51
|
Kelly A, Houston SA, Sherwood E, Casulli J, Travis MA. Regulation of Innate and Adaptive Immunity by TGFβ. Adv Immunol 2017; 134:137-233. [PMID: 28413021 DOI: 10.1016/bs.ai.2017.01.001] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Immune regulation by cytokines is crucial in maintaining immune homeostasis, promoting responses to infection, resolving inflammation, and promoting immunological memory. Additionally, cytokine responses drive pathology in immune-mediated disease. A crucial cytokine in the regulation of all aspects of an immune response is transforming growth factor beta (TGFβ). Although best known as a crucial regulator of T cell responses, TGFβ plays a vital role in regulating responses mediated by virtually every innate and adaptive immune cell, including dendritic cells, B cells, NK cells, innate lymphoid cells, and granulocytes. Here, we review our current knowledge of how TGFβ regulates the immune system, highlighting the multifunctional nature of TGFβ and how its function can change depending on location and context of action.
Collapse
Affiliation(s)
- Aoife Kelly
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, United Kingdom; Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom; Manchester Immunology Group, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Stephanie A Houston
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, United Kingdom; Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom; Manchester Immunology Group, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Eleanor Sherwood
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, United Kingdom; Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom; Manchester Immunology Group, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Joshua Casulli
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, United Kingdom; Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom; Manchester Immunology Group, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Mark A Travis
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, United Kingdom; Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom; Manchester Immunology Group, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
52
|
Nagata Y, Yamamoto T, Hayashi M, Hayashi S, Kadowaki M. Improvement of Therapeutic Efficacy of Oral Immunotherapy in Combination with Regulatory T Cell-Inducer Kakkonto in a Murine Food Allergy Model. PLoS One 2017; 12:e0170577. [PMID: 28107533 PMCID: PMC5249179 DOI: 10.1371/journal.pone.0170577] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/08/2017] [Indexed: 12/14/2022] Open
Abstract
Oral immunotherapy (OIT) has been considered a promising approach for food allergies (FAs). However, the current OIT strategy is limited in terms of the long-term efficacy and safety. We have previously demonstrated that kakkonto, a traditional Japanese herbal medicine, suppresses the occurrence of allergic symptoms in a murine model of ovalbumin (OVA)-induced FA, which is attributed to the induction of the Foxp3+ CD4+ regulatory T cells. In this study, we established an OIT model using the FA mice with already established allergic symptoms and determined whether kakkonto could improve the efficacy of OIT. The OIT method consisted of initially administrating a very small amount of OVA and slowly increasing the amount. Allergic symptoms decreased in the OIT-treated FA mice. OIT significantly downregulated Th2 immune response-related gene expression in the FA mouse colon, and decreased the level of mouse mast cell protease-1, a marker of mast cell degranulation in the FA mouse plasma. Moreover, the concomitant use of kakkonto significantly enhanced the effectiveness of OIT on the allergic symptoms, and the combination therapy further suppressed the Th2 immune responses and the mast cell degranulation. In addition, OIT significantly increased the population of Foxp3+ CD4+ regulatory T cells in the FA mouse colon, and this population was further increased by OIT in combination with kakkonto. Furthermore, the combined therapy with kakkonto reduced the expression of RA-degrading enzyme CYP26B1 mRNA in the FA mouse colon. These findings indicated that the combination of OIT with kakkonto represents a promising approach for FA treatment.
Collapse
Affiliation(s)
- Yuka Nagata
- Division of Gastrointestinal Pathophysiology, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Takeshi Yamamoto
- Division of Gastrointestinal Pathophysiology, Institute of Natural Medicine, University of Toyama, Toyama, Japan
- * E-mail:
| | - Michie Hayashi
- Division of Gastrointestinal Pathophysiology, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Shusaku Hayashi
- Division of Gastrointestinal Pathophysiology, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Makoto Kadowaki
- Division of Gastrointestinal Pathophysiology, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| |
Collapse
|
53
|
Martín-Orozco E, Norte-Muñoz M, Martínez-García J. Regulatory T Cells in Allergy and Asthma. Front Pediatr 2017; 5:117. [PMID: 28589115 PMCID: PMC5440567 DOI: 10.3389/fped.2017.00117] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 05/03/2017] [Indexed: 12/12/2022] Open
Abstract
The immune system's correct functioning requires a sophisticated balance between responses to continuous microbial challenges and tolerance to harmless antigens, such as self-antigens, food antigens, commensal microbes, allergens, etc. When this equilibrium is altered, it can lead to inflammatory pathologies, tumor growth, autoimmune disorders, and allergy/asthma. The objective of this review is to show the existing data on the importance of regulatory T cells (Tregs) on this balance and to underline how intrauterine and postnatal environmental exposures influence the maturation of the immune system in humans. Genetic and environmental factors during embryo development and/or early life will result in a proper or, conversely, inadequate immune maturation with either beneficial or deleterious effects on health. We have focused herein on Tregs as a reflection of the maturity of the immune system. We explain the types, origins, and the mechanisms of action of these cells, discussing their role in allergy and asthma predisposition. Understanding the importance of Tregs in counteracting dysregulated immunity would provide approaches to diminish asthma and other related diseases in infants.
Collapse
Affiliation(s)
- Elena Martín-Orozco
- Department of Biochemistry and Molecular Biology B and Immunology, School of Medicine, Murcia Biohealth Research Institute-University of Murcia (IMIB-UMU), Regional Campus of International Excellence "Campus Mare Nostrum", Murcia, Spain
| | - María Norte-Muñoz
- Department of Biochemistry and Molecular Biology B and Immunology, School of Medicine, Murcia Biohealth Research Institute-University of Murcia (IMIB-UMU), Regional Campus of International Excellence "Campus Mare Nostrum", Murcia, Spain
| | - Javier Martínez-García
- Department of Biochemistry and Molecular Biology B and Immunology, School of Medicine, Murcia Biohealth Research Institute-University of Murcia (IMIB-UMU), Regional Campus of International Excellence "Campus Mare Nostrum", Murcia, Spain
| |
Collapse
|
54
|
Epicutaneous immunotherapy induces gastrointestinal LAP + regulatory T cells and prevents food-induced anaphylaxis. J Allergy Clin Immunol 2016; 139:189-201.e4. [PMID: 27417020 DOI: 10.1016/j.jaci.2016.03.057] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 02/09/2016] [Accepted: 03/22/2016] [Indexed: 11/23/2022]
Abstract
BACKGROUND The attempt to induce oral tolerance as a treatment for food allergy has been hampered by a lack of sustained clinical protection. Immunotherapy by nonoral routes, such as the skin, may be more effective for the development of maintained tolerance to food allergens. OBJECTIVE We sought to determine the efficacy and mechanism of tolerance induced by epicutaneous immunotherapy (EPIT) in a model of food-induced anaphylaxis. METHODS C3H/HeJ mice were sensitized to ovalbumin (OVA) orally or through the skin and treated with EPIT using OVA-Viaskin patches or oral immunotherapy using OVA. Mice were orally challenged with OVA to induce anaphylaxis. Antigen-specific regulatory T (Treg)-cell induction was assessed by flow cytometry using a transgenic T-cell transfer model. RESULTS By using an adjuvant-free model of food allergy generated by epicutaneous sensitization and reactions triggered by oral allergen challenge, we found that EPIT induced sustained protection against anaphylaxis. We show that the gastrointestinal tract is deficient in de novo generation of Treg cells in allergic mice. This defect was tissue-specific, and epicutaneous application of antigen generated a population of gastrointestinal-homing LAP+Foxp3- Treg cells. The mechanism of protection was found to be a novel pathway of direct TGF-β-dependent Treg-cell suppression of mast cell activation, in the absence of modulation of T- or B-cell responses. CONCLUSIONS Our data highlight the immune communication between skin and gastrointestinal tract, and identifies novel mechanisms by which epicutaneous tolerance can suppress food-induced anaphylaxis.
Collapse
|
55
|
Chellappa S, Hugenschmidt H, Hagness M, Line PD, Labori KJ, Wiedswang G, Taskén K, Aandahl EM. Regulatory T cells that co-express RORγt and FOXP3 are pro-inflammatory and immunosuppressive and expand in human pancreatic cancer. Oncoimmunology 2016; 5:e1102828. [PMID: 27141387 PMCID: PMC4839385 DOI: 10.1080/2162402x.2015.1102828] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 09/21/2015] [Accepted: 09/26/2015] [Indexed: 12/20/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is highly infiltrated by CD4+T cells that express RORγt and IL-17 (TH17). Compelling evidence from the tumor microenvironment suggest that regulatory T cells (Treg) contribute to TH17 mediated inflammation. Concurrently, PDAC patients have elevated levels of pro-inflammatory cytokines that may lead to TH17 associated functional plasticity in Treg. In this study, we investigated the phenotype and functional properties of Treg in patients with PDAC. We report that PDAC patients have elevated frequency of FOXP3+Treg, which exclusively occurred within the FOXP3+RORγt+Treg compartment. The FOXP3+RORγt+Treg retained FOXP3+Treg markers and represented an activated subset. The expression of RORγt in Treg may indicate a phenotypic switch toward TH17 cells. However, the FOXP3+RORγt+Treg produced both TH17 and TH2 associated pro-inflammatory cytokines, which corresponded with elevated TH17 and TH2 immune responses in PDAC patients. Both the FOXP3+Treg and FOXP3+RORγt+Treg from PDAC patients strongly suppressed T cell immune responses, but they had impaired anti-inflammatory properties. We conclude that FOXP3+RORγt+Treg have a dual phenotype with combined pro-inflammatory and immunosuppressive activity, which may be involved in the pathogenesis of PDAC.
Collapse
Affiliation(s)
- Stalin Chellappa
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway
- Biotechnology Center, University of Oslo, Oslo, Norway
- K.G. Jebsen Inflammation Research Center, University of Oslo, Oslo, Norway
- K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, Oslo, Norway
| | - Harald Hugenschmidt
- Section for Transplantation Surgery, Oslo University Hospital, Oslo, Norway
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital, Oslo, Norway
| | - Morten Hagness
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway
- Biotechnology Center, University of Oslo, Oslo, Norway
- K.G. Jebsen Inflammation Research Center, University of Oslo, Oslo, Norway
- Section for Transplantation Surgery, Oslo University Hospital, Oslo, Norway
| | - Pål D. Line
- Section for Transplantation Surgery, Oslo University Hospital, Oslo, Norway
| | - Knut J. Labori
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital, Oslo, Norway
| | - Gro Wiedswang
- Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, Norway
| | - Kjetil Taskén
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway
- Biotechnology Center, University of Oslo, Oslo, Norway
- K.G. Jebsen Inflammation Research Center, University of Oslo, Oslo, Norway
- K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Einar M. Aandahl
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway
- Biotechnology Center, University of Oslo, Oslo, Norway
- K.G. Jebsen Inflammation Research Center, University of Oslo, Oslo, Norway
- Section for Transplantation Surgery, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
56
|
Slyepchenko A, Maes M, Köhler CA, Anderson G, Quevedo J, Alves GS, Berk M, Fernandes BS, Carvalho AF. T helper 17 cells may drive neuroprogression in major depressive disorder: Proposal of an integrative model. Neurosci Biobehav Rev 2016; 64:83-100. [PMID: 26898639 DOI: 10.1016/j.neubiorev.2016.02.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 01/04/2016] [Accepted: 02/02/2016] [Indexed: 02/07/2023]
Abstract
The exact pathophysiology of major depressive disorder (MDD) remains elusive. The monoamine theory, which hypothesizes that MDD emerges as a result of dysfunctional serotonergic, dopaminergic and noradrenergic pathways, has guided the therapy of this illness for several decades. More recently, the involvement of activated immune, oxidative and nitrosative stress pathways and of decreased levels of neurotrophic factors has provided emerging insights regarding the pathophysiology of MDD, leading to integrated theories emphasizing the complex interplay of these mechanisms that could lead to neuroprogression. In this review, we propose an integrative model suggesting that T helper 17 (Th17) cells play a pivotal role in the pathophysiology of MDD through (i) microglial activation, (ii) interactions with oxidative and nitrosative stress, (iii) increases of autoantibody production and the propensity for autoimmunity, (iv) disruption of the blood-brain barrier, and (v) dysregulation of the gut mucosa and microbiota. The clinical and research implications of this model are discussed.
Collapse
Affiliation(s)
- Anastasiya Slyepchenko
- Womens Health Concerns Clinic, St. Joseph's Healthcare Hamilton, MiNDS Program, McMaster University; Hamilton, Ontario, Canada
| | - Michael Maes
- IMPACT Strategic Research Centre, Deakin University, School of Medicine and Barwon Health, Geelong, VIC, Australia
| | - Cristiano A Köhler
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - João Quevedo
- Center for Translational Psychiatry, Department of Psychiatry and Behavioral Sciences, The University of Texas Medical School at Houston, Houston, TX, USA; Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Gilberto S Alves
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Michael Berk
- IMPACT Strategic Research Centre, Deakin University, School of Medicine and Barwon Health, Geelong, VIC, Australia; Department of Psychiatry, Florey Institute of Neuroscience and Mental Health, Orygen, The National Centre of Excellence in Youth Mental Health and Orygen Youth Health Research Centre, University of Melbourne, Parkville, VIC, Australia
| | - Brisa S Fernandes
- IMPACT Strategic Research Centre, Deakin University, School of Medicine and Barwon Health, Geelong, VIC, Australia; Laboratory of Calcium Binding Proteins in the Central Nervous System, Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - André F Carvalho
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil.
| |
Collapse
|
57
|
Betto E, Usuelli V, Mandelli A, Badami E, Sorini C, Capolla S, Danelli L, Frossi B, Guarnotta C, Ingrao S, Tripodo C, Pucillo C, Gri G, Falcone M. Mast cells contribute to autoimmune diabetes by releasing interleukin-6 and failing to acquire a tolerogenic IL-10 + phenotype. Clin Immunol 2015; 178:29-38. [PMID: 26732858 DOI: 10.1016/j.clim.2015.12.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/10/2015] [Accepted: 12/24/2015] [Indexed: 12/18/2022]
Abstract
Mast cells (MCs) are innate immune cells that exert positive and negative immune modulatory functions capable to enhance or limit the intensity and/or duration of adaptive immune responses. Although MCs are crucial to regulate T cell immunity, their action in the pathogenesis of autoimmune diseases is still debated. Here we demonstrate that MCs play a crucial role in T1D pathogenesis so that their selective depletion in conditional MC knockout NOD mice protects them from the disease. MCs of diabetic NOD mice are overly inflammatory and secrete large amounts of IL-6 that favors differentiation of IL-17-secreting T cells at the site of autoimmunity. Moreover, while MCs of control mice acquire an IL-10+ phenotype upon interaction with FoxP3+ Treg cells, MCs of NOD mice do not undergo this tolerogenic differentiation. Our data indicate that overly inflammatory MCs unable to acquire a tolerogenic IL-10+ phenotype contribute to the pathogenesis of autoimmune T1D.
Collapse
Affiliation(s)
- Elena Betto
- Department of Biomedical Science and Technology and M.A.T.I. Center of Excellence, University of Udine, Udine, Italy
| | - Vera Usuelli
- Experimental Diabetes Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandra Mandelli
- Experimental Diabetes Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Ester Badami
- Mediterranean Institute for Transplantation and Advanced Specialized Therapies, ISMETT, Palermo, Italy
| | - Chiara Sorini
- Experimental Diabetes Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Sara Capolla
- Department of Biomedical Science and Technology and M.A.T.I. Center of Excellence, University of Udine, Udine, Italy
| | - Luca Danelli
- Department of Biomedical Science and Technology and M.A.T.I. Center of Excellence, University of Udine, Udine, Italy
| | - Barbara Frossi
- Department of Biomedical Science and Technology and M.A.T.I. Center of Excellence, University of Udine, Udine, Italy
| | - Carla Guarnotta
- Department of Human Pathology, University of Palermo, Palermo, Italy
| | - Sabrina Ingrao
- Department of Human Pathology, University of Palermo, Palermo, Italy
| | - Claudio Tripodo
- Department of Human Pathology, University of Palermo, Palermo, Italy
| | - Carlo Pucillo
- Department of Biomedical Science and Technology and M.A.T.I. Center of Excellence, University of Udine, Udine, Italy.
| | - Giorgia Gri
- Department of Biomedical Science and Technology and M.A.T.I. Center of Excellence, University of Udine, Udine, Italy
| | - Marika Falcone
- Department of Biomedical Science and Technology and M.A.T.I. Center of Excellence, University of Udine, Udine, Italy.
| |
Collapse
|
58
|
Abstract
Food allergy is a growing public health problem that is estimated to affect 4% to 8% of children and 5% of adults. In this review, we discuss our current understanding of the pathophysiology of food allergy, from oral tolerance, to sensitization, and lastly the elicitation of an allergic response. As much of the existing evidence for the mechanisms of food allergy is derived from animal models, we include these studies where relevant. In addition, whenever possible, we review similar evidence involved in human disease and provide applications for consideration in clinical practice.
Collapse
|
59
|
Izadi N, Luu M, Ong PY, Tam JS. The Role of Skin Barrier in the Pathogenesis of Food Allergy. CHILDREN (BASEL, SWITZERLAND) 2015; 2:382-402. [PMID: 27417371 PMCID: PMC4928763 DOI: 10.3390/children2030382] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 08/25/2015] [Accepted: 08/25/2015] [Indexed: 02/08/2023]
Abstract
Food allergy is a serious public health problem with an increasing prevalence. Current management is limited to food avoidance and emergency treatment. Research into the pathogenesis of food allergy has helped to shape our understanding of how patients become sensitized to an allergen. Classically, food sensitization was thought to occur through the gastrointestinal tract, but alternative routes of sensitization are being explored, specifically through the skin. Damaged skin barrier may play a crucial role in the development of food sensitization. Better understanding of how patients initially become sensitized may help lead to the development of a safe and effective treatment for food allergies or better prevention strategies.
Collapse
Affiliation(s)
- Neema Izadi
- Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA.
| | - Minnelly Luu
- Department of Dermatology, Keck School of Medicine, University of Southern California.
| | - Peck Y Ong
- Division of Clinical Immunology and Allergy, Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California.
| | - Jonathan S Tam
- Division of Clinical Immunology and Allergy, Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California.
| |
Collapse
|
60
|
Chhiba KD, Singh AM, Bryce PJ. New developments in immunotherapies for food allergy. Immunotherapy 2015; 7:913-22. [DOI: 10.2217/imt.15.55] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Food allergy affects around 10% of the population. As the prevalence of food allergy continues to increase, disproportionately in children, new therapies for food allergy are being investigated. While there are no approved treatments for food allergy, immunotherapy facilitates significant desensitization and protection from accidental exposure. Nevertheless, current immunotherapies do not entirely nor permanently eliminate sensitivity to the food allergen. Since the rates of sustained unresponsiveness are significantly lower than desensitization, future therapies that enhance the rates of long-term tolerance in patients will catalyze progress in this field over the next 5–10 years.
Collapse
Affiliation(s)
- Krishan Dilip Chhiba
- Division of Allergy-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Anne Marie Singh
- Division of Allergy-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Paul J Bryce
- Division of Allergy-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
61
|
Järvinen KM, Suárez-Fariñas M, Savilahti E, Sampson HA, Berin MC. Immune factors in breast milk related to infant milk allergy are independent of maternal atopy. J Allergy Clin Immunol 2015; 135:1390-3.e1-6. [PMID: 25533649 PMCID: PMC4426237 DOI: 10.1016/j.jaci.2014.10.051] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 09/05/2014] [Accepted: 10/28/2014] [Indexed: 11/24/2022]
Affiliation(s)
- Kirsi M Järvinen
- Division of Allergy and Immunology and Center for Immunology and Microbial Diseases, Albany Medical College, Albany, NY; Division of Pediatric Allergy and Immunology and Jaffe Institute for Food Allergy, Icahn School of Medicine at Mt Sinai, New York, NY.
| | | | - Erkki Savilahti
- Division of Paediatric Gastroenterology and Clinical Immunology, Children's Hospital, University of Helsinki, Helsinki, Finland
| | - Hugh A Sampson
- Division of Pediatric Allergy and Immunology and Jaffe Institute for Food Allergy, Icahn School of Medicine at Mt Sinai, New York, NY
| | - M Cecilia Berin
- Division of Pediatric Allergy and Immunology and Jaffe Institute for Food Allergy, Icahn School of Medicine at Mt Sinai, New York, NY
| |
Collapse
|
62
|
|
63
|
Kim DH, Jung WS, Kim ME, Lee HW, Youn HY, Seon JK, Lee HN, Lee JS. Genistein inhibits pro‑inflammatory cytokines in human mast cell activation through the inhibition of the ERK pathway. Int J Mol Med 2014; 34:1669-74. [PMID: 25319548 DOI: 10.3892/ijmm.2014.1956] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 09/22/2014] [Indexed: 11/06/2022] Open
Abstract
Anaphylaxis is a rapidly occurring allergic reaction to any foreign substance, including venom from insects, foods and medications, which may cause fatalities. To prevent anaphylaxis, these triggers must be avoided. However, avoidance of numerous triggers is difficult. For this reason, the development of immunotherapeutic adjuvants that suppress the allergic response is important for anaphylaxis control. Mast cells are one of the major inflammatory cells involved in the inflammatory response, which secrete several inflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1β, and recruits other immune cells. Mast cells are also involved in a number of diseases, such as sinusitis, rheumatoid arthritis and asthma. Genistein, a phytoestrogen, has been reported to have anti-oxidative and anti-inflammatory activities. However, the effects of genistein on the anti-inflammatory response of mast cells remain unknown. In the present study, the anti-inflammatory effects of genistein on mast cells were investigated. Genistein significantly decreased IL-6 and IL-1β mRNA levels, as well as IL-6 production in PMA/A23187-induced mast cells activation. In addition, genistein inhibited the phosphorylation of ERK 1/2 in PMA/A23187-induced mast cell activation. However, phosphorylation of p38 was not altered. Thus, these findings indicate that genistein inhibited the inflammatory status of mast cells through inhibition of the ERK pathway.
Collapse
Affiliation(s)
- Dong Hwan Kim
- Department of Biology, Immunology Research Lab, BK21‑plus Research Team for Bioactive Control Technology, College of Natural Sciences, Chosun University, Gwangju 501‑759, Republic of Korea
| | - Woo-Sung Jung
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 151‑742, Republic of Korea
| | - Mi Eun Kim
- Department of Biology, Immunology Research Lab, BK21‑plus Research Team for Bioactive Control Technology, College of Natural Sciences, Chosun University, Gwangju 501‑759, Republic of Korea
| | - Hee-Woo Lee
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 151‑742, Republic of Korea
| | - Hwa-Young Youn
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 151‑742, Republic of Korea
| | - Jong Keun Seon
- Center for Joint Disease, Chonnam National University Hwasun Hospital, Hwasun‑eup, Hwasun‑gun, Jeollanam‑do 519‑809, Republic of Korea
| | - Haeng-Nam Lee
- Department of Mechanical Engineering, College of Engineering, Chosun University, Gwangju 501‑759, Republic of Korea
| | - Jun Sik Lee
- Department of Biology, Immunology Research Lab, BK21‑plus Research Team for Bioactive Control Technology, College of Natural Sciences, Chosun University, Gwangju 501‑759, Republic of Korea
| |
Collapse
|
64
|
Liu X, Jin H, Zhang G, Lin X, Chen C, Sun J, Zhang Y, Zhang Q, Yu J. Intratumor IL-17-positive mast cells are the major source of the IL-17 that is predictive of survival in gastric cancer patients. PLoS One 2014; 9:e106834. [PMID: 25197971 PMCID: PMC4157802 DOI: 10.1371/journal.pone.0106834] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 08/01/2014] [Indexed: 01/09/2023] Open
Abstract
Interleukin-17 (IL-17) is prevalent in tumor tissue and suppresses effective anti-tumor immune responses. However, the source of the increased tumor-infiltrating IL-17 and its contribution to tumor progression in human gastric cancer remain poorly understood. In this study, we enrolled 112 gastric cancer patients, immunofluorescence was used to evaluate the colocalization of CD3, CD4, CD56, CD20, CD68, and mast cell tryptase (MCT) with IL-17. Immunohistochemistry was used to evaluate the distribution of microvessel density (CD34), CD66b+, CD68+, and FoxP3+ cells in different microanatomical areas. Prognostic value was determined by Kaplan-Meier analysis and a Cox regression model. The results showed that mast cells, but not T cells or macrophages, were the predominant cell type producing IL-17 in gastric cancer. Significant positive correlations were detected between densities of mast cell-derived IL-17 and microvessels, neutrophils, and regulatory T cells (Tregs). Futhermore, we found that the majority of vascular endothelial cells expressing Interleukin-17 receptor (IL-17R). Kaplan-Meier analysis revealed that increasing intratumor infiltrated mast cells and IL-17+ cells, as well as MCT+ IL-17+ cells, were significantly associated with worse overall survival. These findings indicated that mast cells were the major source of IL-17 in gastric cancer, and intratumor IL-17 infiltration may have promoted tumor progression by enhancing angiogenesis in the tumor microenvironment through the axis of IL-17/IL-17R. IL-17-positive mast cells showed a prognostic factor in gastric cancer, indicating that immunotherapy targeting mast cells might be an effective strategy to control intratumor IL-17 infiltration, and consequently reverse immunosuppression in the tumor microenvironment, facilitating cancer immunotherapy.
Collapse
Affiliation(s)
- Xiaosun Liu
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Medical College, Zhejiang University, Hangzhou, China
| | - Hailong Jin
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Medical College, Zhejiang University, Hangzhou, China
| | - Geer Zhang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Medical College, Zhejiang University, Hangzhou, China
| | - Xianke Lin
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Medical College, Zhejiang University, Hangzhou, China
| | - Chao Chen
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Medical College, Zhejiang University, Hangzhou, China
| | - Jianyi Sun
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Medical College, Zhejiang University, Hangzhou, China
| | - Yu Zhang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Medical College, Zhejiang University, Hangzhou, China
| | - Qing Zhang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Medical College, Zhejiang University, Hangzhou, China
| | - Jiren Yu
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Medical College, Zhejiang University, Hangzhou, China
- * E-mail:
| |
Collapse
|
65
|
Johnston LK, Chien KB, Bryce PJ. The immunology of food allergy. THE JOURNAL OF IMMUNOLOGY 2014; 192:2529-34. [PMID: 24610821 DOI: 10.4049/jimmunol.1303026] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Food allergies represent an increasingly prevalent human health problem, and therapeutic options remain limited, with avoidance being mainstay, despite its adverse effects on quality of life. A better understanding of the key immunological mechanisms involved in such responses likely will be vital for development of new therapies. This review outlines the current understanding of how the immune system is thought to contribute to prevention or development of food allergies. Drawing from animal studies, as well as clinical data when available, the importance of oral tolerance in sustaining immunological nonresponsiveness to food Ags, our current understanding of why oral tolerance may fail and sensitization may occur, and the knowledge of pathways that may lead to anaphylaxis and food allergy-associated responses are addressed.
Collapse
Affiliation(s)
- Laura K Johnston
- Division of Allergy-Immunology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago IL 60611
| | | | | |
Collapse
|
66
|
Skaper SD, Facci L, Giusti P. Mast cells, glia and neuroinflammation: partners in crime? Immunology 2014; 141:314-27. [PMID: 24032675 DOI: 10.1111/imm.12170] [Citation(s) in RCA: 180] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 08/31/2013] [Accepted: 09/04/2013] [Indexed: 12/12/2022] Open
Abstract
Glia and microglia in particular elaborate pro-inflammatory molecules that play key roles in central nervous system (CNS) disorders from neuropathic pain and epilepsy to neurodegenerative diseases. Microglia respond also to pro-inflammatory signals released from other non-neuronal cells, mainly those of immune origin such as mast cells. The latter are found in most tissues, are CNS resident, and traverse the blood-spinal cord and blood-brain barriers when barrier compromise results from CNS pathology. Growing evidence of mast cell-glia communication opens new perspectives for the development of therapies targeting neuroinflammation by differentially modulating activation of non-neuronal cells that normally control neuronal sensitization - both peripherally and centrally. Mast cells and glia possess endogenous homeostatic mechanisms/molecules that can be up-regulated as a result of tissue damage or stimulation of inflammatory responses. Such molecules include the N-acylethanolamine family. One such member, N-palmitoylethanolamine is proposed to have a key role in maintenance of cellular homeostasis in the face of external stressors provoking, for example, inflammation. N-Palmitoylethanolamine has proven efficacious in mast-cell-mediated experimental models of acute and neurogenic inflammation. This review will provide an overview of recent progress relating to the pathobiology of neuroinflammation, the role of microglia, neuroimmune interactions involving mast cells and the possibility that mast cell-microglia cross-talk contributes to the exacerbation of acute symptoms of chronic neurodegenerative disease and accelerates disease progression, as well as promoting pain transmission pathways. We will conclude by considering the therapeutic potential of treating systemic inflammation or blockade of signalling pathways from the periphery to the brain in such settings.
Collapse
Affiliation(s)
- Stephen D Skaper
- Dipartimento di Scienze del Farmaco, Largo 'Egidio Meneghetti' 2, Università degli Studi di Padova, Padova, Italy
| | | | | |
Collapse
|
67
|
Oyoshi MK, Oettgen HC, Chatila TA, Geha RS, Bryce PJ. Food allergy: Insights into etiology, prevention, and treatment provided by murine models. J Allergy Clin Immunol 2014; 133:309-17. [PMID: 24636470 DOI: 10.1016/j.jaci.2013.12.1045] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 12/12/2013] [Accepted: 12/13/2013] [Indexed: 12/15/2022]
Abstract
Food allergy is a rapidly growing public health concern because of its increasing prevalence and life-threatening potential. Animal models of food allergy have emerged as a tool for identifying mechanisms involved in the development of sensitization to normally harmless food allergens, as well as delineating the critical immune components of the effector phase of allergic reactions to food. However, the role animal models might play in understanding human diseases remains contentious. This review summarizes how animal models have provided insights into the etiology of human food allergy, experimental corroboration for epidemiologic findings that might facilitate prevention strategies, and validation for the utility of new therapies for food allergy. Improved understanding of food allergy from the study of animal models together with human studies is likely to contribute to the development of novel strategies to prevent and treat food allergy.
Collapse
Affiliation(s)
- Michiko K Oyoshi
- Division of Immunology, Boston Children's Hospital and the Departments of Pediatrics, Harvard Medical School, Boston, Mass.
| | - Hans C Oettgen
- Division of Immunology, Boston Children's Hospital and the Departments of Pediatrics, Harvard Medical School, Boston, Mass
| | - Talal A Chatila
- Division of Immunology, Boston Children's Hospital and the Departments of Pediatrics, Harvard Medical School, Boston, Mass
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital and the Departments of Pediatrics, Harvard Medical School, Boston, Mass
| | - Paul J Bryce
- Division of Allergy-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Ill.
| |
Collapse
|
68
|
Zhang H, Kong H, Zeng X, Guo L, Sun X, He S. Subsets of regulatory T cells and their roles in allergy. J Transl Med 2014; 12:125. [PMID: 24886492 PMCID: PMC4023533 DOI: 10.1186/1479-5876-12-125] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 03/28/2014] [Indexed: 12/17/2022] Open
Abstract
In recent years, it is recognized that acquired immunity is controlled by regulatory T cell (Treg). Since fundamental pathophysiological changes of allergy are mainly caused by hyperresponsiveness of immune system to allergens that acquires after birth, Tregs likely play key roles in the pathogenesis of allergy, particularly during the sensitization phase. However, accumulated information indicate that there are several distinctive subtypes of Tregs in man, and each of them seems to play different role in controlling immune system, which complicates the involvement of Tregs in allergy. The aim of the present study is to attempt to classify subtypes of Tregs and summarize their roles in allergy. Tregs should include natural Tregs (nTreg) including inducible costimulator (ICOS)(+) Tregs, inducible/adaptive Tregs (iTreg), interleukin (IL)-10-producing type 1 Tregs (Tr1 cells), CD8(+) Tregs and IL-17-producing Tregs. These cells share some common features including expression of Foxp3 (except for Tr1 cells), and secretion of inhibitory cytokine IL-10 and/or TGF-β. Furthermore, it is noticeable that Tregs likely contribute to allergic disorders such as dermatitis and airway inflammation, and play a crucial role in the treatment of allergy through their actions on suppression of effector T cells and inhibition of activation of mast cells and basophils. Modulation of functions of Tregs may provide a novel strategy to prevent and treat allergic diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Shaoheng He
- Allergy and Clinical Immunology Research Centre, the First Affiliated Hospital of Liaoning Medical University, No, 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning 121001, People's Republic of China.
| |
Collapse
|
69
|
Abstract
The cause of chronic pelvic pain syndrome (CPPS) has yet to be established. Since the late 1980s, cytokine, chemokine, and immunological classification studies using human samples have focused on identifying biomarkers for CPPS, but no diagnostically beneficial biomarkers have been identified, and these studies have done little to deepen our understanding of the mechanisms underlying chronic prostatic pain. Given the large number of men thought to be affected by this condition and the ineffective nature of current treatments, there is a pressing need to elucidate these mechanisms. Prostatitis types IIIa and IIIb are classified according to the presence of pain without concurrent presence of bacteria; however, it is becoming more evident that, although levels of bacteria are not directly associated with levels of pain, the presence of bacteria might act as the initiating factor that drives primary activation of mast-cell-mediated inflammation in the prostate. Mast cell activation is also known to suppress regulatory T cell (Treg) control of self-tolerance and also activate neural sensitization. This combination of established autoimmunity coupled with peripheral and central neural sensitization can result in the development of multiple symptoms, including pelvic pain and bladder irritation. Identifying these mechanisms as central mediators in CPPS offers new insight into the prospective treatment of the disease.
Collapse
|
70
|
Oldford SA, Marshall JS. Mast cells as targets for immunotherapy of solid tumors. Mol Immunol 2014; 63:113-24. [PMID: 24698842 DOI: 10.1016/j.molimm.2014.02.020] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 02/26/2014] [Accepted: 02/27/2014] [Indexed: 01/09/2023]
Abstract
Mast cells have historically been studied mainly in the context of allergic disease. In recent years, we have come to understand the critical importance of mast cells in tissue remodeling events and their role as sentinel cells in the induction and development of effective immune responses to infection. Studies of the role of mast cells in tumor immunity are more limited. The pro-tumorigenic role of mast cells has been widely reported. However, mast cell infiltration predicts improved prognosis in some cancers, suggesting that their prognostic value may be dependent on other variables. Such factors may include the nature of local mast cell subsets and the various activation stimuli present within the tumor microenvironment. Experimental models have highlighted the importance of mast cells in orchestrating the anti-tumor events that follow immunotherapies that target innate immunity. Mast cells are long-lived tissue resident cells that are abundant around many solid tumors and are radiation resistant making them unique candidates for combined treatment modalities. This review will examine some of the key roles of mast cells in tumor immunity, with a focus on potential immunotherapeutic interventions that harness the sentinel role of mast cells.
Collapse
Affiliation(s)
- Sharon A Oldford
- Dalhousie Inflammation Group, Dalhousie University, Halifax, NS, Canada; Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Jean S Marshall
- Dalhousie Inflammation Group, Dalhousie University, Halifax, NS, Canada; Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
71
|
Interleukin-6 signal transduction and its role in hepatic lipid metabolic disorders. Cytokine 2014; 66:133-42. [PMID: 24491813 DOI: 10.1016/j.cyto.2013.12.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Revised: 12/14/2013] [Accepted: 12/30/2013] [Indexed: 01/12/2023]
Abstract
Hepatic lipid dysregulation can lead to spectrum of metabolic disease conditions including metabolic syndrome (MS), fatty liver and diabetes. Liver lipids are regulated by a complex set of extra-hepatic and intra-hepatic factors including cellular cross-talk with variety of cells, inducing various cytokines. Interleukin 6(IL-6) is a pleiotropic cytokine that exerts both pro-inflammatory and anti-inflammatory effects on hepatic system through either JNK/STAT or ERK/MAPK signaling. Although, IL-6 has shown to protect the liver from fat storage in both rodent and human models and various IL-6(-/-) studies have supported this notion yet a question remains over its deleterious pro-inflammatory effects on hepatocytes. IL-6 ability to produce reactive oxygen species (ROS) and subsequently disturb the hepatic lipid balance has created a conundrum. Furthermore, IL-6 has shown to behave differently under different disease states within hepatocytes and hence, modulating the hepatic lipids accordingly. This review deals with the role of IL-6 on hepatic lipid metabolism and analyzes various data presented on this topic.
Collapse
|
72
|
Graham BB, Chabon J, Kumar R, Kolosionek E, Gebreab L, Debella E, Edwards M, Diener K, Shade T, Bifeng G, Bandeira A, Butrous G, Jones K, Geraci M, Tuder RM. Protective role of IL-6 in vascular remodeling in Schistosoma pulmonary hypertension. Am J Respir Cell Mol Biol 2014; 49:951-9. [PMID: 23815102 DOI: 10.1165/rcmb.2012-0532oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Schistosomiasis is one of the most common causes of pulmonary arterial hypertension worldwide, but the pathogenic mechanism by which the host inflammatory response contributes to vascular remodeling is unknown. We sought to identify signaling pathways that play protective or pathogenic roles in experimental Schistosoma-induced pulmonary vascular disease via whole-lung transcriptome analysis. Wild-type mice were experimentally exposed to Schistosoma mansoni ova by intraperitoneal sensitization followed by tail-vein augmentation, and the phenotype was assessed by right ventricular catheterization and tissue histology, as well as RNA and protein analysis. Whole-lung transcriptome analysis by microarray and RNA sequencing was performed, and RNA sequencing was analyzed according to two bioinformatics methods. Functional testing of the candidate IL-6 pathway was determined using IL-6 knockout mice and the signal transducers and activators of transcription protein-3 (STAT3) inhibitor S3I-201. Wild-type mice exposed to S. mansoni demonstrated increased right ventricular systolic pressure and thickness of the pulmonary vascular media. Whole-lung transcriptome analysis determined that the IL-6-STAT3-nuclear factor of activated T cells c2(NFATc2) pathway was up-regulated, as confirmed by PCR and the immunostaining of lung tissue from S. mansoni-exposed mice and patients who died of the disease. Mice lacking IL-6 or treated with S3I-201 developed pulmonary hypertension, associated with significant intima remodeling after exposure to S. mansoni. Whole-lung transcriptome analysis identified the up-regulation of the IL-6-STAT3-NFATc2 pathway, and IL-6 signaling was found to be protective against Schistosoma-induced intimal remodeling.
Collapse
Affiliation(s)
- Brian B Graham
- 1 Program in Translational Lung Research, Division of Pulmonary Sciences and Critical Care Medicine, and
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Krstic J, Santibanez JF. Transforming growth factor-beta and matrix metalloproteinases: functional interactions in tumor stroma-infiltrating myeloid cells. ScientificWorldJournal 2014; 2014:521754. [PMID: 24578639 PMCID: PMC3918721 DOI: 10.1155/2014/521754] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 10/28/2013] [Indexed: 01/05/2023] Open
Abstract
Transforming growth factor-beta (TGF-β) is a pleiotropic factor with several different roles in health and disease. In tumorigenesis, it may act as a protumorigenic factor and have a profound impact on the regulation of the immune system response. Matrix metalloproteinases (MMPs) are a family that comprises more than 25 members, which have recently been proposed as important regulators acting in tumor stroma by regulating the response of noncellular and cellular microenvironment. Tumor stroma consists of several types of resident cells and infiltrating cells derived from bone marrow, which together play crucial roles in the promotion of tumor growth and metastasis. In cancer cells, TGF-β regulates MMPs expression, while MMPs, produced by either cancer cells or residents' stroma cells, activate latent TGF-β in the extracellular matrix, together facilitating the enhancement of tumor progression. In this review we will focus on the compartment of myeloid stroma cells, such as tumor-associated macrophages, neutrophils, and dendritic and mast cells, which are potently regulated by TGF-β and produce large amounts of MMPs. Their interplay and mutual implications in the generation of pro-tumorigenic cancer microenvironment will be analyzed.
Collapse
Affiliation(s)
- Jelena Krstic
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Dr Subotića 4, 11129 Belgrade, Serbia
| | - Juan F. Santibanez
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Dr Subotića 4, 11129 Belgrade, Serbia
| |
Collapse
|
74
|
Interleukin-25 (IL-25) promotes efficient protective immunity against Trichinella spiralis infection by enhancing the antigen-specific IL-9 response. Infect Immun 2013; 81:3731-41. [PMID: 23897610 DOI: 10.1128/iai.00646-13] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Mammalian hosts often develop distinct immune response against the diverse parasitic helminths that have evolved for immune evasion. Interleukin-25 (IL-25), an IL-17 cytokine family member, plays a key role in initiating the protective immunity against several parasitic helminths; however, the involvement and underlying mechanisms by which IL-25 mediates immune response against Trichinella spiralis infection have not been investigated. Here we showed that IL-25 functions in promoting protective immunity against T. spiralis infection. Mice treated with IL-25 exhibited a lower worm burden and fewer muscle larvae in the later stage of T. spiralis infection. In contrast, mice treated with neutralizing antibody against IL-25 failed to expel T. spiralis effectively. During T. spiralis infection, intestinal IL-25 expression was rapidly elevated before the onset of IL-4 and IL-9 induction. While antigen-specific Th2 and Th9 immune responses were both developed during T. spiralis infection, an antigen-specific Th9 response appeared to be transiently induced in the early stage of infection. Mice into which antigen-specific T cells deficient in IL-9 were transferred were less effective in worm clearance than those given wild-type T cells. The strength of the antigen-specific Th9 immune response against T. spiralis could be enhanced or attenuated after treatment with IL-25 or neutralizing antibody against IL-25, respectively, correlating positively with the levels of intestinal mastocytosis and the expression of IL-9-regulated genes, including mast cell- and Paneth cell-specific genes. Thus, our study demonstrates that intestinal IL-25 promotes protective immunity against T. spiralis infection by inducing antigen-specific Th9 immune response.
Collapse
|
75
|
Kanjarawi R, Dy M, Bardel E, Sparwasser T, Dubois B, Mecheri S, Kaiserlian D. Regulatory CD4+Foxp3+ T cells control the severity of anaphylaxis. PLoS One 2013; 8:e69183. [PMID: 23922690 PMCID: PMC3724852 DOI: 10.1371/journal.pone.0069183] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 06/05/2013] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Anaphylaxis is a life-threatening outcome of immediate-type hypersensitivity to allergen, consecutive to mast cell degranulation by allergen-specific IgE. Regulatory T cells (Treg) can control allergic sensitization and mast cell degranulation, yet their clinical benefit on anaphylactic symptoms is poorly documented. Here we investigated whether Treg action during the effector arm of the allergic response alleviates anaphylaxis. METHODS We used a validated model of IgE-mediated passive systemic anaphylaxis, induced by intravenous challenge with DNP-HSA in mice passively sensitized with DNP-specific IgE. Anaphylaxis was monitored by the drop in body temperature as well as plasma histamine and serum mMCP1 levels. The role of Treg was analyzed using MHC class II-deficient (Aβ(°/°)) mice, treatment with anti-CD25 or anti-CD4 mAbs and conditional ablation of Foxp3(+) Treg in DEREG mice. Therapeutic efficacy of Treg was also evaluated by transfer experiments using FoxP3-eGFP knock-in mice. RESULTS Anaphylaxis did not occur in mast cell-deficient W/W(v) mutant mice and was only moderate and transient in mice deficient for histamine receptor-1. Defects in constitutive Treg, either genetic or induced by antibody or toxin treatment resulted in a more severe and/or sustained hypothermia, associated with a rise in serum mMCP1, but not histamine. Adoptive transfer of Foxp3(+) Treg from either naïve or DNP-sensitized donors similarly alleviated body temperature loss in Treg-deficient DEREG mice. CONCLUSION Constitutive Foxp3(+) Treg can control the symptomatic phase of mast cell and IgE-dependent anaphylaxis in mice. This might open up new therapeutic avenues using constitutive rather than Ag-specific Treg for inducing tolerance in allergic patients.
Collapse
Affiliation(s)
- Reem Kanjarawi
- CIRI, International Center for Infectiology Research, “Mucosal immunity, Vaccination & Biotherapies” Team, Université de Lyon, Lyon, France
- INSERM, U1111, Lyon, France
- École Normale Supérieure de Lyon, Lyon, France
- Université Lyon 1, Centre International de Recherche en Infectiologie, Lyon, France
- CNRS, UMR5308, Lyon, France
| | - Michel Dy
- CNRS/UMR 8147, université René Descartes, Hôpital Necker, Paris, France
| | - Emilie Bardel
- CIRI, International Center for Infectiology Research, “Mucosal immunity, Vaccination & Biotherapies” Team, Université de Lyon, Lyon, France
- INSERM, U1111, Lyon, France
- École Normale Supérieure de Lyon, Lyon, France
- Université Lyon 1, Centre International de Recherche en Infectiologie, Lyon, France
- CNRS, UMR5308, Lyon, France
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Center for Experimental and Clinical Infection Research, Hannover, Germany
| | - Bertrand Dubois
- CIRI, International Center for Infectiology Research, “Mucosal immunity, Vaccination & Biotherapies” Team, Université de Lyon, Lyon, France
- INSERM, U1111, Lyon, France
- École Normale Supérieure de Lyon, Lyon, France
- Université Lyon 1, Centre International de Recherche en Infectiologie, Lyon, France
- CNRS, UMR5308, Lyon, France
| | - Salah Mecheri
- Institut Pasteur, Unité de Biologie des Interactions Hôte Parasites, Paris, France
- Centre National de la Recherche Scientifique, Unité de Recherche Associée 2581, Paris, France
| | - Dominique Kaiserlian
- CIRI, International Center for Infectiology Research, “Mucosal immunity, Vaccination & Biotherapies” Team, Université de Lyon, Lyon, France
- INSERM, U1111, Lyon, France
- École Normale Supérieure de Lyon, Lyon, France
- Université Lyon 1, Centre International de Recherche en Infectiologie, Lyon, France
- CNRS, UMR5308, Lyon, France
- * E-mail:
| |
Collapse
|
76
|
Wisniewski J, Agrawal R, Woodfolk JA. Mechanisms of tolerance induction in allergic disease: integrating current and emerging concepts. Clin Exp Allergy 2013; 43:164-76. [PMID: 23331558 DOI: 10.1111/cea.12016] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The prevalence of atopy and allergic disease continues to escalate worldwide. Defining immune mechanisms that suppress the underlying Th2-driven inflammatory process is critical for the rational design of new treatments to prevent or attenuate disease. Allergen immunotherapy has provided a useful framework for evaluating changes in the immune response that occur during the development of tolerance. Despite this, elucidating the phenotypic and functional properties of regulatory cells, has proven challenging in humans with allergic disease. This article provides an overview of our current understanding of the immune pathways that orchestrate allergen tolerance, with an emphasis on emerging concepts related to human disease. A variety of regulatory cell types, including IL-10-secreting T and B cells, play a pivotal role in suppressing allergic responses to inhaled, ingested and injected allergens. These cells may inhibit Th2 effectors directly, or else indirectly, through other cell types and mediators. Protective antibodies, including IgG4, Fc sialylated IgG, and IgA, have the capacity to modulate the response by preventing allergen binding to surface-bound IgE, or inhibiting dendritic cell maturation. Immune cell plasticity may augment suppression of Th2 cells by T regulatory cells, through mechanisms that involve T cell conversion, or else unconventional roles of classical effector cells. These actions depend upon external cues provided by the in vivo milieu. As such, specific anatomical sites may preferentially favour tolerance induction. Recent scientific advances now allow a global analysis of immune parameters that capture novel markers of tolerance induction in allergic patients. Such markers could provide new molecular targets for assessing tolerance, and for designing treatments that confer long-lasting protection in a safe and efficacious fashion.
Collapse
Affiliation(s)
- J Wisniewski
- Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908-1355, USA
| | | | | |
Collapse
|
77
|
Ganeshan K, Johnston LK, Bryce PJ. TGF-β1 limits the onset of innate lung inflammation by promoting mast cell-derived IL-6. THE JOURNAL OF IMMUNOLOGY 2013; 190:5731-8. [PMID: 23630359 DOI: 10.4049/jimmunol.1203362] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
TGF-β1 is an important suppressive mediator of inflammation, but it can also drive fibrosis and remodeling in the lung. In response to intratracheal LPS, neutrophils migrate into the lung, and TGF-β1 was suggested to protect against the ensuing injury. However, the mechanisms for this protective role remain unknown. Using a model of acute lung injury, we demonstrate that TGF-β1 decreases neutrophil numbers during the onset of injury. This was due to increased apoptosis rather than reduced migration. We demonstrate that TGF-β1 does not directly regulate neutrophil apoptosis but instead functions through IL-6 to promote neutrophil clearance. rIL-6 is sufficient to promote neutrophil apoptosis and reduce neutrophilia in bronchoalveolar lavage fluid, while IL-6 increases rapidly following LPS-induced injury. Mast cells are a critical source of IL-6, because mast cell-deficient mice exhibit increased neutrophil numbers that are reduced by reconstitution with wild-type, but not IL-6(-/-), mast cells. Although IL-6 diminishes neutrophilia in mast cell-deficient mice, TGF-β1 is ineffective, suggesting that these effects were mast cell dependent. Taken together, our findings establish a novel pathway through which TGF-β1, likely derived from resident regulatory T cells, controls the severity and magnitude of early innate inflammation by promoting IL-6 from mast cells.
Collapse
Affiliation(s)
- Kirthana Ganeshan
- Division of Allergy/Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | |
Collapse
|
78
|
Hong GU, Kim NG, Jeoung D, Ro JY. Anti-CD40 Ab- or 8-oxo-dG-enhanced Treg cells reduce development of experimental autoimmune encephalomyelitis via down-regulating migration and activation of mast cells. J Neuroimmunol 2013; 260:60-73. [PMID: 23622820 DOI: 10.1016/j.jneuroim.2013.04.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 03/08/2013] [Accepted: 04/01/2013] [Indexed: 11/18/2022]
Abstract
This study investigated whether anti-CD40 Ab and 8-oxo-dG attenuate mast cell migration and EAE development. Anti-CD40 Ab and 8-oxo-dG reduced EAE scores, mast cell numbers, expression of adhesion molecules, OX40L and Act1, levels of TNF-α, LTs, expression of cytokines, and co-localization of Treg cells and mast cells, all of which are increased in EAE-brain tissues. Each treatment enhanced Treg cells, expression of OX40, and cytokines related to suppressive function of Treg cells in EAE brain tissues. Act-BMMCs with Treg cells reduced expression of OX40L and CCL2/CCR2, VCAM-1, PECAM-1, [Ca²⁺]i levels, release of mediators, various signaling molecules, Act1 related to IL-17a signals versus those in act-BMMCs without Treg cells. The data suggest that IL-10- and IL-35-producing Foxp3⁺-Treg cells, enhanced by anti-CD40 Ab or 8-oxo-dG, suppress migration of mast cells through down-regulating the expression of adhesion molecules, and suppress mast cell activation through cell-to-cell cross-talk via OX40/OX40L in EAE development.
Collapse
Affiliation(s)
- Gwan Ui Hong
- Department of Pharmacology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-746, South Korea
| | | | | | | |
Collapse
|
79
|
Abstract
Hypersensitivity reactions of the immune system have been broadly categorized into the atopic and autoimmune depending on whether the antigen triggering the reaction is endogenous (or self) or exogenous, the types of cellular and humoral components involved, and the clinical symptoms. Research into the pathophysiology of the resultant disease states has focused on a dichotomy between Th1 and Th2 T helper lymphocytes thought to govern autoimmune and atopic disease, respectively. Recent discoveries, however, have served to dispute this paradigm and have provided additional insight into the roles of Th17 cells, B-lymphocytes and T regulatory cells as well as the considerable communication and commonalities between the complex signaling pathways. Furthermore, clinical studies have served to challenge the idea that the presence of atopy and autoimmunity are mutually exclusive states. Finally, application of recent approaches to treatment-biologic targeted therapy in autoimmunity and induction of immune tolerance in atopic disease--to both disease states have shown mixed but promising results.
Collapse
|
80
|
Pezato R, Balsalobre L, Lima M, Bezerra TFP, Voegels RL, Gregório LC, Stamm AC, van Zele T. Convergence of two major pathophysiologic mechanisms in nasal polyposis: immune response to Staphylococcus aureus and airway remodeling. J Otolaryngol Head Neck Surg 2013; 42:27. [PMID: 23663431 PMCID: PMC3651231 DOI: 10.1186/1916-0216-42-27] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 03/18/2013] [Indexed: 12/19/2022] Open
Abstract
This review is addressed two pathophysiologic mechanisms implicated in the pathogenesis of nasal polyposis: the unique remodeling process found in nasal polyp tissue and the immune response of patients with nasal polyposis to Staphylococcus aureus. These two theories converge to the same direction in different aspects, including decreased extracellular matrix production, impaired T regulation and favoring of a Th2 immune response. In patients with nasal polyposis, an exaggerated immune response to Staphylococcus aureus may aggravate the airway remodeling process.
Collapse
Affiliation(s)
- Rogério Pezato
- Department of Otolaryngology - Head and Neck Surgery of Federal University of São Paulo, Rua Maestro Antão Fernandes, 173, Jd São Bento, São Paulo, SP02526-060, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
81
|
Can we produce true tolerance in patients with food allergy? J Allergy Clin Immunol 2013; 131:14-22. [PMID: 23265693 DOI: 10.1016/j.jaci.2012.10.058] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 10/05/2012] [Accepted: 10/05/2012] [Indexed: 11/23/2022]
Abstract
Immune tolerance is defined as nonresponsiveness of the adaptive immune system to antigens. Immune mechanisms preventing inappropriate immune reactivity to innocuous antigens include deletion of reactive lymphocytes and generation of regulatory T (Treg) cells. The normal response to food antigens is the generation of antigen-specific Treg cells. In patients with food allergy, the dominant immune response is a T(H)2-skewed T-cell response and the generation of food-specific IgE antibodies from B cells. It is not known whether a failure of the Treg cell response is behind this inappropriate immune response, but interventions that boost the Treg cell response, such as mucosal immunotherapy, might lead to a restoration of immune tolerance to foods. Tolerance has been notoriously difficult to restore in animal disease models, but limited data from human trials suggest that tolerance (sustained nonresponsiveness) can be re-established in a subset of patients. Furthermore, studies on the natural history of food allergy indicate that spontaneous development of tolerance to foods over time is not uncommon. The current challenge is to understand the mechanisms responsible for restoration of natural or induced tolerance so that interventions can be developed to more successfully induce tolerance in the majority of patients with food allergy.
Collapse
|
82
|
Clinical significance of peritumoral mast cells in esophageal squamous cell carcinoma with neoadjuvant chemoradiotherapy. Esophagus 2013. [DOI: 10.1007/s10388-012-0356-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
83
|
Coomes SM, Pelly VS, Wilson MS. Plasticity within the αβ⁺CD4⁺ T-cell lineage: when, how and what for? Open Biol 2013; 3:120157. [PMID: 23345540 PMCID: PMC3603458 DOI: 10.1098/rsob.120157] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Following thymic output, αβ⁺CD4⁺ T cells become activated in the periphery when they encounter peptide-major histocompatibility complex. A combination of cytokine and co-stimulatory signals instructs the differentiation of T cells into various lineages and subsequent expansion and contraction during an appropriate and protective immune response. Our understanding of the events leading to T-cell lineage commitment has been dominated by a single fate model describing the commitment of T cells to one of several helper (T(H)), follicular helper (T(FH)) or regulatory (T(REG)) phenotypes. Although a single lineage-committed and dedicated T cell may best execute a single function, the view of a single fate for T cells has recently been challenged. A relatively new paradigm in αβ⁺CD4⁺ T-cell biology indicates that T cells are much more flexible than previously appreciated, with the ability to change between helper phenotypes, between helper and follicular helper, or, most extremely, between helper and regulatory functions. In this review, we comprehensively summarize the recent literature identifying when T(H) or T(REG) cell plasticity occurs, provide potential mechanisms of plasticity and ask if T-cell plasticity is beneficial or detrimental to immunity.
Collapse
Affiliation(s)
- Stephanie M Coomes
- Division of Molecular Immunology, National Institute for Medical Research, MRC, London NW7 1AA, UK
| | | | | |
Collapse
|
84
|
A molecular insight of CTLA-4 in food allergy. Immunol Lett 2013; 149:101-9. [DOI: 10.1016/j.imlet.2012.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Accepted: 12/06/2012] [Indexed: 12/31/2022]
|
85
|
Dominguez JA, Xie Y, Dunne WM, Yoseph BP, Burd EM, Coopersmith CM, Davidson NO. Intestine-specific Mttp deletion decreases mortality and prevents sepsis-induced intestinal injury in a murine model of Pseudomonas aeruginosa pneumonia. PLoS One 2012; 7:e49159. [PMID: 23145105 PMCID: PMC3493497 DOI: 10.1371/journal.pone.0049159] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 10/04/2012] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The small intestine plays a crucial role in the pathophysiology of sepsis and has been referred to as the "motor" of the systemic inflammatory response. One proposed mechanism is that toxic gut-derived lipid factors, transported in mesenteric lymph, induce systemic injury and distant organ failure. However, the pathways involved are yet to be defined and the role of intestinal chylomicron assembly and secretion in transporting these lipid factors is unknown. Here we studied the outcome of sepsis in mice with conditional, intestine-specific deletion of microsomal triglyceride transfer protein (Mttp-IKO), which exhibit a block in chylomicron assembly together with lipid malabsorption. METHODOLOGY/PRINCIPAL FINDINGS Mttp-IKO mice and controls underwent intratracheal injection with either Pseudomonas aeruginosa or sterile saline. Mttp-IKO mice exhibited decreased seven-day mortality, with 0/20 (0%) dying compared to 5/17 (29%) control mice (p<0.05). This survival advantage in Mttp-IKO mice, however, was not associated with improvements in pulmonary bacterial clearance or neutrophil infiltration. Rather, Mttp-IKO mice exhibited protection against sepsis-associated decreases in villus length and intestinal proliferation and were also protected against increased intestinal apoptosis, both central features in control septic mice. Serum IL-6 levels, a major predictor of mortality in human and mouse models of sepsis, were elevated 8-fold in septic control mice but remained unaltered in septic Mttp-IKO mice. Serum high density lipoprotein (HDL) levels were reduced in septic control mice but were increased in septic Mttp-IKO mice. The decreased levels of HDL were associated with decreased hepatic expression of apolipoprotein A1 in septic control mice. CONCLUSIONS/SIGNIFICANCE These studies suggest that strategies directed at blocking intestinal chylomicron secretion may attenuate the progression and improve the outcome of sepsis through effects mediated by metabolic and physiological adaptations in both intestinal and hepatic lipid flux.
Collapse
Affiliation(s)
- Jessica A. Dominguez
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Yan Xie
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - W. Michael Dunne
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Benyam P. Yoseph
- Emory Center for Critical Care and Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Eileen M. Burd
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Craig M. Coopersmith
- Emory Center for Critical Care and Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Nicholas O. Davidson
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
86
|
Kim JS, Shin DC, Woo MY, Kwon MH, Kim K, Park S. T Cell Immunoglobulin Mucin Domain (TIM)-3 Promoter Activity in a Human Mast Cell Line. Immune Netw 2012; 12:207-12. [PMID: 23213314 PMCID: PMC3509165 DOI: 10.4110/in.2012.12.5.207] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 09/19/2012] [Accepted: 09/21/2012] [Indexed: 12/14/2022] Open
Abstract
T cell immunoglobulin mucin domain (TIM)-3 is an immunomodulatory molecule and upregulated in T cells by several cytokines. TIM-3 also influences mast cell function but its transcriptional regulation in mast cells has not been clarified. Therefore, we examined the transcript level and the promoter activity of TIM-3 in mast cells. The TIM-3 transcript level was assessed by real-time RT-PCR and promoter activity by luciferase reporter assay. TIM-3 mRNA levels were increased in HMC-1, a human mast cell line by TGF-β1 stimulation but not by stimulation with interferon (IFN)-α, IFN-λ, TNF-α, or IL-10. TIM-3 promoter -349~+144 bp region relative to the transcription start site was crucial for the basal and TGF-β1-induced TIM-3 promoter activities in HMC-1 cells. TIM-3 promoter activity was increased by overexpression of Smad2 and Smad4, downstream molecules of TGF-β1 signaling. Our results localize TIM-3 promoter activity to the region spanning -349 to +144 bp in resting and TGF-β1 stimulated mast cells.
Collapse
Affiliation(s)
- Jung Sik Kim
- Department of Microbiology, Ajou University School of Medicine, Suwon 442-749, Korea. ; Graduate Program of Molecular Medicine, Ajou University School of Medicine, Suwon 442-749, Korea
| | | | | | | | | | | |
Collapse
|
87
|
Okwor I, Onyilagha C, Kuriakose S, Mou Z, Jia P, Uzonna JE. Regulatory T cells enhance susceptibility to experimental Trypanosoma congolense infection independent of mouse genetic background. PLoS Negl Trop Dis 2012; 6:e1761. [PMID: 22860150 PMCID: PMC3409116 DOI: 10.1371/journal.pntd.0001761] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 06/20/2012] [Indexed: 12/22/2022] Open
Abstract
Background BALB/c mice are highly susceptible while C57BL/6 are relatively resistant to experimental Trypanosoma congolense infection. Although regulatory T cells (Tregs) have been shown to regulate the pathogenesis of experimental T. congolense infection, their exact role remains controversial. We wished to determine whether Tregs contribute to distinct phenotypic outcomes in BALB/c and C57BL/6 mice and if so how they operate with respect to control of parasitemia and production of disease-exacerbating proinflammatory cytokines. Methodology/Findings BALB/c and C57BL/6 mice were infected intraperitoneally (i.p) with 103T. congolense clone TC13 and both the kinetics of Tregs expansion and intracellular cytokine profiles in the spleens and livers were monitored directly ex vivo by flow cytometry. In some experiments, mice were injected with anti-CD25 mAb prior or post T. congolense infection or adoptively (by intravenous route) given highly enriched naïve CD25+ T lymphocytes prior to T. congolense infection and the inflammatory cytokine/chemokine levels and survival were monitored. In contrast to a transient and non significant increase in the percentages and absolute numbers of CD4+CD25+Foxp3+ T cells (Tregs) in C57BL/6 mouse spleens and livers, a significant increase in the percentage and absolute numbers of Tregs was observed in spleens of infected BALB/c mice. Ablation or increasing the number of CD25+ cells in the relatively resistant C57BL/6 mice by anti-CD25 mAb treatment or by adoptive transfer of CD25+ T cells, respectively, ameliorates or exacerbates parasitemia and production of proinflammatory cytokines. Conclusion Collectively, our results show that regulatory T cells contribute to susceptibility in experimental murine trypanosomiasis in both the highly susceptible BALB/c and relatively resistant C57BL/6 mice. BALB/c mice are highly susceptible while C57BL/6 is relatively resistant to experimental Trypanosoma congolense infection. Acute death observed in infected BALB/c mice is usually associated with the excessive production of pro-inflammatory cytokines. Regulatory T cells (Tregs) have been shown to play a significant role in the pathogenesis of many diseases including those caused by parasites. However, the role of Tregs in the pathogenesis of T. congolense infection remains unclear. We were interested in addressing the following questions: Do Tregs contribute to the distinct phenotypic outcomes observed in T. congolense-infected BALB/c and C57BL/6 mice? If so, where and how do they operate with respect to parasitemia and cytokine response? By selectively altering the numbers of these cells either by targeted depletion with monoclonal antibody or adoptive transfer of highly enriched naïve CD25+ cells prior to infection, we show that Tregs impairs efficient parasite control and impacts on production of disease-exacerbating proinflammatory cytokines. Collectively, our findings suggest that Tregs contribute to enhanced susceptibility to experimental T. congolense infection in mice.
Collapse
Affiliation(s)
- Ifeoma Okwor
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Chukwunonso Onyilagha
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Shiby Kuriakose
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Zhirong Mou
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ping Jia
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jude E. Uzonna
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- * E-mail:
| |
Collapse
|
88
|
Gri G, Frossi B, D'Inca F, Danelli L, Betto E, Mion F, Sibilano R, Pucillo C. Mast cell: an emerging partner in immune interaction. Front Immunol 2012; 3:120. [PMID: 22654879 PMCID: PMC3360165 DOI: 10.3389/fimmu.2012.00120] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 04/27/2012] [Indexed: 01/09/2023] Open
Abstract
Mast cells (MCs) are currently recognized as effector cells in many settings of the immune response, including host defense, immune regulation, allergy, chronic inflammation, and autoimmune diseases. MC pleiotropic functions reflect their ability to secrete a wide spectrum of preformed or newly synthesized biologically active products with pro-inflammatory, anti-inflammatory and/or immunosuppressive properties, in response to multiple signals. Moreover, the modulation of MC effector phenotypes relies on the interaction of a wide variety of membrane molecules involved in cell–cell or cell-extracellular-matrix interaction. The delivery of co-stimulatory signals allows MC to specifically communicate with immune cells belonging to both innate and acquired immunity, as well as with non-immune tissue-specific cell types. This article reviews and discusses the evidence that MC membrane-expressed molecules play a central role in regulating MC priming and activation and in the modulation of innate and adaptive immune response not only against host injury, but also in peripheral tolerance and tumor-surveillance or -escape. The complex expression of MC surface molecules may be regarded as a measure of connectivity, with altered patterns of cell–cell interaction representing functionally distinct MC states. We will focalize our attention on roles and functions of recently discovered molecules involved in the cross-talk of MCs with other immune partners.
Collapse
Affiliation(s)
- Giorgia Gri
- Immunology Laboratory, Department of Medical and Biological Science, University of Udine Udine, Italy
| | | | | | | | | | | | | | | |
Collapse
|