1
|
Muramatsu MK, Winter SE. Nutrient acquisition strategies by gut microbes. Cell Host Microbe 2024; 32:863-874. [PMID: 38870902 PMCID: PMC11178278 DOI: 10.1016/j.chom.2024.05.011] [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: 03/25/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 06/15/2024]
Abstract
The composition and function of the gut microbiota are intimately tied to nutrient acquisition strategies and metabolism, with significant implications for host health. Both dietary and host-intrinsic factors influence community structure and the basic modes of bacterial energy metabolism. The intestinal tract is rich in carbon and nitrogen sources; however, limited access to oxygen restricts energy-generating reactions to fermentation. By contrast, increased availability of electron acceptors during episodes of intestinal inflammation results in phylum-level changes in gut microbiota composition, suggesting that bacterial energy metabolism is a key driver of gut microbiota function. In this review article, we will illustrate diverse examples of microbial nutrient acquisition strategies in the context of habitat filters and anatomical location and the central role of energy metabolism in shaping metabolic strategies to support bacterial growth in the mammalian gut.
Collapse
Affiliation(s)
- Matthew K Muramatsu
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis, Davis, CA 95616, USA
| | - Sebastian E Winter
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis, Davis, CA 95616, USA.
| |
Collapse
|
2
|
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] [Grants] [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
|
3
|
Lei C, Sun R, Xu G, Tan Y, Feng W, McClain CJ, Deng Z. Enteric VIP-producing neurons maintain gut microbiota homeostasis through regulating epithelium fucosylation. Cell Host Microbe 2022; 30:1417-1434.e8. [PMID: 36150396 PMCID: PMC9588764 DOI: 10.1016/j.chom.2022.09.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/18/2022] [Accepted: 08/31/2022] [Indexed: 12/12/2022]
Abstract
Interactions between the enteric nervous system (ENS) and intestinal epithelium are thought to play a vital role in intestinal homeostasis. How the ENS monitors the frontier with commensal and pathogenic microbes while maintaining epithelial function remains unclear. Here, by combining subdiaphragmatic vagotomy with transcriptomics, chemogenetic strategy, and coculture of enteric neuron-intestinal organoid, we show that enteric neurons expressing VIP shape the α1,2-fucosylation of intestinal epithelial cells (IECs). Mechanistically, neuropeptide VIP activates fut2 expression via the Erk1/2-c-Fos pathway through the VIPR1 receptor on IECs. We further demonstrate that perturbation of enteric neurons leads to gut dysbiosis through α1,2-fucosylation in the steady state and results in increased susceptibility to alcohol-associated liver disease (ALD). This was attributed to an imbalance between beneficial Bifidobacterium and opportunistic pathogenic Enterococcus faecalis in ALD. In addition, Bifidobacterium α1,2-fucosidase may promote Bifidobacterium adhesion to the mucosal surface, which restricts Enterococcus faecalis overgrowth and prevents ALD progression.
Collapse
Affiliation(s)
- Chao Lei
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, USA; Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Rui Sun
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, USA; Brown Cancer Center, University of Louisville, Louisville, KY, USA; Central Laboratory and Department of Oncology, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430033, China
| | - Guangzhong Xu
- Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Yi Tan
- Department of Pediatrics, University of Louisville, Louisville, KY, USA
| | - Wenke Feng
- Department of Medicine, University of Louisville, Louisville, KY, USA; Alcohol Research Center, University of Louisville, Louisville, KY, USA; Hepatobiology & Toxicology Center, University of Louisville, Louisville, KY, USA
| | - Craig J McClain
- Department of Medicine, University of Louisville, Louisville, KY, USA; Alcohol Research Center, University of Louisville, Louisville, KY, USA; Hepatobiology & Toxicology Center, University of Louisville, Louisville, KY, USA; Robley Rex VA Medical Center, Louisville, KY, USA
| | - Zhongbin Deng
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, USA; Brown Cancer Center, University of Louisville, Louisville, KY, USA; Alcohol Research Center, University of Louisville, Louisville, KY, USA; Hepatobiology & Toxicology Center, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
4
|
Tsang DK, Wang RJ, De Sa O, Ayyaz A, Foerster EG, Bayer G, Goyal S, Trcka D, Ghoshal B, Wrana JL, Girardin SE, Philpott DJ. A single cell survey of the microbial impacts on the mouse small intestinal epithelium. Gut Microbes 2022; 14:2108281. [PMID: 35939622 PMCID: PMC9361762 DOI: 10.1080/19490976.2022.2108281] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The small intestinal epithelial barrier inputs signals from the gut microbiota in order to balance physiological inflammation and tolerance, and to promote homeostasis. Understanding the dynamic relationship between microbes and intestinal epithelial cells has been a challenge given the cellular heterogeneity associated with the epithelium and the inherent difficulty of isolating and identifying individual cell types. Here, we used single-cell RNA sequencing of small intestinal epithelial cells from germ-free and specific pathogen-free mice to study microbe-epithelium crosstalk at the single-cell resolution. The presence of microbiota did not impact overall cellular composition of the epithelium, except for an increase in Paneth cell numbers. Contrary to expectations, pattern recognition receptors and their adaptors were not induced by the microbiota but showed concentrated expression in a small proportion of epithelial cell subsets. The presence of the microbiota induced the expression of host defense- and glycosylation-associated genes in distinct epithelial cell compartments. Moreover, the microbiota altered the metabolic gene expression profile of epithelial cells, consequently inducing mTOR signaling thereby suggesting microbe-derived metabolites directly activate and regulate mTOR signaling. Altogether, these findings present a resource of the homeostatic transcriptional and cellular impact of the microbiota on the small intestinal epithelium.
Collapse
Affiliation(s)
- Derek K.L. Tsang
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Ryan J. Wang
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Oliver De Sa
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Arshad Ayyaz
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada,Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | | | - Giuliano Bayer
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Shawn Goyal
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Trcka
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Bibaswan Ghoshal
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Jeffrey L. Wrana
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Stephen E. Girardin
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Dana J. Philpott
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada,CONTACT Dana J. Philpott Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Kamioka M, Goto Y, Nakamura K, Yokoi Y, Sugimoto R, Ohira S, Kurashima Y, Umemoto S, Sato S, Kunisawa J, Takahashi Y, Domino SE, Renauld JC, Nakae S, Iwakura Y, Ernst PB, Ayabe T, Kiyono H. Intestinal commensal microbiota and cytokines regulate Fut2 + Paneth cells for gut defense. Proc Natl Acad Sci U S A 2022; 119:e2115230119. [PMID: 35027453 PMCID: PMC8784097 DOI: 10.1073/pnas.2115230119] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/02/2021] [Indexed: 01/10/2023] Open
Abstract
Paneth cells are intestinal epithelial cells that release antimicrobial peptides, such as α-defensin as part of host defense. Together with mesenchymal cells, Paneth cells provide niche factors for epithelial stem cell homeostasis. Here, we report two subtypes of murine Paneth cells, differentiated by their production and utilization of fucosyltransferase 2 (Fut2), which regulates α(1,2)fucosylation to create cohabitation niches for commensal bacteria and prevent invasion of the intestine by pathogenic bacteria. The majority of Fut2- Paneth cells were localized in the duodenum, whereas the majority of Fut2+ Paneth cells were in the ileum. Fut2+ Paneth cells showed higher granularity and structural complexity than did Fut2- Paneth cells, suggesting that Fut2+ Paneth cells are involved in host defense. Signaling by the commensal bacteria, together with interleukin 22 (IL-22), induced the development of Fut2+ Paneth cells. IL-22 was found to affect the α-defensin secretion system via modulation of Fut2 expression, and IL-17a was found to increase the production of α-defensin in the intestinal tract. Thus, these intestinal cytokines regulate the development and function of Fut2+ Paneth cells as part of gut defense.
Collapse
Affiliation(s)
- Mariko Kamioka
- Department of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Department of Medicine, School of Medicine and Chiba University-University of California San Diego Center for Mucosal Immunology, Allergy and Vaccine (CU-UCSD cMAV), University of California, San Diego, CA 92093
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health, and Nutrition, Osaka 567-0085, Japan
| | - Yoshiyuki Goto
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba 260-8673, Japan
| | - Kiminori Nakamura
- Department of Cell Biological Science, Graduate School of Life Science, Faculty of Advanced Life Science, Hokkaido University, Hokkaido 001-0021, Japan
| | - Yuki Yokoi
- Department of Cell Biological Science, Graduate School of Life Science, Faculty of Advanced Life Science, Hokkaido University, Hokkaido 001-0021, Japan
| | - Rina Sugimoto
- Department of Cell Biological Science, Graduate School of Life Science, Faculty of Advanced Life Science, Hokkaido University, Hokkaido 001-0021, Japan
| | - Shuya Ohira
- Department of Cell Biological Science, Graduate School of Life Science, Faculty of Advanced Life Science, Hokkaido University, Hokkaido 001-0021, Japan
| | - Yosuke Kurashima
- Department of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Department of Medicine, School of Medicine and Chiba University-University of California San Diego Center for Mucosal Immunology, Allergy and Vaccine (CU-UCSD cMAV), University of California, San Diego, CA 92093
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health, and Nutrition, Osaka 567-0085, Japan
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Shingo Umemoto
- Department of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Department of Medicine, School of Medicine and Chiba University-University of California San Diego Center for Mucosal Immunology, Allergy and Vaccine (CU-UCSD cMAV), University of California, San Diego, CA 92093
- Department of Otolaryngology and Head and Neck Surgery, Faculty of Medicine, Oita University, Oita 879-5593, Japan
| | - Shintaro Sato
- Department of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Mucosal Vaccine Project, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
- Department of Immunology and Genomics, Osaka City University, Graduate School of Medicine, Osaka 545-8585, Japan
| | - Jun Kunisawa
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health, and Nutrition, Osaka 567-0085, Japan
| | - Yu Takahashi
- Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Steven E Domino
- Department of Obstetrics and Gynecology, Cellular and Molecular Biology Program, University of Michigan Medical Center, Ann Arbor, MI 48109-5617
| | - Jean-Christophe Renauld
- Ludwig Institute for Cancer Research, Université Catholique de Louvain, Brussels B-1200, Belgium
| | - Susumu Nakae
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan
| | - Yoichiro Iwakura
- Center for Experimental Animal Models, Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan
| | - Peter B Ernst
- Department of Medicine, School of Medicine and Chiba University-University of California San Diego Center for Mucosal Immunology, Allergy and Vaccine (CU-UCSD cMAV), University of California, San Diego, CA 92093
- Division of Comparative Pathology and Medicine, Department of Pathology, University of California, San Diego, CA 92093
- Center for Veterinary Sciences and Comparative Medicine, University of California, San Diego, CA 92093
- Future Medicine Education and Research Organization, Chiba University, Chiba 260-8670, Japan
| | - Tokiyoshi Ayabe
- Department of Cell Biological Science, Graduate School of Life Science, Faculty of Advanced Life Science, Hokkaido University, Hokkaido 001-0021, Japan
| | - Hiroshi Kiyono
- Department of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan;
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Department of Medicine, School of Medicine and Chiba University-University of California San Diego Center for Mucosal Immunology, Allergy and Vaccine (CU-UCSD cMAV), University of California, San Diego, CA 92093
- Future Medicine Education and Research Organization, Chiba University, Chiba 260-8670, Japan
| |
Collapse
|
6
|
Brazil JC, Parkos CA. Finding the sweet spot: glycosylation mediated regulation of intestinal inflammation. Mucosal Immunol 2022; 15:211-222. [PMID: 34782709 PMCID: PMC8591159 DOI: 10.1038/s41385-021-00466-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/11/2021] [Accepted: 10/14/2021] [Indexed: 02/04/2023]
Abstract
Glycans are essential cellular components that facilitate a range of critical functions important for tissue development and mucosal homeostasis. Furthermore, specific alterations in glycosylation represent important diagnostic hallmarks of cancer that contribute to tumor cell dissociation, invasion, and metastasis. However, much less is known about how glycosylation contributes to the pathobiology of inflammatory mucosal diseases. Here we will review how epithelial and immune cell glycosylation regulates gut homeostasis and how inflammation-driven changes in glycosylation contribute to intestinal pathobiology.
Collapse
Affiliation(s)
- Jennifer C. Brazil
- grid.214458.e0000000086837370Department of Pathology, University of Michigan, Ann Arbor, MI 48109 USA
| | - Charles A. Parkos
- grid.214458.e0000000086837370Department of Pathology, University of Michigan, Ann Arbor, MI 48109 USA
| |
Collapse
|
7
|
Fan Q, Wu Y, Li M, An F, Yao L, Wang M, Wang X, Yuan J, Jiang K, Li W, Li M. Lactobacillus spp. create a protective micro-ecological environment through regulating the core fucosylation of vaginal epithelial cells against cervical cancer. Cell Death Dis 2021; 12:1094. [PMID: 34799549 PMCID: PMC8604912 DOI: 10.1038/s41419-021-04388-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 10/30/2021] [Accepted: 11/05/2021] [Indexed: 12/24/2022]
Abstract
Vaginal dysbiosis often occurs in patients with cervical cancer. The fucosylation of mucosal epithelial cells is closely related to microbial colonization, and play an important role in protecting the vaginal mucosal epithelial cells. However, no reports on the relationship between vaginal dysbiosis and abnormal mucosal epithelial cell fucosylation, and their roles in the occurrence and development of cervical cancer are unavailable. Here we report that core fucosylation levels were significantly lower in the serum, exfoliated cervical cells and tumor tissue of cervical cancer patients. Core fucosyltransferase gene (Fut8) knockout promoted the proliferation and migration of cervical cancer cells. In patients with cervical cancer, the vaginal dysbiosis, and the abundance of Lactobacillus, especially L. iners, was significantly reduced. Meanwhile, the abundance of L.iners was positively correlated with core fucosylation levels. The L. iners metabolite lactate can activate the Wnt pathway through the lactate-Gpr81 complex, which increases the level of core fucosylation in epidermal cells, inhibiting the proliferation and migration of cervical cancer cells, and have application prospects in regulating the vaginal microecology and preventing cervical cancer.
Collapse
Affiliation(s)
- Qingjie Fan
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Yuanhang Wu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Mechou Li
- Department of Medical Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Fan An
- The Cancer Stem Cell Research Institute of Dalian Medical University, Dalian, China
| | - Lulu Yao
- Department of Medical Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Meixian Wang
- The Reproductive and Genetics Center of Dalian Women and Children's Medical Center (Group), Dalian, China
| | - Xiuying Wang
- The Gynecology and Oncology Ward of Dalian Maternal and Child Health Hospital, Dalian, China
| | - Jieli Yuan
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Kui Jiang
- Department of Medical Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Wenzhe Li
- College of Basic Medical Science, Dalian Medical University, Dalian, China.
| | - Ming Li
- College of Basic Medical Science, Dalian Medical University, Dalian, China.
| |
Collapse
|
8
|
Li Y, Jiang Y, Zhang L, Qian W, Hou X, Lin R. Exogenous l-fucose protects the intestinal mucosal barrier depending on upregulation of FUT2-mediated fucosylation of intestinal epithelial cells. FASEB J 2021; 35:e21699. [PMID: 34151459 DOI: 10.1096/fj.202002446rrrr] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 05/09/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022]
Abstract
FUT2, a protein that uses l-fucose to mediate fucosylation of intestinal epithelial cells, is one of the detected gene variants in IBD patients. We aimed to investigate whether exogenous l-fucose could be an enteral nutritional supplement to protect intestinal barrier function. The effect of l-fucose on the restoration of epithelial barrier function in both the DSS-induced colitis mouse model and LPS-stimulated Caco-2 cells was investigated, and the impact on fucosylation of epithelial cells was examined. The severity of DSS-induced colitis was significantly reduced by l-fucose. Restoration of epithelial barrier function by l-fucose was detected. Direct l-fucose-mediated protection of tight junctions was observed in Caco-2 cells. Moreover, exogenous l-fucose promoted the exogenous metabolic pathway of l-fucose, and fucosylation of epithelial cells both in vivo and in vitro. Moreover, knockout of the FUT2 gene restrained fucosylation and the protective effect of l-fucose on barrier function. The severity of colitis was not improved by l-fucose in Fut2 knockout mice. Therefore we conclude that exogenous l-fucose protects intestinal barrier function and relieves intestinal inflammation via upregulation of FUT2-mediated fucosylation of intestinal epithelial cells.
Collapse
Affiliation(s)
- Ying Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yudong Jiang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zhang
- 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
| | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Lin
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Kononova S, Litvinova E, Vakhitov T, Skalinskaya M, Sitkin S. Acceptive Immunity: The Role of Fucosylated Glycans in Human Host-Microbiome Interactions. Int J Mol Sci 2021; 22:3854. [PMID: 33917768 PMCID: PMC8068183 DOI: 10.3390/ijms22083854] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/04/2021] [Accepted: 04/05/2021] [Indexed: 02/07/2023] Open
Abstract
The growth in the number of chronic non-communicable diseases in the second half of the past century and in the first two decades of the new century is largely due to the disruption of the relationship between the human body and its symbiotic microbiota, and not pathogens. The interaction of the human immune system with symbionts is not accompanied by inflammation, but is a physiological norm. This is achieved via microbiota control by the immune system through a complex balance of pro-inflammatory and suppressive responses, and only a disturbance of this balance can trigger pathophysiological mechanisms. This review discusses the establishment of homeostatic relationships during immune system development and intestinal bacterial colonization through the interaction of milk glycans, mucins, and secretory immunoglobulins. In particular, the role of fucose and fucosylated glycans in the mechanism of interactions between host epithelial and immune cells is discussed.
Collapse
Affiliation(s)
- Svetlana Kononova
- Department of Microbiology, State Research Institute of Highly Pure Biopreparations, 197110 St. Petersburg, Russia; (T.V.); (M.S.); (S.S.)
- Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Ekaterina Litvinova
- Scientific-Research Institute of Neurosciences and Medicine, 630117 Novosibirsk, Russia;
- Siberian Federal Scientific Center of Agro-BioTechnologies, Russian Academy of Sciences, Krasnoobsk, 633501 Novosibirsk, Russia
| | - Timur Vakhitov
- Department of Microbiology, State Research Institute of Highly Pure Biopreparations, 197110 St. Petersburg, Russia; (T.V.); (M.S.); (S.S.)
| | - Maria Skalinskaya
- Department of Microbiology, State Research Institute of Highly Pure Biopreparations, 197110 St. Petersburg, Russia; (T.V.); (M.S.); (S.S.)
- Department of Internal Diseases, Gastroenterology and Dietetics, North-Western State Medical University Named after I.I. Mechnikov, 191015 St. Petersburg, Russia
| | - Stanislav Sitkin
- Department of Microbiology, State Research Institute of Highly Pure Biopreparations, 197110 St. Petersburg, Russia; (T.V.); (M.S.); (S.S.)
- Department of Internal Diseases, Gastroenterology and Dietetics, North-Western State Medical University Named after I.I. Mechnikov, 191015 St. Petersburg, Russia
- Institute of Perinatology and Pediatrics, Almazov National Medical Research Centre, 197341 St. Petersburg, Russia
| |
Collapse
|
10
|
She X, Du H, Yi C, He Y, Ai Q, Yu J. The decrease of fucosylation in intestinal epithelium is related to the development of necrotizing enterocolitis. Mol Immunol 2021; 131:23-32. [PMID: 33465592 DOI: 10.1016/j.molimm.2020.12.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/06/2020] [Accepted: 12/30/2020] [Indexed: 02/07/2023]
Abstract
Necrotizing enterocolitis (NEC) is a devastating neonatal gastrointestinal emergency. Fucosylated glycans on intestinal epithelial cells (IECs) play a central role in the maintenance of intestinal homeostasis. Nevertheless, its association with necrotizing enterocolitis is not clear. We examined paraffin-embedded intestinal specimens from participants and found that the NEC patients showed lower intestinal epithelial fucosylation levels than the control patients. In the mouse model of NEC, the percentage of fucosylated epithelial cells (F-ECs) and ILC3s was decreased. Also, the expression levels of IL-22 and Fut2 were reduced. Moreover, the critical role of epithelial fucosylation in NEC was further confirmed by administering the anti-IL-22 antibody, which caused an increase in histological damage, body weight loss, intestinal permeability and proinflammatory cytokine release correlated with a reduction of F-ECs. Overall, intestinal fucosylation deficiency led to increased susceptibility and severity of NEC. Further studies are needed to determine whether modification of intestinal fucosylation affects the development of NEC.
Collapse
Affiliation(s)
- Xiang She
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Pediatrics, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China; National Clinical Research Center for Child Health and Disorder, Chongqing, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Hua Du
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Pediatrics, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China; National Clinical Research Center for Child Health and Disorder, Chongqing, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Cong Yi
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Pediatrics, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China; National Clinical Research Center for Child Health and Disorder, Chongqing, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Yu He
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Pediatrics, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China; National Clinical Research Center for Child Health and Disorder, Chongqing, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Qing Ai
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Pediatrics, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China; National Clinical Research Center for Child Health and Disorder, Chongqing, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Jialin Yu
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China; Department of Pediatrics, Shenzhen University General Hospital, Shenzhen, Guangdong, China; Chongqing Key Laboratory of Pediatrics, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China; National Clinical Research Center for Child Health and Disorder, Chongqing, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.
| |
Collapse
|
11
|
Kudelka MR, Stowell SR, Cummings RD, Neish AS. Intestinal epithelial glycosylation in homeostasis and gut microbiota interactions in IBD. Nat Rev Gastroenterol Hepatol 2020; 17:597-617. [PMID: 32710014 PMCID: PMC8211394 DOI: 10.1038/s41575-020-0331-7] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/05/2020] [Indexed: 02/08/2023]
Abstract
Inflammatory bowel disease (IBD) affects 6.8 million people globally. A variety of factors have been implicated in IBD pathogenesis, including host genetics, immune dysregulation and gut microbiota alterations. Emerging evidence implicates intestinal epithelial glycosylation as an underappreciated process that interfaces with these three factors. IBD is associated with increased expression of truncated O-glycans as well as altered expression of terminal glycan structures. IBD genes, glycosyltransferase mislocalization, altered glycosyltransferase and glycosidase expression and dysbiosis drive changes in the glycome. These glycan changes disrupt the mucus layer, glycan-lectin interactions, host-microorganism interactions and mucosal immunity, and ultimately contribute to IBD pathogenesis. Epithelial glycans are especially critical in regulating the gut microbiota through providing bacterial ligands and nutrients and ultimately determining the spatial organization of the gut microbiota. In this Review, we discuss the regulation of intestinal epithelial glycosylation, altered epithelial glycosylation in IBD and mechanisms for how these alterations contribute to disease pathobiology. We hope that this Review provides a foundation for future studies on IBD glycosylation and the emergence of glycan-inspired therapies for IBD.
Collapse
Affiliation(s)
- Matthew R Kudelka
- Medical Scientist Training Program, Emory University School of Medicine, Atlanta, GA, USA
- Department of Internal Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sean R Stowell
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Andrew S Neish
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
12
|
Chen YM, Helm ET, Gabler N, Hostetter JM, Burrough ER. Alterations in Intestinal Innate Mucosal Immunity of Weaned Pigs During Porcine Epidemic Diarrhea Virus Infection. Vet Pathol 2020; 57:642-652. [PMID: 32880235 DOI: 10.1177/0300985820932140] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In the small intestine, localized innate mucosal immunity is critical for intestinal homeostasis. Porcine epidemic diarrhea virus (PEDV) infection induces villus injury and impairs digestive function. Moreover, the infection might comprise localized innate mucosal immunity. This study investigated specific enterocyte subtypes and innate immune components of weaned pigs during PEDV infection. Four-week-old pigs were orally inoculated with PEDV IN19338 strain (n = 40) or sham-inoculated (n = 24). At day post inoculation (DPI) 2, 4, and 6, lysozyme expression in Paneth cells, cellular density of villous and Peyer's patch microfold (M) cells, and the expression of polymeric immunoglobulin receptor (pIgR) were assessed in the jejunum and ileum by immunohistochemistry, and interleukin (IL)-1β and tumor necrosis factor (TNF)-α were measured in the jejunum by ELISA. PEDV infection led to a decrease in the ratios of villus height to crypt depth (VH-CD) in jejunum at DPI 2, 4, and 6 and in ileum at DPI 4. The number of villous M cells was reduced in jejunum at DPI 4 and 6 and in ileum at DPI 6, while the number of Peyer's patch M cells in ileum increased at DPI 2 and then decreased at DPI 6. PEDV-infected pigs also had reduced lysozyme expression in ileal Paneth cells at DPI 2 and increased ileal pIgR expression at DPI 4. There were no significant changes in IL-1β and TNF-α expression in PEDV-infected pigs compared to controls. In conclusion, PEDV infection affected innate mucosal immunity of weaned pigs through alterations in Paneth cells, villous and Peyer's patch M cells, and pIgR expression.
Collapse
|
13
|
Regular alteration of protein glycosylation in skeletal muscles of hibernating Daurian ground squirrels (Spermophilus dauricus). Comp Biochem Physiol B Biochem Mol Biol 2019; 237:110323. [PMID: 31454680 DOI: 10.1016/j.cbpb.2019.110323] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/19/2019] [Accepted: 08/22/2019] [Indexed: 11/21/2022]
Abstract
Glycosylation is one of the most common post-translational protein modifications and is closely associated with muscle atrophy. This study aims to investigate the changes in glycan profiles in the fast-twitch extensor digitorum longus (EDL) muscles of Daurian ground squirrels (Spermophilus dauricus) during hibernation as well as the correlation between protein glycosylation and muscle atrophy prevention in hibernating animals. The results showed that there was no significant change in the muscle-to-body mass ratio, muscle fiber cross-sectional area (CSA), fiber distribution and ultrastructures in the EDL muscles of ground squirrels during hibernation. Alterations of six glycans comprising sialic acid α2-3 galactose (Sia2-3Gal) and Fucα1-2Galβ1-4Glc(NAc) in the EDL muscles were observed. In addition, the observed downregulation of sialyltransferase (ST3Gals) mRNA levels and upregulation of fucosyltransferase (FUT1 and FUT2) mRNA levels during hibernation and the subsequent restoration to normal levels during periodic interbout arousal were consistent with the changes in sialic acid and fucose modifications. Our results indicate that changes in ST3Gals and FUTs in the EDL muscles of Daurian ground squirrels during hibernation can alter sialylation and fucosylation of muscle glycoproteins, which may protect the skeletal muscles of hibernating Daurian ground squirrels from disuse atrophy.
Collapse
|
14
|
Singh RP. Glycan utilisation system in Bacteroides and Bifidobacteria and their roles in gut stability and health. Appl Microbiol Biotechnol 2019; 103:7287-7315. [PMID: 31332487 DOI: 10.1007/s00253-019-10012-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 02/07/2023]
Abstract
Gut residential hundred trillion microbial cells are indispensable for maintaining gut homeostasis and impact on host physiology, development and immune systems. Many of them have displayed excellence in utilising dietary- and host-derived complex glycans and are producing useful postbiotics including short-chain fatty acids to primarily fuel different organs of the host. Therefore, employing individual microbiota is nowadays becoming a propitious target in biomedical for improving gut dysbiosis conditions of the host. Among other gut microbial communities, Bacteroides and Bifidobacteria are coevolved to utilise diverse ranges of diet- and host-derived glycans through harmonising distinct glycan utilisation systems. These gut symbionts frequently share digested oligosaccharides, carbohydrate-active enzymes and fermentable intermediate molecules for sustaining gut microbial symbiosis and improving fitness of own or other communities. Genomics approaches have provided unprecedented insights into these functions, but their precise mechanisms of action have poorly known. Sympathetic glycan-utilising strategy of each gut commensal will provide overview of mechanistic dynamic nature of the gut environment and will then assist in applying aptly personalised nutritional therapy. Thus, the review critically summarises cutting edge understanding of major plant- and host-derived glycan-utilising systems of Bacteroides and Bifidobacteria. Their evolutionary adaptation to gut environment and roles of postbiotics in human health are also highlighted.
Collapse
Affiliation(s)
- Ravindra Pal Singh
- Food and Nutritional Biotechnology Division, National Agri-Food Biotechnology Institute (NABI), SAS, Nagar, Punjab, 140306, India.
| |
Collapse
|
15
|
McKeen S, Young W, Fraser K, Roy NC, McNabb WC. Glycan Utilisation and Function in the Microbiome of Weaning Infants. Microorganisms 2019; 7:microorganisms7070190. [PMID: 31277402 PMCID: PMC6681113 DOI: 10.3390/microorganisms7070190] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 06/23/2019] [Accepted: 06/25/2019] [Indexed: 12/12/2022] Open
Abstract
Glycans are present exogenously in the diet, expressed and secreted endogenously by host cells, and produced by microbes. All of these processes result in them being available to the gut microbiome, firmly placing glycans at the interface of diet–microbe–host interactions. The most dramatic shift in dietary sources of glycans occurs during the transition from the milk-based neonatal diet to the diverse omnivorous adult diet, and this has profound effects on the composition of the gut microbiome, gene expression by microbes and host cells, mucin composition, and immune development from innate towards adaptive responses. Understanding the glycan-mediated interactions occurring during this transitional window may inform dietary recommendations to support gut and immune development during a vulnerable age. This review aims to summarise the current state of knowledge on dietary glycan mediated changes that may occur in the infant gut microbiome and immune system during weaning.
Collapse
Affiliation(s)
- Starin McKeen
- Food Nutrition & Health, AgResearch, Grasslands Research Centre, Private Bag 11008, Palmerston north 4442, New Zealand
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand
- High-Value Nutrition National Science Challenge, Auckland 1023, New Zealand
| | - Wayne Young
- Food Nutrition & Health, AgResearch, Grasslands Research Centre, Private Bag 11008, Palmerston north 4442, New Zealand
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand
- High-Value Nutrition National Science Challenge, Auckland 1023, New Zealand
| | - Karl Fraser
- Food Nutrition & Health, AgResearch, Grasslands Research Centre, Private Bag 11008, Palmerston north 4442, New Zealand
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand
- High-Value Nutrition National Science Challenge, Auckland 1023, New Zealand
| | - Nicole C Roy
- Food Nutrition & Health, AgResearch, Grasslands Research Centre, Private Bag 11008, Palmerston north 4442, New Zealand
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand
- High-Value Nutrition National Science Challenge, Auckland 1023, New Zealand
| | - Warren C McNabb
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland 1023, New Zealand.
| |
Collapse
|
16
|
Suwandi A, Galeev A, Riedel R, Sharma S, Seeger K, Sterzenbach T, García Pastor L, Boyle EC, Gal-Mor O, Hensel M, Casadesús J, Baines JF, Grassl GA. Std fimbriae-fucose interaction increases Salmonella-induced intestinal inflammation and prolongs colonization. PLoS Pathog 2019; 15:e1007915. [PMID: 31329635 PMCID: PMC6675130 DOI: 10.1371/journal.ppat.1007915] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 08/01/2019] [Accepted: 06/13/2019] [Indexed: 12/14/2022] Open
Abstract
Expression of ABO and Lewis histo-blood group antigens by the gastrointestinal epithelium is governed by an α-1,2-fucosyltransferase enzyme encoded by the Fut2 gene. Alterations in mucin glycosylation have been associated with susceptibility to various bacterial and viral infections. Salmonella enterica serovar Typhimurium is a food-borne pathogen and a major cause of gastroenteritis. In order to determine the role of Fut2-dependent glycans in Salmonella-triggered intestinal inflammation, Fut2+/+ and Fut2-/- mice were orally infected with S. Typhimurium and bacterial colonization and intestinal inflammation were analyzed. Bacterial load in the intestine of Fut2-/- mice was significantly lower compared to Fut2+/+ mice. Analysis of histopathological changes revealed significantly lower levels of intestinal inflammation in Fut2-/- mice compared to Fut2+/+ mice and measurement of lipocalin-2 level in feces corroborated histopathological findings. Salmonella express fimbriae that assist in adherence of bacteria to host cells thereby facilitating their invasion. The std fimbrial operon of S. Typhimurium encodes the π-class Std fimbriae which bind terminal α(1,2)-fucose residues. An isogenic mutant of S. Typhimurium lacking Std fimbriae colonized Fut2+/+ and Fut2-/- mice to similar levels and resulted in similar intestinal inflammation. In vitro adhesion assays revealed that bacteria possessing Std fimbriae adhered significantly more to fucosylated cell lines or primary epithelial cells in comparison to cells lacking α(1,2)-fucose. Overall, these results indicate that Salmonella-triggered intestinal inflammation and colonization are dependent on Std-fucose interaction.
Collapse
Affiliation(s)
- Abdulhadi Suwandi
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research (DZIF), Hannover, Germany
| | - Alibek Galeev
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research (DZIF), Hannover, Germany
| | - René Riedel
- Max Planck Institute for Evolutionary Biology, Evolutionary Genomics, Plön, Germany and Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Samriti Sharma
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research (DZIF), Hannover, Germany
| | - Katrin Seeger
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research (DZIF), Hannover, Germany
| | - Torsten Sterzenbach
- Division of Microbiology and CellNanOs–Center for Cellular Nanoanalytics, University of Osnabrück, Osnabrück, Germany
| | - Lucía García Pastor
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - Erin C. Boyle
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Ohad Gal-Mor
- The Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer, Israel
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michael Hensel
- Division of Microbiology and CellNanOs–Center for Cellular Nanoanalytics, University of Osnabrück, Osnabrück, Germany
| | - Josep Casadesús
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - John F. Baines
- Max Planck Institute for Evolutionary Biology, Evolutionary Genomics, Plön, Germany and Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Guntram A. Grassl
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research (DZIF), Hannover, Germany
| |
Collapse
|
17
|
Pathogen Colonization Resistance in the Gut and Its Manipulation for Improved Health. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1300-1310. [PMID: 31100210 DOI: 10.1016/j.ajpath.2019.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/15/2019] [Accepted: 03/05/2019] [Indexed: 02/07/2023]
Abstract
Mammals have coevolved with a large community of symbiotic, commensal, and some potentially pathogenic microbes. The trillions of bacteria and hundreds of species in our guts form a relatively stable community that resists invasion by outsiders, including pathogens. This powerful protective force is referred to as colonization resistance. We discuss the variety of proposed or demonstrated mechanisms that can mediate colonization resistance and some potential ways to manipulate them for improved human health. Instances in which certain bacterial pathogens can overcome colonization resistance are also discussed.
Collapse
|
18
|
Colomb F, Giron LB, Trbojevic-Akmacic I, Lauc G, Abdel-Mohsen M. Breaking the Glyco-Code of HIV Persistence and Immunopathogenesis. Curr HIV/AIDS Rep 2019; 16:151-168. [PMID: 30707400 PMCID: PMC6441623 DOI: 10.1007/s11904-019-00433-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Glycoimmunology is an emerging field focused on understanding how immune responses are mediated by glycans (carbohydrates) and their interaction with glycan-binding proteins called lectins. How glycans influence immunological functions is increasingly well understood. In a parallel way, in the HIV field, it is increasingly understood how the host immune system controls HIV persistence and immunopathogenesis. However, what has mostly been overlooked, despite its potential for therapeutic applications, is the role that the host glycosylation machinery plays in modulating the persistence and immunopathogenesis of HIV. Here, we will survey four areas in which the links between glycan-lectin interactions and immunology and between immunology and HIV are well described. For each area, we will describe these links and then delineate the opportunities for the HIV field in investigating potential interactions between glycoimmunology and HIV persistence/immunopathogenesis. RECENT FINDINGS Recent studies show that the human glycome (the repertoire of human glycan structures) plays critical roles in driving or modulating several cellular processes and immunological functions that are central to maintaining HIV infection. Understanding the links between glycoimmunology and HIV infection may create a new paradigm for discovering novel glycan-based therapies that can lead to eradication, functional cure, or improved tolerance of lifelong infection.
Collapse
Affiliation(s)
- Florent Colomb
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, USA
| | - Leila B Giron
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, USA
| | | | - Gordan Lauc
- Genos Glycoscience Research Laboratory, Borongajska cesta 83h, Zagreb, Croatia
- Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovacica 1, Zagreb, Croatia
| | | |
Collapse
|
19
|
Dias AM, Pereira MS, Padrão NA, Alves I, Marcos-Pinto R, Lago P, Pinho SS. Glycans as critical regulators of gut immunity in homeostasis and disease. Cell Immunol 2018; 333:9-18. [DOI: 10.1016/j.cellimm.2018.07.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 07/04/2018] [Accepted: 07/17/2018] [Indexed: 12/19/2022]
|
20
|
Omata Y, Aoki R, Aoki-Yoshida A, Hiemori K, Toyoda A, Tateno H, Suzuki C, Takayama Y. Reduced fucosylation in the distal intestinal epithelium of mice subjected to chronic social defeat stress. Sci Rep 2018; 8:13199. [PMID: 30181591 PMCID: PMC6123462 DOI: 10.1038/s41598-018-31403-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 08/14/2018] [Indexed: 01/28/2023] Open
Abstract
Psychological stress can cause dysfunction of the gastrointestinal tract by regulating its interaction with central nervous system (brain-gut axis). Chronic social defeat stress (CSDS) is widely used to produce a rodent model of stress-induced human mood disorders and depression. We previously showed that CSDS significantly affects the intestinal ecosystem including cecal and fecal microbiota, intestinal gene expression profiles and cecal metabolite profiles. Here, we investigated whether the glycosylation pattern in the intestinal epithelium was affected in C57BL/6 mice exposed to CSDS (hereinafter referred to as CSDS mice). A lectin microarray analysis revealed that CSDS significantly reduced the reactivity of fucose-specific lectins (rAOL, TJA-II, rAAL, rGC2, AOL, AAL, rPAIIL and rRSIIL) with distal intestinal mucosa, but not with mucosa from proximal intestine and colon. Flow cytometric analysis confirmed the reduced TJA-II reactivity with intestinal epithelial cells in CSDS mice. In addition, distal intestine expression levels of the genes encoding fucosyltransferase 1 and 2 (Fut1 and Fut2) were downregulated in CSDS mice. These findings suggest that CSDS alters the fucosylation pattern in the distal intestinal epithelium, which could be used as a sensitive marker for CSDS exposure.
Collapse
Affiliation(s)
- Yasuhiro Omata
- National Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, 305-0901, Japan
| | - Reiji Aoki
- National Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, 305-0901, Japan
| | - Ayako Aoki-Yoshida
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Keiko Hiemori
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, 305-8568, Japan
| | - Atsushi Toyoda
- College of Agriculture, Ibaraki University, Ami, Ibaraki, 300-0393, Japan
| | - Hiroaki Tateno
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, 305-8568, Japan
| | - Chise Suzuki
- National Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, 305-0901, Japan
| | - Yoshiharu Takayama
- National Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, 305-0901, Japan.
| |
Collapse
|
21
|
Li J, Hsu HC, Mountz JD, Allen JG. Unmasking Fucosylation: from Cell Adhesion to Immune System Regulation and Diseases. Cell Chem Biol 2018. [DOI: 10.1016/j.chembiol.2018.02.005] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
22
|
Diesner SC, Bergmayr C, Wang XY, Heiden D, Exenberger S, Roth-Walter F, Starkl P, Ret D, Pali-Schöll I, Gabor F, Untersmayr E. Characterization of Vibrio cholerae neuraminidase as an immunomodulator for novel formulation of oral allergy immunotherapy. Clin Immunol 2018; 192:30-39. [PMID: 29608970 DOI: 10.1016/j.clim.2018.03.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 09/05/2017] [Accepted: 03/28/2018] [Indexed: 01/22/2023]
Abstract
To improve current mucosal allergen immunotherapy Vibrio cholerae neuraminidase (NA) was evaluated as a novel epithelial targeting molecule for functionalization of allergen-loaded, poly(D,L-lactide-co-glycolide) (PLGA) microparticles (MPs) and compared to the previously described epithelial targeting lectins wheat germ agglutinin (WGA) and Aleuria aurantia lectin (AAL). All targeters revealed binding to Caco-2 cells, but only NA had high binding specificity to α-L fucose and monosialoganglioside-1. An increased transepithelial uptake was found for NA-MPs in a M-cell co-culture model. NA and NA-MPs induced high levels of IFN-γ and IL10 in naive mouse splenocytes and CCL20 expression in Caco-2. Repeated oral gavage of NA-MPs resulted in a modulated, allergen-specific immune response. In conclusion, NA has enhanced M-cell specificity compared to the other targeters. NA functionalized MPs induce a Th1 and T-regulatory driven immune response and avoid allergy effector cell activation. Therefore, it is a promising novel, orally applied formula for allergy therapy.
Collapse
Affiliation(s)
- Susanne C Diesner
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Cornelia Bergmayr
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Xue-Yan Wang
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Denise Heiden
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Sarah Exenberger
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Franziska Roth-Walter
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Philipp Starkl
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Davide Ret
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Institute of Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria
| | - Isabella Pali-Schöll
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Franz Gabor
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Eva Untersmayr
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
23
|
Cervin J, Wands AM, Casselbrant A, Wu H, Krishnamurthy S, Cvjetkovic A, Estelius J, Dedic B, Sethi A, Wallom KL, Riise R, Bäckström M, Wallenius V, Platt FM, Lebens M, Teneberg S, Fändriks L, Kohler JJ, Yrlid U. GM1 ganglioside-independent intoxication by Cholera toxin. PLoS Pathog 2018; 14:e1006862. [PMID: 29432456 PMCID: PMC5825173 DOI: 10.1371/journal.ppat.1006862] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 02/23/2018] [Accepted: 01/08/2018] [Indexed: 11/18/2022] Open
Abstract
Cholera toxin (CT) enters and intoxicates host cells after binding cell surface receptors via its B subunit (CTB). We have recently shown that in addition to the previously described binding partner ganglioside GM1, CTB binds to fucosylated proteins. Using flow cytometric analysis of primary human jejunal epithelial cells and granulocytes, we now show that CTB binding correlates with expression of the fucosylated Lewis X (LeX) glycan. This binding is competitively blocked by fucosylated oligosaccharides and fucose-binding lectins. CTB binds the LeX glycan in vitro when this moiety is linked to proteins but not to ceramides, and this binding can be blocked by mAb to LeX. Inhibition of glycosphingolipid synthesis or sialylation in GM1-deficient C6 rat glioma cells results in sensitization to CT-mediated intoxication. Finally, CT gavage produces an intact diarrheal response in knockout mice lacking GM1 even after additional reduction of glycosphingolipids. Hence our results show that CT can induce toxicity in the absence of GM1 and support a role for host glycoproteins in CT intoxication. These findings open up new avenues for therapies to block CT action and for design of detoxified enterotoxin-based adjuvants.
Collapse
Affiliation(s)
- Jakob Cervin
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Amberlyn M. Wands
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Anna Casselbrant
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Han Wu
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Soumya Krishnamurthy
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Aleksander Cvjetkovic
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Johanna Estelius
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Benjamin Dedic
- Department of Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anirudh Sethi
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Kerri-Lee Wallom
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Rebecca Riise
- Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Malin Bäckström
- Mammalian Protein Expression Core Facility, University of Gothenburg, Gothenburg, Sweden
| | - Ville Wallenius
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Frances M. Platt
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Michael Lebens
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Susann Teneberg
- Department of Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lars Fändriks
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jennifer J. Kohler
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Ulf Yrlid
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
24
|
Bosi E, Bacci G, Mengoni A, Fondi M. Perspectives and Challenges in Microbial Communities Metabolic Modeling. Front Genet 2017; 8:88. [PMID: 28680442 PMCID: PMC5478693 DOI: 10.3389/fgene.2017.00088] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 06/09/2017] [Indexed: 01/31/2023] Open
Abstract
Bacteria have evolved to efficiently interact each other, forming complex entities known as microbial communities. These "super-organisms" play a central role in maintaining the health of their eukaryotic hosts and in the cycling of elements like carbon and nitrogen. However, despite their crucial importance, the mechanisms that influence the functioning of microbial communities and their relationship with environmental perturbations are obscure. The study of microbial communities was boosted by tremendous advances in sequencing technologies, and in particular by the possibility to determine genomic sequences of bacteria directly from environmental samples. Indeed, with the advent of metagenomics, it has become possible to investigate, on a previously unparalleled scale, the taxonomical composition and the functional genetic elements present in a specific community. Notwithstanding, the metagenomic approach per se suffers some limitations, among which the impossibility of modeling molecular-level (e.g., metabolic) interactions occurring between community members, as well as their effects on the overall stability of the entire system. The family of constraint-based methods, such as flux balance analysis, has been fruitfully used to translate genome sequences in predictive, genome-scale modeling platforms. Although these techniques have been initially developed for analyzing single, well-known model organisms, their recent improvements allowed engaging in multi-organism in silico analyses characterized by a considerable predictive capability. In the face of these advances, here we focus on providing an overview of the possibilities and challenges related to the modeling of metabolic interactions within a bacterial community, discussing the feasibility and the perspectives of this kind of analysis in the (near) future.
Collapse
Affiliation(s)
| | | | - Alessio Mengoni
- Department of Biology, University of FlorenceFlorence, Italy
| | | |
Collapse
|
25
|
Kurashima Y, Kiyono H. Mucosal Ecological Network of Epithelium and Immune Cells for Gut Homeostasis and Tissue Healing. Annu Rev Immunol 2017; 35:119-147. [PMID: 28125357 DOI: 10.1146/annurev-immunol-051116-052424] [Citation(s) in RCA: 207] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The intestinal epithelial barrier includes columnar epithelial, Paneth, goblet, enteroendocrine, and tuft cells as well as other cell populations, all of which contribute properties essential for gastrointestinal homeostasis. The intestinal mucosa is covered by mucin, which contains antimicrobial peptides and secretory IgA and prevents luminal bacteria, fungi, and viruses from stimulating intestinal immune responses. Conversely, the transport of luminal microorganisms-mediated by M, dendritic, and goblet cells-into intestinal tissues facilitates the harmonization of active and quiescent mucosal immune responses. The bacterial population within gut-associated lymphoid tissues creates the intratissue cohabitations for harmonized mucosal immunity. Intermolecular and intercellular communication among epithelial, immune, and mesenchymal cells creates an environment conducive for epithelial regeneration and mucosal healing. This review summarizes the so-called intestinal mucosal ecological network-the complex but vital molecular and cellular interactions of epithelial mesenchymal cells, immune cells, and commensal microbiota that achieve intestinal homeostasis, regeneration, and healing.
Collapse
Affiliation(s)
- Yosuke Kurashima
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan; .,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan.,Institute for Global Prominent Research, Chiba University, Chiba 260-8670, Japan.,Department of Mucosal Immunology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan.,Department of Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan.,Chiba University-UC San Diego Center for Mucosal Immunology, Allergy, and Vaccine, La Jolla, CA 92093
| | - Hiroshi Kiyono
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan; .,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan.,Chiba University-UC San Diego Center for Mucosal Immunology, Allergy, and Vaccine, La Jolla, CA 92093.,Department of Immunology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| |
Collapse
|
26
|
Goto Y, Uematsu S, Kiyono H. Epithelial glycosylation in gut homeostasis and inflammation. Nat Immunol 2016; 17:1244-1251. [PMID: 27760104 DOI: 10.1038/ni.3587] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intestinal epithelial cells apically express glycans, especially α1,2-fucosyl linkages, which work as a biological interface for the host-microbe interaction. Emerging studies have shown that epithelial α1,2-fucosylation is regulated by microbes and by group 3 innate lymphoid cells (ILC3s). Dysregulation of the gene (FUT2) encoding fucosyltransferase 2, an enzyme governing epithelial α1,2-fucosylation, is associated with various human disorders, including infection and chronic inflammatory diseases. This suggests a critical role for an interaction between microbes, epithelial cells and ILC3s mediated via glycan residues. In this Review, using α1,2-fucose and Fut2 gene expression as an example, we describe how epithelial glycosylation is controlled by immune cells and luminal microbes. We also address the pathophysiological contribution of epithelial α1,2-fucosylation to pathogenic and commensal microbes as well as the potential of α1,2-fucose and its regulatory pathway as previously unexploited targets in the development of new therapeutic approaches for human diseases.
Collapse
Affiliation(s)
- Yoshiyuki Goto
- Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japan
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Satoshi Uematsu
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Mucosal Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiroshi Kiyono
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Division of Mucosal Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
27
|
Aoki-Yoshida A, Aoki R, Moriya N, Goto T, Kubota Y, Toyoda A, Takayama Y, Suzuki C. Omics Studies of the Murine Intestinal Ecosystem Exposed to Subchronic and Mild Social Defeat Stress. J Proteome Res 2016; 15:3126-38. [PMID: 27482843 DOI: 10.1021/acs.jproteome.6b00262] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The microbiota-gut-brain axis plays an important role in the development of stress-induced mental disorders. We previously established the subchronic and mild social defeat stress (sCSDS) model, a murine experimental model of depression, and investigated the metabolomic profiles of plasma and liver. Here we used omics approaches to identify stress-induced changes in the gastrointestinal tract. Mice exposed to sCSDS for 10 days showed the following changes: (1) elevation of cholic acid and reduction of 5-aminovaleric acid among cecal metabolites; (2) downregulation of genes involved in the immune response in the terminal ileum; (3) a shift in the diversity of the microbiota in cecal contents and feces; and (4) fluctuations in the concentrations of cecal metabolites produced by gut microbiota reflected in plasma and hepatic metabolites. Operational taxonomic units within the family Lachnospiraceae showed an inverse correlation with certain metabolites. The social interaction score correlated with cecal metabolites, IgA, and cecal and fecal microbiota, suggesting that sCSDS suppressed the ileal immune response, altering the balance of microbiota, which together with host cells and host enzymes resulted in a pattern of accumulated metabolites in the intestinal ecosystem distinct from that of control mice.
Collapse
Affiliation(s)
- Ayako Aoki-Yoshida
- Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO) , Tsukuba, Ibaraki 305-0901, Japan
| | - Reiji Aoki
- Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO) , Tsukuba, Ibaraki 305-0901, Japan
| | - Naoko Moriya
- Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO) , Tsukuba, Ibaraki 305-0901, Japan
| | - Tatsuhiko Goto
- College of Agriculture, Ibaraki University , Ami, Ibaraki 300-0393, Japan.,Ibaraki University Cooperation between Agriculture and Medical Science (IUCAM) , Ami, Ibaraki 300-0393, Japan
| | - Yoshifumi Kubota
- College of Agriculture, Ibaraki University , Ami, Ibaraki 300-0393, Japan
| | - Atsushi Toyoda
- College of Agriculture, Ibaraki University , Ami, Ibaraki 300-0393, Japan.,Ibaraki University Cooperation between Agriculture and Medical Science (IUCAM) , Ami, Ibaraki 300-0393, Japan.,United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology , Fuchu-city, Tokyo 183-8509, Japan
| | - Yoshiharu Takayama
- Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO) , Tsukuba, Ibaraki 305-0901, Japan
| | - Chise Suzuki
- Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO) , Tsukuba, Ibaraki 305-0901, Japan
| |
Collapse
|
28
|
IL-10-producing CD4(+) T cells negatively regulate fucosylation of epithelial cells in the gut. Sci Rep 2015; 5:15918. [PMID: 26522513 PMCID: PMC4629126 DOI: 10.1038/srep15918] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 10/01/2015] [Indexed: 12/20/2022] Open
Abstract
Fucosylated glycans on the surface of epithelial cells (ECs) regulate intestinal homeostasis by serving as attachment receptors and a nutrient source for some species of bacteria. We show here that epithelial fucosylation in the ileum is negatively regulated by IL-10-producing CD4+ T cells. The number of fucosylated ECs was increased in the ileum of mice lacking T cells, especially those expressing αβ T cell receptor (TCR), CD4, and IL-10. No such effect was observed in mice lacking B cells. Adoptive transfer of αβTCR+ CD4+ T cells from normal mice, but not IL-10-deficient mice, normalized fucosylation of ECs. These findings suggest that IL-10-producing CD4+ T cells contribute to the maintenance of the function of ECs by regulating their fucosylation.
Collapse
|
29
|
Wang M, Gao Z, Zhang Z, Pan L, Zhang Y. Roles of M cells in infection and mucosal vaccines. Hum Vaccin Immunother 2015; 10:3544-51. [PMID: 25483705 DOI: 10.4161/hv.36174] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The mucosal immune system plays a crucial part in the control of infection. Exposure of humans and animals to potential pathogens generally occurs through mucosal surfaces, thus, strategies that target the mucosa seem rational and efficient vaccination measures. Vaccination through the mucosal immune system can induce effective systemic immune responses simultaneously with mucosal immunity compared with parenteral vaccination. M cells are capable of transporting luminal antigens to the underlying lymphoid tissues and can be exploited by pathogens as an entry portal to invade the host. Therefore, targeting M-cell-specific molecules might enhance antigen entry, initiate the immune response, and induce protection against mucosal pathogens. Here, we outline our understanding of the distribution and function of M cells, and summarize the advances in mucosal vaccine strategies that target M cells.
Collapse
Key Words
- ANX, Annexin; BALT, bronchus-associated lymphoid tissue
- C5aR, C5a receptor
- DCs, dendritic cells
- DENV, dengue virus
- EDIII, envelope domain III
- FAE, follicle-associated epithelium
- GALT, gut-associated lymphoid tissue
- GENALT, genital-associated lymphoid tissue
- GP2, Glycoprotein 2
- Hsp60, heat shock protein 60
- LPS, lipopolysaccharide
- M cells
- M cells, microfold cells
- MALT, mucosa-associated lymphoid tissue
- NALT, nasopharynx- or nose-associated lymphoid tissue
- OVA, ovalbumin
- OmpH, outer membrane protein H
- PP, Peyer's patches
- PRRs, pathogen recognition receptors
- PrPC, cellular prion protein
- SELEX, Systematic Evolution of Ligands by EXponential enrichment
- SIgA secretory IgA
- TLR-4, Toll-like receptor-4
- UEA-1,Ulex europaeus agglutinin-1
- antigen
- infection
- mucosal immunity
- pσ1, reovirus surface protein σ1
- vaccine
Collapse
Affiliation(s)
- Miao Wang
- a State Key Laboratory of Veterinary Etiological Biology; National Foot-and-Mouse Disease Reference Laboratory; Lanzhou Veterinary Research Institute; CAAS ; Lanzhou , Gansu , China
| | | | | | | | | |
Collapse
|
30
|
Pickard JM, Chervonsky AV. Intestinal fucose as a mediator of host-microbe symbiosis. THE JOURNAL OF IMMUNOLOGY 2015; 194:5588-93. [PMID: 26048966 DOI: 10.4049/jimmunol.1500395] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Fucose is an L-configuration sugar found abundantly in the mammalian gut. It has long been known to be induced there by the presence of bacteria, but only recently have some of the molecular mechanisms behind this process been uncovered. New work suggests that fucose can have a protective role in both gut-centered and systemic infection and inflammation. This review highlights recent studies showing that, in addition to acting as a food source for beneficial gut symbionts, host fucose can suppress the virulence of pathogens and pathobionts. The relevance of gut fucosylation to human diseases also is discussed.
Collapse
Affiliation(s)
- Joseph M Pickard
- Department of Pathology and Committee on Immunology, The University of Chicago, Chicago, IL 60637
| | - Alexander V Chervonsky
- Department of Pathology and Committee on Immunology, The University of Chicago, Chicago, IL 60637
| |
Collapse
|
31
|
Abstract
Blood group antigens represent polymorphic traits inherited among individuals and populations. At present, there are 34 recognized human blood groups and hundreds of individual blood group antigens and alleles. Differences in blood group antigen expression can increase or decrease host susceptibility to many infections. Blood groups can play a direct role in infection by serving as receptors and/or coreceptors for microorganisms, parasites, and viruses. In addition, many blood group antigens facilitate intracellular uptake, signal transduction, or adhesion through the organization of membrane microdomains. Several blood groups can modify the innate immune response to infection. Several distinct phenotypes associated with increased host resistance to malaria are overrepresented in populations living in areas where malaria is endemic, as a result of evolutionary pressures. Microorganisms can also stimulate antibodies against blood group antigens, including ABO, T, and Kell. Finally, there is a symbiotic relationship between blood group expression and maturation of the gastrointestinal microbiome.
Collapse
Affiliation(s)
- Laura Cooling
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
32
|
Goto Y, Obata T, Kunisawa J, Sato S, Ivanov II, Lamichhane A, Takeyama N, Kamioka M, Sakamoto M, Matsuki T, Setoyama H, Imaoka A, Uematsu S, Akira S, Domino SE, Kulig P, Becher B, Renauld JC, Sasakawa C, Umesaki Y, Benno Y, Kiyono H. Innate lymphoid cells regulate intestinal epithelial cell glycosylation. Science 2014; 345:1254009. [PMID: 25214634 PMCID: PMC4774895 DOI: 10.1126/science.1254009] [Citation(s) in RCA: 409] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Fucosylation of intestinal epithelial cells, catalyzed by fucosyltransferase 2 (Fut2), is a major glycosylation mechanism of host-microbiota symbiosis. Commensal bacteria induce epithelial fucosylation, and epithelial fucose is used as a dietary carbohydrate by many of these bacteria. However, the molecular and cellular mechanisms that regulate the induction of epithelial fucosylation are unknown. Here, we show that type 3 innate lymphoid cells (ILC3) induced intestinal epithelial Fut2 expression and fucosylation in mice. This induction required the cytokines interleukin-22 and lymphotoxin in a commensal bacteria-dependent and -independent manner, respectively. Disruption of intestinal fucosylation led to increased susceptibility to infection by Salmonella typhimurium. Our data reveal a role for ILC3 in shaping the gut microenvironment through the regulation of epithelial glycosylation.
Collapse
Affiliation(s)
- Yoshiyuki Goto
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan. Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan. Microbe Division/Japan Collection of Microorganisms, RIKEN BioResource Center, Tsukuba 305-0074, Japan
| | - Takashi Obata
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan. Microbe Division/Japan Collection of Microorganisms, RIKEN BioResource Center, Tsukuba 305-0074, Japan
| | - Jun Kunisawa
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan. Laboratory of Vaccine Materials, National Institute of Biomedical Innovation, Osaka 567-0085, Japan. Division of Mucosal Immunology, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Shintaro Sato
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan. Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Ivaylo I Ivanov
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Aayam Lamichhane
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Natsumi Takeyama
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan. Nippon Institute for Biological Science, Tokyo 198-0024, Japan
| | - Mariko Kamioka
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Mitsuo Sakamoto
- Microbe Division/Japan Collection of Microorganisms, RIKEN BioResource Center, Tsukuba 305-0074, Japan
| | | | | | | | - Satoshi Uematsu
- Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan. Department of Mucosal Immunology, School of Medicine, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba, 260-8670, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, WPI Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Steven E Domino
- Department of Obstetrics and Gynecology, Cellular and Molecular Biology Program, University of Michigan Medical Center, Ann Arbor, MI 48109-5617, USA
| | - Paulina Kulig
- Institute of Experimental Immunology, University of Zürich, Winterthurerstrasse 190, Zürich CH-8057, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zürich, Winterthurerstrasse 190, Zürich CH-8057, Switzerland
| | - Jean-Christophe Renauld
- Ludwig Institute for Cancer Research and Université Catholique de Louvain, Brussels B-1200, Belgium
| | - Chihiro Sasakawa
- Nippon Institute for Biological Science, Tokyo 198-0024, Japan. Division of Bacterial Infection, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan. Medical Mycology Research Center, Chiba University, Chiba 260-8673, Japan
| | | | - Yoshimi Benno
- Benno Laboratory, Innovation Center, RIKEN, Wako, Saitama 351-0198, Japan
| | - Hiroshi Kiyono
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan. Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan. Division of Mucosal Immunology, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| |
Collapse
|
33
|
Exploring the influence of the gut microbiota and probiotics on health: a symposium report. Br J Nutr 2014; 112 Suppl 1:S1-18. [PMID: 24953670 PMCID: PMC4077244 DOI: 10.1017/s0007114514001275] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The present report describes the presentations delivered at the 7th International Yakult Symposium, ‘The Intestinal Microbiota and Probiotics: Exploiting Their Influence on Health’, in London on 22–23 April 2013. The following two themes associated with health risks were covered: (1) the impact of age and diet on the gut microbiota and (2) the gut microbiota's interaction with the host. The strong influence of the maternal gut microbiota on neonatal colonisation was reported, as well as rapid changes in the gut microbiome of older people who move from community living to residential care. The effects of dietary changes on gut metabolism were described and the potential influence of inter-individual microbiota differences was noted, in particular the presence/absence of keystone species involved in butyrate metabolism. Several speakers highlighted the association between certain metabolic disorders and imbalanced or less diverse microbiota. Data from metagenomic analyses and novel techniques (including an ex vivo human mucosa model) provided new insights into the microbiota's influence on coeliac, obesity-related and inflammatory diseases, as well as the potential of probiotics. Akkermansia muciniphila and Faecalibacterium prausnitzii were suggested as targets for intervention. Host–microbiota interactions were explored in the context of gut barrier function, pathogenic bacteria recognition, and the ability of the immune system to induce either tolerogenic or inflammatory responses. There was speculation that the gut microbiota should be considered a separate organ, and whether analysis of an individual's microbiota could be useful in identifying their disease risk and/or therapy; however, more research is needed into specific diseases, different population groups and microbial interventions including probiotics.
Collapse
|
34
|
Abstract
Dendritic cells (DCs) are key modulators that shape the immune system. In mucosal tissues, DCs act as surveillance systems to sense infection and also function as professional antigen-presenting cells that stimulate the differentiation of naive T and B cells. On the basis of their molecular expression, DCs can be divided into several subsets with unique functions. In this review, we focus on intestinal DC subsets and their function in bridging the innate signaling and adaptive immune systems to maintain the homeostasis of the intestinal immune environment. We also review the current strategies for manipulating mucosal DCs for the development of efficient mucosal vaccines to protect against infectious diseases.
Collapse
|
35
|
Isozaki T, Ruth JH, Amin MA, Campbell PL, Tsou PS, Ha CM, Haines GK, Edhayan G, Koch AE. Fucosyltransferase 1 mediates angiogenesis, cell adhesion and rheumatoid arthritis synovial tissue fibroblast proliferation. Arthritis Res Ther 2014; 16:R28. [PMID: 24467809 PMCID: PMC3978694 DOI: 10.1186/ar4456] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Accepted: 01/13/2014] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION We previously reported that sialyl Lewis(y), synthesized by fucosyltransferases, is involved in angiogenesis. Fucosyltransferase 1 (fut1) is an α(1,2)-fucosyltransferase responsible for synthesis of the H blood group and Lewis(y) antigens. However, the angiogenic involvement of fut 1 in the pathogenesis of rheumatoid arthritis synovial tissue (RA ST) has not been clearly defined. METHODS Assay of α(1,2)-linked fucosylated proteins in RA was performed by enzyme-linked lectin assay. Fut1 expression was determined in RA ST samples by immunohistological staining. We performed angiogenic Matrigel assays using a co-culture system of human dermal microvascular endothelial cells (HMVECs) and fut1 small interfering RNA (siRNA) transfected RA synovial fibroblasts. To determine if fut1 played a role in leukocyte retention and cell proliferation in the RA synovium, myeloid THP-1 cell adhesion assays and fut1 siRNA transfected RA synovial fibroblast proliferation assays were performed. RESULTS Total α(1,2)-linked fucosylated proteins in RA ST were significantly higher compared to normal (NL) ST. Fut1 expression on RA ST lining cells positively correlated with ST inflammation. HMVECs from a co-culture system with fut1 siRNA transfected RA synovial fibroblasts exhibited decreased endothelial cell tube formation compared to control siRNA transfected RA synovial fibroblasts. Fut1 siRNA also inhibited myeloid THP-1 adhesion to RA synovial fibroblasts and RA synovial fibroblast proliferation. CONCLUSIONS These data show that α(1,2)-linked fucosylated proteins are upregulated in RA ST compared to NL ST. We also show that fut1 in RA synovial fibroblasts is important in angiogenesis, leukocyte-synovial fibroblast adhesion, and synovial fibroblast proliferation, all key processes in the pathogenesis of RA.
Collapse
Affiliation(s)
- Takeo Isozaki
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Currently Department of Internal Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Jeffrey H Ruth
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Mohammad A Amin
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Phillip L Campbell
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Pei-Suen Tsou
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Christine M Ha
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Gautam Edhayan
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Alisa E Koch
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- VA Medical Service, Department of Veterans Affairs Medical Center, Ann Arbor, MI 48108, USA
| |
Collapse
|
36
|
The intestinal microbiota interferes with the microRNA response upon oral Listeria infection. mBio 2013; 4:e00707-13. [PMID: 24327339 PMCID: PMC3870255 DOI: 10.1128/mbio.00707-13] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The intestinal tract is the largest reservoir of microbes in the human body. The intestinal microbiota is thought to be able to modulate alterations of the gut induced by enteropathogens, thereby maintaining homeostasis. Listeria monocytogenes is the agent of listeriosis, an infection transmitted to humans upon ingestion of contaminated food. Crossing of the intestinal barrier is a critical step of the infection before dissemination into deeper organs. Here, we investigated the role of the intestinal microbiota in the regulation of host protein-coding genes and microRNA (miRNA or miR) expression during Listeria infection. We first established the intestinal miRNA signatures corresponding to the 10 most highly expressed miRNAs in the murine ileum of conventional and germfree mice, noninfected and infected with Listeria. Next, we identified 6 miRNAs whose expression decreased upon Listeria infection in conventional mice. Strikingly, five of these miRNA expression variations (in miR-143, miR-148a, miR-200b, miR-200c, and miR-378) were dependent on the presence of the microbiota. In addition, as is already known, protein-coding genes were highly affected by infection in both conventional and germfree mice. By crossing bioinformatically the predicted targets of the miRNAs to our whole-genome transcriptomic data, we revealed an miRNA-mRNA network that suggested miRNA-mediated global regulation during intestinal infection. Other recent studies have revealed an miRNA response to either bacterial pathogens or commensal bacteria. In contrast, our work provides an unprecedented insight into the impact of the intestinal microbiota on host transcriptional reprogramming during infection by a human pathogen. While the crucial role of miRNAs in regulating the host response to bacterial infection is increasingly recognized, the involvement of the intestinal microbiota in the regulation of miRNA expression has not been explored in detail. Here, we investigated the impact of the intestinal microbiota on the regulation of protein-coding genes and miRNA expression in a host infected by L. monocytogenes, a food-borne pathogen. We show that the microbiota interferes with the microRNA response upon oral Listeria infection and identify several protein-coding target genes whose expression correlates inversely with that of the miRNA. Further investigations of the regulatory networks involving miR-143, miR-148a, miR-200b, miR-200c, and miR-378 will provide new insights into the impact of the intestinal microbiota on the host upon bacterial infection.
Collapse
|
37
|
Kurashima Y, Goto Y, Kiyono H. Mucosal innate immune cells regulate both gut homeostasis and intestinal inflammation. Eur J Immunol 2013; 43:3108-15. [PMID: 24414823 DOI: 10.1002/eji.201343782] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 09/25/2013] [Accepted: 09/30/2013] [Indexed: 12/12/2022]
Abstract
Continuous exposure of intestinal mucosal surfaces to diverse microorganisms and their metabolites reflects the biological necessity for a multifaceted, integrated epithelial and immune cell-mediated regulatory system. The development and function of the host cells responsible for the barrier function of the intestinal surface (e.g., M cells, Paneth cells, goblet cells, and columnar epithelial cells) are strictly regulated through both positive and negative stimulation by the luminal microbiota. Stimulation by damage-associated molecular patterns and commensal bacteria-derived microbe-associated molecular patterns provokes the assembly of inflammasomes, which are involved in maintaining the integrity of the intestinal epithelium. Mucosal immune cells located beneath the epithelium play critical roles in regulating both the mucosal barrier and the relative composition of the luminal microbiota. Innate lymphoid cells and mast cells, in particular, orchestrate the mucosal regulatory system to create a mutually beneficial environment for both the host and the microbiota. Disruption of mucosal homeostasis causes intestinal inflammation such as that seen in inflammatory bowel disease. Here, we review the recent research on the biological interplay among the luminal microbiota, epithelial cells, and mucosal innate immune cells in both healthy and pathological conditions.
Collapse
Affiliation(s)
- Yosuke Kurashima
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo, Japan; Division of Infectious Genetics, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | | |
Collapse
|
38
|
Kashyap PC, Marcobal A, Ursell LK, Smits SA, Sonnenburg ED, Costello EK, Higginbottom SK, Domino SE, Holmes SP, Relman DA, Knight R, Gordon JI, Sonnenburg JL. Genetically dictated change in host mucus carbohydrate landscape exerts a diet-dependent effect on the gut microbiota. Proc Natl Acad Sci U S A 2013; 110:17059-64. [PMID: 24062455 PMCID: PMC3800993 DOI: 10.1073/pnas.1306070110] [Citation(s) in RCA: 199] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
We investigate how host mucus glycan composition interacts with dietary carbohydrate content to influence the composition and expressed functions of a human gut community. The humanized gnotobiotic mice mimic humans with a nonsecretor phenotype due to knockout of their α1-2 fucosyltransferase (Fut2) gene. The fecal microbiota of Fut2(-) mice that lack fucosylated host glycans show decreased alpha diversity relative to Fut2(+) mice and exhibit significant differences in community composition. A glucose-rich plant polysaccharide-deficient (PD) diet exerted a strong effect on the microbiota membership but eliminated the effect of Fut2 genotype. Additionally fecal metabolites predicted host genotype in mice on a polysaccharide-rich standard diet but not on a PD diet. A more detailed mechanistic analysis of these interactions involved colonization of gnotobiotic Fut2(+) and Fut2(-) mice with Bacteroides thetaiotaomicron, a prominent member of the human gut microbiota known to adaptively forage host mucosal glycans when dietary polysaccharides are absent. Within Fut2(-) mice, the B. thetaiotaomicron fucose catabolic pathway was markedly down-regulated, whereas BT4241-4247, an operon responsive to terminal β-galactose, the precursor that accumulates in the Fut2(-) mice, was significantly up-regulated. These changes in B. thetaiotaomicron gene expression were only evident in mice fed a PD diet, wherein B. thetaiotaomicron relies on host mucus consumption. Furthermore, up-regulation of the BT4241-4247 operon was also seen in humanized Fut2(-) mice. Together, these data demonstrate that differences in host genotype that affect the carbohydrate landscape of the distal gut interact with diet to alter the composition and function of resident microbes in a diet-dependent manner.
Collapse
Affiliation(s)
- Purna C. Kashyap
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Angela Marcobal
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Luke K. Ursell
- Department of Chemistry and Biochemistry, Howard Hughes Medical Institute, University of Colorado, Boulder, CO 80309
| | - Samuel A. Smits
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Erica D. Sonnenburg
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Elizabeth K. Costello
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Steven K. Higginbottom
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Steven E. Domino
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109
| | - Susan P. Holmes
- Department of Statistics, Stanford University, Stanford, CA 94305
| | - David A. Relman
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304; and
| | - Rob Knight
- Department of Chemistry and Biochemistry, Howard Hughes Medical Institute, University of Colorado, Boulder, CO 80309
| | - Jeffrey I. Gordon
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63108
| | - Justin L. Sonnenburg
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
39
|
Casteleyn C, Van den Broeck W, Gebert A, Tambuyzer BR, Van Cruchten S, Van Ginneken C. M cell specific markers in man and domestic animals: Valuable tools in vaccine development. Comp Immunol Microbiol Infect Dis 2013; 36:353-64. [DOI: 10.1016/j.cimid.2013.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 03/01/2013] [Accepted: 03/21/2013] [Indexed: 12/13/2022]
|
40
|
Sato S, Kaneto S, Shibata N, Takahashi Y, Okura H, Yuki Y, Kunisawa J, Kiyono H. Transcription factor Spi-B-dependent and -independent pathways for the development of Peyer's patch M cells. Mucosal Immunol 2013; 6:838-46. [PMID: 23212199 DOI: 10.1038/mi.2012.122] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Although many of the biological features of microfold cells (M cells) have been known for many years, the molecular mechanisms of M-cell development and antigen recognition have remained unclear. Here, we report that Umod is a novel M-cell-specific gene, the translation products of which might contribute to the uptake function of M cells. Transcription factor Spi-B was also specifically expressed in M cells among non-hematopoietic lineages. Spi-B-deficient mice showed reduced expression of most, but not all, other M-cell-specific genes and M-cell surface markers. Whereas uptake of Salmonella Typhimurium via M cells was obviously reduced in Spi-B-deficient mice, the abundance of intratissue cohabiting bacteria was comparable between wild-type and Spi-B-deficient mice. These data indicate that there is a small M-cell population with developmental regulation that is Spi-B independent; however, Spi-B is probably a candidate master regulator of M-cell functional maturation and development by another pathway.
Collapse
Affiliation(s)
- S Sato
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Orr SL, Le D, Long JM, Sobieszczuk P, Ma B, Tian H, Fang X, Paulson JC, Marth JD, Varki N. A phenotype survey of 36 mutant mouse strains with gene-targeted defects in glycosyltransferases or glycan-binding proteins. Glycobiology 2012; 23:363-80. [PMID: 23118208 DOI: 10.1093/glycob/cws150] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The consortium for functional glycomics (CFG) was a large research initiative providing networking and resources for investigators studying the role of glycans and glycan-binding proteins in health and disease. Starting in 2001, six scientific cores were established to generate data, materials and new technologies. By the end of funding in 2011, the mouse phenotype core (MPC) submitted data to a website from the phenotype screen of 36 mutant mouse strains deficient in a gene for either a glycan-binding protein (GBP) or glycosyltransferase (GT). Each mutant strain was allotted three months for analysis and screened by standard phenotype assays used in the fields of immunology, histology, hematology, coagulation, serum chemistry, metabolism and behavior. Twenty of the deficient mouse strains had been studied in other laboratories, and additional tests were performed on these strains to confirm previous observations and discover new data. The CFG constructed 16 new homozygous mutant mouse strains and completed the initial phenotype screen of the majority of these new mutant strains. In total, >300 phenotype changes were observed, but considering the over 100 assays performed on each strain, most of the phenotypes were unchanged. Phenotype differences include abnormal testis morphology in GlcNAcT9- and Siglec-H-deficient mice and lethality in Pomgnt1-deficient mice. The numerous altered phenotypes discovered, along with the consideration of the significant findings of normality, will provide a platform for future characterization to understand the important roles of glycans and GBPs in the mechanisms of health and disease.
Collapse
Affiliation(s)
- Sally L Orr
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093-0687, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kobayashi A, Donaldson DS, Kanaya T, Fukuda S, Baillie JK, Freeman TC, Ohno H, Williams IR, Mabbott NA. Identification of novel genes selectively expressed in the follicle-associated epithelium from the meta-analysis of transcriptomics data from multiple mouse cell and tissue populations. DNA Res 2012; 19:407-22. [PMID: 22991451 PMCID: PMC3473373 DOI: 10.1093/dnares/dss022] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 08/16/2012] [Indexed: 01/09/2023] Open
Abstract
The follicle-associated epithelium (FAE) overlying the Peyer's patches and the microfold cells (M cells) within it are important sites of antigen transcytosis across the intestinal epithelium. Using a meta-analysis approach, we identified a transcriptional signature that distinguished the FAE from a large collection of mouse cells and tissues. A co-expressed cluster of 21 FAE-specific genes was identified, and the analysis of the transcription factor binding site motifs in their promoter regions indicated that these genes shared an underlying transcriptional programme. This cluster contained known FAE- (Anxa10, Ccl20, Psg18 and Ubd) and M-cell-specific (Gp2) genes, suggesting that the others were novel FAE-specific genes. Some of these novel candidate genes were expressed highly by the FAE and M cells (Calcb, Ces3b, Clca2 and Gjb2), and others only by the FAE (Ascl2, Cftr, Fgf15, Gpr133, Kcna1, Kcnj15, Mycl1, Pgap1 and Rps6kl). We also identified a subset of novel FAE-related genes that were induced in the intestinal epithelium after receptor activator of nuclear factor (NF)-κB ligand stimulation. These included Mfge8 which was specific to FAE enterocytes. This study provides new insight into the FAE transcriptome. Further characterization of the candidate genes identified here will aid the identification of novel regulators of cell function in the FAE.
Collapse
Affiliation(s)
- Atsushi Kobayashi
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
- Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - David S. Donaldson
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Takashi Kanaya
- Research Center for Allergy and Immunology (RCAI), RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama 230-0045, Japan
| | - Shinji Fukuda
- Research Center for Allergy and Immunology (RCAI), RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama 230-0045, Japan
| | - J. Kenneth Baillie
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Tom C. Freeman
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Hiroshi Ohno
- Research Center for Allergy and Immunology (RCAI), RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama 230-0045, Japan
| | - Ifor R. Williams
- Department of Pathology, Emory University School of Medicine, Whitehead Bldg. 105D, 615 Michael St., Atlanta, GA 30322, USA
| | - Neil A. Mabbott
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| |
Collapse
|
43
|
Kunisawa J, Kurashima Y, Kiyono H. Gut-associated lymphoid tissues for the development of oral vaccines. Adv Drug Deliv Rev 2012; 64:523-30. [PMID: 21827802 DOI: 10.1016/j.addr.2011.07.003] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 06/23/2011] [Accepted: 07/10/2011] [Indexed: 01/22/2023]
Abstract
Oral vaccine has been considered to be a prospective vaccine against many pathogens especially invading across gastrointestinal tracts. One key element of oral vaccine is targeting efficient delivery of antigen to gut-associated lymphoid tissue (GALT), the inductive site in the intestine where antigen-specific immune responses are initiated. Various chemical and biological antigen delivery systems have been developed and some are in clinical trials. In this review, we describe the immunological features of GALT and the current status of antigen delivery system candidates for successful oral vaccine.
Collapse
Affiliation(s)
- Jun Kunisawa
- Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Japan.
| | | | | |
Collapse
|
44
|
Goto Y, Kiyono H. Epithelial barrier: an interface for the cross-communication between gut flora and immune system. Immunol Rev 2012; 245:147-63. [PMID: 22168418 DOI: 10.1111/j.1600-065x.2011.01078.x] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Large numbers of environmental antigens, including commensal bacteria and food-derived antigens, constitutively interact with the epithelial layer of the gastrointestinal (GI) tract. Commensal bacteria peacefully cohabit with the host GI tract and exert multiple beneficial or destructive effects on their host. Intestinal epithelial cells (IECs) constitute the first physical and immunological protective wall against invasive pathogens and a cohabitation niche for commensal bacteria. As the physiological homeostasis of IECs is maintained by multiple biological processes such as apoptosis, autophagy, and the handling of endoplasmic reticulum stress, the aberrant kinetics of these biological events, which have genetic and environmental causes, leads to the development of host intestinal pathogenesis such as inflammatory bowel disease. In addition, IECs recognize and interact with commensal bacteria and give instructions to mucosal immune cells to initiate an immunological balance between active and quiescent conditions, eventually establishing intestinal homeostasis. The mucosal immune system regulates the homeostasis of gut microbiota by producing immunological molecules such as secretory immunoglobulin A, the production of which is mediated by IECs. IECs therefore play a central role in the creation and maintenance of a physiologically and immunologically stable intestinal environment.
Collapse
Affiliation(s)
- Yoshiyuki Goto
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
45
|
Wang J, Gusti V, Saraswati A, Lo DD. Convergent and divergent development among M cell lineages in mouse mucosal epithelium. THE JOURNAL OF IMMUNOLOGY 2011; 187:5277-85. [PMID: 21984701 DOI: 10.4049/jimmunol.1102077] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
M cells are specialized epithelial cells mediating immune surveillance of the mucosal lumen by transepithelial delivery of Ags to underlying dendritic cells (DC). At least three M cell phenotypes are known in the airways and intestine, but their developmental relationships are unclear. We used reporter transgenic mouse strains to follow the constitutive development of M cell subsets and their acute induction by cholera toxin (CT). M cells overlying intestinal Peyer's patches (PPs), isolated lymphoid follicles, and nasal-associated lymphoid tissue are induced by distinct settings, yet show convergent phenotypes, such as expression of a peptidoglycan recognition protein-S (PGRP-S) transgene reporter. By contrast, though PP, isolated lymphoid follicle, and villous M cells are all derived from intestinal crypt stem cells, their phenotypes were clearly distinct; for example, PP M cells frequently appeared to form M cell-DC functional units, whereas villous M cells did not consistently engage underlying DC. B lymphocytes are critical to M cell function by forming a basolateral pocket and possible signaling through CD137; however, initial commitment to all M cell lineages is B lymphocyte and CD137 independent. CT causes induction of new M cells in the airway and intestine without cell division, suggesting transdifferentiation from mature epithelial cells. In contrast with intestinal PP M cells, CT-induced nasal-associated lymphoid tissue M cells appear to be generated from ciliated Foxj1(+)PGRP-S(+) cells, indicative of a possible precommitted progenitor. In summary, constitutive and inducible differentiation of M cells is toward strictly defined context-dependent phenotypes, suggesting specialized roles in surveillance of mucosal Ags.
Collapse
Affiliation(s)
- Jing Wang
- Division of Biomedical Sciences, University of California Riverside, Riverside, CA 92521, USA
| | | | | | | |
Collapse
|