Abstract
Abstract
The meninges serve as critical barriers that maintain immune homeostasis in the central nervous system (CNS) and play vital roles in immune surveillance and defense. Traditionally, the brain has been regarded as an “immune-privileged” organ owing to the absence of conventional lymphatic vessels. However, the rediscovery of meningeal lymphatic vessels (MLVs) has revealed a mechanism for the directional transport of cerebrospinal fluid (CSF) to the deep cervical lymph nodes (dCLNs), demonstrating that the brain possesses a distinct fluid communication pathway with the peripheral system that is independent of blood circulation. Additionally, the identification of the glymphatic system has revealed a perivascular mechanism for solute exchange between the CSF and brain parenchyma, primarily mediated by the astrocytic water channel protein aquaporin-4 (AQP4). These discoveries have significantly expanded our understanding of brain fluid dynamics and CNS homeostasis. This review provides a comprehensive overview of the structure, regulation, and function of MLVs and the glymphatic system, which together constitute lymphatic system of the brain. We also discuss recent evidence, particularly conflicting perspectives, on the role of meningeal immunity in various central nervous system (CNS) disorders, such as multiple sclerosis, Parkinson’s disease, and epilepsy. Furthermore, we explore the therapeutic potential of targeting the brain lymphatic system to treat these conditions. Given their critical roles in CNS homeostasis, MLVs and the glymphatic system have emerged as promising therapeutic targets, potentially offering novel treatment strategies for currently incurable neurological diseases.
Graphical Abstract
The figure illustrates the mechanisms and modulators of the cerebral lymphatic system, including recent insights and influencing factors (left); its involvement in six neurological disorders (middle); and associated therapeutic strategies (right)
Keywords: Meningeal lymphatic system, Glymphatic system, Aquaporin-4 (AQP4), The Central Nervous System (CNS) diseases, Neurolymphatic Therapy
Introduction
The brain, the most complex and functionally advanced component of the nervous system, regulates vital human activities. Historically, the brain has been regarded as an “immune-privileged” organ owing to the absence of conventional lymphatic vessels composed of lymphatic endothelial cells. However, Wilhelm His and Gustav Schwalbe first identified a connection between the CSF and the lymphatic system when Berlin Blue was injected into the central nervous system (CNS) of dogs and observed that a network of dural lymphatics facilitated the drainage of interstitial fluid (ISF) from the CNS into peripheral lymph nodes. In the twenty first century, the presence of dural lymphatics has been confirmed in both rodents and humans (Ahn et al. 2019; Louveau et al. 2015), establishing that the brain microenvironment communicates with the peripheral lymphatic system via CSF drainage into the dCLNs. Additionally, recent studies have identified a glymphatic system within the brain responsible for nutrient transport and metabolic waste clearance through a paravascular pathway primarily regulated by the astrocytic water channel protein AQP4 (Iliff et al. 2012). Collectively, these systems contribute to the maintenance of intracerebral homeostasis (Louveau 2018).
The onset and progression of CNS disorders are intricately linked to the maintenance of intracerebral homeostasis. The glymphatic system and meningeal lymphatics play crucial roles in various neurological diseases, including multiple sclerosis (MS; Brosnan & Raine 2013; Maggi et al. 2018), Parkinson’s disease (PD; Ding et al. 2021), cerebral small vessel disease (CSVD; Esposito et al. 2019), epilepsy (Feldman et al. 2018), major depressive disorder (MDD; Roomruangwong et al. 2017), and traumatic brain injury (TBI; Bolte et al. 2020; Piantino et al. 2022). Dysfunction of the glymphatic system and meningeal lymphatics contributes to disease pathogenesis by facilitating the accumulation of neurotoxic molecules, disrupting brain tissue integrity, and modulating immune and inflammatory responses. However, the impact of these systems is not necessarily unidirectional. For instance, in MS, meningeal lymphatics exacerbate neuroinflammation (Hsu et al. 2019; Yang et al. 2024), while on the other hand, they exert protective effects by influencing oligodendrocytes and astrocytes, thereby improving the disease prognosis (Louveau et al. 2018). Moreover, the glymphatic system and meningeal lymphatics are implicated in secondary brain injury in various CNS disorders, particularly in pathological processes such as tau protein aggregation and cerebral edema (Jessen et al. 2015; Mestre et al. 2020). However, conflicting findings regarding the role of these systems in cerebral edema require further investigation. Thus, targeting the cerebral lymphatic system may be a promising therapeutic strategy for treating CNS disorders.
This review aims to explore the physiological functions of meningeal lymphatics in intracerebral homeostasis, the role of the lymphatic system in ISF–CSF exchange, and the implications of these systems in the pathophysiology and therapeutic approaches for various CNS disorders.
Structure and Function of Meningeal Lymphatic Vessels
Meningeal lymphatic vessels express all classical lymphatic endothelial cell markers, including vascular endothelial growth factor receptor 3 (VEGFR3), prospero homeobox protein 1 (Prox1), podoplanin (gp38), lymphatic vessel endothelial hyaluronan receptor 1 (LYVE-1), CD31, and C-C motif chemokine ligand 21 (CCL21; Louveau et al. 2015). Based on their anatomical location, MLVs are classified into the dorsal intracranial surface and ventral base (Hu et al. 2020), each exhibiting distinct structural characteristics and drainage functions.
Ventral basal MLVs are extensively branched, exhibit distinct lymphatic endothelial features such as valves, and demonstrate a clear structural distinction between capillaries and collecting lymphatics, making them particularly suitable for CSF absorption (Hu et al. 2020). Furthermore, their proximity to the subarachnoid space, particularly in areas lacking arachnoid separation at the cribriform plate, facilitates direct CSF drainage. With ongoing research, the pathways involved in MLV-mediated fluid drainage have been elucidated. CSF is primarily produced by the choroid plexus epithelium and flows through the ventricles and subarachnoid space (Spector et al. 2015). The CSF then exchanges with ISF through the PVS of the penetrating arteries and is ultimately drained by arachnoid granulations and MLVs. Macromolecules in the subarachnoid space are transported to dCLNs and superficial cervical lymph nodes via MLVs (Hu et al. 2020).
Emerging evidence suggests that different meningeal layers exhibit varying degrees of involvement in autoimmune processes in the CNS (Merlini et al. 2022). Specifically, effector T cell infiltration and activation predominantly occur in the leptomeninges, whereas the dura mater appears to be relatively excluded from direct CNS autoimmune involvement (Merlini et al. 2022). This is demonstrated in experimental autoimmune encephalomyelitis models, in which myelin basic protein (MBP) injection in mice leads to intense inflammation in the leptomeninges, characterized by substantial infiltration of effector T cells and myeloid cells (Merlini et al. 2022). In contrast, the dura mater exhibits a minimal inflammatory response, likely due to a lack of intrinsic antigen-presenting capacity (Merlini et al. 2022). The dura mater plays a crucial role in meningeal immunity by facilitating CSF drainage and metabolite clearance and modulating central immune cell dynamics.
MLVs, characterized by relatively loose endothelial junctions, function in concert with CNS immune cells to establish a tightly regulated immuno-lymphatic interface (Hu et al. 2020; Mogensen et al. 2021). Vascular endothelial growth factor C (VEGFC) enhances CSF drainage into dCLNs and induces MLVs remodeling by promoting lymphangiogenesis and upregulating neuroprotective signaling pathways, as revealed by single-nucleus RNA sequencing of brain cells (Boisserand et al. 2024). VEGFC administration increases MLVs diameter and density, leading to improved CSF lymphatic drainage, suppression of pro-inflammatory microglial responses, and enhancement of neurotrophic signaling (Boisserand et al. 2024). Notably, VEGFC preconditioning elevates the levels of brain-derived neurotrophic factor, a key regulator of neuronal differentiation, maturation, and synaptic plasticity, which are critical for CNS development and neuroprotection following injury or disease (Boisserand et al. 2024). Conversely, genetic deletion of VEGFC or VEGFR3, pharmacological inhibition of VEGFR signaling via tyrosine kinase inhibitors (e.g., sunitinib), or the use of VEGFC/D traps results in impaired meningeal lymphatic development and reduced CSF drainage into dCLNs (Antila et al. 2017). These findings suggest that VEGFC-mediated modulation of MLVs function has therapeutic potential for neurological disorders, including enhanced clearance of pathological proteins, such as glutamate, amyloid-beta (Aβ), and tau proteins, immune cells, and inflammatory mediators, thereby preventing their pathological accumulation in the CNS.
Glymphatic System: CSF–ISF Communication
The traditional theory of CSF homeostasis presents several challenges. For instance, studies have demonstrated that labeled substances with different molecular weights, such as albumin (69 kDa) and polyethylene glycol (900 kDa), are cleared from the brain within nearly identical timeframes, despite a fivefold difference in their diffusion coefficients (Cserr et al. 1981). The rapid exchange of ISF and CSF within the brain cannot be adequately explained by the conventional diffusion theory, suggesting the presence of a distinct metabolite clearance mechanism unique to the brain that differs from that in other tissues and organs. Based on this hypothesis, Iliff et al. first proposed the existence of a CNS lymphatic-like system in a murine model. Their findings revealed a fluid transport pathway in the CNS that parallels the peripheral lymphatic system (Iliff et al. 2012).
This system facilitates efficient CSF–ISF exchange through the paravascular pathway. The CSF enters this system via the PVS surrounding the choroidal arteries (Iliff et al. 2012). As the CSF follows the branching of blood vessels, it crosses the astrocytic end-feet via AQP4 channels and enters the PVS, where it contributes to nutrient delivery and metabolic waste clearance (Hannocks et al. 2018). After mixing with ISF and undergoing molecular exchange within the brain parenchyma, CSF exits the brain through multiple drainage routes, including MLVs (Kress et al. 2014) (Fig. 1).
Fig. 1.
Mechanisms of CSF–ISF communication and the role of AQP4 involved. Bottom, schematic diagram of a coronal brain section showing the lymphatic pathway along the cerebral vessels; Top, schematic showing the case of CSF–ISF communication in the case of AQP4 polarization and depolarization, respectively. Right, the CSF communicates with the ISF via a paravascular pathway that allows cerebrospinal fluid from the subarachnoid space to enter the lymphatic system via the PVS. Waste products are drained from the ISF within the parenchyma into the CSF, further cleared by the drainage of functional MLVs. The blue dashed box shows the structure of AQP4, a water channel protein composed of six helical transmembrane domains and two highly conserved regions (Asn-Pro-Ala) that form narrow hemipores. Left, mislocalization or depolarization of AQP4 fails to support CSF–ISF exchange, contributing to glymphatic system dysfunction
AQP4 is a water channel protein isoform exclusively expressed in astrocytes and ependymal cells of the ventricular lining. It shows a polarized distribution on over 60% of the astrocytic end-feet surrounding the blood vessels (Nielsen et al. 1993). Under physiological conditions, AQP4 exhibits polarized localization, which is crucial for maintaining the efficiency of glymphatic clearance (Simon et al. 2022). It is predominantly positioned at the plasma membrane of perivascular astrocytic end-feet, facing the PVS, and anchored by the dystrophin-associated protein complex (DAPC). This configuration enables selective water transport and reduces the hydrodynamic resistance of the plasma membrane, facilitating fluid movement from the perivascular space into the ISF and supporting the transport of small molecules and ions (Jessen et al. 2015). Additionally, advanced imaging techniques, such as large-volume three-dimensional scanning electron microscopy and optoelectronic microscopy, reveal gaps in astrocytic coverage of penetrating cortical arterioles in mice. These structural discontinuities permit direct communication between the vasculature and the subcellular structures of adjacent cells, allowing the passage of macromolecular compounds (Zhang et al. 2024a, b) (Fig. 1). In contrast, under pathological conditions, such as neuroinflammation, trauma, and aging, AQP4 depolarization is associated with glymphatic dysfunction.
A growing body of research indicates that AQP4 not only provides structural support to astrocytic and ependymal cell membranes but also serves as a crucial regulator of fluid dynamics within the brain (Ho & Yang 2024). It facilitates fluid exchange between various compartments, including the vasculature, interstitial space, intracellular space, and CSF (Ho & Yang 2024). Moreover, AQP4 plays a significant role in CSF production and circulation. Disruptions in AQP4 localization or polarization at astrocytic end-feet can impair glymphatic drainage, potentially contributing to hydrocephalus and neurodegenerative conditions such as Alzheimer’s disease and idiopathic normal pressure hydrocephalus (Reeves et al. 2020; Simon et al. 2022). Overall, glymphatic system functionality is highly dependent on AQP4 expression and its precise localization within astrocytic end-feet (Palazzo et al. 2019).
In summary, the glymphatic system mediates ISF–CSF exchange, whereas MLVs facilitate CSF drainage to the dCLNs, thereby maintaining intracranial homeostasis. Together, these components form the cerebral lymphatic system, which serves as a crucial interface between the CNS and the peripheral immune system.
Glymphatic Circulation: Influencing Factors
Glymphatic function is critically dependent on CSF circulation and the bulk flow of ISF within the brain, both of which are intrinsic components of the glymphatic system (Chong et al. 2022; Jiang-Xie et al. 2024). Arterial pulsation serves as the primary driving force of CSF circulation, and the rhythmic expansion and contraction of arteries during the cardiac cycle (systole and diastole) dictate the overall direction of CSF flow (Enzmann and Pelc 1991). Notably, the peak velocity of CSF movement coincides with arterial pulsation, providing microscopic evidence that arterial pulsation is a key regulator of CSF inflow (Mestre et al. 2018).
Studies utilizing in vivo two-photon microscopy have demonstrated that unilateral internal carotid artery ligation reduces arterial pulsation and subsequently slows the rate of perivascular CSF–ISF exchange (Mestre et al. 2018). Conversely, systemic administration of the adrenergic agonist dobutamine significantly enhances penetrating artery pulsation, leading to a concomitant increase in the rate of CSF–ISF exchange (Mestre et al. 2018). In addition to arterial pulsation, changes in arterial diameter and fluctuations in venous blood volume resulting from the systolic-diastolic modulation of vascular smooth muscle cells can markedly influence intracerebroventricular CSF flow and metabolite clearance (Mateo et al. 2017). Respiratory movements also contribute to CSF dynamics by modulating intracranial vascular function. The absence of valves in the intracranial venous system renders it highly sensitive to central venous pressure, which fluctuates in response to changes in intrathoracic pressure during the respiratory cycle (Wilson 2016). Additionally, respiratory-induced alterations in the arterial wall are shown to affect CSF circulation (Mestre et al. 2018).
Using two-photon microimaging, Xie et al. observed that the volume fraction of the interstitial space was significantly higher during natural sleep and anesthesia than during wakefulness, suggesting that the glymphatic function is markedly enhanced during sleep (Xie et al. 2013). In awake mice, intracerebroventricular injection of an adrenergic receptor antagonist inhibited central norepinephrine signaling, leading to an increase in ISF volume and enhanced glymphatic activity (Xie et al. 2013). This finding highlights that norepinephrine is a critical neuromodulator of the glymphatic function, linking arousal states to lymphatic circulation. However, recent research measuring the clearance and movement of fluorescent molecules in the brains of male mice has revealed that brain clearance is significantly reduced during sleep and anesthesia, contradicting previous beliefs (Miao et al. 2024). During non-rapid eye movement (NREM) sleep, a coordinated interplay between electrophysiological oscillations, hemodynamic changes, and CSF flow dynamics has been observed, suggesting that neuronal activity, cerebral blood flow, and CSF circulation are functionally interdependent during sleep (Fultz et al. 2019) (Fig. 2). Studies using transcranial optogenetic stimulation to generate synthetic neural network oscillations demonstrated a significant enhancement of CSF perfusion into the ISF, further supporting the hypothesis that neural activity regulates ISF bulk flow (Jiang-Xie et al. 2024).
Fig. 2.
The illustration highlights cellular and material changes within the brain parenchyma and MLVs in both healthy (left side) and diseased or aged states (right side). Pathological changes in the brain parenchyma include AQP4 depolarization, impaired demyelination, Aβ and tau protein deposition, oxidative stress, vascular leakage, loss of dopaminergic neurons, and release of toxic molecules
Aging is strongly correlated with the accumulation of metabolic waste products in the brain, which may be attributable to age-related impairments in the glymphatic function. Ma et al. labeled CSF in the lateral ventricles with an infrared tracer and measured its accumulation in the cervical lymph nodes over a fixed period. They found that 18-month-old mice exhibited significantly slower CSF outflow kinetics compared to 2-month-old mice (Ma et al. 2017). Furthermore, aged mice display markedly reduced clearance of Aβ injected into the brain parenchyma, confirming age-associated glymphatic dysfunction (Kress et al. 2014). Aging is also accompanied by structural and functional changes in cerebral arterioles, including decreased pulsatility of small intracortical arterioles (Kress et al. 2014) and increased vessel tortuosity, both of which increase the resistance to CSF flow within the PVS (Bennett et al. 2024). Additionally, aging is associated with diminished perivascular AQP4 expression and widespread loss of AQP4 polarization, which further impairs glymphatic clearance (Iliff et al. 2012). In aged mice, ligation of the dCLNs exacerbates severe glymphatic dysfunction, accompanied by increased depolarization of astrocyte-activated AQP4, neuronal damage, induced neuroinflammation, behavioral deficits, and memory impairment (Zhu et al. 2024a, b) (Fig. 2).
Given the central role of glymphatic circulation in waste clearance and brain homeostasis, the modulation of its function has emerged as a promising therapeutic strategy for neurodegenerative diseases and other CNS disorders. In the following sections, we explore current research efforts aimed at targeting the glymphatic system for disease prevention and treatment.
The Role of the Cerebral Lymphatic System in CNS Diseases
Parkinson’s Disease
Parkinson’s disease (PD), the second most prevalent neurodegenerative disorder after Alzheimer’s disease, is pathologically characterized by the early degeneration of dopaminergic neurons in the substantia nigra pars compacta, aberrant aggregation and propagation of α-synuclein (αSyn) and tau proteins, and formation of Lewy bodies (Kalia & Lang 2015). Emerging evidence suggests that the cerebral lymphatic system influences PD progression by modulating the accumulation of pathological proteins and neuroinflammatory responses in the brain.
The glymphatic system facilitates metabolic waste clearance, including misfolded αSyn, via CSF–ISF exchange (Iliff et al. 2012). AQP4, a critical water channel in the glymphatic system, plays a pivotal role in PD pathogenesis (Ding et al. 2021). Reduced expression or depolarization of AQP4 has been observed in both patients with PD and A53T-αSyn-overexpressing mice, impairing glymphatic function and diminishing αSyn clearance (Lapshina & Ekimova 2024). This dysfunction is correlated with αSyn accumulation in the brain parenchyma, which is a hallmark of progressive PD. Notably, AQP4-knockout mice exhibit αSyn aggregation in the dorsal striatum, cerebral cortex, and substantia nigra pars compacta, accompanied by dopaminergic neuronal loss, likely because of impaired macromolecular clearance by the glymphatic system (Lapshina & Ekimova 2024).
The glymphatic system also modulates PD progression through inflammatory pathways. In the 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)-induced PD model, AQP4-deficient mice exhibited exacerbate neuroinflammation and dopaminergic neuron (DN) loss compared with wild-type mice (Chi et al. 2011) (Fig. 2). Mechanistically, interferon-gamma (IFNγ) interacts with leucine-rich repeat kinase 2 (LRRK2) to induce neuroinflammation, whereas LRRK2 R1441G-mediated AQP4 phosphorylation leads to depolarization, disruption of IFNγ clearance, and exacerbation of neurodegeneration (Huang et al. 2024). CD4⁺ T cells upregulate major histocompatibility complex class II (MHC-II) expression in CNS myeloid cells during αSyn pathology, and genetic ablation of CD4⁺ T cells ameliorates αSyn-induced neurodegeneration (Fig. 3). Notably, AQP4 dysfunction in PD models reduces CD4⁺CD25⁺ regulatory T cells, thereby amplifying microglial inflammation (Jiang et al. 2024). Furthermore, AQP4 single nucleotide polymorphisms (SNPs) rs162009 and rs2075575 are linked to susceptibility to PD and may serve as genetic markers for glymphatic impairment and cognitive decline in patients with PD (Sun et al. 2023; Jiang et al. 2023).
Fig. 3.
The scheme depicts alterations in the MLVs and cervical lymph nodes in healthy states (left side of the illustration) and central nervous system disorders (right side of the illustration). Pathological changes in MLVs are characterized by augmented MHC-II expression, immune cell proliferation, reduced tau and Aβ clearance, and abnormal secretion of immune molecules. In cervical lymph nodes, these changes include lymph node atrophy, lymphocyte depletion, and abnormal antibody secretion
Meningeal lymphatics have a dual effect on PD progression. Impaired meningeal lymphatic drainage exacerbates pathological protein accumulation (Da Mesquita et al. 2018). Fibrillar αSyn activates pro-inflammatory nuclear factor kappa B (NF-κB) signaling via toll-like receptor (TLR)-dependent and -independent mechanisms or binds to receptors for advanced glycation end products (RAGE) on microglia, driving cytokine production (Bearoff et al. 2023). Meningeal lymphatics mitigate the αSyn burden by draining it to the dCLNs, whereas cervical lymphatic ligation promotes αSyn accumulation and DN loss. Additionally, pathological αSyn deposition disrupts meningeal lymphatic function by inducing meningeal inflammation or compromising endothelial tight junctions (Ding et al. 2021; Zou et al. 2019) (Fig. 3). Paradoxically, some clinical studies reported unchanged αSyn levels in the brains of patients with PD despite meningeal lymphatic dysfunction, potentially due to compensatory mechanisms. First, AQP4-dependent perivascular glymphatic pathways may sustain αSyn clearance via CSF–ISF exchange, particularly when AQP4 polarization remains partially intact (Lapshina & Ekimova 2024). Second, meningeal lymphatic dysfunction may enhance microglial phagocytosis or upregulate molecular chaperones (e.g., heat shock proteins) to counteract αSyn aggregation (Da Mesquita et al. 2021). Finally, Genetic heterogeneity, including AQP4 SNPs (rs162009 and rs2075575), may further obscure αSyn pathology due to inter-individual variability in glymphatic efficiency (Jiang et al. 2023; Sun et al. 2023). These observations underscore the need to evaluate meningeal lymphatic contribution to αSyn metabolism within the broader neuroimmune microenvironment (Fig. 3).
Conversely, meningeal lymphatics exacerbate neuroinflammation by facilitating the infiltration of peripheral immune cells (Williams et al. 2021). CD4⁺ T cells migrate along the meningeal lymphatics via the CCR7-CCL21 signaling axis to the dCLNs (Leser et al. 2025), where they acquire CNS-specific phenotypes. Upon reentering the CNS through the blood–brain barrier (BBB) or choroid plexus–CSF pathways (Cervellati et al. 2020; Salvador et al. 2024), these cells secrete pro-inflammatory cytokines (e.g., TNF-α and IL-17), activate microglia, and perpetuate a neurotoxic feedback loop (Fig. 2). This cascade amplifies neuroinflammation, edema, apoptosis, and neuronal death, highlighting the complex interplay between meningeal lymphatic function and immune dysregulation in PD (Da Mesquita et al. 2018).
Multiple Sclerosis
Multiple sclerosis (MS) is a chronic autoimmune disorder characterized by progressive neurological damage. Its pathogenesis involves motor and cognitive impairment resulting from demyelination, neuronal injury, and astrocyte activation within the CNS (Filippi et al. 2018) (Fig. 2). These pathological changes are believed to be driven by a complex interplay between BBB disruption, microglial activation, localized and diffuse inflammation, and the release of pro-inflammatory cytokines (Magliozzi et al. 2006). Emerging evidence suggests that the cerebral lymphatic system influences MS progression by modulating immune cell trafficking, priming inflammatory responses, and altering the function of oligodendrocytes.
Studies have demonstrated glymphatic system dysfunction in patients with MS. In acute MS lesions, retraction of glial cells delineating the PVS occurs, accompanied by astrocyte damage (Brosnan & Raine 2013). Patients with MS exhibited significantly reduced diffusivity along the PVS, as measured by diffusion tensor imaging analysis along the perivascular space (DTI-ALPS), and dilation of the perispinal veins, both of which are indicative of glymphatic system impairment. In addition, a decreased DTI-ALPS score is associated with greater clinical disability and longer disease duration. These findings suggest that glymphatic system dysfunction may contribute to MS progression (Brosnan & Raine 2013; Carotenuto et al. 2022; Bateman & Bateman 2024).
Inflammatory mediators, such as reactive oxygen species (ROS), produced in the meninges of patients with MS, enhance microglial proliferation and activation, leading to the acquisition of an infiltrative phenotype and subsequent CNS damage (Yang et al. 2024; Hsu et al. 2019). Several studies indicated that the glymphatic system plays a crucial role in microglial clearance, implicating it in MS-related neuroinflammation (Yang et al. 2024; Hsu et al. 2019). In secondary progressive MS, elevated levels of immune cells and inflammatory molecules, including tumor necrosis factor (TNF) and interferon (IFN), within the meninges and CSF contribute to microglial activation and demyelination (Gardner et al. 2013). Peripheral immune-mediated destruction of CNS structures is a key pathogenic mechanism underlying autoimmune diseases (Carotenuto et al. 2022). Impaired lymphatic fluid flow and reduced drainage at the regional level result in the accumulation of inflammatory cells and neurotoxic elements, exacerbating cortical demyelination (Carotenuto et al. 2022). Toxic molecules or metabolic byproducts also play crucial roles in MS pathogenesis. Microbial dysbiosis facilitates the infiltration of bacterial toxins into the brain parenchyma. Among these, ε-toxin (ETX), produced by Clostridium perfringens, has been found to be elevated in the brain tissue of patients with MS. ETX binds to the cerebral vasculature and induces oligodendrocyte death (Cekanaviciute et al. 2018; Berer et al. 2017). Lymphatic system dysfunction impairs the clearance of toxic molecules and metabolites from the ventricles and deep gray matter, contributing to gray matter pathology (De Meo et al. 2021) (Fig. 2).
MLVs have a dual effect on MS progression. MLVs exacerbate MS by promoting inflammatory responses. MLVs facilitate the migration of meningeal T cells to dCLNs via the CCR7-CCL21 signaling pathway (Louveau et al. 2018). In an experimental autoimmune encephalomyelitis mouse model, cribriform plate lymphatic vessels developed, whereas classical inflammation-induced lymphangiogenesis was absent in meningeal lymphatics (Hsu et al. 2019; Louveau et al. 2018). Increased T-cell density has been observed within and around the meningeal lymphatic system, accompanied by enhanced drainage of meningeal immune cells to downstream lymph nodes (Hsu et al. 2019; Louveau et al. 2018). MLVs ablation achieved through nasal lymphatic disruption using Visudyne, lymphatic vessel ligation at deep cervical lymph node input, or inhibition of VEGFR3 with MAZ51 attenuates CD4⁺ T cell infiltration, reduces spinal cord demyelination, and subsequently improves experimental autoimmune encephalomyelitis prognosis (Louveau et al. 2018; Hsu et al. 2019). This protective effect may stem from MAZ51-mediated inhibition of lymphangiogenesis in the cribriform plate, thereby limiting the recruitment of pathogenic CNS antigen-specific T cells (Hsu et al. 2019). Furthermore, impaired lymphatic drainage diminishes T cell-antigen-presenting cell (APC) interactions and disrupts T cell phenotype differentiation, leading to altered cytokine and chemokine signaling (Louveau et al. 2018). Although MLVs play a fundamental role in immune cell drainage, their influence on T cell differentiation and immune activation appears to be more pronounced (Fig. 3).
Conversely, MLVs may exert neuroprotective effects in MS by modulating the function of oligodendrocytes and astrocytes. Oligodendrocytes, which are responsible for myelin formation in the CNS, are depleted in MS lesions (Brosnan & Raine 2013). In a well-established cuprizone-induced demyelination model, meningeal lymphatic ablation led to altered glial gene expression, a reduction in mature oligodendrocyte populations, impaired myelin regeneration, extensive axonal loss, and increased apoptosis, all of which had significant implications for demyelinating disorders such as MS (Das Neves et al. 2024). Additionally, MLVs dysfunction has been associated with astrocytic overexpression of Cst3, suggesting increased oxidative phosphorylation stress prior to demyelination, which may contribute to subsequent demyelination (Das Neves et al. 2024) (Fig. 2).
Epilepsy
Epilepsy is one of the most prevalent neurological disorders, exerting a multitude of effects on the cerebral nerves, cognition, and psychology (Fisher et al. 2014). It is characterized by its spontaneity and enduring predisposition to generate epileptic seizures. However, 75% of patients have responded inadequately to treatment (Fisher et al. 2014).
Most patients with epilepsy have significant GS impairment. In children, a significant increase in PVS counts is observed in various forms of epilepsy, such as idiopathic generalized epilepsy, suggesting that impaired GS function leads to slowed clearance of interstitial fluid and fluid retention in the PVS (Liu et al. 2020a, b; Salimeen et al. 2021). Notably, contradictory experimental results regarding PVS changes, ranging from no significant alterations to increased counts, have been reported in patients with febrile seizures and epilepsy (Salimeen et al. 2021; Spalice et al. 2020). Additionally, some studies have revealed that PVS morphology and size in patients with typical Rolandic epilepsy are comparable to those observed in migraine patients, indicating a likely congenital origin rather than epileptogenic changes (Boxerman et al. 2007). In adults, glymphatic malformations and changes in perivascular lymphatic dysfunction appear to be more prominent, with many epilepsy patients exhibiting marked asymmetrical PVS distribution, particularly ipsilateral to the seizure onset zone. This has been observed in focal epilepsy (Feldman et al. 2018), post-TBI epilepsy (Hlauschek et al. 2024), temporal lobe epilepsy (Lee et al. 2022), and frontal lobe epilepsy (Zhou et al. 2024). The DTI-ALPS index was significantly reduced in temporal lobe epilepsy patients with hippocampal sclerosis compared to healthy controls. However, the index showed no significant association with clinical parameters, such as age at epilepsy onset, disease duration, or anti-seizure medication dosage (Lee et al. 2022). Moreover, in mouse models, status epilepticus (SE) was associated with substantial glymphatic impairment, which positively correlated with the degree of post-SE cerebral edema (Liu et al. 2021a, b). Alleviation of cerebral edema was shown to facilitate earlier restoration of glymphatic function following SE (Liu et al. 2021a, b).
The mechanisms by which glymphatic system dysfunction affects epilepsy include impaired solute clearance, which leads to ion homeostasis disruption, accumulation of toxic substances, and cerebral edema (Fig. 2). During epileptic seizures, the reduced excretion of K⁺ and Ca⁺ ions in the ISF affects the electrophysiological activity of the neurons. AQP4 interacts with molecules such as glutamate transporter 1 to modulate the CSF–ISF osmotic pressure and ion exchange. Dysregulation and mislocalization of AQP4 in patients with epilepsy affect ion exchange, thereby exacerbating epileptic symptoms (Rabinovitch et al. 2019). The deposition of phosphorylated tau and serum proteins leads to axonal damage and neuronal hyperexcitability (Noé & Marchi 2019). Glutamate serves as a precursor for the reduction of inhibitory neurotransmitter synthesis and is thus closely linked to the onset of epilepsy (Hlauschek et al. 2024). Glymphatic system-induced phosphorylation impairment of tau and glutamate deposition also increases neuronal excitability (Hlauschek et al. 2024). Post-epileptic cerebral edema, accompanied by glymphatic system dysfunction and enhanced AQP4 polarization, can be alleviated by glibenclamide treatment or Trpm4 knockout. This intervention significantly facilitates early recovery of impaired glymphatic function and ISF drainage following SE, as evidenced by reduced PT deposition, decreased neuronal degeneration, and improved cognitive function (Liu et al. 2021a, b). Notably, among patients with focal epilepsy, those with poor responses to anti-seizure medications (ASMs) exhibited more severe glymphatic system dysfunction than good responders, suggesting that the glymphatic system may enhance ASM responsiveness and contribute to treatment outcomes in patients with focal epilepsy (Kim et al. 2024).
On the other hand, MLVs play a significant role in regulating immune cells. Leukocyte infiltration in the PVS induces inflammation in the central nervous system after seizures, leading to neuronal damage. Inhibition of the inflammatory receptor CCR2 alleviates the deleterious effects of status epilepticus (Alemán-Ruiz et al. 2023). Impaired MLVs drainage allows brain-derived antigens to reach the periphery, activating cytotoxic CD8⁺ T cells to mediate neuroinflammation, thereby causing neuronal damage (Noé & Marchi 2019). CD8⁺ T-cell-driven limbic encephalitis leads to temporal lobe epilepsy, suggesting that MLVs may play a crucial role in epilepsy development via CD8⁺ T cell regulation (Pitsch et al. 2021).
Major Depressive Disorder
Major depressive disorder (MDD) is a globally prevalent mood disorder characterized by cognitive and behavioral impairments, anhedonia, and suicidal ideation (Monroe & Harkness 2022). It manifests as a wide range of functional disturbances, including dysregulation of sleep, cognition, appetite, and mood, making it one of the most pressing mental health challenges (Monroe & Harkness 2022).
Sleep disturbances are a core symptom of depression (Stickley et al. 2019), and their alleviation is closely associated with the remission of depressive symptoms (Fang et al. 2019). The glymphatic system, which is primarily active during sleep (Xie et al. 2013), has been implicated in the causal relationship with MDD pathophysiology (Nedergaard & Goldman 2020; Zhang et al. 2024a, b). Global blood oxygen level-dependent (gBOLD) signals derived from resting-state functional magnetic resonance imaging (rs-fMRI) provided a noninvasive method for quantifying glymphatic dynamics (Liu et al. 2018). Compared to healthy controls, patients with MDD exhibited weaker gBOLD–CSF coupling, which may reflect glymphatic dysfunction (Zhang et al. 2024a, b).
Astrocytes, a key component of the glymphatic system, are reduced in both number and activity in MDD (Wallensten et al. 2021). The glymphatic system influences the pathogenesis of MDD through mechanisms involving oxidative stress, monoaminergic dysregulation, and neuroinflammation (Fig. 2). The glymphatic system facilitates the clearance of reactive oxygen species (ROS) into the peripheral lymphatic system, which serves as the primary pathway for ROS elimination (Harrison et al. 2020). Given that the brain accounts for 20% of total oxygen consumption, it is particularly vulnerable to ROS-induced damage (Bhatt et al. 2020). In MDD, increased BBB permeability exacerbates neuroinflammation by enhancing the leakage of astrocyte-derived extracellular vesicles (Wallensten et al. 2021). Impaired glymphatic clearance of ROS can activate microglia, leading to the production of pro-inflammatory cytokines that induce apoptosis, thereby contributing to the pathophysiology of MDD (Roomruangwong et al. 2017). While low ROS levels promote neuronal growth, excessive oxidative stress triggers neuroinflammatory damage, apoptosis, and ultimately, MDD progression (Bhatt et al. 2020). Chronic unpredictable mild stress (CUMS)-induced glymphatic dysfunction may act as a bridge between monoamine dysregulation and neuroinflammation, thereby playing a central role in the pathogenesis of depression (Roomruangwong et al. 2018). The glymphatic function is closely linked to monoamine neurotransmitter levels (Liu et al. 2020a, b). During sleep, reductions in monoamine concentrations enhance glymphatic clearance, whereas during wakefulness, increased monoamine release, particularly norepinephrine (NE), inhibits glymphatic function (Liu et al. 2020a, b). Recent studies have shown that CUMS induces excessive NE release, which subsequently disrupts AQP4 polarization in astrocytes, thereby impairing glymphatic function (Yao et al. 2023b, a). This impairment contributes to oxidative stress and neuroinflammation, ultimately leading to the development of depression-like symptoms in affected individuals.
Sex-dependent differences in stress sensitivity are associated with meningeal lymphatic dysfunction in mice. Studies have demonstrated that subchronic variable stress selectively impairs meningeal lymphatic function in female mice, whereas enhancement of meningeal lymphatic function reduces stress susceptibility and mitigates stress-induced alterations in the medial prefrontal cortex (mPFC) and ventral tegmental area (VTA; Dai et al. 2022). Notably, both the mPFC and VTA are critical brain regions involved in emotion regulation (Tye et al. 2013). These findings suggest that meningeal lymphatics play a pivotal role in the sex-dependent modulation of stress susceptibility and resilience.
Cerebral Small Vessel Disease
Diseases such as aging, hypertension, and diabetes influence the pathogenesis of cerebral small vessel disease (CSVD) via the cerebral lymphatic system. Aging is considered a key factor in the regulation of cerebral lymphatic function (Tian et al. 2022). Age-related dysfunction in the glymphatic system and MLVs exacerbates cognitive decline and promotes neurodegenerative pathologies, further contributing to CSVD development (Tian et al. 2022). Type 2 diabetes mellitus, a prevalent metabolic disorder in middle-aged and elderly populations, is a significant risk factor for CSVD (Staals et al. 2014). Compared to non-diabetic rats, rats with type 2 diabetes mellitus exhibit a threefold slower clearance of CSF tracers, as demonstrated by Gd-DTPA contrast-enhanced imaging and fluorescent tracer studies. Impairment of the glymphatic system-induced inhibition of hippocampal and hypothalamic ISF clearance is directly associated with cognitive deficits (Jiang et al. 2017). Clustering analyses further revealed that type 2 diabetes mellitus significantly increased the perivascular space, likely due to diabetes-induced microvascular and macrovascular damage, which in turn heightened CSVD susceptibility (Jiang et al. 2017; Doubal et al. 2010) (Fig. 2).
The formation of cerebral edema in CSVD is influenced by the function of the glymphatic system. Stroke is the leading cause of long-term disability and the second leading cause of mortality worldwide (Steinmetz et al. 2024). Ischemic stroke (IS) accounts for 63% of all stroke subtypes globally and is the primary contributor to non-fatal and fatal burdens in adults (Steinmetz et al. 2024). IS is frequently accompanied by cerebral edema, which exerts mechanical stress on brain tissues and capillaries, exacerbating damage (Simard et al. 2009). Recent studies have shown that the glymphatic system provides a pathway for lymphatic exchange between the CSF and mesenchymal cells, facilitating the rapid influx of CSF into brain tissue and serving as the earliest source of Na⁺ and fluid in cerebral edema (Mestre et al. 2020). Moreover, increased PVS flow is observed during stroke, potentially due to the release of K⁺ and other vasoactive molecules, along with endothelial nitric oxide depletion, leading to significant contraction of parenchymal and pial arterioles and the generation of a pressure gradient. This mechanism may play a crucial role in the development of cerebral edema (Mestre et al. 2020). AQP4 exacerbates post-ischemic cerebral edema by promoting CSF transport to the brain parenchyma. AQP4-knockout mice (Mestre et al. 2020) or treatment with AQP4 inhibitors, such as TGN-020 (Hirt et al. 2017; Sun et al. 2022), significantly reduced CSF tracer influx into the PVS and ISF within 15 min of ischemic onset, thereby mitigating cerebral edema, reducing mortality, and restoring motor function (Mestre et al. 2020; Hirt et al. 2017; Sun et al. 2022). However, some studies found that IS also induced glymphatic system dysfunction. Three hours post-IS onset, reduced CSF inflow and impaired clearance of low-molecular-weight compounds were linked to decreased arterial pulsatility and PVS compression, resulting in glymphatic system impairment (Gaberel et al. 2014). Notably, glymphatic system function was restored 24 h post-IS onset, coinciding with the reperfusion of the middle cerebral artery (Gaberel et al. 2014). Additionally, AQP4 reactivity and depolarization increased at 3 and 14 days after diffuse microinfarcts, disrupting cerebral water homeostasis and impairing edema clearance (Wang et al. 2012). Thus, although glymphatic system dysfunction may limit the extent of AQP4-mediated cerebral edema, its role in stroke-related pathophysiology remains complex.
Glymphatic system impairment also occurs in the acute phase following subarachnoid hemorrhage (SAH), leading to the accumulation of metabolic waste and neurotoxic factors (De Rooij et al. 2013). Fibrinolytic plasminogen activators enhance lymphatic function by dissolving blood clots in the PVS, but their effect on prognosis remains unclear (Gaberel et al. 2014; Goulay et al. 2017). Interestingly, AQP4-knockout models demonstrated increased cerebral edema and neuroinflammation (Liu et al. 2021a, b). However, conflicting findings have been reported regarding the role of AQP4 in SAH-induced cerebral edema. AQP4 expression was significantly elevated in the posterior cerebral arteries following SAH, whereas venous AQP4 levels remain unchanged, potentially due to astrocyte activation (Liu et al. 2021a, b; Chen et al. 2016). Higher AQP4 expression levels in patients with SAH were correlated with more severe cerebral edema (Chen et al. 2016). Notably, atorvastatin-mediated AQP4 inhibition reduced the severity of cerebral edema, Caspase-3 expression, and mortality in SAH models, suggesting potential therapeutic benefits in early brain injury following SAH (Chen et al. 2016). These findings indicate that AQP4’s involvement in SAH-associated cerebral edema is multifaceted and necessitates further investigation.
MLVs play a pivotal role in various CSVDs through immune regulation and drainage. In IS, MLVs mediate immune responses in the nervous system, promoting neuroinflammation by activating macrophages via the VEGFC/VEGFR3 signaling pathway, ultimately contributing to cerebral infarction (Esposito et al. 2019). Additionally, thrombospondin-1 (THBS1) is significantly elevated in the CSF of patients, and THBS1-CD47 ligand–receptor interactions were shown to induce apoptosis in meningeal lymphatic endothelial cells, leading to MLVs dysfunction and poor prognosis in SAH (Wang et al. 2023). In intracerebral hemorrhage (ICH), increased meningeal lymphangiogenesis is observed in the later stages, persisting for up to 60 days. Impaired MLVs function hampers hematoma clearance from the brain parenchyma, whereas MLVs enhancement improves neurological outcomes and accelerates hematoma resolution (Tsai et al. 2022). Furthermore, cilostazol-induced enhancement of meningeal lymphatic drainage following ICH promoted erythrocyte clearance and facilitated hematoma regression (Kimura et al. 2014).
Overall, the interplay between glymphatic system dysfunction and CSVD pathogenesis remains incompletely understood, with variability in experimental timelines, therapeutic interventions, and individual AQP4 responses potentially contributing to the conflicting findings (Hirt et al. 2017; Wang et al. 2012; Gaberel et al. 2014; De Rooij et al. 2013; Chen et al. 2016). Conversely, MLVs consistently exhibit varying degrees of dysfunction, while their enhancement is generally associated with improved CSVD outcomes (Esposito et al. 2019; Wang et al. 2023).
Traumatic Brain Injury
Traumatic brain injury (TBI) is a form of organic brain tissue damage caused by external collisions, blunt trauma, or penetrating injuries (Bolte et al. 2020). Enhancing basic and clinical research on TBI and improving therapeutic interventions are crucial for reducing the high mortality and disability rates associated with severe TBI (Bolte et al. 2020).
Meningeal lymphatic dysfunction is closely related to the development of TBI, and the two influence each other. Intracranial pressure rises significantly within 2 h of TBI onset, leading to dysfunction in key CSF absorption sites within the subarachnoid space, suggesting meningeal lymphatic impairment (Bolte et al. 2020). This dysfunction persists for at least one month after injury, exacerbating neuroinflammation and cognitive deficits (Willis et al. 2020). TBI induces both morphological and functional changes in MLVs, including increased expression of lymphatic vessel endothelial hyaluronan receptor 1 (Lyve-1), a significant increase in the number of endothelial loops and buds within the lymphatic system, and an overall trend toward increased lymphatic vessel diameter (Bolte et al. 2020). Impaired lymphatic drainage following TBI results in the accumulation of Aβ, cellular debris, and other neurotoxic substances, perpetuating immune activation in post-injury brain tissue (Wu et al. 2024). This sustained neuroinflammatory response contributes to the prolonged pathology observed after TBI. Furthermore, pre-existing meningeal lymphatic dysfunction exacerbates neuroinflammation and cognitive impairment following trauma (Bolte et al. 2020). For instance, compared with mice with TBI alone, those with pre-existing meningeal lymphatic injury exhibited significant upregulation of complement-related gene expression, increased glial fibrillary acidic protein immunoreactivity, and elevated ionized calcium-binding adapter molecule 1 immunoreactivity within 24 h after TBI, indicating exacerbation of glial cell damage (Bolte et al. 2020). These findings suggest that promoting meningeal lymphatic recovery after TBI may be an effective strategy for mitigating neuroinflammation and improving neurological outcomes in TBI patients.
The glymphatic system is closely associated with both the initial and secondary pathophysiological processes of TBI. TBI disrupts the function of key regulatory structures, such as the hypothalamus, pineal gland, and brainstem, which impairs glymphatic function by affecting sleep regulation (Piantino et al. 2022). Additionally, post-TBI elevation of blood fibrinogen levels allows fibrinogen to cross the BBB, where it can be deposited and converted into fibrin at the vascular–astrocyte end-foot interface. This process disrupts the paravascular pathways of the glymphatic system, leading to the continuous accumulation of metabolic waste products, ultimately contributing to neurodegeneration (Sulimai & Lominadze 2020). Neurodegenerative features commonly observed in patients with TBI include neurofibrillary tangles composed of tau protein aggregates and Aβ amyloid plaques (Iwata et al. 2002; Zanier et al. 2018) (Fig. 2). Glymphatic circulation was significantly impaired following TBI, with reductions detectable as early as one day post-injury and persisting for at least 28 days post-injury. In severe cases, CSF clearance can decrease by up to 60% (Jessen et al. 2015). Notably, in a moderate TBI animal model, tau protein aggregates were found within the perivenous interstitial space of large veins, aligned with the established paravascular pathways of the glymphatic system. These findings suggest that tau accumulation is closely linked to glymphatic clearance deficits (Jessen et al. 2015). Similarly, studies in veterans with blast-related mild TBI (mTBI) have identified an increased burden of MRI-visible perivascular spaces in the frontal cortex, which is identified as a potential neuroimaging marker of perilymphatic dysfunction. A greater PVS volume was correlated with poorer cognitive performance (Braun et al. 2024; Jung et al. 2024). These findings further support the notion that glymphatic dysfunction, which leads to impaired tau clearance, plays a crucial role in secondary neuronal injury and neurofibrillary tangle formation following TBI.
Cerebral edema is a critical sequela of TBI, and glymphatic dysfunction is implicated as a key contributor to early stage cytotoxic edema (Eide & Ringstad 2024). TBI-induced sympathetic activation and increased noradrenergic tone are identified as major factors disrupting glymphatic function (Hussain et al. 2023). Driven by CSF influx and osmotic pressure gradients, outflow pathways from the venous interstitial space are inhibited by osmotic forces that lead to ISF retention and the development of brain edema (Wu et al. 2021).
Modulating the Cerebral Lymphatic System for Neurological Disorders
Direct Targeting of AQP4
Specifically, targeting AQP4 offers a direct approach to modulating the glymphatic system. In AQP4 antibody-positive patients, immunosuppression has reduced the risk of relapse (Paul et al. 2023). The PPA regimen (prazosin, propranolol, and atipamezole) may alleviate post-traumatic brain edema by blocking adrenergic receptors and enhancing lymphatic drainage (Hussain et al. 2023). Furthermore, AQP4 neutralizing antibodies have been shown to significantly reduce cerebral edema within 24 h and mitigate hippocampal neuronal loss and memory deficits by day 21 (Li et al. 2023). Moreover, AQP4 localization at the blood–spinal cord barrier has been shown to be regulated by calmodulin, which binds to its C-terminal domain during hypoxia-induced swelling, exacerbating CNS edema (Kitchen. et al. 2020; Salman et al. 2017). Inhibiting calmodulin or using AQP4 inhibitors (e.g., TGN-020) has been demonstrated to effectively reduce edema (Vandebroek and Yasui 2020; Sun et al. 2022). Trifluoperazine, a calmodulin inhibitor, has been shown to prevent AQP4 localization to the barrier, reduce edema, and promote functional recovery (Xing et al. 2023).
Indirect Modulation of Glymphatic System
Astrocytic modulation influences the glymphatic system. Knockdown of the transient receptor potential melastatin 4 (TRPM4) has been shown to reduce astrocytosis and microglial proliferation post-status epilepticus, alleviating cerebral edema and neuronal damage (Rolin et al. 2024; Wang et al. 2025a, b). As a result, targeted inhibition of astrocyte proliferation has been shown to increase AQP4 expression and reduce inflammation (Nakano-Kobayashi et al. 2023). Antisense oligonucleotides (ASOs) are promising for the treatment for Huntington’s disease (HD), where HTT gene dysfunction disrupts astrocyte and glymphatic system functions via NF-κB signaling (Dhuri et al. 2020; Träger et al. 2014). Intravenous mesenchymal stem cell (MSC) therapy has been shown to restore glymphatic system function by enhancing ASO distribution in BACHD mice while suppressing mutant HTT (mHTT; Wang et al. 2021; Yen et al. 2020). Consequently, omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been shown to inhibit astrocyte activation, while preserving AQP4 polarization, improving glymphatic system function, and alleviating depressive and cognitive symptoms (Ren et al. 2017). Interstitial space expansion during slow-wave sleep has been demonstrated to correlate with δ-wave activity (Xie et al. 2013). Similarly, transcranial direct current stimulation (tDCS) has been shown to enhance δ-wave activity through the IP3/Ca2⁺ pathway, increasing the CSF–ISF exchange (Smith et al. 2020; Wang et al. 2025a, b).
Regulation via Proteins and Cytokines
Specifically, Per2 expression is inversely correlated with AQP4 polarization. Melatonin maintains AQP4 polarity via circadian regulation (Yao et al. 2023; Cruz-Sanabria et al. 2023). AQP4 polarity also depends on the dystrophin complex (Si et al. 2024), which is degraded by matrix metalloproteinase-9 (MMP-9; Huang 2018). MMP-9 inhibition (GM6001) has been demonstrated to restore perivascular AQP4 and β-dystroglycan levels (Yuan et al. 2024; Zhu et al. 2024a, b). Notably, ultrasound therapy, including continuous theta burst stimulation (cTBS), has been shown to enhance the glymphatic system by upregulating VEGFC, which promotes MLVs dilation and drainage (Yulug et al. 2017; Liu et al. 2017; Song et al. 2020). Similarly, 40 Hz light flicker, acting via the adenosine-A2AR pathway, has been demonstrated to improve lymphatic inflow and AQP4 distribution (Sun et al. 2024; Li et al. 2020; Poderoso et al. 2019).
Modulating MLVs in Neurological Disorders
VEGFC is critical to MLVs development and has therapeutic potential for neurological disorders (Boisserand et al. 2024; Xu et al. 2023). VEGFC-VEGFR3 signaling has been demonstrated to enhance MLVs function, alleviating cognitive impairment, neuroinflammation, and neuronal damage in sepsis-associated encephalopathy (Deng et al. 2015; Leppänen et al. 2013). Impaired MLVs drainage worsens microglial activation in Alzheimer’s disease, thereby reducing the efficacy of immunotherapy. However, VEGFC delivery has been shown to improve Aβ clearance, suggesting that MLV-targeted therapy can enhance AD immunotherapy (Da Mesquita et al. 2021). More importantly, BO nanomicelles (BO-MSs) have been demonstrated to improve MLV permeability and enhance Aβ42 oligomer clearance within minutes (Wu et al. 2023; Ye et al. 2024). Similarly, near-infrared light has been demonstrated to enhance the metabolic homeostasis of mitochondria in meningeal lymphatic endothelial cells, promote cell adhesion and growth, improve the connectivity of meningeal lymphatic endothelial cells, and favor lymphatic transport (Foo et al. 2020; Wang et al. 2024). In a mouse model of chronically elevated intracranial pressure, elevated intracranial pressure has been demonstrated to limit CSF acquisition by MLVs and affect MLV development, maintenance, and functional drainage, whereas stimulation of Piezo1 ion channels in mice enhances MLV coverage, drainage, and cerebral CSF perfusion (Coste et al. 2010; Li et al. 2022; Warren et al. 2024). Some drugs ultimately act on both the glymphatic system and MLVs. In 5XFAD mice (a model of early dementia), focused ultrasound combined with microbubbles (FUS-MB) inhibited microglia and astrocyte proliferation or activation and increased lymphoid clearance of Aβ through meningeal arterioles and veins to the CSF space, and into the dCLN via meningeal lymphatic vessels. This treatment significantly improved working memory, showing potential for preventing cognitive impairment (Daini et al. 2021; Lee et al. 2020; Yang et al. 2023).
Controversies and Discussion
While GS primarily suppresses neuroinflammation in PD (Chi et al. 2011; Huang et al. 2024; Jiang et al. 2024; Sun et al. 2023), MLVs exacerbate neuroinflammation in PD and MS (Leser et al. 2025; Cervellati et al. 2020; Salvador et al. 2024; Da Mesquita et al. 2018; Louveau et al. 2018; Hsu et al. 2019). On the other hand, some studies have found that damage to MLVs in PD patients exacerbates pathological protein deposition (Da Mesquita et al. 2018; Bearoff et al. 2023; Ding et al. 2021; Zou et al. 2019). However, a subset of these studies reported that αSyn levels remained unchanged (Lapshina & Ekimova 2024; Jiang et al. 2023; Sun et al. 2023).
Conflicting findings regarding epilepsy are primarily observed in pediatric patients. Studies have indicated that the number of PVS associated with different types of epilepsy may increase, decrease, or remain relatively unchanged (Liu et al. 2020a, b; Salimeen et al. 2021; Spalice et al. 2020; Boxerman et al. 2007). Many of these studies relied solely on MRI scans to demonstrate alterations in PVS (Liu et al. 2020a, b, 2021a; Spalice et al. 2020; Boxerman et al. 2007; Feldman et al. 2018; Hlauschek et al. 2024). Disruption of GS structures may lead to compensatory restoration of lymphatic drainage through mechanisms such as MLVs, which could explain the divergence from mainstream findings in studies on childhood epilepsy (Spalice et al. 2020). Furthermore, most of these studies had sample sizes of approximately 60 participants (Liu et al. 2020a, b, 2021a; Salimeen et al. 2021; Feldman et al. 2018; Hlauschek et al. 2024). In contrast, in the study in which contradictory results emerged, the sample size was only 30 cases (Spalice et al. 2020). Therefore, a more comprehensive retrospective study is needed to validate these findings.
In CSVD, GS may exacerbate brain edema after IS. However, other studies suggest that severe GS dysfunction associated with IS may instead impair water clearance. This apparent contradiction may be attributable to temporal variations in cerebral lymphatic system function. Early activity of the cerebral lymphatic system primarily facilitates the development of brain edema (Mestre et al. 2020; Hirt et al. 2017; Sun et al. 2022), whereas subsequent dysfunction later in life exacerbates impaired water clearance and further contributes to neurological impairment (Doubal et al. 2010; Wang et al. 2012; Gaberel et al. 2014). Further experimental studies are required to validate this hypothesis. Additionally, various experimental models were employed in these studies, including the Middle Cerebral Artery Occlusion Model (MCAO; Mestre et al. 2020; Hirt et al. 2017; Sun et al. 2022), minimally invasive injection of gadolinium chelate into the cisterna magna (Gaberel et al. 2014), acute stroke patients (Doubal et al. 2010), a mouse model of multiple diffuse microinfarcts induced by unilateral internal carotid artery injection of cholesterol crystals (Wang et al. 2012), among others.
Research on AQP4 in SAH has yielded mixed results. Inhibition of AQP4 with atorvastatin in a rabbit model ameliorated early brain damage after SAH (Chen et al. 2016). AQP4 knockdown in a rat model exacerbated brain edema and neurological deficits (Liu et al. 2021a, b). In contrast, measurement of lymphatic system perfusion using MRI after minimally invasive injection of gadolinium chelates into the large pool using a mouse model indicated that AQP4 may not be directly associated with brain edema or increased intracranial pressure (Gaberel et al. 2014). We hypothesize that this may be related to animal model reactivity, experimental methods for AQP4 inhibition, and methods to detect AQP4.
Based on the above studies, we propose the following hypotheses. First, in most diseases, MLVs and GS function interdependently, i.e., they jointly mediate anti-inflammatory and drainage functions. However, in certain diseases, the two systems may exhibit opposing roles (e.g., PD, MS). We hypothesize that the MLVs and GS may have compensatory roles for each other and cannot be studied in complete isolation. For instance, findings show that impairment of AQP4 in the aging brain is accompanied by increased AQP4 expression, which has been viewed as a physiological compensation, and it is possible that a similar compensatory mechanism may apply to MLVs damage (Lapshina & Ekimova 2024; Jiang et al. 2023; Sun et al. 2023). Other factors such as genetic heterogeneity, including AQP4 SNPs (rs162009 and rs2075575) (Jiang et al. 2023; Sun et al. 2023), enhanced phagocytosis by microglia, or upregulation of molecular chaperones (e.g., heat shock proteins) (Da Mesquita et al. 2021) may compensate for impaired MLV-mediated clearance via GS mechanisms. Most of the above studies target GS or MLVs individually. However, given their possible interdependence, the respective roles of GS and MLVs in drainage and neuroinflammation should not be evaluated in isolation. Therefore, researchers should simultaneously investigate the inflammatory and clearance-related roles of both GS and MLVs in the same model to better elucidate their complex mechanisms of action within the CNS. Second, differences in experimental methods may cause errors in the results. In the above studies, the main methods for observing the lymphatic system were MRI and DTI-ALPS. The exclusive use of MRI can reflect structural changes and cannot directly assess GS dysfunction (Liu et al. 2020a, b, 2021a; Boxerman et al. 2007; Spalice et al. 2020; Feldman et al. 2018; Hlauschek et al. 2024), which in turn contributes to errors. Furthermore, potential compensatory interactions between MLVs and GS may further confound the findings. Therefore, we advocate the combined use of MRI and DTI-ALPS to demonstrate the structural and functional aspects of GS changes and their impact on disease. Third, modeling methods and animal models are different. These studies applied different modeling methods, animal models, or patient samples, and, as a result, their findings were inevitably inconsistent, considering that different species may exhibit varied physiological responses (Mestre et al. 2020; Hirt et al. 2017; Sun et al. 2022; Doubal et al. 2010; Wang et al. 2012; Chen et al. 2016; Liu et al. 2021a, b; Gaberel et al. 2014). Among them, in the experiments conducted using drugs, unknown drug effects may have confounded experimental outcomes (Huang et al. 2024; Liu et al. 2021a, b; Kim et al. 2024; Hirt et al. 2017; Sun et al. 2022; Gaberel et al. 2014; Goulay et al. 2017; Chen et al. 2016). Additionally, other sources of error include small sample sizes, lack of patient follow-up, and variability in observation time points, all of which contribute to inconsistencies.
Conclusion and Outlook
The cerebral lymphatic system, comprising the MLVs and the glymphatic system, plays a crucial role in metabolic waste clearance and the regulation of neuroinflammation. It has become a major focus in neurological disease research. In this review, we summarize recent physiological and functional findings regarding the cerebral lymphatic system, including factors influencing the glymphatic system, pathological alterations, and the pathogenic mechanisms of various neurological disorders. We also discuss the therapeutic targets and approaches aimed at modulating this system, along with conflicting findings across studies.
Many central nervous system (CNS) disorders are closely associated with cerebral lymphatic dysfunction. The cerebral lymphatic system consistently supports recovery in MDD and TBI, with the glymphatic system aiding in the clearance of reactive oxygen species (ROS) in MDD and tau protein clearance and edema resolution in TBI. No significant contradictions were observed between these conditions. However, research in PD, MS, CSVD, and epilepsy has yielded contradictory findings and unexplained discrepancies under different conditions. We analyze contradictory studies on these diseases and find that the reasons for these discrepancies include studying MLVs and GS in isolation while ignoring the compensatory interactions between them; using different research models and observational methods within the same disease; small sample sizes; and lack of patient follow-up. Addressing these uncertainties is critical for optimizing CSF–ISF exchange and developing precise clinical guidelines for therapeutic interventions. These findings also underscore the complex and multifactorial nature of CNS pathogenesis, emphasizing the need for evidence-based research to elucidate disease mechanisms. Despite these challenges, there is no doubt that meningeal lymphatic vessels and glymphatic system represent promising therapeutic targets, offering potential breakthroughs in treating currently incurable neurodegenerative diseases.
Several cerebral lymphatic system-targeted therapies have been developed in recent years, with promising results. However, disease-specific therapeutic strategies, particularly for MS and TBI, remain limited. Notably, we found that the cerebral lymphatic system shares common pathogenic mechanisms with different neurological diseases, suggesting that similar experimental models and therapeutic targets may be applicable to multiple conditions. In addition, research into the glymphatic system and MLVs has progressed unevenly across different disorders. For example, studies on MLVs involvement in epilepsy are scarce compared to those on the glymphatic system. Future investigations should explore the involvement of MLVs in epilepsy by leveraging insights from lymphatic research on other diseases. This review not only synthesizes recent advances in the structure, function, and regulatory factors of the cerebral lymphatic system but also highlights its involvement in major CNS disorders. These findings provide key insights for the development of more effective therapeutic strategies, ultimately paving the way for improved clinical outcomes in neurological diseases.
Acknowledgements
We would like to thank the members of the Institutes of Brain Diseases (Nanfang Hospital, Southern Medical University) for their suggestions on the paper.
Abbreviations
- CNS
Central nervous system
- AQP4
Aquaporin-4
- CSF
Cerebrospinal fluid
- ISF
Interstitial fluid
- PVS
Perivascular space
- BBB
Blood–brain barrier
- MLVs
Meningeal lymphatic vessels
- VEGFR3
Vascular endothelial growth factor receptor 3
- dCLNs
The deep cervical lymph nodes
- Aβ
Amyloid-beta
- IFN
Interferon
- TNF
Tumor necrosis factor
- ROS
Reactive oxygen species
- DTI-ALPS
Diffusion tensor imaging analysis along the perivascular space
- αSyn
α-Synuclein
- THBS1
Increased levels of thrombospondin 1
- VEGFC
Vascular endothelial growth factor C
- MS
Multiple sclerosis
- PD
Parkinson's disease
- CSVD
Cerebral small vessel disease
- MRI
Magnetic resonance imaging
- IS
Ischemic stroke
- SAH
Subarachnoid haemorrhage
- MDD
Major Depressive Disorder
- TBI
Traumatic brain injury
Author Contributions
JXP and YQF: conceived, drafted, edited, rigorously reviewed the manuscript and drafted illustrations; PY: critically revised and integrated the final manuscript; WFL: reviewed and edited the manuscript; ZFL: contributed significantly to the revision; AS: Supervised and gave valuable input to the manuscript; JSD: reviewed and revised the manuscript; FM: gave valuable input to the manuscript; FL: responsible for revision of illustrations; STQ: Supervised and gave valuable input to the manuscript; YB: Supervised and gave valuable input to the manuscript.
Funding
This work was supported by the Clinical Research Program of Nanfang Hospital, Southern Medical University (Grant Numbers 2023CR023, 2018CR029, 2019CR020); and the Guangdong Basic and Applied Basic Research Foundation (Grant Number 2024A1515030069).
Data Availability
No datasets were generated or analysed during the current study.
Declarations
Conflict of interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Ethical Approval
Not applicable.
Illustration Attribution
All illustrations presented in this manuscript were originally created by the authors using Adobe Illustrator 2015 (version 19.0.0). All graphical elements (including schematics, cellular diagrams) were developed de novo based on theoretical frameworks in this study.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Jingxi Pan, Yinqi Fu, and Peng Yang are the co-first authors contributed equally to this work.
Contributor Information
Songtao Qi, Email: qisongtaonfyy@126.com.
Yun Bao, Email: maybe@smu.edu.cn.
References
- Ahn JH, Cho H, Kim J-H, Kim SH, Ham J-S, Park I, Suh SH, Hong SP, Song J-H, Hong Y-K, Jeong Y, Park S-H, Koh GY (2019) Meningeal lymphatic vessels at the skull base drain cerebrospinal fluid. Nature 572(7767):62–66. 10.1038/s41586-019-1419-5 [DOI] [PubMed] [Google Scholar]
- Alemán-Ruiz C, Wang W, Dingledine R, Varvel NH (2023) Pharmacological inhibition of the inflammatory receptor CCR2 relieves the early deleterious consequences of status epilepticus. Sci Rep 13(1):5651. 10.1038/s41598-023-32752-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Antila S, Karaman S, Nurmi H, Airavaara M, Voutilainen MH, Mathivet T, Chilov D, Li Z, Koppinen T, Park J-H, Fang S, Aspelund A, Saarma M, Eichmann A, Thomas J-L, Alitalo K (2017) Development and plasticity of meningeal lymphatic vessels. J Exp Med 214(12):3645–3667. 10.1084/jem.20170391 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bateman GA, Bateman AR (2024) The dilated veins surrounding the cord in multiple sclerosis suggest elevated pressure and obstruction of the glymphatic system. Neuroimage 286:120517. 10.1016/j.neuroimage.2024.120517 [DOI] [PubMed] [Google Scholar]
- Bearoff F, Dhavale D, Kotzbauer P, Kortagere S (2023) Aggregated alpha-synuclein transcriptionally activates pro-inflammatory canonical and non-canonical NF-κB signaling pathways in peripheral monocytic cells. Mol Immunol 154:1–10. 10.1016/j.molimm.2022.12.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bennett HC, Zhang Q, Wu Y, Manjila SB, Chon U, Shin D, Vanselow DJ, Pi H-J, Drew PJ, Kim Y (2024) Aging drives cerebrovascular network remodeling and functional changes in the mouse brain. Nat Commun 15(1):6398. 10.1038/s41467-024-50559-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berer K, Gerdes LA, Cekanaviciute E, Jia X, Xiao L, Xia Z, Liu C, Klotz L, Stauffer U, Baranzini SE, Kümpfel T, Hohlfeld R, Krishnamoorthy G, Wekerle H (2017) Gut microbiota from multiple sclerosis patients enables spontaneous autoimmune encephalomyelitis in mice. Proc Natl Acad Sci U S A 114(40):10719–10724. 10.1073/pnas.1711233114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bhatt S, Nagappa AN, Patil CR (2020) Role of oxidative stress in depression. Drug Discov Today 25(7):1270–1276. 10.1016/j.drudis.2020.05.001 [DOI] [PubMed] [Google Scholar]
- Boisserand LSB, Geraldo LH, Bouchart J, El Kamouh M-R, Lee S, Sanganahalli BG, Spajer M, Zhang S, Lee S, Parent M, Xue Y, Skarica M, Yin X, Guegan J, Boyé K, Saceanu Leser F, Jacob L, Poulet M, Li M et al (2024) VEGF-C prophylaxis favors lymphatic drainage and modulates neuroinflammation in a stroke model. J Exp Med 221(4):e20221983. 10.1084/jem.20221983 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bolte AC, Dutta AB, Hurt ME, Smirnov I, Kovacs MA, McKee CA, Ennerfelt HE, Shapiro D, Nguyen BH, Frost EL, Lammert CR, Kipnis J, Lukens JR (2020) Meningeal lymphatic dysfunction exacerbates traumatic brain injury pathogenesis. Nat Commun 11(1):4524. 10.1038/s41467-020-18113-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boxerman JL, Hawash K, Bali B, Clarke T, Rogg J, Pal DK (2007) Is rolandic epilepsy associated with abnormal findings on cranial MRI? Epilepsy Res 75(2–3):180–185. 10.1016/j.eplepsyres.2007.06.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Braun M, Sevao M, Keil SA, Gino E, Wang MX, Lee J, Haveliwala MA, Klein E, Agarwal S, Pedersen T, Rhodes CH, Jansson D, Cook D, Peskind E, Perl DP, Piantino J, Schindler AG, Iliff JJ (2024) Macroscopic changes in aquaporin-4 underlie blast traumatic brain injury-related impairment in glymphatic function. Brain 147(6):2214–2229. 10.1093/brain/awae065 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brosnan CF, Raine CS (2013) The astrocyte in multiple sclerosis revisited. Glia 61(4):453–465. 10.1002/glia.22443 [DOI] [PubMed] [Google Scholar]
- Carotenuto A, Cacciaguerra L, Pagani E, Preziosa P, Filippi M, Rocca MA (2022) Glymphatic system impairment in multiple sclerosis: relation with brain damage and disability. Brain 145(8):2785–2795. 10.1093/brain/awab454 [DOI] [PubMed] [Google Scholar]
- Cekanaviciute E, Pröbstel AK, Thomann A, Runia TF, Casaccia P, Katz Sand I, Crabtree E, Singh S, Morrissey J, Barba P, Gomez R, Knight R, Mazmanian S, Graves J, Cree BAC, Zamvil SS, Baranzini SE (2018) Multiple sclerosis-associated changes in the composition and immune functions of spore-forming bacteria. mSystems 3(6):e00083-18. 10.1128/mSystems.00083-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cervellati C, Trentini A, Pecorelli A, Valacchi G (2020) Inflammation in neurological disorders: the thin boundary between brain and periphery. Antioxid Redox Signal 33(3):191–210. 10.1089/ars.2020.8076 [DOI] [PubMed] [Google Scholar]
- Chen J-H, Yang L-K, Chen L, Wang Y-H, Wu Y, Jiang B-J, Zhu J, Li P-P (2016) Atorvastatin ameliorates early brain injury after subarachnoid hemorrhage via inhibition of AQP4 expression in rabbits. Int J Mol Med 37(4):1059–1066. 10.3892/ijmm.2016.2506 [DOI] [PubMed] [Google Scholar]
- Chi Y, Fan Y, He L, Liu W, Wen X, Zhou S, Wang X, Zhang C, Kong H, Sonoda L, Tripathi P, Li CJ, Yu MS, Su C, Hu G (2011) Novel role of aquaporin-4 in CD4+ CD25+ T regulatory cell development and severity of Parkinson’s disease: AQP4 in Treg cell development and severity of PD. Aging Cell 10(3):368–382. 10.1111/j.1474-9726.2011.00677.x [DOI] [PubMed] [Google Scholar]
- Chong PLH, Garic D, Shen MD, Lundgaard I, Schwichtenberg AJ (2022) Sleep, cerebrospinal fluid, and the glymphatic system: a systematic review. Sleep Med Rev 61:101572. 10.1016/j.smrv.2021.101572 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coste B, Mathur J, Schmidt M, Earley TJ, Ranade S, Petrus MJ, Dubin AE, Patapoutian A (2010) Piezo1 and Piezo2 are essential components of distinct mechanically activated cation channels. Science (New York NY) 330(6000):55–60. 10.1126/science.1193270 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cruz-Sanabria F, Carmassi C, Bruno S, Bazzani A, Carli M, Scarselli M, Faraguna U (2023) Melatonin as a chronobiotic with sleep-promoting properties. Curr Neuropharmacol 21(4):951–987. 10.2174/1570159X20666220217152617 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cserr HF, Cooper DN, Suri PK, Patlak CS (1981) Efflux of radiolabeled polyethylene glycols and albumin from rat brain. Am J Physiol-Renal Physiol 240(4):F319–F328. 10.1152/ajprenal.1981.240.4.F319 [DOI] [PubMed] [Google Scholar]
- Dai W, Yang M, Xia P, Xiao C, Huang S, Zhang Z, Cheng X, Li W, Jin J, Zhang J, Wu B, Zhang Y, Wu P, Lin Y, Wu W, Zhao H, Zhang Y, Lin W-J, Ye X (2022) A functional role of meningeal lymphatics in sex difference of stress susceptibility in mice. Nat Commun 13(1):4825. 10.1038/s41467-022-32556-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Daini E, Secco V, Liao W, Zoli M, Vilella A (2021) A regional and cellular analysis of the early intracellular and extracellular accumulation of Aβ in the brain of 5XFAD mice. Neurosci Lett 754:135869. 10.1016/j.neulet.2021.135869 [DOI] [PubMed] [Google Scholar]
- Da Mesquita S, Louveau A, Vaccari A, Smirnov I, Cornelison RC, Kingsmore KM, Contarino C, Onengut-Gumuscu S, Farber E, Raper D, Viar KE, Powell RD, Baker W, Dabhi N, Bai R, Cao R, Hu S, Rich SS, Munson JM et al (2018) Functional aspects of meningeal lymphatics in ageing and Alzheimer’s disease. Nature 560(7717):185–191. 10.1038/s41586-018-0368-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Da Mesquita S, Papadopoulos Z, Dykstra T, Brase L, Farias FG, Wall M, Jiang H, Kodira CD, de Lima KA, Herz J, Louveau A, Goldman DH, Salvador AF, Onengut-Gumuscu S, Farber E, Dabhi N, Kennedy T, Milam MG, Baker W et al (2021) Meningeal lymphatics affect microglia responses and anti-Aβ immunotherapy. Nature 593(7858):255–260. 10.1038/s41586-021-03489-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Das Neves SP, Delivanoglou N, Ren Y, Cucuzza CS, Makuch M, Almeida F, Sanchez G, Barber MJ, Rego S, Schrader R, Faroqi AH, Thomas J-L, McLean PJ, Oliveira TG, Irani SR, Piehl F, Da Mesquita S (2024) Meningeal lymphatic function promotes oligodendrocyte survival and brain myelination. Immunity 57(10):2328-2343.e8. 10.1016/j.immuni.2024.08.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Meo E, Storelli L, Moiola L, Ghezzi A, Veggiotti P, Filippi M, Rocca MA (2021) In vivo gradients of thalamic damage in paediatric multiple sclerosis: a window into pathology. Brain 144(1):186–197. 10.1093/brain/awaa379 [DOI] [PubMed] [Google Scholar]
- de Rooij NK, Greving JP, Rinkel GJ, Frijns CJ (2013) Early prediction of delayed cerebral ischemia after subarachnoid hemorrhage: development and validation of a practical risk chart. Stroke 44(5):1288–1294. 10.1161/STROKEAHA.113.001125 [DOI] [PubMed] [Google Scholar]
- Deng Y, Zhang X, Simons M (2015) Molecular controls of lymphatic VEGFR3 signaling. Arterioscler Thromb Vasc Biol 35(2):421–429. 10.1161/ATVBAHA.114.304881 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dhuri K, Bechtold C, Quijano E, Pham H, Gupta A, Vikram A, Bahal R (2020) Antisense oligonucleotides: an emerging area in drug discovery and development. J Clin Med 9(6):2004. 10.3390/jcm9062004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ding X-B, Wang X-X, Xia D-H, Liu H, Tian H-Y, Fu Y, Chen Y-K, Qin C, Wang J-Q, Xiang Z, Zhang Z-X, Cao Q-C, Wang W, Li J-Y, Wu E, Tang B-S, Ma M-M, Teng J-F, Wang X-J (2021) Impaired meningeal lymphatic drainage in patients with idiopathic Parkinson’s disease. Nat Med 27(3):411–418. 10.1038/s41591-020-01198-1 [DOI] [PubMed] [Google Scholar]
- Doubal FN, MacLullich AMJ, Ferguson KJ, Dennis MS, Wardlaw JM (2010) Enlarged perivascular spaces on MRI are a feature of cerebral small vessel disease. Stroke 41(3):450–454. 10.1161/STROKEAHA.109.564914 [DOI] [PubMed] [Google Scholar]
- Eide PK, Ringstad G (2024) Glymphatic-stagnated edema induced by traumatic brain injury. Trends Pharmacol Sci 45(5):388–390. 10.1016/j.tips.2024.01.005 [DOI] [PubMed] [Google Scholar]
- Enzmann DR, Pelc NJ (1991) Normal flow patterns of intracranial and spinal cerebrospinal fluid defined with phase-contrast cine MR imaging. Radiology 178(2):467–474. 10.1148/radiology.178.2.1987610 [DOI] [PubMed] [Google Scholar]
- Esposito E, Ahn BJ, Shi J, Nakamura Y, Park JH, Mandeville ET, Yu Z, Chan SJ, Desai R, Hayakawa A, Ji X, Lo EH, Hayakawa K (2019) Brain-to-cervical lymph node signaling after stroke. Nat Commun 10(1):5306. 10.1038/s41467-019-13324-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fang H, Tu S, Sheng J, Shao A (2019) Depression in sleep disturbance: a review on a bidirectional relationship, mechanisms and treatment. J Cell Mol Med 23(4):2324–2332. 10.1111/jcmm.14170 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feldman RE, Rutland JW, Fields MC, Marcuse LV, Pawha PS, Delman BN, Balchandani P (2018) Quantification of perivascular spaces at 7 T: a potential MRI biomarker for epilepsy. Seizure 54:11–18. 10.1016/j.seizure.2017.11.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Filippi M, Bar-Or A, Piehl F, Preziosa P, Solari A, Vukusic S, Rocca MA (2018) Multiple sclerosis. Nat Rev Dis Primers 4(1):43. 10.1038/s41572-018-0041-4 [DOI] [PubMed] [Google Scholar]
- Fisher RS, Acevedo C, Arzimanoglou A, Bogacz A, Cross JH, Elger CE, Engel J, Forsgren L, French JA, Glynn M, Hesdorffer DC, Lee BI, Mathern GW, Moshé SL, Perucca E, Scheffer IE, Tomson T, Watanabe M, Wiebe S (2014) ILAE official report: a practical clinical definition of epilepsy. Epilepsia 55(4):475–482. 10.1111/epi.12550 [DOI] [PubMed] [Google Scholar]
- Foo ASC, Soong TW, Yeo TT, Lim K-L (2020) Mitochondrial dysfunction and Parkinson’s disease-near-infrared photobiomodulation as a potential therapeutic strategy. Front Aging Neurosci 12:89. 10.3389/fnagi.2020.00089 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fultz NE, Bonmassar G, Setsompop K, Stickgold RA, Rosen BR, Polimeni JR, Lewis LD (2019) Coupled electrophysiological, hemodynamic, and cerebrospinal fluid oscillations in human sleep. Science 366(6465):628–631. 10.1126/science.aax5440 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gaberel T, Gakuba C, Goulay R, De Lizarrondo SM, Hanouz J-L, Emery E, Touze E, Vivien D, Gauberti M (2014) Impaired glymphatic perfusion after strokes revealed by contrast-enhanced MRI: a new target for fibrinolysis? Stroke 45(10):3092–3096. 10.1161/STROKEAHA.114.006617 [DOI] [PubMed] [Google Scholar]
- Gardner C, Magliozzi R, Durrenberger PF, Howell OW, Rundle J, Reynolds R (2013) Cortical grey matter demyelination can be induced by elevated pro-inflammatory cytokines in the subarachnoid space of MOG-immunized rats. Brain 136(12):3596–3608. 10.1093/brain/awt279 [DOI] [PubMed] [Google Scholar]
- Goulay R, Flament J, Gauberti M, Naveau M, Pasquet N, Gakuba C, Emery E, Hantraye P, Vivien D, Aron-Badin R, Gaberel T (2017) Subarachnoid hemorrhage severely impairs brain parenchymal cerebrospinal fluid circulation in nonhuman primate. Stroke 48(8):2301–2305. 10.1161/STROKEAHA.117.017014 [DOI] [PubMed] [Google Scholar]
- Hannocks M-J, Pizzo ME, Huppert J, Deshpande T, Abbott NJ, Thorne RG, Sorokin L (2018) Molecular characterization of perivascular drainage pathways in the murine brain. J Cereb Blood Flow Metab 38(4):669–686. 10.1177/0271678X17749689 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harrison IF, Ismail O, Machhada A, Colgan N, Ohene Y, Nahavandi P, Ahmed Z, Fisher A, Meftah S, Murray TK, Ottersen OP, Nagelhus EA, O’Neill MJ, Wells JA, Lythgoe MF (2020) Impaired glymphatic function and clearance of tau in an Alzheimer’s disease model. Brain 143(8):2576–2593. 10.1093/brain/awaa179 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hirt L, Fukuda AM, Ambadipudi K, Rashid F, Binder D, Verkman A, Ashwal S, Obenaus A, Badaut J (2017) Improved long-term outcome after transient cerebral ischemia in aquaporin-4 knockout mice. J Cereb Blood Flow Metab 37(1):277–290. 10.1177/0271678X15623290 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hlauschek G, Nicolo J, Sinclair B, Law M, Yasuda CL, Cendes F, Lossius MI, Kwan P, Vivash L (2024) Role of the glymphatic system and perivascular spaces as a potential biomarker for post-stroke epilepsy. Epilepsia Open 9(1):60–76. 10.1002/epi4.12877 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ho KO, Yang S (2024) Quantum sensor settles debate about superconductivity in hydrides. Nature 627(8002):45–46. 10.1038/d41586-024-00423-y [DOI] [PubMed] [Google Scholar]
- Hsu M, Rayasam A, Kijak JA, Choi YH, Harding JS, Marcus SA, Karpus WJ, Sandor M, Fabry Z (2019) Neuroinflammation-induced lymphangiogenesis near the cribriform plate contributes to drainage of CNS-derived antigens and immune cells. Nat Commun 10(1):229. 10.1038/s41467-018-08163-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu X, Deng Q, Ma L, Li Q, Chen Y, Liao Y, Zhou F, Zhang C, Shao L, Feng J, He T, Ning W, Kong Y, Huo Y, He A, Liu B, Zhang J, Adams R, He Y et al (2020) Meningeal lymphatic vessels regulate brain tumor drainage and immunity. Cell Rep 30(3):229–243. 10.1038/s41422-020-0287-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang H (2018) Matrix metalloproteinase-9 (MMP-9) as a cancer biomarker and MMP-9 biosensors: recent advances. Sensors (Basel Switz) 18(10):3249. 10.3390/s18103249 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang H, Lin L, Wu T, Wu C, Zhou L, Li G, Su F, Liang F, Guo W, Chen W, Jiang Q, Guan Y, Li X, Xu P, Zhang Y, Smith W, Pei Z (2024) Phosphorylation of AQP4 by LRRK2 R1441G impairs glymphatic clearance of IFNγ and aggravates dopaminergic neurodegeneration. NPJ Parkinsons Dis 10(1):31. 10.1038/s41531-024-00643-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hussain R, Tithof J, Wang W, Cheetham-West A, Song W, Peng W, Sigurdsson B, Kim D, Sun Q, Peng S, Plá V, Kelley DH, Hirase H, Castorena-Gonzalez JA, Weikop P, Goldman SA, Davis MJ, Nedergaard M (2023) Potentiating glymphatic drainage minimizes post-traumatic cerebral oedema. Nature 623(7989):992–1000. 10.1038/s41586-023-06737-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, Benveniste H, Vates GE, Deane R, Goldman SA, Nagelhus EA, Nedergaard M (2012) A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci Transl Med 4(147):147ra111. 10.1126/scitranslmed.3003748 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iwata A, Chen XH, McIntosh TK, Browne KD, Smith DH (2002) Long-term accumulation of amyloid-beta in axons following brain trauma without persistent upregulation of amyloid precursor protein genes. J Neuropathol Exp Neurol 61(12):1056–1068. 10.1093/jnen/61.12.1056 [DOI] [PubMed] [Google Scholar]
- Jessen NA, Munk ASF, Lundgaard I, Nedergaard M (2015) The glymphatic system: a beginner’s guide. Neurochem Res 40(12):2583–2599. 10.1007/s11064-015-1581-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang Q, Zhang L, Ding G, Davoodi-Bojd E, Li Q, Li L, Sadry N, Nedergaard M, Chopp M, Zhang Z (2017) Impairment of the glymphatic system after diabetes. J Cereb Blood Flow Metab 37(4):1326–1337. 10.1177/0271678X16654702 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang M, Fang Y, Dai S, Si X, Wang Z, Tang J, Gao T, Liu Y, Song Z, Pu J, Zhang B (2023) The effects of AQP4 rs162009 on resting-state brain activity in Parkinson’s disease. CNS Neurosci Ther 29(9):2645–2655. 10.1111/cns.14208 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang W, Liu X, Chen Y, Liu M, Yuan J, Nie M, Fan Y, Wu D, Qian Y, Sha Z, Dong S, Wu C, Liu T, Huang J, Zhang J, Gao C, Jiang R (2024) CD4+ CD11b+ T cells infiltrate and aggravate the traumatic brain injury depending on brain-to-cervical lymph node signaling. CNS Neurosci Ther 30(3):e14673. 10.1111/cns.14673 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang-Xie L-F, Drieu A, Bhasiin K, Quintero D, Smirnov I, Kipnis J (2024) Neuronal dynamics direct cerebrospinal fluid perfusion and brain clearance. Nature 627(8002):157–164. 10.1038/s41586-024-07108-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jung LB, Wiegand TLT, Tuz-Zahra F, Tripodis Y, Iliff JJ, Piantino J, Arciniega H, Kim CL, Pankatz L, Bouix S, Lin AP, Alosco ML, Daneshvar DH, Mez J, Sepehrband F, Rathi Y, Pasternak O, Coleman MJ, Adler CH et al (2024) Repetitive head impacts and perivascular space volume in former American football players. JAMA Netw Open 7(8):e2428687. 10.1001/jamanetworkopen.2024.28687 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalia LV, Lang AE (2015) Parkinson’s disease. Lancet (Lond Engl) 386(9996):896–912. 10.1016/S0140-6736(14)61393-3 [DOI] [PubMed] [Google Scholar]
- Kim S, Kim SE, Lee DA, Lee H, Park KM (2024) Anti-seizure medication response and the glymphatic system in patients with focal epilepsy. Eur J Neurol 31(1):e16097. 10.1111/ene.16097 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kimura T, Hamazaki TS, Sugaya M, Fukuda S, Chan T, Tamura-Nakano M, Sato S, Okochi H (2014) Cilostazol improves lymphatic function by inducing proliferation and stabilization of lymphatic endothelial cells. J Dermatol Sci 74(2):150–158. 10.1016/j.jdermsci.2014.01.001 [DOI] [PubMed] [Google Scholar]
- Kitchen P, Salman MM, Halsey AM, Clarke-Bland C, MacDonald JA, Ishida H, Vogel HJ, Almutiri S, Logan A, Kreida S, Al-Jubair T, Missel JW, Gourdon P, Tornroth-Horsefield S, Conner MT, Ahmed Z, Conner AC, Bill RM (2020) Targeting aquaporin-4 subcellular localization to treat central nervous system edema. Cell. 10.1016/j.cell.2020.03.037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kress BT, Iliff JJ, Xia M, Wang M, Wei HS, Zeppenfeld D, Xie L, Kang H, Xu Q, Liew JA, Plog BA, Ding F, Deane R, Nedergaard M (2014) Impairment of paravascular clearance pathways in the aging brain. Ann Neurol 76(6):845–861. 10.1002/ana.24271 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lapshina KV, Ekimova IV (2024) Aquaporin-4 and Parkinson’s disease. Int J Mol Sci 25(3):1672. 10.3390/ijms25031672 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee Y, Choi Y, Park E-J, Kwon S, Kim H, Lee JY, Lee DS (2020) Improvement of glymphatic–lymphatic drainage of beta-amyloid by focused ultrasound in Alzheimer’s disease model. Sci Rep 10(1):16144. 10.1038/s41598-020-73151-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee DA, Park BS, Ko J, Park SH, Lee YJ, Kim IH, Park JH, Park KM (2022) Glymphatic system dysfunction in temporal lobe epilepsy patients with hippocampal sclerosis. Epilepsia Open 7(2):306–314. 10.1002/epi4.12594 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leppänen V-M, Tvorogov D, Kisko K, Prota AE, Jeltsch M, Anisimov A, Markovic-Mueller S, Stuttfeld E, Goldie KN, Ballmer-Hofer K, Alitalo K (2013) Structural and mechanistic insights into VEGF receptor 3 ligand binding and activation. Proc Natl Acad Sci U S A 110(32):12960–12965. 10.1073/pnas.1301415110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leser FS, Júnyor FdeS, Pagnoncelli IB, Delgado AB, Medeiros I, Nóbrega ACC, Andrade BdaS, de Lima MN, da Silva NE, Jacob L, Boyé K, Geraldo LHM, de Souza AMT, Maron-Gutierrez T, Castro-Faria-Neto H, Follmer C, Braga C, Neves GA, Eichmann A et al (2025) Ccl21-CCR7 blockade prevents neuroinflammation and degeneration in Parkinson’s disease models. J Neuroinflamm 22(1):31. 10.1186/s12974-024-03318-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li X, Liu C, Wang R (2020) Light modulation of brain and development of relevant equipment. J Alzheimers Dis 74(1):29–41. 10.3233/JAD-191240 [DOI] [PubMed] [Google Scholar]
- Li X, Zhang Z, Han M, Li Y, He L, Zhou B (2022) Generation of Piezo1-CreER transgenic mice for visualization and lineage tracing of mechanical force responsive cells in vivo. Genesis 60(4–5):e23476. 10.1002/dvg.23476 [DOI] [PubMed] [Google Scholar]
- Li Y, Wei S, Zhou H (2023) Interpret of international Delphi consensus on the management of aquaporin-4 immunoglobulin G seropositive neuromyelitis optica spectrum disorder. Chinese J Ocular Fund Dis 39(7):525–529. 10.3760/cma.j.cn511434-20230621-00281
- Liu D-X, He X, Wu D, Zhang Q, Yang C, Liang F-Y, He X-F, Dai G-Y, Pei Z, Lan Y, Xu G-Q (2017) Continuous theta burst stimulation facilitates the clearance efficiency of the glymphatic pathway in a mouse model of sleep deprivation. Neurosci Lett 653:189–194. 10.1016/j.neulet.2017.05.064 [DOI] [PubMed] [Google Scholar]
- Liu X, De Zwart JA, Schölvinck ML, Chang C, Ye FQ, Leopold DA, Duyn JH (2018) Subcortical evidence for a contribution of arousal to fMRI studies of brain activity. Nat Commun 9(1):395. 10.1038/s41467-017-02815-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu C, Habib T, Salimeen M, Pradhan A, Singh M, Wang M, Wu F, Zhang Y, Gao L, Yang G, Li X, Yang J (2020a) Quantification of visible Virchow-Robin spaces for detecting the functional status of the glymphatic system in children with newly diagnosed idiopathic generalized epilepsy. Seizure 78:12–17. 10.1016/j.seizure.2020.02.015 [DOI] [PubMed] [Google Scholar]
- Liu Y, Feng S, Subedi K, Wang H (2020b) Attenuation of ischemic stroke-caused brain injury by a monoamine oxidase inhibitor involves improved proteostasis and reduced neuroinflammation. Mol Neurobiol 57(2):937–948. 10.1007/s12035-019-01788-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu E, Peng X, Ma H, Zhang Y, Yang X, Zhang Y, Sun L, Yan J (2021a) The involvement of aquaporin-4 in the interstitial fluid drainage impairment following subarachnoid hemorrhage. Front Aging Neurosci 12:611494. 10.3389/fnagi.2020.611494 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu K, Zhu J, Chang Y, Lin Z, Shi Z, Li X, Chen X, Lin C, Pan S, Huang K (2021b) Attenuation of cerebral edema facilitates recovery of glymphatic system function after status epilepticus. JCI Insight 6(17):e151835. 10.1172/jci.insight.151835 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Louveau A (2018) Meningeal immunity, drainage, and tertiary lymphoid structure formation. In: Dieu-Nosjean M-C (ed) Tertiary lymphoid structures, vol 1845. Springer, New York, pp 31–45. 10.1007/978-1-4939-8709-2_3 [DOI] [PubMed] [Google Scholar]
- Louveau A, Smirnov I, Keyes TJ, Eccles JD, Rouhani SJ, Peske JD, Derecki NC, Castle D, Mandell JW, Lee KS, Harris TH, Kipnis J (2015) Structural and functional features of central nervous system lymphatic vessels. Nature 523(7560):337–341. 10.1038/nature14432 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Louveau A, Herz J, Alme MN, Salvador AF, Dong MQ, Viar KE, Herod SG, Knopp J, Setliff JC, Lupi AL, Da Mesquita S, Frost EL, Gaultier A, Harris TH, Cao R, Hu S, Lukens JR, Smirnov I, Overall CC, Oliver G, Kipnis J (2018) CNS lymphatic drainage and neuroinflammation are regulated by meningeal lymphatic vasculature. Nat Neurosci 21(10):1380–1391. 10.1038/s41593-018-0227-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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(1):1434. 10.1038/s41467-017-01484-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maggi P, Absinta M, Grammatico M, Vuolo L, Emmi G, Carlucci G, Spagni G, Barilaro A, Repice AM, Emmi L, Prisco D, Martinelli V, Scotti R, Sadeghi N, Perrotta G, Sati P, Dachy B, Reich DS, Filippi M, Massacesi L (2018) Central vein sign differentiates multiple sclerosis from central nervous system inflammatory vasculopathies. Ann Neurol 83(2):283–294. 10.1002/ana.25146 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Magliozzi R, Howell O, Vora A, Serafini B, Nicholas R, Puopolo M, Reynolds R, Aloisi F (2006) Meningeal B-cell follicles in secondary progressive multiple sclerosis associate with early onset of disease and severe cortical pathology. Brain 130(4):1089–1104. 10.1093/brain/awm038 [DOI] [PubMed] [Google Scholar]
- Mateo C, Knutsen PM, Tsai PS, Shih AY, Kleinfeld D (2017) Entrainment of arteriole vasomotor fluctuations by neural activity is a basis of blood-oxygenation-level-dependent “Resting-State” connectivity. Neuron 96(4):936-948.e3. 10.1016/j.neuron.2017.10.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Merlini A, Haberl M, Strauß J, Hildebrand L, Genc N, Franz J, Chilov D, Alitalo K, Flügel-Koch C, Stadelmann C, Flügel A, Odoardi F (2022) Distinct roles of the meningeal layers in CNS autoimmunity. Nat Neurosci 25(7):887–899. 10.1038/s41593-022-01108-3 [DOI] [PubMed] [Google Scholar]
- Mestre H, Tithof J, Du T, Song W, Peng W, Sweeney AM, Olveda G, Thomas JH, Nedergaard M, Kelley DH (2018) Flow of cerebrospinal fluid is driven by arterial pulsations and is reduced in hypertension. Nat Commun 9(1):4878. 10.1038/s41467-018-07318-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mestre H, Du T, Sweeney AM, Liu G, Samson AJ, Peng W, Mortensen KN, Stæger FF, Bork PAR, Bashford L, Toro ER, Tithof J, Kelley DH, Thomas JH, Hjorth PG, Martens EA, Mehta RI, Solis O, Blinder P et al (2020) Cerebrospinal fluid influx drives acute ischemic tissue swelling. Science 367(6483):eaax7171. 10.1126/science.aax7171 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miao A, Luo T, Hsieh B, Edge CJ, Gridley M, Wong RTC, Constandinou TG, Wisden W, Franks NP (2024) Brain clearance is reduced during sleep and anesthesia. Nat Neurosci 27(6):1046–1050. 10.1038/s41593-024-01638-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mogensen FL-H, Delle C, Nedergaard M (2021) The glymphatic system (en)during inflammation. Int J Mol Sci 22(14):7491. 10.3390/ijms22147491 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Monroe SM, Harkness KL (2022) Major depression and its recurrences: life course matters. Annu Rev Clin Psychol 18(1):329–357. 10.1146/annurev-clinpsy-072220-021440 [DOI] [PubMed] [Google Scholar]
- Nakano-Kobayashi A, Canela A, Yoshihara T, Hagiwara M (2023) Astrocyte-targeting therapy rescues cognitive impairment caused by neuroinflammation via the Nrf2 pathway. Proc Natl Acad Sci U S A 120(33):e2303809120. 10.1073/pnas.2303809120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nedergaard M, Goldman SA (2020) Glymphatic failure as a final common pathway to dementia. Science 370(6512):50–56. 10.1126/science.abb8739 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nielsen S, Smith BL, Christensen EI, Agre P (1993) Distribution of the aquaporin CHIP in secretory and resorptive epithelia and capillary endothelia. Proc Natl Acad Sci USA 90(15):7275–7279. 10.1073/pnas.90.15.7275 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Noé FM, Marchi N (2019) Central nervous system lymphatic unit, immunity, and epilepsy: is there a link? Epilepsia Open 4(1):30–39. 10.1002/epi4.12302 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Palazzo C, Buccoliero C, Mola MG, Abbrescia P, Nicchia GP, Trojano M, Frigeri A (2019) AQP4ex is crucial for the anchoring of AQP4 at the astrocyte end-feet and for neuromyelitis optical antibody binding. Acta Neuropathol Commun 7(1):51. 10.1186/s40478-019-0707-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paul F, Marignier R, Palace J, Arrambide G, Asgari N, Bennett JL, Cree BAC, De Sèze J, Fujihara K, Kim HJ, Hornby R, Huda S, Kissani N, Kleiter I, Kuwabara S, Lana-Peixoto M, Law L, Leite MI, Pandit L, Pittock SJ, Quan C, Ramanathan S, Rotstein D, Saiz A, Sato DK, Vaknin-Dembinsky A (2023) International delphi consensus on the management of AQP4-IgG+ NMOSD: recommendations for eculizumab, inebilizumab, and satralizumab. Neurol Neuroimmunol Neuroinflamm 10(4):e200124. 10.1212/NXI.0000000000200124 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Piantino JA, Iliff JJ, Lim MM (2022) The bidirectional link between sleep disturbances and traumatic brain injury symptoms: a role for glymphatic dysfunction? Biol Psychiatry 91(5):478–487. 10.1016/j.biopsych.2021.06.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pitsch J, Van Loo KMJ, Gallus M, Dik A, Kamalizade D, Baumgart A, Gnatkovsky V, Müller JA, Opitz T, Hicking G, Naik VN, Wachsmuth L, Faber C, Surges R, Kurts C, Schoch S, Melzer N, Becker AJ (2021) CD8+ T-lymphocyte-driven limbic encephalitis results in temporal lobe epilepsy. Ann Neurol 89(4):666–685. 10.1002/ana.26000 [DOI] [PubMed] [Google Scholar]
- Poderoso JJ, Helfenberger K, Poderoso C (2019) The effect of nitric oxide on mitochondrial respiration. Nitric Oxide Biol Chem 88:61–72. 10.1016/j.niox.2019.04.005 [DOI] [PubMed] [Google Scholar]
- Rabinovitch A, Aviramd I, Biton Y, Braunstein D (2019) A combined astrocyte–G-lymphatic model of epilepsy initiation, maintenance and termination. Med Hypotheses 133:109384. 10.1016/j.mehy.2019.109384 [DOI] [PubMed] [Google Scholar]
- Reeves BC, Karimy JK, Kundishora AJ, Mestre H, Cerci HM, Matouk C, Alper SL, Lundgaard I, Nedergaard M, Kahle KT (2020) Glymphatic system impairment in Alzheimer’s disease and idiopathic normal pressure hydrocephalus. Trends Mol Med 26(3):285–295. 10.1016/j.molmed.2019.11.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ren H, Luo C, Feng Y, Yao X, Shi Z, Liang F, Kang JX, Wan J-B, Pei Z, Su H (2017) Omega-3 polyunsaturated fatty acids promote amyloid-β clearance from the brain through mediating the function of the glymphatic system. FASEB J off Publ Fed Am Soc Exp Biol 31(1):282–293. 10.1096/fj.201600896 [DOI] [PubMed] [Google Scholar]
- Rolin C, Zimmer J, Seguin-Devaux C (2024) Bridging the gap with multispecific immune cell engagers in cancer and infectious diseases. Cell Mol Immunol 21(7):643–661. 10.1038/s41423-024-01176-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roomruangwong C, Barbosa DS, Matsumoto AK, Nogueira ADS, Kanchanatawan B, Sirivichayakul S, Carvalho AF, Duleu S, Geffard M, Moreira EG, Maes M (2017) Activated neuro-oxidative and neuro-nitrosative pathways at the end of term are associated with inflammation and physio-somatic and depression symptoms, while predicting outcome characteristics in mother and baby. J Affect Disord 223:49–58. 10.1016/j.jad.2017.07.002 [DOI] [PubMed] [Google Scholar]
- Roomruangwong C, Anderson G, Berk M, Stoyanov D, Carvalho AF, Maes M (2018) A neuro-immune, neuro-oxidative and neuro-nitrosative model of prenatal and postpartum depression. Prog Neuro-Psychopharmacol Biol Psychiatry 81:262–274. 10.1016/j.pnpbp.2017.09.015 [DOI] [PubMed] [Google Scholar]
- Salimeen MSA, Liu C, Li X, Wang M, Singh M, Si S, Li M, Cheng Y, Wang X, Zhao H, Wu F, Zhang Y, Tafawa H, Pradhan A, Yang G, Yang J (2021) Exploring variances of white matter integrity and the glymphatic system in simple febrile seizures and epilepsy. Front Neurol 12:595647. 10.3389/fneur.2021.595647 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salman MM, Kitchen P, Woodroofe MN, Brown JE, Bill RM, Conner AC, Conner MT (2017) Hypothermia increases aquaporin 4 (AQP4) plasma membrane abundance in human primary cortical astrocytes via a calcium/transient receptor potential vanilloid 4 (TRPV4)- and calmodulin-mediated mechanism. Eur J Neurosci 46(9):2542–2547. 10.1111/ejn.13723 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salvador AFM, Abduljawad N, Kipnis J (2024) Meningeal lymphatics in central nervous system diseases. Annu Rev Neurosci 47(1):323–344. 10.1146/annurev-neuro-113023-103045 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Si X, Dai S, Fang Y, Tang J, Wang Z, Li Y, Song Z, Chen Y, Liu Y, Zhao G, Zhang B, Pu J (2024) Matrix metalloproteinase-9 inhibition prevents aquaporin-4 depolarization-mediated glymphatic dysfunction in Parkinson’s disease. J Adv Res 56:125–136. 10.1016/j.jare.2023.03.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simard JM, Yurovsky V, Tsymbalyuk N, Melnichenko L, Ivanova S, Gerzanich V (2009) Protective effect of delayed treatment with low-dose glibenclamide in three models of ischemic stroke. Stroke 40(2):604–609. 10.1161/STROKEAHA.108.522409 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simon M, Wang MX, Ismail O, Braun M, Schindler AG, Reemmer J, Wang Z, Haveliwala MA, O’Boyle RP, Han WY, Roese N, Grafe M, Woltjer R, Boison D, Iliff JJ (2022) Loss of perivascular aquaporin-4 localization impairs glymphatic exchange and promotes amyloid β plaque formation in mice. Alzheimers Res Ther 14(1):59. 10.1186/s13195-022-00999-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith RC, Md WL, Wang Y, Jiang J, Wang J, Szabo V, Faull R, Jin H, Davis JM, Li C (2020) Effects of transcranial direct current stimulation on cognition and symptoms in Chinese patients with schizophrenia✰. Psychiatry Res 284:112617. 10.1016/j.psychres.2019.112617 [DOI] [PubMed] [Google Scholar]
- Song E, Mao T, Dong H, Boisserand LSB, Antila S, Bosenberg M, Alitalo K, Thomas J-L, Iwasaki A (2020) VEGF-C-driven lymphatic drainage enables immunosurveillance of brain tumours. Nature 577(7792):689–694. 10.1038/s41586-019-1912-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spalice A, Guido CA, Nicita F, Biasi CD, Zicari AM, Giannini L (2020) Dilated Virchow-Robin spaces in children with seizures. A possible correlation? Med Hypotheses 136:109481. 10.1016/j.mehy.2019.109481 [DOI] [PubMed] [Google Scholar]
- Spector R, Robert Snodgrass S, Johanson CE (2015) A balanced view of the cerebrospinal fluid composition and functions: focus on adult humans. Exp Neurol 273:57–68. 10.1016/j.expneurol.2015.07.027 [DOI] [PubMed] [Google Scholar]
- Staals J, Makin SDJ, Doubal FN, Dennis MS, Wardlaw JM (2014) Stroke subtype, vascular risk factors, and total MRI brain small-vessel disease burden. Neurology 83(14):1228–1234. 10.1212/WNL.0000000000000837 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Steinmetz JD, Seeher KM, Schiess N, Nichols E, Cao B, Servili C, Cavallera V, Cousin E, Hagins H, Moberg ME, Mehlman ML, Abate YH, Abbas J, Abbasi MA, Abbasian M, Abbastabar H, Abdelmasseh M, Abdollahi M, Abdollahi M, Abdollahifar M-A, Abd-Rabu R, Abdulah DM, Abdullahi A, Abedi A, Abedi V, Abeldańo Zuńiga RA, Abidi H, Abiodun O, Aboagye RG, Abolhassani H, Aboyans V, Abrha WA, Abualhasan A, Abu-Gharbieh E, Aburuz S, Adamu LH, Addo IY, Adebayo OM, Adekanmbi V, Adekiya TA, Adikusuma W, Adnani QES, Adra S, Afework T, Afolabi AA, Afraz A, Afzal S, Aghamiri S, Agodi A, Agyemang-Duah W, Ahinkorah BO, Ahmad A, Ahmad D, Ahmad S, Ahmadzade AM, Ahmed A, Ahmed H, Ahmed JQ, Ahmed LA, Ahmed MB, Ahmed SA, Ajami M, Aji B, Ajumobi O, Akade SE, Akbari M, Akbarialiabad H, Akhlaghi S, Akinosoglou K, Akinyemi RO, Akonde M, Al Hasan SM, Alahdab F, AL-Ahdal TMA, Al-amer RM, Albashtawy M, AlBataineh MT, Aldawsari KA, Alemi H, Alemi S, Algammal AM, Al-Gheethi AAS, Alhalaiqa FAN, Alhassan RK, Ali A, Ali EA, Ali L, Ali MU, Ali MM, Ali R, Ali S, Ali SSS, Ali Z, Alif SM, Alimohamadi Y, Aliyi AA, Aljofan M, Aljunid SM, Alladi S, Almazan JU, Almustanyir S, Al-Omari B, Alqahtani JS, Alqasmi I, Alqutaibi AY, Al-Shahi Salman R, Altaany Z, Al-Tawfiq JA, Altirkawi KA, Alvis-Guzman N, Al-Worafi YM, Aly H, Aly S, Alzoubi KH, Amani R, Amindarolzarbi A, Amiri S, Amirzade-Iranaq MH, Amu H, Amugsi DA, Amusa GA, Amzat J, Ancuceanu R, Anderlini D, Anderson DB, Andrei CL, Androudi S, Angappan D, Angesom TW, Anil A, Ansari-Moghaddam A, Anwer R, Arafat M, Aravkin AY, Areda D, Ariffin H, Arifin H, Arkew M, Ärnlöv J, Arooj M, Artamonov AA, Artanti KD, Aruleba RT, Asadi-Pooya AA, Asena TF, Asghari-Jafarabadi M, Ashraf M, Ashraf T, Atalell KA, Athari SS, Atinafu BTT, Atorkey P, Atout MMW, Atreya A, Aujayeb A, Avan A, Ayala Quintanilla BP, Ayatollahi H, Ayinde OO, Ayyoubzadeh SM, Azadnajafabad S, Azizi Z, Azizian K, Azzam AY, Babaei M, Badar M, Badiye AD, Baghdadi S, Bagherieh S, Bai R, Baig AA, Balakrishnan S, Balalla S, Baltatu OC, Banach M, Bandyopadhyay S, Banerjee I, Baran MF, Barboza MA, Barchitta M, Bardhan M, Barker-Collo SL, Bärnighausen TW, Barrow A, Bashash D, Bashiri H, Bashiru HA, Basiru A, Basso JD, Basu S, Batiha A-M, Batra K, Baune BT, Bedi N, Begde A, Begum T, Behnam B, Behnoush AH, Beiranvand M, Béjot Y, Bekele A, Belete MA, Belgaumi UI, Bemanalizadeh M, Bender RG, Benfor B, Bennett DA, Bensenor IM, Berice B, Bettencourt PJG, Beyene KA, Bhadra A, Bhagat DS, Bhangdia K, Bhardwaj N, Bhardwaj P, Bhargava A, Bhaskar S, Bhat AN, Bhat V, Bhatti GK, Bhatti JS, Bhatti R, Bijani A, Bikbov B, Bilalaga MM, Biswas A, Bitaraf S, Bitra VR, Bjørge T, Bodolica V, Bodunrin AO, Boloor A, Braithwaite D, Brayne C, Brenner H, Briko A, Bringas Vega ML, Brown J, Budke CM, Buonsenso D, Burkart K, Burns RA, Bustanji Y, Butt MH, Butt NS, Butt ZA, Cabral LS, dos Caetano Santos FL, Calina D, Campos-Nonato IR, Cao C, Carabin H, Cárdenas R, Carreras G, Carvalho AF, Castańeda-Orjuela CA, Casulli A, Catalá-López F, Catapano AL, Caye A, Cegolon L, Cenderadewi M, Cerin E, Chacón-Uscamaita PR, Chan JSK, Chanie GS, Charan J, Chattu VK, Chekol Abebe E, Chen H, Chen J, Chi G, Chichagi F, Chidambaram SB, Chimoriya R, Ching PR, Chitheer A, Chong YY, Chopra H, Choudhari SG, Chowdhury EK, Chowdhury R, Christensen H, Chu D-T, Chukwu IS, Chung E, Coberly K, Columbus A, Comachio J, Conde J, Cortesi PA, Costa VM, Couto RAS, Criqui MH, Cruz-Martins N, Dabbagh Ohadi MA, Dadana S, Dadras O, Dai X, Dai Z, D’Amico E, Danawi HA, Dandona L, Dandona R, Darwish AH, Das S, Dascalu AM, Dash NR, Dashti M, De la Hoz FP, de la Torre-Luque A, De Leo D, Dean FE, Dehghan A, Dejene H, Demant D, Demetriades AK, Demissie S, Deng X, Desai HD, Devanbu VGC, Dhama K, Dharmaratne SD, Dhimal M, da Dias Silva D, Diaz D, Dibas M, Ding DD, Dinu M, Dirac MA, Diress M, Do TC, Do THP, Doan KDK, Dodangeh M, Doheim MF, Dokova KG, Dongarwar D, Dsouza HL, Dube J, Duraisamy S, Durojaiye OC, Dutta S, Dziedzic AM, Edinur HA, Eissazade N, Ekholuenetale M, Ekundayo TC, El Nahas N, El Sayed I, Elahi Najafi MA, Elbarazi I, Elemam NM, Elgar FJ, Elgendy IY, Elhabashy HR, Elhadi M, Elilo LT, Ellenbogen RG, Elmeligy OAA, Elmonem MA, Elshaer M, Elsohaby I, Emamverdi M, Emeto TI, Endres M, Esezobor CI, Eskandarieh S, Fadaei A, Fagbamigbe AF, Fahim A, Faramarzi A, Fares J, Farjoud Kouhanjani M, Faro A, Farzadfar F, Fatehizadeh A, Fathi M, Fathi S, Fatima SAF, Feizkhah A, Fereshtehnejad S-M, Ferrari AJ, Ferreira N, Fetensa G, Firouraghi N, Fischer F, Fonseca AC, Force LM, Fornari A, Foroutan B, Fukumoto T, Gadanya MA, Gaidhane AM, Galali Y, Galehdar N, Gan Q, Gandhi AP, Ganesan B, Gardner WM, Garg N, Gau S-Y, Gautam RK, Gebre T, Gebrehiwot M, Gebremeskel GG, Gebreslassie HG, Getacher L, Ghaderi Yazdi B, Ghadirian F, Ghaffarpasand F, Ghanbari R, Ghasemi MohammadReza, Ghazy RM, Ghimire S, Gholami A, Gholamrezanezhad A, Ghotbi E, Ghozy S, Gialluisi A, Gill PS, Glasstetter LM, Gnedovskaya EV, Golchin A, Golechha M, Goleij P, Golinelli D, Gomes-Neto M, Goulart AC, Goyal A, Gray RJ, Grivna M, Guadie HA, Guan B, Guarducci G, Guicciardi S, Gunawardane DA, Guo H, Gupta B, Gupta R, Gupta S, Gupta VB, Gupta VK, Gutiérrez RA, Habibzadeh F, Hachinski V, Haddadi R, Hadei M, Hadi NR, Haep N, Haile TG, Haj-Mirzaian A, Hall BJ, Halwani R, Hameed S, Hamiduzzaman M, Hammoud A, Han H, Hanifi N, Hankey GJ, Hannan MA, Hao J, Harapan H, Hareru HE, Hargono A, Harlianto NI, Haro JM, Hartman NN, Hasaballah AI, Hasan F, Hasani H, Hasanian M, Hassan A, Hassan S, Hassanipour S, Hassankhani H, Hassen MB, Haubold J, Hay SI, Hayat K, Hegazy MI, Heidari G, Heidari M, Heidari-Soureshjani R, Hesami H, Hezam K, Hiraike Y, Hoffman HJ, Holla R, Hopf KP, Horita N, Hossain MM, Hossain MB, Hossain S, Hosseinzadeh H, Hosseinzadeh M, Hostiuc S, Hu C, Huang J, Huda MN, Hussain J, Hussein NR, Huynh H-H, Hwang B-F, Ibitoye SE, Ilaghi M, Ilesanmi OS, Ilic IM, Ilic MD, Immurana M, Iravanpour F, Islam SMS, Ismail F, Iso H, Isola G, Iwagami M, Iwu CCD, Iyer M, Jaan A, Jacob L, Jadidi-Niaragh F, Jafari M, Jafarinia M, Jafarzadeh A, Jahankhani K, Jahanmehr N, Jahrami H, Jaiswal A, Jakovljevic M, Jamora RDG, Jana S, Javadi N, Javed S, Javeed S, Jayapal SK, Jayaram S, Jiang H, Johnson CO, Johnson WD, Jokar M, Jonas JB, Joseph A, Joseph N, Joshua CE, Jürisson M, Kabir A, Kabir Z, Kabito GG, Kadashetti V, Kafi F, Kalani R, Kalantar F, Kaliyadan F, Kamath A, Kamath S, Kanchan T, Kandel A, Kandel H, Kanmodi KK, Karajizadeh M, Karami J, Karanth SD, Karaye IM, Karch A, Karimi A, Karimi H, Karimi Behnagh A, Kasraei H, Kassebaum NJ, Kauppila JH, Kaur H, Kaur N, Kayode GA, Kazemi F, Keikavoosi-Arani L, Keller C, Keykhaei M, Khadembashiri MA, Khader YS, Khafaie MA, Khajuria H, Khalaji A, Khamesipour F, Khammarnia M, Khan M, Khan MAB, Khan YH, Khan Suheb MZ, Khanmohammadi S, Khanna T, Khatab K, Khatatbeh H, Khatatbeh MM, Khateri S, Khatib MN, Khayat Kashani HR, Khonji MS, khorashadizadeh F, Khormali M, Khubchandani J, Kian S, Kim G, Kim J, Kim MS, Kim YJ, Kimokoti RW, Kisa A, Kisa S, Kivimäki M, Kochhar S, Kolahi A-A, Koly KN, Kompani F, Koroshetz WJ, Kosen S, Kourosh Arami M, Koyanagi Ai, Kravchenko MA, Krishan K, Krishnamoorthy V, Kuate Defo B, Kuddus MA, Kumar A, Kumar GA, Kumar M, Kumar N, Kumsa NB, Kundu S, Kurniasari MD, Kusuma D, Kuttikkattu A, Kyu HH, La Vecchia C, Ladan MA, Lahariya C, Laksono T, Lal DK, Lallukka T, Lám J, Lami FH, Landires I, Langguth B, Lasrado S, Latief K, Latifinaibin K, Lau K-M, Laurens MB, Lawal BK, Le LKD, Le TTT, Ledda C, Lee M, Lee S-W, Lee W-C, Lee YH, Leonardi M, Lerango TL, Li M-C, Li W, Ligade VS, Lim SS, Linehan C, Liu C, Liu J, Liu W, Lo C-H, Lo WD, Lobo SW, Logroscino G, Lopes G, Lopukhov PD, Lorenzovici L, Lorkowski S, Loureiro JA, Lubinda J, Lucchetti G, Lutzky Saute R, Ma ZF, Mabrok M, Machoy M, Madadizadeh F, El Magdy Abd Razek M, Maghazachi AA, Maghbouli N, Mahjoub S, Mahmoudi M, Majeed A, Malagón-Rojas JN, Malakan Rad E, Malhotra K, Malik AA, Malik I, Mallhi TH, Malta DC, Manilal A, Mansouri V, Mansournia MA, Marasini BP, Marateb HR, Maroufi SF, Martinez-Raga J, Martini S, Martins-Melo FR, Martorell M, März W, Marzo RR, Massano J, Mathangasinghe Y, Mathews E, Maude RJ, Maugeri A, Maulik PK, Mayeli M, Mazaheri M, McAlinden C, McGrath JJ, Meena JK, Mehndiratta MM, Mendez-Lopez MAM, Mendoza W, Mendoza-Cano O, Menezes RG, Merati M, Meretoja A, Merkin A, Mersha AM, Mestrovic T, Mi T, Miazgowski T, Michalek IM, Mihretie ET, Minh LHN, Mirfakhraie R, Mirica A, Mirrakhimov EM, Mirzaei M, Misganaw A, Misra S, Mithra P, Mizana BA, Mohamadkhani A, Mohamed NS, Mohammadi E, Mohammadi H, Mohammadi S, Mohammadshahi M, Mohammed M, Mohammed S, Mohan S, Mojiri-forushani H, Moka N, Mokdad AH, Molinaro S, Möller H, Monasta L, Moniruzzaman Md, Montazeri F, Moradi M, Moradi Y, Moradi-Lakeh M, Moraga P, Morovatdar N, Morrison SD, Mosapour A, Mosser JF, Mossialos E, Motaghinejad M, Mousavi P, Mousavi SE, Mubarik S, Muccioli L, Mughal F, Mukoro GD, Mulita A, Mulita F, Musaigwa F, Mustafa A, Mustafa G, Muthu S, Nagarajan AJ, Naghavi P, Naik GR, Nainu F, Nair TS, Najmuldeen HHR, Nakhostin Ansari N, Nambi G, Namdar Areshtanab H, Nargus S, Nascimento BR, Naser AY, Nashwan AJJ, Nasoori H, Nasreldein A, Natto ZS, Nauman J, Nayak BP, Nazri-Panjaki A, Negaresh M, Negash H, Negoi I, Negoi RI, Negru SM, Nejadghaderi SA, Nematollahi MH, Nesbit OD, Newton CRJ, Nguyen DH, Nguyen HTH, Nguyen HQ, Nguyen N-T, Nguyen PT, Nguyen VT, Niazi RK, Nikolouzakis TK, Niranjan V, Nnyanzi LA, Noman EA, Noroozi N, Norrving Bo, Noubiap JJ, Nri-Ezedi CA, Ntaios G, Nuńez-Samudio V, Nurrika D, Oancea B, Odetokun IA, O’Donnell MJ, Ogunsakin RE, Oguta JO, Oh I-H, Okati-Aliabad H, Okeke SR, Okekunle AP, Okonji OC, Okwute PG, Olagunju AT, Olaiya MT, Olana MD, Olatubi MI, Oliveira GMM, Olufadewa II, Olusanya BO, Omar Bali A, Ong S, Onwujekwe OE, Ordak M, Orji AU, Ortega-Altamirano DV, Osuagwu UL, Otstavnov N, Otstavnov SS, Ouyahia A, Owolabi MO, P A MP, Pacheco-Barrios K, Padubidri JR, Pal PK, Palange PN, Palladino C, Palladino R, Palma-Alvarez RF, Pan F, Panagiotakos D, Panda-Jonas S, Pandey A, Pandian JD, Pangaribuan HU, Pantazopoulos I, Pardhan S, Parija PP, Parikh RR, Park S, Parthasarathi A, Pashaei A, Patel J, Patil S, Patoulias D, Pawar S, Pedersini P, Pensato U, Pereira DM, Pereira J, Pereira MO, Peres MFP, Perico N, Perna S, Petcu I-R, Petermann-Rocha FE, Pham HT, Phillips MR, Pinilla-Monsalve GD, Piradov MA, Plotnikov E, Poddighe D, Polat B, Poluru R, Pond CD, Poudel GR, Pouramini A, Pourbagher-Shahri AM, Pourfridoni M, Pourtaheri N, Prakash PY, Prakash S, Prakash V, Prates EJS, Pritchett N, Purnobasuki H, Qasim NH, Qattea I, Qian G, Radhakrishnan V, Raee P, Raeisi Shahraki H, Rafique I, Raggi A, Raghav PR, Rahati MM, Rahim F, Rahimi Z, Rahimifard M, Rahman MO, Rahman MHU, Rahman M, Rahman MA, Rahmani AM, Rahmani S, Rahmani Youshanlouei H, Rahmati M, Raj Moolambally S, Rajabpour-Sanati A, Ramadan H, Ramasamy SK, Ramasubramani P, Ramazanu S, Rancic N, Rao IR, Rao SJ, Rapaka D, Rashedi V, Rashid AM, Rashidi M-M, Rashidi Alavijeh M, Rasouli-Saravani A, Rawaf S, Razo C, Redwan EMM, Rekabi Bana A, Remuzzi G, Rezaei N, Rezaeian M, Rhee TG, Riad A, Robinson SR, Rodrigues M, Rodriguez JAB, Roever L, Rogowski ELB, Romoli M, Ronfani L, Roy P, Roy Pramanik K, Rubagotti E, Ruiz MA, Russ TC, S Sunnerhagen K, Saad AMA, Saadatian Z, Saber K, SaberiKamarposhti M, Sacco S, Saddik B, Sadeghi E, Sadeghian S, Saeed U, Safdarian M, Safi SZ, Sagar R, Sagoe D, Saheb Sharif-Askari F, Saheb Sharif-Askari N, Sahebkar A, Sahoo SS, Sahraian MA, Sajedi SA, Sakshaug JW, Saleh MA, Salehi Omran H, Salem MR, Salimi S, Samadi Kafil H, Samadzadeh S, Samargandy S, Samodra YL, Samuel VP, Samy AM, Sanadgol N, Sanjeev RK, Sanmarchi F, Santomauro DF, Santri IN, Santric-Milicevic MM, Saravanan A, Sarveazad A, Satpathy M, Saylan M, Sayyah M, Scarmeas N, Schlaich MP, Schuermans A, Schwarzinger M, Schwebel DC, Selvaraj S, Sendekie AK, Sengupta P, Senthilkumaran S, Serban D, Sergindo MT, Sethi Y, SeyedAlinaghi SeyedAhmad, Seylani A, Shabani M, Shabany M, Shafie M, Shahabi S, Shahbandi A, Shahid S, Shahraki-Sanavi F, Shahsavari HR, Shahwan MJ, Shaikh MA, Shaji KS, Sham S, Shama ATT, Shamim MA, Shams-Beyranvand M, Shamsi MA, Shanawaz M, Sharath M, Sharfaei S, Sharifan A, Sharma M, Sharma R, Shashamo BB, Shayan M, Sheikhi RA, Shekhar S, Shen J, Shenoy SM, Shetty PH, Shiferaw DS, Shigematsu M, Shiri R, Shittu A, Shivakumar KM, Shokri F, Shool S, Shorofi SA, Shrestha S, Siankam Tankwanchi AB, Siddig EE, Sigfusdottir ID, Silva JP, Silva LMLR, Sinaei E, Singh BB, Singh G, Singh P, Singh S, Sirota SB, Sivakumar S, Sohag AAM, Solanki R, Soleimani H, Solikhah S, Solomon Y, Song S, Song Y, Sotoudeh H, Spartalis M, Stark BA, Starnes JR, Starodubova AV, Stein DJ, Steiner TJ, Stovner LJ, Suleman M, Suliankatchi Abdulkader R, Sultana A, Sun J, Sunkersing D, Sunny A, Susianti H, Swain CK, Szeto MD, Tabarés-Seisdedos R, Tabatabaei SM, Tabatabai S, Tabish M, Taheri M, Tahvildari A, Tajbakhsh A, Tampa M, Tamuzi JJLL, Tan K-K, Tang H, Tareke M, Tarigan IU, Tat NY, Tat VY, Tavakoli Oliaee R, Tavangar SM, Tavasol A, Tefera YM, Tehrani-Banihashemi A, Temesgen WA, Temsah M-H, Teramoto M, Tesfaye AH, Tesfaye EG, Tesler R, Thakali O, Thangaraju P, Thapa R, Thapar R, Thomas NK, Thrift AG, Ticoalu JHV, Tillawi T, Toghroli R, Tonelli M, Tovani-Palone MR, Traini E, Tran NM, Tran N-H, Tran PV, Tromans SJ, Truelsen TC, Truyen TTTT, Tsatsakis A, Tsegay GM, Tsermpini EE, Tualeka AR, Tufa DG, Ubah CS, Udoakang AJ, Ulhaq I, Umair M, Umakanthan S, Umapathi KK, Unim B, Unnikrishnan B, Vaithinathan AG, Vakilian A, Valadan Tahbaz S, Valizadeh R, Van den Eynde J, Vart P, Varthya SB, Vasankari TJ, Vaziri S, Vellingiri B, Venketasubramanian N, Verras G-I, Vervoort D, Villafańe JH, Villani L, Vinueza Veloz AF, Viskadourou M, Vladimirov SK, Vlassov V, Volovat SR, Vu LT, Vujcic IS, Wagaye B, Waheed Y, Wahood W, Walde MT, Wang F, Wang S, Wang Y, Wang Y-P, Waqas M, Waris A, Weerakoon KG, Weintraub RG, Weldemariam AH, Westerman R, Whisnant JL, Wickramasinghe DP, Wickramasinghe ND, Willekens B, Wilner LB, Winkler AS, Wolfe CDA, Wu A-M, Wulf Hanson S, Xu S, Xu X, Yadollahpour A, Yaghoubi S, Yahya G, Yamagishi K, Yang L, Yano Y, Yao Y, Yehualashet SS, Yeshaneh A, Yesiltepe M, Yi S, Yiğit A, Yiğit V, Yon DK, Yonemoto N, You Y, Younis MZ, Yu C, Yusuf H, Zadey S, Zahedi M, Zakham F, Zaki N, Zali A, Zamagni G, Zand R, Zandieh GGZ, Zangiabadian M, Zarghami A, Zastrozhin MS, Zeariya MGM, Zegeye ZB, Zeukeng F, Zhai C, Zhang C, Zhang H, Zhang Y, Zhang Z-J, Zhao H, Zhao Y, Zheng P, Zhou H, Zhu B, Zhumagaliuly A, Zielińska M, Zikarg YT, Zoladl M, Murray CJL, Ong KL, Feigin VL, Vos T, Dua T (2024) Global, regional, and national burden of disorders affecting the nervous system, 1990–2021: a systematic analysis for the Global burden of Disease study 2021. Lancet Neurol 23(4):344–381. 10.1016/S1474-4422(24)00038-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stickley A, Leinsalu M, DeVylder JE, Inoue Y, Koyanagi A (2019) Sleep problems and depression among 237 023 community-dwelling adults in 46 low- and middle-income countries. Sci Rep 9(1):12011. 10.1038/s41598-019-48334-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sulimai N, Lominadze D (2020) Fibrinogen and neuroinflammation during traumatic brain injury. Mol Neurobiol 57(11):4692–4703. 10.1007/s12035-020-02012-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun C, Lin L, Yin L, Hao X, Tian J, Zhang X, Ren Y, Li C, Yang Y (2022) Acutely inhibiting AQP4 with TGN-020 improves functional outcome by attenuating edema and peri-infarct astrogliosis after cerebral ischemia. Front Immunol 13:870029. 10.3389/fimmu.2022.870029 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun X, Tian Q, Yang Z, Liu Y, Li C, Hou B, Xie A (2023) Association of AQP4 single nucleotide polymorphisms (rs335929 and rs2075575) with Parkinson’s disease: a case-control study. Neurosci Lett 797:137062. 10.1016/j.neulet.2023.137062 [DOI] [PubMed] [Google Scholar]
- Sun X, Dias L, Peng C et al (2024) 40 Hz light flickering facilitates the glymphatic flow via adenosine signaling in mice. Cell Discov 10:81. 10.1038/s41421-024-00701-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tian Y, Zhao M, Chen Y, Yang M, Wang Y (2022) The underlying role of the glymphatic system and meningeal lymphatic vessels in cerebral small vessel disease. Biomolecules 12(6):748. 10.3390/biom12060748 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Träger U, Andre R, Lahiri N, Magnusson-Lind A, Weiss A, Grueninger S, McKinnon C, Sirinathsinghji E, Kahlon S, Pfister EL, Moser R, Hummerich H, Antoniou M, Bates GP, Luthi-Carter R, Lowdell MW, Björkqvist M, Ostroff GR, Aronin N, Tabrizi SJ (2014) HTT-lowering reverses Huntington’s disease immune dysfunction caused by NFκB pathway dysregulation. Brain 137(Pt 3):819–833. 10.1093/brain/awt355 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsai H-H, Hsieh Y-C, Lin JS, Kuo Z-T, Ho C-Y, Chen C-H, Chang C-F (2022) Functional investigation of meningeal lymphatic system in experimental intracerebral hemorrhage. Stroke 53(3):987–998. 10.1161/STROKEAHA.121.037834 [DOI] [PubMed] [Google Scholar]
- Tye KM, Mirzabekov JJ, Warden MR, Ferenczi EA, Tsai H-C, Finkelstein J, Kim S-Y, Adhikari A, Thompson KR, Andalman AS, Gunaydin LA, Witten IB, Deisseroth K (2013) Dopamine neurons modulate neural encoding and expression of depression-related behaviour. Nature 493(7433):537–541. 10.1038/nature11740 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vandebroek A, Yasui M (2020) Regulation of AQP4 in the central nervous system. Int J Mol Sci 21(5):1603. 10.3390/ijms21051603 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wallensten J, Nager A, Åsberg M, Borg K, Beser A, Wilczek A, Mobarrez F (2021) Leakage of astrocyte-derived extracellular vesicles in stress-induced exhaustion disorder: a cross-sectional study. Sci Rep 11(1):2009. 10.1038/s41598-021-81453-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang M, Iliff JJ, Liao Y, Chen MJ, Shinseki MS, Venkataraman A, Cheung J, Wang W, Nedergaard M (2012) Cognitive deficits and delayed neuronal loss in a mouse model of multiple microinfarcts. J Neurosci 32(50):17948–17960. 10.1523/JNEUROSCI.1860-12.2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang L-T, Liu K-J, Sytwu H-K, Yen M-L, Yen BL (2021) Advances in mesenchymal stem cell therapy for immune and inflammatory diseases: use of cell-free products and human pluripotent stem cell-derived mesenchymal stem cells. Stem Cells Transl Med 10(9):1288–1303. 10.1002/sctm.21-0021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang X, Zhang A, Yu Q, Wang Z, Wang J, Xu P, Liu Y, Lu J, Zheng J, Li H, Qi Y, Zhang J, Fang Y, Xu S, Zhou J, Wang K, Chen S, Zhang J (2023) Single-cell RNA sequencing and spatial transcriptomics reveal pathogenesis of meningeal lymphatic dysfunction after experimental subarachnoid hemorrhage. Adv Sci 10(21):2301428. 10.1002/advs.202301428 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang M, Yan C, Li X, Yang T, Wu S, Liu Q, Luo Q, Zhou F (2024) Non-invasive modulation of meningeal lymphatics ameliorates ageing and Alzheimer’s disease-associated pathology and cognition in mice. Nat Commun 15(1):1453. 10.1038/s41467-024-45656-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang H, Qin N, Maimaitiaili D, Wu J, Wang S, Zhou Y, Lu J, Li Y (2025a) Transcranial electrical stimulation as a therapeutic strategy for Alzheimer’s disease: current uses and challenges. J Alzheimers Dis. 10.1177/13872877251315777 [DOI] [PubMed] [Google Scholar]
- Wang Y, Li G, Wang H, Qi Q, Wang X, Lu H (2025b) Targeted therapeutic strategies for Nectin-4 in breast cancer: recent advances and future prospects. Breast (Edinb Scotl) 79:103838. 10.1016/j.breast.2024.103838 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Warren KE, Hood RJ, Walker FR, Spratt NJ (2024) Intracranial pressure is elevated at 24 h post-stroke in mice. Neuroprotection 2(1):60–64. 10.1002/nep3.36 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williams GP, Schonhoff AM, Jurkuvenaite A, Gallups NJ, Standaert DG, Harms AS (2021) CD4 T cells mediate brain inflammation and neurodegeneration in a mouse model of Parkinson’s disease. Brain 144(7):2047–2059. 10.1093/brain/awab103 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Willis EF, MacDonald KPA, Nguyen QH, Garrido AL, Gillespie ER, Harley SBR, Bartlett PF, Schroder WA, Yates AG, Anthony DC, Rose-John S, Ruitenberg MJ, Vukovic J (2020) Repopulating microglia promote brain repair in an IL-6-dependent manner. Cell 180(5):833-846.e16. 10.1016/j.cell.2020.02.013 [DOI] [PubMed] [Google Scholar]
- Wilson MH (2016) Monro-kellie 2.0: the dynamic vascular and venous pathophysiological components of intracranial pressure. J Cereb Blood Flow Metab 36(8):1338–1350. 10.1177/0271678X16648711 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu H, Zheng J, Xu S, Fang Y, Wu Y, Zeng J, Shao A, Shi L, Lu J, Mei S, Wang X, Guo X, Wang Y, Zhao Z, Zhang J (2021) Mer regulates microglial/macrophage M1/M2 polarization and alleviates neuroinflammation following traumatic brain injury. J Neuroinflamm 18(1):2. 10.1186/s12974-020-02041-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu Y, Zhang T, Li X, Wei Y, Li X, Wang S, Liu J, Li D, Wang S, Ye T (2023) Borneol-driven meningeal lymphatic drainage clears amyloid-β peptide to attenuate Alzheimer-like phenotype in mice. Theranostics 13(1):106–124. 10.7150/thno.76133 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu Y-C, Bogale TA, Koistinaho J, Pizzi M, Rolova T, Bellucci A (2024) The contribution of β-amyloid, Tau and α-synuclein to blood–brain barrier damage in neurodegenerative disorders. Acta Neuropathol 147(1):39. 10.1007/s00401-024-02696-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xie L, Kang H, Xu Q, Chen MJ, Liao Y, Thiyagarajan M, O’Donnell J, Christensen DJ, Nicholson C, Iliff JJ, Takano T, Deane R, Nedergaard M (2013) Sleep drives metabolite clearance from the adult brain. Science 342(6156):373–377. 10.1126/science.1241224 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xing R, Cheng J, Yu J, Li S, Ma H, Zhao Y (2023) Trifluoperazine reduces apoptosis and inflammatory responses in traumatic brain injury by preventing the accumulation of Aquaporin4 on the surface of brain cells. Int J Med Sci 20(6):797–809. 10.7150/ijms.82677 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu J-Q, Liu Q-Q, Huang S-Y, Duan C-Y, Lu H-B, Cao Y, Hu J-Z (2023) The lymphatic system: a therapeutic target for central nervous system disorders. Neural Regen Res 18(6):1249–1256. 10.4103/1673-5374.355741 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang Y, García-Cruzado M, Zeng H, Camprubí-Ferrer L, Bahatyrevich-Kharitonik B, Bachiller S, Deierborg T (2023) LPS priming before plaque deposition impedes microglial activation and restrains Aβ pathology in the 5xFAD mouse model of Alzheimer’s disease. Brain Behav Immun 113:228–247. 10.1016/j.bbi.2023.07.006 [DOI] [PubMed] [Google Scholar]
- Yang T, Tang Y, Liu X, Gong S, Yao E (2024) Microglia synchronizes with the circadian rhythm of the glymphatic system and modulates glymphatic system function. IUBMB Life 76(12):1209–1222. 10.1002/iub.2903 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yao D, Li R, Hao J, Huang H, Wang X, Ran L, Fang Y, He Y, Wang W, Liu X, Wang M (2023a) Melatonin alleviates depression-like behaviors and cognitive dysfunction in mice by regulating the circadian rhythm of AQP4 polarization. Transl Psychiatry. 10.1038/s41398-023-02614-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yao D, Li R, Hao J, Huang H, Wang X, Ran L, Fang Y, He Y, Wang W, Liu X, Wang M (2023b) Melatonin alleviates depression-like behaviors and cognitive dysfunction in mice by regulating the circadian rhythm of AQP4 polarization. Transl Psychiatry 13(1):310. 10.1038/s41398-023-02614-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye T, Yan X, Bai H, Wu Y, Liu J, Zhang X, Wei Y, Wang S (2024) Borneol regulates meningeal lymphatic valve plasticity to clear Aβ aggregates in the prevention of AD-like symptoms. Phytomedicine 130:155753. 10.1016/j.phymed.2024.155753 [DOI] [PubMed] [Google Scholar]
- Yen BL, Yen M-L, Wang L-T, Liu K-J, Sytwu H-K (2020) Current status of mesenchymal stem cell therapy for immune/inflammatory lung disorders: gleaning insights for possible use in COVID-19. Stem Cells Transl Med 9(10):1163–1173. 10.1002/sctm.20-0186 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yuan Y, Peng W, Lei J, Zhao Y, Zhao B, Li Y, Wang J, Qu Q (2024) AQP4 endocytosis-lysosome degradation mediated by MMP-9/β-DG involved in diabetes cognitive impairment. Mol Neurobiol 61(10):8438–8453. 10.1007/s12035-024-04085-9 [DOI] [PubMed] [Google Scholar]
- Yulug B, Hanoglu L, Kilic E (2017) The neuroprotective effect of focused ultrasound: new perspectives on an old tool. Brain Res Bull 131:199–206. 10.1016/j.brainresbull.2017.04.015 [DOI] [PubMed] [Google Scholar]
- Zanier ER, Bertani I, Sammali E, Pischiutta F, Chiaravalloti MA, Vegliante G, Masone A, Corbelli A, Smith DH, Menon DK, Stocchetti N, Fiordaliso F, De Simoni M-G, Stewart W, Chiesa R (2018) Induction of a transmissible tau pathology by traumatic brain injury. Brain. 10.1093/brain/awy193 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang D, Ruan J, Peng S, Li J, Hu X, Zhang Y, Zhang T, Ge Y, Zhu Z, Xiao X, Zhu Y, Li X, Li T, Zhou L, Gao Q, Zheng G, Zhao B, Li X, Zhu Y et al (2024a) Synaptic-like transmission between neural axons and arteriolar smooth muscle cells drives cerebral neurovascular coupling. Nat Neurosci 27(2):232–248. 10.1038/s41593-023-01515-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Y, Peng B, Chen S, Liang Q, Zhang Y, Lin S, Xu Z, Zhang J, Hou G, Qiu Y (2024b) Reduced coupling between global signal and cerebrospinal fluid inflow in patients with depressive disorder: a resting state functional MRI study. J Affect Disord 354:136–142. 10.1016/j.jad.2024.03.023 [DOI] [PubMed] [Google Scholar]
- Zhou K, Peng S, Yao G, Luo Y, Li Q, Huang Y, Zhang Q, Deng L, Song Z, Wang W, Liu D, Liu Y (2024) Association between glymphatic dysfunction and neurocognitive decline in patients with frontal lobe epilepsy. Quant Imaging Med Surg 14(9):6745–6755. 10.21037/qims-24-375 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu B, Cao A, Chen C, Zhou W, Luo W, Gui Y, Wang Q, Xu Z, Wang J (2024a) MMP-9 inhibition alleviates postoperative cognitive dysfunction by improving glymphatic function via regulating AQP4 polarity. Int Immunopharmacol 126:111215. 10.1016/j.intimp.2023.111215 [DOI] [PubMed] [Google Scholar]
- Zhu X, Lin J, Yang P, Wu S, Lin H, He W, Lin D, Cao M (2024b) Surgery induces neurocognitive disorder via neuroinflammation and glymphatic dysfunction in middle-aged mice with brain lymphatic drainage impairment. Front Neurosci 18:1426718. 10.3389/fnins.2024.1426718 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zou W, Pu T, Feng W, Lu M, Zheng Y, Du R, Xiao M, Hu G (2019) Blocking meningeal lymphatic drainage aggravates Parkinson’s disease-like pathology in mice overexpressing mutated α-synuclein. Transl Neurodegener 8:7. 10.1186/s40035-019-0147-y [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No datasets were generated or analysed during the current study.




