Inflammation and Neuroprotection in Traumatic Brain Injury

  • Upload
    cemoara

  • View
    217

  • Download
    0

Embed Size (px)

Citation preview

  • 7/26/2019 Inflammation and Neuroprotection in Traumatic Brain Injury

    1/8

    Copyright 2015 American Medical Association. All rig hts reserved.

    Inflammation andNeuroprotection in Traumatic Brain Injury

    Kara N. Corps, DVM, DACVP; TheodoreL. Roth,MS; DorianB. McGavern,PhD

    IMPORTANCE Traumatic brain injury (TBI) is a significant publichealth concern that affects

    individuals in alldemographics.With increasing interest in themedical and public

    communities, understanding the inflammatory mechanisms thatdrive the pathologic and

    consequentcognitive outcomes can inform future research and clinical decisions for patients

    with TBI.

    OBJECTIVES To review known inflammatory mechanisms in TBIand to highlightclinicaltrials

    and neuroprotective therapeutic manipulations of pathologic and inflammatory mechanisms

    of TBI.

    EVIDENCE REVIEW We searched articles in PubMedpublishedbetween 1960 and August1,

    2014, using the following keywords: traumatic brain injury, sterile injury, inflammation,

    astrocytes, microglia, monocytes, macrophages, neutrophils, T cells, reactive oxygen species,

    alarmins, danger-associated molecular patterns, purinergic receptors, neuroprotection, and

    clinical trials. Previous clinical trials or therapeutic studies that involved manipulation of the

    discussed mechanisms were considered for inclusion. The final list of selected studies was

    assembled based on novelty and direct relevance to theprimary focus of this review.

    FINDINGS Traumatic brain injury is a diverse group of sterile injuries induced by primary and

    secondary mechanismsthat give rise to cell death, inflammation, and neurologic dysfunction

    in patients of all demographics. Pathogenesis is driven by complex, interacting mechanisms

    thatinclude reactive oxygen species, ion channel and gap junction signaling, purinergic

    receptor signaling, excitotoxic neurotransmitter signaling, perturbations in calcium

    homeostasis, and damage-associated molecular pattern molecules, among others. Central

    nervous systemresident and peripherally derived inflammatory cells respond to TBIand can

    provide neuroprotectionor participate in maladaptive secondary injury reactions. The exact

    contribution of inflammatory cells to a TBIlesion is dictated by their anatomical positioning as

    well as the local cues to which they are exposed.

    CONCLUSIONS AND RELEVANCE The mechanisms that drive TBIlesion development as well as

    those that promote repair are exceedingly complex and often superimposed. Because

    pathogenic mechanisms candiversify over time or even differbased on the injury type, it is

    importantthat neuroprotective therapeutics be developed and administered with these

    variables in mind. Due to itscomplexity, TBIhas provenparticularly challenging to treat;

    however, a number of promising therapeutic approaches are now under pre-clinical

    development,and recentclinicaltrials have even yielded a few successes. Given the

    worldwide impactof TBIon thehumanpopulation, it is imperative that research remains

    active in this area andthat we continue to develop therapeutics to improve outcome in

    afflicted patients.

    JAMA Neurol. 2015;72(3):355-362. doi:10.1001/jamaneurol.2014.3558

    Published onlineJanuary 19,2015.

    Video at jamaneurology.com

    Author Affiliations: Viral

    Immunology and IntravitalImaging

    Section, National Institutesof

    NeurologicalDisorders and Stroke,National Institutesof Health,

    Bethesda, Maryland.

    Corresponding Author: DorianB.

    McGavern, PhD, Viral Immunology

    and IntravitalImaging Section,

    National Institutesof Neurological

    Disorders and Stroke, National

    Institutesof Health, 10 Center Dr,

    Bethesda, MD 20892 (mcgavernd

    @mail.nih.gov).

    Section Editor: HassanM.

    Fathallah-Shaykh, MD, PhD.

    Clinical Review & Education

    Clinical Implications of Basic NeuroscienceResearch

    (Reprinted) 355

    Copyright 2015 American Medical Association. All rig hts reserved.

    wnloaded From: http://archneur.jamanetwork.com/ by Cesar Pereira on 03/27/2016

  • 7/26/2019 Inflammation and Neuroprotection in Traumatic Brain Injury

    2/8

    Copyright 2015 American Medical Association. All rig hts reserved.

    Traumatic brain injury (TBI) is a diverse group of brain inju-

    ries thatvary in cause, severity, pathogenesis, and clinical

    outcome. As public awareness of TBI and its conse-

    quences increases, there is a growing need to understand the un-

    derlyingmechanisms anddevelop therapeuticinterventions. Within

    theUnited Statesalone, nearly2 million peoplesustaina TBIannu-

    ally, contributing to one-third of all injury-related deaths. Individu-

    als from all nations and demographics are affected, including ath-letes, military troops, and individuals with unintentional injuries.1-3

    Traumatic brain injury is a significant cause of mortality in children

    and young adults, and the incidence in older individuals has in-

    creased withthe averagelife span.4MildTBI(mTBI)isthemostfre-

    quenttype diagnosed, typically resultingin post-TBI survival. Trau-

    matic brain injury is suspectedto contribute to a variety of chronic

    degenerativeprocesses, including chronic traumaticencephalopa-

    thy, Alzheimerdisease,and Parkinsondisease.5 Traumaticbrain in-

    jury is initiated by the application of mechanical force to the head,

    whichcan occurwith orwithout lossof consciousness.This thentrig-

    gers a series of cerebral events that depend in part on the nature

    andlocationof theinjury. A major challengeassociated with treat-

    ingpatientswithTBIisthediversepathologicandpathogenicmecha-

    nisms that become operational after injuries. For example, TBI of-

    ten promotes disruptionof blood-brain barrier (BBB)integrity and

    theneurovascularunit, which canresultin vascular leakage,edema,

    hemorrhage,and hypoxia.Other pathologicmechanisms includecell

    death within the meninges and brain parenchyma, stretching and

    tearing of axonal fibers, and disruptions at the junctions between

    whiteand graymatter, stemmingfrom rotational forces thatcause

    shearing injuries.6Allthese primary pathologic mechanisms are ac-

    companied by cellularand molecular cascadesleading to inflamma-

    tion andadditionalcell death.Thisreviewfocuses onour currentun-

    derstanding of the sterile immune reaction to TBI and someclinical

    successes in treating patients with TBI.

    We searched articles in PubMed published between 1960

    and August1, 2014, using the following keywords: traumatic braininjury, sterile injury, inflammation, astrocytes, microglia, mono-

    cytes, macrophages, neutrophils, T cells, reactive oxygen species,

    alarmins, danger-associated molecular patterns, purinergic recep-

    tors, neuroprotection,andclinical trials.Clinical trials or therapeu-

    tic studies that involved manipulation of the discussed mecha-

    nisms were considered for inclusion. The final reference list was

    assembled based on novelty and direct relevance to the primary

    focus of this review.

    Sterile Immune Reaction to TBI

    Centralnervous system (CNS)residentand peripherally derived in-flammatory cellsrespond quickly to brain injuriesand caneven par-

    ticipatein the repair process.7,8These responsesare commonlyre-

    ferredto as sterile immune reactions. A previous study9 found that

    the inflammatory gene expression profile is comparable between

    mTBIand severe TBI,suggestinga commonresponse toboth forms

    of injury. The acute cellular reaction to TBIincludesastrocytes, mi-

    croglia,monocytes or macrophages,neutrophils, andT cells, which

    are initiallyactivated in part by purinergicreceptor signaling.10,11 In

    the following paragraphs, we describe the inflammatory response

    toTBI inmore detail,focusingspecificallyon traditional immunecell

    populations.Sterile immune reactionsare at least initiallydesigned

    to be beneficial butcan becomedetrimentalin certain situations.

    Danger Signals

    Pathogens can trigger innate immune activation via pathogen-

    associatedmolecular patternmolecules,which are conserved struc-

    tureswithina classof microbes recognized by Toll-like receptors or

    pathogen-recognition receptors. These innate signaling pathwaysallow plants and animals to respond quickly to invading microbes.

    However, it is nowrecognized thattissuedamagein theabsenceof

    microbial infection can trigger inflammasome and innate immune

    activation throughthe releaseof damage-associatedmolecular pat-

    ternmolecules(DAMPs),sometimesreferredto asdangersignals.12

    Alarmins are endogenousDAMPs released by cellsundergoing no-

    napoptotic death or by cells of the immune system.13 Some ex-

    amples of alarmins include HMGB1, S-100 proteins, adenosine tri-

    phosphate (ATP),uric acid,DNA or RNA, andinterleukin 1, among

    others.AfterTBI, alarminsare undoubtedly released,14andthistrig-

    gers a sterile immune reaction designed to restore tissue homeo-

    stasis. However, theseverityand duration of injury canfoster mal-

    adaptiveimmunereactionsthatbecomeinjurious.Apreviousstudy15

    found thatATPrelease anddetectionvia puringericreceptors elicit

    an acutely neuroprotective inflammatory response after mild cor-

    ticalinjury, butsustainedimmune activationmay notalways beben-

    eficial.For example,therapeuticblockade ofinflammasomeactiva-

    tionreducedinnateimmuneactivationand severe TBIlesionsize.16

    Thus, additional researchis required to better understand therules

    that govern pathogenic vs nonpathogenic innate immune reac-

    tions after DAMP signaling in theinjured brain.

    PurinergicReceptor Signaling

    Purinergic receptors are an evolutionarily ancient family of trans-

    membranemolecules thatdetect ATP, adenosinediphosphate(ADP),

    oradenosine.10,11Thereceptors aredividedinto2 basicclassesbased

    onwhethertheyrespondto adenosine (P1receptors)or ATP or ADP(P2receptors).BecauseATP is a cellularsource ofenergy, it is main-

    tained ata highintracellular concentration during steady-state con-

    ditions. Aftertissue injury, ATPis released fromdamagedcells, which

    triggers an immune reaction via purinergic receptor signaling. This

    reaction canbe amplified by pannexin and connexinhemichannels

    thatpump ATP fromhealthycells intothe extracellularspace. Ster-

    ileimmunereactionsgenerallysubsideasATPisconvertedintoaden-

    osine through a 2-step reaction that involves ectonucleoside tri-

    phosphate diphosphohydrolase1 (CD39) and ecto-5-nucleotidase

    (CD73). Astrocytes and microglia each express at least one these

    ectoenzymes,17,18 allowing them to dampen ATP-mediated neuro-

    inflammation.

    Microglia

    Microglia are highly dynamic CNS resident innate immune senti-

    nels that originate from primitive myeloid progenitor cells during

    development.19,20Microgliaparticipate in a variety of homeostatic

    CNS functions, including synaptic plasticity and learning,21 andare

    often the first responders to any inflammatory event that occurs

    within the parenchyma.20 Microglia mediate neuron removal dur-

    ingdevelopment via release of reactive oxygen species (ROS) and

    can acquire a phagocytic phenotype without an inflammatory

    response.20 Microglial expression of genes associated with neuro-

    Clinical Review & Education ClinicalImplications of BasicNeuroscienceResearch TraumaticBrain Injury

    356 JAMANeurology March2015 Volume 72,Number 3 (Reprinted) jamaneurology.com

    Copyright 2015 American Medical Association. All rig hts reserved.

    wnloaded From: http://archneur.jamanetwork.com/ by Cesar Pereira on 03/27/2016

  • 7/26/2019 Inflammation and Neuroprotection in Traumatic Brain Injury

    3/8

    Copyright 2015 American Medical Association. All rig hts reserved.

    protection is upregulated with age.22 Microglia express a large

    number of surface and cytoplasmic receptors, and cumulative sig-

    naling through these receptors determines whether microglia

    remain in a ramified, sentinel state or take on various configura-

    tions as a result of activation.23 In addition, microglia can sense a

    large repertoire of exogenous and endogenous signals, allowing

    for dynamic responses to sterile injuries and infectious agents

    that can be injurious or neuroprotective, depending on thecontext.22

    During CNS autoimmune disease, activated microglia phago-

    cytose debrisand downregulate cellular metabolism in contrastto

    disease-initiating, peripherally derived macrophages, which ap-

    pearto playa moredestructiverole by promotingdemyelination.24

    These datasuggestthatmicrogliaare notinherently neurotoxicdur-

    ing development of a CNS autoimmune disease. After acute focal

    brain injury in rodents, microglia similarly appear to play a neuro-

    protective role.15 Using 2-photonmicroscopy, we revealed thatmi-

    croglia respond within minutes of brain injury by extending pro-

    cesses tothe glial limitansand circumscribingindividualastrocytes,

    resemblingahexagonalhoneycombstructure(Figure1A-E,Figure2E

    and H,and Videos1 and 2). This reaction was dependenton purin-

    ergic receptor signaling (P2X4and P2Y12) and astrocytic ATP-

    dependent ATP release via connexinhemichannels. In response to

    cell death (eg, astrocytic cell death in the glial limitans), microglia

    transformed into phagocytic cells that resembled jellyfish

    (Figure 1A-E, Figure 2F andI, andVideos1 and 2). Jellyfish microg-

    lia werehighly mobile andoften inserted themselvesinto the dam-

    agedglial limitansin place of dead astrocytes, connecting together

    to form a phagocytic barrier. This reaction was also dependent on

    purinergicreceptorsignaling(P2X4,P2Y6,P2Y12)andconnexinhemi-

    channels. When these microglia responses were inhibited locally

    throughblockadeof purinergic receptorsignalingor connexin hemi-

    channels, the pathologic mechanisms observed after brain injury

    were moresevere.One of themost notable changes wasincreased

    leakage of materialsthrough the gliallimitans intothe brainparen-chyma.These data suggest that microglianot only clean up debris

    fromthe injured brain butalso helpmaintain glial limitans barrier in-

    tegrity by sealing thegapsthatresult from dead or damaged astro-

    cytes.Moreover, our dataare consistent withpreviousstudies25-28

    thatlinkmicrogliainjuryresponses toATPreleaseand purinergic re-

    ceptor signaling. Although it is conceivable that microglia re-

    sponses become maladaptive overtime or afterexposure to differ-

    ent combinations of stimuli,29 we propose that the acute role of

    microglia in the focally injured brain is neuroprotective.

    Monocytes and Macrophages

    Monocytesare a multipotent populationof circulatingbonemarrow

    derived leukocytes capable of differentiating into macrophages ordendriticcells afterinvasionof an infectedor injured tissue.30They

    are also known to participate in diverse functions, such as phago-

    cytosis, cytokine or chemokine release, antigen presentation, im-

    munemodulation,and tissue repair. Inthe naivebrain,thereare also

    populations of specialized macrophages that reside in the menin-

    ges,choroidplexus,andperivascularspaces.31TheirroleinTBIpatho-

    genesisis unknown.Anotherstudy15alsofoundthatmeningeal mac-

    rophagesareamongthefirstcellstodieafterfocalcorticalinjuryand

    may serve as an early source of alarmins and ROS (Figure 1A-C,

    Figure2AandB,and Video1).Monocyte-derivedmacrophagescom-

    ingfromtheblooddonotreachpeaknumbersinthedamagedbrain

    ofanimalsand humansuntil 24to 48 hours after injury.32,33 Mono-

    cytes are capable of crossing the bloodcerebrospinal fluid barrier

    with neutrophils into the injured brain as a result of CCL2 produc-

    tionby choroid plexus epithelium.34 CCL2is significantlyincreased

    in the cerebrospinal fluid of patients with TBI.33 Examination of

    CCL2/miceafterTBI revealedslight alterations incytokine expres-

    sion but no changes in lesion size within the first week of injury.

    33

    However, when followed for a longer timeframe of 2 to 4 weeks,

    CCL2/micehad improved functionalrecovery, suggestinga patho-

    genicrole formacrophages during thechronicphase of TBI.Similar

    results were obtained in CCR2/ mice after TBI.35 CCR2 is the re-

    ceptor for CCL2, and deficiency significantly reduced the number

    of lesion macrophages and increased hippocampal neuronal den-

    sities, spatial learning, and locomotion when measured several

    weeks after brain injury. Collectively, the data obtained in CCL2

    and CCR2 knockout mice suggest that monocyte-derived macro-

    phages play a pathogenic role in the chronic phase after TBI.

    Additional studies are required to determine whether these cells

    can participate in brain repair after TBI similar to what has been

    described in models of spinal cord injury.36 Whether a macro-

    phage is pathogenic or beneficial after tissue injury likely depends

    on its state of differentiation.

    Neutrophils

    Neutrophils arean abundantpopulationof circulating leukocytesthat

    are usually among the first responders to tissue injuries in the pe-

    ripheryandCNS.37Neutrophilsare oftenviewedas a proinflamma-

    tory cell population butareknown toplaya vitalrolein woundheal-

    ing through their involvement in phagocytosis, metalloproteinase

    release, and growth factor production. After tissue injury, neutro-

    phils can help prepare the damaged environment for repair. Neu-

    trophilsare rapidly recruitedto theCNS after TBIand enter through

    meningealblood vessels andthe choroidplexus.15,32,38,39Theycan

    also facilitate the recruitment of monocytes.37

    A previous study40

    focused on sterile injury of the liver found that ATP released from

    the damaged tissue induced inflammasome activation in a P2X7R-

    dependent manner. This activation in turn promoted rapid

    recruitment of neutrophils through release of chemoattractants

    (CXCL1 and CXCL2) and formyl peptides that guided these cells to

    thesite of injury. After focal TBI, we observed that neutrophils are

    similarly recruited in a P2X7R-dependent manner and arrive

    within 1 hour of injury (Figure 2C). 15 Visualization of cellular

    dynamics and localization by 2-photon microscopy revealed that

    neutrophils localized primarily to the damaged meninges (instead

    of theparenchyma), where they swarmed the area andinteracted

    with dead cells. Antagonism of this response by blocking P2X7R

    signalingincreased theamount of cell death in themeninges, sug-gesting a protective role for neutrophils in the meningeal space

    after focal cortical injury.

    Neutrophils are not always neuroprotective and have the ca-

    pacity tobreak downthe BBBby releasing metalloproteinases,pro-

    teases, tumor necrosis factor , and ROS. Inflammatory mediators

    released after brain injury can facilitate this process by inducing a

    hyperactivated state thatallowsneutrophilsto breach theBBB and

    enter the CNS.41 On arrival, neutrophils have the potential to in-

    duceneuronalcelldeathusingthesamesolublemediatorsthatbreak

    down the BBB.42 A previous study43 revealed that neutrophils are

    TraumaticBrain Injury ClinicalImplications of BasicNeuroscienceResearch ClinicalReview & Education

    jamaneurology.com (Reprinted) JAMA Neurology March2015 Volume 72,Number 3 357

    Copyright 2015 American Medical Association. All rig hts reserved.

    wnloaded From: http://archneur.jamanetwork.com/ by Cesar Pereira on 03/27/2016

  • 7/26/2019 Inflammation and Neuroprotection in Traumatic Brain Injury

    4/8

    Copyright 2015 American Medical Association. All rig hts reserved.

    themost abundantcell population in circulation after TBIand cause

    increasedexpression of oxidative enzymes indicative of activation.

    Depletion of neutrophils with antiGr-1 antibodies after controlled

    cortical impact in rodents reduced edema, microglia and macro-

    phageactivation,and TBIlesion size,but didnot affectvascularleak-

    ageat 24 to 48 hoursafter injury.44 These data reveal thatneutro-

    phils can be pathogenic after open-skullcortical impact. However,

    thecontribution ofneutrophilsto a CNSlesionmay depend ontheir

    preciselocalization andstateofactivation. Open-skullcontrolled cor-

    tical impact is highly disruptive tomeningeal architecture andlikely

    favors neutrophil recruitmentto the heavily damaged brainparen-

    chyma. These findings contrast with mild closed-skull cortical in-

    jury,which maintainsmeningeal architectureand fostersa more se-

    lective patternof neutrophil recruitment.15Todefinitively establish

    the contribution of neutrophils to TBI pathogenesis, these cells

    shouldbeevaluatedinmanydifferentmodelsofbraininjury.Itiscon-

    ceivablethattheir contribution willdifferbasedon thenatureof the

    injury.

    Figure 1. Pathogenesisof Traumatic Brain Injury (TBI)

    ATP

    ROS

    UDP

    Glutamate

    Skull

    Normal mTBI

    Dura mater

    Meningealmacrophage

    Blood vesselsArachnoid mater

    Subarachnoidspace ROS

    Fluid leakage,meningealcell death Neutrophils

    Jellyfish microgliaMicroglial process

    extension to the glial limitans

    Glutamate

    NMDA

    Ca2+ Necroticneuron

    UDP

    ATPGlial limitans

    Pia mater

    CSF

    Astrocyte

    Corticalneuron

    Microglia

    A

    20 m

    Parenchymalcell death

    250 m

    Skull bone

    Meningeal macrophages

    Parenchyma

    Meninges

    Microglia

    Microglia

    B

    D

    C

    E

    A, Comparisonof brain anatomy in

    the meninges and superficial

    neocortexbeforeand after focal mild

    TBI(mTBI). Theduramatercontains

    numerous small vessels that arelined

    by thin,elongatedmeningeal

    macrophages.The subarachnoid

    spacecontains vessels, fibroblastlike

    stromal cells,and cerebrospinal fluid

    (CSF).The glial limitans,composedof

    astrocytic foot processes, lies

    beneath thepia mater andformsa

    barrierbetweenthe CSFand

    underlyingparenchyma. Mild focal

    braininjury mechanically compresses

    the meningeal space, compromising

    vascular integrityand inducing rapid

    necrosis of meningeal macrophages

    andstructural cells.Leakageof fluid

    from meningeal blood vessels results

    in edema,and damaged cells within

    the meninges release reactive oxygen

    species (ROS) and adenosine

    triphosphate(ATP),initiating a sterile

    immunereaction. B andC, Maximum

    projections (5-m wide) areshownin

    thexzplaneof 2-photonz-stacks

    captured through thethinned skull of

    CX3CR1GFP/+ mice(original

    magnification20).

    B, A representativeimageof an

    uninjured mousereveals the

    presence of meningeal macrophages

    (green)in theduraand ramified

    microglia (green)in thebrain

    parenchymabeneaththe glial

    limitans (white dotted line).

    C, Thirtyminutes after focal mTBI,

    meningealmacrophagesdie and

    microglia relocate to theinjuredglial

    limitans (arrowheads).Skull boneis

    shown inblue. D and E,

    Histopathologic analysis of the

    superficial neocortexby confocal

    microscopy8 hours after mTBI

    (original magnification20). D, An

    uninjuredbrainis shownfor

    comparison. Deadcells werelabeled

    transcranially with propidium iodide.

    Cell nucleiare blue.E, A large lesion

    consistingof numerous deadcells

    (red)(arrowhead). See Videos1 and

    2. UPD indicates uridinediphosphate.

    Clinical Review & Education ClinicalImplications of BasicNeuroscienceResearch TraumaticBrain Injury

    358 JAMANeurology March2015 Volume 72,Number 3 (Reprinted) jamaneurology.com

    Copyright 2015 American Medical Association. All rig hts reserved.

    wnloaded From: http://archneur.jamanetwork.com/ by Cesar Pereira on 03/27/2016

  • 7/26/2019 Inflammation and Neuroprotection in Traumatic Brain Injury

    5/8

    Copyright 2015 American Medical Association. All rig hts reserved.

    T Cells

    Although T cells play diverse roles in adaptive immune responses

    andthe regulation of inflammation, their role (ifany) in TBIpatho-

    genesis is not clear. It has been proposed that autoreactive T cells

    against CNS antigens, such as myelin basic protein, can be neuro-

    protective after spinal cord injury.45 After brain injury, activated T

    cellsare recruitedto sites ofdamage,46andROS release may facili-

    tate this recruitment by activating endothelial barriers.47 To ad-

    dresstheroleof T cells inTBI, a previousstudy48 examined the re-

    sponseto closed-skullhead injury in RAG1 knockoutmice that lack

    matureT and B cells.No difference in any pathologic or neurologic

    parameters was observed between wild-type andRAG1-deficient

    mice forup to1 week.Thesedatasuggestthat T cellsplay norolein

    early TBI pathogenesis. Additional studies are required to deter-

    mine whether T cells actively participate in chronic TBIlesions (be-

    yond 1 week) and/or thereparativeprocess.

    Therapeutic Modulation of TBI Pathogenesis

    The pathogenesis of TBI is complex as reflected by the number of

    clinical trials thathave failed to improve outcomes in humans.49,50

    The many reasons for these failures have been discussed in other

    reviews.49,50Rather thanfocuson thereasonsfor priorfailures, we

    instead briefly discuss somesuccessesthat pertain to mechanisms

    of pathogenesis and inflammation covered in this review.

    Theconceptof freeradicalmediated damage of CNStissue af-

    terinjury hasexistedfor several decades.51,52Administration of ef-

    fectiveantioxidantshas thepotential to significantlylimit thespread

    of damageand inflammationif given soon after brain injury. In ani-

    malmodels, a number of previous studies53,54 have yielded prom-

    ising results withantioxidants thatneutralizeROS. Forexample,in-

    travenousadministrationof thesmall-moleculefree radicalscavenger

    edaravone at 2 and 12 hours after weight dropinduced TBI re-

    sulted in significantly reduced inflammation, edema, BBB break-

    down,lesion size,and neurologicdeficits.53 Inhibitionof NADPHoxi-

    dasecomplexassemblywithapocyninalsoreducedROSproduction,

    BBB breakdown, and neuronal cell death after weight drop

    induced TBI.54 The only caveatof this study was that the apocynin

    was injected intraperitoneally 15 minutes before injury. Neverthe-

    less,the favorable outcome implicatesNADPH oxidase as a poten-

    tial source of ROSafter brain injury.

    Using a newmodelof mild cortical injury, we found that trans-

    cranial administration of the antioxidant glutathione at 15 minutes

    Figure 2. Inflammatory Reaction to Traumatic Brain Injury

    A

    G

    B

    E

    H

    C

    F

    I

    50 m

    20 m

    50 m

    50 m

    Normal Ram if ie d Microgl ia Hon eycom b Microgl ia Phagocytic Jel lyfish Microgl ia

    Normal mTBI Myelomonocytic Cells

    Meningeal Macrophages

    D

    A-I,The 25-mxymaximum

    projections from CX3CR1GFP/+ (A,B,

    andD-I)or LysMGFP/+ (C)micewere

    captured by 2-photon microscopy

    through a thinned skull.A, Meningeal

    macrophages (green) are thin,

    elongated cellsthat residealong the

    dural blood vessels in theuninjured

    brain.B, After focal mild traumatic

    braininjury (mTBI), meningeal

    macrophages undergo necrosis

    within30 minutes and disappear

    fromthefield of view.

    C, Myelomonocytic cells(green)

    invadethe damaged meninges within

    anhourof braininjury. D and G,In the

    uninjured brain,microglia (green)

    have small cell bodies andare highly

    ramified. Focalbrain injuryinduces

    the rapidtransformation of microglia

    into at least2 distinct morphologic

    patterns.E andH, Honeycomb

    microglia extend processes that

    circumscribethe bordersbetween

    individual astrocytesin the glial

    limitans. F andI, Phagocytic jellyfish

    microglia are generatedin response

    tocelldeathandforma film across

    the damaged glial limitans.

    High-magnificationviews in panels G

    through I aredenoted with white

    boxesin panelsD throughF (original

    magnification20). See Videos1

    and 2.

    TraumaticBrain Injury ClinicalImplications of BasicNeuroscienceResearch ClinicalReview & Education

    jamaneurology.com (Reprinted) JAMA Neurology March2015 Volume 72,Number 3 359

    Copyright 2015 American Medical Association. All rig hts reserved.

    wnloaded From: http://archneur.jamanetwork.com/ by Cesar Pereira on 03/27/2016

  • 7/26/2019 Inflammation and Neuroprotection in Traumatic Brain Injury

    6/8

    Copyright 2015 American Medical Association. All rig hts reserved.

    or 3 hours after injury significantly reduced inflammation, glial limi-

    tansbreakdown,and parenchymal(but notmeningeal)cell deathby

    up to approximately 70%.15Pretreatmentwith glutathione reduced

    meningealcell deathby approximately50%. Thesedataindicate that

    ROS area primary inducer of cell death andinflammationafter focal

    brain injury andthatan antioxidant canhavea major effecton lesion

    expansion if givenearly.The advantageof passing a neuroprotective

    compounddirectlythroughtheskullbone(transcranialdelivery)isthatahighlocaldrugconcentrationcanbeachievedintheCNSwithalim-

    itedoff-target effect on theperiphery.

    Previous studies55,56 have supported antioxidants as neuro-

    protective agents in rats and humans, revealing that administra-

    tionofN-acetylcysteine reduces brain damage andimproves recov-

    eryafterTBI.N-acetylcysteineisthecellularprecursortoglutathione.

    A randomized, double-blind, placebo-controlledclinicaltrial55 was

    performed toassess efficacy inmembers ofthe military whoexpe-

    rienced a mTBI that resulted from blastexposure. Patients whore-

    ceivedN-acetylcysteine within 24 hours had significantly im-

    proved recovery during a 7-day period when compared with a

    placebo control group. These findings were corroborated in 2 dif-

    ferent rodent models of TBI (weight drop and fluid percussion),

    whichrevealed thatN-acetylcysteine reversed the behavioral defi-

    cits associated with mTBI and moderate TBI.56 Further studies are

    needed to determine whether this promising neuroprotective in-

    tervention willbe efficaciousin patientswith diverse typesof brain

    injury.

    Many clinical trials have been completed or are under way to

    assess therole of excitotoxicmechanismsin TBIpathogenesis.49,50

    With the exception of amantadine, all drugs in this class tested to

    datehavenot beeneffective inpromoting recoveryin patientswith

    TBI. Amantadine is thought to act as anN-methyl-D-aspartate re-

    ceptor antagonist and an indirect dopamine agonist. When pa-

    tientswithTBI were treated duringa 4-week periodbeginning4 to

    16 weeksafterinjury, amantadineimproved recoveryrelative tothe

    placebo control. The mechanismunderlying thispositive effectre-mains unclear. Prevention ofN-methyl-D-aspartate receptor

    mediatedexcitatorydamageseemsunlikelygiventhatthe drugwas

    administered a monthor more after theinitial injury.57

    Manipulationof purinergicreceptorsignaling is another thera-

    peutic approach worth considering. Use of specific purinergic re-

    ceptor agonists and antagonists should allow therapeutic amelio-

    rationof differentTBI lesionparameters.A previous study15 found

    that microgliaresponsesafter mTBI weredependent onP2X4,P2Y6,

    andP2Y12receptors,whereasP2X7Rsignalingwasnecessaryforneu-

    trophil recruitment. It might be possible to promote neuroprotec-

    tiveinflammatory responsesthrough therapeutic agonismof these

    pathways after brain injury. The challenge, however, with puriner-

    gic receptor manipulation is that specific receptors are often ex-pressed on multiple cell populations. A purinergicreceptor agonist

    or antagonist will likely affect multiple cell populations simultane-

    ously. As an example, a previous58 study found that P2X7R local-

    ized to astrocytic end feet and antagonism of this receptor re-

    duced astrocyte activation, cerebral edema, and neurobehavioral

    abnormalities after controlled cortical impactinduced TBI. A simi-

    larprotectiveeffectwasobtainedbyblockingP2X 7Rafterspinalcord

    injury, which was linked to receptor expression on spinal cord

    neurons.59However, P2X7R isalso expressed oninflammatory cells,

    and a previous study15 found that antagonism of this pathway in-

    creased meningeal cell death after mTBI, likely due to diminished

    neutrophil recruitment. Thus, purinergic receptor modulation can

    positively affect one CNS environment and negatively affect an-

    other.It willtherefore be important in future studies tomap outthe

    exactcontributions of specific purinergicreceptors to differentTBI

    lesion parameters beforedeciding which (ifany) arebest to target

    therapeutically in patients.

    Discussion

    The pathogenesis of TBI is initially induced by a mechanical injury

    that sets into motion a complex secondary reaction mediated by

    ROS, purines, calcium ions, excitatory amino acids, and DAMPs,

    amongothers.Thispathogenesis in turntriggers a robuststerileim-

    mune reaction that consists of CNS resident and peripherally re-

    cruited inflammatory cells. The response is designed to be neuro-

    protectiveandpromotewoundhealingbutcanbecomemaladaptive

    over time, especially if thelesion remains active for weeks.Among

    the earliest soluble mediators are ROS and purines. Both are re-

    leased within minutes of brain injury and initiate an inflammatory

    cascade. Even after mild focal cortical injury, ROS can damage the

    glial limitans that separate themeningesand parenchyma, which re-

    sults in lesion expansion within brain tissue. Vascular damage and

    leakage represent another early hallmark of TBI pathogenesis that

    can foster edema, hypoxia, and tissue destruction. After brain in-

    jury,the innateimmune systemquickly mobilizesin responseto pu-

    rines andalarmins,and astrocytes helporchestrate this responseby

    serving as inflammatory amplifiers. Within minutes, resident mi-

    crogliaareamongthefirsttoreactbyfortifyingCNSbarriersandpar-

    ticipating in phagocytic cleanup. Neutrophils and monocytes ar-

    rive shortly thereafter and preferentially survey injured meningeal

    spaces if theCNS architectureremainsintact.Focal brain injury elic-

    its an anatomically partitioned immune reaction (at least acutely)

    with myelomonocytic cells tending to the damaged meninges andmicroglia responding within the parenchyma. Eventually, myelo-

    monocyticcellscanenterthedamagedbrain,andstudies40-42have

    found that their presence there is sometimes neurotoxic. How-

    ever, sterileimmunereactionsare notinherentlyneurotoxicand are

    usuallyelicitedtoprepareadamagedtissueforwoundhealing.Thus,

    the entire contribution of immune cellsubsets to TBI lesions needs

    to be considered before targeted therapeutic interventions can

    be intelligently designed. Another important variable is time. The

    exact contribution of immune cells to a TBIlesion mayin fact shift

    over time. For example, an initially neuroprotective immune

    response may become maladaptive as secondary inducers of tis-

    sue destruction diversify.

    Although TBI has proven difficult to treat, promisinginterven-tions lieon thehorizon. Given theimportance of ROSin TBIpatho-

    genesisandthesuccesswithN-acetylcysteine in patients withmTBI,

    clinical pursuit of antioxidant therapy seems warranted. The likely

    key to success is early treatment with antioxidants so that TBI le-

    sionexpansionand subsequentinflammationcan bestoppedas soon

    as they are initiated. Because TBI lesions begin to expand within

    hoursof injury, development of strategies to rapidly preserve brain

    tissueis paramount. Thekineticsof lesionexpansionmustbe simi-

    larlyconsidered whenattemptingto manipulate purinergicand ex-

    citatoryneurotransmitterpathways, whichengagerapidlyafter in-

    Clinical Review & Education ClinicalImplications of BasicNeuroscienceResearch TraumaticBrain Injury

    360 JAMANeurology March2015 Volume 72,Number 3 (Reprinted) jamaneurology.com

    Copyright 2015 American Medical Association. All rig hts reserved.

    wnloaded From: http://archneur.jamanetwork.com/ by Cesar Pereira on 03/27/2016

  • 7/26/2019 Inflammation and Neuroprotection in Traumatic Brain Injury

    7/8

    Copyright 2015 American Medical Association. All rig hts reserved.

    jury. Therapeutic targeti ng of t hese pathways has the greatest

    likelihoodofworkingifadministeredsoonafterinjury.Forthechronic

    phase of TBI pathogenesis, more research is required to under-

    stand lesion dynamics. Over time, it may become necessary to

    dampen maladaptive inflammatory responses andattemptto pro-

    mote wound healing reactions, which would be challenging to

    achieve without having a better understanding of chronic lesion

    dynamics.

    Conclusions

    Traumatic brain injury encompasses a complex spectrum of inju-

    ries that tax the neural-immune interfaceand canresult in perma-

    nent neurologic dysfunction. Detailed knowledge of this interface

    during the acute and chronic phases of TBI will help us design the

    most efficacious interventions.

    ARTICLE INFORMATION

    Accepted for Publication: October 2, 2014.

    Published Online: January19, 2015.

    doi:10.1001/jamaneurol.2014.3558.

    Author Contributions: DrMcGavernhad full access

    to allthe data in thestudy andtakes responsibility

    forthe integrityof thedataand theaccuracyof the

    data analysis.

    Study concept and design: Corps,McGavern.

    Acquisition, analysis, or interpretation of data: All

    authors.

    Draftingof the manuscript: All authors.

    Critical revision of the manuscript for important

    intellectualcontent: All authors.Obtained funding: McGavern.

    Administrative, technical, or material support:

    McGavern.

    Study supervision: McGavern.

    Conflict of Interest Disclosures: Nonereported.

    Funding/Support: This work wassupported bythe

    National Institutesof Healthintramural program.

    Roleof the Funder/Sponsor:Thefunding source

    hadno rolein thedesign andconduct of thestudy;

    collection, management, analysis, and

    interpretation of thedata; preparation,review, or

    approval of themanuscript; andthe decision to

    submitthe manuscriptfor publication.

    Additional Contributions: Ethan Tyler, MA, and

    Alan Hoofring, MS, Medical Arts Design Section,

    National Institutesof Health, helped withtheillustrationshownin Figure1. MessrsTyler and

    Hoofring are salaried employees of the National

    Institutes of Healthand were notdirectly

    compensatedby ourlaboratoryfor their work.

    REFERENCES

    1. FeiginVL, Theadom A, Barker-Collo S,et al;

    BIONIC StudyGroup. Incidenceof traumatic brain

    injuryin NewZealand:a population-based study.

    LancetNeurol. 2013;12(1):53-64.

    2. JordanBD. Theclinical spectrum of sport-related

    traumatic braininjury. NatRev Neurol. 2013;9(4):

    222-230.

    3. LoganBW,Goldman S,Zola M,Mackey A.

    Concussive braininjury in themilitary:September

    2001 to thepresent. Behav SciLaw. 2013;31(6):803-813.

    4. Roozenbeek B, Maas AI,Menon DK.Changing

    patterns in the epidemiologyof traumatic brain

    injury. NatRev Neurol. 2013;9(4):231-236.

    5. SmithDH, Johnson VE,StewartW. Chronic

    neuropathologies of singleand repetitiveTBI:

    substratesof dementia? NatRev Neurol. 2013;9(4):

    211-221.

    6. Blennow K, HardyJ, Zetterberg H.The

    neuropathology and neurobiologyof traumatic

    braininjury. Neuron. 2012;76(5):886-899.

    7. DasM, MohapatraS, MohapatraSS. New

    perspectiveson central and peripheralimmune

    responses to acutetraumaticbrain injury.

    J Neuroinflammation. 2012;9:236.

    8. Finnie JW. Neuroinflammation:beneficial and

    detrimental effects aftertraumaticbrain injury.

    Inflammopharmacology. 2013;21(4):309-320.

    9. Lagraoui M,LatocheJR, CartwrightNG,

    Sukumar G, Dalgard CL, Schaefer BC. Controlled

    cortical impactand craniotomyinduce strikingly

    similarprofiles of inflammatorygene expression,

    but with distinct kinetics. Front Neurol. 2012;3:155.

    10. JungerWG. Immunecellregulation by

    autocrine purinergic signalling. Nat RevImmunol.2011;11(3):201-212.

    11. Eltzschig HK, SitkovskyMV, RobsonSC.

    Purinergic signaling duringinflammation. N EnglJ

    Med. 2012;367(24):2322-2333.

    12. Matzinger P. Tolerance, danger,and the

    extended family.Annu Rev Immunol. 1994;12:991-

    1045.

    13. Bianchi ME.DAMPs, PAMPsand alarmins:all we

    need to know about danger.J Leukoc Biol. 2007;81

    (1):1-5.

    14. MansonJ, ThiemermannC, Brohi K. Trauma

    alarmins as activators of damage-induced

    inflammation. BrJ Surg. 2012;99(suppl 1):12-20.

    15. RothTL, Nayak D,Atanasijevic T, Koretsky AP,

    LatourLL, McGavern DB. Transcranialamelioration

    of inflammationand cell deathafterbraininjury.

    Nature. 2014;505(7482):223-228.

    16. de RiveroVaccariJP, Lotocki G,AlonsoOF,

    Bramlett HM, Dietrich WD,Keane RW. Therapeutic

    neutralization of the NLRP1 inflammasomereduces

    theinnate immune response and improves

    histopathology after traumatic braininjury.J Cereb

    BloodFlow Metab. 2009;29(7):1251-1261.

    17. Niemel J,IferganI, Yegutkin GG,Jalkanen S,

    Prat A, Airas L. IFN-beta regulatesCD73and

    adenosine expressionat the blood-brainbarrier. Eur

    J Immunol. 2008;38(10):2718-2726.

    18. Braun N, Svigny J,Robson SC,et al.

    Assignmentof ecto-nucleoside triphosphate

    diphosphohydrolase-1/cd39 expressionto microglia

    andvasculatureof thebrain. Eur J Neurosci. 2000;

    12(12):4357-4366.

    19. Ginhoux F, GreterM, Leboeuf M,et al.Fate

    mapping analysis reveals thatadult microglia derive

    from primitive macrophages.Science. 2010;330

    (6005):841-845.

    20. Nayak D,RothTL, McGavern DB.Microglia

    development and function.Annu Rev Immunol.

    2014;32:367-402.

    21. Parkhurst CN,Yang G, NinanI, et al.Microglia

    promote learning-dependentsynapse formation

    through brain-derived neurotrophicfactor. Cell.

    2013;155(7):1596-1609.

    22. Hickman SE,Kingery ND, Ohsumi TK,et al.The

    microglial sensome revealed by directRNA

    sequencing. Nat Neurosci. 2013;16(12):1896-1905.

    23. HuX, LiouAK,LeakRK,etal. Neurobiologyof

    microglial actionin CNSinjuries: receptor-mediated

    signaling mechanismsand functional roles. Prog

    Neurobiol. 2014;119-120(Jun):60-84.

    24. Yamasaki R, Lu H,ButovskyO, et al.Differential

    roles of microglia andmonocytes inthe inflamed

    central nervous system.J Exp Med. 2014;211(8):

    1533-1549.

    25. NimmerjahnA, Kirchhoff F,HelmchenF.

    Resting microglial cellsare highly dynamic

    surveillantsof brainparenchyma in vivo.Science.2005;308(5726):1314-1318.

    26. Davalos D,GrutzendlerJ, YangG, etal. ATP

    mediates rapid microglial response to local brain

    injuryin vivo. Nat Neurosci. 2005;8(6):752-758.

    27. Haynes SE,Hollopeter G, YangG, etal. The

    P2Y12receptorregulates microglial activation by

    extracellular nucleotides. Nat Neurosci. 2006;9(12):

    1512-1519.

    28. Koizumi S, Shigemoto-MogamiY, Nasu-Tada K,

    et al.UDP actingat P2Y6 receptorsis a mediator of

    microglial phagocytosis. Nature. 2007;446(7139):

    1091-1095.

    29. Zhang J,MalikA, ChoiHB,Ko RW,

    Dissing-OlesenL, MacVicar BA. Microglial CR3

    activation triggers long-termsynapticdepressionin

    the hippocampusvia NADPHoxidase. Neuron.2014;82(1):195-207.

    30. ShiC, Pamer EG.Monocyte recruitment during

    infection and inflammation. Nat Rev Immunol.

    2011;11(11):762-774.

    31. Nayak D,Zinselmeyer BH,CorpsKN, McGavern

    DB.In vivo dynamics of innateimmune sentinelsin

    the CNS. Intravital. 2012;1(2):95-106.

    32. SoaresHD,HicksRR, SmithD, McIntoshTK.

    Inflammatoryleukocyticrecruitment and diffuse

    neuronal degeneration are separate pathological

    processes resulting from traumatic braininjury.

    J Neurosci. 1995;15(12):8223-8233.

    33. SempleBD, ByeN, RancanM, Ziebell JM,

    Morganti-Kossmann MC. Roleof CCL2(MCP-1) in

    traumatic braininjury (TBI):evidencefrom severe

    TBI patients and CCL2-/- mice.J Cereb Blood Flow

    Metab. 2010;30(4):769-782.

    34. Szmydynger-ChodobskaJ, Strazielle N, Gandy

    JR, et al. Posttraumatic invasion of monocytes

    acrossthe blood-cerebrospinal fluid barrier.J Cereb

    Blood Flow Metab. 2012;32(1):93-104.

    35. HsiehCL, Niemi EC,Wang SH,et al.CCR2

    deficiency impairsmacrophageinfiltration and

    improves cognitive function aftertraumaticbrain

    injury.J Neurotrauma. 2014;31(20):1677-1688.

    36. ShechterR, LondonA, VarolC, et al.Infiltrating

    blood-derived macrophages are vital cellsplaying

    TraumaticBrain Injury ClinicalImplications of BasicNeuroscienceResearch ClinicalReview & Education

    jamaneurology.com (Reprinted) JAMA Neurology March2015 Volume 72,Number 3 361

    Copyright 2015 American Medical Association. All rig hts reserved.

    wnloaded From: http://archneur.jamanetwork.com/ by Cesar Pereira on 03/27/2016

  • 7/26/2019 Inflammation and Neuroprotection in Traumatic Brain Injury

    8/8

    Copyright 2015 American Medical Association. All rig hts reserved.

    an anti-inflammatory role in recovery from spinal

    cordinjury in mice. PLoSMed. 2009;6(7):e1000113.

    37. Kolaczkowska E, KubesP. Neutrophil

    recruitmentand function in healthand

    inflammation. NatRev Immunol. 2013;13(3):159-175.

    38. CarlosTM, ClarkRS, Franicola-Higgins D,

    Schiding JK, KochanekPM. Expression of

    endothelialadhesion molecules and recruitmentof

    neutrophilsafter traumatic braininjury in rats.

    J Leukoc Biol. 1997;61(3):279-285.

    39. Szmydynger-ChodobskaJ, StrazielleN, ZinkBJ,

    Ghersi-Egea JF, Chodobski A. Theroleof the

    choroidplexus in neutrophil invasion after

    traumatic braininjury.J Cereb Blood Flow Metab.

    2009;29(9):1503-1516.

    40. McDonald B, Pittman K, Menezes GB,et al.

    Intravascular danger signals guideneutrophils to

    sitesof sterile inflammation.Science. 2010;330

    (6002):362-366.

    41. ScholzM, CinatlJ, Schdel-HpfnerM, Windolf

    J. Neutrophilsand theblood-brainbarrier

    dysfunctionafter trauma. MedRes Rev. 2007;27(3):

    401-416.

    42. Nguyen HX, OBarr TJ, Anderson AJ.

    Polymorphonuclearleukocytespromote

    neurotoxicity through release of matrix

    metalloproteinases, reactive oxygen species, and

    TNF-alpha.J Neurochem. 2007;102(3):900-912.

    43. LiaoY,Liu P, GuoF, ZhangZY,Zhang Z.

    Oxidative burstof circulatingneutrophilsfollowing

    traumatic braininjury in human. PLoS One. 2013;8

    (7):e68963.

    44. KenneE, ErlandssonA, Lindbom L, Hillered L,

    Clausen F.Neutrophildepletionreduces edema

    formation and tissueloss following traumatic brain

    injuryin mice.J Neuroinflammation. 2012;9:17.

    45. CohenIR, Schwartz M. Autoimmune

    maintenance and neuroprotection of the central

    nervous system.J Neuroimmunol. 1999;100(1-2):

    111-114.

    46. LingC, SandorM, SureshM, Fabry Z.Traumatic

    injuryand thepresence of antigen differentiallycontribute to T-cellrecruitmentin theCNS.J Neurosci.

    2006;26(3):731-741.

    47. ClausenF, LorantT,Lewn A, Hillered L. T

    lymphocytetrafficking: a novel targetfor

    neuroprotection in traumatic braininjury.

    J Neurotrauma. 2007;24(8):1295-1307.

    48. WeckbachS, Neher M,Losacco JT, etal.

    Challenging the role of adaptive immunity in

    neurotrauma: Rag1(-/-) mice lacking matureB andT

    cellsdo not showneuroprotection afterclosed

    headinjury.J Neurotrauma. 2012;29(6):1233-1242.

    49. JanowitzT, Menon DK.Exploring newroutes

    for neuroprotective drug development in traumatic

    braininjury.Sci Transl Med. 2010;2(27):27rv1.

    50. McConeghyKW,HattonJ, HughesL, CookAM.

    A review of neuroprotection pharmacologyand

    therapiesin patients withacute traumatic brain

    injury. CNS Drugs. 2012;26(7):613-636.

    51. KontosHA. Oxygenradicalsin CNSdamage.

    ChemBiol Interact. 1989;72(3):229-255.

    52. Bains M,Hall ED. Antioxidanttherapies in

    traumatic brainand spinalcord injury. Biochim

    Biophys Acta. 2012;1822(5):675-684.

    53. Wang GH,Jiang ZL,Li YC,et al.Free-radical

    scavengeredaravonetreatmentconfers

    neuroprotection against traumatic braininjury in

    rats.J Neurotrauma. 2011;28(10):2123-2134.

    54. Choi BY, Jang BG,Kim JH,et al.Prevention of

    traumatic braininjury-induced neuronal deathby

    inhibition of NADPHoxidaseactivation. BrainRes.

    2012;1481:49-58.

    55. HofferME, BalabanC, Slade MD, TsaoJW,

    HofferB. Amelioration of acutesequelaeof blastinduced mild traumatic braininjury by N-acetyl

    cysteine:a double-blind, placebo controlled study.

    PLoS One. 2013;8(1):e54163.

    56. Eakin K, Baratz-Goldstein R, Pick CG,et al.

    Efficacy of N-acetyl cysteine in traumatic brain

    injury. PLoSOne. 2014;9(4):e90617.

    57. GiacinoJT,Whyte J,BagiellaE, etal.

    Placebo-controlledtrial of amantadinefor severe

    traumatic braininjury. NEngl JMed. 2012;366(9):

    819-826.

    58. KimblerDE, Shields J,YanasakN, Vender JR,

    DhandapaniKM. Activation of P2X7promotes

    cerebral edema and neurologicalinjury after

    traumaticbraininjury inmice. PLoS One. 2012;7(7):

    e41229.

    59. WangX, Arcuino G,TakanoT,et al.P2X7

    receptor inhibitionimproves recovery afterspinal

    cord injury. NatMed. 2004;10(8):821-827.

    Clinical Review & Education ClinicalImplications of BasicNeuroscienceResearch TraumaticBrain Injury

    362 JAMANeurology March2015 Volume 72,Number 3 (Reprinted) jamaneurology.com

    Copyright 2015 American Medical Association. All rig hts reserved.