1
|
Cheng H, Li H, Li Z, Wang Y, Liu L, Wang J, Ma X, Tan B. The role of glycosylated mucins in maintaining intestinal homeostasis and gut health. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 21:439-446. [PMID: 40491555 PMCID: PMC12148640 DOI: 10.1016/j.aninu.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 01/12/2025] [Accepted: 03/12/2025] [Indexed: 06/11/2025]
Abstract
The intestinal mucus barrier is a crucial component of the host's innate defense system, playing a vital role in regulating intestinal microecology and maintaining intestinal homeostasis. Glycosylated mucins, the core components of this barrier, are essential for preserving its integrity by preventing bacterial degradation. Additionally, mucins significantly contribute to establishing a balanced symbiotic relationship between the host and microbes. These mucins have the potential to mitigate intestinal epithelial damage by capturing and transporting cell debris and pathogenic bacteria. Meanwhile, certain bacteria help maintain the equilibrium and stability of the gut microbiome by degrading glycosylated mucins to utilize the carbohydrate chains, thus affecting the cytokine expression to regulate the synthesis and secretion of specific glycans. Investigating the complex connections between the mucus barrier and mucin glycosylation holds great promise for advancing our understanding of gastrointestinal disease mechanisms, paving the way for innovative prevention and treatment strategies.
Collapse
Affiliation(s)
- Hao Cheng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan University of Arts and Science, Changde 415000, China
- Yuelushan Laboratory, Changsha 410128, China
| | - Hao Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan University of Arts and Science, Changde 415000, China
- Yuelushan Laboratory, Changsha 410128, China
| | - Zhong Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Yuelushan Laboratory, Changsha 410128, China
| | - Yun Wang
- Hunan University of Arts and Science, Changde 415000, China
| | - Liangguo Liu
- Hunan University of Arts and Science, Changde 415000, China
| | - Jing Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Yuelushan Laboratory, Changsha 410128, China
| | - Xiaokang Ma
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Yuelushan Laboratory, Changsha 410128, China
| | - Bie Tan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Yuelushan Laboratory, Changsha 410128, China
| |
Collapse
|
2
|
Mondry Cohen N, Krishna Kumar C, Iitoyo H, Rookyard AW, Cain JA, Man L, White MY, Dale AL, Cordwell SJ. Exploring the Targets of Reactive Oxygen Species and Defense against Oxidative Stress in Campylobacter jejuni Using a Multiomics Approach. J Proteome Res 2025. [PMID: 40426317 DOI: 10.1021/acs.jproteome.5c00182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2025]
Abstract
Campylobacter jejuni is a major cause of human gastroenteritis. Pathogenesis depends on survival in reactive oxygen species (ROS) that are produced endogenously and by host phagocytes and microbiota. Label-based proteomics by LC-MS/MS quantified 1347 proteins (83.0% of the predicted proteome) in response to hydrogen peroxide (10 μM/0.5 mM) and superoxide-inducing paraquat (PQ; 2 μM/10 μM). Antioxidants including catalase (KatA) and alkylhydroperoxide reductase (AhpC), were induced, consistent with the oxidative stress response. Changes to nutrient transporters (SdaC/PutP/LctP) correlated with the intracellular abundance of substrates (serine/proline/lactate). ROS significantly elevated the abundance of the outer membrane protein Cj1170c, and Δcj1170c bacteria were compromised for survival in H2O2 and under osmotic stress. PQ induced intracellular accumulation of threonine and homoserine, while Δcj1170c bacteria were depleted of these metabolites. ROS targets cysteine thiols that can be irreversibly modified to sulfinic and sulfonic (SO2H/SO3H) acids. We identified 1334 Cys-SO2H/SO3H-modified peptides (867 sites in 495 proteins) using SCX negative and HILIC positive selection coupled to LC-MS/MS. Many sites were modified without exogenous H2O2, suggesting that C. jejuni maintains an oxidative intracellular environment potentially related to microaerophilicity. Fe-S clusters were the primary targets of ROS. ROS trigger molecular remodeling associated with in-host growth, while overoxidizable Cys sites provide targets for redox-based antimicrobials.
Collapse
Affiliation(s)
- Nova Mondry Cohen
- School of Life and Environmental Sciences, The University of Sydney , Sydney2006, Australia
- Charles Perkins Centre, The University of Sydney , Sydney2006, Australia
| | - Chiranth Krishna Kumar
- School of Life and Environmental Sciences, The University of Sydney , Sydney2006, Australia
- Charles Perkins Centre, The University of Sydney , Sydney2006, Australia
| | - Haruta Iitoyo
- School of Life and Environmental Sciences, The University of Sydney , Sydney2006, Australia
- Charles Perkins Centre, The University of Sydney , Sydney2006, Australia
| | - Alexander W Rookyard
- School of Life and Environmental Sciences, The University of Sydney , Sydney2006, Australia
- Charles Perkins Centre, The University of Sydney , Sydney2006, Australia
- Sydney Mass Spectrometry, The University of Sydney , Sydney2006, Australia
| | - Joel A Cain
- School of Life and Environmental Sciences, The University of Sydney , Sydney2006, Australia
- Charles Perkins Centre, The University of Sydney , Sydney2006, Australia
| | - Lok Man
- School of Life and Environmental Sciences, The University of Sydney , Sydney2006, Australia
- Charles Perkins Centre, The University of Sydney , Sydney2006, Australia
| | - Melanie Y White
- Charles Perkins Centre, The University of Sydney , Sydney2006, Australia
- School of Medical Sciences, The University of Sydney , Sydney2006, Australia
| | - Ashleigh L Dale
- School of Life and Environmental Sciences, The University of Sydney , Sydney2006, Australia
- Charles Perkins Centre, The University of Sydney , Sydney2006, Australia
- Sydney Mass Spectrometry, The University of Sydney , Sydney2006, Australia
| | - Stuart J Cordwell
- School of Life and Environmental Sciences, The University of Sydney , Sydney2006, Australia
- Charles Perkins Centre, The University of Sydney , Sydney2006, Australia
- Sydney Mass Spectrometry, The University of Sydney , Sydney2006, Australia
- School of Medical Sciences, The University of Sydney , Sydney2006, Australia
| |
Collapse
|
3
|
Zhou B, Garber JM, Butcher J, Muszynski A, Casey RL, Huynh S, Archer-hartmann S, Porfírio S, Rogers AM, Azadi P, Parker CT, Ng KKS, Hines KM, Stintzi A, Szymanski CM. Campylobacter jejuni resistance to human milk involves the acyl carrier protein AcpP. mBio 2025; 16:e0399724. [PMID: 39998218 PMCID: PMC11980577 DOI: 10.1128/mbio.03997-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Campylobacter jejuni is a common foodborne pathogen worldwide that is associated with high rates of morbidity and mortality among infants in low- to middle-income countries (LMICs). Human milk provides infants with an important source of nutrients and contains antimicrobial components for protection against infection. However, recent studies, including our own, have found significantly higher levels of Campylobacter in diarrheal stool samples collected from breastfed infants compared to non-breastfed infants in LMICs. We hypothesized that C. jejuni has unique strategies to resist the antimicrobial properties of human milk. Transcriptional profiling found human milk exposure induces genes associated with ribosomal function, iron acquisition, and amino acid utilization in C. jejuni strains 81-176 and 11168. However, unidentified proteinaceous components of human milk prevent bacterial growth. Evolving both C. jejuni isolates to survive in human milk resulted in mutations in genes encoding the acyl carrier protein (AcpP) and the major outer membrane porin (PorA). Introduction of the PorA/AcpP amino acid changes into the parental backgrounds followed by electron microscopy showed distinct membrane architectures, and the AcpP changes not only significantly improved growth in human milk, but also yielded cells surrounded with outer membrane vesicles. Analyses of the phospholipid and lipooligosaccharide (LOS) compositions suggest an imbalance in acyl chain distributions. For strain 11168, these changes protect both evolved and 11168∆acpPG33R strains from bacteriophage infection and polymyxin killing. Taken together, this study provides insights into how C. jejuni may evolve to resist the bactericidal activity of human milk and flourish in the hostile environment of the gastrointestinal tract. IMPORTANCE In this study, we evolved C. jejuni strains which can grow in the presence of human milk and found that cell membrane alterations may be involved in resistance to the antimicrobial properties of human milk. These bacterial membrane changes are predominantly linked to amino acid substitutions within the acyl carrier protein, AcpP, although other bacterial components, including PorA, are likely involved. This study provides some insights into possible strategies for C. jejuni survival and propagation in the gastrointestinal tract of breastfed infants.
Collapse
Affiliation(s)
- Bibi Zhou
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Jolene M. Garber
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - James Butcher
- School of Pharmaceutical Sciences, Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Artur Muszynski
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Rebekah L. Casey
- Department of Chemistry, University of Georgia, Athens, Georgia, USA
| | - Steven Huynh
- Produce Safety and Microbiology Research Unit, Agricultural Research Service, U.S. Department of Agriculture, Albany, California, USA
| | | | - Sara Porfírio
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Ashley M. Rogers
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Craig T. Parker
- Produce Safety and Microbiology Research Unit, Agricultural Research Service, U.S. Department of Agriculture, Albany, California, USA
| | - Kenneth K. S. Ng
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - Kelly M. Hines
- Department of Chemistry, University of Georgia, Athens, Georgia, USA
| | - Alain Stintzi
- School of Pharmaceutical Sciences, Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Christine M. Szymanski
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
4
|
Ghorashi AC, Boucher A, Archer-Hartmann SA, Zalem D, Taherzadeh Ghahfarrokhi M, Murray NB, Konada RSR, Zhang X, Xing C, Teneberg S, Azadi P, Yrlid U, Kohler JJ. Fucosylation of glycoproteins and glycolipids: opposing roles in cholera intoxication. Nat Chem Biol 2025; 21:555-566. [PMID: 39414978 DOI: 10.1038/s41589-024-01748-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/13/2024] [Indexed: 10/18/2024]
Abstract
Cholera toxin (CT) is the etiological agent of cholera. Here we report that multiple classes of fucosylated glycoconjugates function in CT binding and intoxication of intestinal epithelial cells. In Colo205 cells, knockout (KO) of B3GNT5, which encodes an enzyme required for synthesis of lacto and neolacto series glycosphingolipids (GSLs), reduces CT binding but sensitizes cells to intoxication. Overexpressing B3GNT5 to generate more fucosylated GSLs confers protection against intoxication, indicating that fucosylated GSLs act as decoy receptors for CT. KO of B3GALT5 causes increased production of fucosylated O-linked and N-linked glycoproteins and leads to increased CT binding and intoxication. KO of B3GNT5 in B3GALT5-KO cells eliminates production of fucosylated GSLs but increases intoxication, identifying fucosylated glycoproteins as functional receptors for CT. These findings provide insight into the molecular determinants regulating CT sensitivity of host cells.
Collapse
Affiliation(s)
- Atossa C Ghorashi
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Andrew Boucher
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | | | - Dani Zalem
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | | | - Nathan B Murray
- Complex Carbohydrate Research Center, The University of Georgia, Athens, GA, USA
| | | | - Xunzhi Zhang
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA
| | - Chao Xing
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Susann Teneberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, The University of Georgia, Athens, GA, USA
| | - Ulf Yrlid
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Jennifer J Kohler
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
5
|
Abolhasani FS, Vaghefinanekaran N, Yarahmadi A, Akrami S, Mirmahdavi S, Yousefi MH, Afkhami H, Shafiei M. Outer membrane vesicles in gram-negative bacteria and its correlation with pathogenesis. Front Immunol 2025; 16:1541636. [PMID: 40236702 PMCID: PMC11996793 DOI: 10.3389/fimmu.2025.1541636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 03/11/2025] [Indexed: 04/17/2025] Open
Abstract
There is a widespread distribution of gram-negative bacteria worldwide, which are responsible for the deaths of numerous patients each year. The illnesses they cause can be localized and systemic, and these bacteria possess several key virulence factors that contribute to their pathogenicity. In recent years, several distinct mechanisms of pathogenesis have evolved that remain largely unknown to scientists and medical experts. Among these, outer membrane vesicles (OMVs) are undoubtedly one of the most significant factors influencing virulence. OMVs contain various bacterial compounds and can have diverse effects on host organisms and the immune system, potentially exacerbating disease and inflammation while evading immune responses. This review comprehensively examines the role of OMVs in bacterial pathogenesis, their interaction with host cells, and their potential biomedical applications. Understanding the molecular mechanisms governing OMV biogenesis and function could pave the way for novel antimicrobial strategies and therapeutic interventions.
Collapse
Affiliation(s)
- Fatemeh Sadat Abolhasani
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| | - Sousan Akrami
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Solmaz Mirmahdavi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Microbiology, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Hasan Yousefi
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Hamed Afkhami
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, School of Medicine, Shahed University, Tehran, Iran
| | - Morvarid Shafiei
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
6
|
Dessenne C, Mariller C, Vidal O, Huvent I, Guerardel Y, Elass-Rochard E, Rossez Y. Glycan-mediated adhesion mechanisms in antibiotic-resistant bacteria. BBA ADVANCES 2025; 7:100156. [PMID: 40207210 PMCID: PMC11979486 DOI: 10.1016/j.bbadva.2025.100156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 04/11/2025] Open
Abstract
Bacterial adhesins play a central role in host-pathogen interactions, with many specifically targeting glycans to mediate bacterial colonization, influence infection dynamics, and evade host immune responses. In this review, we focus on bacterial pathogens identified by the World Health Organization as critical threats to public health and in urgent need of new treatments. We summarize glycoconjugate targets identified in the literature across 19 bacterial genera and species. This comprehensive review provides a foundation for the development of innovative therapeutic strategies to effectively combat these pathogens.
Collapse
Affiliation(s)
- Clara Dessenne
- Université Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Christophe Mariller
- Université Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Olivier Vidal
- Université Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Isabelle Huvent
- Université Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Yann Guerardel
- Université Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| | - Elisabeth Elass-Rochard
- Université Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Yannick Rossez
- Université Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| |
Collapse
|
7
|
Middendorf PS, Zomer AL, Bergval IL, Jacobs-Reitsma WF, den Besten HMW, Abee T. Host associations of Campylobacter jejuni and Campylobacter coli isolates carrying the L-fucose or d-glucose utilization cluster. Int J Food Microbiol 2024; 425:110855. [PMID: 39191191 DOI: 10.1016/j.ijfoodmicro.2024.110855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 07/06/2024] [Accepted: 08/03/2024] [Indexed: 08/29/2024]
Abstract
Campylobacter was considered asaccharolytic, but is now known to carry saccharide metabolization pathways for L-fucose and d-glucose. We hypothesized that these clusters are beneficial for Campylobacter niche adaptation and may help establish human infection. We investigated the distribution of d-glucose and L-fucose clusters among ∼9600 C. jejuni and C. coli genomes of different isolation sources in the Netherlands, the United Kingdom, the United States of America and Finland. The L-fucose utilization cluster was integrated at the same location in all C. jejuni and C. coli genomes, and was flanked by the genes rpoB, rpoC, rspL, repsG and fusA, which are associated with functions in transcription as well as translation and in acquired drug resistance. In contrast, the flanking regions of the d-glucose utilization cluster were variable among the isolates, and integration sites were located within one of the three different 16S23S ribosomal RNA areas of the C. jejuni and C. coli genomes. In addition, we investigated whether acquisition of the L-fucose utilization cluster could be due to horizontal gene transfer between the two species and found three isolates for which this was the case: one C. jejuni isolate carrying a C. coli L-fucose cluster, and two C. coli isolates which carried a C. jejuni L-fucose cluster. Furthermore, L-fucose utilization cluster alignments revealed multiple frameshift mutations, most of which were commonly found in the non-essential genes for L-fucose metabolism, namely, Cj0484 and Cj0489. These findings support our hypothesis that the L-fucose cluster was integrated multiple times across the C. coli/C. jejuni phylogeny. Notably, association analysis using the C. jejuni isolates from the Netherlands showed a significant correlation between human C. jejuni isolates and C. jejuni isolates carrying the L-fucose utilization cluster. This correlation was even stronger when the Dutch isolates were combined with the isolates from the UK, the USA and Finland. No such correlations were observed for C. coli or for the d-glucose cluster for both species. This research provides insight into the spread and host associations of the L-fucose and d-glucose utilization clusters in C. jejuni and C. coli, and the potential benefits in human infection and/or proliferation in humans, conceivably after transmission from any reservoir.
Collapse
Affiliation(s)
- Pjotr S Middendorf
- Food Microbiology, Wageningen University and Research, Wageningen, Netherlands; National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Aldert L Zomer
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, Netherlands; WHO Collaborating Center for Campylobacter/OIE Reference Laboratory for Campylobacteriosis, Utrecht, Netherlands
| | - Indra L Bergval
- National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | | | | | - Tjakko Abee
- Food Microbiology, Wageningen University and Research, Wageningen, Netherlands.
| |
Collapse
|
8
|
Middendorf PS, Wijnands LM, Boeren S, Zomer AL, Jacobs-Reitsma WF, den Besten HM, Abee T. Activation of the l-fucose utilization cluster in Campylobacter jejuni induces proteomic changes and enhances Caco-2 cell invasion and fibronectin binding. Heliyon 2024; 10:e34996. [PMID: 39220920 PMCID: PMC11365321 DOI: 10.1016/j.heliyon.2024.e34996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 07/16/2024] [Accepted: 07/21/2024] [Indexed: 09/04/2024] Open
Abstract
Most Campylobacter jejuni isolates carry the fucose utilization cluster (Cj0480c-Cj0489) that supports the metabolism of l-fucose and d-arabinose. In this study we quantified l-fucose and d-arabinose metabolism and metabolite production, and the impact on Caco-2 cell interaction and binding to fibronectin, using C. jejuni NCTC11168 and the closely related human isolate C. jejuni strain 286. When cultured with l-fucose and d-arabinose, both isolates showed increased survival and production of acetate, pyruvate and succinate, and the respective signature metabolites lactate and glycolic acid, in line with an overall upregulation of l-fucose cluster proteins. In vitro Caco-2 cell studies and fibronectin-binding experiments showed a trend towards higher invasion and a significantly higher fibronectin binding efficacy of C. jejuni NCTC11168 cells grown with l-fucose and d-arabinose, while no significant differences were found with C. jejuni 286. Both fibronectin binding proteins, CadF and FlpA, were detected in the two isolates, but were not significantly differentially expressed in l-fucose or d-arabinose grown cells. Comparative proteomics analysis linked the C. jejuni NCTC11168 phenotypes uniquely to the more than 135-fold upregulated protein Cj0608, putative TolC-like component MacC, which, together with the detected Cj0606 and Cj0607 proteins, forms the tripartite secretion system MacABC with putative functions in antibiotic resistance, cell envelope stress response and virulence in Gram negative pathogenic bacteria. Further studies are required to elucidate the role of the MacABC system in C. jejuni cell surface structure modulation and virulence.
Collapse
Affiliation(s)
- Pjotr S. Middendorf
- Food Microbiology, Wageningen University and Research, Wageningen, the Netherlands
- National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Lucas M. Wijnands
- National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University and Research, Wageningen, the Netherlands
| | - Aldert L. Zomer
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, the Netherlands
- WHO Collaborating Center for Campylobacter/OIE Reference Laboratory for Campylobacteriosis, Utrecht, the Netherlands
| | | | | | - Tjakko Abee
- Food Microbiology, Wageningen University and Research, Wageningen, the Netherlands
| |
Collapse
|
9
|
Man L, Soh PXY, McEnearney TE, Cain JA, Dale AL, Cordwell SJ. Multi-Omics of Campylobacter jejuni Growth in Chicken Exudate Reveals Molecular Remodelling Associated with Altered Virulence and Survival Phenotypes. Microorganisms 2024; 12:860. [PMID: 38792690 PMCID: PMC11123243 DOI: 10.3390/microorganisms12050860] [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: 03/21/2024] [Revised: 04/15/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Campylobacter jejuni is the leading cause of foodborne human gastroenteritis in the developed world. Infections are largely acquired from poultry produced for human consumption and poor food handling is thus a major risk factor. Chicken exudate (CE) is a liquid produced from defrosted commercial chicken products that facilitates C. jejuni growth. We examined the response of C. jejuni to growth in CE using a multi-omics approach. Changes in the C. jejuni proteome were assessed by label-based liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS). We quantified 1328 and 1304 proteins, respectively, in experiments comparing 5% CE in Mueller-Hinton (MH) medium and 100% CE with MH-only controls. These proteins represent 81.8% and 80.3% of the predicted C. jejuni NCTC11168 proteome. Growth in CE induced profound remodelling of the proteome. These changes were typically conserved between 5% and 100% CE, with a greater magnitude of change observed in 100% CE. We confirmed that CE induced C. jejuni biofilm formation, as well as increasing motility and resistance against oxidative stress, consistent with changes to proteins representing those functions. Assessment of the C. jejuni metabolome showed CE also led to increased intracellular abundances of serine, proline, and lactate that were correlated with the elevated abundances of their respective transporters. Analysis of carbon source uptake showed prolonged culture supernatant retention of proline and succinate in CE-supplemented medium. Metabolomics data provided preliminary evidence for the uptake of chicken-meat-associated dipeptides. C. jejuni exposed to CE showed increased resistance to several antibiotics, including polymyxin B, consistent with changes to tripartite efflux system proteins and those involved in the synthesis of lipid A. The C. jejuni CE proteome was also characterised by very large increases in proteins associated with iron acquisition, while a decrease in proteins containing iron-sulphur clusters was also observed. Our data suggest CE is both oxygen- and iron-limiting and provide evidence of factors required for phenotypic remodelling to enable C. jejuni survival on poultry products.
Collapse
Affiliation(s)
- Lok Man
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Pamela X. Y. Soh
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Tess E. McEnearney
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Joel A. Cain
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Ashleigh L. Dale
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Stuart J. Cordwell
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
- Sydney Mass Spectrometry, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
10
|
Smallman TR, Perlaza-Jiménez L, Wang X, Korman TM, Kotsanas D, Gibson JS, Turni C, Harper M, Boyce JD. Pathogenomic analysis and characterization of Pasteurella multocida strains recovered from human infections. Microbiol Spectr 2024; 12:e0380523. [PMID: 38426766 PMCID: PMC10986470 DOI: 10.1128/spectrum.03805-23] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/03/2024] [Indexed: 03/02/2024] Open
Abstract
Pasteurella multocida is an upper respiratory tract commensal in several mammal and bird species but can also cause severe disease in humans and in production animals such as poultry, cattle, and pigs. In this study, we performed whole-genome sequencing of P. multocida isolates recovered from a range of human infections, from the mouths of cats, and from wounds on dogs. Together with publicly available P. multocida genome sequences, we performed phylogenetic and comparative genomic analyses. While isolates from cats and dogs were spread across the phylogenetic tree, human infections were caused almost exclusively by subsp. septica strains. Most of the human isolates were capsule type A and LPS type L1 and L3; however, some strains lacked a capsule biosynthesis locus, and some strains contained a novel LPS outer-core locus, distinct from the eight LPS loci that can currently be identified using an LPS multiplex PCR. In addition, the P. multocida strains isolated from human infections contained novel mobile genetic elements. We compiled a curated database of known P. multocida virulence factor and antibiotic resistance genes (PastyVRDB) allowing for detailed characterization of isolates. The majority of human P. multocida isolates encoded a reduced range of iron receptors and contained only one filamentous hemagglutinin gene. Finally, gene-trait analysis identified a putative L-fucose uptake and utilization pathway that was over-represented in subsp. septica strains and may represent a novel host predilection mechanism in this subspecies. Together, these analyses have identified pathogenic mechanisms likely important for P. multocida zoonotic infections.IMPORTANCEPasteurella multocida can cause serious infections in humans, including skin and wound infections, pneumonia, peritonitis, meningitis, and bacteraemia. Cats and dogs are known vectors of human pasteurellosis, transmitting P. multocida via bite wounds or contact with animal saliva. The mechanisms that underpin P. multocida human predilection and pathogenesis are poorly understood. With increasing identification of antibiotic-resistant P. multocida strains, understanding these mechanisms is vital for developing novel treatments and control strategies to combat P. multocida human infection. Here, we show that a narrow range of P. multocida strains cause disease in humans, while cats and dogs, common vectors for zoonotic infections, can harbor a wide range of P. multocida strains. We also present a curated P. multocida-specific database, allowing quick and detailed characterization of newly sequenced P. multocida isolates.
Collapse
Affiliation(s)
- Thomas R. Smallman
- Department of Microbiology, Infection Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Laura Perlaza-Jiménez
- Monash Bioinformatics Platform, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Xiaochu Wang
- Department of Microbiology, Infection Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Tony M. Korman
- Monash University and Monash Health, Clayton, Victoria, Australia
| | - Despina Kotsanas
- Monash University and Monash Health, Clayton, Victoria, Australia
| | - Justine S. Gibson
- School of Veterinary Science, University of Queensland, Gatton, Queensland, Australia
| | - Conny Turni
- Queensland Alliance for Agriculture and Food Innovation, University of Queensland, St Lucia, Queensland, Australia
| | - Marina Harper
- Department of Microbiology, Infection Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - John D. Boyce
- Department of Microbiology, Infection Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
11
|
Al Hakeem WG, Cason EE, Adams D, Fathima S, Shanmugasundaram R, Lourenco J, Selvaraj RK. Characterizing the Effect of Campylobacter jejuni Challenge on Growth Performance, Cecal Microbiota, and Cecal Short-Chain Fatty Acid Concentrations in Broilers. Animals (Basel) 2024; 14:473. [PMID: 38338116 PMCID: PMC10854649 DOI: 10.3390/ani14030473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/20/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
This study aimed to understand the effect of C. jejuni challenge on the cecal microbiota and short-chain fatty acid (SCFA) concentration to form a better understanding of the host-pathogen interaction. Sixty broilers were randomly allocated into two treatments: control and challenge. Each treatment was replicated in six pens with five birds per pen. On day 21, birds in the challenge group were orally gavaged with 1 × 108C. jejuni/mL, while the control group was mock challenged with PBS. The C. jejuni challenge had no effect on body weight, feed intake, and feed conversion ratio compared to the control group. On day 28, the C. jejuni challenge decreased the observed features and Shannon index compared to the control group. On the species level, the C. jejuni challenge decreased (p = 0.02) the relative abundance of Sellimonas intestinalis on day 28 and increased (p = 0.04) the relative abundance of Faecalibacterium sp002160895 on day 35 compared to the control group. The C. jejuni challenge did not change the microbial function and the cecal concentrations of SCFA on days 28 and 35 compared to the control group. In conclusion, C. jejuni might alter the gut microbiota's composition and diversity without significantly compromising broilers' growth.
Collapse
Affiliation(s)
- Walid G. Al Hakeem
- Department of Poultry Science, The University of Georgia, Athens, GA 3060, USA; (W.G.A.H.); (E.E.C.); (D.A.); (S.F.)
| | - Emily E. Cason
- Department of Poultry Science, The University of Georgia, Athens, GA 3060, USA; (W.G.A.H.); (E.E.C.); (D.A.); (S.F.)
| | - Daniel Adams
- Department of Poultry Science, The University of Georgia, Athens, GA 3060, USA; (W.G.A.H.); (E.E.C.); (D.A.); (S.F.)
| | - Shahna Fathima
- Department of Poultry Science, The University of Georgia, Athens, GA 3060, USA; (W.G.A.H.); (E.E.C.); (D.A.); (S.F.)
| | - Revathi Shanmugasundaram
- Toxicology and Mycotoxin Research Unit, US National Poultry Research Center, Athens, GA 30605, USA;
| | - Jeferson Lourenco
- Department of Animal and Dairy Science, The University of Georgia, Athens, GA 30602, USA
| | - Ramesh K. Selvaraj
- Department of Poultry Science, The University of Georgia, Athens, GA 3060, USA; (W.G.A.H.); (E.E.C.); (D.A.); (S.F.)
| |
Collapse
|
12
|
Sinha R, LeVeque RM, Callahan SM, Chatterjee S, Stopnisek N, Kuipel M, Johnson JG, DiRita VJ. Gut metabolite L-lactate supports Campylobacter jejuni population expansion during acute infection. Proc Natl Acad Sci U S A 2024; 121:e2316540120. [PMID: 38170751 PMCID: PMC10786315 DOI: 10.1073/pnas.2316540120] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/15/2023] [Indexed: 01/05/2024] Open
Abstract
How the microaerobic pathogen Campylobacter jejuni establishes its niche and expands in the gut lumen during infection is poorly understood. Using 6-wk-old ferrets as a natural disease model, we examined this aspect of C. jejuni pathogenicity. Unlike mice, which require significant genetic or physiological manipulation to become colonized with C. jejuni, ferrets are readily infected without the need to disarm the immune system or alter the gut microbiota. Disease after C. jejuni infection in ferrets reflects closely how human C. jejuni infection proceeds. Rapid growth of C. jejuni and associated intestinal inflammation was observed within 2 to 3 d of infection. We observed pathophysiological changes that were noted by cryptic hyperplasia through the induction of tissue repair systems, accumulation of undifferentiated amplifying cells on the colon surface, and instability of HIF-1α in colonocytes, which indicated increased epithelial oxygenation. Metabolomic analysis demonstrated that lactate levels in colon content were elevated in infected animals. A C. jejuni mutant lacking lctP, which encodes an L-lactate transporter, was significantly decreased for colonization during infection. Lactate also influences adhesion and invasion by C. jejuni to a colon carcinoma cell line (HCT116). The oxygenation required for expression of lactate transporter (lctP) led to identification of a putative thiol-based redox switch regulator (LctR) that may repress lctP transcription under anaerobic conditions. Our work provides better insights into the pathogenicity of C. jejuni.
Collapse
Affiliation(s)
- Ritam Sinha
- Department of Microbiology, Genetics, & Immunology, Michigan State University, East Lansing, MI48824
| | - Rhiannon M. LeVeque
- Department of Microbiology, Genetics, & Immunology, Michigan State University, East Lansing, MI48824
| | - Sean M. Callahan
- Department of Microbiology, University of Tennessee, Knoxville, TN37996
| | - Shramana Chatterjee
- Department of Microbiology, Genetics, & Immunology, Michigan State University, East Lansing, MI48824
| | - Nejc Stopnisek
- Department of Microbiology, Genetics, & Immunology, Michigan State University, East Lansing, MI48824
| | - Matti Kuipel
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI48824
| | | | - Victor J. DiRita
- Department of Microbiology, Genetics, & Immunology, Michigan State University, East Lansing, MI48824
| |
Collapse
|
13
|
Zhou B, Garber JM, Vlach J, Azadi P, Ng KKS, Escalante-Semerena JC, Szymanski CM. Campylobacter jejuni uses energy taxis and a dehydrogenase enzyme for l-fucose chemotaxis. mBio 2023; 14:e0273223. [PMID: 38032212 PMCID: PMC10746189 DOI: 10.1128/mbio.02732-23] [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: 10/11/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
IMPORTANCE In this study, we identify a separate role for the Campylobacter jejuni l-fucose dehydrogenase in l-fucose chemotaxis and demonstrate that this mechanism is not only limited to C. jejuni but is also present in Burkholderia multivorans. We now hypothesize that l-fucose energy taxis may contribute to the reduction of l-fucose-metabolizing strains of C. jejuni from the gastrointestinal tract of breastfed infants, selecting for isolates with increased colonization potential.
Collapse
Affiliation(s)
- Bibi Zhou
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Jolene M. Garber
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Jiri Vlach
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Kenneth K. S. Ng
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | | | - Christine M. Szymanski
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
14
|
Zangara MT, Darwish L, Coombes BK. Characterizing the Pathogenic Potential of Crohn's Disease-Associated Adherent-Invasive Escherichia coli. EcoSal Plus 2023; 11:eesp00182022. [PMID: 37220071 PMCID: PMC10729932 DOI: 10.1128/ecosalplus.esp-0018-2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/04/2023] [Indexed: 01/28/2024]
Abstract
The microbiome of Crohn's disease (CD) patients is composed of a microbial community that is considered dysbiotic and proinflammatory in nature. The overrepresentation of Enterobacteriaceae species is a common feature of the CD microbiome, and much attention has been given to understanding the pathogenic role this feature plays in disease activity. Over 2 decades ago, a new Escherichia coli subtype called adherent-invasive E. coli (AIEC) was isolated and linked to ileal Crohn's disease. Since the isolation of the first AIEC strain, additional AIEC strains have been isolated from both inflammatory bowel disease (IBD) patients and non-IBD individuals using the original in vitro phenotypic characterization methods. Identification of a definitive molecular marker of the AIEC pathotype has been elusive; however, significant advancements have been made in understanding the genetic, metabolic, and virulence determinants of AIEC infection biology. Here, we review the current knowledge of AIEC pathogenesis to provide additional, objective measures that could be considered in defining AIEC and their pathogenic potential.
Collapse
Affiliation(s)
- Megan T. Zangara
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Lena Darwish
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Brian K. Coombes
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, Hamilton, Ontario, Canada
| |
Collapse
|
15
|
Sinha R, LeVeque RM, Callahan SM, Chatterjee S, Stopnisek N, Kuipel M, Johnson JG, DiRita VJ. Gut metabolite L-lactate supports Campylobacter jejuni population expansion during acute infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.02.560557. [PMID: 37873437 PMCID: PMC10592923 DOI: 10.1101/2023.10.02.560557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
How the microaerobic pathogen Campylobacter jejuni establishes its niche and expands in the gut lumen during infection is poorly understood. Using six-week-old ferrets as a natural disease model, we examined this aspect of C. jejuni pathogenicity. Unlike mice, which require significant genetic or physiological manipulation to become colonized with C. jejuni , ferrets are readily infected without the need to disarm the immune system or alter the gut microbiota. Disease after C. jejuni infection in ferrets reflects closely how human C. jejuni infection proceeds. Rapid growth of C. jejuni and associated intestinal inflammation was observed within two-three days of infection. We observed pathophysiological changes that were noted by cryptic hyperplasia through the induction of tissue repair systems, accumulation of undifferentiated amplifying cells on the colon surface, and instability of HIF-1α in colonocytes, which indicated increased epithelial oxygenation. Metabolomic analysis demonstrated that lactate levels in colon content were elevated in infected animals. A C. jejuni mutant lacking lctP , which encodes an L-lactate transporter, was significantly decreased for colonization during infection. Lactate also influences adhesion and invasion by C. jejuni to a colon carcinoma cell line (HCT116). The oxygenation required for expression of lactate transporter ( lctP ) led to discovery of a putative thiol based redox switch regulator (LctR) that may repress lctP transcription under anaerobic conditions. Our work provides new insights into the pathogenicity of C. jejuni . Significance There is a gap in knowledge about the mechanisms by which C. jejuni populations expand during infection. Using an animal model which accurately reflects human infection without the need to alter the host microbiome or the immune system prior to infection, we explored pathophysiological alterations of the gut after C. jejuni infection. Our study identified the gut metabolite L-lactate as playing an important role as a growth substrate for C. jejuni during acute infection. We identified a DNA binding protein, LctR, that binds to the lctP promoter and may repress lctP expression, resulting in decreased lactate transport under low oxygen levels. This work provides new insights about C. jejuni pathogenicity.
Collapse
|
16
|
Ghorashi AC, Boucher A, Archer-Hartmann SA, Murray NB, Konada RSR, Zhang X, Xing C, Azadi P, Yrlid U, Kohler JJ. Fucosylated glycoproteins and fucosylated glycolipids play opposing roles in cholera intoxication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.02.551727. [PMID: 37577488 PMCID: PMC10418270 DOI: 10.1101/2023.08.02.551727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Cholera toxin (CT) is the etiological agent of cholera. Here we report that multiple classes of fucosylated glycoconjugates function in CT binding and intoxication of intestinal epithelial cells. In Colo205 cells, knockout of B3GNT5, the enzyme required for synthesis of lacto- and neolacto-series glycosphingolipids (GSLs), reduces CT binding but sensitizes cells to intoxication. Overexpressing B3GNT5 to generate more fucosylated GSLs confers protection against intoxication, indicating that fucosylated GSLs act as decoy receptors for CT. Knockout (KO) of B3GALT5 causes increased production of fucosylated O-linked and N-linked glycoproteins, and leads to increased CT binding and intoxication. Knockout of B3GNT5 in B3GALT5 KO cells eliminates production of fucosylated GSLs but increases intoxication, identifying fucosylated glycoproteins as functional receptors for CT. These findings provide insight into molecular determinants regulating CT sensitivity of host cells.
Collapse
Affiliation(s)
- Atossa C. Ghorashi
- Department of Biochemistry, UT Southwestern Medical Center, Dallas TX 75390 USA
| | - Andrew Boucher
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | | | - Nathan B. Murray
- Complex Carbohydrate Research Center, The University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | | | - Xunzhi Zhang
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas TX 75390 USA
| | - Chao Xing
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas TX 75390 USA
- Department of Bioinformatics, UT Southwestern Medical Center, Dallas TX 75390 USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, The University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - Ulf Yrlid
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Jennifer J. Kohler
- Department of Biochemistry, UT Southwestern Medical Center, Dallas TX 75390 USA
| |
Collapse
|
17
|
Kim J, Jin YS, Kim KH. L-Fucose is involved in human-gut microbiome interactions. Appl Microbiol Biotechnol 2023:10.1007/s00253-023-12527-y. [PMID: 37148338 DOI: 10.1007/s00253-023-12527-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/30/2023] [Accepted: 04/10/2023] [Indexed: 05/08/2023]
Abstract
L-Fucose is one of the key metabolites in human-gut microbiome interactions. It is continuously synthesized by humans in the form of fucosylated glycans and fucosyl-oligosaccharides and delivered into the gut throughout their lifetime. Gut microorganisms metabolize L-fucose and produce short-chain fatty acids, which are absorbed by epithelial cells and used as energy sources or signaling molecules. Recent studies have revealed that the carbon flux in L-fucose metabolism by gut microorganisms is distinct from that in other sugar metabolisms because of cofactor imbalance and low efficiencies in energy synthesis of L-fucose metabolism. The large amounts of short-chain fatty acids produced during microbial L-fucose metabolism are used by epithelial cells to recover most of the energy used up during L-fucose synthesis. In this review, we present a detailed overview of microbial L-fucose metabolism and a potential solution for disease treatment and prevention using genetically engineered probiotics that modulate fucose metabolism. Our review contributes to the understanding of human-gut microbiome interactions through L-fucose metabolism. KEY POINTS: • Fucose-metabolizing microorganisms produce large amounts of short-chain fatty acids • Fucose metabolism differs from other sugar metabolisms by cofactor imbalance • Modulating fucose metabolism is the key to control host-gut microbiome interactions.
Collapse
Affiliation(s)
- Jungyeon Kim
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yong-Su Jin
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| | - Kyoung Heon Kim
- Department of Biotechnology, Graduate School, Korea University, Seoul, 02841, Republic of Korea.
- Department of Food Bioscience and Technology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
18
|
Zhou B, Szymanski CM, Baylink A. Bacterial chemotaxis in human diseases. Trends Microbiol 2023; 31:453-467. [PMID: 36411201 PMCID: PMC11238666 DOI: 10.1016/j.tim.2022.10.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/16/2022] [Accepted: 10/18/2022] [Indexed: 11/21/2022]
Abstract
To infect and cause disease, bacterial pathogens must localize to specific regions of the host where they possess the metabolic and defensive acumen for survival. Motile flagellated pathogens exercise control over their localization through chemotaxis to direct motility based on the landscape of exogenous nutrients, toxins, and molecular cues sensed within the host. Here, we review advances in understanding the roles chemotaxis plays in human diseases. Chemotaxis drives pathogen colonization to sites of inflammation and injury and mediates fitness advantages through accessing host-derived nutrients from damaged tissue. Injury tropism may worsen clinical outcomes through instigating chronic inflammation and subsequent cancer development. Inhibiting bacterial chemotactic systems could act synergistically with antibacterial medicines for more effective and specific eradication.
Collapse
Affiliation(s)
- Bibi Zhou
- University of Georgia, Department of Microbiology and Complex Carbohydrate Research Center, Athens, GA 30602, USA
| | - Christine M Szymanski
- University of Georgia, Department of Microbiology and Complex Carbohydrate Research Center, Athens, GA 30602, USA
| | - Arden Baylink
- Washington State University, Department of Veterinary Microbiology and Pathology, Pullman, WA 99164, USA.
| |
Collapse
|
19
|
Al Hakeem WG, Acevedo Villanueva KY, Selvaraj RK. The Development of Gut Microbiota and Its Changes Following C. jejuni Infection in Broilers. Vaccines (Basel) 2023; 11:595. [PMID: 36992178 PMCID: PMC10056385 DOI: 10.3390/vaccines11030595] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/22/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
The gut is home to more than millions of bacterial species. The gut bacteria coexist with the host in a symbiotic relationship that can influence the host's metabolism, nutrition, and physiology and even module various immune functions. The commensal gut microbiota plays a crucial role in shaping the immune response and provides a continuous stimulus to maintain an activated immune system. The recent advancements in high throughput omics technologies have improved our understanding of the role of commensal bacteria in developing the immune system in chickens. Chicken meat continues to be one of the most consumed sources of protein worldwide, with the demand expected to increase significantly by the year 2050. Yet, chickens are a significant reservoir for human foodborne pathogens such as Campylobacter jejuni. Understanding the interaction between the commensal bacteria and C. jejuni is essential in developing novel technologies to decrease C. jejuni load in broilers. This review aims to provide current knowledge of gut microbiota development and its interaction with the immune system in broilers. Additionally, the effect of C. jejuni infection on the gut microbiota is addressed.
Collapse
Affiliation(s)
| | | | - Ramesh K. Selvaraj
- Department of Poultry Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
20
|
Structure and function of microbial α-l-fucosidases: a mini review. Essays Biochem 2023; 67:399-414. [PMID: 36805644 PMCID: PMC10154630 DOI: 10.1042/ebc20220158] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 02/23/2023]
Abstract
Fucose is a monosaccharide commonly found in mammalian, insect, microbial and plant glycans. The removal of terminal α-l-fucosyl residues from oligosaccharides and glycoconjugates is catalysed by α-l-fucosidases. To date, glycoside hydrolases (GHs) with exo-fucosidase activity on α-l-fucosylated substrates (EC 3.2.1.51, EC 3.2.1.-) have been reported in the GH29, GH95, GH139, GH141 and GH151 families of the Carbohydrate Active Enzymes (CAZy) database. Microbes generally encode several fucosidases in their genomes, often from more than one GH family, reflecting the high diversity of naturally occuring fucosylated structures they encounter. Functionally characterised microbial α-l-fucosidases have been shown to act on a range of substrates with α-1,2, α-1,3, α-1,4 or α-1,6 fucosylated linkages depending on the GH family and microorganism. Fucosidases show a modular organisation with catalytic domains of GH29 and GH151 displaying a (β/α)8-barrel fold while GH95 and GH141 show a (α/α)6 barrel and parallel β-helix fold, respectively. A number of crystal structures have been solved in complex with ligands, providing structural basis for their substrate specificity. Fucosidases can also be used in transglycosylation reactions to synthesise oligosaccharides. This mini review provides an overview of the enzymatic and structural properties of microbial α-l-fucosidases and some insights into their biological function and biotechnological applications.
Collapse
|
21
|
Yan A, Butcher J, Schramm L, Mack DR, Stintzi A. Multiomic spatial analysis reveals a distinct mucosa-associated virome. Gut Microbes 2023; 15:2177488. [PMID: 36823020 PMCID: PMC9980608 DOI: 10.1080/19490976.2023.2177488] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
The human gut virome has been increasingly explored in recent years. However, nearly all virome-sequencing efforts rely solely on fecal samples and few studies leverage multiomic approaches to investigate phage-host relationships. Here, we combine metagenomics, metaviromics, and metatranscriptomics to study virome-bacteriome interactions at the colonic mucosal-luminal interface in a cohort of three individuals with inflammatory bowel disease; non-IBD controls were not included in this study. We show that the mucosal viral population is distinct from the stool virome and houses abundant crAss-like phages that are undetectable by fecal sampling. Through viral protein prediction and metatranscriptomic analysis, we explore viral gene transcription, prophage activation, and the relationship between the presence of integrase and temperate phages in IBD subjects. We also show the impact of deep sequencing on virus recovery and offer guidelines for selecting optimal sequencing depths in future metaviromic studies. Systems biology approaches such as those presented in this report will enhance our understanding of the human virome and its interactions with our microbiome and our health.
Collapse
Affiliation(s)
- Austin Yan
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - James Butcher
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Laetitia Schramm
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - David R. Mack
- Department of Pediatrics, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada,Inflammatory Bowel Disease Centre and CHEO Research Institute, Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Alain Stintzi
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada,CONTACT Alain Stintzi Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| |
Collapse
|
22
|
Deblais L, Jang H, Kauffman M, Gangiredla J, Sawyer M, Basa S, Poelstra JW, Babu US, Harrison LM, Hiett KL, Balan KV, Rajashekara G. Whole genome characterization of thermophilic Campylobacter species isolated from dairy manure in small specialty crop farms of Northeast Ohio. Front Microbiol 2023; 14:1074548. [PMID: 37025625 PMCID: PMC10071015 DOI: 10.3389/fmicb.2023.1074548] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/30/2023] [Indexed: 04/08/2023] Open
Abstract
Introduction With more public interest in consuming locally grown produce, small specialty crop farms (SSCF) are a viable and growing segment of the food production chain in the United States. Methods The goal of this study was to investigate the genomic diversity of Campylobacter isolated from dairy manure (n = 69) collected from 10 SSCF in Northeast Ohio between 2018 and 2020. Results A total of 56 C. jejuni and 13 C. coli isolates were sequenced. Multi-locus sequence typing (MLST) identified 22 sequence types (STs), with ST-922 (18%) and ST-61 (13%) predominant in C. jejuni and ST-829 (62%) and ST-1068 (38%) predominant in C. coli. Interestingly, isolates with similar genomic and gene contents were detected within and between SSCF over time, suggesting that Campylobacter could be transmitted between farms and may persist in a given SSCF over time. Virulence-associated genes (n = 35) involved in the uptake and utilization of potassium and organic compounds (succinate, gluconate, oxoglutarate, and malate) were detected only in the C. jejuni isolates, while 45 genes associated with increased resistance to environmental stresses (capsule production, cell envelope integrity, and iron uptake) were detected only in the C. coli isolates. Campylobacter coli isolates were also sub-divided into two distinct clusters based on the presence of unique prophages (n = 21) or IncQ conjugative plasmid/type-IV secretion system genes (n = 15). Campylobacter coli isolates harbored genes associated with resistance to streptomycin (aadE-Cc; 54%) and quinolone (gyrA-T86I; 77%), while C. jejuni had resistance genes for kanamycin (aph3'-IIIa; 20%). Both species harbored resistance genes associated with β-lactam (especially, blaOXA-193; up to 100%) and tetracycline (tetO; up to 59%). Discussion/Conclusion Our study demonstrated that Campylobacter genome plasticity associated with conjugative transfer might provide resistance to certain antimicrobials and viral infections via the acquisition of protein-encoding genes involved in mechanisms such as ribosomal protection and capsule modification.
Collapse
Affiliation(s)
- Loic Deblais
- Department of Animal Sciences, Center for Food Animal Health, The Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - Hyein Jang
- Center for Food Safety and Applied Nutrition (CFSAN), Office of Applied Research and Safety Assessment (OARSA), U.S. Food and Drug Administration, Laurel, MD, United States
| | - Mike Kauffman
- Department of Animal Sciences, Center for Food Animal Health, The Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - Jayanthi Gangiredla
- Center for Food Safety and Applied Nutrition (CFSAN), Office of Applied Research and Safety Assessment (OARSA), U.S. Food and Drug Administration, Laurel, MD, United States
| | - Marianne Sawyer
- Center for Food Safety and Applied Nutrition (CFSAN), Office of Applied Research and Safety Assessment (OARSA), U.S. Food and Drug Administration, Laurel, MD, United States
| | - Saritha Basa
- Center for Food Safety and Applied Nutrition (CFSAN), Office of Applied Research and Safety Assessment (OARSA), U.S. Food and Drug Administration, Laurel, MD, United States
| | - Jelmer W. Poelstra
- Molecular and Cellular Imaging Center, The Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - Uma S. Babu
- Center for Food Safety and Applied Nutrition (CFSAN), Office of Applied Research and Safety Assessment (OARSA), U.S. Food and Drug Administration, Laurel, MD, United States
| | - Lisa M. Harrison
- Center for Food Safety and Applied Nutrition (CFSAN), Office of Applied Research and Safety Assessment (OARSA), U.S. Food and Drug Administration, Laurel, MD, United States
| | - Kelli L. Hiett
- Center for Food Safety and Applied Nutrition (CFSAN), Office of Applied Research and Safety Assessment (OARSA), U.S. Food and Drug Administration, Laurel, MD, United States
| | - Kannan V. Balan
- Center for Food Safety and Applied Nutrition (CFSAN), Office of Applied Research and Safety Assessment (OARSA), U.S. Food and Drug Administration, Laurel, MD, United States
| | - Gireesh Rajashekara
- Department of Animal Sciences, Center for Food Animal Health, The Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
- *Correspondence: Gireesh Rajashekara,
| |
Collapse
|
23
|
Taha, Elgamoudi BA, Andrianova EP, Haselhorst T, Day CJ, Hartley-Tassell LE, King RM, Najnin T, Zhulin IB, Korolik V. Diverse Sensory Repertoire of Paralogous Chemoreceptors Tlp2, Tlp3, and Tlp4 in Campylobacter jejuni. Microbiol Spectr 2022; 10:e0364622. [PMID: 36374080 PMCID: PMC9769880 DOI: 10.1128/spectrum.03646-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022] Open
Abstract
Campylobacter jejuni responds to extracellular stimuli via transducer-like chemoreceptors (Tlps). Here, we describe receptor-ligand interactions of a unique paralogue family of dCache_1 (double Calcium channels and chemotaxis) chemoreceptors: Tlp2, Tlp3, and Tlp4. Phylogenetic analysis revealed that Tlp2, Tlp3, and Tlp4 receptors may have arisen through domain duplications, followed by a divergent evolutionary drift, with Tlp3 emerging more recently, and unexpectedly, responded to glycans, as well as multiple organic and amino acids with overlapping specificities. All three Tlps interacted with five monosaccharides and complex glycans, including Lewis's antigens, P antigens, and fucosyl GM1 ganglioside, indicating a potential role in host-pathogen interactions. Analysis of chemotactic motility of single, double, and triple mutants indicated that these chemoreceptors are likely to work together to balance responses to attractants and repellents to modulate chemotaxis in C. jejuni. Molecular docking experiments, in combination with saturation transfer difference nuclear magnetic resonance spectroscopy and competition surface plasmon resonance analysis, illustrated that the ligand-binding domain of Tlp3 possess one major binding pocket with two overlapping, but distinct binding sites able to interact with multiple ligands. A diverse sensory repertoire could provide C. jejuni with the ability to modulate responses to attractant and repellent signals and allow for adaptation in host-pathogen interactions. IMPORTANCE Campylobacter jejuni responds to extracellular stimuli via transducer-like chemoreceptors (Tlps). This remarkable sensory perception mechanism allows bacteria to sense environmental changes and avoid unfavorable conditions or to maneuver toward nutrient sources and host cells. Here, we describe receptor-ligand interactions of a unique paralogue family of chemoreceptors, Tlp2, Tlp3, and Tlp4, that may have arisen through domain duplications, followed by a divergent evolutionary drift, with Tlp3 emerging more recently. Unlike previous reports of ligands interacting with sensory proteins, Tlp2, Tlp3, and Tlp4 responded to many types of chemical compounds, including simple and complex sugars such as those present on human blood group antigens and gangliosides, indicating a potential role in host-pathogen interactions. Diverse sensory repertoire could provide C. jejuni with the ability to modulate responses to attractant and repellent signals and allow for adaptation in host-pathogen interactions.
Collapse
Affiliation(s)
- Taha
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Bassam A. Elgamoudi
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Ekaterina P. Andrianova
- Department of Microbiology and Translational Data Analytics Institute, The Ohio State University, Columbus, Ohio, USA
| | - Thomas Haselhorst
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Christopher J. Day
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | | | - Rebecca M. King
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Tahria Najnin
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Igor B. Zhulin
- Department of Microbiology and Translational Data Analytics Institute, The Ohio State University, Columbus, Ohio, USA
| | - Victoria Korolik
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
- School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
24
|
Cheong YE, Kim J, Jin YS, Kim KH. Elucidation of the fucose metabolism of probiotic Lactobacillus rhamnosus GG by metabolomic and flux balance analyses. J Biotechnol 2022; 360:110-116. [PMID: 36336085 DOI: 10.1016/j.jbiotec.2022.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 10/08/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
Abstract
Lactobacillus rhamnosus GG (LGG) is one of the most widely used probiotics because of its health benefits and safety. Fucose is among the most abundant hexoses in the human intestine, and LGG consumes fucose to produce energy or proliferate. However, no study has elucidated the metabolism by which LGG metabolizes fucose to produce energy, biomass, and extracellular metabolites. We used metabolomics and flux balance analysis to elucidate these mechanisms and highlight how they might affect the host. We found three different metabolic flux modes by which LGG anaerobically metabolizes fucose to produce energy and biomass. These metabolic flux modes differ from homolactic or heterolactic fermentation and account for the production of lactic acid, 1,2-propanediol, acetic acid, formic acid, and carbon dioxide as a result of fucose metabolism in LGG. We also used gas chromatography/time-of-flight mass spectrometry to identify a variety of short-chain fatty acids and organic acids secreted during fucose metabolism by LGG. Our study is the first to elucidate the unique fucose metabolism of LGG in anaerobic condition.
Collapse
Affiliation(s)
- Yu Eun Cheong
- Department of Biotechnology, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Jungyeon Kim
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Yong-Su Jin
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Kyoung Heon Kim
- Department of Biotechnology, Graduate School, Korea University, Seoul 02841, Republic of Korea; Department of Food Bioscience and Technology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
25
|
Kim J, Cheong YE, Yu S, Jin YS, Kim KH. Strain engineering and metabolic flux analysis of a probiotic yeast Saccharomyces boulardii for metabolizing L-fucose, a mammalian mucin component. Microb Cell Fact 2022; 21:204. [PMID: 36207743 PMCID: PMC9541068 DOI: 10.1186/s12934-022-01926-x] [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] [Received: 08/08/2022] [Accepted: 09/19/2022] [Indexed: 11/24/2022] Open
Abstract
Background Saccharomyces boulardii is a probiotic yeast that exhibits antimicrobial and anti-toxin activities. Although S. boulardii has been clinically used for decades to treat gastrointestinal disorders, several studies have reported weak or no beneficial effects of S. boulardii administration in some cases. These conflicting results of S. boulardii efficacity may be due to nutrient deficiencies in the intestine that make it difficult for S. boulardii to maintain its metabolic activity. Results To enable S. boulardii to overcome any nutritional deficiencies in the intestine, we constructed a S. boulardii strain that could metabolize l-fucose, a major component of mucin in the gut epithelium. The fucU, fucI, fucK, and fucA from Escherichia coli and HXT4 from S. cerevisiae were overexpressed in S. boulardii. The engineered S. boulardii metabolized l-fucose and produced 1,2-propanediol under aerobic and anaerobic conditions. It also produced large amounts of 1,2-propanediol under strict anaerobic conditions. An in silico genome-scale metabolic model analysis was performed to simulate the growth of S. boulardii on l-fucose, and elementary flux modes were calculated to identify critical metabolic reactions for assimilating l-fucose. As a result, we found that the engineered S. boulardii consumes l-fucose via (S)-lactaldehyde-(S)-lactate-pyruvate pathway, which is highly oxygen dependent. Conclusion To the best of our knowledge, this is the first study in which S. cerevisiae and S. boulardii strains capable of metabolizing l-fucose have been constructed. This strategy could be used to enhance the metabolic activity of S. boulardii and other probiotic microorganisms in the gut. Supplementary Information The online version contains supplementary material available at 10.1186/s12934-022-01926-x.
Collapse
Affiliation(s)
- Jungyeon Kim
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.,Department of Biotechnology, Graduate School, Korea University, Seoul, 02841, Republic of Korea
| | - Yu Eun Cheong
- Department of Biotechnology, Graduate School, Korea University, Seoul, 02841, Republic of Korea
| | - Sora Yu
- Department of Biotechnology, Graduate School, Korea University, Seoul, 02841, Republic of Korea
| | - Yong-Su Jin
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA. .,Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| | - Kyoung Heon Kim
- Department of Biotechnology, Graduate School, Korea University, Seoul, 02841, Republic of Korea. .,Department of Food Bioscience and Technology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
26
|
Arzamasov AA, Osterman AL. Milk glycan metabolism by intestinal bifidobacteria: insights from comparative genomics. Crit Rev Biochem Mol Biol 2022; 57:562-584. [PMID: 36866565 PMCID: PMC10192226 DOI: 10.1080/10409238.2023.2182272] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 01/11/2023] [Accepted: 02/15/2023] [Indexed: 03/04/2023]
Abstract
Bifidobacteria are early colonizers of the human neonatal gut and provide multiple health benefits to the infant, including inhibiting the growth of enteropathogens and modulating the immune system. Certain Bifidobacterium species prevail in the gut of breastfed infants due to the ability of these microorganisms to selectively forage glycans present in human milk, specifically human milk oligosaccharides (HMOs) and N-linked glycans. Therefore, these carbohydrates serve as promising prebiotic dietary supplements to stimulate the growth of bifidobacteria in the guts of children suffering from impaired gut microbiota development. However, the rational formulation of milk glycan-based prebiotics requires a detailed understanding of how bifidobacteria metabolize these carbohydrates. Accumulating biochemical and genomic data suggest that HMO and N-glycan assimilation abilities vary remarkably within the Bifidobacterium genus, both at the species and strain levels. This review focuses on the delineation and genome-based comparative analysis of differences in respective biochemical pathways, transport systems, and associated transcriptional regulatory networks, providing a foundation for genomics-based projection of milk glycan utilization capabilities across a rapidly growing number of sequenced bifidobacterial genomes and metagenomic datasets. This analysis also highlights remaining knowledge gaps and suggests directions for future studies to optimize the formulation of milk-glycan-based prebiotics that target bifidobacteria.
Collapse
Affiliation(s)
- Aleksandr A Arzamasov
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Andrei L Osterman
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
27
|
Szymanski CM. Bacterial glycosylation, it’s complicated. Front Mol Biosci 2022; 9:1015771. [PMID: 36250013 PMCID: PMC9561416 DOI: 10.3389/fmolb.2022.1015771] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Each microbe has the ability to produce a wide variety of sugar structures that includes some combination of glycolipids, glycoproteins, exopolysaccharides and oligosaccharides. For example, bacteria may synthesize lipooligosaccharides or lipopolysaccharides, teichoic and lipoteichoic acids, N- and O-linked glycoproteins, capsular polysaccharides, exopolysaccharides, poly-N-acetylglycosamine polymers, peptidoglycans, osmoregulated periplasmic glucans, trehalose or glycogen, just to name a few of the more broadly distributed carbohydrates that have been studied. The composition of many of these glycans are typically dissimilar from those described in eukaryotes, both in the seemingly endless repertoire of sugars that microbes are capable of synthesizing, and in the unique modifications that are attached to the carbohydrate residues. Furthermore, strain-to-strain differences in the carbohydrate building blocks used to create these glycoconjugates are the norm, and many strains possess additional mechanisms for turning on and off transferases that add specific monosaccharides and/or modifications, exponentially contributing to the structural heterogeneity observed by a single isolate, and preventing any structural generalization at the species level. In the past, a greater proportion of research effort was directed toward characterizing human pathogens rather than commensals or environmental isolates, and historically, the focus was on microbes that were simple to grow in large quantities and straightforward to genetically manipulate. These studies have revealed the complexity that exists among individual strains and have formed a foundation to better understand how other microbes, hosts and environments further transform the glycan composition of a single isolate. These studies also motivate researchers to further explore microbial glycan diversity, particularly as more sensitive analytical instruments and methods are developed to examine microbial populations in situ rather than in large scale from an enriched nutrient flask. This review emphasizes many of these points using the common foodborne pathogen Campylobacter jejuni as the model microbe.
Collapse
|
28
|
Wang S, Ma M, Liang Z, Zhu X, Yao H, Wang L, Wu Z. Pathogenic investigations of Streptococcus pasteurianus, an underreported zoonotic pathogen, isolated from a diseased piglet with meningitis. Transbound Emerg Dis 2022; 69:2609-2620. [PMID: 34871467 DOI: 10.1111/tbed.14413] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/28/2021] [Accepted: 11/28/2021] [Indexed: 01/14/2023]
Abstract
Streptococcus pasteurianus, an underreported opportunistic pathogen, is considered an increasingly recognized cause of meningitis and bacteremia in many animals and humans worldwide. However, except for some epidemiological studies, there is no report about the gene-deletion mutagenesis, virulence factors, reservoir niches or animal infection models for this pathogen. In this study, we first isolated an S. pasteurianus strain from a newly weaned piglet's brain with meningitis. The genomic sequence of this swine isolate WUSP067 shared high homology with that of two human strains. The comparative genome analysis showed that strain WUSP067 contained a fucose utilization cluster absent in human strains, and it shared 91% identity with that of an integrative and conjugative element (ICE) ICEssuZJ20091101-2 from Streptococcus suis, another important swine bacterial pathogen. Strain WUSP067 was resistant to erythromycin, tulathromycin, lincomycin, clindamycin, doxycycline and gentamycin, and ICEs are vehicles for harbouring antimicrobial resistance genes. The infection model was established using the 3-week-old newly weaned ICR mice. The 50% lethal dose value of strain WUSP067 was 4.0 × 107 colony-forming units per mouse. The infected mice showed severe signs of meningitis and pathological changes in brains. Furthermore, the capsule-deficient mutant was generated using natural transformation, and we showed that capsule was an essential virulence factor for S. pasteurianus. In addition, we found that tonsils and hilar lymph nodes of healthy pigs may be reservoir niches for this bacterium. Thus, our study provided valuable information about the pathogenetic characteristics and antimicrobial resistance of S. pasteurianus and paved the way for studying its pathogenesis.
Collapse
Affiliation(s)
- Shuoyue Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing, China
| | - Miaohang Ma
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing, China
| | - Zijing Liang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing, China
| | - Xinchi Zhu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing, China
| | - Huochun Yao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing, China
| | - Liping Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Zongfu Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing, China
| |
Collapse
|
29
|
Rath A, Rautenschlein S, Rzeznitzeck J, Lalk M, Methling K, Rychlik I, Peh E, Kittler S, Waldmann KH, von Altrock A. Investigation on the colonisation of Campylobacter strains in the pig intestine depending on available metabolites. Comp Immunol Microbiol Infect Dis 2022; 88:101865. [PMID: 35914481 DOI: 10.1016/j.cimid.2022.101865] [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: 02/02/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 10/16/2022]
Abstract
Campylobacter (C.) spp. represent one of the most important causes for food-borne bacterial pathogen in humans worldwide. The aim of this study was to investigate metabolic requirements of two Campylobacter strains of different species based on substrate utilisation (in vitro). Based on these results, a correlation between the colonisation and the available substrates in different intestinal sections was recorded using an animal model. Campylobacter coli (ST-5777) and C. jejuni (ST-122) were used to inoculate 16 pigs, respectively, and one group of 16 pigs was used as control. The strains differed significantly in substrate utilisation - C. coli was able to metabolise various substrates (acetate, asparagine, serine, fucose, and propionate), while C. jejuni only utilised serine. Metabolomic analysis of intestinal content from different gut sections showed the presence of all previously tested metabolites, except for fucose. A significantly larger amount of glucose was found in the jejunum of those pigs infected with C. coli, while neither strain utilised it in vitro. The analysis of the intestinal contents revealed a very low proportion of Campylobacterales in the total microbiome, suggesting that the small percentage of the inoculated Campylobacter strains in the gut microflora of the animals is too low to cause differences between the control and infected groups in the composition of the metabolome. Nevertheless, knowledge of specific nutritional requirements of the pathogens combined with proof of different metabolites in the intestinal segments may provide clues about the site of colonisation in the host and improve our understanding of this zoonotic germ.
Collapse
Affiliation(s)
- Alexandra Rath
- Clinic for Swine and Small Ruminants, Forensic Medicine and Ambulatory Service, University of Veterinary Medicine Hannover, Foundation, 30173 Hannover, Germany.
| | - Silke Rautenschlein
- Clinic for Poultry, University of Veterinary Medicine Hannover Foundation, 30559 Hannover, Germany
| | - Janina Rzeznitzeck
- Clinic for Poultry, University of Veterinary Medicine Hannover Foundation, 30559 Hannover, Germany
| | - Michael Lalk
- Institute for Pharmaceutical Biology, University of Greifswald, Greifswald, Germany
| | - Karen Methling
- Institute for Pharmaceutical Biology, University of Greifswald, Greifswald, Germany
| | - Ivan Rychlik
- Veterinary Research Institute, Brno, Czech Republic
| | - Elisa Peh
- Institute for Food Quality and Food Safety, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Sophie Kittler
- Institute for Food Quality and Food Safety, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Karl-Heinz Waldmann
- Clinic for Swine and Small Ruminants, Forensic Medicine and Ambulatory Service, University of Veterinary Medicine Hannover, Foundation, 30173 Hannover, Germany
| | - Alexandra von Altrock
- Clinic for Swine and Small Ruminants, Forensic Medicine and Ambulatory Service, University of Veterinary Medicine Hannover, Foundation, 30173 Hannover, Germany
| |
Collapse
|
30
|
Stoakes E, Savva GM, Coates R, Tejera N, Poolman MG, Grant AJ, Wain J, Singh D. Substrate Utilisation and Energy Metabolism in Non-Growing Campylobacter jejuni M1cam. Microorganisms 2022; 10:1355. [PMID: 35889074 PMCID: PMC9318392 DOI: 10.3390/microorganisms10071355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/23/2022] [Accepted: 06/30/2022] [Indexed: 02/04/2023] Open
Abstract
Campylobacter jejuni, the major cause of bacterial foodborne illness, is also a fastidious organism that requires strict growth requirements in the laboratory. Our aim was to study substrate utilisation and energy metabolism in non-growing C. jejuni to investigate the ability of these bacteria to survive so effectively in the food chain. We integrated phenotypic microarrays and genome-scale metabolic modelling (GSM) to investigate the survival of C. jejuni on 95 substrates. We further investigated the underlying metabolic re-adjustment associated with varying energy demands on each substrate. We identified amino acids, organic acids and H2, as single substrates supporting survival without growth. We identified several different mechanisms, which were used alone or in combination, for ATP production: substrate-level phosphorylation via acetate kinase, the TCA cycle, and oxidative phosphorylation via the electron transport chain that utilised alternative electron donors and acceptors. The benefit of ATP production through each of these mechanisms was associated with the cost of enzyme investment, nutrient availability and/or O2 utilisation. C. jejuni can utilise a wide range of substrates as energy sources, including organic acids commonly used for marination or preservation of ingredients, which might contribute to the success of their survival in changing environments.
Collapse
Affiliation(s)
- Emily Stoakes
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK; (E.S.); (R.C.); (A.J.G.)
| | - George M. Savva
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK; (G.M.S.); (N.T.)
| | - Ruby Coates
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK; (E.S.); (R.C.); (A.J.G.)
| | - Noemi Tejera
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK; (G.M.S.); (N.T.)
| | - Mark G. Poolman
- Cell System Modelling Group, Oxford Brookes University, Oxford OX3 0BP, UK;
| | - Andrew J. Grant
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK; (E.S.); (R.C.); (A.J.G.)
| | - John Wain
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK; (G.M.S.); (N.T.)
| | - Dipali Singh
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK; (G.M.S.); (N.T.)
| |
Collapse
|
31
|
Xiao M, Ren X, Cui J, Li R, Liu Z, Zhu L, Kong Q, Fu X, Mou H. A novel glucofucobiose with potential prebiotic activity prepared from the exopolysaccharides of Clavibacter michiganensis M1. Food Chem 2022; 377:132001. [PMID: 34999464 DOI: 10.1016/j.foodchem.2021.132001] [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] [Received: 09/18/2021] [Revised: 11/14/2021] [Accepted: 11/18/2021] [Indexed: 11/04/2022]
Abstract
Fucose and fucosylated oligosaccharides have important applications in various industries owing to their prebiotic, anti-inflammatory, anticoagulant, and antiviral activities. Here, we aimed to obtain fucosylated oligosaccharides using the acidolysis method to depolymerize exopolysaccharides extracted from Clavibacter michiganensis M1. Based on structural analysis, the prepared glucofucobiose was found to consist of d-glucose and l-fucose, with a molecular weight of 326 Da and a structure of d-Glcp-β-(1→4)-l-Fucp. The prebiotic activity of glucofucobiose was compared with that of 2'-fucosyllactose (2'-FL), the most abundant oligosaccharide in human milk. According to the results, glucofucobiose could significantly promote the proliferation of six probiotic strains, and short-chain fatty acid production of five probiotic strains on glucofucobiose was substantially higher than that on 2'-FL at 48 h of fermentation. Overall, this study proposed a new technology for obtaining fucosylated oligosaccharides. The prepared glucofucobiose was found to exhibit potential prebiotic activity and should be further assessed.
Collapse
Affiliation(s)
- Mengshi Xiao
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong, People's Republic of China.
| | - Xinmiao Ren
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong, People's Republic of China.
| | - Jinzheng Cui
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, Shandong, People's Republic of China.
| | - Rong Li
- Qingdao Women and Children Hospital, Qingdao 266003, Shandong, People's Republic of China.
| | - Zhemin Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong, People's Republic of China.
| | - Lin Zhu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong, People's Republic of China; Weihai Deepsea Biotechnology Co., Ltd, Weihai 264300, Shandong, People's Republic of China.
| | - Qing Kong
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong, People's Republic of China.
| | - Xiaodan Fu
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi 330047, People's Republic of China.
| | - Haijin Mou
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong, People's Republic of China.
| |
Collapse
|
32
|
Middendorf PS, Jacobs-Reitsma WF, Zomer AL, den Besten HMW, Abee T. Comparative Analysis of L-Fucose Utilization and Its Impact on Growth and Survival of Campylobacter Isolates. Front Microbiol 2022; 13:872207. [PMID: 35572645 PMCID: PMC9100392 DOI: 10.3389/fmicb.2022.872207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/31/2022] [Indexed: 12/04/2022] Open
Abstract
Campylobacter jejuni and Campylobacter coli were previously considered asaccharolytic, but are now known to possess specific saccharide metabolization pathways, including L-fucose. To investigate the influence of the L-fucose utilization cluster on Campylobacter growth, survival and metabolism, we performed comparative genotyping and phenotyping of the C. jejuni reference isolate NCTC11168 (human isolate), C. jejuni Ca1352 (chicken meat isolate), C. jejuni Ca2426 (sheep manure isolate), and C. coli Ca0121 (pig manure isolate), that all possess the L-fucose utilization cluster. All isolates showed enhanced survival and prolonged spiral cell morphology in aging cultures up to day seven in L-fucose-enriched MEMα medium (MEMαF) compared to MEMα. HPLC analysis indicated L-fucose utilization linked to acetate, lactate, pyruvate and succinate production, confirming the activation of the L-fucose pathway in these isolates and its impact on general metabolism. Highest consumption of L-fucose by C. coli Ca0121 is conceivably linked to its enhanced growth performance up to day 7, reaching 9.3 log CFU/ml compared to approximately 8.3 log CFU/ml for the C. jejuni isolates. Genetic analysis of the respective L-fucose clusters revealed several differences, including a 1 bp deletion in the Cj0489 gene of C. jejuni NCTC11168, causing a frameshift in this isolate resulting in two separate genes, Cj0489 and Cj0490, while no apparent phenotype could be linked to the presumed frameshift in this isolate. Additionally, we found that the L-fucose cluster of C. coli Ca0121 was most distant from C. jejuni NCTC11168, but confirmation of links to L-fucose metabolism associated phenotypic traits in C. coli versus C. jejuni isolates requires further studies.
Collapse
Affiliation(s)
- Pjotr S. Middendorf
- Food Microbiology, Wageningen University, Wageningen, Netherlands
- National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | | | - Aldert L. Zomer
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, Netherlands
- WHO Collaborating Center for Campylobacter/OIE Reference Laboratory for Campylobacteriosis, Utrecht, Netherlands
| | - Heidy M. W. den Besten
- Food Microbiology, Wageningen University, Wageningen, Netherlands
- Heidy M. W. den Besten,
| | - Tjakko Abee
- Food Microbiology, Wageningen University, Wageningen, Netherlands
- *Correspondence: Tjakko Abee,
| |
Collapse
|
33
|
Bergstrom K, Xia L. The barrier and beyond: Roles of intestinal mucus and mucin-type O-glycosylation in resistance and tolerance defense strategies guiding host-microbe symbiosis. Gut Microbes 2022; 14:2052699. [PMID: 35380912 PMCID: PMC8986245 DOI: 10.1080/19490976.2022.2052699] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Over the past two decades, our appreciation of the gut mucus has moved from a static lubricant to a dynamic and essential component of the gut ecosystem that not only mediates the interface between host tissues and vast microbiota, but regulates how this ecosystem functions to promote mutualistic symbioses and protect from microbe-driven diseases. By delving into the complex chemistry and biology of the mucus, combined with innovative in vivo and ex vivo approaches, recent studies have revealed novel insights into the formation and function of the mucus system, the O-glycans that make up this system, and how they mediate two major host-defense strategies - resistance and tolerance - to reduce damage caused by indigenous microbes and opportunistic pathogens. This current review summarizes these findings by highlighting the emerging roles of mucus and mucin-type O-glycans in influencing host and microbial physiology with an emphasis on host defense strategies against bacteria in the gastrointestinal tract.
Collapse
Affiliation(s)
- Kirk Bergstrom
- Department of Biology, University of British Columbia, Okanagan Campus, 3333 University Way, Kelowna, British ColumbiaV1V 1V7, Canada,Kirk Bergstrom Department of Biology, University of British Columbia, 3333 University Way, Kelowna, B.C. Canada
| | - Lijun Xia
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, OK, Oklahoma73104, USA,Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, OK, Oklahoma73104, USA,CONTACT Lijun Xia Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, OK, Oklahoma73104, USA
| |
Collapse
|
34
|
Li J, Chen J, Wang L, Lin Y, Zhang X, Liu J, Wang F. Characterization of the response of
Escherichia coli
to
l
‐fucose in bacterial swimming motility. J Basic Microbiol 2022; 62:584-592. [DOI: 10.1002/jobm.202200054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/02/2022] [Accepted: 03/13/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Jingyun Li
- Department of Food and Biotechnology, School of Food and Biological Engineering Hefei University of Technology Baohe District, Hefei Anhui PR China
| | - Juan Chen
- Department of Food and Biotechnology, School of Food and Biological Engineering Hefei University of Technology Baohe District, Hefei Anhui PR China
| | - Lu Wang
- Department of Food and Biotechnology, School of Food and Biological Engineering Hefei University of Technology Baohe District, Hefei Anhui PR China
| | - Yan Lin
- Department of Food and Biotechnology, School of Food and Biological Engineering Hefei University of Technology Baohe District, Hefei Anhui PR China
| | - Xian Zhang
- Department of Food and Biotechnology, School of Food and Biological Engineering Hefei University of Technology Baohe District, Hefei Anhui PR China
| | - Jian Liu
- Department of Food and Biotechnology, School of Food and Biological Engineering Hefei University of Technology Baohe District, Hefei Anhui PR China
| | - Fangbin Wang
- Department of Food and Biotechnology, School of Food and Biological Engineering Hefei University of Technology Baohe District, Hefei Anhui PR China
| |
Collapse
|
35
|
Detection of Bacterial α-l-Fucosidases with an Ortho-Quinone Methide-Based Probe and Mapping of the Probe-Protein Adducts. Molecules 2022; 27:molecules27051615. [PMID: 35268716 PMCID: PMC8911971 DOI: 10.3390/molecules27051615] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 11/24/2022] Open
Abstract
Fucosidases are associated with several pathological conditions and play an important role in the health of the human gut. For example, fucosidases have been shown to be indicators and/or involved in hepatocellular carcinoma, breast cancer, and helicobacter pylori infections. A prerequisite for the detection and profiling of fucosidases is the formation of a specific covalent linkage between the enzyme of interest and the activity-based probe (ABP). The most commonly used fucosidase ABPs are limited to only one of the classes of fucosidases, the retaining fucosidases. New approaches are needed that allow for the detection of the second class of fucosidases, the inverting type. Here, we report an ortho-quinone methide-based probe with an azide mini-tag that selectively labels both retaining and inverting bacterial α-l-fucosidases. Mass spectrometry-based intact protein and sequence analysis of a probe-labeled bacterial fucosidase revealed almost exclusive single labeling at two specific tryptophan residues outside of the active site. Furthermore, the probe could detect and image extracellular fucosidase activity on the surface of live bacteria.
Collapse
|
36
|
Lanzl MI, van Mastrigt O, Zwietering MH, Abee T, den Besten HMW. Role of substrate availability in the growth of Campylobacter co-cultured with extended spectrum beta-lactamase-producing Escherichia coli in Bolton broth. Int J Food Microbiol 2021; 363:109518. [PMID: 34996645 DOI: 10.1016/j.ijfoodmicro.2021.109518] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/17/2021] [Accepted: 12/24/2021] [Indexed: 01/03/2023]
Abstract
It is well-established that Extended-spectrum beta-lactamase-producing (ESBL-) Escherichia coli challenge reliable detection of campylobacters during enrichment in Bolton broth (BB) following ISO 10272-1:2017. The overgrowth of Campylobacter by ESBL-E. coli in the enrichment medium BB can lead to false-negative detection outcomes, but the cause for the growth suppression is yet unknown. A plausible reason could be the competition-induced lack of certain growth substrates. Therefore, this study aimed to investigate whether campylobacters and ESBL-E. coli compete for the same medium components and whether this is the cause for the observed growth repression. The availability of possible growth substrates in BB was determined and changes in their extracellular concentration were measured over time during mono-culture enrichment of C. jejuni, C. coli or ESBL-E. coli as well as in co-culture enrichments of campylobacters and ESBL-E. coli. Comparative analysis showed lactate and fumarate utilization by C. jejuni and C. coli exclusively, whereas ESBL-E. coli rapidly consumed asparagine, glutamine/arginine, lysine, threonine, tryptophan, pyruvate, glycerol, cellobiose, and glucose. Both campylobacters and ESBL-E. coli utilized aspartate, serine, formate, a-ketoglutarate and malate. Trends in compound utilization were similar for C. jejuni and C. coli and trends in compound utilization were rather comparable during enrichment of reference and freeze-stressed campylobacters. Since final cell densities of C. jejuni and C. coli in co-cultures were not enhanced by the addition of surplus l-serine and final cell densities were similar in fresh and spent medium, growth suppression seems not to be caused by a lack of substrates or production of inhibitory compounds. We hypothesized that oxygen availability was limiting growth in co-cultures. Higher oxygen availability increased the competitive fitness of C. jejuni 81-176 in co-culture with ESBL-E. coli in duplicate experiments, as cell concentrations in stationary phase were similar to those without competition. This could indicate the critical role of oxygen availability during the growth of Campylobacter and offers potential for further improvement of Campylobacter spp. enrichment efficacy.
Collapse
Affiliation(s)
- M I Lanzl
- Food Microbiology, Wageningen University & Research, Netherlands
| | - O van Mastrigt
- Food Microbiology, Wageningen University & Research, Netherlands
| | - M H Zwietering
- Food Microbiology, Wageningen University & Research, Netherlands
| | - T Abee
- Food Microbiology, Wageningen University & Research, Netherlands
| | - H M W den Besten
- Food Microbiology, Wageningen University & Research, Netherlands.
| |
Collapse
|
37
|
Fu R, Feng H, Dini-Andreote F, Wang Z, Bo C, Cao W, Yang K, Liu M, Yang T, Shen Q, Xu Y, Wei Z. Modulation of the Tomato Rhizosphere Microbiome via Changes in Root Exudation Mediated by the Ethylene Receptor NR. Microorganisms 2021; 9:2456. [PMID: 34946058 PMCID: PMC8705318 DOI: 10.3390/microorganisms9122456] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 11/17/2022] Open
Abstract
Plant hormones have been recently shown to exert an indirect influence on the recruitment of plant-associated microbiomes. However, it remains unclear the extent to which the disruption of the ethylene (ET) signaling pathway affects the assembly and functioning of plant-root microbiomes. In this study, the Never-ripe tomato mutant (Nr) was profiled for differences compared to the wild type (control). Tomato plants were subjected to root exudate profiling and the characterization of bacterial and fungal communities. Compared to the control, Nr revealed differences in the composition of root exudates, including lower amounts of esculetin, gallic acid, L-fucose, eicosapentaenoic acid, and higher amounts of β-aldehyde. Interestingly, Nr significantly differed in the composition and functioning of the rhizosphere bacterial community. We also identified the taxa that occurred at relatively higher abundances in Nr, including the genus Lysobacter, which displayed a significant negative correlation with changes in eicosapentaenoic acid and esculetin, and a significant positive correlation with changes in β-aldehyde. Taken together, our study provides evidence that a mutation in the ET receptor exerts predictable changes in the root-associated microbial taxa of tomato plants. These indirect effects can potentially be explored towards new strategies to engineer beneficial plant microbiomes via targeted changes in plant genetics and physiology.
Collapse
Affiliation(s)
- Ruixin Fu
- Jiangsu Provincial Key Lab of Solid Organic Waste Utilization, Jiangsu Collaborative Innovation Center of Solid Organic Waste Resource Utilization, National Engineering Research Center for Organic-Based Fertilizers, Nanjing Agricultural University, Nanjing 210095, China; (R.F.); (H.F.); (Z.W.); (C.B.); (W.C.); (K.Y.); (Q.S.); (Y.X.); (Z.W.)
| | - Haichao Feng
- Jiangsu Provincial Key Lab of Solid Organic Waste Utilization, Jiangsu Collaborative Innovation Center of Solid Organic Waste Resource Utilization, National Engineering Research Center for Organic-Based Fertilizers, Nanjing Agricultural University, Nanjing 210095, China; (R.F.); (H.F.); (Z.W.); (C.B.); (W.C.); (K.Y.); (Q.S.); (Y.X.); (Z.W.)
| | - Francisco Dini-Andreote
- Department of Plant Science and Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16801, USA;
| | - Zhen Wang
- Jiangsu Provincial Key Lab of Solid Organic Waste Utilization, Jiangsu Collaborative Innovation Center of Solid Organic Waste Resource Utilization, National Engineering Research Center for Organic-Based Fertilizers, Nanjing Agricultural University, Nanjing 210095, China; (R.F.); (H.F.); (Z.W.); (C.B.); (W.C.); (K.Y.); (Q.S.); (Y.X.); (Z.W.)
| | - Chunbin Bo
- Jiangsu Provincial Key Lab of Solid Organic Waste Utilization, Jiangsu Collaborative Innovation Center of Solid Organic Waste Resource Utilization, National Engineering Research Center for Organic-Based Fertilizers, Nanjing Agricultural University, Nanjing 210095, China; (R.F.); (H.F.); (Z.W.); (C.B.); (W.C.); (K.Y.); (Q.S.); (Y.X.); (Z.W.)
| | - Wenhui Cao
- Jiangsu Provincial Key Lab of Solid Organic Waste Utilization, Jiangsu Collaborative Innovation Center of Solid Organic Waste Resource Utilization, National Engineering Research Center for Organic-Based Fertilizers, Nanjing Agricultural University, Nanjing 210095, China; (R.F.); (H.F.); (Z.W.); (C.B.); (W.C.); (K.Y.); (Q.S.); (Y.X.); (Z.W.)
| | - Keming Yang
- Jiangsu Provincial Key Lab of Solid Organic Waste Utilization, Jiangsu Collaborative Innovation Center of Solid Organic Waste Resource Utilization, National Engineering Research Center for Organic-Based Fertilizers, Nanjing Agricultural University, Nanjing 210095, China; (R.F.); (H.F.); (Z.W.); (C.B.); (W.C.); (K.Y.); (Q.S.); (Y.X.); (Z.W.)
| | - Mingchun Liu
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China;
| | - Tianjie Yang
- Jiangsu Provincial Key Lab of Solid Organic Waste Utilization, Jiangsu Collaborative Innovation Center of Solid Organic Waste Resource Utilization, National Engineering Research Center for Organic-Based Fertilizers, Nanjing Agricultural University, Nanjing 210095, China; (R.F.); (H.F.); (Z.W.); (C.B.); (W.C.); (K.Y.); (Q.S.); (Y.X.); (Z.W.)
| | - Qirong Shen
- Jiangsu Provincial Key Lab of Solid Organic Waste Utilization, Jiangsu Collaborative Innovation Center of Solid Organic Waste Resource Utilization, National Engineering Research Center for Organic-Based Fertilizers, Nanjing Agricultural University, Nanjing 210095, China; (R.F.); (H.F.); (Z.W.); (C.B.); (W.C.); (K.Y.); (Q.S.); (Y.X.); (Z.W.)
| | - Yangchun Xu
- Jiangsu Provincial Key Lab of Solid Organic Waste Utilization, Jiangsu Collaborative Innovation Center of Solid Organic Waste Resource Utilization, National Engineering Research Center for Organic-Based Fertilizers, Nanjing Agricultural University, Nanjing 210095, China; (R.F.); (H.F.); (Z.W.); (C.B.); (W.C.); (K.Y.); (Q.S.); (Y.X.); (Z.W.)
| | - Zhong Wei
- Jiangsu Provincial Key Lab of Solid Organic Waste Utilization, Jiangsu Collaborative Innovation Center of Solid Organic Waste Resource Utilization, National Engineering Research Center for Organic-Based Fertilizers, Nanjing Agricultural University, Nanjing 210095, China; (R.F.); (H.F.); (Z.W.); (C.B.); (W.C.); (K.Y.); (Q.S.); (Y.X.); (Z.W.)
| |
Collapse
|
38
|
Jaakkola K, Virtanen K, Lahti P, Keto-Timonen R, Lindström M, Korkeala H. Comparative Genome Analysis and Spore Heat Resistance Assay Reveal a New Component to Population Structure and Genome Epidemiology Within Clostridium perfringens Enterotoxin-Carrying Isolates. Front Microbiol 2021; 12:717176. [PMID: 34566921 PMCID: PMC8456093 DOI: 10.3389/fmicb.2021.717176] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 08/10/2021] [Indexed: 11/13/2022] Open
Abstract
Clostridium perfringens causes a variety of human and animal enteric diseases including food poisoning, antibiotic-associated diarrhea, and necrotic enteritis. Yet, the reservoirs of enteropathogenic enterotoxin-producing strains remain unknown. We conducted a genomic comparison of 290 strains and a heat resistance phenotyping of 30 C. perfringens strains to elucidate the population structure and ecology of this pathogen. C. perfringens genomes shared a conserved genetic backbone with more than half of the genes of an average genome conserved in >95% of strains. The cpe-carrying isolates were found to share genetic context: the cpe-carrying plasmids had different distribution patterns within the genetic lineages and the estimated pan genome of cpe-carrying isolates had a larger core genome and a smaller accessory genome compared to that of 290 strains. We characterize cpe-negative strains related to chromosomal cpe-carrying strains elucidating the origin of these strains and disclose two distinct groups of chromosomal cpe-carrying strains with different virulence characteristics, spore heat resistance properties, and, presumably, ecological niche. Finally, an antibiotic-associated diarrhea isolate carrying two copies of the enterotoxin cpe gene and the associated genetic lineage with the potential for the emergence of similar strains are outlined. With C. perfringens as an example, implications of input genome quality for pan genome analysis are discussed. Our study furthers the understanding of genome epidemiology and population structure of enteropathogenic C. perfringens and brings new insight into this important pathogen and its reservoirs.
Collapse
Affiliation(s)
- Kaisa Jaakkola
- Department of Food Hygiene and Environmental Health, University of Helsinki, Helsinki, Finland
| | - Kira Virtanen
- Department of Bacteriology and Immunology, Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Northern Finland Laboratory Centre NordLab, Oulu, Finland
| | - Päivi Lahti
- City of Helsinki, Unit of Environmental Services, Helsinki, Finland
| | - Riikka Keto-Timonen
- Department of Food Hygiene and Environmental Health, University of Helsinki, Helsinki, Finland
| | - Miia Lindström
- Department of Food Hygiene and Environmental Health, University of Helsinki, Helsinki, Finland
| | - Hannu Korkeala
- Department of Food Hygiene and Environmental Health, University of Helsinki, Helsinki, Finland
| |
Collapse
|
39
|
Mucolytic bacteria: prevalence in various pathological diseases. World J Microbiol Biotechnol 2021; 37:176. [PMID: 34519941 DOI: 10.1007/s11274-021-03145-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023]
Abstract
All mucins are highly glycosylated and a key constituent of the mucus layer that is vigilant against pathogens in many organ systems of animals and humans. The viscous layer is organized in bilayers, i.e., an outer layer that is loosely arranged, variable in thickness, home to the commensal microbiota that grows in the complex environment, and an innermost layer that is stratified, non-aspirated, firmly adherent to the epithelial cells and devoid of any microorganisms. The O-glycosylation moiety represents the site of adhesion for pathogens and due to the increase of motility, mucolytic activity, and upregulation of virulence factors, some microorganisms can circumvent the component of the mucus layer and cause disruption in organ homeostasis. A dysbiotic microbiome, defective mucus barrier, and altered immune response often result in various diseases. In this review, paramount emphasis is given to the role played by the bacterial species directly or indirectly involved in mucin degradation, alteration in mucus secretion or its composition or mucin gene expression, which instigates many diseases in the digestive, respiratory, and other organ systems. A systematic view can help better understand the etiology of some complex disorders such as cystic fibrosis, ulcerative colitis and expand our knowledge about mucin degraders to develop new therapeutic approaches to correct ill effects caused by these mucin-dwelling pathogens.
Collapse
|
40
|
Aviles FA, Kyndt JA. Cellulosimicrobium fucosivorans sp. nov., isolated from San Elijo Lagoon, contains a fucose metabolic pathway linked to carotenoid production. Arch Microbiol 2021; 203:4525-4538. [PMID: 34148152 DOI: 10.1007/s00203-021-02443-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 06/10/2021] [Accepted: 06/13/2021] [Indexed: 10/21/2022]
Abstract
Cellulosimicrobium strain SE3T was isolated from the San Elijo coastal lagoon near San Diego. A whole genome-based phylogenetic comparison shows great heterogeneity within the Cellulosimicrobium genus. Based on average nucleotide identity, whole genome-based comparison, and the presence of a unique L-fucose metabolic pathway, strain SE3T was shown to belong to a novel species within the genus, together with five other strains. The name Cellulosimicrobium fucosivorans sp. nov. is proposed, with strain SE3T as the type strain. The strain encodes a unique alpha-L-fucosidase and the L-fucose metabolic pathway is homologous to the one recently described in Campylobacter jejuni. C. fucosivorans is able to grow on L-fucose, and interestingly, the biosynthesis of the yellow carotenoid is dependent on the presence of L-fucose in the media. The ability to metabolize fucose and the linked production of carotenoids are expected to provide C. fucosivorans with a competitive advantage in the sunny coastal lagoon area.
Collapse
Affiliation(s)
- Fabiola A Aviles
- College of Science and Technology, Bellevue University, 1000 Galvin Rd. S., Bellevue, NE, 68005, USA
| | - John A Kyndt
- College of Science and Technology, Bellevue University, 1000 Galvin Rd. S., Bellevue, NE, 68005, USA.
| |
Collapse
|
41
|
Luijkx YMCA, Jongkees S, Strijbis K, Wennekes T. Development of a 1,2-difluorofucoside activity-based probe for profiling GH29 fucosidases. Org Biomol Chem 2021; 19:2968-2977. [PMID: 33729259 DOI: 10.1039/d1ob00054c] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
GH29 α-l-fucosidases catalyze hydrolysis of terminal α-l-fucosyl linkages with varying specificity and are expressed by prominent members of the human gut microbiota. Both homeostasis and dysbiosis at the human intestinal microbiota interface have been correlated with altered fucosidase activity. Herein we describe the development of a 2-deoxy-2-fluoro fucosyl fluoride derivative with an azide mini-tag as an activity-based probe (ABP) for selective in vitro labelling of GH29 α-l-fucosidases. Only catalytically active fucosidases are inactivated by this ABP, allowing their functionalization with a biotin reporter group via the CuAAC reaction and subsequent in-gel detection at nanogram levels. The ABP we present here is shown to be active against a GH29 α-l-fucosidase from Bacteroides fragilis and capable of labeling two other GH29 α-l-fucosidases with different linkage specificity, illustrating its broader utility. This novel ABP is a valuable addition to the toolbox of fucosidase probes by allowing identification and functional studies of the wide variety of GH29 fucosidases, including those in the gut microbiota.
Collapse
Affiliation(s)
- Yvette M C A Luijkx
- Department Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
42
|
Galeev A, Suwandi A, Cepic A, Basu M, Baines JF, Grassl GA. The role of the blood group-related glycosyltransferases FUT2 and B4GALNT2 in susceptibility to infectious disease. Int J Med Microbiol 2021; 311:151487. [PMID: 33662872 DOI: 10.1016/j.ijmm.2021.151487] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/01/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022] Open
Abstract
The glycosylation profile of the gastrointestinal tract is an important factor mediating host-microbe interactions. Variation in these glycan structures is often mediated by blood group-related glycosyltransferases, and can lead to wide-ranging differences in susceptibility to both infectious- as well as chronic disease. In this review, we focus on the interplay between host glycosylation, the intestinal microbiota and susceptibility to gastrointestinal pathogens based on studies of two exemplary blood group-related glycosyltransferases that are conserved between mice and humans, namely FUT2 and B4GALNT2. We highlight that differences in susceptibility can arise due to both changes in direct interactions, such as bacterial adhesion, as well as indirect effects mediated by the intestinal microbiota. Although a large body of experimental work exists for direct interactions between host and pathogen, determining the more complex and variable mechanisms underlying three-way interactions involving the intestinal microbiota will be the subject of much-needed future research.
Collapse
Affiliation(s)
- Alibek Galeev
- Max Planck Institute for Evolutionary Biology, Plön, Germany and Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - Abdulhadi Suwandi
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research (DZIF), Hannover, Germany
| | - Aleksa Cepic
- Max Planck Institute for Evolutionary Biology, Plön, Germany and Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - Meghna Basu
- Max Planck Institute for Evolutionary Biology, Plön, Germany and Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - John F Baines
- Max Planck Institute for Evolutionary Biology, Plön, Germany and Institute for Experimental Medicine, Kiel University, Kiel, Germany.
| | - Guntram A Grassl
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research (DZIF), Hannover, Germany.
| |
Collapse
|
43
|
Desantis S, Galosi L, Santamaria N, Roncarati A, Biagini L, Rossi G. Modulation of Morphology and Glycan Composition of Mucins in Farmed Guinea Fowl ( Numida meleagris) Intestine by the Multi-Strain Probiotic Slab51 ®. Animals (Basel) 2021; 11:495. [PMID: 33668637 PMCID: PMC7918860 DOI: 10.3390/ani11020495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 11/24/2022] Open
Abstract
Probiotics have become highly recognized as supplements for poultry.Since gut health can be considered synonymous withanimal health, the effects of probiotic Slab51® on the morphology and the glycan composition of guineafowlintestine were examined. The probiotics were added in drinking water (2 × 1011 UFC/L) throughout the grow-out cycle.Birds were individually weighed andslaughtered after four months. Samples from the duodenum, ileum and caecum were collected and processed for morphological, morphometric, conventional and lectin glycohistochemical studies.The results were analyzed for statistical significance by Student's t test. Compared with control samples, probiotic group revealed (1) significant increase in villus height (p < 0.001 in duodenum and ileum; p < 0.05 in caecum), crypt depth (p < 0.001 in duodenum and caecum; p < 0.05 in ileum) and goblet cells (GCs) per villus (p < 0.001) in all investigated tracts; (2) increase in galactoseβl,3N-acetylgalacyosamine(Galβl,3GalNAc)terminating O-glycans and αl,2-fucosylated glycans secretory GCs in the duodenum; (3) increase in α2,6-sialoglycans and high-mannose N-linked glycans secretory GCs but reduction in GCs-secreting sulfoglycans in the ileum; (4) increase in Galβl,3GalNAc and high-mannose N-linked glycans secretory GCs and decrease in GCs-producing sulfomucins in the caecum; (5) increase in the numbers of crypt cells containing sulfate and non-sulfated acidic glycans. Overall, dietary Slab51® induces morphological and region-specific changes in glycoprotein composition of guinea fowl intestine, promoting gut health.
Collapse
Affiliation(s)
- Salvatore Desantis
- Department of Emergency and Organ Transplantation (DETO), University of Bari Aldo Moro, S.P. 62 per Casamassima Km 3, 70010 Valenzano (Bari), Italy;
| | - Livio Galosi
- School of Biosciences and Veterinary medicine, University of Camerino, Via Circonvallazione 93/95, 62024 Matelica (M.C.), Italy; (A.R.); (L.B.); (G.R.)
| | - Nicoletta Santamaria
- Department of Emergency and Organ Transplantation (DETO), University of Bari Aldo Moro, S.P. 62 per Casamassima Km 3, 70010 Valenzano (Bari), Italy;
| | - Alessandra Roncarati
- School of Biosciences and Veterinary medicine, University of Camerino, Via Circonvallazione 93/95, 62024 Matelica (M.C.), Italy; (A.R.); (L.B.); (G.R.)
| | - Lucia Biagini
- School of Biosciences and Veterinary medicine, University of Camerino, Via Circonvallazione 93/95, 62024 Matelica (M.C.), Italy; (A.R.); (L.B.); (G.R.)
| | - Giacomo Rossi
- School of Biosciences and Veterinary medicine, University of Camerino, Via Circonvallazione 93/95, 62024 Matelica (M.C.), Italy; (A.R.); (L.B.); (G.R.)
| |
Collapse
|
44
|
Garber JM, Hennet T, Szymanski CM. Significance of fucose in intestinal health and disease. Mol Microbiol 2021; 115:1086-1093. [PMID: 33434389 DOI: 10.1111/mmi.14681] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/30/2020] [Accepted: 01/08/2021] [Indexed: 12/14/2022]
Abstract
The deoxyhexose sugar L-fucose is important for many biological processes within the human body and the associated microbiota. This carbohydrate is abundant in host gut mucosal surfaces, numerous microbial cell surface structures, and some dietary carbohydrates. Fucosylated oligosaccharides facilitate the establishment of a healthy microbiota and provide protection from infection. However, there are instances where pathogens can also exploit these fucosylated structures to cause infection. Furthermore, deficiencies in host fucosylation are associated with specific disease outcomes. This review focuses on our current understanding of the impact of fucosylation within the mucosal environment of the gastrointestinal tract with a specific emphasis on the mediatory effects in host-microbe interactions.
Collapse
Affiliation(s)
- Jolene M Garber
- Complex Carbohydrate Research Center and Department of Microbiology, University of Georgia, Athens, GA, USA
| | - Thierry Hennet
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Christine M Szymanski
- Complex Carbohydrate Research Center and Department of Microbiology, University of Georgia, Athens, GA, USA
| |
Collapse
|
45
|
Elmi A, Nasher F, Dorrell N, Wren B, Gundogdu O. Revisiting Campylobacter jejuni Virulence and Fitness Factors: Role in Sensing, Adapting, and Competing. Front Cell Infect Microbiol 2021; 10:607704. [PMID: 33614526 PMCID: PMC7887314 DOI: 10.3389/fcimb.2020.607704] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/11/2020] [Indexed: 12/18/2022] Open
Abstract
Campylobacter jejuni is the leading cause of bacterial foodborne gastroenteritis world wide and represents a major public health concern. Over the past two decades, significant progress in functional genomics, proteomics, enzymatic-based virulence profiling (EBVP), and the cellular biology of C. jejuni have improved our basic understanding of this important pathogen. We review key advances in our understanding of the multitude of emerging virulence factors that influence the outcome of C. jejuni–mediated infections. We highlight, the spatial and temporal dynamics of factors that promote C. jejuni to sense, adapt and survive in multiple hosts. Finally, we propose cohesive research directions to obtain a comprehensive understanding of C. jejuni virulence mechanisms.
Collapse
Affiliation(s)
- Abdi Elmi
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Fauzy Nasher
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Nick Dorrell
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Brendan Wren
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ozan Gundogdu
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
46
|
Higgins MA, Ryan KS. Generating a fucose permease deletion mutant in Bifidobacterium longum subspecies infantis ATCC 15697. Anaerobe 2021; 68:102320. [PMID: 33460787 DOI: 10.1016/j.anaerobe.2021.102320] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/16/2022]
Abstract
Bifidobacterium longum subsp. infantis ATCC 15697 has emerged as a model for infant gut-associated bifidobacterial strains. Here we present a genetic system for B. longum subsp. infantis ATCC 15697 using its own DNA restriction-modification systems and create a fucose permease deletion mutant lacking the ability to use free fucose as a carbon source.
Collapse
Affiliation(s)
- Melanie A Higgins
- Department of Chemistry, The University of British Columbia, Vancouver, British Columbia, V6T 1Z1, Canada
| | - Katherine S Ryan
- Department of Chemistry, The University of British Columbia, Vancouver, British Columbia, V6T 1Z1, Canada.
| |
Collapse
|
47
|
Elgamoudi BA, Andrianova EP, Shewell LK, Day CJ, King RM, Taha, Rahman H, Hartley-Tassell LE, Zhulin IB, Korolik V. The Campylobacter jejuni chemoreceptor Tlp10 has a bimodal ligand-binding domain and specificity for multiple classes of chemoeffectors. Sci Signal 2021; 14:14/664/eabc8521. [PMID: 33402336 DOI: 10.1126/scisignal.abc8521] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Campylobacter jejuni is a bacterial pathogen that is a common cause of enteritis in humans. We identified a previously uncharacterized type of sensory domain in the periplasmic region of the C. jejuni chemoreceptor Tlp10, termed the DAHL domain, that is predicted to have a bimodular helical architecture. Through two independent ligand-binding sites in this domain, Tlp10 responded to molecular aspartate, isoleucine, fumarate, malate, fucose, and mannose as attractants and to arginine, galactose, and thiamine as repellents. Tlp10 also recognized glycan ligands when present as terminal and intermediate residues of complex structures, such as the fucosylated human ganglioside GM1 and Lewisa antigen. A tlp10 mutant strain lacking the ligand-binding sites was attenuated in its ability to colonize avian caeca and to adhere to cultured human intestinal cells, indicating the potential involvement of the DAHL domain in host colonization and disease. The Tlp10 intracellular signaling domain interacted with the scaffolding proteins CheV and CheW, which couple chemoreceptors to intracellular signaling machinery, and with the signaling domains of other chemoreceptors, suggesting a key role for Tlp10 in signal transduction and incorporation into sensory arrays. We identified the DAHL domain in other bacterial signal transduction proteins, including the essential virulence induction protein VirA from the plant pathogen Agrobacterium tumefaciens Together, these results suggest a potential link between Tlp10 and C. jejuni virulence.
Collapse
Affiliation(s)
- Bassam A Elgamoudi
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD 4222, Australia
| | | | - Lucy K Shewell
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD 4222, Australia
| | - Christopher J Day
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD 4222, Australia
| | - Rebecca M King
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD 4222, Australia
| | - Taha
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD 4222, Australia
| | - Hossinur Rahman
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD 4222, Australia
| | | | - Igor B Zhulin
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Victoria Korolik
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD 4222, Australia. .,School of Medical Science, Griffith University, Gold Coast Campus, Southport, QLD 4222, Australia
| |
Collapse
|
48
|
Bailén M, Bressa C, Martínez-López S, González-Soltero R, Montalvo Lominchar MG, San Juan C, Larrosa M. Microbiota Features Associated With a High-Fat/Low-Fiber Diet in Healthy Adults. Front Nutr 2020; 7:583608. [PMID: 33392236 PMCID: PMC7775391 DOI: 10.3389/fnut.2020.583608] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/23/2020] [Indexed: 12/14/2022] Open
Abstract
A high intake of dietary saturated fatty acids (SFAs) is related to an increased risk of obesity, inflammation and cancer-related diseases, and this risk is attenuated only when SFAs are replaced by unsaturated fats and unrefined carbohydrates. The gut microbiota has recently emerged as a new environmental factor in the pathophysiology of these disorders, and is also one of the factors most influenced by diet. We sought to determine whether the gut microbiota of healthy individuals whose intake of SFAs exceeds World Health Organization (WHO) recommendations exhibits features similar to those reported in people with obesity, inflammation, cancer or metabolic disease. Healthy non-obese subjects were divided into two groups based on their SFAs intake. Body composition and gut microbiota composition were analyzed, and associations between bacterial taxa, diet and body fat composition were determined globally and separately by sex. Metagenome functional pathways were predicted by PICRUSt analysis. Subjects whose SFAs intake exceeded WHO recommendations also had a dietary pattern of low fiber intake. This high saturated fat/low fiber diet was associated with a greater sequence abundance of the Anaerotruncus genus, a butyrate producer associated with obesity. Analysis of data of high SFAs intake by sex showed that females presented with a greater abundance of Campylobacter, Blautia, Flavonifractor and Erysipelatoclostridium, whereas males showed higher levels of Anaerotruncus, Eisenbergiella, a genus from the order Clostridiales (FamilyXIIIUCG_001) and two genera from the Lachnospiraceae family. PICRUSt analysis confirmed these data, showing a correlation with a decrease in the abundance of sequences encoding for transporters of some metals such as iron, which is needed to maintain a healthy metabolism. Thus, the microbiota of healthy people on a high SFAs diet contain bacterial taxa (Anaerotruncus, Lachnospiraceae Flavonifractor, Campylobacter, Erysipelotrichacea and Eisenbergiella) that could be related to the development of some diseases, especially obesity and other pro-inflammatory diseases in women. In summary, the present study identifies bacterial taxa that could be considered as early predictors for the onset of different diseases in healthy subjects. Also, sex differences in gut microbiota suggest that women and men differentially benefit from following a specific diet.
Collapse
Affiliation(s)
- María Bailén
- MAS Microbiota Group, School of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
49
|
Dedon LR, Özcan E, Rani A, Sela DA. Bifidobacterium infantis Metabolizes 2'Fucosyllactose-Derived and Free Fucose Through a Common Catabolic Pathway Resulting in 1,2-Propanediol Secretion. Front Nutr 2020; 7:583397. [PMID: 33330584 PMCID: PMC7732495 DOI: 10.3389/fnut.2020.583397] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/09/2020] [Indexed: 01/11/2023] Open
Abstract
Human milk oligosaccharides (HMOs) enrich beneficial bifidobacteria in the infant gut microbiome which produce molecules that impact development and physiology. 2′fucosyllactose (2′FL) is a highly abundant fucosylated HMO which is utilized by Bifidobacterium longum subsp. infantis, despite limited scientific understanding of the underlying mechanism. Moreover, there is not a current consensus on whether free fucose could be metabolized when not incorporated in a larger oligosaccharide structure. Based on metabolic and genomic analyses, we hypothesize that B. infantis catabolizes both free fucose and fucosyl oligosaccharide residues to produce 1,2-propanediol (1,2-PD). Accordingly, systems-level approaches including transcriptomics and proteomics support this metabolic path. Co-fermentation of fucose and limiting lactose or glucose was found to promote significantly higher biomass and 1,2-PD concentrations than individual substrates, suggesting a synergistic effect. In addition, and during growth on 2′FL, B. infantis achieves significantly higher biomass corresponding to increased 1,2-PD. These findings support a singular fucose catabolic pathway in B. infantis that is active on both free and HMO-derived fucose and intimately linked with central metabolism. The impact of fucose and 2′FL metabolism on B. infantis physiology provides insight into the role of fucosylated HMOs in influencing host- and microbe-microbe interactions within the infant gut microbiome.
Collapse
Affiliation(s)
- Liv R Dedon
- Department of Food Science, University of Massachusetts Amherst, Amherst, MA, United States
| | - Ezgi Özcan
- Department of Food Science, University of Massachusetts Amherst, Amherst, MA, United States
| | - Asha Rani
- Department of Food Science, University of Massachusetts Amherst, Amherst, MA, United States
| | - David A Sela
- Department of Food Science, University of Massachusetts Amherst, Amherst, MA, United States.,Department of Microbiology, University of Massachusetts Amherst, Amherst, MA, United States.,Department of Microbiology and Physiological Systems and Center for Microbiome Research, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
50
|
Xu C, Jia Q, Zhang L, Wang Z, Zhu S, Wang X, Liu Y, Li M, Zhang J, Wang X, Zhang J, Sun Q, Wang K, Zhu H, Duan L. Multiomics Study of Gut Bacteria and Host Metabolism in Irritable Bowel Syndrome and Depression Patients. Front Cell Infect Microbiol 2020; 10:580980. [PMID: 33194817 PMCID: PMC7658686 DOI: 10.3389/fcimb.2020.580980] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/07/2020] [Indexed: 12/20/2022] Open
Abstract
Background and Aims Irritable bowel syndrome (IBS) and depression have high tendencies of comorbidity. In particular, diarrhea-predominant IBS (IBS-D) and depression exhibit similar fecal microbiota signatures, yet little is known about their pathogenic mechanism. Here, we propose that the differences in structure and composition of IBS-D and depression gut microbiota give rise to different downstream functions, which lead to distinct clinical phenotypes via host metabolism and further influence the interaction of brain–gut axis. Methods We performed multiomics study, including fecal metagenome-wide sequencing and serum metabolomics profiling in 65 individuals with IBS-D (n=22), depression (n=15), comorbid patients (n=13), and healthy controls (n=15). We analyzed functional genes contributed by the primary genus and evaluated their correlations with clinical indices and host metabolites. Results Metagenomic analysis revealed 26 clusters of orthologous groups of protein (COG) categories consisting of a total of 4,631 functional genes. Trehalose and maltose hydrolase (COG1554) and fucose permease (COG0738) were the most relevant and enriched functional genes in the IBS-D patients; urease accessory proteins UreE (COG2371) was that in the depression patients. Context based genome annotation suggest that an alteration of Escherichia coli and Enterobacter cloacae in IBS-D and depression respectively may be responsible for the enrichment described above. Correlation with host metabolites, such as maltotriose and isomaltose in carbohydrate metabolism and anandamide in neuroactive metabolism, drew further connections between these findings. Conclusions These changes led us to propose a connection between genomic signatures and clinical differences observed in IBS-D and depression. Our findings provide further insights into the involvement of gut microbiota in diseases related to brain–gut disorder.
Collapse
Affiliation(s)
- Congmin Xu
- Department of Biomedical Engineering, College of Engineering, and Center for Quantitative Biology, Peking University, Beijing, China
| | - Qiong Jia
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Lu Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Zhe Wang
- Department of Biomedical Engineering, College of Engineering, and Center for Quantitative Biology, Peking University, Beijing, China
| | - Shiwei Zhu
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Xiaoqi Wang
- Department of Biomedical Engineering, College of Engineering, and Center for Quantitative Biology, Peking University, Beijing, China
| | - Yixuan Liu
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Mo Li
- Department of Biomedical Engineering, College of Engineering, and Center for Quantitative Biology, Peking University, Beijing, China
| | - Jingjing Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Xiangqun Wang
- Department of Psychiatry, Institute of Mental Health, Peking University, Beijing, China
| | - Jindong Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Qinghua Sun
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Kun Wang
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Huaiqiu Zhu
- Department of Biomedical Engineering, College of Engineering, and Center for Quantitative Biology, Peking University, Beijing, China
| | - Liping Duan
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| |
Collapse
|