301
|
Abstract
The suppressive/immunomodulatory function of CD4(+)CD25(+)FOXP3(+) regulatory T (Treg) cells is crucial for the maintenance of immune homeostasis, which helps to prevent autoimmunity and reduce the inflammation induced by pathogens and environmental insults. This review summarizes the current knowledge on the types and mechanisms of action of Treg cells and their role in the immune tolerance to self-antigens, with a particular focus on naturally occurring Treg cells.
Collapse
Affiliation(s)
- Elaine V Lourenço
- Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095-1670, USA
| | | |
Collapse
|
302
|
Vaickus LJ, Bouchard J, Kim J, Natarajan S, Remick DG. Oral tolerance inhibits pulmonary eosinophilia in a cockroach allergen induced model of asthma: a randomized laboratory study. Respir Res 2010; 11:160. [PMID: 21092270 PMCID: PMC3016351 DOI: 10.1186/1465-9921-11-160] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 11/23/2010] [Indexed: 11/10/2022] Open
Abstract
Background Antigen desensitization through oral tolerance is becoming an increasingly attractive treatment option for allergic diseases. However, the mechanism(s) by which tolerization is achieved remain poorly defined. In this study we endeavored to induce oral tolerance to cockroach allergen (CRA: a complex mixture of insect components) in order to ameliorate asthma-like, allergic pulmonary inflammation. Methods We compared the pulmonary inflammation of mice which had received four CRA feedings prior to intratracheal allergen sensitization and challenge to mice fed PBS on the same time course. Respiratory parameters were assessed by whole body unrestrained plethysmography and mechanical ventilation with forced oscillation. Bronchoalveolar lavage fluid (BAL) and lung homogenate (LH) were assessed for cytokines and chemokines by ELISA. BAL inflammatory cells were also collected and examined by light microscopy. Results CRA feeding prior to allergen sensitization and challenge led to a significant improvement in respiratory health. Airways hyperreactivity measured indirectly via enhanced pause (Penh) was meaningfully reduced in the CRA-fed mice compared to the PBS fed mice (2.3 ± 0.4 vs 3.9 ± 0.6; p = 0.03). Directly measured airways resistance confirmed this trend when comparing the CRA-fed to the PBS-fed animals (2.97 ± 0.98 vs 4.95 ± 1.41). This effect was not due to reduced traditional inflammatory cell chemotactic factors, Th2 or other cytokines and chemokines. The mechanism of improved respiratory health in the tolerized mice was due to significantly reduced eosinophil numbers in the bronchoalveolar lavage fluid (43300 ± 11445 vs 158786 ± 38908; p = 0.007) and eosinophil specific peroxidase activity in the lung homogenate (0.59 ± 0.13 vs 1.19 ± 0.19; p = 0.017). The decreased eosinophilia was likely the result of increased IL-10 in the lung homogenate of the tolerized mice (6320 ± 354 ng/mL vs 5190 ± 404 ng/mL, p = 0.02). Conclusion Our results show that oral tolerization to CRA can improve the respiratory health of experimental mice in a CRA-induced model of asthma-like pulmonary inflammation by reducing pulmonary eosinophilia.
Collapse
Affiliation(s)
- Louis J Vaickus
- Boston University School of Medicine, Department of Pathology and Laboratory Medicine, Boston, MA, USA
| | | | | | | | | |
Collapse
|
303
|
Azizi A, Kumar A, Diaz-Mitoma F, Mestecky J. Enhancing oral vaccine potency by targeting intestinal M cells. PLoS Pathog 2010; 6:e1001147. [PMID: 21085599 PMCID: PMC2978714 DOI: 10.1371/journal.ppat.1001147] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The immune system in the gastrointestinal tract plays a crucial role in the control of infection, as it constitutes the first line of defense against mucosal pathogens. The attractive features of oral immunization have led to the exploration of a variety of oral delivery systems. However, none of these oral delivery systems have been applied to existing commercial vaccines. To overcome this, a new generation of oral vaccine delivery systems that target antigens to gut-associated lymphoid tissue is required. One promising approach is to exploit the potential of microfold (M) cells by mimicking the entry of pathogens into these cells. Targeting specific receptors on the apical surface of M cells might enhance the entry of antigens, initiating the immune response and consequently leading to protection against mucosal pathogens. In this article, we briefly review the challenges associated with current oral vaccine delivery systems and discuss strategies that might potentially target mouse and human intestinal M cells.
Collapse
Affiliation(s)
- Ali Azizi
- Infectious Disease and Vaccine Research Center, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada.
| | | | | | | |
Collapse
|
304
|
Abstract
IL-6 is a pleiotropic cytokine involved in the physiology of virtually every organ system. Recent studies have demonstrated that IL-6 has a very important role in regulating the balance between IL-17-producing Th17 cells and regulatory T cells (Treg). The two T-cell subsets play prominent roles in immune functions: Th17 cell is a key player in the pathogenesis of autoimmune diseases and protection against bacterial infections, while Treg functions to restrain excessive effector T-cell responses. IL-6 induces the development of Th17 cells from naïve T cells together with TGF-beta; in contrast, IL-6 inhibits TGF-beta-induced Treg differentiation. Dysregulation or overproduction of IL-6 leads to autoimmune diseases such as multiple sclerosis (MS) and rheumatoid arthritis (RA), in which Th17 cells are considered to be the primary cause of pathology. Given the critical role of IL-6 in altering the balance between Treg and Th17 cells, controlling IL-6 activities is potentially an effective approach in the treatment of various autoimmune and inflammatory diseases. Here, we review the role of IL-6 in regulating Th17/Treg balance and describe the critical functions of IL-6 and Th17 in immunity and immune-pathology.
Collapse
Affiliation(s)
- Akihiro Kimura
- Laboratory of Immune Regulation, Osaka University Graduate School of Frontier Biosciences, Osaka, Japan
| | | |
Collapse
|
305
|
O'Brien K, Gran B, Rostami A. T-cell based immunotherapy in experimental autoimmune encephalomyelitis and multiple sclerosis. Immunotherapy 2010; 2:99-115. [PMID: 20231863 DOI: 10.2217/imt.09.61] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
One of the reasons multiple sclerosis (MS) has been considered a T-cell mediated autoimmune disease is that a similar experimental disease can be induced in certain rodents and primates by immunization with myelin antigens, leading to T-cell-mediated inflammatory demyelination in the CNS. In addition, most if not all pharmacological treatments available for MS are biologically active on T cells. In this article we review the principles of T-cell-based immunotherapies and the specific actions of current and novel treatments on T-cell functions, when these are known. For both licensed and innovative agents, we also discuss biological actions on other immune cell types. Finally, we offer a brief perspective on expected changes in the use of MS immunotherapies in the near future.
Collapse
Affiliation(s)
- Kate O'Brien
- Division of Clinical Neurology, University of Nottingham, UK
| | | | | |
Collapse
|
306
|
Kapp K, Maul J, Hostmann A, Mundt P, Preiss JC, Wenzel A, Thiel A, Zeitz M, Ullrich R, Duchmann R. Modulation of systemic antigen-specific immune responses by oral antigen in humans. Eur J Immunol 2010; 40:3128-37. [PMID: 20957752 DOI: 10.1002/eji.201040701] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2010] [Revised: 07/21/2010] [Accepted: 08/16/2010] [Indexed: 11/06/2022]
Abstract
Oral antigen uptake can induce systemic immune responses ranging from tolerance to immunity. However, the underlying mechanisms are poorly understood, especially in humans. Here, keyhole limpet hemocyanin (KLH), a neoantigen which has been used in earlier studies of oral tolerance, was fed in a repeated low-dose and a single high-dose protocol to healthy volunteers. KLH-specific CD4(+) T-cell proliferation and cytokine production, as well as KLH-specific serum Ab and the effects of oral KLH on a subsequent parenterally induced systemic immune response, were analyzed. Repeated low-dose oral KLH alone induced antigen-specific CD4(+) T cells positive predominantly for the gut-homing receptor integrin β7 and the cytokines IL-2 and TNF-α; some CD4(+) T cells also produced IL-4. Oral feeding of KLH accelerated a subsequent parenterally induced systemic CD4(+) T-cell response. The cytokine pattern of KLH-specific CD4(+) T cells shifted toward more IL-4- and IL-10- and less IFN-γ-, IL-2- and TNF-α-producing cells. The parenterally induced systemic KLH-specific B-cell response was accelerated and amplified by oral KLH. The impact of single high-dose oral KLH on antigen-specific immune responses was less pronounced compared with repeated low-dose oral KLH. These findings suggest that oral antigen can effectively modulate subsequently induced systemic antigen-specific immune responses. Immunomodulation by oral antigen may offer new therapeutic strategies for Th type1-mediated inflammatory diseases and for the development of vaccination strategies.
Collapse
Affiliation(s)
- Kerstin Kapp
- Charité-Campus Benjamin Franklin, Medizinische Klinik I (Gastroenterologie, Infektiologie, Rheumatologie), Berlin, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
307
|
Torres J, Tamimi F, Alkhraisat MH, Manchón A, Linares R, Prados-Frutos JC, Hernández G, López Cabarcos E. Platelet-rich plasma may prevent titanium-mesh exposure in alveolar ridge augmentation with anorganic bovine bone. J Clin Periodontol 2010. [PMID: 20796106 DOI: 10.1111/j.1600] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Bone augmentation with the titanium-mesh (Ti-mesh) technique is susceptible to a large rate of complications such as morbidity of bone graft donor site, and mesh exposure to the oral cavity. The purpose of this study was to evaluate the effectiveness of anorganic bovine bone (ABB) in alveolar bone augmentation with the Ti-mesh technique. In addition, we investigated the effect of platelet-rich plasma (PRP) in preventing mesh exposure by using it to cover the Ti-mesh. PATIENTS AND METHODS Patients included in the clinical trial were randomly allocated by a blinded assistant into two groups. The 30 patients recruited for this study underwent 43 alveolar bone augmentation with the Ti-mesh technique using ABB as graft material in all of them. In 15 patients, the Ti-meshes were covered with PRP (PRP group) whereas in the other 15 the Ti-meshes were not (control group). After 6 months, patients were called for clinical, radiographic, and histological evaluation, and implant placement surgery. A total of 97 implants were placed in the augmented bone and their evolution was followed up for a period of 24 months. RESULTS Significant differences were found between the two study groups in terms of complications and bone formation. In the control group, 28.5% of the cases suffered from mesh exposure, while in the PRP group, no exposures were registered. Radiographic analysis revealed that bone augmentation was higher in the PRP group than in the control group. Overall, 97.3% of implants placed in the control group and 100% of those placed in the PRP group were successful during the monitoring period. We suggest that the positive effect of PRP on the Ti-mesh technique is due to its capacity to improve soft tissue healing, thereby protecting the mesh and graft material secured beneath the gingival tissues. CONCLUSIONS Alveolar bone augmentation using ABB alone in the Ti-mesh technique is sufficient for implant rehabilitation. Besides, covering the Ti-meshes with PRP was a determining factor in avoiding mesh exposure. Ti-mesh exposure provoked significant bone loss, but in most cases it did not affect the subsequent placement of implants.
Collapse
Affiliation(s)
- Jesús Torres
- Ciencias de la Salud III, Universidad Rey Juan Carlos, Alcorcón, Spain
| | | | | | | | | | | | | | | |
Collapse
|
308
|
Rubin IMC, Dabelsteen S, Nielsen MM, White IR, Johansen JD, Geisler C, Bonefeld CM. Repeated exposure to hair dye induces regulatory T cells in mice. Br J Dermatol 2010; 163:992-8. [PMID: 20716223 DOI: 10.1111/j.1365-2133.2010.09988.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND We have recently shown that commercial p-phenylenediamine (PPD)-containing hair dyes are potent immune activators that lead to severe contact hypersensitivity in an animal model. However, only a minority of people exposed to permanent hair dyes develops symptomatic contact hypersensitivity. This suggests that the majority of people exposed to hair dyes does not become sensitized or develop immunological tolerance. OBJECTIVES To study the immune response in mice repeatedly exposed to PPD-containing hair dye in a consumer-like manner. METHODS A commercial hair dye containing PPD was tested in C57BL/6 mice. The local immune response was measured by ear swelling and by histological examinations. The immune response in the draining lymph nodes was analysed by flow cytometry. RESULTS The hair dye induced local inflammation as seen by swelling and cell infiltration of the treated ears. In addition, exposure to hair dye caused T-cell activation as seen by T-cell proliferation and production of interferon-γ and interleukin (IL)-17 within the draining lymph nodes. The inflammatory response peaked at the fourth exposure to hair dye. From this point on, an upregulation of regulatory T cells and IL-10-producing cells was seen. CONCLUSIONS This study shows that PPD-containing hair dyes strongly affect the immune system. In addition to being potent skin sensitizers that activate inflammatory T cells, hair dyes also induce anti-inflammatory mechanisms. This might explain why many consumers can use hair dyes repeatedly without developing noticeable allergies, but it also raises the question whether the immune modulatory effects of hair dyes might influence the development of autoimmune diseases and cancers.
Collapse
Affiliation(s)
- I M C Rubin
- Department of International Health, Immunology and Microbiology, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark
| | | | | | | | | | | | | |
Collapse
|
309
|
Jung C, Hugot JP, Barreau F. Peyer's Patches: The Immune Sensors of the Intestine. Int J Inflam 2010; 2010:823710. [PMID: 21188221 PMCID: PMC3004000 DOI: 10.4061/2010/823710] [Citation(s) in RCA: 278] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2010] [Accepted: 07/11/2010] [Indexed: 12/13/2022] Open
Abstract
The gut-associated lymphoid tissue (GALT) consists of isolated or aggregated lymphoid follicles forming Peyer's patches (PPs). By their ability to transport luminal antigens and bacteria, PPs can be considered as the immune sensors of the intestine. PPs functions like induction of immune tolerance or defense against pathogens result from the complex interplay between immune cells located in the lymphoid follicles and the follicle-associated epithelium. This crosstalk seems to be regulated by pathogen recognition receptors, especially Nod2. Although TLR exerts a limited role in PP homeotasis, Nod2 regulates the number, size, and T-cell composition of PPs, in response to the gut flora. In turn, CD4+ T-cells present in the PP are able to modulate the paracellular and transcellular permeabilities. Two human disorders, Crohn's disease and graft-versus-host disease are thought to be driven by an abnormal response toward the commensal flora. They have been associated with NOD2 mutations and PP dysfunction.
Collapse
Affiliation(s)
- Camille Jung
- UMR843 INSERM, Université Sorbonne Paris Cité-Diderot, Hôpital Robert Debré, 75019 Paris, France
| | | | | |
Collapse
|
310
|
Ye Y, Jin X, Yue M, Chen S, Yu C, Li Y. The protective effect of oral colitis-derived proteins in a murine model of inflammatory bowel disease is associated with an increase in gammadelta T cells in large intestinal mucosa. Int J Colorectal Dis 2010; 25:1055-1062. [PMID: 20571813 DOI: 10.1007/s00384-010-0975-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/25/2010] [Indexed: 02/04/2023]
Abstract
BACKGROUNDS AND AIMS Oral tolerance has previously been shown effective in preventing several immune-mediated disorders in animal models. The aims of this study were to investigate the effect of oral colitis-extracted proteins (CEP) on dextran sulfate sodium (DSS)-induced colitis in BALB/c mice and to explore the relative role of the intestinal mucosal gammadelta T cells. METHODS The effect of five low oral doses of CEP on colitis was evaluated by clinical manifestation and histological lesions. Serum cytokines were measured by enzyme-linked immunosorbent assay. The percentages of the intestinal mucosal gammadelta T cells were evaluated by flow cytometry. RESULTS CEP-fed colitis mice showed less severe symptoms and histological injury than bovine serum albumin (BSA)-fed control mice. Tolerized mice developed an increase in TGF-beta1 and no change in IFN-gamma serum levels. Increases in TCRgammadelta(+) T cells and CD8alpha(+)TCRgammadelta(+) T cells in small intestinal mucosal lymphocytes and no quantitative change in large intestinal mucosal lymphocytes were demonstrated in colitis mice compared to untreated mice. The proportions of TCRgammadelta(+) T cells and CD8alpha(+)TCRgammadelta(+) T cells in large intestinal mucosal lymphocytes from CEP-fed colitis mice were significantly higher compared to BSA-fed controls. The disease activity index negatively correlated with the percentages of large intestinal mucosal gammadelta T cells. Furthermore, mucosal repair in repair-period mice was also accompanied by increases in TCRgammadelta(+) T cells and CD8alpha(+)TCRgammadelta(+) T cells in large intestinal mucosal lymphocytes. CONCLUSION Improvement of DSS-induced colitis that resulted from oral administration of colitis-extracted proteins is associated with an increase in gammadelta T cells in large intestinal mucosa.
Collapse
Affiliation(s)
- Yuefang Ye
- Department of Gastroenterology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, People's Republic of China
| | | | | | | | | | | |
Collapse
|
311
|
Chmill S, Kadow S, Winter M, Weighardt H, Esser C. 2,3,7,8-Tetrachlorodibenzo-p-dioxin impairs stable establishment of oral tolerance in mice. Toxicol Sci 2010; 118:98-107. [PMID: 20729464 DOI: 10.1093/toxsci/kfq232] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The toxic environmental pollutant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a potent immunomodulatory chemical. TCDD activates the aryl hydrocarbon receptor (AhR) and suppresses peripheral humoral and cellular adaptive immune responses. Though the major route of uptake is via food, little is known until now on the immunotoxic effects of TCDD on the gut-associated lymphoid tissue. We show here that AhR is strongly expressed along the small intestine, especially in intestinal epithelial cells (IEC). The AhR marker gene cyp1a1 is induced in IEC by oral TCDD exposure. We asked how TCDD affects oral tolerance, a unique function of mucosal immunity. C57BL/6 mice were injected with 10 μg/kg body weight TCDD and fed with ovalbumin (OVA) in a high-dose tolerization protocol. Mice were immunized and boosted with OVA on days 12, 23, and 55 after tolerization. Five of 14, 6 of 15, and 13 of 14 TCDD-treated mice generated OVA-specific immunoglobulin (Ig)G1 antibodies after the first, second, and third immunization with OVA, respectively. Only one mouse harbored anti-OVA IgG1 antibodies in the control group even after the third immunization with OVA. OVA-specific IgA in fecal samples of tolerized and TCDD-exposed mice could be detected at the levels of nontolerized mice, whereas completely absent in tolerant control mice. Correlated to this, we found in TCDD-treated mice an increase in interleukin-6 producing CD103+ dendritic cells (DC) present in the gut-draining mesenteric lymph nodes (MLN) and a small increase in the frequency of Th17 cells. Neither the frequencies nor the absolute numbers of immune cells in the lamina propria (LP) or in intraepithelial lymphocytes were changed by TCDD treatment. Our data not only have implications for food allergies in settings of environmental exposure but also raise concerns regarding the harmlessness of overdosing potential AhR agonist in food, which needs to be studied further.
Collapse
Affiliation(s)
- Stefanie Chmill
- Institute for Environmental Medical Research, Molecular Immunology Unit, 40225 Düsseldorf, Germany
| | | | | | | | | |
Collapse
|
312
|
Wu HY, Maron R, Tukpah AM, Weiner HL. Mucosal anti-CD3 monoclonal antibody attenuates collagen-induced arthritis that is associated with induction of LAP+ regulatory T cells and is enhanced by administration of an emulsome-based Th2-skewing adjuvant. THE JOURNAL OF IMMUNOLOGY 2010; 185:3401-7. [PMID: 20720210 DOI: 10.4049/jimmunol.1000836] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mucosal (nasal or oral) administration of anti-CD3 mAb is effective in ameliorating animal models of autoimmunity (experimental autoimmune encephalomyelitis, diabetes, and lupus) by inducing LAP(+) regulatory T cells. We tested this approach in an arthritis model using type II collagen. We found that nasal anti-CD3 was more effective than oral anti-CD3 in attenuating the development of arthritis. Nasal anti-CD3 induced a LAP(+) regulatory T cell that secreted high levels of IL-10 and suppressed collagen-specific T cell proliferation and anti-collagen Ab production. However, neither nasal nor oral anti-CD3 attenuated disease when given to animals with ongoing arthritis, and this was associated with a lack of induction of LAP(+) regulatory T cells. We found, however, that coadministration of a novel emulsome adjuvant, which enhances Th2 responses, resulted in the induction of LAP(+) regulatory T cells and suppression of ongoing arthritis by both nasal and oral anti-CD3. Suppression of arthritis by mucosal anti-CD3 was associated with less joint damage, a decrease of TNF-alpha and IFN-gamma mRNA expression in joints, and a reduction in anti-collagen Abs. These results demonstrate that mucosal anti-CD3 therapy may serve as a therapeutic approach in arthritis and that the biologic effect is enhanced by an emulsome-based adjuvant.
Collapse
Affiliation(s)
- Henry Yim Wu
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
313
|
Kunisawa J, Kiyono H. Aberrant interaction of the gut immune system with environmental factors in the development of food allergies. Curr Allergy Asthma Rep 2010; 10:215-21. [PMID: 20424996 DOI: 10.1007/s11882-010-0097-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The gastrointestinal immune system is a major component of the mucosal barrier, which maintains an immunologic homeostasis between the host and the harsh environment of the gut. This homeostasis is achieved by immunologic quiescence, and its dysregulation is thought to result from the development of immune diseases such as food allergies. Recent findings have revealed versatile pathways in the development of intestinal allergies to certain food antigens. In this review, we summarize the regulatory and quiescence mechanisms in the gut immune system and describe aberrant interactions between the host immune system and the gut environment in the development of food allergies.
Collapse
Affiliation(s)
- Jun Kunisawa
- Division of Mucosal Immunology, Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | | |
Collapse
|
314
|
Cholera-like enterotoxins and Regulatory T cells. Toxins (Basel) 2010; 2:1774-95. [PMID: 22069660 PMCID: PMC3153266 DOI: 10.3390/toxins2071774] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Revised: 06/23/2010] [Accepted: 06/28/2010] [Indexed: 01/25/2023] Open
Abstract
Cholera toxin (CT) and the heat-labile enterotoxin of E. coli (LT), as well as their non toxic mutants, are potent mucosal adjuvants of immunization eliciting mucosal and systemic responses against unrelated co-administered antigens in experimental models and in humans (non toxic mutants). These enterotoxins are composed of two subunits, the A subunit, responsible for an ADP-ribosyl transferase activity and the B subunit, responsible for cell binding. Paradoxically, whereas the whole toxins have adjuvant properties, the B subunits of CT (CTB) and of LT (LTB) have been shown to induce antigen specific tolerance when administered mucosally with antigens in experimental models as well as, recently, in humans, making them an attractive strategy to prevent or treat autoimmune or allergic disorders. Immunomodulation is a complex process involving many cell types notably antigen presenting cells and regulatory T cells (Tregs). In this review, we focus on Treg cells and cholera-like enterotoxins and their non toxic derivates, with regard to subtype, in vivo/in vitro effects and possible role in the modulation of immune responses to coadministered antigens.
Collapse
|
315
|
Verhasselt V. Oral tolerance in neonates: from basics to potential prevention of allergic disease. Mucosal Immunol 2010; 3:326-33. [PMID: 20485330 DOI: 10.1038/mi.2010.25] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Oral tolerance refers to the observation that prior feeding of an antigen induces local and systemic immune tolerance to that antigen. Physiologically, this process is probably of central importance for preventing inflammatory responses to the numerous dietary and microbial antigens present in the gut. Defective oral tolerance can lead to gut inflammatory disease, food allergies, and celiac disease. In the last two cases, the diseases develop early in life, stressing the necessity of understanding how oral tolerance is set up in neonates. This article reviews the parameters that have been outlined in adult animal models as necessary for tolerance induction and assesses whether these factors operate in neonates. In addition, we highlight the factors that are specific for this period of life and discuss how they could have an impact on oral tolerance. We pay particular attention to maternal influence on early oral tolerance induction through breast-feeding and outline the major parameters that could be modified to optimize tolerance induction in early life and possibly prevent allergic diseases.
Collapse
|
316
|
Bonagura VR, Hatam LJ, Rosenthal DW, de Voti JA, Lam F, Steinberg BM, Abramson AL. Recurrent respiratory papillomatosis: a complex defect in immune responsiveness to human papillomavirus-6 and -11. APMIS 2010; 118:455-70. [PMID: 20553528 DOI: 10.1111/j.1600-0463.2010.02617.x] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Recurrent respiratory papillomatosis (RRP) is a rare disease of the larynx caused by infection with human papillomaviruses (HPV) -6 or -11, associated with significant morbidity and on occasion mortality. Here we summarize our current understanding of the permissive adaptive and innate responses made by patients with RRP that support chronic HPV infection and prevent immune clearance of these viruses. Furthermore, we provide new evidence of T(H)2-like polarization in papillomas and blood of patients with RRP, restricted CD4 and CD8 Vbeta repertoires, the effect of HPV-11 early protein E6 on T-cell alloreactivity, enriched Langerhans cell presence in papillomas, and evidence that natural killer cells are dysfunctional in RRP. We review the immunogenetic mechanisms that regulate the dysfunctional responses made by patients with RRP in response to HPV infection of the upper airway. In addition, we are identifying T-cell epitopes on HPV-11 early proteins, in the context of human leukocyte antigen (HLA) class II alleles enriched in RRP that should help generate a therapeutic vaccine. Taken together, RRP is a complex, multigene disease manifesting as a tissue and HPV-specific, immune deficiency that prevents effective clearance and/or control of HPV-6 and -11 infection.
Collapse
Affiliation(s)
- Vincent R Bonagura
- Feinstein Institute for Medical Research, North Shore-Long Island Jewish Health System, Manhasset, NY, USA.
| | | | | | | | | | | | | |
Collapse
|
317
|
Abstract
Regulatory T cells (Tregs) and the PD-1: PD-ligand (PD-L) pathway are both critical to terminating immune responses. Elimination of either can result in the breakdown of tolerance and the development of autoimmunity. The PD-1: PD-L pathway can thwart self-reactive T cells and protect against autoimmunity in many ways. In this review, we highlight how PD-1 and its ligands defend against potentially pathogenic self-reactive effector T cells by simultaneously harnessing two mechanisms of peripheral tolerance: (i) the promotion of Treg development and function and (ii) the direct inhibition of potentially pathogenic self-reactive T cells that have escaped into the periphery. Treg cells induced by the PD-1 pathway may also assist in maintaining immune homeostasis, keeping the threshold for T-cell activation high enough to safeguard against autoimmunity. PD-L1 expression on non-hematopoietic cells as well as hematopoietic cells endows PD-L1 with the capacity to promote Treg development and enhance Treg function in lymphoid organs and tissues that are targets of autoimmune attack. At sites where transforming growth factor-beta is present (e.g. sites of immune privilege or inflammation), PD-L1 may promote the de novo generation of Tregs. When considering the consequences of uncontrolled immunity, it would be therapeutically advantageous to manipulate Treg development and sustain Treg function. Thus, this review also discusses how the PD-1 pathway regulates a number of autoimmune diseases and the therapeutic potential of PD-1: PD-L modulation.
Collapse
Affiliation(s)
- Loise M. Francisco
- Departments of Pathology, Harvard Medical School, Boston, MA, USA
- Brigham & Women’s Hospital, Boston, MA, USA
| | - Peter T. Sage
- Departments of Pathology, Harvard Medical School, Boston, MA, USA
| | - Arlene H. Sharpe
- Departments of Pathology, Harvard Medical School, Boston, MA, USA
- Brigham & Women’s Hospital, Boston, MA, USA
| |
Collapse
|
318
|
|
319
|
Pugliese A. Insulin: a critical autoantigen and potential therapeutic agent in Type 1 diabetes. Expert Rev Clin Immunol 2010; 2:419-31. [PMID: 20476913 DOI: 10.1586/1744666x.2.3.419] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Insulin is a polypeptide hormone secreted by pancreatic beta-cells and is critical for glucose homeostasis. Abnormalities in insulin secretion result in various forms of diabetes. Type 1A diabetes is an autoimmune form in which insulin has been identified as a critical autoantigen. Recent studies have identified genetic determinants of insulin-specific autoimmune responses and insulin epitopes targeted by autoreactive T lymphocytes. The study of insulin as an autoantigen has also led to discoveries about basic mechanisms of immunological tolerance and autoimmunity. Experimental and clinical evidence suggests that insulin and insulin-derived peptides may delay and perhaps prevent the development of diabetes. Further clinical trials may identify effective treatment modalities for inhibiting diabetogenic autoimmunity and preventing disease development.
Collapse
Affiliation(s)
- Alberto Pugliese
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Avenue, Miami, FL 33136, USA.
| |
Collapse
|
320
|
Fort MM, Narayanan PK. Manipulation of Regulatory T-Cell Function by Immunomodulators: A Boon or a Curse? Toxicol Sci 2010; 117:253-62. [DOI: 10.1093/toxsci/kfq136] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
321
|
B7-H1/CD80 interaction is required for the induction and maintenance of peripheral T-cell tolerance. Blood 2010; 116:1291-8. [PMID: 20472828 DOI: 10.1182/blood-2010-01-265975] [Citation(s) in RCA: 267] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
T-cell tolerance is the central program that prevents harmful immune responses against self-antigens, in which inhibitory PD-1 signal given by B7-H1 interaction plays an important role. Recent studies demonstrated that B7-H1 binds CD80 besides PD-1, and B7-H1/CD80 interaction also delivers inhibitory signals in T cells. However, a role of B7-H1/CD80 signals in regulation of T-cell tolerance has yet to be explored. We report here that attenuation of B7-H1/CD80 signals by treatment with anti-B7-H1 monoclonal antibody, which specifically blocks B7-H1/CD80 but not B7-H1/PD-1, enhanced T-cell expansion and prevented T-cell anergy induction. In addition, B7-H1/CD80 blockade restored Ag responsiveness in the previously anergized T cells. Experiments using B7-H1 or CD80-deficient T cells indicated that an inhibitory signal through CD80, but not B7-H1, on T cells is responsible in part for these effects. Consistently, CD80 expression was detected on anergic T cells and further up-regulated when they were re-exposed to the antigen (Ag). Finally, blockade of B7-H1/CD80 interaction prevented oral tolerance induction and restored T-cell responsiveness to Ag previously tolerized by oral administration. Taken together, our findings demonstrate that the B7-H1/CD80 pathway is a crucial regulator in the induction and maintenance of T-cell tolerance.
Collapse
|
322
|
Luo X, Herold KC, Miller SD. Immunotherapy of type 1 diabetes: where are we and where should we be going? Immunity 2010; 32:488-99. [PMID: 20412759 DOI: 10.1016/j.immuni.2010.04.002] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 03/22/2010] [Accepted: 03/31/2010] [Indexed: 02/06/2023]
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disorder characterized by destruction of insulin-producing pancreatic beta cells. Many broad-based immunosuppressive and antigen-specific immunoregulatory therapies have been and are currently being evaluated for their utility in the prevention and treatment of T1D. Looking forward, this review discusses the potential therapeutic use of antigen-specific tolerance strategies, including tolerance induced by "tolerogenic" antigen-presenting cells pulsed with diabetogenic antigens and transfer of induced or expanded regulatory T cells, which have demonstrated efficacy in nonobese diabetic (NOD) mice. Depending on the time of therapeutic intervention in the T1D disease process, antigen-specific immunoregulatory strategies may be employed as monotherapies, or in combination with short-term tolerance-promoting immunoregulatory drugs and/or drugs promoting differentiation of insulin-producing beta cells from endogenous progenitors.
Collapse
Affiliation(s)
- Xunrong Luo
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | | |
Collapse
|
323
|
Induction of regulatory T cells decreases adipose inflammation and alleviates insulin resistance in ob/ob mice. Proc Natl Acad Sci U S A 2010; 107:9765-70. [PMID: 20445103 DOI: 10.1073/pnas.0908771107] [Citation(s) in RCA: 272] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Leptin-deficient ob/ob mice are overweight, develop insulin resistance, and serve as a model for type 2 diabetes (T2D). Studies suggest that inflammatory pathways are linked to the development of insulin resistance and T2D both in animals and humans. We asked whether the induction of regulatory T cells (Tregs) could alleviate the pathological and metabolic abnormalities in ob/ob mice. We induced TGF-beta-dependent CD4(+) latency-associated peptide (LAP)-positive Tregs by oral administration of anti-CD3 antibody plus beta-glucosylceramide. We found a decrease in pancreatic islet cell hyperplasia, fat accumulation in the liver, and inflammation in adipose tissue, accompanied by lower blood glucose and liver enzymes. In addition, treated animals had decreased CD11b(+)F4/80(+) macrophages and TNF-alpha in adipose tissue. Adoptive transfer of orally induced CD4(+)LAP(+) Tregs ameliorated metabolic and cytokine abnormalities. Our results demonstrate the importance of inflammation in T2D and identify a unique immunological approach for treatment of T2D by the induction of Tregs.
Collapse
|
324
|
Koeberl DD, Kishnani PS. Immunomodulatory gene therapy in lysosomal storage disorders. Curr Gene Ther 2010; 9:503-10. [PMID: 19807648 DOI: 10.2174/156652309790031094] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Accepted: 09/29/2009] [Indexed: 11/22/2022]
Abstract
Significant advances in therapy for lysosomal storage disorders have occurred with an accelerating pace over the past decade. Although enzyme replacement therapy has improved the outcome of lysosomal storage disorders, antibody responses have occurred and sometimes prevented efficacy, especially in cross-reacting immune material negative patients with Pompe disease. Preclinical gene therapy experiments have revealed the relevance of immune responses to long-term efficacy. The choice of regulatory cassette played a critical role in evading humoral and cellular immune responses to gene therapy in knockout mouse models, at least in adult animals. Liver-specific regulatory cassettes prevented antibody formation and enhanced the efficacy of gene therapy. Regulatory T cells prevented transgene directed immune responses, as shown by adoptive transfer of antigen-specific immune tolerance to enzyme therapy. Immunomodulatory gene therapy with a very low vector dose could enhance the efficacy of enzyme therapy in Pompe disease and other lysosomal storage disorders.
Collapse
Affiliation(s)
- Dwight D Koeberl
- Division of Medical Genetics/Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA.
| | | |
Collapse
|
325
|
Fiocchi A, Brozek J, Schünemann H, Bahna SL, von Berg A, Beyer K, Bozzola M, Bradsher J, Compalati E, Ebisawa M, Guzman MA, Li H, Heine RG, Keith P, Lack G, Landi M, Martelli A, Rancé F, Sampson H, Stein A, Terracciano L, Vieths S. World Allergy Organization (WAO) Diagnosis and Rationale for Action against Cow's Milk Allergy (DRACMA) Guidelines. World Allergy Organ J 2010; 3:57-161. [PMID: 23268426 PMCID: PMC3488907 DOI: 10.1097/wox.0b013e3181defeb9] [Citation(s) in RCA: 194] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
326
|
Merson TD, Binder MD, Kilpatrick TJ. Role of cytokines as mediators and regulators of microglial activity in inflammatory demyelination of the CNS. Neuromolecular Med 2010; 12:99-132. [PMID: 20411441 DOI: 10.1007/s12017-010-8112-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2009] [Accepted: 02/26/2010] [Indexed: 12/11/2022]
Abstract
As the resident innate immune cells of the central nervous system (CNS), microglia fulfil a critical role in maintaining tissue homeostasis and in directing and eliciting molecular responses to CNS damage. The human disease Multiple Sclerosis and animal models of inflammatory demyelination are characterized by a complex interplay between degenerative and regenerative processes, many of which are regulated and mediated by microglia. Cellular communication between microglia and other neural and immune cells is controlled to a large extent by the activity of cytokines. Here we review the role of cytokines as mediators and regulators of microglial activity in inflammatory demyelination, highlighting their importance in potentiating cell damage, promoting neuroprotection and enhancing cellular repair in a context-dependent manner.
Collapse
Affiliation(s)
- Tobias D Merson
- Florey Neuroscience Institutes, Centre for Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia.
| | | | | |
Collapse
|
327
|
Expression of a ricin toxin B subunit: insulin fusion protein in edible plant tissues. Mol Biotechnol 2010; 44:90-100. [PMID: 19898971 DOI: 10.1007/s12033-009-9217-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Onset of juvenile Type 1 diabetes (T1D) occurs when autoreactive lymphocytes progressively destroy the insulin-producing beta-cells in the pancreatic Islets of Langerhans. The increasing lack of insulin and subsequent onset of hyperglycemia results in increased damage to nerves, blood vessels, and tissues leading to the development of a host of severe disease symptoms resulting in premature morbidity and mortality. To enhance restoration of normoglycemia and immunological homeostasis generated by lymphocytes that mediate the suppression of autoimmunity, the non-toxic B chain of the plant AB enterotoxin ricin (RTB), a castor bean lectin binding a variety of epidermal cell receptors, was genetically linked to the coding region of the proinsulin gene (INS) and expressed as a fusion protein (INS-RTB) in transformed potato plants. This study is the first documented example of a plant enterotoxin B subunit linked to an autoantigen and expressed in transgenic plants for enhanced immunological suppression of T1D autoimmunity.
Collapse
|
328
|
Mortimer L, Chadee K. The immunopathogenesis of Entamoeba histolytica. Exp Parasitol 2010; 126:366-80. [PMID: 20303955 DOI: 10.1016/j.exppara.2010.03.005] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2009] [Revised: 03/08/2010] [Accepted: 03/12/2010] [Indexed: 12/17/2022]
Abstract
Amebiasis is the disease caused by the enteric dwelling protozoan parasite Entamoeba histolytica. The WHO considers amebiasis as one of the major health problems in developing countries; it is surpassed by only malaria and schistosomiasis for death caused by parasitic infection. E. histolytica primarily lives in the colon as a harmless commensal, but is capable of causing devastating dysentery, colitis and liver abscess. What triggers the switch to a pathogenic phenotype and the onset of disease is unknown. We are becoming increasingly aware of the complexity of the host-parasite interaction. During chronic stages of amebiasis, the host develops an immune response that is incapable of eliminating tissue resident parasites, while the parasite actively immunosuppresses the host. However, most individuals with symptomatic infections succumb only to an episode of dysentery. Why most halt invasion and a minority progress to chronic disease remains poorly understood. This review presents a current understanding of the immune processes that shape the outcome of E. histolytica infections during its different stages.
Collapse
Affiliation(s)
- Leanne Mortimer
- Faculty of Medicine, Department of Microbiology and Infectious Diseases, University of Calgary Health Sciences Centre, 3330 Hospital Dr. NW, Calgary, Alberta, Canada
| | | |
Collapse
|
329
|
Rottiers P, De Smedt T, Steidler L. Modulation of gut-associated lymphoid tissue functions with genetically modified Lactococcus lactis. Int Rev Immunol 2010; 28:465-86. [PMID: 19954359 DOI: 10.3109/08830180903197498] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Lactic acid bacteria are a group of taxonomically diverse, Gram-positive food-grade bacteria that have been safely consumed throughout history. The lactic acid bacterium Lactococcus lactis, well-known for its use in the manufacture of cheese, can be genetically engineered and orally formulated to deliver therapeutic proteins in the gastrointestinal tract. This review focuses on the genetic engineering of Lactococcus lactis to secrete high-quality, correctly processed bioactive molecules derived from a eukaryotic background. The therapeutic applications of these genetically modified strains are discussed, with special regards to immunomodulation.
Collapse
|
330
|
Abstract
Natural regulatory T cells (nTregs) are defined by their inherent ability to establish and maintain peripheral self-tolerance. In recent years, the development of nTregs has come under close examination with the advent of Forkhead Box P3 protein (FOXP3)-green fluorescent protein reporter mice that pinpointed the initiation of FOXP3 expression within the thymus. The mechanism and pathway of nTreg development has only recently been studied in detail and to a large degree remains unclear. In this review, we will discuss our current understanding of nTreg lineage choice and development from a cellular and intracellular standpoint.
Collapse
Affiliation(s)
- Matthew L Bettini
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105-3678, USA
| | | |
Collapse
|
331
|
Barabas AZ, Cole CD, Barabas AD, Graeff RM, Lafreniere R, Weir DM. Modified Vaccination Technique for Prophylactic and Therapeutic Applications to Combat Endogenous Antigen-Induced Disorders. Scand J Immunol 2010; 71:125-33. [DOI: 10.1111/j.1365-3083.2009.02360.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
332
|
Tremblay R, Wang D, Jevnikar AM, Ma S. Tobacco, a highly efficient green bioreactor for production of therapeutic proteins. Biotechnol Adv 2010; 28:214-21. [PMID: 19961918 PMCID: PMC7132750 DOI: 10.1016/j.biotechadv.2009.11.008] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Revised: 11/06/2009] [Accepted: 11/19/2009] [Indexed: 01/06/2023]
Abstract
Molecular farming of pharmaceuticals in plants has the potential to provide almost unlimited amounts of recombinant proteins for use in disease diagnosis, prevention or treatment. Tobacco has been and will continue to be a major crop for molecular farming and offers several practical advantages over other crops. It produces significant leaf biomass, has high soluble protein content and is a non-food crop, minimizing the risk of food-chain contamination. This, combined with its flexibility and highly-efficient genetic transformation/regeneration, has made tobacco particularly well suited for plant-based production of biopharmaceutical products. The goal of this review is to provide an update on the use of tobacco for molecular farming of biopharmaceuticals as well the technologies developed to enhance protein production/purification/efficacy. We show that tobacco is a robust biological reactor with a multitude of applications and may hold the key to success in plant molecular farming.
Collapse
Affiliation(s)
- Reynald Tremblay
- Department of Biology, University of Western Ontario, London, Ontario, Canada N6A 5B7
| | - David Wang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544-1014, USA
| | - Anthony M. Jevnikar
- Transplantation Immunology Group, Lawson Health Research Institute, London, Ontario, Canada N6A 4G5
| | - Shengwu Ma
- Department of Biology, University of Western Ontario, London, Ontario, Canada N6A 5B7
- Transplantation Immunology Group, Lawson Health Research Institute, London, Ontario, Canada N6A 4G5
- Plantigen Inc., 700 Collip Circle, London, Ontario, Canada N6G 4X8
| |
Collapse
|
333
|
Hasselberg A, Ekman L, Yrlid LF, Schön K, Lycke NY. ADP-ribosylation controls the outcome of tolerance or enhanced priming following mucosal immunization. THE JOURNAL OF IMMUNOLOGY 2010; 184:2776-84. [PMID: 20142362 DOI: 10.4049/jimmunol.0901445] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Accumulating evidence suggests that the dichotomy between tolerance and active IgA immunity in mucosal immune responses is regulated at the APC level. Therefore, immunomodulation of the APC could be an effective mechanism to control the two response patterns. In this study, we demonstrate that ADP-ribosylation controls the outcome of tolerance or active effector T cell immunity to an internal peptide p323-339 from OVA inserted into the cholera toxin (CT)-derived CTA1-OVA-DD adjuvant. We found that a single point mutation, CTA1R7K-OVA-DD, resulting in lack of enzymatic activity, promoted peptide-specific tolerance in TCR transgenic CD4(+) T cells following a single intranasal (i.n.) treatment. The CTA1R7K-OVA-DD-induced tolerance was strong, long-lasting, and impaired the ability of adoptively transferred naive peptide-specific CD4(+) T cells to respond to Ag-challenge, irrespective if this was given i.p or i.n. The tolerance correlated with induction of regulatory T cells of the regulatory T type 1 characterized by CD25(-)Foxp3(-)CD4(+) T cells producing IL-10. In contrast, in IL-10-deficient mice, no peptide-specific tolerance was observed, and these mice exhibited unimpaired CD4(+) T cell responsiveness to recall Ag irrespective of if they were untreated (PBS) or treated i.n. with CTA1R7K-OVA-DD. Thus, for the first time, we can provide unequivocal proof that ADP-ribosylation can control the outcome of mucosal Ag exposure from tolerance to an enhanced effector CD4(+) T cell response. The exploitation of this system for clinical treatment of autoimmune diseases is discussed.
Collapse
Affiliation(s)
- Annemarie Hasselberg
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Research Center, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | |
Collapse
|
334
|
Rescigno M. Functional specialization of antigen presenting cells in the gastrointestinal tract. Curr Opin Immunol 2010; 22:131-6. [DOI: 10.1016/j.coi.2009.12.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Revised: 12/22/2009] [Accepted: 12/23/2009] [Indexed: 01/21/2023]
|
335
|
Verhasselt V. Neonatal tolerance under breastfeeding influence: the presence of allergen and transforming growth factor-beta in breast milk protects the progeny from allergic asthma. J Pediatr 2010; 156:S16-20. [PMID: 20105659 DOI: 10.1016/j.jpeds.2009.11.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Once the umbilical cord has been cut, immunologists have often looked at the neonate as an entity that develops on its own. For years, breast milk was considered mainly as a source of nutrients for the developing child. The extensive observations that breastfeeding affords protection toward infectious diseases and could reduce by more than the half the mortality rate because of common infections have added another key role to breastfeeding. This protection relies in great part on the passive transfer through breast milk of high amounts of microbe-specific immunoglobulins that compensate for the deficiency of immunoglobulins synthesis during the first year of life. Here, we will present and discuss our data showing how breast milk can actively shape the immune response of the progeny, particularly in the context of allergic disease. Indeed, our data obtained in a mouse model suggest that the protection attributed to breastfeeding toward asthma development might rely on immune tolerance induction. For this to occur, the mother mice needed to be exposed to the allergen by aerosol or oral route during the lactation period, which resulted into the transfer of the allergen to breast milk. The presence of the allergen together with transforming growth factor-beta in breast milk was necessary and sufficient to induce the development of regulatory T lymphocytes in the progeny and their protection from asthma development. If confirmed in human beings, this study may suggest new strategies for asthma prevention such as deliberate exposure of mother to allergens during breastfeeding and qualitative modification of artificial milks.
Collapse
|
336
|
Thorburn AN, Hansbro PM. Harnessing regulatory T cells to suppress asthma: from potential to therapy. Am J Respir Cell Mol Biol 2010; 43:511-9. [PMID: 20097830 DOI: 10.1165/rcmb.2009-0342tr] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Regulatory T cells (Tregs) play an essential role in maintaining the homeostatic balance of immune responses. Asthma is an inflammatory condition of the airways that is driven by dysregulated immune responses toward normally innocuous antigens. Individuals with asthma have fewer and less functional Tregs, which may lead to uncontrolled effector cell responses and promote proasthmatic responses of T helper type 2, T helper 17, natural killer T, antigen-presenting, and B cells. Tregs have the capacity to either directly or indirectly suppress these responses. Hence, the induced expansion of functional Tregs in predisposed or individuals with asthma is a potential approach for the prevention and treatment of asthma. Infection by a number of micro-organisms has been associated with reduced prevalence of asthma, and many infectious agents have been shown to induce Tregs and reduce allergic airways disease in mouse models. The translation of the regulatory and therapeutic properties of infectious agents for use in asthma requires the identification of key modulatory components and the development and trial of effective immunoregulatory therapies. Further translational and clinical research is required for the induction of Tregs to be harnessed as a therapeutic strategy for asthma.
Collapse
Affiliation(s)
- Alison N Thorburn
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, the University of Newcastle, Newcastle, Australia
| | | |
Collapse
|
337
|
Transfer of tolerance to collagen type V suppresses T-helper-cell-17 lymphocyte-mediated acute lung transplant rejection. Transplantation 2010; 88:1341-8. [PMID: 20029330 DOI: 10.1097/tp.0b013e3181bcde7b] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Rat lung allograft rejection is mediated by collagen type V (col(V)) specific T-helper-cell 17 (Th17) cells. Adoptive transfer of these cells is sufficient to induce rejection pathology in isografts, whereas tolerance to col(V) suppresses allograft rejection. Therefore, we tested whether regulatory T cells from tolerant rats could suppress the Th17-mediated rejection in the syngeneic model of lung transplantation. METHODS Rats were subjected to syngeneic left lung transplantation, and acute rejection was induced by adoptive transfer of lymph node cells from col(V)-immunized rats. Tolerance was induced by intravenous injection of col(V), and spleen lymphocytes were used for adoptive transfer. CD4+ T cells were depleted using magnetic beads. Lung isografts were analyzed using micro-positron emission tomography imaging and histochemistry. The transvivo delayed type hypersensitivity assay was used to analyze the Th17 response. RESULTS Adoptive cotransfer of col(V)-specific effector cells with cells from col(V)-tolerized rats suppressed severe vasculitis and bronchiolitis with parenchymal inflammation, and the expression of interleukin (IL)-17 transcripts in mediastinal lymph nodes induced by effector cells alone. Analysis by transvivo delayed type hypersensitivity showed that the reactivity to col(V) was dependent on the presence of tumor necrosis factor-alpha and IL-17 but not interferon-gamma. Depletion of CD4+ T cells from the suppressor cell population abrogated the col(V)-specific protection. CONCLUSION Th17-mediated acute rejection after lung transplantation is ameliorated by CD4+ col(V)-specific regulatory T cells. The mechanism for this Th17 suppression is consistent with tolerance induction to col(V). The goal of transplantation treatment, therefore, should target Th17 development and not suppression of T-cell activation by suppressing IL-2.
Collapse
|
338
|
Abstract
PURPOSE OF REVIEW Food allergy is a serious and growing problem. Although the current standard of care for patients with food allergies is based on avoidance of the trigger, increased understanding of the mechanisms involved in oral tolerance has shifted focus of treatment and prevention toward inducing tolerance. Here we discuss the relationship of food allergy to oral tolerance and review recent oral tolerance studies, focusing on the mechanistic role of antigen presenting cells and the generation of regulatory T cells in mice and humans. RECENT FINDINGS Specialized intestinal antigen presenting cells are conditioned by spatial and soluble microenvironmental factors to promote tolerance to dietary antigen primarily via the induction of regulatory T cells. Retinoic acid, a vitamin A metabolite, has been recently identified as a key environmental factor in this process. SUMMARY Future clinical trials for food allergy immunotherapy will benefit from approaches, which target the oral tolerance pathways currently being elucidated.
Collapse
|
339
|
Velosa APP, Teodoro WR, dos Anjos DM, Konno R, Oliveira CC, Katayama MLH, Parra ER, Capelozzi VL, Yoshinari NH. Collagen V-induced nasal tolerance downregulates pulmonary collagen mRNA gene and TGF-beta expression in experimental systemic sclerosis. Respir Res 2010; 11:1. [PMID: 20047687 PMCID: PMC2817654 DOI: 10.1186/1465-9921-11-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Accepted: 01/04/2010] [Indexed: 11/10/2022] Open
Abstract
Background The purpose of this study was to evaluate collagen deposition, mRNA collagen synthesis and TGF-beta expression in the lung tissue in an experimental model of scleroderma after collagen V-induced nasal tolerance. Methods Female New Zealand rabbits (N = 12) were immunized with 1 mg/ml of collagen V in Freund's adjuvant (IM). After 150 days, six immunized animals were tolerated by nasal administration of collagen V (25 μg/day) (IM-TOL) daily for 60 days. The collagen content was determined by morphometry, and mRNA expressions of types I, III and V collagen were determined by Real-time PCR. The TGF-beta expression was evaluated by immunostaining and quantified by point counting methods. To statistic analysis ANOVA with Bonferroni test were employed for multiple comparison when appropriate and the level of significance was determined to be p < 0.05. Results IM-TOL, when compared to IM, showed significant reduction in total collagen content around the vessels (0.371 ± 0.118 vs. 0.874 ± 0.282, p < 0.001), bronchioles (0.294 ± 0.139 vs. 0.646 ± 0.172, p < 0.001) and in the septal interstitium (0.027 ± 0.014 vs. 0.067 ± 0.039, p = 0.026). The lung tissue of IM-TOL, when compared to IM, showed decreased immunostaining of types I, III and V collagen, reduced mRNA expression of types I (0.10 ± 0.07 vs. 1.0 ± 0.528, p = 0.002) and V (1.12 ± 0.42 vs. 4.74 ± 2.25, p = 0.009) collagen, in addition to decreased TGF-beta expression (p < 0.0001). Conclusions Collagen V-induced nasal tolerance in the experimental model of SSc regulated the pulmonary remodeling process, inhibiting collagen deposition and collagen I and V mRNA synthesis. Additionally, it decreased TGF-beta expression, suggesting a promising therapeutic option for scleroderma treatment.
Collapse
Affiliation(s)
- Ana Paula P Velosa
- Rheumatology Division of the School of Medicine of the University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
340
|
Israeli E, Ilan Y. Oral administration of Alequel, a mixture of autologous colon-extracted proteins for the treatment of Crohn's disease. Therap Adv Gastroenterol 2010; 3:23-30. [PMID: 21180587 PMCID: PMC3002565 DOI: 10.1177/1756283x09351733] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The pathogenesis of Crohn's disease involves an immune-mediated damage to the gut mucosa. Current developed therapies are based on the use of immunosuppressive drugs that can lead to significant drug-related adverse responses. There is a need for a therapeutic strategy that is more specific and less global in its effect on the immune system. Oral tolerance is an active process wherein oral administration of antigens is associated with the induction of regulatory cells and the suppression of effector cells directed toward specific and nonspecific antigens. Studies in animal models of experimental colitis suggest that oral administration of proteins extracted from the gut can induce tolerance and alleviate the disease symptoms. Recent clinical trials showed that oral administration of Alequel, an autologous protein-containing colon extract, to patients with Crohn's disease is safe and may be effective as a therapeutic modality for treating the disease. This treatment was associated with disease-associated antigen alterations of the immune response in the patients. Oral administration of Alequel could provide a patient-tailored approach that is side-effect-free for the treatment of patients with Crohn's disease.
Collapse
Affiliation(s)
- Eran Israeli
- Gastroenterology and Liver Units, Department of Medicine, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Yaron Ilan
- Gastroenterology and Liver Units, Department of Medicine, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| |
Collapse
|
341
|
Jabri B, Sollid LM. Tissue-mediated control of immunopathology in coeliac disease. Nat Rev Immunol 2009; 9:858-70. [PMID: 19935805 DOI: 10.1038/nri2670] [Citation(s) in RCA: 234] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Coeliac disease is an inflammatory disorder with autoimmune features that is characterized by destruction of the intestinal epithelium and remodelling of the intestinal mucosa following the ingestion of dietary gluten. A common feature of coeliac disease and many organ-specific autoimmune diseases is a central role for T cells in causing tissue destruction. In this Review, we discuss the emerging hypothesis that, in coeliac disease, intestinal tissue inflammation--induced either by infectious agents or by gluten--is crucial for activating T cells and eliciting their tissue-destructive effector functions.
Collapse
Affiliation(s)
- Bana Jabri
- Department of Medicine, University of Chicago, Chicago, Illinois 60637, USA.
| | | |
Collapse
|
342
|
Wang XT, Zheng PY, Luo Y, Liu ZQ, Zhang LL. TIM4 and TIM1 modulate the function of CD4 +CD25 + T regulatory cells in mice with food allergy. Shijie Huaren Xiaohua Zazhi 2009; 17:3507-3513. [DOI: 10.11569/wcjd.v17.i34.3507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To evaluate the function of T regulatory (Treg) cells and determine the role of T cell immunoglobulin and mucin domain-containing protein 4 (TIM4) and TIM1 in modulating the function of Treg cells in mice with food allergy.
METHODS: Thirty-two BALB/c mice fed an ovalbumin (OVA)-free diet were randomly and equally divided into four groups: normal saline (NS) group, staphylococcal enterotoxin B (SEB) plus OVA group, anti-TIM1 antibody plus SEB and OVA group, and anti-TIM4 antibody plus SEB and OVA group. Mice in the four groups were sensitized by intraperitoneal injections of NS, SEB/OVA, anti-TIM1/SEB/OVA, and anti-TIM4/SEB/OVA on days 0, 3 and 9, respectively. All mice (except the NS group) were challenged by intraperitoneal injections of SEB/OVA on days 7 and 14, respectively. The expression of forkhead box P3 (FOXP3) mRNA in the jejunum and spleen and TIM4 mRNA in the jejunum was measured by reverse transcription-polymerase chain reaction (RT-PCR). The levels of transforming growth factor β1 (TGF-β1) and interleukin-10 (IL-10) in the serum were analyzed by enzyme-linked immunosorbent assay (ELISA). The expression of TGF-β1 and IL-10 proteins in the jejunum was detected by immunohistochemistry.
RESULTS: Compared with the NS group, the expression levels of FOXP3 mRNA in the jejunum and spleen significantly decreased (0.401 ± 0.145 vs 0.732 ± 0.162 and 0.407 ± 0.082 vs 0.691 ± 0.145, respectively; both P < 0.05), the expression level of TIM4 mRNA increased significantly (P < 0.05), and the levels of TGF-β1 in the serum and jejunum decreased significantly (7859.853 ± 126.704 ng/L vs 8342.814 ± 488.461 ng/L and 108.834 ± 9.634 ng/L vs 156.298 ± 12.002 ng/L, respectively; both P < 0.05) in the SEB/OVA group. In contrast, the expression levels of FOXP3 mRNA in the jejunum and spleen and TGF-β1 in the serum and jejunum were significantly higher in the anti-TIM1/SEB/OVA and anti-TIM4/SEB/OVA groups than in the SEB/OVA group (all P < 0.05).
CONCLUSION: Treg cells in SEB/OVA-sensitized mice are dysfunctional. Pretreatment with anti-TIM1 or anti-TIM4 can restore the function of Treg cells, suggesting that the TIM4-TIM1 pathway may play a key role in the development of food allergy.
Collapse
|
343
|
Triggering and modulation of the host-parasite interplay byEchinococcus multilocularis: a review. Parasitology 2009; 137:557-68. [DOI: 10.1017/s0031182009991533] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
SUMMARYAs more facts emerge regarding the ways in whichE. multilocularis-derived molecules trigger the host immune response and modulate the host-parasite interplay, it becomes possible to envisage how the parasite can survive and proliferate in its intermediate host, while in other hosts it dies out. Through effects on cells of both the innate and adaptive arms of the immune response,E. multiloculariscan orchestrate a range of outcomes that are beneficial not only to the parasite, in terms of facilitating its intrahepatic proliferation and maturation, and thus life cycle over all, but also to its intermediate host, in limiting pathology. The present review deals with the role of metacestode surface molecules as well as excretory/secretory (E/S) metabolic products of the parasite in the modulation of the host responses such as to optimize its own survival.
Collapse
|
344
|
Simioni PU, Fernandes LGR, Gabriel DL, Tamashiro WMSC. Effect of aging and oral tolerance on dendritic cell function. Braz J Med Biol Res 2009; 43:68-76. [PMID: 19967261 DOI: 10.1590/s0100-879x2009007500024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Accepted: 11/12/2009] [Indexed: 12/13/2022] Open
Abstract
Oral tolerance can be induced in some mouse strains by gavage or spontaneous ingestion of dietary antigens. In the present study, we determined the influence of aging and oral tolerance on the secretion of co-stimulatory molecules by dendritic cells (DC), and on the ability of DC to induce proliferation and cytokine secretion by naive T cells from BALB/c and OVA transgenic (DO11.10) mice. We observed that oral tolerance could be induced in BALB/c mice (N = 5 in each group) of all ages (8, 20, 40, 60, and 80 weeks old), although a decline in specific antibody levels was observed in the sera of both tolerized and immunized mice with advancing age (40 to 80 weeks old). DC obtained from young, adult and middle-aged (8, 20, and 40 weeks old) tolerized mice were less efficient (65, 17 and 20%, respectively) than DC from immunized mice (P < 0.05) in inducing antigen-specific proliferation of naive T cells from both BALB/c and DO11.10 young mice, or in stimulating IFN-g, IL-4 and IL-10 production. However, TGF-beta levels were significantly elevated in co-cultures carried out with DC from tolerant mice (P < 0.05). DC from both immunized and tolerized old and very old (60 and 80 weeks old) mice were equally ineffective in inducing T cell proliferation and cytokine production (P < 0.05). A marked reduction in CD86+ marker expression was observed in DC isolated from both old and tolerized mice (75 and 50%, respectively). The results indicate that the aging process does not interfere with the establishment of oral tolerance in BALB/c mice, but reduces DC functions, probably due to the decline of the expression of the CD86 surface marker.
Collapse
Affiliation(s)
- P U Simioni
- Departamento de Microbiologia e Imunologia, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, SP, Brasil
| | | | | | | |
Collapse
|
345
|
Amaral JF, Gomes-Santos AC, Paula-Silva J, Nicoli JR, Vieira LQ, Faria AMC, Menezes JS. Antigenic dietary protein guides maturation of the host immune system promoting resistance to Leishmania major infection in C57BL/6 mice. Immunology 2009; 129:455-64. [PMID: 20002788 DOI: 10.1111/j.1365-2567.2009.03198.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The immature immune system requires constant stimulation by foreign antigens during the early stages of life to develop properly and to create efficient immune responses against later infections. We have previously shown that intake of antigenic dietary protein is critical for inducing maturation of the immune system as well as for the development of T helper type 1 (Th1) immunity. In this study, we show that administration of an amino acid (aa)-based diet during the development of the immune system subsequently resulted in inefficient control of Leishmania major infection in adult C57BL/6 mice. Compared with mice fed a control protein-containing diet, adult aa-fed mice showed a decreased interferon (IFN)-gamma response to parasite antigens and insufficient production of nitric oxide (NO), which is crucial to parasite death. However, no deviation towards Th2-specific immunity to L. major was observed. Phenotypic analysis of antigen-presenting cells (APCs) from aa-fed mice revealed deficient levels of the costimulatory molecules CD40 and CD80, and low levels of interleukin (IL)-12 produced by peritoneal macrophages, revealing an early stage of maturation of these cells. APCs isolated from aa-fed mice were unable to stimulate a Th1 response in vitro. Both phenotypic features of T cells from aa-fed mice and their ability to produce a Th1 response in the presence of mature APCs were unaffected when compared with T cells from control mice. The results presented here support the notion that regulation of Th1 immunity to infection includes environmental factors such as dietary proteins, which provide a natural source of stimulation that contributes to the process of maturation of APCs.
Collapse
Affiliation(s)
- Joana F Amaral
- Departamento de Bioquímica e Imunologia, Institutode Ciências Biológicas, Universidade Federalde Minas Gerais - UFMG, Belo Horizonte, Brazil
| | | | | | | | | | | | | |
Collapse
|
346
|
Sonier B, Patrick C, Ajjikuttira P, Scott FW. Intestinal Immune Regulation as a Potential Diet-Modifiable Feature of Gut Inflammation and Autoimmunity. Int Rev Immunol 2009; 28:414-45. [DOI: 10.3109/08830180903208329] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
347
|
Abstract
The rapid rise of allergic disorders in developed countries has been attributed to the hygiene hypothesis, implicating that increased environmental sanitation in early childhood may be associated with higher incidence of hypersensitivity. Intestinal epithelial barriers play a crucial role in the maintenance of gut homeostasis by limiting penetration of luminal bacteria and dietary allergens, yet allowing antigen sampling via the follicle-associated epithelium for generation of tolerance. However, this intricate balance is upset in allergic intestines, whereby luminal proteins with antigenic properties gain access to the subepithelial compartment and stimulate mast cell degranulation. Recent studies demonstrated that food allergens were protected from lysosomal degradation, and were transported in large quantities across the epithelium by binding to cell surface IgE/CD23 (FcepsilonRII) that prevented the antigenic protein from lysosomal degradation in enterocytes. IL-4 (a Th2-type cytokine) not only increased production of IgE from B cells, but also upregulated the expression of CD23 on intestinal epithelial cells. Further studies indicated that CD23 was responsible for the bidirectional transport of IgE across epithelium. The presence of IgE/CD23 opens a gate for intact dietary allergens to transcytose across the epithelial cells, and thus foments the mast cell-dependent anaphylactic responses. The understanding of the molecular mechanism responsible for epithelial barrier defects may be helpful in designing novel therapies to treat food allergy and other allergic diseases.
Collapse
|
348
|
Sasaki N, Yamashita T, Takeda M, Shinohara M, Nakajima K, Tawa H, Usui T, Hirata KI. Oral Anti-CD3 Antibody Treatment Induces Regulatory T Cells and Inhibits the Development of Atherosclerosis in Mice. Circulation 2009; 120:1996-2005. [DOI: 10.1161/circulationaha.109.863431] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Naoto Sasaki
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (N.S., T.Y., M.T., M.S., K.N., H.T., K.H.), and Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Kobe, Japan; and Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan (T.U.)
| | - Tomoya Yamashita
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (N.S., T.Y., M.T., M.S., K.N., H.T., K.H.), and Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Kobe, Japan; and Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan (T.U.)
| | - Masafumi Takeda
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (N.S., T.Y., M.T., M.S., K.N., H.T., K.H.), and Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Kobe, Japan; and Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan (T.U.)
| | - Masakazu Shinohara
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (N.S., T.Y., M.T., M.S., K.N., H.T., K.H.), and Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Kobe, Japan; and Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan (T.U.)
| | - Kenji Nakajima
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (N.S., T.Y., M.T., M.S., K.N., H.T., K.H.), and Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Kobe, Japan; and Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan (T.U.)
| | - Hideto Tawa
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (N.S., T.Y., M.T., M.S., K.N., H.T., K.H.), and Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Kobe, Japan; and Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan (T.U.)
| | - Takashi Usui
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (N.S., T.Y., M.T., M.S., K.N., H.T., K.H.), and Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Kobe, Japan; and Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan (T.U.)
| | - Ken-ichi Hirata
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (N.S., T.Y., M.T., M.S., K.N., H.T., K.H.), and Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Kobe, Japan; and Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan (T.U.)
| |
Collapse
|
349
|
Abstract
Autoimmune diseases represent a group of disorders in which there exists a large unmet medical need for effective treatments, but also where there exists a tremendous responsibility among physicians and drug developers to maintain adequate and acceptable patient safety. Several drugs have been approved and many others are about to be approved for the treatment of autoimmune diseases, but in pushing the envelope of therapeutic efficacy, concerns have been raised about the long-term safety of these new therapies. DNA vaccines provide a method of treating autoimmune diseases in a highly specific manner, and could therefore overcome these safety concerns while still maintaining comparable efficacy. The numerous reports of DNA vaccines in animal models of autoimmune diseases and results from three recent human trials of DNA vaccines in autoimmune diseases are reviewed here.
Collapse
Affiliation(s)
- Hideki Garren
- Bayhill Therapeutics, Inc., Suite 150, San Mateo, CA 94404, USA.
| |
Collapse
|
350
|
Crowley DC, Lau FC, Sharma P, Evans M, Guthrie N, Bagchi M, Bagchi D, Dey DK, Raychaudhuri SP. Safety and efficacy of undenatured type II collagen in the treatment of osteoarthritis of the knee: a clinical trial. Int J Med Sci 2009; 6:312-21. [PMID: 19847319 PMCID: PMC2764342 DOI: 10.7150/ijms.6.312] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Accepted: 10/08/2009] [Indexed: 12/27/2022] Open
Abstract
Previous studies have shown that undenatured type II collagen (UC-II) is effective in the treatment of rheumatoid arthritis, and preliminary human and animal trials have shown it to be effective in treating osteoarthritis (OA). The present clinical trial evaluated the safety and efficacy of UC-II as compared to a combination of glucosamine and chondroitin (G+C) in the treatment of OA of the knee. The results indicate that UC-II treatment was more efficacious resulting in a significant reduction in all assessments from the baseline at 90 days; whereas, this effect was not observed in G+C treatment group. Specifically, although both treatments reduced the Western Ontario McMaster Osteoarthritis Index (WOMAC) score, treatment with UC-II reduced the WOMAC score by 33% as compared to 14% in G+C treated group after 90 days. Similar results were obtained for visual analog scale (VAS) scores. Although both the treatments reduced the VAS score, UC-II treatment decreased VAS score by 40% after 90 days as compared to 15.4% in G+C treated group. The Lequesne's functional index was used to determine the effect of different treatments on pain during daily activities. Treatment with UC-II reduced Lequesne's functional index score by 20% as compared to 6% in G+C treated group at the end of 90-day treatment. Thus, UC-II treated subjects showed significant enhancement in daily activities suggesting an improvement in their quality of life.
Collapse
|