Elsevier

Clinical Immunology

Volume 230, September 2021, 108815
Clinical Immunology

Full Length Article
Long-term cognitive deficits after traumatic brain injury associated with microglia activation

https://doi.org/10.1016/j.clim.2021.108815Get rights and content

Highlights

  • Traumatic brain injury induces long-term cognitive deficits in mice.

  • Injury induces the activation of microglia and CNS infiltration by macrophages.

  • After injury, microglia change phenotypes and the frequency of activated microglia increases with the injury intensity.

  • Activated microglia found in injured brains correlate positively with memory deficit 35 days post injury.

Abstract

Traumatic Brain Injury (TBI) is the most prevalent of all head injuries. Microglia play an essential role in homeostasis and diseases of the central nervous system. We hypothesize that microglia may play a beneficial or detrimental role in TBI depending on their state of activation and duration.

In this study, we evaluated whether TBI results in a spatiotemporal change in microglia phenotype and whether it affects sensory-motor or learning and memory functions in male C57BL/6 mice. We used a panel of neurological and behavioral tests and a multi-color flow cytometry-based data analysis followed by unsupervised clustering to evaluate isolated microglia from injured brain tissue. We characterized several microglial phenotypes and their association with cognitive deficits. TBI results in a spatiotemporal increase in activated microglia that correlated negatively with spatial learning and memory at 35 days post-injury. These observations could define therapeutic windows and accelerate translational research to improve patient outcomes.

Introduction

Microglia are the central nervous system (CNS) resident innate immune cells that play critical physiological roles in the healthy and injured brain. They detect and rapidly respond to any disruption in the status quo of the CNS, including infections or tissue injury, and often act to remove cellular debris [1]. The activation of microglia from their resting surveillance state occurs within minutes of the injury and is critical for recovery. However, prolonged activation may be detrimental and contribute to secondary damage. When the microglia are activated in response to any disruption, this activation alters their gene expression and morphology [2]. Although they have distinctive lineage, microglia resemble blood-derived macrophages that infiltrate the CNS from the periphery in response to tissue damage [3]. Microglia are usually classified into the classical M1 phenotype that is considered proinflammatory, and the M2 anti-inflammatory is thought to be involved in neural repair [4]. However, more recent data suggest that microglia's activation status is on a continuum between anti- and pro-inflammation rather than a rigid dichotomous phenotype [5].

Chronic neuroinflammatory response to an acquired brain insult such as a TBI contributes to the injury and lengthens or halts recovery [6]. In chronic neuroinflammation, microglia remain activated for an extended period during which the production of repair mediators is sustained longer than usual [7]. Of note, chronic microglial activation has also been linked to most, if not all, neurodegenerative diseases like Alzheimer's disease, amyotrophic lateral sclerosis, and Parkinson's disease [[8], [9], [10]]. In humans with traumatic brain injury (TBI), microglial activation has been reported as early as 72 h after injury [11], and it can persist for months after injury [12].

TBI is the most prevalent of all CNS injuries and results in chronic neurologic and cognitive deficits [13]. TBI causes cell death and neurologic dysfunction through secondary injury mechanisms characterized by edema, neuronal cell death, glial activation, and infiltration of peripheral immune cells [14]. Extensive research has been conducted investigating the role of microglia after head injury and its interaction with the neural microenvironment suggesting a role for microglia in the injury process as well as in neurotransmission and maintenance of synaptic integrity [15]. TBI initiates a neuroinflammatory cascade that persists over time and may lead to prolonged cognitive deficits. The regional distribution of microglial activation is yet to be determined. Some studies found chronic activation in the thalamus, others in the hippocampus in addition to the injury site [16]. Despite evidence showing a significant role for microglia in the pathogenesis of TBI, no studies to date have examined the spatiotemporal microglial activation response after injury and explored its potential correlation with cognitive deficits [[17], [18], [19]].

Here we hypothesized that TBI would result in phenotypic changes in the microglial cell population that persists for a long time after the injury. We also hypothesized that chronic microglial activation is associated with cognitive deficits. We analyzed the spatiotemporal course of microglia changes isolated from the injured brain up to 35 days after controlled cortical impact (CCI) in mice. We used conventional flow cytometry techniques followed by bioinformatics-based multi-parametric methods that are not constrained by assumptions or bias. We observed heterogeneity in microglia phenotypes and temporal changes in microglia subpopulations following TBI. A particular subpopulation that we called hyperactivated microglia was correlated with chronic deficits in learning and spatial memory after injury.

Section snippets

Animals

The Institutional Animal Care and Utilization Committee (IACUC) of the American University of Beirut (AUB) approved this study. The study is reported in accordance with ARRIVE guidelines [64]. C57BL/6 mice were obtained from the Animal Care Facility of the American University of Beirut and housed in a controlled environment (12 h reverse light/dark cycles, 22 ± 2 °C). All efforts were made to reduce the number of animals used and their suffering. All animals were handled and fed regular chow

Traumatic brain injury affects motor and sensory performance

We evaluated the performance of mice on the pole climbing test. At 48 h, mice with mild and severe TBI performed significantly worse than sham-injured mice as indicated by the total time taken by the animals to descend the pole (mild:11.7 ± 0.6 and severe: 21.4 ± 3.6 vs. 6.4 ± 0.8 s, P < 0.001 and P < 0.0001 respectively). Less time is indicative of better motor coordination and balance. Both TBI groups showed recovered motor performance at 7- and 35-days post-injury (dpi) in comparison to

Discussion

In the present study, we investigated whether CCI traumatic brain injury results in a spatiotemporal phenotypic change in microglia and if these changes are linked to a neurologic and cognitive deficit in a mouse model. Microglia are the brain immune cells, and they are implicated in almost all physiological processes in the CNS and play an important role in several inflammatory and neurodegenerative diseases. Chronic changes in microglia phenotype and function, support the notion that chronic

Author contribution

Conceptualization: SJK, HD, FK

Data curation, Formal analysis, Investigation, Methodology: ESS, MK, LN, HD, SJK

Writing - original draft, Writing - review & editing.: ESS, HD, SJK

Funding acquisition: SJK, HD

Funding information

This study was funded by the Office of Naval Research (ONR), [ONRG - NICOP - N62909-17-1 2059] AWARD. The funders had no role in study design, data collection, analysis, decision to publish, or manuscript preparation.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References (73)

  • T.C. Thomas et al.

    Does time heal all wounds? Experimental diffuse traumatic brain injury results in persisting histopathology in the thalamus

    Behav. Brain Res.

    (2018)
  • J.M. Ziebell et al.

    Involvement of pro- and anti-inflammatory cytokines and chemokines in the pathophysiology of traumatic brain injury

    Neurotherapeutics

    (2010)
  • S.T. Fujimoto et al.

    Motor and cognitive function evaluation following experimental traumatic brain injury

    Neurosci. Biobehav. Rev.

    (2004)
  • L. Martínez-Drudis et al.

    Delayed voluntary physical exercise restores “when” and “where” object recognition memory after traumatic brain injury

    Behav. Brain Res.

    (2021)
  • K. Matsuura et al.

    Pole test is a useful method for evaluating the mouse movement disorder caused by striatal dopamine depletion

    J. Neurosci. Methods

    (1997)
  • E.K. Hebda-Bauer et al.

    Forebrain glucocorticoid receptor overexpression increases environmental reactivity and produces a stress-induced spatial discrimination deficit

    Neuroscience

    (2010)
  • S.A. Wolf et al.

    Microglia in physiology and disease

    Annu. Rev. Physiol.

    (2017)
  • D. Nayak et al.

    Microglia development and function

    Annu. Rev. Immunol.

    (2014)
  • Y. Tang et al.

    Differential roles of M1 and M2 microglia in neurodegenerative diseases

    Mol. Neurobiol.

    (2016)
  • T.R. Hammond et al.

    Single-Cell RNA sequencing of microglia throughout the mouse lifespan and in the injured brain reveals complex cell-state changes

    Immunity

    (2019)
  • M.R. Nichols et al.

    Inflammatory mechanisms in neurodegeneration

    J. Neurochem.

    (2019)
  • V.H. Perry et al.

    Microglia in neurodegenerative disease

    Nat. Rev. Neurol.

    (2010)
  • D.V. Hansen et al.

    Microglia in Alzheimer’s disease

    J. Cell Biol.

    (2018)
  • M.C. Geloso et al.

    The dual role of microglia in ALS: mechanisms and therapeutic approaches

    Front. Aging Neurosci.

    (2017)
  • M.S. Ho

    Microglia in Parkinson’s disease

    Adv. Exp. Med. Biol.

    (2019)
  • R. Beschorner et al.

    Differential regulation of the monocytic calcium-binding peptides macrophage-inhibiting factor related protein-8 (MRP8/S100A8) and allograft inflammatory factor-1 (AIF-1) following human traumatic brain injury

    Acta Neuropathol.

    (2000)
  • G.G. Whiteneck et al.

    Prevalence of self-reported lifetime history of traumatic brain injury and associated disability: a statewide population-based survey

    J. Head Trauma Rehabil.

    (2016)
  • J.W. Skovira et al.

    Cell cycle inhibition reduces inflammatory responses, neuronal loss, and cognitive deficits induced by hypobaria exposure following traumatic brain injury

    J. Neuroinflammation

    (2016)
  • D.G. Hernandez-Ontiveros et al.

    Microglia activation as a biomarker for traumatic brain injury

    Front Neurol.

    (2013)
  • A.I. Faden

    Microglial activation and traumatic brain injury

    Ann. Neurol.

    (2011)
  • J.B. Redell et al.

    Analysis of functional pathways altered after mild traumatic brain injury

    J. Neurotrauma

    (2013)
  • Y. Wolf et al.

    Microglial MHC class II is dispensable for experimental autoimmune encephalomyelitis and cuprizone-induced demyelination

    Eur. J. Immunol.

    (2018)
  • C.V. Vorhees et al.

    Assessing spatial learning and memory in rodents

    ILAR J.

    (2014)
  • E. Martin et al.

    Analysis of microglia and monocyte-derived macrophages from the central nervous system by flow cytometry

    J. Vis. Exp. JoVE

    (2017)
  • Q. Li et al.

    Expression of Tmem119/Sall1 and Ccr2/CD69 in FACS-sorted microglia- and monocyte/macrophage-enriched cell populations after intracerebral hemorrhage

    Front. Cell. Neurosci.

    (2019)
  • G.B. Fox et al.

    Sustained sensory/motor and cognitive deficits with neuronal apoptosis following controlled cortical impact brain injury in the mouse

    J. Neurotrauma

    (1998)
  • Cited by (11)

    View all citing articles on Scopus
    1

    Esber S Saba and Mona Karout contributed equally to this work as first authors.

    View full text