RhoA Drives T-Cell Activation and Encephalitogenic Potential in an Animal Model of Multiple Sclerosis

T-cells are known to be intimately involved in the pathogenesis of multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). T-cell activation is controlled by a range of intracellular signaling pathways regulating cellular responses such as proliferation, cytokin...

Full description

Bibliographic Details
Main Authors: Alba Manresa-Arraut, Flemming Fryd Johansen, Cord Brakebusch, Shohreh Issazadeh-Navikas, Henrik Hasseldam
Format: Article
Language:English
Published: Frontiers Media S.A. 2018-05-01
Series:Frontiers in Immunology
Subjects:
Online Access:https://www.frontiersin.org/article/10.3389/fimmu.2018.01235/full
id doaj-026acbef7ed748a7bfaf311f9ba1ee89
record_format Article
spelling doaj-026acbef7ed748a7bfaf311f9ba1ee892020-11-25T02:24:45ZengFrontiers Media S.A.Frontiers in Immunology1664-32242018-05-01910.3389/fimmu.2018.01235333447RhoA Drives T-Cell Activation and Encephalitogenic Potential in an Animal Model of Multiple SclerosisAlba Manresa-Arraut0Flemming Fryd Johansen1Cord Brakebusch2Shohreh Issazadeh-Navikas3Henrik Hasseldam4Neuroinflammation Unit, Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DenmarkNeuroinflammation Unit, Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DenmarkCytoskeletal Organization Group, Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DenmarkNeuroinflammation Unit, Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DenmarkNeuroinflammation Unit, Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DenmarkT-cells are known to be intimately involved in the pathogenesis of multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). T-cell activation is controlled by a range of intracellular signaling pathways regulating cellular responses such as proliferation, cytokine production, integrin expression, and migration. These processes are crucial for the T-cells’ ability to mediate inflammatory processes in autoimmune diseases such as MS. RhoA is a ubiquitously expressed small GTPase well described as a regulator of the actin cytoskeleton. It is essential for embryonic development and together with other Rho GTPases controls various cellular processes such as cell development, shaping, proliferation, and locomotion. However, the specific contribution of RhoA to these processes in T-cells in general, and in autoreactive T-cells in particular, has not been fully characterized. Using mice with a T-cell specific deletion of the RhoA gene (RhoAfl/flLckCre+), we investigated the role of RhoA in T-cell development, functionality, and encephalitogenic potential in EAE. We show that lack of RhoA specifically in T-cells results in reduced numbers of mature T-cells in thymus and spleen but normal counts in peripheral blood. EAE induction in RhoAfl/flLckCre+ mice results in significantly reduced disease incidence and severity, which coincides with a reduced CNS T-cell infiltration. Besides presenting reduced migratory capacity, both naïve and autoreactive effector T-cells from RhoAfl/flLckCre+ mice show decreased viability, proliferative capacity, and an activation profile associated with reduced production of Th1 pro-inflammatory cytokines. Our study demonstrates that RhoA is a central regulator of several archetypical T-cell responses, and furthermore points toward RhoA as a new potential therapeutic target in diseases such as MS, where T-cell activity plays a central role.https://www.frontiersin.org/article/10.3389/fimmu.2018.01235/fullRhoAmultiple sclerosisexperimental autoimmune encephalomyelitisT-cellneuroinflammation
collection DOAJ
language English
format Article
sources DOAJ
author Alba Manresa-Arraut
Flemming Fryd Johansen
Cord Brakebusch
Shohreh Issazadeh-Navikas
Henrik Hasseldam
spellingShingle Alba Manresa-Arraut
Flemming Fryd Johansen
Cord Brakebusch
Shohreh Issazadeh-Navikas
Henrik Hasseldam
RhoA Drives T-Cell Activation and Encephalitogenic Potential in an Animal Model of Multiple Sclerosis
Frontiers in Immunology
RhoA
multiple sclerosis
experimental autoimmune encephalomyelitis
T-cell
neuroinflammation
author_facet Alba Manresa-Arraut
Flemming Fryd Johansen
Cord Brakebusch
Shohreh Issazadeh-Navikas
Henrik Hasseldam
author_sort Alba Manresa-Arraut
title RhoA Drives T-Cell Activation and Encephalitogenic Potential in an Animal Model of Multiple Sclerosis
title_short RhoA Drives T-Cell Activation and Encephalitogenic Potential in an Animal Model of Multiple Sclerosis
title_full RhoA Drives T-Cell Activation and Encephalitogenic Potential in an Animal Model of Multiple Sclerosis
title_fullStr RhoA Drives T-Cell Activation and Encephalitogenic Potential in an Animal Model of Multiple Sclerosis
title_full_unstemmed RhoA Drives T-Cell Activation and Encephalitogenic Potential in an Animal Model of Multiple Sclerosis
title_sort rhoa drives t-cell activation and encephalitogenic potential in an animal model of multiple sclerosis
publisher Frontiers Media S.A.
series Frontiers in Immunology
issn 1664-3224
publishDate 2018-05-01
description T-cells are known to be intimately involved in the pathogenesis of multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). T-cell activation is controlled by a range of intracellular signaling pathways regulating cellular responses such as proliferation, cytokine production, integrin expression, and migration. These processes are crucial for the T-cells’ ability to mediate inflammatory processes in autoimmune diseases such as MS. RhoA is a ubiquitously expressed small GTPase well described as a regulator of the actin cytoskeleton. It is essential for embryonic development and together with other Rho GTPases controls various cellular processes such as cell development, shaping, proliferation, and locomotion. However, the specific contribution of RhoA to these processes in T-cells in general, and in autoreactive T-cells in particular, has not been fully characterized. Using mice with a T-cell specific deletion of the RhoA gene (RhoAfl/flLckCre+), we investigated the role of RhoA in T-cell development, functionality, and encephalitogenic potential in EAE. We show that lack of RhoA specifically in T-cells results in reduced numbers of mature T-cells in thymus and spleen but normal counts in peripheral blood. EAE induction in RhoAfl/flLckCre+ mice results in significantly reduced disease incidence and severity, which coincides with a reduced CNS T-cell infiltration. Besides presenting reduced migratory capacity, both naïve and autoreactive effector T-cells from RhoAfl/flLckCre+ mice show decreased viability, proliferative capacity, and an activation profile associated with reduced production of Th1 pro-inflammatory cytokines. Our study demonstrates that RhoA is a central regulator of several archetypical T-cell responses, and furthermore points toward RhoA as a new potential therapeutic target in diseases such as MS, where T-cell activity plays a central role.
topic RhoA
multiple sclerosis
experimental autoimmune encephalomyelitis
T-cell
neuroinflammation
url https://www.frontiersin.org/article/10.3389/fimmu.2018.01235/full
work_keys_str_mv AT albamanresaarraut rhoadrivestcellactivationandencephalitogenicpotentialinananimalmodelofmultiplesclerosis
AT flemmingfrydjohansen rhoadrivestcellactivationandencephalitogenicpotentialinananimalmodelofmultiplesclerosis
AT cordbrakebusch rhoadrivestcellactivationandencephalitogenicpotentialinananimalmodelofmultiplesclerosis
AT shohrehissazadehnavikas rhoadrivestcellactivationandencephalitogenicpotentialinananimalmodelofmultiplesclerosis
AT henrikhasseldam rhoadrivestcellactivationandencephalitogenicpotentialinananimalmodelofmultiplesclerosis
_version_ 1724853515798446080