1
|
Zheng Y, Corrêa-Silva S, Rodrigues RM, Corrêa de Souza E, Macaferri da Fonseca FA, Gilio AE, Carneiro-Sampaio M, Palmeira P. Infant respiratory infections modulate lymphocyte homing to breast milk. Front Immunol 2025; 15:1481416. [PMID: 39867906 PMCID: PMC11757141 DOI: 10.3389/fimmu.2024.1481416] [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: 08/15/2024] [Accepted: 12/16/2024] [Indexed: 01/28/2025] Open
Abstract
Introduction Chemokines and their receptors are essential for leukocyte migration to several tissues, including human milk. Here, we evaluated the homing of T and B lymphocyte subsets to breast milk in response to ongoing respiratory infections in the nursing infant. Methods Blood and mature milk were collected from healthy mothers of nurslings with respiratory infections (Group I) and from healthy mothers of healthy nurslings (Group C). Total lymphocyte, T and B cells, their subset numbers, and the expression of the homing receptors CCR5, CCR6, CCR10, and CXCR3 in these cells were evaluated in milk. Maternal serum and milk chemokine, cytokine, and IgA and IgG antibody levels were also quantified. Results All milk lymphocyte numbers were greater in Group I than in Group C. All CD4 T-cell subsets expressing CCR5, CCR6, and CXCR3 were higher in Group I. Within the CD8 T-cell subsets, only CCR6 and CXCR3 were higher in Group I, while CCR5 expression was higher in Group I exclusively for activated CD8 T cells. Group I showed greater numbers of all CCR6+ B-cell subsets and CXCR3+ naive B cells and plasma cells than did Group C. Infection of the nurslings promoted increased CCL20, CXCL10, IL-6, IL-8, total IgA, and IgG levels in the milk. Conclusion Respiratory infections in nursing infants stimulate an increase in cytokines and chemokines in breast milk, facilitating the recruitment and activation of lymphocytes. This process may promote immunological tolerance and help in the maturation of the infant's immune system, providing an additional strategy for passive maternal-infant protection.
Collapse
Affiliation(s)
- Yingying Zheng
- Department of Pediatrics, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Simone Corrêa-Silva
- Laboratorio de Pediatria Clinica (LIM36), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Regina Maria Rodrigues
- Department of Pediatrics, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Eloisa Corrêa de Souza
- Department of Pediatrics, University Hospital, Medical School, University of São Paulo, São Paulo, Brazil
| | | | - Alfredo Elias Gilio
- Department of Pediatrics, University Hospital, Medical School, University of São Paulo, São Paulo, Brazil
| | - Magda Carneiro-Sampaio
- Department of Pediatrics, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Patricia Palmeira
- Laboratorio de Pediatria Clinica (LIM36), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
2
|
Li J, Simmons AJ, Chiron S, Ramirez-Solano MA, Tasneem N, Kaur H, Xu Y, Revetta F, Vega PN, Bao S, Cui C, Tyree RN, Raber LW, Conner AN, Beaulieu DB, Dalal RL, Horst SN, Pabla BS, Huo Y, Landman BA, Roland JT, Scoville EA, Schwartz DA, Washington MK, Shyr Y, Wilson KT, Coburn LA, Lau KS, Liu Q. A Specialized Epithelial Cell Type Regulating Mucosal Immunity and Driving Human Crohn's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.30.560293. [PMID: 37873404 PMCID: PMC10592875 DOI: 10.1101/2023.09.30.560293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Crohn's disease (CD) is a complex chronic inflammatory disorder that may affect any part of gastrointestinal tract with extra-intestinal manifestations and associated immune dysregulation. To characterize heterogeneity in CD, we profiled single-cell transcriptomics of 170 samples from 65 CD patients and 18 non-inflammatory bowel disease (IBD) controls in both the terminal ileum (TI) and ascending colon (AC). Analysis of 202,359 cells identified a novel epithelial cell type in both TI and AC, featuring high expression of LCN2, NOS2, and DUOX2, and thus is named LND. LND cells, confirmed by high-resolution in-situ RNA imaging, were rarely found in non-IBD controls, but expanded significantly in active CD. Compared to other epithelial cells, genes defining LND cells were enriched in antimicrobial response and immunoregulation. Moreover, multiplexed protein imaging demonstrated that LND cell abundance was associated with immune infiltration. Cross-talk between LND and immune cells was explored by ligand-receptor interactions and further evidenced by their spatial colocalization. LND cells showed significant enrichment of expression specificity of IBD/CD susceptibility genes, revealing its role in immunopathogenesis of CD. Investigating lineage relationships of epithelial cells detected two LND cell subpopulations with different origins and developmental potential, early and late LND. The ratio of the late to early LND cells was related to anti-TNF response. These findings emphasize the pathogenic role of the specialized LND cell type in both Crohn's ileitis and Crohn's colitis and identify novel biomarkers associated with disease activity and treatment response.
Collapse
Affiliation(s)
- Jia Li
- Center for Quantitative Sciences, Vanderbilt Univerity Medical Center, Nashville, TN, USA
- Department of Biostatistics, Vanderbilt Univerity Medical Center, Nashville, TN, USA
| | - Alan J. Simmons
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Sophie Chiron
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Marisol A. Ramirez-Solano
- Center for Quantitative Sciences, Vanderbilt Univerity Medical Center, Nashville, TN, USA
- Department of Biostatistics, Vanderbilt Univerity Medical Center, Nashville, TN, USA
| | - Naila Tasneem
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Harsimran Kaur
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Yanwen Xu
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Frank Revetta
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Paige N. Vega
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Shunxing Bao
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Computer Science, Vanderbilt University, Nashville, TN, USA
| | - Can Cui
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Computer Science, Vanderbilt University, Nashville, TN, USA
| | - Regina N. Tyree
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Larry W. Raber
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Anna N. Conner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dawn B. Beaulieu
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Robin L. Dalal
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sara N. Horst
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Baldeep S. Pabla
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yuankai Huo
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Computer Science, Vanderbilt University, Nashville, TN, USA
| | - Bennett A. Landman
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Computer Science, Vanderbilt University, Nashville, TN, USA
| | - Joseph T. Roland
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Surgery, Vanderbilt University Medical Center, Nashville TN, USA
| | - Elizabeth A. Scoville
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center; Nashville, TN, USA
| | - David A. Schwartz
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M. Kay Washington
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center; Nashville, TN, USA
| | - Yu Shyr
- Center for Quantitative Sciences, Vanderbilt Univerity Medical Center, Nashville, TN, USA
- Department of Biostatistics, Vanderbilt Univerity Medical Center, Nashville, TN, USA
| | - Keith T. Wilson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center; Nashville, TN, USA
| | - Lori A. Coburn
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center; Nashville, TN, USA
| | - Ken S. Lau
- Center for Quantitative Sciences, Vanderbilt Univerity Medical Center, Nashville, TN, USA
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Surgery, Vanderbilt University Medical Center, Nashville TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center; Nashville, TN, USA
| | - Qi Liu
- Center for Quantitative Sciences, Vanderbilt Univerity Medical Center, Nashville, TN, USA
- Department of Biostatistics, Vanderbilt Univerity Medical Center, Nashville, TN, USA
| |
Collapse
|
3
|
Nettleford SK, Liao C, Short SP, Rossi RM, Singh V, Prabhu KS. Selenoprotein W Ameliorates Experimental Colitis and Promotes Intestinal Epithelial Repair. Antioxidants (Basel) 2023; 12:850. [PMID: 37107231 PMCID: PMC10134982 DOI: 10.3390/antiox12040850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 04/05/2023] Open
Abstract
Selenoprotein W (Selenow) is a ~9 kDa selenoprotein suggested to play a beneficial role in resolving inflammation. However, the underlying mechanisms are poorly understood. SELENOW expression in the human GI tract using ScRNAseq Gut Cell Atlas and Gene Expression Omnibus (GEO) databases revealed its expression in the small intestine and colonic epithelial, endothelial, mesenchymal, and stem cells and correlated with a protective effect in ulcerative colitis patients. Selenow KO mice treated with 4% dextran sodium sulfate (DSS) showed exacerbated acute colitis, with greater weight loss, shorter colons, and increased fecal occult blood compared to the WT counterparts. Selenow KO mice expressed higher colonic Tnfα, increased Tnfα+ macrophages in the colonic lamina propria, and exhibited loss in epithelial barrier integrity and decreased zonula occludens 1 (Zo-1) expression following DSS treatment. Expression of epithelial cellular adhesion marker (EpCam), yes-associated protein 1 (Yap1), and epidermal growth factor receptor (Egfr) were decreased along with CD24lo cycling epithelial cells in Selenow KO mice. Colonic lysates and organoids confirmed a crosstalk between Egfr and Yap1 that was regulated by Selenow. Overall, our findings suggest Selenow expression is key for efficient resolution of inflammation in experimental colitis that is mediated through the regulation of Egfr and Yap1.
Collapse
Affiliation(s)
- Shaneice K. Nettleford
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Chang Liao
- Department of Medicine-Infectious Diseases, University of California, San Francisco, CA 94143, USA
| | - Sarah P. Short
- Department of Medicine, Department of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Randall M. Rossi
- Mouse Transgenic Core Facility, Huck Institute of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Vishal Singh
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - K. Sandeep Prabhu
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
4
|
Chen Z, Haus JM, Chen L, Jiang Y, Sverdlov M, DiPietro LA, Xiong N, Wu SC, Koh TJ, Minshall RD. Inhibition of CCL28/CCR10-Mediated eNOS Downregulation Improves Skin Wound Healing in the Obesity-Induced Mouse Model of Type 2 Diabetes. Diabetes 2022; 71:2166-2180. [PMID: 35899992 PMCID: PMC9501665 DOI: 10.2337/db21-1108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 07/21/2022] [Indexed: 11/13/2022]
Abstract
Chronic, nonhealing skin wounds, such as diabetic foot ulcers (DFUs), are common in patients with type 2 diabetes. Here, we investigated the role of chemokine (C-C motif) ligand 28 (CCL28) and its receptor C-C chemokine receptor type 10 (CCR10) in downregulation of endothelial nitric (NO) oxide synthase (eNOS) in association with delayed skin wound healing in the db/db mouse model of type 2 diabetes. We observed reduced eNOS expression and elevated CCL28/CCR10 levels in dorsal skin of db/db mice and subdermal leg biopsy specimens from human subjects with type 2 diabetes. Further interrogation revealed that overexpression of CCR10 reduced eNOS expression, NO bioavailability, and tube formation of human dermal microvascular endothelial cells (HDMVECs) in vitro, which was recapitulated in mouse dorsal skin. In addition, incubation of HDMVECs with CCL28 led to internalization of the CCR10/eNOS complex and colocalization with lysosome-associated membrane protein 1. Finally, topical application of myristoylated CCR10 binding domain 7 amino acid (Myr-CBD7) peptide prevented CCR10-eNOS interaction and subsequent eNOS downregulation, enhanced eNOS/NO levels, eNOS/VEGF-R2+ microvessel density, and blood perfusion, reduced inflammatory cytokine levels, and importantly, decreased wound healing time in db/db mice. Thus, endothelial cell CCR10 activation in genetically obese mice with type 2 diabetes promotes eNOS depletion and endothelial dysfunction, and targeted disruption of CCR10/eNOS interaction improves wound healing.
Collapse
Affiliation(s)
- Zhenlong Chen
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL
| | - Jacob M. Haus
- School of Kinesiology, University of Michigan, Ann Arbor, MI
| | - Lin Chen
- Department of Periodontics, University of Illinois at Chicago, Chicago, IL
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL
| | - Ying Jiang
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL
| | - Maria Sverdlov
- Research Resources Center, Research Histology and Tissue Imaging Collaborative, University of Illinois at Chicago, Chicago, IL
| | - Luisa A. DiPietro
- Department of Periodontics, University of Illinois at Chicago, Chicago, IL
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL
| | - Na Xiong
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Stephanie C. Wu
- Departments of Surgery and Stem Cell and Regenerative Medicine, Center for Lower Extremity Ambulatory Research, Dr. William M. Scholl College of Podiatric Medicine, Rosalind Franklin University of Medicine and Science, North Chicago, IL
| | - Timothy J. Koh
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL
| | - Richard D. Minshall
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
5
|
Gary EN, Tursi NJ, Warner B, Parzych EM, Ali AR, Frase D, Moffat E, Embury-Hyatt C, Smith TRF, Broderick KE, Humeau L, Kobasa D, Patel A, Kulp DW, Weiner DB. Mucosal chemokine adjuvant enhances synDNA vaccine-mediated responses to SARS-CoV-2 and provides heterologous protection in vivo. Cell Rep Med 2022; 3:100693. [PMID: 35839767 PMCID: PMC9237025 DOI: 10.1016/j.xcrm.2022.100693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/16/2022] [Accepted: 06/23/2022] [Indexed: 11/28/2022]
Abstract
The global coronavirus disease 2019 (COVID-19) pandemic has claimed more than 5 million lives. Emerging variants of concern (VOCs) continually challenge viral control. Directing vaccine-induced humoral and cell-mediated responses to mucosal surfaces may enhance vaccine efficacy. Here we investigate the immunogenicity and protective efficacy of optimized synthetic DNA plasmids encoding wild-type severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein (pS) co-formulated with the plasmid-encoded mucosal chemokine cutaneous T cell-attracting chemokine (pCTACK; CCL27). pCTACK-co-immunized animals exhibit increased spike-specific antibodies at the mucosal surface and increased frequencies of interferon gamma (IFNγ)+ CD8+ T cells in the respiratory mucosa. pCTACK co-immunization confers 100% protection from heterologous Delta VOC challenge. This study shows that mucosal chemokine adjuvants can direct vaccine-induced responses to specific immunological sites and have significant effects on heterologous challenge. Further study of this unique chemokine-adjuvanted vaccine approach in the context of SARS-CoV-2 vaccines is likely important.
Collapse
Affiliation(s)
- Ebony N Gary
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Nicholas J Tursi
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bryce Warner
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Elizabeth M Parzych
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Ali R Ali
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Drew Frase
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Estella Moffat
- National Center for Foreign Animal Disease (NCFAD), Canadian Food Inspection Agency, Winnipeg, MB, Canada
| | - Carissa Embury-Hyatt
- National Center for Foreign Animal Disease (NCFAD), Canadian Food Inspection Agency, Winnipeg, MB, Canada
| | | | | | | | - Darwyn Kobasa
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada; Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Ami Patel
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Daniel W Kulp
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - David B Weiner
- The Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Abstract
Epithelial barriers, which include the gastrointestinal, respiratory, and genitourinary mucosa, compose the body’s front line of defense. Since barrier tissues are persistently exposed to microbial challenges, a rapid response that can deal with diverse invading pathogens is crucial. Because B cells have been perceived as indirectly contributing to immune responses through antibody production, B cells functioning in the peripheral organs have been outside the scope of researchers. However, recent evidence supports the existence of tissue-resident memory B cells (BRMs) in the lungs. This population’s defensive response was stronger and faster than that of their circulating counterparts and could resist heterogeneous strains. With such traits, BRMs could be a promising target for vaccine design, but much about them remains to be revealed, including their locations, origin, specific markers, and the mechanisms of their establishment and maintenance. There is evidence for resident B cells in organs other than the lungs, suggesting that B cells are directly involved in the immune reactions of multiple non-lymphoid organs. This review summarizes the history of the discovery of BRMs and discusses important unresolved questions. Unique characteristics of humoral immunity that play an important role in the peripheral organs will be described briefly. Future research on B cells residing in non-lymphoid organs will provide new insights to help solve major problems regarding human health.
Collapse
Affiliation(s)
- Choong Man Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Ji Eun Oh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- BioMedical Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- *Correspondence: Ji Eun Oh,
| |
Collapse
|
7
|
Trevelin SC, Pickering S, Todd K, Bishop C, Pitcher M, Garrido Mesa J, Montorsi L, Spada F, Petrov N, Green A, Shankar-Hari M, Neil SJ, Spencer J. Disrupted Peyer's Patch Microanatomy in COVID-19 Including Germinal Centre Atrophy Independent of Local Virus. Front Immunol 2022; 13:838328. [PMID: 35251032 PMCID: PMC8893224 DOI: 10.3389/fimmu.2022.838328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/27/2022] [Indexed: 12/12/2022] Open
Abstract
Confirmed SARS-coronavirus-2 infection with gastrointestinal symptoms and changes in microbiota associated with coronavirus disease 2019 (COVID-19) severity have been previously reported, but the disease impact on the architecture and cellularity of ileal Peyer's patches (PP) remains unknown. Here we analysed post-mortem tissues from throughout the gastrointestinal (GI) tract of patients who died with COVID-19. When virus was detected by PCR in the GI tract, immunohistochemistry identified virus in epithelium and lamina propria macrophages, but not in lymphoid tissues. Immunohistochemistry and imaging mass cytometry (IMC) analysis of ileal PP revealed depletion of germinal centres (GC), disruption of B cell/T cell zonation and decreased potential B and T cell interaction and lower nuclear density in COVID-19 patients. This occurred independent of the local viral levels. The changes in PP demonstrate that the ability to mount an intestinal immune response is compromised in severe COVID-19, which could contribute to observed dysbiosis.
Collapse
Affiliation(s)
- Silvia C. Trevelin
- Peter Gorer Department of Immunology, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Suzanne Pickering
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Katrina Todd
- National Institute for Health Research (NIHR) Guy’s and St. Thomas Biomedical Research Centre at Guy’s and St. Thomas NHS Foundation Trust and King’s College London, London, United Kingdom
| | - Cynthia Bishop
- National Institute for Health Research (NIHR) Guy’s and St. Thomas Biomedical Research Centre at Guy’s and St. Thomas NHS Foundation Trust and King’s College London, London, United Kingdom
| | - Michael Pitcher
- Peter Gorer Department of Immunology, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Jose Garrido Mesa
- National Institute for Health Research (NIHR) Guy’s and St. Thomas Biomedical Research Centre at Guy’s and St. Thomas NHS Foundation Trust and King’s College London, London, United Kingdom
| | - Lucia Montorsi
- Peter Gorer Department of Immunology, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Filomena Spada
- National Institute for Health Research (NIHR) Guy’s and St. Thomas Biomedical Research Centre at Guy’s and St. Thomas NHS Foundation Trust and King’s College London, London, United Kingdom
| | - Nedyalko Petrov
- National Institute for Health Research (NIHR) Guy’s and St. Thomas Biomedical Research Centre at Guy’s and St. Thomas NHS Foundation Trust and King’s College London, London, United Kingdom
| | - Anna Green
- Department of Histopathology, Guy’s and St. Thomas NHS Foundation Trust and King’s College London, London, United Kingdom
| | - Manu Shankar-Hari
- Centre for Inflammation Research, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Stuart J.D. Neil
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Jo Spencer
- Peter Gorer Department of Immunology, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| |
Collapse
|
8
|
Ligotti ME, Pojero F, Accardi G, Aiello A, Caruso C, Duro G, Candore G. Immunopathology and Immunosenescence, the Immunological Key Words of Severe COVID-19. Is There a Role for Stem Cell Transplantation? Front Cell Dev Biol 2021; 9:725606. [PMID: 34595175 PMCID: PMC8477205 DOI: 10.3389/fcell.2021.725606] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/06/2021] [Indexed: 01/08/2023] Open
Abstract
The outcomes of Coronavirus disease-2019 (COVID-19) vary depending on the age, health status and sex of an individual, ranging from asymptomatic to lethal. From an immunologic viewpoint, the final severe lung damage observed in COVID-19 should be caused by cytokine storm, driven mainly by interleukin-6 and other pro-inflammatory cytokines. However, which immunopathogenic status precedes this "cytokine storm" and why the male older population is more severely affected, are currently unanswered questions. The aging of the immune system, i.e., immunosenescence, closely associated with a low-grade inflammatory status called "inflammageing," should play a key role. The remodeling of both innate and adaptive immune response observed with aging can partly explain the age gradient in severity and mortality of COVID-19. This review discusses how aging impacts the immune response to the virus, focusing on possible strategies to rejuvenate the immune system with stem cell-based therapies. Indeed, due to immunomodulatory and anti-inflammatory properties, multipotent mesenchymal stem cells (MSCs) are a worth-considering option against COVID-19 adverse outcomes.
Collapse
Affiliation(s)
- Mattia Emanuela Ligotti
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
- Institute for Biomedical Research and Innovation, National Research Council of Italy, Palermo, Italy
| | - Fanny Pojero
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Giulia Accardi
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Anna Aiello
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Calogero Caruso
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
- International Society on Aging and Disease, Fort Worth, TX, United States
| | - Giovanni Duro
- Institute for Biomedical Research and Innovation, National Research Council of Italy, Palermo, Italy
| | - Giuseppina Candore
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| |
Collapse
|
9
|
Zhao L, Hu S, Davila ML, Yang J, Lin YD, Albanese JM, Lo Y, Wang Y, Kennett MJ, Liu Q, Xiong N. Coordinated co-migration of CCR10 + antibody-producing B cells with helper T cells for colonic homeostatic regulation. Mucosal Immunol 2021; 14:420-430. [PMID: 32773769 PMCID: PMC7870723 DOI: 10.1038/s41385-020-0333-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 02/04/2023]
Abstract
In the intestine, IgA antibody-secreting B cells (IgA-ASCs) and helper T cells coordinate to maintain local homeostasis while their dysregulation could lead to development of intestinal inflammatory diseases. However, mechanisms underlying the coordinated localization and function of the B and T cells into the intestine, particularly the colon, are poorly understood. We herein report the first evidence that the gut-homing chemokine receptor CCR10+ IgA-ASCs form conjugates with helper T cells, preferentially regulatory T cells, at their differentiation sites of gut-associated lymphoid organs for their coordinated co-localization into the colon to promote local homeostasis. In CCR10-knockout mice, defective migration of IgA-ASCs also resulted in defective T-cell migration and homeostasis, and development of inflammatory symptoms in the colon. Antigen-specific interaction of CCR10+ IgA-ASCs and T cells is crucial for their homeostatic establishment in the colon. On the other hand, in IgA-knockout mice, preferential expansion of CCR10+ IgG1-ASCs with regulatory functions compensated for CCR10+ IgA-ASCs to help maintain colonic homeostasis. The preferential expansion of specific subclasses of CCR10+ IgG-ASCs with regulatory functions was also found in asymptomatic IgA-deficient patients. These findings suggest coordinated cell migration as a novel mechanism underlying localization and function of B and T cells in colonic homeostatic regulation.
Collapse
Affiliation(s)
- Luming Zhao
- Center for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA
| | - Shaomin Hu
- Center for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA.,Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, 111 East 210th Street, Bronx, NY 10467, USA,Current address: Department of Pathology, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Micha L. Davila
- Center for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA.,Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Jie Yang
- Center for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA.,Current address: Precision for Medicine-Houston Site, 2575 West Bellfort, Suite 190, Houston, TX 77054, USA
| | - Yang-Ding Lin
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Joseph M. Albanese
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, 111 East 210th Street, Bronx, NY 10467, USA
| | - Yungtai Lo
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Yanhua Wang
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, 111 East 210th Street, Bronx, NY 10467, USA
| | - Mary J. Kennett
- Center for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA
| | - Qiang Liu
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, 111 East 210th Street, Bronx, NY 10467, USA
| | - Na Xiong
- Center for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA.,Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA,Department of Medicine-Division of Dermatology and Cutaneous Surgery, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.,Correspondence to: Na Xiong, Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229. ; Tel: 2104505362
| |
Collapse
|
10
|
Nettleford SK, Zhao L, Qian F, Herold M, Arner B, Desai D, Amin S, Xiong N, Singh V, Carlson BA, Prabhu KS. The Essential Role of Selenoproteins in the Resolution of Citrobacter rodentium-Induced Intestinal Inflammation. Front Nutr 2020; 7:96. [PMID: 32775340 PMCID: PMC7381334 DOI: 10.3389/fnut.2020.00096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/26/2020] [Indexed: 12/13/2022] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) leads to adverse colonic inflammation associated with poor resolution of inflammation and loss of epithelial integrity. Micronutrient trace element selenium (Se) is incorporated into selenoproteins as the 21st amino acid, selenocysteine (Sec). Previous studies have shown that such an incorporation of Sec into the selenoproteome is key for the anti-inflammatory functions of Se in macrophages and other immune cells. An intriguing mechanism underlying the anti-inflammatory and pro-resolving effects of Se stems from the ability of selenoproteins to skew arachidonic acid metabolism from pro-inflammatory mediators, prostaglandin E2 (PGE2) toward anti-inflammatory mediators derived from PGD2, such as 15-deoxy-Δ12, 14- prostaglandin J2 (15d-PGJ2), via eicosanoid class switching of bioactive lipids. The impact of Se and such an eicosanoid-class switching mechanism was tested in an enteric infection model of gut inflammation by C. rodentium, a murine equivalent of EPEC. C57BL/6 mice deficient in Se (Se-D) experienced higher mortality when compared to those on Se adequate (0.08 ppm Se) and Se supplemented (0.4 ppm Se) diets following infection. Decreased survival was associated with decreased group 3 innate lymphoid cells (ILC3s) and T helper 17 (Th17) cells in colonic lamina propria of Se-D mice along with deceased expression of epithelial barrier protein Zo-1. Inhibition of metabolic inactivation of PGE2 by 15-prostaglandin dehydrogenase blocked the Se-dependent increase in ILC3 and Th17 cells in addition to reducing epithelial barrier integrity, as seen by increased systemic levels of FITC-dextran following oral administration; while 15d-PGJ2 administration in Se-D mice alleviated the effects by increasing ILC3 and Th17 cells. Mice lacking selenoproteins in monocyte/macrophages via the conditional deletion of the tRNA[Sec] showed increased mortality post infection. Our studies indicate a crucial role for dietary Se in the protection against inflammation following enteric infection via immune mechanisms involving epithelial barrier integrity.
Collapse
Affiliation(s)
- Shaneice K Nettleford
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, The Pennsylvania State University, State College, PA, United States
| | - Luming Zhao
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, The Pennsylvania State University, State College, PA, United States
| | - Fenghua Qian
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, The Pennsylvania State University, State College, PA, United States
| | - Morgan Herold
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, The Pennsylvania State University, State College, PA, United States
| | - Brooke Arner
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, The Pennsylvania State University, State College, PA, United States
| | - Dhimant Desai
- Department of Pharmacology, Organic Synthesis Core Laboratory, Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, United States
| | - Shantu Amin
- Department of Pharmacology, Organic Synthesis Core Laboratory, Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, United States
| | - Na Xiong
- Department of Microbiology, Immunology & Molecular Genetics, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Vishal Singh
- Department of Nutritional Sciences, The Pennsylvania State University, State College, PA, United States
| | - Bradley A Carlson
- Molecular Biology of Selenium Section, Mouse Genetics Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - K Sandeep Prabhu
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, The Pennsylvania State University, State College, PA, United States
| |
Collapse
|
11
|
Yang J, Restori KH, Xu M, Song EH, Zhao L, Hu S, Lyu P, Wang WB, Xiong N. Preferential Perinatal Development of Skin-Homing NK1.1 + Innate Lymphoid Cells for Regulation of Cutaneous Microbiota Colonization. iScience 2020; 23:101014. [PMID: 32283522 PMCID: PMC7155142 DOI: 10.1016/j.isci.2020.101014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 03/10/2020] [Accepted: 03/24/2020] [Indexed: 12/30/2022] Open
Abstract
Proper immune cell development at early ontogenic stages is critical for life-long health. How resident immune cells are established in barrier tissues at neonatal stages to provide early protection is an important but still poorly understood question. We herein report that a developmentally programmed preferential generation of skin-homing group 1 innate lymphoid cells (ILC1s) at perinatal stages helps regulate early skin microbiota colonization. We found that a population of skin-homing NK1.1+ ILC1s was preferentially generated in the perinatal thymi of mice. Unique thymic environments and progenitor cells are responsible for the preferential generation of skin-homing NK1.1+ ILC1s at perinatal stages. In the skin, NK1.1+ ILC1s regulate proper microbiota colonization and control the opportunistic pathogen Pseudomonas aeruginosa in neonatal mice. These findings provide insight into the development and function of tissue-specific immune cells at neonatal stages, a critical temporal window for establishment of local tissue immune homeostasis.
Collapse
Affiliation(s)
- Jie Yang
- Department of Veterinary and Biomedical Sciences, Centre for Molecular Immunology and Infectious Disease, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA
| | - Katherine H Restori
- Department of Veterinary and Biomedical Sciences, Centre for Molecular Immunology and Infectious Disease, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA
| | - Ming Xu
- Department of Veterinary and Biomedical Sciences, Centre for Molecular Immunology and Infectious Disease, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA; Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Eun Hyeon Song
- Department of Veterinary and Biomedical Sciences, Centre for Molecular Immunology and Infectious Disease, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA; Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Luming Zhao
- Department of Veterinary and Biomedical Sciences, Centre for Molecular Immunology and Infectious Disease, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA
| | - Shaomin Hu
- Department of Veterinary and Biomedical Sciences, Centre for Molecular Immunology and Infectious Disease, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA
| | - Pingyun Lyu
- Department of Veterinary and Biomedical Sciences, Centre for Molecular Immunology and Infectious Disease, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA
| | - Wei-Bei Wang
- Department of Veterinary and Biomedical Sciences, Centre for Molecular Immunology and Infectious Disease, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA; Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Na Xiong
- Department of Veterinary and Biomedical Sciences, Centre for Molecular Immunology and Infectious Disease, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA; Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; Department of Medicine-Division of Dermatology and Cutaneous Surgery, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| |
Collapse
|
12
|
Gary EN, Kathuria N, Makurumidze G, Curatola A, Ramamurthi A, Bernui ME, Myles D, Yan J, Pankhong P, Muthumani K, Haddad E, Humeau L, Weiner DB, Kutzler MA. CCR10 expression is required for the adjuvant activity of the mucosal chemokine CCL28 when delivered in the context of an HIV-1 Env DNA vaccine. Vaccine 2020; 38:2626-2635. [PMID: 32057572 PMCID: PMC10681704 DOI: 10.1016/j.vaccine.2020.01.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 12/05/2019] [Accepted: 01/07/2020] [Indexed: 01/07/2023]
Abstract
An effective prophylactic vaccine targeting HIV must induce a robust humoral response and must direct the bulk of this response to the mucosa-the primary site of HIV transmission. The chemokine, CCL28, is secreted by epithelial cells at mucosal surfaces and recruits' cells expressing its receptor CCR10. CCR10 is predominantly expressed by IgA + ASCs. We hypothesized that co-immunization with plasmid DNA encoding consensus envelope antigens with plasmid-encoded CCL28 would enhance anti-HIV IgA responses at mucosal surfaces. Indeed, animals receiving pCCL28 and pEnvA/C had significantly increased HIV-specific IgA in fecal extract. Surprisingly, CCL28 co-immunization induced a significant increase in anti-HIV IgG in the serum in mice compared to those receiving pEnvA/C alone. These robust antibody responses were not associated with changes in the frequency of germinal center B cells but depended upon the expression of CCR10, as these responses we abolished in CCR10-deficient animals. Finally, immunization with CCL28 led to increased frequencies in HIV-specific CCR10 + and CCR10 + IgA + B cells in the small intestine and Peyer's patches of vaccinated animals as compared to those receiving pEnvA/C alone. These data indicate that CCL28 administration can enhance antigen-specific humoral responses systemically and at mucosal surfaces.
Collapse
Affiliation(s)
- E N Gary
- The Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - N Kathuria
- The Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - G Makurumidze
- The Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, United States
| | - A Curatola
- The Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - A Ramamurthi
- The Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - M E Bernui
- The Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States; The Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, United States
| | - D Myles
- The Department of Pathology and Laboratory Medicine, The University of Pennsylvania, Philadelphia, PA, United States
| | - J Yan
- Inovio Pharmaceuticals, Blue Bell, PA, United States
| | - P Pankhong
- The Department of Pathology and Laboratory Medicine, The University of Pennsylvania, Philadelphia, PA, United States
| | - K Muthumani
- The Wistar Institute, Philadelphia, PA, United States
| | - E Haddad
- The Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, United States
| | - L Humeau
- Inovio Pharmaceuticals, Blue Bell, PA, United States
| | - D B Weiner
- The Wistar Institute, Philadelphia, PA, United States
| | - M A Kutzler
- The Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States; The Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, United States.
| |
Collapse
|
13
|
Host Factors Affecting Generation of Immunity Against Porcine Epidemic Diarrhea Virus in Pregnant and Lactating Swine and Passive Protection of Neonates. Pathogens 2020; 9:pathogens9020130. [PMID: 32085410 PMCID: PMC7168134 DOI: 10.3390/pathogens9020130] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 02/08/2023] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) is a highly virulent re-emerging enteric coronavirus that causes acute diarrhea, dehydration, and up to 100% mortality in neonatal suckling piglets. Despite this, a safe and effective PEDV vaccine against highly virulent strains is unavailable, making PEDV prevention and control challenging. Lactogenic immunity induced via the gut-mammary gland-secretory IgA (sIgA) axis, remains the most promising and effective way to protect suckling piglets from PEDV. Therefore, a successful PEDV vaccine must induce protective maternal IgA antibodies that passively transfer into colostrum and milk. Identifying variables that influence lymphocyte migration and IgA secretion during gestation and lactation is imperative for designing maternal immunization strategies that generate the highest amount of lactogenic immune protection against PEDV in suckling piglets. Because pregnancy-associated immune alterations influence viral pathogenesis and adaptive immune responses in many different species, a better understanding of host immune responses to PEDV in pregnant swine may translate into improved maternal immunization strategies against enteric pathogens for multiple species. In this review, we discuss the role of host factors during pregnancy on antiviral immunity and their implications for generating protective lactogenic immunity in suckling neonates.
Collapse
|
14
|
Price MJ, Hicks SL, Bradley JE, Randall TD, Boss JM, Scharer CD. IgM, IgG, and IgA Influenza-Specific Plasma Cells Express Divergent Transcriptomes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:2121-2129. [PMID: 31501259 PMCID: PMC6783370 DOI: 10.4049/jimmunol.1900285] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 08/13/2019] [Indexed: 12/31/2022]
Abstract
Ab-secreting cells (ASC) or plasma cells are essential components of the humoral immune system. Although Abs of different isotypes have distinct functions, it is not known if the ASC that secrete each isotype are also distinct. ASC downregulate their surface BCR upon differentiation, hindering analyses that couple BCR information to other molecular characteristics. In this study, we developed a methodology using fixation, permeabilization, and intracellular staining coupled with cell sorting and reversal of the cross-links to allow RNA sequencing of isolated cell subsets. Using hemagglutinin and nucleoprotein Ag-specific B cell tetramers and intracellular staining for IgM, IgG, and IgA isotypes, we were able to derive and compare the gene expression programs of ASC subsets that were responding to the same Ags following influenza infection in mice. Intriguingly, whereas a shared ASC signature was identified, each ASC isotype-specific population expressed distinct transcriptional programs controlling cellular homing, metabolism, and potential effector functions. Additionally, we extracted and compared BCR clonotypes and found that each ASC isotype contained a unique, clonally related CDR3 repertoire. In summary, these data reveal specific complexities in the transcriptional programming of Ag-specific ASC populations.
Collapse
Affiliation(s)
- Madeline J Price
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Sakeenah L Hicks
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - John E Bradley
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294; and
| | - Troy D Randall
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294; and
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322;
| |
Collapse
|
15
|
Lu E, Cyster JG. G-protein coupled receptors and ligands that organize humoral immune responses. Immunol Rev 2019; 289:158-172. [PMID: 30977196 PMCID: PMC6464390 DOI: 10.1111/imr.12743] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 01/22/2019] [Indexed: 12/26/2022]
Abstract
B-cell responses are dynamic processes that depend on multiple types of interactions. Rare antigen-specific B cells must encounter antigen and specialized systems are needed-unique to each lymphoid tissue type-to ensure this happens efficiently. Lymphoid tissue barrier cells act to ensure that pathogens, while being permitted entry for B-cell recognition, are blocked from replication or dissemination. T follicular helper (Tfh) cells often need to be primed by dendritic cells before supporting B-cell responses. For most responses, antigen-specific helper T cells and B cells need to interact, first to initiate clonal expansion and the plasmablast response, and later to support the germinal center (GC) response. Newly formed plasma cells need to travel to supportive niches. GC B cells must become confined to the follicle center, organize into dark and light zones, and interact with Tfh cells. Memory B cells need to be positioned for rapid responses following reinfection. Each of these events requires the actions of multiple G-protein coupled receptors (GPCRs) and their ligands, including chemokines and lipid mediators. This review will focus on the guidance cue code underlying B-cell immunity, with an emphasis on findings from our laboratory and on newer advances in related areas. We will discuss our recent identification of geranylgeranyl-glutathione as a ligand for P2RY8. Our goal is to provide the reader with a focused knowledge about the GPCRs guiding B-cell responses and how they might be therapeutic targets, while also providing examples of how multiple types of GPCRs can cooperate or act iteratively to control cell behavior.
Collapse
Affiliation(s)
- Erick Lu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California
| |
Collapse
|
16
|
Langel SN, Paim FC, Alhamo MA, Buckley A, Van Geelen A, Lager KM, Vlasova AN, Saif LJ. Stage of Gestation at Porcine Epidemic Diarrhea Virus Infection of Pregnant Swine Impacts Maternal Immunity and Lactogenic Immune Protection of Neonatal Suckling Piglets. Front Immunol 2019; 10:727. [PMID: 31068924 PMCID: PMC6491507 DOI: 10.3389/fimmu.2019.00727] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 03/18/2019] [Indexed: 01/22/2023] Open
Abstract
During pregnancy, the maternal immune response changes dramatically over the course of gestation. This has implications for generation of lactogenic immunity and subsequent protection in suckling neonates against enteric viral infections. For example, porcine epidemic diarrhea virus (PEDV) is an alphacoronavirus that causes acute diarrhea in neonatal piglets. Due to the high virulence of PEDV and the naïve, immature immune system of neonatal suckling piglets, passive lactogenic immunity to PEDV induced during pregnancy, via the gut-mammary gland (MG)-secretory IgA (sIgA) axis, is critical for piglet protection. However, the anti-PEDV immune response during pregnancy and stage of gestation required to optimally stimulate the gut-MG-sIgA axis is undefined. We hypothesize that there is a gestational window in which non-lethal PEDV infection of pregnant gilts influences maximum lymphocyte mucosal trafficking to the MG, resulting in optimal passive lactogenic protection in suckling piglets. To understand how the stages of gestation affect maternal immune responses to PEDV, three groups of gilts were orally infected with PEDV in the first, second or third trimester. Control (mock) gilts were inoculated with medium in the third trimester. To determine if lactogenic immunity correlated with protection, all piglets were PEDV-challenged at 3–5 days postpartum. PEDV infection of gilts at different stages of gestation significantly affected multiple maternal systemic immune parameters prepartum, including cytokines, B cells, PEDV antibodies (Abs), and PEDV antibody secreting cells (ASCs). Pregnant second trimester gilts had significantly higher levels of circulating PEDV IgA and IgG Abs and ASCs and PEDV virus neutralizing (VN) Abs post PEDV infection. Coinciding with the significantly higher PEDV Ab responses in second trimester gilts, the survival rate of their PEDV-challenged piglets was 100%, compared with 87.2, 55.9, and 5.7% for first, third, and mock litters, respectively. Additionally, piglet survival positively correlated with PEDV IgA Abs and ASCs and VN Abs in milk and PEDV IgA and IgG Abs in piglet serum. Our findings have implications for gestational timing of oral attenuated PEDV maternal vaccines, whereby PEDV intestinal infection in the second trimester optimally stimulated the gut-MG-sIgA axis resulting in 100% lactogenic immune protection in suckling piglets.
Collapse
Affiliation(s)
- Stephanie N Langel
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, College of Food, Agriculture and Environmental Sciences, College of Veterinary Medicine, The Ohio State University, Wooster, OH, United States
| | - Francine C Paim
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, College of Food, Agriculture and Environmental Sciences, College of Veterinary Medicine, The Ohio State University, Wooster, OH, United States
| | - Moyasar A Alhamo
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, College of Food, Agriculture and Environmental Sciences, College of Veterinary Medicine, The Ohio State University, Wooster, OH, United States
| | - Alexandra Buckley
- National Animal Disease Center, Agricultural Research Service, USDA, Ames, IA, United States
| | - Albert Van Geelen
- National Animal Disease Center, Agricultural Research Service, USDA, Ames, IA, United States
| | - Kelly M Lager
- National Animal Disease Center, Agricultural Research Service, USDA, Ames, IA, United States
| | - Anastasia N Vlasova
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, College of Food, Agriculture and Environmental Sciences, College of Veterinary Medicine, The Ohio State University, Wooster, OH, United States
| | - Linda J Saif
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, College of Food, Agriculture and Environmental Sciences, College of Veterinary Medicine, The Ohio State University, Wooster, OH, United States
| |
Collapse
|
17
|
Burkhardt AM, Perez-Lopez A, Ushach I, Catalan-Dibene J, Nuccio SP, Chung LK, Hernandez-Ruiz M, Carnevale C, Raffatellu M, Zlotnik A. CCL28 Is Involved in Mucosal IgA Responses, Olfaction, and Resistance to Enteric Infections. J Interferon Cytokine Res 2019; 39:214-223. [PMID: 30855201 PMCID: PMC6479244 DOI: 10.1089/jir.2018.0099] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 12/10/2018] [Indexed: 01/14/2023] Open
Abstract
CCL28 is a mucosal chemokine that has been involved in various responses, including IgA production. We have analyzed its production in human tissues using a comprehensive microarray database. Its highest expression is in the salivary gland, indicating that it is an important component of saliva. It is also expressed in the trachea, bronchus, and in the mammary gland upon onset of lactation. We have also characterized a Ccl28-/- mouse that exhibits very low IgA levels in milk, and the IgA levels in feces are also reduced. These observations confirm a role for the CCL28/CCR10 chemokine axis in the recruitment of IgA plasmablasts to the lactating mammary gland. CCL28 is also expressed in the vomeronasal organ. We also detected olfactory defects (anosmia) in a Ccl28-/- mouse suggesting that CCL28 is involved in the function/development of olfaction. Importantly, Ccl28-/- mice are highly susceptible to Salmonella enterica serovar Typhimurium in an acute model of infection, indicating that CCL28 plays a major role in innate immunity against Salmonella in the gut. Finally, microbiome studies revealed modest differences in the gut microbiota between Ccl28-/- mice and their cohoused wild-type littermates. The latter observation suggests that under homeostatic conditions, CCL28 plays a limited role in shaping the gut microbiome.
Collapse
Affiliation(s)
- Amanda M. Burkhardt
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, California
- Institute for Immunology, University of California, Irvine, Irvine, California
| | - Araceli Perez-Lopez
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, California
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, San Diego, California
| | - Irina Ushach
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, California
- Institute for Immunology, University of California, Irvine, Irvine, California
| | - Jovani Catalan-Dibene
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, California
- Institute for Immunology, University of California, Irvine, Irvine, California
| | - Sean-Paul Nuccio
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, California
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, San Diego, California
| | - Lawton K. Chung
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, California
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, San Diego, California
| | - Marcela Hernandez-Ruiz
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, California
- Institute for Immunology, University of California, Irvine, Irvine, California
| | - Christina Carnevale
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, California
- Institute for Immunology, University of California, Irvine, Irvine, California
| | - Manuela Raffatellu
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, California
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, San Diego, California
- Chiba University-UC San Diego Center for Mucosal Immunology, Allergy and Vaccines (CU-UCSD-cMAV), University of California, San Diego, San Diego, California
- Center for Microbiome Innovation, University of California, San Diego, San Diego, California
| | - Albert Zlotnik
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, California
- Institute for Immunology, University of California, Irvine, Irvine, California
| |
Collapse
|
18
|
Benelli R, Venè R, Ferrari N. Prostaglandin-endoperoxide synthase 2 (cyclooxygenase-2), a complex target for colorectal cancer prevention and therapy. Transl Res 2018; 196:42-61. [PMID: 29421522 DOI: 10.1016/j.trsl.2018.01.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/15/2017] [Accepted: 01/09/2018] [Indexed: 12/16/2022]
Abstract
A plentiful literature has linked colorectal cancer (CRC) to inflammation and prostaglandin-endoperoxide synthase (PTGS)2 expression. Accordingly, several nonsteroidal antiinflammatory drugs (NSAIDs) have been tested often successfully in CRC chemoprevention despite their different ability to specifically target PTGS2 and the low or null expression of PTGS2 in early colon adenomas. Some observational studies showed an increased survival for patients with CRC assuming NSAIDs after diagnosis, but no clinical trial has yet demonstrated the efficacy of NSAIDs against established CRC, where PTGS2 is expressed at high levels. The major limits for the application of NSAIDs, or specific PTGS2 inhibitors, as adjuvant drugs in CRC are (1) a frequent confusion about the physiological role of PTGS1 and PTGS2, reflecting in CRC pathology and therapy; (2) the presence of unavoidable side effects linked to the intrinsic function of these enzymes; (3) the need of established criteria and markers for patient selection; and (4) the evaluation of the immunomodulatory potential of PTGS2 inhibitors as possible adjuvants for immunotherapy. This review has been written to rediscover the multifaceted potential of PTGS2 targeting, hoping it could act as a starting point for a new and more aware application of NSAIDs against CRC.
Collapse
Affiliation(s)
- Roberto Benelli
- OU Immunology, Ospedale Policlinico San Martino (Istituto di ricovero e cura a carattere scientifico per l'oncologia), Genoa, Italy.
| | - Roberta Venè
- OU Molecular Oncology & Angiogenesis, Ospedale Policlinico San Martino (Istituto di ricovero e cura a carattere scientifico per l'oncologia), Genoa, Italy
| | - Nicoletta Ferrari
- OU Molecular Oncology & Angiogenesis, Ospedale Policlinico San Martino (Istituto di ricovero e cura a carattere scientifico per l'oncologia), Genoa, Italy
| |
Collapse
|
19
|
Hughes CE, Nibbs RJB. A guide to chemokines and their receptors. FEBS J 2018; 285:2944-2971. [PMID: 29637711 PMCID: PMC6120486 DOI: 10.1111/febs.14466] [Citation(s) in RCA: 883] [Impact Index Per Article: 126.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/25/2018] [Accepted: 04/03/2018] [Indexed: 12/12/2022]
Abstract
The chemokines (or chemotactic cytokines) are a large family of small, secreted proteins that signal through cell surface G protein-coupled heptahelical chemokine receptors. They are best known for their ability to stimulate the migration of cells, most notably white blood cells (leukocytes). Consequently, chemokines play a central role in the development and homeostasis of the immune system, and are involved in all protective or destructive immune and inflammatory responses. Classically viewed as inducers of directed chemotactic migration, it is now clear that chemokines can stimulate a variety of other types of directed and undirected migratory behavior, such as haptotaxis, chemokinesis, and haptokinesis, in addition to inducing cell arrest or adhesion. However, chemokine receptors on leukocytes can do more than just direct migration, and these molecules can also be expressed on, and regulate the biology of, many nonleukocytic cell types. Chemokines are profoundly affected by post-translational modification, by interaction with the extracellular matrix (ECM), and by binding to heptahelical 'atypical' chemokine receptors that regulate chemokine localization and abundance. This guide gives a broad overview of the chemokine and chemokine receptor families; summarizes the complex physical interactions that occur in the chemokine network; and, using specific examples, discusses general principles of chemokine function, focusing particularly on their ability to direct leukocyte migration.
Collapse
Affiliation(s)
- Catherine E Hughes
- Institute of Infection, Inflammation & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Robert J B Nibbs
- Institute of Infection, Inflammation & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| |
Collapse
|
20
|
Thangavadivel S, Zelle-Rieser C, Olivier A, Postert B, Untergasser G, Kern J, Brunner A, Gunsilius E, Biedermann R, Hajek R, Pour L, Willenbacher W, Greil R, Jöhrer K. CCR10/CCL27 crosstalk contributes to failure of proteasome-inhibitors in multiple myeloma. Oncotarget 2018; 7:78605-78618. [PMID: 27732933 PMCID: PMC5346663 DOI: 10.18632/oncotarget.12522] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 10/03/2016] [Indexed: 11/25/2022] Open
Abstract
The bone marrow microenvironment plays a decisive role in multiple myeloma progression and drug resistance. Chemokines are soluble mediators of cell migration, proliferation and survival and essentially modulate tumor progression and drug resistance. Here we investigated bone marrow-derived chemokines of naive and therapy-refractory myeloma patients and discovered that high levels of the chemokine CCL27, known so far for its role in skin inflammatory processes, correlated with worse overall survival of the patients. In addition, chemokine levels were significantly higher in samples from patients who became refractory to bortezomib at first line treatment compared to resistance at later treatment lines. In vitro as well as in an in vivo model we could show that CCL27 triggers bortezomib-resistance of myeloma cells. This effect was strictly dependent on the expression of the respective receptor, CCR10, on stroma cells and involved the modulation of IL-10 expression, activation of myeloma survival pathways, and modulation of proteasomal activity. Drug resistance could be totally reversed by blocking CCR10 by siRNA as well as blocking IL-10 and its receptor. From our data we suggest that blocking the CCR10/CCL27/IL-10 myeloma-stroma crosstalk is a novel therapeutic target that could be especially relevant in early refractory myeloma patients.
Collapse
Affiliation(s)
| | | | | | - Benno Postert
- Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Gerold Untergasser
- Tyrolean Cancer Research Institute, Innsbruck, Austria.,Laboratory of Tumor Angiogenesis and Tumorbiology, Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Austria
| | - Johann Kern
- Laboratory of Tumor Angiogenesis and Tumorbiology, Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Austria
| | - Andrea Brunner
- Department of Pathology, Medical University Innsbruck, Innsbruck, Austria
| | - Eberhard Gunsilius
- Laboratory of Tumor Angiogenesis and Tumorbiology, Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Austria
| | - Rainer Biedermann
- Department of Orthopedic Surgery, Medical University Innsbruck, Innsbruck, Austria
| | - Roman Hajek
- Babak Myeloma Group, Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Department of Clinical Hematology, University Hospital Brno, Brno, Czech Republic.,Department of Hematooncology, Faculty of Medicine, University of Ostrava and University Hospital Ostrava, Ostrava, Czech Republic
| | - Ludek Pour
- Babak Myeloma Group, Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Department of Clinical Hematology, University Hospital Brno, Brno, Czech Republic.,Department of Hematooncology, Faculty of Medicine, University of Ostrava and University Hospital Ostrava, Ostrava, Czech Republic
| | - Wolfgang Willenbacher
- Department of Internal Medicine V, University Hospital Innsbruck, Innsbruck, Austria
| | - Richard Greil
- Tyrolean Cancer Research Institute, Innsbruck, Austria.,Salzburg Cancer Research Institute-Laboratory of Immunological and Molecular Cancer Research, Salzburg, Austria.,Third Medical Department at The Paracelsus Medical University Salzburg, Austria.,Cancer Cluster Salzburg (CCS), Salzburg, Austria
| | - Karin Jöhrer
- Tyrolean Cancer Research Institute, Innsbruck, Austria
| |
Collapse
|
21
|
Zhiming W, Luman W, Tingting Q, Yiwei C. Chemokines and receptors in intestinal B lymphocytes. J Leukoc Biol 2018; 103:807-819. [PMID: 29443417 DOI: 10.1002/jlb.1ru0717-299rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 01/11/2018] [Accepted: 01/11/2018] [Indexed: 02/06/2023] Open
Abstract
Recent studies indicate that chemoattractant cytokines (chemokines) and their receptors modulate intestinal B lymphocytes in different ways, including regulating their maturity and differentiation in the bone marrow and homing to intestinal target tissues. Here, we review several important chemokine/chemokine receptor axes that guide intestinal B cells, focusing on the homing and migration of IgA antibody-secreting cells (IgA-ASCs) to intestinal-associated lymphoid tissues. We describe the selective regulation of these chemokine axes in coordinating the IgA-ASC trafficking in intestinal diseases. Finally, we discuss the role of B cells as chemokine producers serving dual roles in regulating the mucosal immune microenvironment.
Collapse
Affiliation(s)
- Wang Zhiming
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wang Luman
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Biotherapy Research Center, Fudan University, Shanghai, China
| | - Qian Tingting
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chu Yiwei
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Biotherapy Research Center, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Matsuo K, Nagakubo D, Yamamoto S, Shigeta A, Tomida S, Fujita M, Hirata T, Tsunoda I, Nakayama T, Yoshie O. CCL28-Deficient Mice Have Reduced IgA Antibody-Secreting Cells and an Altered Microbiota in the Colon. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:800-809. [PMID: 29237777 DOI: 10.4049/jimmunol.1700037] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 11/14/2017] [Indexed: 02/05/2023]
Abstract
CCL28 induces the migration of IgA Ab-secreting cells (ASCs) via CCR10 and also displays a potent antimicrobial activity in vitro. To explore the role of CCL28 in vivo, we generated CCL28-deficient mice. The mice exhibited a significant reduction and abnormal distribution of IgA ASCs in the lamina propria of the colon. The concentrations of total and Ag-specific IgA in the fecal extracts of CCL28-deficient mice were also drastically reduced. The average amount of IgA secreted by a single IgA ASC derived from the colon was also substantially reduced in CCL28-deficient mice. Furthermore, CCL28 was found to significantly increase the average amount of IgA secreted by a single IgA ASC derived from the colon in vitro. In contrast, the generation of IgA ASCs in Peyer's and cecal patches was not significantly impaired in CCL28-deficient mice. We also found a relative increase in the Class Bacilli in the fecal extracts of CCL28-deficient mice and demonstrated a potent antimicrobial activity of CCL28 against Bacillus cereus and Enterococcus faecalis, both of which belong to Class Bacilli. Thus, CCL28 may also suppress the outgrowth of some bacterial species by its direct antimicrobial activity. Finally, CCL28-deficient mice exhibited a highly aggravated dextran sodium sulfate-induced colitis that was ameliorated by pretreatment with antibiotics. Collectively, CCL28 plays a pivotal role in the homing, distribution, and function of IgA ASCs in the colon and may also affect the intestinal microbiota through its direct antimicrobial activity.
Collapse
Affiliation(s)
- Kazuhiko Matsuo
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Daisuke Nagakubo
- Department of Fundamental Biosciences, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Shinya Yamamoto
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Akiko Shigeta
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-8511, Japan; and
| | - Shuta Tomida
- Department of Biobank, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama 700-8558, Japan
| | - Mitsugu Fujita
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-8511, Japan; and
| | - Takako Hirata
- Department of Fundamental Biosciences, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Ikuo Tsunoda
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-8511, Japan; and
| | - Takashi Nakayama
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan;
| | - Osamu Yoshie
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-8511, Japan; and
| |
Collapse
|
23
|
|
24
|
Zacarías MF, Souza TC, Zaburlín N, Carmona Cara D, Reinheimer J, Nicoli J, Vinderola G. Influence of Technological Treatments on the Functionality ofBifidobacterium lactisINL1, a Breast Milk-Derived Probiotic. J Food Sci 2017; 82:2462-2470. [DOI: 10.1111/1750-3841.13852] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 07/03/2017] [Accepted: 07/25/2017] [Indexed: 12/31/2022]
Affiliation(s)
- María Florencia Zacarías
- Inst. de Lactología Industrial (INLAIN); Univ. Nacional del Litoral; Santiago del Estero 2829 3000 Santa Fe Argentina
| | - Tassia Costa Souza
- Inst. de Lactología Industrial (INLAIN); Univ. Nacional del Litoral; Santiago del Estero 2829 3000 Santa Fe Argentina
| | - Natalia Zaburlín
- Inst. de Lactología Industrial (INLAIN); Univ. Nacional del Litoral; Santiago del Estero 2829 3000 Santa Fe Argentina
| | - Denise Carmona Cara
- the Dept. de Morfologia, Inst. de Ciências Biológicas (ICB); Univ. Federal de Minas Gerais; Belo Horizonte MG Brazil
| | - Jorge Reinheimer
- Inst. de Lactología Industrial (INLAIN); Univ. Nacional del Litoral; Santiago del Estero 2829 3000 Santa Fe Argentina
| | - Jacques Nicoli
- the Dept. de Microbiologia; Inst. de Ciências Biológicas (ICB); Univ. Federal de Minas Gerais; Belo Horizonte MG Brazil
| | - Gabriel Vinderola
- Inst. de Lactología Industrial (INLAIN); Univ. Nacional del Litoral; Santiago del Estero 2829 3000 Santa Fe Argentina
| |
Collapse
|
25
|
Agace WW, McCoy KD. Regionalized Development and Maintenance of the Intestinal Adaptive Immune Landscape. Immunity 2017; 46:532-548. [PMID: 28423335 DOI: 10.1016/j.immuni.2017.04.004] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 04/03/2017] [Accepted: 04/04/2017] [Indexed: 12/14/2022]
Abstract
The intestinal immune system has the daunting task of protecting us from pathogenic insults while limiting inflammatory responses against the resident commensal microbiota and providing tolerance to food antigens. This role is particularly impressive when one considers the vast mucosal surface and changing landscape that the intestinal immune system must monitor. In this review, we highlight regional differences in the development and composition of the adaptive immune landscape of the intestine and the impact of local intrinsic and environmental factors that shape this process. To conclude, we review the evidence for a critical window of opportunity for early-life exposures that affect immune development and alter disease susceptibility later in life.
Collapse
Affiliation(s)
- William W Agace
- Division of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark (DTU), 2800 Kongens Lyngby, Denmark; Immunology Section, Department of Experimental Medical Science, Lund University, BMC D14, Sölvegatan 19, 221 84 Lund, Sweden.
| | - Kathy D McCoy
- Department of Physiology and Pharmacology and Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
26
|
Parra D, Korytář T, Takizawa F, Sunyer JO. B cells and their role in the teleost gut. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 64:150-66. [PMID: 26995768 PMCID: PMC5125549 DOI: 10.1016/j.dci.2016.03.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 03/11/2016] [Accepted: 03/13/2016] [Indexed: 05/03/2023]
Abstract
Mucosal surfaces are the main route of entry for pathogens in all living organisms. In the case of teleost fish, mucosal surfaces cover the vast majority of the animal. As these surfaces are in constant contact with the environment, fish are perpetually exposed to a vast number of pathogens. Despite the potential prevalence and variety of pathogens, mucosal surfaces are primarily populated by commensal non-pathogenic bacteria. Indeed, a fine balance between these two populations of microorganisms is crucial for animal survival. This equilibrium, controlled by the mucosal immune system, maintains homeostasis at mucosal tissues. Teleost fish possess a diffuse mucosa-associated immune system in the intestine, with B cells being one of the main responders. Immunoglobulins produced by these lymphocytes are a critical line of defense against pathogens and also prevent the entrance of commensal bacteria into the epithelium. In this review we will summarize recent literature regarding the role of B-lymphocytes and immunoglobulins in gut immunity in teleost fish, with specific focus on immunoglobulin isotypes and the microorganisms, pathogenic and non-pathogenic that interact with the immune system.
Collapse
Affiliation(s)
- David Parra
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Tomáš Korytář
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Fumio Takizawa
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - J Oriol Sunyer
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
27
|
Abstract
Secondary lymphoid tissues share the important function of bringing together antigens and rare antigen-specific lymphocytes to foster induction of adaptive immune responses. Peyer's patches (PPs) are unique compared to other secondary lymphoid tissues in their continual exposure to an enormous diversity of microbiome- and food-derived antigens and in the types of pathogens they encounter. Antigens are delivered to PPs by specialized microfold (M) epithelial cells and they may be captured and presented by resident dendritic cells (DCs). In accord with their state of chronic microbial antigen exposure, PPs exhibit continual germinal center (GC) activity. These GCs not only contribute to the generation of B cells and plasma cells producing somatically mutated gut antigen-specific IgA antibodies but have also been suggested to support non-specific antigen diversification of the B-cell repertoire. Here, we review current understanding of how PPs foster B-cell encounters with antigen, how they favor isotype switching to the secretory IgA isotype, and how their GC responses may uniquely contribute to mucosal immunity.
Collapse
Affiliation(s)
- Andrea Reboldi
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
28
|
Habtezion A, Nguyen LP, Hadeiba H, Butcher EC. Leukocyte Trafficking to the Small Intestine and Colon. Gastroenterology 2016; 150:340-54. [PMID: 26551552 PMCID: PMC4758453 DOI: 10.1053/j.gastro.2015.10.046] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 10/21/2015] [Accepted: 10/26/2015] [Indexed: 12/14/2022]
Abstract
Leukocyte trafficking to the small and large intestines is tightly controlled to maintain intestinal immune homeostasis, mediate immune responses, and regulate inflammation. A wide array of chemoattractants, chemoattractant receptors, and adhesion molecules expressed by leukocytes, mucosal endothelium, epithelium, and stromal cells controls leukocyte recruitment and microenvironmental localization in intestine and in the gut-associated lymphoid tissues (GALTs). Naive lymphocytes traffic to the gut-draining mesenteric lymph nodes where they undergo antigen-induced activation and priming; these processes determine their memory/effector phenotypes and imprint them with the capacity to migrate via the lymph and blood to the intestines. Mechanisms of T-cell recruitment to GALT and of T cells and plasmablasts to the small intestine are well described. Recent advances include the discovery of an unexpected role for lectin CD22 as a B-cell homing receptor GALT, and identification of the orphan G-protein-coupled receptor 15 (GPR15) as a T-cell chemoattractant/trafficking receptor for the colon. GPR15 decorates distinct subsets of T cells in mice and humans, a difference in species that could affect translation of the results of mouse colitis models to humans. Clinical studies with antibodies to integrin α4β7 and its vascular ligand mucosal vascular addressin cell adhesion molecule 1 are proving the value of lymphocyte trafficking mechanisms as therapeutic targets for inflammatory bowel diseases. In contrast to lymphocytes, cells of the innate immune system express adhesion and chemoattractant receptors that allow them to migrate directly to effector tissue sites during inflammation. We review the mechanisms for innate and adaptive leukocyte localization to the intestinal tract and GALT, and discuss their relevance to human intestinal homeostasis and inflammation.
Collapse
Affiliation(s)
- Aida Habtezion
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California.
| | - Linh P Nguyen
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Husein Hadeiba
- The Center for Molecular Biology and Medicine, Veterans Affairs Palo Alto Health Care System, The Palo Alto Veterans Institute for Research, Palo Alto, California
| | - Eugene C Butcher
- The Center for Molecular Biology and Medicine, Veterans Affairs Palo Alto Health Care System, The Palo Alto Veterans Institute for Research, Palo Alto, California; Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
29
|
Yang J, Hu S, Zhao L, Kaplan DH, Perdew GH, Xiong N. Selective programming of CCR10(+) innate lymphoid cells in skin-draining lymph nodes for cutaneous homeostatic regulation. Nat Immunol 2015; 17:48-56. [PMID: 26523865 PMCID: PMC4838393 DOI: 10.1038/ni.3312] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/29/2015] [Indexed: 12/15/2022]
Abstract
Innate lymphoid cells (ILCs) 'preferentially' localize into barrier tissues, where they function in tissue protection but can also contribute to inflammatory diseases. The mechanisms that regulate the establishment of ILCs in barrier tissues are poorly understood. Here we found that under steady-state conditions, ILCs in skin-draining lymph nodes (sLNs) were continuously activated to acquire regulatory properties and high expression of the chemokine receptor CCR10 for localization into the skin. CCR10(+) ILCs promoted the homeostasis of skin-resident T cells and, reciprocally, their establishment in the skin required T cell-regulated homeostatic environments. CD207(+) dendritic cells expressing the transcription factor Foxn1 were required for the proper generation of CCR10(+) ILCs. These observations reveal mechanisms that underlie the specific programming and priming of skin-homing CCR10(+) ILCs in the sLNs.
Collapse
Affiliation(s)
- Jie Yang
- Center for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Shaomin Hu
- Center for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Luming Zhao
- Center for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Daniel H Kaplan
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Gary H Perdew
- Center for Molecular Toxicology &Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Na Xiong
- Center for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
30
|
Chen Z, Kim SJ, Essani AB, Volin MV, Vila OM, Swedler W, Arami S, Volkov S, Sardin LV, Sweiss N, Shahrara S. Characterising the expression and function of CCL28 and its corresponding receptor, CCR10, in RA pathogenesis. Ann Rheum Dis 2015; 74:1898-906. [PMID: 24833787 PMCID: PMC4282625 DOI: 10.1136/annrheumdis-2013-204530] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 04/16/2014] [Indexed: 12/25/2022]
Abstract
OBJECTIVE This study was conducted to determine the expression pattern, regulation and function of CCL28 and CCR10 in rheumatoid arthritis (RA) pathogenesis. METHODS Expression of CCL28 and CCR10 was assessed in RA compared with other arthritis synovial tissues (STs) or fluids (SFs) by histology or ELISA. The factors modulating CCL28 and CCR10 expression were identified in RA myeloid and endothelial cells by ELISA, FACS and Western blotting. The mechanism by which CCL28 ligation promotes RA angiogenesis was examined in control and CCR10-knockdown endothelial cell chemotaxis and capillary formation. RESULTS CCL28 and/or CCR10 expression levels were accentuated in STs and SFs of patients with joint disease compared with normal controls and they were predominately coexpressed in RA myeloid and endothelial cells. We show that protein expression of CCL28 and CCR10 was modulated by tumour necrosis factor (TNF)-α and toll-like receptor 4 ligation in RA monocytes and endothelial cells and by interleukin (IL)-6 stimulation in RA macrophages. Neutralisation of CCL28 in RA SF or blockade of CCR10 on human endothelial progenitor cells (EPCs) significantly reduced SF-induced endothelial migration and capillary formation, demonstrating that ligation of joint CCL28 to endothelial CCR10+ cells is involved in RA angiogenesis. We discovered that angiogenesis driven by ligation of CCL28 to CCR10 is linked to the extracellular signal regulated kinase (ERK) cascade, as CCR10-knockdown cells exhibit dysfunctional CCL28-induced ERK signalling, chemotaxis and capillary formation. CONCLUSIONS The overexpression of CCL28 and CCR10 in RA ST and their contribution to EPC migration into RA joints support the CCL28/CCR10 cascade as a potential therapeutic target for RA.
Collapse
MESH Headings
- Adult
- Aged
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/immunology
- Cells, Cultured
- Chemokines, CC/biosynthesis
- Chemokines, CC/genetics
- Chemokines, CC/immunology
- Chemotaxis/immunology
- Endothelial Cells/immunology
- Endothelium, Vascular/immunology
- Female
- Gene Expression Regulation/immunology
- Gene Silencing
- Humans
- Joints/blood supply
- MAP Kinase Signaling System/immunology
- MAP Kinase Signaling System/physiology
- Macrophages/immunology
- Male
- Middle Aged
- Monocytes/immunology
- Neovascularization, Pathologic/immunology
- Osteoarthritis/genetics
- Osteoarthritis/immunology
- RNA, Messenger/genetics
- Receptors, CCR10/biosynthesis
- Receptors, CCR10/deficiency
- Receptors, CCR10/genetics
- Receptors, CCR10/immunology
- Signal Transduction/immunology
- Synovial Membrane/immunology
Collapse
Affiliation(s)
- Zhenlong Chen
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612
| | - Seung-jae Kim
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612
| | - Abdul B Essani
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612
| | - Michael V. Volin
- Department of Microbiology & Immunology, Midwestern University, Chicago College of Osteopathic Medicine, Downers Grove, IL 60515
| | - Olga M Vila
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612
| | - William Swedler
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612
| | - Shiva Arami
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612
| | - Suncica Volkov
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612
| | - Latriese V. Sardin
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612
| | - Nadera Sweiss
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612
| | - Shiva Shahrara
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612
| |
Collapse
|
31
|
Pallister KB, Mason S, Nygaard TK, Liu B, Griffith S, Jones J, Linderman S, Hughes M, Erickson D, Voyich JM, Davis MF, Wilson E. Bovine CCL28 Mediates Chemotaxis via CCR10 and Demonstrates Direct Antimicrobial Activity against Mastitis Causing Bacteria. PLoS One 2015; 10:e0138084. [PMID: 26359669 PMCID: PMC4567263 DOI: 10.1371/journal.pone.0138084] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 08/26/2015] [Indexed: 11/19/2022] Open
Abstract
In addition to the well characterized function of chemokines in mediating the homing and accumulation of leukocytes to tissues, some chemokines also exhibit potent antimicrobial activity. Little is known of the potential role of chemokines in bovine mammary gland health and disease. The chemokine CCL28 has previously been shown to play a key role in the homing and accumulation of IgA antibody secreting cells to the lactating murine mammary gland. CCL28 has also been shown to act as an antimicrobial peptide with activity demonstrated against a wide range of pathogens including bacteria, fungi and protozoans. Here we describe the cloning and function of bovine CCL28 and document the concentration of this chemokine in bovine milk. Bovine CCL28 was shown to mediate cellular chemotaxis via the CCR10 chemokine receptor and exhibited antimicrobial activity against a variety of bovine mastitis causing organisms. The concentration of bovine CCL28 in milk was found to be highly correlated with the lactation cycle. Highest concentrations of CCL28 were observed soon after parturition, with levels decreasing over time. These results suggest a potential role for CCL28 in the prevention/resolution of bovine mastitis.
Collapse
Affiliation(s)
- Kyler B. Pallister
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana, United States of America
| | - Sara Mason
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, Utah, United States of America
| | - Tyler K. Nygaard
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana, United States of America
| | - Bin Liu
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, Utah, United States of America
| | - Shannon Griffith
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana, United States of America
| | - Jennifer Jones
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana, United States of America
| | - Susanne Linderman
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, Utah, United States of America
| | - Melissa Hughes
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, Utah, United States of America
| | - David Erickson
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, Utah, United States of America
| | - Jovanka M. Voyich
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana, United States of America
| | - Mary F. Davis
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, Utah, United States of America
| | - Eric Wilson
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, Utah, United States of America
- * E-mail:
| |
Collapse
|
32
|
Xiong N, Hu S. Regulation of intestinal IgA responses. Cell Mol Life Sci 2015; 72:2645-55. [PMID: 25837997 DOI: 10.1007/s00018-015-1892-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 03/18/2015] [Accepted: 03/20/2015] [Indexed: 12/20/2022]
Abstract
The intestine harbors enormous numbers of commensal bacteria and is under frequent attack from food-borne pathogens and toxins. A properly regulated immune response is critical for homeostatic maintenance of commensals and for protection against infection and toxins in the intestine. Immunoglobulin A (IgA) isotype antibodies function specifically in mucosal sites such as the intestines to help maintain intestinal health by binding to and regulating commensal microbiota, pathogens and toxins. IgA antibodies are produced by intestinal IgA antibody-secreting plasma cells generated in gut-associated lymphoid tissues from naïve B cells in response to stimulations of the intestinal bacteria and components. Research on generation, migration, and maintenance of IgA-secreting cells is important in our effort to understand the biology of IgA responses and to help better design vaccines against intestinal infections.
Collapse
Affiliation(s)
- Na Xiong
- Department of Veterinary and Biomedical Sciences, Centre for Molecular Immunology and Infectious Disease, The Pennsylvania State University, 115 Henning Building, University Park, PA, 16802, USA,
| | | |
Collapse
|
33
|
Fagarasan S, Macpherson AJ. The Regulation of IgA Production. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00023-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
34
|
Abstract
The intestine represents the largest compartment of the immune system. It is continually exposed to antigens and immunomodulatory agents from the diet and the commensal microbiota, and it is the port of entry for many clinically important pathogens. Intestinal immune processes are also increasingly implicated in controlling disease development elsewhere in the body. In this Review, we detail the anatomical and physiological distinctions that are observed in the small and large intestines, and we suggest how these may account for the diversity in the immune apparatus that is seen throughout the intestine. We describe how the distribution of innate, adaptive and innate-like immune cells varies in different segments of the intestine and discuss the environmental factors that may influence this. Finally, we consider the implications of regional immune specialization for inflammatory disease in the intestine.
Collapse
|
35
|
Meier D, Docena GH, Ramisch D, Toscanini U, Berardi G, Gondolesi GE, Rumbo M. Immunological status of isolated lymphoid follicles after intestinal transplantation. Am J Transplant 2014; 14:2148-58. [PMID: 25088845 DOI: 10.1111/ajt.12797] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 03/20/2014] [Accepted: 04/17/2014] [Indexed: 01/25/2023]
Abstract
Intestinal transplantation (ITx) faces the challenge of grafting a high immunogenic organ, which is certainly one of the major obstacles for intestinal allograft acceptance. The allograft has to guarantee the proper functioning of the mucosal immune machinery under immunosuppressive conditions. Recently, it has been elucidated that isolated lymphoid follicles (ILFs) are an indispensable part of mucosal immunity to maintain IgA synthesis and consequently to control commensal microflora. No data about these follicular structures in the setting of ITx are available so far. Therefore, we addressed the question whether constitution, integrity and function of allograft ILFs are disturbed by immunosuppressive regimen. We compared allograft ILFs from terminal ileum of transplant patients with ILFs from nontransplant patients via flow cytometry, quantitative real-time polymerase chain reaction and immunohistochemistry. We found that host leukocytes rapidly repopulate allograft ILFs and that maintenance immunosuppressive regimen, tacrolimus and corticosteroids, does not affect their cellular integrity and function. However, allograft ILFs revealed a higher maturation state than control samples and IgA positive plasma cells were increased in number in allograft mucosa. Our results open the path for a better understanding of allograft mucosal immunity.
Collapse
Affiliation(s)
- D Meier
- Laboratory of Translational Research and Transplant Immunology, Multiorgan Transplantation Institute, Favaloro University, Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
36
|
Xia M, Hu S, Fu Y, Jin W, Yi Q, Matsui Y, Yang J, McDowell MA, Sarkar S, Kalia V, Xiong N. CCR10 regulates balanced maintenance and function of resident regulatory and effector T cells to promote immune homeostasis in the skin. J Allergy Clin Immunol 2014; 134:634-644.e10. [PMID: 24767879 DOI: 10.1016/j.jaci.2014.03.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 03/16/2014] [Accepted: 03/19/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND CCR10 and CCL27 make up the most skin-specific chemokine receptor/ligand pair implicated in skin allergy and inflammatory diseases, including atopic dermatitis and psoriasis. This pair is thought to regulate the migration, maintenance, or both of skin T cells and is suggested to be therapeutic targets for treatment of skin diseases. However, the functional importance of CCR10/CCL27 in vivo remains elusive. OBJECTIVE We sought to determine the expression and function of CCR10 in different subsets of skin T cells under both homeostatic and inflammatory conditions to gain a mechanistic insight into the potential roles of CCR10 during skin inflammation. METHODS Using heterozygous and homozygous CCR10 knockout/enhanced green fluorescent protein knockin mice, we assessed the expression of CCR10 on regulatory and effector T cells of healthy and inflamed skin induced by chemicals, pathogens, and autoreactive T cells. In addition, we assessed the effect of CCR10 knockout on the maintenance and functions of different T cells and inflammatory status in the skin during different phases of the immune response. RESULTS CCR10 expression is preferentially induced on memory-like skin-resident T cells and their progenitors for their maintenance in homeostatic skin but not expressed on most skin-infiltrating effector T cells during inflammation. In CCR10 knockout mice the imbalanced presence and dysregulated function of resident regulatory and effector T cells result in over-reactive and prolonged innate and memory responses in the skin, leading to increased clearance of Leishmania species infection in the skin. CONCLUSION CCR10 is a critical regulator of skin immune homeostasis.
Collapse
Affiliation(s)
- Mingcan Xia
- Centre for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pa
| | - Shaomin Hu
- Centre for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pa
| | - Yaoyao Fu
- Centre for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pa
| | - Wensen Jin
- Teaching and Research Section of Nuclear Medicine, Anhui Medical University, Anhui, China
| | - Qiyi Yi
- Teaching and Research Section of Nuclear Medicine, Anhui Medical University, Anhui, China
| | - Yurika Matsui
- Cell and Development Biology Graduate Program, Pennsylvania State University, University Park, Pa
| | - Jie Yang
- Centre for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pa
| | - Mary Ann McDowell
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, Ind
| | - Surojit Sarkar
- Centre for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pa
| | - Vandana Kalia
- Centre for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pa
| | - Na Xiong
- Centre for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pa.
| |
Collapse
|
37
|
Masahata K, Umemoto E, Kayama H, Kotani M, Nakamura S, Kurakawa T, Kikuta J, Gotoh K, Motooka D, Sato S, Higuchi T, Baba Y, Kurosaki T, Kinoshita M, Shimada Y, Kimura T, Okumura R, Takeda A, Tajima M, Yoshie O, Fukuzawa M, Kiyono H, Fagarasan S, Iida T, Ishii M, Takeda K. Generation of colonic IgA-secreting cells in the caecal patch. Nat Commun 2014; 5:3704. [PMID: 24718324 DOI: 10.1038/ncomms4704] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 03/21/2014] [Indexed: 01/22/2023] Open
Abstract
Gut-associated lymphoid tissues are responsible for the generation of IgA-secreting cells. However, the function of the caecal patch, a lymphoid tissue in the appendix, remains unknown. Here we analyse the role of the caecal patch using germ-free mice colonized with intestinal bacteria after appendectomy. Appendectomized mice show delayed accumulation of IgA(+) cells in the large intestine, but not the small intestine, after colonization. Decreased colonic IgA(+) cells correlate with altered faecal microbiota composition. Experiments using photoconvertible Kaede-expressing mice or adoptive transfer show that the caecal patch IgA(+) cells migrate to the large and small intestines, whereas Peyer's patch cells are preferentially recruited to the small intestine. IgA(+) cells in the caecal patch express higher levels of CCR10. Dendritic cells in the caecal patch, but not Peyer's patches, induce CCR10 on cocultured B cells. Thus, the caecal patch is a major site for generation of IgA-secreting cells that migrate to the large intestine.
Collapse
Affiliation(s)
- Kazunori Masahata
- 1] Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan [2] Department of Pediatric Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Eiji Umemoto
- 1] Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan [2] Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Hisako Kayama
- 1] Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan [2] Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Manato Kotani
- Department of Immunology and Cell Biology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Shota Nakamura
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Takashi Kurakawa
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Junichi Kikuta
- Department of Immunology and Cell Biology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kazuyoshi Gotoh
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Daisuke Motooka
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Shintaro Sato
- Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Tomonori Higuchi
- Department of Microbiology, Kinki University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Yoshihiro Baba
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Makoto Kinoshita
- 1] Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan [2] Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Yosuke Shimada
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Taishi Kimura
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Ryu Okumura
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Akira Takeda
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masaru Tajima
- The Institute of Experimental Animal Sciences, Faculty of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Osamu Yoshie
- Department of Microbiology, Kinki University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Masahiro Fukuzawa
- Department of Pediatric Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hiroshi Kiyono
- 1] Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan [2] Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Sidonia Fagarasan
- Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences, RIKEN, Yokohama 230-0045, Japan
| | - Tetsuya Iida
- 1] Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan [2] Laboratory of Genomic Research on Pathogenic Bacteria, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masaru Ishii
- 1] Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan [2] Department of Immunology and Cell Biology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kiyoshi Takeda
- 1] Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan [2] Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan
| |
Collapse
|
38
|
Rajagopal S, Bassoni DL, Campbell JJ, Gerard NP, Gerard C, Wehrman TS. Biased agonism as a mechanism for differential signaling by chemokine receptors. J Biol Chem 2013; 288:35039-48. [PMID: 24145037 DOI: 10.1074/jbc.m113.479113] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Chemokines display considerable promiscuity with multiple ligands and receptors shared in common, a phenomenon that is thought to underlie their biochemical "redundancy." Their receptors are part of a larger seven-transmembrane receptor superfamily, commonly referred to as G protein-coupled receptors, which have been demonstrated to be able to signal with different efficacies to their multiple downstream signaling pathways, a phenomenon referred to as biased agonism. Biased agonism has been primarily reported as a phenomenon of synthetic ligands, and the biologic prevalence and importance of such signaling are unclear. Here, to assess the presence of biased agonism that may underlie differential signaling by chemokines targeting the same receptor, we performed a detailed pharmacologic analysis of a set of chemokine receptors with multiple endogenous ligands using assays for G protein signaling, β-arrestin recruitment, and receptor internalization. We found that chemokines targeting the same receptor can display marked differences in their efficacies for G protein- or β-arrestin-mediated signaling or receptor internalization. This ligand bias correlates with changes in leukocyte migration, consistent with different mechanisms underlying the signaling downstream of these receptors induced by their ligands. These findings demonstrate that biased agonism is a common and likely evolutionarily conserved biological mechanism for generating qualitatively distinct patterns of signaling via the same receptor in response to different endogenous ligands.
Collapse
Affiliation(s)
- Sudarshan Rajagopal
- From the Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710
| | | | | | | | | | | |
Collapse
|
39
|
Hwang IY, Park C, Luong T, Harrison KA, Birnbaumer L, Kehrl JH. The loss of Gnai2 and Gnai3 in B cells eliminates B lymphocyte compartments and leads to a hyper-IgM like syndrome. PLoS One 2013; 8:e72596. [PMID: 23977324 PMCID: PMC3747273 DOI: 10.1371/journal.pone.0072596] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 07/18/2013] [Indexed: 11/22/2022] Open
Abstract
B lymphocytes are compartmentalized within lymphoid organs. The organization of these compartments depends upon signaling initiated by G-protein linked chemoattractant receptors. To address the importance of the G-proteins Gαi2 and Gαi3 in chemoattractant signaling we created mice lacking both proteins in their B lymphocytes. While bone marrow B cell development and egress is grossly intact; mucosal sites, splenic marginal zones, and lymph nodes essentially lack B cells. There is a partial block in splenic follicular B cell development and a 50-60% reduction in splenic B cells, yet normal numbers of splenic T cells. The absence of Gαi2 and Gαi3 in B cells profoundly disturbs the architecture of lymphoid organs with loss of B cell compartments in the spleen, thymus, lymph nodes, and gastrointestinal tract. This results in a severe disruption of B cell function and a hyper-IgM like syndrome. Beyond the pro-B cell stage, B cells are refractory to chemokine stimulation, and splenic B cells are poorly responsive to antigen receptor engagement. Gαi2 and Gαi3 are therefore critical for B cell chemoattractant receptor signaling and for normal B cell function. These mice provide a worst case scenario of the consequences of losing chemoattractant receptor signaling in B cells.
Collapse
Affiliation(s)
- Il-Young Hwang
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Chung Park
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Thuyvi Luong
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kathleen A. Harrison
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lutz Birnbaumer
- Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health/Department of Health and Human Services, Durham, North Carolina, United States of America
| | - John H. Kehrl
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
40
|
Agnello D, Denimal D, Lavaux A, Blondeau-Germe L, Lu B, Gerard NP, Gerard C, Pothier P. Intrarectal immunization and IgA antibody-secreting cell homing to the small intestine. THE JOURNAL OF IMMUNOLOGY 2013; 190:4836-47. [PMID: 23547118 DOI: 10.4049/jimmunol.1202979] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
According to the current paradigm, lymphocyte homing to the small intestine requires the expression of two tissue-specific homing receptors, the integrin α4β7 and the CCL25 receptor CCR9. In this study, we investigated the organ distribution and the homing molecule expression of IgA Ab-secreting cells (ASCs) induced by intrarectal immunization with a particulate Ag, in comparison with other mucosal immunization routes. Intrarectal immunization induces gut-homing IgA ASCs that localize not only in the colon but also in the small intestine, although they are not responsive to CCL25, unlike IgA ASCs induced by oral immunization. The mucosal epithelial chemokine CCL28, known to attract all IgA ASCs, does not compensate for the lack of CCL25 responsiveness, because the number of Ag-specific cells is not decreased in the gut of CCR10-deficient mice immunized by the intrarectal route. However, Ag-specific IgA ASCs induced by intrarectal immunization express the integrin α4β7, and their number is considerably decreased in the gut of β7-deficient mice immunized by the intrarectal route, indicating that α4β7 enables these cells to migrate into the small intestine, even without CCL25 responsiveness. In contrast, IgA ASCs induced by intranasal immunization express low α4β7 levels and are usually excluded from the gut. Paradoxically, after intranasal immunization, Ag-specific IgA ASCs are significantly increased in the small intestine of β7-deficient mice, demonstrating that lymphocyte homing is a competitive process and that integrin α4β7 determines not only the intestinal tropism of IgA ASCs elicited in GALTs but also the intestinal exclusion of lymphocytes primed in other inductive sites.
Collapse
Affiliation(s)
- Davide Agnello
- Laboratoire de Virologie et Centre National de Référence des Virus Entériques, Centre Hospitalier Universitaire de Dijon, 21070 Dijon Cedex, France.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
The intestinal mucosa contains the largest population of antibody-secreting plasma cells in the body, and in humans several grams of secretory immunoglobulin A (SIgA) are released into the intestine each day. In the gut lumen, SIgA serves as a first-line barrier that protects the epithelium from pathogens and toxins. Recently, next-generation sequencing has revolutionized our understanding of the nature of the intestinal microbiota and has also shed new light on the important roles of SIgA in the regulation of host-commensal homeostasis. Here, I discuss pathways of IgA induction in the context of SIgA specificity and function.
Collapse
Affiliation(s)
- Oliver Pabst
- Institute of Immunology, Hannover Medical School, Carl-Neuberg Strae 1, 30625 Hannover, Germany.
| |
Collapse
|
42
|
CCR10 and its ligands in regulation of epithelial immunity and diseases. Protein Cell 2012; 3:571-80. [PMID: 22684736 DOI: 10.1007/s13238-012-2927-3] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 04/11/2012] [Indexed: 01/13/2023] Open
Abstract
Epithelial tissues covering the external and internal surface of a body are constantly under physical, chemical or biological assaults. To protect the epithelial tissues and maintain their homeostasis, multiple layers of immune defense mechanisms are required. Besides the epithelial tissue-resident immune cells that provide the first line of defense, circulating immune cells are also recruited into the local tissues in response to challenges. Chemokines and chemokine receptors regulate tissue-specific migration, maintenance and functions of immune cells. Among them, chemokine receptor CCR10 and its ligands chemokines CCL27 and CCL28 are uniquely involved in the epithelial immunity. CCL27 is expressed predominantly in the skin by keratinocytes while CCL28 is expressed by epithelial cells of various mucosal tissues. CCR10 is expressed by various subsets of innate-like T cells that are programmed to localize to the skin during their developmental processes in the thymus. Circulating T cells might be imprinted by skin-associated antigen- presenting cells to express CCR10 for their recruitment to the skin during the local immune response. On the other hand, IgA antibody-producing B cells generated in mucosa-associated lymphoid tissues express CCR10 for their migration and maintenance at mucosal sites. Increasing evidence also found that CCR10/ligands are involved in regulation of other immune cells in epithelial immunity and are frequently exploited by epithelium-localizing or -originated cancer cells for their survival, proliferation and evasion from immune surveillance. Herein, we review current knowledge on roles of CCR10/ligands in regulation of epithelial immunity and diseases and speculate on related important questions worth further investigation.
Collapse
|
43
|
Knoop KA, Newberry RD. Isolated Lymphoid Follicles are Dynamic Reservoirs for the Induction of Intestinal IgA. Front Immunol 2012; 3:84. [PMID: 22566964 PMCID: PMC3343265 DOI: 10.3389/fimmu.2012.00084] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 04/03/2012] [Indexed: 12/12/2022] Open
Abstract
IgA is one of the most important molecules in the regulation of intestinal homeostasis. Peyer's patches have been traditionally recognized as sites for the induction of intestinal IgA responses, however more recent studies demonstrate that isolated lymphoid follicles (ILFs) can perform this function as well. ILF development is dynamic, changing in response to the luminal microbial burden, suggesting that ILFs play an important role providing an expandable reservoir of compensatory IgA inductive sites. However, in situations of immune dysfunction, ILFs can over-develop in response to uncontrollable enteric flora, resulting in ILF hyperplasia. The ability of ILFs to expand and respond to help control the enteric flora makes this dynamic reservoir an important arm of IgA inductive sites in intestinal immunity.
Collapse
Affiliation(s)
- Kathryn A Knoop
- Department of Internal Medicine, Washington University School of Medicine St. Louis, MO, USA
| | | |
Collapse
|
44
|
IgA synthesis: a form of functional immune adaptation extending beyond gut. Curr Opin Immunol 2012; 24:261-8. [PMID: 22503962 DOI: 10.1016/j.coi.2012.03.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 03/22/2012] [Accepted: 03/23/2012] [Indexed: 12/12/2022]
Abstract
Immunoglobulin A (IgA) is the most abundantly produced antibody isotype in mammals. The primary function of IgA is to maintain homeostasis at mucosal surfaces. IgA is generated in specialized gut associated lymphoid tissues (GALT) by T cell-dependent and T cell-independent mechanisms. Studies in mice have demonstrated that IgA diversification has an essential role in the regulation of gut microbiota. Aberrant bacterial growth, by activating innate and adaptive immune cells, has emerged as a risk factor for inflammatory diseases such as metabolic disorders and autoimmune diseases. Dynamic diversification of IgA shields bacterial antigens preventing inflammatory responses, but when IgA regulation is suboptimal aberrant bacterial growth and inflammation can ensue.
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW This article reviews the most recent publications on innate immunity in the small intestine. We will go over the innate immune receptors that act as sensors of microbial presence or cell injury, Paneth cells as the main epithelial cell type that secrete antimicrobial peptides, and mucosal production of immunoglobulin A (IgA). In addition, we will give an update on examples of imbalance of the innate immune response resulting in clinical disease with the most relevant example being Crohn's disease. RECENT FINDINGS Toll-like receptors (TLRs) are involved in B-cell homing to the intestine, rejection of small intestinal allografts, and recruitment of mast cells. The TLR adaptor Toll/interleukin-1 receptor domain-containing adapter-inducing interferon-β is necessary to activate innate immunity after Yersinia enterocolitica infection. Moreover, MyD88 is required to keep the intestinal microbiota under control and physically separated from the epithelium, and RegIIIγ is responsible for the bacterial segregation from the lining epithelial cells. In Crohn's disease, ATG16L1 T300A variant promotes a proinflammatory response; and miR-196 downregulates a protective immunity-related GTPase family M protein (IRGM) polymorphism leading to impaired clearance of adherent Escherichia coli in the intestine. SUMMARY The intestine is continuously exposed to dietary and microbial antigens. The host has to maintain intestinal homeostasis to keep the commensal and pathogenic bacteria under control. Some of the mechanisms to do so are by expression of innate immune receptors, production of antimicrobial peptides, secretion of IgA, or autophagy of intracellular bacteria. Unfortunately, in some cases the innate immune response fails to protect the host and chronic inflammation, transplant rejection, or other disorders may occur.
Collapse
|
46
|
Bemark M, Boysen P, Lycke NY. Induction of gut IgA production through T cell-dependent and T cell-independent pathways. Ann N Y Acad Sci 2012; 1247:97-116. [PMID: 22260403 DOI: 10.1111/j.1749-6632.2011.06378.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The gut immune system protects against mucosal pathogens, maintains a mutualistic relationship with the microbiota, and establishes tolerance against food antigens. This requires a balance between immune effector responses and induction of tolerance. Disturbances of this strictly regulated balance can lead to infections or the development inflammatory diseases and allergies. Production of secretory IgA is a unique effector function at mucosal surfaces, and basal mechanisms regulating IgA production have been the focus of much recent research. These investigations have aimed at understanding how long-term IgA-mediated mucosal immunity can best be achieved by oral or sublingual vaccination, or at analyzing the relationship between IgA production, the composition of the gut microbiota, and protection from allergies and autoimmunity. This research has lead to a better understanding of the IgA system; but at the same time seemingly conflicting data have been generated. Here, we discuss how gut IgA production is controlled, with special focus on how differences between T cell-dependent and T cell-independent IgA production may explain some of these discrepancies.
Collapse
Affiliation(s)
- Mats Bemark
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden.
| | | | | |
Collapse
|