Skip to main content
Aging and Disease logoLink to Aging and Disease
. 2024 Apr 1;15(2):584–600. doi: 10.14336/AD.2023.0426

Contribution of Direct Cerebral Vascular Transport in Brain Substance Clearance

Qiuting Wen 1,#, Haoyu Wang 2,#, E Mark Haacke 3, Quan Jiang 4,*, Jiani Hu 3,*
PMCID: PMC10917538  PMID: 37611901

Abstract

The accumulation of harmful substances has long been recognized as a likely cause of many neurodegenerative diseases. The two classic brain clearance pathways are cerebrospinal fluid (CSF) and vascular circulation systems. Since the discovery of the glymphatic system, research on the CSF pathway has gained momentum, and impaired CSF clearance has been implicated in virtually all neurodegenerative animal models. However, the contribution of the direct participation of vascular transport across the blood-brain barrier in clearing substances is often ignored in glymphatic papers. Supportive evidence for the direct involvement of parenchymal vasculature in substance clearance is accumulated. First, multiple mechanisms have been proposed for the vascular drainage of exogenous and endogenous substances across the blood-brain barriers. Second, the "traditional" role of arachnoid villi and granulations as the main site for CSF draining into the vasculature system has been questioned. Third, MRI studies using different CSF tracers indicate that parenchymal vasculature directly participates in tracer efflux, consistent with immunohistochemical findings. Here we will review evidence in the literature that supports the direct participation of the parenchymal vascular system in substance clearance, in addition to the CSF clearance pathways.

Keywords: substance clearance, vascular circulation, CSF, glymphatic system

1. Introduction

Harmful substance removal from the brain is essential for maintaining brain homeostasis across the lifespan [1]. A critical function of sleep is to remove harmful substances more effectively [2]. The accumulation of harmful substances, including heavy metals and waste proteins, has long been hypothesized to cause many neuro-degenerative diseases. For instance, Alzheimer's disease (AD) is marked by the accumulation of protein aggregates in the brain parenchyma, including amyloid-β (Aβ) plaques and hyperphosphorylated tau tangles. The imbalance between protein production and clearance has been increasingly recognized as a pathway to the pathogenesis of AD, which starts long before the symptom onsite [3-12]. It is, therefore, critical to understand how harmful substances are cleared from the brain parenchyma and how this process alterates in neurodegenerative diseases.

The brain's anatomy is unique: the parenchyma and all its communication channels with the outside are completely encased in cerebrospinal fluid (CSF) [13], including the parenchyma itself, all 12 pairs of cranial nerves, all vascular-communication channels, and the spinal cord. There are several consequences associated with this unique anatomy. First, it indicates that there are only two physical pathways for parenchymal waste removal: the CSF circulation pathways (Fig. 1a) and the parenchymal vascular pathways (Fig. 1b). Second, it suggests a dual mission of the brain vessels: blood transportation and CSF pumping. The reliance of CSF circulation on pulsation and vasomotion has been shown in both animal and human studies [14-19]. Third, material exchange between the brain parenchyma and the lymphatic system can only happen through the CSF because all brain's communications to the outside are bathed in CSF, including those in paravascular spaces surrounding vascular-communication channels and those in peri-neural spaces surrounding 12 pairs of cranial nerves. This suggests that CSF plays a critical role in regulating brain immunity. Fourth, substance removal from brain parenchyma can be studied with the CSF administration of tracers without direct injection into the brain tissues.

Figure 1.

Figure 1.

Summary of possible efflux routes of interstitial substances (endogenous or exogenous waste products) into the peripheral vascular system. Inside the parenchyma, the endogenous or exogenous substances can efflux either into the paravascular/perineural CSF (a), or into the vascular blood across the blood-brain barrier (b) to rapidly circulate into the peripheral vasculature (eγf). For those drained through the paravascular/perineural CSF (a), it will travel along this space to exit the parenchyma and transport either into connecting lymphatic vessels (e.g., meningeal lymphatic vessels, nasal mucosa, etc.) (c), or into the dura sinus through arachnoid villi/granulations (d). Eventually, all substance will drain into the peripheral vascular system (f).

The traditional view of parenchymal tracers getting into the blood pool is focused on arachnoid villi and granulations [20-22] (Fig. 1d). However, this view has been questioned by accumulating data from in vivo and post-mortem studies [23-25]. If arachnoid villi and granulations are not the main outflow pathway of CSF draining to the vascular system [25], which pathway contributes to the rapid observation of CSF tracer in the peripheral vascular system? Recent MRI results have demonstrated evident participation of the parenchymal venous system in draining the CSF tracers following intra-cisterna magna infusion of different MRI tracers [26]. This is consistent with the results of detecting CSF tracers in cerebral vascular walls by immunohistochemistry [25]. Moreover, various vascular transport mechanisms, including ATP binding cassette (ABC) transporters, solute carrier (SLC), and receptor mediated transcytosis (RMT), have been identified for cerebral clearance of both exogenous and endogenous substances across the blood-brain barrier, further strengthening the participation of parenchymal vascular system in substance clearance. Below, we will review evidence from different perspectives that jointly support an unignorable role of the parenchymal vascular system in waste clearance, aiming to raise awareness of the vasculature route when studying CSF pathways.

2. The consequence of the unique anatomy of the brain parenchyma

Brain parenchyma is the most crucial organ in the human body. In addition to protect it from physical insults, the brain's anatomy must also meet its demands for high energy consumption (3-5% of body weight but consumption of 15-25% of energy) and the corresponding requirement for effective and timely waste removal.

Unlike most peripheral vessels where arterial vessels are paired with venous vessels nearby, arterial vessels are separated from venous vessels inside the brain. This can be considered the anatomical basis for the glymphatic system, in that such a separation facilitates the interstitial fluid drainage into the venous side through the para-arterial to the para-venous flow gradient. In addition to the unique wiring of the vasculature, the brain parenchyma is also completely bathed in CSF: not only the brain parenchyma itself is surrounded by CSF, all its communication channels to the outside are also enclosed by CSF. Specifically, all parenchymal arterial and venous communication channels are surrounded by CSF-filled para-arterial space and para-venous space. The para-arterial and para-venous spaces are also termed influx and efflux pathways in the glymphatic theory, respectively. Similarly, all parenchymal nerve communication channels to the outside, including 12 pairs of cranial nerves and the spinal cord, are also surrounded by CSF-filled perineural space [27]. As a result, waste substances must exit through the CSF pathways (Fig. 1a) and/or through parenchymal vasculature by crossing BBB (Fig. 1b). All substances in these two pathways will eventually drain toward the peripheral vascular system (Fig. 1f).

Such CSF-encapsulated brain's anatomy suggests that there are possibly three main efflux routes by which the interstitial substances can reach the peripheral vascular system: 1) directly transport across BBB into the vascular system within the parenchyma (Fig. 1bγe); 2) travel along the paravascular space to exit the parenchyma and then enter the sinus system through arachnoid villi and granulations (Fig. 1aγd), 3) travel along the paravascular space to exit the parenchyma and then enter the connecting lymphatic vessels which innervate the para-brain area, then into the peripheral lymphatic system (Fig. 1aγc). Anatomically, the connecting lymphatic vessels, including the nasal lymphatics and the recently discovered meningeal lymphatic vessels at the skull base, are outside the brain parenchyma (Fig. 1c). Among perineural drainage pathways, the most studied ones are along the olfactory nerve (CN I), followed by the optic (CN II) nerve, and the trigeminal nerve (CN V). Other less-studied cranial nerve pathways include the glossopharyngeal (CN IX), vagus (CN X), accessory (CN XI) nerves, and facial nerve (CN VII). The unstudied CN III, IV, and VI theoretically may also participate in parenchymal substance clearance. Because the circulation speed of CSF is at least one order slower than that of blood, it would be desirable for the substances to enter the vascular system inside the parenchyma for fast drainage. For these reasons, it is crucial to determine whether the parenchymal vascular system directly participates in substance clearance.

Another consequence of the unique anatomy of the brain parenchyma is that the interaction between the brain parenchyma and the lymphatic system must involve the CSF clearance system, because all communication channels connecting parenchyma to the outside are surrounded by CSF and the brain parenchyma is devoid of lymph. For this reason, the paravascular space is expected to play a crucial role in the immunity of the brain parenchyma, which is consistent with the clinical manifestation of enlarged paravascular space in many neurological diseases [28-31].

Interestingly, the brain's CSF circulation network also offers unique opportunities to study the waste clearance pathways through CSF tracer injection without injuring the parenchymal tissue. Keep parenchymal tissue intact is key to understanding the brain clearance function under normal physiology, as parenchymal clearance is sensitive to many physiological parameters, including blood pressure [7], sleep [32], anesthesia [33-35], body posture [36], and aging [4, 7, 37-43]. For example, a 10 bpm decrease in heart rate (bradycardia) reduces substance clearance and results in a nearly 20% additional accumulation of Aβ in the brain parenchyma, whereas a 30 bpm increase in heart rate (tachycardia) showed a nearly 30% reduction in Aβ levels in the brain parenchyma [7]. Thus, any large perturbation and invasive procedures (i.e., direct injection into the parenchymal tissue) may alter the glymphatic function and, therefore, should be avoided. Fortunately, the unique design of the CSF network allows the intra-cisterna magna infused tracers to be delivered to the brain parenchyma without disturbing brain tissue. This injection approach has become a standard practice for tracking waste clearance pathways and investigating clearance functions in both animal and human studies [44].

3. The debated role of arachnoid villi and granulations in CSF drainage

Arachnoid villi or granulations have long been considered a primary site for CSF circulating back to the bloodstream through the dura sinus [45, 46]. As a result, it has been believed that the vascular participation of parenchymal waste clearance mainly occurs at the dural sinus level through arachnoid villi/granulations that project into the dural venous sinuses. Thus, the current paradigm suggests a dual-outflow system for CSF to reach the blood circulation, one directly to the venous blood through arachnoid projections (Fig. 1aγd) and one indirectly through the lymphatic system (Fig. 1aγc). However, the view of arachnoid villi/granulations as a major drainage site is debated [47, 48] and has been challenged by results from recent animal and human studies.

Recent mice studies have suggested that the arachnoid villi-sinus pathway plays a minimum role in draining the CSF-injected tracers [49, 50]. The studies reported non-detectible tracer outflow in the level of dural sinuses for both large and small molecular tracers, with the majority outflow through perineural routes to the lymphatic system. Their observations were also consistent with earlier MRI studies in dogs and pigs where tracers injected into the cisterna magna were observed across the cribriform plate, nasal (sub)mucosa, and periorbital regions, but no enhancement was observed in proximity to the superior sagittal sinus [51-53]. These findings collectively challenged the traditional view of arachnoid villi as a major outflow route of solute from CSF. In fact, historical data has never reached a consensus on the relative contribution of the arachnoid pathway to drain CSF tracers. In some species, such as rabbits and sheep, lymphatic vessels accounted for around 30-50% of total outflow. The remainder was assumed to be drained through arachnoid villi without direct evidence [54, 55]. The mice studies questioned this assumption and suggested that arachnoid villi are, at most, a secondary pathway for CSF drainage.

A recent human study with a large number of post-mortem patient samples and high-quality of histological and MRI data provided extensive details on the anatomy, morphology, and cytologic composition of arachnoid granulations, which suggested that the main role of arachnoid granulations serve as immune hubs at meningeal interfaces [56]. The study showed that only a subset of human arachnoid granulations associates with dural sinus, yet all arachnoid granulations cores are enriched with internal cytokine and immune cells, highlighting unexplored neuroimmune properties of these structures that localize to the brain-meningeal lymphatic interface. Given the high variability in arachnoid granulations morphology and its partial associations with the dural sinus, it is suggested that arachnoid granulations form a critical interface for immune surveillance at the brain surface, thus likely to be a secondary, rather than the major site, for the extra-parenchymal CSF drainage.

These in vivo and post-mortem findings collectively call into question the role of arachnoid villi as a major site for CSF drainage into the bloodstream. Supporting evidence for the importance of these structures draining CSF has mostly relied on examining post-mortem tissue [45, 57]. Although human studies have reported tracer deposition in the arachnoid/dura sinus regions [58], the findings were made in aged or diseased brains where complex morphological and functional changes of arachnoid granulations likely occur [56]. Anatomically, a continuous lining of endothelial cells with tight junctions exists on the arachnoid villus, which acts as a barrier to large molecules/proteins. It is still unclear how macromolecules are transported through this barrier [59, 60]. The recent discovery of the subarachnoid lymphatic-like membrane (SLYM) - a fourth meningeal layer that separates subarachnoid space into inner and outer layers - revealed an additional barrier for the CSF solute to exit from the inner layer to the outer layer and further to the dural sinuses under normal physiological pressure [61]. These structural barriers prohibit the arachnoid villi from being an efficient site for CSF solute to enter the bloodstream, implying alternative route(s) that facilitate CSF solute into the vascular system for fast circulation.

If the arachnoid villiγsinus pathway is not a major CSF outflow route (Fig. 1d, dashed line), what would be the express entry for the solute into the bloodstream? The rapid appearance of effluxed material in venous blood draining the head (within minutes after CSF tracer injection) has been observed in multiple studies [62-64]. This timeline suggests that vasculature transport across the BBB may play a role (Fig. 1b), as the alternative paravascular CSFγlymphγbloodstream route is too slow to allow the observation of such rapid venous efflux (Fig. 1aγcγf). Clearance of exogenous substances/tracers across the BBB is mainly understudied due to the difficulty of in-vivo detection and quantification of tracer concentration inside the smaller brain vessels. However, BBB is widely accepted as a critical route for removing many metabolic waste solutes (e.g., amino acids, Aβ peptides) from the brain to the blood circulation through different mechanisms [13, 65]. Between the two parenchymal efflux routes, i.e., transport across BBB (Fig. 1b) and efflux along paravascular CSF (Fig. 1aγcγf), BBB is likely the express route for the waste solutes to cycle into the bloodstream.

4. Substance efflux across blood-brain barriers: mechanism studies

The brain's elimination of many substrates (e.g., CO2, glucose, lactate, amino acids, Aβ peptides) are considerably greater than could be supported by paravascular efflux alone, suggesting that BBB is an important site for solute transport [13, 66]. The BBB consists of continuous endothelial cells joined together with tight junctions which are further supported by pericytes, the basement membrane, and the astrocytic endfeet. It makes the vascular pathway in brain parenchyma highly selective in both influx and efflux of substances, as compared to the CSF pathways, due to the tight junctions in the gaps between endothelial cells [67]. The effective efflux of substances from brain parenchyma to blood (brain-to-blood) is critical for rapid transport out of the brain in large quantities to maintain brain homeostasis. Depending on the size, concentration, and chemical compositions, the brain-to-blood efflux across BBB is made through three mechanisms: via passive transfer, via transporters, and via transcytosis [13, 65].

4.1. Efflux via passive, non-selective transfer

The structures of BBB determine that passive, non-selective transfer can occur via a paracellular (around the endothelial cells through tight junctions) or transcellular pathway (through the endothelial cells) [68-70]. The paracellular pathway is more restrictive as blocked by the tight junctions and only allows for small solutes to transport, e.g., Na+ and Cl- [68-70]. The transcellular pathway, on the other hand, is responsible for the majority of the passively removed solutes. These solutes are either more lipid soluble (which allows them to easily diffuse across both the cell membranes and the interior of the endothelial cells) or small neutral substances such as water, methanol, ethanol, isopropanol, glycerol, ethylene glycol, urea, and thiourea [65]. The features of passive, non-selective transfer, as compared to the other two mechanisms, is that this transport does not saturate, is not inhibited by competition by other transported substances, and has no specific inhibitors. This passive permeability is a diffusion-like process that allows both influx and efflux directions [71, 72]. It has been speculated that a combination of hydrostatic and osmotic pressure gradients, known as Starling Forces, drive the cross-BBB passive transfer [73-75]. An increase in the interstitial-to-venous pressure would increase the solute efflux rate into the vein and vice versa. As a result, such efflux is likely to be affected in pathological states such as venous hypertension, where the "hydraulic push" would be reduced with the venous pressure increase.

4.2. Efflux via active BBB Transporters

BBB possesses virious transporters for many types of solutes located on luminal (blood-facing), abluminal (brain-facing) or both surfaces of the brain capillary endothelial cells (BCEC). ATP-binding cassette (ABC) and solute carrier (SLC) are the prominent efflux transporter families at BBB.

4.2.1. ABC transporters

The discovery of ATP-binding cassette (ABC) in the early 1970s and the subsequent demonstration of their expression within the BBB added a critical element to barrier function. ABC efflux transporters belong to an ancient protein superfamily and are highly conserved among different species [76]. By consuming ATP, ABC transporters extrude metabolic wastes produced by brain parenchyma into the blood and prevent the xenobiotics from entering the brain parenchyma against the solute's concentration gradient. Detailed reviews of ABC efflux transporters can be found in [76, 77]. In the context of this review, we only focused on the transporters of ABCA, ABCB, ABCC, and ABCG families located in the membrane of the brain capillary endothelial cells (BCEC) that could efflux the substances from brain parenchyma to blood.

Transporters in ABCA family are involved in apolipoprotein (ApoE)-dependent cholesterol efflux, sterol homeostasis, and lipid metabolism. ACBA1 is the most studied ABCA transporter in CNS. Panzenboeck and colleagues [78] reported that ABCA1 is located in the abluminal membrane of BCEC. They found that ABCA1 contributed to ApoE-dependent cholesterol efflux in porcine BCEC. For other members of the ABCA family, related mRNA or protein expression has been detected in BCEC [79-82].

ABCB1 (P-glycoprotein, Pgp; multidrug resistences1, MDR1) transports amphipathic, cationic, and neutral compounds. ABCB1 was the first ABC transporter identified in a drug-resistant cell line in 1976 [83]. In 1989, Cordon-Cardo and colleagues [84, 85] found that ABCB1 existed in human brain capillary endothelial cells. The ABCB1 transporter protein is highly expressed in the luminal membrane of brain capillary endothelial cells, preventing toxins from entering the brain parenchyma. Interestingly, ABCB1 was also found in the abluminal membrane of BCEC and other locations inside the cells. However, the functions of the ABCB1 expressing in the abluminal membrane and intracellular remain unclear [86, 87]. It is worth noting that ABCB1 transports an extensive range of substrates. These substrates have large structural diversity (small molecules such as morphine, verapamil, and loperamide; peptides such as Aβ) and various classes (such as chemotherapeutics, HIV protease inhibitors, opioids, and so on) [88-96].

ABCC1 (also known as multidrug resistance protein MRP1) was first detected in the CNS in 1998 [97]. The localization of ABCC1 at the BBB is still controversial. Roberts et al. [98] reported that ABCC1 is primarily located in the abluminal membrane of BCEC, but a low level is also present in the luminal membrane. However, Cisternino S. et al. [99] found that efflux mediated by ABCC1 does not seem to occur across the luminal membrane of the BCEC. ABCC1 was reported to be associated with the efflux of 17-β-estradiol-D-17-β-glucuronide (E217βG) across BBB [100]. Besides ABCC1, other transporters in the ABCC family, such as ABCC3, ABCC4, and ABCC5, were also expressed in the membrane of BCEC. ABCC3 and ABCC5 could mediate the transport of methotrexate [101]. And ABCC4 has been demonstrated to mediate the efflux of many substances, including organic anions, glutathione-, sulfate-, or glucuronate-conjugated drugs, prostaglandins, nucleoside analogs, methotrexate, topotecan, and thiopurines [102-105].

ABCG2 (also known as breast cancer resistance protein, BCRP) was found in 1998 by Doyle et al. in a breast cancer cell line (MCF-7) that displayed high resistance to mitoxantrone [106]. In 2002, ABCG2 was detected in BBB by Eisenblätter et al. [107]. The localization of ABCG2 has been found in the luminal membrane of brain capillaries of capillary endothelial cell cultures [98, 108-112]. At BBB, ABCG2 mediates the efflux of daunorubicin, prazosin, and mitoxantrone [109, 113]. ABCG2 has also been demonstrated to efflux several exogenous chemotherapeutic drugs, such as imatinib, dasatinib, and lapatinib [109, 113-118]. Besides ABCG2, ABCG1 (White1), ABCG5 (White3, Sterolin-1), and ABCG8 (Sterolin-2) have been found on the plasma side of BCEC. They are transporters for cholesterol, sterols, and biosynthetic sterol intermediates and are responsible for cholesterol and sterol homeostasis [119].

4.2.2. SLC transporters

More than 500 genes classified in 65 families named solute carrier (SLC) have been reported (see http://slc.bioparadigms.org/). Many SLC transporters have been detected in the membrane of BCEC [120]. While some SLC transporters mediate the transportation of specific substrates, others transport a relatively broad range of organic anions and cations (primarily those in the SLC21 and SLC22 families). The SLC transporters have been reviewed extensively in previous literatures [121-132]. In this review, we only focused on the SLC transporters that could efflux the substances from brain parenchyma to blood. The SLC transporters reviewed in this review included SLC1, SLC6, SLC7, SLC16, SLC21, SLC22, SLC38.

Members of the SLC1 family are glutamate transporters [133]. SLC1A3 (GLAST), SLC1A2 (GLT1), and SLC1A1 (EAAC1) have been detected in the abluminal membrane of bovine brain microvessels and been found to catalyze glutamate efflux from the brain parenchyma [134, 135]. SLC1A5 localized at the abluminal side of BCEC mediates the L-aspartate efflux from the parenchyma [136].

SLC6 is the sodium- and chloride-dependent neurotransmitter transporter family. SLC6A6 (TAUT) has been detected in the rat BCEC [137], which mediates the transport of taurine (2-amino-ethane sulfonic acid). SLC6A13 (GAT2/BGT-1) has been detected in the cultured brain endothelial cells. It mediates the removal of GABA and betaine from the brain parenchyma [138]. SLC6A2 (NET) and SLC6A4 (SERT) have been detected in mouse brain capillary cell line, which mediates the transport of norepinephrine and serotonin, respectively [139].

The SLC7 family mediates the transportation of amino acids. It can be divided into two subfamilies: the cationic amino acid transporters (CATs) and L-type amino acid transporters (LATs). SLC7A5 (LAT1) is a glycoprotein-associated transporter belonging to LATs. Boado and colleagues [140] showed that SLC7A5 mediates the transport of amino acids through the BBB. SLC7A11 (also known as xCT) is a cystine/glutamate antiporter that belongs to LATs. L-Aspartate and L-glutamate are the predominant substrates of SLC7A11 [141]. It has been reported that SLC7A11 expresses in the luminal membrane and mediates the efflux of glutamate into the blood [142].

The SLC16 family has been first detected in the 1970s as a specific L-lactate membrane transporter [143]. SLC16A1 (MCT1) is the first cloned member of its family, which mediates the transportation of short-chain monocarboxylates [144, 145]. Gerhart et al. [146] found SLC16A1 in rat brain endothelium's luminal and abluminal membranes. Besides nutrient transport, MCT1 is also responsible for removing lactate from the brain parenchyma to endothelial cells and plasma, especially under pathological conditions [28].

The SLC21 family mediates the transport of organic anions. In this family, SLCO1A4 (OATP2) has been detected in the rat brain and mouse BCEC [147]. The localization of SLCO1A4 is postulated to be in the luminal and abluminal membranes of the BCEC [148]. It mediates the efflux of various substrates from the brain parenchyma. The substrates of SLCO1A4 include glucuronides, sulfates, glutathione conjugates, δ-opioid receptor agonists, and valproic acid [149-151].

SLC22A5 (OCTN2) is an organic cation transporter in the SLC22 family. It has been detected in the primary culture of the BCEC and the cell lines [152-156]. The localization of SLC22A5 was postulated to be in both the luminal and abluminal sides of the BBB. The substrates of SLC22A5 are several organic cations, such as β-lactam antibiotics [157, 158].

SLC38 is a family of neutral amino acid transporters. SLC38A3 (SNAT3) has been detected in the mouse capillary luminal and abluminal membranes, which mediates glutamine transport in both directions. SLC38A2 (ATA) was seen in the abluminal side of rat BBB. It mediates the efflux of small amino acids, including L-proline, glycine, and L-alanine [159].

In summary, substrate efflux via transporters located on the membranes of BCEC has been extensively investigated. Ample evidence demonstrated that these transporters are a critical pathway for waste clearance from the parenchyma, as substrate efflux through SLC has revealed a clearance volume much greater than the clearance associated with the paravascular route (Table 1 in [65]). The evidence further showed that these transporters are responsible for the quick elimination of solute from the parenchyma, resulting in the rapid appearance of effluxed material in venous blood draining the head.

4.3. Efflux via transcytosis

Transcytosis is an active transcellular efflux mechanism for the transport of large substrates across the BBB through either absorptive-mediated transcytosis (AMT) or receptor-mediated transcytosis (RMT). AMT usually starts with the adsorption of substrates onto the surface of caveolae, while RMT is initialized by binding substrates with specific receptors. These initialization events lead to endocytosis flowed by delivery of the vesicles to the cell membrane of the opposite side [160-162]. There is evidence that transcytosis can occur in either direction, i.e., from the brain to blood and from blood to brain. The large substrates that are transported from brain to blood through the transcytosis pathway include Aβ peptides (via low-density lipoprotein receptor-related protein LRP1 and LRP2) [163, 164], insulin (via insulin receptor), [165, 166], transferrin (via the transferrin receptor, TfR) [167, 168], and IgG molecules (via interaction with an unidentified receptor). Note that while both BBB, paravascular, and periarterial routes have been identified for Aβ efflux, studies have shown that LRP1-dependent transcytosis plays a substantial role in Aβ elimination, with LRP1 knocked out mice showing 48% reduction in Aβ removal [169, 170]. The first glymphatic study also reported a much faster Aβ clearance than mannitol or dextran, where the latter two substances lack specific efflux receptors, supporting receptor-mediated efflux of Aβ across the BBB [6].

Collectively, these well-studied mechanisms demonstrate the ability of cerebral vasculature to drain metabolite waste substance across blood-brain barriers in large quantities. Besides brain's innate waste substance, many "blood-to-brain impermeable" tracers have been observed to drain into the cerebral veins following cisterna-magna tracer injections [25, 26, 171]. While the exact mechanism for their brain-to-blood transport across BBB awaits further investigation, these observations suggest a role of vascular participation in draining exogenous tracers, as discussed in detail below.

5. Parenchymal vasculature participates in substance clearance: Imaging and immunohistochemical studies

The strongest evidence for vascular participation in waste clearance comes from MRI and immunohistochemical studies. In this section, we will discuss the MRI technique and review the evidence for the direct involvement of parenchymal vasculature in substance clearance.

5.1. MRI vs. two-photon microscopy imaging technique

As a hydraulic system, both the paravascular and vascular spaces must be studied intact without any leakage, ideally with minimal perturbation to the system and ultra-high detection sensitivity. Two-photon microscopy and MRI are the most used imaging techniques for studying brain waste clearance. Two-photo microscopy has ultra-high detection sensitivity and has made ground-breaking discoveries, including the glymphatic system and solving the long-time puzzle of why we need sleep [172]. However, due to technical limitations, it can only image the brain surface with a limited penetration depth (i.e., 250µm). Therefore, it can only capture regional/partial paravascular and vascular pathways. As a result, two-photon imaging may miss some critical influx/efflux routes that prevent it from answering whether parenchymal vasculature directly participates in substance clearance. In comparison, the MRI-based superparamagnetic iron oxide enhanced susceptibility weighted imaging (SPIO-SWI) has a whole-brain coverage. It can overcome insufficient spatial resolution in conventional MRI with a marked increase in detection sensitivity. Therefore, SPIO-SWI may provide a more comprehensive picture of influx and efflux pathways.

5.2. The SPIO-SWI technique

Conventional MRI cannot simultaneously study the micro-vessels and their surrounding paravascular space due to limited imaging resolution and the small sizes of these spaces. SWI overcomes the limited imaging resolution by incorporating the phase information of the image. MRI provides both magnitude and phase information [173, 174], yet the phase information is usually discarded in conventional MRI. The susceptibility-induced phase signal can amplify the underlying signal source and boost the detection sensitivity. Combined with the superparamagnetic iron oxide (SPIO), SPIO-SWI can offer a blooming effect that can significantly increase the detection sensitivity of sub-pixel micro-vessels, providing the capability to distinguish between paravascular and adjacent intra-vascular spaces.

It has been demonstrated SPIO-SWI can increase the MRI-measured vessel diameter by 13.3-fold and 19.9-fold with 5.6 mg Fe/kg and 16.8 mg Fe/kg ferumoxytol [175]. Moreover, SPIO-SWI provides excellent contrast between CSF (dark signal due to SPIO) and blood (bright signal due to inflow effect). Therefore, SPIO-SWI becomes a superb tool for studying the clearance pathways along both paravascular and vascular networks simultaneously given its whole-brain coverage, high detection sensitivity, and enhanced contrast between blood and CSF.

5.3. MRI evidence for the direct participation of parenchymal vasculature in substance clearance with the intrathecal administration of CSF tracers

As discussed, imaging the intrathecally administered CSF tracers can reveal clearance pathways due to the unique CSF network. Combined with the SPIO-SWI technique, determining whether parenchymal vasculature directly participates in substance clearance becomes simple. If tracers in venous blood flowing out of the brain parenchyma are more than that in the arterial inflow, we can conclude that tracers in the brain parenchyma enter venous blood [26].

Three experiments were conducted with observations that unanimously support the direct participation of parenchymal vasculature in substance efflux. For the first experiment, the signal intensity profile at the vein and artery and corresponding para-venous and para-arterial space was qualitatively estimated before and after the intra-cisterna magna infusion of 100μg Fe-dextran (100nm). The decrease of signal intensity from both para-arterial and para-venous space indicated the MRI tracers transported through the paravascular space, consistent with the glymphatic theory. More importantly, while the signal intensity from the artery (azygos pericallosal artery) remained unchanged, the signal intensity from the veins (azygos internal cerebral vein) decreased. This indicated that MRI tracers enter the vein but not the artery, suggesting the participation of parenchymal veins in draining the Fe-dextran tracer.

For the second experiment, CSF tracers were quantitatively measured in both the vein and artery using a similar experimental setup but with a 75 μg (Fe) FeREX™ (50-150 nm) tracer. The relative signal intensity changes in the veins decreased by 16.0 (± 4.1) % at 15 min post-tracer infusion compared to baseline (pre-tracer) (p < 0.01). In contrast, the relative signal changes in the arteries were not significantly different (p >0.01) [26]. In other words, the outflow of tracers in venous blood was more than its inflow in arterial blood, suggesting its drainage through the veins.

For the third experiment, the susceptibility of the MRI tracers in both the vein and artery was estimated by quantitative susceptibility mapping (QSM) to reduce possible blooming effects on arteries and veins. Greater QSM changes in the vein than in the artery were detected at both post-15min and post-45min 75μg FeREX™ (100nm) CSF injection, suggesting more tracers in the vein than in the artery. Moreover, the comparison between MRI integral signal in the vein and adjacent para-venous space gave a ratio of 32:100, suggesting a non-ignorable contribution of venous efflux in tracer clearance, in addition to the para-venous drainage.

5.4. Immunofluorescence study indicated the presence of CSF tracer in the BBB

Immunofluorescence imaging was used to study brain tissue two hours after injecting FITC-dextran (molecular weight 10KDa) tracer into the right ventricle. The presence of CSF tracer within the endothelial walls of capillaries and small vessels was observed [25]. An analogous result was reported by Wagner et al [176], where they identified vesicular transport within both vacular endothelial cells and perivascular microglia as primary mechanisms for clearing horseradish peroxidase from CSF into blood plasma. These histology findings echo with the in vivo imaging results [26], which collectively support the vascular involvement in parenchymal substance clearance.

6. Implications for understanding neuro-degenerative diseases

As discussed, substance clearance in the brain parenchyma relies on both the vascular system and the glymphatic system. Impairments of either system have been observed in neurodegenerative diseases such as AD [9, 67, 177-190], Parkinson’s disease [191-201], and traumatic brain injury [202-208] in both animal and human studies. For example, damages to cerebro-vasculature, such as the increased vascular permeability or transporter dysfunction across BBB, were associated with AD pathologies (Aβ and tau) and contributed to the onset and progression of AD [67, 177-183]. And impaired glymphatic system has also been found in AD pathogenesis [9, 184-190]. These studies suggested that integrity of both vascular and glymphatic functions are critical to brain waste clearance. Dysfunction of either one may cause waste buildup and trigger neurodegenerative diseases [209].

Therefore, simultaneous evaulation of cerebro-vascular and glymphatic functions can be beneficial in studying neurodegenerative diseases. Current imaging techniques mainly evaluate cerebrovascular or glymphatic function alone, e.g., targeting either perfusion or CSF [14, 178, 189, 194, 205, 209-213]. SPIO-SWI holds promises in simultaneously evaluating waste drainage along both vascular and glymphatic pathways with clinical translation potential [26, 171]. Also, multimodal imaging techniques could be incorporated to simultaneously assess both clearance pathways to gain a complete view of brain’s clearance function.

Therapeutic strategies that target both vascular and glymphatic systems rather than target either system alone could be explored to achieve better treatment effects. Drugs and intervention strategies targeting either cerebrovascular or glymphatic dysfunctions have been investigated to prevent or alleviate neurodegenerative diseases [214-221]. Yet, the impairments of cerebrovascular and glymphatic function may share common mechanisms. For example, the deficiency of NOTCH3, primarily expressed in vascular smooth muscle cells (vSMCs), could impair cerebral vessel architecture, contractility, and glymphatic function [222]. And disrupted glymphatic function has been observed when cardiovascular functions were altered [15, 16, 18, 223]. Therefore, considering both clearance systems during the development of future therapies could prove to be advantageous.

While our focus is the brain parenchyma, we could speculate that this dual clearance system—the combined vascular and CSF clearance—may apply to waste clearance function in other parts of the central nervous system, including the spinal cord and the neural parts of the eye and inner ear. Dysfunctional water drainage in the ocular and the inner ear may underlie glaucoma and Meniere disease, both of which are characterized by an excess of local water [224]. Recent studies have suggested an ocular glymphatic system based on the observance of Aβ clearance along the optic nerves and intravenously injected GBCA leaking into the CSF surrounding optic nerves [225-227]. The studies further proposed that glymphatic dysfunction may underlie the water accumulation. Based on our previous discussion, we suspect altered vasculature and CSF drainage may contribute to the excess water. Thus, both pathways are worth investigating in future studies of these diseases.

7. Conclusion

Together, the evidence from different perspectives highlights a contribution of the cerebral vascular system in brain clearance: 1) the intra-parenchymal vascular transport may be the express entry for the solute into the bloodstream that underlies the rapid appearance of effluxed material in venous blood observed in many studies, especially given that the arachnoid villi/granulations as the main site for solute entering the vascular system have been questioned; 2) multiple blood-brain barrier transport mechanisms have been identified for the parenchymal vascular drainage; 3) recent MRI studies of CSF tracer efflux pathways support direct participation of the parenchymal venous system in draining the intra-cisterna magna infused tracers, complimenting the paravascular CSF drainage (glymphatic system); Furthermore, the different sizes of CSF tracers can enter the parenchymal venous system but not arterial vessels, reflecting a directional transport of BBB. While the relative contribution of vascular and CSF pathways in waste clearance is yet to be studied and maybe substance dependent, the parenchymal vascular drainage is logically consistent with the notion that the venous system outside the brain drains about 90% of the interstitial fluid, with the remaining 10% drained through the lymphatic system that has the similar circulation speed as the CSF [228]. As the brain is the most bioactive, energy-consuming organ, it is logical that vascular clearance plays a role, particularly considering less bioactive peripheral tissues require both the fast vascular and slow lymphatic systems to remove substances efficiently. In conclusion, the parenchymal vascular contribution to the brain's substance clearance should gain more attention in future studies. Understanding the brain’s reliance on the two pathways is crucial for unveiling the underlying mechanism of neurodegenerative diseases and for developing effective diagnostic and therapeutic approaches.

Footnotes

Disclosure Statement

The authors have no conflicts of interest to declare.

References

  • [1].Kaur J, Fahmy LM, Davoodi-Bojd E, Li Z, Ding GL, Hu JN, et al. (2021). Waste clearance in the brain. Front Neuroanat, 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Frank MG, Heller HC (2019). The function(s) of sleep. Handb Exp Pharmacol, 253:3-34. [DOI] [PubMed] [Google Scholar]
  • [3].Arbel-Ornath M, Hudry E, Eikermann-Haerter K, Hou S, Gregory JL, Zhao L, et al. (2013). Interstitial fluid drainage is impaired in ischemic stroke and Alzheimer's disease mouse models. Acta Neuropathol, 126:353-364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Da Mesquita S, Louveau A, Vaccari A, Smirnov I, Cornelison RC, Kingsmore KM, et al. (2018). Functional aspects of meningeal lymphatics in ageing and Alzheimer's disease. Nature, 560:185-+. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Goodman JR, Adham ZO, Woltjer RL, Lund AW, Iliff JJ (2018). Characterization of dural sinus-associated lymphatic vasculature in human Alzheimer's dementia subjects. Brain Behav Immun, 73:34-40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, et al. (2012). A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med, 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Kyrtsos CR, Baras JS (2015). Modeling the role of the glymphatic pathway and cerebral blood vessel properties in Alzheimer's disease pathogenesis. Plos One, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Louveau A, Da Mesquita S, Kipnis J (2016). Lymphatics in neurological disorders: A neuro-lympho-vascular component of multiple sclerosis and Alzheimer's disease? Neuron, 91:957-973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Peng W, Achariyar TM, Li B, Liao Y, Mestre H, Hitomi E, et al. (2016). Suppression of glymphatic fluid transport in a mouse model of Alzheimer's disease. Neurobiol Dis, 93:215-225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Reeves BC, Karimy JK, Kundishora AJ, Mestre H, Cerci HM, Matouk C, et al. (2020). Glymphatic system impairment in Alzheimer's disease and idiopathic normal pressure hydrocephalus. Trends Mol Med, 26:285-295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Tarasoff-Conway JM, Carare RO, Osorio RS, Glodzik L, Butler T, Fieremans E, et al. (2015). Clearance systems in the brain-implications for Alzheimer disease. Nat Rev Neurol, 11:457-470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Xia M, Yang L, Sun G, Qi S, Li B (2017). Mechanism of depression as a risk factor in the development of Alzheimer's disease: the function of AQP4 and the glymphatic system. Psychopharmacology, 234:365-379. [DOI] [PubMed] [Google Scholar]
  • [13].Hladky SB, Barrand MA (2016). Fluid and ion transfer across the blood-brain and blood-cerebrospinal fluid barriers; a comparative account of mechanisms and roles. Fluids Barriers CNS, 13:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Wen Q, Tong Y, Zhou X, Dzemidzic M, Ho CY, Wu YC (2022). Assessing pulsatile waveforms of paravascular cerebrospinal fluid dynamics within the glymphatic pathways using dynamic diffusion-weighted imaging (dDWI). Neuroimage, 260:119464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Mestre H, Tithof J, Du T, Song W, Peng W, Sweeney AM, et al. (2018). Flow of cerebrospinal fluid is driven by arterial pulsations and is reduced in hypertension. Nat Commun, 9:4878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Iliff JJ, Wang M, Zeppenfeld DM, Venkataraman A, Plog BA, Liao Y, et al. (2013). Cerebral arterial pulsation drives paravascular CSF-interstitial fluid exchange in the murine brain. J Neurosci, 33:18190-18199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Hadaczek P, Yamashita Y, Mirek H, Tamas L, Bohn MC, Noble C, et al. (2006). The "perivascular pump" driven by arterial pulsation is a powerful mechanism for the distribution of therapeutic molecules within the brain. Mol Ther, 14:69-78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Rennels ML, Gregory TF, Blaumanis OR, Fujimoto K, Grady PA (1985). Evidence for a 'paravascular' fluid circulation in the mammalian central nervous system, provided by the rapid distribution of tracer protein throughout the brain from the subarachnoid space. Brain Res, 326:47-63. [DOI] [PubMed] [Google Scholar]
  • [19].Fultz NE, Bonmassar G, Setsompop K, Stickgold RA, Rosen BR, Polimeni JR, et al. (2019). Coupled electrophysiological, hemodynamic, and cerebrospinal fluid oscillations in human sleep. Science, 366:628-631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Davson H (1966). Formation and drainage of the cerebrospinal fluid. Sci Basis Med Annu Rev:238-259. [PubMed] [Google Scholar]
  • [21].Davson H, Domer FR, Hollingsworth JR (1973). Mehcanism of drainage of cerebrospinal-fluid. Brain, 96:329-336. [DOI] [PubMed] [Google Scholar]
  • [22].Johanson CE, Duncan JA 3rd, Klinge PM, Brinker T, Stopa EG, Silverberg GD (2008). Multiplicity of cerebrospinal fluid functions: New challenges in health and disease. Cerebrospinal Fluid Res, 5:10-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Brinker T, Stopa E, Morrison J, Klinge P (2014). A new look at cerebrospinal fluid circulation. Fluids and Barriers of the CNS, 11:10-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Eide PK, Mariussen E, Uggerud H, Pripp AH, Lashkarivand A, Hassel B, et al. (2021). Clinical application of intrathecal gadobutrol for assessment of cerebrospinal fluid tracer clearance to blood. JCI Insight, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Krishnamurthy S, Li J, Shen Y, Duncan TM, Jenrow KA, Haacke EM (2018). Normal macromolecular clearance out of the ventricles is delayed in hydrocephalus. Brain Res, 1678:337-355. [DOI] [PubMed] [Google Scholar]
  • [26].Hu J, Shen Y, Fahmy LM, Krishnamurthy S, Li J, Zhang L, et al. (2023). The role of the parenchymal vascular system in cerebrospinal fluid tracer clearance. Eur Radiol, 33:656-665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Fahmy LM, Chen Y, Xuan S, Haacke EM, Hu J, Jiang Q (2021). All Central Nervous System Neuro- and Vascular-Communication Channels Are Surrounded With Cerebrospinal Fluid. Front Neurol, 12:614636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Shi Y, Wardlaw JM (2016). Update on cerebral small vessel disease: a dynamic whole-brain disease. Stroke Vasc Neurol, 1:83-92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Debette S, Schilling S, Duperron M-G, Larsson SC, Markus HS (2019). Clinical significance of magnetic resonance imaging markers of vascular brain injury A systematic review and meta-analysis. JAMA Neurology, 76:81-94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Das AS, Regenhardt RW, Vernooij MW, Blacker D, Charidimou A, Viswanathan A (2019). Asymptomatic cerebral small vessel disease: Insights from population-based studies. J Stroke, 21:121-138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Wardlaw JM, Benveniste H, Nedergaard M, Zlokovic BV, Mestre H, Lee H, et al. (2020). Perivascular spaces in the brain: anatomy, physiology and pathology. Nat Rev Neurol, 16:137-153. [DOI] [PubMed] [Google Scholar]
  • [32].Nedergaard M, Goldman SA (2016). Brain drain. Sci Am, 314:44-49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Benveniste H, Lee H, Ding F, Sun Q, Al-Bizri E, Makaryus R, et al. (2017). Anesthesia with dexmedetomidine and low-dose isoflurane increases solute transport via the glymphatic pathway in rat brain when compared with high-dose isoflurane. Anesthesiology, 127:976-988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Benveniste H, Heerdt PM, Fontes M, Rothman DL, Volkow ND (2019). Glymphatic System Function in Relation to Anesthesia and Sleep States. Anesth Analg, 128:747-758. [DOI] [PubMed] [Google Scholar]
  • [35].Gakuba C, Gaberel T, Goursaud S, Bourges J, Di Palma C, Quenault A, et al. (2018). General Anesthesia Inhibits the Activity of the "Glymphatic System". Theranostics, 8:710-722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Lee H, Xie L, Yu M, Kang H, Feng T, Deane R, et al. (2015). The effect of body posture on brain glymphatic transport. J Neurosci, 35:11034-11044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Jessen NA, Munk ASF, Lundgaard I, Nedergaard M (2015). The glymphatic system: A beginner's guide. Neurochem Res, 40:2583-2599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Chen RL, Kassem NA, Redzic ZB, Chen CPC, Segal MB, Preston JE (2009). Age-related changes in choroid plexus and blood-cerebrospinal fluid barrier function in the sheep. Exp Gerontol, 44:289-296. [DOI] [PubMed] [Google Scholar]
  • [39].Fleischman D, Berdahl JP, Zaydlarova J, Stinnett S, Fautsch MP, Allingham RR (2012). Cerebrospinal fluid pressure decreases with older age. Plos One, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Iliff JJ, Wang M, Zeppenfeld DM, Venkataraman A, Plog BA, Liao Y, et al. (2013). Cerebral arterial pulsation drives paravascular CSF-interstitial fluid exchange in the murine brain. J Neurosci, 33:18190-18199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Kress BT, Iliff JJ, Xia M, Wang M, Wei HS, Zeppenfeld D, et al. (2014). Impairment of paravascular clearance pathways in the aging brain. Ann Neurol, 76:845-861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Zhou Y, Cai J, Zhang W, Gong X, Yan S, Zhang K, et al. (2020). Impairment of the glymphatic pathway and putative meningeal lymphatic vessels in the aging human. Ann Neurol, 87:357-369. [DOI] [PubMed] [Google Scholar]
  • [43].Zieman SJ, Melenovsky V, Kass DA (2005). Mechanisms, pathophysiology, and therapy of arterial stiffness. Arterioscler Thromb Vasc Biol, 25:932-943. [DOI] [PubMed] [Google Scholar]
  • [44].Xavier ALR, Hauglund NL, von Holstein-Rathlou S, Li QL, Sanggaard S, Lou NH, et al. (2018). Cannula implantation into the cisterna magna of rodents. JOVE. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Key ARG. 1875. Studien in der Anatomie des Nervensystems und des Bindegewebes: Stockholm: Samson & Wallin. [Google Scholar]
  • [46].Woollam DH (1957). The historical significance of the cerebrospinal fluid. Med Hist, 1:91-114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Proulx ST (2021). Cerebrospinal fluid outflow: a review of the historical and contemporary evidence for arachnoid villi, perineural routes, and dural lymphatics. Cell Mol Life Sci, 78:2429-2457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Dandy WE (1929). Where is cerebrospinal fluid absorbed? JAMA, 92:2012-2014. [Google Scholar]
  • [49].Decker Y, Kramer J, Xin L, Muller A, Scheller A, Fassbender K, et al. (2022). Magnetic resonance imaging of cerebrospinal fluid outflow after low-rate lateral ventricle infusion in mice. JCI Insight, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Ma Q, Ineichen BV, Detmar M, Proulx ST (2017). Outflow of cerebrospinal fluid is predominantly through lymphatic vessels and is reduced in aged mice. Nat Commun, 8:1434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Di Chiro G, Girton ME, Frank JA, Dietz MJ, Gansow OA, Wright DC, et al. (1986). Cerebrospinal fluid rhinorrhea: depiction with MR cisternography in dogs. Radiology, 160:221-222. [DOI] [PubMed] [Google Scholar]
  • [52].Di Chiro G, Stein SC, Harrington T (1972). Spontaneous cerebrospinal fluid rhinorrhea in normal dogs. Radioisotope studies of an alternate pathway of CSF drainage. J Neuropathol Exp Neurol, 31:447-453. [DOI] [PubMed] [Google Scholar]
  • [53].Yamada S, Shibata M, Scadeng M, Bluml S, Nguy C, Ross B, et al. (2005). MRI tracer study of the cerebrospinal fluid drainage pathway in normal and hydrocephalic guinea pig brain. Tokai J Exp Clin Med, 30:21-29. [PubMed] [Google Scholar]
  • [54].Koh L, Zakharov A, Johnston M (2005). Integration of the subarachnoid space and lymphatics: is it time to embrace a new concept of cerebrospinal fluid absorption? Cerebrospinal Fluid Res, 2:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Boulton M, Flessner M, Armstrong D, Hay J, Johnston M (1998). Determination of volumetric cerebrospinal fluid absorption into extracranial lymphatics in sheep. Am J Physiol, 274:R88-96. [DOI] [PubMed] [Google Scholar]
  • [56].Shah T, Leurgans SE, Mehta RI, Yang J, Galloway CA, de Mesy Bentley KL, et al. (2023). Arachnoid granulations are lymphatic conduits that communicate with bone marrow and dura-arachnoid stroma. J Exp Med, 220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Weed LH (1914). Studies on Cerebro-Spinal Fluid. No. III : The pathways of escape from the Subarachnoid Spaces with particular reference to the Arachnoid Villi. J Med Res, 31:51-91. [PMC free article] [PubMed] [Google Scholar]
  • [58].Ringstad G, Eide PK (2020). Cerebrospinal fluid tracer efflux to parasagittal dura in humans. Nat Commun, 11:354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Pollay M (2010). The function and structure of the cerebrospinal fluid outflow system. Cerebrospinal Fluid Res, 7:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Tripathi BJ, Tripathi RC (1974). Vacuolar transcellular channels as a drainage pathway for cerebrospinal fluid. J Physiol, 239:195-206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Mollgard K, Beinlich FRM, Kusk P, Miyakoshi LM, Delle C, Pla V, et al. (2023). A mesothelium divides the subarachnoid space into functional compartments. Science, 379:84-88. [DOI] [PubMed] [Google Scholar]
  • [62].Mann JD, Butler AB, Johnson RN, Bass NH (1979). Clearance of macromolecular and particulate substances from the cerebrospinal fluid system of the rat. J Neurosurg, 50:343-348. [DOI] [PubMed] [Google Scholar]
  • [63].Dixon WE, Halliburton WD (1916). The cerebro-spinal fluid: IV. Circulation. J Physiol, 50:198-216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Dandy WE, Blackfan KD (1914). An experimental, clinical and pathological study: Part 1. —experimental studies. AJDC, VIII:406-482. [Google Scholar]
  • [65].Hladky SB, Barrand MA (2018). Elimination of substances from the brain parenchyma: efflux via perivascular pathways and via the blood-brain barrier. Fluids Barriers CNS, 15:30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Pulgar VM (2018). Transcytosis to Cross the Blood Brain Barrier, New Advancements and Challenges. Front Neurosci, 12:1019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV (2019). Blood-Brain Barrier: From Physiology to Disease and Back. Physiol Rev, 99:21-78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Ennis SR, Ren XD, Betz AL (1996). Mechanisms of sodium transport at the blood-brain barrier studied with in situ perfusion of rat brain. J Neurochem, 66:756-763. [DOI] [PubMed] [Google Scholar]
  • [69].Crone C (1986). The blood-brain barrier as a tight epithelium: where is information lacking? Ann N Y Acad Sci, 481:174-185. [DOI] [PubMed] [Google Scholar]
  • [70].Davson H, Segal MB (1970). The effects of some inhibitors and accelerators of sodium transport on the turnover of 22Na in the cerebrospinal fluid and the brain. J Physiol, 209:131-153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Simpson IA, Carruthers A, Vannucci SJ (2007). Supply and demand in cerebral energy metabolism: the role of nutrient transporters. J Cereb Blood Flow Metab, 27:1766-1791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Hladky SB, Barrand MA (2019). Metabolite clearance during wakefulness and sleep. Handb Exp Pharmacol, 253:385-423. [DOI] [PubMed] [Google Scholar]
  • [73].Linninger AA, Xu C, Tangen K, Hartung G (2017). Starling forces drive intracranial water exchange during normal and pathological states. Croat Med J, 58:384-394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Woodcock TE, Michel CC (2021). Advances in the Starling Principle and Microvascular Fluid Exchange; Consequences and Implications for Fluid Therapy. Front Vet Sci, 8:623671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Starling EH (1896). On the Absorption of Fluids from the Connective Tissue Spaces. J Physiol, 19:312-326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Dean M, Allikmets R (1995). Evolution of ATP-binding cassette transporter genes. Curr Opin Genet Dev, 5:779-785. [DOI] [PubMed] [Google Scholar]
  • [77].Hartz AM, Bauer B (2011). ABC transporters in the CNS - an inventory. Curr Pharm Biotechnol, 12:656-673. [DOI] [PubMed] [Google Scholar]
  • [78].Panzenboeck U, Balazs Z, Sovic A, Hrzenjak A, Levak-Frank S, Wintersperger A, et al. (2002). ABCA1 and scavenger receptor class B, type I, are modulators of reverse sterol transport at an in vitro blood-brain barrier constituted of porcine brain capillary endothelial cells. J Biol Chem, 277:42781-42789. [DOI] [PubMed] [Google Scholar]
  • [79].Gosselet F, Candela P, Sevin E, Berezowski V, Cecchelli R, Fenart L (2009). Transcriptional profiles of receptors and transporters involved in brain cholesterol homeostasis at the blood-brain barrier: use of an in vitro model. Brain Res, 1249:34-42. [DOI] [PubMed] [Google Scholar]
  • [80].Ohtsuki S, Watanabe Y, Hori S, Suzuki H, Bhongsatiern J, Fujiyoshi M, et al. (2004). mRNA expression of the ATP-binding cassette transporter subfamily A (ABCA) in rat and human brain capillary endothelial cells. Biol Pharm Bull, 27:1437-1440. [DOI] [PubMed] [Google Scholar]
  • [81].Quezada CA, Garrido WX, González-Oyarzún MA, Rauch MC, Salas MR, San Martín RE, et al. (2008). Effect of tacrolimus on activity and expression of P-glycoprotein and ATP-binding cassette transporter A5 (ABCA5) proteins in hematoencephalic barrier cells. Biol Pharm Bull, 31:1911-1916. [DOI] [PubMed] [Google Scholar]
  • [82].Warren MS, Zerangue N, Woodford K, Roberts LM, Tate EH, Feng B, et al. (2009). Comparative gene expression profiles of ABC transporters in brain microvessel endothelial cells and brain in five species including human. Pharmacol Res, 59:404-413. [DOI] [PubMed] [Google Scholar]
  • [83].Juliano RL, Ling V (1976). A surface glycoprotein modulating drug permeability in Chinese hamster ovary cell mutants. Biochim Biophys Acta, 455:152-162. [DOI] [PubMed] [Google Scholar]
  • [84].Cordon-Cardo C, O'Brien JP, Casals D, Rittman-Grauer L, Biedler JL, Melamed MR, et al. (1989). Multidrug-resistance gene (P-glycoprotein) is expressed by endothelial cells at blood-brain barrier sites. Proc Natl Acad Sci U S A, 86:695-698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Thiebaut F, Tsuruo T, Hamada H, Gottesman MM, Pastan I, Willingham MC (1989). Immunohistochemical localization in normal-tissues of different epitopes in the multidrug transport protein P170 - evidence for localization in brain capillaries and crossreactivity of one antibody with a muscle protein. J Histochem Cytochem, 37:159-164. [DOI] [PubMed] [Google Scholar]
  • [86].Bendayan R, Ronaldson PT, Gingras D, Bendayan M (2006). In situ localization of P-glycoprotein (ABCB1) in human and rat brain. J Histochem Cytochem, 54:1159-1167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Pardridge WM, Golden PL, Kang YS, Bickel U (1997). Brain microvascular and astrocyte localization of P-glycoprotein. J Neurochem, 68:1278-1285. [DOI] [PubMed] [Google Scholar]
  • [88].Beaulieu E, Demeule M, Ghitescu L, Béliveau R (1997). P-glycoprotein is strongly expressed in the luminal membranes of the endothelium of blood vessels in the brain. Biochem J, 326(Pt 2):539-544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Hartz AM, Miller DS, Bauer B (2010). Restoring blood-brain barrier P-glycoprotein reduces brain amyloid-beta in a mouse model of Alzheimer's disease. Mol Pharmacol, 77:715-723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Biegel D, Spencer DD, Pachter JS (1995). Isolation and culture of human brain microvessel endothelial cells for the study of blood-brain barrier properties in vitro. Brain Res, 692:183-189. [DOI] [PubMed] [Google Scholar]
  • [91].Kuhnke D, Jedlitschky G, Grube M, Krohn M, Jucker M, Mosyagin I, et al. (2007). MDR1-P-Glycoprotein (ABCB1) mediates transport of Alzheimer's amyloid-beta peptides--implications for the mechanisms of Abeta clearance at the blood-brain barrier. Brain Pathol, 17:347-353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Shirai A, Naito M, Tatsuta T, Dong J, Hanaoka K, Mikami K, et al. (1994). Transport of cyclosporin A across the brain capillary endothelial cell monolayer by P-glycoprotein. Biochim Biophys Acta, 1222:400-404. [DOI] [PubMed] [Google Scholar]
  • [93].Sugawara I, Hamada H, Tsuruo T, Mori S (1990). Specialized localization of P-glycoprotein recognized by MRK 16 monoclonal antibody in endothelial cells of the brain and the spinal cord. JPN J Cancer Res, 81:727-730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Tanaka Y, Abe Y, Tsugu A, Takamiya Y, Akatsuka A, Tsuruo T, et al. (1994). Ultrastructural localization of P-glycoprotein on capillary endothelial cells in human gliomas. Virchows Arch, 425:133-138. [DOI] [PubMed] [Google Scholar]
  • [95].Tsuji A, Terasaki T, Takabatake Y, Tenda Y, Tamai I, Yamashima T, et al. (1992). P-glycoprotein as the drug efflux pump in primary cultured bovine brain capillary endothelial cells. Life Sci, 51:1427-1437. [DOI] [PubMed] [Google Scholar]
  • [96].Bauer B, Hartz AM, Fricker G, Miller DS (2005). Modulation of p-glycoprotein transport function at the blood-brain barrier. Exp Biol Med (Maywood), 230:118-127. [DOI] [PubMed] [Google Scholar]
  • [97].Regina A, Koman A, Piciotti M, El Hafny B, Center MS, Bergmann R, et al. (1998). MRP1 multidrug resistance-associated protein and P-glycoprotein expression in rat brain microvessel endothelial cells. J Neurochem, 71:705-715. [DOI] [PubMed] [Google Scholar]
  • [98].Roberts LM, Black DS, Raman C, Woodford K, Zhou M, Haggerty JE, et al. (2008). Subcellular localization of transporters along the rat blood-brain barrier and blood-cerebral-spinal fluid barrier by in vivo biotinylation. Neurosci, 155:423-438. [DOI] [PubMed] [Google Scholar]
  • [99].Cisternino S, Rousselle C, Lorico A, Rappa G, Scherrmann JM (2003). Apparent lack of MRP1-mediated efflux at the luminal side of mouse blood-brain barrier endothelial cells. Pharm Res, 20:904-909. [DOI] [PubMed] [Google Scholar]
  • [100].Sugiyama D, Kusuhara H, Lee YJ, Sugiyama Y (2003). Involvement of multidrug resistance associated protein 1 (Mrp1) in the efflux transport of 17beta estradiol-D-17beta-glucuronide (E217betaG) across the blood-brain barrier. Pharm Res, 20:1394-1400. [DOI] [PubMed] [Google Scholar]
  • [101].Poller B, Gutmann H, Krähenbühl S, Weksler B, Romero I, Couraud PO, et al. (2008). The human brain endothelial cell line hCMEC/D3 as a human blood-brain barrier model for drug transport studies. J Neurochem, 107:1358-1368. [DOI] [PubMed] [Google Scholar]
  • [102].Zhou SF, Wang LL, Di YM, Xue CC, Duan W, Li CG, et al. (2008). Substrates and inhibitors of human multidrug resistance associated proteins and the implications in drug development. Curr Med Chem, 15:1981-2039. [DOI] [PubMed] [Google Scholar]
  • [103].Adachi M, Reid G, Schuetz JD (2002). Therapeutic and biological importance of getting nucleotides out of cells: a case for the ABC transporters, MRP4 and 5. Adv Drug Deliv Rev, 54:1333-1342. [DOI] [PubMed] [Google Scholar]
  • [104].Leggas M, Adachi M, Scheffer GL, Sun DX, Wielinga P, Du GQ, et al. (2004). MRP4 confers resistance to topotecan and protects the brain from chemotherapy. Mol Cell Biol, 24:7612-7621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Chen ZS, Lee K, Walther S, Raftogianis RB, Kuwano M, Zeng H, et al. (2002). Analysis of methotrexate and folate transport by multidrug resistance protein 4 (ABCC4): MRP4 is a component of the methotrexate efflux system. Cancer Res, 62:3144-3150. [PubMed] [Google Scholar]
  • [106].Doyle LA, Yang WD, Abruzzo LV, Krogmann T, Gao YM, Rishi AK, et al. (1998). A multidrug resistance transporter from human MCF-7 breast cancer cells. PNAS, 95:15665-15670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Eisenblatter T, Galla HJ (2002). A new multidrug resistance protein at the blood-brain barrier. Biochem Biophys Res Commun, 293:1273-1278. [DOI] [PubMed] [Google Scholar]
  • [108].Aronica E, Gorter JA, Redeker S, van Vliet EA, Ramkema M, Scheffer GL, et al. (2005). Localization of breast cancer resistance protein (BCRP) in microvessel endothelium of human control and epileptic brain. Epilepsia, 46:849-857. [DOI] [PubMed] [Google Scholar]
  • [109].Cisternino S, Mercier C, Bourasset F, Roux F, Scherrmann JM (2004). Expression, up-regulation, and transport activity of the multidrug-resistance protein Abcg2 at the mouse blood-brain barrier. Cancer Res, 64:3296-3301. [DOI] [PubMed] [Google Scholar]
  • [110].Cooray HC, Blackmore CG, Maskell L, Barrand MA (2002). Localisation of breast cancer resistance protein in microvessel endothelium of human brain. Neuroreport, 13:2059-2063. [DOI] [PubMed] [Google Scholar]
  • [111].Hartz AM, Mahringer A, Miller DS, Bauer B (2010). 17-β-Estradiol: a powerful modulator of blood-brain barrier BCRP activity. J Cereb Blood Flow Metab, 30:1742-1755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [112].Hori S, Ohtsuki S, Tachikawa M, Kimura N, Kondo T, Watanabe M, et al. (2004). Functional expression of rat ABCG2 on the luminal side of brain capillaries and its enhancement by astrocyte-derived soluble factor(s). J Neurochem, 90:526-536. [DOI] [PubMed] [Google Scholar]
  • [113].Eisenblatter T, Huwel S, Galla HJ (2003). Characterisation of the brain multidrug resistance protein (BMDP/ABCG2/BCRP) expressed at the blood-brain barrier. Brain Res, 971:221-231. [DOI] [PubMed] [Google Scholar]
  • [114].Breedveld P, Beijnen JH, Schellens JH (2006). Use of P-glycoprotein and BCRP inhibitors to improve oral bioavailability and CNS penetration of anticancer drugs. Trends Pharmacol Sci, 27:17-24. [DOI] [PubMed] [Google Scholar]
  • [115].Chen Y, Agarwal S, Shaik NM, Chen C, Yang Z, Elmquist WF (2009). P-glycoprotein and breast cancer resistance protein influence brain distribution of dasatinib. J Pharmacol Exp Ther, 330:956-963. [DOI] [PubMed] [Google Scholar]
  • [116].Lagas JS, van der Kruijssen CM, van de Wetering K, Beijnen JH, Schinkel AH (2009). Transport of diclofenac by breast cancer resistance protein (ABCG2) and stimulation of multidrug resistance protein 2 (ABCC2)-mediated drug transport by diclofenac and benzbromarone. Drug Metab Dispos, 37:129-136. [DOI] [PubMed] [Google Scholar]
  • [117].Polli JW, Olson KL, Chism JP, John-Williams LS, Yeager RL, Woodard SM, et al. (2009). An unexpected synergist role of P-glycoprotein and breast cancer resistance protein on the central nervous system penetration of the tyrosine kinase inhibitor lapatinib (N-{3-chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methylsulfonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine; GW572016). Drug Metab Dispos, 37:439-442. [DOI] [PubMed] [Google Scholar]
  • [118].Zhou L, Schmidt K, Nelson FR, Zelesky V, Troutman MD, Feng B (2009). The effect of breast cancer resistance protein and P-glycoprotein on the brain penetration of flavopiridol, imatinib mesylate (Gleevec), prazosin, and 2-methoxy-3-(4-(2-(5-methyl-2-phenyloxazol-4-yl)ethoxy)phenyl)propanoic acid (PF-407288) in mice. Drug Metab Dispos, 37:946-955. [DOI] [PubMed] [Google Scholar]
  • [119].Wang N, Yvan-Charvet L, Lütjohann D, Mulder M, Vanmierlo T, Kim TW, et al. (2008). ATP-binding cassette transporters G1 and G4 mediate cholesterol and desmosterol efflux to HDL and regulate sterol accumulation in the brain. Faseb J, 22:1073-1082. [DOI] [PubMed] [Google Scholar]
  • [120].Hediger MA. BioParadigms SLC tables. [Google Scholar]
  • [121].Ashraf T, Ronaldson PT, Bendayan R.2014. Drug transport in the brain. In Drug Transporters. 273-301. [Google Scholar]
  • [122].Bronger H, König J, Kopplow K, Steiner HH, Ahmadi R, Herold-Mende C, et al. (2005). ABCC drug efflux pumps and organic anion uptake transporters in human gliomas and the blood-tumor barrier. Cancer Res, 65:11419-11428. [DOI] [PubMed] [Google Scholar]
  • [123].Farthing CA, Sweet DH (2014). Expression and function of organic cation and anion transporters (SLC22 family) in the CNS. Curr Pharm Des, 20:1472-1486. [DOI] [PubMed] [Google Scholar]
  • [124].Geier EG, Chen EC, Webb A, Papp AC, Yee SW, Sadee W, et al. (2013). Profiling solute carrier transporters in the human blood-brain barrier. Clin Pharmacol Ther, 94:636-639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Hagenbuch B, Stieger B (2013). The SLCO (former SLC21) superfamily of transporters. Mol Aspects Med, 34:396-412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Koepsell H, Endou H (2004). The SLC22 drug transporter family. Pflugers Arch, 447:666-676. [DOI] [PubMed] [Google Scholar]
  • [127].Kovacsics D, Patik I, Özvegy-Laczka C (2017). The role of organic anion transporting polypeptides in drug absorption, distribution, excretion and drug-drug interactions. Expert Opin Drug Metab Toxicol, 13:409-424. [DOI] [PubMed] [Google Scholar]
  • [128].Kusuhara H, Sugiyama Y (2005). Active efflux across the blood-brain barrier: Role of the solute carrier family. NeuroRX, 2:73-85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Obaidat A, Roth M, Hagenbuch B (2012). The expression and function of organic anion transporting polypeptides in normal tissues and in cancer. Annu Rev Pharmacol Toxicol, 52:135-151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Ronaldson PT, Davis TP (2013). Targeted drug delivery to treat pain and cerebral hypoxia. Pharmacol Rev, 65:291-314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Roth M, Obaidat A, Hagenbuch B (2012). OATPs, OATs and OCTs: the organic anion and cation transporters of the SLCO and SLC22A gene superfamilies. Br J of Pharmacol, 165:1260-1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Zhou F, Zhu L, Wang K, Murray M (2017). Recent advance in the pharmacogenomics of human solute carrier transporters (SLCs) in drug disposition. Adv Drug Deliv Rev, 116:21-36. [DOI] [PubMed] [Google Scholar]
  • [133].Grewer C, Gameiro A, Rauen T (2014). SLC1 glutamate transporters. Pflugers Arch, 466:3-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Cederberg HH, Uhd NC, Brodin B (2014). Glutamate efflux at the blood-brain barrier: cellular mechanisms and potential clinical relevance. Arch Med Res, 45:639-645. [DOI] [PubMed] [Google Scholar]
  • [135].O'Kane RL, Martínez-López I, DeJoseph MR, Viña JR, Hawkins RA (1999). Na(+)-dependent glutamate transporters (EAAT1, EAAT2, and EAAT3) of the blood-brain barrier. A mechanism for glutamate removal. J Biol Chem, 274:31891-31895. [DOI] [PubMed] [Google Scholar]
  • [136].Tetsuka K, Takanaga H, Ohtsuki S, Hosoya K, Terasaki T (2003). The l-isomer-selective transport of aspartic acid is mediated by ASCT2 at the blood-brain barrier. J Neurochem, 87:891-901. [DOI] [PubMed] [Google Scholar]
  • [137].Kang YS, Ohtsuki S, Takanaga H, Tomi M, Hosoya K, Terasaki T (2002). Regulation of taurine transport at the blood-brain barrier by tumor necrosis factor-alpha, taurine and hypertonicity. J Neurochem, 83:1188-1195. [DOI] [PubMed] [Google Scholar]
  • [138].Takanaga H, Ohtsuki S, Hosoya K, Terasaki T (2001). GAT2/BGT-1 as a system responsible for the transport of gamma-aminobutyric acid at the mouse blood-brain barrier. J Cereb Blood Flow Metab, 21:1232-1239. [DOI] [PubMed] [Google Scholar]
  • [139].Wakayama K, Ohtsuki S, Takanaga H, Hosoya K, Terasaki T (2002). Localization of norepinephrine and serotonin transporter in mouse brain capillary endothelial cells. Neurosci Res, 44:173-180. [DOI] [PubMed] [Google Scholar]
  • [140].Boado RJ, Li JY, Nagaya M, Zhang C, Pardridge WM (1999). Selective expression of the large neutral amino acid transporter at the blood-brain barrier. Proc Natl Acad Sci U S A, 96:12079-12084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [141].Fotiadis D, Kanai Y, Palacín M (2013). The SLC3 and SLC7 families of amino acid transporters. Mol Aspects Med, 34:139-158. [DOI] [PubMed] [Google Scholar]
  • [142].Hosoya K, Tomi M, Ohtsuki S, Takanaga H, Saeki S, Kanai Y, et al. (2002). Enhancement of L-cystine transport activity and its relation to xCT gene induction at the blood-brain barrier by diethyl maleate treatment. J Pharmacol Exp Ther, 302:225-231. [DOI] [PubMed] [Google Scholar]
  • [143].Halestrap AP, Denton RM (1974). Specific inhibition of pyruvate transport in rat liver mitochondria and human erythrocytes by alpha-cyano-4-hydroxycinnamate. Biochem J, 138:313-316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [144].Halestrap AP, Meredith D (2004). The SLC16 gene family-from monocarboxylate transporters (MCTs) to aromatic amino acid transporters and beyond. Pflugers Arch, 447:619-628. [DOI] [PubMed] [Google Scholar]
  • [145].Halestrap AP, Wilson MC (2012). The monocarboxylate transporter family--role and regulation. IUBMB Life, 64:109-119. [DOI] [PubMed] [Google Scholar]
  • [146].Gerhart DZ, Enerson BE, Zhdankina OY, Leino RL, Drewes LR (1997). Expression of monocarboxylate transporter MCT1 by brain endothelium and glia in adult and suckling rats. Am J Physiol, 273:E207-213. [DOI] [PubMed] [Google Scholar]
  • [147].Asaba H, Hosoya K, Takanaga H, Ohtsuki S, Tamura E, Takizawa T, et al. (2000). Blood-brain barrier is involved in the efflux transport of a neuroactive steroid, dehydroepiandrosterone sulfate, via organic anion transporting polypeptide 2. J Neurochem, 75:1907-1916. [DOI] [PubMed] [Google Scholar]
  • [148].Gao B, Stieger B, Noé B, Fritschy JM, Meier PJ (1999). Localization of the organic anion transporting polypeptide 2 (Oatp2) in capillary endothelium and choroid plexus epithelium of rat brain. J Histochem Cytochem, 47:1255-1264. [DOI] [PubMed] [Google Scholar]
  • [149].Gao B, Hagenbuch B, Kullak-Ublick GA, Benke D, Aguzzi A, Meier PJ (2000). Organic anion-transporting polypeptides mediate transport of opioid peptides across blood-brain barrier. J Pharmacol Exp Ther, 294:73-79. [PubMed] [Google Scholar]
  • [150].Guo Y, Jiang L (2016). Organic anion transporting polypeptide 2 transports valproic acid in rat brain microvascular endothelial cells. Neurol Res, 38:634-639. [DOI] [PubMed] [Google Scholar]
  • [151].Sugiyama D, Kusuhara H, Shitara Y, Abe T, Meier PJ, Sekine T, et al. (2001). Characterization of the efflux transport of 17beta-estradiol-D-17beta-glucuronide from the brain across the blood-brain barrier. J Pharmacol Exp Ther, 298:316-322. [PubMed] [Google Scholar]
  • [152].Berezowski V, Miecz D, Marszałek M, Bröer A, Bröer S, Cecchelli R, et al. (2004). Involvement of OCTN2 and B0,+ in the transport of carnitine through an in vitro model of the blood-brain barrier. J Neurochem, 91:860-872. [DOI] [PubMed] [Google Scholar]
  • [153].Friedrich A, Prasad PD, Freyer D, Ganapathy V, Brust P (2003). Molecular cloning and functional characterization of the OCTN2 transporter at the RBE4 cells, an in vitro model of the blood-brain barrier. Brain Res, 968:69-79. [DOI] [PubMed] [Google Scholar]
  • [154].Kido Y, Tamai I, Ohnari A, Sai Y, Kagami T, Nezu J-i, et al. (2001). Functional relevance of carnitine transporter OCTN2 to brain distribution of l-carnitine and acetyl-l-carnitine across the blood-brain barrier. J Neurochem, 79:959-969. [DOI] [PubMed] [Google Scholar]
  • [155].Miecz D, Januszewicz E, Czeredys M, Hinton BT, Berezowski V, Cecchelli R, et al. (2008). Localization of organic cation/carnitine transporter (OCTN2) in cells forming the blood-brain barrier. J Neurochem, 104:113-123. [DOI] [PubMed] [Google Scholar]
  • [156].Okura T, Kato S, Deguchi Y (2014). Functional expression of organic cation/carnitine transporter 2 (OCTN2/SLC22A5) in human brain capillary endothelial cell line hCMEC/D3, a human blood-brain barrier model. Drug Metab Pharmacokinet, 29:69-74. [DOI] [PubMed] [Google Scholar]
  • [157].Ganapathy ME, Huang W, Rajan DP, Carter AL, Sugawara M, Iseki K, et al. (2000). beta-lactam antibiotics as substrates for OCTN2, an organic cation/carnitine transporter. J Biol Chem, 275:1699-1707. [DOI] [PubMed] [Google Scholar]
  • [158].Wu X, Prasad PD, Leibach FH, Ganapathy V (1998). cDNA sequence, transport function, and genomic organization of human OCTN2, a new member of the organic cation transporter family. Biochem Biophys Res Commun, 246:589-595. [DOI] [PubMed] [Google Scholar]
  • [159].Takanaga H, Tokuda N, Ohtsuki S, Hosoya K, Terasaki T (2002). ATA2 is predominantly expressed as system A at the blood-brain barrier and acts as brain-to-blood efflux transport for L-proline. Mol Pharmacol, 61:1289-1296. [DOI] [PubMed] [Google Scholar]
  • [160].Preston JE, Joan Abbott N, Begley DJ (2014). Transcytosis of macromolecules at the blood-brain barrier. Adv Pharmacol, 71:147-163. [DOI] [PubMed] [Google Scholar]
  • [161].Banks WA (2012). Drug delivery to the brain in Alzheimer's disease: consideration of the blood-brain barrier. Adv Drug Deliv Rev, 64:629-639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [162].Pan W. 2006. CHAPTER 200 - Permeability of the blood—brain barrier to neurotrophic peptides. In Handbook of Biologically Active Peptides. Kastin AJ, editor. Burlington: Academic Press. 1435-1441. [Google Scholar]
  • [163].Shibata M, Yamada S, Kumar SR, Calero M, Bading J, Frangione B, et al. (2000). Clearance of Alzheimer's amyloid-ss(1-40) peptide from brain by LDL receptor-related protein-1 at the blood-brain barrier. J Clin Invest, 106:1489-1499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [164].Qosa H, Abuasal BS, Romero IA, Weksler B, Couraud P-O, Keller JN, et al. (2014). Differences in amyloid-β clearance across mouse and human blood-brain barrier models: Kinetic analysis and mechanistic modeling. Neuropharmacology, 79:668-678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].Duffy KR, Pardridge WM (1987). Blood-brain barrier transcytosis of insulin in developing rabbits. Brain Res, 420:32-38. [DOI] [PubMed] [Google Scholar]
  • [166].Rhea EM, Rask-Madsen C, Banks WA (2018). Insulin transport across the blood-brain barrier can occur independently of the insulin receptor. J Physiol, 596:4753-4765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Banks WA, Kastin AJ, Fasold MB, Barrera CM, Augereau G (1988). Studies of the slow bidirectional transport of iron and transferrin across the blood-brain barrier. Brain Res Bull, 21:881-885. [DOI] [PubMed] [Google Scholar]
  • [168].Zhang Y, Pardridge WM (2001). Rapid transferrin efflux from brain to blood across the blood-brain barrier. J Neurochem, 76:1597-1600. [DOI] [PubMed] [Google Scholar]
  • [169].Storck SE, Meister S, Nahrath J, Meißner JN, Schubert N, Di Spiezio A, et al. (2016). Endothelial LRP1 transports amyloid-β(1-42) across the blood-brain barrier. J Clin Invest, 126:123-136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [170].Zlokovic BV, Deane R, Sagare AP, Bell RD, Winkler EA (2010). Low-density lipoprotein receptor-related protein-1: a serial clearance homeostatic mechanism controlling Alzheimer's amyloid beta-peptide elimination from the brain. J Neurochem, 115:1077-1089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [171].Hu J, Shen Y, Fahmy LM, Krishnamurthy S, Li J, Zhang L, et al. (2022). Quantitative results for the direct participation of the parenchymal vascular system in cerebral waste removal. Veins and Lymphatics, 11. [Google Scholar]
  • [172].Xie LL, Kang HY, Xu QW, Chen MJ, Liao YH, Thiyagarajan M, et al. (2013). Sleep drives metabolite clearance from the adult brain. Science, 342:373-377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [173].Haacke EM, Chengb NYC, House MJ, Liu Q, Neelavalli J, Ogg RJ, et al. (2005). Imaging iron stores in the brain using magnetic resonance imaging. Magn Reson Imaging, 23:1-25. [DOI] [PubMed] [Google Scholar]
  • [174].Haacke EM, Xu YB, Cheng YCN, Reichenbach JR (2004). Susceptibility weighted imaging (SWI). MRM, 52:612-618. [DOI] [PubMed] [Google Scholar]
  • [175].Wang H, Jiang Q, Shen Y, Zhang L, Haacke EM, Ge Y, et al. (2020). The capability of detecting small vessels beyond the conventional MRI sensitivity using iron-based contrast agent enhanced susceptibility weighted imaging. NMR in Biomedicine, 33. [DOI] [PubMed] [Google Scholar]
  • [176].Wagner HJ, Pilgrim C, Brandl J (1974). Penetration and removal of horseradish peroxidase injected into the cerebrospinal fluid: role of cerebral perivascular spaces, endothelium and microglia. Acta Neuropathol, 27:299-315. [DOI] [PubMed] [Google Scholar]
  • [177].Montagne A, Zhao Z, Zlokovic BV (2017). Alzheimer's disease: A matter of blood-brain barrier dysfunction? Journal of Experimental Medicine, 214:3151-3169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [178].Nation DA, Sweeney MD, Montagne A, Sagare AP, D'Orazio LM, Pachicano M, et al. (2019). Blood-brain barrier breakdown is an early biomarker of human cognitive dysfunction. Nature Medicine, 25:270-+. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Zipser BD, Johanson CE, Gonzalez L, Berzin TM, Tavares R, Hulette CM, et al. (2007). Microvascular injury and blood-brain barrier leakage in Alzheimer's disease. Neurobiology of Aging, 28:977-986. [DOI] [PubMed] [Google Scholar]
  • [180].Sweeney MD, Kisler K, Montagne A, Toga AW, Zlokovic BV (2018). The role of brain vasculature in neurodegenerative disorders. Nature Neuroscience, 21:1318-1331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [181].Deane R, Zlokovic BV (2007). Role of the blood-brain barrier in the pathogenesis of Alzheimer's disease. Current Alzheimer Research, 4:191-197. [DOI] [PubMed] [Google Scholar]
  • [182].Ramos-Cejudo J, Wisniewski T, Marmar C, Zetterberg H, Blennow K, de Leon MJ, et al. (2018). Traumatic Brain Injury and Alzheimer's Disease: The Cerebrovascular Link. Ebiomedicine, 28:21-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [183].Sweeney MD, Montagne A, Sagare AP, Nation DA, Schneider LS, Chui HC, et al. (2019). Vascular dysfunction-The disregarded partner of Alzheimer's disease. Alzheimers & Dementia, 15:158-167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [184].Arighi A, Arcaro M, Fumagalli GG, Carandini T, Pietroboni AM, Sacchi L, et al. (2022). Aquaporin-4 cerebrospinal fluid levels are higher in neurodegenerative dementia: looking at glymphatic system dysregulation. Alzheimers Research & Therapy, 14:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [185].Iliff JJ, Lee H, Yu M, Feng T, Logan J, Nedergaard M, et al. (2013). Brain-wide pathway for waste clearance captured by contrast-enhanced MRI. Journal of Clinical Investigation, 123:1299-1309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Municio C, Carrero L, Antequera D, Carro E (2023). Choroid Plexus Aquaporins in CSF Homeostasis and the Glymphatic System: Their Relevance for Alzheimer's Disease. International Journal of Molecular Sciences, 24:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [187].Rasmussen MK, Mestre H, Nedergaard M (2018). The glymphatic pathway in neurological disorders. Lancet Neurology, 17:1016-1024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [188].Silva I, Silva J, Ferreira R, Trigo D (2021). Glymphatic system, AQP4, and their implications in Alzheimer's disease. Neurological research and practice, 3:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [189].Hsu JL, Wei YC, Toh CH, Hsiao IT, Lin KJ, Yen TC, et al. (2023). Magnetic Resonance Images Implicate That Glymphatic Alterations Mediate Cognitive Dysfunction in Alzheimer Disease. Annals of Neurology, 93:164-174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [190].Buccellato FR, D'Anca M, Serpente M, Arighi A, Galimberti D (2022). The Role of Glymphatic System in Alzheimer's and Parkinson's Disease Pathogenesis. Biomedicines, 10:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [191].Segal O, Mardor Y, Orbach L, Hoffman C, Hassin-Baer S (2022). Advanced radiological investigation of levodopa- induced dyskinesia in Parkinson's disease patients: assessment of blood brain barrier function. Neurology Asia, 27:353-362. [Google Scholar]
  • [192].Lan GY, Wang P, Chan RB, Liu ZR, Yu ZW, Liu XD, et al. (2022). Astrocytic VEGFA: An essential mediator in blood-brain-barrier disruption in Parkinson's disease. Glia, 70:337-353. [DOI] [PubMed] [Google Scholar]
  • [193].Khor SLQ, Ng KY, Koh RY, Chye SM (2023). Blood-Brain Barrier and Neurovascular Unit Dysfunction in Parkinson's Disease: From Clinical Insights to Pathogenic Mechanisms and Novel Therapeutic Approaches. CNS & neurological disorders drug targets. [DOI] [PubMed] [Google Scholar]
  • [194].Al-Bachari S, Naish JH, Parker GJM, Emsley HCA, Parkes LM (2020). Blood-Brain Barrier Leakage Is Increased in Parkinson's Disease. Frontiers in Physiology, 11:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [195].Gasca-Salas C, Fernandez-Rodriguez B, Pineda-Pardo JA, Rodriguez-Rojas R, Obeso I, Hernandez-Fernandez F, et al. (2021). Blood-brain barrier opening with focused ultrasound in Parkinson's disease dementia. Nature Communications, 12:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [196].Chen HL, Chen PC, Lu CH, Tsai NW, Yu CC, Chou KH, et al. (2021). Associations among Cognitive Functions, Plasma DNA, and Diffusion Tensor Image along the Perivascular Space (DTI-ALPS) in Patients with Parkinson's Disease. Oxidative Medicine and Cellular Longevity, 2021:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [197].Scott-Massey A, Boag MK, Magnier A, Bispo D, Khoo TK, Pountney DL (2022). Glymphatic System Dysfunction and Sleep Disturbance May Contribute to the Pathogenesis and Progression of Parkinson's Disease. International Journal of Molecular Sciences, 23:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [198].Si X, Dai S, Fang Y, Tang J, Wang Z, Li Y, et al. (2023). Matrix metalloproteinase-9 inhibition prevents aquaporin-4 depolarization-mediated glymphatic dysfunction in Parkinson's disease. Journal of advanced research. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [199].Si XL, Guo T, Wang ZY, Fang Y, Gu LY, Cao LX, et al. (2022). Neuroimaging evidence of glymphatic system dysfunction in possible REM sleep behavior disorder and Parkinson's disease. Npj Parkinsons Disease, 8:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [200].Qin Y, He RC, Chen J, Zhou XX, Zhou X, Liu ZH, et al. Neuroimaging uncovers distinct relationships of glymphatic dysfunction and motor symptoms in Parkinson's disease. Journal of Neurology: 10. [DOI] [PubMed] [Google Scholar]
  • [201].Gu LY, Dai SB, Guo T, Si XL, Lv DY, Wang ZY, et al. (2023). Noninvasive neuroimaging provides evidence for deterioration of the glymphatic system in Parkinson's disease relative to essential tremor. Parkinsonism & Related Disorders, 107:9. [DOI] [PubMed] [Google Scholar]
  • [202].Thal SC, Neuhaus W (2014). The Blood-Brain Barrier as a Target in Traumatic Brain Injury Treatment. Archives of Medical Research, 45:698-710. [DOI] [PubMed] [Google Scholar]
  • [203].Prakash R, Carmichael ST (2015). Blood-brain barrier breakdown and neovascularization processes after stroke and traumatic brain injury. Current Opinion in Neurology, 28:556-564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [204].Tomkins O, Feintuch A, Benifla M, Cohen A, Friedman A, Shelef I (2011). Blood-brain barrier breakdown following traumatic brain injury: a possible role in posttraumatic epilepsy. Cardiovascular psychiatry and neurology, 2011:765923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [205].Ware JB, Sinha S, Morrison J, Walter AE, Gugger JJ, Schneider ALC, et al. (2022). Dynamic contrast enhanced MRI for characterization of blood-brain-barrier dysfunction after traumatic brain injury. Neuroimage-Clinical, 36:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [206].Ferrara M, Bertozzi G, Volonnino G, Di Fazio N, Frati P, Cipolloni L, et al. (2022). Glymphatic System a Window on TBI Pathophysiology: A Systematic Review. International Journal of Molecular Sciences, 23:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [207].Iliff JJ, Chen MJ, Plog BA, Zeppenfeld DM, Soltero M, Yang LJ, et al. (2014). Impairment of Glymphatic Pathway Function Promotes Tau Pathology after Traumatic Brain Injury. Journal of Neuroscience, 34:16180-16193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [208].Piantino J, Lim MM, Newgard CD, Iliff J (2019). Linking Traumatic Brain Injury, Sleep Disruption and Post-Traumatic Headache: a Potential Role for Glymphatic Pathway Dysfunction. Current Pain and Headache Reports, 23:11. [DOI] [PubMed] [Google Scholar]
  • [209].Zhang B, Li W, Zhuo YG, Xiang H, Li WF, Liu HY, et al. (2021). L-3-n-Butylphthalide Effectively Improves the Glymphatic Clearance and Reduce Amyloid-beta Deposition in Alzheimer's Transgenic Mice. Journal of Molecular Neuroscience, 71:1266-1274. [DOI] [PubMed] [Google Scholar]
  • [210].Alghanimy A, Martin C, Gallagher L, Holmes WM (2023). The effect of a novel AQP4 facilitator, TGN-073, on glymphatic transport captured by diffusion MRI and DCE-MRI. PloS one, 18:e0282955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [211].Huang JP, Lan H, Xie CJ, Wei CC, Liu Z, Huang ZX, et al. Pramipexole Protects Against Traumatic Brain Injury-Induced Blood-Brain Barrier (BBB) Dysfunction. Neurotoxicity Research: 9. [DOI] [PubMed] [Google Scholar]
  • [212].Li YT, Zhou P, Hu T, Ren J, Xu YP, Qiu Y, et al. (2021). NAAA inhibitor F96 attenuates BBB disruption and secondary injury after traumatic brain injury (TBI). European Journal of Pharmacology, 912:11. [DOI] [PubMed] [Google Scholar]
  • [213].Gold BT, Shao X, Sudduth TL, Jicha GA, Wilcock DM, Seago ER, et al. (2021). Water exchange rate across the blood-brain barrier is associated with CSF amyloid-β 42 in healthy older adults. Alzheimer's & Dementia, 17:2020-2029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [214].Zhai KF, Duan H, Wang W, Zhao SY, Khan GJ, Wang MT, et al. (2021). Ginsenoside Rg1 ameliorates blood-brain barrier disruption and traumatic brain injury via attenuating macrophages derived exosomes miR-21 release. Acta Pharmaceutica Sinica B, 11:3493-3507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [215].Robinson BD, Isbell CL, Melge AR, Lomas AM, Shaji CA, Mohan CG, et al. (2022). Doxycycline prevents blood-brain barrier dysfunction and microvascular hyperpermeability after traumatic brain injury. Scientific Reports, 12:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [216].Zhang EW, Wan XD, Yang LJ, Wang D, Chen ZS, Chen Y, et al. (2020). Omega-3 Polyunsaturated Fatty Acids Alleviate Traumatic Brain Injury by Regulating the Glymphatic Pathway in Mice. Frontiers in Neurology, 11:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [217].Wu C, Lin T, Ding Q, Zhang N, Ou ZT, Cai GY, et al. (2022). Continuous Theta-Burst Stimulation Promotes Paravascular CSF-Interstitial Fluid Exchange through Regulation of Aquaporin-4 Polarization in APP/PS1 Mice. Mediators of Inflammation, 2022:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [218].Taoka T, Masutani Y, Kawai H, Nakane T, Matsuoka K, Yasuno F, et al. (2017). Evaluation of glymphatic system activity with the diffusion MR technique: diffusion tensor image analysis along the perivascular space (DTI-ALPS) in Alzheimer's disease cases. Japanese Journal of Radiology, 35:172-178. [DOI] [PubMed] [Google Scholar]
  • [219].Ma XX, Li SH, Li CM, Wang R, Chen M, Chen HB, et al. (2021). Diffusion Tensor Imaging Along the Perivascular Space Index in Different Stages of Parkinson's Disease. Frontiers in Aging Neuroscience, 13:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [220].Ruan X, Huang X, Li Y, Li E, Li M, Wei X (2022). Diffusion Tensor Imaging Analysis Along the Perivascular Space Index in Primary Parkinson's Disease Patients With and Without Freezing of Gait. Neuroscience, 506:51-57. [DOI] [PubMed] [Google Scholar]
  • [221].Shen T, Yue YM, Ba F, He TG, Tang XC, Hu XY, et al. (2022). Diffusion along perivascular spaces as marker for impairment of glymphatic system in Parkinson's disease. Npj Parkinsons Disease, 8:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [222].Romay M, Ma FY, Hernandez GE, Salvador J, Mirkov S, Batra A, et al. (2022). Notch3 Deficiency Impairs Vascular Smooth Muscle Cell Contractility And Glymphatic Function In The Brain. Circulation Research, 131:2. [Google Scholar]
  • [223].Li M, Kitamura A, Beverley J, Koudelka J, Duncombe J, Lennen R, et al. (2021). Impaired Glymphatic Function and Pulsation Alterations in a Mouse Model of Vascular Cognitive Impairment. Front Aging Neurosci, 13:788519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [224].Nakashima T, Sone M, Teranishi M, Yoshida T, Terasaki H, Kondo M, et al. (2012). A perspective from magnetic resonance imaging findings of the inner ear: relationships among cerebrospinal, ocular and inner ear fluids. Auris Nasus Larynx, 39:345-355. [DOI] [PubMed] [Google Scholar]
  • [225].Wang X, Lou N, Eberhardt A, Yang Y, Kusk P, Xu Q, et al. (2020). An ocular glymphatic clearance system removes β-amyloid from the rodent eye. Science translational medicine, 12:eaaw3210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [226].Uddin N, Rutar M (2022). Ocular Lymphatic and Glymphatic Systems: Implications for Retinal Health and Disease. International Journal of Molecular Sciences, 23:10139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [227].Thrane VR, Hynnekleiv L, Wang X, Thrane AS, Krohn J, Nedergaard M (2021). Twists and turns of ocular glymphatic clearance-new study reveals surprising findings in glaucoma. Acta ophthalmologica, 99:e283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [228].Schwartz N, Chalasani MLS, Li TM, Feng Z, Shipman WD, Lu TT (2019). Lymphatic function in autoimmune diseases. Front Immunol, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Aging and Disease are provided here courtesy of JKL International LLC

RESOURCES