1
|
Villablanca EJ. Organismal mucosal immunology: A perspective through the eyes of game theory. Mucosal Immunol 2024:S1933-0219(24)00126-0. [PMID: 39672543 DOI: 10.1016/j.mucimm.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
In complex organisms, functional units must interact cohesively to maintain homeostasis, especially within mucosal barriers that house diverse, specialized cell exposed to constant environmental challenges. Understanding how homeostasis at mucosal barriers is maintained and how its disruption can lead to autoimmune diseases or cancer, requires a holistic view. Although omics approaches and systems immunology have become powerful tools, they are not without limitations; interpretations may reflect researchers' assumptions, even if other explanations exist. In this perspective, I propose that applying game theory concepts to mucosal immunology could help interpret complex data, offering fresh perspectives and supporting the exploration of alternative scenarios. By framing the mucosal immune system as a network of strategic interactions with multiple possible outcomes, game theory, which analyzes strategic interactions and decision-making processes, could illuminate novel cell types and functions, cell interactions, and responses to pathogens and commensals, leading to a more comprehensive understanding of immune homeostasis and diseases. In addition, game theory might encourage researchers to consider a broader range of possibilities, reduce the risk of myopic thinking, and ultimately enable a more refined and comprehensive understanding of the complexity of the immune system at mucosal barriers. This perspective aims to introduce game theory as a complementary framework for mucosal immunologists, encouraging them to incorporate these concepts into data interpretation and system modeling.
Collapse
Affiliation(s)
- Eduardo J Villablanca
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institute and University Hospital, Stockholm, Sweden; Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden; Center of Molecular Medicine, Stockholm, Sweden.
| |
Collapse
|
2
|
Yoshida Y, Iijima K, Matsunaga M, Masuda MY, Jheng MJ, Kobayashi T, Kita H. Oral mucosa effectively protects against peanut allergy in mice. J Allergy Clin Immunol 2024; 154:1060-1068. [PMID: 38795733 PMCID: PMC11456774 DOI: 10.1016/j.jaci.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/08/2024] [Accepted: 05/03/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND Oral consumption of peanut products early in life reduces the incidence of peanut allergy in children. However, little is known about whether exposure via the oral mucosa alone is sufficient or whether the gastrointestinal tract must be engaged to protect against peanut allergy. OBJECTIVE We used a mouse model and examined the effects of peanut allergen administration to only the oral cavity on allergy development induced by environmental exposure. METHODS Naive BALB/c mice were administered peanut flour (PNF) sublingually, followed by epicutaneous exposure to PNF to mimic a human condition. The sublingual volume was adjusted to engage only the oral cavity and prevent it from reaching the esophagus or gastrointestinal tract. The efficacy was evaluated by examining the anaphylactic response, antibody titers, and T follicular helper cells. RESULTS The mice exposed epicutaneously to PNF developed peanut allergy, as demonstrated by increased plasma levels of peanut-specific IgE and the manifestation of acute systemic anaphylaxis following intraperitoneal challenge with peanut extract. The development of peanut allergy was suppressed when mice had been given PNF sublingually before epicutaneous exposure. There were fewer T follicular helper cells in the skin-draining lymph nodes of mice that received sublingual PNF than in the mice that received PBS. Suppression of IgE production was observed with sublingual PNF at 1/10 of the intragastric PNF dose. CONCLUSION Administration of peanut allergens only to the oral cavity effectively prevents the development of peanut allergy. The capacity of the oral mucosa to promote immunologic tolerance needs to be evaluated further to prevent food allergy.
Collapse
Affiliation(s)
- Yuya Yoshida
- Division of Allergy, Asthma and Clinical Immunology, and Department of Medicine, Mayo Clinic Arizona, Scottsdale, Ariz; Department of Pathological Biochemistry, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Osaka, Japan
| | - Koji Iijima
- Division of Allergy, Asthma and Clinical Immunology, and Department of Medicine, Mayo Clinic Arizona, Scottsdale, Ariz
| | - Mayumi Matsunaga
- Division of Allergy, Asthma and Clinical Immunology, and Department of Medicine, Mayo Clinic Arizona, Scottsdale, Ariz
| | - Mia Y Masuda
- Immunology Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minn and Scottsdale, Ariz
| | - Min-Jhen Jheng
- Virology and Gene Therapy Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minn and Scottsdale, Ariz
| | - Takao Kobayashi
- Division of Allergy, Asthma and Clinical Immunology, and Department of Medicine, Mayo Clinic Arizona, Scottsdale, Ariz
| | - Hirohito Kita
- Division of Allergy, Asthma and Clinical Immunology, and Department of Medicine, Mayo Clinic Arizona, Scottsdale, Ariz; Department of Immunology, Mayo Clinic Rochester, Rochester, Minn.
| |
Collapse
|
3
|
Seefeld ML, Templeton EL, Lehtinen JM, Sinclair N, Yadav D, Hartwell BL. Harnessing the potential of the NALT and BALT as targets for immunomodulation using engineering strategies to enhance mucosal uptake. Front Immunol 2024; 15:1419527. [PMID: 39286244 PMCID: PMC11403286 DOI: 10.3389/fimmu.2024.1419527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/08/2024] [Indexed: 09/19/2024] Open
Abstract
Mucosal barrier tissues and their mucosal associated lymphoid tissues (MALT) are attractive targets for vaccines and immunotherapies due to their roles in both priming and regulating adaptive immune responses. The upper and lower respiratory mucosae, in particular, possess unique properties: a vast surface area responsible for frontline protection against inhaled pathogens but also simultaneous tight regulation of homeostasis against a continuous backdrop of non-pathogenic antigen exposure. Within the upper and lower respiratory tract, the nasal and bronchial associated lymphoid tissues (NALT and BALT, respectively) are key sites where antigen-specific immune responses are orchestrated against inhaled antigens, serving as critical training grounds for adaptive immunity. Many infectious diseases are transmitted via respiratory mucosal sites, highlighting the need for vaccines that can activate resident frontline immune protection in these tissues to block infection. While traditional parenteral vaccines that are injected tend to elicit weak immunity in mucosal tissues, mucosal vaccines (i.e., that are administered intranasally) are capable of eliciting both systemic and mucosal immunity in tandem by initiating immune responses in the MALT. In contrast, administering antigen to mucosal tissues in the absence of adjuvant or costimulatory signals can instead induce antigen-specific tolerance by exploiting regulatory mechanisms inherent to MALT, holding potential for mucosal immunotherapies to treat autoimmunity. Yet despite being well motivated by mucosal biology, development of both mucosal subunit vaccines and immunotherapies has historically been plagued by poor drug delivery across mucosal barriers, resulting in weak efficacy, short-lived responses, and to-date a lack of clinical translation. Development of engineering strategies that can overcome barriers to mucosal delivery are thus critical for translation of mucosal subunit vaccines and immunotherapies. This review covers engineering strategies to enhance mucosal uptake via active targeting and passive transport mechanisms, with a parallel focus on mechanisms of immune activation and regulation in the respiratory mucosa. By combining engineering strategies for enhanced mucosal delivery with a better understanding of immune mechanisms in the NALT and BALT, we hope to illustrate the potential of these mucosal sites as targets for immunomodulation.
Collapse
Affiliation(s)
- Madison L Seefeld
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Erin L Templeton
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Justin M Lehtinen
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Noah Sinclair
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Daman Yadav
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Brittany L Hartwell
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
4
|
Nagler CR. Immune responses to food. Immunol Rev 2024; 326:5-7. [PMID: 39285805 DOI: 10.1111/imr.13397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Affiliation(s)
- Cathryn R Nagler
- Department of Pathology, University of Chicago, Chicago, Illinois, USA
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
5
|
Rajanala K, Upadhyay AK. Vaccines for Respiratory Viruses-COVID and Beyond. Vaccines (Basel) 2024; 12:936. [PMID: 39204059 PMCID: PMC11360283 DOI: 10.3390/vaccines12080936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
The COVID-19 (coronavirus disease 2019) pandemic had an extensive impact on global morbidity and mortality. Several other common respiratory viruses, such as the influenza virus and respiratory syncytial virus (RSV), are endemic or epidemic agents causing acute respiratory infections that are easily transmissible and pose a significant threat to communities due to efficient person-to-person transmission. These viruses can undergo antigenic variation through genetic mutations, resulting in the emergence of novel strains or variants, thereby diminishing the effectiveness of current vaccines, and necessitating ongoing monitoring and adjustment of vaccine antigens. As the virus-specific immunity is maintained only for several weeks or months after the infection, there is an emergent need to develop effective and durable vaccines. Additionally, specific populations, such as elderly or immunocompromised individuals, may exhibit reduced immune responses to respiratory viruses, posing significant challenges to develop vaccines that elicit durable and potent immunity. We present a comprehensive review of the molecular mechanisms underlying the pathogenesis and virulence of common respiratory viruses, such as RSV, influenza virus, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We discuss several vaccine approaches that are under development. A thorough understanding of the current strategies and the challenges encountered during the vaccine development process can lead to the advancement of effective next-generation vaccines.
Collapse
|
6
|
Kocabiyik O, Amlashi P, Vo AL, Suh H, Rodriguez-Aponte SA, Dalvie NC, Love JC, Andrabi R, Irvine DJ. Vaccine targeting to mucosal lymphoid tissues promotes humoral immunity in the gastrointestinal tract. SCIENCE ADVANCES 2024; 10:eadn7786. [PMID: 38809992 PMCID: PMC11135404 DOI: 10.1126/sciadv.adn7786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/23/2024] [Indexed: 05/31/2024]
Abstract
Viruses, bacteria, and parasites frequently cause infections in the gastrointestinal tract, but traditional vaccination strategies typically elicit little or no mucosal antibody responses. Here, we report a strategy to effectively concentrate immunogens and adjuvants in gut-draining lymph nodes (LNs) to induce gut-associated mucosal immunity. We prepared nanoemulsions (NEs) based on biodegradable oils commonly used as vaccine adjuvants, which encapsulated a potent Toll-like receptor agonist and displayed antigen conjugated to their surface. Following intraperitoneal administration, these NEs accumulated in gut-draining mesenteric LNs, priming strong germinal center responses and promoting B cell class switching to immunoglobulin A (IgA). Optimized NEs elicited 10- to 1000-fold higher antigen-specific IgG and IgA titers in the serum and feces, respectively, compared to free antigen mixed with NE, and strong neutralizing antibody titers against severe acute respiratory syndrome coronavirus 2. Thus, robust gut humoral immunity can be elicited by exploiting the unique lymphatic collection pathways of the gut with a lymph-targeting vaccine formulation.
Collapse
Affiliation(s)
- Ozgun Kocabiyik
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Parastoo Amlashi
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - A. Lina Vo
- Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Heikyung Suh
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sergio A. Rodriguez-Aponte
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Neil C. Dalvie
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - J. Christopher Love
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Raiees Andrabi
- Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Darrell J. Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815 USA
| |
Collapse
|
7
|
Liu Y, Lam DMK, Luan M, Zheng W, Ai H. Recent development of oral vaccines (Review). Exp Ther Med 2024; 27:223. [PMID: 38590568 PMCID: PMC11000446 DOI: 10.3892/etm.2024.12511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 02/08/2024] [Indexed: 04/10/2024] Open
Abstract
Oral immunization can elicit an effective immune response and immune tolerance to specific antigens. When compared with the traditional injection route, delivering antigens via the gastrointestinal mucosa offers superior immune effects and compliance, as well as simplicity and convenience, making it a more optimal route for immunization. At present, various oral vaccine delivery systems exist. Certain modified bacteria, such as Salmonella, Escherichia coli and particularly Lactobacillus, are considered promising carriers for oral vaccines. These carriers can significantly enhance immunization efficiency by actively replicating in the intestinal tract following oral administration. The present review provided a discussion of the main mechanisms of oral immunity and the research progress made in the field of oral vaccines. Additionally, it introduced the advantages and disadvantages of the currently more commonly administered injectable COVID-19 vaccines, alongside the latest advancements in this area. Furthermore, recent developments in oral vaccines are summarized, and their potential benefits and side effects are discussed.
Collapse
Affiliation(s)
- Ying Liu
- Key Laboratory of Follicular Development and Reproductive Health in Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | | | - Mei Luan
- Department of Geriatric Medicine, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Wenfu Zheng
- Chinese Academy of Sciences Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Hao Ai
- Key Laboratory of Follicular Development and Reproductive Health in Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| |
Collapse
|
8
|
Huang M, Shao H, Wang Z, Chen H, Li X. Specific and nonspecific nutritional interventions enhance the development of oral tolerance in food allergy. Crit Rev Food Sci Nutr 2023; 64:10303-10318. [PMID: 37313721 DOI: 10.1080/10408398.2023.2222803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The goal of food allergy (FA) prevention and treatment is to induce oral tolerance (OT). Appropriate nutritional interventions are essential to induce OT to food allergens. This review introduces the mechanism of OT and the importance of early nutritional interventions, and then firstly summarizes specific nutritional factors to induce the development of OT of FA, including proteins, vitamins, fatty acids, saccharides and probiotics. The regulatory mechanism mainly induces the development of tolerance by increasing local or systemic protective regulatory T cells (Tregs) to suppress FA, while the gut microbiota may also be changed to maintain intestinal homeostasis. For allergens-specific OT, the disruption to the structure of proteins and epitopes is critical for the induction of tolerance by hydrolyzed and heated proteins. Vitamins (vitamin A, D), fatty acids, saccharides and probiotics as allergens nonspecific OT also induce the development of OT through immunomodulatory effects. This review contributes to our understanding of OT in FA through nutritional interventions. Nutritional interventions play an important role in the induction of OT, and offer promising approaches to reduce allergy risk and alleviate FA. Moreover, due to the importance and diversity of nutrition, it must be the future trend of induction of OT in FA.
Collapse
Affiliation(s)
- Meijia Huang
- School of Food Science and Technology, Nanchang University, Nanchang, P.R. China
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, P.R. China
| | - Huming Shao
- School of Food Science and Technology, Nanchang University, Nanchang, P.R. China
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, P.R. China
| | - Zhongliang Wang
- School of Food Science and Technology, Nanchang University, Nanchang, P.R. China
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, P.R. China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, P.R. China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, P.R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, P.R. China
| | - Xin Li
- School of Food Science and Technology, Nanchang University, Nanchang, P.R. China
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, P.R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, P.R. China
| |
Collapse
|
9
|
Jonas D, Kirby M, Schenkel AR, Dangelmayr G. Modeling of adaptive immunity uncovers disease tolerance mechanisms. J Theor Biol 2023; 568:111498. [PMID: 37100114 DOI: 10.1016/j.jtbi.2023.111498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/03/2023] [Accepted: 04/17/2023] [Indexed: 04/28/2023]
Abstract
When an organism is challenged with a pathogen a cascade of events unfolds. The innate immune system rapidly mounts a preliminary nonspecific defense, while the acquired immune system slowly develops microbe-killing specialists. These responses cause inflammation, and along with the pathogen cause direct and indirect tissue damage, which anti-inflammatory mediators seek to temper. This interplay of systems is credited for maintaining homeostasis but may produce unexpected results such as disease tolerance. Tolerance is characterized by the persistence of pathogen and damage mitigation, where the relevant mechanisms are poorly understood. In this work we develop an ordinary differential equations model of the immune response to infection in order to identify key components in tolerance. Bifurcation analysis uncovers health, immune- and pathogen-mediated death clinical outcomes dependent on pathogen growth rate. We demonstrate that decreasing the inflammatory response to damage and increasing the strength of the immune system gives rise to a region in which limit cycles, or periodic solutions, are the only biological trajectories. We then describe areas of parameter space corresponding to disease tolerance by varying immune cell decay, pathogen removal, and lymphocyte proliferation rates.
Collapse
Affiliation(s)
- Daniel Jonas
- Colorado State University, Department of Mathematics, Fort Collins, CO, United States.
| | - Michael Kirby
- Colorado State University, Department of Mathematics, Fort Collins, CO, United States; Colorado State University, Department of Computer Science, Fort Collins, CO, United States
| | - Alan R Schenkel
- Colorado State University Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, United States
| | - Gerhard Dangelmayr
- Colorado State University, Department of Mathematics, Fort Collins, CO, United States
| |
Collapse
|
10
|
Wang P, Chen L, McIntosh CM, Lane JI, Li R, Xie SZ, Sattar H, Esterhazy D, Chong AS, Alegre M. Oral alloantigen exposure promotes donor-specific tolerance in a mouse model of minor-mismatched skin transplantation. Am J Transplant 2022; 22:2348-2359. [PMID: 35633180 PMCID: PMC9547964 DOI: 10.1111/ajt.17107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 05/08/2022] [Accepted: 05/23/2022] [Indexed: 01/25/2023]
Abstract
Oral antigen exposure is a powerful, non-invasive route to induce immune tolerance to dietary antigens, and has been modestly successful at prolonging graft survival in rodent models of transplantation. To harness the mechanisms of oral tolerance for promoting long-term graft acceptance, we developed a mouse model where the antigen ovalbumin (OVA) was introduced orally prior to transplantation with skin grafts expressing OVA. Oral OVA treatment pre-transplantation promoted permanent graft acceptance and linked tolerance to skin grafts expressing OVA fused to the additional antigen 2W. Tolerance was donor-specific, as secondary donor-matched, but not third-party allografts were spontaneously accepted. Oral OVA treatment promoted an anergic phenotype in OVA-reactive CD4+ and CD8+ conventional T cells (Tconvs) and expanded OVA-reactive Tregs pre-transplantation. However, skin graft acceptance following oral OVA resisted partial depletion of Tregs and blockade of PD-L1. Mechanistically, we revealed a role for the proximal gut draining lymph nodes (gdLNs) in mediating this effect, as an intestinal infection that drains to the proximal gdLNs prevented tolerance induction. Our study extends previous work applying oral antigen exposure to transplantation and serves as proof of concept that the systemic immune mechanisms supporting oral tolerance are sufficient to promote long-term graft acceptance.
Collapse
Affiliation(s)
- Peter Wang
- Department of Medicine, Section of RheumatologyUniversity of ChicagoChicagoIllinoisUSA
- The CollegeUniversity of ChicagoChicagoIllinoisUSA
| | - Luqiu Chen
- Department of Medicine, Section of RheumatologyUniversity of ChicagoChicagoIllinoisUSA
| | - Christine M. McIntosh
- Department of Medicine, Section of RheumatologyUniversity of ChicagoChicagoIllinoisUSA
- Pritzker School of MedicineUniversity of ChicagoChicagoIllinoisUSA
| | - Jorden I. Lane
- Department of PathologyUniversity of ChicagoChicagoIllinoisUSA
| | - Rena Li
- Department of Medicine, Section of RheumatologyUniversity of ChicagoChicagoIllinoisUSA
- The CollegeUniversity of ChicagoChicagoIllinoisUSA
| | - Stephen Z. Xie
- Department of Medicine, Section of RheumatologyUniversity of ChicagoChicagoIllinoisUSA
- The CollegeUniversity of ChicagoChicagoIllinoisUSA
| | - Husain Sattar
- Department of PathologyUniversity of ChicagoChicagoIllinoisUSA
| | - Daria Esterhazy
- Department of PathologyUniversity of ChicagoChicagoIllinoisUSA
| | - Anita S. Chong
- Department of Surgery, Section of TransplantationUniversity of ChicagoChicagoIllinoisUSA
| | - Maria‐Luisa Alegre
- Department of Medicine, Section of RheumatologyUniversity of ChicagoChicagoIllinoisUSA
| |
Collapse
|
11
|
Kar S, Devnath P, Emran TB, Tallei TE, Mitra S, Dhama K. Oral and intranasal vaccines against SARS-CoV-2: Current progress, prospects, advantages, and challenges. Immun Inflamm Dis 2022; 10:e604. [PMID: 35349752 PMCID: PMC8959423 DOI: 10.1002/iid3.604] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a deadly pandemic in the 21st century, resulting in many deaths, economic loss, and international immobility. Vaccination represents the only mechanism to defeat this virus. Several intramuscular vaccines have been approved and are currently used worldwide. MAIN BODY However, global mass vaccination has not been achieved owing to several limitations, including the need for expertise to administer the injection-based vaccine, improper distribution of the vaccine, and lack of cold chain facilities, particularly in resource-poor, low-income countries. Mucosal vaccines are typically administered either orally or nasally, and several studies have shown promising results for developing these vaccines against SARS-CoV-2 that might serve as viable alternatives to current vaccines. SARS-CoV-2 invades the human body via oral and nasal mucosal surfaces; thus, an oral or nasal vaccine can trigger the immune system to inhibit the virus at the mucosal level, preventing further transmission via a strong mucosal and systematic immune response. Although several approaches toward developing a mucosal vaccine are currently being tested, additional attention is required. CONCLUSION In this article, the current approaches used to develop effective oral and nasal mucosal vaccines against SARS-CoV-2 and their benefits, prospects, and challenges have been summarized.
Collapse
Affiliation(s)
- Sanchita Kar
- Department of Infectious DiseaseInstitute of Developing Science and Health Initiatives, ECB ChattarDhakaBangladesh
- Department of MicrobiologyUniversity of ChittagongChittagongBangladesh
| | - Popy Devnath
- Department of MicrobiologyNoakhali Science and Technology UniversityNoakhaliBangladesh
| | - Talha B. Emran
- Department of PharmacyBGC Trust University BangladeshChittagongBangladesh
| | - Trina E. Tallei
- Department of Biology, Faculty of Mathematics and Natural SciencesSam Ratulangi UniversityManadoNorth SulawesiIndonesia
- Division of Sustainable Use of Wallacea AreaThe University Centre of Excellence for Biotechnology and Conservation of Wallacea, Institute for Research and Community Services, Sam Ratulangi UniversityManadoNorth SulawesiIndonesia
| | - Saikat Mitra
- Department of Pharmacy, Faculty of PharmacyUniversity of DhakaDhakaBangladesh
| | - Kuldeep Dhama
- Division of PathologyICAR‐Indian Veterinary Research Institute, IzatnagarBareillyUttar PradeshIndia
| |
Collapse
|
12
|
Chen LH, Pan CH, Huang SY, Chan CH, Huang HY. The immunomodulatory effects of long-term supplementation with Lactobacillus casei Shirota depend on ovalbumin presentation in BALB/c mice. Sci Rep 2021; 11:19478. [PMID: 34593870 PMCID: PMC8484482 DOI: 10.1038/s41598-021-98791-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/14/2021] [Indexed: 11/09/2022] Open
Abstract
Immunomodulation is an ability of several particular probiotics. However, it still remains unclear whether the immunomodulatory effects of specific probiotics vary for different antigen presentation models with the same antigen. To investigate this matter, six groups of BALB/c mice (n = 10) were exposed to one of two antigen presentation models: ovalbumin (OVA) by injection alone, or injection plus intranasal administration. Moreover, the mice were fed distilled water or Lactobacillus casei Shirota fermented beverage (LcSFB) at low (2.5 × 109 CFU/kg body weight) or high doses (5 × 109 CFU/kg body weight) by gavage for 8 weeks. LcSFB enhanced the proliferation of splenocytes, production of OVA-specific immunoglobulin (Ig)-G and IgA, and the ratio of T-helper (Th)-2/Th1 cytokines in mice injected with OVA. Conversely, in the mice treated with OVA by injection plus intranasal administration, LcSFB attenuated the immune responses against OVA by reducing the proliferation of splenocytes, levels of OVA-specific IgE, IgG, and IgM, and ratio of Th2/Th1 cytokines. Moreover, LcSFB increased the percentage of regulatory T cells in the injection plus intranasal administration group. Taken together, this work indicates the immunoregulatory effects of LcSFB depend on how the antigen is presented. Therefore, the use of probiotics to boost the immune system must be carefully considered.
Collapse
Affiliation(s)
- Li-Han Chen
- Institute of Fisheries Science, National Taiwan University, Taipei, 10617, Taiwan
- Department of Life Science, National Taiwan University, Taipei, 10617, Taiwan
| | - Chun-Hsu Pan
- School of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
| | - Shih-Yi Huang
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei, 11031, Taiwan
| | - Ching-Hung Chan
- Department of Chemical Engineering and Biotechnology, Tatung University, 10452, Taipei, Taiwan
| | - Hui-Yu Huang
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
13
|
Rossi CM, Lenti MV, Merli S, Santacroce G, Di Sabatino A. Allergic manifestations in autoimmune gastrointestinal disorders. Autoimmun Rev 2021; 21:102958. [PMID: 34560305 DOI: 10.1016/j.autrev.2021.102958] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 09/19/2021] [Indexed: 12/14/2022]
Abstract
Allergic disorders target a young population, are increasing in both incidence and prevalence and are associated with significant disease burden. They result from the complex interplay between (epi)genetic and environmental factors, resulting in a Th2 inflammatory process targeting the epithelium of the respiratory tract (allergic rhinitis and asthma), skin (atopic dermatitis), and gastrointestinal tract (food allergy). Although the exact pathogenic mechanisms remain elusive, an altered immune system response in the gut is increasingly recognized as a relevant step. Allergic and gastrointestinal autoimmune disorders share several epidemiological, pathogenic and risk factors and several treatment modalities. Here we revise the current literature and show that allergic disorders are highly prevalent in gastrointestinal autoimmune diseases, including celiac disease, inflammatory bowel disease, autoimmune pancreatitis, and autoimmune cholangiopathies. No data are available for some autoimmune diseases, such as autoimmune gastritis and autoimmune enteropathy. To ensure the comprehensive care of patients with autoimmune gastrointestinal disorders, along with disease-specific factors, the presence of allergic disorders should be evaluated and treated when present, possibly targeting shared molecular pathways. Future studies are needed to define the exact pathogenic mechanisms underpinning the association between allergic and autoimmune diseases of the gastrointestinal tract.
Collapse
Affiliation(s)
- Carlo Maria Rossi
- Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Marco Vincenzo Lenti
- Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Stefania Merli
- Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Giovanni Santacroce
- Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Antonio Di Sabatino
- Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy.
| |
Collapse
|
14
|
Takaiwa F. Next-Generation Allergen-Specific Immunotherapy for Japanese Cedar Pollinosis Using Molecular Approaches. Immunotargets Ther 2021; 10:213-224. [PMID: 34239843 PMCID: PMC8259616 DOI: 10.2147/itt.s276874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/02/2021] [Indexed: 11/23/2022] Open
Abstract
Japanese cedar (JC) pollinosis is the most major IgE-mediated type I allergic disease in Japan. Allergen-specific immunotherapy is the only curative treatment for allergic diseases. Subcutaneous immunotherapy and sublingual immunotherapy have been introduced in Japan for JC pollinosis, but do not avoid some adverse side effects, because the natural allergens used as tolerogens cross-link with specific IgE types on mast cells and basophils. To make immunotherapy for JC pollinosis safer, more effective and convenient, rice-based oral allergy vaccines using hybrid peptides composed of multiple T-cell epitopes or recombinant deconstructed hypoallergenic derivatives derived from major allergens — Cry j 1 and Cry j 2 — have been developed and their efficacy and safety evaluated by oral administration of transgenic rice seeds. Furthermore, recombinant modified JC allergens conjugated with various immunomodulatory molecules and DNA-based vaccines have been created and their efficacy assessed.
Collapse
Affiliation(s)
- Fumio Takaiwa
- Soul Signal Institute, Kojyohama, Shiraoi, Hokkaido, 059-0641, Japan.,Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, 305-8602, Japan
| |
Collapse
|
15
|
Nettersheim FS, De Vore L, Winkels H. Vaccination in Atherosclerosis. Cells 2020; 9:cells9122560. [PMID: 33266027 PMCID: PMC7760548 DOI: 10.3390/cells9122560] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 12/17/2022] Open
Abstract
Atherosclerosis is the major underlying pathology of cardiovascular diseases that together are the leading cause of death worldwide. The formation of atherosclerotic plaques is driven by chronic vascular inflammation. Although several risk factors have been identified and significant progress in disease prevention and treatment has been made, no therapeutic agents targeting inflammation are clinically available. Recent clinical trials established the potential of anti-inflammatory therapies as a treatment of atherosclerosis. However, adverse impacts on host defense have raised safety concerns about these therapies. Scientific evidence during the past 40 years implicated an adaptive immune response against plaque-associated autoantigens in atherogenesis. Preclinical data have underscored the protective potential of immunization against such targets precisely and without the impairment of host defense. In this review, we discuss the current vaccination strategies against atherosclerosis, supposed mechanisms of action, therapeutic potential, and the challenges that must be overcome in translating this idea into clinical practice.
Collapse
|
16
|
Leonard SA. Food allergy prevention, including early food introduction. JOURNAL OF FOOD ALLERGY 2020; 2:69-74. [PMID: 39022133 PMCID: PMC11250506 DOI: 10.2500/jfa.2020.2.200007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
As the prevalence of immunoglobulin E (IgE)-mediated food allergy continues to increase without an imminent cure, prevention has become an urgent need. A breakthrough study that shows that early consumption of peanut can prevent the development of peanut allergy has led for a push in early interventions. Theories associated with the increasing prevalence of food allergy lend themselves to areas of potential intervention, e.g., age at time of food introduction, infant feeding practices, microbiome influences, diet composition, vitamin D deficiency, and increasing rates of eczema. This review focused on the available data from studies that investigated early interventions to decrease the risk of food allergy.
Collapse
Affiliation(s)
- Stephanie A. Leonard
- From the Division of Pediatric Allergy & Immunology, University of California San Diego, Rady Children's Hospital, San Diego, California
| |
Collapse
|
17
|
Endo T, Asaka D, Nakayama T, Saito S, Kodama H, Mitsuyoshi R, Takaishi S, Sugimoto N, Omae S, Takagi H, Wakasa Y, Ozawa K, Takano M, Takaiwa F, Kojima H, Saito S. Immunological and Symptomatic Effects of Oral Intake of Transgenic Rice Containing 7 Linked Major T-Cell Epitopes from Japanese Cedar Pollen Allergens. Int Arch Allergy Immunol 2020; 182:109-119. [PMID: 32854094 DOI: 10.1159/000509996] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/08/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND A rice-based peptide vaccine containing 7 linked human predominant T-cell epitopes (7Crp) derived from Japanese cedar (JC) pollen allergens, Cry j 1 and Cry j 2, was developed. Here, we examined the efficacy and safety of this transgenic rice in JC pollinosis patients. METHODS Transgenic rice (5, 20, and 80 g) was administered orally. We measured the T-cell proliferative activity against 7Crp, Cry j 1, and Cry j 2; the cytokine expression levels; and specific IgE and IgG4 production levels. In addition, the symptom and medication scores were monitored during the pollen season, and quality of life (QOL) was evaluated. RESULTS T-cell proliferative activities to Cry j 1, Cry j 2, and 7Crp were significantly depressed in a dose-dependent manner. Oral intake of 80 g transgenic rice for 20 weeks resulted in significant suppression of allergen-specific T-cell proliferation with downregulation of IL-13 and upregulation of IL-10 levels but no changes to specific IgE and IgG4 levels. The QOL symptom scores for allergic rhinitis were not significantly improved. CONCLUSIONS Allergen-specific T-cell responses were significantly reduced by oral intake of transgenic rice in a dose-dependent manner. However, neither medication score nor QOL symptom scores could be improved during the JC pollen season with oral intake of transgenic rice for 20 weeks.
Collapse
Affiliation(s)
- Tomonori Endo
- Department of Otorhinolaryngology, Jikei University School of Medicine, Tokyo, Japan, .,Department of Otorhinolaryngology, Federation of National Public Service Personnel Mutual Aid Associations, Tokyo Kyosai Hospital, Tokyo, Japan,
| | - Daiya Asaka
- Department of Otorhinolaryngology, Jikei University School of Medicine, Tokyo, Japan
| | - Tsuguhisa Nakayama
- Department of Otorhinolaryngology, Jikei University School of Medicine, Tokyo, Japan
| | - Shota Saito
- Department of Otorhinolaryngology, Jikei University School of Medicine, Tokyo, Japan
| | - Hiroki Kodama
- Department of Otorhinolaryngology, Jikei University School of Medicine, Tokyo, Japan
| | - Ryoto Mitsuyoshi
- Department of Otorhinolaryngology, Jikei University School of Medicine, Tokyo, Japan
| | - Shinya Takaishi
- Department of Otorhinolaryngology, Jikei University School of Medicine, Tokyo, Japan
| | - Naoki Sugimoto
- Department of Otorhinolaryngology, Jikei University School of Medicine, Tokyo, Japan
| | - Sachiko Omae
- Department of Otorhinolaryngology, Jikei University School of Medicine, Tokyo, Japan
| | - Hidenori Takagi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Ibaraki, Japan
| | - Yuhya Wakasa
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Ibaraki, Japan
| | - Kenjiro Ozawa
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Ibaraki, Japan
| | - Makoto Takano
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Ibaraki, Japan
| | - Fumio Takaiwa
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Ibaraki, Japan
| | - Hiromi Kojima
- Department of Otorhinolaryngology, Jikei University School of Medicine, Tokyo, Japan
| | - Saburo Saito
- Division of Molecular Immunology, Research Center for Medical Science, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
18
|
The potential for vaccines against scour worms of small ruminants. Int J Parasitol 2020; 50:533-553. [PMID: 32569640 DOI: 10.1016/j.ijpara.2020.04.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 02/08/2023]
Abstract
This review addresses the research landscape regarding vaccines against scour worms, particularly Trichostrongylus spp. and Teladorsagia circumcincta. The inability of past research to deliver scour-worm vaccines with reliable and reproducible efficacy has been due in part to gaps in knowledge concerning: (i) host-parasite interactions leading to development of type-2 immunity, (ii) definition of an optimal suite of parasite antigens, and (iii) rational formulation and administration to induce protective immunity against gastrointestinal nematodes (GIN) at the site of infestation. Recent 'omics' developments enable more systematic analyses. GIN genomes are reaching completion, facilitating "reverse vaccinology" approaches that have been used successfully for the Rhipicephalus australis vaccine for cattle tick, while methods for gene silencing and editing in GIN enable identification and validation of potential vaccine antigens. We envisage that any efficacious scour worm vaccine(s) would be adopted similarly to "Barbervax™" within integrated parasite management schemes. Vaccines would therefore effectively parallel the use of resistant animals, and reduce the frequency of drenching and pasture contamination. These aspects of integration, efficacy and operation require updated models and validation in the field. The conclusion of this review outlines an approach to facilitate an integrated research program.
Collapse
|
19
|
Sánchez Ramón S, Manzanares M, Candelas G. MUCOSAL anti-infections vaccines: Beyond conventional vaccines. REUMATOLOGIA CLINICA 2020; 16:49-55. [PMID: 30527360 DOI: 10.1016/j.reuma.2018.10.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 10/03/2018] [Accepted: 10/10/2018] [Indexed: 12/24/2022]
Abstract
An urgent search is currently underway for alternatives to antibiotics to prevent infections, due to the accelerated evolution and increase in antibiotic resistance. This problem is more serious for patients with recurrent infections, since they have to use many cycles of antibiotics per year, so the risk for antibiotic resistance is higher and can be life-threatening. In recent years, the use of prophylactic vaccines via the mucosal route for these patients with recurrent infections has been demonstrated as a potentially beneficial and safe alternative to prevent infections. The new knowledge about mucosal immunity and trained immunity, a form of innate immunity memory that can enhance the response to different infectious threads, has made it easier to extend its use. The application of the new concepts of trained immunity may explain the simultaneous pro-tolerogenic and boosting effect or effects of these drugs on diverse immune cells for different infections. In this review, we describe the immunomodulatory mechanisms of mucosal polybacterial vaccines and their connection with trained immunity and its utility in the prevention of recurrent infections in immunosuppressed patients.
Collapse
Affiliation(s)
| | - Mario Manzanares
- Servicio de Inmunología, Hospital Clínico San Carlos, Madrid, España
| | - Gloria Candelas
- Servicio de Reumatología, Hospital Clínico San Carlos, Madrid, España.
| |
Collapse
|
20
|
Quandt JA, Becquart P, Kamma E, Hallenbeck J. Mucosal Administration of E-selectin Limits Disability in Models of Multiple Sclerosis. Front Mol Neurosci 2019; 12:190. [PMID: 31507371 PMCID: PMC6718462 DOI: 10.3389/fnmol.2019.00190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/22/2019] [Indexed: 11/13/2022] Open
Abstract
E-selectin plays an important role in mediating the rolling of leukocytes along and thus, the subsequent extravasation across activated endothelial cells comprising the microvasculature of the blood brain barrier (BBB). In multiple sclerosis (MS) and other inflammatory disorders of the central nervous system (CNS), the microvasculature is altered and immune cells infiltrate the brain and spinal cord contributing to damage, demyelination and ultimately disability. While mucosal administration is typically used to affect lymphocyte hyporesponsiveness or tolerance to suspect autoantigens, intranasal administration to E-selectin has previously been shown to protect against CNS inflammatory insults. We characterized the potential for mucosal administration of E-selectin to modulate CNS autoimmunity in the experimental autoimmune encephalomyelitis (EAE) model of MS. Intranasally administered E-selectin reduced swelling by as much as 50% in delayed-type hypersensitivity reactions compared to ovalbumin-tolerized controls. Intranasal E-selectin delivery prior to disease induction with myelin oligodendrocyte glycoprotein (MOG)35-55 reduced disease severity and total disease burden by more than 50% compared to PBS-tolerized animals; this protection was not associated with differences in the magnitude of the autoimmune response. Examination after the onset of disease showed that protection was associated with significant reductions in inflammatory infiltrates throughout the spinal cord. Tolerization to E-selectin did not influence encephalitogenic characteristics of autoreactive T cells such as IFN-gamma or IL-17 production. Clinical disease was also significantly reduced when E-selectin was first delivered after the onset of clinical symptoms. Splenic and lymph node (LN) populations from E-selectin-tolerized animals showed E-selectin-specific T cell responses and production of the immunomodulatory cytokine IL-10. Transfer of enriched CD4+ T cells from E-selectin tolerized mice limited disability in the passive SJL model of relapsing remitting MS. These results suggest a role for influencing E-selectin specific responses to limit neuroinflammation that warrants further exploration and characterization to better understand its potential to mitigate neurodegeneration in disorders such as MS.
Collapse
Affiliation(s)
- Jacqueline A Quandt
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Pierre Becquart
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Emily Kamma
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - John Hallenbeck
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
21
|
Effect of Oral T2 Antigen on Chronic Prostatitis/Chronic Pelvic Pain Syndrome in Mice Model. Inflammation 2019; 42:2086-2094. [DOI: 10.1007/s10753-019-01072-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
22
|
Phenotype analyses of IL-10-producing Foxp3 - CD4 + T cells increased by subcutaneous immunotherapy in allergic airway inflammation. Int Immunopharmacol 2018; 61:297-305. [PMID: 29909233 DOI: 10.1016/j.intimp.2018.06.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/08/2018] [Accepted: 06/06/2018] [Indexed: 12/31/2022]
Abstract
INTRODUCTION The mechanisms of allergen immunotherapy are not fully elucidated. Here, we sought to develop a murine model to demonstrate the effectiveness of subcutaneous immunotherapy (SCIT) for allergic responses. As excessive antigen dosages may induce immune tolerance in sensitized mice, the effects of SCIT were assessed by varying the antigen dosage. The mechanisms of SCIT were analyzed by focusing on the induction of Foxp3+ Treg cells and IL-10-producing Foxp3- CD4+ T cells, as well as on the phenotype of the latter cells. METHODS Ovalbumin (OVA) + Al(OH)3-sensitized mice received subcutaneous dosages of OVA at 0.01, 0.1 or 1 mg/animal for SCIT, followed by intratracheal challenges with OVA at 5, 50 or 500 μg/animal. RESULTS The maximum effects of SCIT were observed with 1 mg/animal of OVA for airway inflammation induced by 5 μg/animal of OVA, in which airway eosinophilia and Th2 cytokine production were markedly suppressed. The increase in the OVA-specific IgE level was significantly suppressed by SCIT. The development of bronchial epithelial thickening and mucus accumulation were also suppressed by SCIT. Concomitantly, IL-10-producing Foxp3- CD4+ T cells were increased in the lungs by SCIT, but Foxp3+ Treg cells were not. Most of the induced IL-10-producing Foxp3- CD4+ T cells were negative for either IL-5 or LAG-3, but positive for CD49b. CONCLUSION We successfully developed an airway allergic model for SCIT. It was suggested that most of IL-10-producing Foxp3- CD4+ regulatory T cells increased by SCIT in the lungs were CD49b+ CD4+ regulatory T cells, but neither Th2 cells nor Tr1 cells.
Collapse
|
23
|
Cook DP, Gysemans C, Mathieu C. Lactococcus lactis As a Versatile Vehicle for Tolerogenic Immunotherapy. Front Immunol 2018; 8:1961. [PMID: 29387056 PMCID: PMC5776164 DOI: 10.3389/fimmu.2017.01961] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 12/19/2017] [Indexed: 12/22/2022] Open
Abstract
Genetically modified Lactococcus lactis bacteria have been engineered as a tool to deliver bioactive proteins to mucosal tissues as a means to exert both local and systemic effects. They have an excellent safety profile, the result of years of human consumption in the food industry, as well as a lack of toxicity and immunogenicity. Also, containment strategies have been developed to promote further application as clinical protein-based therapeutics. Here, we review technological advancements made to enhanced the potential of L. lactis as live biofactories and discuss some examples of tolerogenic immunotherapies mediated by mucosal drug delivery via L. lactis. Additionally, we highlight their use to induce mucosal tolerance by targeted autoantigen delivery to the intestine as an approach to reverse autoimmune type 1 diabetes.
Collapse
Affiliation(s)
- Dana P Cook
- Laboratory of Clinical and Experimental Endocrinology (CEE), KU Leuven, Leuven, Belgium
| | - Conny Gysemans
- Laboratory of Clinical and Experimental Endocrinology (CEE), KU Leuven, Leuven, Belgium
| | - Chantal Mathieu
- Laboratory of Clinical and Experimental Endocrinology (CEE), KU Leuven, Leuven, Belgium
| |
Collapse
|
24
|
Smole U, Schabussova I, Pickl WF, Wiedermann U. Murine models for mucosal tolerance in allergy. Semin Immunol 2017; 30:12-27. [PMID: 28807539 DOI: 10.1016/j.smim.2017.07.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 07/21/2017] [Indexed: 02/07/2023]
Abstract
Immunity is established by a fine balance to discriminate between self and non-self. In addition, mucosal surfaces have the unique ability to establish and maintain a state of tolerance also against non-self constituents such as those represented by the large numbers of commensals populating mucosal surfaces and food-derived or air-borne antigens. Recent years have seen a dramatic expansion in our understanding of the basic mechanisms and the involved cellular and molecular players orchestrating mucosal tolerance. As a direct outgrowth, promising prophylactic and therapeutic models for mucosal tolerance induction against usually innocuous antigens (derived from food and aeroallergen sources) have been developed. A major theme in the past years was the introduction of improved formulations and novel adjuvants into such allergy vaccines. This review article describes basic mechanisms of mucosal tolerance induction and contrasts the peculiarities but also the interdependence of the gut and respiratory tract associated lymphoid tissues in that context. Particular emphasis is put on delineating the current prophylactic and therapeutic strategies to study and improve mucosal tolerance induction in allergy.
Collapse
Affiliation(s)
- Ursula Smole
- Institute of Immunology, Center for Pathophysiology, Infectiology, and Immunology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Irma Schabussova
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Winfried F Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology, and Immunology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
25
|
de Oliveira SRP, Nomizo A, Frantz FG, Faccioli LH, de Matos APK, Carrilho E, Afonso A, de Freitas Anibal F. Participation of Leukotrienes in the Immune Modulation of Oral Tolerance. Front Microbiol 2017; 8:242. [PMID: 28270799 PMCID: PMC5318402 DOI: 10.3389/fmicb.2017.00242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 02/03/2017] [Indexed: 11/24/2022] Open
Abstract
Oral tolerance (OT) is characterized as a peripheral immune tolerance form, in which, mature lymphocytes in lymphoid tissues associated with mucosa, become non-functional or hypo responsive due to prior oral administration of antigen. OT is an important immunological phenomenon due to its therapeutic potential in inflammatory processes and others diseases. Here we evaluated leukotriene role in the induction of OT, as well as, the production of cytokines IL-5 and IFN-γ in leukotriene deficient animals (knock-out). Our results suggested that even in the presence of OT and leukotrienes absence, cytokine IFN-γ remains being secreted, which gives us an indication of immune system specificity and also that IFN-γ participates in various immune processes.
Collapse
Affiliation(s)
- Sandra R P de Oliveira
- Laboratory of Parasitology, Department of Morphology and Pathology, Universidade Federal de São Carlos São Carlos, Brazil
| | - Auro Nomizo
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto - University of São Paulo Ribeirão Preto, Brazil
| | - Fabiani G Frantz
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto - University of São Paulo Ribeirão Preto, Brazil
| | - Lúcia H Faccioli
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto - University of São Paulo Ribeirão Preto, Brazil
| | - Ana Paula Keller de Matos
- Laboratory of Parasitology, Department of Morphology and Pathology, Universidade Federal de São CarlosSão Carlos, Brazil; Universidade de São Paulo, Escola de Enfermagem de Ribeirão Preto, Ribeirão PretoBrazil
| | - Emanuel Carrilho
- Bioanalytical, Microfabrication, and Separations Group, Instituto de Química de São Carlos, Universidade de São Paulo São Carlos, Brazil
| | - Ana Afonso
- Laboratory of Parasitology, Department of Morphology and Pathology, Universidade Federal de São CarlosSão Carlos, Brazil; Bioanalytical, Microfabrication, and Separations Group, Instituto de Química de São Carlos, Universidade de São PauloSão Carlos, Brazil; Medical Parasitology Unit, Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade Nova de LisboaLisbon, Portugal
| | - Fernanda de Freitas Anibal
- Laboratory of Parasitology, Department of Morphology and Pathology, Universidade Federal de São Carlos São Carlos, Brazil
| |
Collapse
|
26
|
Deshpande V, Krishnan R, Philip S, Faludi I, Ponnusamy T, Thota LNR, Endresz V, Lu X, Kakkar VV, Mundkur LA. Oral administration of recombinant Mycobacterium smegmatis expressing a tripeptide construct derived from endogenous and microbial antigens prevents atherosclerosis in ApoE(-/-) mice. Cardiovasc Ther 2017; 34:314-24. [PMID: 27241889 DOI: 10.1111/1755-5922.12201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
INTRODUCTION Immunotherapy by inducing oral tolerance to atherogenic self-antigens is gaining importance as an alternative treatment modality for atherosclerosis. The use of live bacterial vectors to express the recombinant antigen in vivo will obviate the need for large-scale purification of recombinant protein and may also augment the efficacy of oral tolerance induction. AIM The objective of the study was to explore the use of recombinant Mycobacterium smegmatis as a live vector for oral delivery of antigens to induce immune tolerance. METHOD AND RESULTS We developed a M. smegmatis vector to secrete a recombinant tripeptide construct (AHC; peptides from Apolipoprotein B, Heat-shock protein 60 and Chlamydia pneumoniae outer membrane protein) expressed in a dendroaspin protein scaffold in pJH154 background. Immune response and oral tolerance to the cloned peptides were studied in C57/BL6 mice. The efficacy of this live vaccine to control atherosclerosis was studied in ApoE(-/-) knockout mice in C57/BL6 background. Oral administration of M. smegmatis secreting the cloned AHC antigen was found to induce tolerance to cloned protein and reduce the development of atherosclerosis by 24.0% compared to control. Protection against atherosclerosis was associated with increase in expression of regulatory T cell-associated markers including CTLA4 (1.8-fold), Foxp3 (2.6-fold), TGF-β (2.8-fold), IL10 (2.9-fold), and reduction in lipids, macrophage infiltration, and expression of inflammatory mediators in aorta. CONCLUSIONS Our results suggest that M. smegmatis can be developed as an oral carrier of recombinant proteins to treat inflammatory autoimmune diseases.
Collapse
Affiliation(s)
| | | | - Sheena Philip
- Molecular Immunology, Thrombosis Research Institute, Bangalore, India
| | - Ildiko Faludi
- Department of Medical Microbiology and Immunobiology, University of Szeged, Szeged, Hungary
| | | | | | - Valeria Endresz
- Department of Medical Microbiology and Immunobiology, University of Szeged, Szeged, Hungary
| | - Xinjie Lu
- Molecular Immunology, Thrombosis Research Institute, London, UK
| | - Vijay V Kakkar
- Molecular Immunology, Thrombosis Research Institute, Bangalore, India.,Molecular Immunology, Thrombosis Research Institute, London, UK
| | - Lakshmi A Mundkur
- Molecular Immunology, Thrombosis Research Institute, Bangalore, India.
| |
Collapse
|
27
|
Groeger SE, Meyle J. Epithelial barrier and oral bacterial infection. Periodontol 2000 2017; 69:46-67. [PMID: 26252401 DOI: 10.1111/prd.12094] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2015] [Indexed: 01/11/2023]
Abstract
The oral epithelial barrier separates the host from the environment and provides the first line of defense against pathogens, exogenous substances and mechanical stress. It consists of underlying connective tissue and a stratified keratinized epithelium with a basement membrane, whose cells undergo terminal differentiation resulting in the formation of a mechanically resistant surface. Gingival keratinocytes are connected by various transmembrane proteins, such as tight junctions, adherens junctions and gap junctions, each of which has a specialized structure and specific functions. Periodontal pathogens are able to induce inflammatory responses that lead to attachment loss and periodontal destruction. A number of studies have demonstrated that the characteristics of pathogenic oral bacteria influence the expression and structural integrity of different cell-cell junctions. Tissue destruction can be mediated by host cells following stimulation with cytokines and bacterial products. Keratinocytes, the main cell type in gingival epithelial tissues, express a variety of proinflammatory cytokines and chemokines, including interleukin-1alpha, interleukin-1beta, interleukin-6, interleukin-8 and tumor necrosis factor-alpha. Furthermore, the inflammatory mediators that may be secreted by oral keratinocytes are vascular endothelial growth factor, prostaglandin E2 , interleukin-1 receptor antagonist and chemokine (C-C motif) ligand 2. The protein family of matrix metalloproteinases is able to degrade all types of extracellular matrix protein, and can process a number of bioactive molecules. Matrix metalloproteinase activities under inflammatory conditions are mostly deregulated and often increased, and those mainly relevant in periodontal disease are matrix metalloproteinases 1, 2, 3, 8, 9, 13 and 24. Viral infection may also influence the epithelial barrier. Studies show that the expression of HIV proteins in the mucosal epithelium is correlated with the disruption of epithelial tight junctions, suggesting a possible enhancement of human papilloma virus infection by HIV-associated disruption of tight junctions. Altered expression of matrix metalloproteinases was demonstrated in keratinocytes transformed with human papilloma virus-16 or papilloma virus-18,. To summarize, the oral epithelium is able to react to a variety of exogenous, possibly noxious influences.
Collapse
|
28
|
Ando S, Hasegawa A, Murakami Y, Zeng N, Takatsuka N, Maeda Y, Masuda T, Suehiro Y, Kannagi M. HTLV-1 Tax-Specific CTL Epitope–Pulsed Dendritic Cell Therapy Reduces Proviral Load in Infected Rats with Immune Tolerance against Tax. THE JOURNAL OF IMMUNOLOGY 2016; 198:1210-1219. [DOI: 10.4049/jimmunol.1601557] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 11/28/2016] [Indexed: 11/19/2022]
|
29
|
Embregts CWE, Forlenza M. Oral vaccination of fish: Lessons from humans and veterinary species. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 64:118-37. [PMID: 27018298 DOI: 10.1016/j.dci.2016.03.024] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 03/17/2016] [Indexed: 05/08/2023]
Abstract
The limited number of oral vaccines currently approved for use in humans and veterinary species clearly illustrates that development of efficacious and safe oral vaccines has been a challenge not only for fish immunologists. The insufficient efficacy of oral vaccines is partly due to antigen breakdown in the harsh gastric environment, but also to the high tolerogenic gut environment and to inadequate vaccine design. In this review we discuss current approaches used to develop oral vaccines for mass vaccination of farmed fish species. Furthermore, using various examples from the human and veterinary vaccine development, we propose additional approaches to fish vaccine design also considering recent advances in fish mucosal immunology and novel molecular tools. Finally, we discuss the pros and cons of using the zebrafish as a pre-screening animal model to potentially speed up vaccine design and testing for aquaculture fish species.
Collapse
Affiliation(s)
- Carmen W E Embregts
- Cell Biology and Immunology Group, Department of Animal Sciences, Wageningen University, Wageningen, The Netherlands
| | - Maria Forlenza
- Cell Biology and Immunology Group, Department of Animal Sciences, Wageningen University, Wageningen, The Netherlands.
| |
Collapse
|
30
|
Holechek SA, McAfee MS, Nieves LM, Guzman VP, Manhas K, Fouts T, Bagley K, Blattman JN. Retinaldehyde dehydrogenase 2 as a molecular adjuvant for enhancement of mucosal immunity during DNA vaccination. Vaccine 2016; 34:5629-5635. [PMID: 27670072 DOI: 10.1016/j.vaccine.2016.09.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 08/28/2016] [Accepted: 09/14/2016] [Indexed: 12/12/2022]
Abstract
In order for vaccines to induce efficacious immune responses against mucosally transmitted pathogens, such as HIV-1, activated lymphocytes must efficiently migrate to and enter targeted mucosal sites. We have previously shown that all-trans retinoic acid (ATRA) can be used as a vaccine adjuvant to enhance mucosal CD8+ T cell responses during vaccination and improve protection against mucosal viral challenge. However, the ATRA formulation is incompatible with most recombinant vaccines, and the teratogenic potential of ATRA at high doses limits its usage in many clinical settings. We hypothesized that increasing in vivo production of retinoic acid (RA) during vaccination with a DNA vector expressing retinaldehyde dehydrogenase 2 (RALDH2), the rate-limiting enzyme in RA biosynthesis, could similarly provide enhanced programming of mucosal homing to T cell responses while avoiding teratogenic effects. Administration of a RALDH2- expressing plasmid during immunization with a HIVgag DNA vaccine resulted in increased systemic and mucosal CD8+ T cell numbers with an increase in both effector and central memory T cells. Moreover, mice that received RALDH2 plasmid during DNA vaccination were more resistant to intravaginal challenge with a recombinant vaccinia virus expressing the same HIVgag antigen (VACVgag). Thus, RALDH2 can be used as an alternative adjuvant to ATRA during DNA vaccination leading to an increase in both systemic and mucosal T cell immunity and better protection from viral infection at mucosal sites.
Collapse
Affiliation(s)
- Susan A Holechek
- Biodesign Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, Tempe, AZ 85287-5401, United States; School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, United States; Simon A. Levin Mathematical, Computational and Modeling Sciences Center, Arizona State University, Tempe, AZ 85287-3901, United States
| | - Megan S McAfee
- Biodesign Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, Tempe, AZ 85287-5401, United States; School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, United States
| | - Lizbeth M Nieves
- Biodesign Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, Tempe, AZ 85287-5401, United States; School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, United States
| | - Vanessa P Guzman
- Biodesign Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, Tempe, AZ 85287-5401, United States; School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, United States
| | - Kavita Manhas
- Biodesign Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, Tempe, AZ 85287-5401, United States; School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, United States
| | - Timothy Fouts
- Profectus BioSciences, Inc., Baltimore, MD 21224, United States
| | - Kenneth Bagley
- Profectus BioSciences, Inc., Baltimore, MD 21224, United States
| | - Joseph N Blattman
- Biodesign Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, Tempe, AZ 85287-5401, United States; School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, United States.
| |
Collapse
|
31
|
Epicutaneous and Oral Low-Zone Tolerance Protects from Colitis in Mice. J Invest Dermatol 2016; 136:1831-1839. [DOI: 10.1016/j.jid.2016.04.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 03/07/2016] [Accepted: 04/11/2016] [Indexed: 11/19/2022]
|
32
|
Buerth C, Mausberg AK, Heininger MK, Hartung HP, Kieseier BC, Ernst JF. Oral Tolerance Induction in Experimental Autoimmune Encephalomyelitis with Candida utilis Expressing the Immunogenic MOG35-55 Peptide. PLoS One 2016; 11:e0155082. [PMID: 27159446 PMCID: PMC4861260 DOI: 10.1371/journal.pone.0155082] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/24/2016] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease that attacks myelinated axons in the central nervous system. Induction of oral tolerance is a potent mechanism to prevent autoimmunity. The food yeast Candida utilis was used to test the therapeutic potential of oral tolerance induction in an animal model of human multiple sclerosis (MS). We constructed a C. utilis strain, which displays a fusion peptide composed of the encephalitogenic MOG35-55 peptide and the C. utilis Gas1 cell wall protein on its surface.By immunizing mice with MOG35-55 peptide experimental autoimmune encephalomyelitis (EAE) was induced in a mouse model. Feeding of mice with C. utilis that expresses MOG35-55 peptide on its surface was started seven days prior to immunization and was continued for ten days. Control animals were treated with wild-type fungus or left untreated. Untreated mice developed first clinical symptoms ten days post immunization (p. i.) with an ascending paralysis reaching maximal clinical disability at day 18 to 20 p. i.. Treatment with the wild-type strain demonstrated comparable clinical symptoms. In contrast, oral gavage of MOG35-55-presenting fungus ameliorated the development of EAE. In addition, incidence as well as maximal clinical disease severity were significantly reduced. Interestingly, reduction of disease severity also occurred in animals treated with heat-inactivated C. utilis cells indicating that tolerance induction was independent of fungal viability. Better disease outcome correlated with reduced demyelination and cellular inflammation in the spinal cord, lower T cell proliferation against rechallenge with MOG35-55 and more regulatory T cells in the lymph nodes. Our data demonstrate successful that using the food approved fungus C. utilis presenting the immunogenic MOG35-55 peptide on its surface induced an oral tolerance against this epitope in EAE. Further studies will reveal the nature and extent of an anti-inflammatory environment established by the treatment that prevents the development of an autoimmune disorder affecting the CNS.
Collapse
Affiliation(s)
- Christoph Buerth
- Institute of Molecular Mycology, Department Biology, Heinrich-Heine-University, Düsseldorf, Germany
- * E-mail: (CB); (AKM)
| | - Anne K. Mausberg
- Research Group for Clinical and Experimental Neuroimmunology, Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- * E-mail: (CB); (AKM)
| | - Maximilian K. Heininger
- Research Group for Clinical and Experimental Neuroimmunology, Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Research Group for Clinical and Experimental Neuroimmunology, Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Bernd C. Kieseier
- Research Group for Clinical and Experimental Neuroimmunology, Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Joachim F. Ernst
- Institute of Molecular Mycology, Department Biology, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
33
|
Mojibian M, Glavas MM, Kieffer TJ. Engineering the gut for insulin replacement to treat diabetes. J Diabetes Investig 2016; 7 Suppl 1:87-93. [PMID: 27186362 PMCID: PMC4854511 DOI: 10.1111/jdi.12479] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 01/06/2016] [Indexed: 12/11/2022] Open
Abstract
The gut epithelium's large surface area, its direct exposure to ingested nutrients, its vast stem cell population and its immunotolerogenic environment make it an excellent candidate for therapeutic cells to treat diabetes. Thus, several attempts have been made to coax immature gut cells to differentiate into insulin-producing cells by altering the expression patterns of specific transcription factors. Furthermore, because of similarities in enteroendocrine and pancreatic endocrine cell differentiation pathways, other approaches have used genetically engineered enteroendocrine cells to produce insulin in addition to their endogenous secreted hormones. Several studies support the utility of both of these approaches for the treatment of diabetes. Converting a patient's own gut cells into meal-regulated insulin factories in a safe and immunotolerogenic environment is an attractive approach to treat and potentially cure diabetes. Here, we review work on these approaches and indicate where we feel further advancements are required.
Collapse
Affiliation(s)
- Majid Mojibian
- Laboratory of Molecular and Cellular Medicine Department of Cellular and Physiological Sciences Life Sciences Institute University of British Columbia Vancouver British Columbia Canada
| | - Maria M Glavas
- Laboratory of Molecular and Cellular Medicine Department of Cellular and Physiological Sciences Life Sciences Institute University of British Columbia Vancouver British Columbia Canada
| | - Timothy J Kieffer
- Laboratory of Molecular and Cellular Medicine Department of Cellular and Physiological Sciences Life Sciences Institute University of British Columbia Vancouver British Columbia Canada
| |
Collapse
|
34
|
Watanabe N, Kaminuma O, Kitamura N, Hiroi T. Induced Treg Cells Augment the Th17-Mediated Intestinal Inflammatory Response in a CTLA4-Dependent Manner. PLoS One 2016; 11:e0150244. [PMID: 26950218 PMCID: PMC4780716 DOI: 10.1371/journal.pone.0150244] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 02/11/2016] [Indexed: 12/17/2022] Open
Abstract
Th17 cells and Foxp3+ regulatory T cells (Tregs) are thought to promote and suppress inflammatory responses, respectively. However, whether they counteract each other or synergize in regulating immune reactions remains controversial. To determine their interactions, we describe the results of experiments employing mouse models of intestinal inflammation by transferring antigen-specific Th cells (Th1, Th2, and Th17) differentiated in vitro followed by the administration of the cognate antigen via enema. We show that cotransfer of induced Tregs (iTregs) suppressed Th1- and Th2-mediated colon inflammation. In contrast, colon inflammation induced by transfer of Th17 cells, was augmented by the cotransfer of iTregs. Furthermore, oral delivery of antigen potentiated Th17-mediated colon inflammation. Administration of a blocking antibody against cytotoxic T lymphocyte-associated antigen 4 (CTLA4) abrogated the effects of cotransfer of iTregs, while the injection of a soluble recombinant immunoglobulin (Ig) fusion protein, CTLA4-Ig substituted for the cotransfer of iTregs. These results suggest that antigen-specific activation of iTregs in a local environment stimulates the Th17-mediated inflammatory response in a CTLA4-dependent manner.
Collapse
Affiliation(s)
- Nobumasa Watanabe
- Allergy and Immunology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Osamu Kaminuma
- Allergy and Immunology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Noriko Kitamura
- Allergy and Immunology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takachika Hiroi
- Allergy and Immunology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
- * E-mail:
| |
Collapse
|
35
|
The search for the target antigens of multiple sclerosis, part 1: autoreactive CD4+ T lymphocytes as pathogenic effectors and therapeutic targets. Lancet Neurol 2015; 15:198-209. [PMID: 26724103 DOI: 10.1016/s1474-4422(15)00334-8] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Identification of the target antigens of pathogenic antibodies and T cells is of fundamental importance for understanding the pathogenesis of multiple sclerosis, and for the development of personalised treatments for the disease. Myelin-specific CD4+ T cells emerged long ago as a key player in animal models of multiple sclerosis. Taking a forward-translational approach, autoreactive CD4+ T cells have been studied extensively in patients with multiple sclerosis, and there is evidence, but as yet no direct proof, that autoreactive CD4+ T cells are a key player in the pathogenesis of the disorder. Several therapies that selectively target myelin-specific CD4+ T cells have been investigated in clinical trials up to phase 3. So far, however, none of these (mostly underpowered) therapeutic trials have provided definitive evidence of clinical efficacy. One major obstacle to personalised, highly selective immunotherapy is the absence of standardised and reliable assays to assess antigen-specific human T-cell responses. Such assays would be essential for stratification of patients with multiple sclerosis according to their individual target antigens.
Collapse
|
36
|
Geng S, Zhang H, Zhou X, He Y, Zhang X, Xie X, Li C, He Z, Yu Q, Zhong Y, Lowrie DB, Zheng G, Wang B. Diabetes tolerogenic vaccines targeting antigen-specific inflammation. Hum Vaccin Immunother 2015; 11:522-30. [PMID: 25622092 DOI: 10.1080/21645515.2014.1004024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Tolerance controls the magnitude of inflammation, and balance between beneficial and harmful effects of inflammation is crucial for organ function and survival. Inadequate tolerance leads to various inflammatory diseases. Antigen specific tolerance is ideal for inflammation control as alternative anti-inflammatory interventions are non-specific and consequently increase the risk of infection and tumorigenesis. With inherent antigen specificity, tolerogenic vaccines are potentially ideal for control of inflammation. Although the concept of tolerogenic vaccines is still in its infancy, tolerogenic mucosal vaccines and specific immuno-therapies have long been proven effective in pioneering examples. Now a body of evidence supporting the concept of tolerogenic vaccines has also accumulated. Here we comment on recent successes of the tolerogenic vaccine concept, present new evidence with a type 1 diabetes vaccine as an example and draw conclusions on the advantages and potential for inflammatory disease control at the bedside.
Collapse
Affiliation(s)
- Shuang Geng
- a Key Laboratory of Medical Molecular Virology of MOH and MOE ; Fudan University Shanghai Medical College ; Shanghai , China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Huang MT, Lin BR, Liu WL, Lu CW, Chiang BL. Lymph node trafficking of regulatory T cells is prerequisite for immune suppression. J Leukoc Biol 2015; 99:561-8. [PMID: 26543091 DOI: 10.1189/jlb.1a0715-296r] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 10/19/2015] [Indexed: 12/13/2022] Open
Abstract
Regulatory T cells have a crucial role in health and disease because of their immune regulation function. However, the anatomic sites where regulatory T cells exert optimal immune regulation are open to debate. In our current study with the use of a shear-stress flow assay, we found that regulatory T cells exhibited significantly decreased adhesion to either activated endothelial monolayer or intercellular adhesion molecule 1 or E-selectin-coated surfaces compared with activated effector T cells. The less transmigration capacity of the regulatory T cells prompted our speculation of preferential lymph node localization for the regulatory T cells that endowed these cells with immune regulation function in the most efficient manner. To test this hypothesis, the role of lymph node localization in regulatory T cell-mediated immune suppression was evaluated with a footpad inflammation model. We found that adoptively transferred regulatory T cells inhibited the development of footpad inflammation. In addition, although blockage of CCR7 or CD62L had no effect on the immune suppressive function of the regulatory T cells per se, pretreatment of the regulatory T cells with either CCR7 or CD62L blocking antibodies prevented their recruitment into draining lymph nodes and concomitantly abrogated the immune suppressive effects of adoptively transferred regulatory T cells during footpad inflammation. Our data demonstrate the crucial role of lymph node localization in regulatory T cell-mediated immune suppression and suggest a probable hierarchy in the anatomic sites for optimal immune regulation. Elucidating the relationships between the transmigration characteristics of the regulatory T cells and their immune regulation function will provide insightful information for regulatory T cell-based cell therapy.
Collapse
Affiliation(s)
- Miao-Tzu Huang
- Departments of *Medical Research, Pediatrics, Graduate Institute of Clinical Medicine, School of Medicine, and Department of General Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Been-Ren Lin
- Departments of *Medical Research, Pediatrics, Graduate Institute of Clinical Medicine, School of Medicine, and Department of General Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Wei-Liang Liu
- Departments of *Medical Research, Pediatrics, Graduate Institute of Clinical Medicine, School of Medicine, and Department of General Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Wei Lu
- Departments of *Medical Research, Pediatrics, Graduate Institute of Clinical Medicine, School of Medicine, and Department of General Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Bor-Luen Chiang
- Departments of *Medical Research, Pediatrics, Graduate Institute of Clinical Medicine, School of Medicine, and Department of General Surgery, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
38
|
Takaiwa F, Wakasa Y, Takagi H, Hiroi T. Rice seed for delivery of vaccines to gut mucosal immune tissues. PLANT BIOTECHNOLOGY JOURNAL 2015; 13:1041-55. [PMID: 26100952 DOI: 10.1111/pbi.12423] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 05/14/2015] [Accepted: 05/23/2015] [Indexed: 05/09/2023]
Abstract
Gut-associated lymphoid tissue (GALT) is the biggest lymphoid organ in the body. It plays a role in robust immune responses against invading pathogens while maintaining immune tolerance against nonpathogenic antigens such as foods. Oral vaccination can induce mucosal and systemic antigen-specific immune reactions and has several advantages including ease of administration, no requirement for purification and ease of scale-up of antigen. Thus far, taking advantage of these properties, various plant-based oral vaccines have been developed. Seeds provide a superior production platform over other plant tissues for oral vaccines; they offer a suitable delivery vehicle to GALT due to their high stability at room temperature, ample and stable deposition space, high expression level, and protection from digestive enzymes in gut. A rice seed production system for oral vaccines was established by combining stable deposition in protein bodies or protein storage vacuoles and enhanced endosperm-specific expression. Various types of rice-based oral vaccines for infectious and allergic diseases were generated. Efficacy of these rice-based vaccines was evaluated in animal models.
Collapse
Affiliation(s)
- Fumio Takaiwa
- Functional Crop Research and Development Unit, National Institute of Agrobiological Sciences, Tsukuba, Ibaraki, Japan
| | - Yuhya Wakasa
- Functional Crop Research and Development Unit, National Institute of Agrobiological Sciences, Tsukuba, Ibaraki, Japan
| | - Hidenori Takagi
- Functional Crop Research and Development Unit, National Institute of Agrobiological Sciences, Tsukuba, Ibaraki, Japan
| | - Takachika Hiroi
- Department of Allergy and Immunology, The Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
39
|
Abstract
Antigen-specific immunotherapy is expected to be a desirable treatment for allergic diseases. Currently, antigen-specific immunotherapy is performed by administering disease-causing antigens subcutaneously or sublingually. These approaches induce long-term remission in patients with allergic rhinitis or asthma. The oral route is an alternative to subcutaneous and sublingual routes, and can also induce long-term remission, a phenomenon known as "oral tolerance." The effectiveness of oral tolerance has been reported in the context of autoimmune diseases, food allergies, asthma, atopic dermatitis, and allergic rhinitis in both human patients and animal models. However, few studies have examined its efficacy in animal models of allergic conjunctivitis. Previously, we showed that ovalbumin feeding suppressed ovalbumin-induced experimental allergic conjunctivitis, indicating the induction of oral tolerance is effective in treating experimental allergic conjunctivitis. In recent years, transgenic rice has been developed that can induce oral tolerance and reduce the severity of anaphylaxis. The major Japanese cedar pollen antigens in transgenic rice, Cryptomeria japonica 1 and C. japonica 2, were deconstructed by molecular shuffling, fragmentation, and changes in the oligomeric structure. Thus, transgenic rice may be an effective treatment for allergic conjunctivitis.
Collapse
|
40
|
Hostmann A, Meyer T, Maul J, Preiss J, Boortz B, Thiel A, Duchmann R, Ullrich R. Preexisting antigen-specific immune responses are modulated by oral KLH feeding in humans. Eur J Immunol 2015; 45:1991-6. [DOI: 10.1002/eji.201445024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 03/12/2015] [Accepted: 03/30/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Arwed Hostmann
- Charité-Campus Benjamin Franklin, Medizinische Klinik für Gastroenterologie; Infektiologie und Rheumatologie; Berlin Germany
| | - Tim Meyer
- Charité-Campus Benjamin Franklin, Medizinische Klinik für Gastroenterologie; Infektiologie und Rheumatologie; Berlin Germany
| | - Jochen Maul
- Charité-Campus Benjamin Franklin, Medizinische Klinik für Gastroenterologie; Infektiologie und Rheumatologie; Berlin Germany
| | - Jan Preiss
- Charité-Campus Benjamin Franklin, Medizinische Klinik für Gastroenterologie; Infektiologie und Rheumatologie; Berlin Germany
| | - Bertram Boortz
- Charité-Campus Benjamin Franklin, Medizinische Klinik für Gastroenterologie; Infektiologie und Rheumatologie; Berlin Germany
| | - Andreas Thiel
- Charité-Universitätsmedizin Berlin; Berlin-Brandenburg Center for Regenerative Therapies (BCRT) Virchow-Klinikum; Berlin Germany
| | - Rainer Duchmann
- Medizinische Klinik; Hospital zum Heiligen Geist; Frankfurt a. M. Germany
| | - Reiner Ullrich
- Charité-Campus Benjamin Franklin, Medizinische Klinik für Gastroenterologie; Infektiologie und Rheumatologie; Berlin Germany
| |
Collapse
|
41
|
Simmons KM, Michels AW. Type 1 diabetes: A predictable disease. World J Diabetes 2015; 6:380-390. [PMID: 25897349 PMCID: PMC4398895 DOI: 10.4239/wjd.v6.i3.380] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 11/26/2014] [Accepted: 01/12/2015] [Indexed: 02/05/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease characterized by loss of insulin producing beta cells and reliance on exogenous insulin for survival. T1D is one of the most common chronic diseases in childhood and the incidence is increasing, especially in children less than 5 years of age. In individuals with a genetic predisposition, an unidentified trigger initiates an abnormal immune response and the development of islet autoantibodies directed against proteins in insulin producing beta cells. There are currently four biochemical islet autoantibodies measured in the serum directed against insulin, glutamic decarboxylase, islet antigen 2, and zinc transporter 8. Development of islet autoantibodies occurs before clinical diagnosis of T1D, making T1D a predictable disease in an individual with 2 or more autoantibodies. Screening for islet autoantibodies is still predominantly done through research studies, but efforts are underway to screen the general population. The benefits of screening for islet autoantibodies include decreasing the incidence of diabetic ketoacidosis that can be life threatening, initiating insulin therapy sooner in the disease process, and evaluating safe and specific therapies in large randomized clinical intervention trials to delay or prevent progression to diabetes onset.
Collapse
|
42
|
Concentrated protein body product derived from rice endosperm as an oral tolerogen for allergen-specific immunotherapy--a new mucosal vaccine formulation against Japanese cedar pollen allergy. PLoS One 2015; 10:e0120209. [PMID: 25774686 PMCID: PMC4361645 DOI: 10.1371/journal.pone.0120209] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 01/20/2015] [Indexed: 01/07/2023] Open
Abstract
The endoplasmic reticulum-derived type-I protein body (PB-I) from rice endosperm cells is an ideal candidate formulation for the oral delivery of bioencapsulated peptides as tolerogens for allergen-specific immunotherapy. In the present study, PBs containing the deconstructed Japanese cedar pollen allergens Cryptomeria japonica 1 (Cry j 1) and Cry j 2 were concentrated by treatment with thermostable α-amylase at 90°C to remove the starch from milled rice powder, which resulted in a 12.5-fold reduction of dry weight compared to the starting material. The modified Cry j 1 and Cry j 2 antigens in this concentrated PB product were more resistant to enzymatic digestion than those in the milled seed powder despite the absence of intact cell wall and starch, and remained stable for at least 10 months at room temperature without detectable loss or degradation. The high resistance of these allergens could be attributed to changes in protein physicochemical properties induced by the high temperature concentration process, as suggested by the decreased solubility of the antigens and seed proteins in PBs in step-wise-extraction experiments. Confocal microscopy showed that the morphology of antigen-containing PB-Is was preserved in the concentrated PB product. The concentrated PB product induced specific immune tolerance against Cry j 1 and Cry j 2 in mice when orally administered, supporting its potential use as a novel oral tolerogen formulation.
Collapse
|
43
|
Abadie V, Jabri B. Immunopathology of Celiac Disease. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00080-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
44
|
Diogo GR, Reljic R. Development of a new tuberculosis vaccine: is there value in the mucosal approach? Immunotherapy 2014; 6:1001-13. [DOI: 10.2217/imt.14.62] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
TB is a global health problem, killing 1.5 million people every year. The only currently available vaccine, Mycobacterium bovis BCG, is effective against severe childhood forms, but it demonstrates a variable efficacy against the pulmonary form of TB in adults. Many of these adult TB cases result from the reactivation of an initially controlled, latent Mycobacterium tuberculosis infection. Effective prophylactic vaccination remains the key long-term strategy for combating TB. Continued belief in reaching this goal requires unrelenting innovation in the formulation and delivery of candidate vaccines. It is also based on the assumption, that the failure of recent human vaccine trials could have been due to a suboptimal vaccine design and delivery, and therefore should not erode the key principle that a TB vaccine is an attainable target. This report gives a brief overview of the mucosal immune system in the context of M. tuberculosis infection, and focuses on the most recent advances in the field of mucosal TB vaccine development, with a specific emphasis on subunit TB vaccines.
Collapse
Affiliation(s)
- Gil Reynolds Diogo
- St George's Hospital, Institute of Infection & Immunity, St George's University of London, London, SW17 0RE, UK
| | - Rajko Reljic
- St George's Hospital, Institute of Infection & Immunity, St George's University of London, London, SW17 0RE, UK
| |
Collapse
|
45
|
Robert S, Steidler L. Recombinant Lactococcus lactis can make the difference in antigen-specific immune tolerance induction, the Type 1 Diabetes case. Microb Cell Fact 2014; 13 Suppl 1:S11. [PMID: 25185797 PMCID: PMC4155828 DOI: 10.1186/1475-2859-13-s1-s11] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Especially in western civilizations, immune diseases that are driven by innocuous (auto- or allo-) antigens are gradually evolving to become pandemic threats. A particularly poignant example is type 1 diabetes, where young children are confronted with the perspective and consequences of total pancreatic β-cell destruction. Along these disquieting observations we find ourselves equipped with impressively accumulating molecular immunological knowledge on the ins and outs of these pathologies. Often, however, it is difficult to translate this wealth into efficacious medicines. The molecular understanding, the concept of oral tolerance induction, the benefit of using recombinant Lactococcus lactis therein and recent openings towards their clinical use may well enable turning all colors to their appropriate fields on this Rubik's cube.
Collapse
Affiliation(s)
- Sofie Robert
- Clinical and Experimental Endocrinology (CEE), KU Leuven, 3000, Leuven, Belgium
| | | |
Collapse
|
46
|
Robert S, Gysemans C, Takiishi T, Korf H, Spagnuolo I, Sebastiani G, Van Huynegem K, Steidler L, Caluwaerts S, Demetter P, Wasserfall CH, Atkinson MA, Dotta F, Rottiers P, Van Belle TL, Mathieu C. Oral delivery of glutamic acid decarboxylase (GAD)-65 and IL10 by Lactococcus lactis reverses diabetes in recent-onset NOD mice. Diabetes 2014; 63:2876-87. [PMID: 24677716 DOI: 10.2337/db13-1236] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Growing insight into the pathogenesis of type 1 diabetes (T1D) and numerous studies in preclinical models highlight the potential of antigen-specific approaches to restore tolerance efficiently and safely. Oral administration of protein antigens is a preferred method for tolerance induction, but degradation during gastrointestinal passage can impede such protein-based therapies, reducing their efficacy and making them cost-ineffective. To overcome these limitations, we generated a tolerogenic bacterial delivery technology based on live Lactococcus lactis (LL) bacteria for controlled secretion of the T1D autoantigen GAD65370-575 and the anti-inflammatory cytokine interleukin-10 in the gut. In combination with short-course low-dose anti-CD3, this treatment stabilized insulitis, preserved functional β-cell mass, and restored normoglycemia in recent-onset NOD mice, even when hyperglycemia was severe at diagnosis. Combination therapy did not eliminate pathogenic effector T cells, but increased the presence of functional CD4(+)Foxp3(+)CD25(+) regulatory T cells. These preclinical data indicate a great therapeutic potential of orally administered autoantigen-secreting LL for tolerance induction in T1D.
Collapse
Affiliation(s)
- Sofie Robert
- Clinical and Experimental Endocrinology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Conny Gysemans
- Clinical and Experimental Endocrinology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Tatiana Takiishi
- Clinical and Experimental Endocrinology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Hannelie Korf
- Clinical and Experimental Endocrinology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Isabella Spagnuolo
- Diabetes Unit, Department of Internal Medicine, Endocrine and Metabolic Sciences and Biochemistry, University of Siena and Fondazione Umberto Di Mario ONLUS, Siena, Italy
| | - Guido Sebastiani
- Diabetes Unit, Department of Internal Medicine, Endocrine and Metabolic Sciences and Biochemistry, University of Siena and Fondazione Umberto Di Mario ONLUS, Siena, Italy
| | | | | | | | - Pieter Demetter
- Department of Pathology, Université Libre de Bruxelles, Brussels, Belgium
| | - Clive H Wasserfall
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Francesco Dotta
- Diabetes Unit, Department of Internal Medicine, Endocrine and Metabolic Sciences and Biochemistry, University of Siena and Fondazione Umberto Di Mario ONLUS, Siena, Italy
| | | | - Tom L Van Belle
- Clinical and Experimental Endocrinology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Chantal Mathieu
- Clinical and Experimental Endocrinology, Katholieke Universiteit Leuven, Leuven, Belgium
| |
Collapse
|
47
|
Mundkur L, Ponnusamy T, Philip S, Rao LN, Biradar S, Deshpande V, Kumar R, Lu X, Kakkar VV. Oral dosing with multi-antigenic construct induces atheroprotective immune tolerance to individual peptides in mice. Int J Cardiol 2014; 175:340-51. [PMID: 24962340 DOI: 10.1016/j.ijcard.2014.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 04/26/2014] [Accepted: 06/01/2014] [Indexed: 12/23/2022]
Abstract
Inflammatory immune response to self-antigens plays an important role in the development of atherosclerosis. Restoring immune tolerance to self-proteins reduces the pro-inflammatory response. We previously showed that oral tolerance to a combination of two peptides is atheroprotective. In the present study we expressed epitopes from apolipoprotein B 100 (ApoB), human heat shock protein (HSP60) and Chlamydia pneumonia outer membrane protein (Cpn) in a single protein scaffold and used this multi-antigenic construct to induce tolerance to individual peptides by oral route in ApoBtm2Sgy/Ldlrtm1Her/J mice. Antigen specific tolerance to individual peptides was observed in treated animals as seen by an increase in regulatory T cells. Tolerance to the peptides resulted in a 46.5% (p=0.002) reduction in the development of atherosclerosis compared with control. Atheroprotection was associated with a significant (p<0.05) decrease in plaque inflammation and an increase in the expression of immune regulatory markers in the aorta. CD11c+ cells coexpressing CD11b and CD103 increased in lymphoid organs and were found to activate regulatory T cells and reduce effector T-cell response. Adoptive transfer of CD11c+ cells was atheroprotective. Our results suggest that atheroprotection by oral tolerance to a multi-antigenic construct is mediated by antigen specific regulatory T cells and CD11c+ cells with immune regulatory properties.
Collapse
Affiliation(s)
- Lakshmi Mundkur
- Mary and Gary Western and Tata Molecular Immunology Unit, Thrombosis Research Institute, Bangalore, India.
| | - Thiruvelselvan Ponnusamy
- Mary and Gary Western and Tata Molecular Immunology Unit, Thrombosis Research Institute, Bangalore, India
| | - Sheena Philip
- Mary and Gary Western and Tata Molecular Immunology Unit, Thrombosis Research Institute, Bangalore, India
| | - Lakshmi Narasimha Rao
- Mary and Gary Western and Tata Molecular Immunology Unit, Thrombosis Research Institute, Bangalore, India
| | - Suryakant Biradar
- Mary and Gary Western and Tata Molecular Immunology Unit, Thrombosis Research Institute, Bangalore, India
| | - Vrushali Deshpande
- Mary and Gary Western and Tata Molecular Immunology Unit, Thrombosis Research Institute, Bangalore, India
| | - Ramesh Kumar
- Mary and Gary Western and Tata Molecular Immunology Unit, Thrombosis Research Institute, Bangalore, India
| | - Xinjie Lu
- Molecular Immunology Unit, Thrombosis Research Institute, London, United Kingdom
| | - Vijay V Kakkar
- Mary and Gary Western and Tata Molecular Immunology Unit, Thrombosis Research Institute, Bangalore, India; Molecular Immunology Unit, Thrombosis Research Institute, London, United Kingdom.
| |
Collapse
|
48
|
Stremnitzer C, Manzano‐Szalai K, Starkl P, Willensdorfer A, Schrom S, Singer J, Reichart U, Akira S, Jensen‐Jarolim E. Epicutaneously applied Der p 2 induces a strong TH 2-biased antibody response in C57BL/6 mice, independent of functional TLR4. Allergy 2014; 69:741-51. [PMID: 24735481 PMCID: PMC4023119 DOI: 10.1111/all.12399] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2014] [Indexed: 12/19/2022]
Abstract
Background The major house dust mite allergen Der p 2 is a structural and functional homologue of MD-2 within the TLR4–CD14–MD-2 complex. An asthma mouse model in TLR4-deficient mice recently suggested that the allergic immune response against Der p 2 is solely dependent on TLR4 signaling. We investigated whether similar mechanisms are important for Der p 2 sensitization via the skin. Methods In an epicutaneous sensitization model, the response to recombinant Der p 2 in combination with or without lipopolysaccharide (LPS) was compared between C57BL/6 WT and TLR4-deficient mice. We further analyzed possible adjuvant function of exogenous cysteine proteases. Results Sensitization with rDer p 2 induced similar levels of allergen-specific IgG1 and IgE antibodies in both mouse strains. LPS increased the systemic (antibody levels, cytokine release by restimulated splenocytes) and local (infiltration of immune cells into the skin) Th2 immune responses, which against our expectations were stronger in the absence of functional TLR4 expression. Barrier disruption by papain, a protease with structural homology to Der p 1, did not enhance the sensitization capacity of rDer p 2. However, the presence of LPS increased the stability of rDer p 2 against the protease. Conclusion Our data suggest that rDer p 2 alone can cause a strong TH2-biased response via the skin being enhanced in the presence of LPS. This response is not reliant on functional TLR4, but vice versa TLR4 expression rather protects against epicutaneous sensitization to house dust mite allergen Der p 2.
Collapse
Affiliation(s)
- C. Stremnitzer
- Comparative Immunology and Oncology Department of Pathophysiology and Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of ViennaVienna Austria
| | - K. Manzano‐Szalai
- Comparative Medicine Messerli Research Institute of the University of Veterinary Medicine Vienna Medical University of Vienna and University ViennaVienna Austria
| | - P. Starkl
- Comparative Immunology and Oncology Department of Pathophysiology and Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of ViennaVienna Austria
| | - A. Willensdorfer
- Comparative Medicine Messerli Research Institute of the University of Veterinary Medicine Vienna Medical University of Vienna and University ViennaVienna Austria
| | - S. Schrom
- Comparative Immunology and Oncology Department of Pathophysiology and Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of ViennaVienna Austria
| | - J. Singer
- Comparative Immunology and Oncology Department of Pathophysiology and Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of ViennaVienna Austria
| | - U. Reichart
- Institute of Animal Breeding and Genetics and Biomodels Austria University of Veterinary Medicine Vienna Vienna Austria
| | - S. Akira
- Department of Host Defense Research Institute for Microbial Diseases Osaka University Osaka Japan
| | - E. Jensen‐Jarolim
- Comparative Immunology and Oncology Department of Pathophysiology and Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of ViennaVienna Austria
- Comparative Medicine Messerli Research Institute of the University of Veterinary Medicine Vienna Medical University of Vienna and University ViennaVienna Austria
| |
Collapse
|
49
|
Rosales-Mendoza S, Rubio-Infante N, Zarazúa S, Govea-Alonso DO, Martel-Gallegos G, Moreno-Fierros L. Plant-based vaccines for Alzheimer's disease: an overview. Expert Rev Vaccines 2014; 13:429-41. [PMID: 24405291 DOI: 10.1586/14760584.2014.874948] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Plants are considered advantageous platforms for biomanufacturing recombinant vaccines. This constitutes a field of intensive research and some plant-derived vaccines are expected to be marketed in the near future. In particular, plant-based production of immunogens targeting molecules with implications on the pathology of Alzheimer's has been explored over the last decade. These efforts involve targeting amyloid beta and β-secretase with several immunogen configurations that have been evaluated in test animals. The results of these developments are analyzed in this review. Perspectives on the topic are identified, such as exploring additional antigen configurations and adjuvants in order to improve immunization schemes, characterizing in detail the elicited immune responses, and immunological considerations in the achievement of therapeutic humoral responses via mucosal immunization. Safety concerns related to these therapies will also be discussed.
Collapse
Affiliation(s)
- Sergio Rosales-Mendoza
- Laboratorio de Biofarmacéuticos recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí , Av. Dr. Manuel Nava 6, SLP, 78210 , México
| | | | | | | | | | | |
Collapse
|
50
|
Macchiaverni P, Rekima A, Turfkruyer M, Mascarell L, Airouche S, Moingeon P, Adel-Patient K, Condino-Neto A, Annesi-Maesano I, Prescott SL, Tulic MK, Verhasselt V. Respiratory allergen from house dust mite is present in human milk and primes for allergic sensitization in a mouse model of asthma. Allergy 2014; 69:395-8. [PMID: 24329931 DOI: 10.1111/all.12332] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2013] [Indexed: 01/15/2023]
Abstract
There is an urgent need to identify environmental risk and protective factors in early life for the prevention of allergy. Our study demonstrates the presence of respiratory allergen from house dust mite, Der p 1, in human breast milk. Der p 1 in milk is immunoreactive, present in similar amounts as dietary egg antigen, and can be found in breast milk from diverse regions of the world. In a mouse model of asthma, oral exposure to Der p through breast milk strongly promotes sensitization rather than protect the progeny as we reported with egg antigen. These data highlight that antigen administration to the neonate through the oral route may contribute to child allergic sensitization and have important implications for the design of studies assessing early oral antigen exposure for allergic disease prevention. The up-to-now unknown worldwide presence of respiratory allergen in maternal milk allows new interpretation and design of environmental control epidemiological studies for allergic disease prevention.
Collapse
Affiliation(s)
- P. Macchiaverni
- Institute of Biomedical Sciences; University of São Paulo; São Paulo Brazil
| | - A. Rekima
- University Nice Sophia Antipolis; TIM; Nice France
| | | | - L. Mascarell
- Research and Development; Stallergenes SA; Antony France
| | - S. Airouche
- Research and Development; Stallergenes SA; Antony France
| | - P. Moingeon
- Research and Development; Stallergenes SA; Antony France
| | - K. Adel-Patient
- INRA; UR496 Immuno-Allergie Alimentaire; CEA/IBiTeC-S/SPI; CEA de Saclay; F-91191 Gif sur Yvette cedex; Paris France
| | - A. Condino-Neto
- Institute of Biomedical Sciences; University of São Paulo; São Paulo Brazil
| | - I. Annesi-Maesano
- EPAR UMR-S 707 INSERM; Paris France
- EPAR UMR-S 707 UPMC Paris6; Medical School Saint-Antoine Paris; Paris France
| | - S. L. Prescott
- School of Pediatrics and Child Health; University of Western Australia
- The International Inflammation ‘in-FLAME’ Network; Worldwide Universities Network (WUN); Perth Australia
| | - M. K. Tulic
- University Nice Sophia Antipolis; TIM; Nice France
- The International Inflammation ‘in-FLAME’ Network; Worldwide Universities Network (WUN); Perth Australia
| | - V. Verhasselt
- University Nice Sophia Antipolis; TIM; Nice France
- The International Inflammation ‘in-FLAME’ Network; Worldwide Universities Network (WUN); Perth Australia
| |
Collapse
|