Your browser doesn't support javascript.
loading
Adaptive Immunity and Pathogenesis of Diabetes: Insights Provided by the α4-Integrin Deficient NOD Mouse.
Oulghazi, Salim; Wegner, Sarah K; Spohn, Gabriele; Müller, Nina; Harenkamp, Sabine; Stenzinger, Albrecht; Papayannopoulou, Thalia; Bonig, Halvard.
  • Oulghazi S; Institute for Transfusion Medicine and Immunohematology, School of Medicine, Goethe University, Sandhofstraße 1, 60528 Frankfurt, Germany.
  • Wegner SK; Institute for Transfusion Medicine and Immunohematology, School of Medicine, Goethe University, Sandhofstraße 1, 60528 Frankfurt, Germany.
  • Spohn G; Institute Frankfurt, German Red Cross Blood Service BaWüHe, Sandhofstraße 1, 60528 Frankfurt, Germany.
  • Müller N; Institute Frankfurt, German Red Cross Blood Service BaWüHe, Sandhofstraße 1, 60528 Frankfurt, Germany.
  • Harenkamp S; Institute Frankfurt, German Red Cross Blood Service BaWüHe, Sandhofstraße 1, 60528 Frankfurt, Germany.
  • Stenzinger A; Institute for Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 672, 69120 Heidelberg, Germany.
  • Papayannopoulou T; Department of Medicine/Division of Hematology, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA.
  • Bonig H; Institute for Transfusion Medicine and Immunohematology, School of Medicine, Goethe University, Sandhofstraße 1, 60528 Frankfurt, Germany.
Cells ; 9(12)2020 12 04.
Article en En | MEDLINE | ID: mdl-33291571
ABSTRACT

BACKGROUND:

The spontaneously diabetic "non-obese diabetic" (NOD) mouse is a faithful model of human type-1 diabetes (T1D).

METHODS:

Given the pivotal role of α4 integrin (CD49d) in other autoimmune diseases, we generated NOD mice with α4-deficient hematopoiesis (NOD.α4-/-) to study the role of α4 integrin in T1D.

RESULTS:

NOD.α4-/- mice developed islet-specific T-cells and antibodies, albeit quantitatively less than α4+ counterparts. Nevertheless, NOD.α4-/- mice were completely and life-long protected from diabetes and insulitis. Moreover, transplantation with isogeneic α4-/- bone marrow prevented progression to T1D of pre-diabetic NOD.α4+ mice despite significant pre-existing islet cell injury. Transfer of α4+/CD3+, but not α4+/CD4+ splenocytes from diabetic to NOD.α4-/- mice induced diabetes with short latency. Despite an only modest contribution of adoptively transferred α4+/CD3+ cells to peripheral blood, pancreas-infiltrating T-cells were exclusively graft derived, i.e., α4+. Microbiota of diabetes-resistant NOD.α4-/- and pre-diabetic NOD.α4+ mice were identical. Co- housed diabetic NOD.α4+ mice showed the characteristic diabetic dysbiosis, implying causality of diabetes for dysbiosis. Incidentally, NOD.α4-/- mice were protected from autoimmune sialitis.

CONCLUSION:

α4 is a potential target for primary or secondary prevention of T1D.
Asunto(s)
Palabras clave

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Integrina alfa4 / Inmunidad Adaptativa Tipo de estudio: Etiology_studies / Prognostic_studies Límite: Animals Idioma: En Año: 2020 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Integrina alfa4 / Inmunidad Adaptativa Tipo de estudio: Etiology_studies / Prognostic_studies Límite: Animals Idioma: En Año: 2020 Tipo del documento: Article