Your browser doesn't support javascript.
loading
Murine LRBA deficiency causes CTLA-4 deficiency in Tregs without progression to immune dysregulation.
Burnett, Deborah L; Parish, Ian A; Masle-Farquhar, Etienne; Brink, Robert; Goodnow, Christopher C.
Afiliação
  • Burnett DL; Immunology Division, Garvan Institute for Medical Research, Sydney, NSW, Australia.
  • Parish IA; John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia.
  • Masle-Farquhar E; Immunology Division, Garvan Institute for Medical Research, Sydney, NSW, Australia.
  • Brink R; Immunology Division, Garvan Institute for Medical Research, Sydney, NSW, Australia.
  • Goodnow CC; Immunology Division, Garvan Institute for Medical Research, Sydney, NSW, Australia.
Immunol Cell Biol ; 95(9): 775-788, 2017 10.
Article em En | MEDLINE | ID: mdl-28611475
ABSTRACT
Inherited mutations in lipopolysaccharide-responsive beige-like anchor (LRBA) cause a recessive human immune dysregulation syndrome with memory B-cell and antibody deficiency (common variable immunodeficiency), inflammatory bowel disease, enlarged spleen and lymph nodes, accumulation of activated T cells and multiple autoimmune diseases. To understand the pathogenesis of the syndrome, C57BL/6 mice carrying a homozygous truncating mutation in Lrba were produced using CRISPR/Cas9-mediated gene targeting. These mice revealed that LRBA has a critical, cell-autonomous role in promoting cytotoxic T-lymphocyte antigen-4 (CTLA-4) accumulation within CD4 effector T cells and FOXP3+ T-regulatory cells (Tregs). In young mice, or in chimeric mice where only half of the T cells are LRBA deficient, low CTLA-4 was the only detectable abnormality in Tregs, whereas in old mice FOXP3 was also decreased. Low CTLA-4 did not translate into increased CD86 on B cells unless the LRBA-deficient mice were immunised, and neither immunisation nor chronic lymphocytic choriomeningitis virus infection precipitated immune dysregulation. LRBA deficiency did not alter antigen-specific B-cell activation, germinal centre (GC) formation, isotype switching or affinity maturation. Paradoxically, CD86 was decreased on GC B cells in LRBA-deficient mice, pointing to compensatory mechanisms for controlling CD86 in the face of low CTLA-4. These results add to the experimental rationale for treating LRBA deficiency with the CTLA4-Ig fusion protein, Abatacept, and pose questions about the limitations of laboratory experiments in mice to reproduce human disease in natura.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Imunodeficiência de Variável Comum / Linfócitos T Reguladores / Linfócitos T Auxiliares-Indutores / Proteínas Adaptadoras de Transdução de Sinal / Antígeno CTLA-4 / Coriomeningite Linfocítica / Vírus da Coriomeningite Linfocítica Tipo de estudo: Etiology_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2017 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Imunodeficiência de Variável Comum / Linfócitos T Reguladores / Linfócitos T Auxiliares-Indutores / Proteínas Adaptadoras de Transdução de Sinal / Antígeno CTLA-4 / Coriomeningite Linfocítica / Vírus da Coriomeningite Linfocítica Tipo de estudo: Etiology_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2017 Tipo de documento: Article