Lineage-Specific Analysis of Syk Function in Autoantibody-Induced Arthritis

Autoantibody production and autoantibody-mediated inflammation are hallmarks of a number of autoimmune diseases. The K/BxN serum-transfer arthritis is one of the most widely used models of the effector phase of autoantibody-induced pathology. Several hematopoietic lineages including neutrophils, pla...

Full description

Bibliographic Details
Main Authors: Tamás Németh, Krisztina Futosi, Kata Szilveszter, Olivér Vilinovszki, Levente Kiss-Pápai, Attila Mócsai
Format: Article
Language:English
Published: Frontiers Media S.A. 2018-03-01
Series:Frontiers in Immunology
Subjects:
Syk
Online Access:http://journal.frontiersin.org/article/10.3389/fimmu.2018.00555/full
id doaj-e78286af00874d5d8ec39239254839ad
record_format Article
spelling doaj-e78286af00874d5d8ec39239254839ad2020-11-24T23:27:08ZengFrontiers Media S.A.Frontiers in Immunology1664-32242018-03-01910.3389/fimmu.2018.00555351690Lineage-Specific Analysis of Syk Function in Autoantibody-Induced ArthritisTamás Németh0Tamás Németh1Krisztina Futosi2Krisztina Futosi3Kata Szilveszter4Kata Szilveszter5Olivér Vilinovszki6Levente Kiss-Pápai7Levente Kiss-Pápai8Attila Mócsai9Attila Mócsai10Department of Physiology, Semmelweis University School of Medicine, Budapest, HungaryMTA-SE “Lendület” Inflammation Physiology Research Group of the Hungarian Academy of Sciences and Semmelweis University, Budapest, HungaryDepartment of Physiology, Semmelweis University School of Medicine, Budapest, HungaryMTA-SE “Lendület” Inflammation Physiology Research Group of the Hungarian Academy of Sciences and Semmelweis University, Budapest, HungaryDepartment of Physiology, Semmelweis University School of Medicine, Budapest, HungaryMTA-SE “Lendület” Inflammation Physiology Research Group of the Hungarian Academy of Sciences and Semmelweis University, Budapest, HungaryDepartment of Physiology, Semmelweis University School of Medicine, Budapest, HungaryDepartment of Physiology, Semmelweis University School of Medicine, Budapest, HungaryMTA-SE “Lendület” Inflammation Physiology Research Group of the Hungarian Academy of Sciences and Semmelweis University, Budapest, HungaryDepartment of Physiology, Semmelweis University School of Medicine, Budapest, HungaryMTA-SE “Lendület” Inflammation Physiology Research Group of the Hungarian Academy of Sciences and Semmelweis University, Budapest, HungaryAutoantibody production and autoantibody-mediated inflammation are hallmarks of a number of autoimmune diseases. The K/BxN serum-transfer arthritis is one of the most widely used models of the effector phase of autoantibody-induced pathology. Several hematopoietic lineages including neutrophils, platelets, and mast cells have been proposed to contribute to inflammation and tissue damage in this model. We have previously shown that the Syk tyrosine kinase is critically involved in the development in K/BxN serum-transfer arthritis and bone marrow chimeric experiments indicated that Syk is likely involved in one or more hematopoietic lineages during the disease course. The aim of the present study was to further define the lineage(s) in which Syk expression is required for autoantibody-induced arthritis. To this end, K/BxN serum-transfer arthritis was tested in conditional mutant mice in which Syk was deleted in a lineage-specific manner from neutrophils, platelets, or mast cells. Combination of the MRP8-Cre, PF4-Cre, or Mcpt5-Cre transgene with floxed Syk alleles allowed efficient and selective deletion of Syk from neutrophils, platelets, or mast cells, respectively. This has also been confirmed by defective Syk-dependent in vitro functional responses of the respective cell types. In vivo studies revealed nearly complete defect of the development of K/BxN serum-transfer arthritis upon neutrophil-specific deletion of Syk. By contrast, Syk deletion from platelets or mast cells did not affect the development of K/BxN serum-transfer arthritis. Our results indicate that autoantibody-induced arthritis requires Syk expression in neutrophils, whereas, contrary to prior assumptions, Syk expression in platelets or mast cells is dispensable for disease development in this model.http://journal.frontiersin.org/article/10.3389/fimmu.2018.00555/fullSykarthritisneutrophilsplateletsmast cells
collection DOAJ
language English
format Article
sources DOAJ
author Tamás Németh
Tamás Németh
Krisztina Futosi
Krisztina Futosi
Kata Szilveszter
Kata Szilveszter
Olivér Vilinovszki
Levente Kiss-Pápai
Levente Kiss-Pápai
Attila Mócsai
Attila Mócsai
spellingShingle Tamás Németh
Tamás Németh
Krisztina Futosi
Krisztina Futosi
Kata Szilveszter
Kata Szilveszter
Olivér Vilinovszki
Levente Kiss-Pápai
Levente Kiss-Pápai
Attila Mócsai
Attila Mócsai
Lineage-Specific Analysis of Syk Function in Autoantibody-Induced Arthritis
Frontiers in Immunology
Syk
arthritis
neutrophils
platelets
mast cells
author_facet Tamás Németh
Tamás Németh
Krisztina Futosi
Krisztina Futosi
Kata Szilveszter
Kata Szilveszter
Olivér Vilinovszki
Levente Kiss-Pápai
Levente Kiss-Pápai
Attila Mócsai
Attila Mócsai
author_sort Tamás Németh
title Lineage-Specific Analysis of Syk Function in Autoantibody-Induced Arthritis
title_short Lineage-Specific Analysis of Syk Function in Autoantibody-Induced Arthritis
title_full Lineage-Specific Analysis of Syk Function in Autoantibody-Induced Arthritis
title_fullStr Lineage-Specific Analysis of Syk Function in Autoantibody-Induced Arthritis
title_full_unstemmed Lineage-Specific Analysis of Syk Function in Autoantibody-Induced Arthritis
title_sort lineage-specific analysis of syk function in autoantibody-induced arthritis
publisher Frontiers Media S.A.
series Frontiers in Immunology
issn 1664-3224
publishDate 2018-03-01
description Autoantibody production and autoantibody-mediated inflammation are hallmarks of a number of autoimmune diseases. The K/BxN serum-transfer arthritis is one of the most widely used models of the effector phase of autoantibody-induced pathology. Several hematopoietic lineages including neutrophils, platelets, and mast cells have been proposed to contribute to inflammation and tissue damage in this model. We have previously shown that the Syk tyrosine kinase is critically involved in the development in K/BxN serum-transfer arthritis and bone marrow chimeric experiments indicated that Syk is likely involved in one or more hematopoietic lineages during the disease course. The aim of the present study was to further define the lineage(s) in which Syk expression is required for autoantibody-induced arthritis. To this end, K/BxN serum-transfer arthritis was tested in conditional mutant mice in which Syk was deleted in a lineage-specific manner from neutrophils, platelets, or mast cells. Combination of the MRP8-Cre, PF4-Cre, or Mcpt5-Cre transgene with floxed Syk alleles allowed efficient and selective deletion of Syk from neutrophils, platelets, or mast cells, respectively. This has also been confirmed by defective Syk-dependent in vitro functional responses of the respective cell types. In vivo studies revealed nearly complete defect of the development of K/BxN serum-transfer arthritis upon neutrophil-specific deletion of Syk. By contrast, Syk deletion from platelets or mast cells did not affect the development of K/BxN serum-transfer arthritis. Our results indicate that autoantibody-induced arthritis requires Syk expression in neutrophils, whereas, contrary to prior assumptions, Syk expression in platelets or mast cells is dispensable for disease development in this model.
topic Syk
arthritis
neutrophils
platelets
mast cells
url http://journal.frontiersin.org/article/10.3389/fimmu.2018.00555/full
work_keys_str_mv AT tamasnemeth lineagespecificanalysisofsykfunctioninautoantibodyinducedarthritis
AT tamasnemeth lineagespecificanalysisofsykfunctioninautoantibodyinducedarthritis
AT krisztinafutosi lineagespecificanalysisofsykfunctioninautoantibodyinducedarthritis
AT krisztinafutosi lineagespecificanalysisofsykfunctioninautoantibodyinducedarthritis
AT kataszilveszter lineagespecificanalysisofsykfunctioninautoantibodyinducedarthritis
AT kataszilveszter lineagespecificanalysisofsykfunctioninautoantibodyinducedarthritis
AT olivervilinovszki lineagespecificanalysisofsykfunctioninautoantibodyinducedarthritis
AT leventekisspapai lineagespecificanalysisofsykfunctioninautoantibodyinducedarthritis
AT leventekisspapai lineagespecificanalysisofsykfunctioninautoantibodyinducedarthritis
AT attilamocsai lineagespecificanalysisofsykfunctioninautoantibodyinducedarthritis
AT attilamocsai lineagespecificanalysisofsykfunctioninautoantibodyinducedarthritis
_version_ 1725553303511629824