Skip to main content
Top
Published in: Fluids and Barriers of the CNS 1/2018

Open Access 01-12-2018 | Editorial

Progress in brain barriers and brain fluid research in 2017

Authors: Richard F. Keep, Hazel C. Jones, Lester R. Drewes

Published in: Fluids and Barriers of the CNS | Issue 1/2018

Login to get access

Abstract

The past year, 2017, has seen many important papers published in the fields covered by Fluids and Barriers of the CNS. This article from the Editors highlights some.
Literature
1.
go back to reference Keep RF, Jones HC, Drewes LR. Brain barriers and brain fluid research in 2016: advances, challenges and controversies. Fluids Barriers CNS. 2017;14(1):4.PubMedPubMedCentralCrossRef Keep RF, Jones HC, Drewes LR. Brain barriers and brain fluid research in 2016: advances, challenges and controversies. Fluids Barriers CNS. 2017;14(1):4.PubMedPubMedCentralCrossRef
2.
go back to reference Cho C, Smallwood PM, Nathans J. Reck and Gpr124 are essential receptor cofactors for Wnt7a/Wnt7b-specific signaling in mammalian CNS angiogenesis and blood–brain barrier regulation. Neuron. 2017;95(5):1056.e1055–1073.e1055.CrossRef Cho C, Smallwood PM, Nathans J. Reck and Gpr124 are essential receptor cofactors for Wnt7a/Wnt7b-specific signaling in mammalian CNS angiogenesis and blood–brain barrier regulation. Neuron. 2017;95(5):1056.e1055–1073.e1055.CrossRef
3.
go back to reference Chang J, Mancuso MR, Maier C, Liang X, Yuki K, Yang L, Kwong JW, Wang J, Rao V, Vallon M, et al. Gpr124 is essential for blood–brain barrier integrity in central nervous system disease. Nat Med. 2017;23(4):450–60.PubMedPubMedCentralCrossRef Chang J, Mancuso MR, Maier C, Liang X, Yuki K, Yang L, Kwong JW, Wang J, Rao V, Vallon M, et al. Gpr124 is essential for blood–brain barrier integrity in central nervous system disease. Nat Med. 2017;23(4):450–60.PubMedPubMedCentralCrossRef
4.
go back to reference Nakazato R, Kawabe K, Yamada D, Ikeno S, Mieda M, Shimba S, Hinoi E, Yoneda Y, Takarada T. Disruption of Bmal1 impairs blood–brain barrier integrity via pericyte dysfunction. J Neurosci. 2017;37(42):10052–62.PubMedCrossRef Nakazato R, Kawabe K, Yamada D, Ikeno S, Mieda M, Shimba S, Hinoi E, Yoneda Y, Takarada T. Disruption of Bmal1 impairs blood–brain barrier integrity via pericyte dysfunction. J Neurosci. 2017;37(42):10052–62.PubMedCrossRef
5.
go back to reference Villasenor R, Kuennecke B, Ozmen L, Ammann M, Kugler C, Gruninger F, Loetscher H, Freskgard PO, Collin L. Region-specific permeability of the blood–brain barrier upon pericyte loss. J Cereb Blood Flow Metab. 2017;37(12):3683–94.PubMedCrossRef Villasenor R, Kuennecke B, Ozmen L, Ammann M, Kugler C, Gruninger F, Loetscher H, Freskgard PO, Collin L. Region-specific permeability of the blood–brain barrier upon pericyte loss. J Cereb Blood Flow Metab. 2017;37(12):3683–94.PubMedCrossRef
6.
go back to reference Underly RG, Levy M, Hartmann DA, Grant RI, Watson AN, Shih AY. Pericytes as inducers of rapid, matrix metalloproteinase-9-dependent capillary damage during ischemia. J Neurosci. 2017;37(1):129–40.PubMedPubMedCentralCrossRef Underly RG, Levy M, Hartmann DA, Grant RI, Watson AN, Shih AY. Pericytes as inducers of rapid, matrix metalloproteinase-9-dependent capillary damage during ischemia. J Neurosci. 2017;37(1):129–40.PubMedPubMedCentralCrossRef
7.
go back to reference More VR, Campos CR, Evans RA, Oliver KD, Chan GN, Miller DS, Cannon RE. PPAR-alpha, a lipid-sensing transcription factor, regulates blood–brain barrier efflux transporter expression. J Cereb Blood Flow Metab. 2017;37(4):1199–212.PubMedCrossRef More VR, Campos CR, Evans RA, Oliver KD, Chan GN, Miller DS, Cannon RE. PPAR-alpha, a lipid-sensing transcription factor, regulates blood–brain barrier efflux transporter expression. J Cereb Blood Flow Metab. 2017;37(4):1199–212.PubMedCrossRef
8.
go back to reference Andreone BJ, Chow BW, Tata A, Lacoste B, Ben-Zvi A, Bullock K, Deik AA, Ginty DD, Clish CB, Gu C. Blood–brain barrier permeability is regulated by lipid transport-dependent suppression of caveolae-mediated transcytosis. Neuron. 2017;94(3):581.e585–594.e585.CrossRef Andreone BJ, Chow BW, Tata A, Lacoste B, Ben-Zvi A, Bullock K, Deik AA, Ginty DD, Clish CB, Gu C. Blood–brain barrier permeability is regulated by lipid transport-dependent suppression of caveolae-mediated transcytosis. Neuron. 2017;94(3):581.e585–594.e585.CrossRef
9.
go back to reference Vutukuri R, Brunkhorst R, Kestner RI, Hansen L, Bouzas NF, Pfeilschifter J, Devraj K, Pfeilschifter W. Alteration of sphingolipid metabolism as a putative mechanism underlying LPS-induced BBB disruption. J Neurochem. 2018;144(2):172–85.PubMedCrossRef Vutukuri R, Brunkhorst R, Kestner RI, Hansen L, Bouzas NF, Pfeilschifter J, Devraj K, Pfeilschifter W. Alteration of sphingolipid metabolism as a putative mechanism underlying LPS-induced BBB disruption. J Neurochem. 2018;144(2):172–85.PubMedCrossRef
10.
go back to reference Xi T, Jin F, Zhu Y, Wang J, Tang L, Wang Y, Liebeskind DS, He Z. MicroRNA-126-3p attenuates blood–brain barrier disruption, cerebral edema and neuronal injury following intracerebral hemorrhage by regulating PIK3R2 and Akt. Biochem Biophys Res Commun. 2017;494(1–2):144–51.PubMedCrossRef Xi T, Jin F, Zhu Y, Wang J, Tang L, Wang Y, Liebeskind DS, He Z. MicroRNA-126-3p attenuates blood–brain barrier disruption, cerebral edema and neuronal injury following intracerebral hemorrhage by regulating PIK3R2 and Akt. Biochem Biophys Res Commun. 2017;494(1–2):144–51.PubMedCrossRef
11.
go back to reference Simion V, Nadim WD, Benedetti H, Pichon C, Morisset-Lopez S, Baril P. Pharmacomodulation of microRNA expression in neurocognitive diseases: obstacles and future opportunities. Curr Neuropharmacol. 2017;15(2):276–90.PubMedPubMedCentralCrossRef Simion V, Nadim WD, Benedetti H, Pichon C, Morisset-Lopez S, Baril P. Pharmacomodulation of microRNA expression in neurocognitive diseases: obstacles and future opportunities. Curr Neuropharmacol. 2017;15(2):276–90.PubMedPubMedCentralCrossRef
12.
go back to reference Ochocinska MJ, Zlokovic BV, Searson PC, Crowder AT, Kraig RP, Ljubimova JY, Mainprize TG, Banks WA, Warren RQ, Kindzelski A, et al. NIH workshop report on the trans-agency blood–brain interface workshop 2016: exploring key challenges and opportunities associated with the blood, brain and their interface. Fluids Barriers CNS. 2017;14(1):12.PubMedPubMedCentralCrossRef Ochocinska MJ, Zlokovic BV, Searson PC, Crowder AT, Kraig RP, Ljubimova JY, Mainprize TG, Banks WA, Warren RQ, Kindzelski A, et al. NIH workshop report on the trans-agency blood–brain interface workshop 2016: exploring key challenges and opportunities associated with the blood, brain and their interface. Fluids Barriers CNS. 2017;14(1):12.PubMedPubMedCentralCrossRef
13.
go back to reference Selmaj I, Mycko MP, Raine CS, Selmaj KW. The role of exosomes in CNS inflammation and their involvement in multiple sclerosis. J Neuroimmunol. 2017;306:1–10.PubMedCrossRef Selmaj I, Mycko MP, Raine CS, Selmaj KW. The role of exosomes in CNS inflammation and their involvement in multiple sclerosis. J Neuroimmunol. 2017;306:1–10.PubMedCrossRef
14.
go back to reference Hashimoto Y, Shirakura K, Okada Y, Takeda H, Endo K, Tamura M, Watari A, Sadamura Y, Sawasaki T, Doi T, et al. Claudin-5-binders enhance permeation of solutes across the blood–brain barrier in a mammalian model. J Pharmacol Exp Ther. 2017;363(2):275–83.PubMedCrossRef Hashimoto Y, Shirakura K, Okada Y, Takeda H, Endo K, Tamura M, Watari A, Sadamura Y, Sawasaki T, Doi T, et al. Claudin-5-binders enhance permeation of solutes across the blood–brain barrier in a mammalian model. J Pharmacol Exp Ther. 2017;363(2):275–83.PubMedCrossRef
15.
go back to reference Dithmer S, Staat C, Muller C, Ku MC, Pohlmann A, Niendorf T, Gehne N, Fallier-Becker P, Kittel A, Walter FR, et al. Claudin peptidomimetics modulate tissue barriers for enhanced drug delivery. Ann NY Acad Sci. 2017;1397(1):169–84.PubMedCrossRef Dithmer S, Staat C, Muller C, Ku MC, Pohlmann A, Niendorf T, Gehne N, Fallier-Becker P, Kittel A, Walter FR, et al. Claudin peptidomimetics modulate tissue barriers for enhanced drug delivery. Ann NY Acad Sci. 2017;1397(1):169–84.PubMedCrossRef
16.
go back to reference Menard C, Pfau ML, Hodes GE, Kana V, Wang VX, Bouchard S, Takahashi A, Flanigan ME, Aleyasin H, LeClair KB, et al. Social stress induces neurovascular pathology promoting depression. Nat Neurosci. 2017;20(12):1752–60.PubMedCrossRef Menard C, Pfau ML, Hodes GE, Kana V, Wang VX, Bouchard S, Takahashi A, Flanigan ME, Aleyasin H, LeClair KB, et al. Social stress induces neurovascular pathology promoting depression. Nat Neurosci. 2017;20(12):1752–60.PubMedCrossRef
17.
go back to reference Ambady P, Fu R, Netto JP, Kersch C, Firkins J, Doolittle ND, Neuwelt EA. Patterns of relapse in primary central nervous system lymphoma: inferences regarding the role of the neuro-vascular unit and monoclonal antibodies in treating occult CNS disease. Fluids Barriers CNS. 2017;14(1):16.PubMedPubMedCentralCrossRef Ambady P, Fu R, Netto JP, Kersch C, Firkins J, Doolittle ND, Neuwelt EA. Patterns of relapse in primary central nervous system lymphoma: inferences regarding the role of the neuro-vascular unit and monoclonal antibodies in treating occult CNS disease. Fluids Barriers CNS. 2017;14(1):16.PubMedPubMedCentralCrossRef
18.
go back to reference Dosa E, Heltai K, Radovits T, Molnar G, Kapocsi J, Merkely B, Fu R, Doolittle ND, Toth GB, Urdang Z, et al. Dose escalation study of intravenous and intra-arterial N-acetylcysteine for the prevention of oto- and nephrotoxicity of cisplatin with a contrast-induced nephropathy model in patients with renal insufficiency. Fluids Barriers CNS. 2017;14(1):26.PubMedPubMedCentralCrossRef Dosa E, Heltai K, Radovits T, Molnar G, Kapocsi J, Merkely B, Fu R, Doolittle ND, Toth GB, Urdang Z, et al. Dose escalation study of intravenous and intra-arterial N-acetylcysteine for the prevention of oto- and nephrotoxicity of cisplatin with a contrast-induced nephropathy model in patients with renal insufficiency. Fluids Barriers CNS. 2017;14(1):26.PubMedPubMedCentralCrossRef
19.
go back to reference Haqqani AS, Delaney CE, Brunette E, Baumann E, Farrington GK, Sisk W, Eldredge J, Ding W, Tremblay TL, Stanimirovic DB. Endosomal trafficking regulates receptor-mediated transcytosis of antibodies across the blood brain barrier. J Cereb Blood Flow Metab. 2017. https://doi.org/10.1177/0271678X17740031. Haqqani AS, Delaney CE, Brunette E, Baumann E, Farrington GK, Sisk W, Eldredge J, Ding W, Tremblay TL, Stanimirovic DB. Endosomal trafficking regulates receptor-mediated transcytosis of antibodies across the blood brain barrier. J Cereb Blood Flow Metab. 2017. https://​doi.​org/​10.​1177/​0271678X17740031​.
20.
go back to reference Molino Y, David M, Varini K, Jabes F, Gaudin N, Fortoul A, Bakloul K, Masse M, Bernard A, Drobecq L, et al. Use of LDL receptor-targeting peptide vectors for in vitro and in vivo cargo transport across the blood–brain barrier. FASEB J. 2017;31(5):1807–27.PubMedCrossRef Molino Y, David M, Varini K, Jabes F, Gaudin N, Fortoul A, Bakloul K, Masse M, Bernard A, Drobecq L, et al. Use of LDL receptor-targeting peptide vectors for in vitro and in vivo cargo transport across the blood–brain barrier. FASEB J. 2017;31(5):1807–27.PubMedCrossRef
21.
go back to reference Shimizu F, Schaller KL, Owens GP, Cotleur AC, Kellner D, Takeshita Y, Obermeier B, Kryzer TJ, Sano Y, Kanda T et al. Glucose-regulated protein 78 autoantibody associates with blood–brain barrier disruption in neuromyelitis optica. Sci Transl Med. 2017;9(397):eaai9111.PubMedCrossRef Shimizu F, Schaller KL, Owens GP, Cotleur AC, Kellner D, Takeshita Y, Obermeier B, Kryzer TJ, Sano Y, Kanda T et al. Glucose-regulated protein 78 autoantibody associates with blood–brain barrier disruption in neuromyelitis optica. Sci Transl Med. 2017;9(397):eaai9111.PubMedCrossRef
22.
go back to reference Dogbevia GK, Tollner K, Korbelin J, Broer S, Ridder DA, Grasshoff H, Brandt C, Wenzel J, Straub BK, Trepel M, et al. Gene therapy decreases seizures in a model of Incontinentia pigmenti. Ann Neurol. 2017;82(1):93–104.PubMedCrossRef Dogbevia GK, Tollner K, Korbelin J, Broer S, Ridder DA, Grasshoff H, Brandt C, Wenzel J, Straub BK, Trepel M, et al. Gene therapy decreases seizures in a model of Incontinentia pigmenti. Ann Neurol. 2017;82(1):93–104.PubMedCrossRef
23.
go back to reference Wang S, Karakatsani ME, Fung C, Sun T, Acosta C, Konofagou E. Direct brain infusion can be enhanced with focused ultrasound and microbubbles. J Cereb Blood Flow Metab. 2017;37(2):706–14.PubMedCrossRef Wang S, Karakatsani ME, Fung C, Sun T, Acosta C, Konofagou E. Direct brain infusion can be enhanced with focused ultrasound and microbubbles. J Cereb Blood Flow Metab. 2017;37(2):706–14.PubMedCrossRef
24.
go back to reference Erdo F, Denes L, de Lange E. Age-associated physiological and pathological changes at the blood–brain barrier: a review. J Cereb Blood Flow Metab. 2017;37(1):4–24.PubMedCrossRef Erdo F, Denes L, de Lange E. Age-associated physiological and pathological changes at the blood–brain barrier: a review. J Cereb Blood Flow Metab. 2017;37(1):4–24.PubMedCrossRef
26.
go back to reference Karimy JK, Zhang J, Kurland DB, Theriault BC, Duran D, Stokum JA, Furey CG, Zhou X, Mansuri MS, Montejo J, et al. Inflammation-dependent cerebrospinal fluid hypersecretion by the choroid plexus epithelium in posthemorrhagic hydrocephalus. Nat Med. 2017;23(8):997–1003.PubMed Karimy JK, Zhang J, Kurland DB, Theriault BC, Duran D, Stokum JA, Furey CG, Zhou X, Mansuri MS, Montejo J, et al. Inflammation-dependent cerebrospinal fluid hypersecretion by the choroid plexus epithelium in posthemorrhagic hydrocephalus. Nat Med. 2017;23(8):997–1003.PubMed
27.
go back to reference Xiang J, Routhe LJ, Wilkinson DA, Hua Y, Moos T, Xi G, Keep RF. 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(1):8.PubMedPubMedCentralCrossRef Xiang J, Routhe LJ, Wilkinson DA, Hua Y, Moos T, Xi G, Keep RF. 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(1):8.PubMedPubMedCentralCrossRef
28.
go back to reference Zhang CE, Wong SM, van de Haar HJ, Staals J, Jansen JF, Jeukens CR, Hofman PA, van Oostenbrugge RJ, Backes WH. Blood–brain barrier leakage is more widespread in patients with cerebral small vessel disease. Neurology. 2017;88(5):426–32.PubMedCrossRef Zhang CE, Wong SM, van de Haar HJ, Staals J, Jansen JF, Jeukens CR, Hofman PA, van Oostenbrugge RJ, Backes WH. Blood–brain barrier leakage is more widespread in patients with cerebral small vessel disease. Neurology. 2017;88(5):426–32.PubMedCrossRef
29.
go back to reference Stamatovic SM, Johnson AM, Sladojevic N, Keep RF, Andjelkovic AV. Endocytosis of tight junction proteins and the regulation of degradation and recycling. Ann NY Acad Sci. 2017;1397(1):54–65.PubMedCrossRef Stamatovic SM, Johnson AM, Sladojevic N, Keep RF, Andjelkovic AV. Endocytosis of tight junction proteins and the regulation of degradation and recycling. Ann NY Acad Sci. 2017;1397(1):54–65.PubMedCrossRef
30.
go back to reference Haley MJ, Lawrence CB. The blood–brain barrier after stroke: structural studies and the role of transcytotic vesicles. J Cereb Blood Flow Metab. 2017;37(2):456–70.PubMedCrossRef Haley MJ, Lawrence CB. The blood–brain barrier after stroke: structural studies and the role of transcytotic vesicles. J Cereb Blood Flow Metab. 2017;37(2):456–70.PubMedCrossRef
32.
go back to reference Shi Y, Jiang X, Zhang L, Pu H, Hu X, Zhang W, Cai W, Gao Y, Leak RK, Keep RF, et al. Endothelium-targeted overexpression of heat shock protein 27 ameliorates blood–brain barrier disruption after ischemic brain injury. Proc Natl Acad Sci USA. 2017;114(7):E1243–52.PubMedPubMedCentralCrossRef Shi Y, Jiang X, Zhang L, Pu H, Hu X, Zhang W, Cai W, Gao Y, Leak RK, Keep RF, et al. Endothelium-targeted overexpression of heat shock protein 27 ameliorates blood–brain barrier disruption after ischemic brain injury. Proc Natl Acad Sci USA. 2017;114(7):E1243–52.PubMedPubMedCentralCrossRef
33.
go back to reference Jiang B, Li L, Chen Q, Tao Y, Yang L, Zhang B, Zhang JH, Feng H, Chen Z, Tang J, et al. Role of glibenclamide in brain injury after intracerebral hemorrhage. Transl Stroke Res. 2017;8(2):183–93.PubMedCrossRef Jiang B, Li L, Chen Q, Tao Y, Yang L, Zhang B, Zhang JH, Feng H, Chen Z, Tang J, et al. Role of glibenclamide in brain injury after intracerebral hemorrhage. Transl Stroke Res. 2017;8(2):183–93.PubMedCrossRef
35.
go back to reference Praetorius J, Damkier HH. Transport across the choroid plexus epithelium. Am J Physiol Cell Physiol. 2017;312(6):C673–86.PubMedCrossRef Praetorius J, Damkier HH. Transport across the choroid plexus epithelium. Am J Physiol Cell Physiol. 2017;312(6):C673–86.PubMedCrossRef
36.
go back to reference Llovera G, Benakis C, Enzmann G, Cai R, Arzberger T, Ghasemigharagoz A, Mao X, Malik R, Lazarevic I, Liebscher S, et al. The choroid plexus is a key cerebral invasion route for T cells after stroke. Acta Neuropathol. 2017;134(6):851–68.PubMedCrossRef Llovera G, Benakis C, Enzmann G, Cai R, Arzberger T, Ghasemigharagoz A, Mao X, Malik R, Lazarevic I, Liebscher S, et al. The choroid plexus is a key cerebral invasion route for T cells after stroke. Acta Neuropathol. 2017;134(6):851–68.PubMedCrossRef
37.
go back to reference Koshida R, Oishi H, Hamada M, Takei Y, Takahashi S. MafB is required for development of the hindbrain choroid plexus. Biochem Biophys Res Commun. 2017;483(1):288–93.PubMedCrossRef Koshida R, Oishi H, Hamada M, Takei Y, Takahashi S. MafB is required for development of the hindbrain choroid plexus. Biochem Biophys Res Commun. 2017;483(1):288–93.PubMedCrossRef
38.
go back to reference Engelhardt B, Vajkoczy P, Weller RO. The movers and shapers in immune privilege of the CNS. Nat Immunol. 2017;18(2):123–31.PubMedCrossRef Engelhardt B, Vajkoczy P, Weller RO. The movers and shapers in immune privilege of the CNS. Nat Immunol. 2017;18(2):123–31.PubMedCrossRef
39.
go back to reference Antila S, Karaman S, Nurmi H, Airavaara M, Voutilainen MH, Mathivet T, Chilov D, Li Z, Koppinen T, Park JH, et al. Development and plasticity of meningeal lymphatic vessels. J Exp Med. 2017;214(12):3645–67.PubMedCrossRef Antila S, Karaman S, Nurmi H, Airavaara M, Voutilainen MH, Mathivet T, Chilov D, Li Z, Koppinen T, Park JH, et al. Development and plasticity of meningeal lymphatic vessels. J Exp Med. 2017;214(12):3645–67.PubMedCrossRef
40.
go back to reference Louveau A, Plog BA, Antila S, Alitalo K, Nedergaard M, Kipnis J. Understanding the functions and relationships of the glymphatic system and meningeal lymphatics. J Clin Invest. 2017;127(9):3210–9.PubMedCrossRef Louveau A, Plog BA, Antila S, Alitalo K, Nedergaard M, Kipnis J. Understanding the functions and relationships of the glymphatic system and meningeal lymphatics. J Clin Invest. 2017;127(9):3210–9.PubMedCrossRef
41.
go back to reference Ma Q, Ineichen BV, Detmar M, Proulx ST. Outflow of cerebrospinal fluid is predominantly through lymphatic vessels and is reduced in aged mice. Nat commun. 2017;8(1):1434.PubMedPubMedCentralCrossRef Ma Q, Ineichen BV, Detmar M, Proulx ST. Outflow of cerebrospinal fluid is predominantly through lymphatic vessels and is reduced in aged mice. Nat commun. 2017;8(1):1434.PubMedPubMedCentralCrossRef
43.
go back to reference Burfeind KG, Murchison CF, Westaway SK, Simon MJ, Erten-Lyons D, Kaye JA, Quinn JF, Iliff JJ. The effects of noncoding aquaporin-4 single-nucleotide polymorphisms on cognition and functional progression of Alzheimer’s disease. Alzheimer’s Dement (N Y). 2017;3(3):348–59. Burfeind KG, Murchison CF, Westaway SK, Simon MJ, Erten-Lyons D, Kaye JA, Quinn JF, Iliff JJ. The effects of noncoding aquaporin-4 single-nucleotide polymorphisms on cognition and functional progression of Alzheimer’s disease. Alzheimer’s Dement (N Y). 2017;3(3):348–59.
44.
go back to reference Ringstad G, Vatnehol SAS, Eide PK. Glymphatic MRI in idiopathic normal pressure hydrocephalus. Brain. 2017;140(10):2691–705.PubMedCrossRef Ringstad G, Vatnehol SAS, Eide PK. Glymphatic MRI in idiopathic normal pressure hydrocephalus. Brain. 2017;140(10):2691–705.PubMedCrossRef
45.
go back to reference Schain AJ, Melo-Carrillo A, Strassman AM, Burstein R. Cortical spreading depression closes paravascular space and impairs glymphatic flow: implications for migraine headache. J Neurosci. 2017;37(11):2904–15.PubMedPubMedCentralCrossRef Schain AJ, Melo-Carrillo A, Strassman AM, Burstein R. Cortical spreading depression closes paravascular space and impairs glymphatic flow: implications for migraine headache. J Neurosci. 2017;37(11):2904–15.PubMedPubMedCentralCrossRef
46.
go back to reference Jiang Q, Zhang L, Ding G, Davoodi-Bojd E, Li Q, Li L, Sadry N, Nedergaard M, Chopp M, Zhang Z. Impairment of the glymphatic system after diabetes. J Cereb Blood Flow Metab. 2017;37(4):1326–37.PubMedCrossRef Jiang Q, Zhang L, Ding G, Davoodi-Bojd E, Li Q, Li L, Sadry N, Nedergaard M, Chopp M, Zhang Z. Impairment of the glymphatic system after diabetes. J Cereb Blood Flow Metab. 2017;37(4):1326–37.PubMedCrossRef
47.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
48.
go back to reference Wang M, Ding F, Deng S, Guo X, Wang W, Iliff JJ, Nedergaard M. Focal solute trapping and global glymphatic pathway impairment in a murine model of multiple microinfarcts. J Neurosci. 2017;37(11):2870–7.PubMedPubMedCentralCrossRef Wang M, Ding F, Deng S, Guo X, Wang W, Iliff JJ, Nedergaard M. Focal solute trapping and global glymphatic pathway impairment in a murine model of multiple microinfarcts. J Neurosci. 2017;37(11):2870–7.PubMedPubMedCentralCrossRef
50.
go back to reference Smith AJ, Yao X, Dix JA, Jin BJ, Verkman AS. Test of the ‘glymphatic’ hypothesis demonstrates diffusive and aquaporin-4-independent solute transport in rodent brain parenchyma. eLife. 2017;6:e27679.PubMedPubMedCentral Smith AJ, Yao X, Dix JA, Jin BJ, Verkman AS. Test of the ‘glymphatic’ hypothesis demonstrates diffusive and aquaporin-4-independent solute transport in rodent brain parenchyma. eLife. 2017;6:e27679.PubMedPubMedCentral
52.
go back to reference Pizzo ME, Wolak DJ, Kumar NN, Brunette E, Brunnquell CL, Hannocks MJ, Abbott NJ, Meyerand ME, Sorokin L, Stanimirovic DB, et al. Intrathecal antibody distribution in the rat brain: surface diffusion, perivascular transport, and osmotic enhancement of delivery. J Physiol. 2017. https://doi.org/10.1113/JP275105.PubMed Pizzo ME, Wolak DJ, Kumar NN, Brunette E, Brunnquell CL, Hannocks MJ, Abbott NJ, Meyerand ME, Sorokin L, Stanimirovic DB, et al. Intrathecal antibody distribution in the rat brain: surface diffusion, perivascular transport, and osmotic enhancement of delivery. J Physiol. 2017. https://​doi.​org/​10.​1113/​JP275105.PubMed
53.
go back to reference Chen Z, Liu C, Zhang J, Relkin N, Xing Y, Li Y. Cerebrospinal fluid Abeta42, t-tau, and p-tau levels in the differential diagnosis of idiopathic normal-pressure hydrocephalus: a systematic review and meta-analysis. Fluids Barriers CNS. 2017;14(1):13.PubMedPubMedCentralCrossRef Chen Z, Liu C, Zhang J, Relkin N, Xing Y, Li Y. Cerebrospinal fluid Abeta42, t-tau, and p-tau levels in the differential diagnosis of idiopathic normal-pressure hydrocephalus: a systematic review and meta-analysis. Fluids Barriers CNS. 2017;14(1):13.PubMedPubMedCentralCrossRef
54.
go back to reference Habiyaremye G, Morales DM, Morgan CD, McAllister JP, CreveCoeur TS, Han RH, Gabir M, Baksh B, Mercer D, Limbrick DD Jr. Chemokine and cytokine levels in the lumbar cerebrospinal fluid of preterm infants with post-hemorrhagic hydrocephalus. Fluids Barriers CNS. 2017;14(1):35.PubMedPubMedCentralCrossRef Habiyaremye G, Morales DM, Morgan CD, McAllister JP, CreveCoeur TS, Han RH, Gabir M, Baksh B, Mercer D, Limbrick DD Jr. Chemokine and cytokine levels in the lumbar cerebrospinal fluid of preterm infants with post-hemorrhagic hydrocephalus. Fluids Barriers CNS. 2017;14(1):35.PubMedPubMedCentralCrossRef
55.
go back to reference Handels RLH, Vos SJB, Kramberger MG, Jelic V, Blennow K, van Buchem M, van der Flier W, Freund-Levi Y, Hampel H, Olde Rikkert M, et al. Predicting progression to dementia in persons with mild cognitive impairment using cerebrospinal fluid markers. Alzheimer’s Dement. 2017;13(8):903–12.CrossRef Handels RLH, Vos SJB, Kramberger MG, Jelic V, Blennow K, van Buchem M, van der Flier W, Freund-Levi Y, Hampel H, Olde Rikkert M, et al. Predicting progression to dementia in persons with mild cognitive impairment using cerebrospinal fluid markers. Alzheimer’s Dement. 2017;13(8):903–12.CrossRef
56.
go back to reference Hansson KT, Skillback T, Pernevik E, Kern S, Portelius E, Hoglund K, Brinkmalm G, Holmen-Larsson J, Blennow K, Zetterberg H et al. Expanding the cerebrospinal fluid endopeptidome. Proteomics. 2017;17(5):1600384.CrossRef Hansson KT, Skillback T, Pernevik E, Kern S, Portelius E, Hoglund K, Brinkmalm G, Holmen-Larsson J, Blennow K, Zetterberg H et al. Expanding the cerebrospinal fluid endopeptidome. Proteomics. 2017;17(5):1600384.CrossRef
57.
go back to reference Limbrick DD Jr, Baksh B, Morgan CD, Habiyaremye G, McAllister JP 2nd, Inder TE, Mercer D, Holtzman DM, Strahle J, Wallendorf MJ, et al. Cerebrospinal fluid biomarkers of infantile congenital hydrocephalus. PLoS ONE. 2017;12(2):e0172353.PubMedPubMedCentralCrossRef Limbrick DD Jr, Baksh B, Morgan CD, Habiyaremye G, McAllister JP 2nd, Inder TE, Mercer D, Holtzman DM, Strahle J, Wallendorf MJ, et al. Cerebrospinal fluid biomarkers of infantile congenital hydrocephalus. PLoS ONE. 2017;12(2):e0172353.PubMedPubMedCentralCrossRef
58.
go back to reference Mattsson N, Lonneborg A, Boccardi M, Blennow K, Hansson O. Geneva Task Force for the Roadmap of Alzheimer’s B: clinical validity of cerebrospinal fluid Abeta42, tau, and phospho-tau as biomarkers for Alzheimer’s disease in the context of a structured 5-phase development framework. Neurobiol Aging. 2017;52:196–213.PubMedCrossRef Mattsson N, Lonneborg A, Boccardi M, Blennow K, Hansson O. Geneva Task Force for the Roadmap of Alzheimer’s B: clinical validity of cerebrospinal fluid Abeta42, tau, and phospho-tau as biomarkers for Alzheimer’s disease in the context of a structured 5-phase development framework. Neurobiol Aging. 2017;52:196–213.PubMedCrossRef
59.
go back to reference Patton SM, Wang Q, Hulgan T, Connor JR, Jia P, Zhao Z, Letendre SL, Ellis RJ, Bush WS, Samuels DC, et al. Cerebrospinal fluid (CSF) biomarkers of iron status are associated with CSF viral load, antiretroviral therapy, and demographic factors in HIV-infected adults. Fluids Barriers CNS. 2017;14(1):11.PubMedPubMedCentralCrossRef Patton SM, Wang Q, Hulgan T, Connor JR, Jia P, Zhao Z, Letendre SL, Ellis RJ, Bush WS, Samuels DC, et al. Cerebrospinal fluid (CSF) biomarkers of iron status are associated with CSF viral load, antiretroviral therapy, and demographic factors in HIV-infected adults. Fluids Barriers CNS. 2017;14(1):11.PubMedPubMedCentralCrossRef
60.
go back to reference Nathan PJ, Lim YY, Abbott R, Galluzzi S, Marizzoni M, Babiloni C, Albani D, Bartres-Faz D, Didic M, Farotti L, et al. Association between CSF biomarkers, hippocampal volume and cognitive function in patients with amnestic mild cognitive impairment (MCI). Neurobiol Aging. 2017;53:1–10.PubMedCrossRef Nathan PJ, Lim YY, Abbott R, Galluzzi S, Marizzoni M, Babiloni C, Albani D, Bartres-Faz D, Didic M, Farotti L, et al. Association between CSF biomarkers, hippocampal volume and cognitive function in patients with amnestic mild cognitive impairment (MCI). Neurobiol Aging. 2017;53:1–10.PubMedCrossRef
61.
go back to reference Asgari M, de Zelicourt DA, Kurtcuoglu V. Barrier dysfunction or drainage reduction: differentiating causes of CSF protein increase. Fluids Barriers CNS. 2017;14(1):14.PubMedPubMedCentralCrossRef Asgari M, de Zelicourt DA, Kurtcuoglu V. Barrier dysfunction or drainage reduction: differentiating causes of CSF protein increase. Fluids Barriers CNS. 2017;14(1):14.PubMedPubMedCentralCrossRef
62.
go back to reference Hoshi K, Matsumoto Y, Ito H, Saito K, Honda T, Yamaguchi Y, Hashimoto Y. A unique glycan-isoform of transferrin in cerebrospinal fluid: a potential diagnostic marker for neurological diseases. Biochem Biophys Acta. 2017;1861(10):2473–8.PubMedCrossRef Hoshi K, Matsumoto Y, Ito H, Saito K, Honda T, Yamaguchi Y, Hashimoto Y. A unique glycan-isoform of transferrin in cerebrospinal fluid: a potential diagnostic marker for neurological diseases. Biochem Biophys Acta. 2017;1861(10):2473–8.PubMedCrossRef
63.
go back to reference Bonin S, Zanotta N, Sartori A, Bratina A, Manganotti P, Trevisan G, Comar M. Cerebrospinal fluid cytokine expression profile in multiple sclerosis and chronic inflammatory demyelinating polyneuropathy. Immunol Invest. 2018;47(2):135–45.PubMedCrossRef Bonin S, Zanotta N, Sartori A, Bratina A, Manganotti P, Trevisan G, Comar M. Cerebrospinal fluid cytokine expression profile in multiple sclerosis and chronic inflammatory demyelinating polyneuropathy. Immunol Invest. 2018;47(2):135–45.PubMedCrossRef
64.
go back to reference Hottenrott T, Dersch R, Berger B, Rauer S, Huzly D, Stich O. The MRZ reaction in primary progressive multiple sclerosis. Fluids Barriers CNS. 2017;14(1):2.PubMedPubMedCentralCrossRef Hottenrott T, Dersch R, Berger B, Rauer S, Huzly D, Stich O. The MRZ reaction in primary progressive multiple sclerosis. Fluids Barriers CNS. 2017;14(1):2.PubMedPubMedCentralCrossRef
65.
go back to reference Bache S, Rasmussen R, Rossing M, Laigaard FP, Nielsen FC, Moller K. MicroRNA changes in cerebrospinal fluid after subarachnoid hemorrhage. Stroke. 2017;48(9):2391–8.PubMedPubMedCentralCrossRef Bache S, Rasmussen R, Rossing M, Laigaard FP, Nielsen FC, Moller K. MicroRNA changes in cerebrospinal fluid after subarachnoid hemorrhage. Stroke. 2017;48(9):2391–8.PubMedPubMedCentralCrossRef
66.
go back to reference Marques TM, Kuiperij HB, Bruinsma IB, van Rumund A, Aerts MB, Esselink RAJ, Bloem BR, Verbeek MM. MicroRNAs in cerebrospinal fluid as potential biomarkers for Parkinson’s disease and multiple system atrophy. Mol Neurobiol. 2017;54(10):7736–45.PubMedCrossRef Marques TM, Kuiperij HB, Bruinsma IB, van Rumund A, Aerts MB, Esselink RAJ, Bloem BR, Verbeek MM. MicroRNAs in cerebrospinal fluid as potential biomarkers for Parkinson’s disease and multiple system atrophy. Mol Neurobiol. 2017;54(10):7736–45.PubMedCrossRef
67.
go back to reference Yagi Y, Ohkubo T, Kawaji H, Machida A, Miyata H, Goda S, Roy S, Hayashizaki Y, Suzuki H, Yokota T. Next-generation sequencing-based small RNA profiling of cerebrospinal fluid exosomes. Neurosci Lett. 2017;636:48–57.PubMedCrossRef Yagi Y, Ohkubo T, Kawaji H, Machida A, Miyata H, Goda S, Roy S, Hayashizaki Y, Suzuki H, Yokota T. Next-generation sequencing-based small RNA profiling of cerebrospinal fluid exosomes. Neurosci Lett. 2017;636:48–57.PubMedCrossRef
68.
go back to reference Feldner A, Adam MG, Tetzlaff F, Moll I, Komljenovic D, Sahm F, Bauerle T, Ishikawa H, Schroten H, Korff T, et al. Loss of Mpdz impairs ependymal cell integrity leading to perinatal-onset hydrocephalus in mice. EMBO Mol Med. 2017;9(7):890–905.PubMedPubMedCentralCrossRef Feldner A, Adam MG, Tetzlaff F, Moll I, Komljenovic D, Sahm F, Bauerle T, Ishikawa H, Schroten H, Korff T, et al. Loss of Mpdz impairs ependymal cell integrity leading to perinatal-onset hydrocephalus in mice. EMBO Mol Med. 2017;9(7):890–905.PubMedPubMedCentralCrossRef
69.
go back to reference Saugier-Veber P, Marguet F, Lecoquierre F, Adle-Biassette H, Guimiot F, Cipriani S, Patrier S, Brasseur-Daudruy M, Goldenberg A, Layet V, et al. Hydrocephalus due to multiple ependymal malformations is caused by mutations in the MPDZ gene. Acta Neuropathol Commun. 2017;5(1):36.PubMedPubMedCentralCrossRef Saugier-Veber P, Marguet F, Lecoquierre F, Adle-Biassette H, Guimiot F, Cipriani S, Patrier S, Brasseur-Daudruy M, Goldenberg A, Layet V, et al. Hydrocephalus due to multiple ependymal malformations is caused by mutations in the MPDZ gene. Acta Neuropathol Commun. 2017;5(1):36.PubMedPubMedCentralCrossRef
70.
go back to reference Zega K, Jovanovic VM, Vitic Z, Niedzielska M, Knaapi L, Jukic MM, Partanen J, Friedel RF, Lang R, Brodski C. Dusp16 deficiency causes congenital obstructive hydrocephalus and brain overgrowth by expansion of the neural progenitor pool. Front Mol Neurosci. 2017;10:372.PubMedPubMedCentralCrossRef Zega K, Jovanovic VM, Vitic Z, Niedzielska M, Knaapi L, Jukic MM, Partanen J, Friedel RF, Lang R, Brodski C. Dusp16 deficiency causes congenital obstructive hydrocephalus and brain overgrowth by expansion of the neural progenitor pool. Front Mol Neurosci. 2017;10:372.PubMedPubMedCentralCrossRef
71.
go back to reference Ayala-Valdovinos MA, Galindo-Garcia J, Sanchez-Chipres D, Duifhuis-Rivera T. Genotyping of friesian horses to detect a hydrocephalus-associated c.1423C>T mutation in B3GALNT2 using PCR-RFLP and PCR-PIRA methods: frequency in stallion horses in Mexico. Mol Cell Probes. 2017;32:69–71.PubMedCrossRef Ayala-Valdovinos MA, Galindo-Garcia J, Sanchez-Chipres D, Duifhuis-Rivera T. Genotyping of friesian horses to detect a hydrocephalus-associated c.1423C>T mutation in B3GALNT2 using PCR-RFLP and PCR-PIRA methods: frequency in stallion horses in Mexico. Mol Cell Probes. 2017;32:69–71.PubMedCrossRef
72.
go back to reference McAllister JP, Guerra MM, Ruiz LC, Jimenez AJ, Dominguez-Pinos D, Sival D, den Dunnen W, Morales DM, Schmidt RE, Rodriguez EM, et al. ventricular zone disruption in human neonates with intraventricular hemorrhage. J Neuropathol Exp Neurol. 2017;76(5):358–75.PubMedCrossRef McAllister JP, Guerra MM, Ruiz LC, Jimenez AJ, Dominguez-Pinos D, Sival D, den Dunnen W, Morales DM, Schmidt RE, Rodriguez EM, et al. ventricular zone disruption in human neonates with intraventricular hemorrhage. J Neuropathol Exp Neurol. 2017;76(5):358–75.PubMedCrossRef
73.
go back to reference Rodriguez EM, Guerra MM. Neural stem cells and fetal-onset hydrocephalus. Pediatr Neurosurg. 2017;52(6):446–61.PubMedCrossRef Rodriguez EM, Guerra MM. Neural stem cells and fetal-onset hydrocephalus. Pediatr Neurosurg. 2017;52(6):446–61.PubMedCrossRef
74.
go back to reference Pfanner T, Henri-Bhargava A, Borchert S. Cerebrospinal fluid biomarkers as predictors of shunt response in idiopathic normal pressure hydrocephalus: a systematic review. Can J Neurol Sci. 2018;45(1):3–10.PubMedCrossRef Pfanner T, Henri-Bhargava A, Borchert S. Cerebrospinal fluid biomarkers as predictors of shunt response in idiopathic normal pressure hydrocephalus: a systematic review. Can J Neurol Sci. 2018;45(1):3–10.PubMedCrossRef
75.
go back to reference Qvarlander S, Ambarki K, Wahlin A, Jacobsson J, Birgander R, Malm J, Eklund A. Cerebrospinal fluid and blood flow patterns in idiopathic normal pressure hydrocephalus. Acta Neurol Scand. 2017;135(5):576–84.PubMedCrossRef Qvarlander S, Ambarki K, Wahlin A, Jacobsson J, Birgander R, Malm J, Eklund A. Cerebrospinal fluid and blood flow patterns in idiopathic normal pressure hydrocephalus. Acta Neurol Scand. 2017;135(5):576–84.PubMedCrossRef
76.
go back to reference Perry A, Graffeo CS, Fattahi N, ElSheikh MM, Cray N, Arani A, Ehman RL, Glaser KJ, Manduca A, Meyer FB, et al. Clinical correlation of abnormal findings on magnetic resonance elastography in idiopathic normal pressure hydrocephalus. World Neurosurg. 2017;99(695–700):e691. Perry A, Graffeo CS, Fattahi N, ElSheikh MM, Cray N, Arani A, Ehman RL, Glaser KJ, Manduca A, Meyer FB, et al. Clinical correlation of abnormal findings on magnetic resonance elastography in idiopathic normal pressure hydrocephalus. World Neurosurg. 2017;99(695–700):e691.
77.
go back to reference Virhammar J, Laurell K, Ahlgren A, Larsson EM. Arterial spin-labeling perfusion MR imaging demonstrates regional CBF decrease in idiopathic normal pressure hydrocephalus. AJNR Am J Neuroradiol. 2017;38(11):2081–8.PubMedCrossRef Virhammar J, Laurell K, Ahlgren A, Larsson EM. Arterial spin-labeling perfusion MR imaging demonstrates regional CBF decrease in idiopathic normal pressure hydrocephalus. AJNR Am J Neuroradiol. 2017;38(11):2081–8.PubMedCrossRef
78.
go back to reference Tuniz F, Vescovi MC, Bagatto D, Drigo D, De Colle MC, Maieron M, Skrap M. The role of perfusion and diffusion MRI in the assessment of patients affected by probable idiopathic normal pressure hydrocephalus. A cohort-prospective preliminary study. Fluids Barriers CNS. 2017;14(1):24.PubMedPubMedCentralCrossRef Tuniz F, Vescovi MC, Bagatto D, Drigo D, De Colle MC, Maieron M, Skrap M. The role of perfusion and diffusion MRI in the assessment of patients affected by probable idiopathic normal pressure hydrocephalus. A cohort-prospective preliminary study. Fluids Barriers CNS. 2017;14(1):24.PubMedPubMedCentralCrossRef
79.
go back to reference Kanno S, Saito M, Kashinoura T, Nishio Y, Iizuka O, Kikuchi H, Takagi M, Iwasaki M, Takahashi S, Mori E. A change in brain white matter after shunt surgery in idiopathic normal pressure hydrocephalus: a tract-based spatial statistics study. Fluids Barriers CNS. 2017;14(1):1.PubMedPubMedCentralCrossRef Kanno S, Saito M, Kashinoura T, Nishio Y, Iizuka O, Kikuchi H, Takagi M, Iwasaki M, Takahashi S, Mori E. A change in brain white matter after shunt surgery in idiopathic normal pressure hydrocephalus: a tract-based spatial statistics study. Fluids Barriers CNS. 2017;14(1):1.PubMedPubMedCentralCrossRef
80.
go back to reference Bifari F, Decimo I, Pino A, Llorens-Bobadilla E, Zhao S, Lange C, Panuccio G, Boeckx B, Thienpont B, Vinckier S, et al. Neurogenic radial glia-like cells in meninges migrate and differentiate into functionally integrated neurons in the neonatal cortex. Cell Stem Cell. 2017;20(3):360.e367–373.e367.CrossRef Bifari F, Decimo I, Pino A, Llorens-Bobadilla E, Zhao S, Lange C, Panuccio G, Boeckx B, Thienpont B, Vinckier S, et al. Neurogenic radial glia-like cells in meninges migrate and differentiate into functionally integrated neurons in the neonatal cortex. Cell Stem Cell. 2017;20(3):360.e367–373.e367.CrossRef
81.
go back to reference Suter T, DeLoughery ZJ, Jaworski A. Meninges-derived cues control axon guidance. Dev Biol. 2017;430(1):1–10.PubMedCrossRef Suter T, DeLoughery ZJ, Jaworski A. Meninges-derived cues control axon guidance. Dev Biol. 2017;430(1):1–10.PubMedCrossRef
82.
go back to reference Horng S, Therattil A, Moyon S, Gordon A, Kim K, Argaw AT, Hara Y, Mariani JN, Sawai S, Flodby P, et al. Astrocytic tight junctions control inflammatory CNS lesion pathogenesis. J Clin Invest. 2017;127(8):3136–51.PubMedPubMedCentralCrossRef Horng S, Therattil A, Moyon S, Gordon A, Kim K, Argaw AT, Hara Y, Mariani JN, Sawai S, Flodby P, et al. Astrocytic tight junctions control inflammatory CNS lesion pathogenesis. J Clin Invest. 2017;127(8):3136–51.PubMedPubMedCentralCrossRef
83.
go back to reference Canfield SG, Stebbins MJ, Morales BS, Asai SW, Vatine GD, Svendsen CN, Palecek SP, Shusta EV. An isogenic blood–brain barrier model comprising brain endothelial cells, astrocytes, and neurons derived from human induced pluripotent stem cells. J Neurochem. 2017;140(6):874–88.PubMedCrossRef Canfield SG, Stebbins MJ, Morales BS, Asai SW, Vatine GD, Svendsen CN, Palecek SP, Shusta EV. An isogenic blood–brain barrier model comprising brain endothelial cells, astrocytes, and neurons derived from human induced pluripotent stem cells. J Neurochem. 2017;140(6):874–88.PubMedCrossRef
84.
go back to reference DeStefano JG, Xu ZS, Williams AJ, Yimam N, Searson PC. Effect of shear stress on iPSC-derived human brain microvascular endothelial cells (dhBMECs). Fluids Barriers CNS. 2017;14(1):20.PubMedPubMedCentralCrossRef DeStefano JG, Xu ZS, Williams AJ, Yimam N, Searson PC. Effect of shear stress on iPSC-derived human brain microvascular endothelial cells (dhBMECs). Fluids Barriers CNS. 2017;14(1):20.PubMedPubMedCentralCrossRef
85.
go back to reference Yamamizu K, Iwasaki M, Takakubo H, Sakamoto T, Ikuno T, Miyoshi M, Kondo T, Nakao Y, Nakagawa M, Inoue H, et al. In vitro modeling of blood–brain barrier with human iPSC-derived endothelial cells, pericytes, neurons, and astrocytes via notch signaling. Stem Cell Rep. 2017;8(3):634–47.CrossRef Yamamizu K, Iwasaki M, Takakubo H, Sakamoto T, Ikuno T, Miyoshi M, Kondo T, Nakao Y, Nakagawa M, Inoue H, et al. In vitro modeling of blood–brain barrier with human iPSC-derived endothelial cells, pericytes, neurons, and astrocytes via notch signaling. Stem Cell Rep. 2017;8(3):634–47.CrossRef
86.
go back to reference Patel R, Page S, Al-Ahmad AJ. Isogenic blood–brain barrier models based on patient-derived stem cells display inter-individual differences in cell maturation and functionality. J Neurochem. 2017;142(1):74–88.PubMedCrossRef Patel R, Page S, Al-Ahmad AJ. Isogenic blood–brain barrier models based on patient-derived stem cells display inter-individual differences in cell maturation and functionality. J Neurochem. 2017;142(1):74–88.PubMedCrossRef
87.
go back to reference Hollmann EK, Bailey AK, Potharazu AV, Neely MD, Bowman AB, Lippmann ES. Accelerated differentiation of human induced pluripotent stem cells to blood–brain barrier endothelial cells. Fluids Barriers CNS. 2017;14(1):9.PubMedPubMedCentralCrossRef Hollmann EK, Bailey AK, Potharazu AV, Neely MD, Bowman AB, Lippmann ES. Accelerated differentiation of human induced pluripotent stem cells to blood–brain barrier endothelial cells. Fluids Barriers CNS. 2017;14(1):9.PubMedPubMedCentralCrossRef
88.
go back to reference Phan DT, Bender RHF, Andrejecsk JW, Sobrino A, Hachey SJ, George SC, Hughes CC. Blood–brain barrier-on-a-chip: microphysiological systems that capture the complexity of the blood-central nervous system interface. Exp Biol Med. 2017;242(17):1669–78.CrossRef Phan DT, Bender RHF, Andrejecsk JW, Sobrino A, Hachey SJ, George SC, Hughes CC. Blood–brain barrier-on-a-chip: microphysiological systems that capture the complexity of the blood-central nervous system interface. Exp Biol Med. 2017;242(17):1669–78.CrossRef
89.
go back to reference Terrell-Hall TB, Ammer AG, Griffith JI, Lockman PR. Permeability across a novel microfluidic blood-tumor barrier model. Fluids Barriers CNS. 2017;14(1):3.PubMedPubMedCentralCrossRef Terrell-Hall TB, Ammer AG, Griffith JI, Lockman PR. Permeability across a novel microfluidic blood-tumor barrier model. Fluids Barriers CNS. 2017;14(1):3.PubMedPubMedCentralCrossRef
90.
go back to reference Akassoglou K, Merlini M, Rafalski VA, Real R, Liang L, Jin Y, Dougherty SE, De Paola V, Linden DJ, Misgeld T, et al. In vivo imaging of CNS injury and disease. J Neurosci. 2017;37(45):10808–16.PubMed Akassoglou K, Merlini M, Rafalski VA, Real R, Liang L, Jin Y, Dougherty SE, De Paola V, Linden DJ, Misgeld T, et al. In vivo imaging of CNS injury and disease. J Neurosci. 2017;37(45):10808–16.PubMed
91.
go back to reference Umans RA, Henson HE, Mu F, Parupalli C, Ju B, Peters JL, Lanham KA, Plavicki JS, Taylor MR. CNS angiogenesis and barriergenesis occur simultaneously. Dev Biol. 2017;425(2):101–8.PubMedCrossRef Umans RA, Henson HE, Mu F, Parupalli C, Ju B, Peters JL, Lanham KA, Plavicki JS, Taylor MR. CNS angiogenesis and barriergenesis occur simultaneously. Dev Biol. 2017;425(2):101–8.PubMedCrossRef
92.
93.
go back to reference Braun C, Sakamoto A, Fuchs H, Ishiguro N, Suzuki S, Cui Y, Klinder K, Watanabe M, Terasaki T, Sauer A. Quantification of transporter and receptor proteins in dog brain capillaries and choroid plexus: relevance for the distribution in brain and CSF of selected BCRP and P-gp substrates. Mol Pharm. 2017;14(10):3436–47.PubMedCrossRef Braun C, Sakamoto A, Fuchs H, Ishiguro N, Suzuki S, Cui Y, Klinder K, Watanabe M, Terasaki T, Sauer A. Quantification of transporter and receptor proteins in dog brain capillaries and choroid plexus: relevance for the distribution in brain and CSF of selected BCRP and P-gp substrates. Mol Pharm. 2017;14(10):3436–47.PubMedCrossRef
94.
go back to reference Garcia-Berrocoso T, Llombart V, Colas-Campas L, Hainard A, Licker V, Penalba A, Ramiro L, Simats A, Bustamante A, Martinez-Saez E et al. Single cell immuno-laser microdissection coupled to label-free proteomics to reveal the proteotypes of human brain cells after ischemia. Mol Cell Proteomics. 2018;17(1):175–89.PubMedCrossRef Garcia-Berrocoso T, Llombart V, Colas-Campas L, Hainard A, Licker V, Penalba A, Ramiro L, Simats A, Bustamante A, Martinez-Saez E et al. Single cell immuno-laser microdissection coupled to label-free proteomics to reveal the proteotypes of human brain cells after ischemia. Mol Cell Proteomics. 2018;17(1):175–89.PubMedCrossRef
95.
go back to reference Gomez-Zepeda D, Chaves C, Taghi M, Sergent P, Liu WQ, Chhuon C, Vidal M, Picard M, Thioulouse E, Broutin I, et al. Targeted unlabeled multiple reaction monitoring analysis of cell markers for the study of sample heterogeneity in isolated rat brain cortical microvessels. J Neurochem. 2017;142(4):597–609.PubMedCrossRef Gomez-Zepeda D, Chaves C, Taghi M, Sergent P, Liu WQ, Chhuon C, Vidal M, Picard M, Thioulouse E, Broutin I, et al. Targeted unlabeled multiple reaction monitoring analysis of cell markers for the study of sample heterogeneity in isolated rat brain cortical microvessels. J Neurochem. 2017;142(4):597–609.PubMedCrossRef
97.
go back to reference Zhang Z, Uchida Y, Hirano S, Ando D, Kubo Y, Auriola S, Akanuma SI, Hosoya KI, Urtti A, Terasaki T, et al. Inner blood-retinal barrier dominantly expresses breast cancer resistance protein: comparative quantitative targeted absolute proteomics study of CNS barriers in pig. Mol Pharm. 2017;14(11):3729–38.PubMedCrossRef Zhang Z, Uchida Y, Hirano S, Ando D, Kubo Y, Auriola S, Akanuma SI, Hosoya KI, Urtti A, Terasaki T, et al. Inner blood-retinal barrier dominantly expresses breast cancer resistance protein: comparative quantitative targeted absolute proteomics study of CNS barriers in pig. Mol Pharm. 2017;14(11):3729–38.PubMedCrossRef
98.
go back to reference Lee SJ, Kwon S, Gatti JR, Korcari E, Gresser TE, Felix PC, Keep SG, Pasquale KC, Bai T, Blanchett-Anderson SA, et al. Large-scale identification of human cerebrovascular proteins: inter-tissue and intracerebral vascular protein diversity. PLoS ONE. 2017;12(11):e0188540.PubMedPubMedCentralCrossRef Lee SJ, Kwon S, Gatti JR, Korcari E, Gresser TE, Felix PC, Keep SG, Pasquale KC, Bai T, Blanchett-Anderson SA, et al. Large-scale identification of human cerebrovascular proteins: inter-tissue and intracerebral vascular protein diversity. PLoS ONE. 2017;12(11):e0188540.PubMedPubMedCentralCrossRef
Metadata
Title
Progress in brain barriers and brain fluid research in 2017
Authors
Richard F. Keep
Hazel C. Jones
Lester R. Drewes
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Fluids and Barriers of the CNS / Issue 1/2018
Electronic ISSN: 2045-8118
DOI
https://doi.org/10.1186/s12987-018-0091-8

Other articles of this Issue 1/2018

Fluids and Barriers of the CNS 1/2018 Go to the issue