria J. Orphan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
- Division of Geological and Planetary Sciences, California Institute of Technology, Pasadena, California, Unites Stated of America
| |
Collapse
|
25
|
Eddenden A, Dooda MK, Morrison ZA, Subramanian AS, Howell PL, Troutman JM, Nitz M. The Metabolic Usage and Glycan Destinations of GlcNAz in E. coli. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.17.553294. [PMID: 37645909 PMCID: PMC10462111 DOI: 10.1101/2023.08.17.553294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Bacteria use a diverse range of carbohydrates to generate a profusion of glycans, with amino sugars such as N-acetylglucosamine (GlcNAc) being prevalent in the cell wall and in many exopolysaccharides. The primary substrate for GlcNAc-containing glycans, UDP-GlcNAc, is the product of the bacterial hexosamine pathway, and a key target for bacterial metabolic glycan engineering. Using the strategy of expressing NahK, to circumvent the hexosamine pathway, it is possible to directly feed the analogue of GlcNAc, N-azidoacetylglucosamine (GlcNAz), for metabolic labelling in E. coli. The cytosolic production of UDP-GlcNAz was confirmed using fluorescence assisted polyacrylamide gel electrophoresis. The key question of where GlcNAz is incorporated, was interrogated by analyzing potential sites including: peptidoglycan (PGN), the biofilm-related exopolysaccharide poly-β-1,6-N-acetylglucosamine (PNAG), lipopolysaccharide (LPS) and the enterobacterial common antigen (ECA). The highest levels of incorporation were observed in PGN with lower levels in PNAG and no observable incorporation in LPS or ECA. The promiscuity of the PNAG synthase (PgaCD) towards UDP-GlcNAz in vitro and lack of undecaprenyl-pyrophosphoryl-GlcNAz intermediates generated in vivo confirmed the incorporation preferences. The results of this work will guide the future development of carbohydrate-based probes and metabolic engineering strategies.
Collapse
Affiliation(s)
- Alexander Eddenden
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Manoj K Dooda
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, North Carolina, United States
| | - Zachary A Morrison
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Adithya Shankara Subramanian
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - P Lynne Howell
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Jerry M Troutman
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, North Carolina, United States
| | - Mark Nitz
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
26
|
Chang SC, Kao MR, Saldivar RK, Díaz-Moreno SM, Xing X, Furlanetto V, Yayo J, Divne C, Vilaplana F, Abbott DW, Hsieh YSY. The Gram-positive bacterium Romboutsia ilealis harbors a polysaccharide synthase that can produce (1,3;1,4)-β-D-glucans. Nat Commun 2023; 14:4526. [PMID: 37500617 PMCID: PMC10374906 DOI: 10.1038/s41467-023-40214-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/16/2023] [Indexed: 07/29/2023] Open
Abstract
(1,3;1,4)-β-D-Glucans are widely distributed in the cell walls of grasses (family Poaceae) and closely related families, as well as some other vascular plants. Additionally, they have been found in other organisms, including fungi, lichens, brown algae, charophycean green algae, and the bacterium Sinorhizobium meliloti. Only three members of the Cellulose Synthase-Like (CSL) genes in the families CSLF, CSLH, and CSLJ are implicated in (1,3;1,4)-β-D-glucan biosynthesis in grasses. Little is known about the enzymes responsible for synthesizing (1,3;1,4)-β-D-glucans outside the grasses. In the present study, we report the presence of (1,3;1,4)-β-D-glucans in the exopolysaccharides of the Gram-positive bacterium Romboutsia ilealis CRIBT. We also report that RiGT2 is the candidate gene of R. ilealis that encodes (1,3;1,4)-β-D-glucan synthase. RiGT2 has conserved glycosyltransferase family 2 (GT2) motifs, including D, D, D, QXXRW, and a C-terminal PilZ domain that resembles the C-terminal domain of bacteria cellulose synthase, BcsA. Using a direct gain-of-function approach, we insert RiGT2 into Saccharomyces cerevisiae, and (1,3;1,4)-β-D-glucans are produced with structures similar to those of the (1,3;1,4)-β-D-glucans of the lichen Cetraria islandica. Phylogenetic analysis reveals that putative (1,3;1,4)-β-D-glucan synthase candidate genes in several other bacterial species support the finding of (1,3;1,4)-β-D-glucans in these species.
Collapse
Affiliation(s)
- Shu-Chieh Chang
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Centre, Stockholm, SE10691, Sweden
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Mu-Rong Kao
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Centre, Stockholm, SE10691, Sweden
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Rebecka Karmakar Saldivar
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Centre, Stockholm, SE10691, Sweden
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Sara M Díaz-Moreno
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Centre, Stockholm, SE10691, Sweden
| | - Xiaohui Xing
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB, T1J 4B1, Canada
| | - Valentina Furlanetto
- Department of Industrial Biotechnology, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Centre, Stockholm, SE10691, Sweden
| | - Johannes Yayo
- Department of Industrial Biotechnology, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Centre, Stockholm, SE10691, Sweden
| | - Christina Divne
- Department of Industrial Biotechnology, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Centre, Stockholm, SE10691, Sweden
| | - Francisco Vilaplana
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Centre, Stockholm, SE10691, Sweden
| | - D Wade Abbott
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB, T1J 4B1, Canada
| | - Yves S Y Hsieh
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Centre, Stockholm, SE10691, Sweden.
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
| |
Collapse
|
27
|
Zhang Q, Peng L, Han W, Chen H, Tang H, Chen X, Langford PR, Huang Q, Zhou R, Li L. The morphology and metabolic changes of Actinobacillus pleuropneumoniae during its growth as a biofilm. Vet Res 2023; 54:42. [PMID: 37237397 PMCID: PMC10224306 DOI: 10.1186/s13567-023-01173-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023] Open
Abstract
Actinobacillus pleuropneumoniae is an important swine respiratory pathogen. Previous studies have suggested that growth as a biofilm is a natural state of A. pleuropneumoniae infection. To understand the survival features involved in the biofilm state, the growth features, morphology and gene expression profiles of planktonic and biofilm A. pleuropneumoniae were compared. A. pleuropneumoniae in biofilms showed reduced viability but maintained the presence of extracellular polymeric substances (EPS) after late log-phase. Under the microscope, bacteria in biofilms formed dense aggregated structures that were connected by abundant EPS, with reduced condensed chromatin. By construction of Δpga and ΔdspB mutants, polymeric β-1,6-linked N-acetylglucosamine and dispersin B were confirmed to be critical for normal biofilm formation. RNA-seq analysis indicated that, compared to their planktonic counterparts, A. pleuropneumoniae in biofilms had an extensively altered transcriptome. Carbohydrate metabolism, energy metabolism and translation were significantly repressed, while fermentation and genes contributing to EPS synthesis and translocation were up-regulated. The regulators Fnr (HlyX) and Fis were found to be up-regulated and their binding motifs were identified in the majority of the differentially expressed genes, suggesting their coordinated global role in regulating biofilm metabolism. By comparing the transcriptome of wild-type biofilm and Δpga, the utilization of oligosaccharides, iron and sulfur and fermentation were found to be important in adhesion and aggregation during biofilm formation. Additionally, when used as inocula, biofilm bacteria showed reduced virulence in mouse, compared with planktonic grown cells. Thus, these results have identified new facets of A. pleuropneumoniae biofilm maintenance and regulation.
Collapse
Affiliation(s)
- Qiuhong Zhang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
| | - Lu Peng
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
| | - Weiyao Han
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
| | - Hongyu Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
| | - Hao Tang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
| | - Xiabing Chen
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Sciences, Wuhan, 430070, Hubei, China
| | - Paul R Langford
- Section of Paediatric Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | - Qi Huang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, 430070, Hubei, China
| | - Rui Zhou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, 430070, Hubei, China
| | - Lu Li
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China.
- International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, 430070, Hubei, China.
| |
Collapse
|
28
|
Burnett AJN, Rodriguez E, Constable S, Lowrance B, Fish M, Weadge JT. WssI from the Gram-Negative Bacterial Cellulose Synthase is an O-acetyltransferase that Acts on Cello-oligomers with Several Acetyl Donor Substrates. J Biol Chem 2023:104849. [PMID: 37224964 PMCID: PMC10302187 DOI: 10.1016/j.jbc.2023.104849] [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: 09/06/2022] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/26/2023] Open
Abstract
In microbial biofilms, bacterial cells are encased in a self-produced matrix of polymers (e.g., exopolysaccharides) that enable surface adherence and protect against environmental stressors. For example, the wrinkly spreader phenotype of Pseudomonas fluorescens colonizes food/water sources and human tissue to form robust biofilms that can spread across surfaces. This biofilm largely consists of bacterial cellulose produced by the cellulose synthase proteins encoded by the wss operon, which also occurs in other species, including pathogenic Achromobacter species. Although phenotypic mutant analysis of the wssFGHI genes has previously shown that they are responsible for acetylation of bacterial cellulose, their specific roles remain unknown and distinct from the recently identified cellulose phosphoethanolamine modification found in other species. Here we have purified the C-terminal soluble form of WssI from P. fluorescens and A. insuavis and demonstrated acetyl-esterase activity with chromogenic substrates. The kinetic parameters (kcat/KM values of 13 and 8.0 M-1∙ s-1, respectively) indicate that these enzymes are up to four times more catalytically efficient than the closest characterized homolog, AlgJ from the alginate synthase. Unlike AlgJ and its cognate alginate polymer, WssI also demonstrated acetyltransferase activity onto cellulose oligomers (e.g., cellotetraose to cellohexaose) with multiple acetyl-donor substrates (pNP-Ac, MU-Ac and acetyl-CoA). Finally, a high-throughput screen identified three low micromolar WssI inhibitors that may be useful for chemically interrogating cellulose acetylation and biofilm formation.
Collapse
Affiliation(s)
| | - Emily Rodriguez
- Department of Biology, Wilfrid Laurier University, Waterloo, ON, Canada
| | - Shirley Constable
- Department of Biology, Wilfrid Laurier University, Waterloo, ON, Canada
| | - Brian Lowrance
- Department of Biology, Wilfrid Laurier University, Waterloo, ON, Canada
| | - Michael Fish
- Department of Biology, Wilfrid Laurier University, Waterloo, ON, Canada
| | - Joel T Weadge
- Department of Biology, Wilfrid Laurier University, Waterloo, ON, Canada.
| |
Collapse
|
29
|
Li J, Chen Y, Wan Q, Zhang M. Phosphorus level impacts luteolin effect on Microcystis aeruginosa growth and microcystin-pollution risk - Novel perspective from correlation between exopolymers substances fractions and microcystin-production/release. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 255:114794. [PMID: 36948014 DOI: 10.1016/j.ecoenv.2023.114794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/03/2023] [Accepted: 03/14/2023] [Indexed: 06/18/2023]
Abstract
Luteolin as a phytogenic algicide can inhibit the growth and microcystins (MCs) release of Microcystis, a dominant genus during cyanobacterial blooms, but how phosphorus (P) level impacts luteolin effect on its growth and MC-pollution risk is unclear. By employing Microcystis aeruginosa as test alga, this study addressed this concern and explored response mechanisms from novel insights of relationship between extracellular polysaccharide (ex-poly) and protein (ex-pro) contents and MC-production/release. At each P level (0.05-5 mg/L), rising luteolin dose more greatly inhibited Microcystis growth and MC-pollution risk, with growth inhibition ratio of around 10%-30%, 20%-50% and 40%-90% for 3, 6 and 12 mg/L luteolin, respectively, but almost increasingly enhanced cellular ability of MC-production/conservation and total and bound ex-poly/ex-pro production. Rising P level promoted Microcystis growth and intracellular/extracellular MCs content (IMC, EMC) in test system at each luteolin dose, thus higher P level weakened algicidal and MC-removal effects of luteolin, indicating that P-decrease was required for stronger application outcome of luteolin. Total and bound ex-poly/ex-pro amount were positively correlated with cellular MC-production/conservation ability, IMC and EMC, which constituted cooperative stress-defense of Microcystis at each P level. Besides, rising luteolin dose posed stronger algicidal effect by inactivating gene expression involving peroxidase synthesis (especially at P-limitation), photosynthesis and P acquisition, while rising P level alleviated algicidal and MC-pollution inhibition effects of luteolin by enhancing gene expression involving N acquisition and peroxidase synthesis. This study shed novel insights for P-dependent effect and mechanisms of luteolin on toxigenic Microcystis growth and MC-pollution control, which guided to mitigating toxigenic Microcystis-dominated cyanobacterial blooms in different P-level water areas.
Collapse
Affiliation(s)
- Jieming Li
- College of Resources and Environmental Sciences, China Agricultural University, Beijing 100193, China; Beijing Key Laboratory of Biodiversity and Organic Farming, China Agricultural University, Beijing 100193, China.
| | - Yanran Chen
- College of Resources and Environmental Sciences, China Agricultural University, Beijing 100193, China
| | - Qianruo Wan
- College of Resources and Environmental Sciences, China Agricultural University, Beijing 100193, China
| | - Mingxia Zhang
- College of Resources and Environmental Sciences, China Agricultural University, Beijing 100193, China
| |
Collapse
|
30
|
Stephens Z, Wilson LFL, Zimmer J. Diverse mechanisms of polysaccharide biosynthesis, assembly and secretion across kingdoms. Curr Opin Struct Biol 2023; 79:102564. [PMID: 36870276 DOI: 10.1016/j.sbi.2023.102564] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/16/2023] [Accepted: 01/28/2023] [Indexed: 03/06/2023]
Abstract
Polysaccharides are essential biopolymers produced in all kingdoms of life. On the cell surface, they represent versatile architectural components, forming protective capsules and coats, cell walls, or adhesives. Extracellular polysaccharide (EPS) biosynthesis mechanisms differ based on the cellular localization of polymer assembly. Some polysaccharides are first synthesized in the cytosol and then extruded by ATP powered transporters [1]. In other cases, the polymers are assembled outside the cell [2], synthesized and secreted in a single step [3], or deposited on the cell surface via vesicular trafficking [4]. This review focuses on recent insights into the biosynthesis, secretion, and assembly of EPS in microbes, plants and vertebrates. We focus on comparing the sites of biosynthesis, secretion mechanisms, and higher-order EPS assemblies.
Collapse
Affiliation(s)
- Zachery Stephens
- Howard Hughes Medical Institute, University of Virginia School of Medicine, Charlottesville, VA, USA; Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, 480 Ray C. Hunt Dr., Charlottesville, VA 22908, USA
| | - Louis F L Wilson
- Howard Hughes Medical Institute, University of Virginia School of Medicine, Charlottesville, VA, USA; Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, 480 Ray C. Hunt Dr., Charlottesville, VA 22908, USA
| | - Jochen Zimmer
- Howard Hughes Medical Institute, University of Virginia School of Medicine, Charlottesville, VA, USA; Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, 480 Ray C. Hunt Dr., Charlottesville, VA 22908, USA.
| |
Collapse
|
31
|
Gonçalves ASC, Leitão MM, Simões M, Borges A. The action of phytochemicals in biofilm control. Nat Prod Rep 2023; 40:595-627. [PMID: 36537821 DOI: 10.1039/d2np00053a] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Covering: 2009 to 2021Antimicrobial resistance is now rising to dangerously high levels in all parts of the world, threatening the treatment of an ever-increasing range of infectious diseases. This has becoming a serious public health problem, especially due to the emergence of multidrug-resistance among clinically important bacterial species and their ability to form biofilms. In addition, current anti-infective therapies have low efficacy in the treatment of biofilm-related infections, leading to recurrence, chronicity, and increased morbidity and mortality. Therefore, it is necessary to search for innovative strategies/antibacterial agents capable of overcoming the limitations of conventional antibiotics. Natural compounds, in particular those obtained from plants, have been exhibiting promising properties in this field. Plant secondary metabolites (phytochemicals) can act as antibiofilm agents through different mechanisms of action from the available antibiotics (inhibition of quorum-sensing, motility, adhesion, and reactive oxygen species production, among others). The combination of different phytochemicals and antibiotics have revealed synergistic or additive effects in biofilm control. This review aims to bring together the most relevant reports on the antibiofilm properties of phytochemicals, as well as insights into their structure and mechanistic action against bacterial pathogens, spanning December 2008 to December 2021.
Collapse
Affiliation(s)
- Ariana S C Gonçalves
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Miguel M Leitão
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Manuel Simões
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Anabela Borges
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| |
Collapse
|
32
|
Fullen AR, Gutierrez-Ferman JL, Rayner RE, Kim SH, Chen P, Dubey P, Wozniak DJ, Peeples ME, Cormet-Boyaka E, Deora R. Architecture and matrix assembly determinants of Bordetella pertussis biofilms on primary human airway epithelium. PLoS Pathog 2023; 19:e1011193. [PMID: 36821596 PMCID: PMC9990917 DOI: 10.1371/journal.ppat.1011193] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 03/07/2023] [Accepted: 02/09/2023] [Indexed: 02/24/2023] Open
Abstract
Traditionally, whooping cough or pertussis caused by the obligate human pathogen Bordetella pertussis (Bp) is described as an acute disease with severe symptoms. However, many individuals who contract pertussis are either asymptomatic or show very mild symptoms and yet can serve as carriers and sources of bacterial transmission. Biofilms are an important survival mechanism for bacteria in human infections and disease. However, bacterial determinants that drive biofilm formation in humans are ill-defined. In the current study, we show that Bp infection of well-differentiated primary human bronchial epithelial cells leads to formation of bacterial aggregates, clusters, and highly structured biofilms which are colocalized with cilia. These findings mimic observations from pathological analyses of tissues from pertussis patients. Distinct arrangements (mono-, bi-, and tri-partite) of the polysaccharide Bps, extracellular DNA, and bacterial cells were visualized, suggesting complex heterogeneity in bacteria-matrix interactions. Analyses of mutant biofilms revealed positive roles in matrix production, cell cluster formation, and biofilm maturity for three critical Bp virulence factors: Bps, filamentous hemagglutinin, and adenylate cyclase toxin. Adherence assays identified Bps as a new Bp adhesin for primary human airway cells. Taken together, our results demonstrate the multi-factorial nature of the biofilm extracellular matrix and biofilm development process under conditions mimicking the human respiratory tract and highlight the importance of model systems resembling the natural host environment to investigate pathogenesis and potential therapeutic strategies.
Collapse
Affiliation(s)
- Audra R. Fullen
- The Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Jessica L. Gutierrez-Ferman
- The Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Rachael E. Rayner
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Sun Hee Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Phylip Chen
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Purnima Dubey
- The Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Daniel J. Wozniak
- The Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
- Department of Microbiology, The Ohio State University, Columbus, Ohio, United States of America
| | - Mark E. Peeples
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, United States of America
| | - Estelle Cormet-Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Rajendar Deora
- The Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
- Department of Microbiology, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
33
|
Dueholm MKD, Besteman M, Zeuner EJ, Riisgaard-Jensen M, Nielsen ME, Vestergaard SZ, Heidelbach S, Bekker NS, Nielsen PH. Genetic potential for exopolysaccharide synthesis in activated sludge bacteria uncovered by genome-resolved metagenomics. WATER RESEARCH 2023; 229:119485. [PMID: 36538841 DOI: 10.1016/j.watres.2022.119485] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/08/2022] [Accepted: 12/10/2022] [Indexed: 06/17/2023]
Abstract
A good floc formation of activated sludge (AS) is crucial for solid-liquid separation and production of clean effluent during wastewater treatment. Floc formation is partly controlled by self-produced extracellular polymeric substances (EPS) such as exopolysaccharides, proteins, and nucleic acids. Little is known about the composition, structure, and function of EPS in AS and which bacteria produce them. To address this knowledge gap for the exopolysaccharides, we took advantage of 1083 high-quality metagenome-assembled genomes (MAGs) obtained from 23 Danish wastewater treatment plants. We investigated the genomic potential for exopolysaccharide biosynthesis in bacterial species typical in AS systems based on genome mining and gene synteny analyses. Putative gene clusters associated with the biosynthesis of alginate, cellulose, curdlan, diutan, hyaluronic acids, Pel, poly-β-1,6-N-acetyl-d-glucosamine (PNAG), Psl, S88 capsular polysaccharide, salecan, succinoglycan, and xanthan were identified and linked to individual MAGs, providing a comprehensive overview of the genome-resolved potential for these exopolysaccharides in AS bacteria. The approach and results provide a starting point for a more comprehensive understanding of EPS composition in wastewater treatment systems, which may facilitate a more refined regulation of the activated sludge process for improved stability.
Collapse
Affiliation(s)
- Morten Kam Dahl Dueholm
- Center for Microbial Communities, Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark.
| | - Maaike Besteman
- Department of Agrotechnology and Food Sciences, Wageningen University & Research, Wageningen, Netherlands
| | - Emil Juel Zeuner
- Center for Microbial Communities, Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Marie Riisgaard-Jensen
- Center for Microbial Communities, Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Morten Eneberg Nielsen
- Center for Microbial Communities, Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Sofie Zacho Vestergaard
- Center for Microbial Communities, Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Søren Heidelbach
- Center for Microbial Communities, Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Nicolai Sundgaard Bekker
- Center for Microbial Communities, Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Per Halkjær Nielsen
- Center for Microbial Communities, Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| |
Collapse
|
34
|
Newman KE, Tindall SN, Mader SL, Khalid S, Thomas GH, Van Der Woude MW. A novel fold for acyltransferase-3 (AT3) proteins provides a framework for transmembrane acyl-group transfer. eLife 2023; 12:e81547. [PMID: 36630168 PMCID: PMC9833829 DOI: 10.7554/elife.81547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 12/04/2022] [Indexed: 01/12/2023] Open
Abstract
Acylation of diverse carbohydrates occurs across all domains of life and can be catalysed by proteins with a membrane bound acyltransferase-3 (AT3) domain (PF01757). In bacteria, these proteins are essential in processes including symbiosis, resistance to viruses and antimicrobials, and biosynthesis of antibiotics, yet their structure and mechanism are largely unknown. In this study, evolutionary co-variance analysis was used to build a computational model of the structure of a bacterial O-antigen modifying acetyltransferase, OafB. The resulting structure exhibited a novel fold for the AT3 domain, which molecular dynamics simulations demonstrated is stable in the membrane. The AT3 domain contains 10 transmembrane helices arranged to form a large cytoplasmic cavity lined by residues known to be essential for function. Further molecular dynamics simulations support a model where the acyl-coA donor spans the membrane through accessing a pore created by movement of an important loop capping the inner cavity, enabling OafB to present the acetyl group close to the likely catalytic resides on the extracytoplasmic surface. Limited but important interactions with the fused SGNH domain in OafB are identified, and modelling suggests this domain is mobile and can both accept acyl-groups from the AT3 and then reach beyond the membrane to reach acceptor substrates. Together this new general model of AT3 function provides a framework for the development of inhibitors that could abrogate critical functions of bacterial pathogens.
Collapse
Affiliation(s)
- Kahlan E Newman
- School of Chemistry, University of SouthamptonSouthamptonUnited Kingdom
| | - Sarah N Tindall
- Department of Biology and the York Biomedical Research Institute, University of YorkYorkUnited Kingdom
| | - Sophie L Mader
- Department of Biochemistry, University of OxfordOxfordUnited Kingdom
| | - Syma Khalid
- Department of Biochemistry, University of OxfordOxfordUnited Kingdom
| | - Gavin H Thomas
- Department of Biology and the York Biomedical Research Institute, University of YorkYorkUnited Kingdom
| | - Marjan W Van Der Woude
- Hull York Medical School and the York Biomedical Research Institute, University of YorkYorkUnited Kingdom
| |
Collapse
|
35
|
Kaur N, Dey P. Bacterial Exopolysaccharides as Emerging Bioactive Macromolecules: From Fundamentals to Applications. Res Microbiol 2022; 174:104024. [PMID: 36587857 DOI: 10.1016/j.resmic.2022.104024] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022]
Abstract
Microbial exopolysaccharides (EPS) are extracellular carbohydrate polymers forming capsules or slimy coating around the cells. EPS can be secreted by various bacterial genera that can help bacterial cells in attachment, environmental adaptation, stress tolerance and are an integral part of microbial biofilms. Several gut commensals (e.g., Lactobacillus, Bifidobacterium) produce EPS that possess diverse bioactivities. Bacterial EPS also has extensive commercial applications in the pharmaceutical and food industries. Owing to the structural and functional diversity, genetic and metabolic engineering strategies are currently employed to increase EPS production. Therefore, the current review provides a comprehensive overview of the fundamentals of bacterial exopolysaccharides, including their classification, source, biosynthetic pathways, and functions in the microbial community. The review also provides an overview of the diverse bioactivities of microbial EPS, including immunomodulatory, anti-diabetic, anti-obesity, and anti-cancer properties. Since several gut microbes are EPS producers and gut microbiota helps maintain a functional gut barrier, emphasis has been given to the intestinal-level bioactivities of the gut microbial EPS. Collectively, the review provides a comprehensive overview of microbial bioactive exopolysaccharides.
Collapse
Affiliation(s)
- Navneet Kaur
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, India
| | - Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, India.
| |
Collapse
|
36
|
Gheorghita AA, Li YE, Kitova EN, Bui DT, Pfoh R, Low KE, Whitfield GB, Walvoort MTC, Zhang Q, Codée JDC, Klassen JS, Howell PL. Structure of the AlgKX modification and secretion complex required for alginate production and biofilm attachment in Pseudomonas aeruginosa. Nat Commun 2022; 13:7631. [PMID: 36494359 PMCID: PMC9734138 DOI: 10.1038/s41467-022-35131-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 11/18/2022] [Indexed: 12/13/2022] Open
Abstract
Synthase-dependent secretion systems are a conserved mechanism for producing exopolysaccharides in Gram-negative bacteria. Although widely studied, it is not well understood how these systems are organized to coordinate polymer biosynthesis, modification, and export across both membranes and the peptidoglycan. To investigate how synthase-dependent secretion systems produce polymer at a molecular level, we determined the crystal structure of the AlgK-AlgX (AlgKX) complex involved in Pseudomonas aeruginosa alginate exopolysaccharide acetylation and export. We demonstrate that AlgKX directly binds alginate oligosaccharides and that formation of the complex is vital for polymer production and biofilm attachment. Finally, we propose a structural model for the AlgEKX outer membrane modification and secretion complex. Together, our study provides insight into how alginate biosynthesis proteins coordinate production of a key exopolysaccharide involved in establishing persistent Pseudomonas lung infections.
Collapse
Affiliation(s)
- Andreea A. Gheorghita
- grid.42327.300000 0004 0473 9646Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Biochemistry, University of Toronto, Toronto, ON Canada
| | - Yancheng E. Li
- grid.42327.300000 0004 0473 9646Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Biochemistry, University of Toronto, Toronto, ON Canada ,grid.20861.3d0000000107068890Present Address: Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA USA
| | - Elena N. Kitova
- grid.17089.370000 0001 2190 316XDepartment of Chemistry, University of Alberta, Edmonton, AB Canada
| | - Duong T. Bui
- grid.17089.370000 0001 2190 316XDepartment of Chemistry, University of Alberta, Edmonton, AB Canada
| | - Roland Pfoh
- grid.42327.300000 0004 0473 9646Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON Canada
| | - Kristin E. Low
- grid.42327.300000 0004 0473 9646Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON Canada ,grid.55614.330000 0001 1302 4958Present Address: Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB Canada
| | - Gregory B. Whitfield
- grid.42327.300000 0004 0473 9646Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Biochemistry, University of Toronto, Toronto, ON Canada ,grid.14848.310000 0001 2292 3357Present Address: Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC Canada
| | - Marthe T. C. Walvoort
- grid.5132.50000 0001 2312 1970Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands ,grid.4830.f0000 0004 0407 1981Present Address: Department of Chemical Biology, Stratingh Institute for Chemistry, University of Groningen, Groningen, The Netherlands
| | - Qingju Zhang
- grid.5132.50000 0001 2312 1970Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands ,grid.411862.80000 0000 8732 9757Present Address: National Research Centre for Carbohydrate Synthesis, Jiangxi Normal University, Nanchang, China
| | - Jeroen D. C. Codée
- grid.5132.50000 0001 2312 1970Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - John S. Klassen
- grid.17089.370000 0001 2190 316XDepartment of Chemistry, University of Alberta, Edmonton, AB Canada
| | - P. Lynne Howell
- grid.42327.300000 0004 0473 9646Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Biochemistry, University of Toronto, Toronto, ON Canada
| |
Collapse
|
37
|
Dai P, Hu D. The making of hypervirulent Klebsiella pneumoniae. J Clin Lab Anal 2022; 36:e24743. [PMID: 36347819 PMCID: PMC9757020 DOI: 10.1002/jcla.24743] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/01/2022] [Accepted: 10/09/2022] [Indexed: 10/08/2023] Open
Abstract
Klebsiella pneumoniae is a notorious bacterium in clinical practice. Virulence, carbapenem-resistance and their convergence among K. pneumoniae are extensively discussed in this article. Hypervirulent K. pneumoniae (HvKP) has spread from the Asian Pacific Rim to the world, inducing various invasive infections, such as pyogenic liver abscess, endophthalmitis, and meningitis. Furthermore, HvKP has acquired more and more drug resistance. Among multidrug-resistant HvKP, hypervirulent carbapenem-resistant K. pneumoniae (Hv-CRKP), and carbapenem-resistant hypervirulent K. pneumoniae (CR-HvKP) are both devastating for their extreme drug resistance and virulence. The hypervirulence of HvKP is primarily attributed to hypercapsule, macromolecular exopolysaccharides, or excessive siderophores, although it has many other factors, for example, lipopolysaccharides, fimbriae, and porins. In contrast with classical determination of HvKP, that is, animal lethality test, molecular determination could be an optional and practical method after improvement. HvKP, including Hv-CRKP and CR-HvKP, has been progressing. R-M and CRISPR-Cas systems may play pivotal roles in such evolutions. Hv-CRKP and CR-HvKP, in particular the former, should be of severe concern due to their being more and more prevalent.
Collapse
Affiliation(s)
- Piaopiao Dai
- Department of Laboratory MedicineTaizhou Municipal HospitalTaizhouChina
| | - Dakang Hu
- Department of Laboratory MedicineTaizhou Municipal HospitalTaizhouChina
| |
Collapse
|
38
|
Saïdi F, Mahanta U, Panda A, Kezzo AA, Jolivet NY, Bitazar R, John G, Martinez M, Mellouk A, Calmettes C, Chang YW, Sharma G, Islam ST. Bacterial Outer Membrane Polysaccharide Export (OPX) Proteins Occupy Three Structural Classes with Selective β-Barrel Porin Requirements for Polymer Secretion. Microbiol Spectr 2022; 10:e0129022. [PMID: 36200915 PMCID: PMC9603273 DOI: 10.1128/spectrum.01290-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 09/08/2022] [Indexed: 12/30/2022] Open
Abstract
Secretion of high-molecular-weight polysaccharides across the bacterial envelope is ubiquitous, as it enhances prokaryotic survival in (a)biotic settings. Such polymers are often assembled by Wzx/Wzy- or ABC transporter-dependent schemes implicating outer membrane (OM) polysaccharide export (OPX) proteins in cell-surface polymer translocation. In the social predatory bacterium Myxococcus xanthus, the exopolysaccharide (EPS) pathway WzaX, major spore coat (MASC) pathway WzaS, and biosurfactant polysaccharide (BPS) pathway WzaB were herein found to be truncated OPX homologues of Escherichia coli Wza lacking OM-spanning α-helices. Comparative genomics across all bacteria (>91,000 OPX proteins identified and analyzed), complemented with cryo-electron tomography cell-envelope analyses, revealed such "truncated" WzaX/S/B architecture to be the most common among three defined OPX-protein structural classes independent of periplasm thickness. Fold recognition and deep learning revealed the conserved M. xanthus proteins MXAN_7418/3226/1916 (encoded beside wzaX/S/B, respectively) to be integral OM β-barrels, with structural homology to the poly-N-acetyl-d-glucosamine synthase-dependent pathway porin PgaA. Such bacterial porins were identified near numerous genes for all three OPX protein classes. Interior MXAN_7418/3226/1916 β-barrel electrostatics were found to match properties of their associated polymers. With MXAN_3226 essential for MASC export, and MXAN_7418 herein shown to mediate EPS translocation, we have designated this new secretion machinery component "Wzp" (i.e., Wz porin), with the final step of M. xanthus EPS/MASC/BPS secretion across the OM now proposed to be mediated by WzpX/S/B (i.e., MXAN_7418/3226/1916). Importantly, these data support a novel and widespread secretion paradigm for polysaccharide biosynthesis pathways in which those containing OPX components that cannot span the OM instead utilize β-barrel porins to mediate polysaccharide transport across the OM. IMPORTANCE Diverse bacteria assemble and secrete polysaccharides that alter their physiologies through modulation of motility, biofilm formation, and host immune system evasion. Most such pathways require outer membrane (OM) polysaccharide export (OPX) proteins for sugar-polymer transport to the cell surface. In the prototypic Escherichia coli Group-1-capsule biosynthesis system, eight copies of this canonical OPX protein cross the OM with an α-helix, forming a polysaccharide-export pore. Herein, we instead reveal that most OPX proteins across all bacteria lack this α-helix, raising questions as to the manner by which most secreted polysaccharides actually exit cells. In the model developmental bacterium Myxococcus xanthus, we show this process to depend on OPX-coupled OM-spanning β-barrel porins, with similar porins encoded near numerous OPX genes in diverse bacteria. Knowledge of the terminal polysaccharide secretion step will enable development of antimicrobial compounds targeted to blocking polymer export from outside the cell, thus bypassing any requirements for antimicrobial compound uptake by the cell.
Collapse
Affiliation(s)
- Fares Saïdi
- Institut National de la Recherche Scientifique (INRS), Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Institut Pasteur International Network, Laval, Quebec, Canada
- PROTEO, the Quebec Network for Research on Protein Function, Engineering, and Applications, Université Laval, Québec, Quebec, Canada
| | - Utkarsha Mahanta
- Institute of Bioinformatics and Applied Biotechnology (IBAB), Bengaluru, Karnataka, India
| | - Adyasha Panda
- Institute of Bioinformatics and Applied Biotechnology (IBAB), Bengaluru, Karnataka, India
| | - Ahmad A. Kezzo
- Institut National de la Recherche Scientifique (INRS), Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Institut Pasteur International Network, Laval, Quebec, Canada
- PROTEO, the Quebec Network for Research on Protein Function, Engineering, and Applications, Université Laval, Québec, Quebec, Canada
| | - Nicolas Y. Jolivet
- Institut National de la Recherche Scientifique (INRS), Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Institut Pasteur International Network, Laval, Quebec, Canada
- PROTEO, the Quebec Network for Research on Protein Function, Engineering, and Applications, Université Laval, Québec, Quebec, Canada
| | - Razieh Bitazar
- Institut National de la Recherche Scientifique (INRS), Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Institut Pasteur International Network, Laval, Quebec, Canada
- PROTEO, the Quebec Network for Research on Protein Function, Engineering, and Applications, Université Laval, Québec, Quebec, Canada
| | - Gavin John
- Department of Pediatrics, Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Matthew Martinez
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Abdelkader Mellouk
- Institut National de la Recherche Scientifique (INRS), Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Institut Pasteur International Network, Laval, Quebec, Canada
- PROTEO, the Quebec Network for Research on Protein Function, Engineering, and Applications, Université Laval, Québec, Quebec, Canada
| | - Charles Calmettes
- Institut National de la Recherche Scientifique (INRS), Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Institut Pasteur International Network, Laval, Quebec, Canada
- PROTEO, the Quebec Network for Research on Protein Function, Engineering, and Applications, Université Laval, Québec, Quebec, Canada
| | - Yi-Wei Chang
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gaurav Sharma
- Institute of Bioinformatics and Applied Biotechnology (IBAB), Bengaluru, Karnataka, India
| | - Salim T. Islam
- Institut National de la Recherche Scientifique (INRS), Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Institut Pasteur International Network, Laval, Quebec, Canada
- PROTEO, the Quebec Network for Research on Protein Function, Engineering, and Applications, Université Laval, Québec, Quebec, Canada
| |
Collapse
|
39
|
Vandana, Das S. Genetic regulation, biosynthesis and applications of extracellular polysaccharides of the biofilm matrix of bacteria. Carbohydr Polym 2022; 291:119536. [DOI: 10.1016/j.carbpol.2022.119536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 11/02/2022]
|
40
|
Bps polysaccharide of Bordetella pertussis resists antimicrobial peptides by functioning as a dual surface shield and decoy and converts Escherichia coli into a respiratory pathogen. PLoS Pathog 2022; 18:e1010764. [PMID: 35969621 PMCID: PMC9410548 DOI: 10.1371/journal.ppat.1010764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 08/25/2022] [Accepted: 07/24/2022] [Indexed: 01/01/2023] Open
Abstract
Infections and disease caused by the obligate human pathogen Bordetella pertussis (Bp) are increasing, despite widespread vaccinations. The current acellular pertussis vaccines remain ineffective against nasopharyngeal colonization, carriage, and transmission. In this work, we tested the hypothesis that Bordetella polysaccharide (Bps), a member of the poly-β-1,6-N-acetyl-D-glucosamine (PNAG/PGA) family of polysaccharides promotes respiratory tract colonization of Bp by resisting killing by antimicrobial peptides (AMPs). Genetic deletion of the bpsA-D locus, as well as treatment with the specific glycoside hydrolase Dispersin B, increased susceptibility to AMP-mediated killing. Bps was found to be both cell surface-associated and released during laboratory growth and mouse infections. Addition of bacterial supernatants containing Bps and purified Bps increased B. pertussis resistance to AMPs. By utilizing ELISA, immunoblot and flow cytometry assays, we show that Bps functions as a dual surface shield and decoy. Co-inoculation of C57BL/6J mice with a Bps-proficient strain enhanced respiratory tract survival of the Bps-deficient strain. In combination, the presented results highlight the critical role of Bps as a central driver of B. pertussis pathogenesis. Heterologous production of Bps in a non-pathogenic E. coli K12 strain increased AMP resistance in vitro, and augmented bacterial survival and pathology in the mouse respiratory tract. These studies can serve as a foundation for other PNAG/PGA polysaccharides and for the development of an effective Bp vaccine that includes Bps.
Collapse
|
41
|
The TPR domain of PgaA is a multifunctional scaffold that binds PNAG and modulates PgaB-dependent polymer processing. PLoS Pathog 2022; 18:e1010750. [PMID: 35930610 PMCID: PMC9384988 DOI: 10.1371/journal.ppat.1010750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 08/17/2022] [Accepted: 07/19/2022] [Indexed: 11/30/2022] Open
Abstract
The synthesis of exopolysaccharides as biofilm matrix components by pathogens is a crucial factor for chronic infections and antibiotic resistance. Many periplasmic proteins involved in polymer processing and secretion in Gram-negative synthase dependent exopolysaccharide biosynthetic systems have been individually characterized. The operons responsible for the production of PNAG, alginate, cellulose and the Pel polysaccharide each contain a gene that encodes an outer membrane associated tetratricopeptide repeat (TPR) domain containing protein. While the TPR domain has been shown to bind other periplasmic proteins, the functional consequences of these interactions for the polymer remain poorly understood. Herein, we show that the C-terminal TPR region of PgaA interacts with the de-N-acetylase domain of PgaB, and increases its deacetylase activity. Additionally, we found that when the two proteins form a complex, the glycoside hydrolase activity of PgaB is also increased. To better understand structure-function relationships we determined the crystal structure of a stable TPR module, which has a conserved groove formed by three repeat motifs. Tryptophan quenching, mass spectrometry analysis and molecular dynamics simulation studies suggest that the crystallized TPR module can bind PNAG/dPNAG via its electronegative groove on the concave surface, and potentially guide the polymer through the periplasm towards the porin for export. Our results suggest a scaffolding role for the TPR domain that combines PNAG/dPNAG translocation with the modulation of its chemical structure by PgaB. Exopolysaccharides are an important component of the extracellular matrix of bacterial and fungal biofilms and provide protection against the host immune response and antibiotics. In Gram-negative bacteria, these polymers are synthesized in the inner membrane and translocated across the periplasm before being secreted across the outer membrane. The periplasm presents both a challenge as an additional environment to cross and an opportunity to chemically alter the polymer prior to secretion to render it more effective. This study focuses on a periplasmic alpha-helical repeat domain whose wide-spread homologues are involved in the export of many chemically distinct exopolysaccharides. We found that in E. coli this superhelical TPR domain acts as a scaffold that can bind the polymer PNAG and alter the enzymatic activity of PgaB, thus providing a means to affect the deacetylation level and chain length of the secreted polymer. Scaffold proteins are known as binding hubs within cellular pathways that often have a central regulatory function and facilitate evolution due to their repetitive modular building blocks. Our study sheds light on the principles of polysaccharide modification and export, which we hope will promote the development of applications against bacterial infections.
Collapse
|
42
|
Cyanophycin Granule Polypeptide: a Neglected High Value-Added Biopolymer, Synthesized in Activated Sludge on a Large Scale. Appl Environ Microbiol 2022; 88:e0074222. [PMID: 35862662 PMCID: PMC9317870 DOI: 10.1128/aem.00742-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Recovery of microbial synthetic polymers with high economic value and market demand in activated sludge has attracted extensive attention. This work analyzed the synthesis of cyanophycin granule peptide (CGP) in activated sludge and its adsorption capacity for heavy metals and dyes. The distribution and expression of synthetic genes for eight biopolymers in two wastewater treatment plants (WWTPs) were analyzed by metagenomics and metatranscriptomics. The results indicate that the abundance and expression level of CGP synthase (cphA) are similar to those of polyhydroxyalkanoate polymerase, implying high synthesis of CGP in activated sludges. CGP in activated sludge is mainly polymerized from aspartic acid and arginine, and its secondary structure is mainly β-sheet. The crude yields of CGP are as high as 104 ± 26 and 76 ± 13 mg/g dry sludge in winter and in summer, respectively, comparable to those of polyhydroxyalkanoate and alginate. CGP has a stronger adsorption capacity for anionic pollutants (Cr (VI) and methyl orange) than for cationic pollutants because it is rich in guanidine groups. This study highlights prospects for recovery and application of CGP from WWTPs. IMPORTANCE The conversion of organic pollutants into bioresources by activated sludge can reduce the carbon dioxide emission of wastewater treatment plants. Identification of new high value-added biopolymers produced by activated sludge is beneficial to recover bioresources. Cyanophycin granule polypeptide (CGP), first discovered in cyanobacteria, has unique chemical and material properties suitable for industrial food, medicine, cosmetics, water treatment, and agriculture applications. Here, we revealed for the first time that activated sludge has a remarkable ability to produce CGP. These findings could further facilitate the conversion of wastewater treatment plants into resource recycling plants.
Collapse
|
43
|
The CRISPR-Cas System Differentially Regulates Surface-Attached and Pellicle Biofilm in Salmonella enterica Serovar Typhimurium. Microbiol Spectr 2022; 10:e0020222. [PMID: 35678575 PMCID: PMC9241790 DOI: 10.1128/spectrum.00202-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The CRISPR-Cas mediated regulation of biofilm by Salmonella enterica serovar Typhimurium was investigated by deleting CRISPR-Cas components ΔcrisprI, ΔcrisprII, ΔΔcrisprI crisprII, and Δcas op. We determined that the system positively regulates surface biofilm while inhibiting pellicle biofilm formation. Results of real-time PCR suggest that the flagellar (fliC, flgK) and curli (csgA) genes were repressed in knockout strains, causing reduced surface biofilm. The mutants displayed altered pellicle biofilm architecture. They exhibited bacterial multilayers and a denser extracellular matrix with enhanced cellulose and less curli, ergo weaker pellicles than those of the wild type. The cellulose secretion was more in the knockout strains due to the upregulation of bcsC, which is necessary for cellulose export. We hypothesized that the secreted cellulose quickly integrates into the pellicle, leading to enhanced pellicular cellulose in the knockout strains. We determined that crp is upregulated in the knockout strains, thereby inhibiting the expression of csgD and, hence, also of csgA and bcsA. The conflicting upregulation of bcsC, the last gene of the bcsABZC operon, could be caused by independent regulation by the CRISPR-Cas system owing to a partial match between the CRISPR spacers and bcsC gene. The cAMP-regulated protein (CRP)-mediated regulation of the flagellar genes in the knockout strains was probably circumvented through the regulation of yddx governing the availability of the sigma factor σ28 that further regulates class 3 flagellar genes (fliC, fljB, and flgK). Additionally, the variations in the lipopolysaccharide (LPS) profile and expression of LPS-related genes (rfaC, rfbG, and rfbI) in knockout strains could also contribute to the altered pellicle architecture. Collectively, we establish that the CRISPR-Cas system differentially regulates the formation of surface-attached and pellicle biofilm. IMPORTANCE In addition to being implicated in bacterial immunity and genome editing, the CRISPR-Cas system has recently been demonstrated to regulate endogenous gene expression and biofilm formation. While the function of individual cas genes in controlling Salmonella biofilm has been explored, the regulatory role of CRISPR arrays in biofilm is less studied. Moreover, studies have focused on the effects of the CRISPR-Cas system on surface-associated biofilms, and comprehensive studies on the impact of the system on pellicle biofilm remain an unexplored niche. We demonstrate that the CRISPR array and cas genes modulate the expression of various biofilm genes in Salmonella, whereby surface and pellicle biofilm formation is distinctively regulated.
Collapse
|
44
|
Zhu J, Wang J, Chen YP, Qing T, Zhang P, Feng B. Quantitative proteomics and phosphoproteomics elucidate the molecular mechanism of nanostructured TiO 2-stimulated biofilm formation. JOURNAL OF HAZARDOUS MATERIALS 2022; 432:128709. [PMID: 35325859 DOI: 10.1016/j.jhazmat.2022.128709] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/08/2022] [Accepted: 03/12/2022] [Indexed: 06/14/2023]
Abstract
With the increasing concerns regarding bacterial adaption to nanomaterials, it is critical to explore the main mechanism behind the adaptive morphogenesis of microorganisms. In this work, the biofilms formed from activated sludge exposed to 5 and 50 mg/L nTiO2 in the dark had increased biomass and selectively enriched pathogens. To further elaborate adaptive mechanism of biofilm formation induced by nTiO2, the protein response and protein phosphorylation modification of Escherichia coli K12 were determined using integrative systems biology analyses of proteomics and phosphoproteomics. Results identified that E. coli cultivated with nTiO2 considerably upregulated iron acquisition, and regulated protein phosphorylation states associated with of transcription and translation and biofilm formation relative to unexposed controls. Accordingly, bacteria increased siderophores and exopolysaccharide content (increased by about 57% and 231%, respectively), and enhanced resistance to transcriptional inhibitory antibiotics. Moreover, a dose of an iron chelator, i.e., deferoxamine mesylate salt, effectively retarded the biofilm development of bacteria exposed to 50 mg/L nTiO2. Overall, this work will provide a new insight for biofouling control, and contribute to an improved understanding of microbial adaption to nanomaterials.
Collapse
Affiliation(s)
- Jing Zhu
- College of Environment and Resources, Xiangtan University, Xiangtan 411105, China
| | - Jingyu Wang
- College of Environment and Resources, Xiangtan University, Xiangtan 411105, China
| | - You-Peng Chen
- Key Laboratory of the Three Gorges Reservoir Region's Eco-Environments of MOE, Chongqing University, Chongqing 400045, China
| | - Taiping Qing
- College of Environment and Resources, Xiangtan University, Xiangtan 411105, China
| | - Peng Zhang
- College of Environment and Resources, Xiangtan University, Xiangtan 411105, China.
| | - Bo Feng
- College of Environment and Resources, Xiangtan University, Xiangtan 411105, China
| |
Collapse
|
45
|
Amini N, Milan PB, Sarmadi VH, Derakhshanmehr B, Hivechi A, Khodaei F, Hamidi M, Ashraf S, Larijani G, Rezapour A. Microorganism-derived biological macromolecules for tissue engineering. Front Med 2022; 16:358-377. [PMID: 35687278 DOI: 10.1007/s11684-021-0903-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/23/2021] [Indexed: 11/04/2022]
Abstract
According to literature, certain microorganism productions mediate biological effects. However, their beneficial characteristics remain unclear. Nowadays, scientists concentrate on obtaining natural materials from live creatures as new sources to produce innovative smart biomaterials for increasing tissue reconstruction in tissue engineering and regenerative medicine. The present review aims to introduce microorganism-derived biological macromolecules, such as pullulan, alginate, dextran, curdlan, and hyaluronic acid, and their available sources for tissue engineering. Growing evidence indicates that these materials can be used as biological material in scaffolds to enhance regeneration in damaged tissues and contribute to cosmetic and dermatological applications. These natural-based materials are attractive in pharmaceutical, regenerative medicine, and biomedical applications. This study provides a detailed overview of natural-based biomaterials, their chemical and physical properties, and new directions for future research and therapeutic applications.
Collapse
Affiliation(s)
- Naser Amini
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, 1591639675, Iran.,Institutes of Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Peiman Brouki Milan
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, 1591639675, Iran. .,Institutes of Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran. .,Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran.
| | - Vahid Hosseinpour Sarmadi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, 1591639675, Iran.,Institutes of Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Bahareh Derakhshanmehr
- Institutes of Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Ahmad Hivechi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, 1591639675, Iran.,Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Fateme Khodaei
- Burn Research Center, Department of Plastic and Reconstructive Surgery, Iran University of Medical Sciences, Tehran, 1591639675, Iran
| | - Masoud Hamidi
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, 4477166595, Iran
| | - Sara Ashraf
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, 1477893855, Iran
| | - Ghazaleh Larijani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, 1477893855, Iran
| | - Alireza Rezapour
- Cellular and Molecular Research Centre, Qom University of Medical Sciences, Qom, 3715835155, Iran. .,Department of Tissue Engineering and Regenerative Medicine, School of Medicine, Qom University of Medical Sciences, Qom, 3715835155, Iran.
| |
Collapse
|
46
|
Gaurivaud P, Tardy F. The Mycoplasma spp. ‘Releasome’: A New Concept for a Long-Known Phenomenon. Front Microbiol 2022; 13:853440. [PMID: 35495700 PMCID: PMC9051441 DOI: 10.3389/fmicb.2022.853440] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
The bacterial secretome comprises polypeptides expressed at the cell surface or released into the extracellular environment as well as the corresponding secretion machineries. Despite their reduced coding capacities, Mycoplasma spp. are able to produce and release several components into their environment, including polypeptides, exopolysaccharides and extracellular vesicles. Technical difficulties in purifying these elements from the complex broth media used to grow mycoplasmas have recently been overcome by optimizing growth conditions and switching to chemically defined culture media. However, the secretion pathways responsible for the release of these structurally varied elements are still poorly described in mycoplasmas. We propose the use of the term ‘releasome,’ instead of secretome, to refer to molecules released by mycoplasmas into their environment. The aim of this review is to more precisely delineate the elements that should be considered part of the mycoplasmal releasome and their role in the interplay of mycoplasmas with host cells and tissues.
Collapse
|
47
|
Tian L, Yan X, Wang D, Du Q, Wan Y, Zhou L, Li T, Liao C, Li N, Wang X. Two key Geobacter species of wastewater-enriched electroactive biofilm respond differently to electric field. WATER RESEARCH 2022; 213:118185. [PMID: 35183018 DOI: 10.1016/j.watres.2022.118185] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 06/14/2023]
Abstract
Electroactive biofilms have attracted increasing attention due to their unique ability to exchange electrons with electrodes. Geobacter spp. are widely found to be dominant in biofilms in acetate-rich environments when an appropriate voltage is applied, but it is still largely unknown how these bacteria are selectively enriched. Herein, two key Geobacter spp. that have been demonstrated predominant in wastewater-enriched electroactive biofilm after long-term operation, G. sulfurreducens and G. anodireducens, responded to electric field (EF) differently, leading to a higher abundance of EF-sensitive G. anodireducens in the strong EF region after cocultivation with G. sulfurreducens. Transcriptome analysis indicated that two-component systems containing sensor histidine kinases and response regulators were the key for EF sensing in G. anodireducens rather than in G. sulfurreducens, which are closely connected to chemotaxis, c-di-GMP, fatty acid metabolism, pilus, oxidative phosphorylation and transcription, resulting in an increase in extracellular polymeric substance secretion and rapid cell proliferation. Our data reveal the mechanism by which EF select specific Geobacter spp. over time, providing new insights into Geobacter biofilm formation regulated by electricity.
Collapse
Affiliation(s)
- Lili Tian
- MOE Key Laboratory of Pollution Processes and Environmental Criteria / Tianjin Key Laboratory of Environmental Remediation and Pollution Control / College of Environmental Science and Engineering, Nankai University, No. 38 Tongyan Road, Jinnan District, Tianjin 300350, China
| | - Xuejun Yan
- MOE Key Laboratory of Pollution Processes and Environmental Criteria / Tianjin Key Laboratory of Environmental Remediation and Pollution Control / College of Environmental Science and Engineering, Nankai University, No. 38 Tongyan Road, Jinnan District, Tianjin 300350, China
| | - Dongbin Wang
- School of Public Health, Guangdong Medical University, Xincheng Road, Dongguan 523000, China
| | - Qing Du
- School of Environmental and Municipal Engineering, Tianjin Chengjian University, Tianjin 300384, China
| | - Yuxuan Wan
- MOE Key Laboratory of Pollution Processes and Environmental Criteria / Tianjin Key Laboratory of Environmental Remediation and Pollution Control / College of Environmental Science and Engineering, Nankai University, No. 38 Tongyan Road, Jinnan District, Tianjin 300350, China
| | - Lean Zhou
- School of Hydraulic Engineering, Changsha University of Science and Technology, Changsha 410114, China
| | - Tian Li
- MOE Key Laboratory of Pollution Processes and Environmental Criteria / Tianjin Key Laboratory of Environmental Remediation and Pollution Control / College of Environmental Science and Engineering, Nankai University, No. 38 Tongyan Road, Jinnan District, Tianjin 300350, China
| | - Chengmei Liao
- MOE Key Laboratory of Pollution Processes and Environmental Criteria / Tianjin Key Laboratory of Environmental Remediation and Pollution Control / College of Environmental Science and Engineering, Nankai University, No. 38 Tongyan Road, Jinnan District, Tianjin 300350, China
| | - Nan Li
- School of Environmental Science and Engineering, Tianjin University, No. 35 Yaguan Road, Jinnan District, Tianjin 300350, China
| | - Xin Wang
- MOE Key Laboratory of Pollution Processes and Environmental Criteria / Tianjin Key Laboratory of Environmental Remediation and Pollution Control / College of Environmental Science and Engineering, Nankai University, No. 38 Tongyan Road, Jinnan District, Tianjin 300350, China.
| |
Collapse
|
48
|
Sun L, Cheng L, Ma Y, Lei P, Wang R, Gu Y, Li S, Zhang F, Xu H. Exopolysaccharides from Pantoea alhagi NX-11 specifically improve its root colonization and rice salt resistance. Int J Biol Macromol 2022; 209:396-404. [PMID: 35413311 DOI: 10.1016/j.ijbiomac.2022.04.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/31/2022] [Accepted: 04/03/2022] [Indexed: 12/13/2022]
Abstract
Plant growth-promoting rhizobacteria (PGPR) and their extracellular polymers such as exopolysaccharides can enhance rice salt stress resistance, however, the relevant mechanism remains unclear. In this study, an exopolysaccharides-deficient strain, named ΔpspD, was obtained from Pantoea alhagi NX-11 by chromosomal pspD deletion. The yield and characteristics of ΔpspD exopolysaccharides was obviously different from P. alhagi NX-11 exopolysaccharides (PAPS). Subsequently, hydroponic experiments showed that NX-11 or PAPS could enhance rice salt tolerance, but ΔpspD could not. Furthermore, it was found that PAPS promoted P. alhagi rhizosphere colonization through a direct effect on biofilm formation, as well as through an indirect impact of enhancing the abilities of biofilm formation and chemotaxis by altering rice root exudates. Importantly, the effect of PAPS in promoting the root colonization of NX-11 was specific. Through transcriptome and RT-qPCR analysis, we revealed that this specificity correlated with PAPS-induced lectin overexpression. The specificity between exopolysaccharides and the host microorganism ensures the colonization of the latter, and prevents other microorganisms from hitchhiking to the rice roots.
Collapse
Affiliation(s)
- Liang Sun
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Lifangyu Cheng
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Yuhang Ma
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Peng Lei
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China.
| | - Rui Wang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Yian Gu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China.
| | - Sha Li
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Fuhai Zhang
- Agricultural and Rural Bureau of Yantai, Yantai 264000, China
| | - Hong Xu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China.
| |
Collapse
|
49
|
Ashraf KU, Nygaard R, Vickery ON, Erramilli SK, Herrera CM, McConville TH, Petrou VI, Giacometti SI, Dufrisne MB, Nosol K, Zinkle AP, Graham CLB, Loukeris M, Kloss B, Skorupinska-Tudek K, Swiezewska E, Roper DI, Clarke OB, Uhlemann AC, Kossiakoff AA, Trent MS, Stansfeld PJ, Mancia F. Structural basis of lipopolysaccharide maturation by the O-antigen ligase. Nature 2022; 604:371-376. [PMID: 35388216 PMCID: PMC9884178 DOI: 10.1038/s41586-022-04555-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 02/16/2022] [Indexed: 01/31/2023]
Abstract
The outer membrane of Gram-negative bacteria has an external leaflet that is largely composed of lipopolysaccharide, which provides a selective permeation barrier, particularly against antimicrobials1. The final and crucial step in the biosynthesis of lipopolysaccharide is the addition of a species-dependent O-antigen to the lipid A core oligosaccharide, which is catalysed by the O-antigen ligase WaaL2. Here we present structures of WaaL from Cupriavidus metallidurans, both in the apo state and in complex with its lipid carrier undecaprenyl pyrophosphate, determined by single-particle cryo-electron microscopy. The structures reveal that WaaL comprises 12 transmembrane helices and a predominantly α-helical periplasmic region, which we show contains many of the conserved residues that are required for catalysis. We observe a conserved fold within the GT-C family of glycosyltransferases and hypothesize that they have a common mechanism for shuttling the undecaprenyl-based carrier to and from the active site. The structures, combined with genetic, biochemical, bioinformatics and molecular dynamics simulation experiments, offer molecular details on how the ligands come in apposition, and allows us to propose a mechanistic model for catalysis. Together, our work provides a structural basis for lipopolysaccharide maturation in a member of the GT-C superfamily of glycosyltransferases.
Collapse
Affiliation(s)
- Khuram U Ashraf
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | - Rie Nygaard
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | - Owen N Vickery
- School of Life Sciences, University of Warwick, Coventry, UK
- Department of Chemistry, University of Warwick, Coventry, UK
| | - Satchal K Erramilli
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Carmen M Herrera
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Thomas H McConville
- Department of Medicine, Division of Infectious Diseases, Columbia University Medical Center, New York, NY, USA
| | - Vasileios I Petrou
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers Biomedical Health Sciences, Newark, NJ, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers Biomedical Health Sciences, Newark, NJ, USA
| | - Sabrina I Giacometti
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | - Meagan Belcher Dufrisne
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | - Kamil Nosol
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Allen P Zinkle
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Michael Loukeris
- New York Consortium on Membrane Protein Structure, New York Structural Biology Center, New York, NY, USA
| | - Brian Kloss
- New York Consortium on Membrane Protein Structure, New York Structural Biology Center, New York, NY, USA
| | | | - Ewa Swiezewska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - David I Roper
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Oliver B Clarke
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Anne-Catrin Uhlemann
- Department of Medicine, Division of Infectious Diseases, Columbia University Medical Center, New York, NY, USA
| | - Anthony A Kossiakoff
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - M Stephen Trent
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA.
| | - Phillip J Stansfeld
- School of Life Sciences, University of Warwick, Coventry, UK.
- Department of Chemistry, University of Warwick, Coventry, UK.
| | - Filippo Mancia
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
50
|
Nguyen TT, Nguyen PT, Nguyen TBN, Bui NB, Nguyen HT. Efficacy of the incorporation between self-encapsulation and cryoprotectants on improving the freeze-dried survival of probiotic bacteria. J Appl Microbiol 2022; 132:3217-3225. [PMID: 35119770 DOI: 10.1111/jam.15473] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/01/2022] [Accepted: 02/01/2022] [Indexed: 11/27/2022]
Abstract
AIMS This study aimed to improve the viability of probiotic bacteria during freeze-drying by the combination of self-encapsulation and cryoprotectants. METHODS AND RESULTS Lactiplantibacillus plantarum VAL6 and Lactobacillus acidophilus VAR1 were exposed to environmental stresses including temperature, pH, and increased CO2 concentration before performing freeze-drying with the addition of cryoprotectants. The results proved that tested stresses can stimulate the bacteria to synthesize more extracellular polymeric substances to form self-encapsulation that increases their freeze-dried viability. In combination with cryoprotectants to form double-layered microencapsulation, L. plantarum VAL6 stressed at pH 3.5 in combination with whey protein isolate could achieve the highest Improving Cell Viability of 4,361 fold, while L. acidophilus VAR1 stressed at 25o C in combination with alginate gave a maximum Improving Cell Viability of 73.33 fold. CONCLUSIONS The combination of self-encapsulation and cryoprotectants significantly improves the freeze-dried viability of probiotics. SIGNIFICANCE AND IMPACT OF THE STUDY This is the first report that uses environmental stress to stimulate EPS synthesis for self-encapsulation formation combined with the addition of cryoprotectants to enhance to the freeze-dried survival of probiotics. This could be a novel approach in improving the viability of probiotic strains for various applications.
Collapse
Affiliation(s)
| | - Phu-Tho Nguyen
- Graduate University of Sciences and Technology, Vietnam Academy of Science and Technology, Hanoi, Vietnam.,Department of Biotechnology, An Giang University, Vietnam.,Vietnam National University Ho Chi Minh City, Vietnam
| | -
- Can Tho Science and Technology Application Center, Can Tho City, Vietnam
| | | | - Nhi-Binh Bui
- Can Tho Science and Technology Application Center, Can Tho City, Vietnam
| | - Huu-Thanh Nguyen
- Department of Biotechnology, An Giang University, Vietnam.,Vietnam National University Ho Chi Minh City, Vietnam
| |
Collapse
|