[HTML][HTML] Requirements for allergen-induced airway hyperreactivity in T and B cell-deficient mice

DB Corry, G Grünig, H Hadeiba, VP Kurup… - Molecular …, 1998 - Springer
DB Corry, G Grünig, H Hadeiba, VP Kurup, ML Warnock, D Sheppard, DM Rennick…
Molecular Medicine, 1998Springer
Background The pathogenesis of asthma is believed to reflect antigen-induced airway
inflammation leading to the recruitment of eosinophils and activation of mast cells through
cell-associated IgE. Controversies persist however, regarding the relative importance of
different pathogenic cells and effector molecules. Materials and Methods A variety of gene-
targeted mice were examined for the induction of cholinergic airway hyperresponsiveness
(AH), allergic airway inflammation, mucus production, and serum IgE reactivity following …
Background
The pathogenesis of asthma is believed to reflect antigen-induced airway inflammation leading to the recruitment of eosinophils and activation of mast cells through cell-associated IgE. Controversies persist however, regarding the relative importance of different pathogenic cells and effector molecules.
Materials and Methods
A variety of gene-targeted mice were examined for the induction of cholinergic airway hyperresponsiveness (AH), allergic airway inflammation, mucus production, and serum IgE reactivity following intratracheal challenge with a potent allergen. AH was determined using whole-body plethysmography following acetylcholine challenge. Where possible, results were confirmed using neutralizing antibodies and cell-specific reconstitution of immune deficient mice.
Results
T and B cell-deficient, recombinase-activating-gene-deficient mice (RAG −/−) failed to develop significant allergic inflammation and AH following allergen challenge. Reconstitution of RAG −/− mice with CD4+ T cells alone was sufficient to restore allergen-induced AH, allergic inflammation, and goblet cell hyperplasia, but not IgE reactivity. Sensitized B cell-deficient mice also developed airway hyperreactivity and lung inflammation comparable to that of wild-type animals, confirming that antibodies were dispensable. Treatment with neutralizing anti-IL-4 antibody or sensitization of IL-4-deficient mice resulted in loss of airway hyperreactivity, whereas treatment with anti-IL-5 antibody or sensitization of IL-5-deficient mice had no effect.
Conclusions
In mice, CD4+ T cells are alone sufficient to mediate many of the pathognomonic changes that occur in human asthma by a mechanism dependent upon IL-4, but independent of IL-5, IgE, or both. Clarification of the role played by CD4+ T cells is likely to stimulate important therapeutic advances in treatment of asthma.
Springer