Progesterone and Vitamin D Hormone as a Biologic Treatment of Traumatic Brain Injury in the Aged

11
Biologics Supplement Progesterone and Vitamin D Hormone as a Biologic Treatment of Traumatic Brain Injury in the Aged Donald G. Stein, PhD, Milos M. Cekic, PhD Abstract: There is growing recognition that traumatic brain injury is a highly variable and complex systemic disorder that is refractory to therapies that target individual mechanisms. It is even more complex in elderly persons, in whom frailty, previous comorbidities, altered metabolism, and a long history of medication use are likely to complicate the secondary effects of brain trauma. Progesterone, one of the few neuroprotective agents that has shown promise for the treatment of acute brain injury, is now in national and international phase 3 multicenter trials. New findings show that vitamin D hormone (VDH) and VDH deficiency in the aging process (and across the developmental spectrum) may interact with progester- one and treatment for traumatic brain injury. In this article we review the use of progester- one and VDH as biologics-based therapies along with recent studies demonstrating that the combination of progesterone and VDH may promote better functional outcomes than either treatment independently. PM R 2011;3:S100-S110 INTRODUCTION Traumatic brain injury (TBI) is a very complex, contextual, and dynamic process [1,2]. From the initial insult to the persistent and highly destructive cascade of secondary damage to eventual reorganization and recovery [3,4], each phase of the injury involves a different set of processes that sometimes overlap. Furthermore, the molecular and physiological changes resulting from a trauma to the brain are not spatially limited to the locus of injury, and even a well-circumscribed cerebral insult can have distal effects throughout almost the entire central nervous system (CNS) [5]. These often-subtle alterations in brain chemistry and physiology can persist for years and are associated with complex and sometimes unpredictable behavioral alterations such as posttraumatic stress disorder, mood disorders, or increased mortality from the disability and health complications that can occur long after the initial injury. In addition to the complex injury process within the CNS, head trauma also has systemic and extraneuronal effects that need to be considered in the development of therapies for TBI. These processes, which most often are associated with systemic inflammation and multi-organ dysfunction, are the actual causes of death in most patients who have sustained a brain injury [6]. In a recent review, Masel and DeWitt [7] suggest that TBI “should be classified as the beginning of an ongoing, perhaps lifelong process that impacts multiple organ systems and may be disease causative and accelerative.” Thus TBI should be viewed not as an isolated or static insult but as an event that dynamically alters the systems in which it occurs. Age and age-associated systemic changes can have additional effects on both survival and recovery from severe injury. Although some aspects of the injury may be different, systemic changes in response to brain injury also are likely to occur in children with TBI or hypoxic damage. Such changes include alterations in hormonal and drug metabolism, nutritional status, immune function, and increased frailty, among others. Thus it is not a given that TBIs, or the putative treatments for TBIs, will necessarily behave in the same way in very young children or in older subjects as they do in young adults. This situation is a good example of a “contextual” effect. In this review we focus primarily on elderly subjects, but similar considerations may apply to pediatric TBIs. Elderly patients might require research D.G.S. Department of Emergency Medicine, Emory University School of Medicine, 1365 B Clifton Road NE, Suite 5100, Atlanta, GA 30322. Address correspondence to D.G.S.; e-mail: [email protected] Disclosure: 4A, royalties from the products of BHR Pharma related to some of the research described in this presentation; 7, BHR; 2A, consultant, BHR. The terms of this arrangement have been reviewed and ap- proved by Emory University, which receives the principal share of any royalties and benefits. In accordance with its conflict of interest policies, the University itself is not required to disclose any similar information; 8B, NIH grants R015R01NS048451 and 5R01HD61971-02; 7, gift in support of research from H. Allen and Company M.M.C. Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA Disclosure: nothing to disclose Portions of this article have been discussed previously in the article “Traumatic brain injury and aging: is a combination of progesterone and Vitamin D hormone a simple solution to a complex problem?” (Cekic M, Stein DG. Neu- rotherapeutics 2010;7:81-90) and are pre- sented with the permission of the authors and publisher. Disclosure Key can be found on the Table of Contents and at www.pmrjournal.org Submitted for publication October 29, 2010; accepted March 16, 2011. PM&R © 2011 by the American Academy of Physical Medicine and Rehabilitation 1934-1482/11/$36.00 Vol. 3, S100-S110, June 2011 Printed in U.S.A. DOI: 10.1016/j.pmrj.2011.03.010 S100

Transcript of Progesterone and Vitamin D Hormone as a Biologic Treatment of Traumatic Brain Injury in the Aged

ic

Biologics Supplement

Progesterone and Vitamin D Hormone as a BiologTreatment of Traumatic Brain Injury in the Aged

Donald G. Stein, PhD, Milos M. Cekic, PhD

iable andhanisms.s, alteredecondaryas shownal phase

eficiencyrogester-rogester-g that thean either

100-S110

ss [1,2].damage

diffeiolo

of inmoshemmetisordong

systapietionustahoulmule viein w

vivalsystehyptriti

iveny ins a gjects

y Medicine,ine, 1365 BAtlanta, GA

e to D.G.S.;

the productsome of theentation; 7,terms of thised and ap-ich receivesyalties ands conflict ofitself is notinformation;48451 and

support ofpany

cy Medicine,e, Atlanta, GA

n discussedc brain injuryprogesteronesolution to ain DG. Neu-nd are pre-authors and

the Table ofl.org

Abstract: There is growing recognition that traumatic brain injury is a highly varcomplex systemic disorder that is refractory to therapies that target individual mecIt is even more complex in elderly persons, in whom frailty, previous comorbiditiemetabolism, and a long history of medication use are likely to complicate the seffects of brain trauma. Progesterone, one of the few neuroprotective agents that hpromise for the treatment of acute brain injury, is now in national and internation3 multicenter trials. New findings show that vitamin D hormone (VDH) and VDH din the aging process (and across the developmental spectrum) may interact with pone and treatment for traumatic brain injury. In this article we review the use of pone and VDH as biologics-based therapies along with recent studies demonstratincombination of progesterone and VDH may promote better functional outcomes thtreatment independently.

PM R 2011;3:S

INTRODUCTION

Traumatic brain injury (TBI) is a very complex, contextual, and dynamic proceFrom the initial insult to the persistent and highly destructive cascade of secondaryto eventual reorganization and recovery [3,4], each phase of the injury involves aset of processes that sometimes overlap. Furthermore, the molecular and physchanges resulting from a trauma to the brain are not spatially limited to the locusand even a well-circumscribed cerebral insult can have distal effects throughout alentire central nervous system (CNS) [5]. These often-subtle alterations in brain cand physiology can persist for years and are associated with complex and sounpredictable behavioral alterations such as posttraumatic stress disorder, mood dor increased mortality from the disability and health complications that can occur lthe initial injury.

In addition to the complex injury process within the CNS, head trauma also hasand extraneuronal effects that need to be considered in the development of therTBI. These processes, which most often are associated with systemic inflammamulti-organ dysfunction, are the actual causes of death in most patients who have sa brain injury [6]. In a recent review, Masel and DeWitt [7] suggest that TBI “sclassified as the beginning of an ongoing, perhaps lifelong process that impactsorgan systems and may be disease causative and accelerative.” Thus TBI should bnot as an isolated or static insult but as an event that dynamically alters the systemsit occurs.

Age and age-associated systemic changes can have additional effects on both surrecovery from severe injury. Although some aspects of the injury may be different,changes in response to brain injury also are likely to occur in children with TBI ordamage. Such changes include alterations in hormonal and drug metabolism, nustatus, immune function, and increased frailty, among others. Thus it is not a gTBIs, or the putative treatments for TBIs, will necessarily behave in the same wayoung children or in older subjects as they do in young adults. This situation iexample of a “contextual” effect. In this review we focus primarily on elderly sub

similar considerations may apply to pediatric TBIs. Elderly patients might require rese

PM&R © 2011 by the American Ac1934-1482/11/$36.00

Printed in U.S.A.S100

rentgicaljury,t theistryimesers,

after

emics forandinedd betiplewedhich

andmicoxiconalthatveryood

, but

D.G.S. Department of EmergencEmory University School of MedicClifton Road NE, Suite 5100,30322. Address correspondence-mail: [email protected]: 4A, royalties fromof BHR Pharma related to sresearch described in this presBHR; 2A, consultant, BHR. Thearrangement have been reviewproved by Emory University, whthe principal share of any robenefits. In accordance with itinterest policies, the Universityrequired to disclose any similar8B, NIH grants R015R01NS05R01HD61971-02; 7, gift inresearch from H. Allen and Com

M.M.C. Department of EmergenEmory University School of MedicinDisclosure: nothing to disclose

Portions of this article have beepreviously in the article “Traumatiand aging: is a combination ofand Vitamin D hormone a simplecomplex problem?” (Cekic M, Sterotherapeutics 2010;7:81-90) asented with the permission of thepublisher.

Disclosure Key can be found onContents and at www.pmrjourna

archSubmitted for publication October 29, 2010;accepted March 16, 2011.

ademy of Physical Medicine and RehabilitationVol. 3, S100-S110, June 2011

DOI: 10.1016/j.pmrj.2011.03.010

geriatreatmthis a

proprienetich ansses susystemr comacolo

unseli“comis tont adetabo

thatbstanlthou

reatedevidenmmatogical

rognosmmatoion sy]. Thious a0,11].nal cauund ture of

oftem wto 47

logy che brrachnn failre orgaequeno beiologyis proines t

irectlyema, adamen

es haverculatione signif-ns and ain essen-systemicthe area

s to sys-erally ismatory

ase pro-ncurrentesponse.pposingsence ofnot justly a keyity [20].patients,a brain

ave a lifethat of

ch morerespira-tients inI may be3] exam-300 pa-tisticallyred withaada andlder pa-

such as(IL-1�),n, a sys-the liverimprovet consid-

mationse of theare up-othrom-in, high-tein C).rvival, inortion oferate thed to have

mationole-bodyiator of

S101PM&R Vol. 3, Iss. 6S, 2011

directed at the special nature and consequences ofbrain injury and a potentially different approach to tdesigned specifically to address the physiology ofgroup.

Although a monotherapy approach may be apfor the handful of diseases known to have specific genvironmental causes, experience shows that sucproach is inadequate for more heterogeneous proceas TBI. Here we suggest that because it is a complex,process, TBI can be addressed best by pleiotropic onation therapy (which, in addition to any pharmtreatment, can certainly include rehabilitation, coand other behavioral approaches contributing to thenation” aspect) and that any such therapy, if itsuccessful in human patients, must take into accoutional contextual factors such as age, gender, and mstatus.

TBI IS A MULTI-ORGAN/SYSTEMICINFLAMMATORY DISORDER

Accumulating evidence now favors the hypothesisflammatory processes throughout the body have a suimpact on morbidity and mortality after a TBI. Abrain and spinal cord injury generally have been tisolated and well-localized insults, experimentalincreasingly suggests that elevated levels of inflacytokines, a well-recognized aspect of the physiolosponse to trauma [8], are the most consistent pmarkers of outcomes in patients with systemic inflaresponse syndrome, sepsis, multi-organ dysfunctdrome (MODS), and multi-organ failure (MOF) [9factors usually cause death, and at the very least, seroften permanent disability after CNS trauma [5,6,1

In many cases, systemic failure seems to be the fiof death after CNS injury. For example, Zygun [6] fodysfunction developed in 89% of patients and failleast one non-neurological organ occurred in 35%tients. Failure of one non-neurological organ sysassociated with a 40% death rate, which increasedwith the failure of 2 systems. Multi-organ pathooccur as a consequence of neurologic injury to t[12,13]. In a study of patients with aneurismal subahemorrhages, mortality was 31% when one orga91% when 2 organs failed, and 100% when 3 or mofailed [14]. The non-neurological systems most fraffected in conjunction with CNS injury seem tcardiovascular and pulmonary systems. The key etthe development of dysfunction in these 2 systemsbly the massive release of cytokines and catecholamoccurs after neurologic injury, which can lead dmyocardial dysfunction [15], severe pulmonary edrespiratory failure [16]. Other mechanisms of fun

importance are inflammation, coagulation, and infection [

ricentge

ateor

ap-chic

bi-gicng,bi-bedi-lic

in-tialghascery

re-ticryn-

esend

sehat

atpa-as%an

ainoided,nstlythein

ba-hatto

ndtal

Significant elevations of inflammatory cytokinbeen observed in cerebrospinal fluid, the systemic ci[17,18], and even in the gut [19] after TBI. Given thicant blood flow to the brain under normal conditiodamaged blood�brain barrier after injury, the bratially acts as a filter for the blood, allowing thedissemination of inflammatory mediators away fromof local injury. This phenomenon eventually leadtemic inflammatory response syndrome, which gencharacterized by local and systemic release of inflamcytokines, complement, coagulation and acute phteins (APPs), and immune cell recruitment [10]. Cowith this process is a systemic anti-inflammatory rThe ability to strike a balance between these 2 oforces may determine a patient’s survival. The preinflammation at and near the site of injury doescontribute to local secondary damage—it is probabmechanism in TBI-associated mortality and morbid

For example, in a study of more than 2000Harrison-Felix et al [21] found that patients withinjury who are completing inpatient rehabilitation hexpectancy that is on average 7 years shorter thanpatients without TBI. These patients also are mulikely to die from septicemia, pneumonia, and othertory diseases for up to 1 year after injury than are pathe general population [22]. Even “mild” cases of TBrelated to a greater risk of mortality. Brown et al [2ined death rates in Minnesota for approximately 1tients with mild TBI and found a modest but stasignificant reduction in long-term survival compapatients who did not have a head injury; however, Flcolleagues [24] found a greater risk in adult and otients.

The systemic release of inflammatory cytokinestumor necrosis factor–� (TNF-�), interleukin 1�and especially IL-6 induces the acute phase reactiotemic response to inflammation mainly executed bythat resets a number of homeostatic set points todefense and adaptation [25]. These changes exhibierable variability, from maintaining persistent inflamto promoting adaptive changes. During the acute phaTBI (hours to several days), a number of APPsregulated (eg, C-reactive protein, fibrinogen, and prbin), whereas others are down-regulated (eg, albumdensity lipoproteins, antithrombin III, and proAlthough these changes are presumed to improve sucases of severe injury such as TBI, an increased proppositive to negative APPs has been shown to acceldevelopment of diffuse intravascular coagulation angenerally pathophysiologic effects [10]. Acute inflamalso has been observed to increase levels of whoxidative stress, which is another important med

6]. systemic injury [26].

redictll asll knoase af

ost prr ofwithsuggeer injun failudeve

icatedponsee undr pathfectedpatienrbiditi

rs ins, TBIfact tsts brperso

ncreast decaresulteat bedictorns its peolizatio[37];BI, ranoccur

rain wlevelneuro

chemiffectsangesing cencreasyribonons a

hondrialn pumpicated inprocess

interest-xidativesubjectsan their

ges, thepatternsmation

not onlygenerateon [48],ng.” Al-ited un-inactive

erly sub-ulation

is situa-[48] andned out-re severe

ation

anges inonly oc-ses asso-d with ahich, be-

munitythe onseto mount

ulationof anti-with a

amountsd mono-

with in-lthough

ts withinry cyto-

lution ofoinflam-disrupt

ronmentreducedpears to

S102 Stein and Cekic BRAIN INJURY, AGING, AND NEUROSTEROIDS

Inflammatory biomarkers also may be useful in pthe severity and outcome of injury [9,27], as weprognosis for recovery [28-31]. Although it is wethat levels of the cytokines TNF-� and IL-1� incresevere injury, the level of IL-6 appears to be the mnostically accurate because it is the chief regulatohepatic acute phase reaction [25] and correlatesdegree of systemic inflammation [9]. Evidence alsothat an increased level of IL-6 in the early period aftis a marker of a high risk of complication and orga[32]. Although considerable effort is being made toprognostic biomarkers for TBI, the issue is complthe fact that so many systemic changes occur in resbrain injury that no single biomarker fully reflects thlying pathology [33]. As with inflammation and otheogies, systemic biomarker activity may be further afthe time when tissue or serum samples are taken, thestress levels, previous health status, other comonutritional history, drug use, gender, and age.

AGING, INFLAMMATION, AND BRAININJURY

Because of the focus on youth-related TBI that occusetting of sports, war, and motor vehicle accidentelderly persons receives less attention, despite thethe Centers for Disease Control and Prevention liinjury as a major cause of morbidity and mortality inolder than 70 years [34]. The incidence of TBI has iby 21% in people older than 65 years during the paseven as it decreased for most other age groups as aimproved primary prevention such as the use of sand safety helmets [35]. Age is an independent premortality resulting from TBI, and in elderly persoceeds that of younger victims by a factor of 2 [36]. Areach their mid to late 70s, the frequency of hospitaattributable to TBI has its most pronounced peakgreatest mortality rates from “moderate” to severe Ting from 60.9% to 86.8% (depending on the study),people 75 to 80 years of age [38].

Effects of Aging in the CNS

Although no overt neuronal loss is observed in the bnormal aging, a variety of changes occur at themedium-scale neuronal networks and individualthat entail more subtle metabolic, structural, andalterations [39,40], all of which have significant eneuronal plasticity and adaptation. Age-related chthe CNS are similar to those that occur in other agand include dysfunction of energy homeostasis, ioxidative damage, protein accumulation, and deoxcleic acid (DNA) damage [39]. As they age, neur

become more susceptible to damage caused by glutam

ingthewnterog-thethestsryre

lopbyto

er-ol-byt’ses,

thein

hatainnsedde,ofltsof

ex-plens

theg-in

ithofnscaloninllsedu-

lso

excitotoxicity [39] because of impairment of mitocmetabolism [41], Ca2� homeostasis [39], and iofunction [42]. Because these processes also are implthe development of injury after trauma, the agingitself is likely to exacerbate the effects of TBI. In oneing report, Wagner and colleagues [43] suggest that odamage loads increase with age but that femaleappear to fare better and have less oxidative stress thmale counterparts.

In addition to these structural and intrinsic chanbrains of aged animals also exhibit gene expressioncharacterized by an increased level of neuroinflamand microglial activation [44-47]. The microglia aremore stimulated in older animals but also appear toan amplified response to inflammatory stimulatiresulting in a general neuroinflammatory “primithough the duration of inflammatory changes is limder normal conditions as the microglia return to anstate upon resolution of immune challenges, in eldjects, the microglia are so sensitive to immune stimthat their activation resolves slowly or not at all. Thtion can result in exacerbation of neurologic diseasestill higher levels of neuroinflammation, with worsecomes in older subjects after TBI secondary to a moand persistent systemic immune response [49].

Effects of Aging on Systemic Inflammand Endocrine Function

In addition to CNS-specific changes, age-related chthe function of the systemic immune system commcur and are implicated in virtually all disease procesciated with advanced age [50]. Aging is associategeneral activation of the inflammatory response, wcause of the chronic antigenic stress on innate imexperienced during a lifetime, becomes the basis forof inflammatory diseases [51] and a reduced ability tan appropriate immune response to antigenic stim[50]. With aging, a decrease in the productioninflammatory hormones also occurs [52], alonggeneral tendency toward production of elevatedof proinflammatory cytokines by peripheral bloonuclear cells [53].

The fulminating inflammatory state associatedcreasing age has been dubbed “inflammaging” [53]. Athis condition is systemic, it also has specific effecthe CNS [48]. Given the importance of inflammatokines in both behavioral modulation and the evotraumatic injury, it is likely that an increased neurmatory cytokine response in elderly patients canneuronal synaptic plasticity, establishing a CNS envipredisposed to long-lasting complications and aability to recover from trauma [48]. The aged brain ap

ate exist in a chronic state of inflammation that is associated with

evel p

ng fraflammecess

examiones tbilitat

d in hr of k[55],ulin-lroxyvicreasee imm

gests tphys

g insul. TNF, grow] and cconcmanrone sil elden seruElevadevelon asso

f cont1 billiencyssocianditio

sion a], cane nor

nstratheal

d preme fr-100%men aare se

ny agmponence t

[69,73].immuney be anormone

tment int such as

nt drugshuman

e essen-gic treat-growingnt phaseindicatet benefi-

tion hasst papersand TBIdopreg-bilateralernationendoge-

by cervi-memory

heir nor-amined,ially lesswas re-proges-ral daysesteronejury andoner the

ffective-12], andels from

orted itshologic,

] concludesenjoy good

in D benefitsdepression,

oke and TBI.ubgroups—e dark skin(p. 3). The

S103PM&R Vol. 3, Iss. 6S, 2011

increased immune reactivity and continuous low-lduction of central inflammatory cytokines [54].

Given the apparent correlation between increasiin the elderly and the increase in a chronic level of intory factors in the brain and other organs, it may be nfor the physician, as part of the patient’s physicaltion, to test for levels of these factors and the hormmay be needed to offset the potential for further dein the absence of immediate overt symptoms.

A final factor that must be specifically considereman aging is the decreased activity of a numbesystemic hormones, including thyroid hormonesteroids [56-58], growth hormone (GH) [59], insgrowth factor-I (IGF-I) [59], and possibly 25-hydmin D3 [60-62]. It is important to note that this deendocrine function appears to be associated with thnologic changes discussed previously. Research sugproinflammatory cytokines may down-regulate thelogical responses to a variety of hormones, includinGH, IGF-I, thyroid hormone, and estrogens [63]IL-1�, and IL-6 can modulate the release of GHhormone-releasing hormone, and somatostatin [64reduce concentrations of IGF-I [65] and increase thetrations of glucocorticoids [66] in the serum of hutients. Lower serum IGF-I and dehydroepiandrostefate levels were found in frail compared with nonfraindividuals [67], and an inverse correlation betweeIL-6 and IGF-I was noted only in frail individuals.levels of inflammatory cytokines [68-71] and thement of frailty in older adults [72,73] also have beeated with VDH deficiency.

Aging and VDH Deficiency

Recently VDH deficiency has received quite a bit oversial media attention, partly because an estimatedpeople worldwide are said to exhibit VDH deficinsufficiency [74] and partly because it has been awith autoimmune, inflammatory, and infectious co[75-81], cardiovascular disease [82,83], hypertenatherosclerosis [84], neuromuscular function [85[86], and neurodegenerative diseases [87,88]. In theastern United States, for example, studies demoprevalence of VDH deficiency ranging from 32% inadults [89] to approximately 50% for adolescents anolescents [90-92]. The most dramatic statistics costudies in the elderly, which demonstrate that 40%community-dwelling American and European olderwomen are deficient in VDH. Even higher averagesin the ill and institutionalized patients [93-101].

VDH deficiency appears to be correlated with mafactors, especially those with an inflammatory co[76,77]. This finding is supported by further evid

low levels of serum VDH are associated with elevated levels

ro-

iltya-

aryna-hation

u-ey

sexiketa-inu-

hatio-in,-�,than

en-pa-ul-rlym

tedp-ci-

ro-ionor

tednsndcerth-e athyad-omof

nden

ingenthat

IL-6 [102] and a key marker of frailty in older adultsVDH is known to be a modulator of the cell cycle,function, and calcium homeostasis. As such it maimportant compound not only as an endogenous hbut also in the context of CNS injury, both as a treaits own right and in combination with another agenprogesterone [103].*

PROGESTERONE AS A TREATMENT FORBRAIN INJURY

During the past 30 years, some 50 candidate treatmefor TBI neuroprotection have failed when tested inpatients, and many pharmaceutical companies havtially discontinued the pursuit of acute pharmacoloment for TBI. Despite this pessimism, a large andpreclinical literature, along with the results of 2 rece2 clinical trials [104,105] and a new phase 3 trial,that the hormone progesterone may exert significancial effects in the treatment of TBI.

Preclinical work on progesterone neuroprotecbeen ongoing for more than 20 years. One of the firto demonstrate a beneficial effect of progesteronecame from our laboratory [106]. We tested “pseunant” and normally cycling female rats with largelesions of the frontal cortex on a delayed spatial altlearning task. The pseudopregnant females, whosenous hormone levels were elevated experimentallycal stimulation, performed significantly better inretention of the trained task after injury than did tmal-cycling counterparts. When the brains were exthe pseudopregnant rats also demonstrated substantventriculomegaly, suggesting that traumatic edemaduced by the relatively greater levels of endogenousterone present at the time of surgery and for sevethereafter [107]. We also showed that, in rats, progcan be given up to 24 hours after bilateral cortical instill retain its neuroprotective effects, although the sotreatment is given, the better the outcome [108].

Many publications have now demonstrated the eness of progesterone treatment for TBI [107,109-1more than 170 papers using 22 different injury mod25 laboratories around the world have now repbeneficial effects in attenuating the cytologic, morp

* A recent, apparently controversial report by the Institute of Medicine [103that dietary intake of VDH and calcium is adequate for “most people . . . tobone health.” The report challenges many popular assumptions about vitamin a number of disease conditions including autism, cognitive function, andbut for unexplained reasons ignores the potential role of VDH in cerebral strHowever, the authors do take care to note that “Nonetheless, some sparticularly those who are older and living in institutions or who havpigmentation—may be at increased risk for getting too little vitamin D”

ofauthors also point out that “no central authority has determined which cut-points to use”(in determining sufficiency or deficiency).

n, spi3-128esizede activreceptessionoth va

ate toy antobutyage, ation acell s

nismn diff

ly ande it m

forsystemleast

neuroly withikely

ficacymanble-blite-to-f Neuxaminhours

ion Glr eversignedintra

olutiorogestlogic ae inju

rone w4%), apatie

p. Mem-d by thes adverse

indepen-in whichreatmentPatients

re of 8 orr proges-Glasgowr the in-ndepen-a mod-monthseasures30 days

s adversereportedays after

AL

04,105],y NIH isd States.will en-

hours ofCS scoreof 0.714e or pla-

ours andurs. Theize pro-nts withistration

s grantedstratifiedk mortal-

isabilitynal out-v (http://

f Besinsaceuticala secondpse-trial.sed-headinitiatedthroughtern Eu-

studiesave repoent revns, manterone, fas. Howeauthors

ria stipultudies uuroprotecriteriabrain re

S104 Stein and Cekic BRAIN INJURY, AGING, AND NEUROSTEROIDS

and functional deficits caused by injury to the braicord, and peripheral nerves [107,113-118, 121, 12

Unlike other sex steroids, progesterone is syntholigodendrocytes in the brain, where it modulates thof classical nuclear, membrane-bound, and steroid[123]. Progesterone also can directly affect the exprwater channel proteins (aquaporins) to modulate bgenic and cytotoxic edema [121,124], reduce glutamicity directly, mediate toxic cellular calcium influx bonizing sigma receptors, up-regulate gamma-aminacid A (GABAA) receptors to reduce excitotoxic damreduce apoptosis and necrosis through the up-reguladown-regulation of a variety of genes that controlvival [112,123].

In contrast to the single-receptor, single-mechaproach, we believe it is progesterone’s ability to act oent components of the injury cascade simultaneousexert differential effects over time and place that makeffective than other agents that have been studiedtreatment of TBI. We have shown that, when givencally, progesterone can affect the expression of atgenes thought to play a role in neuroprotection andpair [110]. This pleiotropy, although not fitting neatthe molecular biological paradigm of disease, is lreason for its translational success thus far.

The preclinical data on the neuroprotective efprogesterone after TBI have been confirmed in hutients in a phase 2a, single-center, randomized, douclinical trial for progesterone treatment of moderavere adult TBI sponsored by the National Institute ological Disorders and Stroke [104]. The authors e100 patients enrolled with proxy consent within 11injury and included only those with a postresuscitatgow Coma Scale (GCS) score between 4 and 12. Fopatients assigned to the treatment group, one was asthe placebo control group, and all patients receivednous administration of the test drug or placebo sProxy consent requirements for this study delayed pone treatment for at least 6 hours after injury. Neurofunctional outcomes were evaluated 30 days after thMortality among patients who were given progesteless than half that of control subjects (13.6% vs 30.30-day functional outcomes for moderately injured

† Despite the growing evidence of progesterone’s neuroprotective effects, 2laboratories examining the role of cyclosporin in the treatment of TBI hnegative outcomes [119,120]. In addition, the authors of 2 rec[121a,121b] suggest that, by the authors’ personal, operational definitionot all, preclinical studies testing neuroprotective agents, including progesmeet a number of their criteria for preclinical development of new drugdespite their concerns, given the large number of studies in which theprogesterone, it may be of interest to note that most, if not all, of the criteby Loane and Faden and Gibson et al have been addressed for the sprogesterone, but perhaps to a lesser extent than for most of the other neagents cited in the paper by Loane and Faden. However, even if all theindeed met, no guarantee exists that a completely successful agent for

would be discovered. The review presents a number of important suggestions fodesign of studies in neuroprotection.

nal].†

byityorsof

so-x-

ag-ricndndur-

ap-er-to

orethe

i-90re-in

the

ofpa-ndse-ro-edof

as-y 4to

ve-ns.er-ndry.as

ndnts

were significantly better than for the placebo groubers of the Data Safety Monitoring Board, appointeNational Institutes of Health (NIH), found no seriouevents attributable to the progesterone treatment.

These initial results were confirmed by a seconddent, randomized, double-blind study from Chinathe authors used a 5-day intramuscular course of twith progesterone in 159 patients with severe TBI.who arrived within 8 hours of injury with a GCS scolower were randomly assigned (1:1) to receive eitheterone or placebo. The primary end point wasOutcome Scale (GOS) measurement 3 months aftejury, but the investigators also used Functional Idence Measure scores and repeated the measures onified Functional Independence Measure and GOS 6after the TBI occurred. Similar beneficial outcome mon morbidity and mortality were observed at bothand 6 months after injury, again without any seriouevents caused by the treatment. The authors alsothat intracranial pressure was lower 72 hours and 7 dtreatment with progesterone [105].

PROGESTERONE IN A PHASE 3 CLINICTRIAL

Following up on the positive phase 2 clinical data [1a phase 3, 17-center clinical trial for TBI sponsored bnow currently enrolling patients across the UniteThis 1:1 randomized, double-blind controlled trialroll 1200 patients. Treatment must begin within 4injury in patients at least 18 years of age with a Gbetween 12 and 4. After a 1-hour loading dosemg/kg administered intravenously (IV), progesteroncebo is given by IV infusion at 0.5 mg/kg for 72 hthen tapered over the course of an additional 24 ho4-hour window of treatment was designed to optimgesterone’s neuroprotective effects by treating patieTBI as soon as possible. Food and Drug Adminapproval for exemption from informed consent wafor this purpose. The primary study end point will be adichotomy of the GOS at 6 months. The trial will tracity, extent of adverse and serious adverse events, DRating Score, and cognitive, neurologic, and functiocomes. More details can be found at ClinicalTrials.goclinicaltrials.gov/ct2/show/record/NCT00822900).

In addition to the NIH study, BHR, a branch oPharma, a privately held Belgian/French pharmcompany, also recently began enrolling patients inclinical trial called SyNAPSe (http://www.synacom/). Approximately 1800 patients with severe cloTBI (GCS scores of 4-8) in whom treatment can bewithin 8 hours of injury will be enrolled in the study100-120 medical centers in the United States, Wes

fromrted

iewsy, ifil tover,useatedsingctivewerepair

rope, Israel, and other countries. Patients are randomly as-r the

f progw-up

tudy eial ofis be

in Injutium.

ratesprog

tiallyeby piscusse actbilitysystemrvivalppor

ontusiessionntestiesterontestijury cwhichn repatritiolismvich a).

ING

e preled to

e in agld VDtentia

nt inprog

ne inestero

hat VDeasehat coutcom

progngnifictory phavior

in ag

ged

old rats,ls [128].ll death,y (24-72“humanted withsurgery

e pointsmatory

We alsooints, asunction,esteronetreating

eficient,ith VDHd with orlts con-hat sug-line level

ing theBI. Thisased cellcondaryeatment.by VDH

teractiont of thee level offact thating IL-6

mationvident inl animalsst of thedeficienturs afterprimaryH defi-

e area oflogic ap-IDS andindivid-hanismsy not bembining

S105PM&R Vol. 3, Iss. 6S, 2011

signed to receive a 5-day continuous IV infusion oterone or placebo in a 1:1 allocation, and folloconducted for 6 months. The GOS is the primary spoint. This global, phase 3, multicenter pivotal trprogesterone infusion for patients with severe TBIconducted in collaboration with the American BraConsortium and the European Brain Injury Consor

We suggest that one of the reasons for increasedsurvival in patients with TBI who are treated withterone may be progesterone’s ability to substanduce systemic acute phase inflammation and thervent MODS and MOF [122,125]. Because, as dpreviously, MODS and MOF frequently are thcauses of death in patients with a TBI, the aprogesterone to modulate the development ofinflammation may be one way that it promotes suthe early stages after brain injury. This idea was suby Chen and colleagues [126], who found that cinjury to the cerebral cortex in rats induced exprIL-1� and TNF-� followed by apoptosis in the imucosa. A 5-day course of treatment with progreduced both the cytokines and cell death in the iThis study shows that an experimental brain intrigger a cascade of inflammation in other tissues,turn can produce serious consequences for braiFor example, gut inflammation could affect nuvariables needed to help with the CNS metabovolved in neuroprotection (see the article by PopoMcTigue [5] for a further discussion of this issue

PROGESTERONE, VITAMIN D, AND AGIN BRAIN INJURY

Progesterone’s effectiveness in treating TBI and thlence of VDH deficiency in the elderly populationquestions: First, would progesterone be as effectivanimals as in young adult animals? Second, woudeficiency affect the outcome of a brain injury and pointerfere with the benefits of progesterone treatmerats? And finally, would a combination of VDH andterone be more effective than either compound alosame aged rat population? We speculated that progwould be effective in treating TBI in old rats but tdeficiency would increase inflammation and decrbenefits of treatment [127]. We also hypothesized tbining VDH with progesterone would improve oBecause the early onset of inflammation is a reliabletic indicator of mortality in human patients with sitrauma, we measured a number of acute inflammateins, cell death, DNA damage, and short-term beindicators of inflammation and secondary damage

and VDH-deficient aged animals after TBI.

es-is

ndIV

ingry

ofes-re-re-ed

ualoficin

tedonof

nalne

ne.aninir.

nalin-nd

va-3

edH

llyoldes-theneH

them-e.

os-antro-ased

Progesterone and VDH after TBI in ARats

Progesterone was indeed effective in treating TBI inalthough at a higher dose than in young adult animaWe measured levels of inflammatory proteins, ceedema, and behavior during the acute phase of injurhours after TBI) in 20-month-old rats (which is theequivalent” of 60 years of age). Injured animals treaprogesterone beginning within the first hour aftershowed decreased inflammatory markers at all timexamined, indicating a reduction in the acute inflamprocess compared with the old rats given vehicle.observed decreased markers of cell death at all time pwell as decreased edema and improved motor fwhich suggests that the pleiotropic effects of progwere also effective despite the added complexity ofan aged organism.

When we tested aged rats that were made VDH dwe observed increased inflammation compared wreplete animals in all groups, with or without TBI anwithout progesterone treatment [127]. Our resufirmed findings of previous studies [76,77,129] tgested that VDH deficiency establishes a higher baseof inflammation even before injury, in effect primsystem for an increased immune response after Televated acute-phase response correlated with incredeath and DNA damage, indicating a more severe seinjury process after injury even with progesterone tr

Both TNF-� and IL-6 were significantly increaseddeficiency, and both cytokines demonstrated an inbetween deficiency and treatment. Although mosvariability in TNF-� was accounted for by injury, thIL-6 was primarily affected by VDH deficiency, acorresponds to other independent data connectlevels with VDH deficiency, frailty, and inflam[73,102,130,131]. The elevation in IL-6 was most eour data comparing VDH-deficient and VDH-normaafter TBI and progesterone treatment. Although moother cytokines were elevated 2- or 3-fold in VDH-animals, IL-6 was increased nearly 5-fold by 72 hothe injury occurred, suggesting that IL-6 may be thecytokine involved in the detrimental effects of VDciency after TBI.

Combination Therapy in TBI

Combination therapy is a relatively novel idea in thbrain injury, but it is a well-established pharmacoproach [4] in other disease processes such as HIV/Atuberculosis. Most drug development still focuses onual agents and is targeted toward a few specific mecat most. As discussed previously, this approach maappropriate in the setting of TBI. Rationales for co

drugs include targeting multiple divergent mechanisms and

kinetiThis idBI [131) dru, wherouslyed tome effis casetmentto ovinatort mayf huml monin injueffect

TBI

ciencyDH desteroeiotroeceptoion eve criteesterooundstion, bTBI,

h a br

act nohe sas progtors a, VDHhe devRamar bindH. Thsites

30 gento VDly impponse

anti-er tissumpanin be

reducesin otherare also

DH ad-rotectivedifferentrogester-esized innts that

own dis-es, takenery goodesting ofstudy a

ical trial,

planningments. Atric TBIAppliede shape.an havetion mayffective-rone for

ged

nation ofodels inas effec-cortical

hallenge.roducedbinationore neu-

at their-inducedecause itd spinal

ent con-bilateralent thatbinationse) com-ne. Thesubstan-nditionscould be

S106 Stein and Cekic BRAIN INJURY, AGING, AND NEUROSTEROIDS

overcoming single-drug limitations such as receptorpharmacology, and signaling pathways [132,133].has recently gained ground in the treatment of T“Polytherapy” approaches can proceed in 2 ways: (can be designed for target promiscuity or pleiotropysingle agent affects multiple pathways simultanesequentially or both; or (2) drugs can be designadministered in combination, which achieves the saof targeting multiple processes, but separately in thhybrid approach, which may be wise for the treaTBI, is to combine pleiotropic drugs themselvescome the limitations of monotherapy. A combapproach to treatment is not only reasonable buessential given the complexity and heterogeneity oTBI and the fact that, of the various preclinicatherapy drugs that have shown promise for braneuroprotection, all have been shown to be inwhen taken to clinical trial [134].

Combining Progesterone and VDH in

Given our results with progesterone and VDH defiaged rats, we speculated that acute correction of Vciency with VDH would restore the efficacy of progCombining progesterone and VDH, which are pldrugs that work primarily through intracellular rtranscription factors, also merits serious consideratoutside the context of VDH-deficiency as meeting thfor combinatorial therapies [4]. Because both progand VDH are pleiotropic and multifunctional compis difficult to predict specific mechanisms of interacboth agents could be used to target cell death afterexample, as one of the many mechanisms by whicinjury evolves.

It is worth emphasizing here that VDH is in fvitamin but rather a secosteroid hormone with tcholesterol backbone as other sex steroids (such aterone) and has its own class of nuclear steroid recepsignaling mechanisms [135,136]. Like progesteronea pleiotropic agent with important implications for topment and treatment of systemic disease. Recentlypalan and colleagues [137] examined VDH receptoin lymphoblastoid cells after treatment with VDfound more than 2700 gene positions with VDHsignaling gene transcription and approximately 2that changed their level of expression in responseMany of these genes (like progesterone) are directcated in modulating the immune/inflammatory resinjury and neurodegenerative diseases.

Importantly, progesterone complements VDH’sflammatory effects not only in the brain but in othaffected by the inflammatory cascade that often accoTBI or stroke. To reiterate, one of the hormone’s ma

fits and probably one of the main reasons it is so effective

cs,ea4].gse aorbeect. Aof

er-ialbeano-ry

ive

inefi-ne.picrs/enriane, itut

forain

t amees-ndis

el-go-ingeyforesH.li-to

in-es

iesne-

a systemic treatment for serious injury is because itthe inflammatory cascade not only in the brain buttissues such as gut, heart, spleen, and thymus thatsubject to cytokine damage [138-143].

Evidence also suggests that, like progesterone, Vministered after a brain injury can have neuropeffects, although a number of its actions involvepathways from those of progesterone [144]. Both pone and VDH are natural hormones that are synthboth men and women, and both can act as ageproduce other metabolites in the brain with theirtinct modes of action in CNS repair. These propertiin the context of the clinical trial results showing vprogesterone safety and efficacy in TBI, makes the tprogesterone and VDH in a preclinical combinedcompelling approach to consider for use in a clinespecially for elderly persons.

Perhaps similar consideration should be given tofor vitamin D supplementation in pediatric TBI treatsafety and efficacy trial for progesterone in pediaunder the auspices of the Pediatric Emergency CareResearch Network (PECARN) is beginning to takGrowing evidence suggests that many children cVDH insufficiency or even deficiency, and this condiaffect long-term disease outcome and possibly the eness of neuroprotective treatments such as progesteearly brain injury [145,146].

Progesterone/VDH Combination in ARats with TBI

We have demonstrated the effectiveness of a combiprogesterone and VDH in both in vitro and in vivo mour laboratory. Combining VDH and progesterone wtive in vitro in increasing the survival of primaryneurons under 24 hours of excitatory glutamate cAlthough VDH alone and progesterone alone both pbeneficial effects on neuronal cell survival, the comof VDH and progesterone produced significantly mroprotection than either compound given alone andbest individual doses [147]. We used glutamateexcitotoxicity as our primary outcome measure breliably triggers cell death in models of TBI, stroke, ancord injury.

Interestingly, when we applied the same treatmcept in vivo to VDH-deficient aged rats with largecontusions of the frontal cortex, the only treatmreduced inflammatory marker proteins was the comof progesterone and VDH (5 mg/kg in a single dopared with vehicle or either compound given alocombination treatment was also the only one thattially improved behavioral outcomes. If these coapply to human patients, the clinical implications

as important: a combination treatment of progesterone and

ould ialoneernatiasona

teroneicator

in injution aaged rouldors; arever

e infla

cationD inj

ve mahat shen inars towe uitamine expetaken

can, oenefitsent fu

c inflaesteroy whould sients.nt wegly clcountents pdults,nt willtic evee wit

s strotaboliconc

apy wdysfupriate)netwonctio

gesterone:pair. Phar-

Traumaticve. J Head

eurologicalil 2005;20:

eurothera-

m: Beware736-737.itical care:n Crit Care

ess, not an

Pape HC.y total carein severely-454.eitzman A.ters. Injury

y 2005;36:

on-neuro-t Care Med

quences of3:877-884.al systemsImmuno-

the brain.

ysfunctionand patho-

ac sequelae? Intensive

ranscranialacute brain

Takaku A.kines, andwith sub-5.s traumaticvation andrs Inflamm

: A clinical

M, Jha A.jury model

M, Jha A.duals with

S107PM&R Vol. 3, Iss. 6S, 2011

VDH administered to elderly patients with TBI shprove survival compared with progesterone giventhe same population. Because few, if any, better altexist, a clinical investigation would appear to be a restep to take.

Briefly summarized, we believe that: (1) progeseffective in reducing acute inflammation, a key indsurvival in human patients and in aged rats with bra(2) D deficiency increases acute-phase inflammaattenuates the benefits of progesterone treatment inwith TBI, which suggests that such a deficiency ccrease mortality and worsen outcome in TBI surviv(3) a combination of progesterone and VDH partiallythe effects of D deficiency and reduces post-TBI acutmation in old rats.

CONCLUSION

Should our results translate to the clinic, the implithe research discussed here is that a simple vitamintion combined with progesterone could help sahuman lives. Extrapolating from the animal data tthat D deficiency enhances inflammatory activity evintact rats, and because vitamin D insufficiency appeprevalent in so many elderly and frail patients,rehabilitation physicians to screen routinely for vlevels in their patients, especially those who havenced a stroke or TBI. Our animal data also can besuggest that ignoring D deficiency or insufficiencytime, exacerbate the injury, probably reduce the bphysical and cognitive therapy, and delay or prevtional recovery.

As we noted previously, the reduction in systemimation resulting from combined VDH and progtreatment may be one of the key mechanisms bprogesterone and VDH, in combination or alone, cnificantly increase survival after TBI in human patthough eventually better alternatives to the treatmeproposing may be found, it is becoming increasinthat no single receptor, gene, or mechanism can acthe complex cascade of focal and systemic toxic evduced by a brain injury, whether it is in children, aelderly persons. It is highly unlikely that a single ageable to trigger all the neuroprotective and neuroplasrequired for the damaged brain to reorganize to coptraumatic injury, degenerative disorder, or seriouDespite the evidence that progesterone and its memight work well, their effectiveness has not yet beensively demonstrated. Individual or combination therpleiotropic agents capable of addressing multiorgantion is becoming more acceptable (and more approwe begin to understand the complex systems andinvolved in the mediation of brain repair and fu

recovery.

m-to

vesble

isof

ry;ndatsin-ndsesm-

ofec-nyowoldbe

rgeD

ri-to

verof

nc-

m-neichig-Al-areearforro-orbentsh ake.teslu-ithnc-as

rksnal

REFERENCES1. Schumacher M, Guennoun R, Stein DG, De Nicola AF. Pro

Therapeutic opportunities for neuroprotection and myelin remacol Ther 2007;116:77-106.

2. Thurman DJ, Alverson C, Dunn KA, Guerrero J, Sniezek JE.brain injury in the United States: A public health perspectiTrauma Rehabil 1999;14:602-615.

3. Povlishock JT, Katz DI. Update of neuropathology and nrecovery after traumatic brain injury. J Head Trauma Rehab76-94.

4. Vink R, Nimmo AJ. Multifunctional drugs for head injury. Npeutics 2009;6:28-42.

5. Popovich P, McTigue D. Damage control in the nervous systethe immune system in spinal cord injury. Nat Med 2009;15:

6. Zygun D. Non-neurological organ dysfunction in neurocrImpact on outcome and etiological considerations. Curr Opi2005;11:139-143.

7. Masel BE, DeWitt DS. Traumatic brain injury: A disease procevent. J Neurotrauma 2010;27:1529-1540.

8. Harwood PJ, Giannoudis PV, van Griensven M, Krettek C,Alterations in the systemic inflammatory response after earland damage control procedures for femoral shaft fractureinjured patients. J Trauma 2005;58:446-452; discussion 452

9. Pape HC, Tsukamoto T, Kobbe P, Tarkin I, Katsoulis S, PAssessment of the clinical course with inflammatory parame2007;38:1358-1364.

10. Keel M, Trentz O. Pathophysiology of polytrauma. Injur691-709.

11. Zygun DA, Kortbeek JB, Fick GH, Laupland KB, Doig CJ. Nlogic organ dysfunction in severe traumatic brain injury. Cri2005;33:654-660.

12. Catania A, Lonati C, Sordi A, Gatti S. Detrimental consebrain injury on peripheral cells. Brain Behav Immun 2009;2

13. Vodovotz Y, Constantine G, Faeder J, et al. Translationapproaches to the biology of inflammation and healing.pharmacol Immunotoxicol 2010;32:181-195.

14. Gruber CJ, Huber JC. Differential effects of progestins onMaturitas 2003;46(Suppl 1):S71-S75.

15. Dujardin KS, McCully RB. Wijdicks EF, et al. Myocardial dassociated with brain death: Clinical, echocardiographic,logic features. J Heart Lung Transplant 2001;20:350-357.

16. Macmillan CS, Grant IS, Andrews PJ. Pulmonary and cardiof subarachnoid haemorrhage: Time for active managementCare Med 2002;28:1012-1023.

17. McKeating EG, Andrews PJ, Signorini DF, Mascia L. Tcytokine gradients in patients requiring intensive care afterinjury. Br J Anaesth 1997;78:520-523.

18. Hirashima Y, Nakamura S, Endo S, Kuwayama N, Naruse Y,Elevation of platelet activating factor, inflammatory cytocoagulation factors in the internal jugular vein of patientsarachnoid hemorrhage. Neurochem Res 1997;22:1249-125

19. Chen G, Shi J, Ding Y, Yin H, Hang C. Progesterone preventbrain injury-induced intestinal nuclear factor kappa B actiproinflammatory cytokines expression in male rats. Mediato2007;2007:93431.

20. Lim HB, Smith MA. Systemic complications after head injuryreview. Anaesthesia 2007;62:474-482.

21. Harrison-Felix C, Whiteneck G, DeVivo M, Hammond FMortality following rehabilitation in the traumatic brain insystems of care. NeuroRehabilitation 2004;19:45-54.

22. Harrison-Felix C, Whiteneck G, DeVivo M, Hammond FCauses of death following 1 year postinjury among indivi

traumatic brain injury. J Head Trauma Rehabil 2006;21:22-33.

Larsonation-ba

rtality aof age

ystemic.losed hll reduc

, Scaleassion inuma 20

T. Backy respoa 2001

mic infltly pred00;49:6

ators ofn failurdiscuss

dy infl1988;1

ucknering m

d its apnn Neu

njury inand de

braininj

eillance8. MM

atic by outco

e traumic persp

, de la Chir (W

ty. Nat

L, HofAging

ns. Tre

: Abundative di

hanek

t of gender,

ammationthe periph-

atkins LR,-inflamma-:717-722.

n of micro-

class II in-biol Aging

lifetime of521-538.ation after

erstandingmunologic

etworks in2007;128:

hormonesunosenes-

nti-inflam-erged from

nd the dis-peripheral

-pituitary-

incidencemates frometab 2004;

d estrogen4;39:1603-

stradiol in

io M, ArvatEndocrinol

gical func-etab 2008;

evention ofAm J Clin

ngiotensin19-426.alnik JM. A3.arabedian

in-inducedpregulatedNeuropsy-

llanua MA,tuitary GHts. J Endo-

S108 Stein and Cekic BRAIN INJURY, AGING, AND NEUROSTEROIDS

23. Brown AW, Leibson CL, Malec JF, Perkins PK, Diehl NN,Long-term survival after traumatic brain injury: A populanalysis. NeuroRehabilitation 2004;19:37-43.

24. Flaada JT, Leibson CL, Mandrekar J, et al. Relative risk of motraumatic brain injury: A population-based study of the roleinjury severity. J Neurotrauma 2007;24:435-445.

25. Gabay C, Kushner I. Acute-phase proteins and other ssponses to inflammation. N Engl J Med 1999;340:448-454

26. Shohami E, Gati I, Beit-Yannai E, Trembovler V, Kohen R. Cinjury in the rat induces whole body oxidative stress: Overaantioxidant profile. J Neurotrauma 1999;16:365-376.

27. Bochicchio GV, Napolitano LM, Joshi M, McCarter RJ JrSystemic inflammatory response syndrome score at admipendently predicts infection in blunt trauma patients. J Tra50:817-820.

28. Malone DL, Kuhls D, Napolitano LM, McCarter R, Scaleabasics: Validation of the admission systemic inflammatorsyndrome score in predicting outcome in trauma. J Traum458-463.

29. Napolitano LM, Ferrer T, McCarter RJ Jr, Scalea TM. Systematory response syndrome score at admission independenmortality and length of stay in trauma patients. J Trauma 20652; discussion 652-653.

30. Nast-Kolb D, Waydhas C, Gippner-Steppert C, et al. Indicposttraumatic inflammatory response correlate with orgapatients with multiple injuries. J Trauma 1997;42:446-454;454-455.

31. Nuytinck HK, Offermans XJ, Kubat K, Goris JA. Whole-bomation in trauma patients. An autopsy study. Arch Surg1519-1524.

32. Gebhard F, Pfetsch H, Steinbach G, Strecker W, Kinzl L, BrIs interleukin 6 an early marker of injury severity followtrauma in humans? Arch Surg 2000;135:291-295.

33. Villoslada P, Steinman L, Baranzini SE. Systems biology ancation to the understanding of neurological diseases. A2009;65:124-139.

34. Centers for Disease Control and Prevention. Traumatic brain iUnited States: Emergency department visits, hospitalizations2002-2006, 2010. Available at: http://www.cdc.gov/traumatictbi_ed.html. Accessed March 29, 2011.

35. Adekoya N, Thurman DJ, White DD, Webb KW. Survtraumatic brain injury deaths—United States, 1989-199Surveill Summ 2002;51:1-14.

36. Mosenthal AC, Lavery RF, Addis M, et al. Isolated trauminjury: Age is an independent predictor of mortality and earlJ Trauma 2002;52:907-911.

37. McArthur DL, Chute DJ, Villablanca JP. Moderate and severbrain injury: Epidemiologic, imaging and neuropathologtives. Brain Pathol 2004;14:185-194.

38. Gomez PA, Lobato RD, Boto GR, De la Lama A, Gonzalez PJJ. Age and outcome after severe head injury. Acta Neuroc2000;142:373-380; discussion 380-381.

39. Mattson MP, Magnus T. Ageing and neuronal vulnerabiliNeurosci 2006;7:278-294.

40. Dickstein DL, Kabaso D, Rocher AB, Luebke JI, Wearne SChanges in the structural complexity of the aged brain.2007;6:275-284.

41. Melov S. Modeling mitochondrial function in aging neuroNeurosci 2004;27:601-606.

42. Mattson MP. Excitotoxic and excitoprotective mechanismstargets for the prevention and treatment of neurodegenerders. Neuromolecular Med 2003;3:65-94.

43. Wagner AK, Bayir H, Ren D, Puccio A, Zafonte DA, Koc

Relationships between cerebrospinal fluid markers of excitotoxic

DR.sed

fterand

re-

eading

T.de-01;

tonse;51:

am-icts47-

thee inion

am-23:

UB.ajor

pli-rol

theathsury/

forWR

rainme.

aticec-

ruzien)

Rev

PR.Cell

nds

antsor-

PM.

ischemia, and oxidative damage after severe TBI: The impacage, and hypothermia. J Neurotrauma 2004;21:125-136.

44. Godbout JP, Chen J, Abraham J, et al. Exaggerated neuroinfland sickness behavior in aged mice following activation oferal innate immune system. FASEB J 2005;19:1329-1331.

45. Frank MG, Barrientos RM, Biedenkapp JC, Rudy JW, WMaier SF. mRNA up-regulation of MHC II and pivotal protory genes in normal brain aging. Neurobiol Aging 2006;27

46. Perry VH, Matyszak MK, Fearn S. Altered antigen expressioglia in the aged rodent CNS. Glia 1993;7:60-67.

47. Sheffield LG, Berman NE. Microglial expression of MHCcreases in normal aging of nonhuman primates. Neuro1998;19:47-55.

48. Godbout JP, Johnson RW. Age and neuroinflammation: Apsychoneuroimmune consequences. Neurol Clin 2006;24:

49. Lenz A, Franklin GA, Cheadle WG. Systemic inflammtrauma. Injury 2007;38:1336-1345.

50. Bulati M, Pellicano M, Vasto S, Colonna-Romano G. Undageing: Biomedical and bioengineering approaches, the imview. Immun Ageing 2008;5:9.

51. Vasto S, Candore G, Balistreri CR, et al. Inflammatory nageing, age-related diseases and longevity. Mech Ageing Dev83-91.

52. Straub RH, Miller LE, Scholmerich J, Zietz B. Cytokines andas possible links between endocrinosenescence and immcence. J Neuroimmunol 2000;109:10-15.

53. Franceschi C, Capri M, Monti D, et al. Inflammaging and amaging: A systemic perspective on aging and longevity emstudies in humans. Mech Ageing Dev 2007;128:92-105.

54. Dilger RN, Johnson RW. Aging, microglial cell priming, acordant central inflammatory response to signals from theimmune system. J Leukoc Biol 2008;84:932-939.

55. Vermeulen A, Kaufman JM. Ageing of the hypothalamotesticular axis in men. Horm Res 1995;43:25-28.

56. Araujo AB, O’Donnell AB, Brambilla DJ, et al. Prevalence andof androgen deficiency in middle-aged and older men: Estithe Massachusetts Male Aging Study. J Clin Endocrinol M89:5920-5926.

57. Jiang M, Huhtaniemi I. Polymorphisms in androgen anreceptor genes: Effects on male aging. Exp Gerontol 2001611.

58. Vermeulen A, Kaufman JM, Goemaere S, van Pottelberg I. Eelderly men. Aging Male 2002;5:98-102.

59. Lanfranco F, Gianotti L, Giordano R, Pellegrino M, MaccarE. Ageing, growth hormone and physical performance. JInvest 2003;26:861-872.

60. Elmadfa I, Meyer AL. Body composition, changing physiolotions and nutrient requirements of the elderly. Ann Nutr M52(Suppl 1):2-5.

61. Holick MF. Sunlight and vitamin D for bone health and prautoimmune diseases, cancers, and cardiovascular disease.Nutr 2004;80(6 Suppl):1678S-1688S.

62. Rammos G, Tseke P, Ziakka S. Vitamin D, the renin-asystem, and insulin resistance. Int Urol Nephrol 2008;40:4

63. Ferrucci L, Ble A, Bandinelli S, Lauretani F, Suthers K, Gurflame burning within. Aging Clin Exp Res 2004;16:240-24

64. Talhouk RS, Saade NE, Mouneimne G. Masaad CA, Safieh-GB. Growth hormone releasing hormone reverses endotoxlocalized inflammatory hyperalgesia without reducing the ucytokines, nerve growth factor and gelatinase activity. Progchopharmacol Biol Psychiatry 2004;28:625-631.

65. Priego T, Granado M, Ibanez de Caceres I, Martin Ai, ViLopez-Calderon A. Endotoxin at low doses stimulates piwhereas it decreases IGF-I and IGF-binding protein-3 in ra

ity, crinol 2003;179:107-117.

o-pituit

s: Evide46-M1vitami

etab 20

hat vitapocamp

vitamire lengt

associawalk

6:454-4North

266-28in D. N

unologl (Noisy

atus, 1,Nutr 20

s regulaNutr 20

ults Can

of vita4 positiv

hem 20

scularthe Unxamina

oxyvitaesults fr8;28:11

al effectl, magnurotrau

e funct

dispose04., Wion

em. Tre

ety in m1-1274D insu2002;1

Prevaleediatr A

nd gaseousal Toxicol

AdolescentDiet Assoc

deficiencyern Med J

in internal

in D3 and

ribution ofan brain.

xylase and.new hori-.Roque M.residents:lin (Barc)

. VitaminMol Biol

Adams JS,oxyvitaminntion and

the acutein preven-

5;64:1022-

on dietarynstitute ofinol Metab

CT: A ran-rain injury.

es from theevere trau-e 2008;12:

ts recoveryeural Biol

rone treat-traumatic

ne rapidlyurs is still

anatomicnse study.

he neuros-ath, gliosis,ats. J Neu-

progester-sion of the

S109PM&R Vol. 3, Iss. 6S, 2011

66. Beishuizen A, Thijs LG. Endotoxin and the hypothalamadrenal (HPA) axis. J Endotoxin Res 2003;9:3-24.

67. Fried LP, Tangen CM, Walston J, et al. Frailty in older adultfor a phenotype. J Gerontol A Biol Sci Med Sci 2001;56:M1

68. Nagpal S, Na S, Rathnachalam R. Noncalcemic actions ofreceptor ligands. Endocr Rev 2005;26:662-687.

69. Topinkova E. Aging, disability and frailty. Ann Nutr M52(Suppl 1):6-11.

70. Moore ME, Piazza A, McCartney Y, Lynch MA. Evidence tD3 reverses age-related inflammatory changes in the rat hipBiochem Soc Trans 2005;33(Pt 4):573-577.

71. Richards JB, Valdes AM. Gardner JP, et al. Higher serumconcentrations are associated with longer leukocyte telomewomen. Am J Clin Nutr 2007;86:1420-1425.

72. Boxer RS, Dauser DA, Walsh SJ, Hager WD, Kenny AM. Thebetween vitamin D and inflammation with the 6-minutefrailty in patients with heart failure. J Am Geriatr Soc 2008;5

73. Morley JE, Haren MT, Rolland Y, Kim MJ. Frailty. Med Clin2006;90:837-847.

74. Holick MF. Vitamin D deficiency. N Engl J Med 2007;357:75. DeLuca HF, Zierold C. Mechanisms and functions of vitam

Rev 1998;56(2 Pt 2):S4-S10; discussion S54-S75.76. Hayes CE, Nashold FE. Spach KM, Pedersen LB. The imm

functions of the vitamin D endocrine system. Cell Mol Biogrand) 2003;49:277-300.

77. Cantorna MT, Zhu Y, Froicu M, Wittke A. Vitamin D stdihydroxyvitamin D3, and the immune system. Am J Clin80(6 Suppl):1717S-1720S.

78. Griffin MD, Xing N, Kumar R. Vitamin D and its analogs aof immune activation and antigen presentation. Annu Rev23:117-145.

79. Holick MF. Evolution and function of vitamin D. Recent ResRes 2003;164:3-28.

80. Mahon BD, Wittke A, Weaver V, Cantorna MT. The targetsD depend on the differentiation and activation status of CDcells. J Cell Biochem 2003;89:922-932.

81. Holick MF, Vitamin D: A millenium perspective. J Cell Bioc88:296-307.

82. Martins D, Wolf M, Pan D, et al. Prevalence of cardiovafactors and the serum levels of 25-hydroxyvitamin D inStates: Data from the Third National Health and Nutrition ESurvey. Arch Intern Med 2007;167:1159-1165.

83. Melamed ML, Muntner P, Michos ED, et al. Serum 25-hydrD levels and the prevalence of peripheral arterial disease: RNHANES 2001 to 2004. Arterioscler Thromb Vasc Biol 2001185.

84. Onyszchuk G, He YY, Berman NE, Brooks WM. Detrimentaging on outcome from traumatic brain injury: A behavioraresonance imaging, and histological study in mice. J Ne2008;25:153-171.

85. Pfeifer M, Begerow B, Minne HW. Vitamin D and musclOsteoporos Int 2002;13:187-194.

86. Peterlik M, Cross HS. Vitamin D and calcium deficits premultiple chronic diseases. Eur J Clin Invest 2005;35:290-3

87. Garcion E, Wion-Barbot N, Montero-Menei CN, Berger FNew clues about vitamin D functions in the nervous systEndocrinol Metab 2002;13:100-105.

88. Kalueff AV, Lou YR, Laaksi I, Tuohimaa P. Increased anxilacking vitamin D receptor gene. Neuroreport 2004;15:127

89. Tangpricha V, Pearce EN, Chen TC, Holick MF. Vitaminciency among free-living healthy young adults. Am J Med659-662.

90. Gordon CM, DePeter KC, Feldman HA, Grace E, Emans SJ.of vitamin D deficiency among healthy adolescents. Arch P

lesc Med 2004;158:531-537.

ary-

nce56.n D

08;

minus.

n Dh in

tionand61.Am

1.utr

ical-le-

25-04;

tors03;

cer

mine T

03;

riskitedtion

minom79-

s ofeticma

ion.

for

D.nds

ice.ffi-12:

ncedo-

91. Nadziejko C, Fang K, Narciso S, et al. Effect of particulate apollutants on spontaneous arrhythmias in aged rats. Inh2004;16:373-380.

92. Sullivan SS, Rosen CJ, Halteman WA, Chen TC, Holick MF.girls in Maine are at risk for vitamin D insufficiency. J Am2005;105:971–874.

93. Chatfield SM, Brand C, Ebeling PR, Russell DM. Vitamin Din general medical inpatients in summer and winter. Int2007;37:377-382.

94. Corino A, D’Amelio P, Gancia R, et al. Hypovitaminosis Dmedicine inpatients. Calcif Tissue Int 2007;80:76-80.

95. Eyles D, Brown J, Mackay-Sim A, McGrath J, Feron F. Vitambrain development. Neuroscience 2003;118:641-653.

96. Eyles DW, Smith S, Kinobe R, Hewison M, McGrath JJ. Distthe vitamin D receptor and 1 alpha-hydroxylase in humJ Chem Neuroanat 2005;29:21-30.

97. Hewison M, Zehnder D, Bland R, Stewart PM. 1alpha-Hydrothe action of vitamin D. J Mol Endocrinol 2000;25:141-148

98. Holick MF. McCollum Award Lecture, 1994: Vitamin D—zons for the 21st century. Am J Clin Nutr 1994;60:619-630

99. Larrosa M, Gratacos J, Vaqueiro M, Prat M, Campos F,Prevalence of hypovitaminosis D in elderly institutionalizedInfluence of a substitutive treatment [in Spanish]. Med C2001;117:611-614.

100. McGrath JJ, Feron FP. Burne TH, Mackay-Sim A, Eyles DWD3-implications for brain development. J Steroid Biochem2004;89–90:557-560.

101. Townsend K, Evans KN, Campbell MJ, Colston KW,Hewison M. Biological actions of extra-renal 25-hydrD-1alpha-hydroxylase and implications for chemoprevetreatment. J Steroid Biochem Mol Biol 2005;97:103-109.

102. McCarty MF. Secondary hyperparathyroidism promotesphase response—a rationale for supplemental vitamin Dtion of vascular events in the elderly. Med Hypotheses 2001026.

103. Ross AC, Manson JE, Abrams SA, et al. The 2011 Reportreference intakes for calcium and vitamin d from the IMedicine: What clinicians need to know. J Clin Endocr2011;96:53-58.

104. Wright DW, Kellermann AL, Hertzberg VS, et al. ProTEdomized clinical trial of progesterone for acute traumatic bAnn Emerg Med 2007;49:391-402.

105. Xiao G, Wei J, Yan W, Wang W, Lu Z. Improved outcomadministration of progesterone for patients with acute smatic brain injury: A randomized controlled trial. Crit CarR61.

106. Attella MJ, Nattinville A, Stein DG. Hormonal state affecfrom frontal cortex lesions in adult female rats. Behav N1987;48:352-367.

107. Pettus EH, Wright DW, Stein DG, Hoffman SW. Progestement inhibits the inflammatory agents that accompanybrain injury. Brain Res 2005;1049:112-119.

108. Roof RL, Duvdevani R. Heyburn JW, Stein DG. Progesterodecreases brain edema: Treatment delayed up to 24 hoeffective. Exp Neurol 1996;138:246-251.

109. Goss CW, Hoffman SW, Stein DG. Behavioral effects andcorrelates after brain injury: A progesterone dose-respoPharmacol Biochem Behav 2003;76:231-242.

110. Djebaili M, Guo Q, Pettus EH. Hoffman SW, Stein G. Tteroids progesterone and allopregnanolone reduce cell deand functional deficits after traumatic brain injury in rrotrauma 2005;22:106-118.

111. Djebaili M, Hoffman SW, Stein DG. Allopregnanolone andone decrease cell death and cognitive deficits after a contu

rat pre-frontal cortex. Neuroscience 2004;123:349-359.

after b

gesteroury in m

ne protcaused

protect28-235.ew role6-391.

contex8:317-3uroproatr Reh

Scheff S. Brain

e followuma 20

one, a pin reductery oc

ain injies. Tre

candidroke? N

insightelevanc

erspectecial re

9., Steinand ede6;198:4

Stein D55 ind

esteroneations,J Surg

Vitaminafter b

JW, Steinc brain in-

6;92:4-8.m vitamin

cture: The7;62:1402-

ters PJ, Decal illness:3-4632.

The search

amage—is

atic brain6:925-939.ysiol Renal

droxyvita-97:93-101.eq definedations with

d allopreg-ing perma-ix metallo-

ogesteroneJ Obstet

one exertsonse after

icoids andus disease.

hibition oftive mech-

revent cellnt middleav Immun.

a combina-lution to a

ds a vision1045.al nervous

Vitamin Dltured cor-

S110 Stein and Cekic BRAIN INJURY, AGING, AND NEUROSTEROIDS

112. Stein DG. Progesterone exerts neuroprotective effectsinjury. Brain Res Rev 2008;57:386-397.

113. Guo Q, Baronne L, Hoffman S, Guennoun R, Stein D. Proeffects on aquaporin-4 expression after traumatic brain injrats. J Neurotrauma 2004;21:1303.

114. Shear DA, Galani R, Hoffman SW, Stein DG. Progesteroagainst necrotic damage and behavioral abnormalitiestraumatic brain injury. Exp Neurol 2002;178:59-67.

115. McCullough LD, Hurn PD. Estrogen and ischemic neuroAn integrated view. Trends Endocrinol Metab 2003;14:2

116. Stein DG. Brain damage, sex hormones and recovery: A nprogesterone and estrogen? Trends Neurosci 2001;24:38

117. Stein DG, Hoffman SW. Concepts of CNS plasticity in thebrain damage and repair. J Head Trauma Rehabil 2003;1

118. Stein DG, Hoffman SW. Estrogen and progesterone as netive agents in the treatment of acute brain injuries. Pedi2003;6:13-22.

119. Fee DB, Swartz KR, Joy KM, Roberts KN, Scheff NN,Effects of progesterone on experimental spinal cord injury2007;1137:146-152.

120. Gilmer LK, Roberts KN, Scheff SW. Efficacy of progesterona moderate unilateral cortical contusion injury. J Neurotra25:593-602.

121. Sayeed I, Guo Q, Hoffman SW, Stein DG Allopregnanolgesterone metabolite, is more effective than progesteronecortical infarct volume after transient middle cerebral arsion. Ann Emerg Med 2006;47:381-389.

121a.Loane DJ, Faden AI. Neuroprotection for traumatic brtranslational challenges and emerging therapeutic strategPharmacol Sci 2010;31:596-604.

121b.Gibson CL, Coomber B, Rathbone J. Is progesterone aneuroprotective factor for treatment following ischemic stroscientist 2009;15:324-332.

122. Ehrnthaller C, Ignatius A, Gebhard F, Huber-Lang M. Newan old defense system: Structure, function, and clinical rthe complement system. Mol Med 2011;17:317-329.

123. Schumacher M, Guennoun R, Ghoumari A, et al. Novel pfor progesterone in hormone replacement therapy, with spence to the nervous system. Endocr Rev 2007;28:387-43

124. Guo Q, Sayeed I, Baronne LM, Hoffman SW, Guennoun RProgesterone administration modulates AQP4 expressionafter traumatic brain injury in male rats. Exp Neurol 200478.

125. VanLandingham JW, Cekic M, Cutler S, Hoffman SW,Neurosteroids reduce inflammation after TBI through CDtion. Neurosci Lett 2007;425:94-98.

126. Chen G, Shi JX, Qi M, Wang HX, Hang CH. Effects of progintestinal inflammatory response, mucosa structure alterapoptosis following traumatic brain injury in male rats.2008;147:92-98.

127. Cekic M, Cutler SM. Vanlandingham JW, Stein DG.deficiency reduces the benefits of progesterone treatment

injury in aged rats. Neurobiol Aging. In press.

rain

ne’sale

ectsby

ion:

for

t of41.tec-abil

W,Res

ing08;

ro-ing

clu-

ury:nds

ateeu-

s ofe of

ivesfer-

DG.ma69-

G.uc-

onandRes

Drain

128. Cutler SM, Cekic M, Miller DM, Wali B, VanLandinghamDG. Progesterone improves acute recovery after traumatijury in the aged rat. J Neurotrauma 2007;24:1475-1486.

129. Lips P. Vitamin D physiology. Prog Biophys Mol Biol 200130. Miller RR, Hicks GE, Shardell MD, et al. Association of seru

D levels with inflammatory response following hip fraBaltimore Hip Studies. J Gerontol A Biol Sci Med Sci 2001406.

131. Van den Berghe G, Van Roosbroeck D, Vanhove P, WouPourcq L, Bouillon R. Bone turnover in prolonged critiEffect of vitamin D. J Clin Endocrinol Metab 2003;88:462

132. Faden AI. Neuroprotection and traumatic brain injury:continues. Arch Neurol 2001;58:1553-1555.

133. Gingrich MB, Traynelis SF. Serine proteases and brain dthere a link? Trends Neurosci 2000;23:399-407.

134. Margulies S, Hicks R. Combination therapies for trauminjury: Prospective considerations. J Neurotrauma 2009;2

135. Dusso AS, Brown AJ, Slatopolsky E. Vitamin D. Am J PhPhysiol 2005;289:F8-F28.

136. van Etten E, Mathieu C. Immunoregulation by 1,25-dihymin D3: Basic concepts. J Steroid Biochem Mol Biol 2005;

137. Ramagopalan SV, Heger A, Berlanga AJ, et al. A ChIP-sgenome-wide map of vitamin D receptor binding: Associdisease and evolution. Genome Res 2010;20:1352-1360.

138. Ishrat T, Sayeed I, Atif F, Hua F, Stein DG. Progesterone annanolone attenuate blood-brain barrier dysfunction follownent focal ischemia by regulating the expression of matrproteinases. Exp Neurol 2010;226:183-190.

139. Schwartz N, Xue X, Elovitz MA, Dowling O, Metz CN. Prsuppresses the fetal inflammatory response ex vivo. AmGynecol 2009;201:211 e1-e9.

140. Jiang C, Wang J, Li X, Liu C, Chen N, Hao Y. Progesterneuroprotective effects by inhibiting inflammatory respstroke. Inflamm Res 2009;58:619-624.

141. Tait AS, Butts CL, Sternberg EM. The role of glucocortprogestins in inflammatory, autoimmune, and infectioJ Leukoc Biol 2008;84:924-931.

142. Luoma JI, Kelley BG, Mermelstein PG. Progesterone Involtage-gated calcium channels is a potential neuroprotecanism against excitotoxicity. Steroids. In press.

143. Dang J, Mitkari B, Kipp M, Beyer C. Gonadal steroids pdamage and stimulate behavioral recovery after transiecerebral artery occlusion in male and female rats. Brain BehIn press.

144. Cekic M, Stein DG. Traumatic brain injury and aging: Istion of progesterone and vitamin D hormone a simple socomplex problem? Neurotherapeutics 2010;7:81-90.

145. Norman AW, Bouillon R. Vitamin D nutritional policy neefor the future. Exp Biol Med (Maywood) 2010;235:1034-

146. Holmoy T, Moen SM. Assessing vitamin D in the centrsystem. Acta Neurol Scand Suppl 2010;(190):88-92.

147. Atif F, Sayeed I, Ishrat T, Stein DG. Progesterone withaffords better neuroprotection against excitotoxicity in cu

tical neurons than progesterone alone. Mol Med 2009;15:328-336.