Skip to main content

A year in review: brain barriers and brain fluids research in 2022

Abstract

This aim of this editorial is to highlight progress made in brain barrier and brain fluid research in 2022. It covers studies on the blood-brain, blood-retina and blood-CSF barriers (choroid plexus and meninges), signaling within the neurovascular unit and elements of the brain fluid systems. It further discusses how brain barriers and brain fluid systems are impacted in CNS diseases, their role in disease progression and progress being made in treating such diseases.

Introduction

Research related to blood-brain barriers and brain fluids continues to thrive. Here the Editors-in-Chief of Fluids and Barriers of the CNS highlight some of the wide range of important articles published in 2022, as well as some controversies. As always, such a review cannot cover the multitude of excellent studies that have been published.

Elements of the blood-brain barrier and the neurovascular unit

The brain endothelial cells and the surrounding neurovascular unit (NVU) have a crucial role in protecting the brain, forming the blood-brain barrier (BBB), maintaining a homeostatic microenvironment, and in facilitating normal brain function (e.g., neurovascular coupling). The development and maintenance of the BBB, the role of cellular and acellular elements of the NVU in regulating the barrier, and differences from non-barrier areas of the brain (circumventricular organs) have recently been reviewed by Ben-Zvi & Liebner [1]. Crouch et al. [2] have also described the process of angiogenesis and endothelial/mural cell interactions in the 2nd trimester in human brain. It should be noted that the NVU interacts with systemic biology (e.g., the brain-gut axis and the systemic immune system) forming a ‘wider’ NVU [3].

One technique that has had a major impact on our understanding of the cerebrovasculature is the use of single cell transcriptomics. Application of this methodology has provided a mass of publicly available information on the different cell types and sub-types in the NVU (including potential cell specific markers), the existence of arterio-venous zonation, and the impact of disease. Such data is available for humans [4,5,6] as well as prior and new data in mice (e.g. [7]).

Brain endothelial cells

Tight junctions (TJ) that link cerebral endothelial cells are an essential component of the BBB and are comprised of transmembrane proteins including claudin-5 and occludin and cytoplasmic plaque proteins including ZO-1. Hashimoto et al. [8] have described a claudin-5 mutation in two patients with alternating hemiplegia with microcephaly that changes this normally barrier forming claudin to an anion-selective channel. Greene et al. [9] have demonstrated the importance of claudin-5 in epilepsy. They report that claudin-5 is reduced and BBB permeability enhanced in patients with drug-resistant epilepsy, and that reducing claudin-5 exacerbates kainate-induced seizures in mice and can induce spontaneous seizures, neuroinflammation and death. Importantly, RepSox, a regulator of claudin-5 could prevent seizures in a mouse epileptic model. Boye et al. [10] have identified the role of Netrin-1-Unc5B signaling in regulating claudin-5 via the Wnt/β-catenin pathway. Endothelial-specific loss of Unc5B reduced claudin-5 expression and disrupted the BBB.

Evidence indicates that beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) has a role in cerebrovascular injury in cerebral amyloid angiopathy and Alzheimer’s disease. Occludin was discovered as a new substrate of BACE1 and leads to cerebral small vessel disease pathogenesis ([11]). Toth et al. [12] have described sortilin as another novel junction regulator. Rats lacking this receptor in brain endothelial cells exhibited a leaky BBB that was associated with a relocalization of the junction proteins ZO-1, VE-cadherin and β-catenin. Loss of sortilin reduced phosphorylation of Akt and increased phosphorylation of Erk1/2.

Major facilitator superfamily domain-containing protein 2 (MFSD2A) plays a major role at the brain endothelium. This dual-function protein transports lysophosphatidylcholines and omega-3 fatty acids and regulates transcytosis. Using cryo-electron microscopy, Martinez-Molledo et al. [13] elucidated the structure of MFSD2A in relation to its transport function. This structural information may allow strategies for development of MFSD2A inhibitors for modulating BBB permeability.

Multiple mechanisms are involved in regulating cerebral blood flow to meet the demands of neural metabolism (neurovascular coupling). In addition, changes in luminal shear stress alter endothelial function as shown by Harraz et al. [14] who identified Piezo 1 channels in endothelial cells that act as mechanosensors and regulate intracellular Ca2+ channels using ex vivo retinal preparations and isolated endothelial cells.

Pericytes and astrocytes at the neurovascular unit (NVU)

Pericytes play an important role at the NVU in controlling brain endothelial function. Thus, Berthiaume et al. [15] have examined the effects of optically ablating pericytes on capillary function in mice and found dilation of capillaries and altered capillary blood flow with no disruption of vascular permeability. However, remodeling of neighboring pericytes was observed and resulted in restored pericyte coverage and capillary tone, a recovery that was slower in aged mice. Ayloo et al. [16] found that vitronectin secreted by pericytes is an important regulator of BBB and the blood-retinal barrier function via interaction with endothelial integrin α5 receptors. The interaction suppressed endothelial transcytosis. Pericytes can produce factors that protect the BBB from hypoxia [17]. Sancho et al. [18] have recently provided evidence that KATP channels are important regulators of pericyte and brain endothelial cell membrane potentials and that adenosine activates those channels via A2A receptors with important implications for brain blood flow regulation.

Astrocytes are another major regulator of NVU and BBB function. Morales et al. [19] have developed a new way of specifically tracking and ablating perivascular astrocytes using the endogenous gene megalencephalic leukoencephalopathy with subcortical cysts 1 to drive expression of Cre fused to a mutated estrogen ligand-binding domain. They found that loss of perivascular astrocytes led to abnormal location of endothelial junction proteins and microgliosis. This approach should be a useful tool to study the role of perivascular astrocytes. Mills et al. [20] have used two-photon chemical ablation of astrocytes and found that neighboring astrocytes extended their processes to cover denuded areas but this was reduced with aging. Yshii et al. [21] have used a viral gene delivery system (adeno associated virus, AAV) to enhance interleukin-2 production in reactive astrocytes and thereby regulate brain-resident Treg cells. This method was protective in mouse stroke, traumatic brain injury and multiple sclerosis models. Importantly, it did not impact the peripheral immune system.

Aquaporin-4 (AQP4), a water channel, is highly expressed in astrocyte endfeet where it forms large arrays (orthogonal arrays of particles). Zhu et al. [22] have examined the impact of a mutant form of AQP4 (AQP4-A25Q) in mice. They found it greatly reduced the number of arrays without affecting overall mRNA and protein levels. Importantly, they found that the mutant had reduced brain edema, astrocyte endfeet swelling and neurological deficits as well as improved survival after stroke. This emphasizes the functional importance of these physical arrays. Evidence regarding the importance of AQP4 subcellular localization in brain water homeostasis in health and disease is reviewed in Salman et al. [23]. They propose that targeting AQP4 localization may be an alternative strategy to developing AQP4 inhibitors. Evidence on the importance of AQP4 also comes from Mader et al. [24] who found that in utero exposure to maternal anti-AQP4 antibodies affected brain and vascular development and BBB leakiness in male mice.

Astrocytes and brain endothelial cells express connexins important for cell:cell communication. De Bock et al. [25] have shown the importance of connexin 43 in both cell types in BBB disruption following lipopolysaccharide (LPS) administration. Kim et al. [26] have examined the pathways by which inflammatory mediators induce a reactive astrocyte phenotype to then induce BBB disruption. Tumor necrosis factor-α induced a reactive phenotype via STAT3 and SERPINA3 upregulation (encoding alpha 1-antichymotrypsin). Apoliprotein E (ApoE) is produced in the brain by astrocytes and microglia as well as systemically by the liver. In humans, the ApoE4 allele is associated with increased risk of Alzheimer’s disease and a leaky BBB. Jackson et al. [27] have found that mice expressing human APOE4 have a leaky BBB, including impaired TJs and reduced astrocyte end-foot coverage. Removing astrocyte-produced ApoE4 reduced that phenotype indicating an important role of that cell type. Barisano et al. [28] have used a multi-omics approach to examine the effects of knocking-in human APOE4 into mice particularly focusing on brain endothelial dysfunction and pericyte damage. While APOE4 is abundant in brain, it is also present in the periphery. Liu et al. [29] have found that peripheral APOE4 has adverse effects on the cerebrovasculature and cognition in mice. This suggests that targeting peripheral APOE4 may be a therapeutic strategy for Alzheimer’s disease.

Gerrits et al. [30] have used single-nucleus RNA sequencing to examine changes in the NVU in patients with frontotemporal dementia with heterozygous mutations in the progranulin gene. They identified disease-associated endothelial and astrocyte subtypes and a loss of pericytes.

While most studies have focused on how components of the NVU impact brain endothelial cell function, there is important communication in the opposite direction. For example, Li et al. [31] found brain endothelial cells release microvesicles containing Ascl1 during stroke-like conditions in vitro that convert astrocytes into neural progenitors. Endothelial specific overexpression of Ascl1 improved behavioral outcomes in mice after stroke. Endothelial platelet-derived growth factor B (PDGFB) plays a crucial role in brain angiogenesis by recruiting pericytes. Vazquez-Liebanas et al. [32] have examined the effects of loss of PDGFB in adult mice after angiogenesis and found that PDGFB loss led to a gradual reduction in endothelial pericyte coverage and increased BBB permeability. Lee et al. [33] have also found that endothelial-derived lactate is a substrate fueling pericyte metabolism.

One important function of signaling at the NVU is in neurovascular development. This requires co-ordination so that vascular development meets the metabolic needs of neural cells. Rattner et al. [34] describe current knowledge of the signaling pathways involved.

Oligodendrocytes

While attention has focused on the NVU in grey matter, there is also an analogous structure in white matter termed the oligovascular unit (OVU). The OVU contains brain endothelial cells, oligodendrocytes and oligodendrocyte progenitor cells (amongst other components). Xiao et al. [35] have found that IL-17/CXCL5 signaling in the OVU plays an important role in white matter injury in humans and mice.

Microglia

In contrast to the role of astrocytes and pericytes at the NVU, the role of microglia has received relatively little attention. Knopp et al. [36] has reviewed evidence on the connection between microglia and the cerebral capillaries (~ 30% of microglia are connected to a capillary) and the effects of disease states on microglia-endothelial communication. Very recently, Csaszar et al. [37] have described the importance of microglia in neurovascular coupling under normal and hypoperfusion conditions, roles mediated by purinergic signaling.

Fibroblasts

One often neglected CNS cell type are the fibroblasts. They are present in the perivascular space, the choroid plexus stroma and the meninges. They also play a key role in scar formation after brain injury. The role of these cells has recently been reviewed [38].

Extracellular matrix

The extracellular matrix is an important component of the NVU. Halder et al. [39] have recently reviewed how genetically manipulating laminins and their integrin receptors impact BBB development/maturation and their role in maintaining BBB integrity. Nirwane and Yao [40] have also recently reviewed the cell specific expression of different laminins within the NVU and their function. De et al. [41] have found that β8-integrin activates extracellular matrix adhesion to promote cerebrovascular development. β8-integrin is essential for transforming growth factor β-dependent gene expression in the endothelium.

The glycocalyx on the luminal surface of the cerebral endothelium also has many important functions. Zhu et al. [42] have found that glycocalyx destruction, such as in stroke, is associated with increased transcellular, caveolin-1 mediated, transport at the BBB.

Elements of the blood-CSF barrier and the CSF and glymphatic systems

Choroid plexus

The choroid plexus is thought by most to be the primary site of CSF secretion with a lesser contribution from the neurovasculature. In terms of CSF secretion, there has been particular focus on the role of the sodium/potassium/chloride cotransporter, NKCC1 [43,44,45,46], and transient receptor potential vanilloid 4 (TRPV4) channels [44, 47] at the choroid plexus epithelium. Such research may lead to a clinical method for reducing CSF production. There is still debate about the mechanism(s) by which fluid is transported across the choroid plexus epithelium. That is reviewed by MacAulay et al. [48] with a particular focus on the hypothesis that certain transporters (e.g., NKCC1) may serve as a water channel as well as transport solutes. There has been great interest in how fluid movement in the brain varies diurnally particularly in relation to removal of waste or toxic metabolites (e.g., via the glymphatic system) and it should be noted that the choroid plexus has a circadian rhythm that may impact CSF homeostasis and flow (reviewed in [49]).

The choroid plexus is also increasingly understood to play a major role in neuroinflammation. For instance, Alzheimer’s disease causes marked changes in choroid plexus myeloid cells [50]. The NLR family pyrin domain containing 3 (NLRP3) inflammasome is involved in CP hypersecretion after intraventricular hemorrhage [46]. The choroid plexus is also a target for reducing T cell infiltration in experimental autoimmune encephalitis [51] and there are very early inflammatory changes at the choroid plexus after intracerebral hemorrhage [52].

As with the cerebral endothelium, choroid plexus development is regulated by Wnt/b-catenin signaling. Parichha et al. [53] have examined the effects of disrupting such signaling in mice and organoids from human embryonic stem cells. Both loss and gain of function of β-catenin disrupted choroid plexus epithelium development indicating the importance of tight regulation of Wnt/β-catenin signaling for correct choroid plexus development.

There have been an increasing number of reports using MRI to examine changes in the choroid plexus in different human disease states. That includes increases in human choroid plexus size or volume in multiple sclerosis (including pre-symptomatic) [54,55,56], Alzheimer’s disease [57], depression [58] and obesity [59]. Such changes may reflect neuroinflammatory events at the choroid plexus as evidenced by positron emission tomography [56, 58]. MRI has also been used to examine changes in choroid plexus function in patients including perfusion [60], water movement [61] and contrast permeability [57].

Understanding the roles of the choroid plexus would be greatly assisted by developing methods to specifically target choroid plexus epithelial cells to induce loss or gain of function. Jang & Lehtinen [62] have recently reviewed progress in that area. It would also be assisted by developing new in vitro choroid plexus epithelium models. Hulme et al. [47] have described a human choroid plexus papilloma (HIBCPP) cell line that had morphology similar to the epithelium in vivo, intermediate transepithelial electrical resistance and correct ion transporter location. Very importantly, these cells secreted fluid when transient receptor potential vanilloid 4 (TRPV4) channels were activated. In contrast, the same group found the porcine choroid plexus-Riems (PCP-R) cell line, while having a high electrical resistance, had incorrect ion transporter location [63].

Ciliated ependyma

Ependymal cell motile cilia produce CSF flow patterns on the cell surface which may provide a communication route between different periventricular regions. Faubel et al. [64] have been examining the CDKL5 deficiency disorder (CDD), the most common cause of infant epilepsy. CDKL5 regulates cilia length and function, and Faubel et al. found longer cilia, abnormal cilia motion and altered CSF flow patterns in the ventral 3rd ventricle in Cdkl5 knockout mice. Mutations in another epilepsy-related gene, Yes1, produced similar alterations in cilia function and CSF flow patterns. Animals with these altered flow patterns had increased risk of anesthesia-induced seizures and the authors suggest that altered motile cilia function has an essential role in CDD related seizures and that cilia may be a therapeutic target. Changes in the ependymal/cilia function are hypothesized to be crucial in hydrocephalus and recent studies are described in the ‘Hydrocephalus’ section below.

Cerebrospinal fluid

The importance of CSF composition has recently been stressed by results from Iram et al. [65]. They found that infusing CSF from young mice enhanced oligodendrogenesis and memory in aged mice. Effects on oligodendrocyte progenitor cells were mediated by serum response factor (a transcription factor) which was activated by fibroblast growth factor 17 in the CSF of young mice.

There continue to be many studies examining CSF for biomarkers of disease. This has been a particular focus in Alzheimer’s disease where studies have examined the correlation between CSF biomarkers and disease pathology [66,67,68], examining potential biomarkers that are upregulated before the onset of cognitive impairment [69] and distinguishing between different types of dementia [70, 71]. Apart from β-amyloid (Aβ) and tau proteins, there has been particular interest in microglia-derived sTREM2 (soluble triggering receptor expressed on myeloid cells 2) [72,73,74]. One biomarker-related question is how well CSF and plasma levels correlate? Shahim et al. [75] examined that question in professional athletes with repetitive head injury. They found that plasma levels of T-tau, GFAP, Aβ40, and Aβ42 do not correlate with CSF levels and concluded that the plasma levels of those biomarkers are not informative for diagnosing the late effects of repetitive brain injury.

Eninger et al. [76] have used non-directed liquid chromatography/mass spectrometry to examine the age-related CSF proteome in mouse models of β-amyloidosis and α-synucleinopathy. They found most of the CSF proteome changes were linked to microglia and astrocytes, identifying a panel of 20 glial-derived proteins. They found a good correlation between those proteins and disease-associated glial genes previously identified by single-cell transcriptomics. Lobanova et al. [77] have shown that it is possible to identify protein aggregates in CSF and plasma using optical single-molecule imaging and high-resolution atomic force microscopy. They compared samples from control and Parkinson’s disease patients and found larger aggregates with a different composition in Parkinson’s disease (50% α-synuclein and 50% Aβ versus 30%:70% in controls).

Cerebrospinal fluid is also an alternative to a brain biopsy for examining brain tumor DNA and RNA [78, 79]. The advantages (and disadvantages) of this approach and comparisons to using blood circulating tumor DNA have recently been reviewed by Tivey et al. [80].

A variety of techniques are available to profile immune cell changes in the CSF in disease states, including cell transcriptomics. Piehl et al. [81] have used RNA analysis to examine CSF changes during normal aging and cognitive impairment. They found that monocytes signal to clonal CD8 + T cells via CXCL16-CXCR6 in patients with cognitive impairment. Yazdani et al. [82] have used the same technique to examine the CSF of patients with amyotrophic lateral sclerosis (ALS) and described clonally expanded CD4+ and CD8+ T cells with characteristic patterns of gene expression.

Meninges

The functions of the arachnoid membrane (blood-arachnoid barrier) have been relatively understudied. However, recent studies have indicated that the arachnoid membrane highly expresses many solute transporters regulating CSF composition. For example, Takeuchi et al. [83] have used quantitative targeted absolute proteomics to compare the expression of a variety of transporters at the blood-arachnoid barrier and the blood-spinal cord barrier. Some organic anionic and cationic drug transporters were only expressed in the former. Sangha et al. [84] have found two important folate transporters, the reduced folate-carrier and the proton-coupled folate transporter, are present at the blood-arachnoid barrier.

CSF circulation and outflow

Two recent reviews have dealt with methods of measuring CSF secretion rate [85] and using different imaging techniques to examine CSF flow [86]. There is debate about the relative importance of different CSF outflow pathways (e.g., across cribriform plate, along cranial nerves, via dural lymphatics or arachnoid villi). Decker et al. [87] used MRI to track CSF outflow in young and old mice and found that it was predominantly via the cribriform plate to the nasopharyngeal lymphatics and that CSF turnover was reduced in aged mice. Albayram et al. [88] have also used MRI to track brain lymphatic networks in human. Using lymph albumin as a marker, they identified lymphatic structures along the dural venous sinuses and along cranial nerves in the dorsal and ventral regions, respectively. They also noted cervical lymph node atrophy and thickening of lymphatics channels with aging.

As well as being involved in CSF drainage, the meningeal lymphatic endothelial cells have phagocytic activity that can take up proteins, polysaccharides and virus particles [89]. Li et al. [90] have also found that several neurotropic viruses drain from the brain via the meningeal lymphatics to the cervical lymph nodes and blocking such drainage increased neurological damage and mortality in virus-infected mice.

There also continues to be great interest in the newly described skull/dural channels linking the subarachnoid space to the skull bone marrow. Pulous et al. [91] have reported that in mice CSF tracers can exit the subarachnoid space and migrate to the skull bone marrow. In meningitis, they found that bacteria use this route to invade the skull hematopoietic niche. Similarly, Mazzitelli et al. [92] described that after a spinal cord injury in mice CSF migrates to the bone marrow hematopoietic niche promoting myelopoiesis and migration of myeloid cells into meninges.

The relationship between cardiac cycle and CSF flow is poorly understood. Yang et al. [93] have used functional MRI (fMRI) to examine the temporal relationship between hemodynamic changes (tissue blood volume) and CSF flow.

Glymphatics

The glymphatic system continues to generate much interest and some controversy (see [94,95,96,97,98] for recent reviews). Impairments in the rate of perivascular penetrance of CSF (or CSF tracers) into brain has been reported in numerous neurological conditions including multiple sclerosis [99], small vessel disease [100], chronic poor sleep [101], and raised intracranial pressure [102]. Also, movement of CSF into brain has been proposed to be a significant source of early brain edema after anoxia [103].

Drieu et al. [104] have recently identified a sub-population of perivascular macrophages closely associated with the brain arterial tree that regulate arterial motion and associated CSF flow. Loss of the perivascular macrophages impaired access of CSF to the perivascular space. Mestre et al. [105] have examined pial structure in mice, how it may influence perivascular fluid flow (and CSF filtration), and how pial structure is altered with aging and Alzheimer’s disease.

Genetics and the blood-brain barriers

In 2022, FBCNS published a series of articles in a thematic series entitled, ‘Genetic Disorders and Genetic Manipulation at the Blood-brain Barriers’ (Genetic disorders and genetic manipulation at the blood-brain barriers (biomedcentral.com)). Studies on patients with genetic disorders, and the use of animals with genetic manipulation have provided insight into the role of particular proteins in blood-brain, blood-retina and blood-CSF barrier function in health and disease. Hopefully, such information will provide ways to treat genetic disorders and manipulate barrier function to treat neurological disease.

An example of such research is the use of induced pluripotent stem cells (iPSCs) from patients with genetic disorders to produce brain microvascular endothelial cell (BMEC)-like cells to examine changes in BBB function in vitro. For example, Linville et al. [106] have used such cells to examine the impact of Huntington’s disease mutations on the function and properties of the BBB.

Gene delivery is one potential method for treating genetic disorders. Sundaram et al. [107] have found that using an AAV directed to deliver the monocarboxylic acid transporter 8 (AAV-BBR1-Mct8) to the brain endothelium of mouse models of Allan-Herndon-Dudley syndrome could increase brain tri-iodothyronine (T3) levels and ameliorate morphological and behavioral deficits.

Transgenic mice have proven a valuable tool for determining the role of specific proteins at the blood-brain, blood-retina and blood-CSF barriers and the NVU. For example, Goncalves & Antonetti [108] have recently reviewed the insights gained from such animals on BBB and blood-retina barriers and their regulation. Halder et al. [39] have also recently described the impact of genetically manipulating laminins and integrins.

Apart from DNA mutations, changes in gene (and protein) expression can occur at multiple levels, The role of epigenetics at the blood-brain barriers is relatively understudied. There is recent evidence that the demethylation factor Tet methylcytosine dioxygenase 2 (TET2) regulates the tight junction protein ZO-1 [109] and p-glycoprotein (ABCB1) [110] and that differences in methylation of the claudin-5 gene are associated with the trajectory of cognitive decline in patients [111].

Another level of regulation is by non-coding RNAs (including microRNAs, long non-coding RNAs and circular RNAs. Sun et al. [112] have reviewed the roles of such RNAs at the BBB in different neurological disorders. One mechanism by which extracellular vesicles shed by one cell can affect other cells is by encapsulating micro RNAs. Brain endothelial cell-derived vesicles are an example of this mechanism [113].

Neurological disorders

BBB/NVU function and brain fluid dynamics are altered in many neurological disorders. Indeed, there is increasing evidence that targeting such dysfunction can ameliorate the effects of several diseases. This section highlights progress in understanding such changes in select diseases.

SARSCoV-2/COVID-19

There continue to be extensive studies examining the neurological and neuropsychiatric consequences of SARS-CoV-2 (COVID-19) infection, including long-term effects (long COVID). Potential underlying mechanisms have recently been reviewed [114]. In patients dying with infection, Agrawal et al. [115] found evidence of vascular pathology and microglial activation in the majority (80%) of subjects. In the elderly, pathological changes were superimposed on pre-existing brain disease, making that population more at risk of neurological sequelae. Etter et al. [116] found that COVID-19 patients with severe neurological effects had BBB impairment, markers of microglia activation and a polyclonal B cell response. Similarly, in CVID-19 patients who died with minimal respiratory involvement, Lee et al. [117] reported multifocal vascular damage with BBB disruption and activation of endothelial cells and the classical complement cascade. Jarius et al. [118] found evidence of blood-CSF barrier disruption in patients with neurological sequelae that was still present 30 days post-infection.

Understanding the neurological effects of COVID-19 may be aided by studies on non-human primates. In such a model, Rutkai et al. [119] found evidence that COVID-19 induces hypoxia-ischemia injury with brain microhemorrhages, hypoxia, neuroinflammation and neurodegeneration. Virus was only detected sparsely in brain endothelial cells and did not correlate with brain injury severity. It should be noted, however, that Altmayer et al. [120] found increased plasma endothelial biomarkers in patients with COVID-19-associated encephalitis, although whether those effects are linked to direct endothelium infection or secondary effects is uncertain. Krasemann et al. [121] found SARS-CoV-2 can infect human iPSC-derived BMEC-like cells in vitro.

Hydrocephalus

Hydrocephalus is defined as enlarged cerebral ventricles with or without raised intracranial pressure and is a disorder of brain fluid circulation. Several major causes exist for hydrocephalus: congenital, usually due to genetic or developmental defects, brain hemorrhage, brain trauma or anoxia, and infections. Additionally, normal pressure hydrocephalus of the elderly has uncertain causes. The major causes of hydrocephalus may vary globally. Aukrust et al. [122] recently analyzed studies from Africa and found that post-infectious hydrocephalus is the single most common cause (28% of cases). The prevalence of hydrocephalus is also strongly age dependent. Thus, Isaacs et al. [123] reported an 8-fold decline between pediatric (perinatal to 18 years old) and adult (19 to 64) populations, but a 17-fold increase from adult to the elderly (65 years and above).

Genetic factors

Genetic factors frequently involve altered ependymal cell and cilia function as an underlying cause of hydrocephalus. For example, Han et al. [124] have examined the effects of the deletion of the RNA binding protein Hu antigen R (HuR) in mice. HuR deletion resulted in impaired ependymal cell development, defective motile ciliogenesis and hydrocephalus. They found that HuR binds to mRNA transcripts related to ciliogenesis, including cilia and flagella-associated protein 52. Harkins et al. [125] have examined the effects of loss of nuclear factor one X (NFIX) in mice which is a transcription factor involved in normal ependymal development and the constitutive loss of NFIX results in hydrocephalus. They found loss of ependymal NFIX in adult brains causes enlarged ventricles, abnormal localization of adhesion molecules and shedding of ependymal cells. Prh mice have a point mutation in the Ccdc39 gene causing ciliary dyskinesia and progressive hydrocephalus. The mutation also causes periventricular white matter injury and neuroinflammation. Iwasawa et al. [126] found that an anti-inflammatory agent, Bindarit, improved cortical development and reduced white matter injury in these mice while having only a mild effect on ventricular volume.

It should be noted that the hypothesis that ependymal motile cilia dysfunction causes hydrocephalus by altering CSF flow has been questioned by Duy et al. [127]. They suggest that some of the mutations that induce motile cilia dysfunction and induce hydrocephalus may also have independent effects on neurodevelopment and non-CNS effects that may result in the ventricular enlargement. They also note that motile ciliopathies rarely cause hydrocephalus in humans (in contrast to mice). In line with this, the same group examined genetic mutations linked to congenital hydrocephalus [128]. They found convergence of risk genes in embryonic neuroepithelial stem cells. TRIM71 harbored the most de novo mutations and it is specifically expressed in neuroepithelial cells. In mice, neuroepithelial-specific Trim71 deletion or mutation induced hydrocephalus. This may be due to premature neuroepithelial cell differentiation, reduced neurogenesis and cortical hypoplasia. The latter may increase cortical compliance and induce secondary ventricular enlargement.

Intraventricular hemorrhage

One major cause of hydrocephalus is intraventricular hemorrhage (IVH). In premature infants IVH is due to germinal matrix hemorrhage and survivors can have significant developmental delay [129]. Blood products in the ventricles are suspected to be responsible for the dilatation and Miller et al. [129] have shown that hemoglobin injected into the ventricles of neonatal rats can induce hydrocephalus. Neuroinflammation is also involved in IVH-induced hydrocephalus (reviewed in [130]). Inflammatory markers in the CSF are associated with hypersecretion of CSF via hyperactivity of choroid plexus transporters, raising the potential for future pharmaceutical treatment [43]. Methods for mitigation of the effects of IVH are being explored and Vinukonda and La Gamma [131] used stem cell lines to improve outcome in animal models. In adults, the CLEAR III trial provided evidence that intraventricular fibrinolytics improve mortality after IVH, but there was no significant difference in functional outcome [132]. Kuramatsu et al. [133] have now performed a meta-analysis of studies using intraventricular fibrinolytics for intracerebral hemorrhage-induced IVH in adults and found significant improvement in functional outcome and that this was linked to early (< 48 h) treatment. This may inform future trials.

Normal pressure hydrocephalus

Normal pressure hydrocephalus (NPH) is a condition occurring in the elderly showing dilated ventricles, together with gait, urinary and cognitive dysfunctions, but without consistent raised CSF pressure. The problem of identifying patients that would benefit from shunt treatment remains ongoing and CSF biomarkers are being investigated extensively. Among a series of biomarkers tested, phosphorylated-tau and total-tau have been found to be significantly increased in lumbar CSF of shunt non-responsive patients using a meta-analysis [134], although earlier studies have refuted this and more sophisticated analytical methods may be helpful in the future. Neurofilament light (Nfl), a protein of axonal degeneration, has also been suggested as a biomarker for NPH [135]. Alzheimer’s disease (AD) and NPH often co-exist but after shunt, patients with AD have a worse outcome cognitively [136].

Magnetic imaging studies have shown that NPH is associated with a decrease in grey matter in some brain areas and increase in others and in particular the cerebellum was adversely affected [137]. MRI analysis (cine-PC) has been used to evaluate CSF and brain recovery, including tissue stiffness, over 15 months after shunting [138]. Intracranial compliance after shunting is slow to recover and shows oscillatory changes over 12 months and does not follow measured changes in intracranial pressure or CSF volume [139]. Such information could be used in the future for post shunt patient management. EEG analysis together with normalized power variance as a measure of cortical activity, is also being explored to distinguish shunt responsive NPH patients [140]. The underlying cause for NPH is elusive although dysfunction of the glymphatic system may be implicated (reviewed in [141]).

Idiopathic intracranial hypertension

Idiopathic intracranial hypertension (IIH) is a condition of raised intracranial pressure without dilatation of the ventricles most frequently found in obese females. The most common symptoms are severe headache and papilledema which can lead to visual loss. Ultrasound scans of the optic nerve are a reliable diagnostic test [142]. Magnetic resonance elastography has detected changes in the pituitary position and increase in brain stiffness in patients with IIH [143]. A general metabolic dysregulation has been detected in IIH patients [144] and Westgate et al. [145] found changes in systemic glucocorticoid metabolism with an increase in11β-hydroxysteroid dehydrogenase (11β-HSD1) and 5α-reductase activity which decreased after weight loss and a fall in ICP. Changes in the pituitary and pineal glands have also been implicated [146]. Treatment for IIH is designed to reduce CSF pressure and frequently involves CSF shunting but this can result in over-drainage and has a high revision rate (reviewed in [147]).

Fenestration of the optic nerve to release pressure has been tested as a treatment but with limited effect [148]. Causes for this condition are elusive and dural venous sinus stenosis treated by stenting has received much attention (reviewed in [149]). Diet-induced obesity leads to a similar condition in rats providing a reliable animal model [150].

Stroke

Neuroinflammation plays a central role in brain injury and brain repair following stroke (reviewed in [151]). One important element of the inflammatory response is leukocyte influx across the BBB. Multiple approaches have been used to try and reduce such extravasation. Recently, Arias et al. [152] found that a PECAM-1 neutralizing antibody reduces leukocyte influx and affects their spatial distribution within brain. BBB disruption occurs after stroke and it contributes to neuroinflammation and edema formation. Thus, for example, Ng et al. [153] found that midline shift (a measure of brain edema on imaging) correlates with the degree of BBB disruption as well as poor patient outcome after large vessel ischemic stroke.

Sphingosine-1-phosphate (S1P) regulates BBB permeability with differential effects via the S1P receptors 1 and 2. Hansen et al. [154] have found that the S1P receptor 4 is also present at the cerebral endothelium (apical membrane) in multiple species. Using KO mice, siRNA as well as antagonists and agonists, they found this receptor protects against BBB disruption and that receptor expression is reduced after stroke. Recent evidence points to osteopontin having a role in stroke [155]. Osteopontin disrupted the BBB in vitro and a neutralizing antibody reduced glial activation, BBB disruption and brain edema as well as improving neurological outcome and survival after stroke in mice.

Cerebral small vessel disease is a major cause of stroke, cognitive decline and dementia. Wardlaw et al. [156] have reviewed human and preclinical data indicating how altered endothelial cell dysfunction in cerebral small vessel disease impacts cell:cell interactions in the NVU and leads to brain injury. Mishra et al. [157] have used gene mapping to identify mutations in TRIM47 as being important in cerebral small vessel disease pathology. Using a novel rat KO line, Quick et al. [158] have provided evidence that loss of the phospholipase flippase, ATP11B, causes endothelial dysfunction and white matter injury similar to human cerebral small vessel disease.

Wnt/β-catenin signaling is an instrumental pathway for inducing barrier properties of brain endothelial cells. Ji et al. [159] tested lithium, a known activator of Wnt signaling, and found a protective effect against ischemic stroke and reperfusion injury. Therefore, agents that activate Wnt signaling may have therapeutic value following cerebral vascular insults. This concept was further developed by Martin et al. [160] who report an elegant study in which Wnt7a ligands were genetically engineered into BBB-specific Wnt activators that mitigated ischemic stroke infarction and glioblastoma expansion. They demonstrated that the signaling specificity of Wnt ligands is adjustable and targetable, thus, defining the normalizing of the BBB/NVU as a new modality to treat CNS disorders.

Ischemic and hemorrhagic stroke induce different injury mechanisms. Solar et al. [161] have recently written a very extensive review on the effects of subarachnoid hemorrhage on the BBB and NVU.

Cerebral cavernous malformations (CCMs) and arteriovenous malformations (AVMs)

Inherited CCMs are due to loss-of-function mutations in three genes, CCM1, CCM2 and CCM3. Animal and cellular models have been generated to examine how these loss of function mutations lead to CCMs [162, 163]. Such models allow examination of potential therapeutics and investigation of factors that may exacerbate CCM lesion development. Thus, McCurdy et al. [164] have found that a β1-integrin monoclonal antibody reduces CCM1 lesion development in mice, and Lai et al. [165] have examined the role of neuroinflammatory astrocytes in CCM3 lesion development. Interestingly, Fang et al. [166] found that loss of the NOGOB receptor in brain endothelial cells results in CCM-like lesions with dilated vessels, BBB hyperpermeability and cerebral hemorrhage. This was linked to an impairment in histone acetylation-mediated CCM1/2 expression.

To understand the abnormal vasculature in AVMs, Winkler et al. [6] compared single cell transcriptomics of the normal human cerebrovasculature and AVMs. They particularly identified pathological endothelial transformations and vascularly derived inflammation.

Aging and dementias

Normal aging in patients impacts the BBB resulting in small leaks detectable on dynamic contrast-enhanced magnetic resonance imaging [167]. The effects of aging can be exacerbated in patients with Alzheimer’s disease and other dementia’s such as frontotemporal dementia [30, 167, 168]. Yang et al. [5] have developed a ‘vessel isolation and nuclei extraction for sequencing’ technique to examine vascular and perivascular cell transcriptomes (and arteriovenous zonation) in brains from individuals with Alzheimer’s disease and those without cognitive impairment. They found evidence that there is selective pericyte vulnerability and blood flow dysregulation in Alzheimer’s disease brains; that two thirds of the top genes associated with risk of Alzheimer’s disease are expressed in the vasculature; and that those genes are linked to endothelial protein transport, adaptive immunity and extracellular matrix pathways. Consequently, activation of the Wnt/β-catenin pathway may be one way of ameliorating the vascular effects of Alzheimer’s disease [169].

Two recent studies have examined how the vascular transcriptome is changed in mice with cerebral amyloid angiopathy (CAA; Swedish mutation) during aging [170, 171]. Both studies highlight the inflammatory changes, alterations in endothelial biology and the impact of age on the transcriptome. There are always concerns over how well mouse genetic models replicate human CAA (and Alzheimer’s disease in general). Tachida et al. [172] have created a new mouse model with expanded CAA pathology by crossing mice expressing human amyloid precursor protein (APP) in endothelial cells with knock-in mice expressing a construct containing a humanized Aβ region along with two pathogenic mutations (Swedish and Iberian). The relevance of this model remains to be evaluated.

Neuroinflammation, multiple sclerosis (MS) and amyotrophic lateral sclerosis (ALS)

Proulx and Engelhardt [173] recently reviewed evidence on immune privilege and immune surveillance in the CNS, with the latter being primarily within the subarachnoid and perivascular spaces. Activation of the immune system has a detrimental effect in a variety of neurological disorders. For example, Frieser et al. [174] identified an important role of resident CD8+ T cells in autoimmune encephalitis and Vincenti et al. [175] found that after clearance of an infection with attenuated lymphocytic choriomeningitis virus reactivation of tissue-resident CD8+ T cells may cause immunopathology, but cooperation of CD4+ T cells is required.

Neuroinflammation is a crucial component of MS with autoreactive lymphocytes attacking CNS myelin. For example, the antibody natalizumab reduces leukocyte entry across the BBB and is used in patients with relapsing-remitting MS. Charabati et al. [176] have identified dual immunoglobulin domain containing cell adhesion molecule (DICAM) as another potential target for impeding leukocyte entry into brain in MS. Epstein-Barr virus infection has been epidemiological linked to MS and Lanz et al. [177] found that a transcription factor (EBNA1) in that virus molecularly mimics glial cell adhesion molecule (GlialCAM).Interestingly, they found cross-reactive antibodies in CSF from MS patients. The gut-brain axis may also be important in MS. Ntranos et al. [178] have identified microbially derived metabolites of tryptophan and phenylalanine that can enter the brain and may induce neurotoxicity in MS.

Surprisingly, the lung also harbors a microbiome that appears to influence significantly the susceptibility for developing CNS autoimmune disease such as multiple sclerosis. Using selective antibiotics to manipulate the lung microbial population, Hosang et al. [179] demonstrated that the pulmonary microbiome regulates the immune reactivity of central nervous tissue and thereby influences its susceptibility to autoimmune disease development. Thus, a lung-brain axis may emerge as an important link between the periphery and CNS disease.

In an animal MS model (experimental autoimmune encephalitis), Hermans et al. [180] have identified a role of the oncostatin-M/CCL20 axis in BBB impairment and T helper 17 cell recruitment. While BBB dysfunction may be secondary to neuroinflammation, Nishihara et al. [181] found evidence of an intrinsic BBB impairment with MS when comparing iPSC-derived BMEC-like cells from control and MS patients. This suggests the brain endothelium is a therapeutic target.

In ALS, Yazdani et al. [82] have found that T cell phenotypes in blood and CSF are good predictors of disease progression with high CD4+FOXP3 effector T cell expression being associated with poor survival, whereas a high frequency of Treg cells was associated with better survival. They also found evidence of clonally expanded CD4+ and CD8+ cells in CSF and propose that modulating adaptive immunity is a therapeutic option for ALS.

Psychiatric disorders

There is growing awareness that altered BBB function may play an important role in psychiatric disorders such as depression [182]. For example, Dion-Albert et al. [183] have found that chronic social and sub-chronic variable stress promotes BBB alterations in female mice and that inducing localized BBB disruption in the prefrontal cortex induces anxiety- and depression-like behaviors. Importantly, they have identified soluble E-selectin (an adhesion molecule expressed on endothelial cells) in the serum of women with major depressive disorders. Matsuno et al. [184] have identified a role of vascular endothelial growth factor (VEGF) and its receptor VEGFR2 in stress-induced depression-like behavior in mice.

Dai et al. [185] have found an impairment in meningeal lymphatics in a mouse model of depression (sub-chronic variable stress) which only occurred in females. Manipulating the meningeal lymphatics altered this sex-difference in susceptibility to stress-induced depression- and anxiety-like behaviors. Together with the work at the BBB, this suggests that changes in brain extracellular composition (either due to greater influx at the BBB or reductions in clearance systems) may contribute to depression- and anxiety-like behaviors.

Drug delivery

Enhancing drug delivery across the blood-brain barriers

There continues to be great interest in the use of focused ultrasound with microbubbles to transiently open the BBB for drug delivery (see [186] for a recent review) and it is being used in clinical trials. For example, Epelbaum et al. [187] have developed an implantable ultrasound device that they have used to repeatedly disrupt the BBB in patients with mild Alzheimer’s disease (NCT03119961). There has been less interest in disrupting the blood-CSF barrier at the choroid plexus but Kung et al. [188] report that a single low energy shockwave pulse can transiently disrupt that barrier in rats.

As described earlier, increasing claudin-5 expression to reduce BBB permeability may be of benefit in a number of neurological diseases. By contrast, reducing claudin-5 expression/function may enhance drug delivery across the BBB. Wakayama et al. [189] review the current state of such research and potential safety concerns. While transient reductions in claudin-5 can enhance drug delivery, chronic reductions cause adverse effects in mice and non-human primates.

There has been research into using extracellular vesicles to carry cargo across the BBB although more in vivo work is required in mammals [190]. The choroid plexus epithelial cell is one source of extracellular vesicles and Pauwells et al. [191] found such vesicles home to the brain and choroid plexus when delivered systemically. The vesicles can deliver cargo to astrocytes and microglia. Marra et al. [113] have found that extracellular vesicles from a subset of endothelial progenitor cells can help protect the NVU in ischemic/neurodegenerative retinopathy.

Delivery of therapeutic agents to brain tumors remains challenging with the blood-brain tumor barrier being very heterogeneous (reviewed in [192]). A wide range of innovative techniques are being developed to deliver effective drug concentrations to tumors (e.g., [193, 194]).

For gene therapy, AAVs that specifically target the brain and cross the BBB would be of great use. Goertsen et al. [195] have described an AAV capsid variant that causes brain-wide transgene expression and has reduced delivery to liver after systemic administration. The variant was effective in mice and non-human primates. AAV delivery of hexosaminidase has been employed in a clinical trial for Tay-Sachs disease, although direct delivery to the CSF/brain was utilized [196]. Duan et al. [197] have used an AAV that penetrates the BBB to deliver Cas9 and a single-guide RNA to target a mutant APP allele in a mouse Alzheimer’s disease model. Brain Aβ deposition, microgliosis, neurite degeneration and cognitive impairment were ameliorated. Recently, Grashoff et al. [198] have used AAVs to deliver short regulatory DNA sequences to enhance tight junction and transporter expression at the brain endothelium.

The blood-brain barriers as therapeutic targets

While most attention has focused on enhancing drug delivery across the blood-brain barriers, those barriers themselves may be therapeutic targets. Martin et al. [160] have targeted Wnt signaling at the BBB by genetically engineering Wnt7a ligands into BBB-specific Wnt activators to avoid pleiotropic effects. They found benefit in both glioblastoma and ischemic stroke models.

Jang et al. [199] found that the chemotherapeutic agent methotrexate impacted non-cancerous choroid plexus cells. A choroid plexus-targeted AAV was used in mice to increase expression of the antioxidant enzyme superoxide dismutase 3. This strategy appeared to increase the antioxidant defense capacity of CSF and reduce methotrexate-induced hippocampal lipid peroxidation and the associated learning and memory deficits.

Conclusions

The Editors-in-Chief of Fluids and Barriers of the CNS thank the authors, reviewers and editorial board members for their contributions to advancing the field in the past year and their support for the journal.

Data Availability

Not applicable.

References

  1. Ben-Zvi A, Liebner S. Developmental regulation of barrier- and non-barrier blood vessels in the CNS. J Intern Med. 2022;292(1):31–46.

    Article  CAS  PubMed  Google Scholar 

  2. Crouch EE, Bhaduri A, Andrews MG, Cebrian-Silla A, Diafos LN, Birrueta JO, Wedderburn-Pugh K, Valenzuela EJ, Bennett NK, Eze UC, et al. Ensembles of endothelial and mural cells promote angiogenesis in prenatal human brain. Cell. 2022;185(20):3753–3769e3718.

    Article  CAS  PubMed  Google Scholar 

  3. Tiedt S, Buchan AM, Dichgans M, Lizasoain I, Moro MA, Lo EH. The neurovascular unit and systemic biology in stroke - implications for translation and treatment. Nat Reviews Neurol. 2022;18(10):597–612.

    Article  Google Scholar 

  4. Garcia FJ, Sun N, Lee H, Godlewski B, Mathys H, Galani K, Zhou B, Jiang X, Ng AP, Mantero J, et al. Single-cell dissection of the human brain vasculature. Nature. 2022;603(7903):893–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Yang AC, Vest RT, Kern F, Lee DP, Agam M, Maat CA, Losada PM, Chen MB, Schaum N, Khoury N, et al. A human brain vascular atlas reveals diverse mediators of Alzheimer’s risk. Nature. 2022;603(7903):885–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Winkler EA, Kim CN, Ross JM, Garcia JH, Gil E, Oh I, Chen LQ, Wu D, Catapano JS, Raygor K, et al. A single-cell atlas of the normal and malformed human brain vasculature. Science. 2022;375(6584):eabi7377.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Jeong HW, Dieguez-Hurtado R, Arf H, Song J, Park H, Kruse K, Sorokin L, Adams RH. Single-cell transcriptomics reveals functionally specialized vascular endothelium in brain. eLife. 2022;11(10):05.

    Google Scholar 

  8. Hashimoto Y, Poirier K, Boddaert N, Hubert L, Aubart M, Kaminska A, Alison M, Desguerre I, Munnich A, Campbell M. Recurrent de novo mutations in CLDN5 induce an anion-selective blood-brain barrier and alternating hemiplegia. Brain. 2022;145(10):3374–82.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Greene C, Hanley N, Reschke CR, Reddy A, Mae MA, Connolly R, Behan C, O’Keeffe E, Bolger I, Hudson N, et al. Microvascular stabilization via blood-brain barrier regulation prevents seizure activity. Nat Commun. 2022;13(1):2003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Boye K, Geraldo LH, Furtado J, Pibouin-Fragner L, Poulet M, Kim D, Nelson B, Xu Y, Jacob L, Maissa N, et al. Endothelial Unc5B controls blood-brain barrier integrity. Nat Commun. 2022;13(1):1169.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Zhou H, Gao F, Yang X, Lin T, Li Z, Wang Q, Yao Y, Li L, Ding X, Shi K, et al. Endothelial BACE1 impairs cerebral small vessels via tight Junctions and eNOS. Circul Res. 2022;130(9):1321–41.

    Article  CAS  Google Scholar 

  12. Toth AE, Helms HC, Harazin A, Johnsen KB, Goldeman C, Burkhart A, Thomsen MS, Kempen PJ, Klepe A, Lipka DV, et al. Sortilin regulates blood-brain barrier integrity. FEBS J. 2022;289(4):1062–79.

    Article  CAS  PubMed  Google Scholar 

  13. Martinez-Molledo M, Nji E, Reyes N. Structural insights into the lysophospholipid brain uptake mechanism and its inhibition by syncytin-2. Nat Struct Mol Biol. 2022;29(6):604–12.

    Article  CAS  PubMed  Google Scholar 

  14. Harraz OF, Klug NR, Senatore AJ, Hill-Eubanks DC, Nelson MT. Piezo1 is a Mechanosensor Channel in Central Nervous System Capillaries. Circul Res. 2022;130(10):1531–46.

    Article  CAS  Google Scholar 

  15. Berthiaume AA, Schmid F, Stamenkovic S, Coelho-Santos V, Nielson CD, Weber B, Majesky MW, Shih AY. Pericyte remodeling is deficient in the aged brain and contributes to impaired capillary flow and structure. Nat Commun. 2022;13(1):5912.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ayloo S, Lazo CG, Sun S, Zhang W, Cui B, Gu C. Pericyte-to-endothelial cell signaling via vitronectin-integrin regulates blood-CNS barrier. Neuron. 2022;110(10):1641–1655e1646.

    Article  CAS  PubMed  Google Scholar 

  17. Jamieson JJ, Lin Y, Malloy N, Soto D, Searson PC, Gerecht S. Hypoxia-induced blood-brain barrier dysfunction is prevented by pericyte-conditioned media via attenuated actomyosin contractility and claudin-5 stabilization. FASEB J. 2022;36(5):e22331.

    Article  CAS  PubMed  Google Scholar 

  18. Sancho M, Klug NR, Mughal A, Koide M, Huerta de la Cruz S, Heppner TJ, Bonev AD, Hill-Eubanks D, Nelson MT. Adenosine signaling activates ATP-sensitive K < sup>+ channels in endothelial cells and pericytes in CNS capillaries. Sci Signal [Electronic Resource]. 2022;15(727):eabl5405.

    Article  CAS  PubMed  Google Scholar 

  19. Morales JE, De A, Miller AA, Chen Z, McCarty JH. Mlc1-Expressing perivascular astrocytes promote blood-brain Barrier Integrity. J Neurosci. 2022;42(8):1406–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Mills WA 3rd, Woo AM, Jiang S, Martin J, Surendran D, Bergstresser M, Kimbrough IF, Eyo UB, Sofroniew MV, Sontheimer H. Astrocyte plasticity in mice ensures continued endfoot coverage of cerebral blood vessels following injury and declines with age. Nat Commun. 2022;13(1):1794.

  21. Yshii L, Pasciuto E, Bielefeld P, Mascali L, Lemaitre P, Marino M, Dooley J, Kouser L, Verschoren S, Lagou V, et al. Astrocyte-targeted gene delivery of interleukin 2 specifically increases brain-resident regulatory T cell numbers and protects against pathological neuroinflammation. Nat Immunol. 2022;23(6):878–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Zhu DD, Yang G, Huang YL, Zhang T, Sui AR, Li N, Su WH, Sun HL, Gao JJ, Ntim M, et al. AQP4-A25Q point mutation in mice depolymerizes orthogonal arrays of particles and decreases polarized expression of AQP4 protein in Astrocytic Endfeet at the blood-brain barrier. J Neurosci. 2022;42(43):8169–83.

    Article  CAS  PubMed  Google Scholar 

  23. Salman MM, Kitchen P, Halsey A, Wang MX, Tornroth-Horsefield S, Conner AC, Badaut J, Iliff JJ, Bill RM. Emerging roles for dynamic aquaporin-4 subcellular relocalization in CNS water homeostasis. Brain. 2022;145(1):64–75.

    Article  PubMed  Google Scholar 

  24. Mader S, Brimberg L, Vo A, Strohl JJ, Crawford JM, Bonnin A, Carrion J, Campbell D, Huerta TS, La Bella A, et al. In utero exposure to maternal anti-aquaporin-4 antibodies alters brain vasculature and neural dynamics in male mouse offspring. Sci Transl Med. 2022;14(641):eabe9726.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. De Bock M, De Smet M, Verwaerde S, Tahiri H, Schumacher S, Van Haver V, Witschas K, Steinhauser C, Rouach N, Vandenbroucke RE, et al. Targeting gliovascular connexins prevents inflammatory blood-brain barrier leakage and astrogliosis. Jci Insight. 2022;7(16):22.

    Google Scholar 

  26. Kim H, Leng K, Park J, Sorets AG, Kim S, Shostak A, Embalabala RJ, Mlouk K, Katdare KA, Rose IVL, et al. Reactive astrocytes transduce inflammation in a blood-brain barrier model through a TNF-STAT3 signaling axis and secretion of alpha 1-antichymotrypsin. Nat Commun. 2022;13(1):6581.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Jackson RJ, Meltzer JC, Nguyen H, Commins C, Bennett RE, Hudry E, Hyman BT. APOE4 derived from astrocytes leads to blood-brain barrier impairment. Brain. 2022;145(10):3582–93.

    Article  PubMed  Google Scholar 

  28. Barisano G, Kisler K, Wilkinson B, Nikolakopoulou AM, Sagare AP, Wang Y, Gilliam W, Huuskonen MT, Hung ST, Ichida JK, et al. A “multi-omics” analysis of blood-brain barrier and synaptic dysfunction in APOE4 mice. J Exp Med. 2022;219(11):07.

    Article  Google Scholar 

  29. Liu CC, Zhao J, Fu Y, Inoue Y, Ren Y, Chen Y, Doss SV, Shue F, Jeevaratnam S, Bastea L, et al. Peripheral apoE4 enhances Alzheimer’s pathology and impairs cognition by compromising cerebrovascular function. Nat Neurosci. 2022;25(8):1020–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Gerrits E, Giannini LAA, Brouwer N, Melhem S, Seilhean D, Le Ber I, Brainbank Neuro CEBNN, Kamermans A, Kooij G, de Vries HE, et al. Neurovascular dysfunction in GRN-associated frontotemporal dementia identified by single-nucleus RNA sequencing of human cerebral cortex. Nat Neurosci. 2022;25(8):1034–48.

    Article  CAS  PubMed  Google Scholar 

  31. Li W, Mandeville ET, Duran-Laforet V, Fukuda N, Yu Z, Zheng Y, Held A, Park JH, Nakano T, Tanaka M, et al. Endothelial cells regulate astrocyte to neural progenitor cell trans-differentiation in a mouse model of stroke. Nat Commun. 2022;13(1):7812.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Vazquez-Liebanas E, Nahar K, Bertuzzi G, Keller A, Betsholtz C, Mae MA. Adult-induced genetic ablation distinguishes PDGFB roles in blood-brain barrier maintenance and development. J Cereb Blood Flow Metabolism. 2022;42(2):264–79.

    Article  CAS  Google Scholar 

  33. Lee HW, Xu Y, Zhu X, Jang C, Choi W, Bae H, Wang W, He L, Jin SW, Arany Z, et al. Endothelium-derived lactate is required for pericyte function and blood-brain barrier maintenance. EMBO J. 2022;41(9):e109890.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Rattner A, Wang Y, Nathans J. Signaling pathways in Neurovascular Development. Annu Rev Neurosci. 2022;45:87–108.

    Article  PubMed  Google Scholar 

  35. Xiao G, Kumar R, Komuro Y, Burguet J, Kakarla V, Azizkhanian I, Sheth SA, Williams CK, Zhang XR, Macknicki M, et al. IL-17/CXCL5 signaling within the oligovascular niche mediates human and mouse white matter injury. Cell Rep. 2022;41(12):111848.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Knopp RC, Banks WA, Erickson MA. Physical associations of microglia and the vascular blood-brain barrier and their importance in development, health, and disease. Curr Opin Neurobiol. 2022;77:102648.

    Article  CAS  PubMed  Google Scholar 

  37. Csaszar E, Lenart N, Cserep C, Kornyei Z, Fekete R, Posfai B, Balazsfi D, Hangya B, Schwarcz AD, Szabadits E, et al. Microglia modulate blood flow, neurovascular coupling, and hypoperfusion via purinergic actions. J Exp Med. 2022;219(3):07.

    Article  Google Scholar 

  38. Dorrier CE, Jones HE, Pintaric L, Siegenthaler JA, Daneman R. Emerging roles for CNS fibroblasts in health, injury and disease. Nat Rev Neurosci. 2022;23(1):23–34.

    Article  CAS  PubMed  Google Scholar 

  39. Halder SK, Sapkota A, Milner R. The impact of genetic manipulation of laminin and integrins at the blood-brain barrier. Fluids and barriers of the CNS. 2022;19(1):50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Nirwane A, Yao Y. Cell-specific expression and function of laminin at the neurovascular unit. J Cereb Blood Flow Metabolism. 2022;42(11):1979–99.

    Article  CAS  Google Scholar 

  41. De A, Morales JE, Chen Z, Sebastian S, McCarty JH. The beta8 integrin cytoplasmic domain activates extracellular matrix adhesion to promote brain neurovascular development. Development. 2022;149(6):15.

    Article  Google Scholar 

  42. Zhu J, Li Z, Ji Z, Wu Y, He Y, Liu K, Chang Y, Peng Y, Lin Z, Wang S, et al. Glycocalyx is critical for blood-brain barrier integrity by suppressing caveolin1-dependent endothelial transcytosis following ischemic stroke. Brain Pathol. 2022;32(1):e13006.

    Article  CAS  PubMed  Google Scholar 

  43. Lolansen SD, Rostgaard N, Barbuskaite D, Capion T, Olsen MH, Norager NH, Vilhardt F, Andreassen SN, Toft-Bertelsen TL, Ye F, et al. Posthemorrhagic hydrocephalus associates with elevated inflammation and CSF hypersecretion via activation of choroidal transporters. Fluids and barriers of the CNS. 2022;19(1):62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Toft-Bertelsen TL, Barbuskaite D, Heerfordt EK, Lolansen SD, Andreassen SN, Rostgaard N, Olsen MH, Norager NH, Capion T, Rath MF, et al. Lysophosphatidic acid as a CSF lipid in posthemorrhagic hydrocephalus that drives CSF accumulation via TRPV4-induced hyperactivation of NKCC1. Fluids and barriers of the CNS. 2022;19(1):69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Xu H, Miyajima M, Nakajima M, Ogino I, Kawamura K, Akiba C, Kamohara C, Sakamoto K, Karagiozov K, Nakamura E, et al. Ptpn20 deletion in H-Tx rats enhances phosphorylation of the NKCC1 cotransporter in the choroid plexus: an evidence of genetic risk for hydrocephalus in an experimental study. Fluids and barriers of the CNS. 2022;19(1):39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Zhang Z, Tan Q, Guo P, Huang S, Jia Z, Liu X, Feng H, Chen Y. NLRP3 inflammasome-mediated choroid plexus hypersecretion contributes to hydrocephalus after intraventricular hemorrhage via phosphorylated NKCC1 channels. J Neuroinflamm. 2022;19(1):163.

    Article  Google Scholar 

  47. Hulme L, Hochstetler A, Schwerk C, Schroten H, Ishikawa H, Tung CY, Perrin B, Blazer-Yost B. Characterization of TRPV4-mediated signaling pathways in an optimized human choroid plexus epithelial cell line. Am J Physiol - Cell Physiol. 2022;323(6):C1823–42.

    Article  CAS  PubMed  Google Scholar 

  48. MacAulay N, Keep RF, Zeuthen T. Cerebrospinal fluid production by the choroid plexus: a century of barrier research revisited. Fluids and barriers of the CNS. 2022;19(1):26.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Christensen J, Li C, Mychasiuk R. Choroid plexus function in neurological homeostasis and disorders: the awakening of the circadian clocks and orexins. J Cereb Blood Flow Metabolism. 2022;42(7):1163–75.

    Article  CAS  Google Scholar 

  50. Fernandez Zapata C, Giacomello G, Spruth EJ, Middeldorp J, Gallaccio G, Dehlinger A, Dames C, Leman JKH, van Dijk RE, Meisel A, et al. Differential compartmentalization of myeloid cell phenotypes and responses towards the CNS in Alzheimer’s disease. Nat Commun. 2022;13(1):7210.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zheng W, Feng Y, Zeng Z, Ye M, Wang M, Liu X, Tang P, Shang H, Sun X, Lin X, et al. Choroid plexus-selective inactivation of adenosine A < sub > 2A receptors protects against T cell infiltration and experimental autoimmune encephalomyelitis. J Neuroinflamm. 2022;19(1):52.

    Article  CAS  Google Scholar 

  52. Akeret K, Buzzi RM, Thomson BR, Schwendinger N, Klohs J, Schulthess-Lutz N, Baselgia L, Hansen K, Regli L, Vallelian F, et al. MyD88-TLR4-dependent choroid plexus activation precedes perilesional inflammation and secondary brain edema in a mouse model of intracerebral hemorrhage. J Neuroinflamm. 2022;19(1):290.

    Article  CAS  Google Scholar 

  53. Parichha A, Suresh V, Chatterjee M, Kshirsagar A, Ben-Reuven L, Olender T, Taketo MM, Radosevic V, Bobic-Rasonja M, Trnski S, et al. Constitutive activation of canonical wnt signaling disrupts choroid plexus epithelial fate. Nat Commun. 2022;13(1):633.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Klistorner S, Barnett MH, Parratt J, Yiannikas C, Graham SL, Klistorner A. Choroid plexus volume in multiple sclerosis predicts expansion of chronic lesions and brain atrophy. Annals of Clinical & Translational Neurology. 2022;9(10):1528–37.

    Article  Google Scholar 

  55. Muller J, Sinnecker T, Wendebourg MJ, Schlager R, Kuhle J, Schadelin S, Benkert P, Derfuss T, Cattin P, Jud C, et al. Choroid Plexus volume in multiple sclerosis vs Neuromyelitis Optica Spectrum disorder: a Retrospective, cross-sectional analysis. Neurol Neuroimmunol neuroinflammation. 2022;9(3):05.

    Article  Google Scholar 

  56. Ricigliano VAG, Louapre C, Poirion E, Colombi A, Yazdan Panah A, Lazzarotto A, Morena E, Martin E, Bottlaender M, Bodini B, et al. Imaging characteristics of Choroid Plexuses in Presymptomatic multiple sclerosis: a retrospective study. Neurol Neuroimmunol neuroinflammation. 2022;9(6):11.

    Article  Google Scholar 

  57. Choi JD, Moon Y, Kim HJ, Yim Y, Lee S, Moon WJ. Choroid Plexus volume and permeability at Brain MRI within the Alzheimer Disease Clinical Spectrum. Radiology. 2022;304(3):635–45.

    Article  PubMed  Google Scholar 

  58. Althubaity N, Schubert J, Martins D, Yousaf T, Nettis MA, Mondelli V, Pariante C, Harrison NA, Bullmore ET, Dima D, et al. Choroid plexus enlargement is associated with neuroinflammation and reduction of blood brain barrier permeability in depression. NeuroImage Clin. 2022;33:102926.

    Article  PubMed  Google Scholar 

  59. Alisch JSR, Egan JM, Bouhrara M. Differences in the choroid plexus volume and microstructure are associated with body adiposity. Front Endocrinol. 2022;13:984929.

    Article  Google Scholar 

  60. Lee H, Ozturk B, Stringer MS, Koundal S, MacIntosh BJ, Rothman D, Benveniste H. Choroid plexus tissue perfusion and blood to CSF barrier function in rats measured with continuous arterial spin labeling. NeuroImage. 2022;261:119512.

    Article  PubMed  Google Scholar 

  61. Anderson VC, Tagge IJ, Doud A, Li X, Springer CS Jr, Quinn JF, Kaye JA, Wild KV, Rooney WD. DCE-MRI of Brain Fluid Barriers: in vivo Water Cycling at the human choroid plexus. Tissue Barriers. 2022;10(1):1963143.

    Article  PubMed  Google Scholar 

  62. Jang A, Lehtinen MK. Experimental approaches for manipulating choroid plexus epithelial cells. Fluids and barriers of the CNS. 2022;19(1):36.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Hochstetler A, Hulme L, Delpire E, Schwerk C, Schroten H, Preston D, Simpson S, Blazer-Yost BL. Porcine choroid plexus-riems cell line demonstrates altered polarization of transport proteins compared with the native epithelium. Am J Physiol - Cell Physiol. 2022;323(1):C1–C13.

    Article  CAS  PubMed  Google Scholar 

  64. Faubel RJ, Santos Canellas VS, Gaesser J, Beluk NH, Feinstein TN, Wang Y, Yankova M, Karunakaran KB, King SM, Ganapathiraju MK, et al. Flow blockage disrupts cilia-driven fluid transport in the epileptic brain. Acta Neuropathol. 2022;144(4):691–706.

    Article  CAS  PubMed  Google Scholar 

  65. Iram T, Kern F, Kaur A, Myneni S, Morningstar AR, Shin H, Garcia MA, Yerra L, Palovics R, Yang AC, et al. Young CSF restores oligodendrogenesis and memory in aged mice via Fgf17. Nature. 2022;605(7910):509–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Bridel C, Somers C, Sieben A, Rozemuller A, Niemantsverdriet E, Struyfs H, Vermeiren Y, Van Broeckhoven C, De Deyn PP, Bjerke M, et al. Associating Alzheimer’s disease pathology with its cerebrospinal fluid biomarkers. Brain. 2022;145(11):4056–64.

    Article  PubMed  Google Scholar 

  67. Dumurgier J, Sabia S, Zetterberg H, Teunissen CE, Hanseeuw B, Orellana A, Schraen S, Gabelle A, Boada M, Lebouvier T, et al. A pragmatic, Data-Driven Method to Determine Cutoffs for CSF biomarkers of Alzheimer Disease based on Validation against PET Imaging. Neurology. 2022;99(7):e669–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Jansen IE, van der Lee SJ, Gomez-Fonseca D, de Rojas I, Dalmasso MC, Grenier-Boley B, Zettergren A, Mishra A, Ali M, Andrade V, et al. Genome-wide meta-analysis for Alzheimer’s disease cerebrospinal fluid biomarkers. Acta Neuropathol. 2022;144(5):821–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Brosseron F, Maass A, Kleineidam L, Ravichandran KA, Gonzalez PG, McManus RM, Ising C, Santarelli F, Kolbe CC, Hasler LM, et al. Soluble TAM receptors sAXL and sTyro3 predict structural and functional protection in Alzheimer’s disease. Neuron. 2022;110(6):1009–1022e1004.

    Article  CAS  PubMed  Google Scholar 

  70. Mattsson-Carlgren N, Grinberg LT, Boxer A, Ossenkoppele R, Jonsson M, Seeley W, Ehrenberg A, Spina S, Janelidze S, Rojas-Martinex J, et al. Cerebrospinal fluid biomarkers in autopsy-confirmed Alzheimer Disease and Frontotemporal Lobar Degeneration. Neurology. 2022;98(11):e1137–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Snellman A, Lantero-Rodriguez J, Emersic A, Vrillon A, Karikari TK, Ashton NJ, Gregoric Kramberger M, Cucnik S, Paquet C, Rot U, et al. N-terminal and mid-region tau fragments as fluid biomarkers in neurological diseases. Brain. 2022;145(8):2834–48.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Li TR, Lyu DY, Liu FQ. Alzheimer’s Disease Neuroimaging I: cerebrospinal fluid sTREM2 in Alzheimer’s Disease is Associated with both amyloid and tau pathologies but not with cognitive status. J Alzheimers Dis. 2022;90(3):1123–38.

    Article  CAS  PubMed  Google Scholar 

  73. Morenas-Rodriguez E, Li Y, Nuscher B, Franzmeier N, Xiong C, Suarez-Calvet M, Fagan AM, Schultz S, Gordon BA, Benzinger TLS, et al. Soluble TREM2 in CSF and its association with other biomarkers and cognition in autosomal-dominant Alzheimer’s disease: a longitudinal observational study. Lancet Neurol. 2022;21(4):329–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Zhao A, Jiao Y, Ye G, Kang W, Tan L, Li Y, Deng Y, Liu J. Alzheimer’s Disease Neuroimaging I: Soluble TREM2 levels associate with conversion from mild cognitive impairment to Alzheimer’s disease. J Clin Invest. 2022;132(24):15.

    Article  Google Scholar 

  75. Shahim P, Zetterberg H, Simren J, Ashton NJ, Norato G, Scholl M, Tegner Y, Diaz-Arrastia R, Blennow K. Association of plasma biomarker levels with their CSF concentration and the number and severity of Concussions in Professional athletes. Neurology. 2022;99(4):e347–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Eninger T, Muller SA, Bacioglu M, Schweighauser M, Lambert M, Maia LF, Neher JJ, Hornfeck SM, Obermuller U, Kleinberger G, et al. Signatures of glial activity can be detected in the CSF proteome. Proc Natl Acad Sci USA. 2022;119(24):e2119804119.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Lobanova E, Whiten D, Ruggeri FS, Taylor CG, Kouli A, Xia Z, Emin D, Zhang YP, Lam JYL, Williams-Gray CH, et al. Imaging protein aggregates in the serum and cerebrospinal fluid in Parkinson’s disease. Brain. 2022;145(2):632–43.

    Article  PubMed  Google Scholar 

  78. Lee B, Mahmud I, Pokhrel R, Murad R, Yuan M, Stapleton S, Bettegowda C, Jallo G, Eberhart CG, Garrett T, et al. Medulloblastoma cerebrospinal fluid reveals metabolites and lipids indicative of hypoxia and cancer-specific RNAs. Acta Neuropathol Commun. 2022;10(1):25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Miller AM, Szalontay L, Bouvier N, Hill K, Ahmad H, Rafailov J, Lee AJ, Rodriguez-Sanchez MI, Yildirim O, Patel A, et al. Next-generation sequencing of cerebrospinal fluid for clinical molecular diagnostics in pediatric, adolescent and young adult brain tumor patients. Neurooncology. 2022;24(10):1763–72.

    Google Scholar 

  80. Tivey A, Church M, Rothwell D, Dive C, Cook N. Circulating tumour DNA - looking beyond the blood. Nat Reviews Clin Oncol. 2022;19(9):600–12.

    Article  CAS  Google Scholar 

  81. Piehl N, van Olst L, Ramakrishnan A, Teregulova V, Simonton B, Zhang Z, Tapp E, Channappa D, Oh H, Losada PM, et al. Cerebrospinal fluid immune dysregulation during healthy brain aging and cognitive impairment. Cell. 2022;185(26):5028–5039e5013.

    Article  CAS  PubMed  Google Scholar 

  82. Yazdani S, Seitz C, Cui C, Lovik A, Pan L, Piehl F, Pawitan Y, Klappe U, Press R, Samuelsson K, et al. T cell responses at diagnosis of amyotrophic lateral sclerosis predict disease progression. Nat Commun. 2022;13(1):6733.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Takeuchi H, Suzuki M, Goto R, Tezuka K, Fuchs H, Ishiguro N, Terasaki T, Braun C, Uchida Y. Regional differences in the Absolute abundance of transporters, receptors and tight Junction Molecules at the blood-arachnoid barrier and blood-spinal cord barrier among cervical, thoracic and lumbar spines in Dogs. Pharm Res. 2022;39(7):1393–413.

    Article  CAS  PubMed  Google Scholar 

  84. Sangha V, Hoque MT, Henderson JT, Bendayan R. Novel localization of folate transport systems in the murine central nervous system. Fluids and barriers of the CNS. 2022;19(1):92.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Liu G, Ladron-de-Guevara A, Izhiman Y, Nedergaard M, Du T. Measurements of cerebrospinal fluid production: a review of the limitations and advantages of current methodologies. Fluids Barriers CNS. 2022;19(1):101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Mehta NH, Suss RA, Dyke JP, Theise ND, Chiang GC, Strauss S, Saint-Louis L, Li Y, Pahlajani S, Babaria V, et al. Quantifying cerebrospinal fluid dynamics: a review of human neuroimaging contributions to CSF physiology and neurodegenerative disease. Neurobiol Dis. 2022;170:105776.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Decker Y, Kramer J, Xin L, Muller A, Scheller A, Fassbender K, Proulx ST. Magnetic resonance imaging of cerebrospinal fluid outflow after low-rate lateral ventricle infusion in mice. Jci Insight. 2022;7(3):08.

    Article  Google Scholar 

  88. Albayram MS, Smith G, Tufan F, Tuna IS, Bostanciklioglu M, Zile M, Albayram O. Non-invasive MR imaging of human brain lymphatic networks with connections to cervical lymph nodes. Nat Commun. 2022;13(1):203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Huisman Y, Uphoff K, Berger M, Dobrindt U, Schelhaas M, Zobel T, Bussmann J, van Impel A, Schulte-Merker S. Meningeal lymphatic endothelial cells fulfill scavenger endothelial cell function and cooperate with microglia in waste removal from the brain. GLIA. 2022;70(1):35–49.

    Article  CAS  PubMed  Google Scholar 

  90. Li X, Qi L, Yang D, Hao S, Zhang F, Zhu X, Sun Y, Chen C, Ye J, Yang J, et al. Meningeal lymphatic vessels mediate neurotropic viral drainage from the central nervous system. Nat Neurosci. 2022;25(5):577–87.

    Article  CAS  PubMed  Google Scholar 

  91. Pulous FE, Cruz-Hernandez JC, Yang C, Kaya Z, Paccalet A, Wojtkiewicz G, Capen D, Brown D, Wu JW, Schloss MJ, et al. Cerebrospinal fluid can exit into the skull bone marrow and instruct cranial hematopoiesis in mice with bacterial meningitis. Nat Neurosci. 2022;25(5):567–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Mazzitelli JA, Smyth LCD, Cross KA, Dykstra T, Sun J, Du S, Mamuladze T, Smirnov I, Rustenhoven J, Kipnis J. Cerebrospinal fluid regulates skull bone marrow niches via direct access through dural channels. Nat Neurosci. 2022;25(5):555–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Yang HS, Inglis B, Talavage TM, Nair VV, Yao JF, Fitzgerald B, Schwichtenberg AJ, Tong Y. Coupling between cerebrovascular oscillations and CSF flow fluctuations during wakefulness: an fMRI study. J Cereb Blood Flow Metabolism. 2022;42(6):1091–103.

    Article  Google Scholar 

  94. Rasmussen MK, Mestre H, Nedergaard M. Fluid transport in the brain. Physiol Rev. 2022;102(2):1025–151.

    Article  CAS  PubMed  Google Scholar 

  95. Carlstrom LP, Eltanahy A, Perry A, Rabinstein AA, Elder BD, Morris JM, Meyer FB, Graffeo CS, Lundgaard I, Burns TC. A clinical primer for the glymphatic system. Brain. 2022;145(3):843–57.

    Article  PubMed  Google Scholar 

  96. Lohela TJ, Lilius TO, Nedergaard M. The glymphatic system: implications for drugs for central nervous system diseases. Nat Rev Drug Discovery. 2022;21(10):763–79.

    Article  CAS  PubMed  Google Scholar 

  97. Hladky SB, Barrand MA. The glymphatic hypothesis: the theory and the evidence. Fluids and barriers of the CNS. 2022;19(1):9.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Zhao L, Tannenbaum A, Bakker E, Benveniste H. Physiology of Glymphatic Solute Transport and Waste Clearance from the brain. Physiology. 2022;37(6):0.

    Article  PubMed  Google Scholar 

  99. Carotenuto A, Cacciaguerra L, Pagani E, Preziosa P, Filippi M, Rocca MA. Glymphatic system impairment in multiple sclerosis: relation with brain damage and disability. Brain. 2022;145(8):2785–95.

    Article  PubMed  Google Scholar 

  100. Benveniste H, Nedergaard M. Cerebral small vessel disease: a glymphopathy? Curr Opin Neurobiol. 2022;72:15–21.

    Article  CAS  PubMed  Google Scholar 

  101. Eide PK, Pripp AH, Berge B, Hrubos-Strom H, Ringstad G, Valnes LM. Altered glymphatic enhancement of cerebrospinal fluid tracer in individuals with chronic poor sleep quality. J Cereb Blood Flow Metabolism. 2022;42(9):1676–92.

    Article  CAS  Google Scholar 

  102. Xiang T, Feng D, Zhang X, Chen Y, Wang H, Liu X, Gong Z, Yuan J, Liu M, Sha Z, et al. Effects of increased intracranial pressure on cerebrospinal fluid influx, cerebral vascular hemodynamic indexes, and cerebrospinal fluid lymphatic efflux. J Cereb Blood Flow Metabolism. 2022;42(12):2287–302.

    Article  Google Scholar 

  103. Du T, Mestre H, Kress BT, Liu G, Sweeney AM, Samson AJ, Rasmussen MK, Mortensen KN, Bork PAR, Peng W, et al. Cerebrospinal fluid is a significant fluid source for anoxic cerebral oedema. Brain. 2022;145(2):787–97.

    Article  PubMed  Google Scholar 

  104. Drieu A, Du S, Storck SE, Rustenhoven J, Papadopoulos Z, Dykstra T, Zhong F, Kim K, Blackburn S, Mamuladze T, et al. Parenchymal border macrophages regulate the flow dynamics of the cerebrospinal fluid. Nature. 2022;611(7936):585–93.

    Article  CAS  PubMed  Google Scholar 

  105. Mestre H, Verma N, Greene TD, Lin LA, Ladron-de-Guevara A, Sweeney AM, Liu G, Thomas VK, Galloway CA, de Mesy Bentley KL, et al. Periarteriolar spaces modulate cerebrospinal fluid transport into brain and demonstrate altered morphology in aging and Alzheimer’s disease. Nat Commun. 2022;13(1):3897.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Linville RM, Nerenberg RF, Grifno G, Arevalo D, Guo Z, Searson PC. Brain microvascular endothelial cell dysfunction in an isogenic juvenile iPSC model of Huntington’s disease. Fluids and barriers of the CNS. 2022;19(1):54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Sundaram SM, Arrulo Pereira A, Muller-Fielitz H, Kopke H, De Angelis M, Muller TD, Heuer H, Korbelin J, Krohn M, Mittag J, et al. Gene therapy targeting the blood-brain barrier improves neurological symptoms in a model of genetic MCT8 deficiency. Brain. 2022;145(12):4264–74.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Goncalves A, Antonetti DA. Transgenic animal models to explore and modulate the blood brain and blood retinal barriers of the CNS. Fluids and barriers of the CNS. 2022;19(1):86.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Wang L, Mao B, Fan K, Sun R, Zhang J, Liang H, Liu Y. ROS attenuates TET2-dependent ZO-1 epigenetic expression in cerebral vascular endothelial cells. Fluids and barriers of the CNS. 2022;19(1):73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Kong FC, Lang LQ, Hu J, Zhang XL, Zhong MK, Ma CL. A novel epigenetic marker, ten-eleven translocation family member 2 (TET2), is identified in the intractable epileptic brain and regulates ATP binding cassette subfamily B member 1 (ABCB1) in the blood-brain barrier. Bioengineered. 2022;13(3):6638–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Huls A, Robins C, Conneely KN, Edgar R, De Jager PL, Bennett DA, Wingo AP, Epstein MP, Wingo TS. Brain DNA methylation patterns in CLDN5 Associated with Cognitive decline. Biol Psychiatry. 2022;91(4):389–98.

    Article  CAS  PubMed  Google Scholar 

  112. Sun P, Hamblin MH, Yin KJ. Non-coding RNAs in the regulation of blood-brain barrier functions in central nervous system disorders. Fluids and barriers of the CNS. 2022;19(1):27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Marra KV, Aguilar E, Guoqin W, Usui-Ouchi A, Ideguchi Y, Sakimoto S, Friedlander M. Bioactive extracellular vesicles from a subset of endothelial progenitor cells rescue retinal ischemia and neurodegeneration. Jci Insight. 2022;7(12):22.

    Article  Google Scholar 

  114. Monje M, Iwasaki A. The neurobiology of long COVID. Neuron. 2022;110(21):3484–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Agrawal S, Farfel JM, Arfanakis K, Al-Harthi L, Shull T, Teppen TL, Evia AM, Patel MB, Ely EW, Leurgans SE, et al. Brain autopsies of critically ill COVID-19 patients demonstrate heterogeneous profile of acute vascular injury, inflammation and age-linked chronic brain diseases. Acta Neuropathol Commun. 2022;10(1):186.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Etter MM, Martins TA, Kulsvehagen L, Possnecker E, Duchemin W, Hogan S, Sanabria-Diaz G, Muller J, Chiappini A, Rychen J, et al. Severe Neuro-COVID is associated with peripheral immune signatures, autoimmunity and neurodegeneration: a prospective cross-sectional study. Nat Commun. 2022;13(1):6777.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Lee MH, Perl DP, Steiner J, Pasternack N, Li W, Maric D, Safavi F, Horkayne-Szakaly I, Jones R, Stram MN, et al. Neurovascular injury with complement activation and inflammation in COVID-19. Brain. 2022;145(7):2555–68.

    Article  PubMed  Google Scholar 

  118. Jarius S, Pache F, Kortvelyessy P, Jelcic I, Stettner M, Franciotta D, Keller E, Neumann B, Ringelstein M, Senel M, et al. Cerebrospinal fluid findings in COVID-19: a multicenter study of 150 lumbar punctures in 127 patients. J Neuroinflamm. 2022;19(1):19.

    Article  CAS  Google Scholar 

  119. Rutkai I, Mayer MG, Hellmers LM, Ning B, Huang Z, Monjure CJ, Coyne C, Silvestri R, Golden N, Hensley K, et al. Neuropathology and virus in brain of SARS-CoV-2 infected non-human primates. Nat Commun. 2022;13(1):1745.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Altmayer V, Ziveri J, Frere C, Salem JE, Weiss N, Cao A, Marois C, Rohaut B, Demeret S, Bourdoulous S, et al. Endothelial cell biomarkers in critically ill COVID-19 patients with encephalitis. J Neurochem. 2022;161(6):492–505.

    Article  CAS  PubMed  Google Scholar 

  121. Krasemann S, Haferkamp U, Pfefferle S, Woo MS, Heinrich F, Schweizer M, Appelt-Menzel A, Cubukova A, Barenberg J, Leu J, et al. The blood-brain barrier is dysregulated in COVID-19 and serves as a CNS entry route for SARS-CoV-2. Stem Cell Reports. 2022;17(2):307–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Aukrust CG, Paulsen AH, Uche EO, Kamalo PD, Sandven I, Fjeld HE, Stromme H, Eide PK. Aetiology and diagnostics of paediatric hydrocephalus across Africa: a systematic review and meta-analysis. The Lancet Global Health. 2022;10(12):e1793–806.

    Article  CAS  PubMed  Google Scholar 

  123. Isaacs AM, Riva-Cambrin J, Yavin D, Hockley A, Pringsheim TM, Jette N, Lethebe BC, Lowerison M, Dronyk J, Hamilton MG. Age-specific global epidemiology of hydrocephalus: systematic review, metanalysis and global birth surveillance. PLoS ONE. 2018;13(10):e0204926.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Han X, Shen X, Wang M, Wang X, Jian Y, Yang C, Guo W. Loss of RNA-Binding protein HuR leads to defective Ependymal cells and Hydrocephalus. J Neurosci. 2022;42(2):202–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Harkins D, Harvey TJ, Atterton C, Miller I, Currey L, Oishi S, Kasherman M, Davila RA, Harris L, Green K, et al. Hydrocephalus in Nfix < sup>-/- mice is underpinned by changes in Ependymal Cell Physiology. Cells. 2022;11(15):02.

    Article  Google Scholar 

  126. Iwasawa E, Brown FN, Shula C, Kahn F, Lee SH, Berta T, Ladle DR, Campbell K, Mangano FT, Goto J. The anti-inflammatory Agent Bindarit attenuates the impairment of neural development through suppression of Microglial activation in a neonatal Hydrocephalus Mouse Model. J Neurosci. 2022;42(9):1820–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Duy PQ, Greenberg ABW, Butler WE, Kahle KT. Rethinking the cilia hypothesis of hydrocephalus. Neurobiol Dis. 2022;175:105913.

    Article  CAS  PubMed  Google Scholar 

  128. Duy PQ, Weise SC, Marini C, Li XJ, Liang D, Dahl PJ, Ma S, Spajic A, Dong W, Juusola J, et al. Impaired neurogenesis alters brain biomechanics in a neuroprogenitor-based genetic subtype of congenital hydrocephalus. Nat Neurosci. 2022;25(4):458–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Shah V, Musrap N, Maharaj K, Afifi J, El-Naggar W, Kelly E, Mukerji A, Shah P, Vincer M. Grading of Intraventricular Hemorrhage and Neurodevelopment in Preterm < 29 Weeks’ GA in Canada.Children (Basel)2022, 9(12).

  130. Holste KG, Xia F, Ye F, Keep RF, Xi G. Mechanisms of neuroinflammation in hydrocephalus after intraventricular hemorrhage: a review. Fluids and barriers of the CNS. 2022;19(1):28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Vinukonda G, La Gamma EF. Emerging therapies for brain recovery after IVH in neonates: cord blood derived mesenchymal stem cells (MSC) and unrestricted somatic stem cells (USSC). Semin Perinatol. 2022;46(5):151598.

    Article  PubMed  Google Scholar 

  132. Hanley DF, Lane K, McBee N, Ziai W, Tuhrim S, Lees KR, Dawson J, Gandhi D, Ullman N, Mould WA, et al. Thrombolytic removal of intraventricular haemorrhage in treatment of severe stroke: results of the randomised, multicentre, multiregion, placebo-controlled CLEAR III trial. Lancet. 2017;389(10069):603–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Kuramatsu JB, Gerner ST, Ziai W, Bardutzky J, Sembill JA, Sprugel MI, Mrochen A, Kolbl K, Ram M, Avadhani R, et al. Association of Intraventricular Fibrinolysis with Clinical Outcomes in Intracerebral Hemorrhage: an individual Participant Data Meta-Analysis. Stroke. 2022;53(9):2876–86.

    Article  CAS  PubMed  Google Scholar 

  134. Thavarajasingam SG, El-Khatib M, Vemulapalli KV, Iradukunda HAS, Laleye J, Russo S, Eichhorn C, Eide PK. Cerebrospinal fluid and venous biomarkers of shunt-responsive idiopathic normal pressure hydrocephalus: a systematic review and meta-analysis. Acta Neurochir (Wien). 2022;164(7):1719–46.

    Article  PubMed  Google Scholar 

  135. Lukkarinen H, Jeppsson A, Wikkelso C, Blennow K, Zetterberg H, Constantinescu R, Remes AM, Herukka SK, Hiltunen M, Rauramaa T, et al. Cerebrospinal fluid biomarkers that reflect clinical symptoms in idiopathic normal pressure hydrocephalus patients. Fluids and barriers of the CNS. 2022;19(1):11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Gold D, Wisialowski C, Piryatinsky I, Malloy P, Correia S, Salloway S, Klinge P, Gaudet CE, Niermeyer M, Lee A. Longitudinal post-shunt outcomes in idiopathic normal pressure hydrocephalus with and without comorbid Alzheimer’s disease.J Int Neuropsychol Soc2022:1–12.

  137. Lv M, Yang X, Zhou X, Chen J, Wei H, Du D, Lin H, Xia J. Gray matter volume of cerebellum associated with idiopathic normal pressure hydrocephalus: a cross-sectional analysis. Front Neurol. 2022;13:922199.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Gholampour S, Frim D, Yamini B. Long-term recovery behavior of brain tissue in hydrocephalus patients after shunting. Commun Biology. 2022;5(1):1198.

    Article  Google Scholar 

  139. Gholampour S, Yamini B, Droessler J, Frim D. A New Definition for Intracranial Compliance to Evaluate Adult Hydrocephalus after shunting. Front Bioeng Biotechnol. 2022;10:900644.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Aoki Y, Kazui H, Pascual-Marqui RD, Bruña R, Yoshiyama K, Wada T, Kanemoto H, Suzuki Y, Suehiro T, Satake Y et al. Normalized Power Variance: A new Field Orthogonal to Power in EEG Analysis.Clin EEG Neurosci2022:15500594221088736.

  141. Verghese JP, Terry A, de Natale ER, Politis M. Research Evidence of the Role of the Glymphatic System and Its Potential Pharmacological Modulation in Neurodegenerative Diseases.J Clin Med2022, 11(23).

  142. De Bernardo M, Vitiello L, De Pascale I, Capasso L, Cornetta P, Rosa N. Optic nerve ultrasound evaluation in idiopathic intracranial hypertension. Front Med (Lausanne). 2022;9:845554.

    Article  PubMed  Google Scholar 

  143. Cogswell PM, Murphy MC, Madhavan AA, Bhatti MT, Cutsforth-Gregory JK, Senjem ML, Huston J 3rd, Chen JJ. Features of idiopathic intracranial hypertension on MRI with MR Elastography: prospective comparison with control individuals and Assessment of Postintervention Changes. AJR Am J Roentgenol. 2022;219(6):940–51.

  144. Grech O, Seneviratne SY, Alimajstorovic Z, Yiangou A, Mitchell JL, Smith TB, Mollan SP, Lavery GG, Ludwig C, Sinclair AJ. Nuclear magnetic resonance spectroscopy metabolomics in idiopathic intracranial hypertension to identify markers of Disease and Headache. Neurology. 2022;99(16):e1702–1714.

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Westgate CSJ, Markey K, Mitchell JL, Yiangou A, Singhal R, Stewart P, Tomlinson JW, Lavery GG, Mollan SP, Sinclair AJ. Increased systemic and adipose 11β-HSD1 activity in idiopathic intracranial hypertension. Eur J Endocrinol. 2022;187(2):323–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Mraja SN, Akdogan O, Mraja HM, Emre U, Karagöz Y. Evaluation of magnetic resonance findings of Circumventricular Organs in idiopathic intracranial hypertension patients. Cureus. 2022;14(11):e31795.

    PubMed  PubMed Central  Google Scholar 

  147. Salih M, Enriquez-Marulanda A, Khorasanizadeh M, Moore J, Prabhu VC, Ogilvy CS. Cerebrospinal fluid shunting for idiopathic intracranial hypertension: a systematic review, Meta-analysis, and implications for a Modern Management Protocol. Neurosurgery. 2022;91(4):529–40.

    Article  PubMed  Google Scholar 

  148. Mullen M, Scofield-Kaplan SM, Ford WC, Mancini R. The Effect of Optic nerve sheath fenestration on intraocular pressure in patients with idiopathic intracranial hypertension. J Neuroophthalmol. 2022;42(1):97–100.

    Article  PubMed  Google Scholar 

  149. Kabanovski A, Kisilevsky E, Yang Y, Margolin E. Dural venous sinus stenting in the treatment of idiopathic intracranial hypertension: a systematic review and critique of literature. Surv Ophthalmol. 2022;67(1):271–87.

    Article  PubMed  Google Scholar 

  150. Westgate CSJ, Hagen SM, Israelsen IME, Hamann S, Jensen RH, Eftekhari S. The impact of obesity-related raised intracranial pressure in rodents. Sci Rep. 2022;12(1):9102.

    Article  PubMed  PubMed Central  Google Scholar 

  151. Candelario-Jalil E, Dijkhuizen RM, Magnus T. Neuroinflammation, Stroke, blood-brain barrier dysfunction, and Imaging Modalities. Stroke. 2022;53(5):1473–86.

    Article  CAS  PubMed  Google Scholar 

  152. Arias E, Nadkarni N, Fang R, Haynes M, Batra A, Muller W, Sullivan D. Inhibition of PECAM-1 significantly delays leukocyte extravasation into the Subcortex Post-Stroke. FASEB J. 2022;36(1):05.

    Google Scholar 

  153. Ng FC, Churilov L, Yassi N, Kleinig TJ, Thijs V, Wu TY, Shah DG, Dewey HM, Sharma G, Desmond PM, et al. Microvascular dysfunction in blood-brain barrier disruption and hypoperfusion within the Infarct Posttreatment are Associated with cerebral edema. Stroke. 2022;53(5):1597–605.

    Article  CAS  PubMed  Google Scholar 

  154. Hansen L, Lohfink N, Vutukuri R, Kestner RI, Trautmann S, Hecht M, Wagner PV, Spitzer D, Khel MI, Macas J, et al. Endothelial sphingosine-1-Phosphate receptor 4 regulates blood-brain barrier permeability and promotes a homeostatic endothelial phenotype. J Neurosci. 2022;42(10):1908–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Spitzer D, Guerit S, Puetz T, Khel MI, Armbrust M, Dunst M, Macas J, Zinke J, Devraj G, Jia X, et al. Profiling the neurovascular unit unveils detrimental effects of osteopontin on the blood-brain barrier in acute ischemic stroke. Acta Neuropathol. 2022;144(2):305–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Wardlaw JM, Benveniste H, Williams A. Cerebral vascular dysfunctions detected in human small Vessel Disease and Implications for Preclinical Studies. Annu Rev Physiol. 2022;84:409–34.

    Article  PubMed  Google Scholar 

  157. Mishra A, Duplaa C, Vojinovic D, Suzuki H, Sargurupremraj M, Zilhao NR, Li S, Bartz TM, Jian X, Zhao W, et al. Gene-mapping study of extremes of cerebral small vessel disease reveals TRIM47 as a strong candidate. Brain. 2022;145(6):1992–2007.

    Article  PubMed  PubMed Central  Google Scholar 

  158. Quick S, Procter TV, Moss J, Seeker L, Walton M, Lawson A, Baker S, Beletski A, Garcia DJ, Mohammad M, et al. Loss of the heterogeneous expression of flippase ATP11B leads to cerebral small vessel disease in a normotensive rat model. Acta Neuropathol. 2022;144(2):283–303.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Ji YB, Gao Q, Tan XX, Huang XW, Ma YZ, Fang C, Wang SN, Qiu LH, Cheng YX, Guo FY, et al. Lithium alleviates blood-brain barrier breakdown after cerebral ischemia and reperfusion by upregulating endothelial Wnt/beta-catenin signaling in mice. Neuropharmacology. 2021;186:108474.

    Article  CAS  PubMed  Google Scholar 

  160. Martin M, Vermeiren S, Bostaille N, Eubelen M, Spitzer D, Vermeersch M, Profaci CP, Pozuelo E, Toussay X, Raman-Nair J, et al. Engineered wnt ligands enable blood-brain barrier repair in neurological disorders. Science. 2022;375(6582):eabm4459.

    Article  CAS  PubMed  Google Scholar 

  161. Solar P, Zamani A, Lakatosova K, Joukal M. The blood-brain barrier and the neurovascular unit in subarachnoid hemorrhage: molecular events and potential treatments. Fluids and barriers of the CNS. 2022;19(1):29.

    Article  PubMed  PubMed Central  Google Scholar 

  162. Phillips CM, Stamatovic SM, Keep RF, Andjelkovic AV. Cerebral cavernous malformation pathogenesis: investigating lesion formation and progression with animal models. Int J Mol Sci. 2022;23(9):30.

    Article  Google Scholar 

  163. Yang X, Dai Z, Gao C, Yin Y, Shi C, Liu R, Zhuge Q, Huang Y, Zhou B, Han Z, et al. Cerebral cavernous malformation development in chronic mouse models driven by dual recombinases induced gene deletion in brain endothelial cells. J Cereb Blood Flow Metabolism. 2022;42(12):2230–44.

    Article  Google Scholar 

  164. McCurdy S, Lin J, Shenkar R, Moore T, Lightle R, Faurobert E, Lopez-Ramirez MA, Awad I. Ginsberg MH: beta1 integrin monoclonal antibody treatment ameliorates cerebral cavernous malformations. FASEB J. 2022;36(12):e22629.

    Article  PubMed  Google Scholar 

  165. Lai CC, Nelsen B, Frias-Anaya E, Gallego-Gutierrez H, Orecchioni M, Herrera V, Ortiz E, Sun H, Mesarwi OA, Ley K, et al. Neuroinflammation plays a critical role in cerebral cavernous malformation disease. Circul Res. 2022;131(11):909–25.

    Article  CAS  Google Scholar 

  166. Fang Z, Sun X, Wang X, Ma J, Palaia T, Rana U, Miao B, Ragolia L, Hu W, Miao QR. NOGOB receptor deficiency increases cerebrovascular permeability and hemorrhage via impairing histone acetylation-mediated CCM1/2 expression. J Clin Invest. 2022;132(9):02.

    Article  Google Scholar 

  167. Montagne A, Barnes SR, Nation DA, Kisler K, Toga AW, Zlokovic BV. Imaging subtle leaks in the blood-brain barrier in the aging human brain: potential pitfalls, challenges, and possible solutions. GeroScience. 2022;44(3):1339–51.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Kurz C, Walker L, Rauchmann BS, Perneczky R. Dysfunction of the blood-brain barrier in Alzheimer’s disease: evidence from human studies. Neuropathology & Applied Neurobiology. 2022;48(3):e12782.

    Article  CAS  Google Scholar 

  169. Wang Q, Huang X, Su Y, Yin G, Wang S, Yu B, Li H, Qi J, Chen H, Zeng W, et al. Activation of Wnt/beta-catenin pathway mitigates blood-brain barrier dysfunction in Alzheimer’s disease. Brain. 2022;145(12):4474–88.

    Article  PubMed  PubMed Central  Google Scholar 

  170. Deng W, Guo S, van Veluw SJ, Yu Z, Chan SJ, Takase H, Arai K, Ning M, Greenberg SM, Lo EH, et al. Effects of cerebral amyloid angiopathy on the brain vasculome. Aging Cell. 2022;21(8):e13503.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Situ M, Citalan-Madrid AF, Stamatovic SM, Keep RF, Andjelkovic AV. Transcriptomic Profile of blood-brain barrier remodeling in cerebral amyloid Angiopathy. Front Cell Neurosci. 2022;16:931247.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Tachida Y, Miura S, Muto Y, Takuwa H, Sahara N, Shindo A, Matsuba Y, Saito T, Taniguchi N, Kawaguchi Y, et al. Endothelial expression of human amyloid precursor protein leads to amyloid beta in the blood and induces cerebral amyloid angiopathy in knock-in mice. J Biol Chem. 2022;298(6):101880.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Proulx ST, Engelhardt B. Central nervous system zoning: how brain barriers establish subdivisions for CNS immune privilege and immune surveillance. J Intern Med. 2022;292(1):47–67.

    Article  PubMed  PubMed Central  Google Scholar 

  174. Frieser D, Pignata A, Khajavi L, Shlesinger D, Gonzalez-Fierro C, Nguyen XH, Yermanos A, Merkler D, Hoftberger R, Desestret V, et al. Tissue-resident CD8 < sup>+ T cells drive compartmentalized and chronic autoimmune damage against CNS neurons. Sci Transl Med. 2022;14(640):eabl6157.

    Article  CAS  PubMed  Google Scholar 

  175. Vincenti I, Page N, Steinbach K, Yermanos A, Lemeille S, Nunez N, Kreutzfeldt M, Klimek B, Di Liberto G, Egervari K, et al. Tissue-resident memory CD8 < sup>+ T cells cooperate with CD4 < sup>+ T cells to drive compartmentalized immunopathology in the CNS. Sci Transl Med. 2022;14(640):eabl6058.

    Article  CAS  PubMed  Google Scholar 

  176. Charabati M, Grasmuck C, Ghannam S, Bourbonniere L, Fournier AP, Lecuyer MA, Tastet O, Kebir H, Rebillard RM, Hoornaert C, et al. DICAM promotes T < sub > H 17 lymphocyte trafficking across the blood-brain barrier during autoimmune neuroinflammation. Sci Transl Med. 2022;14(626):eabj0473.

    Article  CAS  PubMed  Google Scholar 

  177. Lanz TV, Brewer RC, Ho PP, Moon JS, Jude KM, Fernandez D, Fernandes RA, Gomez AM, Nadj GS, Bartley CM, et al. Clonally expanded B cells in multiple sclerosis bind EBV EBNA1 and GlialCAM. Nature. 2022;603(7900):321–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Ntranos A, Park HJ, Wentling M, Tolstikov V, Amatruda M, Inbar B, Kim-Schulze S, Frazier C, Button J, Kiebish MA, et al. Bacterial neurotoxic metabolites in multiple sclerosis cerebrospinal fluid and plasma. Brain. 2022;145(2):569–83.

    Article  PubMed  Google Scholar 

  179. Hosang L, Canals RC, van der Flier FJ, Hollensteiner J, Daniel R, Flugel A, Odoardi F. The lung microbiome regulates brain autoimmunity. Nature. 2022;603(7899):138–44.

    Article  CAS  PubMed  Google Scholar 

  180. Hermans D, Houben E, Baeten P, Slaets H, Janssens K, Hoeks C, Hosseinkhani B, Duran G, Bormans S, Gowing E, et al. Oncostatin M triggers brain inflammation by compromising blood-brain barrier integrity. Acta Neuropathol. 2022;144(2):259–81.

    Article  CAS  PubMed  Google Scholar 

  181. Nishihara H, Perriot S, Gastfriend BD, Steinfort M, Cibien C, Soldati S, Matsuo K, Guimbal S, Mathias A, Palecek SP, et al. Intrinsic blood-brain barrier dysfunction contributes to multiple sclerosis pathogenesis. Brain. 2022;145(12):4334–48.

    Article  PubMed  Google Scholar 

  182. Medina-Rodriguez EM, Beurel E. Blood brain barrier and inflammation in depression. Neurobiol Dis. 2022;175:105926.

    Article  PubMed  Google Scholar 

  183. Dion-Albert L, Cadoret A, Doney E, Kaufmann FN, Dudek KA, Daigle B, Parise LF, Cathomas F, Samba N, Hudson N, et al. Vascular and blood-brain barrier-related changes underlie stress responses and resilience in female mice and depression in human tissue. Nat Commun. 2022;13(1):164.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Matsuno H, Tsuchimine S, O’Hashi K, Sakai K, Hattori K, Hidese S, Nakajima S, Chiba S, Yoshimura A, Fukuzato N, et al. Association between vascular endothelial growth factor-mediated blood-brain barrier dysfunction and stress-induced depression. Mol Psychiatry. 2022;27(9):3822–32.

    Article  CAS  PubMed  Google Scholar 

  185. Dai W, Yang M, Xia P, Xiao C, Huang S, Zhang Z, Cheng X, Li W, Jin J, Zhang J, et al. A functional role of meningeal lymphatics in sex difference of stress susceptibility in mice. Nat Commun. 2022;13(1):4825.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Gorick CM, Breza VR, Nowak KM, Cheng VWT, Fisher DG, Debski AC, Hoch MR, Demir ZEF, Tran NM, Schwartz MR, et al. Applications of focused ultrasound-mediated blood-brain barrier opening. Adv Drug Deliv Rev. 2022;191:114583.

    Article  CAS  PubMed  Google Scholar 

  187. Epelbaum S, Burgos N, Canney M, Matthews D, Houot M, Santin MD, Desseaux C, Bouchoux G, Stroer S, Martin C, et al. Pilot study of repeated blood-brain barrier disruption in patients with mild Alzheimer’s disease with an implantable ultrasound device. Alzheimers Res Ther. 2022;14(1):40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Kung Y, Chen KY, Liao WH, Hsu YH, Wu CH, Hsiao MY, Huang AP, Chen WS. Facilitating drug delivery in the central nervous system by opening the blood-cerebrospinal fluid barrier with a single low energy shockwave pulse. Fluids and barriers of the CNS. 2022;19(1):3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Wakayama E, Kuzu T, Tachibana K, Hirayama R, Okada Y, Kondoh M. Modifying the blood-brain barrier by targeting claudin-5: safety and risks. Ann N Y Acad Sci. 2022;1514(1):62–9.

    Article  CAS  PubMed  Google Scholar 

  190. Ramos-Zaldivar HM, Polakovicova I, Salas-Huenuleo E, Corvalan AH, Kogan MJ, Yefi CP, Andia ME. Extracellular vesicles through the blood-brain barrier: a review. Fluids and barriers of the CNS. 2022;19(1):60.

    Article  PubMed  PubMed Central  Google Scholar 

  191. Pauwels MJ, Xie J, Ceroi A, Balusu S, Castelein J, Van Wonterghem E, Van Imschoot G, Ward A, Menheniott TR, Gustafsson O, et al. Choroid plexus-derived extracellular vesicles exhibit brain targeting characteristics. Biomaterials. 2022;290:121830.

    Article  CAS  PubMed  Google Scholar 

  192. Rathi S, Griffith JI, Zhang W, Zhang W, Oh JH, Talele S, Sarkaria JN, Elmquist WF. The influence of the blood-brain barrier in the treatment of brain tumours. J Intern Med. 2022;292(1):3–30.

    Article  PubMed  Google Scholar 

  193. Lu G, Wang X, Li F, Wang S, Zhao J, Wang J, Liu J, Lyu C, Ye P, Tan H, et al. Engineered biomimetic nanoparticles achieve targeted delivery and efficient metabolism-based synergistic therapy against glioblastoma. Nat Commun. 2022;13(1):4214.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Zhang D, Tian S, Liu Y, Zheng M, Yang X, Zou Y, Shi B, Luo L. Near infrared-activatable biomimetic nanogels enabling deep tumor drug penetration inhibit orthotopic glioblastoma. Nat Commun. 2022;13(1):6835.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Goertsen D, Flytzanis NC, Goeden N, Chuapoco MR, Cummins A, Chen Y, Fan Y, Zhang Q, Sharma J, Duan Y, et al. AAV capsid variants with brain-wide transgene expression and decreased liver targeting after intravenous delivery in mouse and marmoset. Nat Neurosci. 2022;25(1):106–15.

    Article  CAS  PubMed  Google Scholar 

  196. Flotte TR, Cataltepe O, Puri A, Batista AR, Moser R, McKenna-Yasek D, Douthwright C, Gernoux G, Blackwood M, Mueller C, et al. AAV gene therapy for Tay-Sachs disease. Nat Med. 2022;28(2):251–9.

    Article  CAS  PubMed  Google Scholar 

  197. Duan Y, Ye T, Qu Z, Chen Y, Miranda A, Zhou X, Lok KC, Chen Y, Fu AKY, Gradinaru V, et al. Brain-wide Cas9-mediated cleavage of a gene causing familial Alzheimer’s disease alleviates amyloid-related pathologies in mice. Nat Biomedical Eng. 2022;6(2):168–80.

    Article  CAS  Google Scholar 

  198. Grashoff H, Muller-Fielitz H, Dogbevia GK, Korbelin J, Bannach J, Vahldieck CM, Kusche-Vihrog K, Johren O, Muller OJ, Nogueiras R, et al. Short regulatory DNA sequences to target brain endothelial cells for gene therapy. J Cereb Blood Flow Metabolism. 2022;42(1):104–20.

    Article  Google Scholar 

  199. Jang A, Petrova B, Cheong TC, Zawadzki ME, Jones JK, Culhane AJ, Shipley FB, Chiarle R, Wong ET, Kanarek N, et al. Choroid plexus-CSF-targeted antioxidant therapy protects the brain from toxicity of cancer chemotherapy. Neuron. 2022;110(20):3288–3301e3288.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

None.

Funding

None.

Author information

Authors and Affiliations

Authors

Contributions

RFK wrote the initial draft. HCJ, MGH and LRD modified that draft. All authors have read and approved the final manuscript.

Corresponding author

Correspondence to Richard F. Keep.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

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

Additional information

Publisher’s Note

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

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. 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 in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Keep, R.F., Jones, H.C., Hamilton, M.G. et al. A year in review: brain barriers and brain fluids research in 2022. Fluids Barriers CNS 20, 30 (2023). https://doi.org/10.1186/s12987-023-00429-0

Download citation

  • Received:

  • Published:

  • DOI: https://doi.org/10.1186/s12987-023-00429-0

Keywords