Skip to main content

Progress in brain barriers and brain fluid research in 2017

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.

Editorial

The purpose of this editorial is to highlight advances in brain barriers and brain fluids research in 2017 as well as areas of debate. As always, it is not possible to cover all important progress and, as we have mentioned before [1], such choices are idiosyncratic. However, we hope this editorial is useful for informing our readership and identifying promising areas for study as well as areas where technological advances are needed.

Blood–brain barrier (BBB)/neurovascular unit (NVU)

There continues to be progress in identifying pathways important for the development of NVU/BBB properties. Thus, Cho et al. found [2] that the endothelial G-protein-coupled receptor (GPCR) Gpr124 and a glycosylphosphatidylinositol-anchored membrane protein, Reck, are required for forebrain angiogenesis and acquisition of brain barrier properties in mouse development. Both molecules appear to function by regulating Wnt signaling. Chang et al. [3] further examined the effect of a conditional knockout of Gpr124 in adult mice. No BBB effect was observed under normal conditions, but increased barrier disruption occurred in ischemic stroke and glioblastoma models. The effects of Gpr124 were again via the Wnt-βcatenin pathway, with activation of Wnt-βcatenin signaling reversing the effect of the Gpr124 conditional knockout.

Pericytes continued to be a major focus of current NVU/BBB research. Nakazato et al. [4] reported that a circadian clock transcriptional activator, brain and muscle aryl hydrocarbon receptor nuclear translocator-like protein 1 (Bmal1), is an important regulator of pericyte function. Bmal1 deletion caused pericyte dysfunction, age-dependent loss of pericytes, and endothelium hyperpermeability. The possible role of pericytes in circadian changes in barrier function merits further investigation. Some of the evidence for the importance of pericytes in NVU/BBB regulation has come from mice with a mutation in the retention motif for platelet derived growth factor (pdgf)-β (pdgf-bret/ret) which have pericyte depletion. A recent study found that the effects of such depletion on NVU/BBB permeability were brain region-dependent (e.g. greater in cortex, striatum and hippocampus) [5]. There has been interest in whether NVU/BBB function differs between brain regions and these results suggest that there are differences in regulation. Although most studies on pericytes described beneficial effects of pericytes on BBB function, they may have potentially detrimental effects. Underly et al. [6] found that pericytes caused early BBB damage after cerebral ischemia via a matrix metalloproteinase9-dependent mechanism.

A potential theme is developing on the importance of lipid regulation in NVU/BBB function. More et al. found that peroxisome proliferator-activated receptor (PPAR) α is not only a lipid sensor, but it also regulates the expression and activity of brain endothelial efflux transporters [7]. In addition, Andreone et al. found that lipid transport by Mfsd2a inhibits caveolae vesicle formation in brain endothelial cells suppressing transcytosis [8]. These results suggested that the Mfsd2a gene product may have a dual role in lipid metabolism/transport and transcytosis. Similarly, recent evidence suggested the importance of alterations in sphingosine-1-phosphate signaling in BBB dysfunction after endotoxemia [9].

As with other tissues, there was great interest in the role of microRNAs and exosomes in brain and cells of the NVU. Thus, for example, Xi et al. [10] found that microRNA-126-3p promotes barrier integrity in the setting of intracerebral hemorrhage. The therapeutic use of using microRNAs to treat neurological conditions is potentially a beneficial area of research, but delivery into the endothelial cells and brain parenchyma remains a major issue [11]. The use of exosomes as a delivery system is one approach being vigorously pursued [12, 13]. For both exosomes and/or microRNA, the cerebral endothelium may be an easier therapeutic target than the brain parenchyma.

Modifying the BBB for drug delivery

Currently, different approaches are being tested to modify brain endothelial tight junctions and thereby enhance drug delivery to the brain. Hashimoto et al. have shown that a monoclonal antibody targeting claudin-5 can increase barrier permeability in an in vitro endothelial barrier model [14]. Similarly, Dithmer et al. showed that peptidomimetics that have nanomolar affinity for claudin-5 increase barrier permeability in vitro and in vivo [15], a feature that was reversible in 12–48 h. This is important considering the potential side-effects of modifying claudin-5 [16] (see below).

Clinically, osmotic-induced blood–brain disruption is currently used to enhance delivery of anti-neoplastic agents to brain tumors [17]. An important consideration is the potential effect of the anti-neoplastic agent on normal tissue. Dosa et al. [18] reported the results of an early stage clinical trial of N-acetyl cysteine to reduce the ototoxic side-effects induced by cisplatin.

A number of different approaches were investigated to improve drug transfer to the brain: peptide vectors including antibodies may be used to target the LDL receptor and transfer ligands by receptor-mediated transcytosis [19, 20]. Shimizu et al. [21] observed that an antibody against an endothelial membrane protein (glucose-regulated protein 78) led to tight junction disruption and enhanced permeability to high molecular weight proteins. Thus, development of a strategy for controlled delivery of biologics, such as proteins or genes, to the brain may be possible. Also, specific gene therapy with adeno-associated viral vectors was tested in mice to control seizures [22]. Another approach to focally enhance brain vascular permeability was the use of ultrasound and microbubbles [23], currently in clinical investigation for Alzheimer’s disease therapy: Blood Brain Barrier Opening in Alzheimer’ Disease trial (BOREAL1; NCT03119961).

Barriers in disease

Many neurological conditions (e.g. ischemic and hemorrhagic stroke, multiple sclerosis and neurodegenerative diseases) impact the NVU/BBB [24, 25] and blood-CSF barriers [26, 27]. Recently, Menard et al. [16] extended such findings by examining the effects of social stress in mice (a model of depression). They found that stress induced by chronic social defeat reduced brain microvessel claudin-5 expression in the nucleus accumbens, and that reducing claudin-5 with a short hairpin RNA caused depression-like symptoms and increased entry of interleukin-6 into brain. Interestingly, the effects on claudin-5 were reversed by antidepressant treatment. Other studies have indicated that there are subtle NVU/BBB changes in a variety of conditions including aging [24] and cerebral small vessel disease [28]. The impact of these changes (e.g. low level neuroinflammation) is an important area for investigation.

Over the decades there has been a longstanding debate over the relative importance of alterations in the paracellular and transcellular pathways in disease-induced modification of blood–brain transport. Currently, there is a debate about the relative importance of tight junction modification vs. transcytosis. It should be noted that there may be important interactions between tight junctions and the vesicular system (e.g. in internalization of tight junction proteins from the plasma membrane [29]) complicating data interpretation and that there may be differing results dependent upon which markers are being used to assess barrier function. In ischemic stroke, the importance of changes in transcytosis [30] and tight junctions [31] was recently highlighted. An issue with regards to changes in NVU/BBB function in neurological conditions is whether it is a consequence of the condition or whether it contributes to the injury. It is important, therefore, that Shi et al. [32] found that ameliorating BBB disruption in ischemia/reperfusion injury in mice by overexpressing heat shock protein-27 specifically in the endothelium, reduced overall stroke-induced brain injury (infarct size and neurological deficits). Such results indicate that the BBB is a therapeutic target for stroke.

The ultimate goal of brain barrier and brain fluid research is to improve patient outcome. In this regard, the potential use of glibenclamide (glyburide) to reduce brain edema for a variety of neurological conditions is noteworthy. Glibenclamide is a Sur1-TRPM4 channel inhibitor that has been shown to reduce brain edema in a variety of preclinical models (e.g. [33, 34]). It is in clinical trial for stroke-induced brain edema (NCT02864953) and to reduce edema in metastatic brain tumor patients receiving radiosurgery (NCT02460874).

Choroid plexus, CSF secretion and CSF outflow

There has been some debate of the relative role of the choroid plexus in CSF secretion [1]. Praetorius and Dimiker [35] produced a comprehensive review of vectorial ion transport at the choroid plexus epithelium forming the basis for fluid secretion. Such ion transport is not only important for fluid secretion it is also involved in CSF ion homeostasis and secondary active transport. One focus of that review was Na, K and Cl transport by the choroid plexus epithelium. Interestingly, recent evidence indicated that stimulation of choroid plexus Na–K–Cl cotransporter-1 (NKCC-1; Slc12a2) contributed to post-hemorrhagic hydrocephalus [26] (see below). There was also growing evidence for the role of the choroid plexus in neuroinflammation. Thus, results indicated that the choroid plexus is a key site for the entry of T cells into brain in both animal and human stroke [36]. A potentially interesting model for studying choroid plexus development and function was described by Koshida et al. [37]. They found that mice with the transcription factor MafB gene knocked out had delayed differentiation and hypoplasia of the hindbrain choroid plexus, along with increased apoptosis and reduced proliferation in the epithelium.

There has long been substantial data that much CSF absorption is not via the arachnoid granulations/villi into the blood stream but rather into the lymph system via multiple routes [38]. The latter includes CSF drainage through the cribriform plate to the nasal lymphatics and the cervical lymph nodes and drainage via the spinal nerve roots to the lumbar lymph nodes [38], as well as via lymph vessels within the dura [39, 40]. Recently, Ma et al. [41] used noninvasive imaging techniques to quantify the transport of different sized tracers from CSF to the lymph nodes or blood in mice. For that species, they found that the lymph route predominated for both large and small tracers and that such drainage decreased with age.

Fluid and solute flow within the brain

The proposed brain glymphatic system for the brain continued to generate much interest [42], with ~ 80 papers in PubMed in 2017. It is proposed that fluid flow within the brain occurs via the perivascular space around the arterial system, then through astrocytes, with water movement via aquaporin-4, leaving the brain via the perivascular space around the venous system. Altered flow was proposed to occur and contribute to a multitude of neurological conditions (e.g. Alzheimer’s disease, idiopathic normal pressure hydrocephalus, migraine, diabetes, traumatic brain injury and stroke [43,44,45,46,47,48]). Burfeind et al. examined whether five aquaporin-4 single-nucleotide polymorphisms (SNPs) were associated with Alzheimer’s pathology or rate of cognitive decline after diagnosis. While none of the SNPs were associated with degree of pathology, two were associated with accelerated cognitive decline and two with slower decline [43].

While the glymphatic hypothesis engendered much interest, alternative hypotheses for fluid movement within the brain were proposed. Smith et al. [49, 50] recently questioned the experimental underpinning of the glymphatic hypothesis and provided evidence that solute movement through the brain is by diffusion rather than convection. In addition, Hannocks et al. and Pizzo et al. [51, 52] provided evidence that a perivascular space is present in all vessel calibers and that fluid/solute flow may occur through that space from arteriole to capillary to venule.

Although great progress was made in imaging of the perivascular pathways, there is a need for methods to quantify the importance of different pathways within the brain parenchyma. Currently, importance is attached to experiments manipulating aquaporin-4. There are questions, however, over the impact of such manipulations on not only movement of fluid through astrocytes, but also on extracellular diffusion (e.g. volume/tortuosity of the extracellular space).

CSF analysis

CSF analysis to aid in disease identification, progression and prognosis as well as for elucidating therapeutic targets continued to be a major focus across a wide range of neurological conditions (e.g. [53,54,55,56,57,58,59]). One area receiving especial attention was mild cognitive impairment and transition to dementia, particularly in relation to Aβ42 and tau. Some of the practical and theoretical difficulties in using such markers were outlined in recent reviews [55, 58, 60]. Aβ42, total tau and phosphorylated tau were also examined in relation to idiopathic normal pressure hydrocephalus [53]. One issue in CSF analysis is determining the underlying causes of altered CSF protein concentrations: changes may be due to altered barrier function or altered drainage, or both. However, a recent study found that increased CSF proteins are most probably derived from barrier dysfunction [61]. In this regard, it may be possible to determine the source of proteins based on their glycosylation state [62]. This is important because concentrations of CSF components in diagnostic studies need to be normalized to total protein content.

Studies using CSF for diagnosis are ongoing: for example, cytokines were measured in multiple sclerosis and polyneuropathy [63]. Analysis of CSF Aβ42, t-tau and p-tau may be used to distinguish Alzheimer’s disease from normal pressure hydrocephalus [53] and positive MRZ-1 antibody in CSF is a good indicator for multiple sclerosis [64]. Also, CSF chemo- and cytokines were measured in infants with post-hemorrhagic hydrocephalus [54]. There was also growing interest in disease-related changes in microRNAs, which may be encapsulated within CSF exosomes [65,66,67]. As well as being markers of disease processes, microRNAs may be important in cellular communication.

Hydrocephalus

Genetic causes for congenital hydrocephalus involving abnormal brain development continue to be reported: for example loss or mutations in the MPDZ gene affected ependymal cells and led to hydrocephalus [68, 69]. Also, mice lacking the Dusp16 gene developed hydrocephalus with brain overgrowth [70] and a mutation in B3GALNT2 gene led to hydrocephalus in Mexican horses [71].

Post-hemorrhagic hydrocephalus is a major problem in infants that survive at even earlier stages of prematurity. A post mortem study of human infants found that there was extensive disruption of the ventricular zone with loss of ependyma and infiltration of astrocytes [72], consistent with a common finding in models of congenital hydrocephalus of abnormal cell junction pathology and abnormal neurogenesis (reviewed by Rodriguez and Guerra [73]). An interesting insight into post-hemorrhagic hydrocephalus was provided by Karimy et al. [26]. They found an inflammation-mediated hypersecretion of CSF after intraventricular hemorrhage in rats. This was mediated by Toll-like receptor-4 activation at the choroid plexus and resulted in activation of Ste20-type stress kinase that phosphorylated and stimulated Na–K–Cl cotransporter-1 at the choroid plexus, thus, increasing CSF secretion. Such hypersecretion may help clear blood-derived neurotoxic compounds (e.g. hemoglobin and iron) from the CSF but also may participate in generating hydrocephalus, if flow pathways are impeded.

Idiopathic normal pressure hydrocephalus (NPH) continues to generate much interest mainly because of a large increased incidence in the elderly population and the variable response to shunt surgery. A meta-analysis of published papers on CSF biomarkers concluded that CSF Aβ42, t-tau and p-tau were increased compared to the normal state [53] and that Aβ42, tau and p-tau, neurofilament light chain and leucine-rich alpha-2-glycoprotein have the greatest predictive value for improvement with shunt surgery [74]. Evolving magnetic resonance techniques showed that in NPH the CSF pulsatility was increased in the aqueduct [75], that the brain parenchyma became stiffer [76], that cerebral blood flow in selected regions including the periventricular white matter was reduced and correlated with decline in cognitive function [77]. It was found that white matter perfusion increased after shunt surgery [78], an observation consistent with improvement in fractional anisotropy of white matter tracts after shunt surgery [79].

The meninges and other barriers

Historically, most studies on ‘barrier’ tissues focused on the cerebral endothelium or the choroid plexus. Relatively few studies examined the meninges, another site of the blood-CSF barrier. There were, however, a number of interesting studies this year that focused on the meninges and novel functions. Thus, recent evidence indicated that the meninges of perinatal mice contain neurogenic progenitor cells (radial glia-like) that can migrate into the cerebral cortex and form functional neurons [80]. In addition, Suter et al. [81] found that meninges from spinal cord produced both attractive and repulsive factors that help guide different types of axons and may regulate which axons traverse the boundary between the central and peripheral nervous systems.

Similar to the meninges, few studies have focused on glial barrier functions. Interestingly, Horng et al. [82] recently showed that reactive astrocytes around inflammatory lesions express claudin-1 and -4 and junctional adhesion molecule-A. Importantly, they found mice with astrocyte-specific knockout of claudin-4 had greater leukocyte infiltration and worse outcome in models of neuroinflammation.

Technological advances

Many advances in brain barriers and brain fluids research are driven by technological progress. Thus, for example, efforts to improve in vitro NVU/BBB models continue. These included increased use of induced pluripotent stem cells (iPSCs) to create models of the human neurovasculature (endothelial cells alone or in co-culture with derived pericytes, astrocytes and neurons) [83,84,85]. In addition to expression of many classic brain endothelial markers, such models exhibited very high transendothelial electrical resistances. Recently, in vitro models were derived from iPSCs from single patients [83, 86], a system with great potential for examining the impact of patient genetics on barrier properties. The production of such cells has required a lengthy protocol, but efforts to reduce that time were reported [87]. A major area that still remains to be resolved is how well these models replicate transport at the in vivo brain endothelial cell, e.g. efflux transporter activity. Efforts also progressed using microfluidics to produce a ‘BBB-on-a-chip’ [12, 88]. Such models were extended into disease-relevant models [89].

Much new insight into barrier function and brain fluid dynamics in health and disease were derived from advances in imaging. Advances in in vivo optical imaging was the subject of a Society for Neuroscience mini-symposium [90]. In vivo imaging can be facilitated by the choice of animal models. This was exemplified by an elegant study on angiogenesis and barrier-genesis in zebrafish [91]. Advances in the use of clinical and preclinical imaging for examining the NVU was also the subject of another conference [92]. As mentioned earlier, improved techniques and better resolution in magnetic resonance images have great potential for understanding the pathology of neurological diseases with, for example, observation of disturbances in white matter tracts [79] and tracking of transependymal and periarterial flow with the aid of a contrast enhancement agent [44].

There have been initial studies involving extensive genomics and large-scale proteomics that focused on the NVU, the cerebral endothelium and the choroid plexus in health and disease [93,94,95,96,97]. Advances in metabolomics have yet to be extensively applied and may provide important information. In an interesting alternative approach to using liquid chromatography coupled to tandem mass spectrometry (LC–MS) based proteomics, Lee et al. [98] used the publically-accessible Human Protein Atlas (mostly immunohistochemistry based) to examine the human cerebrovascular distribution of 20,000+ proteins. An affiliated database allowed comparisons between cell types within the brain and across organs. It also provided information on endothelial heterogeneity (e.g. by vessel caliber or adjacent cells), an understudied area.

Future directions

Major progress is being made in our basic science of the brain barriers and brain fluids, although there are major controversies. The ultimate goal of such understanding is, however, translating that information to the clinic. While there are some clinical trials, history shows us the difficulties in such translation.

References

  1. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  2. 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.

    Article  Google Scholar 

  3. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. 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.

    Article  PubMed  Google Scholar 

  5. 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.

    Article  PubMed  Google Scholar 

  6. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. 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.

    Article  PubMed  Google Scholar 

  8. 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.

    Article  Google Scholar 

  9. 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.

    Article  PubMed  Google Scholar 

  10. 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.

    Article  CAS  PubMed  Google Scholar 

  11. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  13. 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.

    Article  CAS  PubMed  Google Scholar 

  14. 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.

    Article  PubMed  Google Scholar 

  15. 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.

    Article  CAS  PubMed  Google Scholar 

  16. 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.

    Article  PubMed  Google Scholar 

  17. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  18. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  19. 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.

    Google Scholar 

  20. 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.

    Article  CAS  PubMed  Google Scholar 

  21. 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.

    Article  PubMed  Google Scholar 

  22. 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.

    Article  CAS  PubMed  Google Scholar 

  23. 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.

    Article  CAS  PubMed  Google Scholar 

  24. 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.

    Article  CAS  PubMed  Google Scholar 

  25. Jiang X, Andjelkovic AV, Zhu L, Yang T, Bennett MVL, Chen J, Keep RF, Shi Y. Blood–brain barrier dysfunction and recovery after ischemic stroke. Prog Neurobiol. 2017. https://doi.org/10.1016/j.pneurobio.2017.10.001.

    Google Scholar 

  26. 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.

    CAS  PubMed  Google Scholar 

  27. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  28. 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.

    Article  CAS  PubMed  Google Scholar 

  29. 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.

    Article  CAS  PubMed  Google Scholar 

  30. 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.

    Article  PubMed  Google Scholar 

  31. Lv J, Hu W, Yang Z, Li T, Jiang S, Ma Z, Chen F, Yang Y. Focusing on claudin-5: a promising candidate in the regulation of BBB to treat ischemic stroke. Prog Neurobiol. 2017. https://doi.org/10.1016/j.pneurobio.2017.12.001.

    PubMed  Google Scholar 

  32. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. 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.

    Article  CAS  PubMed  Google Scholar 

  34. Nakayama S, Taguchi N, Isaka Y, Nakamura T, Tanaka M. Glibenclamide and therapeutic hypothermia have comparable effect on attenuating global cerebral edema following experimental cardiac arrest. Neurocrit Care. 2017. https://doi.org/10.1007/s12028-017-0479-3.

    PubMed  Google Scholar 

  35. Praetorius J, Damkier HH. Transport across the choroid plexus epithelium. Am J Physiol Cell Physiol. 2017;312(6):C673–86.

    Article  PubMed  Google Scholar 

  36. 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.

    Article  CAS  PubMed  Google Scholar 

  37. 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.

    Article  CAS  PubMed  Google Scholar 

  38. Engelhardt B, Vajkoczy P, Weller RO. The movers and shapers in immune privilege of the CNS. Nat Immunol. 2017;18(2):123–31.

    Article  CAS  PubMed  Google Scholar 

  39. 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.

    Article  PubMed  Google Scholar 

  40. 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.

    Article  PubMed  Google Scholar 

  41. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Plog BA, Nedergaard M. The glymphatic system in central nervous system health and disease: past, present, and future. Ann Rev Pathol. 2017. https://doi.org/10.1146/annurev-pathol-051217-111018.

    Google Scholar 

  43. 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.

    Google Scholar 

  44. Ringstad G, Vatnehol SAS, Eide PK. Glymphatic MRI in idiopathic normal pressure hydrocephalus. Brain. 2017;140(10):2691–705.

    Article  PubMed  Google Scholar 

  45. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. 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.

    Article  PubMed  Google Scholar 

  47. 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.

    Article  PubMed  Google Scholar 

  48. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Smith AJ, Verkman AS. The “glymphatic” mechanism for solute clearance in Alzheimer’s disease: game changer or unproven speculation? FASEB J. 2017. https://doi.org/10.1096/fj.201700999.

    Google Scholar 

  50. 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.

    PubMed  PubMed Central  Google Scholar 

  51. Hannocks MJ, Pizzo ME, Huppert J, Despande T, Abbott NJ, Thorne RG, Sorokin L. Molecular characterization of perivascular drainage pathways in the murine brain. J Cereb Blood Flow Metab. 2017. https://doi.org/10.1177/0271678X17749689.

    PubMed  Google Scholar 

  52. 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  Google Scholar 

  53. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  54. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  55. 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.

    Article  Google Scholar 

  56. 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.

    Article  Google Scholar 

  57. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  58. 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.

    Article  CAS  PubMed  Google Scholar 

  59. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  60. 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.

    Article  CAS  PubMed  Google Scholar 

  61. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  62. 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.

    Article  CAS  PubMed  Google Scholar 

  63. 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.

    Article  PubMed  Google Scholar 

  64. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  65. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. 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.

    Article  CAS  PubMed  Google Scholar 

  67. 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.

    Article  CAS  PubMed  Google Scholar 

  68. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  70. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  71. 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.

    Article  CAS  PubMed  Google Scholar 

  72. 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.

    Article  PubMed  Google Scholar 

  73. Rodriguez EM, Guerra MM. Neural stem cells and fetal-onset hydrocephalus. Pediatr Neurosurg. 2017;52(6):446–61.

    Article  PubMed  Google Scholar 

  74. 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.

    Article  PubMed  Google Scholar 

  75. 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.

    Article  CAS  PubMed  Google Scholar 

  76. 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.

    Google Scholar 

  77. 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.

    Article  CAS  PubMed  Google Scholar 

  78. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  79. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  80. 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.

    Article  Google Scholar 

  81. Suter T, DeLoughery ZJ, Jaworski A. Meninges-derived cues control axon guidance. Dev Biol. 2017;430(1):1–10.

    Article  CAS  PubMed  Google Scholar 

  82. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  83. 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.

    Article  CAS  PubMed  Google Scholar 

  84. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  85. 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.

    Article  CAS  Google Scholar 

  86. 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.

    Article  CAS  PubMed  Google Scholar 

  87. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  88. 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.

    Article  CAS  Google Scholar 

  89. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  90. 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  Google Scholar 

  91. 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.

    Article  CAS  PubMed  Google Scholar 

  92. Netto JP, Iliff J, Stanimirovic D, Krohn KA, Hamilton B, Varallyay C, Gahramanov S, Daldrup-Link H, d’Esterre C, Zlokovic B, et al. Neurovascular unit: basic and clinical imaging with emphasis on advantages of ferumoxytol. Neurosurgery. 2017. https://doi.org/10.1093/neuros/nyx357.

    PubMed  Google Scholar 

  93. 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.

    Article  CAS  PubMed  Google Scholar 

  94. 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.

    Article  CAS  PubMed  Google Scholar 

  95. 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.

    Article  CAS  PubMed  Google Scholar 

  96. Jha MK, Kim JH, Song GJ, Lee WH, Lee IK, Lee HW, An SSA, Kim S, Suk K. Functional dissection of astrocyte-secreted proteins: implications in brain health and diseases. Prog Neurobiol. 2017. https://doi.org/10.1016/j.pneurobio.2017.12.003.

    PubMed  Google Scholar 

  97. 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.

    Article  CAS  PubMed  Google Scholar 

  98. 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.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Authors’ contributions

RFK wrote the initial draft. HCJ and LRD added sections and edited the manuscript. All authors read and approved the final manuscript.

Acknowledgements

None.

Competing interests

RFK, HCJ and LRD are co-Editors-in-Chief of Fluids Barriers CNS.

Availability of data and materials

Not applicable.

Consent for publication

Not applicable.

Ethics approval and consent to participate

Not applicable.

Funding

Not applicable.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Richard F. Keep.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Keep, R.F., Jones, H.C. & Drewes, L.R. Progress in brain barriers and brain fluid research in 2017. Fluids Barriers CNS 15, 6 (2018). https://doi.org/10.1186/s12987-018-0091-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12987-018-0091-8