1
|
Gámez-Belmonte R, Wagner Y, Mahapatro M, Wang R, Erkert L, González-Acera M, Cineus R, Hainbuch S, Patankar JV, Voehringer D, Hegazy AN, Neurath MF, Wirtz S, Becker C. Intestinal epithelial Gasdermin C is induced by IL-4R/STAT6 signaling but is dispensable for gut immune homeostasis. Sci Rep 2024; 14:26522. [PMID: 39489845 PMCID: PMC11532336 DOI: 10.1038/s41598-024-78336-z] [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: 06/19/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024] Open
Abstract
Gasdermin C is one of the least studied members of the gasdermin family of proteins, known for their critical involvement in pyroptosis and host defense. Furthermore, evidence for the role of Gasdermin C in the intestine is scarce and partly controversial. Here, we tested the functional role of Gasdermin C in intestinal homeostasis, inflammation and tumorigenesis. : We studied Gasdermin C in response to cytokines in intestinal organoids. We evaluated epithelial differentiation, cell death and immune infiltration under steady state conditions in a new mouse line deficient in Gasdermin C. The role of Gasdermin C was analyzed in acute colitis, infection and colitis-associated cancer. Gasdemin C is highly expressed in the intestinal epithelium and strongly induced by the type 2 cytokines IL-4 and IL-13 in a STAT6-dependent manner. Gasdermin C-deficient mice show no changes in tissue architecture and epithelial homeostasis. Epithelial organoids deficient in Gasdermin C develop normally and show no alterations in proliferation or cell death. No changes were found in models of acute colitis, type 2 intestinal infection and colitis-associated cancer. Gasdermin C genes are upregulated by type 2 immunity, yet appear dispensable for the development of intestinal inflammation, infection and colitis-associated cancer.
Collapse
Affiliation(s)
- Reyes Gámez-Belmonte
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Yara Wagner
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Mousumi Mahapatro
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ru Wang
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Lena Erkert
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Miguel González-Acera
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Roodline Cineus
- Department of Gastroenterology, Infectiology and Rheumatology, Charité Universitätsmedizin, Berlin, Germany
- Deutsches Rheumaforschungszentrum Berlin (DRFZ), An Institute of the Leibniz Association, Berlin, Germany
| | - Saskia Hainbuch
- Deutsches Rheumaforschungszentrum Berlin (DRFZ), An Institute of the Leibniz Association, Berlin, Germany
| | - Jay V Patankar
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - David Voehringer
- Institute of Immunology, Ludwig-Maximilians-Universität München, 80336, München, Germany
- Department of Infection Biology, University of Erlangen, 91054, Erlangen, Germany
| | - Ahmed N Hegazy
- Department of Gastroenterology, Infectiology and Rheumatology, Charité Universitätsmedizin, Berlin, Germany
- Deutsches Rheumaforschungszentrum Berlin (DRFZ), An Institute of the Leibniz Association, Berlin, Germany
- The Transregio 241 IBDome Consortium, Berlin, Germany
| | - Markus F Neurath
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- The Transregio 241 IBDome Consortium, Berlin, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- The Transregio 241 IBDome Consortium, Berlin, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Christoph Becker
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.
- The Transregio 241 IBDome Consortium, Berlin, Germany.
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany.
- Department of Medicine 1, University Medical Centre Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
2
|
Billipp TE, Fung C, Webeck LM, Sargent DB, Gologorsky MB, Chen Z, McDaniel MM, Kasal DN, McGinty JW, Barrow KA, Rich LM, Barilli A, Sabat M, Debley JS, Wu C, Myers R, Howitt MR, von Moltke J. Tuft cell-derived acetylcholine promotes epithelial chloride secretion and intestinal helminth clearance. Immunity 2024; 57:1243-1259.e8. [PMID: 38744291 PMCID: PMC11168877 DOI: 10.1016/j.immuni.2024.03.023] [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: 04/05/2023] [Revised: 02/05/2024] [Accepted: 03/29/2024] [Indexed: 05/16/2024]
Abstract
Epithelial cells secrete chloride to regulate water release at mucosal barriers, supporting both homeostatic hydration and the "weep" response that is critical for type 2 immune defense against parasitic worms (helminths). Epithelial tuft cells in the small intestine sense helminths and release cytokines and lipids to activate type 2 immune cells, but whether they regulate epithelial secretion is unknown. Here, we found that tuft cell activation rapidly induced epithelial chloride secretion in the small intestine. This response required tuft cell sensory functions and tuft cell-derived acetylcholine (ACh), which acted directly on neighboring epithelial cells to stimulate chloride secretion, independent of neurons. Maximal tuft cell-induced chloride secretion coincided with immune restriction of helminths, and clearance was delayed in mice lacking tuft cell-derived ACh, despite normal type 2 inflammation. Thus, we have uncovered an epithelium-intrinsic response unit that uses ACh to couple tuft cell sensing to the secretory defenses of neighboring epithelial cells.
Collapse
Affiliation(s)
- Tyler E Billipp
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Connie Fung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lily M Webeck
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Derek B Sargent
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Matthew B Gologorsky
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Zuojia Chen
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Margaret M McDaniel
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Darshan N Kasal
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - John W McGinty
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Kaitlyn A Barrow
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA
| | - Lucille M Rich
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA
| | | | - Mark Sabat
- Takeda Pharmaceuticals, San Diego, CA, USA
| | - Jason S Debley
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | | | - Michael R Howitt
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jakob von Moltke
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
3
|
Mosala P, Mpotje T, Abdel Aziz N, Ndlovu H, Musaigwa F, Nono JK, Brombacher F. Cysteinyl leukotriene receptor-1 as a potential target for host-directed therapy during chronic schistosomiasis in murine model. Front Immunol 2024; 15:1279043. [PMID: 38840916 PMCID: PMC11150569 DOI: 10.3389/fimmu.2024.1279043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 04/25/2024] [Indexed: 06/07/2024] Open
Abstract
Schistosomiasis remains the most devastating neglected tropical disease, affecting over 240 million people world-wide. The disease is caused by the eggs laid by mature female worms that are trapped in host's tissues, resulting in chronic Th2 driven fibrogranulmatous pathology. Although the disease can be treated with a relatively inexpensive drug, praziquantel (PZQ), re-infections remain a major problem in endemic areas. There is a need for new therapeutic drugs and alternative drug treatments for schistosomiasis. The current study hypothesized that cysteinyl leukotrienes (cysLTs) could mediate fibroproliferative pathology during schistosomiasis. Cysteinyl leukotrienes (cysLTs) are potent lipid mediators that are known to be key players in inflammatory diseases, such as asthma and allergic rhinitis. The present study aimed to investigate the role of cysLTR1 during experimental acute and chronic schistosomiasis using cysLTR1-/- mice, as well as the use of cysLTR1 inhibitor (Montelukast) to assess immune responses during chronic Schistosoma mansoni infection. Mice deficient of cysLTR1 and littermate control mice were infected with either high or low dose of Schistosoma mansoni to achieve chronic or acute schistosomiasis, respectively. Hepatic granulomatous inflammation, hepatic fibrosis and IL-4 production in the liver was significantly reduced in mice lacking cysLTR1 during chronic schistosomiasis, while reduced liver pathology was observed during acute schistosomiasis. Pharmacological blockade of cysLTR1 using montelukast in combination with PZQ reduced hepatic inflammation and parasite egg burden in chronically infected mice. Combination therapy led to the expansion of Tregs in chronically infected mice. We show that the disruption of cysLTR1 is dispensable for host survival during schistosomiasis, suggesting an important role cysLTR1 may play during early immunity against schistosomiasis. Our findings revealed that the combination of montelukast and PZQ could be a potential prophylactic treatment for chronic schistosomiasis by reducing fibrogranulomatous pathology in mice. In conclusion, the present study demonstrated that cysLTR1 is a potential target for host-directed therapy to ameliorate fibrogranulomatous pathology in the liver during chronic and acute schistosomiasis in mice.
Collapse
Affiliation(s)
- Paballo Mosala
- Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
| | - Thabo Mpotje
- Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
| | - Nada Abdel Aziz
- Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
- Immuno-Biotechnology Lab, Biotechnology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Hlumani Ndlovu
- Division of Chemical and System Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Fungai Musaigwa
- Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
| | - Justin Komguep Nono
- Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
- Unit of Immunobiology and Helminth Infections, Laboratory of Molecular Biology and Biotechnology, Institute of Medical Research and Medicinal Plant Studies (IMPM), Ministry of Scientific Research and Innovation, Yaoundé, Cameroon
| | - Frank Brombacher
- Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
- Welcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
4
|
Srisai P, Chaiyadet S, Jumnainsong A, Suttiprapa S, Leelayuwat C, Saichua P. T helper cell responses to Opisthorchis viverrini infection associate with host susceptibility. Parasitol Res 2024; 123:135. [PMID: 38363354 DOI: 10.1007/s00436-024-08154-9] [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: 10/26/2023] [Accepted: 02/02/2024] [Indexed: 02/17/2024]
Abstract
Opisthorchis viverrini infection is endemic in the lower Mekong subregion. The liver is an organ that worms are drawn to and cause damage. However, the immune-related susceptibility in the liver is poorly understood. In this study, we investigated T helper (Th) cell responses in the liver of BALB/c mice and golden Syrian hamsters during 2-28 days post-infection (DPI). We found that Th cell responses were distinct between mice and hamsters in terms of dynamics and polarization. Mice exhibited the early induction of Th1, Th2, Th17, and regulatory T (Treg) cells responses after the presence of O. viverrini worms at 2 DPI. In hamsters, the late induction of Th1/Th17, downregulation of Th2/Treg responses and early elevation of suppressive cytokine interleukin (IL)-10 were found together with swift reduction of Th cell numbers. Interestingly, expressions of IL-4 (Th2 functional cytokine) and Foxp3 (Treg lineage) were completely different between mice and hamsters which elevated in mice but suppressed in hamsters. These results suggest that early induction and well-regulation are related to host resistance. In contrast, late induction of Th cell response might allow immature worms to develop in the host. Our findings provide a greater understanding in Th cell response-related susceptibility in O. viverrini infection which would be targeting immunity for the development of immune-based intervention such as vaccine.
Collapse
Affiliation(s)
- Pattaraporn Srisai
- Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Tropical Disease Research Center, WHO Collaborating Centre for Research and Control of Opisthorchiasis, Khon Kaen University, Khon Kaen, Thailand
| | - Sujittra Chaiyadet
- Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Tropical Disease Research Center, WHO Collaborating Centre for Research and Control of Opisthorchiasis, Khon Kaen University, Khon Kaen, Thailand
| | - Amonrat Jumnainsong
- The Centre for Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Sutas Suttiprapa
- Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Tropical Disease Research Center, WHO Collaborating Centre for Research and Control of Opisthorchiasis, Khon Kaen University, Khon Kaen, Thailand
| | - Chanvit Leelayuwat
- The Centre for Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Prasert Saichua
- Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.
- Tropical Disease Research Center, WHO Collaborating Centre for Research and Control of Opisthorchiasis, Khon Kaen University, Khon Kaen, Thailand.
| |
Collapse
|
5
|
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
|
6
|
Wang X, Hallen NR, Lee M, Samuchiwal S, Ye Q, Buchheit KM, Maxfield AZ, Roditi RE, Bergmark RW, Bhattacharyya N, Ryan T, Gakpo D, Raychaudhuri S, Dwyer D, Laidlaw TM, Boyce JA, Gutierrez-Arcelus M, Barrett NA. Type 2 inflammation drives an airway basal stem cell program through insulin receptor substrate signaling. J Allergy Clin Immunol 2023; 151:1536-1549. [PMID: 36804595 PMCID: PMC10784786 DOI: 10.1016/j.jaci.2023.01.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 01/17/2023] [Accepted: 01/26/2023] [Indexed: 02/18/2023]
Abstract
BACKGROUND Chronic rhinosinusitis with nasal polyposis (CRSwNP) is a type 2 (T2) inflammatory disease associated with an increased number of airway basal cells (BCs). Recent studies have identified transcriptionally distinct BCs, but the molecular pathways that support or inhibit human BC proliferation and differentiation are largely unknown. OBJECTIVE We sought to determine the role of T2 cytokines in regulating airway BCs. METHODS Single-cell and bulk RNA sequencing of sinus and lung airway epithelial cells was analyzed. Human sinus BCs were stimulated with IL-4 and IL-13 in the presence and absence of inhibitors of IL-4R signaling. Confocal analysis of human sinus tissue and murine airway was performed. Murine BC subsets were sorted for RNA sequencing and functional assays. Fate labeling was performed in a murine model of tracheal injury and regeneration. RESULTS Two subsets of BCs were found in human and murine respiratory mucosa distinguished by the expression of basal cell adhesion molecule (BCAM). BCAM expression identifies airway stem cells among P63+KRT5+NGFR+ BCs. In the sinonasal mucosa, BCAMhi BCs expressing TSLP, IL33, CCL26, and the canonical BC transcription factor TP63 are increased in patients with CRSwNP. In cultured BCs, IL-4/IL-13 increases the expression of BCAM and TP63 through an insulin receptor substrate-dependent signaling pathway that is increased in CRSwNP. CONCLUSIONS These findings establish BCAM as a marker of airway stem cells among the BC pool and demonstrate that airway epithelial remodeling in T2 inflammation extends beyond goblet cell metaplasia to the support of a BC stem state poised to perpetuate inflammation.
Collapse
Affiliation(s)
- Xin Wang
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Nils R Hallen
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Minkyu Lee
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Sachin Samuchiwal
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Qihua Ye
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Kathleen M Buchheit
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Alice Z Maxfield
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women's Hospital, Boston, Mass
| | - Rachel E Roditi
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women's Hospital, Boston, Mass
| | - Regan W Bergmark
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women's Hospital, Boston, Mass
| | - Neil Bhattacharyya
- Department of Otolaryngology, Head and Neck Surgery, Massachusetts Eye and Ear Infirmary, Boston, Mass
| | - Tessa Ryan
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Deb Gakpo
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital, Boston, Mass; Divisions of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass; Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Mass; Versus Arthritis Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Dan Dwyer
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Tanya M Laidlaw
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Joshua A Boyce
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Maria Gutierrez-Arcelus
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Mass; Division of Immunology, Boston Children's Hospital, Boston, Mass
| | - Nora A Barrett
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass.
| |
Collapse
|
7
|
Gologorsky MB, Mechler CM, Forgó E, Charville GW, Howitt MR. The abundance and morphology of human large intestinal goblet and tuft cells during chronic schistosomiasis. Parasite Immunol 2023; 45:e12981. [PMID: 37038837 PMCID: PMC10192203 DOI: 10.1111/pim.12981] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/23/2023] [Accepted: 03/24/2023] [Indexed: 04/12/2023]
Abstract
Schistosomiasis affects nearly 240 million people in predominately low- and middle-income countries and ranks second in the number of cases and socio-economic burden among all parasitic diseases. Despite the enormous burden posed by schistosomes, our understanding of how schistosomiasis impacts infected human tissues remains limited. Intestinal schistosomiasis in animal models leads to goblet cell hyperplasia, likely increasing mucus production and reflecting an intestinal type 2 immune response. However, it is unknown whether these same changes occur in schistosome-infected humans. Using immunofluorescence and light microscopy, we compared the abundance and morphology of goblet cells in patients diagnosed with schistosomiasis to uninfected controls. The mucin-containing vesicles in goblet cells from schistosome-infected patients were significantly larger (hypertrophic) than uninfected individuals, although goblet cell hyperplasia was absent in chronic human schistosomiasis. In addition, we examined tuft cells in the large intestinal epithelium of control and schistosome-infected patients. Tuft cell numbers expand during helminth infection in mice, but these cells have not been characterized in human parasite infections. We found no evidence of tuft cell hyperplasia during human schistosome infection. Thus, our study provides novel insight into schistosome-associated changes to the intestinal epithelium in humans, suggesting an increase in mucus production by large intestinal goblet cells but relatively minor effects on tuft cell numbers.
Collapse
Affiliation(s)
| | | | - Erna Forgó
- Stanford University School of Medicine, Department of Pathology
| | | | - Michael R. Howitt
- Stanford University School of Medicine, Department of Pathology
- Stanford University School of Medicine, Department of Microbiology & Immunology
| |
Collapse
|
8
|
Lo Conte M, Cosorich I, Ferrarese R, Antonini Cencicchio M, Nobili A, Palmieri V, Massimino L, Lamparelli LA, Liang W, Riba M, Devecchi E, Bolla AM, Pedone E, Scavini M, Bosi E, Fasano A, Ungaro F, Diana J, Mancini N, Falcone M. Alterations of the intestinal mucus layer correlate with dysbiosis and immune dysregulation in human Type 1 Diabetes. EBioMedicine 2023; 91:104567. [PMID: 37062177 PMCID: PMC10139895 DOI: 10.1016/j.ebiom.2023.104567] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 03/09/2023] [Accepted: 03/30/2023] [Indexed: 04/18/2023] Open
Abstract
BACKGROUND In preclinical models of Type 1 Diabetes (T1D) the integrity of the gut barrier (GB) is instrumental to avoid dysregulated crosstalk between the commensal microbiota and immune cells and to prevent autoimmunity. The GB is composed of the intestinal epithelial barrier (IEB) and of the mucus layer containing mucins and antimicrobial peptides (AMPs) that are crucial to maintain immune tolerance. In preclinical models of T1D the alterations of the GB primarily affect the mucus layer. In human T1D increased gut permeability and IEB damage have been demonstrated but the integrity of the mucus layer was never assessed. METHODS We evaluated GB integrity by measuring serological markers of IEB damage (serological levels of zonulin) and bacterial translocation such as lipopolysaccharide binding protein (LBP) and myeloid differentiation protein 2 (MD2), and mRNA expression of tight junction proteins, mucins and AMPs in intestinal tissue of T1D patients and healthy controls (HC). Simultaneously, we performed immunological profiling on intestinal tissue and 16S rRNA analysis on the mucus-associated gut microbiota (MAGM). FINDINGS Our data show a GB damage with mucus layer alterations and reduced mRNA expression of several mucins (MUC2, MUC12, MUC13, MUC15, MUC20, MUC21) and AMPs (HD4 and HD5) in T1D patients. Mucus layer alterations correlated with reduced relative abundance of short chain fatty acids (SCFA)-producing bacteria such as Bifidobacterium dentium, Clostridium butyricum and Roseburia intestinalis that regulate mucin expression and intestinal immune homeostasis. In T1D patients we also found intestinal immune dysregulation with higher percentages of effector T cells such as T helper (Th) 1, Th17 and TNF-α+ T cells. INTERPRETATION Our data show that mucus layer alterations are present in T1D subjects and associated with dysbiosis and immune dysregulation. FUNDING Research Grants from the Juvenile Diabetes Foundation (Grant 1-INO-2018-640-A-N to MF and 2-SRA-2019-680-S-B to JD) and from the Italian Ministry of Health (Grant RF19-12370721 to MF).
Collapse
Affiliation(s)
- Marta Lo Conte
- Autoimmune Pathogenesis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ilaria Cosorich
- Autoimmune Pathogenesis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Roberto Ferrarese
- Virology and Microbiology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Martina Antonini Cencicchio
- Autoimmune Pathogenesis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angelica Nobili
- Autoimmune Pathogenesis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vittoria Palmieri
- Autoimmune Pathogenesis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Massimino
- Experimental Gastroenterology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | | | - Michela Riba
- Center for OMICS Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisabetta Devecchi
- Clinical Nutrition Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Mario Bolla
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Erika Pedone
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marina Scavini
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Emanuele Bosi
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy; San Raffaele Vita Salute University, Milan, Italy
| | - Alessio Fasano
- Department of Pediatrics, Harvard Medical School, MA, USA
| | - Federica Ungaro
- Experimental Gastroenterology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Nicasio Mancini
- Virology and Microbiology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; San Raffaele Vita Salute University, Milan, Italy
| | - Marika Falcone
- Autoimmune Pathogenesis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
9
|
Billipp TE, Fung C, Webeck LM, Sargent DB, Gologorsky MB, McDaniel MM, Kasal DN, McGinty JW, Barrow KA, Rich LM, Barilli A, Sabat M, Debley JS, Myers R, Howitt MR, von Moltke J. Tuft cell-derived acetylcholine regulates epithelial fluid secretion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.17.533208. [PMID: 36993541 PMCID: PMC10055254 DOI: 10.1101/2023.03.17.533208] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Tuft cells are solitary chemosensory epithelial cells that can sense lumenal stimuli at mucosal barriers and secrete effector molecules to regulate the physiology and immune state of their surrounding tissue. In the small intestine, tuft cells detect parasitic worms (helminths) and microbe-derived succinate, and signal to immune cells to trigger a Type 2 immune response that leads to extensive epithelial remodeling spanning several days. Acetylcholine (ACh) from airway tuft cells has been shown to stimulate acute changes in breathing and mucocilliary clearance, but its function in the intestine is unknown. Here we show that tuft cell chemosensing in the intestine leads to release of ACh, but that this does not contribute to immune cell activation or associated tissue remodeling. Instead, tuft cell-derived ACh triggers immediate fluid secretion from neighboring epithelial cells into the intestinal lumen. This tuft cell-regulated fluid secretion is amplified during Type 2 inflammation, and helminth clearance is delayed in mice lacking tuft cell ACh. The coupling of the chemosensory function of tuft cells with fluid secretion creates an epithelium-intrinsic response unit that effects a physiological change within seconds of activation. This response mechanism is shared by tuft cells across tissues, and serves to regulate the epithelial secretion that is both a hallmark of Type 2 immunity and an essential component of homeostatic maintenance at mucosal barriers.
Collapse
Affiliation(s)
- Tyler E. Billipp
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Connie Fung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lily M. Webeck
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Derek B. Sargent
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Matthew B. Gologorsky
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Margaret M. McDaniel
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Darshan N. Kasal
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - John W. McGinty
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Kaitlyn A. Barrow
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Lucille M. Rich
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
| | | | - Mark Sabat
- Takeda Pharmaceuticals, San Diego, California, USA
| | - Jason S. Debley
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Seattle Children’s Hospital, University of Washington, Seattle, WA, USA
| | | | - Michael R. Howitt
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jakob von Moltke
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
10
|
Hao HN, Lu QQ, Wang Z, Li YL, Long SR, Dan Liu R, Cui J, Wang ZQ. Mannose facilitates Trichinella spiralis expulsion from the gut and alleviates inflammation of intestines and muscles in mice. Acta Trop 2023; 241:106897. [PMID: 36931335 DOI: 10.1016/j.actatropica.2023.106897] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/25/2022] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
Trichinellosis is a major zoonotic parasitosis which is a vital risk to meat food safety. It is requisite to exploit new strategy to interdict food animal Trichinella infection and to obliterate Trichinella from food animals to ensure meat safety. Mannose is an oligosaccharide that specifically binds to the carbohydrate-recognition domain of C-type lectin; it has many physiological functions including reliving inflammation and regulating immune reaction. The purpose of this study was to investigate the suppressive role of mannose on T. spiralis larval invasion and infection, its effect on intestinal and muscle inflammation, and immune responses after challenge. The results showed that compared to the saline-treated infected mice, the mannose-treated infected mice had less intestinal adult and muscle worm burdens, mild inflammation of intestine and muscle of infected mice. The levels of specific anti-Trichinella IgG (IgG1/IgG2a), IgA and sIgA in mannose-treated infected mice were obviously inferior to saline-treated infected mice (P < 0.01). Furthermore, the levels of two cytokines (IFN-γ and IL-4) in mannose-treated infected mice were also significantly lower than the saline-treated infected mice (P < 0.01). The protective effect of the mannose against Trichinella infection might be not related to specific antibody and cellular immune responses. The above results demonstrated that mannose could be considered as a novel adjuvant therapeutic agent for anti-Trichinella drugs to block larval invasion at early stage of Trichinella infection.
Collapse
Affiliation(s)
- Hui Nan Hao
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, China
| | - Qi Qi Lu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, China
| | - Zhen Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, China
| | - Yang Li Li
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, China
| | - Shao Rong Long
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, China
| | - Ruo Dan Liu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, China
| | - Jing Cui
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, China.
| | - Zhong Quan Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
11
|
Ariyaratne A, Kim SY, Pollo SMJ, Perera S, Liu H, Nguyen WNT, Coria AL, Luzzi MDC, Bowron J, Szabo EK, Patel KD, Wasmuth JD, Nair MG, Finney CAM. Trickle infection with Heligmosomoides polygyrus results in decreased worm burdens but increased intestinal inflammation and scarring. Front Immunol 2022; 13:1020056. [PMID: 36569914 PMCID: PMC9773095 DOI: 10.3389/fimmu.2022.1020056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 11/03/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction Intestinal roundworms cause chronic debilitating disease in animals, including humans. Traditional experimental models of these types of infection use a large single-dose infection. However, in natural settings, hosts are exposed to parasites on a regular basis and when mice are exposed to frequent, smaller doses of Heligmosomoides polygyrus, the parasites are cleared more quickly. Whether this more effective host response has any negative consequences for the host is not known. Results Using a trickle model of infection, we found that worm clearance was associated with known resistance-related host responses: increased granuloma and tuft cell numbers, increased levels of granuloma IgG and decreased intestinal transit time, as well as higher serum IgE levels. However, we found that the improved worm clearance was also associated with an inflammatory phenotype in and around the granuloma, increased smooth muscle hypertrophy/hyperplasia, and elevated levels of Adamts gene expression. Discussion To our knowledge, we are the first to identify the involvement of this protein family of matrix metalloproteinases (MMPs) in host responses to helminth infections. Our results highlight the delicate balance between parasite clearance and host tissue damage, which both contribute to host pathology. When continually exposed to parasitic worms, improved clearance comes at a cost.
Collapse
Affiliation(s)
- Anupama Ariyaratne
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
| | - Sang Yong Kim
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, United States
| | - Stephen M. J. Pollo
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Shashini Perera
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
| | - Hongrui Liu
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
| | - William N. T. Nguyen
- Departments of Physiology and Pharmacology, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - Aralia Leon Coria
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
| | - Mayara de Cassia Luzzi
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
| | - Joel Bowron
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
| | - Edina K. Szabo
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
| | - Kamala D. Patel
- Departments of Physiology and Pharmacology, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - James D. Wasmuth
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Meera G. Nair
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, United States
| | - Constance A. M. Finney
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
12
|
Schumacher MA, Liu CY, Katada K, Thai MH, Hsieh JJ, Hansten BJ, Waddell A, Rosen MJ, Frey MR. Deep Crypt Secretory Cell Differentiation in the Colonic Epithelium Is Regulated by Sprouty2 and Interleukin 13. Cell Mol Gastroenterol Hepatol 2022; 15:971-984. [PMID: 36414210 PMCID: PMC9982040 DOI: 10.1016/j.jcmgh.2022.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND & AIMS Deep crypt secretory (DCS) cells are a critical component of the colonic stem cell niche. However, the regulatory mechanisms controlling DCS cell numbers and function are not well understood. Sprouty2 is an inflammation-responsive regulator of intracellular signaling that influences colonic secretory cell numbers in colitis via an epithelial-stromal interleukin (IL)33/IL13 signaling loop. Here, we tested the hypothesis that IL13, induced by epithelial Sprouty2 down-regulation, promotes DCS cell differentiation and function. METHODS Distal colons from mice with an intestinal epithelial-specific Sprouty2 deletion (Spry2ΔIE) and littermate controls were analyzed by in situ hybridization for Reg4+ DCS cells. Single-cell RNA sequencing and immunostaining were used to identify DCS cell-derived host defense peptides (HDPs) and localization of IL13 and IL13 receptor; bulk RNA sequencing and quantitative polymerase chain reaction were used to quantify changes in expression of identified HDPs. Cytokine-treated colonoids were assessed for DCS cells. A requirement for an IL33/IL13 signaling loop in the regulation of DCS cells was assessed in vivo using IL13 null mice. RESULTS Reg4+ DCS cell numbers were increased 2-fold in distal colons of Spry2ΔIE mice with a concomitant overall increase in DCS cell marker expression (Reg4, Spink4, and Agr2). Single-cell transcriptomics showed the HDP Retnlb/Resistin Like Beta (RELMβ) is highly enriched in DCS cells. Retnlb/RELMβ expression was increased in Spry2ΔIE colons. IL13, but not IL33, induced Reg4 and Retnlb expression in colonic epithelial organoids, and IL33-mediated expansion of the DCS cell population in vivo was dependent on IL13, which was expressed predominantly by type II innate lymphoid cells in the colonic mucosa. CONCLUSIONS Sprouty2 limits colonic DCS cell differentiation through suppression of IL13 signaling. At homeostasis, DCS cells are marked by high levels of the HDP RELMβ. Loss of epithelial Sprouty2 activates type II innate lymphoid cells to release IL13, promoting expansion of the DCS cell population and increased colonic RELMβ levels.
Collapse
Affiliation(s)
- Michael A Schumacher
- Department of Pediatrics, University of Southern California Keck School of Medicine, Los Angeles, California; The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California.
| | - Cambrian Y Liu
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Kay Katada
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Megan H Thai
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Jonathan J Hsieh
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Britany J Hansten
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Amanda Waddell
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Michael J Rosen
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Mark R Frey
- Department of Pediatrics, University of Southern California Keck School of Medicine, Los Angeles, California; The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California; Department of Biochemistry and Molecular Medicine, University of Southern California Keck School of Medicine, Los Angeles, California.
| |
Collapse
|
13
|
Therapeutic Effect and Immune Changes after Treatment of Hymenolepis nana-Infected BALB/c Mice with Compounds Isolated from Leucaena leucocephala. Vet Sci 2022; 9:vetsci9070368. [PMID: 35878385 PMCID: PMC9316369 DOI: 10.3390/vetsci9070368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/07/2022] [Accepted: 07/16/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary This article is mainly about the development of natural medicines against the zoonotic tapeworm (Hymenolepis nana), which is caused by tapeworm eggs in infected mice, through the isolation of the components of jumbay (Leucaena leucocephala). This study allowed for an understanding of the drug therapy through the testing of these isolated components in vitro and in vivo. The authors assessed the survival and motility, and they confirmed that the two components had good effects against tapeworm. One of these components has a promising therapeutic effect in tapeworm-infected mice in the response to egg production, worm numbers, and immune responses. This article highlights the immune-enhancing and therapeutic effects of 132-hydroxy-(132-S)-pheophytin a against tapeworms in infected mice. Abstract Background/Purpose: Hymenolepis nana is globally distributed. Leucaena leucocephala has been studied as a treatment, including the nematodes and protozoa, but no research results are related to cestodes. Therefore, the aim of this study was to target H. nana. Methods: The natural components of L. leucocephala were isolated and added to H. nana, which was cultured in vitro, to observe changes in the mortality, motility, and morphology. BALB/c male mice infected with H. nana were treated with effective components of L. leucocephala for 10 days, and the changes were recorded. After the mice were sacrificed, the spleen weight was measured, and a primary culture was performed for the subsequent cytokine and chemokine testing. Results: The experiment found that 132-hydroxy-(132-S)-pheophytin a and aristophyll-C have clear cestocidal effects in vitro. 132-hydroxy-(132-S)-pheophytin a has been shown to be effective at reducing parasite populations and eliciting host immune responses in vivo. IL-2, IL-4, IL-5, IL-6, IL-10, IL-13, IL-17, MCP-1, IFN-γ, TNF-α, MIP-1α, and GM-CSF in 132-hydroxy-(132-S)-pheophytin a were significantly increased after stimulation, while IL-1α, IL-1β, IL-3, IL-12p70, and RANTES were unchanged. Conclusions: The investigation shows that components of L. leucocephala have actual cestocidal activity against H. nana.
Collapse
|
14
|
Vacca F, Le Gros G. Tissue-specific immunity in helminth infections. Mucosal Immunol 2022; 15:1212-1223. [PMID: 35680972 PMCID: PMC9178325 DOI: 10.1038/s41385-022-00531-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/25/2022] [Accepted: 05/16/2022] [Indexed: 02/04/2023]
Abstract
A characteristic feature of host responses to helminth infections is the development of profound systemic and tissue-localised Type 2 immune responses that play critical roles in immunity, tissue repair and tolerance of the parasite at tissue sites. These same Type 2 responses are also seen in the tissue-associated immune-pathologies seen in asthma, atopic dermatitis and many forms of allergies. The recent identification of new subtypes of immune cells and cytokine pathways that influence both immune and non-immune cells and tissues creates the opportunity for reviewing helminth parasite-host responses in the context of tissue specific immunity. This review focuses on the new discoveries of the cells and cytokines involved in tissue specific immune responses to helminths and how these contribute to host immunity against helminth infection and allow the host to accommodate the presence of parasites when they cannot be eliminated.
Collapse
Affiliation(s)
- Francesco Vacca
- grid.250086.90000 0001 0740 0291Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Graham Le Gros
- grid.250086.90000 0001 0740 0291Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
15
|
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: 2.3] [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
|
16
|
The Interplay between Nutrition, Innate Immunity, and the Commensal Microbiota in Adaptive Intestinal Morphogenesis. Nutrients 2021; 13:nu13072198. [PMID: 34206809 PMCID: PMC8308283 DOI: 10.3390/nu13072198] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/20/2021] [Accepted: 06/23/2021] [Indexed: 12/15/2022] Open
Abstract
The gastrointestinal tract is a functionally and anatomically segmented organ that is colonized by microbial communities from birth. While the genetics of mouse gut development is increasingly understood, how nutritional factors and the commensal gut microbiota act in concert to shape tissue organization and morphology of this rapidly renewing organ remains enigmatic. Here, we provide an overview of embryonic mouse gut development, with a focus on the intestinal vasculature and the enteric nervous system. We review how nutrition and the gut microbiota affect the adaptation of cellular and morphologic properties of the intestine, and how these processes are interconnected with innate immunity. Furthermore, we discuss how nutritional and microbial factors impact the renewal and differentiation of the epithelial lineage, influence the adaptation of capillary networks organized in villus structures, and shape the enteric nervous system and the intestinal smooth muscle layers. Intriguingly, the anatomy of the gut shows remarkable flexibility to nutritional and microbial challenges in the adult organism.
Collapse
|
17
|
Darlan DM, Rozi MF, Yulfi H. Overview of Immunological Responses and Immunomodulation Properties of Trichuris sp.: Prospects for Better Understanding Human Trichuriasis. Life (Basel) 2021; 11:188. [PMID: 33673676 PMCID: PMC7997218 DOI: 10.3390/life11030188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/17/2021] [Accepted: 02/24/2021] [Indexed: 12/23/2022] Open
Abstract
Trichuris sp. infection has appeared as a pathological burden in the population, but the immunomodulation features could result in an opportunity to discover novel treatments for diseases with prominent inflammatory responses. Regarding the immunological aspects, the innate immune responses against Trichuris sp. are also responsible for determining subsequent immune responses, including the activation of innate lymphoid cell type 2 (ILC2s), and encouraging the immune cell polarization of the resistant host phenotype. Nevertheless, this parasite can establish a supportive niche for worm survival and finally avoid host immune interference. Trichuris sp. could skew antigen recognition and immune cell activation and proliferation through the generation of specific substances, called excretory/secretory (ESPs) and soluble products (SPs), which mainly mediate its immunomodulation properties. Through this review, we elaborate and discuss innate-adaptive immune responses and immunomodulation aspects, as well as the clinical implications for managing inflammatory-based diseases, such as inflammatory bowel diseases, allergic, sepsis, and other autoimmune diseases.
Collapse
Affiliation(s)
- Dewi Masyithah Darlan
- Department of Parasitology, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia; (D.M.D.); (H.Y.)
| | | | - Hemma Yulfi
- Department of Parasitology, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia; (D.M.D.); (H.Y.)
| |
Collapse
|
18
|
Zhang M, Wu C. The relationship between intestinal goblet cells and the immune response. Biosci Rep 2020; 40:BSR20201471. [PMID: 33017020 PMCID: PMC7569202 DOI: 10.1042/bsr20201471] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 02/06/2023] Open
Abstract
Goblet cells (GCs) are single-cell glands that produce and secrete mucin. Mucin forms a mucus layer, which can separate the materials in cavities from the intestinal epithelium and prevent the invasion of pathogenic microorganisms in various ways. GCs can also participate in the immune response through nonspecific endocytosis and goblet cell-associated antigen passages (GAPs). GCs endocytose soluble substances from the lumen and transmit antigens to the underlying antigen-presenting cells (APCs). A variety of immuno-regulatory factors can promote the differentiation, maturation of GCs, and the secretion of mucin. The mucin secreted by GCs forms a mucus layer, which plays an important role in resisting the invasion of foreign bacteria and intestinal inherent microorganisms, regulating the immune performance of the body. Therefore, the present study mainly reviews the barrier function of the mucus layer, the mucus secreted by goblet cells, the protective effect against pathogenic bacteria, the delivery of luminal substances through GAPs and the relationship between GCs and the immune response.
Collapse
Affiliation(s)
- Mingming Zhang
- College of Animal Veterinary Medicine, Northwest A & F University, Yangling 712100, Shaanxi, People’s Republic of China
| | - Chenchen Wu
- College of Animal Veterinary Medicine, Northwest A & F University, Yangling 712100, Shaanxi, People’s Republic of China
| |
Collapse
|
19
|
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: 210] [Impact Index Per Article: 52.5] [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
|
20
|
Pionnier N, Sjoberg H, Furlong-Silva J, Marriott A, Halliday A, Archer J, Steven A, Taylor MJ, Turner JD. Eosinophil-Mediated Immune Control of Adult Filarial Nematode Infection Can Proceed in the Absence of IL-4 Receptor Signaling. THE JOURNAL OF IMMUNOLOGY 2020; 205:731-740. [PMID: 32571840 PMCID: PMC7372315 DOI: 10.4049/jimmunol.1901244] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 05/17/2020] [Indexed: 12/19/2022]
Abstract
Immunity to chronic filarial worm infection is apparent in IL-4Rα–deficient mice. Delayed immunity in IL-4Rα−/− mice is due to suboptimal tissue eosinophilia. Eosinophil recruitment in the absence of IL-4R signaling requires CCR3 and IL-5.
Helminth infections are accompanied by eosinophilia in parasitized tissues. Eosinophils are effectors of immunity to tissue helminths. We previously reported that in the context of experimental filarial nematode infection, optimum tissue eosinophil recruitment was coordinated by local macrophage populations following IL-4R–dependent in situ proliferation and alternative activation. However, in the current study, we identify that control of chronic adult filarial worm infection is evident in IL-4Rα–deficient (IL-4Rα−/−) mice, whereby the majority of infections do not achieve patency. An associated residual eosinophilia was apparent in infected IL-4Rα−/− mice. By treating IL-4Rα−/− mice serially with anti-CCR3 Ab or introducing a compound deficiency in CCR3 within IL-4Rα−/− mice, residual eosinophilia was ablated, and susceptibility to chronic adult Brugia malayi infection was established, promoting a functional role for CCR3-dependent eosinophil influx in immune control in the absence of IL-4/IL-13–dependent immune mechanisms. We investigated additional cytokine signals involved in residual eosinophilia in the absence IL-4Rα signaling and defined that IL-4Rα−/−/IL-5−/− double-knockout mice displayed significant eosinophil deficiency compared with IL-4Rα−/− mice and were susceptible to chronic fecund adult filarial infections. Contrastingly, there was no evidence that either IL-4R–dependent or IL-4R–independent/CCR3/IL-5–dependent immunity influenced B. malayi microfilarial loads in the blood. Our data demonstrate multiplicity of Th2-cytokine control of eosinophil tissue recruitment during chronic filarial infection and that IL-4R–independent/IL-5– and CCR3-dependent pathways are sufficient to control filarial adult infection via an eosinophil-dependent effector response prior to patency.
Collapse
Affiliation(s)
- Nicolas Pionnier
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool L3 5QA, United Kingdom
| | - Hanna Sjoberg
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool L3 5QA, United Kingdom
| | - Julio Furlong-Silva
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool L3 5QA, United Kingdom
| | - Amy Marriott
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool L3 5QA, United Kingdom
| | - Alice Halliday
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool L3 5QA, United Kingdom
| | - John Archer
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool L3 5QA, United Kingdom
| | - Andrew Steven
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool L3 5QA, United Kingdom
| | - Mark J Taylor
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool L3 5QA, United Kingdom
| | - Joseph D Turner
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool L3 5QA, United Kingdom
| |
Collapse
|
21
|
Ryan N, Anderson K, Volpedo G, Varikuti S, Satoskar M, Satoskar S, Oghumu S. The IL-33/ST2 Axis in Immune Responses Against Parasitic Disease: Potential Therapeutic Applications. Front Cell Infect Microbiol 2020; 10:153. [PMID: 32363166 PMCID: PMC7180392 DOI: 10.3389/fcimb.2020.00153] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 03/23/2020] [Indexed: 12/15/2022] Open
Abstract
Parasitic infections pose a wide and varying threat globally, impacting over 25% of the global population with many more at risk of infection. These infections are comprised of, but not limited to, toxoplasmosis, malaria, leishmaniasis and any one of a wide variety of helminthic infections. While a great deal is understood about the adaptive immune response to each of these parasites, there remains a need to further elucidate the early innate immune response. Interleukin-33 is being revealed as one of the earliest players in the cytokine milieu responding to parasitic invasion, and as such has been given the name "alarmin." A nuclear cytokine, interleukin-33 is housed primarily within epithelial and fibroblastic tissues and is released upon cellular damage or death. Evidence has shown that interleukin-33 seems to play a crucial role in priming the immune system toward a strong T helper type 2 immune response, necessary in the clearance of some parasites, while disease exacerbating in the context of others. With the possibility of being a double-edged sword, a great deal remains to be seen in how interleukin-33 and its receptor ST2 are involved in the immune response different parasites elicit, and how those parasites may manipulate or evade this host mechanism. In this review article we compile the current cutting-edge research into the interleukin-33 response to toxoplasmosis, malaria, leishmania, and helminthic infection. Furthermore, we provide insight into directions interleukin-33 research may take in the future, potential immunotherapeutic applications of interleukin-33 modulation and how a better clarity of early innate immune system responses involving interleukin-33/ST2 signaling may be applied in development of much needed treatment options against parasitic invaders.
Collapse
Affiliation(s)
- Nathan Ryan
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Division of Anatomy, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Kelvin Anderson
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Greta Volpedo
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Microbiology, The Ohio State University, Columbus, OH, United States
| | - Sanjay Varikuti
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Monika Satoskar
- Northeast Ohio Medical University, Rootstown, OH, United States
| | - Sanika Satoskar
- Northeast Ohio Medical University, Rootstown, OH, United States
| | - Steve Oghumu
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
22
|
Engevik MA, Luk B, Chang-Graham AL, Hall A, Herrmann B, Ruan W, Endres BT, Shi Z, Garey KW, Hyser JM, Versalovic J. Bifidobacterium dentium Fortifies the Intestinal Mucus Layer via Autophagy and Calcium Signaling Pathways. mBio 2019; 10:e01087-19. [PMID: 31213556 PMCID: PMC6581858 DOI: 10.1128/mbio.01087-19] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 05/11/2019] [Indexed: 02/07/2023] Open
Abstract
Much remains unknown about how the intestinal microbiome interfaces with the protective intestinal mucus layer. Bifidobacterium species colonize the intestinal mucus layer and can modulate mucus production by goblet cells. However, select Bifidobacterium strains can also degrade protective glycans on mucin proteins. We hypothesized that the human-derived species Bifidobacterium dentium would increase intestinal mucus synthesis and expulsion, without extensive degradation of mucin glycans. In silico data revealed that B. dentium lacked the enzymes necessary to extensively degrade mucin glycans. This finding was confirmed by demonstrating that B. dentium could not use naive mucin glycans as primary carbon sources in vitro To examine B. dentium mucus modulation in vivo, Swiss Webster germfree mice were monoassociated with live or heat-killed B. dentium Live B. dentium-monoassociated mice exhibited increased colonic expression of goblet cell markers Krüppel-like factor 4 (Klf4), Trefoil factor 3 (Tff3), Relm-β, Muc2, and several glycosyltransferases compared to both heat-killed B. dentium and germfree counterparts. Likewise, live B. dentium-monoassociated colon had increased acidic mucin-filled goblet cells, as denoted by Periodic Acid-Schiff-Alcian Blue (PAS-AB) staining and MUC2 immunostaining. In vitro, B. dentium-secreted products, including acetate, were able to increase MUC2 levels in T84 cells. We also identified that B. dentium-secreted products, such as γ-aminobutyric acid (GABA), stimulated autophagy-mediated calcium signaling and MUC2 release. This work illustrates that B. dentium is capable of enhancing the intestinal mucus layer and goblet cell function via upregulation of gene expression and autophagy signaling pathways, with a net increase in mucin production.IMPORTANCE Microbe-host interactions in the intestine occur along the mucus-covered epithelium. In the gastrointestinal tract, mucus is composed of glycan-covered proteins, or mucins, which are secreted by goblet cells to form a protective gel-like structure above the epithelium. Low levels of mucin or alterations in mucin glycans are associated with inflammation and colitis in mice and humans. Although current literature links microbes to the modulation of goblet cells and mucins, the molecular pathways involved are not yet fully understood. Using a combination of gnotobiotic mice and mucus-secreting cell lines, we have identified a human-derived microbe, Bifidobacterium dentium, which adheres to intestinal mucus and secretes metabolites that upregulate the major mucin MUC2 and modulate goblet cell function. Unlike other Bifidobacterium species, B. dentium does not extensively degrade mucin glycans and cannot grow on mucin alone. This work points to the potential of using B. dentium and similar mucin-friendly microbes as therapeutic agents for intestinal disorders with disruptions in the mucus barrier.
Collapse
Affiliation(s)
- Melinda A Engevik
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Berkley Luk
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Alexandra L Chang-Graham
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Anne Hall
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Beatrice Herrmann
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Wenly Ruan
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Bradley T Endres
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, Texas, USA
| | - Zhongcheng Shi
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Kevin W Garey
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, Texas, USA
| | - Joseph M Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - James Versalovic
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| |
Collapse
|
23
|
Tougaard P, Martinsen LO, Zachariassen LF, Krych L, Nielsen DS, Buus TB, Pedersen AE, Hansen AK, Skov S, Hansen CHF. TL1A Aggravates Cytokine-Induced Acute Gut Inflammation and Potentiates Infiltration of Intraepithelial Natural Killer Cells in Mice. Inflamm Bowel Dis 2019; 25:510-523. [PMID: 30462201 DOI: 10.1093/ibd/izy351] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND The tumor necrosis factor alpha (TNFα)-homologous cytokine TL1A is emerging as a major player in intestinal inflammation. From in vitro experiments on human lymphocytes, TNF-like molecule 1A (TL1A) is known to activate a highly inflammatory lymphoid response in synergy with interleukin (IL)-12 and IL-18. Carriers of specific genetic polymorphisms associated with IL-12, IL-18, or TL1A signaling have increased Crohn's disease risk, and all 3 cytokines are upregulated during active disease. The study aim was to investigate whether the type 1-polarizing cytokines IL-12 and IL-18 could directly initiate intestinal pathology in mice and how TL1A would influence the resulting inflammatory response. METHODS Conventional barrier-bred and germ-free mice were randomly allocated to different groups and injected twice with different combinations of IL-12, IL-18, and TL1A, and killed 3 days after the first injection. All treatment groups were co-housed and fed a piroxicam-supplemented chow diet. RESULTS Intestinal pathology was evident in IL-12- and IL-18-treated mice and highly exacerbated by TL1A in both the colon and ileum. The cytokine-induced intestinal inflammation was characterized by epithelial damage, increased colonic levels of TNFα, IL-1β, IFN-γ, and IL-6, and various chemokines along with gut microbiota alterations exhibiting high abundance of Enterobacteriaceae. Furthermore, the inflamed ileum and colon exhibited a TL1A-specific increased infiltration of intraepithelial natural killer cells co-expressing NKG2D and IL-18Ra and a higher frequency of unconventional T cells in the colonic epithelium. Upon cytokine injection, germ-free mice exhibited similar intraepithelial lymphoid infiltration and increased colonic levels of IFNγ and TNFα. CONCLUSIONS This study demonstrates that TL1A aggravates IL-12- and IL-18-induced intestinal inflammation in the presence and absence of microbiota.
Collapse
Affiliation(s)
- Peter Tougaard
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.,Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Louise Otterstrøm Martinsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Line Fisker Zachariassen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Lukasz Krych
- Department of Food Science, Faculty of Science, University of Copenhagen, Denmark
| | | | - Terkild Brink Buus
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Anders Elm Pedersen
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Axel Kornerup Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Søren Skov
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Camilla Hartmann Friis Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
24
|
Duque-Correa MA, Karp NA, McCarthy C, Forman S, Goulding D, Sankaranarayanan G, Jenkins TP, Reid AJ, Cambridge EL, Ballesteros Reviriego C, Müller W, Cantacessi C, Dougan G, Grencis RK, Berriman M. Exclusive dependence of IL-10Rα signalling on intestinal microbiota homeostasis and control of whipworm infection. PLoS Pathog 2019; 15:e1007265. [PMID: 30640950 PMCID: PMC6347331 DOI: 10.1371/journal.ppat.1007265] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 01/25/2019] [Accepted: 12/04/2018] [Indexed: 12/28/2022] Open
Abstract
The whipworm Trichuris trichiura is a soil-transmitted helminth that dwells in the epithelium of the caecum and proximal colon of their hosts causing the human disease, trichuriasis. Trichuriasis is characterized by colitis attributed to the inflammatory response elicited by the parasite while tunnelling through intestinal epithelial cells (IECs). The IL-10 family of receptors, comprising combinations of subunits IL-10Rα, IL-10Rβ, IL-22Rα and IL-28Rα, modulates intestinal inflammatory responses. Here we carefully dissected the role of these subunits in the resistance of mice to infection with T. muris, a mouse model of the human whipworm T. trichiura. Our findings demonstrate that whilst IL-22Rα and IL-28Rα are dispensable in the host response to whipworms, IL-10 signalling through IL-10Rα and IL-10Rβ is essential to control caecal pathology, worm expulsion and survival during T. muris infections. We show that deficiency of IL-10, IL-10Rα and IL-10Rβ results in dysbiosis of the caecal microbiota characterised by expanded populations of opportunistic bacteria of the families Enterococcaceae and Enterobacteriaceae. Moreover, breakdown of the epithelial barrier after whipworm infection in IL-10, IL-10Rα and IL-10Rβ-deficient mice, allows the translocation of these opportunistic pathogens or their excretory products to the liver causing organ failure and lethal disease. Importantly, bone marrow chimera experiments indicate that signalling through IL-10Rα and IL-10Rβ in haematopoietic cells, but not IECs, is crucial to control worm expulsion and immunopathology. These findings are supported by worm expulsion upon infection of conditional mutant mice for the IL-10Rα on IECs. Our findings emphasize the pivotal and complex role of systemic IL-10Rα signalling on immune cells in promoting microbiota homeostasis and maintaining the intestinal epithelial barrier, thus preventing immunopathology during whipworm infections.
Collapse
Affiliation(s)
| | - Natasha A Karp
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Catherine McCarthy
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Simon Forman
- 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, United Kingdom
| | - David Goulding
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | | | - Timothy P Jenkins
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Adam J Reid
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Emma L Cambridge
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | | | - Werner Müller
- Lydia Becker Institute of Immunology and Inflammation and Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Cinzia Cantacessi
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Gordon Dougan
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - 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, United Kingdom
| | - Matthew Berriman
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| |
Collapse
|
25
|
Rausch S, Midha A, Kuhring M, Affinass N, Radonic A, Kühl AA, Bleich A, Renard BY, Hartmann S. Parasitic Nematodes Exert Antimicrobial Activity and Benefit From Microbiota-Driven Support for Host Immune Regulation. Front Immunol 2018; 9:2282. [PMID: 30349532 PMCID: PMC6186814 DOI: 10.3389/fimmu.2018.02282] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/14/2018] [Indexed: 12/04/2022] Open
Abstract
Intestinal parasitic nematodes live in intimate contact with the host microbiota. Changes in the microbiome composition during nematode infection affect immune control of the parasites and shifts in the abundance of bacterial groups have been linked to the immunoregulatory potential of nematodes. Here we asked if the small intestinal parasite Heligmosomoides polygyrus produces factors with antimicrobial activity, senses its microbial environment and if the anti-nematode immune and regulatory responses are altered in mice devoid of gut microbes. We found that H. polygyrus excretory/secretory products exhibited antimicrobial activity against gram+/− bacteria. Parasites from germ-free mice displayed alterations in gene expression, comprising factors with putative antimicrobial functions such as chitinase and lysozyme. Infected germ-free mice developed increased small intestinal Th2 responses coinciding with a reduction in local Foxp3+RORγt+ regulatory T cells and decreased parasite fecundity. Our data suggest that nematodes sense their microbial surrounding and have evolved factors that limit the outgrowth of certain microbes. Moreover, the parasites benefit from microbiota-driven immune regulatory circuits, as an increased ratio of intestinal Th2 effector to regulatory T cells coincides with reduced parasite fitness in germ-free mice.
Collapse
Affiliation(s)
- Sebastian Rausch
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Ankur Midha
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Matthias Kuhring
- Bioinformatics Unit (MF 1), Robert Koch Institute, Berlin, Germany.,Core Unit Bioinformatics, Berlin Institute of Health (BIH), Berlin, Germany.,Berlin Institute of Health Metabolomics Platform, Berlin Institute of Health (BIH), Berlin, Germany.,Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Nicole Affinass
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Aleksandar Radonic
- Centre for Biological Threats and Special Pathogens (ZBS 1), Robert Koch Institute, Berlin, Germany.,Genome Sequencing Unit (MF 2), Robert Koch Institute, Berlin, Germany
| | - Anja A Kühl
- iPATH.Berlin, Core Unit for Immunopathology for Experimental Models, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - André Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | | | - Susanne Hartmann
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
26
|
Brosschot TP, Reynolds LA. The impact of a helminth-modified microbiome on host immunity. Mucosal Immunol 2018; 11:1039-1046. [PMID: 29453411 DOI: 10.1038/s41385-018-0008-5] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 12/19/2017] [Accepted: 12/22/2017] [Indexed: 02/04/2023]
Abstract
Intestinal helminths have well-characterized modulatory effects on mammalian immune pathways. Ongoing helminth infection has been associated with both the suppression of allergies and an altered susceptibility to microbial infections. Enteric helminths share a niche with the intestinal microbiota, and the presence of helminths alters the microbiota composition and the metabolic signature of the host. Recent studies have demonstrated that the helminth-modified intestinal microbiome has the capacity to modify host immune responses even in the absence of live helminth infection. This article discusses the mechanisms by which helminths modify the intestinal microbiome of mammals, and reviews the evidence for a helminth-modified microbiome directly influencing host immunity during infectious and inflammatory diseases. Understanding the multifaceted mechanisms that underpin helminth immunomodulation will pave the way for novel therapies to combat infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Tara P Brosschot
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, V8P 5C2, Canada
| | - Lisa A Reynolds
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, V8P 5C2, Canada.
| |
Collapse
|
27
|
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: 64] [Impact Index Per Article: 10.7] [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
|
28
|
Volpe EA, Henriksson JT, Wang C, Barbosa FL, Zaheer M, Zhang X, Pflugfelder SC, de Paiva CS. Interferon-gamma deficiency protects against aging-related goblet cell loss. Oncotarget 2018; 7:64605-64614. [PMID: 27623073 PMCID: PMC5323102 DOI: 10.18632/oncotarget.11872] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/01/2016] [Indexed: 11/25/2022] Open
Abstract
Aging is a well-recognized risk factor for dry eye. Interferon-gamma (IFN-γ) has been implicated in conjunctival keratinization and goblet cell loss in dry eye. We investigated the role of IFN-γ in age-related dry eye by evaluating young (8 weeks) and aged (15 months; 15M) C57BL/6 (B6) and IFN-γKO mice. Age effects on the conjunctiva and cornea epithelium were assessed with PAS staining and corneal staining, respectively. Expression of T cell-related cytokines (IL-17A, IFN-γ), chemokines (CXCL10 and CCL20), in the ocular surface epithelium was evaluated by real time PCR. A significant decrease in filled goblet cells was noted in 15M B6 mice and this was significantly lower than age and sex-matched IFN-γKO mice. Aged male B6 had significantly higher IFN-γ, and CXCL10 mRNA in their conjunctiva than female B6 mice. Aged IFN-γKO females had significantly higher IL-17A mRNA in conjunctiva than IFN-γKO males and B6 mice. Corneal barrier dysfunction was observed in 15M female B6 and aged IFN-γKO mice of both sexes; however it was significantly higher in IFN-γKO compared to B6 mice. While there was a significant increase in IL 17A, and CCL20 in corneas of aged female B6 and IFN-γKO mice compared to males, these changes were more evident in aged female IFN-γKO group. Partial resistance of IFN-γKO mice to aging-induced goblet cell loss indicates IFN-γ is involved in the age-related decline in conjunctival goblet cells. Increased corneal IL-17A expression paralleled corneal barrier disruption in aging female of both strains. IFN-γ appears to suppress IL-17A on the ocular surface.
Collapse
Affiliation(s)
- Eugene A Volpe
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, Texas, USA
| | - Johanna Tukler Henriksson
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, Texas, USA
| | - Changjun Wang
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, Texas, USA.,Eye Center, Second Affiliated Hospital of Zhejiang University, School of Medicine Zhejiang Provincial Key Laboratory of Ophthalmology, Hangzhou, Zhejiang, China
| | - Flavia L Barbosa
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, Texas, USA
| | - Mahira Zaheer
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, Texas, USA
| | - Xiaobo Zhang
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, Texas, USA.,Eye Institute of Xiamen University, Xiamen, Fujian, China
| | - Stephen C Pflugfelder
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, Texas, USA
| | - Cintia S de Paiva
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
29
|
Pierce ES. Could Mycobacterium avium subspecies paratuberculosis cause Crohn's disease, ulcerative colitis…and colorectal cancer? Infect Agent Cancer 2018; 13:1. [PMID: 29308085 PMCID: PMC5753485 DOI: 10.1186/s13027-017-0172-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 12/12/2017] [Indexed: 01/07/2023] Open
Abstract
Infectious agents are known causes of human cancers. Schistosoma japonicum and Schistosoma mansoni cause a percentage of colorectal cancers in countries where the respective Schistosoma species are prevalent. Colorectal cancer is a complication of ulcerative colitis and colonic Crohn’s disease, the two main forms of idiopathic inflammatory bowel disease (IIBD). Mycobacterium avium subspecies paratuberculosis (MAP), the cause of a chronic intestinal disease in domestic and wild ruminants, is one suspected cause of IIBD. MAP may therefore be involved in the pathogenesis of IIBD-associated colorectal cancer as well as colorectal cancer in individuals without IIBD (sporadic colorectal cancer) in countries where MAP infection of domestic livestock is prevalent and MAP’s presence in soil and water is extensive. MAP organisms have been identified in the intestines of patients with sporadic colorectal cancer and IIBD when high magnification, oil immersion light microscopy (×1000 total magnification rather than the usual ×400 total magnification) is used. Research has demonstrated MAP’s ability to invade intestinal goblet cells and cause acute and chronic goblet cell hyperplasia. Goblet cell hyperplasia is the little-recognized initial pathologic lesion of sporadic colorectal cancer, referred to as transitional mucosa, aberrant crypt foci, goblet cell hyperplastic polyps or transitional polyps. It is the even lesser-recognized initial pathologic feature of IIBD, referred to as hypermucinous mucosa, hyperplastic-like mucosal change, serrated epithelial changes, flat serrated changes, goblet cell rich mucosa or epithelial hyperplasia. Goblet cell hyperplasia is the precursor lesion of adenomas and dysplasia in the classical colorectal cancer pathway, of sessile serrated adenomas and serrated dysplasia in the serrated colorectal cancer pathway, and of flat and elevated dysplasia and dysplasia-associated lesions or masses in IIBD-associated intestinal cancers. MAP’s invasion of intestinal goblet cells may result in the initial pathologic lesion of IIBD and sporadic colorectal cancer. MAP’s persistence in infected intestines may result in the eventual development of both IIBD-associated and sporadic colorectal cancer.
Collapse
|
30
|
Tsubokawa D, Ishiwata K, Goso Y, Nakamura T, Hatta T, Ishihara K, Kanuka H, Tsuji N. Interleukin-13/interleukin-4 receptor pathway is crucial for production of Sd a-sialomucin in mouse small intestinal mucosa by Nippostrongylus brasiliensis infection. Parasitol Int 2017; 66:731-734. [PMID: 28802865 DOI: 10.1016/j.parint.2017.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/26/2017] [Accepted: 08/08/2017] [Indexed: 11/25/2022]
Abstract
Mucin is a major component of mucus in gastrointestinal mucosa. Increase of specific sialomucins having Sda blood group antigen, NeuAcα2-3(GalNAcβ1-4)Galβ1-4GlcNAcβ-, is considered to be associated with expulsion of the parasitic intestinal nematode Nippostrongylus brasiliensis. In this study, we examined the relationship between interleukin (IL)-13 pathway and expression of Sda-sialomucins in small intestinal mucosa with N. brasiliensis infection. Nematode infection induced marked increases in small intestinal mucins that reacted with anti-Sda antibody in wild type (wt) mice. However, this increase due to infection was supressed in IL-4 receptor α deficient (IL-4Rα-/-) mice, which lack both IL-4 and IL-13 signaling via IL-4R, and severe combined immunodeficient (SCID) mice, which have defects in B- and T-lymphocytes. Analysis using tandem mass spectroscopy showed that Sda-glycans were not expressed in small intestinal mucins in IL-4Rα-/- and SCID mice after infection despite the appearance of Sda-glycans in the infected wt mice. Inoculation of recombinant IL-13 into the infected SCID mice restored expression of Sda-glycan. Our results suggest that the IL-13/IL-4R axis is important for the production of Sda-sialomucins in the host intestinal mucosa with parasitic nematode infection.
Collapse
Affiliation(s)
- Daigo Tsubokawa
- Department of Parasitology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0374, Japan
| | - Kenji Ishiwata
- Department of Tropical Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shinbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Yukinobu Goso
- Department of Biochemistry, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0374, Japan
| | - Takeshi Nakamura
- Department of Parasitology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0374, Japan
| | - Takeshi Hatta
- Department of Parasitology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0374, Japan
| | - Kazuhiko Ishihara
- Kitasato Junior College of Health and Hygienic Sciences, 500 Kurotsuchishinden Minamiuonuma, Niigata 949-7241, Japan
| | - Hirotaka Kanuka
- Department of Tropical Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shinbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Naotoshi Tsuji
- Department of Parasitology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0374, Japan.
| |
Collapse
|
31
|
Villalobos-Hernández EC, Barajas-López C, Martínez-Salazar EA, Salgado-Delgado RC, Miranda-Morales M. Cholinergic signaling plasticity maintains viscerosensory responses during Aspiculuris tetraptera infection in mice small intestine. Auton Neurosci 2017. [PMID: 28641950 DOI: 10.1016/j.autneu.2017.06.001] [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: 11/18/2022]
Abstract
Intestinal parasites alter gastrointestinal (GI) functions like the cholinergic function. Aspiculuris tetraptera is a pinworm frequently observed in laboratory facilities, which infests the mice cecum and proximal colon. However, little is known about the impact of this infection on the GI sensitivity. Here, we investigated possible changes in spontaneous mesenteric nerve activity and on the mechanosensitivity function of worm-free regions of naturally infected mice with A. tetraptera. Infection increased the basal firing of mesenteric afferent nerves in jejunum. Our findings indicate that nicotinic but not muscarinic receptors, similarly affect spontaneous nerve firing in control and infected animals; these axons are mainly vagal. No difference between groups was observed on spontaneous activity after nicotinic receptor inhibition. However, and contrary to the control group, during infection, the muscarinic signaling was shown to be elevated during mechanosensory experiments. In conclusion, we showed for the first time that alterations induced by infection of the basal afferent activity were independent of the cholinergic function but changes in mechanosensitivity were mediated by muscarinic, but not nicotinic, receptors and specifically by high threshold nerve fibers (activated above 20mmHg), known to play a role in nociception. These plastic changes within the muscarinic signaling would function as a compensatory mechanism to maintain a full mechanosensory response and the excitability of nociceptors during infection. These changes indicate that pinworm colonic infection can target other tissues away from the colon.
Collapse
Affiliation(s)
- Egina C Villalobos-Hernández
- División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica, San Luís Potosí, SLP, Mexico
| | - Carlos Barajas-López
- División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica, San Luís Potosí, SLP, Mexico.
| | - Elizabeth A Martínez-Salazar
- Laboratorio de Colecciones Biológicas y Sistemática Molecular, Unidad Académica de Ciencias Biológicas, Universidad Autónoma de Zacatecas, Mexico
| | | | | |
Collapse
|
32
|
Abstract
杯状细胞(goblet cell, GC)由肠黏膜基底干细胞分化而来, 形似高脚杯, 内含黏液颗粒, 黏液颗粒的组成成分主要为黏蛋白. MUC2是一种重要的拥有特殊网状结构的黏蛋白. GC分泌黏液到肠上皮细胞形成黏液层填补细胞间隙. 黏液层使得GC能在肠黏膜抵御内外源侵袭时发挥重要作用, 尤其是将上皮与肠道菌分离, 从而能维持肠道微生态平衡. 除此之外, GC还能接受和参与免疫调节. 更重要的是GC及其分泌黏蛋白的缺陷与肠道多种疾病密切相关. 总之, GC对肠道健康的作用不容小视.
Collapse
|
33
|
Reyes JL, Fernando MR, Lopes F, Leung G, Mancini NL, Matisz CE, Wang A, McKay DM. IL-22 Restrains Tapeworm-Mediated Protection against Experimental Colitis via Regulation of IL-25 Expression. PLoS Pathog 2016; 12:e1005481. [PMID: 27055194 PMCID: PMC4824453 DOI: 10.1371/journal.ppat.1005481] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 02/09/2016] [Indexed: 12/27/2022] Open
Abstract
Interleukin (IL)-22, an immune cell-derived cytokine whose receptor expression is restricted to non-immune cells (e.g. epithelial cells), can be anti-inflammatory and pro-inflammatory. Mice infected with the tapeworm Hymenolepis diminuta are protected from dinitrobenzene sulphonic acid (DNBS)-induced colitis. Here we assessed expulsion of H. diminuta, the concomitant immune response and the outcome of DNBS-induced colitis in wild-type (WT) and IL-22 deficient mice (IL-22-/-) ± infection. Interleukin-22-/- mice had a mildly impaired ability to expel the worm and this correlated with reduced or delayed induction of TH2 immunity as measured by splenic and mesenteric lymph node production of IL-4, IL-5 and IL-13 and intestinal Muc-2 mRNA and goblet cell hyperplasia; in contrast, IL-25 increased in the small intestine of IL-22-/- mice 8 and 12 days post-infection compared to WT mice. In vitro experiments revealed that H. diminuta directly evoked epithelial production of IL-25 that was inhibited by recombinant IL-22. Also, IL-10 and markers of regulatory T cells were increased in IL-22-/- mice that displayed less DNBS (3 mg, ir. 72h)-induced colitis. Wild-type mice infected with H. diminuta were protected from colitis, as were infected IL-22-/- mice and the latter to a degree that they were almost indistinguishable from control, non-DNBS treated mice. Finally, treatment with anti-IL-25 antibodies exaggerated DNBS-induced colitis in IL-22-/- mice and blocked the anti-colitic effect of infection with H. diminuta. Thus, IL-22 is identified as an endogenous brake on helminth-elicited TH2 immunity, reducing the efficacy of expulsion of H. diminuta and limiting the effectiveness of the anti-colitic events mobilized following infection with H. diminuta in a non-permissive host.
Collapse
Affiliation(s)
- José L. Reyes
- Gastrointestinal Research Group, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Maria R. Fernando
- Gastrointestinal Research Group, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Fernando Lopes
- Gastrointestinal Research Group, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gabriella Leung
- Gastrointestinal Research Group, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nicole L. Mancini
- Gastrointestinal Research Group, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Chelsea E. Matisz
- Gastrointestinal Research Group, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Arthur Wang
- Gastrointestinal Research Group, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Derek M. McKay
- Gastrointestinal Research Group, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
34
|
Birchenough GMH, Johansson MEV, Gustafsson JK, Bergström JH, Hansson GC. New developments in goblet cell mucus secretion and function. Mucosal Immunol 2015; 8:712-9. [PMID: 25872481 PMCID: PMC4631840 DOI: 10.1038/mi.2015.32] [Citation(s) in RCA: 506] [Impact Index Per Article: 56.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/20/2015] [Indexed: 02/06/2023]
Abstract
Goblet cells and their main secretory product, mucus, have long been poorly appreciated; however, recent discoveries have changed this and placed these cells at the center stage of our understanding of mucosal biology and the immunology of the intestinal tract. The mucus system differs substantially between the small and large intestine, although it is built around MUC2 mucin polymers in both cases. Furthermore, that goblet cells and the regulation of their secretion also differ between these two parts of the intestine is of fundamental importance for a better understanding of mucosal immunology. There are several types of goblet cell that can be delineated based on their location and function. The surface colonic goblet cells secrete continuously to maintain the inner mucus layer, whereas goblet cells of the colonic and small intestinal crypts secrete upon stimulation, for example, after endocytosis or in response to acetyl choline. However, despite much progress in recent years, our understanding of goblet cell function and regulation is still in its infancy.
Collapse
Affiliation(s)
- G M H Birchenough
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| | - M E V Johansson
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| | - J K Gustafsson
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| | - J H Bergström
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| | - G C Hansson
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
35
|
Oeser K, Schwartz C, Voehringer D. Conditional IL-4/IL-13-deficient mice reveal a critical role of innate immune cells for protective immunity against gastrointestinal helminths. Mucosal Immunol 2015; 8:672-82. [PMID: 25336167 DOI: 10.1038/mi.2014.101] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 09/23/2014] [Indexed: 02/04/2023]
Abstract
Approximately one-third of the world population is infected with gastrointestinal helminths. Studies in mouse models have demonstrated that the cytokines interleukin (IL)-4 and IL-13 are essential for worm expulsion, but the critical cellular source of these cytokines is poorly defined. Here, we compared the immune response to Nippostrongylus brasiliensis in wild-type, T cell-specific IL-4/IL-13-deficient and general IL-4/IL-13-deficient mice. We show that T cell-derived IL-4/IL-13 promoted T helper 2 (Th2) polarization in a paracrine manner, differentiation of alternatively activated macrophages, and tissue recruitment of innate effector cells. However, innate IL-4/IL-13 played the critical role for induction of goblet cell hyperplasia and secretion of effector molecules like Mucin5ac and RELMβ in the small intestine. Surprisingly, T cell-specific IL-4/IL-13-deficient and wild-type mice cleared the parasite with comparable efficiency, whereas IL-4/IL-13-deficient mice showed impaired expulsion. These findings demonstrate that IL-4/IL-13 produced by cells of the innate immune system is required and sufficient to initiate effective type 2 immune responses resulting in protective immunity against N. brasiliensis.
Collapse
Affiliation(s)
- K Oeser
- Department of Infection Biology, Institute of Microbiology, Universitätsklinikum Erlangen and Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - C Schwartz
- Department of Infection Biology, Institute of Microbiology, Universitätsklinikum Erlangen and Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - D Voehringer
- Department of Infection Biology, Institute of Microbiology, Universitätsklinikum Erlangen and Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
36
|
Scheer S, Krempl C, Kallfass C, Frey S, Jakob T, Mouahid G, Moné H, Schmitt-Gräff A, Staeheli P, Lamers MC. S. mansoni bolsters anti-viral immunity in the murine respiratory tract. PLoS One 2014; 9:e112469. [PMID: 25398130 PMCID: PMC4232382 DOI: 10.1371/journal.pone.0112469] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 10/03/2014] [Indexed: 01/17/2023] Open
Abstract
The human intestinal parasite Schistosoma mansoni causes a chronic disease, schistosomiasis or bilharzia. According to the current literature, the parasite induces vigorous immune responses that are controlled by Th2 helper cells at the expense of Th1 helper cells. The latter cell type is, however, indispensable for anti-viral immune responses. Remarkably, there is no reliable literature among 230 million patients worldwide describing defective anti-viral immune responses in the upper respiratory tract, for instance against influenza A virus or against respiratory syncitial virus (RSV). We therefore re-examined the immune response to a human isolate of S. mansoni and challenged mice in the chronic phase of schistosomiasis with influenza A virus, or with pneumonia virus of mice (PVM), a mouse virus to model RSV infections. We found that mice with chronic schistosomiasis had significant, systemic immune responses induced by Th1, Th2, and Th17 helper cells. High serum levels of TNF-α, IFN-γ, IL-5, IL-13, IL-2, IL-17, and GM-CSF were found after mating and oviposition. The lungs of diseased mice showed low-grade inflammation, with goblet cell hyperplasia and excessive mucus secretion, which was alleviated by treatment with an anti-TNF-α agent (Etanercept). Mice with chronic schistosomiasis were to a relative, but significant extent protected from a secondary viral respiratory challenge. The protection correlated with the onset of oviposition and TNF-α-mediated goblet cell hyperplasia and mucus secretion, suggesting that these mechanisms are involved in enhanced immune protection to respiratory viruses during chronic murine schistosomiasis. Indeed, also in a model of allergic airway inflammation mice were protected from a viral respiratory challenge with PVM.
Collapse
Affiliation(s)
- Sebastian Scheer
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- International Max Planck Research School of Molecular and Cellular Biology, Freiburg, Germany
- University of Freiburg, Freiburg, Germany
| | - Christine Krempl
- Institute of Virology and Immunology, University of Wuerzburg, Wuerzburg, Germany
| | - Carsten Kallfass
- Institute for Virology, Department of Medical Microbiology and Hygiene, University Medical Center Freiburg, Freiburg, Germany
| | - Stefanie Frey
- Allergy Research Group, Department of Dermatology, University Medical Center Freiburg, Freiburg, Germany
| | - Thilo Jakob
- Allergy Research Group, Department of Dermatology, University Medical Center Freiburg, Freiburg, Germany
| | - Gabriel Mouahid
- Univ. Perpignan Via Domitia, Ecologie et Evolution des Interactions, UMR 5244, F-66860, Perpignan, France
| | - Hélène Moné
- CNRS, Ecologie et Evolution des Interactions, UMR 5244, F-66860, Perpignan, France
| | | | - Peter Staeheli
- Institute for Virology, Department of Medical Microbiology and Hygiene, University Medical Center Freiburg, Freiburg, Germany
| | - Marinus C. Lamers
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- * E-mail:
| |
Collapse
|
37
|
The role of antibody in parasitic helminth infections. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 828:1-26. [PMID: 25253025 DOI: 10.1007/978-1-4939-1489-0_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
38
|
Topical interferon-gamma neutralization prevents conjunctival goblet cell loss in experimental murine dry eye. Exp Eye Res 2013; 118:117-24. [PMID: 24315969 DOI: 10.1016/j.exer.2013.11.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 11/11/2013] [Accepted: 11/20/2013] [Indexed: 01/26/2023]
Abstract
Evidence suggests that the cytokine interferon (IFN)-γ released by natural killer and CD4(+) T cells contributes to the conjunctival goblet cell (GC) loss in dry eye. The purpose of this study was to investigate if topical neutralization of IFN-γ prevents or alleviates GC loss in an experimental desiccating stress (DS) model of dry eye. In this study, we found that topical IFN-γ neutralization significantly decreased DS-induced conjunctival GC loss. This was accompanied by decreased epithelial apoptosis, and increased IL-13 and decreased FoxA2 expression in the forniceal conjunctiva. To establish that IFN-γ produced by pathogenic CD4(+) T cells contributes to DS-induced GC loss, adoptive transfer of CD4(+) T cells isolated from DS exposed donors to naïve RAG-1(-/-) recipient mice was performed. Similar to the donor mice, topical IFN-γ neutralization decreased conjunctival GC loss, suppressed apoptosis and increased IL-13 expression in adoptive transfer recipients. In summary, this study demonstrated that topical neutralization of IFN-γ prevents GC loss via modulating apoptosis and maintaining IL-13 signaling.
Collapse
|
39
|
Turner JE, Stockinger B, Helmby H. IL-22 mediates goblet cell hyperplasia and worm expulsion in intestinal helminth infection. PLoS Pathog 2013; 9:e1003698. [PMID: 24130494 PMCID: PMC3795034 DOI: 10.1371/journal.ppat.1003698] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 08/26/2013] [Indexed: 02/07/2023] Open
Abstract
Type 2 immune responses are essential in protection against intestinal helminth infections. In this study we show that IL-22, a cytokine important in defence against bacterial infections in the intestinal tract, is also a critical mediator of anti-helminth immunity. After infection with Nippostrongylus brasiliensis, a rodent hookworm, IL-22-deficient mice showed impaired worm expulsion despite normal levels of type 2 cytokine production. The impaired worm expulsion correlated with reduced goblet cell hyperplasia and reduced expression of goblet cell markers. We further confirmed our findings in a second nematode model, the murine whipworm Trichuris muris. T.muris infected IL-22-deficient mice had a similar phenotype to that seen in N.brasiliensis infection, with impaired worm expulsion and reduced goblet cell hyperplasia. Ex vivo and in vitro analysis demonstrated that IL-22 is able to directly induce the expression of several goblet cell markers, including mucins. Taken together, our findings reveal that IL-22 plays an important role in goblet cell activation, and thus, a key role in anti-helminth immunity.
Collapse
Affiliation(s)
- Jan-Eric Turner
- Division of Molecular Immunology, MRC National Institute for Medical Research, Mill Hill, London, United Kingdom
| | - Brigitta Stockinger
- Division of Molecular Immunology, MRC National Institute for Medical Research, Mill Hill, London, United Kingdom
| | - Helena Helmby
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
40
|
Sant'Ana DMG, Góis MB, Zanoni JN, da Silva AV, da Silva CJT, Araújo EJA. Intraepithelial lymphocytes, goblet cells and VIP-IR submucosal neurons of jejunum rats infected with Toxoplasma gondii. Int J Exp Pathol 2012; 93:279-86. [PMID: 22804764 DOI: 10.1111/j.1365-2613.2012.00824.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Toxoplasma gondii (T. gondii) crosses the intestinal barrier in oral infections and can lead to changes in different cell types, including the neurons located there. In the gastrointestinal system, the autonomous nervous system component that regulate blood flow and mucous secretion is the submucosal plexus. The aim of this study was to examine the effects of T. gondii infection on intraepithelial lymphocytes (IELs), goblet cells and submucosal neurons that are immunoreactive to vasoactive intestinal peptide (VIP-IR) of rat jejunum. Twenty male rats distributed as a control group (CG) and an infected group (IG), which received a suspension with 500 parasite oocysts (strain ME-49, genotype II) orally, were assessed. Routine histological sections were used to quantify IELs and to detect mucins secreted by goblet cells. Whole mounts including the submucosal layer were examined using immunofluorescence to detect the VIP neurotransmitter. Quantitative alterations in IELs were not observed. However, the reduction (P < 0.05) in the number of goblet cells that produce neutral mucins (PAS+) and sulphomucins (AB pH 1.0) and the maintenance of sialomucin-secreting cells (AB pH 2.5) resulting in a more fluid mucous were observed. Concerning the VIP-IR submucosal neurons, an increase in fluorescence on IG animals was observed. There was a reduction (P < 0.05) in the number of VIP-IR submucosal neurons and atrophy of their cell bodies in IG rats. Infection with T. gondii caused alterations in the chemical composition of the intestinal mucous and reduction in the neuron number and atrophy of the remaining neurons in this cell subpopulation.
Collapse
|
41
|
Pastorelli L, De Salvo C, Cominelli MA, Vecchi M, Pizarro TT. Novel cytokine signaling pathways in inflammatory bowel disease: insight into the dichotomous functions of IL-33 during chronic intestinal inflammation. Therap Adv Gastroenterol 2011; 4:311-23. [PMID: 21922030 PMCID: PMC3165208 DOI: 10.1177/1756283x11410770] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
In 2010, four independent groups almost simultaneously reported the association of the novel interleukin-1 (IL-1) family member, IL-33, with inflammatory bowel disease (IBD). The findings were remarkably consistent and demonstrated that IL-33 is markedly upregulated in, and specific to, ulcerative colitis (UC). In addition, although a variety of gut-associated immune cell subsets express IL-33, the primary source appears to be the intestinal epithelium. IL-33's receptor, ST2, a formerly orphaned IL-1 receptor-related protein, was also found to be increased in UC patients, although the cellular source of ST2 appears to be somewhat more ambiguous. In fact, emerging evidence indicates that the IL-33/ST2 axis plays a critical role in several other chronic inflammatory and immune disorders. In the gut, IL-33 has been shown to be important in the clearance of intestinal parasites, and inducing epithelial cell hyperplasia, mucus production and mucosal eosinophilic infiltration. However, despite the established trend of increased IL-33 and ST2 expression during IBD, specifically UC, the precise pathophysiologic relevance of these findings has yet to be determined. Interestingly, IL-33 has the ability to potentiate pathogenic Th2 and Th17 responses in gut-associated lymphoid tissues, while also promoting healing of damaged mucosa following inflammatory insults. Indeed, further mechanistic studies are warranted to confirm the possible dichotomous functions of IL-33 during chronic intestinal inflammation and better define its precise role in the pathogenesis of IBD. Herein, we discuss what is currently known about IL-33/ST2 in the gut and speculate as to the potential role of the IL-33/ST2 system in IBD.
Collapse
Affiliation(s)
- Luca Pastorelli
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA; IRCCS Policlinico San Donato, Gastroenterology and Gastrointestinal Endoscopy Unit, San Donato Milanese, Italy; University of Milan School of Medicine, Medical and Surgical Sciences, Milan, Italy
| | - Carlo De Salvo
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Marissa A. Cominelli
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Maurizio Vecchi
- IRCCS Policlinico San Donato, Gastroenterology and Gastrointestinal Endoscopy Unit, San Donato Milanese, Italy; University of Milan School of Medicine, Medical and Surgical Sciences, Milan, Italy
| | - Theresa T. Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
42
|
Hasnain SZ, Thornton DJ, Grencis RK. Changes in the mucosal barrier during acute and chronic Trichuris muris infection. Parasite Immunol 2011; 33:45-55. [PMID: 21155842 PMCID: PMC3020324 DOI: 10.1111/j.1365-3024.2010.01258.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Accepted: 07/19/2010] [Indexed: 01/28/2023]
Abstract
The intestinal mucosal barrier, part of the innate immune defence, is responsive to the external environment and changes in response to infection. There is disparate evidence for the epithelial and goblet cell products within the intrinsic barrier being part of a response to resolve infection. We comprehensively analysed the changes of mucosal glycoconjugates during acute and chronic infection by utilising the Trichuris muris (T. muris) model. Transcription factors, atonal homolog 1 (Math-1) and SAM pointed domain containing ETS transcription factor (Spdef) were activated during acute infection, which promoted stem cell fate towards a secretory cell phenotype. The thickness of the intermediate barrier, the carbohydrate-rich glycocalyx, composed of cell surface mucins increased with exposure to T. muris, with an increase in Muc4, Muc13 and Muc17. Overall, hypersecretion of glycoproteins into the extrinsic barrier (mediated by IL-13) via the gamma amino-butyric acid-α3 receptor (GABA-α3), was observed during acute infection. Furthermore, altered glycosylation was observed during acute and chronic infection; mucins were more highly charged during acute infection than during chronic infection. This study readdresses the changes within the mucosal barrier, in particular in the cell surface and secreted mucins during acute and chronic nematode infection.
Collapse
Affiliation(s)
- S Z Hasnain
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK.
| | | | | |
Collapse
|
43
|
Hünig T, Lühder F, Elflein K, Gogishvili T, Fröhlich M, Guler R, Cutler A, Brombacher F. CD28 and IL-4: two heavyweights controlling the balance between immunity and inflammation. Med Microbiol Immunol 2010; 199:239-46. [PMID: 20390297 PMCID: PMC3128750 DOI: 10.1007/s00430-010-0156-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Indexed: 12/25/2022]
Abstract
The costimulatory receptor CD28 and IL-4Rα-containing cytokine receptors play key roles in controlling the size and quality of pathogen-specific immune responses. Thus, CD28-mediated costimulation is needed for effective primary T-cell expansion and for the generation and activation of regulatory T-cells (Treg cells), which protect from immunopathology. Similarly, IL-4Rα signals are required for alternative activation of macrophages, which counteract inflammation by type 1 responses. Furthermore, immune modulation by CD28 and IL-4 is interconnected through the promotion of IL-4 producing T-helper 2 cells by CD28 signals. Using conditionally IL-4Rα and CD28 deleting mice, as well as monoclonal antibodies, which block or stimulate CD28, or mAb that deplete Treg cells, we have studied the roles of CD28 and IL-4Rα in experimental mouse models of virus (influenza), intracellular bacteria (L. monocytogenes, M. tuberculosis), and parasite infections (T. congolense, L. major). We observed that in some, but not all settings, Treg cells and type 2 immune deviation, including activation of alternative macrophages can be manipulated to protect the host either from infection or from immunopathology with an overall beneficial outcome. Furthermore, we provide direct evidence that secondary CD8 T-cell responses to i.c. bacteria are dependent on CD28-mediated costimulation.
Collapse
Affiliation(s)
- Thomas Hünig
- Institute for Virology and Immunobiology, University of Würzburg, Versbacher Str. 7, Würzburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Reyes JL, Terrazas CA, Alonso-Trujillo J, van Rooijen N, Satoskar AR, Terrazas LI. Early removal of alternatively activated macrophages leads to Taenia crassiceps cysticercosis clearance in vivo. Int J Parasitol 2010; 40:731-42. [PMID: 20045000 DOI: 10.1016/j.ijpara.2009.11.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2009] [Revised: 11/23/2009] [Accepted: 11/24/2009] [Indexed: 12/14/2022]
Abstract
To determine the role of alternatively activated macrophages in modulating the outcome of experimental cysticercosis caused by Taenia crassiceps, we investigated the effect of removal of alternatively activated macrophage by injecting clodronate-loaded liposomes into susceptible BALB/c mice. Following T. crassiceps infection, mice receiving PBS-loaded liposomes developed a dominant Th2-type response associated with the presence of alternatively activated macrophages together with antigen-specific hyporesponsiveness and high parasite burden. In contrast, similarly infected mice treated with clodronate-loaded liposomes mounted a mixed Th1/Th2-type response, reversed antigen-specific hyporesponsiveness and did not carry notable alternatively activated macrophage populations. These factors were associated with increased resistance to T. crassiceps cysticercosis. Interestingly, early AAM phi depletion was enough to limit parasite growth. However, if macrophages were depleted late in the infection, no effect on parasite burden was observed. These findings demonstrate that alternatively activated macrophages play a critical role in mediating susceptibility to experimental cysticercosis in which their early recruitment may favor parasite survival.
Collapse
Affiliation(s)
- José L Reyes
- Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México, Mexico
| | | | | | | | | | | |
Collapse
|
45
|
Localized Th1-, Th2-, T regulatory cell-, and inflammation-associated hepatic and pulmonary immune responses in Ascaris suum-infected swine are increased by retinoic acid. Infect Immun 2009; 77:2576-87. [PMID: 19332534 DOI: 10.1128/iai.00827-07] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Pigs infected with Ascaris suum or controls were given 100 microg (low-dose) or 1,000 microg (high-dose) all-trans retinoic acid (ATRA)/kg body weight in corn oil or corn oil alone per os on days after inoculation (DAI) -1, +1, and +3 with infective eggs. Treatment with ATRA increased interleukin 4 (IL4) and IL12p70 in plasma of infected pigs at 7 DAI and augmented bronchoalveolar lavage (BAL) eosinophilia observed at 7 and 14 DAI. To explore potential molecular mechanisms underlying these observations, a quantitative real-time reverse transcription (RT)-PCR array was used to examine mRNA expression in tissue. Ascaris-infected pigs had increased levels of liver mRNA for T-helper-2 (Th2)-associated cytokines, mast cell markers, and T regulatory (Treg) cells, while infected pigs given ATRA had higher IL4, IL13, CCL11, CCL26, CCL17, CCL22, and TPSB1 expression. Gene expression for Th1-associated markers (IFNG, IL12B, and TBX21), the CXCR3 ligand (CXCL9), IL1B, and the putative Treg marker TNFRSF18 was also increased. Expression of IL4, IL13, IL1B, IL6, CCL11, and CCL26 was increased in the lungs of infected pigs treated with ATRA. To determine a putative cellular source of eosinophil chemoattractants, alveolar macrophages were treated with IL4 and/or ATRA in vitro. IL4 induced CCL11, CCL17, CCL22, and CCL26 mRNA, and ATRA increased the basal and IL4-stimulated expression of CCL17 and CCL22. Thus, ATRA augments a diverse Th1-, Th2-, Treg-, and inflammation-associated response in swine infected with A. suum, and the increased BAL eosinophilia may be related to enhanced induction of eosinophil chemokine activity by alveolar macrophages.
Collapse
|