Cargando…

Deletion of voltage-gated calcium channels in astrocytes decreases neuroinflammation and demyelination in a murine model of multiple sclerosis

The experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis was used in combination with a Cav1.2 conditional knock-out mouse (Cav1.2(KO)) to study the role of astrocytic voltage-gated Ca(++) channels in autoimmune CNS inflammation and demyelination. Cav1.2 channels were specific...

Descripción completa

Detalles Bibliográficos
Autores principales: Denaroso, G. E., Smith, Z., Angeliu, C. G., Cheli, V. T., Wang, C., Paez, P. M.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10644533/
https://www.ncbi.nlm.nih.gov/pubmed/37964385
http://dx.doi.org/10.1186/s12974-023-02948-x
_version_ 1785147248209821696
author Denaroso, G. E.
Smith, Z.
Angeliu, C. G.
Cheli, V. T.
Wang, C.
Paez, P. M.
author_facet Denaroso, G. E.
Smith, Z.
Angeliu, C. G.
Cheli, V. T.
Wang, C.
Paez, P. M.
author_sort Denaroso, G. E.
collection PubMed
description The experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis was used in combination with a Cav1.2 conditional knock-out mouse (Cav1.2(KO)) to study the role of astrocytic voltage-gated Ca(++) channels in autoimmune CNS inflammation and demyelination. Cav1.2 channels were specifically ablated in Glast-1-positive astrocytes by means of the Cre-lox system before EAE induction. After immunization, motor activity was assessed daily, and a clinical score was given based on the severity of EAE symptoms. Cav1.2 deletion in astrocytes significantly reduced the severity of the disease. While no changes were found in the day of onset and peak disease severity, EAE mean clinical score was lower in Cav1.2(KO) animals during the chronic phase of the disease. This corresponded to better performance on the rotarod and increased motor activity in Cav1.2(KO) mice. Furthermore, decreased numbers of reactive astrocytes, activated microglia, and infiltrating lymphocytes were found in the lumbar section of the spinal cord of Cav1.2(KO) mice 40 days after immunization. The degree of myelin protein loss and size of demyelinated lesions were also attenuated in Cav1.2(KO) spinal cords. Similar results were found in EAE animals treated with nimodipine, a Cav1.2 Ca(++) channel inhibitor with high affinity to the CNS. Mice injected with nimodipine during the acute and chronic phases of the disease exhibited lower numbers of reactive astrocytes, activated microglial, and infiltrating immune cells, as well as fewer demyelinated lesions in the spinal cord. These changes were correlated with improved clinical scores and motor performance. In summary, these data suggest that antagonizing Cav1.2 channels in astrocytes during EAE alleviates neuroinflammation and protects the spinal cord from autoimmune demyelination.
format Online
Article
Text
id pubmed-10644533
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-106445332023-11-14 Deletion of voltage-gated calcium channels in astrocytes decreases neuroinflammation and demyelination in a murine model of multiple sclerosis Denaroso, G. E. Smith, Z. Angeliu, C. G. Cheli, V. T. Wang, C. Paez, P. M. J Neuroinflammation Research The experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis was used in combination with a Cav1.2 conditional knock-out mouse (Cav1.2(KO)) to study the role of astrocytic voltage-gated Ca(++) channels in autoimmune CNS inflammation and demyelination. Cav1.2 channels were specifically ablated in Glast-1-positive astrocytes by means of the Cre-lox system before EAE induction. After immunization, motor activity was assessed daily, and a clinical score was given based on the severity of EAE symptoms. Cav1.2 deletion in astrocytes significantly reduced the severity of the disease. While no changes were found in the day of onset and peak disease severity, EAE mean clinical score was lower in Cav1.2(KO) animals during the chronic phase of the disease. This corresponded to better performance on the rotarod and increased motor activity in Cav1.2(KO) mice. Furthermore, decreased numbers of reactive astrocytes, activated microglia, and infiltrating lymphocytes were found in the lumbar section of the spinal cord of Cav1.2(KO) mice 40 days after immunization. The degree of myelin protein loss and size of demyelinated lesions were also attenuated in Cav1.2(KO) spinal cords. Similar results were found in EAE animals treated with nimodipine, a Cav1.2 Ca(++) channel inhibitor with high affinity to the CNS. Mice injected with nimodipine during the acute and chronic phases of the disease exhibited lower numbers of reactive astrocytes, activated microglial, and infiltrating immune cells, as well as fewer demyelinated lesions in the spinal cord. These changes were correlated with improved clinical scores and motor performance. In summary, these data suggest that antagonizing Cav1.2 channels in astrocytes during EAE alleviates neuroinflammation and protects the spinal cord from autoimmune demyelination. BioMed Central 2023-11-14 /pmc/articles/PMC10644533/ /pubmed/37964385 http://dx.doi.org/10.1186/s12974-023-02948-x Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Denaroso, G. E.
Smith, Z.
Angeliu, C. G.
Cheli, V. T.
Wang, C.
Paez, P. M.
Deletion of voltage-gated calcium channels in astrocytes decreases neuroinflammation and demyelination in a murine model of multiple sclerosis
title Deletion of voltage-gated calcium channels in astrocytes decreases neuroinflammation and demyelination in a murine model of multiple sclerosis
title_full Deletion of voltage-gated calcium channels in astrocytes decreases neuroinflammation and demyelination in a murine model of multiple sclerosis
title_fullStr Deletion of voltage-gated calcium channels in astrocytes decreases neuroinflammation and demyelination in a murine model of multiple sclerosis
title_full_unstemmed Deletion of voltage-gated calcium channels in astrocytes decreases neuroinflammation and demyelination in a murine model of multiple sclerosis
title_short Deletion of voltage-gated calcium channels in astrocytes decreases neuroinflammation and demyelination in a murine model of multiple sclerosis
title_sort deletion of voltage-gated calcium channels in astrocytes decreases neuroinflammation and demyelination in a murine model of multiple sclerosis
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10644533/
https://www.ncbi.nlm.nih.gov/pubmed/37964385
http://dx.doi.org/10.1186/s12974-023-02948-x
work_keys_str_mv AT denarosoge deletionofvoltagegatedcalciumchannelsinastrocytesdecreasesneuroinflammationanddemyelinationinamurinemodelofmultiplesclerosis
AT smithz deletionofvoltagegatedcalciumchannelsinastrocytesdecreasesneuroinflammationanddemyelinationinamurinemodelofmultiplesclerosis
AT angeliucg deletionofvoltagegatedcalciumchannelsinastrocytesdecreasesneuroinflammationanddemyelinationinamurinemodelofmultiplesclerosis
AT chelivt deletionofvoltagegatedcalciumchannelsinastrocytesdecreasesneuroinflammationanddemyelinationinamurinemodelofmultiplesclerosis
AT wangc deletionofvoltagegatedcalciumchannelsinastrocytesdecreasesneuroinflammationanddemyelinationinamurinemodelofmultiplesclerosis
AT paezpm deletionofvoltagegatedcalciumchannelsinastrocytesdecreasesneuroinflammationanddemyelinationinamurinemodelofmultiplesclerosis