1
|
Li C, Han T, Zhong P, Zhang Y, Zhao T, Wang S, Wang X, Tian Y, Gong G, Liu Y, Huang L, Lu Y, Wang Z. α2,6-linked sialylated oligosaccharides riched in goat milk alleviate food allergy by regulating the gut flora and mucin O-glycosylation. Carbohydr Polym 2025; 350:123049. [PMID: 39647952 DOI: 10.1016/j.carbpol.2024.123049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 12/10/2024]
Abstract
The nutritious goat milk has low allergenicity. Oligosaccharides represent one of the crucial functional constituents in goat milk, which are structurally similar to human milk oligosaccharides (HMOs). Currently, the anti-allergic activity of GMOs has not been reported. In this study, GMOs were efficiently separated into neutral (NGMOs) and sialylated (SGMOs) fractions, following by qualitative and quantitative analysis at the isomer level using online LC-MS/MS. Fifteen NGMOs and 28 SGMOs were detected in goat milk, with 10 SGMOs reported for the first time. Distinctly, α2,6-linked SGMOs were 3.9 times more abundant in goat milk than in bovine milk, with the total relative content of 6'SL, 3'SLN and 6'NGL in SGMOs approach to 60%, which is more similar to HMOs. Orally administering GMOs, especially α2,6-linked sialylated oligosaccharides, significantly alleviated food allergy in ovalbumin-induced BALB/c mice. SGMOs restored the balance of Lachnospiraceae, Erysipelotrichaceae, and Bacteroidaceae, reconstructed the intestinal mucosal barrier, especially restored the levels of fucosylation, sialylation, and sulfation of mucin O-glycans, increased the expression of four core type 2 O-glycans (F1H2N2, F2H2N2, S1F2H2N2, and A1F1H2N2) significantly. This is the first comprehensive study of the anti-allergic activity of GMOs, and the results lay the foundation for the development of GMOs-based natural anti-allergic components.
Collapse
Affiliation(s)
- Cheng Li
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Tianjiao Han
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Peiyun Zhong
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Yuyang Zhang
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Tong Zhao
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Shukai Wang
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Xiaoqin Wang
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Yang Tian
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Guiping Gong
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Yuxia Liu
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Linjuan Huang
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Yu Lu
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China.
| | - Zhongfu Wang
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China.
| |
Collapse
|
2
|
Yang Y, Holck J, Thorhallsson AT, Hunt CJ, Yang H, Morth JP, Meyer AS, Zeuner B. Structural elucidation and characterization of GH29A α-l-fucosidases and the effect of pH on their transglycosylation. FEBS J 2024. [PMID: 39658312 DOI: 10.1111/febs.17347] [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: 06/19/2024] [Revised: 10/11/2024] [Accepted: 11/27/2024] [Indexed: 12/12/2024]
Abstract
GH29A α-l-fucosidases (EC 3.2.1.51) catalyze the release of α-l-fucosyl moieties from the nonreducing end of glycoconjugates by hydrolysis and some also catalyze transglycosylation. The latter is particularly interesting with regard to designing enzymatic synthesis of human milk oligosaccharides (HMOs). We combined the bioinformatics tool conserved unique peptide patterns (CUPP) and phylogenetic clustering to discover new microbial GH29A α-l-fucosidases of the underexplored CUPP group GH29:13.1. Three uncharacterized bacterial enzymes (EaGH29, SeGH29, and PmGH29) and two previously identified GH29A α-l-fucosidases (BF3242 and TfFuc1) were selected for reaction optimization, biochemical, and structural characterization. Kinetics, pH-temperature optima, and substrate preference for 2-chloro-4-nitrophenyl-α-l-fucopyranoside (CNP-α-l-Fuc) and 2'-fucosyllactose (2'FL) were determined. Transglycosylation was favored at high neutral to alkaline pH, especially for EaGH29, SeGH29, TfFuc1, and BF3242, mainly because hydrolysis was decreased. The α-l-fucosidases exhibited medium regioselectivity in transglycosylation, generally forming two out of five detected lacto-N-fucopentaose (LNFP) isomers from 2'FL and lacto-N-tetraose (LNT). Alkaline pH also affected the transglycosylation product regioselectivity of SeGH29, which was also affected by a Leu/Phe exchange in the acceptor binding site. New crystal structures of TfFuc1 and BF3242 showed congruence in active site topology between these two enzymes and contributed to understanding the function of GH29A α-l-fucosidases. Notably, the structural data provide new insight into the role of an Asn residue located between the two catalytic residues in the active site.
Collapse
Affiliation(s)
- Yaya Yang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, DTU Bioengineering, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Jesper Holck
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, DTU Bioengineering, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Albert Thor Thorhallsson
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, DTU Bioengineering, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Cameron J Hunt
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, DTU Bioengineering, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Huan Yang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Jens Preben Morth
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, DTU Bioengineering, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Anne S Meyer
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, DTU Bioengineering, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Birgitte Zeuner
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, DTU Bioengineering, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
3
|
Wang R, Song J, Cai M, Xue Y, Liu J, Zuo N, De Felici M, Wang J, Shen W, Sun X. Gut microbiota modulation by L-Fucose as a strategy to alleviate Ochratoxin A toxicity on primordial follicle formation. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136469. [PMID: 39536347 DOI: 10.1016/j.jhazmat.2024.136469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/16/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
In this study, we investigated the potential benefits of L-Fucose administration to pregnant mice exposed to Ochratoxin A (OTA), a widespread mycotoxin, producing ovarian damage in offspring. The results showed that administration of 3.5 μg/d OTA induced alterations in intestinal tissues and gut microbiota of pregnant mice, leading to heightened local and systemic inflammation. This inflammatory affected the ovaries of their 3 dpp offspring, in which elevated levels of LPS and ROS were found associated to significant decreased oocyte count and impaired primordial follicle assembly. Moreover, mRNA-Seq analysis showed significant changes in ovarian transcriptomes linked to various GO terms and KEGG pathways, notably ferroptosis, a recognized form of cell death observed. Interestingly, administration of 0.3 g/kg b. w. L-Fucose following OTA exposure mitigated these effects on intestinal tissues and gut microbiota in mothers and on the offspring's ovaries. Similar benefits were obtained by gut microbiota transplantation from L-Fucose-treated pregnant females into OTA-exposed mothers. These findings suggest that inflammatory impact of OTA on maternal intestine/gut can pass to the fetus causing offspring ovary defects and support the use of L-Fucose as adjuvant to counteract the adverse effects of mycotoxins on the gut microbiota, particularly reference to those affecting reproductive organs.
Collapse
Affiliation(s)
- Ruiting Wang
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Jie Song
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Muyu Cai
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Yuan Xue
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Jing Liu
- Central laboratory of Qingdao Agricultural University, Qingdao 266109, China
| | - Ning Zuo
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Massimo De Felici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome 00133, Italy
| | - Junjie Wang
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Wei Shen
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Xiaofeng Sun
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China.
| |
Collapse
|
4
|
Mancini A, Vitucci D, Lasorsa VA, Lupo C, Brustio PR, Capasso M, Orrù S, Rainoldi A, Schena F, Buono P. Six months of different exercise type in sedentary primary schoolchildren: impact on physical fitness and saliva microbiota composition. Front Nutr 2024; 11:1465707. [PMID: 39512522 PMCID: PMC11542257 DOI: 10.3389/fnut.2024.1465707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/15/2024] [Indexed: 11/15/2024] Open
Abstract
Introduction Lifestyle influences microbiota composition. We previously reported a healthier microbiota composition in saliva from active schoolchildren compared to sedentary. In the present study, we evaluated the effects of 6 months of different exercise types on physical fitness and saliva microbiota composition in 8-11-years-old sedentary schoolchildren. Methods Sixty-four sedentary children from five primary schools in Turin, Italy, were divided into three groups: one continued normal curricular activity while two underwent different exercise protocols for 6 months. The Structured Exercise (Sa) group did 2 h per week of muscle activation, strength and coordination exercises supervised by a kinesiologist. The Daily Mile (Dm) group did 1 h per week of Sa plus 15 min of walking/running outdoors four times a week, supervised by a class teacher; control group (Ct) did 2 h a week of curricular exercise supervised by a class teacher. Physical fitness was evaluated before and after the intervention. Saliva samples were collected post-intervention in all participants and analyzed using PCR amplification of 16S rRNA bacterial genes. The Amplicon Sequence Variants were filtered, decontaminated, and phylogenetically classified using DADA2 software. Differential abundance analysis of microbiome taxa and pathway data was conducted using the LEfSe algorithm and PICRUSt. Results The Sa group showed better performances in lower limb power and sprint performance while both the Sa and Dm groups improved in endurance and balance at the end of the intervention; only balance resulted slightly improved in the Ct group. Among the genera differently enriched in saliva after the training intervention, we found that the Prevotella, the Dubosiella and the Family XIII AD3011 group were the most abundant in the Sa group; differently, the Neisseria and the Abiotrophia in Ct group. Four species showed significant the Prevotella melaninogenica and the Prevotella nanceiensis were more abundant in the Sa, conversely, Gemella sanguinis was enriched in Dm and Abiotrophia defectiva in Ct saliva group. Conclusion We demonstrated that Sa and Dm, not curricular exercise, improve the physical fitness components in sedentary schoolchildren correlated to health and promote an enrichment in saliva microbiota species associated to a healthier profile.
Collapse
Affiliation(s)
- Annamaria Mancini
- Department of Medicine, Movement Sciences and Wellness, University Parthenope, Naples, Italy
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”, Napoli, Italy
| | - Daniela Vitucci
- Department of Medicine, Movement Sciences and Wellness, University Parthenope, Naples, Italy
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”, Napoli, Italy
| | | | - Corrado Lupo
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Mario Capasso
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”, Napoli, Italy
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Stefania Orrù
- Department of Medicine, Movement Sciences and Wellness, University Parthenope, Naples, Italy
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”, Napoli, Italy
| | - Alberto Rainoldi
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Federico Schena
- Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Pasqualina Buono
- Department of Medicine, Movement Sciences and Wellness, University Parthenope, Naples, Italy
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”, Napoli, Italy
| |
Collapse
|
5
|
Suparan K, Trirattanapa K, Piriyakhuntorn P, Sriwichaiin S, Thonusin C, Nawara W, Kerdpoo S, Chattipakorn N, Tantiworawit A, Chattipakorn SC. Exploring alterations of gut/blood microbes in addressing iron overload-induced gut dysbiosis and cognitive impairment in thalassemia patients. Sci Rep 2024; 14:24951. [PMID: 39438708 PMCID: PMC11496663 DOI: 10.1038/s41598-024-76684-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024] Open
Abstract
Iron overload causes cognitive impairment in thalassemia patients. The gut-brain axis plays an important role in cognitive function. However, the association between gut/blood microbiome, cognition, and iron burden in thalassemia patients has not been thoroughly investigated. We aimed to determine those associations in thalassemia patients with different blood-transfusion regimens. Sixty participants: healthy controls, transfusion-dependent thalassemia (TDT) patients, and non-transfusion-dependent (NTDT) patients, were recruited to evaluate iron overload, cognition, and gut/blood microbiome. TDT patients exhibited greater iron overload than NTDT patients. Most thalassemia patients developed gut dysbiosis, and approximately 25% of the patients developed minor cognitive impairment. Increased Fusobacteriota and Verrucomicrobiota with decreased Fibrobacterota were observed in both TDT and NTDT groups. TDT patients showed more abundant beneficial bacteria: Verrucomicrobia. Iron overload was correlated with cognitive impairment. Increased Butyricimonas and decreased Paraclostridium were associated with higher cognitive function. No trace of blood microbiota was observed. Differences in blood bacterial profiles of thalassemia patients and controls were insignificant. These findings suggest iron overload plays a role in the imbalance of gut microbiota and impaired cognitive function in thalassemia patients. Harnessing probiotic potential from those microbes could prevent the gut-brain disturbance in thalassemia patients.
Collapse
Affiliation(s)
- Kanokphong Suparan
- Immunology Unit, Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Kornkanok Trirattanapa
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Pokpong Piriyakhuntorn
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sirawit Sriwichaiin
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Chanisa Thonusin
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Wichwara Nawara
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sasiwan Kerdpoo
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| | - Adisak Tantiworawit
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
6
|
Ghorashi AC, Boucher A, Archer-Hartmann SA, Zalem D, Taherzadeh Ghahfarrokhi M, Murray NB, Konada RSR, Zhang X, Xing C, Teneberg S, Azadi P, Yrlid U, Kohler JJ. Fucosylation of glycoproteins and glycolipids: opposing roles in cholera intoxication. Nat Chem Biol 2024:10.1038/s41589-024-01748-5. [PMID: 39414978 DOI: 10.1038/s41589-024-01748-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/13/2024] [Indexed: 10/18/2024]
Abstract
Cholera toxin (CT) is the etiological agent of cholera. Here we report that multiple classes of fucosylated glycoconjugates function in CT binding and intoxication of intestinal epithelial cells. In Colo205 cells, knockout (KO) of B3GNT5, which encodes an enzyme required for synthesis of lacto and neolacto series glycosphingolipids (GSLs), reduces CT binding but sensitizes cells to intoxication. Overexpressing B3GNT5 to generate more fucosylated GSLs confers protection against intoxication, indicating that fucosylated GSLs act as decoy receptors for CT. KO of B3GALT5 causes increased production of fucosylated O-linked and N-linked glycoproteins and leads to increased CT binding and intoxication. KO of B3GNT5 in B3GALT5-KO cells eliminates production of fucosylated GSLs but increases intoxication, identifying fucosylated glycoproteins as functional receptors for CT. These findings provide insight into the molecular determinants regulating CT sensitivity of host cells.
Collapse
Affiliation(s)
- Atossa C Ghorashi
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Andrew Boucher
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | | | - Dani Zalem
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | | | - Nathan B Murray
- Complex Carbohydrate Research Center, The University of Georgia, Athens, GA, USA
| | | | - Xunzhi Zhang
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA
| | - Chao Xing
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Susann Teneberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, The University of Georgia, Athens, GA, USA
| | - Ulf Yrlid
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Jennifer J Kohler
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
7
|
Starosta RT, Lee AJ, Toolan ER, He M, Wongkittichote P, Daniel EJP, Radenkovic S, Budhraja R, Pandey A, Sharma J, Morava E, Nguyen H, Dickson PI. D-mannose as a new therapy for fucokinase deficiency-related congenital disorder of glycosylation (FCSK-CDG). Mol Genet Metab 2024; 142:108488. [PMID: 38735264 DOI: 10.1016/j.ymgme.2024.108488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 04/22/2024] [Accepted: 05/02/2024] [Indexed: 05/14/2024]
Abstract
INTRODUCTION Fucokinase deficiency-related congenital disorder of glycosylation (FCSK-CDG) is a rare autosomal recessive inborn error of metabolism characterized by a decreased flux through the salvage pathway of GDP-fucose biosynthesis due to a block in the recycling of L-fucose that exits the lysosome. FCSK-CDG has been described in 5 individuals to date in the medical literature, with a phenotype comprising global developmental delays/intellectual disability, hypotonia, abnormal myelination, posterior ocular disease, growth and feeding failure, immune deficiency, and chronic diarrhea, without clear therapeutic recommendations. PATIENT AND METHODS In a so far unreported FCSK-CDG patient, we studied proteomics and glycoproteomics in vitro in patient-derived fibroblasts and also performed in vivo glycomics, before and after treatment with either D-Mannose or L-Fucose. RESULTS We observed a marked increase in fucosylation after D-mannose supplementation in fibroblasts compared to treatment with L-Fucose. The patient was then treated with D-mannose at 850 mg/kg/d, with resolution of the chronic diarrhea, resolution of oral aversion, improved weight gain, and observed developmental gains. Serum N-glycan profiles showed an improvement in the abundance of fucosylated glycans after treatment. No treatment-attributed adverse effects were observed. CONCLUSION D-mannose is a promising new treatment for FCSK-CDG.
Collapse
Affiliation(s)
- Rodrigo Tzovenos Starosta
- Division of Medical Genetics and Genomics, Washington University School of Medicine, St. Louis, MO, USA; Division of Clinical Genetics and Metabolism, University of Colorado Anschutz, Aurora, CO, USA; Graduate Program in Science: Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Angela J Lee
- Division of Medical Genetics and Genomics, Washington University School of Medicine, St. Louis, MO, USA
| | - Elizabeth R Toolan
- Division of Medical Genetics and Genomics, Washington University School of Medicine, St. Louis, MO, USA
| | - Miao He
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Parith Wongkittichote
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Earnest James Paul Daniel
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Rohit Budhraja
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, MN, USA
| | - Akhilesh Pandey
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jaiprakash Sharma
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Eva Morava
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| | - Hoanh Nguyen
- Division of Medical Genetics and Genomics, Washington University School of Medicine, St. Louis, MO, USA
| | - Patricia I Dickson
- Division of Medical Genetics and Genomics, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
8
|
Yang X, Zhang J, Zhu J, Yang R, Tong Y. Molecular insights into FucR transcription factor to control the metabolism of L-fucose in Bifidobacterium longum subsp. infantis. Microbiol Res 2024; 283:127709. [PMID: 38593579 DOI: 10.1016/j.micres.2024.127709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/27/2024] [Accepted: 03/30/2024] [Indexed: 04/11/2024]
Abstract
Bifidobacterium longum subsp. infantis commonly colonizes the human gut and is capable of metabolizing L-fucose, which is abundant in the gut. Multiple studies have focused on the mechanisms of L-fucose utilization by B. longum subsp. infantis, but the regulatory pathways governing the expression of these catabolic processes are still unclear. In this study, we have conducted a structural and functional analysis of L-fucose metabolism transcription factor FucR derived from B. longum subsp. infantis Bi-26. Our results indicated that FucR is a L-fucose-sensitive repressor with more α-helices, fewer β-sheets, and β-turns. Transcriptional analysis revealed that FucR displays weak negative self-regulation, which is counteracted in the presence of L-fucose. Isothermal titration calorimetry indicated that FucR has a 2:1 stoichiometry with L-fucose. The key amino acid residues for FucR binding L-fucose are Asp280 and Arg331, with mutation of Asp280 to Ala resulting in a decrease in the affinity between FucR and L-fucose with the Kd value from 2.58 to 11.68 μM, and mutation of Arg331 to Ala abolishes the binding ability of FucR towards L-fucose. FucR specifically recognized and bound to a 20-bp incomplete palindrome sequence (5'-ACCCCAATTACGAAAATTTTT-3'), and the affinity of the L-fucose-loaded FucR for the DNA fragment was lower than apo-FucR. The results provided new insights into the regulating L-fucose metabolism by B. longum subsp. infantis.
Collapse
Affiliation(s)
- Xiaojun Yang
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jing Zhang
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jing Zhu
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ruijin Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yanjun Tong
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
9
|
Li Y, Ning X, Zhao Z, He X, Xue Q, Zhou M, Li W, Li M. Core fucosylation of maternal milk N-glycans imparts early-life immune tolerance through gut microbiota-dependent regulation in RORγt + Treg cells. Food Funct 2024; 15:4140-4153. [PMID: 38445991 DOI: 10.1039/d4fo00230j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Milk glycans play key roles in shaping and maintaining a healthy infant gut microbiota. Core fucosylation catalyzed by fucosyltransferase (Fut8) is the major glycosylation pattern on human milk N-glycan, which was crucial for promoting the colonization and dominant growth of Bifidobacterium and Lactobacillus spp. in neonates. However, the influence of core-fucose in breast milk on the establishment of early-life immune tolerance remains poorly characterized. In this study, we found that the deficiency of core-fucose in the milk of maternal mice caused by Fut8 gene heterozygosity (Fut8+/-) resulted in poor immune tolerance towards the ovalbumin (OVA) challenge, accompanied by a reduced proportion of intestinal RORγt+ Treg cells and the abundance of Lactobacillus spp., especially L. reuteri and L. johnsonii, in their breast-fed neonates. The administration of the L. reuteri and L. johnsonii mixture to neonatal mice compromised the OVA-induced allergy and up-regulated the intestinal RORγt+ Treg cell proportions. However, Lactobacillus mixture supplementation did not alleviate allergic responses in RORγt+ Treg cell-deficient mice caused by Rorc gene heterozygosity (Rorc+/-) post OVA challenge, indicating that the intervention effects depend on the RORγt+ Treg cells. Interestingly, instead of L. reuteri and L. johnsonii, we found that the relative abundance of another Lactobacillus spp., L. murinus, in the gut of the offspring mice was significantly promoted by intervention, which showed enhancing effects on the proliferation of splenic and intestinal RORγt+ Treg cells in in vitro studies. The above results indicate that core fucosylation of breast milk N-glycans is beneficial for the establishment of RORγt+ Treg cell mediated early-life immune tolerance through the manipulation of symbiotic bacteria in mice.
Collapse
Affiliation(s)
- Yuyuan Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China.
| | - Xixi Ning
- College of Basic Medical Science, Dalian Medical University, Dalian, China.
| | - Zihui Zhao
- Pelvic Floor Repair Center, Dalian Women and Children's Medical Group, Dalian, China
| | - Xi He
- College of Basic Medical Science, Dalian Medical University, Dalian, China.
| | - Qidi Xue
- College of Basic Medical Science, Dalian Medical University, Dalian, China.
| | - Manlin Zhou
- College of Basic Medical Science, Dalian Medical University, Dalian, China.
| | - Wenzhe Li
- Shantou University Medical College, Shantou, Guangdong, China.
| | - Ming Li
- College of Basic Medical Science, Dalian Medical University, Dalian, China.
| |
Collapse
|
10
|
Li W, Wu P, Jin T, Jia J, Chen B, Liu T, Liu Y, Mei J, Luo B, Zhang Z. L-fucose and fucoidan alleviate high-salt diet-promoted acute inflammation. Front Immunol 2024; 15:1333848. [PMID: 38596683 PMCID: PMC11002173 DOI: 10.3389/fimmu.2024.1333848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/05/2024] [Indexed: 04/11/2024] Open
Abstract
Excessive salt intake is a widespread health issue observed in almost every country around the world. A high salt diet (HSD) has a strong correlation with numerous diseases, including hypertension, chronic kidney disease, and autoimmune disorders. However, the mechanisms underlying HSD-promotion of inflammation and exacerbation of these diseases are not fully understood. In this study, we observed that HSD consumption reduced the abundance of the gut microbial metabolite L-fucose, leading to a more substantial inflammatory response in mice. A HSD led to increased peritonitis incidence in mice, as evidenced by the increased accumulation of inflammatory cells and elevated levels of inflammatory cytokines, such as tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6), and monocyte chemotactic protein-1 (MCP-1, also known as C-C motif chemokine ligand 2 or CCL2), in peritoneal lavage fluid. Following the administration of broad-spectrum antibiotics, HSD-induced inflammation was abolished, indicating that the proinflammatory effects of HSD were not due to the direct effect of sodium, but rather to HSD-induced alterations in the composition of the gut microbiota. By using untargeted metabolomics techniques, we determined that the levels of the gut microbial metabolite L-fucose were reduced by a HSD. Moreover, the administration of L-fucose or fucoidan, a compound derived from brown that is rich in L-fucose, normalized the level of inflammation in mice following HSD induction. In addition, both L-fucose and fucoidan inhibited LPS-induced macrophage activation in vitro. In summary, our research showed that reduced L-fucose levels in the gut contributed to HSD-exacerbated acute inflammation in mice; these results indicate that L-fucose and fucoidan could interfere with HSD-promotion of the inflammatory response.
Collapse
Affiliation(s)
- Wenhua Li
- Institute of Immunology, Third Military Medical University, Chongqing, China
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pengfei Wu
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Tianrong Jin
- Medical College of Chongqing University, Chongqing, China
| | - Jialin Jia
- Medical College of Chongqing University, Chongqing, China
| | - Bo Chen
- College of Acupuncture and Tuina, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Tingting Liu
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Yu Liu
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Jie Mei
- College of Acupuncture and Tuina, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Bangwei Luo
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Zhiren Zhang
- Institute of Immunology, Third Military Medical University, Chongqing, China
| |
Collapse
|
11
|
Krupinskaitė A, Stanislauskienė R, Serapinas P, Rutkienė R, Gasparavičiūtė R, Meškys R, Stankevičiūtė J. α-L-Fucosidases from an Alpaca Faeces Metagenome: Characterisation of Hydrolytic and Transfucosylation Potential. Int J Mol Sci 2024; 25:809. [PMID: 38255883 PMCID: PMC10815079 DOI: 10.3390/ijms25020809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/05/2024] [Accepted: 01/07/2024] [Indexed: 01/24/2024] Open
Abstract
In various life forms, fucose-containing glycans play vital roles in immune recognition, developmental processes, plant immunity, and host-microbe interactions. Together with glucose, galactose, N-acetylglucosamine, and sialic acid, fucose is a significant component of human milk oligosaccharides (HMOs). Fucosylated HMOs benefit infants by acting as prebiotics, preventing pathogen attachment, and potentially protecting against infections, including HIV. Although the need for fucosylated derivatives is clear, their availability is limited. Therefore, synthesis methods for various fucosylated oligosaccharides are explored, employing enzymatic approaches and α-L-fucosidases. This work aimed to characterise α-L-fucosidases identified in an alpaca faeces metagenome. Based on bioinformatic analyses, they were confirmed as members of the GH29A subfamily. The recombinant α-L-fucosidases were expressed in Escherichia coli and showed hydrolytic activity towards p-nitrophenyl-α-L-fucopyranoside and 2'-fucosyllactose. Furthermore, the enzymes' biochemical properties and kinetic characteristics were also determined. All four α-L-fucosidases could catalyse transfucosylation using a broad diversity of fucosyl acceptor substrates, including lactose, maltotriose, L-serine, and L-threonine. The results contribute insights into the potential use of α-L-fucosidases for synthesising fucosylated amino acids.
Collapse
Affiliation(s)
- Agnė Krupinskaitė
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Sauletekio av. 7, LT-10257 Vilnius, Lithuania; (R.S.); (P.S.); (R.R.); (R.G.); (R.M.)
| | | | | | | | | | | | - Jonita Stankevičiūtė
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Sauletekio av. 7, LT-10257 Vilnius, Lithuania; (R.S.); (P.S.); (R.R.); (R.G.); (R.M.)
| |
Collapse
|
12
|
Wang Y, Ma W, Mehta R, Nguyen LH, Song M, Drew DA, Asnicar F, Huttenhower C, Segata N, Wolf J, Spector T, Berry S, Staller K, Chan AT. Diet and gut microbial associations in irritable bowel syndrome according to disease subtype. Gut Microbes 2023; 15:2262130. [PMID: 37786251 PMCID: PMC10549191 DOI: 10.1080/19490976.2023.2262130] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/15/2023] [Indexed: 10/04/2023] Open
Abstract
The role of diet and the gut microbiome in the etiopathogenesis of irritable bowel syndrome (IBS) is not fully understood. Therefore, we investigated the interplay between dietary risk factors and gut microbiota in IBS subtypes using a food frequency questionnaire and stool metagenome data from 969 participants aged 18-65 years in the ZOE PREDICT 1 study, an intervention study designed to predict postprandial metabolic responses. We identified individuals with IBS subtype according to the Rome III criteria based on predominant bowel habits during symptom onset: diarrhea (i.e. looser), constipation (i.e. harder), and mixed. Participants with IBS-D (n = 59) consumed more healthy plant-based foods (e.g. whole grains, leafy vegetables) and fiber, while those with IBS-C (n = 49) tended to consume more unhealthy plant-based foods (e.g. refined grains, fruit juice) than participants without IBS (n = 797). Microbial diversity was nominally lower in patients with IBS-D than in participants without IBS or with IBS-C. Using multivariable-adjusted linear regression, we identified specific microbiota variations in IBS subtypes, including slight increases in pro-inflammatory taxa in IBS-C (e.g. Escherichia coli) and loss of strict anaerobes in IBS-D (e.g. Faecalibacterium prausnitzii). Our analysis also revealed intriguing evidence of interactions between diet and Faecalibacterium prausnitzii. The positive associations between fiber and iron intake and IBS-diarrhea were stronger among individuals with a higher relative abundance of Faecalibacterium prausnitzii, potentially driven by carbohydrate metabolic pathways, including the superpathway of β-D-glucuronide and D-glucuronate degradation. In conclusion, our findings suggest subtype-specific variations in dietary habits, gut microbial composition and function, and diet-microbiota interactions in IBS, providing insights into potential microbiome-informed dietary interventions.
Collapse
Affiliation(s)
- Yiqing Wang
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Wenjie Ma
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Raaj Mehta
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Long H. Nguyen
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mingyang Song
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, USA
| | - David A. Drew
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Francesco Asnicar
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nicola Segata
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
- European Institute of Oncology Scientific Institute for Research, Hospitalization and Healthcare, Milan, Italy
| | | | - Tim Spector
- Department of Twin Research, King’s College London, London, UK
| | - Sarah Berry
- Department of Nutritional Sciences, King’s College London, London, UK
| | - Kyle Staller
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew T. Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, USA
| |
Collapse
|
13
|
Djouina M, Waxin C, Caboche S, Lecointe K, Steimle A, Beury D, Desai MS, Hot D, Dubuquoy L, Launay D, Vignal C, Body-Malapel M. Low dose dietary contamination with deoxynivalenol mycotoxin exacerbates enteritis and colorectal cancer in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 900:165722. [PMID: 37482350 DOI: 10.1016/j.scitotenv.2023.165722] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 07/25/2023]
Abstract
BACKGROUND The mycotoxin deoxynivalenol (DON) is a frequent contaminant of grain and cereal products worldwide. Exposure to DON can cause gastrointestinal inflammation, disturb gut barrier function, and induce gut dysbiosis in vivo under basal conditions, but little is known about the effects of DON ingestion in individuals with pre-existing gastrointestinal disease. OBJECTIVES Mice were orally exposed to 10 and 100 μg/kg bw/day of DON, corresponding to 10 to 100-fold human tolerable daily intake concentrations, and to the translation in mice of current human daily intake. The effects of DON exposure were explored under steady-state conditions, and in murine models of enteritis and colorectal cancer (CRC). RESULTS After 8 days of DON exposure, an increase of histomorphological and molecular parameters of epithelial proliferation were observed in normal mice, from the duodenum to the colon. The same exposure in a murine model of indomethacin-induced enteritis led to exacerbation of lesion development and induction of ileal cytokines. DON exposure also worsened the development of colitis-associated CRC in mice as shown by increases in endoscopic and histological colitis scores, tumor grades, and histological hyperplasia. In colon of DON-exposed mice, upstream and downstream ERK signaling genes were upregulated including Mapk1, Mapk3, Map 2k1, Map2k2 core ERK pathway effectors, and Bcl2 and Bcl2l1 antiapoptotic genes. The effects observed in the CRC model were associated with alterations in cecal microbiota taxonomic composition and metabolism of bacterial fucose and rhamnose. Strong Spearman's correlations were revealed between the relative abundance of the changed bacterial genera and CRC-related variables. DISCUSSION Ingestion of DON mycotoxin at concentrations representative of human real-world exposure worsened the development of indomethacin-induced enteritis and colitis-associated CRC in mice. Our results suggest that even at low doses, which are currently tolerated in the human diet, DON could promote the development of intestinal inflammatory diseases and CRC.
Collapse
Affiliation(s)
- Madjid Djouina
- Univ. Lille, Inserm, CHU Lille, U1286- INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Christophe Waxin
- Univ. Lille, Inserm, CHU Lille, U1286- INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Ségolène Caboche
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US41-UAR 2014-PLBS, F-59000 Lille, France
| | - Karine Lecointe
- Inserm U1285, Univ. Lille, CHU de Lille, UMR CNRS 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59000 Lille, France
| | - Alexander Steimle
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Delphine Beury
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US41-UAR 2014-PLBS, F-59000 Lille, France
| | - Mahesh S Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - David Hot
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US41-UAR 2014-PLBS, F-59000 Lille, France
| | - Laurent Dubuquoy
- Univ. Lille, Inserm, CHU Lille, U1286- INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - David Launay
- Univ. Lille, Inserm, CHU Lille, U1286- INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Cécile Vignal
- Univ. Lille, Inserm, CHU Lille, U1286- INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Mathilde Body-Malapel
- Univ. Lille, Inserm, CHU Lille, U1286- INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France.
| |
Collapse
|
14
|
Orłowska M, Barua D, Piłsyk S, Muszewska A. Fucose as a nutrient ligand for Dikarya and a building block of early diverging lineages. IMA Fungus 2023; 14:17. [PMID: 37670396 PMCID: PMC10481521 DOI: 10.1186/s43008-023-00123-8] [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: 02/13/2023] [Accepted: 08/07/2023] [Indexed: 09/07/2023] Open
Abstract
Fucose is a deoxyhexose sugar present and studied in mammals. The process of fucosylation has been the primary focus in studies relating to fucose in animals due to the presence of fucose in Lewis antigens. Very few studies have reported its presence in Fungi, mostly in Mucoromycotina. The constitution of 25% and 12% of this sugar in the carbohydrates of cell wall in the respective Umbelopsis and Mucorales strains boosts the need to bridge the gap of knowledge on fucose metabolism across the fungal tree of life. In the absence of a network map involving fucose proteins, we carried out an in-silico approach to construct the fucose metabolic map in Fungi. We analyzed the taxonomic distribution of 85 protein families in Fungi including diverse early diverging fungal lineages. The expression of fucose-related protein-coding genes proteins was validated with the help of transcriptomic data originating from representatives of early diverging fungi. We found proteins involved in several metabolic activities apart from fucosylation such as synthesis, transport and binding. Most of the identified protein families are shared with Metazoa suggesting an ancestral origin in Opisthokonta. However, the overall complexity of fucose metabolism is greater in Metazoa than in Fungi. Massive gene loss has shaped the evolutionary history of these metabolic pathways, leading to a repeated reduction of these pathways in most yeast-forming lineages. Our results point to a distinctive mode of utilization of fucose among fungi belonging to Dikarya and the early diverging lineages. We speculate that, while Dikarya used fucose as a source of nutrients for metabolism, the early diverging group of fungi depended on fucose as a building block and signaling compound.
Collapse
Affiliation(s)
- Małgorzata Orłowska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5A, 02-106, Warsaw, Poland.
- Institute of Evolutionary Biology, Faculty of Biology, Biological and Chemical Research Centre, University of Warsaw, Zwirki i Wigury 101, 02-089, Warsaw, Poland.
| | - Drishtee Barua
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5A, 02-106, Warsaw, Poland
| | - Sebastian Piłsyk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5A, 02-106, Warsaw, Poland
| | - Anna Muszewska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5A, 02-106, Warsaw, Poland.
| |
Collapse
|
15
|
Ghorashi AC, Boucher A, Archer-Hartmann SA, Murray NB, Konada RSR, Zhang X, Xing C, Azadi P, Yrlid U, Kohler JJ. Fucosylated glycoproteins and fucosylated glycolipids play opposing roles in cholera intoxication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.02.551727. [PMID: 37577488 PMCID: PMC10418270 DOI: 10.1101/2023.08.02.551727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Cholera toxin (CT) is the etiological agent of cholera. Here we report that multiple classes of fucosylated glycoconjugates function in CT binding and intoxication of intestinal epithelial cells. In Colo205 cells, knockout of B3GNT5, the enzyme required for synthesis of lacto- and neolacto-series glycosphingolipids (GSLs), reduces CT binding but sensitizes cells to intoxication. Overexpressing B3GNT5 to generate more fucosylated GSLs confers protection against intoxication, indicating that fucosylated GSLs act as decoy receptors for CT. Knockout (KO) of B3GALT5 causes increased production of fucosylated O-linked and N-linked glycoproteins, and leads to increased CT binding and intoxication. Knockout of B3GNT5 in B3GALT5 KO cells eliminates production of fucosylated GSLs but increases intoxication, identifying fucosylated glycoproteins as functional receptors for CT. These findings provide insight into molecular determinants regulating CT sensitivity of host cells.
Collapse
Affiliation(s)
- Atossa C. Ghorashi
- Department of Biochemistry, UT Southwestern Medical Center, Dallas TX 75390 USA
| | - Andrew Boucher
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | | | - Nathan B. Murray
- Complex Carbohydrate Research Center, The University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | | | - Xunzhi Zhang
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas TX 75390 USA
| | - Chao Xing
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas TX 75390 USA
- Department of Bioinformatics, UT Southwestern Medical Center, Dallas TX 75390 USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, The University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - Ulf Yrlid
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Jennifer J. Kohler
- Department of Biochemistry, UT Southwestern Medical Center, Dallas TX 75390 USA
| |
Collapse
|
16
|
Kim J, Jin YS, Kim KH. L-Fucose is involved in human-gut microbiome interactions. Appl Microbiol Biotechnol 2023:10.1007/s00253-023-12527-y. [PMID: 37148338 DOI: 10.1007/s00253-023-12527-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/30/2023] [Accepted: 04/10/2023] [Indexed: 05/08/2023]
Abstract
L-Fucose is one of the key metabolites in human-gut microbiome interactions. It is continuously synthesized by humans in the form of fucosylated glycans and fucosyl-oligosaccharides and delivered into the gut throughout their lifetime. Gut microorganisms metabolize L-fucose and produce short-chain fatty acids, which are absorbed by epithelial cells and used as energy sources or signaling molecules. Recent studies have revealed that the carbon flux in L-fucose metabolism by gut microorganisms is distinct from that in other sugar metabolisms because of cofactor imbalance and low efficiencies in energy synthesis of L-fucose metabolism. The large amounts of short-chain fatty acids produced during microbial L-fucose metabolism are used by epithelial cells to recover most of the energy used up during L-fucose synthesis. In this review, we present a detailed overview of microbial L-fucose metabolism and a potential solution for disease treatment and prevention using genetically engineered probiotics that modulate fucose metabolism. Our review contributes to the understanding of human-gut microbiome interactions through L-fucose metabolism. KEY POINTS: • Fucose-metabolizing microorganisms produce large amounts of short-chain fatty acids • Fucose metabolism differs from other sugar metabolisms by cofactor imbalance • Modulating fucose metabolism is the key to control host-gut microbiome interactions.
Collapse
Affiliation(s)
- Jungyeon Kim
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yong-Su Jin
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| | - Kyoung Heon Kim
- Department of Biotechnology, Graduate School, Korea University, Seoul, 02841, Republic of Korea.
- Department of Food Bioscience and Technology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
17
|
Structure and function of microbial α-l-fucosidases: a mini review. Essays Biochem 2023; 67:399-414. [PMID: 36805644 PMCID: PMC10154630 DOI: 10.1042/ebc20220158] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 02/23/2023]
Abstract
Fucose is a monosaccharide commonly found in mammalian, insect, microbial and plant glycans. The removal of terminal α-l-fucosyl residues from oligosaccharides and glycoconjugates is catalysed by α-l-fucosidases. To date, glycoside hydrolases (GHs) with exo-fucosidase activity on α-l-fucosylated substrates (EC 3.2.1.51, EC 3.2.1.-) have been reported in the GH29, GH95, GH139, GH141 and GH151 families of the Carbohydrate Active Enzymes (CAZy) database. Microbes generally encode several fucosidases in their genomes, often from more than one GH family, reflecting the high diversity of naturally occuring fucosylated structures they encounter. Functionally characterised microbial α-l-fucosidases have been shown to act on a range of substrates with α-1,2, α-1,3, α-1,4 or α-1,6 fucosylated linkages depending on the GH family and microorganism. Fucosidases show a modular organisation with catalytic domains of GH29 and GH151 displaying a (β/α)8-barrel fold while GH95 and GH141 show a (α/α)6 barrel and parallel β-helix fold, respectively. A number of crystal structures have been solved in complex with ligands, providing structural basis for their substrate specificity. Fucosidases can also be used in transglycosylation reactions to synthesise oligosaccharides. This mini review provides an overview of the enzymatic and structural properties of microbial α-l-fucosidases and some insights into their biological function and biotechnological applications.
Collapse
|
18
|
Meng J, Zhu Y, Chen R, Liu Y, Zhang W, Mu W. Microbial Synthesis of l-Fucose with High Productivity by a Metabolically Engineered Escherichia coli. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:2464-2471. [PMID: 36700831 DOI: 10.1021/acs.jafc.2c08906] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
l-Fucose is a natural deoxy hexose found in a variety of organisms. It possesses many physiological effects and has potential applications in pharmaceutical, cosmetic, and food industries. Microbial synthesis via metabolic engineering attracts increasing attention for efficient production of important chemicals. Previously, we reported the construction of a metabolically engineered Escherichia coli strain with high 2'-fucosyllactose productivity. Herein, we further introduced Bifidobacterium bifidum α-l-fucosidase via both plasmid expression and genomic integration and blocked the l-fucose assimilation pathway by deleting fucI, fucK, and rhaA. The highest l-fucose titers reached 6.31 and 51.05 g/L in shake-flask and fed-batch cultivation, respectively. l-Fucose synthesis was little affected by lactose added, and there was almost no 2'-fucosyllactose residue throughout the cultivation processes. The l-fucose productivity reached 0.76 g/L/h, indicating significant potential for large-scale industrial applications.
Collapse
Affiliation(s)
- Jiawei Meng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Yingying Zhu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Roulin Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Yuanlin Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Wenli Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| |
Collapse
|
19
|
Janapatla RP, Dudek A, Chen CL, Chuang CH, Chien KY, Feng Y, Yeh YM, Wang YH, Chang HJ, Lee YC, Chiu CH. Marine prebiotics mediate decolonization of Pseudomonas aeruginosa from gut by inhibiting secreted virulence factor interactions with mucins and enriching Bacteroides population. J Biomed Sci 2023; 30:9. [PMID: 36732731 PMCID: PMC9896862 DOI: 10.1186/s12929-023-00902-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/12/2023] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Pseudomonas aeruginosa intestinal carriage rates are significantly higher in immunosuppressed individuals and hospitalized patients who therefore have increased risk of infections and antibiotic-associated diarrhea. To combat intestinal dysbiosis and decolonize P. aeruginosa from gastrointestinal tract, we investigated the anti-adherence and gut microbiota modulation properties of marine prebiotic fucoidans. METHODS Proteomic analysis of culture supernatant was performed by LC-MS/MS. Using lectin-based enzyme-linked immunosorbent assay, hemagglutinin domain interaction and inhibition with biomolecules were studied. We investigated the role of nutritional grade fucoidans in a mouse model and used 16S ribosomal RNA sequencing to examine fecal microbiota composition. RESULTS Analysis of culture supernatant proteins indicated the secretion of two-partner secretion (TPS) family proteins, including TpsA1/CdiA2 and TpsA2/CdiA1. Lectin like activity at the N-terminal of TpsA due to a conserved hemagglutinin domain (Pfam identifier [ID] PF05860) mediates binding to mucins that carry multiple fucosylated glycans. Fucose-rich sulfated polysaccharides (fucoidans) and sulfated dextrans were found to be potent inhibitors of the recombinant N-terminal hemagglutinin domain of TpsA (TpsA-NT-HAD) binding to mucins. In a mouse model, antibiotic-induced dysbiosis was essential for P. aeruginosa gastrointestinal colonization. After prophylactic oral fucoidans supplementation, a higher proportion (60%) of the mice were decolonized over time and resisted re-colonization, this was associated with remarkable expansion of Bacteroides (post-infection day-3 abundance, 29-50%) and consequential reductions in bloom of Enterobacteriaceae and Enterococcaceae populations. In the non-supplemented group, Parabacteroides mediated recovery from dysbiosis but failed to decolonize P. aeruginosa. CONCLUSIONS Supplementing diet with marine prebiotic fucoidans can mediate earlier recovery from dysbiosis and decolonization of P. aeruginosa from gut by inhibiting secreted virulence factor (TpsA/CdiA) interaction with mucins and promoting the growth of beneficial Bacteroides population. We suggest the prophylactic use of nutritional grade fucoidans to decolonize P. aeruginosa from gastrointestinal tract of at-risk individuals to prevent infection and transmission of colonizing P. aeruginosa.
Collapse
Affiliation(s)
- Rajendra Prasad Janapatla
- grid.413801.f0000 0001 0711 0593Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Anna Dudek
- grid.413801.f0000 0001 0711 0593Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chyi-Liang Chen
- grid.413801.f0000 0001 0711 0593Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | | | - Kun-Yi Chien
- grid.145695.a0000 0004 1798 0922Graduate Institute of Biomedical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Ye Feng
- grid.13402.340000 0004 1759 700XInstitute for Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuan-Ming Yeh
- grid.413801.f0000 0001 0711 0593Chang Gung Microbiota Therapy Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yi-Hsin Wang
- grid.413801.f0000 0001 0711 0593Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Hsin-Ju Chang
- grid.413801.f0000 0001 0711 0593Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yuan-Chuan Lee
- grid.21107.350000 0001 2171 9311Department of Biology, Johns Hopkins University, Baltimore, MD USA
| | - Cheng-Hsun Chiu
- grid.413801.f0000 0001 0711 0593Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan ,grid.413801.f0000 0001 0711 0593Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| |
Collapse
|
20
|
Lin SJH, Helm ET, Gabler NK, Burrough ER. Acute infection with Brachyspira hyodysenteriae affects mucin expression, glycosylation, and fecal MUC5AC. Front Cell Infect Microbiol 2023; 12:1042815. [PMID: 36683692 PMCID: PMC9852840 DOI: 10.3389/fcimb.2022.1042815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/28/2022] [Indexed: 01/08/2023] Open
Abstract
Introduction Infection with strongly β-hemolytic strains of Brachyspira hyodysenteriae leads to swine dysentery (SD), a production-limiting disease that causes mucohemorrhagic diarrhea and typhlocolitis in pigs. This pathogen has strong chemotactic activity toward mucin, and infected pigs often have a disorganized mucus layer and marked de novo expression of MUC5AC, which is not constitutively expressed in the colon. It has been shown that fucose is chemoattractant for B. hyodysenteriae, and a highly fermentable fiber diet can mitigate and delay the onset of SD. Methods We used lectins targeting sialic acids in α-2,6 or α-2,3 linkages, N-acetylglucosamine (GlcNAc), α-linked L-fucose, and an immunohistochemical stain targeting N-glycolylneuraminic acid (NeuGc) to investigate the local expression of these mucin glycans in colonic tissues of pigs with acute SD. We used a commercial enzyme-linked immunosorbent assay (ELISA) to quantify fecal MUC5AC in infected pigs and assess its potential as a diagnostic monitoring tool and RNA in situ hybridization to detect IL-17A in the colonic mucosa. Results Colonic mucin glycosylation during SD has an overall increase in fucose, a spatially different distribution of GlcNAc with more expression within the crypt lumens of the upper colonic mucosa, and decreased expression or a decreased trend of sialic acids in α-2,6 or α-2,3 linkages, and NeuGc compared to the controls. The degree of increased fucosylation was less in the colonic mucosa of pigs with SD and fed the highly fermentable fiber diet. There was a significant increase in MUC5AC in fecal and colonic samples of pigs with SD at the endpoint compared to the controls, but the predictive value for disease progression was limited. Discussion Fucosylation and the impact of dietary fiber may play important roles in the pathogenesis of SD. The lack of predictive value for fecal MUC5AC quantification by ELISA is possibly due to the presence of other non-colonic sources of MUC5AC in the feces. The moderate correlation between IL-17A, neutrophils and MUC5AC confirms its immunoregulatory and mucin stimulatory role. Our study characterizes local alteration of mucin glycosylation in the colonic mucosa of pigs with SD after B. hyodysenteriae infection and may provide insight into host-pathogen interaction.
Collapse
Affiliation(s)
- Susanne Je-Han Lin
- Department of Veterinary Pathology, Iowa State University, Ames, IA, United States
| | - Emma T Helm
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Nicholas K Gabler
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Eric R Burrough
- Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
21
|
Chun J, Kang T, Seo JP, Jeong H, Kim M, Kim BS, Ahn M, Kim J, Shin T. Glycoconjugate-Specific Developmental Changes in the Horse Vomeronasal Organ. Cells Tissues Organs 2023; 213:147-160. [PMID: 36599327 DOI: 10.1159/000528883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 12/22/2022] [Indexed: 01/05/2023] Open
Abstract
The vomeronasal organ (VNO) is a tubular pheromone-sensing organ in which the lumen is covered with sensory and non-sensory epithelia. This study used immunohistochemistry and lectin histochemistry techniques to evaluate developmental changes, specifically of the glycoconjugate profile, in the horse VNO epithelium. Immunostaining analysis revealed PGP9.5 expression in some vomeronasal non-sensory epithelium (VNSE) cells and in the vomeronasal receptor cells of the vomeronasal sensory epithelium (VSE) in fetuses, young foals, and adult horses. Olfactory marker protein expression was exclusively localized in receptor cells of the VSE in fetuses, young foals, and adult horses and absent in VNSE. To identify the glycoconjugate type, lectin histochemistry was performed using 21 lectins. Semi-quantitative analysis revealed that the intensities of glycoconjugates labeled with WGA, DSL, LEL, and RCA120 were significantly higher in adult horse VSE than those in foal VSE, whereas the intensities of glycoconjugates labeled with LCA and PSA were significantly lower in adult horse VSE. The intensities of glycoconjugates labeled with s-WGA, WGA, BSL-II, DSL, LEL, STL, ConA, LCA, PSA, DBA, SBA, SJA, RCA120, jacalin, and ECL were significantly higher in adult horse VNSE than those in foal VNSE, whereas the intensity of glycoconjugates labeled with UEA-I was lower in adult horse VNSE. Histochemical analysis of each lectin revealed that various glycoconjugates in the VSE were present in the receptor, supporting, and basal cells of foals and adult horses. A similar pattern of lectin histochemistry was also observed in the VNSE of foals and adult horses. In conclusion, these results suggest that there is an increase in the level of N-acetylglucosamine (labeled by WGA, DSL, LEL) and galactose (labeled by RCA120) in horse VSE during postnatal development, implying that they may influence the function of VNO in adult horses.
Collapse
Affiliation(s)
- Jiyoon Chun
- Department of Veterinary Anatomy, College of Veterinary Medicine, Veterinary Medical Research Institute, Jeju National University, Jeju, Republic of Korea
| | - Taeyoung Kang
- Department of Veterinary Obstetrics, College of Veterinary Medicine, Veterinary Medical Research Institute, Jeju National University, Jeju, Republic of Korea
| | - Jong-Pil Seo
- Department of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, Republic of Korea
| | - Hyohoon Jeong
- Department of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, Republic of Korea
| | - Minhan Kim
- Veterinary Research Institute, Jeju Special Self-Governing Province, Jeju, Republic of Korea
| | - Byung Sun Kim
- Department of Equine Science, Cheju Halla University, Jeju, Republic of Korea
| | - Meejung Ahn
- Department of Animal Science, College of Life Science, Sangji University, Wonju, Republic of Korea
| | - Jeongtae Kim
- Department of Anatomy, Kosin University College of Medicine, Busan, Republic of Korea
| | - Taekyun Shin
- Department of Veterinary Anatomy, College of Veterinary Medicine, Veterinary Medical Research Institute, Jeju National University, Jeju, Republic of Korea
| |
Collapse
|
22
|
Duan C, Hou L, Deng X, Wu J, Qian W, Han C, Hou X. Fucose ameliorates the proinflammatory property of Fusobacterium nucleatum in colitis via altering its metabolism. Front Cell Infect Microbiol 2023; 13:1190602. [PMID: 37197204 PMCID: PMC10183584 DOI: 10.3389/fcimb.2023.1190602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 04/19/2023] [Indexed: 05/19/2023] Open
Abstract
Introduction Previous studies reported that fucose plays a protective role in inhibiting pathogens. Fusobacterium nucleatum (Fn) was recently found to promote the progression of colitis. However, the effects of fucose on Fn are poorly understood. This study aimed to explore whether fucose could ameliorate the proinflammatory property of Fn in colitis and the underlying mechanisms. Methods To validate our hypothesis, mice were administrated with Fn and fucose-treated Fn (Fnf) before dextran sulfate sodium (DSS) treatment to establish Fn related colitis model. The metabolism variation of Fn was detected by metabolomic analysis. To verify the effects of bacterial metabolites on intestinal epithelial cells (IECs), Caco-2 cells were treated with bacterial supernatant. Results More severe inflammation, intestinal barrier damage, autophagy block, and apoptosis in the colon were noted in DSS mice that were administrated with Fn or Fnf. However, the severity degree in Fnf+DSS group was less compared to Fn+DSS group. Metabolic pathways of Fn were altered after fucose treatment and proinflammatory metabolites were decreased. The supernatant of Fnf induced a lower level of inflammation than Fn in Caco-2 cells. One of the decreased metabolites, homocysteine thiolactone (HT), was proven to induce inflammatory effects in Caco-2 cells. Discussion In conclusion, fucose ameliorates the proinflammatory property of Fn via altering its metabolism and these findings provide evidence for the application of fucose as functional food or prebiotic in the treatment of Fn related colitis.
Collapse
Affiliation(s)
- Caihan Duan
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingzhi Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohua Deng
- Hubei Center of Industrial Culture Collection and Research, Wuhan, China
| | - Junhao Wu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Qian
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaoqun Han
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Chaoqun Han, ; Xiaohua Hou,
| | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Chaoqun Han, ; Xiaohua Hou,
| |
Collapse
|
23
|
Roager HM, Stanton C, Hall LJ. Microbial metabolites as modulators of the infant gut microbiome and host-microbial interactions in early life. Gut Microbes 2023; 15:2192151. [PMID: 36942883 PMCID: PMC10038037 DOI: 10.1080/19490976.2023.2192151] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 03/06/2023] [Indexed: 03/23/2023] Open
Abstract
The development of infant gut microbiome is a pivotal process affecting the ecology and function of the microbiome, as well as host health. While the establishment of the infant microbiome has been of interest for decades, the focus on gut microbial metabolism and the resulting small molecules (metabolites) has been rather limited. However, technological and computational advances are now enabling researchers to profile the plethora of metabolites in the infant gut, allowing for improved understanding of how gut microbial-derived metabolites drive microbiome community structuring and host-microbial interactions. Here, we review the current knowledge on development of the infant gut microbiota and metabolism within the first year of life, and discuss how these microbial metabolites are key for enhancing our basic understanding of interactions during the early life developmental window.
Collapse
Affiliation(s)
- Henrik M. Roager
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg, Denmark
| | - Catherine Stanton
- APC Microbiome Ireland, Teagasc Moorepark Food Research Centre, Fermoy, Co. Cork, Ireland
| | - Lindsay J. Hall
- Gut Microbes & Health, Quadram Institute Biosciences, Norwich, UK
- Intestinal Microbiome, School of Life Sciences, ZIEL – Institute for Food & Health, Technical University of Munich, Freising, Germany
- Norwich Medical School, University of East Anglia, Norwich, UK
| |
Collapse
|
24
|
Godefroy E, Barbé L, Le Moullac-Vaidye B, Rocher J, Breiman A, Leuillet S, Mariat D, Chatel JM, Ruvoën-Clouet N, Carton T, Jotereau F, Le Pendu J. Microbiota-induced regulatory T cells associate with FUT2-dependent susceptibility to rotavirus gastroenteritis. Front Microbiol 2023; 14:1123803. [PMID: 36922975 PMCID: PMC10008897 DOI: 10.3389/fmicb.2023.1123803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/03/2023] [Indexed: 03/03/2023] Open
Abstract
The FUT2 α1,2fucosyltransferase contributes to the synthesis of fucosylated glycans used as attachment factors by several pathogens, including noroviruses and rotaviruses, that can induce life-threatening gastroenteritis in young children. FUT2 genetic polymorphisms impairing fucosylation are strongly associated with resistance to dominant strains of both noroviruses and rotaviruses. Interestingly, the wild-type allele associated with viral gastroenteritis susceptibility inversely appears to be protective against several inflammatory or autoimmune diseases for yet unclear reasons, although a FUT2 influence on microbiota composition has been observed. Here, we studied a cohort of young healthy adults and showed that the wild-type FUT2 allele was associated with the presence of anti-RVA antibodies, either neutralizing antibodies or serum IgA, confirming its association with the risk of RVA gastroenteritis. Strikingly, it was also associated with the frequency of gut microbiota-induced regulatory T cells (Tregs), so-called DP8α Tregs, albeit only in individuals who had anti-RVA neutralizing antibodies or high titers of anti-RVA IgAs. DP8α Tregs specifically recognize the human symbiont Faecalibacterium prausnitzii, which strongly supports their induction by this anti-inflammatory bacterium. The proportion of F. prausnitzii in feces was also associated with the FUT2 wild-type allele. These observations link the FUT2 genotype with the risk of RVA gastroenteritis, the microbiota and microbiota-induced DP8α Treg cells, suggesting that the anti-RVA immune response might involve an induction/expansion of these T lymphocytes later providing a balanced immunological state that confers protection against inflammatory diseases.
Collapse
Affiliation(s)
- Emmanuelle Godefroy
- Inserm, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1303/EMR6001, Nantes Université, Nantes, France
| | - Laure Barbé
- Inserm, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1303/EMR6001, Nantes Université, Nantes, France
| | - Béatrice Le Moullac-Vaidye
- Inserm, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1303/EMR6001, Nantes Université, Nantes, France
| | - Jézabel Rocher
- Inserm, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1303/EMR6001, Nantes Université, Nantes, France
| | - Adrien Breiman
- Inserm, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1303/EMR6001, Nantes Université, Nantes, France.,CHU de Nantes, Nantes, France
| | | | - Denis Mariat
- INRAE, AgroParisTech, UMR1319, MICALIS, Université Paris Saclay, Jouy en Josas, France
| | - Jean-Marc Chatel
- INRAE, AgroParisTech, UMR1319, MICALIS, Université Paris Saclay, Jouy en Josas, France
| | - Nathalie Ruvoën-Clouet
- Inserm, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1303/EMR6001, Nantes Université, Nantes, France.,ONIRIS, Ecole Nationale Vétérinaire, Agroalimentaire et de l'Alimentation, Nantes, France
| | | | - Francine Jotereau
- Inserm, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1303/EMR6001, Nantes Université, Nantes, France
| | - Jacques Le Pendu
- Inserm, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1303/EMR6001, Nantes Université, Nantes, France
| |
Collapse
|
25
|
Moya-Gonzálvez EM, Peña-Gil N, Rubio-del-Campo A, Coll-Marqués JM, Gozalbo-Rovira R, Monedero V, Rodríguez-Díaz J, Yebra MJ. Infant Gut Microbial Metagenome Mining of α-l-Fucosidases with Activity on Fucosylated Human Milk Oligosaccharides and Glycoconjugates. Microbiol Spectr 2022; 10:e0177522. [PMID: 35943155 PMCID: PMC9430343 DOI: 10.1128/spectrum.01775-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/01/2022] [Indexed: 11/21/2022] Open
Abstract
The gastrointestinal microbiota members produce α-l-fucosidases that play key roles in mucosal, human milk, and dietary oligosaccharide assimilation. Here, 36 open reading frames (ORFs) coding for putative α-l-fucosidases belonging to glycosyl hydrolase family 29 (GH29) were identified through metagenome analysis of breast-fed infant fecal microbiome. Twenty-two of those ORFs showed a complete coding sequence with deduced amino acid sequences displaying the highest degree of identity with α-l-fucosidases from Bacteroides thetaiotaomicron, Bacteroides caccae, Phocaeicola vulgatus, Phocaeicola dorei, Ruminococcus gnavus, and Streptococcus parasanguinis. Based on sequence homology, 10 α-l-fucosidase genes were selected for substrate specificity characterization. The α-l-fucosidases Fuc18, Fuc19A, Fuc35B, Fuc39, and Fuc1584 showed hydrolytic activity on α1,3/4-linked fucose present in Lewis blood antigens and the human milk oligosaccharide (HMO) 3-fucosyllactose. In addition, Fuc1584 also hydrolyzed fucosyl-α-1,6-N-acetylglucosamine (6FN), a component of the core fucosylation of N-glycans. Fuc35A and Fuc193 showed activity on α1,2/3/4/6 linkages from H type-2, Lewis blood antigens, HMOs and 6FN. Fuc30 displayed activity only on α1,6-linked l-fucose, and Fuc5372 showed a preference for α1,2 linkages. Fuc2358 exhibited a broad substrate specificity releasing l-fucose from all the tested free histo-blood group antigens, HMOs, and 6FN. This latest enzyme also displayed activity in glycoconjugates carrying lacto-N-fucopentaose II (Lea) and lacto-N-fucopentaose III (Lex) and in the glycoprotein mucin. Fuc18, Fuc19A, and Fuc39 also removed l-fucose from neoglycoproteins and human α-1 acid glycoprotein. These results give insight into the great diversity of α-l-fucosidases from the infant gut microbiota, thus supporting the hypothesis that fucosylated glycans are crucial for shaping the newborn microbiota composition. IMPORTANCE α-l-Fucosyl residues are frequently present in many relevant glycans, such as human milk oligosaccharides (HMOs), histo-blood group antigens (HBGAs), and epitopes on cell surface glycoconjugate receptors. These fucosylated glycans are involved in a number of mammalian physiological processes, including adhesion of pathogens and immune responses. The modulation of l-fucose content in such processes may provide new insights and knowledge regarding molecular interactions and may help to devise new therapeutic strategies. Microbial α-l-fucosidases are exoglycosidases that remove α-l-fucosyl residues from free oligosaccharides and glycoconjugates and can be also used in transglycosylation reactions to synthesize oligosaccharides. In this work, α-l-fucosidases from the GH29 family were identified and characterized from the metagenome of fecal samples of breastfed infants. These enzymes showed different substrate specificities toward HMOs, HBGAs, naturally occurring glycoproteins, and neoglycoproteins. These novel glycosidase enzymes from the breast-fed infant gut microbiota, which resulted in a good source of α-l-fucosidases, have great biotechnological potential.
Collapse
Affiliation(s)
- Eva M. Moya-Gonzálvez
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Valencia, Spain
| | - Nazaret Peña-Gil
- Departamento de Microbiología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
- INCLIVA, Instituto de Investigación Sanitaría del Hospital Clínico de Valencia, Valencia, Spain
| | - Antonio Rubio-del-Campo
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Valencia, Spain
| | - José M. Coll-Marqués
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Valencia, Spain
| | - Roberto Gozalbo-Rovira
- Departamento de Microbiología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
- INCLIVA, Instituto de Investigación Sanitaría del Hospital Clínico de Valencia, Valencia, Spain
| | - Vicente Monedero
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Valencia, Spain
| | - Jesús Rodríguez-Díaz
- Departamento de Microbiología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
- INCLIVA, Instituto de Investigación Sanitaría del Hospital Clínico de Valencia, Valencia, Spain
| | - María J. Yebra
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Valencia, Spain
| |
Collapse
|
26
|
In Silico Study of Cell Surface Structures of Parabacteroides distasonis Involved in Its Maintenance within the Gut Microbiota. Int J Mol Sci 2022; 23:ijms23169411. [PMID: 36012685 PMCID: PMC9409006 DOI: 10.3390/ijms23169411] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
The health-promoting Parabacteroides distasonis, which is part of the core microbiome, has recently received a lot of attention, showing beneficial properties for its host and potential as a new biotherapeutic product. However, no study has yet investigated the cell surface molecules and structures of P. distasonis that allow its maintenance within the gut microbiota. Moreover, although P. distasonis is strongly recognized as an intestinal commensal species with benefits for its host, several works displayed controversial results, showing it as an opportunistic pathogen. In this study, we reported gene clusters potentially involved in the synthesis of capsule, fimbriae-like and pili-like cell surface structures in 26 P. distasonis genomes and applied the new RfbA-typing classification in order to better understand and characterize the beneficial/pathogenic behavior related to P. distasonis strains. Two different types of fimbriae, three different types of pilus and up to fourteen capsular polysaccharide loci were identified over the 26 genomes studied. Moreover, the addition of data to the rfbA-type classification modified the outcome by rearranging rfbA genes and adding a fifth group to the classification. In conclusion, the strain variability in terms of external proteinaceous structure could explain the inter-strain differences previously observed of P. distasonis adhesion capacities and its potential pathogenicity, but no specific structure related to P. distasonis beneficial or detrimental activity was identified.
Collapse
|
27
|
Almahayni K, Spiekermann M, Fiore A, Yu G, Pedram K, Möckl L. Small molecule inhibitors of mammalian glycosylation. Matrix Biol Plus 2022; 16:100108. [PMID: 36467541 PMCID: PMC9713294 DOI: 10.1016/j.mbplus.2022.100108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/10/2022] [Accepted: 03/10/2022] [Indexed: 01/06/2023] Open
Abstract
Glycans are one of the fundamental biopolymers encountered in living systems. Compared to polynucleotide and polypeptide biosynthesis, polysaccharide biosynthesis is a uniquely combinatorial process to which interdependent enzymes with seemingly broad specificities contribute. The resulting intracellular cell surface, and secreted glycans play key roles in health and disease, from embryogenesis to cancer progression. The study and modulation of glycans in cell and organismal biology is aided by small molecule inhibitors of the enzymes involved in glycan biosynthesis. In this review, we survey the arsenal of currently available inhibitors, focusing on agents which have been independently validated in diverse systems. We highlight the utility of these inhibitors and drawbacks to their use, emphasizing the need for innovation for basic research as well as for therapeutic applications.
Collapse
Affiliation(s)
- Karim Almahayni
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany
| | - Malte Spiekermann
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany
| | - Antonio Fiore
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Guoqiang Yu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Kayvon Pedram
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA,Corresponding authors.
| | - Leonhard Möckl
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany,Corresponding authors.
| |
Collapse
|
28
|
Tian X, Jiang H, Cai B, Feng H, Wang X, Yu G. Comparative Proteomic Analysis of Fucosylated Glycoproteins Produced by Bacteroides thetaiotaomicron Under Different Polysaccharide Nutrition Conditions. Front Microbiol 2022; 13:826942. [PMID: 35308349 PMCID: PMC8931616 DOI: 10.3389/fmicb.2022.826942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Bacteroides thetaiotaomicron, one of the most eminent representative gut commensal Bacteroides species, is able to use the L-fucose in host-derived and dietary polysaccharides to modify its capsular polysaccharides and glycoproteins through a mammalian-like salvage metabolic pathway. This process is essential for the colonization of the bacteria and for symbiosis with the host. However, despite the importance of fucosylated proteins (FGPs) in B. thetaiotaomicron, their types, distribution, and functions remain unclear. In this study, the effects of different polysaccharide (corn starch, mucin, and fucoidan) nutrition conditions on newly synthesized FGPs expressions and fucosylation are investigated using a chemical biological method based on metabolic labeling and bioorthogonal reaction. According to the results of label-free quantification, 559 FGPs (205 downregulated and 354 upregulated) are affected by the dietary conditions. Of these differentially expressed proteins, 65 proteins show extremely sensitive to polysaccharide nutrition conditions (FGPs fold change/global protein fold change ≥2.0 or ≤0.5). Specifically, the fucosylation of the chondroitin sulfate ABC enzyme, Sus proteins, and cationic efflux system proteins varies significantly upon the addition of mucin, corn starch, or fucoidan. Moreover, these polysaccharides can trigger an appreciable increase in the fucosylation level of the two-component system and ammonium transport proteins. These results highlight the efficiency of the combined metabolic glycan labeling and bio-orthogonal reaction in enriching the intestinal Bacteroides glycoproteins. Moreover, it emphasizes the sensitivity of Bacteroides fucosylation to polysaccharide nutrition conditions, which allows for the regulation of bacterial growth.
Collapse
Affiliation(s)
- Xiao Tian
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Hao Jiang
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Binbin Cai
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Huxin Feng
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Xuan Wang
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Guangli Yu
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
29
|
The Serum and Fecal Metabolomic Profiles of Growing Kittens Treated with Amoxicillin/Clavulanic Acid or Doxycycline. Animals (Basel) 2022; 12:ani12030330. [PMID: 35158655 PMCID: PMC8833518 DOI: 10.3390/ani12030330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary This study investigated the impact of antibiotic treatment οn the serum and fecal metabolome (the collection of all small molecules produced by the gut bacteria and the host) of young cats. Thirty 2-month-old cats with an upper respiratory tract infection were treated with either amoxicillin/clavulanic acid for 20 days or doxycycline for 28 days. In addition, another 15 control cats that did not receive antibiotics were included. Blood was collected on days 0 (before treatment), 20/28 (last day of treatment), and 300 (10 months after the end of treatment), while feces were collected on days 0, 20/28, 60, 120, and 300. Seven serum and fecal metabolites differed between cats treated with antibiotics and control cats at the end of treatment period. Ten months after treatment, no metabolites differed from healthy cats, suggesting that amoxicillin/clavulanic acid or doxycycline treatment only temporarily affects the abundance of the serum and fecal metabolome. Abstract The long-term impact of antibiotics on the serum and fecal metabolome of kittens has not yet been investigated. Therefore, the objective of this study was to evaluate the serum and fecal metabolome of kittens with an upper respiratory tract infection (URTI) before, during, and after antibiotic treatment and compare it with that of healthy control cats. Thirty 2-month-old cats with a URTI were randomly assigned to receive either amoxicillin/clavulanic acid for 20 days or doxycycline for 28 days, and 15 cats of similar age were enrolled as controls. Fecal samples were collected on days 0, 20/28, 60, 120, and 300, while serum was collected on days 0, 20/28, and 300. Untargeted and targeted metabolomic analyses were performed on both serum and fecal samples. Seven metabolites differed significantly in antibiotic-treated cats compared to controls on day 20/28, with two differing on day 60, and two on day 120. Alterations in the pattern of serum amino acids, antioxidants, purines, and pyrimidines, as well as fecal bile acids, sterols, and fatty acids, were observed in antibiotic-treated groups that were not observed in control cats. However, the alterations caused by either amoxicillin/clavulanic acid or doxycycline of the fecal and serum metabolome were only temporary and were resolved by 10 months after their withdrawal.
Collapse
|
30
|
Alteration of the Intestinal Permeability Are Reflected by Changes in the Urine Metabolome of Young Autistic Children: Preliminary Results. Metabolites 2022; 12:metabo12020104. [PMID: 35208179 PMCID: PMC8875518 DOI: 10.3390/metabo12020104] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 12/11/2022] Open
Abstract
Several metabolomics-based studies have provided evidence that autistic subjects might share metabolic abnormalities with gut microbiota dysbiosis and alterations in gut mucosal permeability. Our aims were to explore the most relevant metabolic perturbations in a group of autistic children, compared with their healthy siblings, and to investigate whether the increased intestinal permeability may be mirrored by specific metabolic perturbations. We enrolled 13 autistic children and 14 unaffected siblings aged 2–12 years; the evaluation of the intestinal permeability was estimated by the lactulose:mannitol test. The urine metabolome was investigated by proton nuclear magnetic resonance (1H-NMR) spectroscopy. The lactulose:mannitol test unveiled two autistic children with altered intestinal permeability. Nine metabolites significantly discriminated the urine metabolome of autistic children from that of their unaffected siblings; however, in the autistic children with increased permeability, four additional metabolites—namely, fucose, phenylacetylglycine, nicotinurate, and 1-methyl-nicotinamide, strongly discriminated their urine metabolome from that of the remaining autistic children. Our preliminary data suggest the presence of a specific urine metabolic profile associated with the increase in intestinal permeability.
Collapse
|
31
|
The Fast Track for Intestinal Tumor Cell Differentiation and In Vitro Intestinal Models by Inorganic Topographic Surfaces. Pharmaceutics 2022; 14:pharmaceutics14010218. [PMID: 35057113 PMCID: PMC8781367 DOI: 10.3390/pharmaceutics14010218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 11/17/2022] Open
Abstract
Three-dimensional (3D) complex in vitro cell systems are well suited to providing meaningful and translatable results in drug screening, toxicity measurements, and biological studies. Reliable complex gastrointestinal in vitro models as a testbed for oral drug administration and toxicity are very valuable in achieving predictive results for clinical trials and reducing animal testing. However, producing these models is time-consuming due to the lengthy differentiation of HT29 or other cells into mucus-producing goblet cells or other intestinal cell lineages. In the present work, HT29 cells were grown on an inorganic topographic surface decorated with a periodic pattern of micrometre-sized amorphous SiO2 structures for up to 35 days. HT29 cells on topographic surfaces were compared to undifferentiated HT29 in glucose-containing medium on glass or culture dish and with HT29 cells differentiated for 30 days in the presence of methotrexate (HT29-MTX). The cells were stained with Alcian blue for mucus, antibodies for mucus 2 (goblet cells), villin (enterocytes), lysozyme (Paneth cells), and FITC-labeled lectins to identify different cells, glycomic profiles, and cell features. We observed that HT29 cells on topographic surfaces showed more similarities with the differentiated HT29-MTX than with undifferentiated HT29. They formed islands of cell clusters, as observed for HT29-MTX. Already after 2 days, the first mucus secretion was shown by Alcian blue stain and FITC-wheat germ agglutinin. After 4–6 days, mucus was observed on the cell surface and in the intercellular space. The cell layer was undulated, and in 3D reconstruction, the cells showed a clear polarisation with a strong actin signal to one membrane. The lectins and the antibody-staining confirmed the heterogeneous composition of differentiated HT29 cells on topographic surfaces after 6–8 days, or after 6–8 days following MTX differentiation (30 days).
Collapse
|
32
|
Zhao F, Xie S, Li B, Zhang X. Functional nucleic acids in glycobiology: A versatile tool in the analysis of disease-related carbohydrates and glycoconjugates. Int J Biol Macromol 2022; 201:592-606. [PMID: 35031315 DOI: 10.1016/j.ijbiomac.2022.01.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/05/2022] [Accepted: 01/07/2022] [Indexed: 12/12/2022]
Abstract
As significant components of the organism, carbohydrates and glycoconjugates play indispensable roles in energy supply, cell signaling, immune modulation, and tumor cell invasion, and function as biomarkers since aberrance of them has been proved to be associated with the emergence and development of certain diseases. Functional nucleic acids (FNAs) have properties including easy-to-synthesize, good stability, good biocompatibility, low cost, and high programmability, they have attracted significant research attention and been incorporated into biosensors for detecting disease-related carbohydrates and glycoconjugates. This review summarizes the construction strategies and biosensing applications of FNAs-based biosensors in glycobiology in terms of target recognition and signal transduction. By illustrating the mechanisms and comparing the performances, the challenges and development opportunities in this area have been critically elaborated. We believe that this review will provide a better understanding of the role of FNAs in the analysis of disease-related carbohydrates and glycoconjugates, and inspire further discovery in fields that include glycobiology, chemical biology, clinical diagnosis, and drug development.
Collapse
Affiliation(s)
- Furong Zhao
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Siying Xie
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Bingzhi Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China.
| | - Xing Zhang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
33
|
Tomek MB, Janesch B, Braun ML, Taschner M, Figl R, Grünwald-Gruber C, Coyne MJ, Blaukopf M, Altmann F, Kosma P, Kählig H, Comstock LE, Schäffer C. A Combination of Structural, Genetic, Phenotypic and Enzymatic Analyses Reveals the Importance of a Predicted Fucosyltransferase to Protein O-Glycosylation in the Bacteroidetes. Biomolecules 2021; 11:1795. [PMID: 34944439 PMCID: PMC8698959 DOI: 10.3390/biom11121795] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/21/2021] [Accepted: 11/23/2021] [Indexed: 12/20/2022] Open
Abstract
Diverse members of the Bacteroidetes phylum have general protein O-glycosylation systems that are essential for processes such as host colonization and pathogenesis. Here, we analyzed the function of a putative fucosyltransferase (FucT) family that is widely encoded in Bacteroidetes protein O-glycosylation genetic loci. We studied the FucT orthologs of three Bacteroidetes species-Tannerella forsythia, Bacteroides fragilis, and Pedobacter heparinus. To identify the linkage created by the FucT of B. fragilis, we elucidated the full structure of its nine-sugar O-glycan and found that l-fucose is linked β1,4 to glucose. Of the two fucose residues in the T. forsythia O-glycan, the fucose linked to the reducing-end galactose was shown by mutational analysis to be l-fucose. Despite the transfer of l-fucose to distinct hexose sugars in the B. fragilis and T. forsythia O-glycans, the FucT orthologs from B. fragilis, T. forsythia, and P. heparinus each cross-complement the B. fragilis ΔBF4306 and T. forsythia ΔTanf_01305 FucT mutants. In vitro enzymatic analyses showed relaxed acceptor specificity of the three enzymes, transferring l-fucose to various pNP-α-hexoses. Further, glycan structural analysis together with fucosidase assays indicated that the T. forsythia FucT links l-fucose α1,6 to galactose. Given the biological importance of fucosylated carbohydrates, these FucTs are promising candidates for synthetic glycobiology.
Collapse
Affiliation(s)
- Markus B. Tomek
- NanoGlycobiology Unit, Institute of Biologically Inspired Materials, Department of NanoBiotechnology, Universität für Bodenkultur Wien, Muthgasse 11, A-1190 Vienna, Austria; (M.B.T.); (B.J.); (M.L.B.); (M.T.)
| | - Bettina Janesch
- NanoGlycobiology Unit, Institute of Biologically Inspired Materials, Department of NanoBiotechnology, Universität für Bodenkultur Wien, Muthgasse 11, A-1190 Vienna, Austria; (M.B.T.); (B.J.); (M.L.B.); (M.T.)
| | - Matthias L. Braun
- NanoGlycobiology Unit, Institute of Biologically Inspired Materials, Department of NanoBiotechnology, Universität für Bodenkultur Wien, Muthgasse 11, A-1190 Vienna, Austria; (M.B.T.); (B.J.); (M.L.B.); (M.T.)
| | - Manfred Taschner
- NanoGlycobiology Unit, Institute of Biologically Inspired Materials, Department of NanoBiotechnology, Universität für Bodenkultur Wien, Muthgasse 11, A-1190 Vienna, Austria; (M.B.T.); (B.J.); (M.L.B.); (M.T.)
| | - Rudolf Figl
- Institute of Biochemistry, Department of Chemistry, Universität für Bodenkultur Wien, Muthgasse 18, A-1190 Vienna, Austria; (R.F.); (C.G.-G.); (F.A.)
| | - Clemens Grünwald-Gruber
- Institute of Biochemistry, Department of Chemistry, Universität für Bodenkultur Wien, Muthgasse 18, A-1190 Vienna, Austria; (R.F.); (C.G.-G.); (F.A.)
| | - Michael J. Coyne
- Department of Microbiology and the Duchossois Family Institute, University of Chicago, KCBD, 900 E. 57th Street, Chicago, IL 60637, USA; (M.J.C.); (L.E.C.)
| | - Markus Blaukopf
- Institute of Organic Chemistry, Department of Chemistry, Universität für Bodenkultur Wien, Muthgasse 18, A-1190 Vienna, Austria; (M.B.); (P.K.)
| | - Friedrich Altmann
- Institute of Biochemistry, Department of Chemistry, Universität für Bodenkultur Wien, Muthgasse 18, A-1190 Vienna, Austria; (R.F.); (C.G.-G.); (F.A.)
| | - Paul Kosma
- Institute of Organic Chemistry, Department of Chemistry, Universität für Bodenkultur Wien, Muthgasse 18, A-1190 Vienna, Austria; (M.B.); (P.K.)
| | - Hanspeter Kählig
- Department of Organic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Strasse 38, A-1090 Vienna, Austria;
| | - Laurie E. Comstock
- Department of Microbiology and the Duchossois Family Institute, University of Chicago, KCBD, 900 E. 57th Street, Chicago, IL 60637, USA; (M.J.C.); (L.E.C.)
| | - Christina Schäffer
- NanoGlycobiology Unit, Institute of Biologically Inspired Materials, Department of NanoBiotechnology, Universität für Bodenkultur Wien, Muthgasse 11, A-1190 Vienna, Austria; (M.B.T.); (B.J.); (M.L.B.); (M.T.)
| |
Collapse
|
34
|
Achasova KM, Kozhevnikova EN, Borisova MA, Litvinova EA. Fucose Ameliorates Tritrichomonas sp.-Associated Illness in Antibiotic-Treated Muc2-/- Mice. Int J Mol Sci 2021; 22:ijms221910699. [PMID: 34639039 PMCID: PMC8509520 DOI: 10.3390/ijms221910699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
The mucus layer in the intestine plays a critical role in regulation of host–microbe interactions and maintaining homeostasis. Disruptions of the mucus layer due to genetic, environmental, or immune factors may lead to inflammatory bowel diseases (IBD). IBD frequently are accompanied with infections, and therefore are treated with antibiotics. Hence, it is important to evaluate risks of antibiotic treatment in individuals with vulnerable gut barrier and chronic inflammation. Mice with a knockout of the Muc2 gene, encoding the main glycoprotein component of the mucus, demonstrate a close contact of the microbes with the gut epithelium which leads to chronic inflammation resembling IBD. Here we demonstrate that the Muc2−/− mice harboring a gut protozoan infection Tritrichomonas sp. are susceptible to an antibiotic-induced depletion of the bacterial microbiota. Suppression of the protozoan infection with efficient metronidazole dosage or L-fucose administration resulted in amelioration of an illness observed in antibiotic-treated Muc2−/− mice. Fucose is a monosaccharide presented abundantly in gut glycoproteins, including Mucin2, and is known to be involved in host–microbe interactions, in particular in microbe adhesion. We suppose that further investigation of the role of fucose in protozoan adhesion to host cells may be of great value.
Collapse
Affiliation(s)
- Kseniya M. Achasova
- Siberian Federal Scientific Centre of Agro-BioTechnologies of the Russian Academy of Sciences, 630501 Krasnoobsk, Russia; (K.M.A.); (E.N.K.)
- Scientific Research Institute of Neurosciences and Medicine, 630117 Novosibirsk, Russia
- Institute of Molecular and Cellular Biology, The Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Elena N. Kozhevnikova
- Siberian Federal Scientific Centre of Agro-BioTechnologies of the Russian Academy of Sciences, 630501 Krasnoobsk, Russia; (K.M.A.); (E.N.K.)
- Scientific Research Institute of Neurosciences and Medicine, 630117 Novosibirsk, Russia
- Institute of Molecular and Cellular Biology, The Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
- The Laboratory of Biotechnology, Novosibirsk State Agrarian University, 630039 Novosibirsk, Russia
| | - Mariya A. Borisova
- Federal Research Center Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia;
| | - Ekaterina A. Litvinova
- Siberian Federal Scientific Centre of Agro-BioTechnologies of the Russian Academy of Sciences, 630501 Krasnoobsk, Russia; (K.M.A.); (E.N.K.)
- Scientific Research Institute of Neurosciences and Medicine, 630117 Novosibirsk, Russia
- Correspondence: ; Tel.: +7-923-147-94-64
| |
Collapse
|
35
|
Szymanski CM, Koropatkin NM. Microbial transformation of the host glycobiome. Glycobiology 2021; 31:664-666. [PMID: 34213552 PMCID: PMC8252863 DOI: 10.1093/glycob/cwab045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 12/27/2022] Open
Affiliation(s)
- Christine M Szymanski
- Complex Carbohydrate Research Center and Department of Microbiology, University of Georgia, Athens, GA, USA
| | - Nicole M Koropatkin
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|