scholarly journals Development of the Th2 Immune Response Models to Evaluate Allergenicity of Milk Proteins

2021 ◽  
Author(s):  
◽  
Marcus James Robinson

<p>Food allergy, defined as an adverse immune response to food, is increasing in prevalence. It can be broadly separated into phases of sensitization, in which allergy-triggering Immunoglobulin E (IgE) is generated, and the post-sensitization allergic response, in which the allergic response is triggered by sensitizing allergen. While much is known about the specific mediators that cause allergies, the immune processes that underlie disease progression are less clear. This project has employed mouse models of Th2 immunity to clarify the factors involved in the initiation and maintenance of allergic disease.  At the centre of allergic disease is the Interleukin (IL)-4-producing CD4+ T helper type 2 (Th2) cell. One of the key inducers of Th2 cell development in vitro is IL-4, but its involvement in Th2 cell development in vivo is controversial. In our studies, we saw that Th2 cell development could be initiated in vivo by primary, adjuvant-free allergen immunisation in the absence of IL-4. However, Th2 cells were more frequent in IL-4-sufficient conditions. We also determined that genetic lesions that result in loss of one, or both, IL-4 alleles impaired the Th2 cell-mediated allergic process, such that IL-4-heterozygous mice can be considered haplo-insufficient for IL-4 in allergic disease contexts.  In addition to the generation of IgE antibody, Th2 cells are implicated in the post-sensitization phase of allergy. Multiple oral challenges of sensitized mice induces elevations in Th2-associated cytokines and elevates intestinal mast cell frequencies. It was the second aim of this project to clarify the role of CD4+ T cells in the post-sensitization intestinal allergic process. We demonstrate a key role for CD4+ T cells in this jejunal mast cell recruitment, and identify that this is required in addition to their established contribution to IgE production. Our investigations also reveal a previously unappreciated role for the CD4+ T cell-derived cytokine IL-3 in oral food allergy. These findings suggest that intestinally localised mast cell-inducer Th2 (Th2m) cells are required for allergic responses generated in the intestine. We also investigated whether specific components of ruminant milks influence the allergic process. While goat and cow milks share significant protein homology, goat milk has lower sensitizing and response-evoking capacity, or allergenicity, than cow milk, in numerous experimental systems. In this project, we compared dominant allergens purified from cow and goat milks for their ability to initiate Th2 cell development. We also examined the ability of one of these allergens to initiate the intestinal allergic process. In these studies, we observed similar Th2 cell development and intestinal mast cell activity in response to both cow and goat milk proteins. These responses indicate that the intrinsic allergenicity of the proteins analysed is not sufficient to explain the differential allergenicity attributed to cow and goat milk.  These studies examine the endogenous and exogenous factors that contribute to the development of allergic disease. This project clarifies the role of IL-4 in in vivo Th2 cell development, identifies functional segregation of CD4+ Th2 cells in the intestinal allergic process and further illustrates some of the similarities in the allergenicity of isolated cow and goat milk proteins. Collectively, these studies uncover fundamental aspects of the allergic process which may be useful targets for disease intervention in both prophylactic and therapeutic settings.</p>

2021 ◽  
Author(s):  
◽  
Marcus James Robinson

<p>Food allergy, defined as an adverse immune response to food, is increasing in prevalence. It can be broadly separated into phases of sensitization, in which allergy-triggering Immunoglobulin E (IgE) is generated, and the post-sensitization allergic response, in which the allergic response is triggered by sensitizing allergen. While much is known about the specific mediators that cause allergies, the immune processes that underlie disease progression are less clear. This project has employed mouse models of Th2 immunity to clarify the factors involved in the initiation and maintenance of allergic disease.  At the centre of allergic disease is the Interleukin (IL)-4-producing CD4+ T helper type 2 (Th2) cell. One of the key inducers of Th2 cell development in vitro is IL-4, but its involvement in Th2 cell development in vivo is controversial. In our studies, we saw that Th2 cell development could be initiated in vivo by primary, adjuvant-free allergen immunisation in the absence of IL-4. However, Th2 cells were more frequent in IL-4-sufficient conditions. We also determined that genetic lesions that result in loss of one, or both, IL-4 alleles impaired the Th2 cell-mediated allergic process, such that IL-4-heterozygous mice can be considered haplo-insufficient for IL-4 in allergic disease contexts.  In addition to the generation of IgE antibody, Th2 cells are implicated in the post-sensitization phase of allergy. Multiple oral challenges of sensitized mice induces elevations in Th2-associated cytokines and elevates intestinal mast cell frequencies. It was the second aim of this project to clarify the role of CD4+ T cells in the post-sensitization intestinal allergic process. We demonstrate a key role for CD4+ T cells in this jejunal mast cell recruitment, and identify that this is required in addition to their established contribution to IgE production. Our investigations also reveal a previously unappreciated role for the CD4+ T cell-derived cytokine IL-3 in oral food allergy. These findings suggest that intestinally localised mast cell-inducer Th2 (Th2m) cells are required for allergic responses generated in the intestine. We also investigated whether specific components of ruminant milks influence the allergic process. While goat and cow milks share significant protein homology, goat milk has lower sensitizing and response-evoking capacity, or allergenicity, than cow milk, in numerous experimental systems. In this project, we compared dominant allergens purified from cow and goat milks for their ability to initiate Th2 cell development. We also examined the ability of one of these allergens to initiate the intestinal allergic process. In these studies, we observed similar Th2 cell development and intestinal mast cell activity in response to both cow and goat milk proteins. These responses indicate that the intrinsic allergenicity of the proteins analysed is not sufficient to explain the differential allergenicity attributed to cow and goat milk.  These studies examine the endogenous and exogenous factors that contribute to the development of allergic disease. This project clarifies the role of IL-4 in in vivo Th2 cell development, identifies functional segregation of CD4+ Th2 cells in the intestinal allergic process and further illustrates some of the similarities in the allergenicity of isolated cow and goat milk proteins. Collectively, these studies uncover fundamental aspects of the allergic process which may be useful targets for disease intervention in both prophylactic and therapeutic settings.</p>


1999 ◽  
Vol 190 (5) ◽  
pp. 617-628 ◽  
Author(s):  
Takashi Nishimura ◽  
Kenji Iwakabe ◽  
Masashi Sekimoto ◽  
Yasushi Ohmi ◽  
Takashi Yahata ◽  
...  

The role of T helper type 1 (Th1) and Th2 cells in tumor immunity was investigated using Th cells induced from ovalbumin (OVA)-specific T cell receptor transgenic mice. Although Th1 cells exhibited stronger cytotoxicity than Th2 cells, both cell types completely eradicated tumors when transferred into mice bearing A20 tumor cells transfected with the OVA gene (A20-OVA). Th1 cells eradicated the tumor mass by inducing cellular immunity, whereas Th2 cells destroyed the tumor by inducing tumor necrosis. Both Th1 and Th2 cells required CD8+ T cells to eliminate tumors, and neither of these cells were able to completely eliminate A20-OVA tumors from T and B cell–deficient RAG2−/− mice. Mice cured from tumors by Th1 and Th2 cell therapy rejected A20-OVA upon rechallenge, but CD8+ cytotoxic T lymphocytes were induced only from spleen cells prepared from cured mice by Th1 cell therapy. Moreover, we demonstrated that Th1 and Th2 cells used distinct adhesion mechanisms during tumor eradication: the leukocyte function-associated antigen (LFA)-1–dependent cell–cell adhesion step was essential for Th1 cell therapy, but not for Th2 cell therapy. These findings demonstrated for the first time the distinct role of antigen-specific Th1 and Th2 cells during eradication of established tumors in vivo.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 793-793
Author(s):  
Yuji Nakata ◽  
Alan M. Gewirtz

Abstract c-Myb is an obligate hematopoietic transcription factor which is highly expressed in immature hematopoietic cells. It plays a critical role in both myeloid and lymphoid cell development, and specifically in regard to this communication, at multiple points during early T cell development. While the role of c-Myb in developing cells has been intensively studied, we noted that there is a relative paucity of investigations focused on c-myb function in peripheral blood T cells. This situation exists despite the relatively high level of c-myb expression we observe in unstimulated cells, and the increase that occurs when such cells are stimulated. Very recently (Embo J, Aug 2007), Maurice et al demonstrated that c-myb regulates T helper cell lineage commitment in developing mouse thymocytes, at the same time that it appears to block development of cytotoxic T cells, via regulation of GATA-3. However, the role of c-Myb and GATA-3 in normal human peripheral blood lymphocytes was not explored. Here we show that c-myb regulates GATA-3 expression directly in peripheral blood CD4+ cells and has a critical role in human Th2 cell development. To explore the role of c-myb expression in human peripheral blood naive CD4+ cells we employed c-Myb targeted, and control, short hairpinRNA (shRNA) expressed from a lentivirus vector. This strategy yielded a sequence specific ~ 80–90% knockdown of c-Myb expression. Stimulation of naive peripheral blood CD4+ T cells in which the c-Myb directed shRNA was expressed, with a cocktail designed to promote Th2 cell formation (IL-4, IL-2, and anti-IL-12 antibody) blocked the up-regulation of GATA-3 mRNA expression ~90% compared to cells in which a control shRNA had been expressed. Flow cytometric analysis showed that intracellular interleukin-4 expression was also diminished in CD4+ cells stimulated under Th2 promoting conditions. In contrast, silencing c-myb did not affect T-beta mRNA expression, or intracellular interferon-γ expression in the cells induced to undergo Th1 cell formation with IL-12, IL-2 and anti-IL-4 antibody. A ChIP assay showed that c-myb bound to the GATA-3 promoter in human primary CD4+ cells stimulated under Th2 cell promoting conditions, but not under Th1 promoting conditions. A reporter assay demonstrated that c-myb over-expression increased GATA-3 promoter activity by ~5 fold in 293T cells, and approximately 3 fold in human primary T cells. Silencing c-myb in primary human T cells with shRNA resulted in an approximately 50% decrease in GATA-3 promoter activity. These results demonstrate that c-myb plays an important role in Th2 cell development at least in part through direct regulation of GATA-3 expression. In primary human effector/memory CD4+ T cells, which includes established Th2 cells, c-myb suppression with shRNA also decreased GATA-3 promoter activity by approximately 85%, but the suppression of IL-4 expression was only moderate (~50%). These results suggest that c-myb may also play a role in the homeostasis of established Th2 cells. Finally, and as might be expected, silencing c-myb suppressed proliferation of naive CD4+ cells. We conclude that c-Myb plays multiple roles in human peripheral blood T lymphocytes, including the generation and maintainence of Th2 cells, in addition to regulation of cell proliferation. It performs these functions, at least in part, through direct regulation of GATA-3.


2021 ◽  
Vol 22 (7) ◽  
pp. 3687
Author(s):  
Joanna Homa ◽  
Alina Klosowska ◽  
Magdalena Chadzinska

Arginase is the manganese metalloenzyme catalyzing the conversion of l-arginine to l-ornithine and urea. In vertebrates, arginase is involved in the immune response, tissue regeneration, and wound healing and is an important marker of alternative anti-inflammatory polarization of macrophages. In invertebrates, data concerning the role of arginase in these processes are very limited. Therefore, in the present study, we focused on the changes in arginase activity in the coelomocytes of Eisenia andrei. We studied the effects of lipopolysaccharide (LPS), hydrogen peroxide (H2O2), heavy metals ions (e.g., Mn2+), parasite infection, wound healing, and short-term fasting (5 days) on arginase activity. For the first time in earthworms, we described arginase activity in the coelomocytes and found that it can be up-regulated upon in vitro stimulation with LPS and H2O2 and in the presence of Mn2+ ions. Moreover, arginase activity was also up-regulated in animals in vivo infected with nematodes or experiencing segment amputation, but not in fasting earthworms. Furthermore, we confirmed that the activity of coelomocyte arginase can be suppressed by l-norvaline. Our studies strongly suggest that similarly to the vertebrates, also in the earthworms, coelomocyte arginase is an important element of the immune response and wound healing processes.


PEDIATRICS ◽  
1995 ◽  
Vol 96 (2) ◽  
pp. 391-391
Author(s):  
Leon S. Greos

Alveolar macrophages are infected by RSV in vivo and coexpress potent immunomodulatory molecules that potentially regulate local immune response or lung injury caused by RSV infection.


2016 ◽  
Vol 113 (5) ◽  
pp. E568-E576 ◽  
Author(s):  
Jimena Perez-Lloret ◽  
Isobel S. Okoye ◽  
Riccardo Guidi ◽  
Yashaswini Kannan ◽  
Stephanie M. Coomes ◽  
...  

There is a paucity of new therapeutic targets to control allergic reactions and forestall the rising trend of allergic diseases. Although a variety of immune cells contribute to allergy, cytokine-secreting αβ+CD4+ T-helper 2 (TH2) cells orchestrate the type-2–driven immune response in a large proportion of atopic asthmatics. To identify previously unidentified putative targets in pathogenic TH2 cells, we performed in silico analyses of recently published transcriptional data from a wide variety of pathogenic TH cells [Okoye IS, et al. (2014) Proc Natl Acad Sci USA 111(30):E3081–E3090] and identified that transcription intermediary factor 1 regulator-alpha (Tif1α)/tripartite motif-containing 24 (Trim24) was predicted to be active in house dust mite (HDM)- and helminth-elicited Il4gfp+αβ+CD4+ TH2 cells but not in TH1, TH17, or Treg cells. Testing this prediction, we restricted Trim24 deficiency to T cells by using a mixed bone marrow chimera system and found that T-cell–intrinsic Trim24 is essential for HDM-mediated airway allergy and antihelminth immunity. Mechanistically, HDM-elicited Trim24−/− T cells have reduced expression of many TH2 cytokines and chemokines and were predicted to have compromised IL-1–regulated signaling. Following this prediction, we found that Trim24−/− T cells have reduced IL-1 receptor (IL-1R) expression, are refractory to IL-1β–mediated activation in vitro and in vivo, and fail to respond to IL-1β–exacerbated airway allergy. Collectively, these data identify a previously unappreciated Trim24-dependent requirement for IL-1R expression on TH2 cells and an important nonredundant role for T-cell–intrinsic Trim24 in TH2-mediated allergy and antihelminth immunity.


1996 ◽  
Vol 271 (5) ◽  
pp. R1344-R1352 ◽  
Author(s):  
M. Fleshner ◽  
F. X. Brennan ◽  
K. Nguyen ◽  
L. R. Watkins ◽  
S. F. Maier

Exposure to stressors can affect various aspects of immune function, including the antibody response. We have previously reported that rats exposed to an acute session of inescapable tail shock (IS) show long-term reductions in anti-keyhole limpet hemocyanin (KLH) immunoglobulin (Ig) M and IgG and a failure to expand Th1-like cells in response to KLH. To further investigate the potential role of decreased Th1-like cells in the IS-induced reduction of anti-KLH Ig, we examined two isotypes of IgG, IgG1 and IgG2a. Isotype switching is under cytokine control. Interleukin-4 helps B cells switch from making IgM to making IgG1, whereas interferon (IFN)-gamma helps B cells switch from making IgM to making IgG2a. In this paper we report that IS exposure reduces IFN-gamma levels 4 days after exposure to IS+KLH compared with immunized home cage controls. In addition, IS exposure reduced the Th1 cytokine-sensitive anti-KLH IgG2a but not Th2 cytokine-sensitive anti-KLH IgG1. This pattern of isotype reduction suggests that a failure to expand the Th1 cell, which results in less IFN-gamma, may contribute to the the IS-induced reduction in anti-KLH Ig. Glucocorticoids (GCs) differentially regulate Th1 and Th2 cells. Administration of the type II GC receptor antagonist RU-486 before IS blocked the IS-induced suppression in anti-KLH IgM, IgG, and IgG2a. Corticosterone (2.5 mg/kg), however, did not produce the suppression in anti-KLH Ig. These results support a role of corticosterone in mediating IS-induced reductions in in vivo antibody.


Sign in / Sign up

Export Citation Format

Share Document