51
|
Guerra PV, Andrade CM, Nunes IV, Gama BC, Tibúrcio R, Santos WLC, Azevedo VA, Tavares NM, Rebouças JDS, Maiolii TU, Faria AMC, Brodskyn CI. Oral Tolerance Induced by Heat Shock Protein 65-Producing Lactococcus lactis Mitigates Inflammation in Leishmania braziliensis Infection. Front Immunol 2021; 12:647987. [PMID: 34248935 PMCID: PMC8264454 DOI: 10.3389/fimmu.2021.647987] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 06/07/2021] [Indexed: 12/05/2022] Open
Abstract
Cutaneous leishmaniasis caused by L. braziliensis induces a pronounced Th1 inflammatory response characterized by IFN-γ production. Even in the absence of parasites, lesions result from a severe inflammatory response in which inflammatory cytokines play an important role. Different approaches have been used to evaluate the therapeutic potential of orally administrated heat shock proteins (Hsp). These proteins are evolutionarily preserved from bacteria to humans, highly expressed under inflammatory conditions and described as immunodominant antigens. Tolerance induced by the oral administration of Hsp65 is capable of suppressing inflammation and inducing differentiation in regulatory cells, and has been successfully demonstrated in several experimental models of autoimmune and inflammatory diseases. We initially administered recombinant Lactococcus lactis (L. lactis) prior to infection as a proof of concept, in order to verify its immunomodulatory potential in the inflammatory response arising from L. braziliensis. Using this experimental approach, we demonstrated that the oral administration of a recombinant L. lactis strain, which produces and secretes Hsp65 from Mycobacterium leprae directly into the gut, mitigated the effects of inflammation caused by L. braziliensis infection in association or not with PAM 3CSK4 (N-α-Palmitoyl-S-[2,3-bis(palmitoyloxy)-(2RS)-propyl]-L-cysteine, a TLR2 agonist). This was evidenced by the production of anti-inflammatory cytokines and the expansion of regulatory T cells in the draining lymph nodes of BALB/c mice. Our in vitro experimental results suggest that IL-10, TLR-2 and LAP are important immunomodulators in L. braziliensis infection. In addition, recombinant L. lactis administered 4 weeks after infection was observed to decrease lesion size, as well as the number of parasites, and produced a higher IL-10 production and decrease IFN-γ secretion. Together, these results indicate that Hsp65-producing L. lactis can be considered as an alternative candidate for treatment in both autoimmune diseases, as well as in chronic infections that cause inflammatory disease.
Collapse
Affiliation(s)
- Priscila Valera Guerra
- Laboratório da Interação Parasita-Hospedeiro e Epidemiologia (LAIPHE) Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Curso de Medicina, Centro Universitário Christus, Fortaleza, Brazil
| | - Camila Mattos Andrade
- Laboratório da Interação Parasita-Hospedeiro e Epidemiologia (LAIPHE) Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Ivanéia Valeriano Nunes
- Laboratório da Interação Parasita-Hospedeiro e Epidemiologia (LAIPHE) Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Brena Cardoso Gama
- Laboratório da Interação Parasita-Hospedeiro e Epidemiologia (LAIPHE) Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Rafael Tibúrcio
- Laboratório da Interação Parasita-Hospedeiro e Epidemiologia (LAIPHE) Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Washington Luis Conrado Santos
- Laboratório de Patologia Estrutural e Molecular (LAPEM), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Departamento de Patologia e Medicina Legal Faculdade de Medicina da Universidade Federal da Bahia, Salvador, Brazil
| | - Vasco Ariston Azevedo
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biomédicas, Universidade Federal de Minais Gerais, Belo Horizonte, Brazil
| | - Natalia Machado Tavares
- Laboratório da Interação Parasita-Hospedeiro e Epidemiologia (LAIPHE) Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| | - Juliana de Souza Rebouças
- Instituto de Ciências Biológicas, Programa de Pós Graduação em Ciências da Saúde, Universidade de Pernambuco, Recife, Brazil
| | - Tatiani Uceli Maiolii
- Departamento de Nutrição, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Maria Caetano Faria
- Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil.,Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Cláudia Ida Brodskyn
- Laboratório da Interação Parasita-Hospedeiro e Epidemiologia (LAIPHE) Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| |
Collapse
|
52
|
Voisine J, Abadie V. Interplay Between Gluten, HLA, Innate and Adaptive Immunity Orchestrates the Development of Coeliac Disease. Front Immunol 2021; 12:674313. [PMID: 34149709 PMCID: PMC8206552 DOI: 10.3389/fimmu.2021.674313] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/18/2021] [Indexed: 12/26/2022] Open
Abstract
Several environmental, genetic, and immune factors create a "perfect storm" for the development of coeliac disease: the antigen gluten, the strong association of coeliac disease with HLA, the deamidation of gluten peptides by the enzyme transglutaminase 2 (TG2) generating peptides that bind strongly to the predisposing HLA-DQ2 or HLA-DQ8 molecules, and the ensuing unrestrained T cell response. T cell immunity is at the center of the disease contributing to the inflammatory process through the loss of tolerance to gluten and the differentiation of HLA-DQ2 or HLA-DQ8-restricted anti-gluten inflammatory CD4+ T cells secreting pro-inflammatory cytokines and to the killing of intestinal epithelial cells by cytotoxic intraepithelial CD8+ lymphocytes. However, recent studies emphasize that the individual contribution of each of these cell subsets is not sufficient and that interactions between these different populations of T cells and the simultaneous activation of innate and adaptive immune pathways in distinct gut compartments are required to promote disease immunopathology. In this review, we will discuss how tissue destruction in the context of coeliac disease results from the complex interactions between gluten, HLA molecules, TG2, and multiple innate and adaptive immune components.
Collapse
Affiliation(s)
- Jordan Voisine
- Department of Medicine, The University of Chicago, Chicago, IL, United States.,Committee on Immunology, The University of Chicago, Chicago, IL, United States
| | - Valérie Abadie
- Department of Medicine, The University of Chicago, Chicago, IL, United States.,Section of Gastroenterology, Nutrition and Hepatology, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
53
|
Shi T, Li N, He Y, Feng J, Mei Z, Du Y, Jie Z. Th17/Treg cell imbalance plays an important role in respiratory syncytial virus infection compromising asthma tolerance in mice. Microb Pathog 2021; 156:104867. [PMID: 33957244 DOI: 10.1016/j.micpath.2021.104867] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/03/2021] [Accepted: 03/09/2021] [Indexed: 12/14/2022]
Abstract
Mucosal tolerance is induced early in life and is an important mechanism of protection from diseases, such as asthma. Respiratory syncytial virus (RSV) is a main cause of bronchiolitis and pneumonia in infants. Clinical studies have found that there is a strong association between RSV infection in infancy and later development of asthma, but the underlying mechanisms are unclear. A mouse model of immune tolerance induced by oral feeding of ovalbumin(OVA) was successfully established in our previous studies. We found that RSV infection could break the oral immune tolerance state.RSV infection increased the mRNA expression of IL-17A and IL-17A/Foxp3(the transcription factor forkhead box P3) in OT mice, but the mRNA expression of IL-4 and other T helper(Th)2 cytokines did not change significantly. As detected by flow cytometry analysis, RSV infection elevated Th17 cell levels and correspondingly decreased Regulatory T(Treg) cell levels in the hilar lymph nodes (HLNs) and mesenteric lymph nodes (MLNs), but there were no significant differences in the spleen or peripheral blood.We hypothesized that an imbalance in Th cells played an important role in RSV infection compromising asthma tolerance.RSV infection disrupted asthma tolerance by increasing the Th17/Treg ratio rather than the Th1/Th2 ratio'.Therefore, altering the Th17/Treg ratio has been identified as a potential therapeutic target in asthma caused by RSV or another virus.
Collapse
Affiliation(s)
- Tianyun Shi
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China; Center of Community-Based Health Research, Fudan University, China
| | - Na Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China; Center of Community-Based Health Research, Fudan University, China
| | - Yanchao He
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China; Center of Community-Based Health Research, Fudan University, China
| | - Jingjing Feng
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China; Center of Community-Based Health Research, Fudan University, China
| | - Zhoufang Mei
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China; Center of Community-Based Health Research, Fudan University, China
| | - Yong Du
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China; Center of Community-Based Health Research, Fudan University, China
| | - Zhijun Jie
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China; Center of Community-Based Health Research, Fudan University, China.
| |
Collapse
|
54
|
Krempski JW, Warren C, Han X, Zhang W, He Z, Lejeune S, Nadeau K. Food Allergies: An Example of Translational Research. Immunol Allergy Clin North Am 2021; 41:143-163. [PMID: 33863476 DOI: 10.1016/j.iac.2021.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Food allergies have been rising in prevalence since the 1990s, imposing substantial physical, psychosocial, and economic burdens on affected patients and their families. Until recently, the only therapy for food allergy was strict avoidance of the allergenic food. Recent advances in translational studies, however, have led to insights into allergic sensitization and tolerance. This article provides an overview of cutting-edge research into food allergy and immune tolerance mechanisms utilizing mouse models, human studies, and systems biology approaches. This research is being translated and implemented in the clinical setting to improve diagnosis and reduce food allergy's public health burden.
Collapse
Affiliation(s)
- James Walter Krempski
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA.
| | - Christopher Warren
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA
| | - Xiaorui Han
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA
| | - Wenming Zhang
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA
| | - Ziyuan He
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA
| | - Stéphanie Lejeune
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA
| | - Kari Nadeau
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA
| |
Collapse
|
55
|
Li C, Chen X, Luo X, Wang H, Zhu Y, Du G, Chen W, Chen Z, Hao X, Zhang Z, Sun X. Nanoemulsions Target to Ectopic Lymphoids in Inflamed Joints to Restore Immune Tolerance in Rheumatoid Arthritis. NANO LETTERS 2021; 21:2551-2561. [PMID: 33687217 DOI: 10.1021/acs.nanolett.0c05110] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Inducing immune tolerance through repeated administration of self-antigens is a promising strategy for treating rheumatoid arthritis (RA), and current research indicates that coadministration of immunomodulators can further orchestrate the tolerogenic response. However, most of the clinical trials based on tolerance induction have negligible therapeutic effects. Peripheral lymphoid organs play critical roles in immunotherapy. Here, we design an engineered nanoemulsion for targeted codelivery of self-antigens and an immunomodulator to ectopic lymphoid structures (ELSs) in inflamed joints of RA. Namely, a citrullinated multiepitope self-antigen (CitP) and rapamycin are incorporated into the nanoemulsions (NEs@CitP/Rapa), which are fabricated by a facial method using commercialized pharmaceutical excipients. After intravenous administration, the nanoemulsion shows satisfactory accumulation in the inflamed paws and provides enhanced anti-inflammatory effect in various experimental murine models of RA. Our study provides a promising targeting strategy to induce immune tolerance for the treatment of RA.
Collapse
Affiliation(s)
- Chenglong Li
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Xiaoyan Chen
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Xianjin Luo
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Hairui Wang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Yining Zhu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Guangshen Du
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Wenfei Chen
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Zhengjun Chen
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Xinyan Hao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| |
Collapse
|
56
|
Lutterotti A, Hayward-Koennecke H, Sospedra M, Martin R. Antigen-Specific Immune Tolerance in Multiple Sclerosis-Promising Approaches and How to Bring Them to Patients. Front Immunol 2021; 12:640935. [PMID: 33828551 PMCID: PMC8019937 DOI: 10.3389/fimmu.2021.640935] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 02/26/2021] [Indexed: 01/28/2023] Open
Abstract
Antigen-specific tolerance induction aims at treating multiple sclerosis (MS) at the root of its pathogenesis and has the prospect of personalization. Several promising tolerization approaches using different technologies and modes of action have already advanced to clinical testing. The prerequisites for successful tolerance induction include the knowledge of target antigens, core pathomechanisms, and how to pursue a clinical development path that is distinct from conventional drug development. Key aspects including patient selection, outcome measures, demonstrating the mechanisms of action as well as the positioning in the rapidly growing spectrum of MS treatments have to be considered to bring this therapy to patients.
Collapse
Affiliation(s)
- Andreas Lutterotti
- Neuroimmunology and MS Research Section, Neurology Clinic, University Hospital Zurich & University of Zurich, Zurich, Switzerland
| | - Helen Hayward-Koennecke
- Neuroimmunology and MS Research Section, Neurology Clinic, University Hospital Zurich & University of Zurich, Zurich, Switzerland
| | - Mireia Sospedra
- Neuroimmunology and MS Research Section, Neurology Clinic, University Hospital Zurich & University of Zurich, Zurich, Switzerland
| | - Roland Martin
- Neuroimmunology and MS Research Section, Neurology Clinic, University Hospital Zurich & University of Zurich, Zurich, Switzerland
| |
Collapse
|
57
|
Ness S, Lin S, Gordon JR. Regulatory Dendritic Cells, T Cell Tolerance, and Dendritic Cell Therapy for Immunologic Disease. Front Immunol 2021; 12:633436. [PMID: 33777019 PMCID: PMC7988082 DOI: 10.3389/fimmu.2021.633436] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DC) are antigen-presenting cells that can communicate with T cells both directly and indirectly, regulating our adaptive immune responses against environmental and self-antigens. Under some microenvironmental conditions DC develop into anti-inflammatory cells which can induce immunologic tolerance. A substantial body of literature has confirmed that in such settings regulatory DC (DCreg) induce T cell tolerance by suppression of effector T cells as well as by induction of regulatory T cells (Treg). Many in vitro studies have been undertaken with human DCreg which, as a surrogate marker of antigen-specific tolerogenic potential, only poorly activate allogeneic T cell responses. Fewer studies have addressed the abilities of, or mechanisms by which these human DCreg suppress autologous effector T cell responses and induce infectious tolerance-promoting Treg responses. Moreover, the agents and properties that render DC as tolerogenic are many and varied, as are the cells’ relative regulatory activities and mechanisms of action. Herein we review the most current human and, where gaps exist, murine DCreg literature that addresses the cellular and molecular biology of these cells. We also address the clinical relevance of human DCreg, highlighting the outcomes of pre-clinical mouse and non-human primate studies and early phase clinical trials that have been undertaken, as well as the impact of innate immune receptors and symbiotic microbial signaling on the immunobiology of DCreg.
Collapse
Affiliation(s)
- Sara Ness
- Department of Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Shiming Lin
- Department of Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - John R Gordon
- Department of Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada.,Division of Respirology, Critical Care and Sleep Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
58
|
Rodriguez-Sillke Y, Visekruna A, Glauben R, Siegmund B, Steinhoff U. Recognition of food antigens by the mucosal and systemic immune system: Consequences for intestinal development and homeostasis. Int J Med Microbiol 2021; 311:151493. [PMID: 33652373 DOI: 10.1016/j.ijmm.2021.151493] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 02/19/2021] [Accepted: 02/23/2021] [Indexed: 12/16/2022] Open
Abstract
The impact of nutrition on systemic and intestinal immune responses remains controversially discussed and yet not fully understood. The majority of studies investigating the effects of dietary antigens focused to understand how local and systemic unresponsiveness is induced by innocuous food antigens. Moreover, it has been shown that both, microbial and dietary antigens are essential for the normal development of the mucosal immune system. Based on experimental findings from animals and IBD patients, we propose a model how the intestinal immune system performs the balancing act between recognition and tolerance of dietary antigens at the same time: In the healthy gut, repetitive uptake of dietary antigens by Peyer's patches leads to increasing activation of CD4+ T cells till hyper-activated lymphocytes undergo apoptosis. In contrast to healthy controls, this mechanism was disturbed in Crohn's disease patients. This observation might help to better understand beneficial effects of dietary intervention therapy.
Collapse
Affiliation(s)
- Yasmina Rodriguez-Sillke
- Medical Department for Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Germany
| | - Alexander Visekruna
- Institute for Medical Microbiology and Hygiene, Philipps University of Marburg, Marburg, Germany
| | - Rainer Glauben
- Medical Department for Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Germany
| | - Britta Siegmund
- Medical Department for Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Germany
| | - Ulrich Steinhoff
- Institute for Medical Microbiology and Hygiene, Philipps University of Marburg, Marburg, Germany.
| |
Collapse
|
59
|
Barroso A, Mahler JV, Fonseca-Castro PH, Quintana FJ. Therapeutic induction of tolerogenic dendritic cells via aryl hydrocarbon receptor signaling. Curr Opin Immunol 2021; 70:33-39. [PMID: 33607496 DOI: 10.1016/j.coi.2021.02.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/28/2021] [Accepted: 02/02/2021] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DCs) are potent antigen-presenting cells (APCs), which sample the exogenous and endogenous cues to control adaptive immunity, balancing effector and regulatory components of the immune response. Multiple subsets of DCs, such as plasmacytoid and conventional DCs, have been defined based on specific phenotypic markers, functions and regulatory transcriptional programs. Tolerogenic DCs (tolDCs) have been functionally defined based on their ability to expand the regulatory T-cell compartment and suppress immune responses. However, it is still unclear whether tolDCs represent a homogeneous population, a specific DC subset and/or a heterogeneous collection of DC activation/maturation states. The ligand-activated transcription factor aryl hydrocarbon receptor (AHR) has been shown to control transcriptional programs associated to tolDCs. In this review, we discuss the role of AHR in the control of tolDCs, and also AHR-targeted approaches for the therapeutic induction of tolDCs in autoimmune diseases and allergy.
Collapse
Affiliation(s)
- Andreia Barroso
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - João V Mahler
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Pedro H Fonseca-Castro
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
60
|
Ssemakalu CC, Ulaszewska M, Elias S, Spencer AJ. Solar inactivated Salmonella Typhimurium induces an immune response in BALB/c mice. Heliyon 2021; 7:e05903. [PMID: 33553721 PMCID: PMC7855330 DOI: 10.1016/j.heliyon.2021.e05903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/16/2020] [Accepted: 12/31/2020] [Indexed: 12/20/2022] Open
Abstract
Salmonella is contracted through the consumption of untreated water and contaminated food. The contraction and spread of water-related Salmonella in resource-poor communities can be reduced by using solar disinfection (SODIS) to treat the water before its consumption. SODIS is a water sanitizing technique that relies on natural sunshine. It is a cost-effective, inexpensive, environmentally, and user-friendly means of treating microbiologically contaminated water. This water disinfection method has saved many lives in communities vulnerable to water-related infections worldwide. At present, the success of SODIS has mainly been attributed to permanent inactivation of water pathogens ability to grow. However, little to no information exists as to whether immune responses to the solar inactivated pathogens are induced in SODIS water consumers. This study assessed the potential for solar inactivated S. Typhimurium to induce an immune response in mice. Results show that solar inactivated S. Typhimurium can induce bactericidal antibodies against S. Typhimurium. Furthermore, an increase in the secretion of interferon-gamma (IFN-γ) was observed in mice given the solar inactivated S. Typhimurium. These findings suggest that solar inactivated S. Typhimurium induces a humoral and cellular immune response. However, the level of protection afforded by these responses requires further investigation.
Collapse
Affiliation(s)
- Cornelius C Ssemakalu
- Cell Biology Research Unit, Department of Biotechnology, Faculty of Applied and Computer Sciences, Vaal University of Technology, Vanderbijlpark, 1911, South Africa
| | - Marta Ulaszewska
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, United Kingdom
| | - Sean Elias
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, United Kingdom
| | - Alexandra J Spencer
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, United Kingdom
| |
Collapse
|
61
|
Feline lymphoplasmacytic rhinitis (FLPCR): Severity of inflammation correlates with reduced mucosal IgA expression. Vet Immunol Immunopathol 2021; 234:110193. [PMID: 33611160 DOI: 10.1016/j.vetimm.2021.110193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 11/23/2022]
Abstract
Feline lymphoplasmacytic rhinitis (FLPCR) is a rare disease with an unclear pathogenesis characterized by lymphoplasmacytic (LPC) inflammation and progressive tissue destruction. Aims were to evaluate specific FLPCR clinical and pathological features to gain insights into disease pathogenesis. Signalment, clinical signs, serology and 47 pin. h biopsies were retrospectively collected from 33 FLPCR and 3 normal cats. Microscopical lesions and immunohistochemistry results utilizing anti-CD3, anti-CD20, anti-FOXP3, anti-feline-IgA, IgG, IgE and anti-FeLV (p27 and gp70), FIV, FCV and, FHV were scored and most were analyzed statistically. The majority of cats were domestic short haired (26/31) with median age of 11 years and a 0.35 F/M ratio. Serology evidenced 3/22 FIV and 1/22 FeLV positive cats. Immunohistochemistry evidenced 1/33 FeLV-p27 positive cats. Common clinical signs were sneezing (19/24 [79 %]), mucous discharge (13/24 [54 %]) and stertor (10/24 [42 %]). In normal tissues, IgAs were expressed in mucin, apical and lateral cell membrane of columnar cells and in periglandular plasma cells. IgGs were expressed in 20-30 % of columnar cells. Number of clinical signs was statistically significantly higher in female cats (p < 0.0001) and was significantly correlated with chronicity (p = 0.004), and IgG scores (p = 0.01). LPC severity scores correlated positively with infiltration of neutrophils (p = 0.015), gland destruction (p = 0.019) and angiogenesis (p = 0.016) and negatively with fibrosis (p < 0.0001). LPC severity scores were also significantly associated to female sex (p = 0.01) and to IgA (p = 0.03), with higher IgA scores associated to lower LPC scores. FLPCR associated to disruption of mucosal defense mechanisms generating cycles of tissue inflammation, tissue damage and repair with progressive loss of function independent from viral infections.
Collapse
|
62
|
Lei TY, Ye YZ, Zhu XQ, Smerin D, Gu LJ, Xiong XX, Zhang HF, Jian ZH. The immune response of T cells and therapeutic targets related to regulating the levels of T helper cells after ischaemic stroke. J Neuroinflammation 2021; 18:25. [PMID: 33461586 PMCID: PMC7814595 DOI: 10.1186/s12974-020-02057-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/09/2020] [Indexed: 12/21/2022] Open
Abstract
Through considerable effort in research and clinical studies, the immune system has been identified as a participant in the onset and progression of brain injury after ischaemic stroke. Due to the involvement of all types of immune cells, the roles of the immune system in stroke pathology and associated effects are complicated. Past research concentrated on the functions of monocytes and neutrophils in the pathogenesis of ischaemic stroke and tried to demonstrate the mechanisms of tissue injury and protection involving these immune cells. Within the past several years, an increasing number of studies have elucidated the vital functions of T cells in the innate and adaptive immune responses in both the acute and chronic phases of ischaemic stroke. Recently, the phenotypes of T cells with proinflammatory or anti-inflammatory function have been demonstrated in detail. T cells with distinctive phenotypes can also influence cerebral inflammation through various pathways, such as regulating the immune response, interacting with brain-resident immune cells and modulating neurogenesis and angiogenesis during different phases following stroke. In view of the limited treatment options available following stroke other than tissue plasminogen activator therapy, understanding the function of immune responses, especially T cell responses, in the post-stroke recovery period can provide a new therapeutic direction. Here, we discuss the different functions and temporal evolution of T cells with different phenotypes during the acute and chronic phases of ischaemic stroke. We suggest that modulating the balance between the proinflammatory and anti-inflammatory functions of T cells with distinct phenotypes may become a potential therapeutic approach that reduces the mortality and improves the functional outcomes and prognosis of patients suffering from ischaemic stroke.
Collapse
Affiliation(s)
- Tian-Yu Lei
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Ying-Ze Ye
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Xi-Qun Zhu
- Department of Head and Neck and Neurosurgery, Hubei Cancer Hospital, Wuhan, 430079, Hubei Province, People's Republic of China
| | - Daniel Smerin
- University of Central Florida College of Medicine, Orlando, FL, 32827, USA
| | - Li-Juan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Xiao-Xing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Hong-Fei Zhang
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| | - Zhi-Hong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China.
| |
Collapse
|
63
|
Efficacy of Feed-Based Formalin-Killed Vaccine of Streptococcus iniae Stimulates the Gut-Associated Lymphoid Tissues and Immune Response of Red Hybrid Tilapia. Vaccines (Basel) 2021; 9:vaccines9010051. [PMID: 33466950 PMCID: PMC7830294 DOI: 10.3390/vaccines9010051] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/14/2022] Open
Abstract
Red hybrid tilapia were fed a formalin-killed oral Streptococcus iniae vaccine (FKV) in the present study was assessed. Three hundred Red hybrid tilapia 80 ± 10 g were divided into five groups (1A, 1B, 2A, 2B, and Cx), each consisting of 60 fish. Fish from Groups 1A, 1B, 2A, and 2B were fed with FKV over different periods of administration, while Group 2B was the only group of fish to receive an oral booster vaccination on day 14- and 21-days post-vaccination (dpv). Group Cx was fed with normal pellets containing no vaccine as a control group. At four weeks post-vaccination (wpv), all fish were experimentally infected with S. iniae. Groups 2A and 2B had the lowest level of mortalities following vaccination (45% and 30%, respectively) compared to Groups 1A and 1B (80% and 55%, respectively), while the level of mortalities in Group Cx was 100%. All vaccinated groups showed a significant increase in anti-S. iniae IgM levels (p < 0.05) in serum, mucus, and gut-lavage, while Group Cx did not (p > 0.05) and all fish in this group died by five weeks post-infection. In conclusion, fish fed with the S. iniae FKV had a greater level of protection against S. iniae, with increased specific antibody response to the vaccine and there was also evidence of GALT stimulation by the vaccine.
Collapse
|
64
|
Neamah WH, Busbee PB, Alghetaa H, Abdulla OA, Nagarkatti M, Nagarkatti P. AhR Activation Leads to Alterations in the Gut Microbiome with Consequent Effect on Induction of Myeloid Derived Suppressor Cells in a CXCR2-Dependent Manner. Int J Mol Sci 2020; 21:ijms21249613. [PMID: 33348596 PMCID: PMC7767008 DOI: 10.3390/ijms21249613] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/27/2020] [Accepted: 12/12/2020] [Indexed: 02/06/2023] Open
Abstract
Aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a potent ligand for AhR and a known carcinogen. While AhR activation by TCDD leads to significant immunosuppression, how this translates into carcinogenic signal is unclear. Recently, we demonstrated that activation of AhR by TCDD in naïve C57BL6 mice leads to massive induction of myeloid derived-suppressor cells (MDSCs). In the current study, we investigated the role of the gut microbiota in TCDD-mediated MDSC induction. TCDD caused significant alterations in the gut microbiome, such as increases in Prevotella and Lactobacillus, while decreasing Sutterella and Bacteroides. Fecal transplants from TCDD-treated donor mice into antibiotic-treated mice induced MDSCs and increased regulatory T-cells (Tregs). Injecting TCDD directly into antibiotic-treated mice also induced MDSCs, although to a lesser extent. These data suggested that TCDD-induced dysbiosis plays a critical role in MDSC induction. Interestingly, treatment with TCDD led to induction of MDSCs in the colon and undetectable levels of cysteine. MDSCs suppressed T cell proliferation while reconstitution with cysteine restored this response. Lastly, blocking CXC chemokine receptor 2 (CXCR2) impeded TCDD-mediated MDSC induction. Our data demonstrate that AhR activation by TCDD triggers dysbiosis which, in turn, regulates, at least in part, induction of MDSCs.
Collapse
|
65
|
Feng X, Li L, Feng J, He W, Li N, Shi T, Jie Z, Su X. Vagal-α7nAChR signaling attenuates allergic asthma responses and facilitates asthma tolerance by regulating inflammatory group 2 innate lymphoid cells. Immunol Cell Biol 2020; 99:206-222. [PMID: 32893406 DOI: 10.1111/imcb.12400] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/12/2020] [Accepted: 09/04/2020] [Indexed: 12/20/2022]
Abstract
Disorders of immune tolerance may lead to allergic asthma. Group 2 innate lymphoid cells (ILC2s) and inflammatory ILC2s (iILC2s) are key players in asthma. The vagus nerve innervating the airways releases acetylcholine or neuropeptides (i.e. calcitonin gene-related peptide) via pulmonary C-fibers (PCFs), which could regulate ILC2 activity upon binding the α7 nicotinic acetylcholine receptor (α7nAChR, coded by Chrna7) or neuropeptide receptors. Whether and how α7nAChR and PCFs regulate asthma and the formation of asthma tolerance via ILC2s or iILC2s are poorly understood. We used vagotomized, PCF degeneration and Chrna7 knockout mice to investigate ovalbumin (OVA)-induced asthma and oral OVA feeding-induced asthma tolerance. Our results revealed that vagotomy could generally suppress lung ILC2s and iILC2s, which mitigated allergic asthma responses but disrupted asthmatic tolerance. Removal of neuropeptides by PCF degeneration also reduced lung ILC2s and iILC2s, attenuating asthma responses, but did not affect asthma tolerance. In comparison, deletion of Chrna7 increased resident ILC2s and trafficking iILC2s in the lung, worsened allergic inflammation and disrupted oral tolerance. Mechanistically, deletion of Chrna7 in asthma-tolerant conditions upregulated T helper 2 cytokine- (Il4, Il13 and Il25) and sphingosine-1-phosphate (S1P)-related genes (S1pr1 and Sphk1). Blockade of S1P reduced iILC2 recruitment into asthmatic lungs. Our work is the first to demonstrate that vagal-α7nAChR signaling engaging with iILC2s and S1P not only alleviates asthma but also facilitates asthma tolerance. These findings may provide a novel therapeutic target for attenuating asthma by enhancing asthmatic tolerance.
Collapse
Affiliation(s)
- Xintong Feng
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Ling Li
- Unit of Respiratory Infection and Immunity, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Jingjing Feng
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Wei He
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Na Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Tianyun Shi
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Zhijun Jie
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Xiao Su
- Unit of Respiratory Infection and Immunity, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
66
|
Wróblewska B, Kaliszewska-Suchodoła A, Fuc E, Markiewicz LH, Ogrodowczyk AM, Złotkowska D, Wasilewska E. Effect of Low-Immunogenic Yogurt Drinks and Probiotic Bacteria on Immunoreactivity of Cow's Milk Proteins and Tolerance Induction-In Vitro and In Vivo Studies. Nutrients 2020; 12:E3390. [PMID: 33158132 PMCID: PMC7694189 DOI: 10.3390/nu12113390] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/28/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
There is no effective therapy for milk allergy. The role of lactic acid bacteria (LAB) and probiotics in protection against allergy-related outcomes is still under investigation. The aim of the study was to evaluate the immunomodulative and therapeutic potential of yogurt drinks in cow's milk allergy (CMA) management. We compared immunoreactivity of α-casein (α-CN), β-casein (β-CN), κ-casein (κ-CN), α-lactalbumin (α-LA), and β-lactoglobulin (β-LG) in 27 yogurt drinks fermented with different basic yogurt cultures, or yogurt cultures enriched with Lactobacillus plantarum and/or Bifidobacterium lactis strains, by competitive ELISA assay. Drinks with the lowest antigenic potential were used as allergoids for CMA therapy. BALB/c mice were sensitized via intraperitoneal injection of α-CN + β-LG mixture with aluminum adjuvant, and gavaged with increasing doses of selected low-immunogenic drinks (YM-basic, or YM-LB-enriched with L. plantarum and B. lactis) to induce tolerance. Milk- or phosphate-buffered saline (PBS)-dosed mice served as controls. Compared to milk, the immunoreactivity of proteins in drinks increased or decreased, depending on the bacterial sets applied for fermentation. Only a few sets acted synergistically in reducing immunoreactivity. The selected low-immunogenic drinks stimulated allergic mice for profiling Th2 to Th1 response and acquire tolerance, and the effect was greater with YM-LB drink, which during long-lasting interventional feeding strongly increased the secretion of regulatory cytokines, i.e., IL-10 and TGF-β, and IgA and decreased IL-4, IgE, and anti-(α-CN + β-LG) IgG1. The studies revealed variations in the potency of yogurt bacteria to change allergenicity of milk proteins and the need for their strict selection to obtain a safe product for allergy sufferers. The YM-LB drink with reduced antigenic potential may be a source of allergoids used in the immunotherapy of IgE mediated CMA, but further clinical or volunteer studies are required.
Collapse
Affiliation(s)
- Barbara Wróblewska
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10 Str., 10-748 Olsztyn, Poland; (B.W.); (E.F.); (L.H.M.); (A.M.O.); (D.Z.)
| | | | - Ewa Fuc
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10 Str., 10-748 Olsztyn, Poland; (B.W.); (E.F.); (L.H.M.); (A.M.O.); (D.Z.)
| | - Lidia Hanna Markiewicz
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10 Str., 10-748 Olsztyn, Poland; (B.W.); (E.F.); (L.H.M.); (A.M.O.); (D.Z.)
| | - Anna Maria Ogrodowczyk
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10 Str., 10-748 Olsztyn, Poland; (B.W.); (E.F.); (L.H.M.); (A.M.O.); (D.Z.)
| | - Dagmara Złotkowska
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10 Str., 10-748 Olsztyn, Poland; (B.W.); (E.F.); (L.H.M.); (A.M.O.); (D.Z.)
| | - Ewa Wasilewska
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10 Str., 10-748 Olsztyn, Poland; (B.W.); (E.F.); (L.H.M.); (A.M.O.); (D.Z.)
| |
Collapse
|
67
|
Sosa AC, Kariuki B, Gan Q, Knutsen AP, Bellone CJ, Guzmán MA, Barrera LA, Tomatsu S, Chauhan AK, Armbrecht E, Montaño AM. Oral immunotherapy tolerizes mice to enzyme replacement therapy for Morquio A syndrome. J Clin Invest 2020; 130:1288-1300. [PMID: 31743109 DOI: 10.1172/jci125607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 11/13/2019] [Indexed: 12/14/2022] Open
Abstract
Immune response to therapeutic enzymes poses a detriment to patient safety and treatment outcome. Enzyme replacement therapy (ERT) is a standard therapeutic option for some types of mucopolysaccharidoses, including Morquio A syndrome caused by N-acetylgalactosamine-6-sulfate sulfatase (GALNS) deficiency. Current protocols tolerize patients using cytotoxic immunosuppressives, which can cause adverse effects. Here we show development of tolerance in Morquio A mice via oral delivery of peptide or GALNS for 10 days prior to ERT. Our results show that using an immunodominant peptide (I10) or the complete GALNS enzyme to orally induce tolerance to GALNS prior to ERT resulted in several improvements to ERT in mice: (a) decreased splenocyte proliferation after in vitro GALNS stimulation, (b) modulation of the cytokine secretion profile, (c) decrease in GALNS-specific IgG or IgE in plasma, (d) decreased GAG storage in liver, and (e) fewer circulating immune complexes in plasma. This model could be extrapolated to other lysosomal storage disorders in which immune response hinders ERT.
Collapse
Affiliation(s)
- Angela C Sosa
- Department of Pediatrics, Division of Medical Genetics, School of Medicine, Saint Louis University, St. Louis, Missouri, USA.,Instituto de Errores Innatos del Metabolismo, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Barbara Kariuki
- Department of Pediatrics, Division of Allergy and Immunology
| | - Qi Gan
- Department of Pediatrics, Division of Medical Genetics, School of Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Alan P Knutsen
- Department of Pediatrics, Division of Allergy and Immunology
| | | | - Miguel A Guzmán
- Department of Pathology, School of Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Luis A Barrera
- Instituto de Errores Innatos del Metabolismo, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware, USA
| | - Anil K Chauhan
- Department of Internal Medicine, Division of Rheumatology, School of Medicine
| | | | - Adriana M Montaño
- Department of Pediatrics, Division of Medical Genetics, School of Medicine, Saint Louis University, St. Louis, Missouri, USA.,Department of Biochemistry and Molecular Biology, School of Medicine, Saint Louis University, St. Louis, Missouri, USA
| |
Collapse
|
68
|
Shang L, Deng D, Roffel S, Gibbs S. Differential influence of Streptococcus mitis on host response to metals in reconstructed human skin and oral mucosa. Contact Dermatitis 2020; 83:347-360. [PMID: 32677222 PMCID: PMC7693211 DOI: 10.1111/cod.13668] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/10/2020] [Accepted: 07/15/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Skin and oral mucosa are continuously exposed to potential metal sensitizers while hosting abundant microbes, which may influence the host response to sensitizers. This host response may also be influenced by the route of exposure that is skin or oral mucosa, due to their different immune properties. OBJECTIVE Determine how commensal Streptococcus mitis influences the host response to nickel sulfate (sensitizer) and titanium(IV) bis(ammonium lactato)dihydroxide (questionable sensitizer) in reconstructed human skin (RHS) and gingiva (RHG). METHODS RHS/RHG was exposed to nickel or titanium, in the presence or absence of S. mitis for 24 hours. Histology, cytokine secretion, and Toll-like receptors (TLRs) expression were assessed. RESULTS S. mitis increased interleukin (IL)-6, CXCL8, CCL2, CCL5, and CCL20 secretion in RHS but not in RHG; co-application with nickel further increased cytokine secretion. In contrast, titanium suppressed S. mitis-induced cytokine secretion in RHS and had no influence on RHG. S. mitis and metals differentially regulated TLR1 and TLR4 in RHS, and predominantly TLR4 in RHG. CONCLUSION Co-exposure of S. mitis and nickel resulted in a more potent innate immune response in RHS than in RHG, whereas titanium remained inert. These results indicate the important influence of commensal microbes and the route of exposure on the host's response to metals.
Collapse
Affiliation(s)
- Lin Shang
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA)University of Amsterdam and Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA)University of Amsterdam and Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Sanne Roffel
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA)University of Amsterdam and Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Susan Gibbs
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA)University of Amsterdam and Vrije Universiteit AmsterdamAmsterdamThe Netherlands
- Department of Molecular Cell Biology and ImmunologyAmsterdam UMC, Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
69
|
Mast Cell Biology and Linkages for Non-clonal Mast Cell Activation and Autoimmune/Inflammatory Syndrome Induced by Adjuvants. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s42399-020-00494-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
70
|
Gusmao-Silva G, Aguiar SLF, Miranda MCG, Guimarães MA, Alves JL, Vieira AT, Cara DC, Miyoshi A, Azevedo VA, Oliveira RP, Faria AMC. Hsp65-Producing Lactococcocus lactis Prevents Antigen-Induced Arthritis in Mice. Front Immunol 2020; 11:562905. [PMID: 33072101 PMCID: PMC7538670 DOI: 10.3389/fimmu.2020.562905] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 08/18/2020] [Indexed: 01/22/2023] Open
Abstract
Oral tolerance is the physiological process that enables the immune system to differentiate between harmless dietary and microbiota antigens from pathogen derived antigens. It develops at the mucosal surfaces and can result in local and systemic regulatory and anti-inflammatory effects. Translation of these benefits to the clinical practice faces limitations involving specificity and doses of antigen as well as regimens of feeding. To circumvent these problems, we developed a recombinant Hsp65 delivered by the acid lactic bacteria Lactococcus lactis NCDO 2118 directy in the intestinal mucosa. Hsp65 is a ubiquitous protein overexpressed in inflamed tissues and capable of inducing immunoregulatory mechanisms. L. lactis has probiotic properties and is commonly and safely used in dairy products. In this study, we showed that continuous delivery of HSP65 in the gut mucosa by L. lactis is a potent tolerogenic stimulus inducing regulatory CD4+LAP+ T cells that prevented collagen-induced and methylated bovine serum albumin-induced arthritis in mice. Clinical and histological signs of arthritis were inhibited as well as levels of inflammatory cytokines such as IL-17 and IFN-γ, serum titers of anti-collagen antibodies and rheumatoid factor. Oral administration of L. lactis induced alterations in microbiota composition toward an increased abundance of anaerobic bacteria such as Bifidobacterium and Lactobacillus. Tolerance to HSP65 and arthritis prevention induced by the recombinant L. lactis was associated with increase in IL-10 production by B cells and it was dependent on LAP+ T cells, IL-10 and TLR2 signaling. Therefore, HSP65-producing treatment induced effective tolerance and prevented arthritis development suggesting it can be used as a therapeutic tool for autoimmune diseases.
Collapse
Affiliation(s)
- Guilherme Gusmao-Silva
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Sarah Leão Fiorini Aguiar
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Mauro Andrade Guimarães
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juliana Lima Alves
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Angélica Thomaz Vieira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Denise Carmona Cara
- Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Anderson Miyoshi
- Departamento de Genética, Evolução e Ecologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vasco Ariston Azevedo
- Departamento de Genética, Evolução e Ecologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Ana Maria Caetano Faria
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Instituto de Investigação em Imunologia, São Paulo, Brazil
| |
Collapse
|
71
|
Suprabasin-null mice retain skin barrier function and show high contact hypersensitivity to nickel upon oral nickel loading. Sci Rep 2020; 10:14559. [PMID: 32884021 PMCID: PMC7471289 DOI: 10.1038/s41598-020-71536-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 08/06/2020] [Indexed: 12/22/2022] Open
Abstract
Suprabasin (SBSN) is expressed not only in epidermis but also in epithelial cells of the upper digestive tract where metals such as nickel are absorbed. We have recently shown that SBSN level is decreased in the stratum corneum and serum of atopic dermatitis (AD) patients, especially in intrinsic AD, which is characterized by metal allergy. By using SBSN-null (Sbsn–/–) mice, this study was conducted to investigate the outcome of SBSN deficiency in relation to AD. Sbsn–/– mice exhibited skin barrier dysfunction on embryonic day 16.5, but after birth, their barrier function was not perturbed despite the presence of ultrastructural changes in stratum corneum and keratohyalin granules. Sbsn–/– mice showed a comparable ovalbumin-specific skin immune response to wild type (WT) mice and rather lower contact hypersensitivity (CHS) responses to haptens than did WT mice. The blood nickel level after oral feeding of nickel was significantly higher in Sbsn–/– mice than in WT mice, and CHS to nickel was elevated in Sbsn–/– mice under nickel-loading condition. Our study suggests that the completely SBSN deficient mice retain normal barrier function, but harbor abnormal upper digestive tract epithelium that promotes nickel absorption and high CHS to nickel, sharing the features of intrinsic AD.
Collapse
|
72
|
Abstract
The understanding of the pathogenesis of any disease is the key to effective and specific treatment of the disease. immunoglobulin A (IgA) nephropathy is an autoimmune disease of the kidney. Oxford MEST classification is commonly used to stratify patients according to the severity of the disease. Patients with IgA nephropathy seem to produce anti-GalNAc antibodies against a particularly defective IgA1. This immune complex deposits in the kidneys, leading to a type 3 hypersensitivity reaction which ultimately damages the kidneys. People of a certain genetic background and who experience upregulation of certain defective receptors seem to develop primary IgA nephropathy. Secondary IgA nephropathy could be due to dysbiosis of the microbiota in the gut, compromised gut immunity or other gut pathologies, pulmonary function abnormalities, or amyloidosis. Overproduction of IgA due to plasma cell dyscrasia or reduced clearance of IgA due to liver abnormalities could also be potential causes. Genes that predispose individuals to IgA nephropathy and intestinal abnormalities, such as Celiac disease, seem to overlap and these people tend to have a poorer prognosis and need to be placed on more intensive treatment regimens. IgA Vasculitis seems to be a systemic form of IgA nephropathy, whereby IgA deposits systemically and leads to multiple disease manifestations. Patients in high-risk groups could also be prophylactically screened for the disease and closely monitored by immunohistochemical methods such as an enzyme-linked immunosorbent assay (ELISA) or identified by genetic testing. Currently, the major treatment regimens involve supportive therapy or immunosuppressive therapy which has major side effects. More specific treatment methods such as monoclonal antibodies, immunoglobulin replacement therapy, or low-antigen-content diet could also be looked into as potential treatment options. Stem cell replacement, by way of bone marrow transplant and tonsillectomy, has been suggested as a treatment option in patients with indications.
Collapse
Affiliation(s)
- Jemima C Stanley
- Pathology, Zhejiang University School of Medicine, Hangzhou, CHN
| | - Hong Deng
- Pathology, Zhejiang University School of Medicine, Hangzhou, CHN
| |
Collapse
|
73
|
Biram A, Shulman Z. T cell help to B cells: Cognate and atypical interactions in peripheral and intestinal lymphoid tissues. Immunol Rev 2020; 296:36-47. [PMID: 32557712 DOI: 10.1111/imr.12890] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022]
Abstract
Enduring immunity against harmful pathogens depends on the generation of immunological memory. Serum immunoglobulins are constantly secreted by long-lived antibody-producing cells, which provide extended protection from recurrent exposures. These cells originate mainly from germinal center structures, wherein B cells introduce mutations to their immunoglobulin genes followed by affinity-based selection. Generation of high-affinity antibodies relies on physical contacts between T and B cells, a process that facilitates the delivery of fate decision signals. T-B cellular engagements are mediated through interactions between the T cell receptor and its cognate peptide presented on B cell major histocompatibility class II molecules. Here, we describe the cellular and molecular aspects of these cognate T-B interactions, and highlight exceptional cases, especially those arising at intestinal lymphoid organs, at which T cells provide help to B cells in an atypical manner, independent of T cell specificity.
Collapse
Affiliation(s)
- Adi Biram
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Ziv Shulman
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
74
|
Immune Profile of the Nasal Mucosa in Patients with Cutaneous Leishmaniasis. Infect Immun 2020; 88:IAI.00881-19. [PMID: 32094254 DOI: 10.1128/iai.00881-19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 02/10/2020] [Indexed: 11/20/2022] Open
Abstract
Localized skin lesions are characteristic of cutaneous leishmaniasis (CL); however, Leishmania (Viannia) species, which are responsible for most CL cases in the Americas, can spread systemically, sometimes resulting in mucosal disease. Detection of Leishmania has been documented in healthy mucosal tissues (conjunctiva, tonsils, and nasal mucosa) and healthy skin of CL patients and in individuals with asymptomatic infection in areas of endemicity of L (V) panamensis and L (V) braziliensis transmission. However, the conditions and mechanisms that favor parasite persistence in healthy mucosal tissues are unknown. In this descriptive study, we compared the cell populations of the nasal mucosa (NM) of healthy donors and patients with active CL and explored the immune gene expression signatures related to molecular detection of Leishmania in this tissue in the absence of clinical signs or symptoms of mucosal disease. The cellular composition and gene expression profiles of NM samples from active CL patients were similar to those of healthy volunteers, with a predominance of epithelial over immune cells, and within the CD45+ cell population, a higher frequency of CD66b+ followed by CD14+ and CD3+ cells. In CL patients with molecular evidence of Leishmania persistence in the NM, genes characteristic of an anti-inflammatory and tissue repair responses (IL4R, IL5RA, POSTN, and SATB1) were overexpressed relative to NM samples from CL patients in which Leishmania was not detected. Here, we report the first immunological description of subclinically infected NM tissues of CL patients and provide evidence of a local anti-inflammatory environment favoring parasite persistence in the NM.
Collapse
|
75
|
Zhu M, Ma Y, Tan K, Zhang L, Wang Z, Li Y, Chen Y, Guo J, Yan G, Qi Z. Thalidomide with blockade of co-stimulatory molecules prolongs the survival of alloantigen-primed mice with cardiac allografts. BMC Immunol 2020; 21:19. [PMID: 32299357 PMCID: PMC7164359 DOI: 10.1186/s12865-020-00352-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 04/07/2020] [Indexed: 01/20/2023] Open
Abstract
Background Miscellaneous memory cell populations that exist before organ transplantation are crucial barriers to transplantation. In the present study, we used a skin-primed heart transplantation model in mouse to evaluate the abilities of Thalidomide (TD), alone or in combination with co-stimulatory blockade, using monoclonal antibodies (mAbs) against memory T cells and alloantibodies to prolong the second cardiac survival. Results In the skin-primed heart transplantation model, TD combined with mAbs significantly prolonged the second cardiac survival, accompanied by inhibition of memory CD8+ T cells. This combined treatment enhanced the CD4+Foxp3+ regulatory T cells ratio in the spleen, restrained the infiltration of lymphocytes into the allograft, and suppressed the allo-response of spleen T cells in the recipient. The levels of allo-antibodies also decreased in the recipient serum. In addition, we detected low levels of the constitutions of the lytic machinery of cytotoxic cells, which cause allograft damage. Conclusions Our study indicated a potential synergistic action of TD in combination with with mAbs to suppress the function of memory T cells and increase the survival of second allografts in alloantigen-primed mice.
Collapse
Affiliation(s)
- Maoshu Zhu
- Xiang'an Branch, The First Affiliated Hospital of Xiamen University, Xiamen, 361100, Fujian, China.,The Fifth Hospital of Xiamen, Xiamen, 361100, Fujian, China
| | - Yunhan Ma
- Organ Transplantation institute, School of Medicine, Xiamen University, Xiamen, 361100, Fujian, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, 361100, Fujian, China
| | - Kai Tan
- Grade 2015 Clinical Medicine, Fuzhou Medical College of Nanchang University, Fuzhou, 344000, Jiangxi, China
| | - Liyi Zhang
- Organ Transplantation institute, School of Medicine, Xiamen University, Xiamen, 361100, Fujian, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, 361100, Fujian, China
| | - Zhaowei Wang
- Xiang'an Branch, The First Affiliated Hospital of Xiamen University, Xiamen, 361100, Fujian, China.,The Fifth Hospital of Xiamen, Xiamen, 361100, Fujian, China
| | - Yongsheng Li
- Xiang'an Branch, The First Affiliated Hospital of Xiamen University, Xiamen, 361100, Fujian, China.,The Fifth Hospital of Xiamen, Xiamen, 361100, Fujian, China
| | - Yingyu Chen
- Organ Transplantation institute, School of Medicine, Xiamen University, Xiamen, 361100, Fujian, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, 361100, Fujian, China
| | - Junjun Guo
- Organ Transplantation institute, School of Medicine, Xiamen University, Xiamen, 361100, Fujian, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, 361100, Fujian, China
| | - Guoliang Yan
- Organ Transplantation institute, School of Medicine, Xiamen University, Xiamen, 361100, Fujian, China. .,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, 361100, Fujian, China.
| | - Zhongquan Qi
- Organ Transplantation institute, School of Medicine, Xiamen University, Xiamen, 361100, Fujian, China. .,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, 361100, Fujian, China. .,School of Medicine, Guangxi University, Nanning, 530004, Guangxi, China.
| |
Collapse
|
76
|
Vojdani A. Reaction of food‐specific antibodies with different tissue antigens. Int J Food Sci Technol 2020. [DOI: 10.1111/ijfs.14467] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Aristo Vojdani
- Immunosciences Lab., Inc. 822 S. Robertson Blvd., Ste. 312 Los Angeles CA 90035 USA
- Department of Preventive Medicine Loma Linda University School of Medicine 24785 Stewart St., Evans Hall, Ste. 111 Loma Linda CA 92354 USA
| |
Collapse
|
77
|
Cormier M, Batty P, Tarrant J, Lillicrap D. Advances in knowledge of inhibitor formation in severe haemophilia A. Br J Haematol 2020; 189:39-53. [DOI: 10.1111/bjh.16377] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Matthew Cormier
- Department of Pathology and Molecular Medicine Richardson Laboratory Queen’s University Kingston ON Canada
| | - Paul Batty
- Department of Pathology and Molecular Medicine Richardson Laboratory Queen’s University Kingston ON Canada
| | - Julie Tarrant
- Department of Pathology and Molecular Medicine Richardson Laboratory Queen’s University Kingston ON Canada
| | - David Lillicrap
- Department of Pathology and Molecular Medicine Richardson Laboratory Queen’s University Kingston ON Canada
| |
Collapse
|
78
|
Miqdady M, Al Mistarihi J, Azaz A, Rawat D. Prebiotics in the Infant Microbiome: The Past, Present, and Future. Pediatr Gastroenterol Hepatol Nutr 2020; 23:1-14. [PMID: 31988871 PMCID: PMC6966216 DOI: 10.5223/pghn.2020.23.1.1] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 12/23/2019] [Indexed: 12/26/2022] Open
Abstract
The latest definition of a prebiotic is "a substrate that is selectively utilized by host microorganisms conferring a health benefit"; it now includes non-food elements and is applicable to extra-intestinal tissues. Prebiotics are recognized as a promising tool in the promotion of general health and in the prevention and treatment of numerous juvenile diseases. Prebiotics are considered an immunoactive agent, with the potential for long-lasting effects extending past active administration of the prebiotic. Because of its extremely low risk of serious adverse effects, ease of administration, and strong potential for influencing the composition and function of the microbiota in the gut and beyond, the beneficial clinical applications of prebiotics are expanding. Prebiotics are the third largest component of human breast milk. Preparations including galactooligosaccharides (GOS), fructooligosaccharides (FOS), 2'-fucosyllactose, lacto-N-neo-tetraose are examples of commonly used and studied products for supplementation in baby formula. In particular, the GOS/FOS combination is the most studied. Maintaining a healthy microbiome is essential to promote homeostasis of the gut and other organs. With more than 1,000 different microbial species in the gut, it is likely more feasible to modify the gut microbiota through the use of certain prebiotic mixtures rather than supplementing with a particular probiotic strain. In this review, we discuss the latest clinical evidence regarding prebiotics and its role in gut immunity, allergy, infections, inflammation, and functional gastrointestinal disorders.
Collapse
Affiliation(s)
- Mohamad Miqdady
- Department of Pediatric, Sheikh Khalifa Medical City, Abu Dhabi, UAE
| | | | - Amer Azaz
- Department of Pediatric, Sheikh Khalifa Medical City, Abu Dhabi, UAE
| | - David Rawat
- Department of Pediatric, Sheikh Khalifa Medical City, Abu Dhabi, UAE
| |
Collapse
|
79
|
Rivillas-Reyes JF, Hernández-Durán EF, Morales-Manrique JL, Rivillas MP, Lozano-Marquez E, Lozano-Marquez H. Inducción de tolerancia por vía oral en trasplante de órganos y tejidos. Revisión de la Literatura. REVISTA DE LA FACULTAD DE MEDICINA 2020. [DOI: 10.15446/revfacmed.v68n1.72108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Introducción. La tolerancia oral es la supresión de la respuesta inmune a antígenos administrados con anterioridad por vía oral; su inducción tiene el propósito de evitar el uso de fármacos inmunosupresores, los cuales, dado que son poco específicos a antígenos, vuelven al huésped más susceptible de contraer infecciones y desarrollar neoplasias.Objetivos. Realizar una revisión de la literatura sobre los referentes teóricos más relevantes de la inducción de a tolerancia oral en lo que respecta al trasplante de órganos y tejidos para demostrar que el uso de esta alternativa terapéutica es viable en pacientes trasplantados.Materiales y métodos. Se realizó una revisión de la literatura en PubMed, MEDLINE, LILACS y Embase mediante la siguiente estrategia de búsqueda: periodo de publicación: sin límites; idiomas: Inglés y Español; tipo de artículos: estudios caso-control, revisiones sistemáticas y de la literatura; términos de búsqueda: “T-Lymphocytes, Regulatory”, “Autoimmunity”, Immunosuppression”, “Immune system” and “Immune Tolerance”, y sus equivalentes en español.Resultados. La búsqueda inicial arrojó 719 registros, sin embargo solo 99 abordaban la inducción de la tolerancia oral. Una vez los registros duplicados y los artículos sin acceso a texto completo fueron removidos, se incluyeron 72 estudios en la revisión.Conclusiones. La administración oral de antígenos es una opción efectiva para inducir tolerancia inmunológica en pacientes trasplantados (modelos murinos), pues elimina los efectos adversos que conlleva la terapia inmunosupresora actualmente utilizada.
Collapse
|
80
|
Wilson HL, Gerdts V, Babiuk LA. Mucosal Vaccine Development for Veterinary and Aquatic Diseases. MUCOSAL VACCINES 2020. [PMCID: PMC7149622 DOI: 10.1016/b978-0-12-811924-2.00048-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Because most pathogens and food antigens enter the host via the mucosal surfaces, effective mucosal immunity is critical for maintaining homeostasis through immune regulation, tolerance, and induction of effective immune responses when needed. Thus the mucosa-associated lymphoid tissues represent an important target for vaccination. Indeed, more than 20 years of research have clearly demonstrated the benefits of mucosal vaccination versus systemic vaccination. Such benefits include local induction of secretory immunoglobulin A (SIgA) as well as activation and maturation of mucosal dendritic cells, homing of effector cells to the mucosal surfaces, expression of specific host defense peptides, and other innate effector molecules. In addition, mucosal vaccination offers the opportunity to induce colostral and lactogenic immunity during pregnancy and the possibility of avoiding neutralization of early life vaccines by maternal antibodies, both of which are critical for protecting the most susceptible from infectious diseases. Moreover, mucosal administration offers the advantage of inducing both effective systemic immunity and mucosal immunity, enhancing vaccine efficacy and providing improved protection. A number of animal vaccines are already administered via the mucosal surfaces, with many more to come over the next few years. It is gratifying to see that veterinary vaccine development has yet again taken a leadership role in exploring innovative approaches and technologies to mucosal vaccination. For the veterinary field, considerations for mucosal vaccine development and use necessarily include costs (often pennies per dose), mass delivery that preferably avoids animal restraint, and economic and trade considerations. In this chapter, we provide an overview of some of the existing vaccine technologies and discuss their advantages and disadvantages.
Collapse
|
81
|
Chan CJ, Yong YS, Song AAL, Abdul Rahim R, In LLA, Lim RLH. Lactococcus lactis harbouring Ara h 2.02 alleviates allergen-specific Th2-associated responses in sensitized mice. J Appl Microbiol 2019; 128:862-874. [PMID: 31758869 DOI: 10.1111/jam.14524] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/28/2019] [Accepted: 11/19/2019] [Indexed: 12/21/2022]
Abstract
AIM To study the prophylactic effect of recombinant Lactococcus lactis (rLl) harbouring Ara h 2.02 peanut allergen, in sensitized and challenged mice. METHODS AND RESULTS Ara h 2.02 cDNA was cloned into pNZ8048 for heterologous expression in L. lactis. The purified recombinant allergen showed IgE binding comparable with native Ara h 2. Balb/c mice were fed with either recombinant (rLl), nonrecombinant L. lactis (Ll) or NaHCO3 (Sham) prior to sensitization and challenged with rAra h 2.02, whereas the baseline group was only fed with Ll. Allergen-specific immunoglobulin and splenocyte cytokines responses were determined for each mouse. Mice fed with either Ll or rLl showed significant alleviation of IgE and IgG1 compared to the Sham group. Despite no significant decrease in Th2 (IL-4, IL-13, IL-6) or increase in Th1 (IFN-γ) cytokines, both groups showed lower IL-10 level, while the IL-4 : IFN-γ ratio was significantly lower for rLl compared to Ll group. CONCLUSIONS Oral administration of rLl harbouring Ara h 2.02 demonstrated alleviation of Th2-associated responses in allergen-challenged mice and a possible added allergen-specific prophylactic effect. SIGNIFICANCE AND IMPACT OF THE STUDY Ara h 2.02 coupled with the intrinsic properties of probiotic L. lactis as a delivery vehicle can be explored for the development of a commercially scalable vaccine.
Collapse
Affiliation(s)
- C J Chan
- Faculty of Applied Sciences, UCSI University, UCSI Heights, Cheras, Kuala Lumpur, Malaysia
| | - Y S Yong
- Faculty of Applied Sciences, UCSI University, UCSI Heights, Cheras, Kuala Lumpur, Malaysia
| | - A A L Song
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, University Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - R Abdul Rahim
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, University Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - L L A In
- Faculty of Applied Sciences, UCSI University, UCSI Heights, Cheras, Kuala Lumpur, Malaysia
| | - R L H Lim
- Faculty of Applied Sciences, UCSI University, UCSI Heights, Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
82
|
Ivancic R, Iqbal H, deSilva B, Pan Q, Matrka L. Immunological tolerance of low-risk HPV in recurrent respiratory papillomatosis. Clin Exp Immunol 2019; 199:131-142. [PMID: 31628850 DOI: 10.1111/cei.13387] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2019] [Indexed: 12/12/2022] Open
Abstract
Recurrent respiratory papillomatosis (RRP) is characterized by benign exophytic lesions of the respiratory tract caused by the human papillomavirus (HPV), in particular low-risk HPV6 and HPV11. Aggressiveness varies greatly among patients. Surgical excision is the current standard of care for RRP, with adjuvant therapy used when surgery cannot control disease recurrence. Numerous adjuvant therapies have been used to control RRP with some success, but none are curative. Current literature supports a polarization of the adaptive immune response to a T helper type 2 (Th2)-like or T regulatory phenotype, driven by a complex interplay between innate immunity, adaptive immunity and HPV6/11 proteins. Additionally, certain immunogenetic polymorphisms can predispose individuals to an HPV6/11-tolerant microenvironment. As a result, immunomodulatory efforts are being made to restore the host immune system to a more balanced T cell phenotype and clear viral infection. Literature has shown exciting evidence for the role of HPV vaccination with Gardasil or Gardasil-9 as both primary prevention, by decreasing incidence through childhood vaccinations, and secondary prevention, by treating active RRP disease. Multi-institution randomized clinical trials are needed to better assess their efficacy as treatment for active disease. Interestingly, a DNA vaccine has recently shown in-vitro success in generating a more robust CD8+ T cell response. Furthermore, clinical trials for programmed death 1 (PD-1) inhibitors are under investigation for RRP management. Molecular insights into RRP, in particular the interplay between RRP and the immune system, are needed to advance our understanding of this disease and may lead to the identification of immunomodulatory agents to better manage RRP.
Collapse
Affiliation(s)
- R Ivancic
- College of Medicine, The Ohio State University, OH, USA
| | - H Iqbal
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| | - B deSilva
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Q Pan
- Case Comprehensive Cancer Center, Cleveland, OH
| | - L Matrka
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| |
Collapse
|
83
|
Horwitz DA, Fahmy TM, Piccirillo CA, La Cava A. Rebalancing Immune Homeostasis to Treat Autoimmune Diseases. Trends Immunol 2019; 40:888-908. [PMID: 31601519 DOI: 10.1016/j.it.2019.08.003] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 08/01/2019] [Accepted: 08/14/2019] [Indexed: 12/18/2022]
Abstract
During homeostasis, interactions between tolerogenic dendritic cells (DCs), self-reactive T cells, and T regulatory cells (Tregs) contribute to maintaining mammalian immune tolerance. In response to infection, immunogenic DCs promote the generation of proinflammatory effector T cell subsets. When complex homeostatic mechanisms maintaining the balance between regulatory and effector functions become impaired, autoimmune diseases can develop. We discuss some of the newest advances on the mechanisms of physiopathologic homeostasis that can be employed to develop strategies to restore a dysregulated immune equilibrium. Some of these designs are based on selectively activating regulators of immunity and inflammation instead of broadly suppressing these processes. Promising approaches include the use of nanoparticles (NPs) to restore Treg control over self-reactive cells, aiming to achieve long-term disease remission, and potentially to prevent autoimmunity in susceptible individuals.
Collapse
Affiliation(s)
- David A Horwitz
- General Nanotherapeutics, LLC, Santa Monica, CA, USA; Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Tarek M Fahmy
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, Yale University, New Haven, CT, USA; Chemical and Environmental Engineering, School of Engineering and Applied Sciences, Yale University, New Haven, CT, USA; Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Ciriaco A Piccirillo
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada; Program in Infectious Disease and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montréal, QC, Canada; Centre of Excellence in Translational Immunology (CETI), Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Antonio La Cava
- Department of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
84
|
Steinman L, Ho PP, Robinson WH, Utz PJ, Villoslada P. Antigen-specific tolerance to self-antigens in protein replacement therapy, gene therapy and autoimmunity. Curr Opin Immunol 2019; 61:46-53. [PMID: 31476445 DOI: 10.1016/j.coi.2019.07.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 07/24/2019] [Accepted: 07/30/2019] [Indexed: 12/23/2022]
Abstract
Trials of antigen-specific tolerance have been undertaken in the clinic for over fifty years and the results of these antigen-specific clinical trials are described in this review. Antigen-specific tolerization of the immune system in protein replacement therapy for hemophilia A is an accepted treatment. Clinical trials are ongoing for autoimmune conditions such as type 1 diabetes, multiple sclerosis, neuromyelitis optica, and rheumatoid arthritis with various antigen-specific strategies. Trials for tolerization in celiac disease aim for antigen specific tolerance to gluten, an environmental trigger, which may then halt the progression to autoimmunity targeting a self-antigen, tissue transglutaminase. Although many promising approaches have been demonstrated in pre-clinical models, this review will focus primarily on clinical trials of antigen-specific tolerance that have been taken to the clinic and with initial results reported in the peer reviewed literature. A separate article on approaches with CAR-T cells appears in this volume.
Collapse
Affiliation(s)
- Lawrence Steinman
- Stanford University School of Medicine, Stanford, CA 94305, United States.
| | - Peggy P Ho
- Stanford University School of Medicine, Stanford, CA 94305, United States
| | - William H Robinson
- Stanford University School of Medicine, Stanford, CA 94305, United States
| | - Paul J Utz
- Stanford University School of Medicine, Stanford, CA 94305, United States
| | - Pablo Villoslada
- Stanford University School of Medicine, Stanford, CA 94305, United States
| |
Collapse
|
85
|
Parenky AC, Akalkotkar A, Mulla NS, D'Souza MJ. Harnessing T-cell activity against prostate cancer: A therapeutic microparticulate oral cancer vaccine. Vaccine 2019; 37:6085-6092. [PMID: 31477437 DOI: 10.1016/j.vaccine.2019.08.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 07/16/2019] [Accepted: 08/16/2019] [Indexed: 10/26/2022]
Abstract
Prostate Cancer specific immunotherapy in combination with immune stimulating adjuvants may serve as a viable strategy for facilitating tumor regression and preventing recurrence. In this study, an oral microparticulate vaccine encapsulating tumor associated antigens (TAA) extracted from a murine prostate cancer cell line, TRAMP-C2, was formulated with the help of a spray dryer. Microparticles were characterized in vitro to determine their physicochemical properties and antigenicity. Formulated microparticles had an average size of 4.92 ± 0.5 μm with a zeta potential of 7.92 ± 1.2 mV. In order to test our formulation for its ability to demonstrate adequate antigen presentation and co-stimulation, microparticles were tested in vitro on murine dendritic cells. In vitro biological characterization demonstrated the activation of specific immune system markers such as CD80/86, CD40, MHC-I and MHC-II. Following in vitro characterization, in vivo anti-tumor efficacy of the oral microparticulate vaccine was evaluated in C57BL/6 male mice. Combination therapy of vaccine microparticles with cyclophosphamide and granulocyte macrophage-colony stimulating factor (GM-CSF) demonstrated a five-fold reduction in tumor volume as compared to non-vaccinated mice. At the cellular level, cyclophosphamide and GM-CSF augmented the vaccine response as indicated by the reduced tumor volume and significant elevation of cytotoxic T-cell (CTL) CD8+ and (T-helper) CD4+ T-cells compared to mice receiving vaccine microparticles alone. Furthermore, our studies indicate a significant reduction in T-regulatory cells (T-regs) in mice receiving vaccine along with GM-CSF and cyclophosphamide, one of the immune escape mechanisms linked to tumor growth and progression. Thus, oral microparticulate vaccines have the potential to trigger a robust anti-tumor cellular response, and in combination with clinically relevant agents, significantly resist tumor growth and progression.
Collapse
Affiliation(s)
- Ashwin C Parenky
- Mercer University, Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, Atlanta, GA 30341, USA.
| | - Archana Akalkotkar
- Mercer University, Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, Atlanta, GA 30341, USA; Charles River Laboratories, Ashland, OH 44805, USA.
| | - Nihal S Mulla
- Mercer University, Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, Atlanta, GA 30341, USA; Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Drake University, 2507 University Ave, Des Moines, IA 50311, USA
| | - Martin J D'Souza
- Mercer University, Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, Atlanta, GA 30341, USA.
| |
Collapse
|
86
|
Strainic MG, Liu J, An F, Bailey E, Esposito A, Hamann J, Heeger PS, Medof ME. CD55 Is Essential for CD103 + Dendritic Cell Tolerogenic Responses that Protect against Autoimmunity. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1386-1401. [PMID: 31103439 DOI: 10.1016/j.ajpath.2019.04.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/10/2019] [Accepted: 04/01/2019] [Indexed: 12/23/2022]
Abstract
Recent studies traced inflammatory bowel disease in some patients to deficiency of CD55 [decay-accelerating factor (DAF)], but the mechanism underlying the linkage remained unclear. Herein, we studied the importance of DAF in enabling processes that program tolerance in the gut and the eye, two immune-privileged sites where immunosuppressive responses are continuously elicited. Unlike oral feeding or ocular injection of ovalbumin in wild-type (WT) mice, which induced dominant immune tolerance, identical treatment of DAF-/- mice or DAF-/- to WT bone marrow chimeras did not. While 10% to 30% of mesenteric and submandibular lymph node CD4+ cells became robust T-regulatory cells (Tregs) in WT forkhead box P3 (Foxp3)-green fluorescent protein mice, few in either site became Tregs with little suppressor activity in DAF-/- Foxp3-green fluorescent protein mice. Phenotyping of CD103+ dendritic cells (DCs) from the ovalbumin-fed DAF-/- mice showed impaired expression of inducer of costimulation (ICOS) ligand, programmed death receptor 1-ligand 1 (PD1-L1), CxxxC chemokine receptor 1 (Cx3CR1), CCR7, and CCR9. Analyses of elicited DAF-/- Foxp3+ Tregs showed reduced expression of interferon regulatory factor 8 (IRF-8)/aldehyde dehydrogenase 1 family member A2 (Aldh1a2) and glycoprotein A repetitions predominant/latency-associated protein associated with Treg transforming growth factor-β production and presentation, as well as integrin β6/integrin β8 associated with Treg and CD103+ DC transforming growth factor-β release. Thus, DAF is required for the properties of CD103+ DCs and their naïve CD4+ cell partners that together program tolerance.
Collapse
Affiliation(s)
- Michael G Strainic
- Institute of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Jinbo Liu
- Institute of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Fengqi An
- Institute of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Erin Bailey
- Institute of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Andrew Esposito
- Department of Ophthalmology, Case Western Reserve University, Cleveland, Ohio
| | - Jörg Hamann
- Department of Experimental Immunology, University of Amsterdam, Academic Medical Center, Amsterdam, the Netherlands
| | - Peter S Heeger
- Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - M Edward Medof
- Institute of Pathology, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
87
|
Rosenthal KS, Carambula R, Zimmerman DH. Why Don't We Have a Vaccine Against Autoimmune Diseases? - A Review. JOURNAL OF CLINICAL & CELLULAR IMMUNOLOGY 2019; 10:574. [PMID: 31328022 PMCID: PMC6640150 DOI: 10.4172/2155-9899.1000574] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
This review examines some of the reasons why we don't have a vaccine against autoimmune diseases and highlights the progress that has been made. Many autoimmune diseases, such as rheumatoid arthritis (RA), multiple sclerosis (MS) and type 1 diabetes (T1D), are driven by autoimmune T cell responses. Unlike vaccines for most infectious diseases, which elicit antibody responses, are intended for immuno-naive individuals and considered preventative, a vaccine for an autoimmune disease must be therapeutic and resolve or control the on-going autoimmune response and condition in the diseased host. Despite these differences, many of the same considerations for infectious disease vaccines must also be addressed to develop a therapeutic vaccine for autoimmune diseases. The disease initiator/triggers, antigens and autoantigens, nature of the immunopathogenic and protective/therapeutic immune response will be compared for infectious and autoimmune diseases as will approaches for developing vaccines including formulations, animal models and indicators of success. The rationale for a therapeutic vaccine for RA will be discussed in greater detail with a relatively limited discussion of T1D, MS and other autoimmune diseases.
Collapse
Affiliation(s)
- Ken S Rosenthal
- Roseman University College of Medicine, 10530 Discovery Dr, Las Vegas, USA
- Northeast Ohio Medical University, Rootstown, OH, USA
| | | | | |
Collapse
|
88
|
Das S, Mohakud NK, Suar M, Sahu BR. Vaccine development for enteric bacterial pathogens: Where do we stand? Pathog Dis 2019; 76:5040763. [PMID: 30052916 DOI: 10.1093/femspd/fty057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/19/2018] [Indexed: 01/06/2023] Open
Abstract
Gut infections triggered by pathogenic bacteria lead to most frequently occurring diarrhea in humans accounting for million deaths annually. Currently, only a few licensed vaccines are available against these pathogens for mostly travelers moving to diarrheal endemic areas. Besides commercialized vaccines, there are many formulations that are either under clinical or pre-clinical stages of development and despite several efforts to improve safety, immunogenicity and efficacy, none of them can confer long-term protective immunity, for which repeated booster doses are always recommended. Further in many countries, financial, social and political constraints have jeopardized vaccine development program against these pathogens that enforce us to gather knowledge on safety, tolerability, immunogenicity and protective efficacy regarding the same. In this review, we analyze safety and efficacy issues of vaccines against five major gut bacteria causing enteric infections. The article also simultaneously describes several barriers for vaccine development and further discusses possible strategies to enhance immunogenicity and efficacy.
Collapse
Affiliation(s)
- Susmita Das
- Infection Biology Lab, KIIT School of Biotechnology, Campus XI, Bhubaneswar 751024, India
| | - Nirmal K Mohakud
- Department of Pediatrics, Kalinga Institute of Medical Sciences, Patia, Bhubaneswar 751024, India
| | - Mrutyunjay Suar
- Infection Biology Lab, KIIT School of Biotechnology, Campus XI, Bhubaneswar 751024, India
| | - Bikash R Sahu
- Infection Biology Lab, KIIT School of Biotechnology, Campus XI, Bhubaneswar 751024, India
| |
Collapse
|
89
|
Dawod B, Marshall JS. Cytokines and Soluble Receptors in Breast Milk as Enhancers of Oral Tolerance Development. Front Immunol 2019; 10:16. [PMID: 30723472 PMCID: PMC6349727 DOI: 10.3389/fimmu.2019.00016] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/04/2019] [Indexed: 12/13/2022] Open
Abstract
The postpartum period is an important window during which environmental factors can shape the life-long health of the infant. This time period often coincides with substantial milk consumption either in the form of breast milk or from cow's milk sources, such as infant formulas. Although breast milk is the most beneficial source of nutrients for infants during the first 6 months after birth, its role in regulating food allergy development, through regulation of oral tolerance, is still controversial. Breast milk contains several factors that can impact mucosal immune function, including immune cells, antibodies, microbiota, oligosaccharides, cytokines, and soluble receptors. However, there is considerable variation in the assessed levels of cytokines and soluble receptors between studies and across the lactation period. Most of these cytokines and soluble receptors are absent, or only found in limited quantities, in commercial baby formulas. Differences in content of these pluripotent factors, which impact on both the mother and the neonate, could contribute to the controversy surrounding the role of breast milk regulating oral tolerance. This review highlights current knowledge about the importance of cytokines and soluble receptors in breast milk on the development of oral tolerance and tolerance-relateddisorders. Understanding the mechanisms by which such milk components might promote oral tolerance could aid in the development of improved strategies for allergy prevention.
Collapse
Affiliation(s)
- Bassel Dawod
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Jean S Marshall
- Department of Pathology, Dalhousie University, Halifax, NS, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
90
|
Abstract
Finely tuned mechanisms enable the gastrointestinal tract to break down dietary components into nutrients without mounting, in the majority of cases, a dysregulated immune or functional host response. However, adverse reactions to food have been steadily increasing, and evidence suggests that this process is environmental. Adverse food reactions can be divided according to their underlying pathophysiology into food intolerances, when, for instance, there is deficiency of a host enzyme required to digest the food component, and food sensitivities, when immune mechanisms are involved. In this Review, we discuss the clinical and experimental evidence for enteric infections and/or alterations in the gut microbiota in inciting food sensitivity. We focus on mechanisms by which microorganisms might provide direct pro-inflammatory signals to the host promoting breakdown of oral tolerance to food antigens or indirect pathways that involve the metabolism of protein antigens and other dietary components by gut microorganisms. Better understanding of these mechanisms will help in the development of preventive and therapeutic strategies for food sensitivities.
Collapse
|
91
|
Feng RY, Chen Q, Yang WJ, Tong XG, Sun ZM, Yan H. Immune Tolerance Therapy: A New Method for Treatment of Traumatic Brain Injury. Chin Med J (Engl) 2018; 131:1990-1998. [PMID: 30082532 PMCID: PMC6085845 DOI: 10.4103/0366-6999.238147] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective: Due to the special anatomical structure and pathophysiological mechanism of the central nervous system (CNS), there is a big difference between the repair of brain injury and other systems of the body. More and more evidence shows that targetedly reducing the autoimmune response of brain tissue without affecting the immune function in other parts of the body will be the best optimized treatment for brain injury. Data Sources: This review was based on data in articles published in PubMed up to June 5, 2017, with the following keywords: “immune tolerance”, “traumatic brain injury”, and “central nervous system”. Study Selection: Original articles and critical reviews on immune tolerance and brain damage were selected for this review. References of the retrieved articles were also screened to search for potentially relevant papers. Results: The CNS is isolated from the immune system through the blood-brain barrier. After brain injury, brain antigens are released into the systemic circulation to induce damaging immune responses. Immune tolerance can effectively reduce the brain edema and neurological inflammatory response after brain injury, which is beneficial to the recovery of neurological function. The clinical application prospect and theoretical research value of the treatment of immune tolerance on traumatic brain injury (TBI) is worth attention. Conclusions: The establishment of immune tolerance mechanism has a high clinical value in the treatment of TBI. It opens up new opportunities for the treatment of brain damage.
Collapse
Affiliation(s)
- Ruo-Yang Feng
- Department of Neurosurgery, Tianjin Medical University, Tianjin 300070, China
| | - Qian Chen
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases; Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Wei-Jian Yang
- Department of Neurosurgery, Tianjin Medical University, Tianjin 300070, China
| | - Xiao-Guang Tong
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Zhi-Ming Sun
- Department of Spine Surgery, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Hua Yan
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin 300350, China
| |
Collapse
|
92
|
McKim JM, Willoughby JA, Blakemore WR, Weiner ML. Clarifying the confusion between poligeenan, degraded carrageenan, and carrageenan: A review of the chemistry, nomenclature, and in vivo toxicology by the oral route. Crit Rev Food Sci Nutr 2018; 59:3054-3073. [DOI: 10.1080/10408398.2018.1481822] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
93
|
Bouziat R, Biering SB, Kouame E, Sangani KA, Kang S, Ernest JD, Varma M, Brown JJ, Urbanek K, Dermody TS, Ng A, Hinterleitner R, Hwang S, Jabri B. Murine Norovirus Infection Induces T H1 Inflammatory Responses to Dietary Antigens. Cell Host Microbe 2018; 24:677-688.e5. [PMID: 30392830 PMCID: PMC6326098 DOI: 10.1016/j.chom.2018.10.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 09/03/2018] [Accepted: 10/08/2018] [Indexed: 12/20/2022]
Abstract
Intestinal reovirus infection can trigger T helper 1 (TH1) immunity to dietary antigen, raising the question of whether other viruses can have a similar impact. Here we show that the acute CW3 strain of murine norovirus, but not the persistent CR6 strain, induces TH1 immunity to dietary antigen. This property of CW3 is dependent on its major capsid protein, a virulence determinant. Transcriptional profiling of mesenteric lymph nodes following infection reveals an immunopathological signature that does not segregate with protective immunity but with loss of oral tolerance, in which interferon regulatory factor 1 is critical. These data show that viral capacity to trigger specific inflammatory pathways at sites where T cell responses to dietary antigens take place interferes with the development of tolerance to an oral antigen. Collectively, these data provide a foundation for the development of therapeutic strategies to prevent TH1-mediated complex immune disorders triggered by viral infections.
Collapse
Affiliation(s)
- Romain Bouziat
- Department of Medicine, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Scott B Biering
- Committee on Microbiology, University of Chicago, Chicago, IL, USA
| | - Elaine Kouame
- Department of Medicine, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Kishan A Sangani
- Department of Medicine, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Soowon Kang
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Jordan D Ernest
- Department of Medicine, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Mukund Varma
- Division of Gastroenterology, Department of Medicine, Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Judy J Brown
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kelly Urbanek
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Terence S Dermody
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Aylwin Ng
- Division of Gastroenterology, Department of Medicine, Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Reinhard Hinterleitner
- Department of Medicine, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Seungmin Hwang
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Committee on Microbiology, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA.
| | - Bana Jabri
- Department of Medicine, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA; Section of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
94
|
Abbring S, Hols G, Garssen J, van Esch BCAM. Raw cow's milk consumption and allergic diseases - The potential role of bioactive whey proteins. Eur J Pharmacol 2018; 843:55-65. [PMID: 30439365 DOI: 10.1016/j.ejphar.2018.11.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/06/2018] [Accepted: 11/09/2018] [Indexed: 12/18/2022]
Abstract
The prevalence of allergic diseases has increased significantly in Western countries in the last decades. This increase is often explained by the loss of rural living conditions and associated changes in diet and lifestyle. In line with this 'hygiene hypothesis', several epidemiological studies have shown that growing up on a farm lowers the risk of developing allergic diseases. The consumption of raw, unprocessed, cow's milk seems to be one of the factors contributing to this protective effect. Recent evidence indeed shows an inverse relation between raw cow's milk consumption and the development of asthma and allergies. However, the consumption of raw milk is not recommended due to the possible contamination with pathogens. Cow's milk used for commercial purposes is therefore processed, but this milk processing is shown to abolish the allergy-protective effects of raw milk. This emphasizes the importance of understanding the components and mechanisms underlying the allergy-protective capacity of raw cow's milk. Only then, ways to produce a safe and protective milk can be developed. Since mainly heat treatment is shown to abolish the allergy-protective effects of raw cow's milk, the heat-sensitive whey protein fraction of raw milk is an often-mentioned source of the protective components. In this review, several of these whey proteins, their potential contribution to the allergy-protective effects of raw cow's milk and the consequences of heat treatment will be discussed. A better understanding of these bioactive whey proteins might eventually contribute to the development of new nutritional approaches for allergy management.
Collapse
Affiliation(s)
- Suzanne Abbring
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Gert Hols
- Danone Nutricia Research, Utrecht, the Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands; Danone Nutricia Research, Utrecht, the Netherlands
| | - Betty C A M van Esch
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands; Danone Nutricia Research, Utrecht, the Netherlands.
| |
Collapse
|
95
|
Ren Z, Zhao Y, Liu J, Ji X, Meng L, Wang T, Sun W, Zhang K, Sang X, Yu Z, Li Y, Feng N, Wang H, Yang S, Yang Z, Wang Z, Gao Y, Xia X. Inclusion of membrane-anchored LTB or flagellin protein in H5N1 virus-like particles enhances protective responses following intramuscular and oral immunization of mice. Vaccine 2018; 36:5990-5998. [PMID: 30172635 DOI: 10.1016/j.vaccine.2018.08.053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 08/10/2018] [Accepted: 08/18/2018] [Indexed: 12/24/2022]
Abstract
We previously demonstrated that intramuscular immunization with virus-like particles (VLPs) composed of the haemagglutinin (HA), neuraminidase (NA), and matrix (M1) proteins of A/meerkat/Shanghai/SH-1/2012 (clade 2.3.2.1) protected mice from lethal challenge with viruses from other H5 HPAI clades. The inclusion of additional proteins that can serve as immunological adjuvants in VLPs may enhance adaptive immune responses following vaccination, and oral vaccines may represent the safest choice. Here, we report the generation of H5N1 VLPs composed of the viral HA, NA, and M1 proteins and membrane-anchored forms of the Escherichia coli heat-labile enterotoxin B subunit protein (LTB) or the Toll-like receptor 5 ligand flagellin (Flic). Mice intramuscularly or orally immunized with VLPs containing LTB or Flic generated greater humoural and cellular immune responses than those administered H5N1 VLPs without LTB or Flic. Intramuscular immunization with VLPs protected mice from lethal challenge with homologous or heterologous H5N1 viruses irrespective of whether the VLPs additionally included LTB or Flic. In contrast, oral immunization of mice with LTB- or Flic-VLPs conferred substantial protection against lethal challenge with both homologous and heterologous H5N1 influenza viruses, whereas mice immunized orally with VLPs lacking LTB and Flic universally succumbed to infection. Mice immunized orally with LTB- or Flic-VLPs showed 10-fold higher virus-specific IgG titres than mice immunized with H5N1-VLPs lacking LTB or Flic. Collectively, these results indicate that the inclusion of immunostimulatory proteins, such as LTB and Flic, in VLP-based vaccines may represent a promising new approach for the control of current H5N1 HPAI outbreaks by eliciting higher humoural and cellular immune responses and conferring improved cross-clade protection.
Collapse
Affiliation(s)
- Zhiguang Ren
- Joint National Laboratory for Antibody Drug Engineering, Henan University, School of Basic Medical Sciences, Kaifeng, Henan Province, China; Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China; Key Lab of Cellular and Molecular Immunology, Henan University, School of Basic Medical Sciences, Kaifeng, Henan Province, China.
| | - Yongkun Zhao
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Jing Liu
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China; Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xianliang Ji
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Lingnan Meng
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Tiecheng Wang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Weiyang Sun
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Kun Zhang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Xiaoyu Sang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Zhijun Yu
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Yuanguo Li
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Na Feng
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Hualei Wang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Songtao Yang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Zhengyan Yang
- Joint National Laboratory for Antibody Drug Engineering, Henan University, School of Basic Medical Sciences, Kaifeng, Henan Province, China; Key Lab of Cellular and Molecular Immunology, Henan University, School of Basic Medical Sciences, Kaifeng, Henan Province, China
| | - Zhizeng Wang
- Joint National Laboratory for Antibody Drug Engineering, Henan University, School of Basic Medical Sciences, Kaifeng, Henan Province, China; Key Lab of Cellular and Molecular Immunology, Henan University, School of Basic Medical Sciences, Kaifeng, Henan Province, China
| | - Yuwei Gao
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu Province, China; Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, China.
| | - Xianzhu Xia
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu Province, China; Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, China.
| |
Collapse
|
96
|
Matsubara T, Iwamoto H, Nakazato Y, Okamoto T, Ehara T, Izumi H, Takeda Y. Ingestion of partially hydrolyzed whey protein suppresses epicutaneous sensitization to β-lactoglobulin in mice. Pediatr Allergy Immunol 2018. [PMID: 29518286 DOI: 10.1111/pai.12887] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Epicutaneous sensitization to food allergens can occur through defective skin barriers. However, the relationship between oral tolerance and epicutaneous sensitization remains to be elucidated. We aimed to determine whether prior oral exposure to whey proteins or their hydrolysates prevents epicutaneous sensitization and subsequent food-allergic reaction to the whey protein, β-lactoglobulin (β-LG), and investigated the underlying mechanisms. METHODS BALB/c mice were given whey protein concentrate (WPC), two kinds of partial whey protein hydrolysate (PWH1 or PWH2), or extensive whey protein hydrolysate (EWH) in drinking water for 21 days. The mice were then epicutaneously sensitized with β-LG on tape-stripped skin. Sensitization was assessed by basophil activation tests and by measuring the level of serum β-LG-specific antibodies and cytokines secreted from β-LG-restimulated spleen and mesenteric lymph node (MLN) cells. Development of an allergic reaction was assessed by monitoring body temperature and by measuring mast cell protease-1 level in plasma after the β-LG oral challenge. Activated T-cell population among β-LG-restimulated MLN cells was also analyzed. RESULTS In mice fed with WPC, PWH1, or PWH2, sensitization and the development of an allergic reaction were totally reduced. The acceleration of cytokine release from the spleen and MLN cells or T-cell activation was not evident after β-LG restimulation. In EWH-fed mice, a suppressive effect, though milder than that in WPC-, PWH1-, or PWH2-fed mice, was observed during the development of the allergic reaction. CONCLUSIONS Prior oral exposure to partially hydrolyzed whey protein prevents epicutaneous sensitization and subsequent allergic response to β-LG in mice.
Collapse
Affiliation(s)
- Takeshi Matsubara
- Wellness & Nutrition Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Hiroshi Iwamoto
- Wellness & Nutrition Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Yuki Nakazato
- Wellness & Nutrition Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Tomoyuki Okamoto
- Wellness & Nutrition Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Tatsuya Ehara
- Wellness & Nutrition Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Hirohisa Izumi
- Wellness & Nutrition Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Yasuhiro Takeda
- Wellness & Nutrition Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| |
Collapse
|
97
|
Hosseini A, Dolati S, Hashemi V, Abdollahpour‐Alitappeh M, Yousefi M. Regulatory T and T helper 17 cells: Their roles in preeclampsia. J Cell Physiol 2018; 233:6561-6573. [DOI: 10.1002/jcp.26604] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/16/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Arezoo Hosseini
- Stem Cell Research CenterTabriz University of Medical SciencesTabrizIran
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyFaculty of MedicineTabriz University of Medical SciencesTabrizIran
- Student's Research CommitteeTabriz University of Medical SciencesTabrizIran
| | - Sanam Dolati
- Stem Cell Research CenterTabriz University of Medical SciencesTabrizIran
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyFaculty of MedicineTabriz University of Medical SciencesTabrizIran
- Student's Research CommitteeTabriz University of Medical SciencesTabrizIran
| | - Vida Hashemi
- Department of Basic ScienceFaculty of MedicineMaragheh University of Medical SciencesMaraghehIran
| | - Meghdad Abdollahpour‐Alitappeh
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Mehdi Yousefi
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyFaculty of MedicineTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
98
|
|
99
|
Kim YS, Kim MN, Lee KE, Hong JY, Oh MS, Kim SY, Kim KW, Sohn MH. Activated leucocyte cell adhesion molecule (ALCAM/CD166) regulates T cell responses in a murine model of food allergy. Clin Exp Immunol 2018; 192:151-164. [PMID: 29363753 DOI: 10.1111/cei.13104] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2018] [Indexed: 12/14/2022] Open
Abstract
Food allergy is a major public health problem. Studies have shown that long-term interactions between activated leucocyte cell adhesion molecule (ALCAM/CD166) on the surface of antigen-presenting cells, and CD6, a co-stimulatory molecule, influence immune responses. However, there are currently no studies on the functions of ALCAM in food allergy. Therefore, we aimed to identify the functions of ALCAM in ovalbumin (OVA)-induced food allergy using ALCAM-deficient mice. Wild-type (WT) and ALCAM-deficient (ALCAM-/- ) mice were sensitized intraperitoneally and with orally fed OVA. The mice were killed, and parameters related to food allergy and T helper type 2 (Th2) immune responses were analysed. ALCAM serum levels increased and mRNA expression decreased in OVA-challenged WT mice. Serum immunoglobulin (Ig)E levels, Th2 cytokine mRNA and histological injuries were higher in OVA-challenged WT mice than in control mice, and these were attenuated in ALCAM-/- mice. T cell proliferation of total cells, CD3+ CD4+ T cells and activated T cells in immune tissues were diminished in OVA-challenged ALCAM-/- mice. Proliferation of co-cultured T cells and dendritic cells (DCs) was decreased by the anti-CD6 antibody. In addition, WT mice sensitized by adoptive transfer of OVA-pulsed ALCAM-/- BM-derived DCs showed reduced immune responses. Lastly, serum ALCAM levels were higher in children with food allergy than in control subjects. In this study, serum levels of ALCAM were elevated in food allergy-induced WT mice and children with food allergy. Moreover, immune responses and T cell activation were attenuated in OVA-challenged ALCAM-/- mice. These results indicate that ALCAM regulates food allergy by affecting T cell activation.
Collapse
Affiliation(s)
- Y S Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - M N Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - K E Lee
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - J Y Hong
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - M S Oh
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - S Y Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - K W Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - M H Sohn
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
100
|
Efficacy of oral immunotherapy with a rice-based edible vaccine containing hypoallergenic Japanese cedar pollen allergens for treatment of established allergic conjunctivitis in mice. Allergol Int 2018; 67:119-123. [PMID: 28676431 DOI: 10.1016/j.alit.2017.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 05/23/2017] [Accepted: 05/25/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND We have previously shown that prophylactic oral administration of transgenic rice seeds expressing hypoallergenic modified antigens suppressed the development of allergic conjunctivitis induced by Japanese cedar pollen. We have now investigated the efficacy of oral immunotherapy with such transgenic rice for established allergic conjunctivitis in mice. METHODS BALB/c mice were sensitized with two intraperitoneal injections of Japanese cedar pollen in alum, challenged with pollen in eyedrops, and then fed for 16 days with transgenic rice seeds expressing modified Japanese cedar pollen allergens Cry j 1 and Cry j 2 or with nontransgenic rice seeds as a control. They were then challenged twice with pollen in eyedrops, with clinical signs being evaluated at 15 min after the first challenge and the eyes, blood, spleen, and lymph nodes being isolated at 24 h after the second challenge. RESULTS The number of eosinophils in the conjunctiva and the clinical score for conjunctivitis were both significantly lower in mice fed the transgenic rice than in those fed nontransgenic rice. Oral vaccination with transgenic rice seeds also resulted in a significant increase in the production of IFN-γ by splenocytes, whereas it had no effect on the number of CD4+CD25+Foxp3+ regulatory T cells in the spleen or submandibular or mesenteric lymph nodes. CONCLUSIONS Oral administration of transgenic rice seeds expressing hypoallergenic allergens ameliorated allergic conjunctivitis in the established setting. Such a rice-based edible vaccine is potentially both safe and effective for oral immunotherapy in individuals with allergic conjunctivitis.
Collapse
|