Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Feb 1.
Published in final edited form as: Neuropharmacology. 2018 Jun 20;145(Pt B):153–159. doi: 10.1016/j.neuropharm.2018.06.025

Therapeutic strategies to target acute and long-term sequelae of pediatric traumatic brain injury

Jimmy W Huh 1, Ramesh Raghupathi 2
PMCID: PMC6309300  NIHMSID: NIHMS979037  PMID: 29933010

Abstract

Pediatric traumatic brain injury (TBI) remains one of the leading causes of morbidity and mortality in children. Experimental and clinical studies demonstrate that the developmental age, the type of injury (diffuse vs. focal) and sex may play important roles in the response of the developing brain to a traumatic injury. Advancements in acute neurosurgical interventions and neurocritical care have improved and led to a decrease in mortality rates over the past decades. However, survivors are left with life-long behavioral deficits underscoring the need to better define the cellular mechanisms underlying these functional changes. A better understanding of these mechanisms some of which begin in the acute post-traumatic period may likely lead to targeted treatment strategies. Key considerations in designing pre-clinical experiments to test therapeutic strategies in pediatric TBI include the use of age-appropriate and pathologically-relevant models, functional outcomes that are tested as animals age into adolescence and beyond, sex as a biological variable and the recognition that doses and dosing strategies that have been demonstrated to be effective in animal models of adult TBI may not be effective in the developing brain.

Introduction

Traumatic brain injury (TBI) in infants and children remains one of the leading causes of long-term disability and mortality worldwide17 and occurs as a result of either accidental or inflicted causes (abusive head trauma)8,9. Advancements in pediatric neurosurgical interventions and neurocritical care has reduced mortality in cases of severe pediatric TBI10. However, survivors face life-long behavioral problems exacerbated by the paucity of pharmacological interventions that are aimed at reversing or attenuating specific cellular mechanisms in order to limit these behavioral pathologies. Pediatric TBI is a “chronic” disease, as long-term intellectual and psychosocial deficits are observed in adult survivors following childhood injury11,12. A number of age-appropriate animal models have been developed over the past 2 decades which have validated some of the acute and chronic behavioral deficits and the structural and cellular alterations typically observed in patients13. These animal models are important for testing hypotheses for the underlying cellular mechanisms, cellular alterations and behavioral deficits. In this review, we will briefly describe human pathologies and their validation in animal models of pediatric TBI and describes some of the attempts to attenuate these pathologies using standard pharmacological approaches.

Functional deficits following pediatric TBI

The negative consequences of TBI early in life manifests during childhood and extends throughout their life as these individuals have difficulty in developing new cognitive or social skills1416. Age-appropriate behavior and levels of arousal and responsiveness were significantly decreased in infants and young children that sustained moderate to severe TBI17. Intelligent quotient measures did not improve between 6 months and 24 months post-injury indicating a lack of improvement and developmental arrest18. Children with brain trauma also exhibit deficits in verbal working memory, visuo-spatial memory, and attention that may contribute to difficulties in a school setting1921. Psychosocial problems such as depression, anxiety, and sleep disturbances become more apparent as these children become older15,16. Brain injury sustained in early childhood led to decreased social competencies, an increase in irritability and aggression associated with disinhibited verbalizations, and the development of novel depressive and anxiety disorders2224. Children with severe brain injuries had significantly decreased motor scores at the initial testing time which only improved in the first 6 months25. Children that sustain severe injuries demonstrate an increased incidence of seizures in the early stages following injury which are typically observed in those under the age of 1 and/or following abusive head trauma26,27.

Cognitive function in animal models of pediatric TBI have been restricted to simple cognitive tasks such as spatial navigation learning and memory in the Morris water maze or visual recognition memory in the novel object recognition test, although these deficits were evident for weeks to months after the initial injury2835. It is important that more complex cognitive function such as retrograde or anterograde spatial memory, episodic memory and working memory be evaluated as neonate animals age into adolescence and adulthood36. Contusive brain trauma in the neonate mouse resulted in prolonged hyperactivity, a decrease in general anxiety-like behavior, and deficits in social behavior weeks to months after the injury3037. Motor function deficits in animal models of pediatric TBI are typically observed in only the first few weeks after injury34. Video-electroencephalograph recordings and susceptibility to seizure-inducing agents have been utilized in animal models to demonstrate the incidence of seizure activity in the acute and chronic post-traumatic periods following pediatric TBI3840.

Cellular pathology following pediatric TBI

Macroscopic structural pathology following TBI include focal contusions which are related to impact forces or diffuse injury which may occur due to rotational forces41. The pliancy and relatively thinner skull of the infant results in a decreased ability to absorb energy during the application of an external force leading to a greater deformations than the area of direct application and a more diffuse pattern of injury in the developing brain42. Skull fractures occur in both accidental and abusive head trauma, with subdural hematomas present more frequently in children who sustained abusive TBI and epidural hematomas much more likely in children with accidental TBI43,44. Evidence of brain atrophy and ex-vacuo ventriculomegaly (enlarged ventricular space) has been reported following accidental or abusive TBI43,45,46. Subarachnoid hemorrhage and subdural hematomas have been observed in animal models in which impact occurred on the skull surface either via a weight-drop method47,48 or using the controlled cortical impact device49,50, or as a result of non-impact, rotational-acceleration51,52; in some cases ex-vacuo ventriculomegaly was observed49,50,53.

Microscopic alterations to the traumatically-injured pediatric brain include axonal injury, cell death, gliosis and inflammation. Damage to the white matter tracts resulting in traumatic axonal injury (TAI) has been identified as the predominant cellular pathology in cases of infant TBI. Magnetic resonance imaging has revealed evidence of shearing and impaired myelination in the corpus callosum which is associated with slower interhemispheric transfer times measured using event-related potentials and indicative of impaired function20,5457. Diffuse tensor imaging has demonstrated reduced fractional anisotropy in the corpus callosum, internal capsule, and longitudinal fasciculus, which has been associated with cognitive and psychosocial deficits in patients who sustained a TBI in early childhood20,54,55,58,59. Post-mortem evaluation using silver staining or beta-amyloid precursor protein (β-APP) immunohistochemistry60,61 has detected axonal injury in the corpus callosum, internal capsule, midbrain and brainstem6264. These clinical observations have been validated in multiple animal models of impact and non-impact TBI using either imaging65 or immunostaining for β-APP and/or neurofilament35,4952,6668. A decrease in axonal function in the white matter tracts (measured using compound action potential in ex vivo preparations) has also been documented65,69, along with observations of atrophy in the white matter tracts over time associated with axonal degeneration70,71. Evidence of brain atrophy in head-injured children suggests that neuronal cell death may be a component of TBI pathology45,72,73. Direct indication of cell death in various pre-clinical pediatric TBI models has been facilitated via the use of markers specifically associated with necrosis, apoptosis, and excitotoxicity48,49,74,75. Associated with neuronal loss is evidence of glial reactivity. Severe TBI in children is associated with an increase in pro-and anti-inflammatory cytokines such as the interleukins −1β, −6, and −1076,77, chemokines interleukin-8 and macrophage inflammatory protein-1α indicating migratory signaling to other immune cells77, and significantly higher concentrations of quinolinic acid indicative of microglia/macrophage activation78. These are by no means an exhaustive list of cytokines and chemokines underscoring a need to extend these observations to other chemokines such as chemokine C-C motif ligand (CCL) and CXCL families that have demonstrated roles in mediating the cellular immune response. Recent evidence indicates that TBI in neonate and juvenile animals is associated with an increase in tissue and serum concentrations of various cytokines and chemokines such as tumor necrosis factor-α, IL-1β, IL-6, CXCL1, CCL5 and CCL3 to name a few7981. Head trauma in the immature animal also resulted in microglial activation along with infiltration of peripheral immune cells in multiple brain regions such as the cortex, thalamus, hippocampus and subcortical white matter tracts which demonstrate evidence of neurodegeneration and axonal injury28,70,71,74,79,82.

Treatment strategies in pediatric TBI focused on specific cellular pathology

Axonal injury

Despite the identification of axonal injury as a predominant cellular pathology in pediatric TBI, surprisingly few studies have focused on attenuating axonal degeneration. Proposed secondary mechanisms underlying traumatic axonal injury include ionic dysregulation, impaired axonal transport, calpain-mediated proteolysis, calcineurin-mediated dephosphorylation, microtubule degradation, and neurofilament compaction8389. Calpain activation has been documented within injured axons in the immature rat75,90 although administration of calpain inhibitor III (MDL28,170) at doses of either 30mg/kg or 60mg/kg over the first 24 hours following TBI in the juvenile rat reduced calpain activation but not axonal function deficits (unpublished observations). Calcineurin-mediated neurofilament dephosphorylation and compaction has been reported following diffuse brain injury in the immature rat67,69 although the calcineurin inhibitor tacrolimus (FK-506) did not improve axonal function despite reducing neurofilament dephosphorylation69. These observations underscore the possibility that mechanisms of TBI-induced axonal damage and degeneration may not occur via similar mechanisms in the immature and adult brains89,9193.

Cell death

High concentrations of glutamate have been reported in the cerebrospinal fluid of children that sustained a severe TBI94, suggesting that excitotoxicity leading to either apoptotic or necrotic cell death may occur in the pediatric brain95,96 and may be identified using activation of caspases and calpains, respectively97. Traumatic injury to the 9-day-old, 11-day-old or 17-day-old rat demonstrated activation of calpains and caspases in multiple brain regions75,98,99. Similarly, decreased expression of the anti-apoptotic protein Bcl-2 along with activation of the pro-apoptotic protease caspase-3 suggests that apoptotic cascades are activated following trauma to the developing brain and discovered that these apoptotic markers were most severe in the youngest animals48. As observed in models of adult TBI100,101, glutamate receptor antagonists administered acutely following neonate TBI appears to provide neuroprotection102. However, this strategy must be considered with caution as NMDA receptor antagonists were observed to increase apoptosis in the developing brain following TBI103. Moreover, excitatory neurotransmission is critically important for proper development of the immature brain104; in this regard, the co-agonist of the N-methyl-D-aspartate receptor (NMDAR), D-cycloserine has been observed to limit post-traumatic decreases in NMDAR expression, novel object recognition deficits and improve the response of the brain to experience-dependent plasticity105.

Inflammation

Targeting the activation of astrocytes, microglia and infiltrating leukocytes and monocytes for therapeutic purposes in models of pediatric TBI has only been a recent development106,107, albeit with limited success. Decreasing the expression of the astrocytic protein aquaporin-4 via RNA silencing reduced edema and blood-brain barrier disruption in the acute post-traumatic period and spatial learning deficits in the months after injury but did increase microglial activation107. The protease elastase is released from activated neutrophils that enter the brain after trauma but inhibition of this protease in the acute post-traumatic period only served to reduce cell death and had no effect on long-term behavioral outcomes108. Similarly, manipulation of infiltrating mast cells after trauma to the neonate mouse using a degranulation inhibitor or with genetic approaches had little to no effect on cell death, axonal injury or microglial activation109. Short-term minocycline administration in the first week following trauma to the neonate rodent reduced microglial proliferation and activation but was accompanied by increased neurodegeneration and no attenuation of spatial learning and memory deficits70,71,79. These observations are suggestive of a differential age-at-injury inflammatory response because similar strategies in the adult TBI models have demonstrated marked efficacy in increasing neuroprotection, reducing microglial activation and attenuating locomotor and spatial learning and memory deficits110116. Because microglia in the developing brain are important for sculpting of neuronal circuits, synapse and axonal remodeling, and pruning of unwanted or excess cells and clearance of unwanted cellular debris117120, a better approach may be targeting the pro-inflammatory cytokines that may mediate secondary damage after trauma. In this regard, the interleukin-1 receptor antagonist (anakinra) has been reported to reduce post-traumatic seizure susceptibility in a mouse model of pediatric TBI38.

Treatment strategies in pediatric TBI focused on multiple cellularpathologies

Pediatric TBI, as in the case of adult TBI, exhibits complex pathologic alterations indicative of multiple mechanisms13. Whereas strategies targeting a single mechanism may yet be viable, a second approach is using either a combination of pharmacologic agents, or a single pleiotropic agent/intervention that targets multiple mechanisms. The former strategy has had limited success in models of pediatric TBI and is reviewed by Margulies et al121. Here we describe a few examples of the latter approach based on their successes in models of adult TBI.

Progesterone

The steroid hormone progesterone (PROG) has multiple mechanisms of neuroprotection from reducing edema, inflammation, oxidative damage to apoptosis and excitotoxic cell death122. It has been extensively used in models of adult TBI but failed in phase III clinical trials in moderate-severe adult TBI123126. Despite this setback, PROG has been tested in a few different models of pediatric TBI albeit with limited success. In male but not female immature rats, PROG reversed mitochondrial dysfunction and improved anti-oxidant reserves127. Similarly, PROG treatment following contusive trauma in 4-week old mice improved motor function in male mice while worsening performance in female mice and did not reduce neurodegeneration in either sex128. When 4-week-old male and female rats were subjected to bilateral frontal lobe contusive trauma, PROG attenuated spatial learning and locomotor deficits in both sexes, along with a reduction in lesion volume129,130. It must be noted that few, if any, of these studies have evaluated the mechanistic basis of the actions of PROG but do highlight the importance of the differential efficacy of interventions on the basis of sex.

Erythropoietin

The pleiotropic cytokine, erythropoietin (EPO) is currently in use as a neuroprotective agent in preterm infants at risk for central nervous system injury131,132. EPO, like PROG, has multiple mechanisms of action including limiting apoptosis and promoting neural tissue repair133. EPO, particularly at high doses administered over an extended period of time after injury, has been effective in reducing post-traumatic pathology and behavioral deficits in multiple animal models of adult TBI134. In either 12- or 17-day-old rat pups, EPO was administered over the first week post-injury and was observed to reverse motor deficits, cortex and white matter damage, attenuate hippocampal apoptosis and improve recognition memory80,135. A recent study reported that the improvement in recognition memory was maintained into adulthood136. However, neither the specific mechanism of action of EPO in the injured neonate brain nor the dependence of efficacy on sex are currently known.

Future directions for treating pediatric TBI

The importance of evaluating multiple outcome measures (cognitive, motor, psychosocial) during the chronic phase of injury following a therapeutic intervention cannot be overstated, as most pediatric TBI patients survive but are left with profound morbidity in adulthood. Preclinical pediatric TBI studies have successfully modeled acute (days to weeks) cellular pathologies observed in the human condition and have associated these changes with simple behavioral tasks of learning and memory. The investigation of long-term (weeks to months) pathologic and functional outcomes and evaluation of psychosocial behaviors have only recently been implemented and need additional evaluation. Intervention strategies aimed at specific cellular pathologies such as axonal injury, cell death or inflammation that have worked in adult TBI models are not always effective in attenuating functional deficits in the immature animal and therefore must be carefully examined in future studies. These data underscore the need for further studies to have a much better understanding of age-dependent mechanistic responses to trauma. In addition, mechanisms of brain damage sustained after trauma at the different maturational stages in development (infant, toddler, adolescent) need to be evaluated with a view to developing age-specific treatment strategies. For example, the decrease in glucose utilization that occurs over time after pediatric TBI137 allows ketone bodies to be administered as an alternative substrate although this approach was effective in reducing lesion size and behavioral deficits in a narrow range of ages (post-natal days 35–45)138,139. Future research aimed at attenuating deleterious processes using pharmacological means may also be augmented by using strategies to enhance reparative (plasticity) responses, not only during the acute post-traumatic period, but also at the subacute or chronic post-traumatic period. In this regard, environmental enrichment or transcranial magnetic stimulation offer promising leads to improve recovery of the immature brain from a traumatic injury140,141. It is becoming increasingly apparent that the gender affects outcome following pediatric TBI142,143 and the efficacy of the intervention paradigm in the acute post-traumatic period to limit pathology in preclinical studies127,128,144. In addition to the preliminary studies with progesterone127,128, vasopressor studies have demonstrated that phenylephrine (Phe) resulted in a greater reduction in metabolic crisis than with norepinephrine in female brain-injured piglets145, whereas Phe exacerbated cerebrovascular dysregulation in male brain-injured piglets144. Further studies on the role of sex and gender at different stages of maturation and the role of circulating sex hormones especially during the adolescent stages of TBI may dictate effective therapeutic strategies. Finally, as the field continues to develop mechanism-specific pharmacologic strategies, care must be given to exploring the potential for age-dependent differences in pharmacodynamics (target engagement) and pharmacokinetics (drug metabolism). Despite these challenges, it is encouraging to see the growth in preclinical pediatric TBI research.

Supplementary Material

Highlights.

Pediatric TBI is a leading cause of morbidity in children below the age of 14.

As brain-injured children age into adulthood, they exhibit multiple cognitive and behavioral deficits.

The cellular pathology of brain injury in children includes axonal injury, neurodegeneration and inflammation.

Pleiotropic agents may be better suited to treat pediatric TBI.

Factors such as age, sex and pharmacokinetics need to be considered.

Long-term behavioral measures need to be incorporated into the study design.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Thurman DJ. The Epidemiology of Traumatic Brain Injury in Children and Youths: A Review of Research Since 1990. J Child Neurol 2016;31(1):20–27. [DOI] [PubMed] [Google Scholar]
  • 2.Dewan MC, Mummareddy N, Wellons JC 3rd, Bonfield CM. Epidemiology of Global Pediatric Traumatic Brain Injury: Qualitative Review. World Neurosurg 2016;91:497–509 e491. [DOI] [PubMed] [Google Scholar]
  • 3.Taylor CA, Bell JM, Breiding MJ, Xu L. Traumatic Brain Injury-Related Emergency Department Visits, Hospitalizations, and Deaths -United States, 2007 and 2013. MMWR Surveill Summ 2017;66(9):1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Selassie AW, Zaloshnja E, Langlois JA, Miller T, Jones P, Steiner C. Incidence of long-term disability following traumatic brain injury hospitalization, United States, 2003. J Head Trauma Rehabil 2008;23(2):123–131. [DOI] [PubMed] [Google Scholar]
  • 5.Shaklai S, Peretz R, Spasser R, Simantov M, Groswasser Z. Long-term functional outcome after moderate-to-severe paediatric traumatic brain injury. Brain Inj 2014;28(7):915–921. [DOI] [PubMed] [Google Scholar]
  • 6.Rivara FP, Vavilala MS, Durbin D, et al. Persistence of disability 24 to 36 months after pediatric traumatic brain injury: a cohort study. J Neurotrauma 2012;29(15):2499–2504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Anderson V, Godfrey C, Rosenfeld JV, Catroppa C. Predictors of cognitive function and recovery 10 years after traumatic brain injury in young children. Pediatrics 2012;129(2):e254–261. [DOI] [PubMed] [Google Scholar]
  • 8.Ewing-Cobbs L, Kramer L, Prasad M, et al. Neuroimaging, physical, and developmental findings after inflicted and noninflicted traumatic brain injury in young children. Pediatrics 1998;102(2 Pt 1):300–307. [DOI] [PubMed] [Google Scholar]
  • 9.Keenan HT, Bratton SL. Epidemiology and outcomes of pediatric traumatic brain injury. Dev Neurosci 2006;28(4–5):256–263. [DOI] [PubMed] [Google Scholar]
  • 10.Kochanek PM, Carney N, Adelson PD, et al. Guidelines for the acute medical management of severe traumatic brain injury in infants, children, and adolescents--second edition. Pediatr Crit Care Med 2012;13 Suppl 1:S1–82. [DOI] [PubMed] [Google Scholar]
  • 11.Anderson V, Brown S, Newitt H, Hoile H. Educational, vocational, psychosocial, and quality-of-life outcomes for adult survivors of childhood traumatic brain injury. J Head Trauma Rehabil 2009;24(5):303–312. [DOI] [PubMed] [Google Scholar]
  • 12.Anderson V, Brown S, Newitt H, Hoile H. Long-term outcome from childhood traumatic brain injury: intellectual ability, personality, and quality of life. Neuropsychology 2011;25(2):176–184. [DOI] [PubMed] [Google Scholar]
  • 13.Kochanek PM, Wallisch JS, Bayir H, Clark RSB. Pre-clinical models in pediatric traumatic brain injury-challenges and lessons learned. Childs Nerv Syst 2017;33(10):1693–1701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Beauchamp MH, Anderson V. Cognitive and psychopathological sequelae of pediatric traumatic brain injury. Handb Clin Neurol 2013;112:913–920. [DOI] [PubMed] [Google Scholar]
  • 15.Choe MC, Valino H, Fischer J, et al. Targeting the Epidemic: Interventions and Follow-up Are Necessary in the Pediatric Traumatic Brain Injury Clinic. J Child Neurol 2016;31(1):109–115. [DOI] [PubMed] [Google Scholar]
  • 16.Anderson VA, Spencer-Smith MM, Coleman L, et al. Predicting neurocognitive and behavioural outcome after early brain insult. Dev Med Child Neurol 2014;56(4):329–336. [DOI] [PubMed] [Google Scholar]
  • 17.Ewing-Cobbs L, Prasad M, Kramer L, Landry S. Inflicted traumatic brain injury: relationship of developmental outcome to severity of injury. Pediatr Neurosurg 1999;31(5):251–258. [DOI] [PubMed] [Google Scholar]
  • 18.Ewing-Cobbs L, Fletcher JM, Levin HS, Francis DJ, Davidson K, Miner ME. Longitudinal neuropsychological outcome in infants and preschoolers with traumatic brain injury. J Int Neuropsychol Soc 1997;3(6):581–591. [PubMed] [Google Scholar]
  • 19.Treble A, Hasan KM, Iftikhar A, et al. Working memory and corpus callosum microstructural integrity after pediatric traumatic brain injury: a diffusion tensor tractography study. J Neurotrauma 2013;30(19):1609–1619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ewing-Cobbs L, Prasad MR, Swank P, et al. Arrested development and disrupted callosal microstructure following pediatric traumatic brain injury: relation to neurobehavioral outcomes. Neuroimage 2008;42(4):1305–1315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Catroppa C, Anderson V. A prospective study of the recovery of attention from acute to 2 years following pediatric traumatic brain injury. J Int Neuropsychol Soc 2005;11(1):84–98. [DOI] [PubMed] [Google Scholar]
  • 22.Max JE, Keatley E, Wilde EA, et al. Depression in children and adolescents in the first 6 months after traumatic brain injury. Int J Dev Neurosci 2012;30(3):239–245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Max JE, Lopez A, Wilde EA, et al. Anxiety disorders in children and adolescents in the second six months after traumatic brain injury. J Pediatr Rehabil Med 2015;8(4):345–355. [DOI] [PubMed] [Google Scholar]
  • 24.Ewing-Cobbs L, Prasad MR, Mendez D, Barnes MA, Swank P. Social interaction in young children with inflicted and accidental traumatic brain injury: relations with family resources and social outcomes. J Int Neuropsychol Soc 2013;19(5):497–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ewing-Cobbs L, Miner ME, Fletcher JM, Levin HS. Intellectual, motor, and language sequelae following closed head injury in infants and preschoolers. J Pediatr Psychol 1989;14(4):531–547. [DOI] [PubMed] [Google Scholar]
  • 26.Arndt DH, Lerner JT, Matsumoto JH, et al. Subclinical early posttraumatic seizures detected by continuous EEG monitoring in a consecutive pediatric cohort. Epilepsia 2013;54(10):1780–1788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hasbani DM, Topjian AA, Friess SH, et al. Nonconvulsive electrographic seizures are common in children with abusive head trauma*. Pediatr Crit Care Med 2013;14(7):709–715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Zhang Z, Saraswati M, Koehler RC, Robertson C, Kannan S. A New Rabbit Model of Pediatric Traumatic Brain Injury. J Neurotrauma 2015;32(17):1369–1379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Adelson PD, Fellows-Mayle W, Kochanek PM, Dixon CE. Morris water maze function and histologic characterization of two age-at-injury experimental models of controlled cortical impact in the immature rat. Childs Nerv Syst 2013;29(1):43–53. [DOI] [PubMed] [Google Scholar]
  • 30.Pullela R, Raber J, Pfankuch T, et al. Traumatic injury to the immature brain results in progressive neuronal loss, hyperactivity and delayed cognitive impairments. Dev Neurosci 2006;28(4–5):396–409. [DOI] [PubMed] [Google Scholar]
  • 31.Gurkoff GG, Giza CC, Hovda DA. Lateral fluid percussion injury in the developing rat causes an acute, mild behavioral dysfunction in the absence of significant cell death. Brain Res 2006;1077(1):24–36. [DOI] [PubMed] [Google Scholar]
  • 32.Friess SH, Ichord RN, Owens K, et al. Neurobehavioral functional deficits following closed head injury in the neonatal pig. Exp Neurol 2007;204(1):234–243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Adelson PD, Dixon CE, Robichaud P, Kochanek PM. Motor and cognitive functional deficits following diffuse traumatic brain injury in the immature rat. J Neurotrauma 1997;14(2):99–108. [DOI] [PubMed] [Google Scholar]
  • 34.Adelson PD, Dixon CE, Kochanek PM. Long-term dysfunction following diffuse traumatic brain injury in the immature rat. J Neurotrauma 2000;17(4):273–282. [DOI] [PubMed] [Google Scholar]
  • 35.Raghupathi R, Huh JW. Diffuse brain injury in the immature rat: evidence for an age-at-injury effect on cognitive function and histopathologic damage. J Neurotrauma 2007;24(10):1596–1608. [DOI] [PubMed] [Google Scholar]
  • 36.Paterno R, Folweiler KA, Cohen AS. Pathophysiology and Treatment of Memory Dysfunction After Traumatic Brain Injury. Curr Neurol Neurosci Rep 2017;17(7):52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Semple BD, Canchola SA, Noble-Haeusslein LJ. Deficits in social behavior emerge during development after pediatric traumatic brain injury in mice. J Neurotrauma 2012;29(17):2672–2683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Semple BD, O’Brien TJ, Gimlin K, et al. Interleukin-1 Receptor in Seizure Susceptibility after Traumatic Injury to the Pediatric Brain. J Neurosci 2017;37(33):7864–7877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Statler KD, Scheerlinck P, Pouliot W, Hamilton M, White HS, Dudek FE. A potential model of pediatric posttraumatic epilepsy. Epilepsy Res 2009;86(2–3):221–223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Gurkoff GG, Giza CC, Shin D, Auvin S, Sankar R, Hovda DA. Acute neuroprotection to pilocarpine-induced seizures is not sustained after traumatic brain injury in the developing rat. Neuroscience 2009;164(2):862–876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Andriessen TM, Jacobs B, Vos PE. Clinical characteristics and pathophysiological mechanisms of focal and diffuse traumatic brain injury. J Cell Mol Med 2010;14(10):2381–2392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Margulies SS, Thibault KL. Infant skull and suture properties: measurements and implications for mechanisms of pediatric brain injury. J Biomech Eng 2000;122(4):364–371. [DOI] [PubMed] [Google Scholar]
  • 43.Ewing-Cobbs L, Prasad M, Kramer L, et al. Acute neuroradiologic findings in young children with inflicted or noninflicted traumatic brain injury. Child’s nervous system : ChNS : official journal of the International Society for Pediatric Neurosurgery 2000;16(1):25–33; discussion 34. [DOI] [PubMed] [Google Scholar]
  • 44.Keenan HT, Runyan DK, Marshall SW, Nocera MA, Merten DF. A population-based comparison of clinical and outcome characteristics of young children with serious inflicted and noninflicted traumatic brain injury. Pediatrics 2004;114(3):633–639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Wilde EA, Hunter JV, Newsome MR, et al. Frontal and temporal morphometric findings on MRI in children after moderate to severe traumatic brain injury. J Neurotrauma 2005;22(3):333–344. [DOI] [PubMed] [Google Scholar]
  • 46.Ghosh A, Wilde EA, Hunter JV, et al. The relation between Glasgow Coma Scale score and later cerebral atrophy in paediatric traumatic brain injury. Brain Inj 2009;23(3):228–233. [DOI] [PubMed] [Google Scholar]
  • 47.Adelson PD, Robichaud P, Hamilton RL, Kochanek PM. A model of diffuse traumatic brain injury in the immature rat. J Neurosurg 1996;85(5):877–884. [DOI] [PubMed] [Google Scholar]
  • 48.Bittigau P, Sifringer M, Pohl D, et al. Apoptotic neurodegeneration following trauma is markedly enhanced in the immature brain. Ann Neurol 1999;45(6):724–735. [DOI] [PubMed] [Google Scholar]
  • 49.Huh JW, Raghupathi R. Chronic cognitive deficits and long-term histopathological alterations following contusive brain injury in the immature rat. J Neurotrauma 2007;24(9):1460–1474. [DOI] [PubMed] [Google Scholar]
  • 50.Huh JW, Widing AG, Raghupathi R. Midline brain injury in the immature rat induces sustained cognitive deficits, bihemispheric axonal injury and neurodegeneration. Exp Neurol 2008;213(1):84–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Eucker SA, Smith C, Ralston J, Friess SH, Margulies SS. Physiological and histopathological responses following closed rotational head injury depend on direction of head motion. Exp Neurol 2011;227(1):79–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Raghupathi R, Margulies SS. Traumatic axonal injury after closed head injury in the neonatal pig. J Neurotrauma 2002;19(7):843–853. [DOI] [PubMed] [Google Scholar]
  • 53.Adelson PD, Jenkins LW, Hamilton RL, Robichaud P, Tran MP, Kochanek PM. Histopathologic response of the immature rat to diffuse traumatic brain injury. J Neurotrauma 2001;18(10):967–976. [DOI] [PubMed] [Google Scholar]
  • 54.Wilde EA, Chu Z, Bigler ED, et al. Diffusion tensor imaging in the corpus callosum in children after moderate to severe traumatic brain injury. J Neurotrauma 2006;23(10):1412–1426. [DOI] [PubMed] [Google Scholar]
  • 55.Konigs M, Pouwels PJ, Ernest van Heurn LW, et al. Relevance of neuroimaging for neurocognitive and behavioral outcome after pediatric traumatic brain injury. Brain Imaging Behav 2018;12(1):29–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Dennis EL, Rashid F, Ellis MU, et al. Diverging white matter trajectories in children after traumatic brain injury: The RAPBI study. Neurology 2017;88(15):1392–1399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Dennis EL, Ellis MU, Marion SD, et al. Callosal Function in Pediatric Traumatic Brain Injury Linked to Disrupted White Matter Integrity. J Neurosci 2015;35(28):10202–10211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Yuan W, Holland SK, Schmithorst VJ, et al. Diffusion tensor MR imaging reveals persistent white matter alteration after traumatic brain injury experienced during early childhood. AJNR Am J Neuroradiol 2007;28(10):1919–1925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wozniak JR, Krach L, Ward E, et al. Neurocognitive and neuroimaging correlates of pediatric traumatic brain injury: a diffusion tensor imaging (DTI) study. Arch Clin Neuropsychol 2007;22(5):555–568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Vowles GH, Scholtz CL, Cameron JM. Diffuse axonal injury in early infancy. J Clin Pathol 1987;40(2):185–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Shannon P, Smith CR, Deck J, Ang LC, Ho M, Becker L. Axonal injury and the neuropathology of shaken baby syndrome. Acta Neuropathol 1998;95(6):625–631. [DOI] [PubMed] [Google Scholar]
  • 62.Geddes JF, Hackshaw AK, Vowles GH, Nickols CD, Whitwell HL. Neuropathology of inflicted head injury in children. I. Patterns of brain damage. Brain 2001;124(Pt 7):1290–1298. [DOI] [PubMed] [Google Scholar]
  • 63.Geddes JF, Vowles GH, Hackshaw AK, Nickols CD, Scott IS, Whitwell HL. Neuropathology of inflicted head injury in children. II. Microscopic brain injury in infants. Brain 2001;124(Pt 7):1299–1306. [DOI] [PubMed] [Google Scholar]
  • 64.Gleckman AM, Bell MD, Evans RJ, Smith TW. Diffuse axonal injury in infants with nonaccidental craniocerebral trauma: enhanced detection by beta-amyloid precursor protein immunohistochemical staining. Arch Pathol Lab Med 1999;123(2):146–151. [DOI] [PubMed] [Google Scholar]
  • 65.Ajao DO, Pop V, Kamper JE, et al. Traumatic brain injury in young rats leads to progressive behavioral deficits coincident with altered tissue properties in adulthood. J Neurotrauma 2012;29(11):2060–2074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Adelson PD, Jenkins LW, Hamilton RL, Robichaud P, Tran MP, Kochanek PM. Histopathologic response of the immature rat to diffuse traumatic brain injury. J Neurotrauma 2001;18(10):967–976. [DOI] [PubMed] [Google Scholar]
  • 67.DiLeonardi AM, Huh JW, Raghupathi R. Impaired axonal transport and neurofilament compaction occur in separate populations of injured axons following diffuse brain injury in the immature rat. Brain Res 2009;1263:174–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Huh JW, Widing AG, Raghupathi R. Differential effects of injury severity on cognition and cellular pathology after contusive brain trauma in the immature rat. J Neurotrauma 2011;28(2):245–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Dileonardi AM, Huh JW, Raghupathi R. Differential effects of FK506 on structural and functional axonal deficits after diffuse brain injury in the immature rat. J Neuropathol Exp Neurol 2012;71(11):959–972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Hanlon LA, Raghupathi R, Huh JW. Differential effects of minocycline on microglial activation and neurodegeneration following closed head injury in the neonate rat. Exp Neurol 2017;290:1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Hanlon LA, Huh JW, Raghupathi R. Minocycline Transiently Reduces Microglia/Macrophage Activation but Exacerbates Cognitive Deficits Following Repetitive Traumatic Brain Injury in the Neonatal Rat. J Neuropathol Exp Neurol 2016;75(3):214–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Verger K, Junque C, Levin HS, et al. Correlation of atrophy measures on MRI with neuropsychological sequelae in children and adolescents with traumatic brain injury. Brain Inj 2001;15(3):211–221. [DOI] [PubMed] [Google Scholar]
  • 73.Suskauer SJ, Huisman TA. Neuroimaging in pediatric traumatic brain injury: current and future predictors of functional outcome. Dev Disabil Res Rev 2009;15(2):117–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Tong W, Igarashi T, Ferriero DM, Noble LJ. Traumatic brain injury in the immature mouse brain: characterization of regional vulnerability. Exp Neurol 2002;176(1):105–116. [DOI] [PubMed] [Google Scholar]
  • 75.Huh JW, Franklin MA, Widing AG, Raghupathi R. Regionally distinct patterns of calpain activation and traumatic axonal injury following contusive brain injury in immature rats. Dev Neurosci 2006;28(4–5):466–476. [DOI] [PubMed] [Google Scholar]
  • 76.Bell MJ, Kochanek PM, Doughty LA, et al. Interleukin-6 and interleukin-10 in cerebrospinal fluid after severe traumatic brain injury in children. J Neurotrauma 1997;14(7):451–457. [DOI] [PubMed] [Google Scholar]
  • 77.Buttram SD, Wisniewski SR, Jackson EK, et al. Multiplex assessment of cytokine and chemokine levels in cerebrospinal fluid following severe pediatric traumatic brain injury: effects of moderate hypothermia. J Neurotrauma 2007;24(11):1707–1717. [DOI] [PubMed] [Google Scholar]
  • 78.Berger RP, Heyes MP, Wisniewski SR, Adelson PD, Thomas N, Kochanek PM. Assessment of the macrophage marker quinolinic acid in cerebrospinal fluid after pediatric traumatic brain injury: insight into the timing and severity of injury in child abuse. J Neurotrauma 2004;21(9):1123–1130. [DOI] [PubMed] [Google Scholar]
  • 79.Chhor V, Moretti R, Le Charpentier T, et al. Role of microglia in a mouse model of paediatric traumatic brain injury. Brain Behav Immun 2017;63:197–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Robinson S, Winer JL, Berkner J, et al. Imaging and serum biomarkers reflecting the functional efficacy of extended erythropoietin treatment in rats following infantile traumatic brain injury. J Neurosurg Pediatr 2016;17(6):739–755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Gu X, Wei ZZ, Espinera A, et al. Pharmacologically induced hypothermia attenuates traumatic brain injury in neonatal rats. Exp Neurol 2015;267:135–142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Claus CP, Tsuru-Aoyagi K, Adwanikar H, et al. Age is a determinant of leukocyte infiltration and loss of cortical volume after traumatic brain injury. Dev Neurosci 2010;32(5–6):454–465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Pettus EH, Christman CW, Giebel ML, Povlishock JT. Traumatically induced altered membrane permeability: its relationship to traumatically induced reactive axonal change. J Neurotrauma 1994;11(5):507–522. [DOI] [PubMed] [Google Scholar]
  • 84.Pettus EH, Povlishock JT. Characterization of a distinct set of intra-axonal ultrastructural changes associated with traumatically induced alteration in axolemmal permeability. Brain Res 1996;722(1–2):1–11. [DOI] [PubMed] [Google Scholar]
  • 85.Saatman KE, Bozyczko-Coyne D, Marcy V, Siman R, McIntosh TK. Prolonged calpain-mediated spectrin breakdown occurs regionally following experimental brain injury in the rat. J Neuropathol Exp Neurol 1996;55(7):850–860. [DOI] [PubMed] [Google Scholar]
  • 86.Okonkwo DO, Pettus EH, Moroi J, Povlishock JT. Alteration of the neurofilament sidearm and its relation to neurofilament compaction occurring with traumatic axonal injury. Brain Res 1998;784(1–2):1–6. [DOI] [PubMed] [Google Scholar]
  • 87.Buki A, Siman R, Trojanowski JQ, Povlishock JT. The role of calpain-mediated spectrin proteolysis in traumatically induced axonal injury. J Neuropathol Exp Neurol 1999;58(4):365–375. [DOI] [PubMed] [Google Scholar]
  • 88.Stone JR, Singleton RH, Povlishock JT. Intra-axonal neurofilament compaction does not evoke local axonal swelling in all traumatically injured axons. Exp Neurol 2001;172(2):320–331. [DOI] [PubMed] [Google Scholar]
  • 89.Reeves TM, Phillips LL, Lee NN, Povlishock JT. Preferential neuroprotective effect of tacrolimus (FK506) on unmyelinated axons following traumatic brain injury. Brain Res 2007;1154:225–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Huh JW, Widing AG, Raghupathi R. Basic science; repetitive mild non-contusive brain trauma in immature rats exacerbates traumatic axonal injury and axonal calpain activation: a preliminary report. J Neurotrauma 2007;24(1):15–27. [DOI] [PubMed] [Google Scholar]
  • 91.Marmarou CR, Povlishock JT. Administration of the immunophilin ligand FK506 differentially attenuates neurofilament compaction and impaired axonal transport in injured axons following diffuse traumatic brain injury. Exp Neurol 2006;197(2):353–362. [DOI] [PubMed] [Google Scholar]
  • 92.Singleton RH, Stone JR, Okonkwo DO, Pellicane AJ, Povlishock JT. The immunophilin ligand FK506 attenuates axonal injury in an impact-acceleration model of traumatic brain injury. J Neurotrauma 2001;18(6):607–614. [DOI] [PubMed] [Google Scholar]
  • 93.Saatman KE, Creed J, Raghupathi R. Calpain as a therapeutic target in traumatic brain injury. Neurotherapeutics 2010;7(1):31–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Ruppel RA, Kochanek PM, Adelson PD, et al. Excitatory amino acid concentrations in ventricular cerebrospinal fluid after severe traumatic brain injury in infants and children: the role of child abuse. J Pediatr 2001;138(1):18–25. [DOI] [PubMed] [Google Scholar]
  • 95.Portera-Cailliau C, Price DL, Martin LJ. Excitotoxic neuronal death in the immature brain is an apoptosis-necrosis morphological continuum. J Comp Neurol 1997;378(1):70–87. [PubMed] [Google Scholar]
  • 96.Huh JW, Raghupathi R. New concepts in treatment of pediatric traumatic brain injury. Anesthesiol Clin 2009;27(2):213–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Wang KK. Calpain and caspase: can you tell the difference? Trends Neurosci 2000;23(1):20–26. [DOI] [PubMed] [Google Scholar]
  • 98.Schober ME, Requena DF, Davis LJ, et al. Alpha II Spectrin breakdown products in immature Sprague Dawley rat hippocampus and cortex after traumatic brain injury. Brain Res 2014;1574:105–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Aikman J, O’Steen B, Silver X, et al. Alpha-II-spectrin after controlled cortical impact in the immature rat brain. Dev Neurosci 2006;28(4–5):457–465. [DOI] [PubMed] [Google Scholar]
  • 100.Rao VL, Dogan A, Todd KG, Bowen KK, Dempsey RJ. Neuroprotection by memantine, a non-competitive NMDA receptor antagonist after traumatic brain injury in rats. Brain Res 2001;911(1):96–100. [DOI] [PubMed] [Google Scholar]
  • 101.Belayev L, Alonso OF, Liu Y, et al. Talampanel, a novel noncompetitive AMPA antagonist, is neuroprotective after traumatic brain injury in rats. J Neurotrauma 2001;18(10):1031–1038. [DOI] [PubMed] [Google Scholar]
  • 102.Sonmez A, Sayin O, Gurgen SG, Calisir M. Neuroprotective effects of MK-801 against traumatic brain injury in immature rats. Neurosci Lett 2015;597:137–142. [DOI] [PubMed] [Google Scholar]
  • 103.Pohl D, Bittigau P, Ishimaru MJ, et al. N-Methyl-D-aspartate antagonists and apoptotic cell death triggered by head trauma in developing rat brain. Proc Natl Acad Sci U S A 1999;96(5):2508–2513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Giza CC, Mink RB, Madikians A. Pediatric traumatic brain injury: not just little adults. Curr Opin Crit Care 2007;13(2):143–152. [DOI] [PubMed] [Google Scholar]
  • 105.Sta Maria NS, Reger ML, Cai Y, et al. D-Cycloserine Restores Experience-Dependent Neuroplasticity after Traumatic Brain Injury in the Developing Rat Brain. J Neurotrauma 2017;34(8):1692–1702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Potts MB, Koh SE, Whetstone WD, et al. Traumatic injury to the immature brain: inflammation, oxidative injury, and iron-mediated damage as potential therapeutic targets. NeuroRx 2006;3(2):143–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Fukuda AM, Adami A, Pop V, et al. Posttraumatic reduction of edema with aquaporin-4 RNA interference improves acute and chronic functional recovery. J Cereb Blood Flow Metab 2013;33(10):1621–1632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Semple BD, Trivedi A, Gimlin K, Noble-Haeusslein LJ. Neutrophil elastase mediates acute pathogenesis and is a determinant of long-term behavioral recovery after traumatic injury to the immature brain. Neurobiol Dis 2015;74:263–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Moretti R, Chhor V, Bettati D, et al. Contribution of mast cells to injury mechanisms in a mouse model of pediatric traumatic brain injury. J Neurosci Res 2016;94(12):1546–1560. [DOI] [PubMed] [Google Scholar]
  • 110.Homsi S, Federico F, Croci N, et al. Minocycline effects on cerebral edema: relations with inflammatory and oxidative stress markers following traumatic brain injury in mice. Brain Res 2009;1291:122–132. [DOI] [PubMed] [Google Scholar]
  • 111.Homsi S, Piaggio T, Croci N, et al. Blockade of acute microglial activation by minocycline promotes neuroprotection and reduces locomotor hyperactivity after closed head injury in mice: a twelve-week follow-up study. J Neurotrauma 2010;27(5):911–921. [DOI] [PubMed] [Google Scholar]
  • 112.Siopi E, Cho AH, Homsi S, et al. Minocycline restores sAPPalpha levels and reduces the late histopathological consequences of traumatic brain injury in mice. J Neurotrauma 2011;28(10):2135–2143. [DOI] [PubMed] [Google Scholar]
  • 113.Siopi E, Llufriu-Daben G, Fanucchi F, Plotkine M, Marchand-Leroux C, Jafarian-Tehrani M. Evaluation of late cognitive impairment and anxiety states following traumatic brain injury in mice: the effect of minocycline. Neurosci Lett 2012;511(2):110–115. [DOI] [PubMed] [Google Scholar]
  • 114.Abdel Baki SG, Schwab B, Haber M, Fenton AA, Bergold PJ. Minocycline synergizes with N-acetylcysteine and improves cognition and memory following traumatic brain injury in rats. PLoS One 2010;5(8):e12490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Sangobowale MA, Grin’kina NM, Whitney K, et al. Minocycline plus N-Acetylcysteine Reduce Behavioral Deficits and Improve Histology with a Clinically Useful Time Window. J Neurotrauma 2018. [DOI] [PubMed]
  • 116.Lam TI, Bingham D, Chang TJ, et al. Beneficial effects of minocycline and botulinum toxin-induced constraint physical therapy following experimental traumatic brain injury. Neurorehabil Neural Repair 2013;27(9):889–899. [DOI] [PubMed] [Google Scholar]
  • 117.Salter MW, Beggs S. Sublime microglia: expanding roles for the guardians of the CNS. Cell 2014;158(1):15–24. [DOI] [PubMed] [Google Scholar]
  • 118.Schlegelmilch T, Henke K, Peri F. Microglia in the developing brain: from immunity to behaviour. Curr Opin Neurobiol 2011;21(1):5–10. [DOI] [PubMed] [Google Scholar]
  • 119.Harry GJ, Kraft AD. Microglia in the developing brain: a potential target with lifetime effects. Neurotoxicology 2012;33(2):191–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Loane DJ, Kumar A. Microglia in the TBI brain: The good, the bad, and the dysregulated. Exp Neurol 2016;275 Pt 3:316–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Margulies S, Anderson G, Atif F, et al. Combination Therapies for Traumatic Brain Injury: Retrospective Considerations. J Neurotrauma 2016;33(1):101–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Robertson CL, Fidan E, Stanley RM, Noje C, Bayir H. Progesterone for neuroprotection in pediatric traumatic brain injury. Pediatr Crit Care Med 2015;16(3):236–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Stein DG. Brain damage, sex hormones and recovery: a new role for progesterone and estrogen? Trends Neurosci 2001;24(7):386–391. [DOI] [PubMed] [Google Scholar]
  • 124.Wright DW, Yeatts SD, Silbergleit R, et al. Very early administration of progesterone for acute traumatic brain injury. N Engl J Med 2014;371(26):2457–2466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Stein DG. Embracing failure: What the Phase III progesterone studies can teach about TBI clinical trials. Brain Inj 2015;29(11):1259–1272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Schumacher M, Denier C, Oudinet JP, Adams D, Guennoun R. Progesterone neuroprotection: The background of clinical trial failure. J Steroid Biochem Mol Biol 2016;160:53–66. [DOI] [PubMed] [Google Scholar]
  • 127.Robertson CL, Saraswati M. Progesterone protects mitochondrial function in a rat model of pediatric traumatic brain injury. J Bioenerg Biomembr 2015;47(1–2):43–51. [DOI] [PubMed] [Google Scholar]
  • 128.Mannix R, Berglass J, Berkner J, et al. Sex differences in the effect of progesterone after controlled cortical impact in adolescent mice: a preliminary study. J Neurosurg 2014;121(6):1337–1341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Geddes RI, Sribnick EA, Sayeed I, Stein DG. Progesterone treatment shows benefit in a pediatric model of moderate to severe bilateral brain injury. PLoS One 2014;9(1):e87252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Geddes RI, Peterson BL, Stein DG, Sayeed I. Progesterone Treatment Shows Benefit in Female Rats in a Pediatric Model of Controlled Cortical Impact Injury. PLoS One 2016;11(1):e0146419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Ohls RK, Kamath-Rayne BD, Christensen RD, et al. Cognitive outcomes of preterm infants randomized to darbepoetin, erythropoietin, or placebo. Pediatrics 2014;133(6):1023–1030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Messier AM, Ohls RK. Neuroprotective effects of erythropoiesis-stimulating agents in term and preterm neonates. Curr Opin Pediatr 2014;26(2):139–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Brines M, Cerami A. Emerging biological roles for erythropoietin in the nervous system. Nat Rev Neurosci 2005;6(6):484–494. [DOI] [PubMed] [Google Scholar]
  • 134.Peng W, Xing Z, Yang J, Wang Y, Wang W, Huang W. The efficacy of erythropoietin in treating experimental traumatic brain injury: a systematic review of controlled trials in animal models. J Neurosurg 2014;121(3):653–664. [DOI] [PubMed] [Google Scholar]
  • 135.Schober ME, Requena DF, Block B, et al. Erythropoietin improved cognitive function and decreased hippocampal caspase activity in rat pups after traumatic brain injury. J Neurotrauma 2014;31(4):358–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Schober ME, Requena DF, Rodesch CK. EPO improved neurologic outcome in rat pups late after traumatic brain injury. Brain Dev 2018;40(5):367–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Prins ML, Matsumoto JH. The collective therapeutic potential of cerebral ketone metabolism in traumatic brain injury. J Lipid Res 2014;55(12):2450–2457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Prins ML, Fujima LS, Hovda DA. Age-dependent reduction of cortical contusion volume by ketones after traumatic brain injury. J Neurosci Res 2005;82(3):413–420. [DOI] [PubMed] [Google Scholar]
  • 139.Appelberg KS, Hovda DA, Prins ML. The effects of a ketogenic diet on behavioral outcome after controlled cortical impact injury in the juvenile and adult rat. J Neurotrauma 2009;26(4):497–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Giza CC, Griesbach GS, Hovda DA. Experience-dependent behavioral plasticity is disturbed following traumatic injury to the immature brain. Behav Brain Res 2005;157(1):11–22. [DOI] [PubMed] [Google Scholar]
  • 141.Lu H, Kobilo T, Robertson C, Tong S, Celnik P, Pelled G. Transcranial magnetic stimulation facilitates neurorehabilitation after pediatric traumatic brain injury. Sci Rep 2015;5:14769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Scott C, McKinlay A, McLellan T, Britt E, Grace R, MacFarlane M. A comparison of adult outcomes for males compared to females following pediatric traumatic brain injury. Neuropsychology 2015;29(4):501–508. [DOI] [PubMed] [Google Scholar]
  • 143.Covassin T, Elbin RJ, Harris W, Parker T, Kontos A. The role of age and sex in symptoms, neurocognitive performance, and postural stability in athletes after concussion. Am J Sports Med 2012;40(6):1303–1312. [DOI] [PubMed] [Google Scholar]
  • 144.Armstead WM, Kiessling JW, Kofke WA, Vavilala MS. Impaired cerebral blood flow autoregulation during posttraumatic arterial hypotension after fluid percussion brain injury is prevented by phenylephrine in female but exacerbated in male piglets by extracellular signal-related kinase mitogen-activated protein kinase upregulation. Crit Care Med 2010;38(9):1868–1874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Friess SH, Bruins B, Kilbaugh TJ, Smith C, Margulies SS. Differing effects when using phenylephrine and norepinephrine to augment cerebral blood flow after traumatic brain injury in the immature brain. J Neurotrauma 2015;32(4):237–243. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

RESOURCES