Skip to main content
Erschienen in: Fluids and Barriers of the CNS 1/2017

Open Access 01.12.2017 | Editorial

Brain barriers and brain fluid research in 2016: advances, challenges and controversies

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

Erschienen in: Fluids and Barriers of the CNS | Ausgabe 1/2017

Abstract

This editorial highlights some of the advances that occurred in relation to brain barriers and brain fluid research in 2016. It also aims to raise some of the attendant controversies and challenges in such research.

Editorial

There continues to be a tremendous interest in brain barriers and brain fluids research. Thus, for example, in 2016 PubMed cites ~4000 papers on CSF, ~2950 papers on the blood–brain barrier (BBB)/neurovascular unit (NVU) and ~1200 papers on hydrocephalus. The purpose of this editorial is to highlight some of the advances made in 2016, as well as some of the attendant controversies. With regards to the latter, we also wish to highlight some of the challenges in such research. With the amount of research published in 2016, it is impossible to cover the breadth of important advances. The choices may seem idiosyncratic but hopefully useful.

Crossing the blood–brain barrier

One major aim of NVU/BBB research is to develop methods to improve the delivery of therapeutics to the brain. In that regard, there have been several advances in 2016. Deverman et al. used Cre recombination-based adeno-associated virus (AAV) targeted evolution (CREATE) to develop AAV capsids that better target the CNS [1]. That approach resulted in the generation of an AAV variant that widely transfected the mouse brain (neurons and astrocytes) after intravenous injection. The overall CREATE approach may greatly enhance gene transfer into the brain parenchyma for a variety of neurological conditions although there are some issues with the loading capacity of AAVs for large genes.
The delivery of proteins, including antibodies, at therapeutic concentrations across the highly impermeable blood–brain interface has been a major impediment in the development of treatments for neurological conditions. It is, therefore, noteworthy that Sevigny et al. [2] using an antibody, aducanumab, that targets aggregated amyloid-β managed to achieve a brain:plasma area under the curve (AUC) of 1.3% and to dose-dependently reduce both brain soluble and insoluble amyloid-β in a mouse Alzheimer’s disease model. In addition, in a phase 1b clinical trial, aducanumab reduced brain amyloid-β and slowed clinical deterioration in Alzheimer’s patients.
The use of focused ultrasound in conjunction with intravenously administered microbubbles to disrupt endothelial cell tight junctions and enhance blood–brain permeability of therapeutics has been the focus of multiple groups around the world (e.g. [35]). The feasibility of this approach has now been shown in non-human primates [5]. Focused ultrasound to enhance drug delivery is now in clinical trial for brain cancers (NCT02253212 and NCT02343991). Considerable effort has been made in such studies to determine the safety window for the ultrasound. A concern is that the safety window may vary if the cerebrovasculature is impacted by a neurological condition (e.g. stroke), particularly as that impact may not be homogeneous. Interestingly, Cho et al. also found that focused ultrasound with microbubbles locally down-regulated brain endothelial cell P-glycoprotein expression in rats [6]. This suggests that focused ultrasound can potentially impact brain drug delivery by both inducing structural changes and altering drug transport mechanisms. It does, however, raise the question as to what other endothelial properties, including gene expression, may be affected by the ultrasound. Overall, much effort is being directed towards enhancing delivery of therapeutics to the brain using multiple approaches. The readership is referred to a recent volume of Drug Discov Today Technol [7] on some of those approaches.

Fluid flow within the brain

The concept of a glymphatic system was first proposed by Iliff et al. [8]. It is proposed that there is a rapid movement of CSF from the subarachnoid space into brain along the artery/arteriole paravascular space (linked to arterial pulsation), a movement of fluid from that space through the brain parenchyma where astrocyte aquaporin4 plays an important role, and a return of fluid to subarachnoid space along the paravascular space of venules/veins. This circulation may provide a way of clearing potentially toxic waste products, such as amyloid-β, from the brain. Since its first proposal, the concept of a glymphatic system has gained great traction with ~40 papers addressing glymphatics in 2016 alone (PubMed) and there have been excellent reviews on the subject [911]. It has been the spur for innovative research, including the impact of sleep on the system [12] and even its potential role in regards to the eye [13]. The application of ultrafast MRI techniques has led to the suggestion that in addition to arterial pulsations, low frequency vasomotor activity may provide assistance to glymphatic propulsion through the interstitial space [14] and also has enabled visualization of the perivascular spaces with sub millimeter resolution [15]. It should be noted, however, that there have been recent challenges to the concepts underlying the proposed glymphatic system in particular related to the role of arterial pulsation [16] and the role of astrocyte water permeability/aquaporin4 [17].
There also continues to be some controversy as the relative role of the choroid plexus in CSF production. The readership is referred to a number of recent reviews addressing that subject [9, 1822]. In models of CSF flow there is generally a vectoral component (e.g. through the ventricular system), although there can be a large oscillation around that flow based on the respiratory and cardiac cycles [23]. However, recent evidence shows that within the ventricular system there are complex flow patterns generated by cilia networks [24]. Interestingly, those flow patterns change not only spatially, but also temporally. These flow patterns may be important in regulating communication between different periventricular areas.
A related question to how much fluid is produced by the choroid plexus is how much fluid is produced across the cerebral capillaries, by what mechanisms, and how is fluid production affected by disease states? Fluid production and ion transport by cells of the NVU have been dealt with in depth in a recent review by Hladky and Barrand [19]. While such fluid transport cannot result from filtration across capillaries and needs to be linked to ion transport, defining the precise mechanisms involved (and how to manipulate them) has proven very difficult [19]. Studies of brain endothelial transport in vivo are complicated by the presence of other components of the NVU. Perhaps progress can be made on vectoral ion and fluid transport with the newer generation of in vitro models with higher transendothelial electrical resistance (TEER) and lower paracellular permeability (see below). Choroid plexus epithelial cell cultures with high TEERs have been used to demonstrate fluid transport [25]. An understanding of NVU/BBB fluid transport in disease states is vital for developing methods to reduce brain edema formation [26]. Methods of treating cerebral edema have not changed in decades.

Choroid plexus

Apart from fluid secretion and ion transport, there has been an upsurge in interest in other functions of the choroid plexus. Evidence now indicates that it has important functions in immune surveillance [27], in stem cell regulation [28] and in brain signaling via extracellular vesicles [29]. The choroid plexus may also be a potential route for drug delivery into the CSF by transcytosis [30].

Hydrocephalus

Fetal hydrocephalus can be induced in mice by intracerebral injection of blood or lysophosphatidic acid (LPA) and such hydrocephalus can be blocked by an LPA receptor antagonist [31]. Interestingly, Park et al. [32] have recently investigated the molecular mechanisms underlying LPA-induced hydrocephalus and found that LPA is an upstream regulator of the protein YAP which is important in the development of ependymal junctions. Further dissection of these pathways may give insights into the potential causes of some forms of hydrocephalus.
Although congenital hydrocephalus can have non-genetic causes, such as intraventricular hemorrhage, over 100 genetic causes have now been identified [33]. Several biochemical pathways are implicated including Wnt signaling, cell junction integrity and especially cilia function (e.g. [34, 35]). However, despite our advances in understanding underlying causes, shunt treatment or ventriculoscopy continue to be the only treatment available for hydrocephalus [36]. Despite efforts to improve shunt design, treatment still carries large risks.
Much effort continues to be directed towards idiopathic normal pressure hydrocephalus (iNPH) defined by enlarged ventricles and a triad of clinical signs in older people. Many theories of pathogenesis exist for this complex condition and are comprehensively reviewed by Keong et al. [37]. Problems exist in distinguishing which patients will respond to shunt treatment (e.g. [38, 39]). A multimodal analysis including clinical signs, MRI parameters, intracranial pressure and CSF biomarkers did not predict shunt responders in a cohort of 284 patients [40]. Diffusion tensor imaging (DTI) has been used to determine white matter pathology in hydrocephalus [41] and changes after shunt treatment [42].

The blood–brain barrier/neurovascular unit in disease

Many neurological conditions are accompanied by BBB dysfunction (e.g. stroke, traumatic brain injury and brain neoplasms) with increased transfer of even proteins from blood to brain. There has always been a question of whether the BBB dysfunction is a secondary result of parenchymal injury or a primary site of injury. Recent results from Shi et al. found that endothelial-specific overexpression of a mutant form of actin depolymerizing factor reduced barrier disruption after stroke and ameliorated acute and chronic brain injury [43]. These results strongly suggest that barrier dysfunction may be a cause rather than just result from parenchymal damage after stroke and that it is an important therapeutic target.
Currently, the only therapeutic intervention for ischemic stroke is thrombolysis-induced reperfusion with tissue plasminogen activator (tPA) and now thrombectomy [44]. One major concern about the use of tPA to induce reperfusion is potential barrier dysfunction and hemorrhagic transformation. There has, therefore, been interest in combining tPA with an agent that protects the cerebrovasculature. Wahlgren et al. [45] conducted a phase II randomized clinical trial combining intravenous thrombolysis with tPA and imatinib, an agent that protects the cells of the NVU and reduces hemorrhagic transformation in animal stroke models [46, 47]. Wahlgren et al. found that imatinib was safe and that it showed some evidence of improved outcome in the stroke patients [45].
One function of brain endothelial cells is to supply essential nutrients to the brain. Thus, those cells express high levels of the glucose transport GLUT 1 (SLC2A1) and the large neutral amino acid transporter LAT1 (SLC7A5). The importance of mutations in GLUT1 have long been known (glucose transporter type 1 deficiency syndrome; [48]) but Tarlungeanu et al. [49] have recently found that homozygous mutations in LAT1 are associated with autistic traits and that endothelial deletion of LAT1 in mice causes severe neurological defects that can be ameliorated by intracerebroventricular administration of branch-chain amino acids.
The use of serum (and CSF) biomarkers for neurological conditions continues to be an area of intense research. For example, one particular focus has been the use of biomarkers for diagnosis and prognosis in different grades of traumatic brain injury; e.g. concussion [50]. A recent article by Dadas et al. [51] in Fluids Barriers CNS highlights the multiplicity of factors that impact biomarker levels in blood and presents modeling to enhance interpretation.
The cerebrovasculature has functions outside what are normally considered as ‘barrier’ properties. One such function is to form a vascular niche for neural stem cells promoting self-renewal and inhibiting differentiation [52] and there has been considerable interest in manipulating that niche to target cancer stem cells in glioblastoma and other brain cancers [53]. The cerebrovasculature can, however, also be a route for tumor invasion into surrounding tissue and Yadav et al. recently showed that such invasion can be reduced by targeting a chemokine receptor, CXCR4, improving survival in a mouse glioma model [54].

In vitro blood–brain barrier/neurovascular unit modeling

Helms et al. recently provided an excellent review of current in vitro NVU/BBB models, their uses and limitations [55]. Progress continues to be made. Of note, Canfield et al. [56] recently showed that induced pluripotent stem cells (iPSCs) from a single patient can be used to derive endothelial cells, astrocytes and neurons and produce a BBB/NVU model with transendothelial electrical resistances and permeabilities close to in vivo. An ultimate goal for a number of groups is to produce microfluidic devices, a ‘BBB on a chip’ (e.g. [5760]). An advantage of those models, as well some other larger scale models, is the incorporation of shear stress, an important determinant of endothelial characteristics. Big strides are being made in developing in vitro NVU/BBB models with high TEERs and low passive permeabilities to hydrophilic molecules. A remaining challenge is whether those models can also recapitulate other NVU/BBB characteristics; e.g. relevant expression/activity of transporters and the metabolizing enzymes that form the metabolic barrier, as well as rates of transcytosis.

The future

As can be seen from this brief summary of just some of the research into the brain barriers and brain fluids, major strides have been made in 2016. It is a vibrant area of research and one that is always associated with areas of controversy. Technical advances are being made that will hopefully address those controversies and open new avenues for research.

Authors contributions

RFK, HCJ and LRD contributed equally to 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.
Open AccessThis 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.
Literatur
1.
Zurück zum Zitat Deverman BE, Pravdo PL, Simpson BP, Kumar SR, Chan KY, Banerjee A, Wu WL, Yang B, Huber N, Pasca SP, et al. Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain. Nat Biotechnol. 2016;34(2):204–9.CrossRefPubMedPubMedCentral Deverman BE, Pravdo PL, Simpson BP, Kumar SR, Chan KY, Banerjee A, Wu WL, Yang B, Huber N, Pasca SP, et al. Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain. Nat Biotechnol. 2016;34(2):204–9.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Sevigny J, Chiao P, Bussiere T, Weinreb PH, Williams L, Maier M, Dunstan R, Salloway S, Chen T, Ling Y, et al. The antibody aducanumab reduces Abeta plaques in Alzheimer’s disease. Nature. 2016;537(7618):50–6.CrossRefPubMed Sevigny J, Chiao P, Bussiere T, Weinreb PH, Williams L, Maier M, Dunstan R, Salloway S, Chen T, Ling Y, et al. The antibody aducanumab reduces Abeta plaques in Alzheimer’s disease. Nature. 2016;537(7618):50–6.CrossRefPubMed
3.
Zurück zum Zitat Chu PC, Chai WY, Tsai CH, Kang ST, Yeh CK, Liu HL. Focused Ultrasound-induced blood–brain barrier opening: association with mechanical index and cavitation index analyzed by dynamic contrast-enhanced magnetic-resonance imaging. Sci Rep. 2016;6:33264.CrossRefPubMedPubMedCentral Chu PC, Chai WY, Tsai CH, Kang ST, Yeh CK, Liu HL. Focused Ultrasound-induced blood–brain barrier opening: association with mechanical index and cavitation index analyzed by dynamic contrast-enhanced magnetic-resonance imaging. Sci Rep. 2016;6:33264.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Samiotaki G, Karakatsani ME, Buch A, Papadopoulos S, Wu SY, Jambawalikar S, Konofagou EE. Pharmacokinetic analysis and drug delivery efficiency of the focused ultrasound-induced blood–brain barrier opening in non-human primates. Magn Reson Imaging. 2016;37:273–81.CrossRefPubMed Samiotaki G, Karakatsani ME, Buch A, Papadopoulos S, Wu SY, Jambawalikar S, Konofagou EE. Pharmacokinetic analysis and drug delivery efficiency of the focused ultrasound-induced blood–brain barrier opening in non-human primates. Magn Reson Imaging. 2016;37:273–81.CrossRefPubMed
5.
Zurück zum Zitat Wu SY, Sanchez CS, Samiotaki G, Buch A, Ferrera VP, Konofagou EE. Characterizing focused-ultrasound mediated drug delivery to the heterogeneous primate brain in vivo with acoustic monitoring. Sci Rep. 2016;6:37094.CrossRefPubMedPubMedCentral Wu SY, Sanchez CS, Samiotaki G, Buch A, Ferrera VP, Konofagou EE. Characterizing focused-ultrasound mediated drug delivery to the heterogeneous primate brain in vivo with acoustic monitoring. Sci Rep. 2016;6:37094.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Cho H, Lee HY, Han M, Choi JR, Ahn S, Lee T, Chang Y, Park J. Localized down-regulation of P-glycoprotein by focused ultrasound and microbubbles induced blood–brain barrier disruption in rat brain. Sci Rep. 2016;6:31201.CrossRefPubMedPubMedCentral Cho H, Lee HY, Han M, Choi JR, Ahn S, Lee T, Chang Y, Park J. Localized down-regulation of P-glycoprotein by focused ultrasound and microbubbles induced blood–brain barrier disruption in rat brain. Sci Rep. 2016;6:31201.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Gaillard PJ. BBB crossing assessment and BBB crossing technologies in CNS drug discovery. Drug Discov Today Technol. 2016;20:1–3.CrossRefPubMed Gaillard PJ. BBB crossing assessment and BBB crossing technologies in CNS drug discovery. Drug Discov Today Technol. 2016;20:1–3.CrossRefPubMed
8.
Zurück zum Zitat Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, Benveniste H, Vates GE, Deane R, Goldman SA, et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med. 2012;4(147):147ra111.CrossRefPubMedPubMedCentral Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, Benveniste H, Vates GE, Deane R, Goldman SA, et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med. 2012;4(147):147ra111.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Hladky SB, Barrand MA. Mechanisms of fluid movement into, through and out of the brain: evaluation of the evidence. Fluids Barriers CNS. 2014;11(1):26.CrossRefPubMedPubMedCentral Hladky SB, Barrand MA. Mechanisms of fluid movement into, through and out of the brain: evaluation of the evidence. Fluids Barriers CNS. 2014;11(1):26.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Xie L, Kang H, Xu Q, Chen MJ, Liao Y, Thiyagarajan M, O’Donnell J, Christensen DJ, Nicholson C, Iliff JJ, et al. Sleep drives metabolite clearance from the adult brain. Science (New York, NY). 2013;342(6156):373–7.CrossRef Xie L, Kang H, Xu Q, Chen MJ, Liao Y, Thiyagarajan M, O’Donnell J, Christensen DJ, Nicholson C, Iliff JJ, et al. Sleep drives metabolite clearance from the adult brain. Science (New York, NY). 2013;342(6156):373–7.CrossRef
13.
Zurück zum Zitat Wostyn P, Van Dam D, Audenaert K, Killer HE, De Deyn PP, De Groot V. A new glaucoma hypothesis: a role of glymphatic system dysfunction. Fluids Barriers CNS. 2015;12:16.CrossRefPubMedPubMedCentral Wostyn P, Van Dam D, Audenaert K, Killer HE, De Deyn PP, De Groot V. A new glaucoma hypothesis: a role of glymphatic system dysfunction. Fluids Barriers CNS. 2015;12:16.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Kiviniemi V, Wang X, Korhonen V, Keinanen T, Tuovinen T, Autio J, LeVan P, Keilholz S, Zang YF, Hennig J, et al. Ultra-fast magnetic resonance encephalography of physiological brain activity—glymphatic pulsation mechanisms? J Cereb Blood Flow Metab. 2016;36(6):1033–45.CrossRefPubMed Kiviniemi V, Wang X, Korhonen V, Keinanen T, Tuovinen T, Autio J, LeVan P, Keilholz S, Zang YF, Hennig J, et al. Ultra-fast magnetic resonance encephalography of physiological brain activity—glymphatic pulsation mechanisms? J Cereb Blood Flow Metab. 2016;36(6):1033–45.CrossRefPubMed
15.
Zurück zum Zitat Zong X, Park SH, Shen D, Lin W. Visualization of perivascular spaces in the human brain at 7T: sequence optimization and morphology characterization. NeuroImage. 2016;125:895–902.CrossRefPubMed Zong X, Park SH, Shen D, Lin W. Visualization of perivascular spaces in the human brain at 7T: sequence optimization and morphology characterization. NeuroImage. 2016;125:895–902.CrossRefPubMed
17.
Zurück zum Zitat Jin BJ, Smith AJ, Verkman AS. Spatial model of convective solute transport in brain extracellular space does not support a “glymphatic” mechanism. J Gen Physiol. 2016;148(6):489–501.CrossRefPubMed Jin BJ, Smith AJ, Verkman AS. Spatial model of convective solute transport in brain extracellular space does not support a “glymphatic” mechanism. J Gen Physiol. 2016;148(6):489–501.CrossRefPubMed
19.
Zurück zum Zitat Hladky SB, Barrand MA. Fluid and ion transfer across the blood–brain and blood-cerebrospinal fluid barriers; a comparative account of mechanisms and roles. Fluids Barriers CNS. 2016;13(1):19.CrossRefPubMed Hladky SB, Barrand MA. Fluid and ion transfer across the blood–brain and blood-cerebrospinal fluid barriers; a comparative account of mechanisms and roles. Fluids Barriers CNS. 2016;13(1):19.CrossRefPubMed
20.
Zurück zum Zitat Spector R, Keep RF, Robert Snodgrass S, Smith QR, Johanson CE. A balanced view of choroid plexus structure and function: focus on adult humans. Exp Neurol. 2015;267:78–86.CrossRefPubMed Spector R, Keep RF, Robert Snodgrass S, Smith QR, Johanson CE. A balanced view of choroid plexus structure and function: focus on adult humans. Exp Neurol. 2015;267:78–86.CrossRefPubMed
21.
Zurück zum Zitat Oreskovic D, Rados M, Klarica M. New concepts of cerebrospinal fluid physiology and development of hydrocephalus. Pediatr Neurosurg. 2016; doi:10.1159/000452169. [Epub ahead of print].PubMed Oreskovic D, Rados M, Klarica M. New concepts of cerebrospinal fluid physiology and development of hydrocephalus. Pediatr Neurosurg. 2016; doi:10.​1159/​000452169. [Epub ahead of print].PubMed
22.
Zurück zum Zitat Yamada S, Kelly E. Cerebrospinal fluid dynamics and the pathophysiology of hydrocephalus: new concepts. Semin Ultrasound CT MR. 2016;37(2):84–91.CrossRefPubMed Yamada S, Kelly E. Cerebrospinal fluid dynamics and the pathophysiology of hydrocephalus: new concepts. Semin Ultrasound CT MR. 2016;37(2):84–91.CrossRefPubMed
23.
Zurück zum Zitat Dreha-Kulaczewski S, Joseph AA, Merboldt KD, Ludwig HC, Gartner J, Frahm J. Inspiration is the major regulator of human CSF flow. J Neurosci. 2015;35(6):2485–91.CrossRefPubMed Dreha-Kulaczewski S, Joseph AA, Merboldt KD, Ludwig HC, Gartner J, Frahm J. Inspiration is the major regulator of human CSF flow. J Neurosci. 2015;35(6):2485–91.CrossRefPubMed
24.
Zurück zum Zitat Faubel R, Westendorf C, Bodenschatz E, Eichele G. Cilia-based flow network in the brain ventricles. Science (New York, NY). 2016;353(6295):176–8.CrossRef Faubel R, Westendorf C, Bodenschatz E, Eichele G. Cilia-based flow network in the brain ventricles. Science (New York, NY). 2016;353(6295):176–8.CrossRef
25.
Zurück zum Zitat Hakvoort A, Haselbach M, Galla HJ. Active transport properties of porcine choroid plexus cells in culture. Brain Res. 1998;795(1–2):247–56.CrossRefPubMed Hakvoort A, Haselbach M, Galla HJ. Active transport properties of porcine choroid plexus cells in culture. Brain Res. 1998;795(1–2):247–56.CrossRefPubMed
26.
Zurück zum Zitat O’Donnell ME. Blood–brain barrier Na transporters in ischemic stroke. Adv Pharmacol (San Diego, Calif). 2014;71:113–46.CrossRef O’Donnell ME. Blood–brain barrier Na transporters in ischemic stroke. Adv Pharmacol (San Diego, Calif). 2014;71:113–46.CrossRef
27.
Zurück zum Zitat Strazielle N, Creidy R, Malcus C, Boucraut J, Ghersi-Egea JF. T-lymphocytes traffic into the brain across the blood-CSF barrier: evidence using a reconstituted choroid plexus epithelium. PLoS ONE. 2016;11(3):e0150945.CrossRefPubMedPubMedCentral Strazielle N, Creidy R, Malcus C, Boucraut J, Ghersi-Egea JF. T-lymphocytes traffic into the brain across the blood-CSF barrier: evidence using a reconstituted choroid plexus epithelium. PLoS ONE. 2016;11(3):e0150945.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Silva-Vargas V, Maldonado-Soto AR, Mizrak D, Codega P, Doetsch F. Age-dependent niche signals from the choroid plexus regulate adult neural stem cells. Cell Stem Cell. 2016;19(5):643–52.CrossRefPubMed Silva-Vargas V, Maldonado-Soto AR, Mizrak D, Codega P, Doetsch F. Age-dependent niche signals from the choroid plexus regulate adult neural stem cells. Cell Stem Cell. 2016;19(5):643–52.CrossRefPubMed
29.
Zurück zum Zitat Balusu S, Van Wonterghem E, De Rycke R, Raemdonck K, Stremersch S, Gevaert K, Brkic M, Demeestere D, Vanhooren V, Hendrix A, et al. Identification of a novel mechanism of blood–brain communication during peripheral inflammation via choroid plexus-derived extracellular vesicles. EMBO Mol Med. 2016;8(10):1162–83.CrossRefPubMedPubMedCentral Balusu S, Van Wonterghem E, De Rycke R, Raemdonck K, Stremersch S, Gevaert K, Brkic M, Demeestere D, Vanhooren V, Hendrix A, et al. Identification of a novel mechanism of blood–brain communication during peripheral inflammation via choroid plexus-derived extracellular vesicles. EMBO Mol Med. 2016;8(10):1162–83.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Strazielle N, Ghersi-Egea JF. Potential pathways for CNS drug delivery across the blood-cerebrospinal fluid barrier. Curr Pharm Des. 2016;22(35):5463–76.CrossRefPubMed Strazielle N, Ghersi-Egea JF. Potential pathways for CNS drug delivery across the blood-cerebrospinal fluid barrier. Curr Pharm Des. 2016;22(35):5463–76.CrossRefPubMed
31.
Zurück zum Zitat Yung YC, Mutoh T, Lin ME, Noguchi K, Rivera RR, Choi JW, Kingsbury MA, Chun J. Lysophosphatidic acid signaling may initiate fetal hydrocephalus. Science Transl Med. 2011;3:99ra87.CrossRef Yung YC, Mutoh T, Lin ME, Noguchi K, Rivera RR, Choi JW, Kingsbury MA, Chun J. Lysophosphatidic acid signaling may initiate fetal hydrocephalus. Science Transl Med. 2011;3:99ra87.CrossRef
32.
Zurück zum Zitat Park R, Moon UY, Park JY, Hughes LJ, Johnson RL, Cho SH, Kim S. Yap is required for ependymal integrity and is suppressed in LPA-induced hydrocephalus. Nat Commun. 2016;7:10329.CrossRefPubMedPubMedCentral Park R, Moon UY, Park JY, Hughes LJ, Johnson RL, Cho SH, Kim S. Yap is required for ependymal integrity and is suppressed in LPA-induced hydrocephalus. Nat Commun. 2016;7:10329.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Kousi M, Katsanis N. The genetic basis of hydrocephalus. Annu Rev Neurosci. 2016;39:409–35.CrossRefPubMed Kousi M, Katsanis N. The genetic basis of hydrocephalus. Annu Rev Neurosci. 2016;39:409–35.CrossRefPubMed
34.
Zurück zum Zitat Foerster P, Daclin M, Asm S, Faucourt M, Boletta A, Genovesio A, Spassky N. mTORC1 signaling and primary cilia are required for brain ventricle morphogenesis. Development. 2017;144(2):201–10.CrossRefPubMed Foerster P, Daclin M, Asm S, Faucourt M, Boletta A, Genovesio A, Spassky N. mTORC1 signaling and primary cilia are required for brain ventricle morphogenesis. Development. 2017;144(2):201–10.CrossRefPubMed
35.
Zurück zum Zitat Wang X, Zhou Y, Wang J, Tseng IC, Huang T, Zhao Y, Zheng Q, Gao Y, Luo H, Zhang X, et al. SNX27 deletion causes hydrocephalus by impairing ependymal cell differentiation and ciliogenesis. J Neurosci. 2016;36(50):12586–97.CrossRefPubMed Wang X, Zhou Y, Wang J, Tseng IC, Huang T, Zhao Y, Zheng Q, Gao Y, Luo H, Zhang X, et al. SNX27 deletion causes hydrocephalus by impairing ependymal cell differentiation and ciliogenesis. J Neurosci. 2016;36(50):12586–97.CrossRefPubMed
36.
37.
Zurück zum Zitat Keong NC, Pena A, Price SJ, Czosnyka M, Czosnyka Z, Pickard JD. Imaging normal pressure hydrocephalus: theories, techniques, and challenges. Neurosurg Focus. 2016;41(3):E11.CrossRefPubMed Keong NC, Pena A, Price SJ, Czosnyka M, Czosnyka Z, Pickard JD. Imaging normal pressure hydrocephalus: theories, techniques, and challenges. Neurosurg Focus. 2016;41(3):E11.CrossRefPubMed
38.
Zurück zum Zitat Djukic M, Spreer A, Lange P, Bunkowski S, Wiltfang J, Nau R. Small cisterno-lumbar gradient of phosphorylated Tau protein in geriatric patients with suspected normal pressure hydrocephalus. Fluids Barriers CNS. 2016;13(1):15.CrossRefPubMedPubMedCentral Djukic M, Spreer A, Lange P, Bunkowski S, Wiltfang J, Nau R. Small cisterno-lumbar gradient of phosphorylated Tau protein in geriatric patients with suspected normal pressure hydrocephalus. Fluids Barriers CNS. 2016;13(1):15.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Ishikawa M, Yamada S, Yamamoto K. Early and delayed assessments of quantitative gait measures to improve the tap test as a predictor of shunt effectiveness in idiopathic normal pressure hydrocephalus. Fluids Barriers CNS. 2016;13(1):20.CrossRefPubMedPubMedCentral Ishikawa M, Yamada S, Yamamoto K. Early and delayed assessments of quantitative gait measures to improve the tap test as a predictor of shunt effectiveness in idiopathic normal pressure hydrocephalus. Fluids Barriers CNS. 2016;13(1):20.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Luikku AJ, Hall A, Nerg O, Koivisto AM, Hiltunen M, Helisalmi S, Herukka SK, Sutela A, Kojoukhova M, Mattila J, et al. Multimodal analysis to predict shunt surgery outcome of 284 patients with suspected idiopathic normal pressure hydrocephalus. Acta Neurochir (Wien). 2016;158(12):2311–9.CrossRef Luikku AJ, Hall A, Nerg O, Koivisto AM, Hiltunen M, Helisalmi S, Herukka SK, Sutela A, Kojoukhova M, Mattila J, et al. Multimodal analysis to predict shunt surgery outcome of 284 patients with suspected idiopathic normal pressure hydrocephalus. Acta Neurochir (Wien). 2016;158(12):2311–9.CrossRef
41.
Zurück zum Zitat Aojula A, Botfield H, McAllister JP 2nd, Gonzalez AM, Abdullah O, Logan A, Sinclair A. Diffusion tensor imaging with direct cytopathological validation: characterisation of decorin treatment in experimental juvenile communicating hydrocephalus. Fluids Barriers CNS. 2016;13(1):9.CrossRefPubMedPubMedCentral Aojula A, Botfield H, McAllister JP 2nd, Gonzalez AM, Abdullah O, Logan A, Sinclair A. Diffusion tensor imaging with direct cytopathological validation: characterisation of decorin treatment in experimental juvenile communicating hydrocephalus. Fluids Barriers CNS. 2016;13(1):9.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat 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. doi:10.1186/s12987-016-0048-8. (this issue)PubMed 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. doi:10.​1186/​s12987-016-0048-8. (this issue)PubMed
43.
Zurück zum Zitat Shi Y, Zhang L, Pu H, Mao L, Hu X, Jiang X, Xu N, Stetler RA, Zhang F, Liu X, et al. Rapid endothelial cytoskeletal reorganization enables early blood–brain barrier disruption and long-term ischaemic reperfusion brain injury. Nat Commun. 2016;7:10523.CrossRefPubMedPubMedCentral Shi Y, Zhang L, Pu H, Mao L, Hu X, Jiang X, Xu N, Stetler RA, Zhang F, Liu X, et al. Rapid endothelial cytoskeletal reorganization enables early blood–brain barrier disruption and long-term ischaemic reperfusion brain injury. Nat Commun. 2016;7:10523.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Goyal M, Menon BK, van Zwam WH, Dippel DW, Mitchell PJ, Demchuk AM, Davalos A, Majoie CB, van der Lugt A, de Miquel MA, et al. Endovascular thrombectomy after large-vessel ischaemic stroke: a meta-analysis of individual patient data from five randomised trials. Lancet (London, England). 2016;387(10029):1723–31.CrossRef Goyal M, Menon BK, van Zwam WH, Dippel DW, Mitchell PJ, Demchuk AM, Davalos A, Majoie CB, van der Lugt A, de Miquel MA, et al. Endovascular thrombectomy after large-vessel ischaemic stroke: a meta-analysis of individual patient data from five randomised trials. Lancet (London, England). 2016;387(10029):1723–31.CrossRef
45.
Zurück zum Zitat Wahlgren N, Thoren M, Hojeberg B, Kall TB, Laska AC, Sjostrand C, Hoijer J, Almqvist H, Holmin S, Lilja A, et al. Randomized assessment of imatinib in patients with acute ischaemic stroke treated with intravenous thrombolysis. J Intern Med. 2016;. doi:10.1111/joim.12576. [ Epub ahead of print]PubMed Wahlgren N, Thoren M, Hojeberg B, Kall TB, Laska AC, Sjostrand C, Hoijer J, Almqvist H, Holmin S, Lilja A, et al. Randomized assessment of imatinib in patients with acute ischaemic stroke treated with intravenous thrombolysis. J Intern Med. 2016;. doi:10.​1111/​joim.​12576. [ Epub ahead of print]PubMed
46.
Zurück zum Zitat Merali Z, Leung J, Mikulis D, Silver F, Kassner A. Longitudinal assessment of imatinib’s effect on the blood–brain barrier after ischemia/reperfusion injury with permeability MRI. Transl Stroke Res. 2015;6(1):39–49.CrossRefPubMed Merali Z, Leung J, Mikulis D, Silver F, Kassner A. Longitudinal assessment of imatinib’s effect on the blood–brain barrier after ischemia/reperfusion injury with permeability MRI. Transl Stroke Res. 2015;6(1):39–49.CrossRefPubMed
47.
Zurück zum Zitat Su EJ, Fredriksson L, Geyer M, Folestad E, Cale J, Andrae J, Gao Y, Pietras K, Mann K, Yepes M, et al. Activation of PDGF-CC by tissue plasminogen activator impairs blood–brain barrier integrity during ischemic stroke. Nat Med. 2008;14(7):731–7.CrossRefPubMedPubMedCentral Su EJ, Fredriksson L, Geyer M, Folestad E, Cale J, Andrae J, Gao Y, Pietras K, Mann K, Yepes M, et al. Activation of PDGF-CC by tissue plasminogen activator impairs blood–brain barrier integrity during ischemic stroke. Nat Med. 2008;14(7):731–7.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Leen WG, Taher M, Verbeek MM, Kamsteeg EJ, van de Warrenburg BP, Willemsen MA. GLUT1 deficiency syndrome into adulthood: a follow-up study. J Neurol. 2014;261(3):589–99.CrossRefPubMed Leen WG, Taher M, Verbeek MM, Kamsteeg EJ, van de Warrenburg BP, Willemsen MA. GLUT1 deficiency syndrome into adulthood: a follow-up study. J Neurol. 2014;261(3):589–99.CrossRefPubMed
49.
Zurück zum Zitat Tarlungeanu DC, Deliu E, Dotter CP, Kara M, Janiesch PC, Scalise M, Galluccio M, Tesulov M, Morelli E, Sonmez FM, et al. Impaired amino acid transport at the blood brain barrier is a cause of autism spectrum disorder. Cell. 2016;167(6):1481–94.CrossRefPubMed Tarlungeanu DC, Deliu E, Dotter CP, Kara M, Janiesch PC, Scalise M, Galluccio M, Tesulov M, Morelli E, Sonmez FM, et al. Impaired amino acid transport at the blood brain barrier is a cause of autism spectrum disorder. Cell. 2016;167(6):1481–94.CrossRefPubMed
50.
Zurück zum Zitat Zetterberg H, Blennow K. Fluid biomarkers for mild traumatic brain injury and related conditions. Nat Rev Neurol. 2016;12(10):563–74.CrossRefPubMed Zetterberg H, Blennow K. Fluid biomarkers for mild traumatic brain injury and related conditions. Nat Rev Neurol. 2016;12(10):563–74.CrossRefPubMed
51.
Zurück zum Zitat Dadas A, Washington J, Marchi N, Janigro D. Improving the clinical management of traumatic brain injury through the pharmacokinetic modeling of peripheral blood biomarkers. Fluids Barriers CNS. 2016;13(1):21.CrossRefPubMed Dadas A, Washington J, Marchi N, Janigro D. Improving the clinical management of traumatic brain injury through the pharmacokinetic modeling of peripheral blood biomarkers. Fluids Barriers CNS. 2016;13(1):21.CrossRefPubMed
52.
Zurück zum Zitat Shen Q, Goderie SK, Jin L, Karanth N, Sun Y, Abramova N, Vincent P, Pumiglia K, Temple S. Endothelial cells stimulate self-renewal and expand neurogenesis of neural stem cells. Science (New York, NY). 2004;304(5675):1338–40.CrossRef Shen Q, Goderie SK, Jin L, Karanth N, Sun Y, Abramova N, Vincent P, Pumiglia K, Temple S. Endothelial cells stimulate self-renewal and expand neurogenesis of neural stem cells. Science (New York, NY). 2004;304(5675):1338–40.CrossRef
53.
Zurück zum Zitat Gilbertson RJ, Rich JN. Making a tumour’s bed: glioblastoma stem cells and the vascular niche. Nat Rev Cancer. 2007;7(10):733–6.CrossRefPubMed Gilbertson RJ, Rich JN. Making a tumour’s bed: glioblastoma stem cells and the vascular niche. Nat Rev Cancer. 2007;7(10):733–6.CrossRefPubMed
54.
Zurück zum Zitat Yadav VN, Zamler D, Baker GJ, Kadiyala P, Erdreich-Epstein A, DeCarvalho AC, Mikkelsen T, Castro MG, Lowenstein PR. CXCR4 increases in vivo glioma perivascular invasion, and reduces radiation induced apoptosis: a genetic knockdown study. Oncotarget. 2016. (in press) Yadav VN, Zamler D, Baker GJ, Kadiyala P, Erdreich-Epstein A, DeCarvalho AC, Mikkelsen T, Castro MG, Lowenstein PR. CXCR4 increases in vivo glioma perivascular invasion, and reduces radiation induced apoptosis: a genetic knockdown study. Oncotarget. 2016. (in press)
55.
Zurück zum Zitat Helms HC, Abbott NJ, Burek M, Cecchelli R, Couraud PO, Deli MA, Forster C, Galla HJ, Romero IA, Shusta EV, et al. In vitro models of the blood–brain barrier: an overview of commonly used brain endothelial cell culture models and guidelines for their use. J Cereb Blood Flow Metab. 2016;36(5):862–90.CrossRefPubMed Helms HC, Abbott NJ, Burek M, Cecchelli R, Couraud PO, Deli MA, Forster C, Galla HJ, Romero IA, Shusta EV, et al. In vitro models of the blood–brain barrier: an overview of commonly used brain endothelial cell culture models and guidelines for their use. J Cereb Blood Flow Metab. 2016;36(5):862–90.CrossRefPubMed
56.
Zurück zum Zitat 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. 2016;. doi:10.1111/jnc.13923. [Epub ahead of print]PubMed 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. 2016;. doi:10.​1111/​jnc.​13923. [Epub ahead of print]PubMed
57.
Zurück zum Zitat Adriani G, Ma D, Pavesi A, Kamm RD, Goh EL. A 3D neurovascular microfluidic model consisting of neurons, astrocytes and cerebral endothelial cells as a blood–brain barrier. Lab Chip. 2016;. doi:10.1039/c6lc00638h. [Epub ahead of print] Adriani G, Ma D, Pavesi A, Kamm RD, Goh EL. A 3D neurovascular microfluidic model consisting of neurons, astrocytes and cerebral endothelial cells as a blood–brain barrier. Lab Chip. 2016;. doi:10.​1039/​c6lc00638h. [Epub ahead of print]
58.
Zurück zum Zitat Tourovskaia A, Fauver M, Kramer G, Simonson S, Neumann T. Tissue-engineered microenvironment systems for modeling human vasculature. Exp Biol Med (Maywood, NJ). 2014;239(9):1264–71.CrossRef Tourovskaia A, Fauver M, Kramer G, Simonson S, Neumann T. Tissue-engineered microenvironment systems for modeling human vasculature. Exp Biol Med (Maywood, NJ). 2014;239(9):1264–71.CrossRef
59.
Zurück zum Zitat van der Helm MW, van der Meer AD, Eijkel JC, van den Berg A, Segerink LI. Microfluidic organ-on-chip technology for blood–brain barrier research. Tissue Barriers. 2016;4(1):e1142493.CrossRefPubMedPubMedCentral van der Helm MW, van der Meer AD, Eijkel JC, van den Berg A, Segerink LI. Microfluidic organ-on-chip technology for blood–brain barrier research. Tissue Barriers. 2016;4(1):e1142493.CrossRefPubMedPubMedCentral
60.
Zurück zum Zitat Wang YI, Abaci HE, Shuler ML. Microfluidic blood–brain barrier model provides in vivo-like barrier properties for drug permeability screening. Biotechnol Bioeng. 2017;114(1):184–94.CrossRefPubMed Wang YI, Abaci HE, Shuler ML. Microfluidic blood–brain barrier model provides in vivo-like barrier properties for drug permeability screening. Biotechnol Bioeng. 2017;114(1):184–94.CrossRefPubMed
Metadaten
Titel
Brain barriers and brain fluid research in 2016: advances, challenges and controversies
verfasst von
Richard F. Keep
Hazel C. Jones
Lester R. Drewes
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Fluids and Barriers of the CNS / Ausgabe 1/2017
Elektronische ISSN: 2045-8118
DOI
https://doi.org/10.1186/s12987-017-0052-7

Weitere Artikel der Ausgabe 1/2017

Fluids and Barriers of the CNS 1/2017 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.