11
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Neurons and glial cells acquire a senescent signature after repeated mild traumatic brain injury in a sex-dependent manner

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Mild traumatic brain injury (mTBI) is an important public health issue, as it can lead to long-term neurological symptoms and risk of neurodegenerative disease. The pathophysiological mechanisms driving this remain unclear, and currently there are no effective therapies for mTBI. In this study on repeated mTBI (rmTBI), we have induced three mild closed-skull injuries or sham procedures, separated by 24 h, in C57BL/6 mice. We show that rmTBI mice have prolonged righting reflexes and astrogliosis, with neurological impairment in the Morris water maze (MWM) and the light dark test. Cortical and hippocampal tissue analysis revealed DNA damage in the form of double-strand breaks, oxidative damage, and R-loops, markers of cellular senescence including p16 and p21, and signaling mediated by the cGAS-STING pathway. This study identified novel sex differences after rmTBI in mice. Although these markers were all increased by rmTBI in both sexes, females had higher levels of DNA damage, lower levels of the senescence protein p16, and lower levels of cGAS-STING signaling proteins compared to their male counterparts. Single-cell RNA sequencing of the male rmTBI mouse brain revealed activation of the DNA damage response, evidence of cellular senescence, and pro-inflammatory markers reminiscent of the senescence-associated secretory phenotype (SASP) in neurons and glial cells. Cell-type specific changes were also present with evidence of brain immune activation, neurotransmission alterations in both excitatory and inhibitory neurons, and vascular dysfunction. Treatment of injured mice with the senolytic drug ABT263 significantly reduced markers of senescence only in males, but was not therapeutic in females. The reduction of senescence by ABT263 in male mice was accompanied by significantly improved performance in the MWM. This study provides compelling evidence that senescence contributes to brain dysfunction after rmTBI, but may do so in a sex-dependent manner.

          Related collections

          Most cited references95

          • Record: found
          • Abstract: found
          • Article: not found

          Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles

          Although genomewide RNA expression analysis has become a routine tool in biomedical research, extracting biological insight from such information remains a major challenge. Here, we describe a powerful analytical method called Gene Set Enrichment Analysis (GSEA) for interpreting gene expression data. The method derives its power by focusing on gene sets, that is, groups of genes that share common biological function, chromosomal location, or regulation. We demonstrate how GSEA yields insights into several cancer-related data sets, including leukemia and lung cancer. Notably, where single-gene analysis finds little similarity between two independent studies of patient survival in lung cancer, GSEA reveals many biological pathways in common. The GSEA method is embodied in a freely available software package, together with an initial database of 1,325 biologically defined gene sets.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Integrated analysis of multimodal single-cell data

            Summary The simultaneous measurement of multiple modalities represents an exciting frontier for single-cell genomics and necessitates computational methods that can define cellular states based on multimodal data. Here, we introduce “weighted-nearest neighbor” analysis, an unsupervised framework to learn the relative utility of each data type in each cell, enabling an integrative analysis of multiple modalities. We apply our procedure to a CITE-seq dataset of 211,000 human peripheral blood mononuclear cells (PBMCs) with panels extending to 228 antibodies to construct a multimodal reference atlas of the circulating immune system. Multimodal analysis substantially improves our ability to resolve cell states, allowing us to identify and validate previously unreported lymphoid subpopulations. Moreover, we demonstrate how to leverage this reference to rapidly map new datasets and to interpret immune responses to vaccination and coronavirus disease 2019 (COVID-19). Our approach represents a broadly applicable strategy to analyze single-cell multimodal datasets and to look beyond the transcriptome toward a unified and multimodal definition of cellular identity.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              The senescence-associated secretory phenotype: the dark side of tumor suppression.

              Cellular senescence is a tumor-suppressive mechanism that permanently arrests cells at risk for malignant transformation. However, accumulating evidence shows that senescent cells can have deleterious effects on the tissue microenvironment. The most significant of these effects is the acquisition of a senescence-associated secretory phenotype (SASP) that turns senescent fibroblasts into proinflammatory cells that have the ability to promote tumor progression.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Neurosci
                Front Neurosci
                Front. Neurosci.
                Frontiers in Neuroscience
                Frontiers Media S.A.
                1662-4548
                1662-453X
                03 November 2022
                2022
                : 16
                : 1027116
                Affiliations
                [1] 1Department of Laboratory Medicine and Pathobiology, University of Toronto , Toronto, ON, Canada
                [2] 2Program in Neurosciences and Mental Health, The Hospital for Sick Children , Toronto, ON, Canada
                [3] 3The Donnelly Centre, University of Toronto , Toronto, ON, Canada
                [4] 4Department of Molecular Genetics, University of Toronto , Toronto, ON, Canada
                Author notes

                Edited by: Allison B. Reiss, New York University, United States

                Reviewed by: Rachel Knopp, University of Washington, United States; Gemma Llovera, LMU Munich University Hospital, Germany; Laura B. Tucker, Uniformed Services University of the Health Sciences, United States; Wai Hang Cheng, University of British Columbia, Canada

                *Correspondence: Lili-Naz Hazrati, lili-naz.hazrati@ 123456sickkids.ca

                This article was submitted to Translational Neuroscience, a section of the journal Frontiers in Neuroscience

                Article
                10.3389/fnins.2022.1027116
                9669743
                36408415
                352bd6b0-7708-4752-ba3d-3dc1c7da28af
                Copyright © 2022 Schwab, Taskina, Leung, Innes, Bader and Hazrati.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 24 August 2022
                : 23 September 2022
                Page count
                Figures: 8, Tables: 1, Equations: 0, References: 95, Pages: 22, Words: 15537
                Funding
                Funded by: Canadian Institutes of Health Research, doi 10.13039/501100000024;
                Award ID: PJT166031
                Categories
                Neuroscience
                Original Research

                Neurosciences
                mild traumatic brain injury,cellular senescence,senolytic,concussion,sex difference
                Neurosciences
                mild traumatic brain injury, cellular senescence, senolytic, concussion, sex difference

                Comments

                Comment on this article