151
|
Abstract
Ideal therapy for inflammatory disease in the nervous system would preserve normal immune function, while suppressing only the pathologic immune responses that damage tissue and allowing for repair. In principle, antigen-specific therapy would eradicate unwanted adaptive immune responses-antibody and T-cell mediated--while preserving the integrity of other adaptive responses to infectious agents and retaining the ability to fight malignancy. However, at this time, for multiple sclerosis (MS) we do not have compelling evidence that would support any particular dominant immune response to any specific antigen or even a limited group of antigens. In fact, there are adaptive immune responses to a wide swathe of proteins and lipids found on neurons and myelin in MS. Unless controlling a few of the known immune responses is sufficient, antigen-specific therapy in MS may not have enough of an impact to modulate clinical outcome. However, in other neuroinflammatory conditions, such as neuromyelitis optica, the adaptive immune response is highly focused. Trials of antigen-specific therapy for neuroinflammatory disease might first be tested in diseases with a more limited adaptive immune response like neuromyelitis optica. The likelihood of a significant success for this therapeutic strategy might then ensue.
Collapse
|
152
|
Liberal R, Grant CR, Longhi MS, Mieli-Vergani G, Vergani D. Regulatory T cells: Mechanisms of suppression and impairment in autoimmune liver disease. IUBMB Life 2015; 67:88-97. [PMID: 25850692 DOI: 10.1002/iub.1349] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 11/11/2015] [Indexed: 12/24/2022]
Abstract
There are three classic liver diseases with probable autoimmune etiology: primary biliary cirrhosis, primary sclerosing cholangitis, and autoimmune hepatitis. The occurrence of these autoimmune conditions is determined by the breakdown of immune-regulatory mechanisms that in health are responsible for maintaining immunological tolerance against self-antigens. Among the multiple T cell subsets with suppressive function, the regulatory T cells (Tregs), defined by the expression of CD4, the IL-2 receptor α chain (CD25), and the transcription factor FOXP3, have emerged as having a central role in maintaining immune-tolerance to autoantigens. Tregs are equipped with an array of mechanisms of suppression, including the modulation of antigen presenting cell maturation and function, the killing of target cells, the disruption of metabolic pathways, and the production of anti-inflammatory cytokines. In all the three autoimmune liver diseases mentioned above, there is evidence pointing for either a reduced frequency and/or function of Tregs. Here, we review the definition, phenotypic characteristics, and mechanisms of suppression employed by Tregs and then we discuss the evidence available pointing to their impairment in patients with autoimmune liver disease.
Collapse
Affiliation(s)
- Rodrigo Liberal
- Institute of Liver Studies and Paediatric Liver, GI & Nutrition Centre, King's College London School of Medicine at King's College Hospital, London, UK
| | | | | | | | | |
Collapse
|
153
|
Abstract
A large body of evidence produced during decades of research indicates that myocardial injury activates innate immunity. On the one hand, innate immunity both aggravates ischemic injury and impedes remodeling after myocardial infarction (MI). On the other hand, innate immunity activation contributes to myocardial healing, as exemplified by monocytes' central role in the formation of a stable scar and protection against intraventricular thrombi after acute infarction. Although innate leukocytes can recognize a wide array of self-antigens via pattern recognition receptors, adaptive immunity activation requires highly specific cooperation between antigen-presenting cells and distinct antigen-specific receptors on lymphocytes. We have only recently begun to examine lymphocyte activation's relationship to adaptive immunity and significance in the context of ischemic myocardial injury. There is some experimental evidence that CD4(+) T-cells contribute to ischemia-reperfusion injury. Several studies have shown that CD4(+) T-cells, especially CD4(+) T-regulatory cells, improve wound healing after MI, whereas depleting B-cells is beneficial post MI. That T-cell activation after MI is induced by T-cell receptor signaling implicates autoantigens that have not yet been identified in this context. Also, the significance of lymphocytes in humans post MI remains unclear, primarily as a result of methodology. This review summarizes current experimental evidence of lymphocytes' activation, functional role, and crosstalk with innate leukocytes in myocardial ischemia-reperfusion injury, wound healing, and remodeling after myocardial infarction.
Collapse
Affiliation(s)
- Ulrich Hofmann
- From the Department of Internal Medicine I, University Hospital Würzburg, and Comprehensive Heart Failure Center, University of Würzburg, Germany (U.H.); and Universitätsklinik und Poliklinik für Innere Medizin III, Universitätsklinikum Halle (Saale), Halle/Saale, Germany (S.F.).
| | - Stefan Frantz
- From the Department of Internal Medicine I, University Hospital Würzburg, and Comprehensive Heart Failure Center, University of Würzburg, Germany (U.H.); and Universitätsklinik und Poliklinik für Innere Medizin III, Universitätsklinikum Halle (Saale), Halle/Saale, Germany (S.F.).
| |
Collapse
|
154
|
Effect of a protein-free diet in the development of food allergy and oral tolerance in BALB/c mice. Br J Nutr 2015; 113:935-43. [PMID: 25759975 DOI: 10.1017/s0007114515000173] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The aim of the present study was to investigate the effect of a protein-free diet in the induction of food allergy and oral tolerance in BALB/c mice. The experimental model used was mice that were fed, since weaning up to adulthood, a balanced diet in which all dietary proteins were replaced by amino acid diet (Aa). The absence of dietary proteins did not prevent the development of food allergy to ovalbumin (OVA) in these mice. However, Aa-fed mice produced lower levels of IgE, secretory IgA and cytokines. In addition, when compared with mice from control group, Aa-fed mice had a milder aversive reaction to the allergen measured by consumption of OVA-containing solution and weight loss during food allergy development. In addition, mice that did not have dietary proteins in their diets were less susceptible to induction of oral tolerance. One single oral administration was not enough to suppress specific serum Ig and IgG1 levels in the Aa-fed group, although it was efficient to induce suppression in the control group. The present results indicate that the stimulation by dietary proteins alters both inflammatory reactivity and regulatory immune reactivity in mice probably due to their effect in the maturation of the immune system.
Collapse
|
155
|
Pasternak JA, Ng SH, Buchanan RM, Mertins S, Mutwiri GK, Gerdts V, Wilson HL. Oral antigen exposure in newborn piglets circumvents induction of oral tolerance in response to intraperitoneal vaccination in later life. BMC Vet Res 2015; 11:50. [PMID: 25889479 PMCID: PMC4357157 DOI: 10.1186/s12917-015-0350-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 02/05/2015] [Indexed: 11/10/2022] Open
Abstract
Background We previously determined that newborn piglets orally gavaged with Ovalbumin (OVA) responded to systemic OVA re-exposure with tolerance; if adjuvants were included in oral vaccine, piglets responded with antibody-mediated immunity (Vet Immunol Immunopathol 161(3–4):211–21, 2014). Here, we will investigate whether newborn piglets gavaged with a vaccine comprised of OVA plus unmethylated CpG oligodeoxynucleotides (CpG; soluble component; OVA/CpG) combined with OVA plus CpG encapsulated within polyphosphazene microparticles (MP; particulate component) responded with systemic and mucosal immunity. To monitor the response to systemic antigen re-exposure, piglets were i.p.-immunized with OVA plus Incomplete Freund’s Adjuvant (IFA) one month later. Results Newborn piglets (n = 5/group) were gavaged with a combined soluble and particulate vaccine consisting of OVA (0.5-0.05 mg) plus 50 μg CpG and 0.5 mg OVA plus 50 μg CpG encapsulated within a polyphosphazene MP (0.5 mg) referred to as OVA/CpG + MP. Control piglets were gavaged with saline alone. Piglets were i.p. immunized with 10 mg OVA (or saline) in IFA at four weeks of age and then euthanized at eight weeks of age. We observed significantly higher titres of serum anti-OVA immunoglobulin (Ig) IgM, IgA, IgG, IgG1, IgG2 and IgG in piglets immunized with 0.05 mg OVA/CpG + MP relative to saline control animals. Thus, a single oral exposure at birth to a combined soluble and particulate OVA vaccine including adjuvants can circumvent induction of oral tolerance which impacts response to i.p. vaccination in later life. Further, piglets gavaged with 0.05 mg OVA/CpG + MP generated significant anti-OVA IgG and IgG1 titres in lung compared to saline control piglets but results were comparable to titres measured in parenteral control piglets. Peripheral blood mononuclear cells (PBMCs) ex vivo-stimulated with OVA showed markedly decreased production of IL-10 cytokine after 72 hours relative to animal-matched cells incubated with media alone. No production of IFN-γ was observed from any groups. Conclusion Newborn piglets gavaged with low dose soluble and particulate OVA plus CpG ODN and polyphosphazene adjuvants produced antigen-specific antibodies in serum and lung after systemic re-exposure in later life. These data indicate circumvention of oral tolerance but not induction of oral immunity.
Collapse
Affiliation(s)
- J Alex Pasternak
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK, S7N 5E3, Canada.
| | - Siew Hon Ng
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK, S7N 5E3, Canada.
| | - Rachelle M Buchanan
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK, S7N 5E3, Canada.
| | - Sonja Mertins
- Current address: Klinikum der Universität zu Köln, Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Goldenfelsstraße 19-21, 50935, Köln, Germany.
| | - George K Mutwiri
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK, S7N 5E3, Canada.
| | - Volker Gerdts
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK, S7N 5E3, Canada.
| | - Heather L Wilson
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK, S7N 5E3, Canada.
| |
Collapse
|
156
|
Oliveira RP, Santiago AF, Ficker SM, Gomes-Santos AC, Faria AMC. Antigen administration by continuous feeding enhances oral tolerance and leads to long-lasting effects. J Immunol Methods 2015; 421:36-43. [PMID: 25707356 DOI: 10.1016/j.jim.2015.02.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/14/2015] [Accepted: 02/15/2015] [Indexed: 11/16/2022]
Abstract
The ability to avoid inflammatory responses to dietary components and microbiota antigens in the gut mucosa is achieved by a mechanism termed oral tolerance. This phenomenon is crucial to maintain the physiological immune activity in the gut and to prevent inflammatory disorders such as food allergy and inflammatory bowel diseases. Moreover, orally administered antigens induce regulatory cells that control systemic inflammatory responses as well. Given its specific, systemic and long-lasting effects, oral tolerance represents a promising approach for immunotherapies that aim to modulate inflammatory and autoimmune diseases. However, there are different protocols of feeding for induction of oral tolerance, and they have an impact in tolerance efficiency and length. Herein, we present and discuss different experimental feeding protocols and how they influence the outcome of oral administration of antigens.
Collapse
Affiliation(s)
- Rafael Pires Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Andrezza Fernanda Santiago
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Sabine Madsen Ficker
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Cristina Gomes-Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Maria Caetano Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
157
|
Ruff KJ, Durham PL, O’Reilly A, Long FD. Eggshell membrane hydrolyzates activate NF-κB in vitro: possible implications for in vivo efficacy. J Inflamm Res 2015; 8:49-57. [PMID: 25709492 PMCID: PMC4332312 DOI: 10.2147/jir.s78118] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
PURPOSE Eggshell membrane (ESM) has been shown to contain naturally occurring bioactive components, and biological activities such as reducing proinflammatory cytokines, liver fibrosis, and joint pain in osteoarthritis sufferers have also been reported for ESM matrix as a whole. Nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-κB) is a signaling protein found in the cytoplasm of nearly all human and animal cell types and is a primary regulator of immune function. The studies reported herein were designed to investigate the possible role that NF-κB activity might play in the reported biological activities of ESM. METHODS Three ESM hydrolyzates produced via fermentation, enzymatic, or chemical hydrolysis were evaluated in vitro in either human peripheral blood mononuclear cell or THP-1 (human leukemic monocyte) cell cultures for NF-κB activity following 4-hour exposure. The hydrolyzates were compared with untreated control cells or cells incubated with lipopolysaccharide or ascorbic acid. The source of ESM activity was also evaluated. RESULTS NF-κB levels were increased above levels found in untreated cells at all three dilutions (1:100, 1:1,000, and 1:10,000) for the fermentation hydrolyzate of ESM (ESM-FH) (P=0.021, P=0.020, P=0.009, respectively) in peripheral blood mononuclear cells. The enzymatic hydrolyzate of ESM (ESM-EH) also produced statistically significant levels of activated NF-κB at the 1:100 and 1:1,000 dilutions (P=0.004, P=0.006, respectively) but fell just shy of significance at the 1:10,000 dilution (P=0.073). Similarly, ESM-FH (P=0.021, P=0.002) and ESM-EH (P=0.007, P=0.007) activated NF-κB in THP-1 cells at 1:1,000 and 1:10,000 dilutions, respectively. The chemical hydrolyzate of ESM (ESM-CH) showed statistically significant levels of activation at the 1:1,000 dilution (P=0.005) but failed to differ from untreated cells at the 1:10,000 dilution (P=0.193) in THP-1 cells. CONCLUSION Results from our studies provide evidence that ESM hydrolyzates significantly activate NF-κB, and the source of this activity was investigated to confirm that it is inherent to ESM and not derived from bacterial contamination. Based on our findings, we propose a plausible hypothesis as to how increased NF-κB activity might translate into the in vivo efficacy that has been observed with ESM via an "oral tolerance" mechanism.
Collapse
Affiliation(s)
| | - Paul L Durham
- Center for Biomedical and Life Sciences, Missouri State University, Springfield, MO, USA
| | - Austin O’Reilly
- Center for Biomedical and Life Sciences, Missouri State University, Springfield, MO, USA
| | | |
Collapse
|
158
|
Xu Y, Tang H, Wang H, Liu Y. Blockade of oral tolerance to ovalbumin in mice by silver nanoparticles. Nanomedicine (Lond) 2015; 10:419-31. [DOI: 10.2217/nnm.14.111] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Aim: The authors investigated the effect of silver nanoparticles (Ag NPs) on oral tolerance to dietary antigen ovalbumin (OVA) in mice. Materials & methods: Ag NPs were orally administered for 7 days before feeding of OVA and the immunological responses were assayed. Results: Polyvinyl pyrrolidone-coated Ag NPs (Ag NPs-PVP) were found to block the induction of oral tolerance to OVA. The physicochemical properties of the three kinds of Ag NPs were assessed and silver ions were found to inhibit the induction of oral tolerance, which might be the dominating factor in the blockade of oral tolerance by Ag NPs-PVP. Conclusion: Ag NPs with certain physicochemical properties can block oral tolerance to dietary antigen that may induce food allergy in humans.
Collapse
Affiliation(s)
- Yingying Xu
- Beijing National Laboratory for Molecular Sciences, Department of Chemical Biology, College of Chemistry & Molecular Engineering, Peking University, Beijing, China
| | - Huan Tang
- Beijing National Laboratory for Molecular Sciences, Department of Chemical Biology, College of Chemistry & Molecular Engineering, Peking University, Beijing, China
| | - Haifang Wang
- Institute of Nanochemistry & Nanobiology, Shanghai University, Shanghai, China
| | - Yuanfang Liu
- Beijing National Laboratory for Molecular Sciences, Department of Chemical Biology, College of Chemistry & Molecular Engineering, Peking University, Beijing, China
- Institute of Nanochemistry & Nanobiology, Shanghai University, Shanghai, China
| |
Collapse
|
159
|
Jones RGA, Martino A. Targeted localized use of therapeutic antibodies: a review of non-systemic, topical and oral applications. Crit Rev Biotechnol 2015; 36:506-20. [PMID: 25600465 DOI: 10.3109/07388551.2014.992388] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Therapeutic antibodies provide important tools in the "medicine chest" of today's clinician for the treatment of a range of disorders. Typically monoclonal or polyclonal antibodies are administered in large doses, either directly or indirectly into the circulation, via a systemic route which is well suited for disseminated ailments. Diseases confined within a specific localized tissue, however, may be treated more effectively and at reduced cost by a delivery system which targets directly the affected area. To explore the advantages of the local administration of antibodies, we reviewed current alternative, non-systemic delivery approaches which are in clinical use, being trialed or developed. These less conventional approaches comprise: (a) local injections, (b) topical and (c) peroral administration routes. Local delivery includes intra-ocular injections into the vitreal humor (i.e. Ranibizumab for age-related macular degeneration), subconjunctival injections (e.g. Bevacizumab for corneal neovascularization), intra-articular joint injections (i.e. anti-TNF alpha antibody for persistent inflammatory monoarthritis) and intratumoral or peritumoral injections (e.g. Ipilimumab for cancer). A range of other strategies, such as the local use of antibacterial antibodies, are also presented. Local injections of antibodies utilize doses which range from 1/10th to 1/100th of the required systemic dose therefore reducing both side-effects and treatment costs. In addition, any therapeutic antibody escaping from the local site of disease into the systemic circulation is immediately diluted within the large blood volume, further lowering the potential for unwanted effects. Needle-free topical application routes become an option when the condition is restricted locally to an external surface. The topical route may potentially be utilized in the form of eye drops for infections or corneal neovascularization or be applied to diseased skin for psoriasis, dermatitis, pyoderma gangrenosum, antibiotic resistant bacterial infections or ulcerated wounds. Diseases confined to the gastrointestinal tract can be targeted directly by applying antibody via the injection-free peroral route. The gastrointestinal tract is unusual in that its natural immuno-tolerant nature ensures the long-term safety of repeatedly ingesting heterologous antiserum or antibody materials. Without the stringent regulatory, purity and clean room requirements of manufacturing parenteral (injectable) antibodies, production costs are minimal, with the potential for more direct low-cost targeting of gastrointestinal diseases, especially with those caused by problematic antibiotic resistant or toxigenic bacteria (e.g. Clostridium difficile, Helicobacter pylori), viruses (e.g. rotavirus, norovirus) or inflammatory bowel disease (e.g. ulcerative colitis, Crohn's disease). Use of the oral route has previously been hindered by excessive antibody digestion within the gastrointestinal tract; however, this limitation may be overcome by intelligently applying one or more strategies (i.e. decoy proteins, masking therapeutic antibody cleavage sites, pH modulation, enzyme inhibition or encapsulation). These aspects are additionally discussed in this review and novel insights also provided. With the development of new applications via local injections, topical and peroral routes, it is envisaged that an extended range of ailments will increasingly fall within the clinical scope of therapeutic antibodies further expanding this market.
Collapse
Affiliation(s)
| | - Angela Martino
- a Department of Chemistry , University of Warwick , Coventry , UK
| |
Collapse
|
160
|
Batista NV, Pereira RVS, Noviello MLM, Dourado LPA, Perez DA, Foureaux G, Ferreira AJ, Ferreira AVM, Cara DC. Prolonged ingestion of ovalbumin diet by sensitized mice improves the metabolic consequences induced by experimental food allergy. Clin Exp Immunol 2015; 178:416-27. [PMID: 25112154 DOI: 10.1111/cei.12435] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2014] [Indexed: 01/22/2023] Open
Abstract
The prevalence of food allergy is rising in the western world. Allergen restriction is the chosen treatment in this condition, but continuous ingestion of the antigen has shown positive results in clinical trials. In a previous study, we have shown several allergic and metabolic alterations after 7 days of ovalbumin (OVA) ingestion by sensitized mice. The aim of this study was to investigate whether prolonged ingestion of antigen by sensitized mice would reverse the metabolic consequences caused by experimental food allergy. For this, allergic and metabolic parameters were analysed after prolonged ingestion of an OVA diet by OVA-sensitized mice. As shown previously, after 7 days of OVA consumption, sensitized mice showed increased serum levels of anti-OVA immunoglobulin (Ig)E and IgG1, aversion to the antigen ingestion, marked body and adipose tissue weight loss, followed by adipose tissue inflammation and decreased serum levels of adipokines, glucose and triglycerides. However, after 14 days of oral challenge, sensitized mice showed an anti-OVA IgE level similar to the mice that were only sensitized, but the specific IgG1 did not change. With this prolonged ingestion of OVA, sensitized mice were protected from OVA-induced anaphylaxis when the antigen was given systemically at a dose of 2 mg/animal. Moreover, various parameters analysed were significantly ameliorated, including adipose tissue inflammation, body and adipose tissue loss, as well as serum levels of adipokines and triglycerides. Therefore, our data suggest that prolonged ingestion of OVA by sensitized mice results in an improvement of the metabolic consequences caused by experimental food allergy.
Collapse
Affiliation(s)
- N V Batista
- Department of Biochemistry and Immunology, Biological Sciences Institute, Belo Horizonte, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
161
|
Fukuda K, Ishida W, Harada Y, Wakasa Y, Takagi H, Takaiwa F, Fukushima A. Prevention of allergic conjunctivitis in mice by a rice-based edible vaccine containing modified Japanese cedar pollen allergens. Br J Ophthalmol 2015; 99:705-9. [PMID: 25563761 DOI: 10.1136/bjophthalmol-2014-305842] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 12/14/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND/AIMS To determine whether oral immunotherapy with transgenic rice seeds expressing hypoallergenic modified antigens suppresses cedar pollen-induced allergic conjunctivitis by eliciting immune tolerance in mice. METHODS BALB/c mice were fed once a day for 20 days with 220 mg of transgenic rice expressing modified Japanese cedar pollen allergens Cry j 1 and Cry j 2 or with non-transgenic rice seeds as a control. They were then sensitised with two intraperitoneal injections of Japanese cedar pollen in alum before challenge twice with pollen in eye drops. Twenty-four hours after the second challenge, the conjunctiva, spleen, and blood were isolated for histological analysis, cytokine production assays, and measurement of serum immunoglobulin E concentrations, respectively. RESULTS The numbers of eosinophils and total inflammatory cells in the conjunctiva were significantly lower in mice fed the transgenic rice than in those fed non-transgenic rice. The clinical score evaluated at 15 min after antigen challenge was also significantly lower in mice fed the transgenic rice than in those fed non-transgenic rice. The serum concentrations of both total and allergen-specific immunoglobulin E were also significantly lower in mice fed the transgenic rice. Oral vaccination with transgenic rice resulted in significant down-regulation of the allergen-induced production of interleukin (IL)-2, IL-4, IL-5, IL-12p70, interferon-γ, and IL-17A by splenocytes. CONCLUSIONS Oral immunotherapy with transgenic rice expressing modified Japanese cedar pollen allergens suppressed pollen-induced experimental allergic conjunctivitis in mice by eliciting immune tolerance. This novel prophylactic approach is potentially safe and effective for allergen-specific oral immunotherapy in allergic conjunctivitis.
Collapse
Affiliation(s)
- Ken Fukuda
- Department of Ophthalmology and Visual Science, Kochi Medical School, Nankoku City, Kochi, Japan
| | - Waka Ishida
- Department of Ophthalmology and Visual Science, Kochi Medical School, Nankoku City, Kochi, Japan
| | - Yosuke Harada
- Department of Ophthalmology and Visual Science, Kochi Medical School, Nankoku City, Kochi, Japan
| | - Yuhya Wakasa
- Functional Transgenic Crop Research Unit, National Institute of Agrobiological Sciences, Tsukuba, Ibaraki, Japan
| | - Hidenori Takagi
- Functional Transgenic Crop Research Unit, National Institute of Agrobiological Sciences, Tsukuba, Ibaraki, Japan
| | - Fumio Takaiwa
- Functional Transgenic Crop Research Unit, National Institute of Agrobiological Sciences, Tsukuba, Ibaraki, Japan
| | - Atsuki Fukushima
- Department of Ophthalmology and Visual Science, Kochi Medical School, Nankoku City, Kochi, Japan
| |
Collapse
|
162
|
Increasing small intestinal permeability worsens colitis in the IL-10-/- mouse and prevents the induction of oral tolerance to ovalbumin. Inflamm Bowel Dis 2015; 21:8-18. [PMID: 25517593 DOI: 10.1097/mib.0000000000000253] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Increased intestinal permeability is found in noninflamed portions of the gut of inflammatory bowel disease patients and in their first-degree relatives, suggesting that it is not a consequence of inflammation. Additionally, increased small intestinal permeability precedes colonic disease in animal models of inflammatory bowel disease. However, it is not known how small intestinal permeability modulates disease in the colon. The aim of this study was to determine if increasing small intestinal permeability modulates colonic inflammation in interleukin (IL)-10 mice and if an increase in permeability is sufficient to prevent oral tolerance to a dietary antigen. METHODS IL-10 mice were treated with the zonula occludens toxin pathway agonist AT-1002 for 8 weeks, and colitis severity was measured at 12 weeks of age. Wild-type mice were also treated with AT-1002 and fed ovalbumin (OVA) to determine the local and systemic immune response to this antigen under increased small intestinal permeability conditions. RESULTS IL-10 mice treated with AT-1002 showed exacerbated colitis at 12 weeks of age. AT-1002 also induced a significant OVA-specific humoral response compared with mice that received OVA alone. In addition, the intestinal production of IL-10 and TGF-β in response to oral OVA was prevented when OVA was given with AT-1002. CONCLUSIONS Increasing small intestinal permeability worsens colitis in IL-10 mice, and it prevents the development of oral tolerance to OVA in wild-type mice. This study suggests that small intestinal permeability is not merely a consequence of inflammation but a condition that leads to two of the main pathological features of inflammatory bowel disease.
Collapse
|
163
|
Kant CD, Akiyama Y, Tanaka K, Shea S, Yamada Y, Connolly SE, Marino J, Tocco G, Benichou G. Both rejection and tolerance of allografts can occur in the absence of secondary lymphoid tissues. THE JOURNAL OF IMMUNOLOGY 2014; 194:1364-71. [PMID: 25535285 DOI: 10.4049/jimmunol.1401157] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In this study, we showed that aly/aly mice, which are devoid of lymph nodes and Peyer's patches, acutely rejected fully allogeneic skin and heart grafts. They mounted potent inflammatory direct alloresponses but failed to develop indirect alloreactivity after transplantation. Remarkably, skin allografts also were rejected acutely by splenectomized aly/aly (aly/aly-spl(-)) mice devoid of all secondary lymphoid organs. In these recipients, the rejection was mediated by alloreactive CD8(+) T cells presumably primed in the bone marrow. In contrast, cardiac transplants were not rejected by aly/aly-spl(-) mice. Actually, aly/aly-spl(-) mice that spontaneously accepted a heart allotransplant and displayed donor-specific tolerance also accepted skin grafts from the same, but not a third-party, donor via a mechanism involving CD4(+) regulatory T cells producing IL-10 cytokine. Therefore, direct priming of alloreactive T cells, as well as rejection and regulatory tolerance of allogeneic transplants, can occur in recipient mice lacking secondary lymphoid organs.
Collapse
Affiliation(s)
- Cavit D Kant
- Transplantation Research Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Yoshinobu Akiyama
- Transplantation Research Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Katsunori Tanaka
- Transplantation Research Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Susan Shea
- Transplantation Research Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Yohei Yamada
- Transplantation Research Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Sarah E Connolly
- Transplantation Research Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Jose Marino
- Transplantation Research Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Georges Tocco
- Transplantation Research Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Gilles Benichou
- Transplantation Research Center, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| |
Collapse
|
164
|
The mucosal immune system for vaccine development. Vaccine 2014; 32:6711-23. [DOI: 10.1016/j.vaccine.2014.08.089] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 08/28/2014] [Indexed: 12/16/2022]
|
165
|
Wilson HL, Obradovic MR. Evidence for a common mucosal immune system in the pig. Mol Immunol 2014; 66:22-34. [PMID: 25242212 PMCID: PMC7132386 DOI: 10.1016/j.molimm.2014.09.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 08/15/2014] [Accepted: 09/01/2014] [Indexed: 12/03/2022]
Abstract
There is evidence that the common mucosal immune system exists in pigs. Vaccination at an easily accessible mucosal site may assist in providing protection at other mucosal sites. Local and distal mucosal sites should be sampled after vaccinations to define the optimal dose and formulation which promotes the common mucosal immune system in pigs.
The majority of lymphocytes activated at mucosal sites receive instructions to home back to the local mucosa, but a portion also seed distal mucosa sites. By seeding distal sites with antigen-specific effector or memory lymphocytes, the foundation is laid for the animal's mucosal immune system to respond with a secondary response should to this antigen be encountered at this site in the future. The common mucosal immune system has been studied quite extensively in rodent models but less so in large animal models such as the pig. Reasons for this paucity of reported induction of the common mucosal immune system in this species may be that distal mucosal sites were examined but no induction was observed and therefore it was not reported. However, we suspect that the majority of investigators simply did not sample distal mucosal sites and therefore there is little evidence of immune response induction in the literature. It is our hope that more pig immunologists and infectious disease experts who perform mucosal immunizations or inoculations on pigs will sample distal mucosal sites and report their findings, whether results are positive or negative. In this review, we highlight papers that show that immunization/inoculation using one route triggers mucosal immune system induction locally, systemically, and within at least one distal mucosal site. Only by understanding whether immunizations at one site triggers immunity throughout the common mucosal immune system can we rationally develop vaccines for the pig, and through these works we can gather evidence about the mucosal immune system that may be extrapolated to other livestock species or humans.
Collapse
Affiliation(s)
- Heather L Wilson
- Vaccine and Infectious Disease Organization (VIDO), Home of the International Vaccine Centre (InterVac), 120 Veterinary Road, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E3, Canada.
| | - Milan R Obradovic
- Vaccine and Infectious Disease Organization (VIDO), Home of the International Vaccine Centre (InterVac), 120 Veterinary Road, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E3, Canada.
| |
Collapse
|
166
|
Tunis MC, Marshall JS. Toll-like receptor 2 as a regulator of oral tolerance in the gastrointestinal tract. Mediators Inflamm 2014; 2014:606383. [PMID: 25309051 PMCID: PMC4182894 DOI: 10.1155/2014/606383] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 09/02/2014] [Accepted: 09/04/2014] [Indexed: 02/07/2023] Open
Abstract
Food allergy, other adverse immune responses to foods, inflammatory bowel disease, and eosinophilic esophagitis have become increasingly common in the last 30 years. It has been proposed in the "hygiene hypothesis" that dysregulated immune responses to environmental microbial stimuli may modify the balance between tolerance and sensitization in some patients. Of the pattern recognition receptors that respond to microbial signals, toll-like receptors (TLRs) represent the most investigated group. The relationship between allergy and TLR activation is currently at the frontier of immunology research. Although TLR2 is abundant in the mucosal environment, little is known about the complex relationship between bystander TLR2 activation by the commensal microflora and the processing of oral antigens. This review focuses on recent advances in our understanding of the relationship between TLR2 and oral tolerance, with an emphasis on regulatory T cells, eosinophils, B cells, IgA, intestinal regulation, and commensal microbes.
Collapse
Affiliation(s)
- Matthew C. Tunis
- Department of Microbiology and Immunology, Dalhousie University, 5850 College Street, Halifax, NS, Canada B3H 1X5
- Dalhousie Inflammation Group, Dalhousie University, 5850 College Street, Halifax, NS, Canada B3H 1X5
| | - Jean S. Marshall
- Department of Microbiology and Immunology, Dalhousie University, 5850 College Street, Halifax, NS, Canada B3H 1X5
- Dalhousie Inflammation Group, Dalhousie University, 5850 College Street, Halifax, NS, Canada B3H 1X5
| |
Collapse
|
167
|
Xiong Y, Svingen PA, Sarmento OO, Smyrk TC, Dave M, Khanna S, Lomberk GA, Urrutia RA, Faubion WA. Differential coupling of KLF10 to Sin3-HDAC and PCAF regulates the inducibility of the FOXP3 gene. Am J Physiol Regul Integr Comp Physiol 2014; 307:R608-R620. [PMID: 24944246 PMCID: PMC4166759 DOI: 10.1152/ajpregu.00085.2014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 06/13/2014] [Indexed: 12/30/2022]
Abstract
Inducible gene expression, which requires chromatin remodeling on gene promoters, underlies the epigenetically inherited differentiation program of most immune cells. However, chromatin-mediated mechanisms that underlie these events in T regulatory cells remain to be fully characterized. Here, we report that inducibility of FOXP3, a key transcription factor for the development of T regulatory cells, depends upon Kruppel-like factor 10 (KLF10) interacting with two antagonistic histone-modifying systems. We utilized chromatin immunoprecipitation, genome-integrated reporter assays, and functional domain KLF10 mutant proteins, to characterize reciprocal interactions between this transcription factor and either the Sin3-histone deacetylase complex or the histone acetyltransferase, p300/CBP-associated factor (PCAF). We characterize a Sin3-interacting repressor domain on the NH2 terminus of KLF10, which works to limit the activating function of this transcription factor. Indeed, inactivation of this Sin3-interacting domain renders KLF10 able to physically associate with PCAF as to induce FOXP3 gene transcription. We show that this biochemical data derived from studying our genome-integrated reporter cell system are recapitulated in primary murine lymphocytes. Collectively, these results advance our understanding of how a single transcription factor, namely KLF10, functions as a toggle to integrate antagonistic signals regulating FOXP3 and, thus, immune activation.
Collapse
Affiliation(s)
- Yuning Xiong
- Epigenetics and Chromatin Dynamics Laboratory, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Phyllis A Svingen
- Epigenetics and Chromatin Dynamics Laboratory, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Olga O Sarmento
- Epigenetics and Chromatin Dynamics Laboratory, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Thomas C Smyrk
- Department of Anatomic Pathology, Mayo Clinic, Rochester, Minnesota
| | - Maneesh Dave
- Epigenetics and Chromatin Dynamics Laboratory, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Sahil Khanna
- Epigenetics and Chromatin Dynamics Laboratory, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Gwen A Lomberk
- Epigenetics and Chromatin Dynamics Laboratory, Mayo Clinic, Rochester, Minnesota; Translational Epigenomic Program, Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota; and
| | - Raul A Urrutia
- Epigenetics and Chromatin Dynamics Laboratory, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota; Translational Epigenomic Program, Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota; and
| | - William A Faubion
- Epigenetics and Chromatin Dynamics Laboratory, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota; Translational Epigenomic Program, Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota; and
| |
Collapse
|
168
|
Pasternak JA, Ng SH, Wilson HL. A single, low dose oral antigen exposure in newborn piglets primes mucosal immunity if administered with CpG oligodeoxynucleotides and polyphosphazene adjuvants. Vet Immunol Immunopathol 2014; 161:211-21. [PMID: 25194591 DOI: 10.1016/j.vetimm.2014.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Revised: 08/05/2014] [Accepted: 08/12/2014] [Indexed: 10/24/2022]
Abstract
By definition, soluble antigens ingested orally trigger mucosal tolerance such that any subsequent re-exposure by a systemic route results in suppression of immunity. We propose that antigens introduced in extreme early life can readily traverse the gut wall and therefore circumvent induction of mucosal tolerance and instead induce immunity. Piglets were drenched with low-doses of ovalbumin (OVA; 5mg or 0.05 mg) alone, OVA plus adjuvants (CpG oligodeoxynucleotides and PCEP polyphosphazene) or saline within 6h of birth. At 28 days of age, they were administered 10mg OVA plus 1:1 Montanide adjuvant (or saline) via the intraperitoneal (i.p.) route or via the oral route. Serum was obtained on day 28 and day 49 to measure OVA-specific antibodies titres. All piglets boosted orally with OVA plus Montanide, regardless of prior OVA exposure, failed to induce immunity. As expected, piglets drenched with saline but boosted via the i.p. route with OVA plus Montanide showed significant induction of anti-OVA IgA, IgG, IgG1 and IgG2 relative to saline control piglets. Newborn animals drenched with 5mg or 0.05 mg OVA failed to induce oral immunity. A second intramuscular injection in adulthood triggered immunity in the piglets that were drenched with 0.05 mg OVA and boosted initially by the i.p. route suggesting that some systemic lymphocytes were primed despite initial lack of induction of humoral immunity. In contrast, piglets orally immunized with 5mg or 0.05 mg OVA plus adjuvants resulted in significant induction of anti-OVA IgA (5mg only), IgM, IgG, IgG1 and IgG2 in serum relative to saline control piglets as well as significant induction of anti-OVA IgA, IgM (5mg only) IgG, IgG1 (5mg only) or IgG2 relative to piglets drenched with OVA alone. These data clearly show that the response was sensitive to the oral vaccine components and was not simply a response to the i.p. immunization at day 28. This work demonstrates that newborn piglets respond to oral antigens with immunity if re-exposure to the antigen occurs via a systemic route and if adjuvants are included with the oral vaccine administered at birth. These results should be further explored to establish whether early life oral vaccination can be exploited to protect this susceptible population against infectious diseases.
Collapse
Affiliation(s)
- J Alex Pasternak
- Vaccine and Infectious Disease Organization, home of the International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK, S7N 5E3, Canada.
| | - Siew Hon Ng
- Vaccine and Infectious Disease Organization, home of the International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK, S7N 5E3, Canada.
| | - Heather L Wilson
- Vaccine and Infectious Disease Organization, home of the International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK, S7N 5E3, Canada.
| |
Collapse
|
169
|
Luerce TD, Gomes-Santos AC, Rocha CS, Moreira TG, Cruz DN, Lemos L, Sousa AL, Pereira VB, de Azevedo M, Moraes K, Cara DC, LeBlanc JG, Azevedo V, Faria AMC, Miyoshi A. Anti-inflammatory effects of Lactococcus lactis NCDO 2118 during the remission period of chemically induced colitis. Gut Pathog 2014; 6:33. [PMID: 25110521 PMCID: PMC4126083 DOI: 10.1186/1757-4749-6-33] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 07/20/2014] [Indexed: 02/07/2023] Open
Abstract
Background Many probiotic bacteria have been described as promising tools for the treatment and prevention of inflammatory bowel diseases (IBDs). Most of these bacteria are lactic acid bacteria, which are part of the healthy human microbiota. However, little is known about the effects of transient bacteria present in normal diets, including Lactococcus lactis. Methods In the present study, we analysed the immunomodulatory effects of three L. lactis strains in vitro using intestinal epithelial cells. L. lactis NCDO 2118 was administered for 4 days to C57BL/6 mice during the remission period of colitis induced by dextran sodium sulphate (DSS). Results Only one strain, L. lactis NCDO 2118, was able to reduce IL-1β-induced IL-8 secretion in Caco-2 cells, suggesting a potential anti-inflammatory effect. Oral treatment using L. lactis NCDO 2118 resulted in a milder form of recurrent colitis than that observed in control diseased mice. This protective effect was not attributable to changes in secretory IgA (sIgA); however, NCDO 2118 administration was associated with an early increase in IL-6 production and sustained IL-10 production in colonic tissue. Mice fed L. lactis NCDO 2118 had an increased number of regulatory CD4+ T cells (Tregs) bearing surface TGF-β in its latent form (Latency-associated peptide-LAP) in the mesenteric lymph nodes and spleen. Conclusions Here, we identified a new probiotic strain with a potential role in the treatment of IBD, and we elucidated some of the mechanisms underlying its anti-inflammatory effect.
Collapse
Affiliation(s)
- Tessalia Diniz Luerce
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627 - 31270-901 Belo Horizonte, MG, Brazil
| | - Ana Cristina Gomes-Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Clarissa Santos Rocha
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627 - 31270-901 Belo Horizonte, MG, Brazil
| | - Thais Garcias Moreira
- Departamento de Ciência de Alimentos, Faculdade de Farmácia, Belo Horizonte, MG, Brazil
| | - Déborah Nogueira Cruz
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luísa Lemos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Adna Luciana Sousa
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vanessa Bastos Pereira
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627 - 31270-901 Belo Horizonte, MG, Brazil
| | - Marcela de Azevedo
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627 - 31270-901 Belo Horizonte, MG, Brazil
| | - Kátia Moraes
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627 - 31270-901 Belo Horizonte, MG, Brazil
| | - Denise Carmona Cara
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jean Guy LeBlanc
- Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán, Argentina
| | - Vasco Azevedo
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627 - 31270-901 Belo Horizonte, MG, Brazil
| | - Ana Maria Caetano Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Anderson Miyoshi
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627 - 31270-901 Belo Horizonte, MG, Brazil
| |
Collapse
|
170
|
Hetta HF, Mehta MJ, Shata MTM. Gut immune response in the presence of hepatitis C virus infection. World J Immunol 2014; 4:52-62. [DOI: 10.5411/wji.v4.i2.52] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 05/22/2014] [Accepted: 06/20/2014] [Indexed: 02/05/2023] Open
Abstract
Hepatitis C virus (HCV) is an important etiologic agent of hepatitis and a major cause of chronic liver infection that often leads to cirrhosis, fibrosis and hepatocellular carcinoma. Although, HCV is a hepatotropic virus, there is strong evidence that HCV could replicate extra-hepatic in the gastrointestinal tissue which could serve as a reservoir for HCV. The outcome of HCV infection depends mainly on the host innate and adaptive immune responses. Innate immunity against HCV includes mainly nuclear factor cells and activation of IFN-related genes. There is an immunologic link between the gut and the liver through a population of T-cells that are capable of homing to both the liver and gut via the portal circulation. However, little is known on the role of Gut immune response in HCV. In this review we discussed the immune regulation of Gut immune cells and its association with HCV pathogenesis, various outcomes of anti-HCV therapy, viral persistence and degree of liver inflammation. Additionally, we investigated the relationship between Gut immune responses to HCV and IL28B genotypes, which were identified as a strong predictor for HCV pathogenesis and treatment outcome after acute infection.
Collapse
|
171
|
Nussenblatt RB, Lee RW, Chew E, Wei L, Liu B, Sen HN, Dick AD, Ferris FL. Immune responses in age-related macular degeneration and a possible long-term therapeutic strategy for prevention. Am J Ophthalmol 2014; 158:5-11.e2. [PMID: 24709810 DOI: 10.1016/j.ajo.2014.03.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 03/27/2014] [Accepted: 03/28/2014] [Indexed: 12/21/2022]
Abstract
PURPOSE To describe the immune alterations associated with age-related macular degeneration (AMD); and, based on these findings, to offer an approach to possibly prevent the expression of late disease. DESIGN Perspective. METHODS Review of the existing literature dealing with epidemiology, models, and immunologic findings in patients. RESULTS Significant genetic associations have been identified and reported, but environmentally induced (including epigenetic) changes are also an important consideration. Immune alterations include a strong interleukin 17 family signature as well as marked expression of these molecules in the eye. Oxidative stress as well as other homeostatic altering mechanisms occur throughout life. With this immune dysregulation there is a rationale for considering immunotherapy. Indeed, immunotherapy has been shown to affect the late stages of AMD. CONCLUSION Immune dysregulation appears to be an underlying alteration in AMD, as in other diseases thought to be degenerative and attributable to aging. Para-inflammation and immunosenescence may importantly contribute to the development of disease. The role of complement factor H still needs to be better defined, but in light of its association with ocular inflammatory conditions such as sarcoidosis, it does not appear to be unique to AMD but rather may be a marker for retinal pigment epithelium function. With the strong interleukin 17 family signature and the need to treat early on in the disease process, oral tolerance may be considered to prevent disease progression.
Collapse
|
172
|
Andrieu JM, Chen S, Lai C, Guo W, Lu W. Mucosal SIV Vaccines Comprising Inactivated Virus Particles and Bacterial Adjuvants Induce CD8(+) T-Regulatory Cells that Suppress SIV-Positive CD4(+) T-Cell Activation and Prevent SIV Infection in the Macaque Model. Front Immunol 2014; 5:297. [PMID: 25071760 PMCID: PMC4074992 DOI: 10.3389/fimmu.2014.00297] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 06/11/2014] [Indexed: 01/12/2023] Open
Abstract
A new paradigm of mucosal vaccination against human immunodeficiency virus (HIV) infection has been investigated in the macaque model. A vaccine consisting of inactivated simian immunodeficiency virus (SIV)mac239 particles together with a living bacterial adjuvant (either the Calmette and Guerin bacillus, Lactobacillus plantarum or Lactobacillus rhamnosus) was administered to macaques via the vaginal or oral/intragastric route. In contrast to all established human and veterinary vaccines, these three vaccine regimens did not elicit SIV-specific antibodies nor cytotoxic T-lymphocytes but induced a previously unrecognized population of non-cytolytic MHCIb/E-restricted CD8(+) T-regulatory cells that suppressed the activation of SIV-positive CD4(+) T-lymphocytes. SIV reverse transcription was thereby blocked in inactivated CD4(+) T-cells; the initial burst of virus replication was prevented and the vaccinated macaques were protected from a challenge infection. For 3-14 months after intragastric immunization, 24 macaques were challenged intrarectally with a high dose of SIVmac239 or with the heterologous strain SIV B670 (both strains grown on macaques PBMC). Twenty-three of these animals were found to be protected for up to 48 months while all 24 control macaques became infected. This protective effect against SIV challenge together with the concomitant identification of a robust ex vivo correlate of protection suggests a new approach for developing an HIV vaccine in humans. The induction of this new class of CD8(+) T-regulatory cells could also possibly be used therapeutically for suppressing HIV replication in infected patients and this novel tolerogenic vaccine paradigm may have potential applications for treating a wide range of immune disorders and is likely to may have profound implications across immunology generally.
Collapse
Affiliation(s)
- Jean-Marie Andrieu
- Institut de Recherches sur les Vaccins et l'Immunothérapie des Cancers et du SIDA, Centre Universitaire des Saints Peres, Université de Paris-Descartes , Paris , France
| | - Song Chen
- Tropical Medicine Institute, Guangzhou University of Chinese Medicine , Guangzhou , China
| | - Chunhui Lai
- Tropical Medicine Institute, Guangzhou University of Chinese Medicine , Guangzhou , China
| | - Weizhong Guo
- Tropical Medicine Institute, Guangzhou University of Chinese Medicine , Guangzhou , China
| | - Wei Lu
- Institut de Recherches sur les Vaccins et l'Immunothérapie des Cancers et du SIDA, Centre Universitaire des Saints Peres, Université de Paris-Descartes , Paris , France
| |
Collapse
|
173
|
Hiraide E, Nakajima-Adachi H, Hachimura S. Oral Administration of T Cell Epitope Peptide Inhibits the Systemic IL-4 Response Elicited by an Egg-White Diet in a TCR Transgenic Mouse Model. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2014; 33:47-51. [PMID: 24936382 PMCID: PMC4034323 DOI: 10.12938/bmfh.33.47] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 09/04/2013] [Indexed: 12/05/2022]
Abstract
Oral immunotherapy with T cell epitope peptides is a promising treatment for food
allergy. We examined the effect of oral administration of an ovalbumin T cell epitope
peptide (OVA323-339) in a TCR transgenic mouse model (OVA23-3 mice). OVA23-3 mice were fed
egg-white diet containing ovalbumin and subsequently orally administrated the OVA323-339
peptide. Cytokine measurements revealed that the IL-4 production of splenic
CD4+ T cells was significantly decreased by feeding the OVA323-339 peptide.
Our study suggested that oral administration of the OVA323-339 T cell epitope peptide was
capable of inhibiting systemic IL-4 response after elicitation of predominant Th2
responses.
Collapse
Affiliation(s)
- Erika Hiraide
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan ; Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Haruyo Nakajima-Adachi
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Satoshi Hachimura
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan ; Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
174
|
Vaz NM, Carvalho CR. On the origin of immunopathology. J Theor Biol 2014; 375:61-70. [PMID: 24937801 DOI: 10.1016/j.jtbi.2014.06.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 06/02/2014] [Accepted: 06/04/2014] [Indexed: 12/19/2022]
Abstract
Stranded between medicine and experimental biology, immunology is buried in its own problems and remains distant from important areas of current biology, such as evolutionary theory, developmental biology and cognitive sciences. Immunology has treated the living system merely as the place or dimension in which immune activity takes place, inserted on a misleading axis (progressive responsiveness versus no response; memory versus tolerance) which neglects the analysis of a robustly stable dynamics which is always present and is neither tolerance nor immunity-a problem currently approached as one of "regulatory" activity. However, a regulatory response also demands regulation, leading to an endless recursion and the adoption of a stimulus-response framework inevitably drives us away from the physiological processes in which lymphocytes are involved. Herein, we propose that immunological physiology, like everything else in the body is dynamic and conservative. Immunopathology, including inherited immunodeficiencies, severe forms of infectious diseases, allergy and autoimmune diseases, are interferences with this stability which frequently include oligoclonal expansions of T lymphocytes. We suggest that this decrease in clonal diversity results from a loss of the stabilizing connectivity among lymphocytes and are not simply markers of immunopathology, but are rather expressions of basic pathogenic mechanisms. The so-called autoimmune diseases are examples of this disequilibrium. In the last decade the characterization of an enormous and diversified commensal microbiota has posed a new and pressing problem: how to explain the harmonic conviviality with trillions of foreign macromolecules. In addition, robustly stable relations towards macromolecular diet can be established by simple ingestion, a state presently labeled as "oral tolerance", a problem that has been buffered for decades as anti-inflammatory protection of the gut. A major change in terminology is necessary to describe this new panorama. We focus on two important gaps in immunological discussions: (a) the organism, seen simultaneously as the medium with which the immune system is constantly in touch and as the entity that mediates the contact with external materials; and (b) the observer, the immunologist, who operates as a human being in human languaging with other human beings, and characterizes immunological specificity. We acknowledge that we are proposing radical departures from current dogma and that we should justify them. Most of what we propose stem form a way of seeing called Biology of Cognition and Language, that derives from ideas of the neurobiologist/philosopher Humberto Maturana, also known as "autopoiesis theory".
Collapse
Affiliation(s)
- Nelson M Vaz
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil.
| | - Claudia R Carvalho
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil
| |
Collapse
|
175
|
Pesek RD, Jones SM. Immunomodulatory Effect of Active Treatment Options in Food Allergy. CURRENT TREATMENT OPTIONS IN ALLERGY 2014. [DOI: 10.1007/s40521-014-0016-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
176
|
Dietary Melibiose Regulates Th Cell Response and Enhances the Induction of Oral Tolerance. Biosci Biotechnol Biochem 2014; 71:2774-80. [DOI: 10.1271/bbb.70372] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
177
|
Batista-Duharte A, Portuondo D, Pérez O, Carlos IZ. Systemic immunotoxicity reactions induced by adjuvanted vaccines. Int Immunopharmacol 2014; 20:170-80. [DOI: 10.1016/j.intimp.2014.02.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 02/04/2014] [Accepted: 02/21/2014] [Indexed: 02/08/2023]
|
178
|
Principato M, Qian BF. Staphylococcal enterotoxins in the etiopathogenesis of mucosal autoimmunity within the gastrointestinal tract. Toxins (Basel) 2014; 6:1471-89. [PMID: 24776983 PMCID: PMC4052247 DOI: 10.3390/toxins6051471] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 04/18/2014] [Accepted: 04/22/2014] [Indexed: 01/22/2023] Open
Abstract
The staphylococcal enterotoxins (SEs) are the products of Staphylococcus aureus and are recognized as the causative agents of classical food poisoning in humans following the consumption of contaminated food. While illness evoked by ingestion of the SE or its producer organism in tainted food are often self-limited, our current understanding regarding the evolution of S. aureus provokes the utmost concern. The organism and its associated toxins, has been implicated in a wide variety of disease states including infections of the skin, heart, sinuses, inflammatory gastrointestinal disease, toxic shock, and Sudden Infant Death Syndrome. The intricate relationship between the various subsets of immunocompetent T cells and accessory cells and the ingested material found within the gastrointestinal tract present daunting challenges to the maintenance of immunologic homeostasis. Dysregulation of the intricate balances within this environment has the potential for extreme consequences within the host, some of which are long-lived. The focus of this review is to evaluate the relevance of staphylococcal enterotoxin in the context of mucosal immunity, and the underlying mechanisms that contribute to the pathogenesis of gastrointestinal autoimmune disease.
Collapse
Affiliation(s)
- MaryAnn Principato
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, 8301 Muirkirk Road, Laurel, MD 20708, USA.
| | - Bi-Feng Qian
- Commissioner's Fellowship Program, Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, 8301 Muirkirk Road, Laurel, MD 20708, USA.
| |
Collapse
|
179
|
Lombardi VC, Khaiboullina SF. Plasmacytoid dendritic cells of the gut: relevance to immunity and pathology. Clin Immunol 2014; 153:165-77. [PMID: 24769378 DOI: 10.1016/j.clim.2014.04.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/11/2014] [Accepted: 04/12/2014] [Indexed: 12/15/2022]
Abstract
Plasmacytoid dendritic cells (pDCs) are bone marrow-derived immune cells with the ability to express copious amounts of type I and III interferon (IFN) and can differentiate into antigen-presenting dendritic cells as a result of stimulation by pathogen-derived nucleic acid. These powerful combined functionalities allow pDCs to bridge the innate and adaptive immune systems resulting in a concerted pathogen response. The contribution of pDCs to gastrointestinal immunity is only now being elucidated and is proving to be a critical component in systemic immunity. This review will explore the immunology of pDCs and will discuss their involvement in human disease and tolerance with an emphasis on those in the gastrointestinal lymphoid tissue.
Collapse
Affiliation(s)
- Vincent C Lombardi
- Department of Biochemistry and Molecular Biology, University of Nevada School of Medicine, WPI, University of Nevada, Reno, 1664 N Virginia St. MS 0552, Reno, NV 89557, USA.
| | - Svetlana F Khaiboullina
- Department of Biochemistry and Molecular Biology, University of Nevada School of Medicine, WPI, University of Nevada, Reno, 1664 N Virginia St. MS 0552, Reno, NV 89557, USA; Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.
| |
Collapse
|
180
|
Gorantala J, Grover S, Rahi A, Chaudhary P, Rajwanshi R, Sarin NB, Bhatnagar R. Generation of protective immune response against anthrax by oral immunization with protective antigen plant-based vaccine. J Biotechnol 2014; 176:1-10. [PMID: 24548460 DOI: 10.1016/j.jbiotec.2014.01.033] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 12/30/2013] [Accepted: 01/29/2014] [Indexed: 01/04/2023]
Abstract
In concern with frequent recurrence of anthrax in endemic areas and inadvertent use of its spores as biological weapon, the development of an effective anthrax vaccine suitable for both human and veterinary needs is highly desirable. A simple oral delivery through expression in plant system could offer promising alternative to the current methods that rely on injectable vaccines extracted from bacterial sources. In the present study, we have expressed protective antigen (PA) gene in Indian mustard by Agrobacterium-mediated transformation and in tobacco by plastid transformation. Putative transgenic lines were verified for the presence of transgene and its expression by molecular analysis. PA expressed in transgenic lines was biologically active as evidenced by macrophage lysis assay. Intraperitoneal (i.p.) and oral immunization with plant PA in murine model indicated high serum PA specific IgG and IgA antibody titers. PA specific mucosal immune response was noted in orally immunized groups. Further, antibodies indicated lethal toxin neutralizing potential in-vitro and conferred protection against in-vivo toxin challenge. Oral immunization experiments demonstrated generation of immunoprotective response in mice. Thus, our study examines the feasibility of oral PA vaccine expressed in an edible plant system against anthrax.
Collapse
Affiliation(s)
- Jyotsna Gorantala
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Sonam Grover
- Molecular Technology Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Amit Rahi
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Prerna Chaudhary
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Ravi Rajwanshi
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Neera Bhalla Sarin
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rakesh Bhatnagar
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
181
|
Grignolio A, Mishto M, Faria AMC, Garagnani P, Franceschi C, Tieri P. Towards a liquid self: how time, geography, and life experiences reshape the biological identity. Front Immunol 2014; 5:153. [PMID: 24782860 PMCID: PMC3988364 DOI: 10.3389/fimmu.2014.00153] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 03/24/2014] [Indexed: 01/08/2023] Open
Abstract
The conceptualization of immunological self is amongst the most important theories of modern biology, representing a sort of theoretical guideline for experimental immunologists, in order to understand how host constituents are ignored by the immune system (IS). A consistent advancement in this field has been represented by the danger/damage theory and its subsequent refinements, which at present represents the most comprehensive conceptualization of immunological self. Here, we present the new hypothesis of "liquid self," which integrates and extends the danger/damage theory. The main novelty of the liquid self hypothesis lies in the full integration of the immune response mechanisms into the host body's ecosystems, i.e., in adding the temporal, as well as the geographical/evolutionary and environmental, dimensions, which we suggested to call "immunological biography." Our hypothesis takes into account the important biological changes occurring with time (age) in the IS (including immunosenescence and inflammaging), as well as changes in the organismal context related to nutrition, lifestyle, and geography (populations). We argue that such temporal and geographical dimensions impinge upon, and continuously reshape, the antigenicity of physical entities (molecules, cells, bacteria, viruses), making them switching between "self" and "non-self" states in a dynamical, "liquid" fashion. Particular attention is devoted to oral tolerance and gut microbiota, as well as to a new potential source of unexpected self epitopes produced by proteasome splicing. Finally, our framework allows the set up of a variety of testable predictions, the most straightforward suggesting that the immune responses to defined molecules representing potentials antigens will be quantitatively and qualitatively quite different according to the immuno-biographical background of the host.
Collapse
Affiliation(s)
- Andrea Grignolio
- Interdepartmental Center "Luigi Galvani" for Bioinformatics, Biophysics and Biocomplexity, University of Bologna , Bologna , Italy
| | - Michele Mishto
- Centro Interdipartimentale di Ricerca sul Cancro "G. Prodi", University of Bologna , Bologna , Italy ; Institut für Biochemie, Charité - Universitätsmedizin Berlin , Berlin , Germany
| | - Ana Maria Caetano Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais , Belo Horizonte , Brazil
| | - Paolo Garagnani
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna , Bologna , Italy
| | - Claudio Franceschi
- Interdepartmental Center "Luigi Galvani" for Bioinformatics, Biophysics and Biocomplexity, University of Bologna , Bologna , Italy ; Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna , Bologna , Italy ; IRCCS of Neurological Science , Bologna , Italy ; Institute of Organic Synthesis and Photoreactivity, National Research Council , Bologna , Italy
| | - Paolo Tieri
- Institute for Applied Mathematics "M. Picone", National Research Council , Rome , Italy
| |
Collapse
|
182
|
Garofalo S, Mussa A, Mostert M, Suteu L, Vinardi S, Gamba S, Lonati L, Teruzzi E, Tommasoni N, Bassignana M, Masi G, Marenzi G, Sammartino G, Mortellaro C. Successful medical treatment for ranula in children. Oral Surg Oral Med Oral Pathol Oral Radiol 2014; 117:e289-97. [DOI: 10.1016/j.oooo.2012.07.430] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 06/12/2012] [Accepted: 07/10/2012] [Indexed: 10/27/2022]
|
183
|
Regulatory T-cell vaccination independent of auto-antigen. Exp Mol Med 2014; 46:e82. [PMID: 24626168 PMCID: PMC3972794 DOI: 10.1038/emm.2014.4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 12/06/2013] [Indexed: 12/21/2022] Open
Abstract
To date, efforts to treat autoimmune diseases have primarily focused on the disease symptoms rather than on the cause of the disease. In large part, this is attributed to not knowing the responsible auto-antigens (auto-Ags) for driving the self-reactivity coupled with the poor success of treating autoimmune diseases using oral tolerance methods. Nonetheless, if tolerogenic approaches or methods that stimulate regulatory T (Treg) cells can be devised, these could subdue autoimmune diseases. To forward such efforts, our approach with colonization factor antigen I (CFA/I) fimbriae is to establish bystander immunity to ultimately drive the development of auto-Ag-specific Treg cells. Using an attenuated Salmonella vaccine expressing CFA/I fimbriae, fimbriae-specific Treg cells were induced without compromising the vaccine's capacity to protect against travelers' diarrhea or salmonellosis. By adapting the vaccine's anti-inflammatory properties, it was found that it could also dampen experimental inflammatory diseases resembling multiple sclerosis (MS) and rheumatoid arthritis. Because of this bystander effect, disease-specific Treg cells are eventually induced to resolve disease. Interestingly, this same vaccine could elicit the required Treg cell subset for each disease. For MS-like disease, conventional CD25+ Treg cells are stimulated, but for arthritis CD39+ Treg cells are induced instead. This review article will examine the potential of treating autoimmune diseases without having previous knowledge of the auto-Ag using an innocuous antigen to stimulate Treg cells via the production of transforming growth factor-β and interleukin-10.
Collapse
|
184
|
Peine KJ, Guerau-de-Arellano M, Lee P, Kanthamneni N, Severin M, Probst GD, Peng H, Yang Y, Vangundy Z, Papenfuss TL, Lovett-Racke AE, Bachelder EM, Ainslie KM. Treatment of experimental autoimmune encephalomyelitis by codelivery of disease associated Peptide and dexamethasone in acetalated dextran microparticles. Mol Pharm 2014; 11:828-35. [PMID: 24433027 PMCID: PMC3993881 DOI: 10.1021/mp4005172] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune, demyelinating disease of the central nervous system that can cause loss of motor function and is thought to result, in part, from chronic inflammation due to an antigen-specific T cell immune response. Current treatments suppress the immune system without antigen specificity, increasing the risks of cancer, chronic infection, and other long-term side effects. In this study, we show treatment of experimental autoimmune encephalomyelitis (EAE), a model of MS, by coencapsulating the immunodominant peptide of myelin oligodendrocyte glycoprotein (MOG) with dexamethasone (DXM) into acetalated dextran (Ac-DEX) microparticles (DXM/MOG/MPs) and administering the microparticles subcutaneously. The clinical score of the mice was reduced from 3.4 to 1.6 after 3 injections 3 days apart with the coencapsulated microparticulate formulation (MOG 17.6 μg and DXM 8 μg). This change in clinical score was significantly greater than observed with phosphate-buffered saline (PBS), empty MPs, free DXM and MOG, DXM/MPs, and MOG/MPs. Additionally, treatment with DXM/MOG/MPs significantly inhibited disease-associated cytokine (e.g., IL-17, GM-CSF) expression in splenocytes isolated in treated mice. Here we show a promising approach for the therapeutic treatment of MS using a polymer-based microparticle delivery platform.
Collapse
Affiliation(s)
- Kevin J Peine
- Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University , Columbus, Ohio, 43210, United States
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Santos Rocha C, Gomes-Santos AC, Garcias Moreira T, de Azevedo M, Diniz Luerce T, Mariadassou M, Longaray Delamare AP, Langella P, Maguin E, Azevedo V, Caetano de Faria AM, Miyoshi A, van de Guchte M. Local and systemic immune mechanisms underlying the anti-colitis effects of the dairy bacterium Lactobacillus delbrueckii. PLoS One 2014; 9:e85923. [PMID: 24465791 PMCID: PMC3897545 DOI: 10.1371/journal.pone.0085923] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 12/03/2013] [Indexed: 02/07/2023] Open
Abstract
Several probiotic bacteria have been proposed for treatment or prevention of inflammatory bowel diseases (IBD), showing a protective effect in animal models of experimental colitis and for some of them also in human clinical trials. While most of these probiotic bacteria are isolated from the digestive tract, we recently reported that a Lactobacillus strain isolated from cheese, L. delbrueckii subsp. lactis CNRZ327 (Lb CNRZ327), also possesses anti-inflammatory effects in vitro and in vivo, demonstrating that common dairy bacteria may be useful in the treatment or prevention of IBD. Here, we studied the mechanisms underlying the protective effects of Lb CNRZ327 in vivo, in a mouse dextran sodium sulfate (DSS) colitis model. During colitis, Lb CNRZ327 modulated the production of TGF-β, IL-6, and IL-12 in colonic tissue and of TGF-β and IL-6 in the spleen, and caused an expansion of CD4+Foxp3+ regulatory T cells in the cecal lymph nodes. Moreover, a strong tendency to CD4+Foxp3+ expansion was also observed in the spleen. The results of this study for the first time show that orally administered dairy lactobacilli can not only modulate mucosal but also systemic immune responses and constitute an effective treatment of IBD.
Collapse
Affiliation(s)
- Clarissa Santos Rocha
- Department of General Biology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- INRA, UMR1319 Micalis, Jouy-en-Josas, France
- AgroParisTech, UMR Micalis, Jouy-en-Josas, France
| | - Ana Cristina Gomes-Santos
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thais Garcias Moreira
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marcela de Azevedo
- Department of General Biology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Tessalia Diniz Luerce
- Department of General Biology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | - Philippe Langella
- INRA, UMR1319 Micalis, Jouy-en-Josas, France
- AgroParisTech, UMR Micalis, Jouy-en-Josas, France
| | - Emmanuelle Maguin
- INRA, UMR1319 Micalis, Jouy-en-Josas, France
- AgroParisTech, UMR Micalis, Jouy-en-Josas, France
| | - Vasco Azevedo
- Department of General Biology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Maria Caetano de Faria
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Anderson Miyoshi
- Department of General Biology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maarten van de Guchte
- INRA, UMR1319 Micalis, Jouy-en-Josas, France
- AgroParisTech, UMR Micalis, Jouy-en-Josas, France
- * E-mail:
| |
Collapse
|
186
|
Li MO, Flavell RA. TGF-β, T-cell tolerance and immunotherapy of autoimmune diseases and cancer. Expert Rev Clin Immunol 2014; 2:257-65. [DOI: 10.1586/1744666x.2.2.257] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
187
|
|
188
|
Park KS, Park MJ, Cho ML, Kwok SK, Ju JH, Ko HJ, Park SH, Kim HY. Type II collagen oral tolerance; mechanism and role in collagen-induced arthritis and rheumatoid arthritis. Mod Rheumatol 2014. [DOI: 10.3109/s10165-009-0210-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
189
|
Bellavite P, Marzotto M, Olioso D, Moratti E, Conforti A. High-dilution effects revisited. 2. Pharmacodynamic mechanisms. HOMEOPATHY 2014; 103:22-43. [DOI: 10.1016/j.homp.2013.08.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 08/12/2013] [Indexed: 11/30/2022]
|
190
|
Salisbury EM, Game DS, Lechler RI. Transplantation tolerance. Pediatr Nephrol 2014; 29:2263-72. [PMID: 24213880 PMCID: PMC4212135 DOI: 10.1007/s00467-013-2659-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 10/01/2013] [Accepted: 10/04/2013] [Indexed: 01/26/2023]
Abstract
Although transplantation has been a standard medical practice for decades, marked morbidity from the use of immunosuppressive drugs and poor long-term graft survival remain important limitations in the field. Since the first solid organ transplant between the Herrick twins in 1954, transplantation immunology has sought to move away from harmful, broad-spectrum immunosuppressive regimens that carry with them the long-term risk of potentially life-threatening opportunistic infections, cardiovascular disease, and malignancy, as well as graft toxicity and loss, towards tolerogenic strategies that promote long-term graft survival. Reports of "transplant tolerance" in kidney and liver allograft recipients whose immunosuppressive drugs were discontinued for medical or non-compliant reasons, together with results from experimental models of transplantation, provide the proof-of-principle that achieving tolerance in organ transplantation is fundamentally possible. However, translating the reconstitution of immune tolerance into the clinical setting is a daunting challenge fraught with the complexities of multiple interacting mechanisms overlaid on a background of variation in disease. In this article, we explore the basic science underlying mechanisms of tolerance and review the latest clinical advances in the quest for transplantation tolerance.
Collapse
Affiliation(s)
- Emma M. Salisbury
- Section of Immunobiology, Division of Immunology and Inflammation, Department of Medicine, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, Exhibition Road, London, SW7 2AZ UK
| | - David S. Game
- Department of Renal Medicine, Guy’s and St. Thomas’ NHS Foundation Trust, Guy’s Hospital, Great Maze Pond, London, SE1 9RT UK
| | - Robert I. Lechler
- King’s Health Partners Academic Health Sciences Centre, King’s College London, London, WC2R 2LS UK
| |
Collapse
|
191
|
Sun H, Liu X, Wang YZ, Liu JX, Feng J. Allergen-specific immunoglobulin, histamine and T-cell responses induced by soybean glycinin and β-conglycinin in BALB/c mice of oral sensitisation. FOOD AGR IMMUNOL 2013. [DOI: 10.1080/09540105.2012.730501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
192
|
Food allergen selective thermal processing regimens may change oral tolerance in infancy. Allergol Immunopathol (Madr) 2013; 41:407-17. [PMID: 23253679 DOI: 10.1016/j.aller.2012.08.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Accepted: 08/03/2012] [Indexed: 12/20/2022]
Abstract
Food allergy can be considered a failure in the induction of oral tolerance. Recently, great interest has been focused on understanding the mechanisms and the contributing factors of oral tolerance development, hoping for new definitive interventions in the prevention and treatment of food allergy. Given that food processing may modify the properties and the nature of dietary proteins, several food processing methods could affect the allergenicity of these proteins and consequently may favour oral tolerance induction to food allergic children. Indeed, effective thermal food processing regimens of altering food proteins to reduce allergenicity have been recently reported in the literature. This article is mainly focused on the effect of selective thermal processing regimens on the main infant allergenic foods, with a potential clinical relevance on their allergenicity and therefore on oral tolerance induction. In the light of recent findings, the acquisition of tolerance in younger age and consequently the ability of young children to "outgrow" food allergy could be achieved through the application of selective thermal processing regimens on certain allergenic foods. Therefore, the ability of processed foods to circumvent clinical disease and at the same time to have an impact on the immune system and facilitate tolerance induction could be invaluable as a component of a successful therapeutic strategy. The opening in the new avenues of research in the use of processed foods in clinical practice for the amelioration of the impact on the quality of life of patients and possibly in food allergy prevention is warranted.
Collapse
|
193
|
Lutterotti A, Martin R. Antigen-specific tolerization approaches in multiple sclerosis. Expert Opin Investig Drugs 2013; 23:9-20. [PMID: 24151958 DOI: 10.1517/13543784.2014.844788] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Inhibition of self-reactive T cells through induction of antigen-specific immune tolerance holds the promise of effective treatment of autoimmune pathology with few side effects and preservation of normal immune functions. In multiple sclerosis (MS) several approaches have been tested already in clinical trials or are currently ongoing with the aim to inhibit myelin-reactive immune responses. AREAS COVERED This article provides an overview of the recent and ongoing strategies to inhibit specific immune responses in MS, including different applications of myelin peptide-based approaches, T-cell vaccination, DNA vaccination and antigen-coupled cells. EXPERT OPINION Despite difficulties in translation of antigen-specific therapies in MS, novel approaches have the potential to effectively induce immune tolerance and ameliorate the disease. To improve efficacy of treatments, future trials should include patients in the early phases of the disease, when the autoimmune response is predominant and immune reactivity still focused. The target antigens are not fully defined yet, and robust immunomonitoring assays should developed to provide mechanistic proof of concept in parallel to showing efficacy with respect to inhibiting inflammatory disease activity in the central nervous system (CNS).
Collapse
Affiliation(s)
- Andreas Lutterotti
- Clinical Department of Neurology, Innsbruck Medical University, Innsbruck , Austria
| | | |
Collapse
|
194
|
|
195
|
Lehtimäki S, Lahesmaa R. Regulatory T Cells Control Immune Responses through Their Non-Redundant Tissue Specific Features. Front Immunol 2013; 4:294. [PMID: 24069022 PMCID: PMC3780303 DOI: 10.3389/fimmu.2013.00294] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 09/07/2013] [Indexed: 01/11/2023] Open
Abstract
Regulatory T cells (Treg) are needed in the control of immune responses and to maintain immune homeostasis. Of this subtype of regulatory lymphocytes, the most potent are Foxp3 expressing CD4+ T cells, which can be roughly divided into two main groups; natural Treg cells (nTreg), developing in the thymus, and induced or adaptive Treg cells (iTreg), developing in the periphery from naïve, conventional T cells. Both nTreg cells and iTreg cells have their own, non-redundant roles in the immune system, with nTreg cells mainly maintaining tolerance toward self-structures, and iTreg developing in response to externally delivered antigens or commensal microbes. In addition, Treg cells acquire tissue specific features and are adapted to function in the tissue they reside. This review will focus on some specific features of Treg cells in different compartments of the body.
Collapse
Affiliation(s)
- Sari Lehtimäki
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University , Turku , Finland
| | | |
Collapse
|
196
|
Ricciardi L, Carni A, Loschiavo G, Gangemi S, Tigano V, Arena E, Mannucci C, Calapai G. Systemic nickel allergy: oral desensitization and possible role of cytokines interleukins 2 and 10. Int J Immunopathol Pharmacol 2013; 26:251-7. [PMID: 23527730 DOI: 10.1177/039463201302600127] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Nickel ingested with food can elicit either systemic cutaneous or gastrointestinal symptoms causing a systemic nickel allergy syndrome (SNAS) that can be treated with tolerance by oral ingestion of the metal. It has been suggested that interleukins 2 (IL-2) and 10 (IL-10) are involved in the mechanisms underlying oral tolerance. We evaluated the clinical efficacy of oral desensitization therapy in SNAS consisting in the administration of nickel sulphate. Because nickel allergy prevalently affects women, only female subjects (N = 22) were recruited. Oral nickel desensitizing therapy was associated with low-nickel diet for three months. Before and after therapy, clinical conditions were evaluated, and circulating cytokines IL-2 and IL-10 were measured. After the two-year treatment, visual analogue scale (VAS) scores for symptoms were significantly reduced (P less than 0.001). Patients were released by either cutaneous or gastrointestinal symptoms and by tolerating nickel-containing food. At the end of the treatment, nickel oral challenge test was negative in 18 patients, and IL-2 level in the serum was significantly reduced while IL-10 was increased, although this datum was not statistically significant. Our study confirms the clinical efficacy of nickel oral immunotherapy and focuses on the mechanisms triggered by oral tolerance indicating that reduction of IL-2 can be associated with success of oral nickel desensitizing therapy.
Collapse
|
197
|
Buchanan RM, Mertins S, Wilson HL. Oral antigen exposure in extreme early life in lambs influences the magnitude of the immune response which can be generated in later life. BMC Vet Res 2013; 9:160. [PMID: 23937675 PMCID: PMC3751536 DOI: 10.1186/1746-6148-9-160] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 08/08/2013] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Previous investigations in newborn lambs determined that adenovirus-mediated expression of antigen to a localized region of the gut induced antigen-specific mucosal and systemic immunity. These experiments were limited in that the localized region of the gut to which antigen was introduced was sterile and the influence of colostrum on the antigen was not assessed but they do suggest that mucosal vaccines may be an effective vaccination strategy to protect neonatal lambs. We propose that persistent oral antigen exposure introduced in extreme early life can induce immunity in lambs, despite the presence of commensal bacteria and colostrum. RESULTS To test this hypothesis, conventionally raised newborn lambs (n = 4 per group) were gavaged with ovalbumin (OVA) starting the day after birth for either a single day (2.27 g), every day for 3 days (0.23 g/day), or every day for 3 days then every second day until nine days of age (0.023 g/day). Lambs gavaged with OVA for 3 to 9 days developed significant serum anti-OVA IgG titres (p < 0.05), but not IgA titres, relative to control lambs (n = 4) after 3 and 4 weeks. At 4 weeks of age, lambs were immunized with OVA in Incomplete Freund's Adjuvant via intraperitoneal (i.p.) injection then lambs were euthanized at 7 weeks. Serum anti-OVA IgG titres were further augmented after i.p. immunization indicating immunity persisted and tolerance was not induced. Serum IgA titres remained low regardless of treatment. It is known that i.p. priming of sheep with antigen in Freund's complete adjuvant leads to an enhanced number of IgA and IgG antibody containing cells in the respiratory mucosa (Immunology 53(2):375-384, 1984). Lambs gavaged with a single bolus of 2.27 g OVA prior to i.p. immunization showed very low titres of anti-OVA IgA in the lung lavage. These data suggest that a single, high dose exposure to OVA can promote tolerance which impacts response to systemic vaccination in later life. Lambs gavaged with 0.023 g OVA for 9 days (Group C) generated significant anti-OVA IgA titres in lung (p < 0.001) compared to negative control lambs but no additive effect was observed compared to parenteral control lambs. When splenocytes were re-stimulated with OVA ex vivo, all groups failed to show increased lymphocyte proliferation or interferon (IFN)-γ production relative to the parenteral control group. CONCLUSIONS In agreement with our hypothesis, persistent low dose antigen exposure primes humoral antibody production in serum in conventionally raised newborn lambs. In contrast, a single high dose bolus of antigen triggered oral tolerance which negatively impacted the quality and magnitude of the immune response to i.p. immunization in later life. These tangential responses are important as they indicate that the dose and/or repeated oral exposure to antigen, such as that which may be found in the neonate's environment, may promote immunity or alternatively it may negatively impact responses to parenteral vaccination.
Collapse
|
198
|
Noviello MDLM, Batista NV, Dourado LPA, Pereira RVS, Oliveira AG, Menezes GB, Cara DC. Prolonged ingestion of ovalbumin diet by Ova sensitized mice suppresses mBSA-induced arthritis. Cell Immunol 2013; 284:20-8. [PMID: 23916876 DOI: 10.1016/j.cellimm.2013.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 06/24/2013] [Accepted: 07/10/2013] [Indexed: 12/26/2022]
Abstract
Concomitant chronic diseases are a common finding in clinics and may consist in a major issue in therapeutics. Here, we investigated whether prolonged ingestion of ovalbumin (Ova) by sensitized mice would reduce the severity of an associated concurrent immunomediated condition such as antigen-induced arthritis (AIA). AIA was induced by administration of methylated bovine albumin (mBSA) into the knee joints of previously immunized mice, and evaluated by articular leukocyte trafficking and levels of cytokines (TNF-α, IL-1β) and chemokine (CXCL-1) in the periarticular tissue. Continuous Ova feeding by Ova sensitized mice decreased serum levels of anti-Ova IgE, and led to a significant suppression of leukocyte adhesion and infiltration into synovial tissue and cavity. Also, a marked cytokine reduction was observed, suggesting that prolonged ingestion of ovalbumin by sensitized mice suppresses specific IgE production with concomitant reduction in peripheral T cells, which may impact in the pathogenesis of AIA, a non-related condition.
Collapse
|
199
|
Kant CD, Akiyama Y, Tanaka K, Shea S, Connolly SE, Germana S, Winn HJ, LeGuern C, Tocco G, Benichou G. Primary vascularization of allografts governs their immunogenicity and susceptibility to tolerogenesis. THE JOURNAL OF IMMUNOLOGY 2013; 191:1948-56. [PMID: 23833234 DOI: 10.4049/jimmunol.1202092] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We investigated the influence of allograft primary vascularization on alloimmunity, rejection, and tolerance in mice. First, we showed that fully allogeneic primarily vascularized and conventional skin transplants were rejected at the same pace. Remarkably, however, short-term treatment of mice with anti-CD40L Abs achieved long-term survival of vascularized skin and cardiac transplants but not conventional skin grafts. Nonvascularized skin transplants triggered vigorous direct and indirect proinflammatory type 1 T cell responses (IL-2 and IFN-γ), whereas primarily vascularized skin allografts failed to trigger a significant indirect alloresponse. A similar lack of indirect alloreactivity was also observed after placement of different vascularized organ transplants, including hearts and kidneys, whereas hearts placed under the skin (nonvascularized) triggered potent indirect alloresponses. Altogether, these results suggest that primary vascularization of allografts is associated with a lack of indirect T cell alloreactivity. Finally, we show that long-term survival of vascularized skin allografts induced by anti-CD40L Abs was associated with a combined lack of indirect alloresponse and a shift of the direct alloresponse toward a type 2 cytokine (IL-4, IL-10)-secretion pattern but no activation/expansion of Foxp3(+) regulatory T cells. Therefore, primary vascularization of allografts governs their immunogenicity and tolerogenicity.
Collapse
Affiliation(s)
- Cavit D Kant
- Transplantation Unit and Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
200
|
Chaudhari AA, Lee JH. Evaluation of the adjuvant effect of Salmonella-based Escherichia coli heat-labile toxin B subunits on the efficacy of a live Salmonella-delivered avian pathogenic Escherichia coli vaccine. Avian Pathol 2013; 42:365-72. [PMID: 23815619 DOI: 10.1080/03079457.2013.811466] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The present study evaluated the adjuvant effect of live attenuated salmonella organisms expressing the heat-labile toxin of Escherichia coli B subunit (LTB) on the efficacy of an avian pathogenic Escherichia coli (APEC) vaccine. The Asd(+) (aspartate semialdehyde dehydrogenase) plasmid pMMP906 containing the LTB gene was introduced into a Salmonella enterica Typhimurium strain lacking the lon, cpxR and asd genes to generate the adjuvant strain. Live recombinant Salmonella-delivered APEC vaccine candidates were used for this study. The birds were divided into three groups: group A, non-vaccinated controls; group B, immunized with vaccine candidates only; and group C, immunized with vaccine candidates and the LTB strain. The immune responses were measured and the birds were challenged at 21 days of age with a virulent APEC strain. Group C showed a significant increase in plasma IgG and intestinal IgA levels and a significantly higher lymphocyte proliferation response compared with the other groups. Upon challenge with the virulent APEC strain, group C showed effective protection whereas group B did not. We also attempted to optimize the effective dose of the adjuvant. The birds were immunized with the vaccine candidates together with 1×10⁷ or 1×10⁸ colony-forming units of the LTB strain and were subsequently challenged at 3 weeks of age. The 1×10⁷ colony-forming units of the LTB strain showed a greater adjuvant effect with increased levels of serum IgG, intestinal IgA and a potent lymphocyte proliferation response, and yielded higher protection against challenge. Overall, the LTB strain increased the efficacy of the Salmonella -delivered APEC vaccine, indicating that vaccination for APEC along with the LTB strain appears to increase the efficacy for protection against colibacillosis in broiler chickens.
Collapse
Affiliation(s)
- Atul A Chaudhari
- College of Veterinary Medicine, Chonbuk National University, Jeonju, Republic of Korea
| | | |
Collapse
|