1
|
Meng X, Zheng X, Mai W, Gao J, Fan Y, Fu J, Xu J. Micro- and nanoplastics differ in particle-mucus interactions: The sight on rheological properties, barrier dysfunction and microbiota dysbiosis. JOURNAL OF HAZARDOUS MATERIALS 2025; 492:138130. [PMID: 40220393 DOI: 10.1016/j.jhazmat.2025.138130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/14/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025]
Abstract
Micro- and nanoplastics (MNPs) in food can cross the intestinal barrier and accumulate in multiple organs. Mucus serves as a vital defense against such invaders, but the nature of its interaction with MNPs remains unclear. In this study, we investigated changes in the rheological properties of mucus and the physicochemical properties of MNPs in co-incubation. The effects of MNPs on the mucus layer and gut microbiota were also assessed in vivo at environmentally relevant doses. MNPs adsorbed proteins in mucus, increasing apparent particle size, and reducing the surface charges. They broke the selective permeability of barrier and destroyed the histomorphology and microenvironment of microbiota in mice. Notably, nanoplastics were wrapped in mucus. They induced mucus secretion, crosstalk of microbiota, and reactive oxygen species (ROS) burst. Microplastics reduced the composite viscosity of mucus and thinned the mucus layer, facilitating diversification of harmful bacteria. Size plays a crucial role in particle-mucus interactions: nanoplastics tend to penetrate the mucus layer and disrupt microbial colonization, while microplastics contribute to mucus depletion. The physicochemical properties of MNPs and mucus characteristics affect microbial community, modulating the MNPs biotoxicity. These findings provide insights into mucus barrier homeostasis in health risk of MNPs.
Collapse
Affiliation(s)
- Xuemei Meng
- School of Food Science and Engineering, Ningxia University, Ningxia, Yinchuan 750021, PR China
| | - Xue Zheng
- School of Food Science and Engineering, Ningxia University, Ningxia, Yinchuan 750021, PR China
| | - Wenting Mai
- School of Food Science and Engineering, Ningxia University, Ningxia, Yinchuan 750021, PR China
| | - Jianying Gao
- School of Food Science and Engineering, Ningxia University, Ningxia, Yinchuan 750021, PR China
| | - Yanli Fan
- School of Food Science and Engineering, Ningxia University, Ningxia, Yinchuan 750021, PR China
| | - Jing Fu
- School of Food Science and Engineering, Ningxia University, Ningxia, Yinchuan 750021, PR China
| | - Junnan Xu
- School of Food Science and Engineering, Ningxia University, Ningxia, Yinchuan 750021, PR China.
| |
Collapse
|
2
|
Gustafsson JK, Hansson GC. Immune Regulation of Goblet Cell and Mucus Functions in Health and Disease. Annu Rev Immunol 2025; 43:169-189. [PMID: 39752567 DOI: 10.1146/annurev-immunol-101721-065224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
The mucosal surfaces of the body are the most vulnerable points for infection because they are lined by single or multiple layers of very active epithelial cells. The main protector of these cells is the mucus system generated by the specialized goblet cell secreting its main components, the gel-forming mucins. The organization of the mucus varies from an attached mucus that is impenetrable to bacteria in the large intestine to a nonattached, more penetrable mucus in the small intestine. The respiratory tract mucus system clears particles and microorganisms from healthy lungs but causes disease if reorganized to an attached mucus that cannot be efficiently transported. Similarly, transformation of large intestine mucus from impenetrable to penetrable causes chronic inflammation directed toward the intestinal microbiota. Mucus-producing goblet cells are regulated by and responsive to signals from immune cells, and at the same time signal back to the immune system. In this review we focus on the relationship of immune cells with intestinal goblet cells and mucus, making parallels to the respiratory tract.
Collapse
Affiliation(s)
| | - Gunnar C Hansson
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden;
| |
Collapse
|
3
|
Wendo WD, Thongrin T, Tangkawattana P, Sukon P, Suttiprapa S, Saichua P, Suyapoh W, Tangkawattana S. Goblet cell metaplasia and mucin alterations in biliary epithelial cells during Opisthorchis viverrini infection in rodent models: Insights into host susceptibility and defense mechanisms. Vet World 2025; 18:534-546. [PMID: 40342755 PMCID: PMC12056906 DOI: 10.14202/vetworld.2025.534-546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/23/2025] [Indexed: 05/11/2025] Open
Abstract
Background and Aim Chronic Opisthorchis viverrini (OV) infection induces significant biliary changes and is a major risk factor for cholangiocarcinoma. However, the role of goblet cell metaplasia (GCM) and mucin dynamics in host defense and parasite persistence remains poorly understood. This study aims to characterize biliary histological changes, particularly mucin types, and compare responses between susceptible (hamsters) and non-susceptible (mice) hosts during early to chronic OV infection. Materials and Methods Thirty-five male golden Syrian hamsters and 35 male BALB/c mice were divided into infected and control groups. Infected animals received 50 OV metacercariae through gastric intubation and were sacrificed on days 1, 2, 7, 14, 28, and 56 post-infection. Histological, histochemical (Alcian Blue, periodic Acid-Schiff, and high iron diamine), and immunohistochemical (Bromodeoxyuridine [BrdU]) analyses were performed to assess mucin production, GCM, and bile duct proliferation. Results Mice demonstrated an early, robust biliary response with pronounced hyperplasia and GCM characterized by acid mucin overproduction during the acute phase (days 1-28). Conversely, hamsters exhibited delayed biliary proliferation and GCM, with predominant sulfated mucins appearing during the chronic phase (days 28-56). BrdU immunoreactivity indicated earlier and stronger bile duct epithelial proliferation in mice, correlating with worm clearance by day 28. In hamsters, mucosal changes supported worm survival, as evidenced by continued parasite presence and egg production. Statistical analyses confirmed significant differences in mucin types and hyperplasia between species across infection stages. Conclusion Distinct mucosal responses in hamsters and mice reflect their susceptibility to OV infection. Acid mucins in mice facilitate worm expulsion, while sulfated mucins in hamsters appear to promote parasite persistence. These findings highlight the dual roles of mucins in host defense and parasite survival, providing insight into mechanisms underlying susceptibility and resistance in OV infections.
Collapse
Affiliation(s)
- Woro Danur Wendo
- Graduate School, Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Theerayut Thongrin
- Graduate School, Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Prasarn Tangkawattana
- Department of Anatomy, Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Peerapol Sukon
- Department of Anatomy, Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Sutas Suttiprapa
- Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Prasert Saichua
- Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Watcharapol Suyapoh
- Department of Veterinary Science, Faculty of Veterinary Science, Prince of Songkla University, Songkhla, 90110, Thailand
| | - Sirikachorn Tangkawattana
- Department of Pathobiology, Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| |
Collapse
|
4
|
Cheng H, Li H, Zhao Y, Yang K, Wang J, Tan B, Ma X. Transcriptome analysis reveals modulations in glycosylation profiles of the mucosal barrier and their potential interaction with gut microbiota in weaned piglets. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 20:226-238. [PMID: 39990197 PMCID: PMC11846933 DOI: 10.1016/j.aninu.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/09/2024] [Accepted: 12/02/2024] [Indexed: 02/25/2025]
Abstract
The current study aims to investigate the potential interaction between glycosylation profiles of the Ningxiang breed (NX) and Western Duroc × Landrace × Yorkshire breed (DLY) weaned piglets, and their characteristic microbes, employing integrated analyses of transcriptomics and metagenomics. Twenty-four (12 NX and 12 DLY) at 28 days of age were transported into an experimental house and fed the same weaned piglet diet. The trail period was 7 days. Results revealed that the NX piglets had a higher growth-to-feed ratio, body weight gain scale, and lower pathological score of intestinal injury compared with the DLY piglets (P < 0.01). DLY piglets displayed elevated mRNA expression levels of MUC2 and MUC5AC in colonic mucosal tissue than NX piglets (P < 0.05). Within the O-linked glycosylated differentially expressed genes (DEGs), FNTA, GALNT18, POMGNT1, POMGNT2, and POMT1 were significantly upregulated in DLY piglets relative to NX piglets (P < 0.05). Conversely, C1GALT2, GALNT1, KMT2C, and OGT were significantly downregulated in DLY piglets compared to NX piglets (P < 0.05). The KMT2C gene was hardly expressed in the transcriptome of DLY piglets. At the phylum taxonomic level, NX piglets had a higher abundance of Firmicutes, while DLY piglets had a higher abundance of Proteobacteria. At the genus taxonomic level, NX piglets had a higher abundance of Lactobacillus, whereas DLY piglets had a higher abundance of Collinsella, Enterococcus and Escherichia. The results of the correlation between intestinal differential bacteria and O-chain glycosylated DEG showed that C1GALT2, GALNT1 and KMT2 were associated with Lactobacillus_pontis showed a positive correlation (R = 0.67). Through comparative analysis of differentially glycosylated genes and their associated functions, this study highlights the potential role of reduced expression of GALNT1 and KMT2C genes, involved in O-linked protein and glycan reactions, in impairing the intestinal barrier function of DLY piglets. Furthermore, members of the Lactobacillus and Prevotella genera may actively contribute to the regulation of piglet colon glycosylation profiles.
Collapse
Affiliation(s)
- Hao Cheng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Yuelushan Laboratory, Changsha 410128, China
| | - Hao Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Yuelushan Laboratory, Changsha 410128, China
| | - Yujie Zhao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Yuelushan Laboratory, Changsha 410128, China
| | - Kai Yang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Yuelushan Laboratory, Changsha 410128, China
| | - Jing Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Yuelushan Laboratory, Changsha 410128, China
| | - Bie Tan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Yuelushan Laboratory, Changsha 410128, China
| | - Xiaokang Ma
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Yuelushan Laboratory, Changsha 410128, China
| |
Collapse
|
5
|
Mao H, Liu Y, Lv Q, Li C, Yang Y, Wu F, Xu N, Jin X. The effect of β-Glucan induced intestinal trained immunity against Trichinella spiralis infection. Vet Parasitol 2025; 333:110238. [PMID: 38944590 DOI: 10.1016/j.vetpar.2024.110238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/01/2024]
Abstract
Parasitic helminth Trichinella spiralis (Ts) induce mixed Th1/Th2 response with predominant type 2 immune responses, with protective immunity mediated by interleukin (IL)-4, IL-5, and IL-13. β-Glucan (BG) has been shown to have the ability to induce trained immunity, confers non-specific protection from secondary infections. However, whether BG-induced trained immunity played a role in protective type 2 immunity against Ts infection is unclear. In this study, BG was administered five days before Ts infection to induce trained immunity. Our findings demonstrate that BG pretreatment effectively reduced the number of T. spiralis adults and muscle larvae, whereas inhibition of trained immunity abolished the effect of BG. Additionally, we observed a significant increase in goblet cells and mucus production as evidenced by Alcian blue periodic acid-Schiff staining. Furthermore, quantitative real-time PCR analysis revealed a significant upregulation of IL-4, IL-5, and IL-13 expression in response to BG. Conversely, the inhibitor of trained immunity reversed these effects, suggesting that BG-induced trained immunity confers strong protection against Ts infection. In conclusion, these findings suggest that BG-induced trained immunity may play a role in protection against infections caused by other helminths.
Collapse
Affiliation(s)
- Hanhai Mao
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yi Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Qingbo Lv
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Chengyao Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yaming Yang
- Department of Helminth, Yunnan Institute of Parasitic Diseases, Puer, China
| | - Fangwei Wu
- Department of Helminth, Yunnan Institute of Parasitic Diseases, Puer, China
| | - Ning Xu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Xuemin Jin
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China.
| |
Collapse
|
6
|
Mules TC, Vacca F, Cait A, Yumnam B, Schmidt A, Lavender B, Maclean K, Noble SL, Gasser O, Camberis M, Le Gros G, Inns S. A Small Intestinal Helminth Infection Alters Colonic Mucus and Shapes the Colonic Mucus Microbiome. Int J Mol Sci 2024; 25:12015. [PMID: 39596084 PMCID: PMC11593901 DOI: 10.3390/ijms252212015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Infecting humans with controlled doses of small intestinal helminths, such as human hookworm, is proposed as a therapy for the colonic inflammatory disease ulcerative colitis. Strengthening the colonic mucus barrier is a potential mechanism by which small intestinal helminths could treat ulcerative colitis. In this study, we compare C57BL/6 mice infected with the small intestinal helminth Heligmosomoides polygyrus and uninfected controls to investigate changes in colonic mucus. Histology, gene expression, and immunofluorescent analysis demonstrate that this helminth induces goblet cell hyperplasia, and an upregulation of mucin sialylation, and goblet-cell-derived functional proteins resistin-like molecule-beta (RELM-β) and trefoil factors (TFFs), in the colon. Using IL-13 knockout mice, we reveal that these changes are predominantly IL-13-dependent. The assessment of the colonic mucus microbiome demonstrates that H. polygyrus infection increases the abundance of Ruminococcus gnavus, a commensal bacterium capable of utilising sialic acid as an energy source. This study also investigates a human cohort experimentally challenged with human hookworm. It demonstrates that TFF blood levels increase in individuals chronically infected with small intestinal helminths, highlighting a conserved mucus response between humans and mice. Overall, small intestinal helminths modify colonic mucus, highlighting this as a plausible mechanism by which human hookworm therapy could treat ulcerative colitis.
Collapse
Affiliation(s)
- Thomas C. Mules
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
- Department of Medicine, University of Otago, 23A Mein St., Newtown, Wellington 6242, New Zealand
| | - Francesco Vacca
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
| | - Alissa Cait
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
| | - Bibek Yumnam
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
| | - Alfonso Schmidt
- Hugh Green Technology Centre, Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| | - Brittany Lavender
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
| | - Kate Maclean
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
| | - Sophia-Louise Noble
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
| | - Olivier Gasser
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
| | - Mali Camberis
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
| | - Graham Le Gros
- Malaghan Institute of Medical Research, Kelburn, Wellington 6012, New Zealand
| | - Stephen Inns
- Department of Medicine, University of Otago, 23A Mein St., Newtown, Wellington 6242, New Zealand
| |
Collapse
|
7
|
Mair I, Bennett AR, Forman R, Othman AA, Logunova L, Smith H, Lowe AE, Bradley JE, Thornton DJ, Else KJ. T. Muris Infection Dynamics of a Fresh, Wild Isolate: Is the Established E Isolate Still Relevant? Parasite Immunol 2024; 46:e13072. [PMID: 39480064 DOI: 10.1111/pim.13072] [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: 06/16/2024] [Revised: 09/18/2024] [Accepted: 10/08/2024] [Indexed: 11/02/2024]
Abstract
For decades, parasitic worms such as Trichuris muris have been maintained in laboratory animals, providing insights into host-parasite interactions and host immune responses. The most used T. muris isolate is the E isolate, established in the laboratory in 1954. However, one concern with these model systems is the potential for laboratory-induced selection and therefore changes in host-parasite interactions. To address these concerns, we compare the E isolate with a recently isolated T. muris isolate (M isolate), established from wild house mice (Mus musculus domesticus, Isle of May, UK), in their capacity to infect laboratory mice. High dose infection of C57BL/6 mice revealed that significantly more parasites of the M isolate survived to the adult stage compared to the E isolate. Worm persistence was associated with heightened TNF-α and IL-10 secretion upon parasite-specific re-stimulation, and higher serum IgG1 and IgG2c levels, concomitant with an increase in T-bet+ and ICOS+ CD4+ T effector-memory cells. Differences in host response to the isolates were not as pronounced during low dose infection. Our study highlights the need for regular evaluation of lab-maintained parasite isolates against freshly isolated parasites to understand whether the established lab strains remain relevant model systems for our understanding of parasitic infections.
Collapse
Affiliation(s)
- Iris Mair
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Alexander R Bennett
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
- Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Ruth Forman
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Abdulrazzag A Othman
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Larisa Logunova
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Hannah Smith
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Ann E Lowe
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | | | - David J Thornton
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
- Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Kathryn J Else
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| |
Collapse
|
8
|
Stanforth KJ, Zakhour MI, Chater PI, Wilcox MD, Adamson B, Robson NA, Pearson JP. The MUC2 Gene Product: Polymerisation and Post-Secretory Organisation-Current Models. Polymers (Basel) 2024; 16:1663. [PMID: 38932019 PMCID: PMC11207715 DOI: 10.3390/polym16121663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
MUC2 mucin, the primary gel-forming component of intestinal mucus, is well researched and a model of polymerisation and post-secretory organisation has been published previously. Recently, several significant developments have been made which either introduce new ideas or challenge previous theories. New ideas include an overhaul of the MUC2 C-terminal globular structure which is proposed to harbour several previously unobserved domains, and include a site for an extra intermolecular disulphide bridge dimer between the cysteine 4379 of adjacent MUC2 C-termini. MUC2 polymers are also now thought to be secreted attached to the epithelial surface of goblet cells in the small intestine and removed following secretion via a metalloprotease meprin β-mediated cleavage of the von Willebrand D2 domain of the N-terminus. It remains unclear whether MUC2 forms intermolecular dimers, trimers, or both, at the N-termini during polymerisation, with several articles supporting either trimer or dimer formation. The presence of a firm inner mucus layer in the small intestine is similarly unclear. Considering this recent research, this review proposes an update to the previous model of MUC2 polymerisation and secretion, considers conflicting theories and data, and highlights the importance of this research to the understanding of MUC2 mucus layers in health and disease.
Collapse
Affiliation(s)
- Kyle J. Stanforth
- Aelius Biotech, The Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK; (P.I.C.); (M.D.W.); (B.A.); (N.A.R.)
| | - Maria I. Zakhour
- Biosciences Institute, Newcastle University Biosciences Institute, Catherine Cookson Building, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK; (M.I.Z.); (J.P.P.)
| | - Peter I. Chater
- Aelius Biotech, The Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK; (P.I.C.); (M.D.W.); (B.A.); (N.A.R.)
| | - Matthew D. Wilcox
- Aelius Biotech, The Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK; (P.I.C.); (M.D.W.); (B.A.); (N.A.R.)
| | - Beth Adamson
- Aelius Biotech, The Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK; (P.I.C.); (M.D.W.); (B.A.); (N.A.R.)
| | - Niamh A. Robson
- Aelius Biotech, The Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK; (P.I.C.); (M.D.W.); (B.A.); (N.A.R.)
| | - Jeffrey P. Pearson
- Biosciences Institute, Newcastle University Biosciences Institute, Catherine Cookson Building, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK; (M.I.Z.); (J.P.P.)
| |
Collapse
|
9
|
Bennett AR, Mair I, Muir A, Smith H, Logunova L, Wolfenden A, Fenn J, Lowe AE, Bradley JE, Else KJ, Thornton DJ. Sex drives colonic mucin sialylation in wild mice. Sci Rep 2024; 14:6954. [PMID: 38521809 PMCID: PMC10960830 DOI: 10.1038/s41598-024-57249-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 03/15/2024] [Indexed: 03/25/2024] Open
Abstract
Mucin protein glycosylation is important in determining biological properties of mucus gels, which form protective barriers at mucosal surfaces of the body such as the intestine. Ecological factors including: age, sex, and diet can change mucus barrier properties by modulating mucin glycosylation. However, as our understanding stems from controlled laboratory studies in house mice, the combined influence of ecological factors on mucin glycosylation in real-world contexts remains limited. In this study, we used histological staining with 'Alcian Blue, Periodic Acid, Schiff's' and 'High-Iron diamine' to assess the acidic nature of mucins stored within goblet cells of the intestine, in a wild mouse population (Mus musculus). Using statistical models, we identified sex as among the most influential ecological factors determining the acidity of intestinal mucin glycans in wild mice. Our data from wild mice and experiments using laboratory mice suggest estrogen signalling associates with an increase in the relative abundance of sialylated mucins. Thus, estrogen signalling may underpin sex differences observed in the colonic mucus of wild and laboratory mice. These findings highlight the significant influence of ecological parameters on mucosal barrier sites and the complementary role of wild populations in augmenting standard laboratory studies in the advancement of mucus biology.
Collapse
Affiliation(s)
- Alexander R Bennett
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK.
| | - Iris Mair
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Andrew Muir
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Hannah Smith
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Larisa Logunova
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Andrew Wolfenden
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Jonathan Fenn
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Ann E Lowe
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | | | - Kathryn J Else
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK.
| | - David J Thornton
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK.
| |
Collapse
|
10
|
Mules TC, Inns S, Le Gros G. Helminths' therapeutic potential to treat intestinal barrier dysfunction. Allergy 2023; 78:2892-2905. [PMID: 37449458 DOI: 10.1111/all.15812] [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: 05/16/2023] [Revised: 06/20/2023] [Accepted: 07/02/2023] [Indexed: 07/18/2023]
Abstract
The intestinal barrier is a dynamic multi-layered structure which can adapt to environmental changes within the intestinal lumen. It has the complex task of allowing nutrient absorption while limiting entry of harmful microbes and microbial antigens present in the intestinal lumen. Excessive entry of microbial antigens via microbial translocation due to 'intestinal barrier dysfunction' is hypothesised to contribute to the increasing incidence of allergic, autoimmune and metabolic diseases, a concept referred to as the 'epithelial barrier theory'. Helminths reside in the intestinal tract are in intimate contact with the mucosal surfaces and induce a range of local immunological changes which affect the layers of the intestinal barrier. Helminths are proposed to prevent, or even treat, many of the diseases implicated in the epithelial barrier theory. This review will focus on the effect of helminths on intestinal barrier function and explore whether this could explain the proposed health benefits delivered by helminths.
Collapse
Affiliation(s)
- Thomas C Mules
- Malaghan Institute of Medical Research, Wellington, New Zealand
- University of Otago, Wellington, New Zealand
| | | | - Graham Le Gros
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
11
|
Wang J, Liu X, Sun R, Mao H, Liu M, Jin X. Akkermansia muciniphila participates in the host protection against helminth-induced cardiac fibrosis via TLR2. PLoS Pathog 2023; 19:e1011683. [PMID: 37788279 PMCID: PMC10547169 DOI: 10.1371/journal.ppat.1011683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 09/12/2023] [Indexed: 10/05/2023] Open
Abstract
Helminth Trichinella spiralis (Ts) is one of the major pathogens of human infective myocarditis that can lead to cardiac fibrosis (CF). The gut microbiota involved in this pathology are of interest. Here, we use mice infected with Ts as a model to examine the interactions between gut microbes and host protection to CF. Infected mice show enhanced CF severity. We find that antibiotics treatment to deplete the microbiota aggravates the disease phenotype. Attempts to restore microbiota using fecal microbiota transplantation ameliorates helminth-induced CF. 16S rRNA gene sequencing and metagenomics sequencing reveal a higher abundance of Akkermansia muciniphila in gut microbiomes of Ts-infected mice. Oral supplementation with alive or pasteurized A. muciniphila improves CF via TLR2. This work represents a substantial advance toward our understanding of causative rather than correlative relationships between the gut microbiota and CF.
Collapse
Affiliation(s)
- Jiaqi Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Animal Sciences, Jilin University, Changchun, China
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiaolei Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ruohang Sun
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hanhai Mao
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Mingyuan Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Xuemin Jin
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
12
|
Hubbard IC, Thompson JS, Else KJ, Shears RK. Another decade of Trichuris muris research: An update and application of key discoveries. ADVANCES IN PARASITOLOGY 2023; 121:1-63. [PMID: 37474238 DOI: 10.1016/bs.apar.2023.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
The mouse whipworm, Trichuris muris, has been used for over 60 years as a tractable model for human trichuriasis, caused by the related whipworm species, T. trichiura. The history of T. muris research, from the discovery of the parasite in 1761 to understanding the lifecycle and outcome of infection with different doses (high versus low dose infection), as well as the immune mechanisms associated with parasite expulsion and chronic infection have been detailed in an earlier review published in 2013. Here, we review recent advances in our understanding of whipworm biology, host-parasite interactions and basic immunology brought about using the T. muris mouse model, focussing on developments from the last decade. In addition to the traditional high/low dose infection models that have formed the mainstay of T. muris research to date, novel models involving trickle (repeated low dose) infection in laboratory mice or infection in wild or semi-wild mice have led to important insights into how immunity develops in situ in a multivariate environment, while the use of novel techniques such as the development of caecal organoids (enabling the study of larval development ex vivo) promise to deliver important insights into host-parasite interactions. In addition, the genome and transcriptome analyses of T. muris and T. trichiura have proven to be invaluable tools, particularly in the context of vaccine development and identification of secreted products including proteins, extracellular vesicles and micro-RNAs, shedding further light on how these parasites communicate with their host and modulate the immune response to promote their own survival.
Collapse
Affiliation(s)
- Isabella C Hubbard
- Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom; Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Jacob S Thompson
- Lydia Becker Institute for Immunology and Inflammation, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kathryn J Else
- Lydia Becker Institute for Immunology and Inflammation, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Rebecca K Shears
- Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom; Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom.
| |
Collapse
|
13
|
Abstract
Type 2 immunity mediates protective responses to helminths and pathological responses to allergens, but it also has broad roles in the maintenance of tissue integrity, including wound repair. Type 2 cytokines are known to promote fibrosis, an overzealous repair response, but their contribution to healthy wound repair is less well understood. This review discusses the evidence that the canonical type 2 cytokines, IL-4 and IL-13, are integral to the tissue repair process through two main pathways. First, essential for the progression of effective tissue repair, IL-4 and IL-13 suppress the initial inflammatory response to injury. Second, these cytokines regulate how the extracellular matrix is modified, broken down, and rebuilt for effective repair. IL-4 and/or IL-13 amplifies multiple aspects of the tissue repair response, but many of these pathways are highly redundant and can be induced by other signals. Therefore, the exact contribution of IL-4Rα signaling remains difficult to unravel.
Collapse
Affiliation(s)
- Judith E Allen
- Lydia Becker Institute for Immunology and Inflammation and Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom;
| |
Collapse
|
14
|
Sutherland TE, Dyer DP, Allen JE. The extracellular matrix and the immune system: A mutually dependent relationship. Science 2023; 379:eabp8964. [PMID: 36795835 DOI: 10.1126/science.abp8964] [Citation(s) in RCA: 178] [Impact Index Per Article: 89.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 12/22/2022] [Indexed: 02/18/2023]
Abstract
For decades, immunologists have studied the role of circulating immune cells in host protection, with a more recent appreciation of immune cells resident within the tissue microenvironment and the intercommunication between nonhematopoietic cells and immune cells. However, the extracellular matrix (ECM), which comprises at least a third of tissue structures, remains relatively underexplored in immunology. Similarly, matrix biologists often overlook regulation of complex structural matrices by the immune system. We are only beginning to understand the scale at which ECM structures determine immune cell localization and function. Additionally, we need to better understand how immune cells dictate ECM complexity. This review aims to highlight the potential for biological discovery at the interface of immunology and matrix biology.
Collapse
Affiliation(s)
- Tara E Sutherland
- Wellcome Centre for Cell-Matrix Research, Lydia Becker Institute for Immunology & Infection, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Center, University of Manchester, Manchester M13 9PT, UK
- School of Medicine, Medical Sciences and Dentistry, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Douglas P Dyer
- Wellcome Centre for Cell-Matrix Research, Lydia Becker Institute for Immunology & Infection, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Center, University of Manchester, Manchester M13 9PT, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Salford M6 8HD, UK
| | - Judith E Allen
- Wellcome Centre for Cell-Matrix Research, Lydia Becker Institute for Immunology & Infection, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Center, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
15
|
The Milk Active Ingredient, 2'-Fucosyllactose, Inhibits Inflammation and Promotes MUC2 Secretion in LS174T Goblet Cells In Vitro. Foods 2023; 12:foods12010186. [PMID: 36613400 PMCID: PMC9818439 DOI: 10.3390/foods12010186] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 01/03/2023] Open
Abstract
In several mice inflammatory models, human milk oligosaccharides (HMOs) were shown to protect the intestinal barrier by promoting mucin secretion and suppressing inflammation. However, the functions of the individual HMOs in enhancing mucin expression in vivo have not been compared, and the related mechanisms are not yet to be clarified. In this study, we investigated the modulatory effects of 2′-fucosyllactose (2′-FL), 3′-sialyllactose (3′-SL), galacto-oligosaccharide (GOS) and lactose (Lac) on goblet cells’ functions in vitro. The appropriate dosage of the four chemicals was assessed in LS174T cells using the CCK-8 method. Then they were supplemented into a homeostasis and inflammatory environment to further investigate their effects under different conditions. Mucin secretion-related genes, including mucin 2 (MUC2), trefoil factor family 3 (TFF3), resistin-like β (RETNLB), carbohydrate sulfotransferase 5 (CHST5) and galactose-3-O-sulfotransferase 2 (GAL3ST2), in LS174T cells were detected using quantitative RT-qPCR. The results showed that 2′-FL (2.5 mg/mL, 72 h) was unable to increase MUC2 secretion in a steady-state condition. Comparatively, it exhibited a greater ability to improve mucin secretion under an inflammatory condition compared with GOS, demonstrated by a significant increase in TFF3 and CHST5 mRNA expression levels (p > 0.05). However, 3′-SL and Lac exhibited no effects on mucin secretion. To further investigate the underlying mechanism via which 2′-FL enhanced goblet cells’ secretion function, the NOD-like receptor family pyrin domain containing 6 (NLRP6) gene, which is closely related to MUC2 secretion, was silenced using the siRNA method. After silencing the NLRP6 gene, the mRNA expression levels of MUC2, TFF3 and CHST5 in the (2′-FL + tumor necrosis factor α (TNF-α) + NLRP6 siRNA) group were significantly decreased compared with the (2′-FL + TNF-α) group (p > 0.05), indicating that NLRP6 was essential for MUC2 expression in goblet cells. We further found that 2′-FL could significantly decrease toll-like receptor 4 (TLR4, p < 0.05), myeloid differential protein-88 (MyD88, p < 0.05) and nuclear factor kappa-B (NF-κB, p < 0.05) levels in LS174T inflammatory cells, even when the NLRP6 was silenced. Altogether, these results indicated that in goblet cells, 2′-FL exerts its function via multiple processes, i.e., by promoting mucin secretion through NLRP6 and suppressing inflammation by inhibiting the TLR4/MyD88/NF-κB pathway.
Collapse
|
16
|
Inclan-Rico JM, Rossi HL, Herbert DR. "Every cell is an immune cell; contributions of non-hematopoietic cells to anti-helminth immunity". Mucosal Immunol 2022; 15:1199-1211. [PMID: 35538230 PMCID: PMC9646929 DOI: 10.1038/s41385-022-00518-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/04/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023]
Abstract
Helminths are remarkably successful parasites that can invade various mammalian hosts and establish chronic infections that can go unnoticed for years despite causing severe tissue damage. To complete their life cycles, helminths migrate through multiple barrier sites that are densely populated by a complex array of hematopoietic and non-hematopoietic cells. While it is clear that type 2 cytokine responses elicited by immune cells promote worm clearance and tissue healing, the actions of non-hematopoietic cells are increasingly recognized as initiators, effectors and regulators of anti-helminth immunity. This review will highlight the collective actions of specialized epithelial cells, stromal niches, stem, muscle and neuroendocrine cells as well as peripheral neurons in the detection and elimination of helminths at mucosal sites. Studies dissecting the interactions between immune and non-hematopoietic cells will truly provide a better understanding of the mechanisms that ensure homeostasis in the context of helminth infections.
Collapse
Affiliation(s)
- Juan M Inclan-Rico
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Heather L Rossi
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - De'Broski R Herbert
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
17
|
Duque-Correa MA, Goulding D, Rodgers FH, Gillis JA, Cormie C, Rawlinson KA, Bancroft AJ, Bennett HM, Lotkowska ME, Reid AJ, Speak AO, Scott P, Redshaw N, Tolley C, McCarthy C, Brandt C, Sharpe C, Ridley C, Moya JG, Carneiro CM, Starborg T, Hayes KS, Holroyd N, Sanders M, Thornton DJ, Grencis RK, Berriman M. Defining the early stages of intestinal colonisation by whipworms. Nat Commun 2022; 13:1725. [PMID: 35365634 PMCID: PMC8976045 DOI: 10.1038/s41467-022-29334-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 03/08/2022] [Indexed: 01/08/2023] Open
Abstract
Whipworms are large metazoan parasites that inhabit multi-intracellular epithelial tunnels in the large intestine of their hosts, causing chronic disease in humans and other mammals. How first-stage larvae invade host epithelia and establish infection remains unclear. Here we investigate early infection events using both Trichuris muris infections of mice and murine caecaloids, the first in-vitro system for whipworm infection and organoid model for live helminths. We show that larvae degrade mucus layers to access epithelial cells. In early syncytial tunnels, larvae are completely intracellular, woven through multiple live dividing cells. Using single-cell RNA sequencing of infected mouse caecum, we reveal that progression of infection results in cell damage and an expansion of enterocytes expressing of Isg15, potentially instigating the host immune response to the whipworm and tissue repair. Our results unravel intestinal epithelium invasion by whipworms and reveal specific host-parasite interactions that allow the whipworm to establish its multi-intracellular niche.
Collapse
Affiliation(s)
- María A Duque-Correa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK.
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK.
| | - David Goulding
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Faye H Rodgers
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
- Mogrify Ltd, 25 Cambridge Science Park, Milton Road, Cambridge, CB4 0FW, UK
| | - J Andrew Gillis
- Department of Zoology, University of Cambridge, Cambridge, CB2 3EJ, UK
| | - Claire Cormie
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Kate A Rawlinson
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Allison J Bancroft
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell Matrix Research and Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Hayley M Bennett
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
- Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Magda E Lotkowska
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Adam J Reid
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Anneliese O Speak
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Paul Scott
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Nicholas Redshaw
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Charlotte Tolley
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Catherine McCarthy
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Cordelia Brandt
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Catherine Sharpe
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell Matrix Research and Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
- InstilBio, UMIC Bio-Incubator, Manchester, M13 9XX, UK
| | - Caroline Ridley
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell Matrix Research and Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
- Prime Global Medical Communications, Knutsford, WA16 8GP, UK
| | - Judit Gali Moya
- Faculty of Biology, University of Barcelona, Barcelona, 08028, Spain
| | - Claudia M Carneiro
- Immunopathology Laboratory, NUPEB, Federal University of Ouro Preto, Campus Universitario Morro do Cruzeiro, Ouro Preto, MG, 35400-000, Brazil
| | - Tobias Starborg
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell Matrix Research and Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
- Rosalind Franklin Institute, Harwell Campus, Didcot, OX11 0FA, UK
| | - Kelly S Hayes
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell Matrix Research and Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Nancy Holroyd
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Mandy Sanders
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - David J Thornton
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell Matrix Research and Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Richard K Grencis
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell Matrix Research and Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Matthew Berriman
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK.
| |
Collapse
|
18
|
The Role of the Intestinal Epithelium in the "Weep and Sweep" Response during Gastro-Intestinal Helminth Infections. Animals (Basel) 2022; 12:ani12020175. [PMID: 35049796 PMCID: PMC8772803 DOI: 10.3390/ani12020175] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/25/2021] [Accepted: 01/10/2022] [Indexed: 02/08/2023] Open
Abstract
Simple Summary The immune system actively combats intruders such as bacteria, viruses, fungi, and protozoan and metazoan parasites using leukocytes. During an infection white blood cells are activated to internalize bacteria or viruses and release a number of molecules to kill pathogens. Unfortunately, those mechanisms are ineffective against larger intruders like helminths, which are too large to be killed by a single immune cell. To eliminate gastro-intestinal helminths an integrated response involving the nervous, endocrine, and immune systems are used to expel the parasites. This is achieved through increased gut hydration and muscle contractions which detach worms from the gut and lead to release outside the body in a “weep and sweep” response. Epithelial cells of the intestine are significant players in this process, being responsible for detecting the presence of helminths in the gut and participating in the regulation of parasite expulsion. This paper describes the role of the gut epithelium in detecting and eliminating helminths from the intestine. Abstract Helminths are metazoan parasites infecting around 1.5 billion people all over the world. During coevolution with hosts, worms have developed numerous ways to trick and evade the host immune response, and because of their size, they cannot be internalized and killed by immune cells in the same way as bacteria or viruses. During infection, a substantial Th2 component to the immune response is evoked which helps restrain Th1-mediated tissue damage. Although an enhanced Th2 response is often not enough to kill the parasite and terminate an infection in itself, when tightly coordinated with the nervous, endocrine, and motor systems it can dislodge parasites from tissues and expel them from the gut. A significant role in this “weep and seep” response is attributed to intestinal epithelial cells (IEC). This review highlights the role of various IEC lineages (enterocytes, tuft cells, Paneth cells, microfold cells, goblet cells, and intestine stem cells) during the course of helminth infections and summarizes their roles in regulating gut architecture and permeability, and muscle contractions and interactions with the immune and nervous system.
Collapse
|
19
|
Chen HL, Xing X, Zhang B, Huang HB, Shi CW, Yang GL, Wang CF. Higher mucosal type II immunity is associated with increased gut microbiota diversity in BALB/c mice after Trichinella spiralis infection. Mol Immunol 2021; 138:87-98. [PMID: 34364076 DOI: 10.1016/j.molimm.2021.07.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 01/22/2023]
Abstract
Understanding the interaction between the gut microbiota and Trichinella spiralis is of interest for the early diagnosis and development of therapeutics for trichinellosis and to reveal the potential role of microbiota in the mechanism of immunomodulation of this tissue-dwelling helminth. In this study, we utilized 16S rRNA gene sequencing to monitor the dynamics of the microbes in BALB/c mice challenged with T. spiralis. Flow cytometry and ELISA were used to analyze cytokines at the same time. Histopathological analysis of the duodenum was also conducted. We found that microbial perturbations occurred during infection. The abundance of the Lachnospiraceae NK4A136 group, Ruminococcus 1 and Lactococcus decreased. However, the abundance of proinflammatory Parabacteroides increased over time after infection. T. spiralis infection also tended to inhibit IFN-γ production, and promote IL-4 and IL-10 levels. In total, T. spiralis disrupts gut homeostasis and impairs the development of the intestinal ecosystem. Defining the bacterial populations affected by T. spiralis infection might help identify microbial markers for diagnosis of the disease, and the populations could also be further exploited as a novel option to treat T. spiralis infection.
Collapse
Affiliation(s)
- Hong-Liang Chen
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Xin Xing
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Bo Zhang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Hai-Bin Huang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chun-Wei Shi
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Gui-Lian Yang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China.
| | - Chun-Feng Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China.
| |
Collapse
|
20
|
Yousefi Y, Haq S, Banskota S, Kwon YH, Khan WI. Trichuris muris Model: Role in Understanding Intestinal Immune Response, Inflammation and Host Defense. Pathogens 2021; 10:pathogens10080925. [PMID: 34451389 PMCID: PMC8399713 DOI: 10.3390/pathogens10080925] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 12/16/2022] Open
Abstract
Several parasites have evolved to survive in the human intestinal tract and over 1 billion people around the world, specifically in developing countries, are infected with enteric helminths. Trichuris trichiura is one of the world’s most common intestinal parasites that causes human parasitic infections. Trichuris muris, as an immunologically well-defined mouse model of T. trichiura, is extensively used to study different aspects of the innate and adaptive components of the immune system. Studies on T. muris model offer insights into understanding host immunity, since this parasite generates two distinct immune responses in resistant and susceptible strains of mouse. Apart from the immune cells, T. muris infection also influences various components of the intestinal tract, especially the gut microbiota, mucus layer, epithelial cells and smooth muscle cells. Here, we reviewed the different immune responses generated by innate and adaptive immune components during acute and chronic T. muris infections. Furthermore, we discussed the importance of studying T. muris model in understanding host–parasite interaction in the context of alteration in the host’s microbiota, intestinal barrier, inflammation, and host defense, and in parasite infection-mediated modulation of other immune and inflammatory diseases.
Collapse
Affiliation(s)
- Yeganeh Yousefi
- Farncombe Family Digestive Health Research Institute, McMaster University Health Sciences Centre Room 3N7, 1280 Main St. W, Hamilton, ON L8N 3Z5, Canada; (Y.Y.); (S.H.); (S.B.); (Y.H.K.)
- Department of Pathology and Molecular Medicine, McMaster University, 1200 Main St. W, Hamilton, ON L8N 3Z5, Canada
| | - Sabah Haq
- Farncombe Family Digestive Health Research Institute, McMaster University Health Sciences Centre Room 3N7, 1280 Main St. W, Hamilton, ON L8N 3Z5, Canada; (Y.Y.); (S.H.); (S.B.); (Y.H.K.)
- Department of Pathology and Molecular Medicine, McMaster University, 1200 Main St. W, Hamilton, ON L8N 3Z5, Canada
| | - Suhrid Banskota
- Farncombe Family Digestive Health Research Institute, McMaster University Health Sciences Centre Room 3N7, 1280 Main St. W, Hamilton, ON L8N 3Z5, Canada; (Y.Y.); (S.H.); (S.B.); (Y.H.K.)
- Department of Pathology and Molecular Medicine, McMaster University, 1200 Main St. W, Hamilton, ON L8N 3Z5, Canada
| | - Yun Han Kwon
- Farncombe Family Digestive Health Research Institute, McMaster University Health Sciences Centre Room 3N7, 1280 Main St. W, Hamilton, ON L8N 3Z5, Canada; (Y.Y.); (S.H.); (S.B.); (Y.H.K.)
- Department of Pathology and Molecular Medicine, McMaster University, 1200 Main St. W, Hamilton, ON L8N 3Z5, Canada
| | - Waliul I. Khan
- Farncombe Family Digestive Health Research Institute, McMaster University Health Sciences Centre Room 3N7, 1280 Main St. W, Hamilton, ON L8N 3Z5, Canada; (Y.Y.); (S.H.); (S.B.); (Y.H.K.)
- Department of Pathology and Molecular Medicine, McMaster University, 1200 Main St. W, Hamilton, ON L8N 3Z5, Canada
- Correspondence: ; Tel.: +1-905-521-2100 (ext. 22846)
| |
Collapse
|
21
|
Donskow-Łysoniewska K, Maruszewska-Cheruiyot M, Stear M. The interaction of host and nematode galectins influences the outcome of gastrointestinal nematode infections. Parasitology 2021; 148:648-654. [PMID: 33461629 PMCID: PMC11010190 DOI: 10.1017/s003118202100007x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/12/2021] [Accepted: 01/12/2021] [Indexed: 12/16/2022]
Abstract
Galectins are a family of proteins that bind β-galactosides and play key roles in a variety of cellular processes including host defence. They have been well studied in hosts but less so in gastrointestinal nematodes. Both host and parasite galectins are present in the gastrointestinal tract following infection. Parasite galectins can both bind antibody, especially highly glycosylated IgE and be bound by antibody. Parasite galectins may act as molecular sponges that soak up antibody. Host galectins promote mast cell degranulation while parasite galectins inhibit degranulation. Host and parasite galectins can also bind mucins and influence mucus viscosity. As the protective response against gastrointestinal nematode infection is partly dependent on IgE mediated mast cell degranulation and mucus, the interactions between host and parasite galectins play key roles in determining the outcome of infection.
Collapse
Affiliation(s)
- Katarzyna Donskow-Łysoniewska
- Laboratory of Parasitology, General Karol Kaczkowski Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163Warsaw, Poland
| | - Marta Maruszewska-Cheruiyot
- Laboratory of Parasitology, General Karol Kaczkowski Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163Warsaw, Poland
| | - Michael Stear
- Department of Animal, Plant and Soil Science, Agribio, La Trobe University, Bundoora, VIC3086, Australia
| |
Collapse
|
22
|
The effects of diet and gut microbiota on the regulation of intestinal mucin glycosylation. Carbohydr Polym 2021; 258:117651. [DOI: 10.1016/j.carbpol.2021.117651] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/06/2021] [Accepted: 01/11/2021] [Indexed: 12/13/2022]
|
23
|
Figueroa-Lozano S, Ren C, Yin H, Pham H, van Leeuwen S, Dijkhuizen L, de Vos P. The impact of oligosaccharide content, glycosidic linkages and lactose content of galacto-oligosaccharides (GOS) on the expression of mucus-related genes in goblet cells. Food Funct 2021; 11:3506-3515. [PMID: 32253406 DOI: 10.1039/d0fo00064g] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Galacto-oligosaccharides (GOS) have been reported to modulate the function of intestinal goblet cells and to improve mucus barrier function. However, GOS is available in many structurally different compositions and it is unknown how GOS structural diversity impacts this modulation of goblet cells. This study aims to investigate the effects of oligosaccharide content and glycosidic linkages of GOS on expression of genes associated with the secretory function of goblet cells. To investigate the effect of oligosaccharide content, LS174T cells were incubated with (β1 → 4)GOS of variable transgalactosylated oligosaccharides and lactose (Lac) composition. To investigate the effect of glycosidic linkages, we compared the effects of (β1 → 4)GOS with (β1 → 3)GOS, and with a mixture of α-linked oligosaccharides (lactose-derived oligosaccharides-LDO). The changes in mRNA expression of mucus-related genes were assessed by RT-PCR. GOS containing Lac significantly enhanced the expression of MUC2, TFF3 and RETNLB but not of Golgi sulfotransferases genes. In contrast, GOS without Lac did not impact these genes. Lac alone significantly enhanced MUC2, TFF3, RETNLB, CHST5, and GAL3ST2 genes suggesting that Lac might be responsible for goblet cell modulation in (β1 → 4)GOS preparations. (β1 → 3)GOS induced the expression of MUC2 and TFF3, and downregulated the RETNLB gene. Compared with the (β1 → 3) and GOS (β1 → 4)GOS, the α-linked LDO significantly upregulated the expression MUC2, TFF3, RETNLB and the Golgi sulfotransferases genes. We identify structural features of GOS that contribute to enhanced mucus integrity. Our study might lead to better GOS formulations for foods to prevent or treat different types of intestinal disorders.
Collapse
Affiliation(s)
- Susana Figueroa-Lozano
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen (RUG-UMCG), Hanzeplein 1, 9700 RB Groningen, The Netherlands.
| | - Chengcheng Ren
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen (RUG-UMCG), Hanzeplein 1, 9700 RB Groningen, The Netherlands.
| | - Huifang Yin
- Microbial Physiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Hien Pham
- Microbial Physiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Sander van Leeuwen
- Microbial Physiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), Nijenborgh 7, 9747 AG Groningen, The Netherlands and Department of Laboratory Medicine, RUG-UMCG, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Lubbert Dijkhuizen
- Microbial Physiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), Nijenborgh 7, 9747 AG Groningen, The Netherlands and CarbExplore Research BV, Zernikepark 12, 9747 AN Groningen, The Netherlands
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen (RUG-UMCG), Hanzeplein 1, 9700 RB Groningen, The Netherlands.
| |
Collapse
|
24
|
Chen H, Mozzicafreddo M, Pierella E, Carletti V, Piersanti A, Ali SM, Ame SM, Wang C, Miceli C. Dissection of the gut microbiota in mothers and children with chronic Trichuris trichiura infection in Pemba Island, Tanzania. Parasit Vectors 2021; 14:62. [PMID: 33468220 PMCID: PMC7814639 DOI: 10.1186/s13071-021-04580-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 01/05/2021] [Indexed: 12/21/2022] Open
Abstract
Background Soil-transmitted helminthiases are important neglected tropical diseases that result in a notably high number of disability-adjusted life years worldwide. Characterizing the interactions between the human intestinal microbiome and helminths is of interest in the development of alternative treatments that do not rely on chemotherapeutics and do not lead to drug resistance. Methods We recruited and obtained fecal samples from 32 pairs of mothers and children on Pemba Island and monitored their intestinal microbiota using 16S rRNA gene sequencing. Results We observed that microbial changes occur in the gut microbiota of infected mothers and children. Some short-chain fatty acid (SCFA)-producing bacteria and carbohydrate-degrading bacteria exhibited lower abundance in the infected individuals. Potentially pathogenic Campylobacter and proinflammatory Methanobrevibacter in infected mothers and opportunistic Enterococcus in infected children exhibited greater abundance. Conclusions Our findings could reveal the microbiota profiling in T. trichiura-infected individuals, indicate the potential roles of key microbiota in the host and aid to the development of novel strategies to control T. trichiura infection. Graphic abstract ![]()
Collapse
Affiliation(s)
- Hongliang Chen
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China.,School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Matteo Mozzicafreddo
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Elisa Pierella
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Vanessa Carletti
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Angela Piersanti
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Said M Ali
- Public Health Laboratory Ivo de Carneri, Pemba Island, Chake Chake, Tanzania
| | - Shaali M Ame
- Public Health Laboratory Ivo de Carneri, Pemba Island, Chake Chake, Tanzania
| | - Chunfeng Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China.
| | - Cristina Miceli
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy.
| |
Collapse
|
25
|
Stoldt M, Klein L, Beros S, Butter F, Jongepier E, Feldmeyer B, Foitzik S. Parasite Presence Induces Gene Expression Changes in an Ant Host Related to Immunity and Longevity. Genes (Basel) 2021; 12:95. [PMID: 33451085 PMCID: PMC7828512 DOI: 10.3390/genes12010095] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/22/2020] [Accepted: 01/12/2021] [Indexed: 12/14/2022] Open
Abstract
Most species are either parasites or exploited by parasites, making parasite-host interactions a driver of evolution. Parasites with complex life cycles often evolve strategies to facilitate transmission to the definitive host by manipulating their intermediate host. Such manipulations could explain phenotypic changes in the ant Temnothorax nylanderi, the intermediate host of the cestode Anomotaenia brevis. In addition to behavioral and morphological alterations, infected workers exhibit prolonged lifespans, comparable to that of queens, which live up to two decades. We used transcriptomic data from cestodes and ants of different castes and infection status to investigate the molecular underpinnings of phenotypic alterations in infected workers and explored whether the extended lifespan of queens and infected workers has a common molecular basis. Infected workers and queens commonly upregulated only six genes, one of them with a known anti-aging function. Both groups overexpressed immune genes, although not the same ones. Our findings suggest that the lifespan extension of infected workers is not achieved via the expression of queen-specific genes. The analysis of the cestodes' transcriptome revealed dominant expression of genes of the mitochondrial respiratory transport chain, which indicates an active metabolism and shedding light on the physiology of the parasite in its cysticercoid stage.
Collapse
Affiliation(s)
- Marah Stoldt
- Institute of Organismic and Molecular Evolution, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; (L.K.); (S.F.)
| | - Linda Klein
- Institute of Organismic and Molecular Evolution, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; (L.K.); (S.F.)
| | - Sara Beros
- Max Planck Institute for the Biology of Ageing, 50931 Cologne, Germany;
| | - Falk Butter
- Institute for Molecular Biology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany;
| | - Evelien Jongepier
- Institute for Evolution and Biodiversity, University of Münster, 48149 Münster, Germany;
| | - Barbara Feldmeyer
- Senckenberg Biodiversity and Climate Research Center (SBiK-F), Molecular Ecology, 60325 Frankfurt, Germany;
| | - Susanne Foitzik
- Institute of Organismic and Molecular Evolution, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; (L.K.); (S.F.)
| |
Collapse
|
26
|
Grondin JA, Kwon YH, Far PM, Haq S, Khan WI. Mucins in Intestinal Mucosal Defense and Inflammation: Learning From Clinical and Experimental Studies. Front Immunol 2020; 11:2054. [PMID: 33013869 PMCID: PMC7500085 DOI: 10.3389/fimmu.2020.02054] [Citation(s) in RCA: 252] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/28/2020] [Indexed: 12/24/2022] Open
Abstract
Throughout the gastrointestinal (GI) tract, a distinct mucus layer composed of highly glycosylated proteins called mucins plays an essential role in providing lubrication for the passage of food, participating in cell signaling pathways and protecting the host epithelium from commensal microorganisms and invading pathogens, as well as toxins and other environmental irritants. These mucins can be broadly classified into either secreted gel-forming mucins, those that provide the structural backbone for the mucus barrier, or transmembrane mucins, those that form the glycocalyx layer covering the underlying epithelial cells. Goblet cells dispersed among the intestinal epithelial cells are chiefly responsible for the synthesis and secretion of mucins within the gut and are heavily influenced by interactions with the immune system. Evidence from both clinical and animal studies have indicated that several GI conditions, including inflammatory bowel disease (IBD), colorectal cancer, and numerous enteric infections are accompanied by considerable changes in mucin quality and quantity. These changes include, but are not limited to, impaired goblet cell function, synthesis dysregulation, and altered post-translational modifications. The current review aims to highlight the structural and functional features as well as the production and immunological regulation of mucins and the impact these key elements have within the context of barrier function and host defense in intestinal inflammation.
Collapse
Affiliation(s)
- Jensine A Grondin
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Yun Han Kwon
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Parsa Mehraban Far
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Sabah Haq
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Waliul I Khan
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
27
|
Coakley G, Harris NL. The Intestinal Epithelium at the Forefront of Host-Helminth Interactions. Trends Parasitol 2020; 36:761-772. [PMID: 32713764 DOI: 10.1016/j.pt.2020.07.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 02/06/2023]
Abstract
Gastrointestinal helminth infection still constitutes a major public health issue, particularly in the developing world. As these parasites can undergo a large part of their lifecycle within the intestinal tract the host has developed various structural and cellular specializations at the epithelial barrier to contend with infection. Detailed characterization of these cells will provide important insights about their contributions to the protective responses mediated against helminths. Here, we discuss how key components of the intestinal epithelium may function to limit the initial establishment of helminths, and how these cells are altered during an active response to infection.
Collapse
Affiliation(s)
- Gillian Coakley
- Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, Victoria, Australia.
| | - Nicola L Harris
- Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| |
Collapse
|
28
|
Quin C, Vicaretti SD, Mohtarudin NA, Garner AM, Vollman DM, Gibson DL, Zandberg WF. Influence of sulfonated and diet-derived human milk oligosaccharides on the infant microbiome and immune markers. J Biol Chem 2020; 295:4035-4048. [PMID: 32014993 DOI: 10.1074/jbc.ra119.011351] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/23/2020] [Indexed: 12/14/2022] Open
Abstract
Human milk oligosaccharides (HMOs) promote the development of the neonatal intestinal, immune, and nervous systems and has recently received considerable attention. Here we investigated how the maternal diet affects HMO biosynthesis and how any diet-induced HMO alterations influence the infant gut microbiome and immunity. Using capillary electrophoresis and MS-based analyses, we extracted and measured HMOs from breast milk samples and then correlated their levels with results from validated 24-h diet recall surveys and breast milk fatty acids. We found that fruit intake and unsaturated fatty acids in breast milk were positively correlated with an increased absolute abundance of numerous HMOs, including 16 sulfonated HMOs we identified here in humans for the first time. The diet-derived monosaccharide 5-N-glycolyl-neuraminic acid (Neu5Gc) was unambiguously detected in all samples. To gain insights into the potential impact of Neu5Gc on the infant microbiome, we used a constrained ordination approach and identified correlations between Neu5Gc levels and Bacteroides spp. in infant stool. However, Neu5Gc was not associated with marked changes in infant immune markers, in contrast with sulfonated HMOs, whose expression correlated with suppression of two major Th2 cytokines, IL-10 and IL-13. The findings of our work highlight the importance of maternal diet for HMO biosynthesis and provide as yet unexplored targets for future studies investigating interactions between HMOs and the intestinal microbiome and immunity in infants.
Collapse
Affiliation(s)
- Candice Quin
- Department of Biology, I. K. Barber School of Arts and Sciences, University of British Columbia, 1177 Research Road, Kelowna, British Columbia, Canada V1V 1V7
| | - Sara D Vicaretti
- Department of Chemistry, I. K. Barber School of Arts and Sciences, University of British Columbia, 3247 University Way, Kelowna, British Columbia, Canada V1V 1V7
| | - Nina A Mohtarudin
- Department of Biology, I. K. Barber School of Arts and Sciences, University of British Columbia, 1177 Research Road, Kelowna, British Columbia, Canada V1V 1V7
| | - Alexander M Garner
- Department of Biology, I. K. Barber School of Arts and Sciences, University of British Columbia, 1177 Research Road, Kelowna, British Columbia, Canada V1V 1V7
| | - Deanna M Vollman
- Department of Biology, I. K. Barber School of Arts and Sciences, University of British Columbia, 1177 Research Road, Kelowna, British Columbia, Canada V1V 1V7
| | - Deanna L Gibson
- Department of Biology, I. K. Barber School of Arts and Sciences, University of British Columbia, 1177 Research Road, Kelowna, British Columbia, Canada V1V 1V7 .,Department of Medicine, Faculty of Medicine, 317-2194 Health Sciences Mall, Vancouver, British Columbia, Canada V6T 1Z3
| | - Wesley F Zandberg
- Department of Chemistry, I. K. Barber School of Arts and Sciences, University of British Columbia, 3247 University Way, Kelowna, British Columbia, Canada V1V 1V7
| |
Collapse
|
29
|
Glover M, Colombo SAP, Thornton DJ, Grencis RK. Trickle infection and immunity to Trichuris muris. PLoS Pathog 2019; 15:e1007926. [PMID: 31730667 PMCID: PMC6881069 DOI: 10.1371/journal.ppat.1007926] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 11/27/2019] [Accepted: 10/29/2019] [Indexed: 01/16/2023] Open
Abstract
The majority of experiments investigating the immune response to gastrointestinal helminth infection use a single bolus infection. However, in situ individuals are repeatedly infected with low doses. Therefore, to model natural infection, mice were repeatedly infected (trickle infection) with low doses of Trichuris muris. Trickle infection resulted in the slow acquisition of immunity reflected by a gradual increase in worm burden followed by partial expulsion. Flow cytometry revealed that the CD4+ T cell response shifted from Th1 dominated to Th2 dominated, which coincided with an increase in Type 2 cytokines. The development of resistance following trickle infection was associated with increased worm expulsion effector mechanisms including goblet cell hyperplasia, Muc5ac production and increased epithelial cell turn over. Depletion of CD4+ T cells reversed resistance confirming their importance in protective immunity following trickle infection. In contrast, depletion of group 2 innate lymphoid cells did not alter protective immunity. T. muris trickle infection resulted in a dysbiotic mircrobiota which began to recover alpha diversity following the development of resistance. These data establish trickle infection as a robust and informative model for analysis of immunity to chronic intestinal helminth infection more akin to that observed under natural infection conditions and confirms the importance of CD4+ T cell adaptive immunity in host protection.
Collapse
Affiliation(s)
- Maya Glover
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
- Wellcome Centre for Cell Matrix Research, University of Manchester, Manchester, United Kingdom
| | - Stefano A. P. Colombo
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
- Wellcome Centre for Cell Matrix Research, University of Manchester, Manchester, United Kingdom
| | - David J. Thornton
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
- Wellcome Centre for Cell Matrix Research, University of Manchester, Manchester, United Kingdom
| | - Richard K. Grencis
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
- Wellcome Centre for Cell Matrix Research, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
30
|
MacMillan JL, Vicaretti SD, Noyovitz B, Xing X, Low KE, Inglis GD, Zaytsoff SJ, Boraston AB, Smith SP, Uwiera RR, Selinger LB, Zandberg WF, Abbott DW. Structural analysis of broiler chicken small intestinal mucin O-glycan modification by Clostridium perfringens. Poult Sci 2019; 98:5074-5088. [DOI: 10.3382/ps/pez297] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/03/2019] [Indexed: 12/12/2022] Open
|
31
|
Etienne-Mesmin L, Chassaing B, Desvaux M, De Paepe K, Gresse R, Sauvaitre T, Forano E, de Wiele TV, Schüller S, Juge N, Blanquet-Diot S. Experimental models to study intestinal microbes–mucus interactions in health and disease. FEMS Microbiol Rev 2019; 43:457-489. [DOI: 10.1093/femsre/fuz013] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/31/2019] [Indexed: 02/06/2023] Open
Abstract
ABSTRACT
A close symbiotic relationship exists between the intestinal microbiota and its host. A critical component of gut homeostasis is the presence of a mucus layer covering the gastrointestinal tract. Mucus is a viscoelastic gel at the interface between the luminal content and the host tissue that provides a habitat to the gut microbiota and protects the intestinal epithelium. The review starts by setting up the biological context underpinning the need for experimental models to study gut bacteria-mucus interactions in the digestive environment. We provide an overview of the structure and function of intestinal mucus and mucins, their interactions with intestinal bacteria (including commensal, probiotics and pathogenic microorganisms) and their role in modulating health and disease states. We then describe the characteristics and potentials of experimental models currently available to study the mechanisms underpinning the interaction of mucus with gut microbes, including in vitro, ex vivo and in vivo models. We then discuss the limitations and challenges facing this field of research.
Collapse
Affiliation(s)
- Lucie Etienne-Mesmin
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Benoit Chassaing
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303 , USA
- Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave, Atlanta, GA 30303 , USA
| | - Mickaël Desvaux
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Kim De Paepe
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Raphaële Gresse
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Thomas Sauvaitre
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Evelyne Forano
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Stephanie Schüller
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR7UQ, United Kingdom
| | - Nathalie Juge
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR7UQ, United Kingdom
| | - Stéphanie Blanquet-Diot
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| |
Collapse
|
32
|
Ren C, Dokter‐Fokkens J, Figueroa Lozano S, Zhang Q, de Haan BJ, Zhang H, Faas MM, de Vos P. Fibroblasts Impact Goblet Cell Responses to Lactic Acid Bacteria After Exposure to Inflammatory Cytokines and Mucus Disruptors. Mol Nutr Food Res 2019; 63:e1801427. [PMID: 30977971 PMCID: PMC6618108 DOI: 10.1002/mnfr.201801427] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/21/2019] [Indexed: 12/15/2022]
Abstract
SCOPE Mucus produced by goblet cells contributes to gut barrier function. Lactic acid bacteria (LAB) have been shown to impact mucus production. It is not completely known whether mucus production is influenced by the abundantly present fibroblasts in the intestine. METHODS AND RESULTS The influence of fibroblasts on mucus-related genes including mucin-2 (MUC2), trefoil factor 3 (TFF3), resistin-like molecule β (RETNLB), carbohydrate sulfotransferase 5 (CHST5), and galactose-3-O-sulfotransferase 2 (GAL3ST2) is examined after co-culture of LS174T-goblet cells and CCD-18Co colonic fibroblasts in the presence and absence of LAB-strains known to impact mucus function. This is also tested after exposure to TNF-α, IL-13, or the mucin synthesis inhibitor tunicamycin (Tm). Effects of fibroblasts are treatment duration- and bacterial species-dependent under homeostatic conditions. During TNF-α challenge, fibroblasts reverse Lactobacillus (L.) rhamnosus CCFM237-elicited declined TFF3 expression. After IL-13 exposure, L. rhamnosus CCFM237 and L. fermentum CCFM787 attenuate enhanced TFF3 and RETNLB expression, respectively, only in the presence of fibroblasts. LAB has no effects on Tm-induced decreased expression of goblet cell-related genes regardless of the presence of fibroblasts. CONCLUSION It is demonstrated that goblet cell-fibroblast crosstalk impacts mucus synthesis and influences the effects of LAB on goblet cell-related genes. Effects are LAB-species and stressor dependent.
Collapse
Affiliation(s)
- Chengcheng Ren
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center Groningen9700RBGroningenThe Netherlands
- School of Food Science and TechnologyJiangnan UniversityWuxi214122China
| | - Jelleke Dokter‐Fokkens
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center Groningen9700RBGroningenThe Netherlands
| | - Susana Figueroa Lozano
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center Groningen9700RBGroningenThe Netherlands
| | - Qiuxiang Zhang
- School of Food Science and TechnologyJiangnan UniversityWuxi214122China
| | - Bart J. de Haan
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center Groningen9700RBGroningenThe Netherlands
| | - Hao Zhang
- School of Food Science and TechnologyJiangnan UniversityWuxi214122China
| | - Marijke M. Faas
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center Groningen9700RBGroningenThe Netherlands
| | - Paul de Vos
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center Groningen9700RBGroningenThe Netherlands
| |
Collapse
|
33
|
Rapin A, Harris NL. Helminth-Bacterial Interactions: Cause and Consequence. Trends Immunol 2018; 39:724-733. [PMID: 29941203 DOI: 10.1016/j.it.2018.06.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/22/2018] [Accepted: 06/02/2018] [Indexed: 01/16/2023]
Abstract
Intestinal helminths, along with mutualistic microbes, have cohabited the intestine of mammals throughout evolution. Interactions between helminths, bacteria, and their mammalian hosts may shape not only host-helminth and host-microbiome interactions, but also the relationship between helminths and the microbiome. This 'ménage à trois' situation may not be completely balanced in that it may favor either the host or the parasite, possibly at the cost of the other partner. Similarly, helminths may favor the establishment of a particular microbiome with either positive or negative consequences for the overall health and well-being of the host. Recent studies indicate that infection with intestinal helminths can and does impact the intestinal microbiome, with important consequences for each partner in this tripartite relationship.
Collapse
Affiliation(s)
- Alexis Rapin
- Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Nicola L Harris
- Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland; Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC, Australia.
| |
Collapse
|
34
|
Alabbas SY, Begun J, Florin TH, Oancea I. The role of IL-22 in the resolution of sterile and nonsterile inflammation. Clin Transl Immunology 2018; 7:e1017. [PMID: 29713472 PMCID: PMC5905349 DOI: 10.1002/cti2.1017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/25/2018] [Accepted: 03/26/2018] [Indexed: 12/14/2022] Open
Abstract
In a broad sense, inflammation can be conveniently characterised by two phases: the first phase, which is a pro-inflammatory, has evolved to clear infection and/or injured tissue; and the second phase concerns regeneration of normal tissue and restitution of normal physiology. Innate immune cell-derived pro-inflammatory cytokines and chemokines activate and recruit nonresident immune cells to the site of infection, thereby amplifying the inflammatory responses to clear infection or injury. This phase is followed by a cytokine milieu that promotes tissue regeneration. There is no absolute temporal distinction between these two phases, and cytokines may have dual pleiotropic effects depending on the timing of release, inflammatory microenvironment or concentrations. IL-22 is a cytokine with reported pro- and anti-inflammatory roles; in this review, we contend that this protein has primarily a function in restitution of normal tissue and physiology.
Collapse
Affiliation(s)
- Saleh Y Alabbas
- Faculty of MedicineSchool of Clinical MedicineThe University of QueenslandBrisbaneQLDAustralia
- Chronic Disease Biology and Care Group at Mater Research InstituteTranslational Research InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Jakob Begun
- Chronic Disease Biology and Care Group at Mater Research InstituteTranslational Research InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Timothy H Florin
- Chronic Disease Biology and Care Group at Mater Research InstituteTranslational Research InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Iulia Oancea
- Faculty of MedicineSchool of Clinical MedicineThe University of QueenslandBrisbaneQLDAustralia
- Chronic Disease Biology and Care Group at Mater Research InstituteTranslational Research InstituteThe University of QueenslandBrisbaneQLDAustralia
| |
Collapse
|
35
|
Wang R, Moniruzzaman M, Shuffle E, Lourie R, Hasnain SZ. Immune regulation of the unfolded protein response at the mucosal barrier in viral infection. Clin Transl Immunology 2018; 7:e1014. [PMID: 29632667 PMCID: PMC5881172 DOI: 10.1002/cti2.1014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 01/12/2023] Open
Abstract
Protein folding in the endoplasmic reticulum (ER) is subject to stringent quality control. When protein secretion demand exceeds the protein folding capacity of the ER, the unfolded protein response (UPR) is triggered as a consequence of ER stress. Due to the secretory function of epithelial cells, UPR plays an important role in maintaining epithelial barrier function at mucosal sites. ER stress and activation of the UPR are natural mechanisms by which mucosal epithelial cells combat viral infections. In this review, we discuss the important role of UPR in regulating mucosal epithelium homeostasis. In addition, we review current insights into how the UPR is involved in viral infection at mucosal barriers and potential therapeutic strategies that restore epithelial cell integrity following acute viral infections via cytokine and cellular stress manipulation.
Collapse
Affiliation(s)
- Ran Wang
- Translational Research Institute Immunopathology Group at Mater Research Institute - The University of Queensland Brisbane QLD Australia.,Faculty of Medicine The University of Queensland Brisbane QLD Australia
| | - Md Moniruzzaman
- Translational Research Institute Immunopathology Group at Mater Research Institute - The University of Queensland Brisbane QLD Australia.,Faculty of Medicine The University of Queensland Brisbane QLD Australia
| | - Eric Shuffle
- Translational Research Institute Immunopathology Group at Mater Research Institute - The University of Queensland Brisbane QLD Australia
| | - Rohan Lourie
- Translational Research Institute Immunopathology Group at Mater Research Institute - The University of Queensland Brisbane QLD Australia.,Translational Research Institute Inflammatory Bowel Disease Group at Mater Research Institute - The University of Queensland Brisbane QLD Australia
| | - Sumaira Z Hasnain
- Translational Research Institute Immunopathology Group at Mater Research Institute - The University of Queensland Brisbane QLD Australia.,Faculty of Medicine The University of Queensland Brisbane QLD Australia
| |
Collapse
|
36
|
Sharpe C, Thornton DJ, Grencis RK. A sticky end for gastrointestinal helminths; the role of the mucus barrier. Parasite Immunol 2018; 40:e12517. [PMID: 29355990 PMCID: PMC5900928 DOI: 10.1111/pim.12517] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 01/15/2018] [Indexed: 12/20/2022]
Abstract
Gastrointestinal (GI) nematodes are a group of successful multicellular parasites that have evolved to coexist within the intestinal niche of multiple species. It is estimated that over 10% of the world's population are chronically infected by GI nematodes, making this group of parasitic nematodes a major burden to global health. Despite the large number of affected individuals, there are few effective treatments to eradicate these infections. Research into GI nematode infections has primarily focused on defining the immunological and pathological consequences on host protection. One important but neglected aspect of host protection is mucus, and the concept that mucus is just a simple barrier is no longer tenable. In fact, mucus is a highly regulated and dynamic-secreted matrix, underpinned by a physical hydrated network of highly glycosylated mucins, which is increasingly recognized to have a key protective role against GI nematode infections. Unravelling the complex interplay between mucins, the underlying epithelium and immune cells during infection are a major challenge and are required to fully define the protective role of the mucus barrier. This review summarizes the current state of knowledge on mucins and the mucus barrier during GI nematode infections, with particular focus on murine models of infection.
Collapse
Affiliation(s)
- C Sharpe
- Manchester Immunology Group, Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - D J Thornton
- Manchester Immunology Group, Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - R K Grencis
- Manchester Immunology Group, Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| |
Collapse
|
37
|
Ren C, Dokter‐Fokkens J, Figueroa Lozano S, Zhang Q, de Haan BJ, Zhang H, Faas MM, de Vos P. Lactic Acid Bacteria May Impact Intestinal Barrier Function by Modulating Goblet Cells. Mol Nutr Food Res 2018; 62:e1700572. [PMID: 29333697 PMCID: PMC5900975 DOI: 10.1002/mnfr.201700572] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 12/11/2017] [Indexed: 12/19/2022]
Abstract
SCOPE Lactic acid bacteria (LAB) are recognized to promote gastrointestinal health by mechanisms that are not fully understood. LABs might modulate the mucus and thereby enhance intestinal barrier function. Herein, we investigate effects of different LAB strains and species on goblet cell genes involved in mucus synthesis. METHODS AND RESULTS Gene expression profiles of goblet-cell-associated products (mucin MUC2, trefoil factor 3, resistin-like molecule β, carbohydrate sulfotransferase 5, and galactose-3-O-sulfotransferase 2) induced by LAB or their derived conditioned medium in human goblet cell line LS174T are studied. Effects of LAB on gene transcription are assessed with or without exposure to TNF-α, IL-13, or the mucus damaging agent tunicamycin. LAB do impact the related genes in a species- and strain-specific fashion and their effects are different in the presence of the cytokines and tunicamycin. Bioactive factors secreted by some strains are also found to regulate goblet cell-related genes. CONCLUSION Our findings provide novel insights in differences in modulatory efficacy on mucus genes between LAB species and strains. This study further unravels direct interactions between LAB and intestinal goblet cells, and highlights the importance of rationally selecting appropriate LAB candidates to achieve specific benefits in the gut.
Collapse
Affiliation(s)
- Chengcheng Ren
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center GroningenHanzeplein 1The Netherlands
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Jelleke Dokter‐Fokkens
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center GroningenHanzeplein 1The Netherlands
| | - Susana Figueroa Lozano
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center GroningenHanzeplein 1The Netherlands
| | - Qiuxiang Zhang
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Bart J. de Haan
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center GroningenHanzeplein 1The Netherlands
| | - Hao Zhang
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Marijke M. Faas
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center GroningenHanzeplein 1The Netherlands
| | - Paul de Vos
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center GroningenHanzeplein 1The Netherlands
| |
Collapse
|
38
|
Immunity to gastrointestinal nematode infections. Mucosal Immunol 2018; 11:304-315. [PMID: 29297502 DOI: 10.1038/mi.2017.113] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 11/20/2017] [Indexed: 02/06/2023]
Abstract
Numerous species of nematodes have evolved to inhabit the gastrointestinal tract of animals and humans, with over a billion of the world's population infected with at least one species. These large multicellular pathogens present a considerable and complex challenge to the host immune system given that individuals are continually exposed to infective stages, as well as the high prevalence in endemic areas. This review summarizes our current understanding of host-parasite interactions, detailing induction of protective immunity, mechanisms of resistance, and resolution of the response. It is clear from studies of well-defined laboratory model systems that these responses are dominated by innate and adaptive type 2 cytokine responses, regulating cellular and soluble effectors that serve to disrupt the niche in which the parasites live by strengthening the physical mucosal barrier and ultimately promoting tissue repair.
Collapse
|
39
|
Wang R, Hasnain SZ. Analyzing the Properties of Murine Intestinal Mucins by Electrophoresis and Histology. Bio Protoc 2017; 7:e2394. [PMID: 34541128 DOI: 10.21769/bioprotoc.2394] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/01/2017] [Accepted: 06/01/2017] [Indexed: 11/02/2022] Open
Abstract
Specialized secretory cells known as goblet cells in the intestine and respiratory epithelium are responsible for the secretion of mucins. Mucins are large heavily glycosylated proteins and typically have a molecular mass higher than 106 Da. These large proteins are densely substituted with short glycan chains, which have many important functional roles including determining the hydration and viscoelastic properties of the mucus gel that lines and protects the intestinal epithelium. In this protocol, we comprehensively describe the method for extraction of murine mucus and its analysis by agarose gel electrophoresis. Additionally we describe the use of High Iron Diamine-Alcian Blue, Periodic Acid Schiff's-Alcian Blue and immune-staining methods to identify and differentiate between the different states of glycosylation on these mucin glycoproteins, in particular with a focus on sulphation and sialylation.
Collapse
Affiliation(s)
- Ran Wang
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Sumaira Z Hasnain
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Australia
| |
Collapse
|
40
|
|