1
|
Kashima T, Akama M, Wakinaka T, Arakawa T, Ashida H, Fushinobu S. Crystal Structure of Bifidobacterium bifidum Glycoside Hydrolase Family 110 α-Galactosidase Specific for Blood Group B Antigen. J Appl Glycosci (1999) 2024; 71:81-90. [PMID: 39234034 PMCID: PMC11368712 DOI: 10.5458/jag.jag.jag-2024_0005] [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: 03/04/2024] [Accepted: 04/18/2024] [Indexed: 09/06/2024] Open
Abstract
To overcome incompatibility issues and increase the possibility of blood transfusion, technologies that enable efficient conversion of A- and B-type red blood cells to the universal donor O-type is desirable. Although several blood type-converting enzymes have been identified, detailed understanding about their molecular functions is limited. α-Galactosidase from Bifidobacterium bifidum JCM 1254 (AgaBb), belonging to glycoside hydrolase (GH) 110 subfamily A, specifically acts on blood group B antigen. Here we present the crystal structure of AgaBb, including the catalytic GH110 domain and part of the C-terminal uncharacterized regions. Based on this structure, we deduced a possible binding mechanism of blood group B antigen to the active site. Site-directed mutagenesis confirmed that R270 and E380 recognize the fucose moiety in the B antigen. Thermal shift assay revealed that the C-terminal uncharacterized region significantly contributes to protein stability. This region is shared only among GH110 enzymes from B. bifidum and some Ruminococcus species. The elucidation of the molecular basis for the specific recognition of blood group B antigen is expected to lead to the practical application of blood group conversion enzymes in the future.
Collapse
Affiliation(s)
- Toma Kashima
- Department of Biotechnology, The University of Tokyo
| | - Megumi Akama
- Department of Biotechnology, The University of Tokyo
| | | | | | - Hisashi Ashida
- Faculty of Biology-Oriented Science and Technology, Kindai University
| | - Shinya Fushinobu
- Department of Biotechnology, The University of Tokyo
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo
| |
Collapse
|
2
|
Bachar-Wikstrom E, Thomsson KA, Sihlbom C, Abbo L, Tartor H, Lindén SK, Wikstrom JD. Identification of Novel Glycans in the Mucus Layer of Shark and Skate Skin. Int J Mol Sci 2023; 24:14331. [PMID: 37762632 PMCID: PMC10532229 DOI: 10.3390/ijms241814331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/13/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
The mucus layer covering the skin of fish has several roles, including protection against pathogens and mechanical damage. While the mucus layers of various bony fish species have been investigated, the composition and glycan profiles of shark skin mucus remain relatively unexplored. In this pilot study, we aimed to explore the structure and composition of shark skin mucus through histological analysis and glycan profiling. Histological examination of skin samples from Atlantic spiny dogfish (Squalus acanthias) sharks and chain catsharks (Scyliorhinus retifer) revealed distinct mucin-producing cells and a mucus layer, indicating the presence of a functional mucus layer similar to bony fish mucus albeit thinner. Glycan profiling using liquid chromatography-electrospray ionization tandem mass spectrometry unveiled a diverse repertoire of mostly O-glycans in the mucus of the two sharks as well as little skate (Leucoraja erinacea). Elasmobranch glycans differ significantly from bony fish, especially in being more sulfated, and some bear resemblance to human glycans, such as gastric mucin O-glycans and H blood group-type glycans. This study contributes to the concept of shark skin having unique properties and provides a foundation for further research into the functional roles and potential biomedical implications of shark skin mucus glycans.
Collapse
Affiliation(s)
- Etty Bachar-Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, 17177 Stockholm, Sweden
- Whitman Center, Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Kristina A. Thomsson
- Proteomics Core Facility of Sahlgrenska Academy, University of Gothenburg, 41390 Gothenburg, Sweden
| | - Carina Sihlbom
- Proteomics Core Facility of Sahlgrenska Academy, University of Gothenburg, 41390 Gothenburg, Sweden
| | - Lisa Abbo
- Whitman Center, Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Haitham Tartor
- Department of Fish Health and Welfare, Norwegian Veterinary Institute, P.O. Box 750, Sentrum, 0106 Oslo, Norway
| | - Sara K. Lindén
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, P.O. Box 440, Medicinaregatan 9C, 40530 Gothenburg, Sweden
| | - Jakob D. Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, 17177 Stockholm, Sweden
- Whitman Center, Marine Biological Laboratory, Woods Hole, MA 02543, USA
- Dermato-Venereology Clinic, Karolinska University Hospital, 17176 Stockholm, Sweden
| |
Collapse
|
3
|
Gui B, Yao L, Qu M, Zhang W, Li M, Jiang Y, Wang L. Cloning, Expression, and Functional Characterization of FUT1, a Key Gene for Histo-Blood Group Antigens Synthesis in Crassostrea gigas. Curr Issues Mol Biol 2023; 45:4200-4213. [PMID: 37232736 DOI: 10.3390/cimb45050267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/27/2023] Open
Abstract
Histo-blood group antigens (HBGAs) comprise a family of cell-surface carbohydrates that are considered norovirus-specific binding receptors or ligands. HBGA-like molecules have also been detected in oysters as common norovirus carriers, although the pathway involved in the synthesis of these molecules in oysters has yet to be elucidated. We isolated and identified a key gene involved in the synthesis of HBGA-like molecules, FUT1, from Crassostrea gigas, named CgFUT1. Real-time quantitative polymerase chain reaction analysis showed that CgFUT1 mRNA was expressed in the mantle, gill, muscle, labellum, and hepatopancreatic tissues of C. gigas, with the hepatopancreas exhibiting the highest expression level. A recombinant CgFUT1 protein with a molecular mass of 38.0 kDa was expressed in Escherichia coli using a prokaryotic expression vector. A eukaryotic expression plasmid was constructed and transfected into Chinese hamster ovary (CHO) cells. The expression of CgFUT1 and membrane localization of type H-2 HBGA-like molecules in CHO cells were detected using Western blotting and cellular immunofluorescence, respectively. This study indicated that CgFUT1, expressed in C. gigas tissues, can synthesize type H-2 HBGA-like molecules. This finding provides a new perspective for analyzing the source and synthetic pathway of HBGA-like molecules in oysters.
Collapse
Affiliation(s)
- Binbin Gui
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
- Key Laboratory of Testing and Evaluation for Aquatic Product Safety and Quality, Ministry of Agriculture and Rural Affairs, Qingdao 266071, China
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Lin Yao
- Key Laboratory of Testing and Evaluation for Aquatic Product Safety and Quality, Ministry of Agriculture and Rural Affairs, Qingdao 266071, China
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Meng Qu
- Key Laboratory of Testing and Evaluation for Aquatic Product Safety and Quality, Ministry of Agriculture and Rural Affairs, Qingdao 266071, China
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Weiran Zhang
- Key Laboratory of Testing and Evaluation for Aquatic Product Safety and Quality, Ministry of Agriculture and Rural Affairs, Qingdao 266071, China
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Mingyu Li
- Key Laboratory of Testing and Evaluation for Aquatic Product Safety and Quality, Ministry of Agriculture and Rural Affairs, Qingdao 266071, China
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Yanhua Jiang
- Key Laboratory of Testing and Evaluation for Aquatic Product Safety and Quality, Ministry of Agriculture and Rural Affairs, Qingdao 266071, China
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Lianzhu Wang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
- Key Laboratory of Testing and Evaluation for Aquatic Product Safety and Quality, Ministry of Agriculture and Rural Affairs, Qingdao 266071, China
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| |
Collapse
|
4
|
Pap A, Kiraly IE, Medzihradszky KF, Darula Z. Multiple Layers of Complexity in O-Glycosylation Illustrated With the Urinary Glycoproteome. Mol Cell Proteomics 2022; 21:100439. [PMID: 36334872 PMCID: PMC9758497 DOI: 10.1016/j.mcpro.2022.100439] [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: 05/25/2022] [Revised: 10/17/2022] [Accepted: 10/31/2022] [Indexed: 11/11/2022] Open
Abstract
While N-glycopeptides are relatively easy to characterize, O-glycosylation analysis is more complex. In this article, we illustrate the multiple layers of O-glycopeptide characterization that make this task so challenging. We believe our carefully curated dataset represents perhaps the largest intact human glycopeptide mixture derived from individuals, not from cell lines. The samples were collected from healthy individuals, patients with superficial or advanced bladder cancer (three of each group), and a single bladder inflammation patient. The data were scrutinized manually and interpreted using three different search engines: Byonic, Protein Prospector, and O-Pair, and the tool MS-Filter. Despite all the recent advances, reliable automatic O-glycopeptide assignment has not been solved yet. Our data reveal such diversity of site-specific O-glycosylation that has not been presented before. In addition to the potential biological implications, this dataset should be a valuable resource for software developers in the same way as some of our previously released data has been used in the development of O-Pair and O-Glycoproteome Analyzer. Based on the manual evaluation of the performance of the existing tools with our data, we lined up a series of recommendations that if implemented could significantly improve the reliability of glycopeptide assignments.
Collapse
Affiliation(s)
- Adam Pap
- Laboratory of Proteomics Research, Biological Research Centre, Eotvos Lorand Research Network (ELKH) Szeged, Hungary
| | | | - Katalin F. Medzihradszky
- Laboratory of Proteomics Research, Biological Research Centre, Eotvos Lorand Research Network (ELKH) Szeged, Hungary,For correspondence: Zsuzsanna Darula; Katalin F. Medzihradszky
| | - Zsuzsanna Darula
- Laboratory of Proteomics Research, Biological Research Centre, Eotvos Lorand Research Network (ELKH) Szeged, Hungary,Single Cell Omics Advanced Core Facility, Hungarian Centre of Excellence for Molecular Medicine Szeged, Hungary,For correspondence: Zsuzsanna Darula; Katalin F. Medzihradszky
| |
Collapse
|
5
|
Wardman JF, Bains RK, Rahfeld P, Withers SG. Carbohydrate-active enzymes (CAZymes) in the gut microbiome. Nat Rev Microbiol 2022; 20:542-556. [PMID: 35347288 DOI: 10.1038/s41579-022-00712-1] [Citation(s) in RCA: 215] [Impact Index Per Article: 71.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2022] [Indexed: 12/13/2022]
Abstract
The 1013-1014 microorganisms present in the human gut (collectively known as the human gut microbiota) dedicate substantial percentages of their genomes to the degradation and uptake of carbohydrates, indicating the importance of this class of molecules. Carbohydrates function not only as a carbon source for these bacteria but also as a means of attachment to the host, and a barrier to infection of the host. In this Review, we focus on the diversity of carbohydrate-active enzymes (CAZymes), how gut microorganisms use them for carbohydrate degradation, the different chemical mechanisms of these CAZymes and the roles that these microorganisms and their CAZymes have in human health and disease. We also highlight examples of how enzymes from this treasure trove have been used in manipulation of the microbiota for improved health and treatment of disease, in remodelling the glycans on biopharmaceuticals and in the potential production of universal O-type donor blood.
Collapse
Affiliation(s)
- Jacob F Wardman
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada.,Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rajneesh K Bains
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Peter Rahfeld
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephen G Withers
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada. .,Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada. .,Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
6
|
Muthusamy S, Jan HM, Hsieh MY, Mondal S, Liu WC, Ko YA, Yang WY, Mong KKT, Chen GC, Lin CH. Enhanced enzymatic production of cholesteryl 6'-acylglucoside impairs lysosomal degradation for the intracellular survival of Helicobacter pylori. J Biomed Sci 2021; 28:72. [PMID: 34706729 PMCID: PMC8549234 DOI: 10.1186/s12929-021-00768-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 10/08/2021] [Indexed: 01/15/2023] Open
Abstract
Background During autophagy defense against invading microbes, certain lipid types are indispensable for generating specialized membrane-bound organelles. The lipid composition of autophagosomes remains obscure, as does the issue of how specific lipids and lipid-associated enzymes participate in autophagosome formation and maturation. Helicobacter pylori is auxotrophic for cholesterol and converts cholesterol to cholesteryl glucoside derivatives, including cholesteryl 6ʹ-O-acyl-α-d-glucoside (CAG). We investigated how CAG and its biosynthetic acyltransferase assist H. pylori to escape host-cell autophagy. Methods We applied a metabolite-tagging method to obtain fluorophore-containing cholesteryl glucosides that were utilized to understand their intracellular locations. H. pylori 26695 and a cholesteryl glucosyltransferase (CGT)-deletion mutant (ΔCGT) were used as the standard strain and the negative control that contains no cholesterol-derived metabolites, respectively. Bacterial internalization and several autophagy-related assays were conducted to unravel the possible mechanism that H. pylori develops to hijack the host-cell autophagy response. Subcellular fractions of H. pylori-infected AGS cells were obtained and measured for the acyltransferase activity. Results The imaging studies of fluorophore-labeled cholesteryl glucosides pinpointed their intracellular localization in AGS cells. The result indicated that CAG enhances the internalization of H. pylori in AGS cells. Particularly, CAG, instead of CG and CPG, is able to augment the autophagy response induced by H. pylori. How CAG participates in the autophagy process is multifaceted. CAG was found to intervene in the degradation of autophagosomes and reduce lysosomal biogenesis, supporting the idea that intracellular H. pylori is harbored by autophago-lysosomes in favor of the bacterial survival. Furthermore, we performed the enzyme activity assay of subcellular fractions of H. pylori-infected AGS cells. The analysis showed that the acyltransferase is mainly distributed in autophago-lysosomal compartments. Conclusions Our results support the idea that the acyltransferase is mainly distributed in the subcellular compartment consisting of autophagosomes, late endosomes, and lysosomes, in which the acidic environment is beneficial for the maximal acyltransferase activity. The resulting elevated level of CAG can facilitate bacterial internalization, interfere with the autophagy flux, and causes reduced lysosomal biogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s12929-021-00768-w.
Collapse
Affiliation(s)
- Sasikala Muthusamy
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan.,Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, National Chung-Hsing University and Academia Sinica, Taipei, 11529, Taiwan.,Graduate Institute of Biotechnology, National Chung-Hsing University, Taichung, 40227, Taiwan.,Biotechnology Center, National Chung-Hsing University, Taichung, 40227, Taiwan
| | - Hau-Ming Jan
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
| | - Ming-Yen Hsieh
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
| | - Soumik Mondal
- Department of Applied Chemistry, National Chiao Tung University, Hsin-Chu, 30010, Taiwan
| | - Wen-Chun Liu
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Yi-An Ko
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Wei-Yuan Yang
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan.,Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan
| | - Kwok-Kong Tony Mong
- Department of Applied Chemistry, National Chiao Tung University, Hsin-Chu, 30010, Taiwan
| | - Guang-Chao Chen
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan.,Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan
| | - Chun-Hung Lin
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan. .,Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, National Chung-Hsing University and Academia Sinica, Taipei, 11529, Taiwan. .,Graduate Institute of Biotechnology, National Chung-Hsing University, Taichung, 40227, Taiwan. .,Biotechnology Center, National Chung-Hsing University, Taichung, 40227, Taiwan. .,Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan. .,Department of Chemistry, National Taiwan University, Taipei, 10617, Taiwan.
| |
Collapse
|
7
|
Goel R, Bloch EM, Pirenne F, Al‐Riyami AZ, Crowe E, Dau L, Land K, Townsend M, Jecko T, Rahimi‐Levene N, Patidar G, Josephson CD, Arora S, Vermeulen M, Vrielink H, Montemayor C, Oreh A, Hindawi S, van den Berg K, Serrano K, So‐Osman C, Wood E, Devine DV, Spitalnik SL. ABO blood group and COVID-19: a review on behalf of the ISBT COVID-19 Working Group. Vox Sang 2021; 116:849-861. [PMID: 33578447 PMCID: PMC8014128 DOI: 10.1111/vox.13076] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 12/30/2022]
Abstract
Growing evidence suggests that ABO blood group may play a role in the immunopathogenesis of SARS-CoV-2 infection, with group O individuals less likely to test positive and group A conferring a higher susceptibility to infection and propensity to severe disease. The level of evidence supporting an association between ABO type and SARS-CoV-2/COVID-19 ranges from small observational studies, to genome-wide-association-analyses and country-level meta-regression analyses. ABO blood group antigens are oligosaccharides expressed on red cells and other tissues (notably endothelium). There are several hypotheses to explain the differences in SARS-CoV-2 infection by ABO type. For example, anti-A and/or anti-B antibodies (e.g. present in group O individuals) could bind to corresponding antigens on the viral envelope and contribute to viral neutralization, thereby preventing target cell infection. The SARS-CoV-2 virus and SARS-CoV spike (S) proteins may be bound by anti-A isoagglutinins (e.g. present in group O and group B individuals), which may block interactions between virus and angiotensin-converting-enzyme-2-receptor, thereby preventing entry into lung epithelial cells. ABO type-associated variations in angiotensin-converting enzyme-1 activity and levels of von Willebrand factor (VWF) and factor VIII could also influence adverse outcomes, notably in group A individuals who express high VWF levels. In conclusion, group O may be associated with a lower risk of SARS-CoV-2 infection and group A may be associated with a higher risk of SARS-CoV-2 infection along with severe disease. However, prospective and mechanistic studies are needed to verify several of the proposed associations. Based on the strength of available studies, there are insufficient data for guiding policy in this regard.
Collapse
Affiliation(s)
- Ruchika Goel
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMDUSA
- Division of Hematology/OncologySimmons Cancer Institute at SIU School of Medicine and Mississippi Valley Regional Blood CenterSpringfieldILUSA
| | - Evan M. Bloch
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - France Pirenne
- Etablissement Français du Sang Ile de FranceHôpital Henri MondorCréteilFrance
| | - Arwa Z. Al‐Riyami
- Department of HematologySultan Qaboos University HospitalMuscatSultanate of Oman
| | - Elizabeth Crowe
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Laetitia Dau
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Kevin Land
- VitalantScottsdaleAZUSA
- Department of PathologyUT, San AntonioTXUSA
| | | | | | | | - Gopal Patidar
- Department of Transfusion MedicineAll India Institute of Medical SciencesNew DelhiIndia
| | | | - Satyam Arora
- Super Speciality Pediatric Hospital and Post Graduate Teaching InstituteNoidaIndia
| | - Marion Vermeulen
- The South African National Blood ServicePort ElizabethSouth Africa
| | - Hans Vrielink
- Dept Unit Transfusion MedicineSanquin BloodbankAmsterdamthe Netherlands
| | | | - Adaeze Oreh
- National Blood Transfusion ServiceDepartment of Hospital ServicesFederal Ministry of HealthAbujaNigeria
| | | | - Karin van den Berg
- Translational Research DepartmentMedical DivisionSouth African National Blood ServicePort ElizabethSouth Africa
- Division of Clinical HaematologyDepartment of MedicineUniversity of Cape TownCape TownSouth Africa
| | - Katherine Serrano
- Canadian Blood ServicesVancouverBCCanada
- Department of Pathology & Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
| | - Cynthia So‐Osman
- Dept Unit Transfusion MedicineSanquin BloodbankAmsterdamthe Netherlands
- Dept. of HaematologyErasmus Medical CenterRotterdamthe Netherlands
| | | | - Dana V. Devine
- Canadian Blood ServicesVancouverBCCanada
- Department of Pathology & Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
| | | | | |
Collapse
|
8
|
Giampaoli O, Conta G, Calvani R, Miccheli A. Can the FUT2 Non-secretor Phenotype Associated With Gut Microbiota Increase the Children Susceptibility for Type 1 Diabetes? A Mini Review. Front Nutr 2020; 7:606171. [PMID: 33425974 PMCID: PMC7785815 DOI: 10.3389/fnut.2020.606171] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
The global toll of type 1 diabetes (T1D) has steadily increased over the last decades. It is now widely acknowledged that T1D pathophysiology is more complex than expected. Indeed, a multifaceted interplay between genetic, metabolic, inflammatory and environmental factors exists that leads to heterogeneous clinical manifestations across individuals. Children with non-secretor phenotype and those affected by T1D share low abundance of bifidobacteria, low content of short-chain fatty acids, intestinal phosphatase alkaline and a high incidence of inflammatory bowel diseases. In this context, host-gut microbiota dyad may represent a relevant contributor to T1D development and progression due to its crucial role in shaping host immunity and susceptibility to autoimmune conditions. The FUT2 gene is responsible for the composition and functional properties of glycans in mucosal tissues and bodily secretions, including human milk. FUT2 polymorphisms may profoundly influence gut microbiota composition and host susceptibility to viral infections and chronic inflammatory disease. In this minireview, the possible interplay between mothers' phenotype, host FUT2 genetic background and gut microbiota composition will be discussed in perspective of the T1D onset. The study of FUT2-gut microbiota interaction may add a new piece on the puzzling T1D etiology and unveil novel targets of intervention to contrast T1D development and progression. Dietary interventions, including the intake of α-(1, 2)-fucosyl oligosaccharides in formula milk and the use of specific prebiotics and probiotics, could be hypothesized.
Collapse
Affiliation(s)
- Ottavia Giampaoli
- Department of Chemistry, Sapienza University of Rome, Rome, Italy.,NMR-Based Metabolomics Laboratory (NMLab), Sapienza University of Rome, Rome, Italy
| | - Giorgia Conta
- Department of Chemistry, Sapienza University of Rome, Rome, Italy.,NMR-Based Metabolomics Laboratory (NMLab), Sapienza University of Rome, Rome, Italy
| | - Riccardo Calvani
- Fondazione Policlinico Universitario 'Agostino Gemelli' IRCCS, Rome, Italy
| | - Alfredo Miccheli
- NMR-Based Metabolomics Laboratory (NMLab), Sapienza University of Rome, Rome, Italy.,Department of Environmental Biology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
9
|
González-Morelo KJ, Vega-Sagardía M, Garrido D. Molecular Insights Into O-Linked Glycan Utilization by Gut Microbes. Front Microbiol 2020; 11:591568. [PMID: 33224127 PMCID: PMC7674204 DOI: 10.3389/fmicb.2020.591568] [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: 08/04/2020] [Accepted: 10/20/2020] [Indexed: 12/16/2022] Open
Abstract
O-linked glycosylation is a post-translational modification found mainly in eukaryotic cells, which covalently attaches oligosaccharides to secreted proteins in certain threonine or serine residues. Most of O-glycans have N-acetylgalactosamine (GalNAc) as a common core. Several glycoproteins, such as mucins (MUCs), immunoglobulins, and caseins are examples of O-glycosylated structures. These glycans are further elongated with other monosaccharides and sulfate groups. Some of them could be found in dairy foods, while others are produced endogenously, in both cases interacting with the gut microbiota. Interestingly, certain gut microbes can access, release, and consume O-linked glycans as a carbon source. Among these, Akkermansia muciniphila, Bifidobacterium bifidum, and Bacteroides thetaiotaomicron are prominent O-linked glycan utilizers. Their consumption strategies include specialized α-fucosidases and α-sialidases, in addition to endo-α-N-acetylgalactosaminidases that release galacto-N-biose (GNB) from peptides backbones. O-linked glycan utilization by certain gut microbes represents an important niche that allows them to predominate and modulate host responses such as inflammation. Here, we focus on the distinct molecular mechanisms of consumption of O-linked GalNAc glycans by prominent gut microbes, especially from mucin and casein glycomacropeptide (GMP), highlighting the potential of these structures as emerging prebiotics.
Collapse
Affiliation(s)
| | | | - Daniel Garrido
- Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
10
|
Butyrate producing colonic Clostridiales metabolise human milk oligosaccharides and cross feed on mucin via conserved pathways. Nat Commun 2020; 11:3285. [PMID: 32620774 PMCID: PMC7335108 DOI: 10.1038/s41467-020-17075-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 06/11/2020] [Indexed: 01/07/2023] Open
Abstract
The early life human gut microbiota exerts life-long health effects on the host, but the mechanisms underpinning its assembly remain elusive. Particularly, the early colonization of Clostridiales from the Roseburia-Eubacterium group, associated with protection from colorectal cancer, immune- and metabolic disorders is enigmatic. Here, we describe catabolic pathways that support the growth of Roseburia and Eubacterium members on distinct human milk oligosaccharides (HMOs). The HMO pathways, which include enzymes with a previously unknown structural fold and specificity, were upregulated together with additional glycan-utilization loci during growth on selected HMOs and in co-cultures with Akkermansia muciniphila on mucin, suggesting an additional role in enabling cross-feeding and access to mucin O-glycans. Analyses of 4599 Roseburia genomes underscored the preponderance and diversity of the HMO utilization loci within the genus. The catabolism of HMOs by butyrate-producing Clostridiales may contribute to the competitiveness of this group during the weaning-triggered maturation of the microbiota.
Collapse
|
11
|
Jan HM, Chen YC, Yang TC, Ong LL, Chang CC, Muthusamy S, Abera AB, Wu MS, Gervay-Hague J, Mong KKT, Lin CH. Cholesteryl α-D-glucoside 6-acyltransferase enhances the adhesion of Helicobacter pylori to gastric epithelium. Commun Biol 2020; 3:120. [PMID: 32170208 PMCID: PMC7069968 DOI: 10.1038/s42003-020-0855-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 02/20/2020] [Indexed: 12/18/2022] Open
Abstract
Helicobacter pylori, the most common etiologic agent of gastric diseases including gastric cancer, is auxotrophic for cholesterol and has to hijack it from gastric epithelia. Upon uptake, the bacteria convert cholesterol to cholesteryl 6′-O-acyl-α-D-glucopyranoside (CAG) to promote lipid raft clustering in the host cell membranes. However, how CAG appears in the host to exert the pathogenesis still remains ambiguous. Herein we identified hp0499 to be the gene of cholesteryl α-D-glucopyranoside acyltransferase (CGAT). Together with cholesteryl glucosyltransferase (catalyzing the prior step), CGAT is secreted via outer membrane vesicles to the host cells for direct synthesis of CAG. This significantly enhances lipid rafts clustering, gathers adhesion molecules (including Lewis antigens and integrins α5, β1), and promotes more bacterial adhesion. Furthermore, the clinically used drug amiodarone was shown as a potent inhibitor of CGAT to effectively reduce the bacterial adhesion, indicating that CGAT is a potential target of therapeutic intervention. Jan et al. identify cholesteryl α-D- glucopyranoside acyltransferase as a key enzyme in Helicobacter pylori’s synthesis of cholesteryl 6’-O-acyl-α-D-glucopyranoside, which promotes bacterial adhesion. This study provides insights into the H. pylori-induced pathogenesis and therapeutic strategies against it.
Collapse
Affiliation(s)
- Hau-Ming Jan
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academic Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
| | - Yi-Chi Chen
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academic Road Section 2, Nan-Kang, Taipei, 11529, Taiwan.,Department of Chemistry and Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan
| | - Tsai-Chen Yang
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academic Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
| | - Lih-Lih Ong
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academic Road Section 2, Nan-Kang, Taipei, 11529, Taiwan.,Department of Applied Chemistry, National Chiao Tung University, Hsin-Chu, 30010, Taiwan.,Sustainable Chemical Science and Technology, Taiwan International Graduate Program, Academia Sinica and National Chiao Tung University, Taipei, 11529, Taiwan
| | - Chia-Chen Chang
- Department of Applied Chemistry, National Chiao Tung University, Hsin-Chu, 30010, Taiwan
| | - Sasikala Muthusamy
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academic Road Section 2, Nan-Kang, Taipei, 11529, Taiwan.,Molecular and Biological Agricultural Sciences, Taiwan International Graduate Program, Academia Sinica and National Chung-Hsing University, Taipei, 11529, Taiwan.,Graduate Institute of Biotechnology, National Chung-Hsing University, Taichung, 40227, Taiwan
| | - Andualem Bahiru Abera
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academic Road Section 2, Nan-Kang, Taipei, 11529, Taiwan.,Molecular and Biological Agricultural Sciences, Taiwan International Graduate Program, Academia Sinica and National Chung-Hsing University, Taipei, 11529, Taiwan.,Graduate Institute of Biotechnology, National Chung-Hsing University, Taichung, 40227, Taiwan
| | - Ming-Shiang Wu
- Division of Gastroenterology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, 10002, Taiwan
| | | | - Kwok-Kong Tony Mong
- Department of Applied Chemistry, National Chiao Tung University, Hsin-Chu, 30010, Taiwan.
| | - Chun-Hung Lin
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academic Road Section 2, Nan-Kang, Taipei, 11529, Taiwan. .,Department of Chemistry and Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan.
| |
Collapse
|
12
|
Pap A, Tasnadi E, Medzihradszky KF, Darula Z. Novel O-linked sialoglycan structures in human urinary glycoproteins. Mol Omics 2020; 16:156-164. [PMID: 32022078 DOI: 10.1039/c9mo00160c] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Glycopeptides represent cross-linked structures between chemically and physically different biomolecules. Mass spectrometric analysis of O-glycopeptides may reveal the identity of the peptide, the composition of the glycan and even the connection between certain sugar units, but usually only the combination of different MS/MS techniques provides sufficient information for reliable assignment. Currently, HCD analysis followed by diagnostic sugar fragment-triggered ETD or EThcD experiments is the most promising data acquisition protocol. However, the information content of the different MS/MS data is handled separately by search engines. We are convinced that these data should be used in concert, as we demonstrate in the present study. First, glycopeptides bearing the most common glycans can be identified from EThcD and/or HCD data. Then, searching for Y0 (the gas-phase deglycosylated peptide) in HCD spectra, the potential glycoforms of these glycopeptides could be lined up. Finally, these spectra and the corresponding EThcD data can be used to verify or discard the tentative assignments and to obtain further structural information about the glycans. We present 18 novel human urinary sialoglycan structures deciphered using this approach. To accomplish this in an automated fashion further software development is necessary.
Collapse
Affiliation(s)
- Adam Pap
- Laboratory of Proteomics Research, Biological Research Centre, Temesvari krt. 62, H-6726 Szeged, Hungary.
| | | | | | | |
Collapse
|
13
|
Heuer F, Stürmer R, Heuer J, Kalinski T, Lemke A, Meyer F, Hoffmann W. Different Forms of TFF2, A Lectin of the Human Gastric Mucus Barrier: In Vitro Binding Studies. Int J Mol Sci 2019; 20:ijms20235871. [PMID: 31771101 PMCID: PMC6928932 DOI: 10.3390/ijms20235871] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/13/2019] [Accepted: 11/21/2019] [Indexed: 12/19/2022] Open
Abstract
Trefoil factor family 2 (TFF2) and the mucin MUC6 are co-secreted from human gastric and duodenal glands. TFF2 binds MUC6 as a lectin and is a constituent of the gastric mucus. Herein, we investigated human gastric extracts by FPLC and identified mainly high- but also low-molecular-mass forms of TFF2. From the high-molecular-mass forms, TFF2 can be completely released by boiling in SDS or by harsh denaturing extraction. The low-molecular-mass form representing monomeric TFF2 can be washed out in part from gastric mucosa specimens with buffer. Overlay assays with radioactively labeled TFF2 revealed binding to the mucin MUC6 and not MUC5AC. This binding is modulated by Ca2+ and can be blocked by the lectin GSA-II and the monoclonal antibody HIK1083. TFF2 binding was also inhibited by Me-β-Gal, but not the α anomer. Thus, both the α1,4GlcNAc as well as the juxtaperipheral β-galactoside residues of the characteristic GlcNAcα1→4Galβ1→R moiety of human MUC6 are essential for TFF2 binding. Furthermore, there are major differences in the TFF2 binding characteristics when human is compared with the porcine system. Taken together, TFF2 appears to fulfill an important role in stabilizing the inner insoluble gastric mucus barrier layer, particularly by its binding to the mucin MUC6.
Collapse
Affiliation(s)
- Franziska Heuer
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - René Stürmer
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Jörn Heuer
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Thomas Kalinski
- Institute of Pathology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Antje Lemke
- Institute of Pathology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Frank Meyer
- Department of Surgery, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Werner Hoffmann
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
- Correspondence:
| |
Collapse
|
14
|
MacMillan JL, Vicaretti SD, Noyovitz B, Xing X, Low KE, Inglis GD, Zaytsoff SJ, Boraston AB, Smith SP, Uwiera RR, Selinger LB, Zandberg WF, Abbott DW. Structural analysis of broiler chicken small intestinal mucin O-glycan modification by Clostridium perfringens. Poult Sci 2019; 98:5074-5088. [DOI: 10.3382/ps/pez297] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/03/2019] [Indexed: 12/12/2022] Open
|
15
|
Rahfeld P, Wardman JF, Mehr K, Huff D, Morgan-Lang C, Chen HM, Hallam SJ, Withers SG. Prospecting for microbial α- N-acetylgalactosaminidases yields a new class of GH31 O-glycanase. J Biol Chem 2019; 294:16400-16415. [PMID: 31530641 DOI: 10.1074/jbc.ra119.010628] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/16/2019] [Indexed: 12/18/2022] Open
Abstract
α-Linked GalNAc (α-GalNAc) is most notably found at the nonreducing terminus of the blood type-determining A-antigen and as the initial point of attachment to the peptide backbone in mucin-type O-glycans. However, despite their ubiquity in saccharolytic microbe-rich environments such as the human gut, relatively few α-N-acetylgalactosaminidases are known. Here, to discover and characterize novel microbial enzymes that hydrolyze α-GalNAc, we screened small-insert libraries containing metagenomic DNA from the human gut microbiome. Using a simple fluorogenic glycoside substrate, we identified and characterized a glycoside hydrolase 109 (GH109) that is active on blood type A-antigen, along with a new subfamily of glycoside hydrolase 31 (GH31) that specifically cleaves the initial α-GalNAc from mucin-type O-glycans. This represents a new activity in this GH family and a potentially useful new enzyme class for analysis or modification of O-glycans on protein or cell surfaces.
Collapse
Affiliation(s)
- Peter Rahfeld
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada .,Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Jacob F Wardman
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Life Sciences Centre, Vancouver, British Columbia V6T 1Z3, Canada
| | - Kevin Mehr
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.,Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Drew Huff
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.,Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Connor Morgan-Lang
- Department of Microbiology and Immunology, University of British Columbia, Life Sciences Centre, Vancouver, British Columbia V6T 1Z3, Canada.,Graduate Program in Bioinformatics, University of British Columbia, Genome Sciences Centre, Vancouver, British Columbia V5Z 4S6, Canada
| | - Hong-Ming Chen
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Steven J Hallam
- Department of Microbiology and Immunology, University of British Columbia, Life Sciences Centre, Vancouver, British Columbia V6T 1Z3, Canada.,Graduate Program in Bioinformatics, University of British Columbia, Genome Sciences Centre, Vancouver, British Columbia V5Z 4S6, Canada.,ECOSCOPE Training Program, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.,Peter Wall Institute for Advanced Studies, University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada
| | - Stephen G Withers
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada .,Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Life Sciences Centre, Vancouver, British Columbia V6T 1Z3, Canada
| |
Collapse
|
16
|
Etienne-Mesmin L, Chassaing B, Desvaux M, De Paepe K, Gresse R, Sauvaitre T, Forano E, de Wiele TV, Schüller S, Juge N, Blanquet-Diot S. Experimental models to study intestinal microbes–mucus interactions in health and disease. FEMS Microbiol Rev 2019; 43:457-489. [DOI: 10.1093/femsre/fuz013] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/31/2019] [Indexed: 02/06/2023] Open
Abstract
ABSTRACT
A close symbiotic relationship exists between the intestinal microbiota and its host. A critical component of gut homeostasis is the presence of a mucus layer covering the gastrointestinal tract. Mucus is a viscoelastic gel at the interface between the luminal content and the host tissue that provides a habitat to the gut microbiota and protects the intestinal epithelium. The review starts by setting up the biological context underpinning the need for experimental models to study gut bacteria-mucus interactions in the digestive environment. We provide an overview of the structure and function of intestinal mucus and mucins, their interactions with intestinal bacteria (including commensal, probiotics and pathogenic microorganisms) and their role in modulating health and disease states. We then describe the characteristics and potentials of experimental models currently available to study the mechanisms underpinning the interaction of mucus with gut microbes, including in vitro, ex vivo and in vivo models. We then discuss the limitations and challenges facing this field of research.
Collapse
Affiliation(s)
- Lucie Etienne-Mesmin
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Benoit Chassaing
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303 , USA
- Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave, Atlanta, GA 30303 , USA
| | - Mickaël Desvaux
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Kim De Paepe
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Raphaële Gresse
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Thomas Sauvaitre
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Evelyne Forano
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Stephanie Schüller
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR7UQ, United Kingdom
| | - Nathalie Juge
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR7UQ, United Kingdom
| | - Stéphanie Blanquet-Diot
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| |
Collapse
|
17
|
Stowell CP, Stowell SR. Biologic roles of the ABH and Lewis histo-blood group antigens Part I: infection and immunity. Vox Sang 2019; 114:426-442. [PMID: 31070258 DOI: 10.1111/vox.12787] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 04/08/2019] [Accepted: 04/10/2019] [Indexed: 12/22/2022]
Abstract
The ABH and Lewis antigens were among the first of the human red blood cell polymorphisms to be identified and, in the case of the former, play a dominant role in transfusion and transplantation. But these two therapies are largely twentieth century innovations, and the ABH and related carbohydrate antigens are not only expressed on a very wide range of human tissues, but were present in primates long before modern humans evolved. Although we have learned a great deal about the biochemistry and genetics of these structures, the biological roles that they play in human health and disease are incompletely understood. This review and its companion, to appear in a later issue of Vox Sanguinis, will focus on a few of the biologic and pathologic processes which appear to be affected by histo-blood group phenotype. The first of the two reviews will explore the interactions of two bacteria with the ABH and Lewis glycoconjugates of their human host cells, and describe the possible connections between the immune response of the human host to infection and the development of the AB-isoagglutinins. The second review will describe the relationship between ABO phenotype and thromboembolic disease, cardio-vascular disease states, and general metabolism.
Collapse
Affiliation(s)
- Christopher P Stowell
- Blood Transfusion Service, Massachusetts General Hospital, Boston, MA, USA.,Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Sean R Stowell
- Center for Apheresis, Center for Transfusion and Cellular Therapies, Emory Hospital, Emory University School of Medicine, Atlanta, GA, USA.,Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
18
|
Newton GR, Lewis SK, Avendano J, Williams EA, Ribeiro FRB, Nuti LC, Foxworth WB, Ing NH. Fucosyltransferase gene expression in goat endometrium during the estrous cycle and early pregnancy. Theriogenology 2019; 132:118-127. [PMID: 31022601 DOI: 10.1016/j.theriogenology.2019.04.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 04/10/2019] [Accepted: 04/15/2019] [Indexed: 11/28/2022]
Abstract
Regulation of the expression of the alpha(1,2)fucosyltransferase (FUT) genes and their enzymatic products, including the H-type 1 antigen (HT1), on the luminal surface of the uterus is believed to be critical for establishment of pregnancy in mammals. The FUT1 gene is a marker for conception rates in dairy cows and HT1 is a marker for uterine receptivity in rodents. To determine the spatiotemporal expression patterns of FUT1 and FUT2 genes in goats, endometrial tissues were obtained on six days spanning the estrous cycle (Days 5, 11, 13, 15, 17 and 19) and seven days spanning early pregnancy (Days 5, 11, 13, 15, 17, 19 and 25). In all data, we found no effect of status (cyclic or pregnant; P > 0.1) and pooled data where appropriate. We cloned FUT1 cDNA from goat endometrium and made probes from it for Northern and slot blot analyses. The analyses indicated that FUT1 gene expression was high until Day 13, and then declined. In situ hybridization revealed a change in the cell-specificity of FUT1 gene expression over the estrous cycle and early pregnancy. In situ hybridization signal intensity scores indicated that FUT1 expression by uterine epithelium was high on Day 5, moderate on Day 11, and minimal on subsequent days. In situ hybridization signals in uterine glandular epithelial cells remained high from Day 5 to Day 13, with weaker signals thereafter. Quantitative reverse transcription-PCR (RT-qPCR) assays were used for quantitation of FUT1 and FUT2 mRNAs. Quantitative RT-qPCR data were generated from endometrium collected from cyclic and pregnant animals on Days 5, 11 and 17. Relative levels of FUT1 mRNA were high on Days 5 and 11, but then fell 5-fold by Day 17 (P < 0.01). FUT2 mRNA concentrations were below the accurate detectable limit of the assay. High levels of HT1 were observed on the apical surface of uterine luminal epithelia on Days 5, 15, 17 and 19, with much lower levels on Days 11 and 13. Thus, data suggests that FUT1 is the primary enzyme responsible for the high levels of HT1 antigen present on the uterine luminal epithelium between Days 5 and 11 of the estrous cycle and early pregnancy. But changes in the expression of the FUT1 gene does not directly correlate to HT1 staining, which increased from Day 13-15. Future studies are required to understand the regulation of the HT1 antigen on the luminal surface of endometrium.
Collapse
Affiliation(s)
- G R Newton
- Prairie View A&M University, Cooperative Agricultural Research Center, Prairie View, TX, 77446-0519, USA.
| | - S K Lewis
- Prairie View A&M University, Cooperative Agricultural Research Center, Prairie View, TX, 77446-0519, USA
| | - J Avendano
- Prairie View A&M University, Cooperative Agricultural Research Center, Prairie View, TX, 77446-0519, USA
| | - E A Williams
- Prairie View A&M University, Cooperative Agricultural Research Center, Prairie View, TX, 77446-0519, USA
| | - F R B Ribeiro
- Prairie View A&M University, Cooperative Agricultural Research Center, Prairie View, TX, 77446-0519, USA
| | - L C Nuti
- Prairie View A&M University, Cooperative Agricultural Research Center, Prairie View, TX, 77446-0519, USA
| | - W B Foxworth
- Prairie View A&M University, Cooperative Agricultural Research Center, Prairie View, TX, 77446-0519, USA
| | - N H Ing
- Department of Animal Science, Texas A&M University, 2471 TAMU, College Station, TX, 77843-2471, USA
| |
Collapse
|
19
|
Chakrani Z, Robinson K, Taye B. Association Between ABO Blood Groups and Helicobacter pylori Infection: A Meta-Analysis. Sci Rep 2018; 8:17604. [PMID: 30514875 PMCID: PMC6279815 DOI: 10.1038/s41598-018-36006-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 11/14/2018] [Indexed: 12/11/2022] Open
Abstract
There is no consensus among the existing literature on the relationship between ABO blood groups and risk of Helicobacter pylori infection. However, histo-blood group carbohydrates are proposed to influence the risk of acquiring this pathogen via effects on adhesion to the gastric mucosa. The objective of this meta-analysis was to evaluate the association between ABO blood groups and H. pylori infection. All relevant epidemiological studies published in English (up to October 2017) was retrieved through an extensive systematic literature search of MEDLINE/PubMed databases. Pooled estimates of effects were obtained through the use of fixed and random effects meta-analyses. Individuals with O blood group were more likely to be infected with H. pylori (pooled odds ratio (OR) 1.163; 95% confidence interval (CI) 1.074-1.259; P < 0.001). While individuals with B and AB blood group were less likely to be infected with H. pylori (OR 0.831; 95% CI 0.738-0.935; P = 0.002 and OR 0.709; 95% CI 0.605-0.832; P < 0.001, respectively). The results from this meta-analysis of observational studies suggest an estimated 16.3% increased odds of H. pylori infection amongst individuals with the O blood group. If this observed association is causal, a better understanding of the underlying mechanisms could provide indications to potential prevention strategies for H. pylori infection.
Collapse
Affiliation(s)
- Zakaria Chakrani
- Department of Biology, Colgate University, 13 Oak Dr, Hamilton, 13346, NY, USA
| | - Karen Robinson
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and The University of Nottingham, Nottingham, UK
- Nottingham Digestive Diseases Centre, The University of Nottingham, Nottingham, UK
| | - Bineyam Taye
- Department of Biology, Colgate University, 13 Oak Dr, Hamilton, 13346, NY, USA.
| |
Collapse
|
20
|
Darula Z, Pap Á, Medzihradszky KF. Extended Sialylated O-Glycan Repertoire of Human Urinary Glycoproteins Discovered and Characterized Using Electron-Transfer/Higher-Energy Collision Dissociation. J Proteome Res 2018; 18:280-291. [PMID: 30407017 DOI: 10.1021/acs.jproteome.8b00587] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A relatively novel activation technique, electron-transfer/higher-energy collision dissociation (EThcD) was used in the LC-MS/MS analysis of tryptic glycopeptides enriched with wheat germ agglutinin from human urine samples. We focused on the characterization of mucin-type O-glycopeptides. EThcD in a single spectrum provided information on both the peptide modified and the glycan carried. Unexpectedly, glycan oxonium ions indicated the presence of O-acetyl, and even O-diacetyl-sialic acids. B and Y fragment ions revealed that (i) in core 1 structures the Gal residue featured the O-acetyl-sialic acid, when there was only one in the glycan; (ii) several glycopeptides featured core 1 glycans with disialic acids, in certain instances O-acetylated; (iii) the disialic acid was linked to the GalNAc residue whatever the degree of O-acetylation; (iv) core 2 isomers with a single O-acetyl-sialic acid were chromatographically resolved. Glycan fragmentation also helped to decipher additional core 2 oligosaccharides: a LacdiNAc-like structure, glycans carrying sialyl LewisX/A at different stages of O-acetylation, and blood antigens. A sialo core 3 structure was also identified. We believe this is the first study when such structures were characterized from a very complex mixture and were linked not only to a specific protein, but also the sites of modifications have been determined.
Collapse
Affiliation(s)
- Zsuzsanna Darula
- Biological Research Centre of the Hungarian Academy of Sciences , Temesvari krt. 62. , H-6726 Szeged , Hungary
| | - Ádám Pap
- Biological Research Centre of the Hungarian Academy of Sciences , Temesvari krt. 62. , H-6726 Szeged , Hungary.,Doctoral School in Biology, Faculty of Science and Informatics , University of Szeged , Kozep fasor 52. , H-6726 Szeged , Hungary
| | - Katalin F Medzihradszky
- Biological Research Centre of the Hungarian Academy of Sciences , Temesvari krt. 62. , H-6726 Szeged , Hungary
| |
Collapse
|
21
|
El Jellas K, Johansson BB, Fjeld K, Antonopoulos A, Immervoll H, Choi MH, Hoem D, Lowe ME, Lombardo D, Njølstad PR, Dell A, Mas E, Haslam SM, Molven A. The mucinous domain of pancreatic carboxyl-ester lipase (CEL) contains core 1/core 2 O-glycans that can be modified by ABO blood group determinants. J Biol Chem 2018; 293:19476-19491. [PMID: 30315106 DOI: 10.1074/jbc.ra118.001934] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 09/11/2018] [Indexed: 12/20/2022] Open
Abstract
Carboxyl-ester lipase (CEL) is a pancreatic fat-digesting enzyme associated with human disease. Rare mutations in the CEL gene cause a syndrome of pancreatic exocrine and endocrine dysfunction denoted MODY8, whereas a recombined CEL allele increases the risk for chronic pancreatitis. Moreover, CEL has been linked to pancreatic ductal adenocarcinoma (PDAC) through a postulated oncofetal CEL variant termed feto-acinar pancreatic protein (FAPP). The monoclonal antibody mAb16D10 was previously reported to detect a glycotope in the highly O-glycosylated, mucin-like C terminus of CEL/FAPP. We here assessed the expression of human CEL in malignant pancreatic lesions and cell lines. CEL was not detectably expressed in neoplastic cells, implying that FAPP is unlikely to be a glycoisoform of CEL in pancreatic cancer. Testing of the mAb16D10 antibody in glycan microarrays then demonstrated that it recognized structures containing terminal GalNAc-α1,3(Fuc-α1,2)Gal (blood group A antigen) and also repeated protein sequences containing GalNAc residues linked to Ser/Thr (Tn antigen), findings that were supported by immunostainings of human pancreatic tissue. To examine whether the CEL glycoprotein might be modified by blood group antigens, we used high-sensitivity MALDI-TOF MS to characterize the released O-glycan pool of CEL immunoprecipitated from human pancreatic juice. We found that the O-glycome of CEL consisted mainly of core 1/core 2 structures with a composition depending on the subject's FUT2 and ABO gene polymorphisms. Thus, among digestive enzymes secreted by the pancreas, CEL is a glycoprotein with some unique characteristics, supporting the view that it could serve additional biological functions to its cholesteryl esterase activity in the duodenum.
Collapse
Affiliation(s)
- Khadija El Jellas
- From the Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway.,Department of Pathology, Haukeland University Hospital, N-5021 Bergen, Norway.,KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway
| | - Bente B Johansson
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway.,Department of Pediatrics and Adolescent Medicine, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Karianne Fjeld
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway.,Center for Medical Genetics, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Aristotelis Antonopoulos
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - Heike Immervoll
- From the Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway.,Department of Pathology, Ålesund Hospital, N-6017 Ålesund, Norway
| | - Man H Choi
- From the Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway.,Department of Pathology, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Dag Hoem
- Department of Gastrointestinal Surgery, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Mark E Lowe
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110, and
| | - Dominique Lombardo
- INSERM, CRO2, Center for Research in Biological Oncology and Oncopharmacology, Aix-Marseille University, 13284 Marseille Cedex 07, France
| | - Pål R Njølstad
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway.,Department of Pediatrics and Adolescent Medicine, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Anne Dell
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - Eric Mas
- INSERM, CRO2, Center for Research in Biological Oncology and Oncopharmacology, Aix-Marseille University, 13284 Marseille Cedex 07, France
| | - Stuart M Haslam
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - Anders Molven
- From the Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway, .,Department of Pathology, Haukeland University Hospital, N-5021 Bergen, Norway.,KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway
| |
Collapse
|
22
|
Zúñiga M, Monedero V, Yebra MJ. Utilization of Host-Derived Glycans by Intestinal Lactobacillus and Bifidobacterium Species. Front Microbiol 2018; 9:1917. [PMID: 30177920 PMCID: PMC6109692 DOI: 10.3389/fmicb.2018.01917] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/30/2018] [Indexed: 12/27/2022] Open
Abstract
Members of the genus Lactobacillus are commonly found at the gastrointestinal tract and other mucosal surfaces of humans. This genus includes various species with a great number of potentially probiotic bacteria. Other often-used probiotic species belong to Bifidobacterium, a genus almost exclusively associated with the gut. As probiotics must survive and be metabolically active at their target sites, namely host mucosal surfaces, consumption of host-produced glycans is a key factor for their survival and activity. The ability to metabolize glycans such as human milk oligosaccharides (HMOs), glycosaminoglycans and the glycan moieties of glycoproteins and glycolipids found at the mucosal surfaces grants a competitive advantage to lactobacilli and bifidobacteria. The analyses of the great number of sequenced genomes from these bacteria have revealed that many of them encode a wide assortment of genes involved in the metabolism and transport of carbohydrates, including several glycoside hydrolases required for metabolizing the carbohydrate moieties of mucins and HMOs. Here, the current knowledge on the genetic mechanisms, known catabolic pathways and biochemical properties of enzymes involved in the utilization of host-produced glycans by lactobacilli and bifidobacteria will be summarized.
Collapse
Affiliation(s)
- Manuel Zúñiga
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, Instituto de Agroquímica y Tecnología de Alimentos-Consejo Superior de Investigaciones Científicas, Valencia, Spain
| | - Vicente Monedero
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, Instituto de Agroquímica y Tecnología de Alimentos-Consejo Superior de Investigaciones Científicas, Valencia, Spain
| | - María J Yebra
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, Instituto de Agroquímica y Tecnología de Alimentos-Consejo Superior de Investigaciones Científicas, Valencia, Spain
| |
Collapse
|
23
|
Fernandez N, Wrzosek L, Radziwill-Bienkowska JM, Ringot-Destrez B, Duviau MP, Noordine ML, Laroute V, Robert V, Cherbuy C, Daveran-Mingot ML, Cocaign-Bousquet M, Léonard R, Robbe-Masselot C, Rul F, Ogier-Denis E, Thomas M, Mercier-Bonin M. Characterization of Mucus-Related Properties of Streptococcus thermophilus: From Adhesion to Induction. Front Physiol 2018; 9:980. [PMID: 30087622 PMCID: PMC6067005 DOI: 10.3389/fphys.2018.00980] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/03/2018] [Indexed: 12/21/2022] Open
Abstract
Mucus is a major component of the intestinal barrier involved both in the protection of the host and the fitness of commensals of the gut. Streptococcus thermophilus is consumed world-wide in fermented dairy products and is also recognized as a probiotic, as its consumption is associated with improved lactose digestion. We determined the overall effect of S. thermophilus on the mucus by evaluating its ability to adhere, degrade, modify, or induce the production of mucus and/or mucins. Adhesion was analyzed in vitro using two types of mucins (from pig or human biopsies) and mucus-producing intestinal HT29-MTX cells. The induction of mucus was characterized in two different rodent models, in which S. thermophilus is the unique bacterial species in the digestive tract or transited as a sub-dominant bacterium through a complex microbiota. S. thermophilus LMD-9 and LMG18311 strains did not grow in sugars used to form mucins as the sole carbon source and displayed weak binding to mucus/mucins relative to the highly adhesive TIL448 Lactococcus lactis. The presence of S. thermophilus as the unique bacteria in the digestive tract of gnotobiotic rats led to accumulation of lactate and increased the number of Alcian-Blue positive goblet cells and the amount of the mucus-inducer KLF4 transcription factor. Lactate significantly increased KLF4 protein levels in HT29-MTX cells. Introduction of S. thermophilusvia transit as a sub-dominant bacterium (103 CFU/g feces) in a complex endogenous microbiota resulted in a slight increase in lactate levels in the digestive tract, no induction of overall mucus production, and moderate induction of sulfated mucin production. We thus show that although S. thermophilus is a poor mucus-adhesive bacterium, it can promote mucus pathway at least in part by producing lactate in the digestive tract.
Collapse
Affiliation(s)
- Neïké Fernandez
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Laura Wrzosek
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | | | - Belinda Ringot-Destrez
- Université de Lille, Lille, France.,USTL, UGSF, IFR 147, CNRS, Villeneuve-d'Ascq, France.,UMR 8576, Unité de Glycobiologie Structurale et Fonctionnelle, Villeneuve-d'Ascq, France
| | | | - Marie-Louise Noordine
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Valérie Laroute
- LISBP, CNRS, INRA, INSA, Université de Toulouse, Toulouse, France
| | - Véronique Robert
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Claire Cherbuy
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | | | | | - Renaud Léonard
- Université de Lille, Lille, France.,USTL, UGSF, IFR 147, CNRS, Villeneuve-d'Ascq, France.,UMR 8576, Unité de Glycobiologie Structurale et Fonctionnelle, Villeneuve-d'Ascq, France
| | - Catherine Robbe-Masselot
- Université de Lille, Lille, France.,USTL, UGSF, IFR 147, CNRS, Villeneuve-d'Ascq, France.,UMR 8576, Unité de Glycobiologie Structurale et Fonctionnelle, Villeneuve-d'Ascq, France
| | - Françoise Rul
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Eric Ogier-Denis
- INSERM, Research Centre of Inflammation BP 416, Paris, France.,University Paris Diderot, Sorbonne Paris Cité, Paris, France.,Laboratory of Excellence Labex INFLAMEX, Université Sorbonne Paris Cité, Paris, France
| | - Muriel Thomas
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Muriel Mercier-Bonin
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
24
|
Talbot P, Radziwill-Bienkowska JM, Kamphuis JBJ, Steenkeste K, Bettini S, Robert V, Noordine ML, Mayeur C, Gaultier E, Langella P, Robbe-Masselot C, Houdeau E, Thomas M, Mercier-Bonin M. Food-grade TiO 2 is trapped by intestinal mucus in vitro but does not impair mucin O-glycosylation and short-chain fatty acid synthesis in vivo: implications for gut barrier protection. J Nanobiotechnology 2018; 16:53. [PMID: 29921300 PMCID: PMC6009062 DOI: 10.1186/s12951-018-0379-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/14/2018] [Indexed: 12/15/2022] Open
Abstract
Background Titanium dioxide (TiO2) particles are commonly used as a food additive (E171 in the EU) for its whitening and opacifying properties. However, the risk of gut barrier disruption is an increasing concern because of the presence of a nano-sized fraction. Food-grade E171 may interact with mucus, a gut barrier protagonist still poorly explored in food nanotoxicology. To test this hypothesis, a comprehensive approach was performed to evaluate in vitro and in vivo interactions between TiO2 and intestinal mucus, by comparing food-grade E171 with NM-105 (Aeroxyde P25) OECD reference nanomaterial. Results We tested E171-trapping properties of mucus in vitro using HT29-MTX intestinal epithelial cells. Time-lapse confocal laser scanning microscopy was performed without labeling to avoid modification of the particle surface. Near-UV irradiation of E171 TiO2 particles at 364 nm resulted in fluorescence emission in the visible range, with a maximum at 510 nm. The penetration of E171 TiO2 into the mucoid area of HT29-MTX cells was visualized in situ. One hour after exposure, TiO2 particles accumulated inside “patchy” regions 20 µm above the substratum. The structure of mucus produced by HT29-MTX cells was characterized by MUC5AC immunofluorescence staining. The mucus layer was thin and organized into regular “islands” located approximately 20 µm above the substratum. The region-specific trapping of food-grade TiO2 particles was attributed to this mucus patchy structure. We compared TiO2-mediated effects in vivo in rats after acute or sub-chronic oral daily administration of food-grade E171 and NM-105 at relevant exposure levels for humans. Cecal short-chain fatty acid profiles and gut mucin O-glycosylation patterns remained unchanged, irrespective of treatment. Conclusions Food-grade TiO2 is trapped by intestinal mucus in vitro but does not affect mucin O-glycosylation and short-chain fatty acid synthesis in vivo, suggesting the absence of a mucus barrier impairment under “healthy gut” conditions. Electronic supplementary material The online version of this article (10.1186/s12951-018-0379-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pauline Talbot
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | | | - Jasper B J Kamphuis
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Karine Steenkeste
- Institut des Sciences Moléculaires d'Orsay (ISMO), CNRS, Université Paris-Sud, Université Paris-Saclay, Orsay, France
| | - Sarah Bettini
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Véronique Robert
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Marie-Louise Noordine
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Camille Mayeur
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Eric Gaultier
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Philippe Langella
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Catherine Robbe-Masselot
- Univ.lille, CNRS, UMR8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F59000, Lille, France
| | - Eric Houdeau
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Muriel Thomas
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Muriel Mercier-Bonin
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France.
| |
Collapse
|
25
|
Hanisch FG, Hansman GS, Morozov V, Kunz C, Schroten H. Avidity of α-fucose on human milk oligosaccharides and blood group-unrelated oligo/polyfucoses is essential for potent norovirus-binding targets. J Biol Chem 2018; 293:11955-11965. [PMID: 29858242 DOI: 10.1074/jbc.ra117.001369] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 05/11/2018] [Indexed: 12/13/2022] Open
Abstract
There is agreement with respect to norovirus infection routes in humans regarding binding of the pathogen to gastrointestinal epithelia via recognition of blood group-active mucin-typeO-glycans as the initiating and essential event. Among food additives playing a potential role in applications to protect newborns, human milk oligosaccharides (HMOs) as competitors are of major importance. By focusing on fractions of high-molecular mass HMOs with high fucose contents, we attempted to identify the structural elements required for norovirus GII.4 (Sydney 2012, JX459908) capsid binding in neoglycolipid-based arrays. We provide evidence that HMO fractions with the strongest binding capacities contained hepta- to decasaccharides expressing branches with terminal blood group H1 or Lewis-b antigen. H2 antigen, as recognized by UEA-I lectin, is apparently not expressed in high-mass HMOs. Beyond affinity, sterical and valency effects contribute more to virus-like particle binding, as revealed for oligovalent fucose conjugates of α-cyclodextrin and oligofucoses from fucoidan. Accordingly, high-mass HMOs with oligovalent fucose can exhibit stronger binding capacities compared with monovalent fucose HMOs. The above features were revealed for the most clinically relevant and prevalent GII.4 strain and are distinct from other strains, like GII.10 (Vietnam 026, AF504671), which showed a preference for blood group Lewis-a positive glycans.
Collapse
Affiliation(s)
- Franz-Georg Hanisch
- From the Institute of Biochemistry II, Medical Faculty, University of Cologne, Joseph-Stelzmann-Strasse 52, 50931 Köln,
| | - Grant S Hansman
- the Schaller Research Group at University of Heidelberg and DKFZ, and Department of Infectious Diseases, Virology, University of Heidelberg, Im Neuenheimer Feld 242, 69120 Heidelberg
| | - Vasily Morozov
- the Schaller Research Group at University of Heidelberg and DKFZ, and Department of Infectious Diseases, Virology, University of Heidelberg, Im Neuenheimer Feld 242, 69120 Heidelberg.,the Pediatric Infectious Diseases Unit, University Children's Hospital Mannheim, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, and
| | - Clemens Kunz
- the Institute of Nutritional Science, University of Giessen, Wilhelmstrasse 20, 35392 Giessen, Germany
| | - Horst Schroten
- the Pediatric Infectious Diseases Unit, University Children's Hospital Mannheim, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, and
| |
Collapse
|
26
|
Ringot-Destrez B, D'Alessandro Z, Lacroix JM, Mercier-Bonin M, Léonard R, Robbe-Masselot C. A Sensitive and Rapid Method to Determin the Adhesion Capacity of Probiotics and Pathogenic Microorganisms to Human Gastrointestinal Mucins. Microorganisms 2018; 6:E49. [PMID: 29844291 PMCID: PMC6027390 DOI: 10.3390/microorganisms6020049] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/22/2018] [Accepted: 05/28/2018] [Indexed: 12/25/2022] Open
Abstract
Mucus is the habitat for the microorganisms, bacteria and yeast that form the commensal flora. Mucins, the main macromolecules of mucus, and more specifically, the glycans that cover them, play essential roles in microbial gastrointestinal colonization. Probiotics and pathogens must also colonize mucus to have lasting positive or deleterious effects. The question of which mucin-harboured glycan motifs favour the adhesion of specific microorganisms remains very poorly studied. In the current study, a simple test based on the detection of fluorescent-labeled microorganisms raised against microgram amounts of mucins spotted on nitrocellulose was developed. The adhesion of various probiotic, commensal and pathogenic microorganisms was evaluated on a panel of human purified gastrointestinal mucins and compared with that of commercially available pig gastric mucins (PGM) and of mucins secreted by the colonic cancer cell line HT29-MTX. The latter two proved to be very poor indicators of adhesion capacity on intestinal mucins. Our results show that the nature of the sialylated cores of O-glycans, determined by MALDI MS-MS analysis, potentially enables sialic acid residues to modulate the adhesion of microorganisms either positively or negatively. Other identified factors affecting the adhesion propensity were O-glycan core types and the presence of blood group motifs. This test should help to select probiotics with enhanced adhesion capabilities as well as deciphering the role of specific mucin glycotopes on microbial adhesion.
Collapse
Affiliation(s)
- Bélinda Ringot-Destrez
- Univ.lille, CNRS, UMR8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F59000 Lille, France.
| | - Zéa D'Alessandro
- Univ.lille, CNRS, UMR8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F59000 Lille, France.
| | - Jean-Marie Lacroix
- Univ.lille, CNRS, UMR8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F59000 Lille, France.
| | - Muriel Mercier-Bonin
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, 31000 Toulouse, France.
| | - Renaud Léonard
- Univ.lille, CNRS, UMR8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F59000 Lille, France.
| | - Catherine Robbe-Masselot
- Univ.lille, CNRS, UMR8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F59000 Lille, France.
- Unité de Glycobiologie Structurale et Fonctionnelle, Campus CNRS de la Haute Borne, 50 avenue de Halley, 59658 Villeneuve d'Ascq, France.
| |
Collapse
|
27
|
Binding of Helicobacter pylori to Human Gastric Mucins Correlates with Binding of TFF1. Microorganisms 2018; 6:microorganisms6020044. [PMID: 29783620 PMCID: PMC6027488 DOI: 10.3390/microorganisms6020044] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/23/2018] [Accepted: 05/01/2018] [Indexed: 02/06/2023] Open
Abstract
Helicobacter pylori binds to the gastric mucin, MUC5AC, and to trefoil factor, TFF1, which has been shown to interact with gastric mucin. We examined the interactions of TFF1 and H. pylori with purified gastrointestinal mucins from different animal species and from humans printed on a microarray platform to investigate whether TFF1 may play a role in locating H. pylori in gastric mucus. TFF1 bound almost exclusively to human gastric mucins and did not interact with human colonic mucins. There was a strong correlation between binding of TFF1 and H. pylori to human gastric mucins, and between binding of both TFF1 and H. pylori to gastric mucins with that of Griffonia simplicifolia lectin-II, which is specific for terminal non-reducing α- or β-linked N-acetyl-d-glucosamine. These results suggest that TFF1 may help to locate H. pylori in a discrete layer of gastric mucus and hence restrain their interactions with epithelial cells.
Collapse
|
28
|
Arend P. Position of human blood group O(H) and phenotype-determining enzymes in growth and infectious disease. Ann N Y Acad Sci 2018; 1425:5-18. [PMID: 29754430 PMCID: PMC7676429 DOI: 10.1111/nyas.13694] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 02/22/2018] [Accepted: 03/06/2018] [Indexed: 12/12/2022]
Abstract
The human ABO(H) blood group phenotypes arise from the evolutionarily oldest genetic system found in primate populations. While the blood group antigen A is considered the ancestral primordial structure, under the selective pressure of life‐threatening diseases blood group O(H) came to dominate as the most frequently occurring blood group worldwide. Non‐O(H) phenotypes demonstrate impaired formation of adaptive and innate immunoglobulin specificities due to clonal selection and phenotype formation in plasma proteins. Compared with individuals with blood group O(H), blood group A individuals not only have a significantly higher risk of developing certain types of cancer but also exhibit high susceptibility to malaria tropica or infection by Plasmodium falciparum. The phenotype‐determining blood group A glycotransferase(s), which affect the levels of anti‐A/Tn cross‐reactive immunoglobulins in phenotypic glycosidic accommodation, might also mediate adhesion and entry of the parasite to host cells via trans‐species O‐GalNAc glycosylation of abundantly expressed serine residues that arise throughout the parasite's life cycle, while excluding the possibility of antibody formation against the resulting hybrid Tn antigen. In contrast, human blood group O(H), lacking this enzyme, is indicated to confer a survival advantage regarding the overall risk of developing cancer, and individuals with this blood group rarely develop life‐threatening infections involving evolutionarily selective malaria strains.
Collapse
Affiliation(s)
- Peter Arend
- Department of Medicine, Philipps University Marburg, Marburg/Lahn, Germany. Gastroenterology Research Laboratory, College of Medicine, University of Iowa, Iowa City, Iowa. Research Laboratories, Chemie Grünenthal GmbH, Aachen, Germany
| |
Collapse
|
29
|
The Double Face of Mucin-Type O-Glycans in Lectin-Mediated Infection and Immunity. Molecules 2018; 23:molecules23051151. [PMID: 29751628 PMCID: PMC6100456 DOI: 10.3390/molecules23051151] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/03/2018] [Accepted: 05/07/2018] [Indexed: 02/07/2023] Open
Abstract
Epithelial human blood group antigens (HBGAs) on O-glycans play roles in pathogen binding and the initiation of infection, while similar structures on secretory mucins exert protective functions. These double-faced features of O-glycans in infection and innate immunity are reviewed based on two instructive examples of bacterial and viral pathogens. Helicobacter pylori represents a class 1 carcinogen in the human stomach. By expressing blood group antigen-binding adhesin (BabA) and LabA adhesins that bind to Lewis-b and LacdiNAc, respectively, H. pylori colocalizes with the mucin MUC5AC in gastric surface epithelia, but not with MUC6, which is cosecreted with trefoil factor family 2 (TFF2) by deep gastric glands. Both components of the glandular secretome are concertedly up-regulated upon infection. While MUC6 expresses GlcNAc-capped glycans as natural antibiotics for H. pylori growth control, TFF2 may function as a probiotic lectin. In viral infection human noroviruses of the GII genogroup interact with HBGAs via their major capsid protein, VP1. HBGAs on human milk oligosaccharides (HMOs) may exert protective functions by binding to the P2 domain pocket on the capsid. We discuss structural details of the P2 carbohydrate-binding pocket in interaction with blood group H/Lewis-b HMOs and fucoidan-derived oligofucoses as effective interactors for the most prevalent norovirus strains, GII.4 and GII.17.
Collapse
|
30
|
Radziejewska I, Borzym-Kluczyk M, Leszczyńska K, Wosek J, Bielawska A. Lotus tetragonolobus and Maackia amurensis lectins influence phospho-IκBα, IL-8, Lewis b and H type 1 glycoforms levels in H. pylori infected CRL-1739 gastric cancer cells. Adv Med Sci 2018; 63:205-211. [PMID: 29197783 DOI: 10.1016/j.advms.2017.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 06/01/2017] [Accepted: 06/13/2017] [Indexed: 12/20/2022]
Abstract
PURPOSE Attachment of Helicobacter pylori to the mucous epithelial cells and the mucous layer is said to be a crucial step for infection development. Sugar antigens of gastric mucins (MUC5AC, MUC1) can act as receptors for bacterial adhesins. The aim of the study was to investigate if Lotus tetragonolobus and Maackia amurensis lectins influence the level of MUC1, MUC5AC, Lewis b, H type 1, sialyl Lewis x, phospho-IκBα and interleukin 8 in Helicobacter pylori infected gastric cancer cells. MATERIALS AND METHODS The study was performed with one clinical H. pylori strain and CRL-1739 gastric cancer cells. To assess the levels of mentioned factors immunosorbent ELISA assays were used. RESULTS Coculture of cells with bacteria had no clear effect on almost all examined structures. After coculture with H. pylori and lectins, a decrease of the level of both mucins, Lewis b and H type 1 antigens was observed. Lectins addition had no effect on sialyl Lewis x. Maackia amurensis caused slight increase of phospho-IκBα while interleukin 8 level was decreased. CONCLUSIONS Lotus tetragonolobus and Maackia amurensis lectins can mediate in binding of Helicobacter pylori to gastric epithelium.
Collapse
|
31
|
Demouveaux B, Gouyer V, Gottrand F, Narita T, Desseyn JL. Gel-forming mucin interactome drives mucus viscoelasticity. Adv Colloid Interface Sci 2018; 252:69-82. [PMID: 29329667 DOI: 10.1016/j.cis.2017.12.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 12/14/2017] [Accepted: 12/15/2017] [Indexed: 12/31/2022]
Abstract
Mucus is a hydrogel that constitutes the first innate defense in all mammals. The main organic component of mucus, gel-forming mucins, forms a complex network through both reversible and irreversible interactions that drive mucus gel formation. Significant advances in the understanding of irreversible gel-forming mucins assembly have been made using recombinant protein approaches. However, little is known about the reversible interactions that may finely modulate mucus viscoelasticity, which can be characterized using rheology. This approach can be used to investigate both the nature of gel-forming mucins interactions and factors that influence hydrogel formation. This knowledge is directly relevant to the development of new drugs to modulate mucus viscoelasticity and to restore normal mucus functions in diseases such as in cystic fibrosis. The aim of the present review is to summarize the current knowledge about the relationship between the mucus protein matrix and its functions, with emphasis on mucus viscoelasticity.
Collapse
Affiliation(s)
| | - Valérie Gouyer
- Univ. Lille, Inserm, CHU Lille, LIRIC UMR 995, F-59000 Lille, France
| | - Frédéric Gottrand
- Univ. Lille, Inserm, CHU Lille, LIRIC UMR 995, F-59000 Lille, France
| | - Tetsuharu Narita
- Laboratoire Sciences et Ingénierie de la Matière Molle, PSL Research University, UPMC Univ Paris 06, ESPCI Paris, CNRS, 10 rue Vauquelin, 75231 Paris Cedex 05, France; Global Station for Soft Matter, Global Institution for Collaborative Research and Education, Hokkaido University, Sapporo, Japan
| | - Jean-Luc Desseyn
- Univ. Lille, Inserm, CHU Lille, LIRIC UMR 995, F-59000 Lille, France.
| |
Collapse
|
32
|
Bansil R, Turner BS. The biology of mucus: Composition, synthesis and organization. Adv Drug Deliv Rev 2018; 124:3-15. [PMID: 28970050 DOI: 10.1016/j.addr.2017.09.023] [Citation(s) in RCA: 362] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 09/24/2017] [Accepted: 09/27/2017] [Indexed: 02/06/2023]
Abstract
In this review we discuss mucus, the viscoelastic secretion from goblet or mucous producing cells that lines the epithelial surfaces of all organs exposed to the external world. Mucus is a complex aqueous fluid that owes its viscoelastic, lubricating and hydration properties to the glycoprotein mucin combined with electrolytes, lipids and other smaller proteins. Electron microscopy of mucosal surfaces reveals a highly convoluted surface with a network of fibers and pores of varying sizes. The major structural and functional component, mucin is a complex glycoprotein coded by about 20 mucin genes which produce a protein backbone having multiple tandem repeats of Serine, Threonine (ST repeats) where oligosaccharides are covalently O-linked. The N- and C-terminals of this apoprotein contain other domains with little or no glycosylation but rich in cysteines leading to dimerization and further multimerization via SS bonds. The synthesis of this complex protein starts in the endoplasmic reticulum with the formation of the apoprotein and is further modified via glycosylation in the cis and medial Golgi and packaged into mucin granules via Ca2+ bridging of the negative charges on the oligosaccharide brush in the trans Golgi. The mucin granules fuse with the plasma membrane of the secretory cells and following activation by signaling molecules release Ca2+ and undergo a dramatic change in volume due to hydration of the highly negatively charged polymer brush leading to exocytosis from the cells and forming the mucus layer. The rheological properties of mucus and its active component mucin and its mucoadhesivity are briefly discussed in light of their importance to mucosal drug delivery.
Collapse
|
33
|
Darula Z, Medzihradszky KF. Analysis of Mammalian O-Glycopeptides-We Have Made a Good Start, but There is a Long Way to Go. Mol Cell Proteomics 2018; 17:2-17. [PMID: 29162637 PMCID: PMC5750848 DOI: 10.1074/mcp.mr117.000126] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Indexed: 12/18/2022] Open
Abstract
Glycosylation is perhaps the most common post-translational modification. Recently there has been growing interest in cataloging the glycan structures, glycoproteins, and specific sites modified and deciphering the biological functions of glycosylation. Although the results are piling up for N-glycosylation, O-glycosylation is seriously trailing behind. In our review we reiterate the difficulties researchers have to overcome in order to characterize O-glycosylation. We describe how an ingenious cell engineering method delivered exciting results, and what could we gain from "wild-type" samples. Although we refer to the biological role(s) of O-glycosylation, we do not provide a complete inventory on this topic.
Collapse
Affiliation(s)
- Zsuzsanna Darula
- From the ‡Laboratory of Proteomics Research, Biological Research Centre, Hungarian Academy of Sciences, H-6726, 62 Temesvari krt, Szeged, Hungary
| | - Katalin F Medzihradszky
- From the ‡Laboratory of Proteomics Research, Biological Research Centre, Hungarian Academy of Sciences, H-6726, 62 Temesvari krt, Szeged, Hungary;
- §Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, Genentech Hall, N472A, MC 2240, 600 16th Street, San Francisco, California 94158-2517
| |
Collapse
|
34
|
Li Z, Gao C, Zhang Y, Palma AS, Childs RA, Silva LM, Liu Y, Jiang X, Liu Y, Chai W, Feizi T. O-Glycome Beam Search Arrays for Carbohydrate Ligand Discovery. Mol Cell Proteomics 2017; 17:121-133. [PMID: 29183914 PMCID: PMC5750842 DOI: 10.1074/mcp.ra117.000285] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/13/2017] [Indexed: 11/10/2022] Open
Abstract
O-glycosylation is a post-translational modification of proteins crucial to molecular mechanisms in health and disease. O-glycans are typically highly heterogeneous. The involvement of specific O-glycan sequences in many bio-recognition systems is yet to be determined because of a lack of efficient methodologies. We describe here a targeted microarray approach: O-glycome beam search that is both robust and efficient for O-glycan ligand-discovery. Substantial simplification of the complex O-glycome profile and facile chromatographic resolution is achieved by arraying O-glycans as branches, monitoring by mass spectrometry, focusing on promising fractions, and on-array immuno-sequencing. This is orders of magnitude more sensitive than traditional methods. We have applied beam search approach to porcine stomach mucin and identified extremely minor components previously undetected within the O-glycome of this mucin that are ligands for the adhesive proteins of two rotaviruses. The approach is applicable to O-glycome recognition studies in a wide range of biological settings to give insights into glycan recognition structures in natural microenvironments.
Collapse
Affiliation(s)
- Zhen Li
- From the ‡Glycosciences Laboratory, Department of Medicine, Imperial College London, W12 0NN, UK
| | - Chao Gao
- From the ‡Glycosciences Laboratory, Department of Medicine, Imperial College London, W12 0NN, UK.,§Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| | - Yibing Zhang
- From the ‡Glycosciences Laboratory, Department of Medicine, Imperial College London, W12 0NN, UK
| | - Angelina S Palma
- From the ‡Glycosciences Laboratory, Department of Medicine, Imperial College London, W12 0NN, UK.,¶Department of Chemistry, UCIBIO-NOVA University of Lisbon, 1099085, Portugal
| | - Robert A Childs
- From the ‡Glycosciences Laboratory, Department of Medicine, Imperial College London, W12 0NN, UK
| | - Lisete M Silva
- From the ‡Glycosciences Laboratory, Department of Medicine, Imperial College London, W12 0NN, UK
| | - Yang Liu
- ‖Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center and
| | - Xi Jiang
- ‖Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center and.,**University of Cincinnati College of Medicine, Cincinnati, Ohio 45229
| | - Yan Liu
- From the ‡Glycosciences Laboratory, Department of Medicine, Imperial College London, W12 0NN, UK
| | - Wengang Chai
- From the ‡Glycosciences Laboratory, Department of Medicine, Imperial College London, W12 0NN, UK;
| | - Ten Feizi
- From the ‡Glycosciences Laboratory, Department of Medicine, Imperial College London, W12 0NN, UK;
| |
Collapse
|
35
|
Mall AS, Habte H, Mthembu Y, Peacocke J, de Beer C. Mucus and Mucins: do they have a role in the inhibition of the human immunodeficiency virus? Virol J 2017; 14:192. [PMID: 28985745 PMCID: PMC5639604 DOI: 10.1186/s12985-017-0855-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/22/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mucins are large O-linked glycosylated proteins which give mucus their gel-forming properties. There are indications that mucus and mucins in saliva, breast milk and in the cervical plug inhibit the human immunodeficiency virus (HIV-1) in an in vitro assay. Crude mucus gels form continuous layers on the epithelial surfaces of the major internal tracts of the body and protect these epithelial surfaces against aggressive luminal factors such as hydrochloric acid and pepsin proteolysis in the stomach lumen, the movement of hard faecal pellets in the colon at high pressure, the effects of shear against the vaginal epithelium during intercourse and the presence of foreign substances in the respiratory airways. Tumour-associated epitopes on mucins make them suitable as immune-targets on malignant epithelial cells, rendering mucins important as diagnostic and prognostic markers for various diseases, even influencing the design of mucin-based vaccines. Sub-Saharan Africa has the highest prevalence of HIV-AIDS in the world. The main points of viral transmission are via the vaginal epithelium during sexual intercourse and mother-to-child transmission during breast-feeding. There have been many studies showing that several body fluids have components that prevent the transmission of HIV-1 from infected to non-infected persons through various forms of contact. Crude saliva and its purified mucins, MUC5B and MUC7, and the purified mucins from breast milk, MUC1 and MUC4 and pregnancy plug cervical mucus (MUC2, MUC5AC, MUC5B and MUC6), inhibit HIV-1 in an in vitro assay. There are conflicting reports of whether crude breast-milk inhibits HIV-1 in an in vitro assay. However studies with a humanised BLT mouse show that breast-milk does inhibit HIV and that breast-feeding is still advisable even amongst HIV-positive women in under-resourced areas, preferably in conjunction with anti-retroviral treatment. CONCLUSION These findings raise questions of how such a naturally occurring biological substance such as mucus, with remarkable protective properties of epithelial surfaces against aggressive luminal factors in delicate locations, could be used as a tool in the fight against HIV-AIDS, which has reached epidemic proportions in sub-Saharan Africa.
Collapse
Affiliation(s)
- Anwar Suleman Mall
- Division of General Surgery, University of Cape Town and Immune Modulation and Biotherapeutics Discovery, Boehringer- Ingelheim, Danbury, USA
| | - Habtom Habte
- Discipline of Medical Virology, University of Stellenbosch & Tygerberg Hospital, Parow, South Africa
| | - Yolanda Mthembu
- Division of General Surgery, University of Cape Town and Immune Modulation and Biotherapeutics Discovery, Boehringer- Ingelheim, Danbury, USA
| | - Julia Peacocke
- Division of General Surgery, University of Cape Town and Immune Modulation and Biotherapeutics Discovery, Boehringer- Ingelheim, Danbury, USA
| | - Corena de Beer
- Department of Surgery, Division of General Surgery, University of Cape Town, Observatory Cape, 7925 South Africa
| |
Collapse
|
36
|
Histo-blood group carbohydrates as facilitators for infection by Helicobacter pylori. INFECTION GENETICS AND EVOLUTION 2017; 53:167-174. [DOI: 10.1016/j.meegid.2017.05.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/06/2017] [Accepted: 05/30/2017] [Indexed: 02/07/2023]
|
37
|
Ravcheev DA, Thiele I. Comparative Genomic Analysis of the Human Gut Microbiome Reveals a Broad Distribution of Metabolic Pathways for the Degradation of Host-Synthetized Mucin Glycans and Utilization of Mucin-Derived Monosaccharides. Front Genet 2017; 8:111. [PMID: 28912798 PMCID: PMC5583593 DOI: 10.3389/fgene.2017.00111] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 08/11/2017] [Indexed: 12/27/2022] Open
Abstract
The colonic mucus layer is a dynamic and complex structure formed by secreted and transmembrane mucins, which are high-molecular-weight and heavily glycosylated proteins. Colonic mucus consists of a loose outer layer and a dense epithelium-attached layer. The outer layer is inhabited by various representatives of the human gut microbiota (HGM). Glycans of the colonic mucus can be used by the HGM as a source of carbon and energy when dietary fibers are not sufficiently available. Both commensals and pathogens can utilize mucin glycans. Commensals are mostly involved in the cleavage of glycans, while pathogens mostly utilize monosaccharides released by commensals. This HGM-derived degradation of the mucus layer increases pathogen susceptibility and causes many other health disorders. Here, we analyzed 397 individual HGM genomes to identify pathways for the cleavage of host-synthetized mucin glycans to monosaccharides as well as for the catabolism of the derived monosaccharides. Our key results are as follows: (i) Genes for the cleavage of mucin glycans were found in 86% of the analyzed genomes, which significantly higher than a previous estimation. (ii) Genes for the catabolism of derived monosaccharides were found in 89% of the analyzed genomes. (iii) Comparative genomic analysis identified four alternative forms of the monosaccharide-catabolizing enzymes and four alternative forms of monosaccharide transporters. (iv) Eighty-five percent of the analyzed genomes may be involved in potential feeding pathways for the monosaccharides derived from cleaved mucin glycans. (v) The analyzed genomes demonstrated different abilities to degrade known mucin glycans. Generally, the ability to degrade at least one type of mucin glycan was predicted for 81% of the analyzed genomes. (vi) Eighty-two percent of the analyzed genomes can form mutualistic pairs that are able to degrade mucin glycans and are not degradable by any of the paired organisms alone. Taken together, these findings provide further insight into the inter-microbial communications of the HGM as well as into host-HGM interactions.
Collapse
Affiliation(s)
- Dmitry A Ravcheev
- Luxembourg Centre for Systems Biomedicine, University of LuxembourgEsch-sur-Alzette, Luxembourg
| | - Ines Thiele
- Luxembourg Centre for Systems Biomedicine, University of LuxembourgEsch-sur-Alzette, Luxembourg
| |
Collapse
|
38
|
How do they stick together? Bacterial adhesins implicated in the binding of bacteria to the human gastrointestinal mucins. Biochem Soc Trans 2017; 45:389-399. [PMID: 28408479 DOI: 10.1042/bst20160167] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/16/2016] [Accepted: 01/23/2017] [Indexed: 01/04/2023]
Abstract
The gastrointestinal mucosal surface is the primary interface between internal host tissues and the vast microbiota. Mucins, key components of mucus, are high-molecular-weight glycoproteins characterized by the presence of many O-linked oligosaccharides to the core polypeptide. They play many biological functions, helping to maintain cellular homeostasis and to establish symbiotic relationships with complex microbiota. Mucin O-glycans exhibit a huge variety of peripheral sequences implicated in the binding of bacteria to the mucosal tissues, thereby playing a key role in the selection of specific species and in the tissue tropism displayed by commensal and pathogenic bacteria. Bacteria have evolved numerous strategies to colonize host mucosae, and among these are modulation of expression of cell surface adhesins which allow bacteria to bind to mucins. However, despite well structurally characterized adhesins and lectins, information on the nature and structure of oligosaccharides recognized by bacteria is still disparate. This review summarizes the current knowledge on the structure of epithelial mucin O-glycans and the interaction between host and commensal or pathogenic bacteria mediated by mucins.
Collapse
|
39
|
Jin C, Kenny DT, Skoog EC, Padra M, Adamczyk B, Vitizeva V, Thorell A, Venkatakrishnan V, Lindén SK, Karlsson NG. Structural Diversity of Human Gastric Mucin Glycans. Mol Cell Proteomics 2017; 16:743-758. [PMID: 28461410 DOI: 10.1074/mcp.m116.067983] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Indexed: 01/16/2023] Open
Abstract
The mucin O-glycosylation of 10 individuals with and without gastric disease was examined in depth in order to generate a structural map of human gastric glycosylation. In the stomach, these mucins and their O-glycosylation protect the epithelial surface from the acidic gastric juice and provide the first point of interaction for pathogens such as Helicobacter pylori, reported to cause gastritis, gastric and duodenal ulcers and gastric cancer. The rational of the present study was to map the O-glycosylation that the pathogen may come in contact with. An enormous diversity in glycosylation was found, which varied both between individuals and within mucins from a single individual: mucin glycan chain length ranged from 2-13 residues, each individual carried 34-103 O-glycan structures and in total over 258 structures were identified. The majority of gastric O-glycans were neutral and fucosylated. Blood group I antigens, as well as terminal α1,4-GlcNAc-like and GalNAcβ1-4GlcNAc-like (LacdiNAc-like), were common modifications of human gastric O-glycans. Furthemore, each individual carried 1-14 glycan structures that were unique for that individual. The diversity and alterations in gastric O-glycosylation broaden our understanding of the human gastric O-glycome and its implications for gastric cancer research and emphasize that the high individual variation makes it difficult to identify gastric cancer specific structures. However, despite the low number of individuals, we could verify a higher level of sialylation and sulfation on gastric O-glycans from cancerous tissue than from healthy stomachs.
Collapse
Affiliation(s)
- Chunsheng Jin
- From the ‡Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, Medicinaregatan 9A, 405 30 Gothenburg, Sweden
| | - Diarmuid T Kenny
- From the ‡Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, Medicinaregatan 9A, 405 30 Gothenburg, Sweden
| | - Emma C Skoog
- From the ‡Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, Medicinaregatan 9A, 405 30 Gothenburg, Sweden
| | - Médea Padra
- From the ‡Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, Medicinaregatan 9A, 405 30 Gothenburg, Sweden
| | - Barbara Adamczyk
- From the ‡Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, Medicinaregatan 9A, 405 30 Gothenburg, Sweden
| | - Varvara Vitizeva
- From the ‡Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, Medicinaregatan 9A, 405 30 Gothenburg, Sweden
| | - Anders Thorell
- §Karolinska Institute, Department for Clinical Science and Department of Surgery, Ersta Hospital, Stockholm, Sweden
| | - Vignesh Venkatakrishnan
- From the ‡Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, Medicinaregatan 9A, 405 30 Gothenburg, Sweden
| | - Sara K Lindén
- From the ‡Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, Medicinaregatan 9A, 405 30 Gothenburg, Sweden
| | - Niclas G Karlsson
- From the ‡Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 440, Medicinaregatan 9A, 405 30 Gothenburg, Sweden;
| |
Collapse
|
40
|
Jin C, Kenny DT, Skoog EC, Padra M, Adamczyk B, Vitizeva V, Thorell A, Venkatakrishnan V, Lindén SK, Karlsson NG. Structural diversity of human gastric mucin glycans. Mol Cell Proteomics 2017; 16:mcp.M117.067983. [PMID: 28289177 PMCID: PMC5417818 DOI: 10.1074/mcp.m117.067983] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 03/13/2017] [Indexed: 01/14/2023] Open
Abstract
The mucin O-glycosylation of 10 individuals with and without gastric disease was examined in depth in order to generate a structural map of human gastric glycosylation. In the stomach, these mucins and their O-glycosylation protect the epithelial surface from the acidic gastric juice and provide the first point of interaction for pathogens such as Helicobacter pylori, reported to cause gastritis, gastric and duodenal ulcers and gastric cancer. The rational of the present study was to map the O-glycosylation that the pathogen may come in contact with. An enormous diversity in glycosylation was found, which varied both between individuals and within mucins from a single individual: mucin glycan chain length ranged from 2-13 residues, each individual carried 34-103 O-glycan structures and in total over 258 structures were identified. The majority of gastric O-glycans were neutral and fucosylated. Blood group I antigens, as well as terminal α1,4-GlcNAc-like and GalNAcβ1-4GlcNAc-like (LacdiNAc-like), were common modifications of human gastric O-glycans. Furthemore, each individual carried 1-14 glycan structures that were unique for that individual. The diversity and alterations in gastric O-glycosylation broaden our understanding of the human gastric O-glycome and its implications for gastric cancer research and emphasize that the high individual variation makes it difficult to identify gastric cancer specific structures. However, despite the low number of individuals, we could verify a higher level of sialylation and sulfation on gastric O-glycans from cancerous tissue than from healthy stomachs.
Collapse
Affiliation(s)
- Chunsheng Jin
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy,, Sweden
| | - Diarmuid T Kenny
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy,, Sweden
| | - Emma C Skoog
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy,, Sweden
| | - Medéa Padra
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy,, Sweden
| | - Barbara Adamczyk
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy,, Sweden
| | - Varvara Vitizeva
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy,, Sweden
| | - Anders Thorell
- Karolinska Institute, Department for Clinical Science and Department of Surgery, Ersta Hospital, Sweden
| | - Vignesh Venkatakrishnan
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy,, Sweden
| | | | | |
Collapse
|
41
|
Magalhães A, Rossez Y, Robbe-Masselot C, Maes E, Gomes J, Shevtsova A, Bugaytsova J, Borén T, Reis CA. Muc5ac gastric mucin glycosylation is shaped by FUT2 activity and functionally impacts Helicobacter pylori binding. Sci Rep 2016; 6:25575. [PMID: 27161092 PMCID: PMC4861914 DOI: 10.1038/srep25575] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 04/19/2016] [Indexed: 12/21/2022] Open
Abstract
The gastrointestinal tract is lined by a thick and complex layer of mucus that protects the mucosal epithelium from biochemical and mechanical aggressions. This mucus barrier confers protection against pathogens but also serves as a binding site that supports a sheltered niche of microbial adherence. The carcinogenic bacteria Helicobacter pylori colonize the stomach through binding to host glycans present in the glycocalyx of epithelial cells and extracellular mucus. The secreted MUC5AC mucin is the main component of the gastric mucus layer, and BabA-mediated binding of H. pylori to MUC5AC confers increased risk for overt disease. In this study we unraveled the O-glycosylation profile of Muc5ac from glycoengineered mice models lacking the FUT2 enzyme and therefore mimicking a non-secretor human phenotype. Our results demonstrated that the FUT2 determines the O-glycosylation pattern of Muc5ac, with Fut2 knock-out leading to a marked decrease in α1,2-fucosylated structures and increased expression of the terminal type 1 glycan structure Lewis-a. Importantly, for the first time, we structurally validated the expression of Lewis-a in murine gastric mucosa. Finally, we demonstrated that loss of mucin FUT2-mediated fucosylation impairs gastric mucosal binding of H. pylori BabA adhesin, which is a recognized feature of pathogenicity.
Collapse
Affiliation(s)
- Ana Magalhães
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology of University of Porto, Ipatimup, Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - Yannick Rossez
- Structural and Functional Glycobiology Unit, UMR CNRS 8576, University of Lille, 59655 Villeneuve d’Ascq, France
| | - Catherine Robbe-Masselot
- Structural and Functional Glycobiology Unit, UMR CNRS 8576, University of Lille, 59655 Villeneuve d’Ascq, France
| | - Emmanuel Maes
- Structural and Functional Glycobiology Unit, UMR CNRS 8576, University of Lille, 59655 Villeneuve d’Ascq, France
| | - Joana Gomes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology of University of Porto, Ipatimup, Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - Anna Shevtsova
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-901 87 Umeå, Sweden
| | - Jeanna Bugaytsova
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-901 87 Umeå, Sweden
| | - Thomas Borén
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-901 87 Umeå, Sweden
| | - Celso A. Reis
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology of University of Porto, Ipatimup, Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
- Medical Faculty, University of Porto, Al. Prof. Hernâni Monteiro, 4200–319 Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira no 228, 4050-313 Porto, Portugal
| |
Collapse
|
42
|
Mereiter S, Balmaña M, Gomes J, Magalhães A, Reis CA. Glycomic Approaches for the Discovery of Targets in Gastrointestinal Cancer. Front Oncol 2016; 6:55. [PMID: 27014630 PMCID: PMC4783390 DOI: 10.3389/fonc.2016.00055] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 02/24/2016] [Indexed: 12/22/2022] Open
Abstract
Gastrointestinal (GI) cancer is the most common group of malignancies and many of its types are among the most deadly. Various glycoconjugates have been used in clinical practice as serum biomarker for several GI tumors, however, with limited diagnose application. Despite the good accessibility by endoscopy of many GI organs, the lack of reliable serum biomarkers often leads to late diagnosis of malignancy and consequently low 5-year survival rates. Recent advances in analytical techniques have provided novel glycoproteomic and glycomic data and generated functional information and putative biomarker targets in oncology. Glycosylation alterations have been demonstrated in a series of glycoconjugates (glycoproteins, proteoglycans, and glycosphingolipids) that are involved in cancer cell adhesion, signaling, invasion, and metastasis formation. In this review, we present an overview on the major glycosylation alterations in GI cancer and the current serological biomarkers used in the clinical oncology setting. We further describe recent glycomic studies in GI cancer, namely gastric, colorectal, and pancreatic cancer. Moreover, we discuss the role of glycosylation as a modulator of the function of several key players in cancer cell biology. Finally, we address several state-of-the-art techniques currently applied in this field, such as glycomic and glycoproteomic analyses, the application of glycoengineered cell line models, microarray and proximity ligation assay, and imaging mass spectrometry, and provide an outlook to future perspectives and clinical applications.
Collapse
Affiliation(s)
- Stefan Mereiter
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal; Institute of Biomedical Sciences of Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Meritxell Balmaña
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona , Girona , Spain
| | - Joana Gomes
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Ana Magalhães
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Celso A Reis
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal; Institute of Biomedical Sciences of Abel Salazar (ICBAS), University of Porto, Porto, Portugal; Medical Faculty, University of Porto, Porto, Portugal
| |
Collapse
|
43
|
Mereiter S, Magalhães A, Adamczyk B, Jin C, Almeida A, Drici L, Ibáñez-Vea M, Gomes C, Ferreira JA, Afonso LP, Santos LL, Larsen MR, Kolarich D, Karlsson NG, Reis CA. Glycomic analysis of gastric carcinoma cells discloses glycans as modulators of RON receptor tyrosine kinase activation in cancer. Biochim Biophys Acta Gen Subj 2015; 1860:1795-808. [PMID: 26721331 DOI: 10.1016/j.bbagen.2015.12.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/18/2015] [Accepted: 12/19/2015] [Indexed: 01/16/2023]
Abstract
BACKGROUND Terminal α2-3 and α2-6 sialylation of glycans precludes further chain elongation, leading to the biosynthesis of cancer relevant epitopes such as sialyl-Lewis X (SLe(X)). SLe(X) overexpression is associated with tumor aggressive phenotype and patients' poor prognosis. METHODS MKN45 gastric carcinoma cells transfected with the sialyltransferase ST3GAL4 were established as a model overexpressing sialylated terminal glycans. We have evaluated at the structural level the glycome and the sialoproteome of this gastric cancer cell line applying liquid chromatography and mass spectrometry. We further validated an identified target expression by proximity ligation assay in gastric tumors. RESULTS Our results showed that ST3GAL4 overexpression leads to several glycosylation alterations, including reduced O-glycan extension and decreased bisected and increased branched N-glycans. A shift from α2-6 towards α2-3 linked sialylated N-glycans was also observed. Sialoproteomic analysis further identified 47 proteins with significantly increased sialylated N-glycans. These included integrins, insulin receptor, carcinoembryonic antigens and RON receptor tyrosine kinase, which are proteins known to be key players in malignancy. Further analysis of RON confirmed its modification with SLe(X) and the concomitant activation. SLe(X) and RON co-expression was validated in gastric tumors. CONCLUSION The overexpression of ST3GAL4 interferes with the overall glycophenotype of cancer cells affecting a multitude of key proteins involved in malignancy. Aberrant glycosylation of the RON receptor was shown as an alternative mechanism of oncogenic activation. GENERAL SIGNIFICANCE This study provides novel targets and points to an integrative tumor glycomic/proteomic-profiling for gastric cancer patients' stratification. This article is part of a Special Issue entitled "Glycans in personalised medicine" Guest Editor: Professor Gordan Lauc.
Collapse
Affiliation(s)
- Stefan Mereiter
- I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal; Institute of Biomedical Sciences of Abel Salazar - ICBAS, University of Porto, Portugal
| | - Ana Magalhães
- I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal
| | - Barbara Adamczyk
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Chunsheng Jin
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Andreia Almeida
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany; Free University Berlin, Berlin, Germany
| | - Lylia Drici
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Maria Ibáñez-Vea
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Catarina Gomes
- I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal
| | - José A Ferreira
- I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal; Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology of Porto, Portugal
| | - Luis P Afonso
- Department of Pathology, Portuguese Institute of Oncology of Porto, Portugal
| | - Lúcio L Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology of Porto, Portugal; Department of Surgical Oncology, Portuguese Institute of Oncology of Porto, Portugal
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Daniel Kolarich
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Niclas G Karlsson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Celso A Reis
- I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal; Institute of Biomedical Sciences of Abel Salazar - ICBAS, University of Porto, Portugal; Medical Faculty, University of Porto, Portugal.
| |
Collapse
|
44
|
Merging carbohydrate chemistry with lectin histochemistry to study inhibition of lectin binding by glycoclusters in the natural tissue context. Histochem Cell Biol 2015; 145:185-99. [PMID: 26553286 DOI: 10.1007/s00418-015-1383-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2015] [Indexed: 01/22/2023]
Abstract
Recognition of glycans by lectins leads to cell adhesion and growth regulation. The specificity and selectivity of this process are determined by carbohydrate structure (sequence and shape) and topology of its presentation. The synthesis of (neo)glycoconjugates with bi- to oligo-valency (glycoclusters) affords tools to delineate structure-activity relationships by blocking lectin binding to an artificial matrix, often a glycoprotein, or cultured cell lines. The drawback of these assays is that glycan presentation is different from that in tissues. In order to approach the natural context, we here introduce lectin histochemistry on fixed tissue sections to determine the susceptibility of binding of two plant lectins, i.e., GSA-II and WGA, to a series of 10 glycoclusters. Besides valency, this panel covers changes in the anomeric position (α/β) and the atom at the glycosidic linkage (O/S). Flanked by cell and solid-phase assays with human tumor lines and two mucins, respectively, staining (intensity and profile) was analyzed in sections of murine jejunum, stomach and epididymis as a function of glycocluster presence. The marked and differential sensitivity of signal generation to structural aspects of the glycoclusters proves the applicability of this method. This enables comparisons between data sets obtained by using (neo)glycoconjugates, cells and the tissue context as platforms. The special advantage of processing tissue sections is the monitoring of interference with lectin association at sites that are relevant for functionality. Testing glycoclusters in lectin histochemistry will especially be attractive in cases of multi-target recognition (glycans, proteins and lipids) by a tissue lectin.
Collapse
|
45
|
Hoffmann W. TFF2, a MUC6-binding lectin stabilizing the gastric mucus barrier and more (Review). Int J Oncol 2015. [PMID: 26201258 DOI: 10.3892/ijo.2015.3090] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The peptide TFF2 (formerly 'spasmolytic polypeptide'), a member of the trefoil factor family (TFF) containing two TFF domains, is mainly expressed together with the mucin MUC6 in the gastric epithelium and duodenal Brunner's glands. Pathologically, TFF2 expression is observed ectopically during stone diseases, chronic inflammatory conditions and in several metaplastic and neoplastic epithelia; most prominent being the 'spasmolytic polypeptide-expressing metaplasia' (SPEM), which is an established gastric precancerous lesion. TFF2 plays a critical role in maintaining gastric mucosal integrity and appears to restrain tumorigenesis in the stomach. Recently, porcine TFF2 has been shown to interact with the gastric mucin MUC6 and thus stabilize the gastric mucus barrier. On the one hand, TFF2 binds to MUC6 via non-covalent lectin interactions with the glycotope GlcNAcα1→4Galβ1→R. On the other hand, TFF2 is probably also covalently bound to MUC6 via disulfide bridges. Thus, implications for the complex multimeric assembly, cross-linking, and packaging of MUC6 as well as the rheology of gastric mucus are discussed in detail in this review. Furthermore, TFF2 is also expressed in minor amounts in the immune and nervous systems. Thus, similar to galectins, its lectin activity would perfectly enable TFF2 to form multivalent complexes and cross-linked lattices with a plethora of transmembrane glycoproteins and thus modulate different signal transduction processes. This could explain the multiple and diverse biological effects of TFF2 [e.g., motogenic, (anti)apoptotic, and angiogenic effects]. Finally, a function during fertilization is also possible for TFF domains because they occur as shuffled modules in certain zona pellucida proteins.
Collapse
Affiliation(s)
- Werner Hoffmann
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke-University Magdeburg, D-39120 Magdeburg, Germany
| |
Collapse
|
46
|
Abstract
Blood group antigens represent polymorphic traits inherited among individuals and populations. At present, there are 34 recognized human blood groups and hundreds of individual blood group antigens and alleles. Differences in blood group antigen expression can increase or decrease host susceptibility to many infections. Blood groups can play a direct role in infection by serving as receptors and/or coreceptors for microorganisms, parasites, and viruses. In addition, many blood group antigens facilitate intracellular uptake, signal transduction, or adhesion through the organization of membrane microdomains. Several blood groups can modify the innate immune response to infection. Several distinct phenotypes associated with increased host resistance to malaria are overrepresented in populations living in areas where malaria is endemic, as a result of evolutionary pressures. Microorganisms can also stimulate antibodies against blood group antigens, including ABO, T, and Kell. Finally, there is a symbiotic relationship between blood group expression and maturation of the gastrointestinal microbiome.
Collapse
Affiliation(s)
- Laura Cooling
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
47
|
Vitiazeva V, Kattla JJ, Flowers SA, Lindén SK, Premaratne P, Weijdegård B, Sundfeldt K, Karlsson NG. The O-Linked Glycome and Blood Group Antigens ABO on Mucin-Type Glycoproteins in Mucinous and Serous Epithelial Ovarian Tumors. PLoS One 2015; 10:e0130197. [PMID: 26075384 PMCID: PMC4468167 DOI: 10.1371/journal.pone.0130197] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/16/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Mucins are heavily O-glycosylated proteins where the glycosylation has been shown to play an important role in cancer. Normal epithelial ovarian cells do not express secreted mucins, but their abnormal expression has previously been described in epithelial ovarian cancer and may relate to tumor formation and progression. The cyst fluids were shown to be a rich source for acidic glycoproteins. The study of these proteins can potentially lead to the identification of more effective biomarkers for ovarian cancer. METHODS In this study, we analyzed the expression of the MUC5AC and the O-glycosylation of acidic glycoproteins secreted into ovarian cyst fluids. The samples were obtained from patients with serous and mucinous ovarian tumors of different stages (benign, borderline, malignant) and grades. The O-linked oligosaccharides were released and analyzed by negative-ion graphitized carbon Liquid Chromatography (LC) coupled to Electrospray Ionization tandem Mass Spectrometry (ESI-MSn). The LC-ESI-MSn of the oligosaccharides from ovarian cyst fluids displayed differences in expression of fucose containing structures such as blood group ABO antigens and Lewis-type epitopes. RESULTS The obtained data showed that serous and mucinous benign adenomas, mucinous low malignant potential carcinomas (LMPs, borderline) and mucinous low-grade carcinomas have a high level of blood groups and Lewis type epitopes. In contrast, this type of fucosylated structures were low abundant in the high-grade mucinous carcinomas or in serous carcinomas. In addition, the ovarian tumors that showed a high level of expression of blood group antigens also revealed a strong reactivity towards the MUC5AC antibody. To visualize the differences between serous and mucinous ovarian tumors based on the O-glycosylation, a hierarchical cluster analysis was performed using mass spectrometry average compositions (MSAC). CONCLUSION Mucinous benign and LMPs along with mucinous low-grade carcinomas appear to be different from serous and high-grade mucinous carcinomas based on their O-glycan profiles.
Collapse
Affiliation(s)
- Varvara Vitiazeva
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
- * E-mail:
| | - Jayesh J. Kattla
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Sarah A. Flowers
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Sara K. Lindén
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Pushpa Premaratne
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Birgitta Weijdegård
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Karin Sundfeldt
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Niclas G. Karlsson
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
48
|
Delivery of a mucin domain enriched in cysteine residues strengthens the intestinal mucous barrier. Sci Rep 2015; 5:9577. [PMID: 25974250 PMCID: PMC4431476 DOI: 10.1038/srep09577] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 01/26/2015] [Indexed: 12/22/2022] Open
Abstract
A weakening of the gut mucous barrier permits an increase in the access of intestinal luminal contents to the epithelial cells, which will trigger the inflammatory response. In inflammatory bowel diseases, there is an inappropriate and ongoing activation of the immune system, possibly because the intestinal mucus is less protective against the endogenous microflora. General strategies aimed at improving the protection of the intestinal epithelium are still missing. We generated a transgenic mouse that secreted a molecule consisting of 12 consecutive copies of a mucin domain into its intestinal mucus, which is believed to modify the mucus layer by establishing reversible interactions. We showed that the mucus gel was more robust and that mucin O-glycosylation was altered. Notably, the gut epithelium of transgenic mice housed a greater abundance of beneficial Lactobacillus spp. These modifications were associated with a reduced susceptibility of transgenic mice to chemically induced colitis. Furthermore, transgenic mice cleared faster Citrobacter rodentium bacteria which were orally given and mice were more protected against bacterial translocation induced by gavage with adherent–invasive Escherichia coli. Our data show that delivering the mucin CYS domain into the gut lumen strengthens the intestinal mucus blanket which is impaired in inflammatory bowel diseases.
Collapse
|
49
|
Joncquel Chevalier Curt M, Lecointe K, Mihalache A, Rossez Y, Gosset P, Léonard R, Robbe-Masselot C. Alteration or adaptation, the two roads for human gastric mucin glycosylation infected by Helicobacter pylori. Glycobiology 2015; 25:617-31. [DOI: 10.1093/glycob/cwv004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 01/06/2015] [Indexed: 12/24/2022] Open
|
50
|
Rossez Y, Wolfson EB, Holmes A, Gally DL, Holden NJ. Bacterial flagella: twist and stick, or dodge across the kingdoms. PLoS Pathog 2015; 11:e1004483. [PMID: 25590430 PMCID: PMC4295861 DOI: 10.1371/journal.ppat.1004483] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The flagellum organelle is an intricate multiprotein assembly best known for its rotational propulsion of bacteria. However, recent studies have expanded our knowledge of other functions in pathogenic contexts, particularly adherence and immune modulation, e.g., for Salmonella enterica, Campylobacter jejuni, Pseudomonas aeruginosa, and Escherichia coli. Flagella-mediated adherence is important in host colonisation for several plant and animal pathogens, but the specific interactions that promote flagella binding to such diverse host tissues has remained elusive. Recent work has shown that the organelles act like probes that find favourable surface topologies to initiate binding. An emerging theme is that more general properties, such as ionic charge of repetitive binding epitopes and rotational force, allow interactions with plasma membrane components. At the same time, flagellin monomers are important inducers of plant and animal innate immunity: variation in their recognition impacts the course and outcome of infections in hosts from both kingdoms. Bacteria have evolved different strategies to evade or even promote this specific recognition, with some important differences shown for phytopathogens. These studies have provided a wider appreciation of the functions of bacterial flagella in the context of both plant and animal reservoirs.
Collapse
Affiliation(s)
- Yannick Rossez
- Cellular and Molecular Sciences, James Hutton Institute, Dundee, United Kingdom
| | - Eliza B. Wolfson
- Division of Infection and Immunity, The Roslin Institute, University of Edinburgh, Easter Bush, United Kingdom
| | - Ashleigh Holmes
- Cellular and Molecular Sciences, James Hutton Institute, Dundee, United Kingdom
| | - David L. Gally
- Division of Infection and Immunity, The Roslin Institute, University of Edinburgh, Easter Bush, United Kingdom
| | - Nicola J. Holden
- Cellular and Molecular Sciences, James Hutton Institute, Dundee, United Kingdom
- * E-mail:
| |
Collapse
|