A New Murine Model of Primary Autoimmune Hemolytic Anemia (AIHA)

Loss of humoral tolerance to red blood cells (RBCs) can lead to autoimmune hemolytic anemia (AIHA), a severe, and sometimes fatal disease. Patients with AIHA present with pallor, fatigue, decreased hematocrit, and splenomegaly. While secondary AIHA is associated with lymphoproliferative disorders, i...

Descripción completa

Guardado en:
Detalles Bibliográficos
Autores principales: Flavia Dei Zotti, Annie Qiu, Francesca La Carpia, Chiara Moriconi, Krystalyn E. Hudson
Formato: article
Lenguaje:EN
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://doaj.org/article/f4eb8fe2dcc7496e83c3f3ff8010c4d7
Etiquetas: Agregar Etiqueta
Sin Etiquetas, Sea el primero en etiquetar este registro!
id oai:doaj.org-article:f4eb8fe2dcc7496e83c3f3ff8010c4d7
record_format dspace
spelling oai:doaj.org-article:f4eb8fe2dcc7496e83c3f3ff8010c4d72021-11-15T06:08:41ZA New Murine Model of Primary Autoimmune Hemolytic Anemia (AIHA)1664-322410.3389/fimmu.2021.752330https://doaj.org/article/f4eb8fe2dcc7496e83c3f3ff8010c4d72021-11-01T00:00:00Zhttps://www.frontiersin.org/articles/10.3389/fimmu.2021.752330/fullhttps://doaj.org/toc/1664-3224Loss of humoral tolerance to red blood cells (RBCs) can lead to autoimmune hemolytic anemia (AIHA), a severe, and sometimes fatal disease. Patients with AIHA present with pallor, fatigue, decreased hematocrit, and splenomegaly. While secondary AIHA is associated with lymphoproliferative disorders, infections, and more recently, as an adverse event secondary to cancer immunotherapy, the etiology of primary AIHA is unknown. Several therapeutic strategies are available; however, there are currently no licensed treatments for AIHA and few therapeutics offer treatment-free durable remission. Moreover, supportive care with RBC transfusions can be challenging as most autoantibodies are directed against ubiquitous RBC antigens; thus, virtually all RBC donor units are incompatible. Given the severity of AIHA and the lack of treatment options, understanding the cellular and molecular mechanisms that facilitate the breakdown in tolerance would provide insight into new therapeutics. Herein, we report a new murine model of primary AIHA that reflects the biology observed in patients with primary AIHA. Production of anti-erythrocyte autoantibodies correlated with sex and age, and led to RBC antigen modulation, complement fixation, and anemia, as determined by decreased hematocrit and hemoglobin values and increased reticulocytes in peripheral blood. Moreover, autoantibody-producing animals developed splenomegaly, with altered splenic architecture characterized by expanded white pulp areas and nearly diminished red pulp areas. Additional analysis suggested that compensatory extramedullary erythropoiesis occurred as there were increased frequencies of RBC progenitors detectable in the spleen. No significant correlations between AIHA onset and inflammatory status or microbiome were observed. To our knowledge, this is the first report of a murine model that replicates observations made in humans with idiopathic AIHA. Thus, this is a tractable murine model of AIHA that can serve as a platform to identify key cellular and molecular pathways that are compromised, thereby leading to autoantibody formation, as well as testing new therapeutics and management strategies.Flavia Dei ZottiAnnie QiuFrancesca La CarpiaChiara MoriconiKrystalyn E. HudsonFrontiers Media S.A.articlered blood cellerythrocyteautoimmune hemolytic anemiatoleranceautoimmune diseaseprimary autoimmune hemolytic anemiaImmunologic diseases. AllergyRC581-607ENFrontiers in Immunology, Vol 12 (2021)
institution DOAJ
collection DOAJ
language EN
topic red blood cell
erythrocyte
autoimmune hemolytic anemia
tolerance
autoimmune disease
primary autoimmune hemolytic anemia
Immunologic diseases. Allergy
RC581-607
spellingShingle red blood cell
erythrocyte
autoimmune hemolytic anemia
tolerance
autoimmune disease
primary autoimmune hemolytic anemia
Immunologic diseases. Allergy
RC581-607
Flavia Dei Zotti
Annie Qiu
Francesca La Carpia
Chiara Moriconi
Krystalyn E. Hudson
A New Murine Model of Primary Autoimmune Hemolytic Anemia (AIHA)
description Loss of humoral tolerance to red blood cells (RBCs) can lead to autoimmune hemolytic anemia (AIHA), a severe, and sometimes fatal disease. Patients with AIHA present with pallor, fatigue, decreased hematocrit, and splenomegaly. While secondary AIHA is associated with lymphoproliferative disorders, infections, and more recently, as an adverse event secondary to cancer immunotherapy, the etiology of primary AIHA is unknown. Several therapeutic strategies are available; however, there are currently no licensed treatments for AIHA and few therapeutics offer treatment-free durable remission. Moreover, supportive care with RBC transfusions can be challenging as most autoantibodies are directed against ubiquitous RBC antigens; thus, virtually all RBC donor units are incompatible. Given the severity of AIHA and the lack of treatment options, understanding the cellular and molecular mechanisms that facilitate the breakdown in tolerance would provide insight into new therapeutics. Herein, we report a new murine model of primary AIHA that reflects the biology observed in patients with primary AIHA. Production of anti-erythrocyte autoantibodies correlated with sex and age, and led to RBC antigen modulation, complement fixation, and anemia, as determined by decreased hematocrit and hemoglobin values and increased reticulocytes in peripheral blood. Moreover, autoantibody-producing animals developed splenomegaly, with altered splenic architecture characterized by expanded white pulp areas and nearly diminished red pulp areas. Additional analysis suggested that compensatory extramedullary erythropoiesis occurred as there were increased frequencies of RBC progenitors detectable in the spleen. No significant correlations between AIHA onset and inflammatory status or microbiome were observed. To our knowledge, this is the first report of a murine model that replicates observations made in humans with idiopathic AIHA. Thus, this is a tractable murine model of AIHA that can serve as a platform to identify key cellular and molecular pathways that are compromised, thereby leading to autoantibody formation, as well as testing new therapeutics and management strategies.
format article
author Flavia Dei Zotti
Annie Qiu
Francesca La Carpia
Chiara Moriconi
Krystalyn E. Hudson
author_facet Flavia Dei Zotti
Annie Qiu
Francesca La Carpia
Chiara Moriconi
Krystalyn E. Hudson
author_sort Flavia Dei Zotti
title A New Murine Model of Primary Autoimmune Hemolytic Anemia (AIHA)
title_short A New Murine Model of Primary Autoimmune Hemolytic Anemia (AIHA)
title_full A New Murine Model of Primary Autoimmune Hemolytic Anemia (AIHA)
title_fullStr A New Murine Model of Primary Autoimmune Hemolytic Anemia (AIHA)
title_full_unstemmed A New Murine Model of Primary Autoimmune Hemolytic Anemia (AIHA)
title_sort new murine model of primary autoimmune hemolytic anemia (aiha)
publisher Frontiers Media S.A.
publishDate 2021
url https://doaj.org/article/f4eb8fe2dcc7496e83c3f3ff8010c4d7
work_keys_str_mv AT flaviadeizotti anewmurinemodelofprimaryautoimmunehemolyticanemiaaiha
AT annieqiu anewmurinemodelofprimaryautoimmunehemolyticanemiaaiha
AT francescalacarpia anewmurinemodelofprimaryautoimmunehemolyticanemiaaiha
AT chiaramoriconi anewmurinemodelofprimaryautoimmunehemolyticanemiaaiha
AT krystalynehudson anewmurinemodelofprimaryautoimmunehemolyticanemiaaiha
AT flaviadeizotti newmurinemodelofprimaryautoimmunehemolyticanemiaaiha
AT annieqiu newmurinemodelofprimaryautoimmunehemolyticanemiaaiha
AT francescalacarpia newmurinemodelofprimaryautoimmunehemolyticanemiaaiha
AT chiaramoriconi newmurinemodelofprimaryautoimmunehemolyticanemiaaiha
AT krystalynehudson newmurinemodelofprimaryautoimmunehemolyticanemiaaiha
_version_ 1718428593330061312