Cargando…

DSS-induced inflammation in the colon drives a proinflammatory signature in the brain that is ameliorated by prophylactic treatment with the S100A9 inhibitor paquinimod

BACKGROUND: Inflammatory bowel disease (IBD) is established to drive pathological sequelae in organ systems outside the intestine, including the central nervous system (CNS). Many patients exhibit cognitive deficits, particularly during disease flare. The connection between colonic inflammation and...

Descripción completa

Detalles Bibliográficos
Autores principales: Talley, Sarah, Valiauga, Rasa, Anderson, Lillian, Cannon, Abigail R., Choudhry, Mashkoor A., Campbell, Edward M.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8578918/
https://www.ncbi.nlm.nih.gov/pubmed/34758843
http://dx.doi.org/10.1186/s12974-021-02317-6
_version_ 1784596338197921792
author Talley, Sarah
Valiauga, Rasa
Anderson, Lillian
Cannon, Abigail R.
Choudhry, Mashkoor A.
Campbell, Edward M.
author_facet Talley, Sarah
Valiauga, Rasa
Anderson, Lillian
Cannon, Abigail R.
Choudhry, Mashkoor A.
Campbell, Edward M.
author_sort Talley, Sarah
collection PubMed
description BACKGROUND: Inflammatory bowel disease (IBD) is established to drive pathological sequelae in organ systems outside the intestine, including the central nervous system (CNS). Many patients exhibit cognitive deficits, particularly during disease flare. The connection between colonic inflammation and neuroinflammation remains unclear and characterization of the neuroinflammatory phenotype in the brain during colitis is ill-defined. METHODS: Transgenic mice expressing a bioluminescent reporter of active caspase-1 were treated with 2% dextran sodium sulfate (DSS) for 7 days to induce acute colitis, and colonic, systemic and neuroinflammation were assessed. In some experiments, mice were prophylactically treated with paquinimod (ABR-215757) to inhibit S100A9 inflammatory signaling. As a positive control for peripheral-induced neuroinflammation, mice were injected with lipopolysaccharide (LPS). Colonic, systemic and brain inflammatory cytokines and chemokines were measured by cytokine bead array (CBA) and Proteome profiler mouse cytokine array. Bioluminescence was quantified in the brain and caspase activation was confirmed by immunoblot. Immune cell infiltration into the CNS was measured by flow cytometry, while light sheet microscopy was used to monitor changes in resident microglia localization in intact brains during DSS or LPS-induced neuroinflammation. RNA sequencing was performed to identify transcriptomic changes occurring in the CNS of DSS-treated mice. Expression of inflammatory biomarkers were quantified in the brain and serum by qRT-PCR, ELISA and WB. RESULTS: DSS-treated mice exhibited clinical hallmarks of colitis, including weight loss, colonic shortening and inflammation in the colon. We also detected a significant increase in inflammatory cytokines in the serum and brain, as well as caspase and microglia activation in the brain of mice with ongoing colitis. RNA sequencing of brains isolated from DSS-treated mice revealed differential expression of genes involved in the regulation of inflammatory responses. This inflammatory phenotype was similar to the signature detected in LPS-treated mice, albeit less robust and transient, as inflammatory gene expression returned to baseline following cessation of DSS. Pharmacological inhibition of S100A9, one of the transcripts identified by RNA sequencing, attenuated colitis severity and systemic and neuroinflammation. CONCLUSIONS: Our findings suggest that local inflammation in the colon drives systemic inflammation and neuroinflammation, and this can be ameliorated by inhibition of the S100 alarmin, S100A9. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12974-021-02317-6.
format Online
Article
Text
id pubmed-8578918
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-85789182021-11-10 DSS-induced inflammation in the colon drives a proinflammatory signature in the brain that is ameliorated by prophylactic treatment with the S100A9 inhibitor paquinimod Talley, Sarah Valiauga, Rasa Anderson, Lillian Cannon, Abigail R. Choudhry, Mashkoor A. Campbell, Edward M. J Neuroinflammation Research BACKGROUND: Inflammatory bowel disease (IBD) is established to drive pathological sequelae in organ systems outside the intestine, including the central nervous system (CNS). Many patients exhibit cognitive deficits, particularly during disease flare. The connection between colonic inflammation and neuroinflammation remains unclear and characterization of the neuroinflammatory phenotype in the brain during colitis is ill-defined. METHODS: Transgenic mice expressing a bioluminescent reporter of active caspase-1 were treated with 2% dextran sodium sulfate (DSS) for 7 days to induce acute colitis, and colonic, systemic and neuroinflammation were assessed. In some experiments, mice were prophylactically treated with paquinimod (ABR-215757) to inhibit S100A9 inflammatory signaling. As a positive control for peripheral-induced neuroinflammation, mice were injected with lipopolysaccharide (LPS). Colonic, systemic and brain inflammatory cytokines and chemokines were measured by cytokine bead array (CBA) and Proteome profiler mouse cytokine array. Bioluminescence was quantified in the brain and caspase activation was confirmed by immunoblot. Immune cell infiltration into the CNS was measured by flow cytometry, while light sheet microscopy was used to monitor changes in resident microglia localization in intact brains during DSS or LPS-induced neuroinflammation. RNA sequencing was performed to identify transcriptomic changes occurring in the CNS of DSS-treated mice. Expression of inflammatory biomarkers were quantified in the brain and serum by qRT-PCR, ELISA and WB. RESULTS: DSS-treated mice exhibited clinical hallmarks of colitis, including weight loss, colonic shortening and inflammation in the colon. We also detected a significant increase in inflammatory cytokines in the serum and brain, as well as caspase and microglia activation in the brain of mice with ongoing colitis. RNA sequencing of brains isolated from DSS-treated mice revealed differential expression of genes involved in the regulation of inflammatory responses. This inflammatory phenotype was similar to the signature detected in LPS-treated mice, albeit less robust and transient, as inflammatory gene expression returned to baseline following cessation of DSS. Pharmacological inhibition of S100A9, one of the transcripts identified by RNA sequencing, attenuated colitis severity and systemic and neuroinflammation. CONCLUSIONS: Our findings suggest that local inflammation in the colon drives systemic inflammation and neuroinflammation, and this can be ameliorated by inhibition of the S100 alarmin, S100A9. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12974-021-02317-6. BioMed Central 2021-11-10 /pmc/articles/PMC8578918/ /pubmed/34758843 http://dx.doi.org/10.1186/s12974-021-02317-6 Text en © The Author(s) 2021 https://creativecommons.org/licenses/by/4.0/Open AccessThis 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
Talley, Sarah
Valiauga, Rasa
Anderson, Lillian
Cannon, Abigail R.
Choudhry, Mashkoor A.
Campbell, Edward M.
DSS-induced inflammation in the colon drives a proinflammatory signature in the brain that is ameliorated by prophylactic treatment with the S100A9 inhibitor paquinimod
title DSS-induced inflammation in the colon drives a proinflammatory signature in the brain that is ameliorated by prophylactic treatment with the S100A9 inhibitor paquinimod
title_full DSS-induced inflammation in the colon drives a proinflammatory signature in the brain that is ameliorated by prophylactic treatment with the S100A9 inhibitor paquinimod
title_fullStr DSS-induced inflammation in the colon drives a proinflammatory signature in the brain that is ameliorated by prophylactic treatment with the S100A9 inhibitor paquinimod
title_full_unstemmed DSS-induced inflammation in the colon drives a proinflammatory signature in the brain that is ameliorated by prophylactic treatment with the S100A9 inhibitor paquinimod
title_short DSS-induced inflammation in the colon drives a proinflammatory signature in the brain that is ameliorated by prophylactic treatment with the S100A9 inhibitor paquinimod
title_sort dss-induced inflammation in the colon drives a proinflammatory signature in the brain that is ameliorated by prophylactic treatment with the s100a9 inhibitor paquinimod
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8578918/
https://www.ncbi.nlm.nih.gov/pubmed/34758843
http://dx.doi.org/10.1186/s12974-021-02317-6
work_keys_str_mv AT talleysarah dssinducedinflammationinthecolondrivesaproinflammatorysignatureinthebrainthatisamelioratedbyprophylactictreatmentwiththes100a9inhibitorpaquinimod
AT valiaugarasa dssinducedinflammationinthecolondrivesaproinflammatorysignatureinthebrainthatisamelioratedbyprophylactictreatmentwiththes100a9inhibitorpaquinimod
AT andersonlillian dssinducedinflammationinthecolondrivesaproinflammatorysignatureinthebrainthatisamelioratedbyprophylactictreatmentwiththes100a9inhibitorpaquinimod
AT cannonabigailr dssinducedinflammationinthecolondrivesaproinflammatorysignatureinthebrainthatisamelioratedbyprophylactictreatmentwiththes100a9inhibitorpaquinimod
AT choudhrymashkoora dssinducedinflammationinthecolondrivesaproinflammatorysignatureinthebrainthatisamelioratedbyprophylactictreatmentwiththes100a9inhibitorpaquinimod
AT campbelledwardm dssinducedinflammationinthecolondrivesaproinflammatorysignatureinthebrainthatisamelioratedbyprophylactictreatmentwiththes100a9inhibitorpaquinimod