Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Front Physiol ; 14: 1215535, 2023.
Article in English | MEDLINE | ID: mdl-37440997

ABSTRACT

Introduction: The response of the brain to space radiation is an important concern for astronauts during space missions. Therefore, we assessed the response of the brain to 28Si ion irradiation (600 MeV/n), a heavy ion present in the space environment, on cognitive performance and whether the response is associated with altered DNA methylation in the hippocampus, a brain area important for cognitive performance. Methods: We determined the effects of 28Si ion irradiation on object recognition, 6-month-old mice irradiated with 28Si ions (600 MeV/n, 0.3, 0.6, and 0.9 Gy) and cognitively tested two weeks later. In addition, we determined if those effects were associated with alterations in hippocampal networks and/or hippocampal DNA methylation. Results: At 0.3 Gy, but not at 0.6 Gy or 0.9 Gy, 28Si ion irradiation impaired cognition that correlated with altered gene expression and 5 hmC profiles that mapped to specific gene ontology pathways. Comparing hippocampal DNA hydroxymethylation following proton, 56Fe ion, and 28Si ion irradiation revealed a general space radiation synaptic signature with 45 genes that are associated with profound phenotypes. The most significant categories were glutamatergic synapse and postsynaptic density. Discussion: The brain's response to space irradiation involves novel excitatory synapse and postsynaptic remodeling.

2.
Eur J Neurosci ; 50(3): 2134-2145, 2019 08.
Article in English | MEDLINE | ID: mdl-30118561

ABSTRACT

Traumatic brain injury (TBI) has been linked to the development of numerous psychiatric diseases, including substance use disorder. However, it can be difficult to ascertain from clinical data whether the TBI is cause or consequence of increased addiction vulnerability. Surprisingly few studies have taken advantage of animal models to investigate the causal nature of this relationship. In terms of a plausible neurobiological mechanism through which TBI could magnify the risk of substance dependence, numerous studies indicate that TBI can cause widespread disruption to monoaminergic signaling in striatal regions, and also increases neuroinflammation. In the current study, male Long-Evans rats received either a mild or severe TBI centered over the frontal cortex via controlled cortical impact, and were subsequently trained to self-administer cocaine over 10 6-hour sessions. At the end of the study, markers of striatal dopaminergic function, and levels of inflammatory cytokine levels in the frontal lobes, were assessed via western blot and multiplex ELISA, respectively. There was significantly higher cocaine intake in a subset of animals with either mild or severe TBI. However, many animals within both TBI groups failed to acquire self-administration. Principal components analysis suggested that both dopaminergic and neuroinflammatory proteins were associated with overall cocaine intake, yet only an inflammatory component was associated with acquisition of self-administration, suggesting neuroinflammation may make a more substantial contribution to the likelihood of drug-taking. Should neuroinflammation play a causal role in mediating TBI-induced addiction risk, anti-inflammatory therapy may reduce the likelihood of substance abuse in TBI populations.


Subject(s)
Brain Injuries, Traumatic/physiopathology , Cocaine/pharmacology , Frontal Lobe/physiopathology , Inflammation/complications , Animals , Brain Injuries, Traumatic/complications , Cocaine/administration & dosage , Disease Models, Animal , Frontal Lobe/metabolism , Male , Rats, Long-Evans , Rats, Sprague-Dawley , Self Administration
3.
Int J Mol Sci ; 19(12)2018 Nov 26.
Article in English | MEDLINE | ID: mdl-30486287

ABSTRACT

Traumatic brain injury (TBI) is of particular concern for the aging community since there is both increased incidence of TBI and decreased functional recovery in this population. In addition, TBI is the strongest environmental risk factor for development of Alzheimer's disease and other dementia-related neurodegenerative disorders. Critical changes that affect cognition take place over time following the initial insult. Our previous work identified immune system activation as a key contributor to cognitive deficits observed in aged animals. Using a focal contusion model in the current study, we demonstrate a brain lesion and cavitation formation, as well as prolonged blood⁻brain barrier breakdown. These changes were associated with a prolonged inflammatory response, characterized by increased microglial cell number and phagocytic activity 30 days post injury, corresponding to significant memory deficits. We next aimed to identify the injury-induced cellular and molecular changes that lead to chronic cognitive deficits in aged animals, and measured increases in complement initiation components C1q, C3, and CR3, which are known to regulate microglial⁻synapse interactions. Specifically, we found significant accumulation of C1q on synapses within the hippocampus, which was paralleled by synapse loss 30 days post injury. We used genetic and pharmacological approaches to determine the mechanistic role of complement initiation on cognitive loss in aging animals after TBI. Notably, both genetic and pharmacological blockade of the complement pathway prevented memory deficits in aged injured animals. Thus, therapeutically targeting early components of the complement cascade represents a significant avenue for possible clinical intervention following TBI in the aging population.


Subject(s)
Aging/pathology , Brain Injuries, Traumatic/complications , Complement System Proteins/metabolism , Memory Disorders/etiology , Microglia/pathology , Synapses/pathology , Animals , Blood-Brain Barrier/pathology , Brain/pathology , Brain Injuries, Traumatic/pathology , Cell Count , Chronic Disease , Contusions , Disease Progression , Female , Magnetic Resonance Imaging , Male , Memory Disorders/pathology , Mice, Inbred C57BL , Microglia/metabolism , Models, Biological , Phagocytosis , Synapses/metabolism
4.
Sci Rep ; 8(1): 10297, 2018 Jul 03.
Article in English | MEDLINE | ID: mdl-29967490

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

5.
Int J Mol Sci ; 19(6)2018 May 30.
Article in English | MEDLINE | ID: mdl-29848996

ABSTRACT

Traumatic brain injury (TBI) is a leading cause for neurological disabilities world-wide. TBI occurs most frequently among the elderly population, and elderly TBI survivors suffer from reduced recovery and poorer quality of life. The effect of age on the pathophysiology of TBI is still poorly understood. We previously established that peripherally-derived monocytes (CCR2⁺) infiltrate the injured brain and contribute to chronic TBI-induced cognitive deficits in young animals. Furthermore, age was shown to amplify monocyte infiltration acutely after injury. In the current study, we investigated the impact of age on the subchronic response of peripherally-derived monocytes (CD45hi; CCR2⁺) and their role in the development of chronic cognitive deficits. In the aged brain, there was a significant increase in the number of peripherally-derived monocytes after injury compared to young, injured animals. The infiltration rate of peripherally-derived monocytes remained elevated subchronically and corresponded with enhanced expression of CCR2 chemotactic ligands. Interestingly, the myeloid cell populations observed in injured aged brains had impaired anti-inflammatory responses compared to those in young animals. Additionally, in the aged animals, there was an expansion of the blood CCR2⁺ monocyte population after injury that was not present in the young animals. Importantly, knocking out CCR2 to inhibit infiltration of peripherally-derived monocytes prevented chronic TBI-induced spatial memory deficits in the aged mice. Altogether, these results demonstrate the critical effects of age on the peripherally-derived monocyte response during the progression of TBI pathophysiology.


Subject(s)
Aging/physiology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/physiopathology , Brain/metabolism , Brain/physiology , Monocytes/physiology , Animals , Cognition/physiology , Disease Models, Animal , Flow Cytometry , Inflammation/immunology , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Receptors, CCR2/genetics , Receptors, CCR2/metabolism
6.
Sci Rep ; 8(1): 7857, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29777152

ABSTRACT

Microglia are the main immune component in the brain that can regulate neuronal health and synapse function. Exposure to cosmic radiation can cause long-term cognitive impairments in rodent models thereby presenting potential obstacles for astronauts engaged in deep space travel. The mechanism/s for how cosmic radiation induces cognitive deficits are currently unknown. We find that temporary microglia depletion, one week after cosmic radiation, prevents the development of long-term memory deficits. Gene array profiling reveals that acute microglia depletion alters the late neuroinflammatory response to cosmic radiation. The repopulated microglia present a modified functional phenotype with reduced expression of scavenger receptors, lysosome membrane protein and complement receptor, all shown to be involved in microglia-synapses interaction. The lower phagocytic activity observed in the repopulated microglia is paralleled by improved synaptic protein expression. Our data provide mechanistic evidence for the role of microglia in the development of cognitive deficits after cosmic radiation exposure.


Subject(s)
Cognitive Dysfunction/prevention & control , Cosmic Radiation/adverse effects , Microglia/radiation effects , Phagocytosis/radiation effects , Animals , Behavior, Animal/drug effects , Behavior, Animal/radiation effects , Chemokines/genetics , Chemokines/metabolism , Cognitive Dysfunction/etiology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Macrophages/cytology , Macrophages/metabolism , Male , Memory Disorders/pathology , Memory Disorders/prevention & control , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/metabolism , Organic Chemicals/pharmacology , Phagocytosis/drug effects , Receptor, Anaphylatoxin C5a/metabolism , Synapses/metabolism , Whole-Body Irradiation
7.
Sci Rep ; 7(1): 17525, 2017 12 13.
Article in English | MEDLINE | ID: mdl-29235509

ABSTRACT

Complex alterations in cerebral energetic metabolism arise after traumatic brain injury (TBI). To date, methods allowing for metabolic evaluation are highly invasive, limiting our understanding of metabolic impairments associated with TBI pathogenesis. We investigated whether 13C MRSI of hyperpolarized (HP) [1-13C] pyruvate, a non-invasive metabolic imaging method, could detect metabolic changes in controlled cortical injury (CCI) mice (n = 57). Our results show that HP [1-13C] lactate-to-pyruvate ratios were increased in the injured cortex at acute (12/24 hours) and sub-acute (7 days) time points after injury, in line with decreased pyruvate dehydrogenase (PDH) activity, suggesting impairment of the oxidative phosphorylation pathway. We then used the colony-stimulating factor-1 receptor inhibitor PLX5622 to deplete brain resident microglia prior to and after CCI, in order to confirm that modulations of HP [1-13C] lactate-to-pyruvate ratios were linked to microglial activation. Despite CCI, the HP [1-13C] lactate-to-pyruvate ratio at the injury cortex of microglia-depleted animals at 7 days post-injury remained unchanged compared to contralateral hemisphere, and PDH activity was not affected. Altogether, our results demonstrate that HP [1-13C] pyruvate has great potential for in vivo non-invasive detection of cerebral metabolism post-TBI, providing a new tool to monitor the effect of therapies targeting microglia/macrophages activation after TBI.


Subject(s)
Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/metabolism , Brain/diagnostic imaging , Brain/metabolism , Magnetic Resonance Imaging/methods , Animals , Brain/drug effects , Carbon Isotopes , Disease Models, Animal , Lactic Acid/metabolism , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Organic Chemicals/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyruvic Acid/metabolism , Receptors, Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Colony-Stimulating Factor/metabolism , Spectrophotometry , Superior Sagittal Sinus , Time Factors
8.
Sci Rep ; 7(1): 10227, 2017 08 31.
Article in English | MEDLINE | ID: mdl-28860502

ABSTRACT

The brain's response to radiation exposure is an important concern for patients undergoing cancer therapy and astronauts on long missions in deep space. We assessed whether this response is specific and prolonged and is linked to epigenetic mechanisms. We focused on the response of the hippocampus at early (2-weeks) and late (20-week) time points following whole body proton irradiation. We examined two forms of DNA methylation, cytosine methylation (5mC) and hydroxymethylation (5hmC). Impairments in object recognition, spatial memory retention, and network stability following proton irradiation were observed at the two-week time point and correlated with altered gene expression and 5hmC profiles that mapped to specific gene ontology pathways. Significant overlap was observed between DNA methylation changes at the 2 and 20-week time points demonstrating specificity and retention of changes in response to radiation. Moreover, a novel class of DNA methylation change was observed following an environmental challenge (i.e. space irradiation), characterized by both increased and decreased 5hmC levels along the entire gene body. These changes were mapped to genes encoding neuronal functions including postsynaptic gene ontology categories. Thus, the brain's response to proton irradiation is both specific and prolonged and involves novel remodeling of non-random regions of the epigenome.


Subject(s)
DNA Methylation/radiation effects , Epigenomics/methods , Hippocampus/radiation effects , Whole-Body Irradiation/methods , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/analysis , 5-Methylcytosine/radiation effects , Animals , Gene Expression Profiling , Gene Expression Regulation/radiation effects , Gene Regulatory Networks/radiation effects , Hippocampus/chemistry , Male , Maze Learning/radiation effects , Mice , Protons/adverse effects , Sequence Analysis, RNA , Spatial Learning/radiation effects , Time Factors
9.
J Neurotrauma ; 34(19): 2790-2800, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28376700

ABSTRACT

Traumatic brain injury (TBI) is associated with the development of numerous psychiatric diseases. Of particular concern for TBI patients is the impact of chronic impulsivity on daily functioning. Despite the scope of the human problem, little has been done to address impulsivity in animal models of brain injury. In the current study, we examined the effects of either a severe or a milder bilateral frontal controlled cortical impact injury on impulsivity using the Delay Discounting Task (DDT), in which preference for smaller-sooner over larger-later rewards is indicative of greater impulsive choice. Both milder and severe TBI caused a significant, chronic increase in impulsive decision making. Despite these pronounced changes in performance of the DDT, memory function, as assessed by the Morris Water Maze, was not impaired in more mildly injured rats and only transiently impacted in the severe TBI group. Whereas a significant lesion was only evident in severely injured rats, analysis of cytokine levels within the frontal cortex revealed a selective increase in interleukin (IL)-12 that was associated with the magnitude of the change in impulsive choice caused by both milder and severe TBI. These findings suggest that tissue loss alone cannot explain the increased impulsivity observed, and that inflammatory pathways mediated by IL-12 may be a contributing factor. The findings from this study highlight the sensitivity of sophisticated behavioral measures designed to assess neuropsychiatric dysfunction in the detection of TBI-induced cognitive impairments and their utility in identifying potential mechanistic pathways and therapeutic targets.


Subject(s)
Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/pathology , Decision Making , Impulsive Behavior , Interleukin-12/metabolism , Animals , Behavior, Animal/physiology , Brain Injuries, Traumatic/psychology , Frontal Lobe/injuries , Frontal Lobe/metabolism , Frontal Lobe/pathology , Male , Rats , Rats, Long-Evans
10.
BMC Genomics ; 17(1): 825, 2016 10 24.
Article in English | MEDLINE | ID: mdl-27776477

ABSTRACT

BACKGROUND: Astronauts are exposed to 56Fe ions that may pose a significant health hazard during and following prolonged missions in deep space. We showed previously that object recognition requiring the hippocampus, a structure critical for cognitive function, is affected in 2-month-old mice irradiated with 56Fe ions. Here we examined object recognition in 6-month-old mice irradiated with 56Fe ions, a biological age more relevant to the typical ages of astronauts. Moreover, because the mechanisms mediating the detrimental effects of 56Fe ions on hippocampal function are unclear, we examined changes in hippocampal networks involved in synaptic plasticity and memory, gene expression, and epigenetic changes in cytosine methylation (5mC) and hydroxymethylation (5hmC) that could accompany changes in gene expression. We assessed the effects of whole body 56Fe ion irradiation at early (2 weeks) and late (20 weeks) time points on hippocampus-dependent memory and hippocampal network stability, and whether these effects are associated with epigenetic changes in hippocampal DNA methylation (both 5mC and 5hmC) and gene expression. RESULTS: At the two-week time point, object recognition and network stability were impaired following irradiation at the 0.1 and 0.4 Gy dose, but not following irradiation at the 0.2 Gy dose. No impairments in object recognition or network stability were seen at the 20-week time point at any irradiation dose used. Consistent with this pattern, the significance of pathways for gene categories for 5hmC was lower, though not eliminated, at the 20-week time point compared to the 2-week time point. Similarly, significant changes were observed for 5mC gene pathways at the 2-week time point, but no significant gene categories were observed at the 20-week time point. Only the 5hmC changes tracked with gene expression changes. CONCLUSIONS: Dose- and time-dependent epigenomic remodeling in the hippocampus following 56Fe ion exposure correlates with behavioral changes.


Subject(s)
Cognition/radiation effects , DNA Methylation/radiation effects , Epigenesis, Genetic/radiation effects , Gene Expression Regulation/radiation effects , Hippocampus/metabolism , Hippocampus/radiation effects , Iron , Radiation, Ionizing , Animals , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/radiation effects , Cluster Analysis , Gene Expression Profiling , Gene Ontology , Immunohistochemistry , Male , Maze Learning , Mice , Psychomotor Performance/radiation effects
11.
ACS Chem Neurosci ; 7(11): 1531-1542, 2016 11 16.
Article in English | MEDLINE | ID: mdl-27525447

ABSTRACT

Traumatic brain injury (TBI) affects millions yearly, and is increasingly associated with chronic neuropsychiatric symptoms. We assessed the long-term effects of different bilateral frontal controlled cortical impact injury severities (mild, moderate, and severe) on the five-choice serial reaction time task, a paradigm with relatively independent measurements of attention, motor impulsivity, and motivation. Moderately- and severely injured animals exhibited impairments across all cognitive domains that were still evident 14 weeks postinjury, while mild-injured animals only demonstrated persistent deficits in impulse control. However, recovery of function varied considerably between subjects such that some showed no impairment ("TBI-resilient"), some demonstrated initial deficits that recovered ("TBI-vulnerable"), and some never recovered ("chronically-impaired"). Three clinically relevant treatments for impulse-control or TBI, amphetamine, atomoxetine, and amantadine, were assessed for efficacy in treating injury-induced deficits. Susceptibility to TBI affected the response to pharmacological challenge with amphetamine. Whereas sham and TBI-resilient animals showed characteristic impairments in impulse control at higher doses, amphetamine had the opposite effect in chronically impaired rats, improving task performance. In contrast, atomoxetine and amantadine reduced premature responding but increased omissions, suggesting psychomotor slowing. Analysis of brain tissue revealed that generalized neuroinflammation was associated with impulsivity even when accounting for the degree of brain damage. This is one of the first studies to characterize psychiatric-like symptoms in experimental TBI. Our data highlight the importance of testing pharmacotherapies in TBI models in order to predict efficacy, and suggest that neuroinflammation may represent a treatment target for impulse control problems following injury.


Subject(s)
Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/physiopathology , Disruptive, Impulse Control, and Conduct Disorders/drug therapy , Disruptive, Impulse Control, and Conduct Disorders/physiopathology , Frontal Lobe/injuries , Acute Disease , Adrenergic Uptake Inhibitors/pharmacology , Amantadine/pharmacology , Amphetamine/pharmacology , Animals , Atomoxetine Hydrochloride/pharmacology , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/psychology , Central Nervous System Stimulants/pharmacology , Chronic Disease , Disease Models, Animal , Disease Progression , Disruptive, Impulse Control, and Conduct Disorders/etiology , Disruptive, Impulse Control, and Conduct Disorders/pathology , Dopamine Agents/pharmacology , Frontal Lobe/drug effects , Frontal Lobe/immunology , Frontal Lobe/pathology , Male , Motor Activity/drug effects , Motor Activity/physiology , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/physiology , Rats, Long-Evans , Severity of Illness Index
12.
J Neuroinflammation ; 13(1): 80, 2016 Apr 18.
Article in English | MEDLINE | ID: mdl-27090212

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is a major risk factor for the development of multiple neurodegenerative diseases, including Alzheimer's disease (AD) and numerous recent reports document the development of dementia after TBI. Age is a significant factor in both the risk of and the incidence of acquired brain injury. TBI-induced inflammatory response is associated with activation of brain resident microglia and accumulation of infiltrating monocytes, which plays a pivotal role in chronic neurodegeneration and loss of neurological function after TBI. Despite the extensive clinical evidence implicating neuroinflammation with the TBI-related sequelae, the specific role of these different myeloid cells and the influence of age on TBI-initiated innate immune response remain unknown and poorly studied. METHODS: We used gene profiling and pathway analysis to define the effect of age on inflammatory response at the time of injury. The recruitment of peripheral CCR2(+) macrophages was delineated using the CX3CR1 (GFP/+) CCR2 (RFP/+) reporter mouse. These responses were examined in the context of CCR2/5 antagonism using cenicriviroc. RESULTS: Unsupervised gene clustering and pathway analysis revealed that age predisposes exacerbated inflammatory response related to the recruitment and activation of peripheral monocytes to the injured brain. Using a unique reporter animal model able to discriminate resident versus peripherally derived myeloid cells, we demonstrate that in the aged brain, there is an increased accumulation of peripherally derived CCR2(+) macrophages after TBI compared to young animals. Exaggerated recruitment of this population of cells was associated with an augmented inflammatory response in the aged TBI animals. Targeting this cellular response with cenicriviroc, a dual CCR2/5 antagonist, significantly ameliorated injury-induced sequelae in the aged TBI animals. CONCLUSIONS: Importantly, these findings demonstrate that peripheral monocytes play a non-redundant and contributing role to the etiology of trauma-induced inflammatory sequelae in the aged brain.


Subject(s)
Aging/pathology , Brain Injuries, Traumatic/pathology , Inflammation/pathology , Macrophages/pathology , Aging/immunology , Aging/metabolism , Animals , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/metabolism , Cluster Analysis , Disease Models, Animal , Female , Flow Cytometry , Inflammation/immunology , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Real-Time Polymerase Chain Reaction , Receptors, CCR2/metabolism , Receptors, CCR5/metabolism
13.
PLoS One ; 11(1): e0148001, 2016.
Article in English | MEDLINE | ID: mdl-26808663

ABSTRACT

Following the primary mechanical impact, traumatic brain injury (TBI) induces the simultaneous production of a variety of pro- and anti-inflammatory molecular mediators. Given the variety of cell types and their requisite expression of cognate receptors this creates a highly complex inflammatory milieu. Increasingly in neurotrauma research there has been an effort to define injury-induced inflammatory responses within the context of in vitro defined macrophage polarization phenotypes, known as "M1" and "M2". Herein, we expand upon our previous work in a rodent model of TBI to show that the categorization of inflammatory response cannot be so easily delineated using this nomenclature. Specifically, we show that TBI elicited a wide spectrum of concurrent expression responses within both pro- and anti-inflammatory arms. Moreover, we show that the cells principally responsible for the production of these inflammatory mediators, microglia/macrophages, simultaneously express both "M1" and "M2" phenotypic markers. Overall, these data align with recent reports suggesting that microglia/macrophages cannot adequately switch to a polarized "M1-only" or "M2-only" phenotype, but display a mixed phenotype due to the complex signaling events surrounding them.


Subject(s)
Brain Injuries/immunology , Brain Injuries/physiopathology , Inflammation/metabolism , Macrophages/metabolism , Animals , Inflammation/etiology , Male , Mice , Signal Transduction
14.
J Neurosci ; 35(2): 748-60, 2015 Jan 14.
Article in English | MEDLINE | ID: mdl-25589768

ABSTRACT

Traumatic brain injury (TBI) is a major risk factor for the development of multiple neurodegenerative diseases. With respect to the increasing prevalence of TBI, new therapeutic strategies are urgently needed that will prevent secondary damage to primarily unaffected tissue. Consistently, neuroinflammation has been implicated as a key mediator of secondary damage following the initial mechanical insult. Following injury, there is uncertainty regarding the role that accumulating CCR2(+) macrophages play in the injury-induced neuroinflammatory sequelae and cognitive dysfunction. Using CX3CR1(GFP/+)CCR2(RFP/+) reporter mice, we show that TBI initiated a temporally restricted accumulation of peripherally derived CCR2(+) macrophages, which were concentrated in the hippocampal formation, a region necessary for learning and memory. Multivariate analysis delineated CCR2(+) macrophages' neuroinflammatory response while identifying a novel therapeutic treatment window. As a proof of concept, targeting CCR2(+) macrophages with CCX872, a novel Phase I CCR2 selective antagonist, significantly reduced TBI-induced inflammatory macrophage accumulation. Concomitantly, there was a significant reduction in multiple proinflammatory and neurotoxic mediators with this treatment paradigm. Importantly, CCR2 antagonism resulted in a sparing of TBI-induced hippocampal-dependent cognitive dysfunction and reduced proinflammatory activation profile 1 month after injury. Thus, therapeutically targeting the CCR2(+) subset of monocytes/macrophages may provide a new avenue of clinical intervention following TBI.


Subject(s)
Brain Injuries/drug therapy , Cognition , Macrophage Activation , Macrophages/drug effects , Receptors, CCR2/agonists , Animals , CX3C Chemokine Receptor 1 , Female , Hippocampus/cytology , Hippocampus/physiopathology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...