Your browser doesn't support javascript.
loading
Adoptive transfer of allergen-expressing B cells prevents IgE-mediated allergy.
Prickler, Lisa; Baranyi, Ulrike; Mengrelis, Konstantinos; Weijler, Anna Marianne; Kainz, Verena; Kratzer, Bernhard; Steiner, Romy; Mucha, Jasmin; Rudoph, Elisa; Pilat, Nina; Bohle, Barbara; Strobl, Herbert; Pickl, Winfried Franz; Valenta, Rudolf; Linhart, Birgit; Wekerle, Thomas.
Afiliação
  • Prickler L; Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria.
  • Baranyi U; Cardiac Surgery Research Laboratory, Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria.
  • Mengrelis K; Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria.
  • Weijler AM; Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria.
  • Kainz V; Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria.
  • Kratzer B; Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
  • Steiner R; Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria.
  • Mucha J; Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria.
  • Rudoph E; Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria.
  • Pilat N; Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria.
  • Bohle B; Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
  • Strobl H; Division of Immunology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria.
  • Pickl WF; Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
  • Valenta R; Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria.
  • Linhart B; Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
  • Wekerle T; Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria.
Front Immunol ; 14: 1286638, 2023.
Article em En | MEDLINE | ID: mdl-38077381
ABSTRACT

Introduction:

Prophylactic strategies to prevent the development of allergies by establishing tolerance remain an unmet medical need. We previously reported that the transfer of autologous hematopoietic stem cells (HSC) expressing the major timothy grass pollen allergen, Phl p 5, on their cell surface induced allergen-specific tolerance in mice. In this study, we investigated the ability of allergen-expressing immune cells (dendritic cells, CD4+ T cells, CD8+ T cells, and CD19+ B cells) to induce allergen-specific tolerance in naive mice and identified CD19+ B cells as promising candidates for allergen-specific cell therapy.

Methods:

For this purpose, CD19+ B cells were isolated from Phl p 5-transgenic BALB/c mice and transferred to naive BALB/c mice, pre-treated with a short course of rapamycin and an anti-CD40L antibody. Subsequently, the mice were subcutaneously sensitized three times at 4-week intervals to Phl p 5 and Bet v 1 as an unrelated control allergen. Allergen-expressing cells were followed in the blood to monitor molecular chimerism, and sera were analyzed for Phl p 5- and Bet v 1-specific IgE and IgG1 levels by RBL assay and ELISA, respectively. In vivo allergen-induced lung inflammation was measured by whole-body plethysmography, and mast cell degranulation was determined by skin testing.

Results:

The transfer of purified Phl p 5-expressing CD19+ B cells to naive BALB/c mice induced B cell chimerism for up to three months and prevented the development of Phl p 5-specific IgE and IgG1 antibody responses for a follow-up period of 26 weeks. Since Bet v 1 but not Phl p 5-specific antibodies were detected, the induction of tolerance was specific for Phl p 5. Whole-body plethysmography revealed preserved lung function in CD19+ B cell-treated mice in contrast to sensitized mice, and there was no Phl p 5-induced mast cell degranulation in treated mice.

Discussion:

Thus, we demonstrated that the transfer of Phl p 5-expressing CD19+ B cells induces allergen-specific tolerance in a mouse model of grass pollen allergy. This approach could be further translated into a prophylactic regimen for the prevention of IgE-mediated allergy in humans.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Alérgenos / Hipersensibilidade Limite: Animals / Humans Idioma: En Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Alérgenos / Hipersensibilidade Limite: Animals / Humans Idioma: En Ano de publicação: 2023 Tipo de documento: Article