Abstract
Traumatic brain injury (TBI) is a complex and often-devastating condition. This disease involves damage to cerebral structures: meninges (dura, arachnoid, pia), cerebral cortex, white matter tracts, and deeper structures (basal ganglia, brainstem), along with mechanisms including contusions, hematomas (epidural/subdural), diffuse axonal injury from shear forces, secondary edema compromising blood-brain barrier, and ischemia/hemorrhage caused by vascular disruption. The pathophysiological process of TBI above varies significantly among individuals. However, prevalent TBI treatments still focus on symptomatic management, such as surgical intervention represented by craniotomy, medical management represented by osmotic agents for cerebral edema, supportive care represented by oxygen therapy, and adjuvant therapies represented by hypothermia. Worse still, traditional therapies often yield unfavorable outcomes and indulge the potential onset of long-term neurodegenerative diseases (NDDs). On the other side, Glymphatic System (GS), discovered as a clearance system in the brain, has made tremendous progress over the past decade. Dysfunction of the GS has been implicated in various central nervous system (CNS) diseases including TBI. The discovery of the GS offers new perspectives for the pathophysiological process of TBI, particularly unveiling the truth of the development of diphasic brain edema following TBI. Impressively, with the GS maturing, unprecedented therapeutic strategies ensue. For instance, the GS might explain sleep deprivation after TBI strikes in part and strongly validate the prospect of sleep therapy, then provide insights into the enigma of sleep. Also, nor-adrenergic inhibition facilitates CSF-ISF exchange and glymphatic outflow, significantly attenuating brain edema. AQP4, the guardian and regulator of brain capacity at the end-foot of astrocyte, which can modulate its array and amounts aligning with nor-adrenergic signal, is indispensable in this process. Moreover, neurons have gained prominence in the brain’s clearance system. Exploring the relationship between the GS and TBI will likely to blaze the new trail for advancing our understanding of TBI.
This is a preview of subscription content, access via your institution
Access options
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout




Similar content being viewed by others
Data availability
Data sharing is not applicable to this article as no datasets were generated or analysed during the current study. However, relevant searching criteria pertinent to the specific part in this review are available from the corresponding author on reasonable request.
References
Maas AIR, Stocchetti N, Bullock R. Moderate and severe traumatic brain injury in adults. Lancet Neurol. 2008;7:728–41.
Brazinova A, Rehorcikova V, Taylor MS, Buckova V, Majdan M, Psota M, et al. Epidemiology of traumatic brain injury in europe: a living systematic review. J Neurotrauma. 2021;38:1411–40.
GBD 2016 Traumatic Brain Injury and Spinal Cord Injury Collaborators. Global, regional, and national burden of traumatic brain injury and spinal cord injury, 1990-2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019;18:56–87.
Simon DW, McGeachy MJ, Bayır H, Clark RSB, Loane DJ, Kochanek PM. The far-reaching scope of neuroinflammation after traumatic brain injury. Nat Rev Neurol. 2017;13:171–91.
Bramlett HM, Dietrich WD. Long-Term consequences of traumatic brain injury: current status of potential mechanisms of injury and neurological outcomes. J Neurotrauma. 2015;32:1834–48.
Maas AIR, Menon DK, Manley GT, Abrams M, Åkerlund C, Andelic N, et al. Traumatic brain injury: progress and challenges in prevention, clinical care, and research. Lancet Neurol. 2022;21:1004–60.
Robba C, Banzato E, Rebora P, Iaquaniello C, Huang C-Y, Wiegers EJA, et al. Acute kidney injury in traumatic brain injury patients: results from the collaborative european neurotrauma effectiveness research in traumatic brain injury study. Crit Care Med. 2021;49:112–26.
Cohan P, Wang C, McArthur DL, Cook SW, Dusick JR, Armin B, et al. Acute secondary adrenal insufficiency after traumatic brain injury: a prospective study. Crit Care Med. 2005;33:2358–66.
Krishnamoorthy V, Manley GT, Jain S, Sun S, Foreman B, Komisarow J, et al. Incidence and clinical impact of myocardial injury following traumatic brain injury: a pilot TRACK-TBI study. J Neurosurg Anesthesiol. 2022;34:233–7.
Krishnamoorthy V, Temkin N, Barber J, Foreman B, Komisarow J, Korley FK, et al. Association of early multiple organ dysfunction with clinical and functional outcomes over the year following traumatic brain injury: a transforming research and clinical knowledge in traumatic brain injury study. Critical Care Medicine. 2021;49:1769–78.
McAllister TW. Neurobiological consequences of traumatic brain injury. Dialogues Clin Neurosci. 2011;13:287–300.
Molteni E, Pagani E, Strazzer S, Arrigoni F, Beretta E, Boffa G, et al. Fronto-temporal vulnerability to disconnection in paediatric moderate and severe traumatic brain injury. Eur J Neurol. 2019;26:1183–90.
Missori P, Paolini S, Peschillo S, Mancarella C, Scafa AK, Rastelli E, et al. Temporal horn enlargements predict secondary hydrocephalus diagnosis earlier than Evans’ index. Tomography (Ann Arbor, Mich). 2022;8:1429–36.
Jassam YN, Izzy S, Whalen M, McGavern DB, El Khoury J. Neuroimmunology of traumatic brain injury: time for a Paradigm Shift. Neuron. 2017;95:1246–65.
Ladak AA, Enam SA, Ibrahim MT. A review of the molecular mechanisms of traumatic brain injury. World Neurosurg. 2019;131:126–32.
Hansen KB, Yi F, Perszyk RE, Furukawa H, Wollmuth LP, Gibb AJ, et al. Structure, function, and allosteric modulation of NMDA receptors. J Gen Physiol. 2018;150:1081–105.
Viviani B, Boraso M, Marchetti N, Marinovich M. Perspectives on neuroinflammation and excitotoxicity: a neurotoxic conspiracy? Neurotoxicology. 2014;43:10–20.
Witkin JM, Shafique H, Cerne R, Smith JL, Marini AM, Lipsky RH, et al. Mechanistic and therapeutic relationships of traumatic brain injury and γ-amino-butyric acid (GABA). Pharmacol Ther. 2024;256:108609.
Gonzalez-Fierro C, Fonte C, Dufourd E, Cazaentre V, Aydin S, Engelhardt B, et al. Effects of a small-molecule perforin inhibitor in a mouse model of CD8 T cell-mediated neuroinflammation. Neurol Neuroimmunol Neuroinflamm. 2023;10:e200117.
Spaethling JM, Klein DM, Singh P, Meaney DF. Calcium-permeable AMPA receptors appear in cortical neurons after traumatic mechanical injury and contribute to neuronal fate. J Neurotrauma. 2008;25:1207–16.
Guerriero RM, Giza CC, Rotenberg A. Glutamate and GABA imbalance following traumatic brain injury. Curr Neurol Neurosci Rep. 2015;15:27.
Mr D. Mitochondria, calcium-dependent neuronal death and neurodegenerative disease. Pflugers Archiv. 2012;464:111–21.
McGovern AJ, Barreto GE. Network pharmacology identifies IL6 as an important hub and target of tibolone for drug repurposing in traumatic brain injury. Biomed Pharmacother. 2021;140:11769.
Lucke-Wold BP, Logsdon AF, Nguyen L, Eltanahay A, Turner RC, Bonasso P, et al. Supplements, nutrition, and alternative therapies for the treatment of traumatic brain injury. Nutr Neurosci. 2018;21:79–91.
Barro C, Chitnis T, Weiner HL. Blood neurofilament light: a critical review of its application to neurologic disease. Ann Clin Transl Neurol. 2020;7:2508–23.
Petzold A. The 2022 Lady Estelle Wolfson lectureship on neurofilaments. J Neurochem. 2022;163:179–219.
Cooper DJ, Rosenfeld JV, Murray L, Arabi YM, Davies AR, D’Urso P, et al. Decompressive craniectomy in diffuse traumatic brain injury. N Engl J Med. 2011;364:1493–502.
van Essen TA, van Erp IAM, Lingsma HF, Pisică D, Yue JK, Singh RD, et al. Comparative effectiveness of decompressive craniectomy versus craniotomy for traumatic acute subdural hematoma (CENTER-TBI): an observational cohort study. EClinicalMedicine. 2023;63:102161.
Kolias AG, Adams H, Timofeev IS, Corteen EA, Hossain I, Czosnyka M, et al. Evaluation of outcomes among patients with traumatic intracranial hypertension treated with decompressive craniectomy vs standard medical care at 24 months: a secondary analysis of the RESCUEicp randomized clinical trial. JAMA Neurol. 2022;79:664–71.
Cooper DJ, Nichol AD, Bailey M, Bernard S, Cameron PA, Pili-Floury S, et al. Effect of early sustained prophylactic hypothermia on neurologic outcomes among patients with severe traumatic brain injury: the POLAR randomized clinical trial. JAMA. 2018;320:2211–20.
Olah E, Poto L, Rumbus Z, Pakai E, Romanovsky AA, Hegyi P, et al. POLAR study revisited: therapeutic hypothermia in severe brain trauma should not be abandoned. J Neurotrauma. 2021;38:2772–6.
Yan A, Torpey A, Morrisroe E, Andraous W, Costa A, Bergese S. Clinical management in traumatic brain injury. Biomedicines. 2024;12:781.
Postolache TT, Wadhawan A, Can A, Lowry CA, Woodbury M, Makkar H, et al. Inflammation in traumatic brain injury. J Alzheimers Dis. 2020;74:1–28.
Ye Z, Li Z, Zhong S, Xing Q, Li K, Sheng W, et al. The recent two decades of traumatic brain injury: a bibliometric analysis and systematic review. Int J Surg. 2024;110:3745–59. https://doi.org/10.1097/JS9.0000000000001367
Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci Transl Med. 2012;4:147ra111.
Hablitz LM, Nedergaard M. The glymphatic system: a novel component of fundamental neurobiology. J Neurosci. 2021;41:7698–711.
Aspelund A, Antila S, Proulx ST, Karlsen TV, Karaman S, Detmar M, et al. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J Exp Med. 2015;212:991–9.
Louveau A, Smirnov I, Keyes TJ, Eccles JD, Rouhani SJ, Peske JD, et al. Structural and functional features of central nervous system lymphatic vessels. Nature. 2015;523:337–41.
Jessen NA, Munk ASF, Lundgaard I, Nedergaard M. The glymphatic system: a beginner’s guide. Neurochem Res. 2015;40:2583–99.
Rasmussen MK, Mestre H, Nedergaard M. The glymphatic pathway in neurological disorders. Lancet Neurol. 2018;17:1016–24.
Plog BA, Nedergaard M. The glymphatic system in central nervous system health and disease: past, present, and future. Annu Rev Pathol. 2018;13:379–94.
Harrison IF, Ismail O, Machhada A, Colgan N, Ohene Y, Nahavandi P, et al. Impaired glymphatic function and clearance of tau in an Alzheimer’s disease model. Brain. 2020;143:2576–93.
Rasmussen MK, Mestre H, Nedergaard M. Fluid transport in the brain. Physiol Rev. 2022;102:1025–151.
Opel RA, Christy A, Boespflug EL, Weymann KB, Case B, Pollock JM, et al. Effects of traumatic brain injury on sleep and enlarged perivascular spaces. J Cereb Blood Flow Metab. 2019;39:2258–67.
Peng S, Liu J, Liang C, Yang L, Wang G. Aquaporin-4 in glymphatic system, and its implication for central nervous system disorders. Neurobiol Dis. 2023;179:106035.
Tian Y, Cai X, Zhou Y, Jin A, Wang S, Yang Y, et al. Impaired glymphatic system as evidenced by low diffusivity along perivascular spaces is associated with cerebral small vessel disease: a population-based study. Stroke Vasc Neurol. 2023;8:413–23.
Kamagata K, Andica C, Takabayashi K, Saito Y, Taoka T, Nozaki H, et al. Association of MRI indices of glymphatic system with amyloid deposition and cognition in mild cognitive impairment and Alzheimer disease. Neurology. 2022;99:e2648–e2660.
Zhang W, Zhou Y, Wang J, Gong X, Chen Z, Zhang X, et al. Glymphatic clearance function in patients with cerebral small vessel disease. Neuroimage. 2021;238:118257.
Shokri-Kojori E, Wang G-J, Wiers CE, Demiral SB, Guo M, Kim SW, et al. β-Amyloid accumulation in the human brain after one night of sleep deprivation. Proc Natl Acad Sci USA. 2018;115:4483–8.
Covington NV, Duff MC. Heterogeneity is a hallmark of traumatic brain injury, not a limitation: a new perspective on study design in rehabilitation research. Am J Speech Lang Pathol. 2021;30:974–85.
Piantino J, Lim MM, Newgard CD, Iliff J. Linking traumatic brain injury, sleep disruption and post-traumatic headache: a potential role for glymphatic pathway dysfunction. Curr Pain Headache Rep. 2019;23:62.
Park JH, Bae YJ, Kim JS, Jung WS, Choi JW, Roh TH, et al. Glymphatic system evaluation using diffusion tensor imaging in patients with traumatic brain injury. Neuroradiology. 2023;65:551–7.
Iliff JJ, Chen MJ, Plog BA, Zeppenfeld DM, Soltero M, Yang L, et al. Impairment of glymphatic pathway function promotes tau pathology after traumatic brain injury. J Neurosci. 2014;34:16180–93.
Sullan MJ, Asken BM, Jaffee MS, DeKosky ST, Bauer RM. Glymphatic system disruption as a mediator of brain trauma and chronic traumatic encephalopathy. Neurosci Biobehav Rev. 2018;84:316–24.
Chen K-H, Lee C-P, Yang Y-H, Yang Y-H, Chen C-M, Lu M-L, et al. Incidence of hydrocephalus in traumatic brain injury: a nationwide population-based cohort study. Medicine (Baltimore). 2019;98:e17568.
Lalou AD, Levrini V, Czosnyka M, Gergelé L, Garnett M, Kolias A, et al. Cerebrospinal fluid dynamics in non-acute post-traumatic ventriculomegaly. Fluids Barriers CNS. 2020;17:24.
Bodnar CN, Watson JB, Higgins EK, Quan N, Bachstetter AD. Inflammatory regulation of CNS barriers after traumatic brain injury: a tale directed by interleukin-1. Front Immunol. 2021;12:688254.
Podvin S, Gonzalez A-M, Miller MC, Dang X, Botfield H, Donahue JE, et al. Esophageal cancer related gene-4 is a choroid plexus-derived injury response gene: evidence for a biphasic response in early and late brain injury. PLoS One. 2011;6:e24609.
Johanson C, Stopa E, Baird A, Sharma H. Traumatic brain injury and recovery mechanisms: peptide modulation of periventricular neurogenic regions by the choroid plexus-CSF nexus. J Neural Transm (Vienna). 2011;118:115–33.
Ahluwalia M, Mcmichael H, Kumar M, Espinosa MP, Bosomtwi A, Lu Y, et al. Altered endocannabinoid metabolism compromises the brain-CSF barrier and exacerbates chronic deficits after traumatic brain injury in mice. Exp Neurol. 2023;361:114320.
Kaur C, Rathnasamy G, Ling E-A. The choroid plexus in healthy and diseased brain. J Neuropathol Exp Neurol. 2016;75:198–213.
Xiang J, Routhe LJ, Wilkinson DA, Hua Y, Moos T, Xi G, et al. The choroid plexus as a site of damage in hemorrhagic and ischemic stroke and its role in responding to injury. Fluids Barriers CNS. 2017;14:8.
Yasmin A, Pitkänen A, Andrade P, Paananen T, Gröhn O, Immonen R. Post-injury ventricular enlargement associates with iron in choroid plexus but not with seizure susceptibility nor lesion atrophy-6-month MRI follow-up after experimental traumatic brain injury. Brain Struct Funct. 2022;227:145–58.
Mestre H, Mori Y, Nedergaard M. The Brain’s glymphatic system: current controversies. Trends Neurosci. 2020;43:458–66.
Plog BA, Dashnaw ML, Hitomi E, Peng W, Liao Y, Lou N, et al. Biomarkers of traumatic injury are transported from brain to blood via the glymphatic system. J Neurosci. 2015;35:518–26.
Johnson VE, Weber MT, Xiao R, Cullen DK, Meaney DF, Stewart W, et al. Mechanical disruption of the blood-brain barrier following experimental concussion. Acta Neuropathol. 2018;135:711–26.
Thrane AS, Rangroo Thrane V, Nedergaard M. Drowning stars: reassessing the role of astrocytes in brain edema. Trends Neurosci. 2014;37:620–8.
Borha A, Chagnot A, Goulay R, Emery E, Vivien D, Gaberel T. Cranioplasty reverses dysfunction of the solutes distribution in the brain parenchyma after decompressive craniectomy. Neurosurgery. 2020;87:1064–9.
Wardlaw JM, Benveniste H, Nedergaard M, Zlokovic BV, Mestre H, Lee H, et al. Perivascular spaces in the brain: anatomy, physiology and pathology. Nat Rev Neurol. 2020;16:137–53.
Hussain R, Tithof J, Wang W, Cheetham-West A, Song W, Peng W, et al. Potentiating glymphatic drainage minimizes post-traumatic cerebral oedema. Nature. 2023;623:992–1000.
Patabendige A, Janigro D. The role of the blood-brain barrier during neurological disease and infection. Biochem Soc Trans. 2023;51:613–26.
Inglese M, Bomsztyk E, Gonen O, Mannon LJ, Grossman RI, Rusinek H. Dilated perivascular spaces: hallmarks of mild traumatic brain injury. AJNR Am J Neuroradiol. 2005;26:719–24.
Hicks AJ, Sinclair B, Shultz SR, Pham W, Silbert LC, Schwartz DL, et al. Associations of enlarged perivascular spaces with brain lesions, brain age, and clinical outcomes in chronic traumatic brain injury. Neurology. 2023;101:e63–e73.
Braun M, Sevao M, Keil SA, Gino E, Wang MX, Lee J, et al. Macroscopic changes in aquaporin-4 underlie blast traumatic brain injury-related impairment in glymphatic function. Brain. 2024;147:2214–29.
Sandsmark DK, Elliott JE, Lim MM. Sleep-wake disturbances after traumatic brain injury: synthesis of human and animal studies. Sleep. 2017;40:zsx044.
Zhuo J, Raghavan P, Shao M, Roys S, Liang X, Tchoquessi RLN, et al. Automatic quantification of enlarged perivascular space in patients with traumatic brain injury using super-resolution of T2-weighted images. J Neurotrauma. 2024;41:407–19.
Butler T, Zhou L, Ozsahin I, Wang XH, Garetti J, Zetterberg H, et al. Glymphatic clearance estimated using diffusion tensor imaging along perivascular spaces is reduced after traumatic brain injury and correlates with plasma neurofilament light, a biomarker of injury severity. Brain Commun. 2023;5:fcad134.
Beschorner R, Nguyen TD, Gözalan F, Pedal I, Mattern R, Schluesener HJ, et al. CD14 expression by activated parenchymal microglia/macrophages and infiltrating monocytes following human traumatic brain injury. Acta Neuropathol. 2002;103:541–9.
Bigler ED. Neuropsychological results and neuropathological findings at autopsy in a case of mild traumatic brain injury. J Int Neuropsychol Soc. 2004;10:794–806.
Buonora JE, Mousseau M, Jacobowitz DM, Lazarus RC, Yarnell AM, Olsen CH, et al. Autoimmune profiling reveals peroxiredoxin 6 as a candidate traumatic brain injury biomarker. J Neurotrauma. 2015;32:1805–14.
Aj S AM-C, Am, S RB. Cortical spreading depression closes paravascular space and impairs glymphatic flow: implications for migraine headache. J Neurosci. 2017;37:2904–15.
Xiang T, Feng D, Zhang X, Chen Y, Wang H, Liu X, et al. Effects of increased intracranial pressure on cerebrospinal fluid influx, cerebral vascular hemodynamic indexes, and cerebrospinal fluid lymphatic efflux. J Cereb Blood Flow Metab. 2022;42:2287–302.
Ren Z, Iliff JJ, Yang L, Yang J, Chen X, Chen MJ, et al. ‘Hit & Run’ model of closed-skull traumatic brain injury (TBI) reveals complex patterns of post-traumatic AQP4 dysregulation. J Cereb Blood Flow Metab. 2013;33:834–45.
Kitchen P, Salman MM, Halsey AM, Clarke-Bland C, MacDonald JA, Ishida H, et al. Targeting aquaporin-4 subcellular localization to treat central nervous system edema. Cell. 2020;181:784–.e19.
Shively SB, Horkayne-Szakaly I, Jones RV, Kelly JP, Armstrong RC, Perl DP. Characterisation of interface astroglial scarring in the human brain after blast exposure: a post-mortem case series. Lancet Neurol. 2016;15:944–53.
Pease M, Gupta K, Moshé SL, Correa DJ, Galanopoulou AS, Okonkwo DO, et al. Insights into epileptogenesis from post-traumatic epilepsy. Nat Rev Neurol. 2024;20:298–312.
Sharma R, Leung WL, Zamani A, O’Brien TJ, Casillas Espinosa PM, Semple BD. Neuroinflammation in post-traumatic epilepsy: pathophysiology and tractable therapeutic targets. Brain Sci. 2019;9:318.
MacAulay N. Molecular mechanisms of brain water transport. Nat Rev Neurosci. 2021;22:326–44.
Vizcarra VS, Fame RM, Hablitz LM. Circadian mechanisms in brain fluid biology. Circ Res. 2024;134:711–26.
Dadgostar E, Rahimi S, Nikmanzar S, Nazemi S, Naderi Taheri M, Alibolandi Z, et al. Aquaporin 4 in traumatic brain injury: from molecular pathways to therapeutic target. Neurochem Res. 2022;47:860–71.
Chen L-H, Zhang H-T, Xu R-X, Li W-D, Zhao H, Yang Y, et al. Interaction of aquaporin 4 and N-methyl-D-aspartate NMDA receptor 1 in traumatic brain injury of rats. Iran J Basic Med Sci. 2018;21:1148–54.
Szczygielski J, Glameanu C, Müller A, Klotz M, Sippl C, Hubertus V, et al. Changes in posttraumatic brain edema in craniectomy-selective brain hypothermia model are associated with modulation of aquaporin-4 level. Front Neurol. 2018;9:799.
Rodriguez-Grande B, Obenaus A, Ichkova A, Aussudre J, Bessy T, Barse E, et al. Gliovascular changes precede white matter damage and long-term disorders in juvenile mild closed head injury. Glia. 2018;66:1663–77.
Jia J, Chen F, Wu Y. Recombinant PEP-1-SOD1 improves functional recovery after neural stem cell transplantation in rats with traumatic brain injury. Exp Ther Med. 2018;15:2929–35.
Lopez-Rodriguez AB, Acaz-Fonseca E, Viveros M-P, Garcia-Segura LM. Changes in cannabinoid receptors, aquaporin 4 and vimentin expression after traumatic brain injury in adolescent male mice. Association with edema and neurological deficit. PLoS One. 2015;10:e0128782.
Laird MD, Shields JS, Sukumari-Ramesh S, Kimbler DE, Fessler RD, Shakir B, et al. High mobility group box protein-1 promotes cerebral edema after traumatic brain injury via activation of toll-like receptor 4. Glia. 2014;62:26–38.
Neri M, Frati A, Turillazzi E, Cantatore S, Cipolloni L, Di Paolo M, et al. Immunohistochemical evaluation of aquaporin-4 and its correlation with CD68, IBA-1, HIF-1α, GFAP, and CD15 expressions in fatal traumatic brain injury. Int J Mol Sci. 2018;19:3544.
Ding JY, Kreipke CW, Speirs SL, Schafer P, Schafer S, Rafols JA. Hypoxia-inducible factor-1alpha signaling in aquaporin upregulation after traumatic brain injury. Neurosci Lett. 2009;453:68–72.
Yang L, Chen Z, Wan X, Liu M, Wu J, Chen Y, et al. Angiotensin II type 1 receptor deficiency protects against the impairment of blood-brain barrier in a mouse model of traumatic brain injury. Int J Neurosci. 2023;133:604–11.
Linnerbauer M, Wheeler MA, Quintana FJ. Astrocyte crosstalk in CNS inflammation. Neuron. 2020;108:608–22.
Guilfoyle MR, Carpenter KLH, Helmy A, Pickard JD, Menon DK, Hutchinson PJA. Matrix metalloproteinase expression in contusional traumatic brain injury: a paired microdialysis study. J Neurotrauma. 2015;32:1553–9.
Si X, Dai S, Fang Y, Tang J, Wang Z, Li Y, et al. Matrix metalloproteinase-9 inhibition prevents aquaporin-4 depolarization-mediated glymphatic dysfunction in Parkinson’s disease. J Adv Res. 2024;56:125–36.
Zhang E, Wan X, Yang L, Wang D, Chen Z, Chen Y, et al. Omega-3 polyunsaturated fatty acids alleviate traumatic brain injury by regulating the glymphatic pathway in mice. Front Neurol. 2020;11:707.
Rosu G-C, Catalin B, Balseanu TA, Laurentiu M, Claudiu M, Kumar-Singh S, et al. Inhibition of aquaporin 4 decreases amyloid Aβ40 drainage around cerebral vessels. Mol Neurobiol. 2020;57:4720–34.
Liu X, Xie Y, Wan X, Wu J, Fan Z, Yang L. Protective effects of aquaporin-4 deficiency on longer-term neurological outcomes in a mouse model. Neurochem Res. 2021;46:1380–9.
Ciappelloni S, Bouchet D, Dubourdieu N, Boué-Grabot E, Kellermayer B, Manso C, et al. Aquaporin-4 surface trafficking regulates astrocytic process motility and synaptic activity in health and autoimmune disease. Cell Rep. 2019;27:3860–.e4.
Salman MM, Kitchen P, Iliff JJ, Bill RM. Aquaporin 4 and glymphatic flow have central roles in brain fluid homeostasis. Nat Rev Neurosci. 2021;22:650–1.
Huber VJ, Igarashi H, Ueki S, Kwee IL, Nakada T. Aquaporin-4 facilitator TGN-073 promotes interstitial fluid circulation within the blood-brain barrier: [17O]H2O JJVCPE MRI study. Neuroreport. 2018;29:697–703.
Bordone MP, Salman MM, Titus HE, Amini E, Andersen JV, Chakraborti B, et al. The energetic brain - A review from students to students. J Neurochem. 2019;151:139–65.
Shan F, Huang Z, Xiong R, Huang Q-Y, Li J. HIF1α-induced upregulation of KLF4 promotes migration of human vascular smooth muscle cells under hypoxia. J Cell Physiol. 2020;235:141–50.
Xiong A, Xiong R, Yu J, Liu Y, Liu K, Jin G, et al. Aquaporin-4 is a potential drug target for traumatic brain injury via aggravating the severity of brain edema. Burns Trauma. 2021;9:tkaa050.
Tang G, Yang G-Y. Aquaporin-4: a potential therapeutic target for cerebral edema. Int J Mol Sci. 2016;17:1413.
Xiong A, Li J, Xiong R, Xia Y, Jiang X, Cao F, et al. Inhibition of HIF-1α-AQP4 axis ameliorates brain edema and neurological functional deficits in a rat controlled cortical injury (CCI) model. Sci Rep. 2022;12:2701.
Marmarou A. A review of progress in understanding the pathophysiology and treatment of brain edema. Neurosurg Focus. 2007;22:E1.
Krueger M, Härtig W, Reichenbach A, Bechmann I, Michalski D. Blood-brain barrier breakdown after embolic stroke in rats occurs without ultrastructural evidence for disrupting tight junctions. PLoS One. 2013;8:e56419.
Kawoos U, Abutarboush R, Gu M, Chen Y, Statz JK, Goodrich SY, et al. Blast-induced temporal alterations in blood-brain barrier properties in a rodent model. Sci Rep. 2021;11:5906.
Main BS, Villapol S, Sloley SS, Barton DJ, Parsadanian M, Agbaegbu C, et al. Apolipoprotein E4 impairs spontaneous blood brain barrier repair following traumatic brain injury. Mol Neurodegener. 2018;13:17.
Wang Z-G, Cheng Y, Yu X-C, Ye L-B, Xia Q-H, Johnson NR, et al. bFGF protects against blood-brain barrier damage through junction protein regulation via PI3K-Akt-Rac1 pathway following traumatic brain injury. Mol Neurobiol. 2016;53:7298–311.
Feng D, Liu T, Zhang X, Xiang T, Su W, Quan W, et al. Fingolimod improves diffuse brain injury by promoting AQP4 polarization and functional recovery of the glymphatic system. CNS Neurosci Ther. 2024;30:e14669.
Eisenbaum M, Pearson A, Ortiz C, Koprivica M, Cembran A, Mullan M, et al. Repetitive head trauma and apoE4 induce chronic cerebrovascular alterations that impair tau elimination from the brain. Exp Neurol. 2024;374:114702.
Clark AT, Abrahamson EE, Harper MM, Ikonomovic MD. Chronic effects of blast injury on the microvasculature in a transgenic mouse model of Alzheimer’s disease related Aβ amyloidosis. Fluids Barriers CNS. 2022;19:5.
Jha RM, Kochanek PM, Simard JM. Pathophysiology and treatment of cerebral edema in traumatic brain injury. Neuropharmacology. 2019;145:230–46.
McKee AC, Cairns NJ, Dickson DW, Folkerth RD, Keene CD, Litvan I, et al. The first NINDS/NIBIB consensus meeting to define neuropathological criteria for the diagnosis of chronic traumatic encephalopathy. Acta Neuropathol. 2016;131:75–86.
Jain A, Ang PS, Matrongolo MJ, Tischfield MA. Understanding the development, pathogenesis, and injury response of meningeal lymphatic networks through the use of animal models. Cell Mol Life Sci. 2023;80:332.
Bolte AC, Shapiro DA, Dutta AB, Ma WF, Bruch KR, Kovacs MA, et al. The meningeal transcriptional response to traumatic brain injury and aging. Elife. 2023;12:e81154.
Shimada R, Tatara Y, Kibayashi K. Gene expression in meningeal lymphatic endothelial cells following traumatic brain injury in mice. PLoS One. 2022;17:e0273892.
Bolte AC, Dutta AB, Hurt ME, Smirnov I, Kovacs MA, McKee CA, et al. Meningeal lymphatic dysfunction exacerbates traumatic brain injury pathogenesis. Nat Commun. 2020;11:4524.
Koh BI, Lee HJ, Kwak PA, Yang MJ, Kim J-H, Kim H-S, et al. VEGFR2 signaling drives meningeal vascular regeneration upon head injury. Nat Commun. 2020;11:3866.
Liu M, Huang J, Liu T, Yuan J, Lv C, Sha Z, et al. Exogenous interleukin 33 enhances the brain’s lymphatic drainage and toxic protein clearance in acute traumatic brain injury mice. Acta Neuropathol Commun. 2023;11:61.
Liao J, Zhang M, Shi Z, Lu H, Wang L, Fan W, et al. Improving the function of meningeal lymphatic vessels to promote brain edema absorption after traumatic brain injury. J Neurotrauma. 2023;40:383–94.
Nonomura K, Lukacs V, Sweet DT, Goddard LM, Kanie A, Whitwam T, et al. Mechanically activated ion channel PIEZO1 is required for lymphatic valve formation. Proc Natl Acad Sci USA. 2018;115:12817–22.
Choi D, Park E, Jung E, Cha B, Lee S, Yu J, et al. Piezo1 incorporates mechanical force signals into the genetic program that governs lymphatic valve development and maintenance. JCI Insight. 2019;4:e125068.
Urner S, Planas-Paz L, Hilger LS, Henning C, Branopolski A, Kelly-Goss M, et al. Identification of ILK as a critical regulator of VEGFR3 signalling and lymphatic vascular growth. EMBO J. 2019;38:e99322.
Song E, Mao T, Dong H, Boisserand LSB, Antila S, Bosenberg M, et al. VEGF-C-driven lymphatic drainage enables immunosurveillance of brain tumours. Nature. 2020;577:689–94.
Ma Q, Ineichen BV, Detmar M, Proulx ST. Outflow of cerebrospinal fluid is predominantly through lymphatic vessels and is reduced in aged mice. Nature Communications. 2017;8:1434.
Ahn JH, Cho H, Kim JH, Kim SH, Ham JS, Park I, et al. Meningeal lymphatic vessels at the skull base drain cerebrospinal fluid. Nature. 2019;572:62–66.
Da Mesquita S, Louveau A, Vaccari A, Smirnov I, Cornelison RC, Kingsmore KM, et al. Functional aspects of meningeal lymphatics in ageing and Alzheimer’s disease. Nature. 2018;560:185–91.
Bolte AC, Lukens JR. Neuroimmune cleanup crews in brain injury. Trends Immunol. 2021;42:480–94.
Iliff JJ, Lee H, Yu M, Feng T, Logan J, Nedergaard M, et al. Brain-wide pathway for waste clearance captured by contrast-enhanced MRI. J Clin Invest. 2013;123:1299–309.
Temmel AFP, Quint C, Schickinger-Fischer B, Klimek L, Stoller E, Hummel T. Characteristics of olfactory disorders in relation to major causes of olfactory loss. Arch Otolaryngol Head Neck Surg. 2002;128:635–41.
Moran DT, Jafek BW, Rowley JC, Eller PM. Electron microscopy of olfactory epithelia in two patients with anosmia. Arch Otolaryngol. 1985;111:122–6.
Jafek BW, Eller PM, Esses BA, Moran DT. Post-traumatic anosmia. Ultrastructural correlates. Arch Neurol. 1989;46:300–4.
Kobayashi M, Costanzo RM. Olfactory nerve recovery following mild and severe injury and the efficacy of dexamethasone treatment. Chem Senses. 2009;34:573–80.
Christensen J, Wright DK, Yamakawa GR, Shultz SR, Mychasiuk R. Repetitive mild traumatic brain injury alters glymphatic clearance rates in limbic structures of adolescent female rats. Sci Rep. 2020;10:6254.
Hsu M, Rayasam A, Kijak JA, Choi YH, Harding JS, Marcus SA, et al. Neuroinflammation-induced lymphangiogenesis near the cribriform plate contributes to drainage of CNS-derived antigens and immune cells. Nature Communications. 2019;10:229.
Brøchner CB, Holst CB, Møllgård K. Outer brain barriers in rat and human development. Front Neurosci. 2015;9:75.
Iadecola C. The neurovascular unit coming of age: a journey through neurovascular coupling in health and disease. Neuron. 2017;96:17–42.
Holstein-Rønsbo S, Gan Y, Giannetto MJ, Rasmussen MK, Sigurdsson B, Beinlich FRM, et al. Glymphatic influx and clearance are accelerated by neurovascular coupling. Nat Neurosci. 2023;26:1042–53.
Aboghazleh R, Parker E, Yang LT, Kaufer D, Dreier JP, Friedman A, et al. Brainstem and cortical spreading depolarization in a closed head injury rat model. Int J Mol Sci. 2021;22:11642.
Dreier JP. The role of spreading depression, spreading depolarization and spreading ischemia in neurological disease. Nat Med. 2011;17:439–47.
Major S, Huo S, Lemale CL, Siebert E, Milakara D, Woitzik J, et al. Direct electrophysiological evidence that spreading depolarization-induced spreading depression is the pathophysiological correlate of the migraine aura and a review of the spreading depolarization continuum of acute neuronal mass injury. Geroscience. 2020;42:57–80.
Leao AAP. Further observations on the spreading depression of activity in the cerebral cortex. J Neurophysiol. 1947;10:409–14.
Mukherjee S, Mirzaee M, Tithof J. Quantifying the relationship between spreading depolarization and perivascular cerebrospinal fluid flow. Sci Rep. 2023;13:12405.
van Hameren G, Muradov J, Minarik A, Aboghazleh R, Orr S, Cort S, et al. Mitochondrial dysfunction underlies impaired neurovascular coupling following traumatic brain injury. Neurobiol Dis. 2023;186:106269.
Parker E, Aboghazleh R, Mumby G, Veksler R, Ofer J, Newton J, et al. Concussion susceptibility is mediated by spreading depolarization-induced neurovascular dysfunction. Brain. 2022;145:2049–63.
Mestre H, Du T, Sweeney AM, Liu G, Samson AJ, Peng W, et al. Cerebrospinal fluid influx drives acute ischemic tissue swelling. Science. 2020;367:eaax7171.
Andrew RD, Hartings JA, Ayata C, Brennan KC, Dawson-Scully KD, Farkas E, et al. The critical role of spreading depolarizations in early brain injury: consensus and contention. Neurocrit Care. 2022;37:83–101.
Longden TA, Dabertrand F, Koide M, Gonzales AL, Tykocki NR, Brayden JE, et al. Capillary K+-sensing initiates retrograde hyperpolarization to increase local cerebral blood flow. Nat Neurosci. 2017;20:717–26.
Farr H, David T. Models of neurovascular coupling via potassium and EET signalling. J Theor Biol. 2011;286:13–23.
Mughal A, Sackheim AM, Sancho M, Longden TA, Russell S, Lockette W, et al. Impaired capillary-to-arteriolar electrical signaling after traumatic brain injury. J Cereb Blood Flow Metab. 2021;41:1313–27.
Hamel E. Perivascular nerves and the regulation of cerebrovascular tone. J Appl Physiol (1985). 2006;100:1059–64.
Elder GA, Gama Sosa MA, De Gasperi R, Perez Garcia G, Perez GM, Abutarboush R, et al. The neurovascular unit as a locus of injury in low-level blast-induced neurotrauma. Int J Mol Sci. 2024;25:1150.
Akbar MN, Ruf SF, Singh A, Faghihpirayesh R, Garner R, Bennett A, et al. Advancing post-traumatic seizure classification and biomarker identification: Information decomposition based multimodal fusion and explainable machine learning with missing neuroimaging data. Comput Med Imaging Graph. 2024;115:102386.
Tran CHT, George AG, Teskey GC, Gordon GR. Seizures elevate gliovascular unit Ca2+ and cause sustained vasoconstriction. JCI Insight. 2020;5:e136469.
Zhang H, Roman RJ, Fan F. Hippocampus is more susceptible to hypoxic injury: has the Rosetta Stone of regional variation in neurovascular coupling been deciphered? Geroscience. 2022;44:127–30.
Shaw K, Bell L, Boyd K, Grijseels DM, Clarke D, Bonnar O, et al. Neurovascular coupling and oxygenation are decreased in hippocampus compared to neocortex because of microvascular differences. Nat Commun. 2021;12:3190.
Shao J, Zhu H, Yao H, Stallones L, Yeates K, Wheeler K, et al. Characteristics and trends of pediatric traumatic brain injuries treated at a large pediatric medical center in China, 2002-2011. PLoS One. 2012;7:e51634.
Hawryluk GWJ, Rubiano AM, Totten AM, O’Reilly C, Ullman JS, Bratton SL, et al. Guidelines for the management of severe traumatic brain injury: 2020 update of the decompressive craniectomy recommendations. Neurosurgery. 2020;87:427–34.
Zhang D, Xue Q, Chen J, Dong Y, Hou L, Jiang Y, et al. Decompressive craniectomy in the management of intracranial hypertension after traumatic brain injury: a systematic review and meta-analysis. Sci Rep. 2017;7:8800.
Walcott BP, Nahed BV, Sheth SA, Yanamadala V, Caracci JR, Asaad WF. Bilateral hemicraniectomy in non-penetrating traumatic brain injury. J Neurotrauma. 2012;29:1879–85.
Hutchinson PJ, Kolias AG, Timofeev IS, Corteen EA, Czosnyka M, Timothy J, et al. Trial of decompressive craniectomy for traumatic intracranial hypertension. N Engl J Med. 2016;375:1119–30.
Plog BA, Lou N, Pierre CA, Cove A, Kenney HM, Hitomi E, et al. When the air hits your brain: decreased arterial pulsatility after craniectomy leading to impaired glymphatic flow. J Neurosurg. 2020;133:210–23.
Bai Y, Yuan M, Mi H, Zhang F, Liu X, Lu C, et al. Hypothermia reduces glymphatic transportation in traumatic edematous brain assessed by intrathecal dynamic contrast-enhanced MRI. Front Neurol. 2022;13:957055.
Gu W, Bai Y, Cai J, Mi H, Bao Y, Zhao X, et al. Hypothermia impairs glymphatic drainage in traumatic brain injury as assessed by dynamic contrast-enhanced MRI with intrathecal contrast. Front Neurosci. 2023;17:1061039.
Eide PK, Vinje V, Pripp AH, Mardal K-A, Ringstad G. Sleep deprivation impairs molecular clearance from the human brain. Brain. 2021;144:863–74.
Xie L, Kang H, Xu Q, Chen MJ, Liao Y, Thiyagarajan M, et al. Sleep drives metabolite clearance from the adult brain. Science. 2013;342:373–7.
Sherpa AD, Xiao F, Joseph N, Aoki C, Hrabetova S. Activation of β-adrenergic receptors in rat visual cortex expands astrocytic processes and reduces extracellular space volume. Synapse. 2016;70:307–16.
Zepernick A-L, Metodieva V, Pelegrina-Hidalgo N, Lippert AH, Horrocks MH, Varela JA. Single-molecule imaging of aquaporin-4 array dynamics in astrocytes. Nanoscale. 2024;16:9576–82.
Furman CS, Gorelick-Feldman DA, Davidson KGV, Yasumura T, Neely JD, Agre P, et al. Aquaporin-4 square array assembly: opposing actions of M1 and M23 isoforms. Proc Natl Acad Sci USA. 2003;100:13609–14.
de Bellis M, Cibelli A, Mola MG, Pisani F, Barile B, Mastrodonato M, et al. Orthogonal arrays of particle assembly are essential for normal aquaporin-4 expression level in the brain. Glia. 2021;69:473–88.
Zhu DD, Yang G, Huang YL, Zhang T, Sui AR, Li N, et al. AQP4-A25Q point mutation in mice depolymerizes orthogonal arrays of particles and decreases polarized expression of AQP4 protein in astrocytic endfeet at the blood-brain barrier. J Neurosci. 2022;42:8169–83.
Ding F, O’Donnell J, Xu Q, Kang N, Goldman N, Nedergaard M. Changes in the composition of brain interstitial ions control the sleep-wake cycle. Science. 2016;352:550–5.
Pantazopoulos H, Gisabella B, Rexrode L, Benefield D, Yildiz E, Seltzer P, et al. Circadian rhythms of perineuronal net composition. eNeuro. 2020;7:ENEURO.0034-19.2020.
Lohela TJ, Lilius TO, Nedergaard M. The glymphatic system: implications for drugs for central nervous system diseases. Nat Rev Drug Discov. 2022;21:763–79.
Jiang-Xie L-F, Drieu A, Bhasiin K, Quintero D, Smirnov I, Kipnis J. Neuronal dynamics direct cerebrospinal fluid perfusion and brain clearance. Nature. 2024;627:157–64.
Chen R, Gore F, Nguyen Q-A, Ramakrishnan C, Patel S, Kim SH, et al. Deep brain optogenetics without intracranial surgery. Nat Biotechnol. 2021;39:161–4.
Marshel JH, Kim YS, Machado TA, Quirin S, Benson B, Kadmon J, et al. Cortical layer-specific critical dynamics triggering perception. Science. 2019;365:eaaw5202.
Scammell TE, Arrigoni E, Lipton JO. Neural circuitry of wakefulness and sleep. Neuron. 2017;93:747–65.
Weber F, Dan Y. Circuit-based interrogation of sleep control. Nature. 2016;538:51–59.
Turner KL, Gheres KW, Proctor EA, Drew PJ. Neurovascular coupling and bilateral connectivity during NREM and REM sleep. Elife. 2020;9:e62071.
Harris KD, Thiele A. Cortical state and attention. Nat Rev Neurosci. 2011;12:509–23.
Fultz NE, Bonmassar G, Setsompop K, Stickgold RA, Rosen BR, Polimeni JR, et al. Coupled electrophysiological, hemodynamic, and cerebrospinal fluid oscillations in human sleep. Science. 2019;366:628–31.
Bojarskaite L, Vallet A, Bjørnstad DM, Gullestad Binder KM, Cunen C, Heuser K, et al. Sleep cycle-dependent vascular dynamics in male mice and the predicted effects on perivascular cerebrospinal fluid flow and solute transport. Nat Commun. 2023;14:953.
Hauglund NL, Andersen M, Tokarska K, Radovanovic T, Kjaerby C, Sørensen FL, et al. Norepinephrine-mediated slow vasomotion drives glymphatic clearance during sleep. Cell. 2025;188:606–622.e17.
Osorio-Forero A, Cardis R, Vantomme G, Guillaume-Gentil A, Katsioudi G, Devenoges C, et al. Noradrenergic circuit control of non-REM sleep substates. Current Biol. 2021;31:5009–5023.e7.
Kjaerby C, Andersen M, Hauglund N, Untiet V, Dall C, Sigurdsson B, et al. Memory-enhancing properties of sleep depend on the oscillatory amplitude of norepinephrine. Nat Neurosci. 2022;25:1059–70.
Hablitz LM, Vinitsky HS, Sun Q, Stæger FF, Sigurdsson B, Mortensen KN, et al. Increased glymphatic influx is correlated with high EEG delta power and low heart rate in mice under anesthesia. Sci Adv. 2019;5:eaav5447.
Benveniste H, Heerdt PM, Fontes M, Rothman DL, Volkow ND. Glymphatic system function in relation to anesthesia and sleep states. Anesth Analg. 2019;128:747–58.
Gao C, Qian Y, Huang J, Wang D, Su W, Wang P, et al. A three-day consecutive fingolimod administration improves neurological functions and modulates multiple immune responses of CCI mice. Mol Neurobiol. 2017;54:8348–60.
Yousaf M, Chang D, Liu Y, Liu T, Zhou X. Neuroprotection of cannabidiol, its synthetic derivatives and combination preparations against microglia-mediated neuroinflammation in neurological disorders. Molecules. 2022;27:4961.
Mannucci C, Navarra M, Calapai F, Spagnolo EV, Busardò FP, Cas RD, et al. Neurological aspects of medical use of cannabidiol. CNS Neurol Disord Drug Targets. 2017;16:541–53.
Dong S, Zhao H, Nie M, Sha Z, Feng J, Liu M, et al. Cannabidiol alleviates neurological deficits after traumatic brain injury by improving intracranial lymphatic drainage. J Neurotrauma. 2024;41:e2009–e2025. https://doi.org/10.1089/neu.2023.0539
Muscogiuri G, DeFronzo RA, Gastaldelli A, Holst JJ. Glucagon-like peptide-1 and the central/peripheral nervous system: crosstalk in diabetes. Trends Endocrinol Metab. 2017;28:88–103.
Lv C, Han S, Sha Z, Liu M, Dong S, Zhang C, et al. Cerebral glucagon-like peptide-1 receptor activation alleviates traumatic brain injury by glymphatic system regulation in mice. CNS Neurosci Ther. 2023;29:3876–88.
Stirling DP, Koochesfahani KM, Steeves JD, Tetzlaff W. Minocycline as a neuroprotective agent. Neuroscientist. 2005;11:308–22.
Lu Q, Xiong J, Yuan Y, Ruan Z, Zhang Y, Chai B, et al. Minocycline improves the functional recovery after traumatic brain injury via inhibition of aquaporin-4. Int J Biol Sci. 2022;18:441–58.
Wang Z, Nong J, Shultz RB, Zhang Z, Kim T, Tom VJ, et al. Local delivery of minocycline from metal ion-assisted self-assembled complexes promotes neuroprotection and functional recovery after spinal cord injury. Biomaterials. 2017;112:62–71.
Korhonen P, Kanninen KM, Lehtonen Š, Lemarchant S, Puttonen KA, Oksanen M, et al. Immunomodulation by interleukin-33 is protective in stroke through modulation of inflammation. Brain Behav Immun. 2015;49:322–36.
Fu AKY, Hung K-W, Yuen MYF, Zhou X, Mak DSY, Chan ICW, et al. IL-33 ameliorates Alzheimer’s disease-like pathology and cognitive decline. Proc Natl Acad Sci USA. 2016;113:E2705–2713.
Jiang H-R, Milovanović M, Allan D, Niedbala W, Besnard A-G, Fukada SY, et al. IL-33 attenuates EAE by suppressing IL-17 and IFN-γ production and inducing alternatively activated macrophages. Eur J Immunol. 2012;42:1804–14.
Liang C-S, Su K-P, Tsai C-L, Lee J-T, Chu C-S, Yeh T-C, et al. The role of interleukin-33 in patients with mild cognitive impairment and Alzheimer’s disease. Alzheimers Res. Ther. 2020;12:86.
Johansson PI, Sørensen AM, Perner A, Welling K-L, Wanscher M, Larsen CF, et al. Elderly trauma patients have high circulating noradrenaline levels but attenuated release of adrenaline, platelets, and leukocytes in response to increasing injury severity. Crit Care Med. 2012;40:1844–50.
Hamill RW, Woolf PD, McDonald JV, Lee LA, Kelly M. Catecholamines predict outcome in traumatic brain injury. Ann Neurol. 1987;21:438–43.
Rizoli SB, Jaja BNR, Di Battista AP, Rhind SG, Neto AC, da Costa L, et al. Catecholamines as outcome markers in isolated traumatic brain injury: the COMA-TBI study. Crit Care. 2017;21:37.
Mayer CL, Savage PJ, Engle CK, Groh SS, Shofer JB, Hargrove AM, et al. Randomized controlled pilot trial of prazosin for prophylaxis of posttraumatic headaches in active-duty service members and veterans. Headache. 2023;63:751–62.
George KC, Kebejian L, Ruth LJ, Miller CWT, Himelhoch S. Meta-analysis of the efficacy and safety of prazosin versus placebo for the treatment of nightmares and sleep disturbances in adults with posttraumatic stress disorder. J Trauma Dissociation. 2016;17:494–510.
Nissinen J, Andrade P, Natunen T, Hiltunen M, Malm T, Kanninen K, et al. Disease-modifying effect of atipamezole in a model of post-traumatic epilepsy. Epilepsy Res. 2017;136:18–34.
Carney N, Totten AM, O’Reilly C, Ullman JS, Hawryluk GWJ, Bell MJ, et al. Guidelines for the management of severe traumatic brain injury, fourth edition. Neurosurgery. 2017;80:6–15.
Eide PK, Ringstad G. Glymphatic-stagnated edema induced by traumatic brain injury. Trends Pharmacol Sci. 2024;45:388–90.
Meyfroidt G, Baguley IJ, Menon DK. Paroxysmal sympathetic hyperactivity: the storm after acute brain injury. Lancet Neurol. 2017;16:721–9.
Tschuor C, Asmis LM, Lenzlinger PM, Tanner M, Härter L, Keel M, et al. In vitro norepinephrine significantly activates isolated platelets from healthy volunteers and critically ill patients following severe traumatic brain injury. Crit Care. 2008;12:R80.
Rubenstein R, Chang B, Yue JK, Chiu A, Winkler EA, Puccio AM, et al. Comparing plasma phospho tau, total tau, and phospho tau-total tau ratio as acute and chronic traumatic brain injury biomarkers. JAMA Neurol. 2017;74:1063–72.
Jiang T, Xie L, Zhou S, Liu Y, Huang Y, Mei N, et al. Metformin and histone deacetylase inhibitor based anti-inflammatory nanoplatform for epithelial-mesenchymal transition suppression and metastatic tumor treatment. J Nanobiotechnol. 2022;20:394.
Tong S, Xie L, Xie X, Xu J, You Y, Sun Y, et al. Nano-Plumber reshapes glymphatic-lymphatic system to sustain microenvironment homeostasis and improve long-term prognosis after traumatic brain injury. Adv Sci. 2023;10:e2304284.
Melloni A, Liu L, Kashinath V, Abdi R, Shah K. Meningeal lymphatics and their role in CNS disorder treatment: moving past misconceptions. Front Neurosci. 2023;17:1184049.
Iaccarino HF, Singer AC, Martorell AJ, Rudenko A, Gao F, Gillingham TZ, et al. Gamma frequency entrainment attenuates amyloid load and modifies microglia. Nature. 2016;540:230–5.
Chan D, Suk H-J, Jackson BL, Milman NP, Stark D, Klerman EB, et al. Gamma frequency sensory stimulation in mild probable Alzheimer’s dementia patients: results of feasibility and pilot studies. PLoS One. 2022;17:e0278412.
Murdock MH, Yang C-Y, Sun N, Pao P-C, Blanco-Duque C, Kahn MC, et al. Multisensory gamma stimulation promotes glymphatic clearance of amyloid. Nature. 2024;627:149–56.
Blanco-Duque C, Chan D, Kahn MC, Murdock MH, Tsai L-H. Audiovisual gamma stimulation for the treatment of neurodegeneration. J Intern Med. 2024;295:146–70.
Martorell AJ, Paulson AL, Suk H-J, Abdurrob F, Drummond GT, Guan W, et al. Multi-sensory gamma stimulation ameliorates Alzheimer’s-Associated pathology and improves cognition. Cell. 2019;177:256–271.e22.
Mateo C, Knutsen PM, Tsai PS, Shih AY, Kleinfeld D. Entrainment of arteriole vasomotor fluctuations by neural activity is a basis of blood-oxygenation-level-dependent ‘Resting-State’ connectivity. Neuron. 2017;96:936–948.e3.
van Veluw SJ, Hou SS, Calvo-Rodriguez M, Arbel-Ornath M, Snyder AC, Frosch MP, et al. Vasomotion as a driving force for paravascular clearance in the awake mouse brain. Neuron. 2020;105:549–561.e5.
Aldea R, Weller RO, Wilcock DM, Carare RO, Richardson G. Cerebrovascular smooth muscle cells as the drivers of intramural periarterial drainage of the brain. Front Aging Neurosci. 2019;11:1.
Munting LP, Bonnar O, Kozberg MG, Auger CA, Hirschler L, Hou SS, et al. Spontaneous vasomotion propagates along pial arterioles in the awake mouse brain like stimulus-evoked vascular reactivity. J Cereb Blood Flow Metab. 2023;43:1752–63.
Kedarasetti RT, Drew PJ, Costanzo F. Arterial vasodilation drives convective fluid flow in the brain: a poroelastic model. Fluids Barriers CNS. 2022;19:34.
Albargothy NJ, Johnston DA, MacGregor-Sharp M, Weller RO, Verma A, Hawkes CA, et al. Convective influx/glymphatic system: tracers injected into the CSF enter and leave the brain along separate periarterial basement membrane pathways. Acta Neuropathol. 2018;136:139–52.
Mestre H, Tithof J, Du T, Song W, Peng W, Sweeney AM, et al. Flow of cerebrospinal fluid is driven by arterial pulsations and is reduced in hypertension. Nat Commun. 2018;9:4878.
Han G, Jiao B, Zhang Y, Wang Z, Liang C, Li Y, et al. Arterial pulsation dependence of perivascular cerebrospinal fluid flow measured by dynamic diffusion tensor imaging in the human brain. Neuroimage. 2024;297:120653.
Asgari M, de Zélicourt D, Kurtcuoglu V. Glymphatic solute transport does not require bulk flow. Sci Rep. 2016;6:38635.
Rey J, Sarntinoranont M. Pulsatile flow drivers in brain parenchyma and perivascular spaces: a resistance network model study. Fluids Barriers CNS. 2018;15:20.
Edwards G, Zhao J, Dash PK, Soto C, Moreno-Gonzalez I. Traumatic brain injury induces tau aggregation and spreading. J Neurotrauma. 2020;37:80–92.
Johnson VE, Stewart W, Smith DH. Traumatic brain injury and amyloid-β pathology: a link to Alzheimer’s disease? Nat Rev Neurosci. 2010;11:361–70.
Nedergaard M. Neuroscience. Garbage truck of the brain. Science. 2013;340:1529–30.
Brett BL, Gardner RC, Godbout J, Dams-O'Connor K, Keene CD. Traumatic brain injury and risk of neurodegenerative disorder. Biol Psychiatry. 2022;91:498–507.
Tran HT, LaFerla FM, Holtzman DM, Brody DL. Controlled cortical impact traumatic brain injury in 3xTg-AD mice causes acute intra-axonal amyloid-β accumulation and independently accelerates the development of tau abnormalities. J Neurosci. 2011;31:9513–25.
Albayram O, Kondo A, Mannix R, Smith C, Tsai C-Y, Li C, et al. Cis P-tau is induced in clinical and preclinical brain injury and contributes to post-injury sequelae. Nat Commun. 2017;8:1000.
Taoka T, Masutani Y, Kawai H, Nakane T, Matsuoka K, Yasuno F, et al. Evaluation of glymphatic system activity with the diffusion MR technique: diffusion tensor image analysis along the perivascular space (DTI-ALPS) in Alzheimer’s disease cases. Jpn J Radiol. 2017;35:172–8.
Huang SY, Zhang YR, Guo Y, Du J, Ren P, Wu BS, et al. Glymphatic system dysfunction predicts amyloid deposition, neurodegeneration, and clinical progression in Alzheimer’s disease. Alzheimers Dement. 2024;20:3251–69.
Obenaus A, Rodriguez-Grande B, Lee JB, Dubois CJ, Fournier M-L, Cador M, et al. A single mild juvenile TBI in male mice leads to regional brain tissue abnormalities at 12 months of age that correlate with cognitive impairment at the middle age. Acta Neuropathol Commun. 2023;11:32.
Ulv Larsen SM, Landolt H-P, Berger W, Nedergaard M, Knudsen GM, Holst SC. Haplotype of the astrocytic water channel AQP4 is associated with slow wave energy regulation in human NREM sleep. PLoS Biol. 2020;18:e3000623.
Wilson L, Stewart W, Dams-O’Connor K, Diaz-Arrastia R, Horton L, Menon DK, et al. The chronic and evolving neurological consequences of traumatic brain injury. Lancet Neurol. 2017;16:813–25.
Morissette MP, Prior HJ, Tate RB, Wade J, Leiter JRS. Associations between concussion and risk of diagnosis of psychological and neurological disorders: a retrospective population-based cohort study. Fam Med Community Health. 2020;8:e000390.
Gardner RC, Burke JF, Nettiksimmons J, Goldman S, Tanner CM, Yaffe K. Traumatic brain injury in later life increases risk for Parkinson disease. Ann Neurol. 2015;77:987–95.
Raj R, Kaprio J, Korja M, Mikkonen ED, Jousilahti P, Siironen J. Risk of hospitalization with neurodegenerative disease after moderate-to-severe traumatic brain injury in the working-age population: a retrospective cohort study using the Finnish national health registries. PLoS Med. 2017;14:e1002316.
Gardner RC, Byers AL, Barnes DE, Li Y, Boscardin J, Yaffe K. Mild TBI and risk of Parkinson disease: a Chronic Effects of Neurotrauma Consortium Study. Neurology. 2018;90:e1771–e1779.
Szlufik S, Kopeć K, Szleszkowski S, Koziorowski D. Glymphatic system pathology and neuroinflammation as two risk factors of neurodegeneration. Cells. 2024;13:286.
Song J, Li Z-H, Xue X-Y, Meng J-C, Zhu W-X, Hu S, et al. Neonatal stress disrupts the glymphatic system development and increases the susceptibility to Parkinson’s disease in later life. CNS Neurosci Ther. 2024;30:e14587.
Yue Y, Zhang X, Lv W, Lai HY, Shen T. Interplay between the glymphatic system and neurotoxic proteins in Parkinson’s disease and related disorders: current knowledge and future directions. Neural Regen Res. 2024;19:1973–80.
Pang H, Wang J, Yu Z, Yu H, Li X, Bu S, et al. Glymphatic function from diffusion-tensor MRI to predict conversion from mild cognitive impairment to dementia in Parkinson’s disease. J Neurol. 2024;271:5598–609. https://doi.org/10.1007/s00415-024-12525-8
Graham NS, Sharp DJ. Understanding neurodegeneration after traumatic brain injury: from mechanisms to clinical trials in dementia. J Neurol Neurosurg Psychiatry. 2019;90:1221–33.
LoBue C, Cullum CM, Didehbani N, Yeatman K, Jones B, Kraut MA, et al. Neurodegenerative dementias after traumatic brain injury. J Neuropsychiatry Clin Neurosci. 2018;30:7–13.
Crane PK, Gibbons LE, Dams-O’Connor K, Trittschuh E, Leverenz JB, Keene CD, et al. Association of traumatic brain injury with late-life neurodegenerative conditions and neuropathologic findings. JAMA Neurol. 2016;73:1062–9.
Eisen A, Nedergaard M, Gray E, Kiernan MC. The glymphatic system and Amyotrophic lateral sclerosis. Prog Neurobiol. 2024;234:102571.
Wu T-T, Su F-J, Feng Y-Q, Liu B, Li M-Y, Liang F-Y, et al. Mesenchymal stem cells alleviate AQP-4-dependent glymphatic dysfunction and improve brain distribution of antisense oligonucleotides in BACHD mice. Stem Cells. 2020;38:218–30.
Vittorini MG, Sahin A, Trojan A, Yusifli S, Alashvili T, Bonifácio GV, et al. The glymphatic system in migraine and other headaches. J Headache Pain. 2024;25:34.
Georgiopoulos C, Tisell A, Holmgren RT, Eleftheriou A, Rydja J, Lundin F, et al. Noninvasive assessment of glymphatic dysfunction in idiopathic normal pressure hydrocephalus with diffusion tensor imaging. J Neurosurg. 2024;140:612–20.
Kim M, Hwang I, Park JH, Chung JW, Kim SM, Kim J-H, et al. Comparative analysis of glymphatic system alterations in multiple sclerosis and neuromyelitis optica spectrum disorder using MRI indices from diffusion tensor imaging. Hum Brain Mapp. 2024;45:e26680.
Margoni M, Pagani E, Meani A, Preziosa P, Mistri D, Gueye M, et al. Cognitive impairment is related to glymphatic system dysfunction in pediatric multiple sclerosis. Ann Neurol. 2024;95:1080–92.
Lopes DM, Wells JA, Ma D, Wallis L, Park D, Llewellyn SK, et al. Glymphatic inhibition exacerbates tau propagation in an Alzheimer’s disease model. Alzheimer’s Res. Ther. 2024;16:71.
Lloyd-Donald P, Spencer W, Cheng J, Romero L, Jithoo R, Udy A, et al. In adult patients with severe traumatic brain injury, does the use of norepinephrine for augmenting cerebral perfusion pressure improve neurological outcome? A systematic review. Injury. 2020;51:2129–34.
Acknowledgements
This research was supported by National High Level Hospital Clinical Research Funding (2022-PUMCH-C-032), Science and Technology Major Project of Stroke Prevention and Treatment of the NHC-Million Disability Reduction Initiative (2024PSPT0903102), CAMS Innovation Fund for Medical Sciences (2024-I2M-C&T-B-022), Peking Union Medical College Hospital Postdoctoral Incubation Fund (kyfyjj202401), and Beijing Science and Technology Innovation Foundation for University students (2024dcxm047). The formats in this review were supported by biorender. The views expressed in this Review are those of the authors and do not necessarily represent the official policy or position of the Department of Neurosurgery, Peking Union Medical College Hospital. The funding source had no input in the conception or preparation of this Review.
Author information
Authors and Affiliations
Contributions
Data acquisition, Mechanism concept and diagram, review writing: ZYY; Revised version: PTL; Source of funds: JJW. All authors have read and approved the manuscript.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Ethics approval and consent to participate
All methods were performed in accordance with the relevant guidelines and regulations. All research described herein was conducted with approval from ethics committees, programmes for the protection of human subjects research, and institutional review boards at all participating sites. During our process of completing this review article, no human participants and data from humans are obtained.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Yang, Z., Li, P. & Wei, J. Perceiving traumatic brain injury from glymphatic system. Mol Psychiatry (2025). https://doi.org/10.1038/s41380-025-03126-6
Received:
Revised:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41380-025-03126-6