Skip to main content
Aging and Disease logoLink to Aging and Disease
. 2024 Feb 1;15(1):115–152. doi: 10.14336/AD.2023.0516

Connecting the Dots: The Cerebral Lymphatic System as a Bridge Between the Central Nervous System and Peripheral System in Health and Disease

Hongxiang Zhao 1,3,#, Meiyan Sun 1,3,#, Yue Zhang 3,#, Wenwen Kong 3, Lulu Fan 3, Kaifang Wang 3, Qing Xu 5, Baiyan Chen 3, Jianxin Dong 3, Yanan Shi 3, Zhengyan Wang 3, ShiQi Wang 3, Xiaoli Zhuang 6, Qi Li 7, Feihong Lin 8, Xinyu Yao 9, WenBo Zhang 10, Chang Kong 11, Rui Zhang 2,3,*, Dayun Feng 4,*, Xiaoyong Zhao 1,2,3,*
PMCID: PMC10796102  PMID: 37307828

Abstract

As a recently discovered waste removal system in the brain, cerebral lymphatic system is thought to play an important role in regulating the homeostasis of the central nervous system. Currently, more and more attention is being focused on the cerebral lymphatic system. Further understanding of the structural and functional characteristics of cerebral lymphatic system is essential to better understand the pathogenesis of diseases and to explore therapeutic approaches. In this review, we summarize the structural components and functional characteristics of cerebral lymphatic system. More importantly, it is closely associated with peripheral system diseases in the gastrointestinal tract, liver, and kidney. However, there is still a gap in the study of the cerebral lymphatic system. However, we believe that it is a critical mediator of the interactions between the central nervous system and the peripheral system.

Keywords: Cerebral lymphatic system, peripheral system, meningeal lymphatic vessels, glymphatic system, gastrointestinal tract, liver, kidney

1. Introduction

For a long time, the brain was thought to lack the presence of the lymphatic system. As a result, it may be in an immune-exempt state. So how exactly does the brain maintain its normal state of physiological function? A further understanding of the anatomy of the brain may be the key to solving this question. Numerous studies have shown that there may be a special lymphatic system in the brain - the cerebral lymphatic system. Paolo Mascagni first discovered the existence of a special structure, the meningeal lymphatic vessels in the brain, as early as the end of the 18th century [1]. However, due to the existence of paradigmatic thought and the limitation of science and technology, this theory has not been widely accepted by academia. Finally, in 2015, the existence of meningeal lymphatic vessels was confirmed and became an accepted concept [2]. At the same time, in 2012, Maiken Nedergaard and his team introduced the concept of the glymphatic system and corroborated the existence of this lymphatic structure in mouse models. Over the following years, the existence of this structure has also been demonstrated in the human brain [3-5]. Our understanding of the cerebral lymphatic system continues to improve with the development of imaging techniques. This has provided conclusive evidence for its existence and revolutionised our understanding of the hydrodynamics of fluid flow in the brain and of brain immunity. In 2023, the study of the cerebral lymphatic system entered a new historical phase with the discovery of the subarachnoidal lymphatic-like membrane [6]. As a novel concept, the cerebral lymphatic system has successfully changed our traditional perception of the anatomy of the brain and its functions [2, 3, 7, 8]. Several studies have demonstrated that it has similar functions to the peripheral lymphatic system, such as the efficient removal of metabolic waste and immune monitoring. At the same time, however, the unique structure of the cerebral lymphatic system and its drainage patterns break down our traditional perception of the lymphatic system. For example, the glymphatic system lacks the walls of typical lymphatic vessels and the space within its lumen is largely occupied by blood vessels. These findings have given us new insights into the pathogenesis of a variety of central nervous system diseases, such as brain tumour, cerebral haemorrhage, hydrocephalus, stroke and neurodegenerative diseases [9-12]. More interestingly, the cerebral lymphatic system has been shown to play an essential role in the interaction of the central nervous system with peripheral system such as the liver, gastrointestinal tract and kidney [13-16]. At the same time, it also performs the function of transporting brain antigens and immune cells. This provides a channel for immune-related communication between the central nervous system and the peripheral system [11, 17]. Currently, two main directions have been derived for the study of the cerebral lymphatic system: the meningeal lymphatics and the glymphatic system, which focus on the functional characteristics of the cerebral lymphatic system in physiological and pathological conditions [18-21]. In this paper, we review the current literature on the structure, physiological functions and properties of the cerebral lymphatic system and present a theoretical framework based on the structure and function of the cerebral lymphatic system as a whole. More importantly, the role of the cerebral lymphatic system as a bridge between the central nervous system and the peripheral system was reviewed and summarised, which may provide ideas for future research in this field and further improve the body of knowledge on the cerebral lymphatic system.

2. Structural components of the cerebral lymphatic system

Brain parenchyma produces various metabolic wastes during physiological or pathological processes. If these metabolic wastes are not removed effectively and in a timely manner, abnormalities in brain function may be induced. This can ultimately lead to the development and progression of a range of brain diseases [22]. Various studies have shown that the cerebral lymphatic system plays an important role in the transport and elimination of metabolic wastes in the central nervous system (CNS) [11, 23, 24]. The cerebral lymphatic system can exert its drainage role through two pathways - the glymphatic system pathway and the cerebrospinal fluid drainage pathway. The former consists mainly of the perivascular space (Virchow-Robin space) and the cerebrospinal fluid-interstitial fluid flow in the brain parenchyma. The latter consists mainly of meningeal lymphatics and olfactory/ cervical lymphatic pathways [22]. However, the precise anatomical pathways of the cerebral lymphatic system drainage are still unclear. According to the current knowledge of the cerebral lymphatic system, it consists mainly of the meningeal lymphatic vessels and the glymphatic system. Some metabolites in the central nervous system first enter the meningeal lymphatics from the deep brain through the glymphatic system and then enter the peripheral lymphatic system [2]. Substance exchange is the main function of the cerebral lymphatic system. Its purpose is to drain metabolic wastes accumulated in the brain parenchyma and to import nutrients. The meningeal lymphatic vessels and the glymphatic system are closely related but have subtle functional differences, and they function together by pairing each other. Further understanding and study of the anatomy of the cerebral lymphatic system may provide new ideas for the treatment of central nervous system diseases. We will present them separately next.

2.1. Meningeal lymphatic vessels

Meningeal lymphatic vessels (MLVs) are the main route for the removal of intracranial cerebrospinal fluid (CSF) and interstitial fluid (ISF) to the deep cervical lymph nodes (dCLN) outside the skull. To some extent, it can be considered as a bridge between intracranial and extracranial areas [25]. Meningeal lymphatics have typical characteristics of lymphatic vessels, mainly due to the presence of typical fully differentiated lymphatic endothelial cells, which are PROX1+/VEGFR3+/ LYVE1+/PDPN+/CCL21+/PECAM1low. The meningeal lymphatics are mainly distributed along the superior sagittal sinus, surrounding the transverse sinus, sigmoid sinus, posterior cranial fossa veins and middle meningeal artery. The meningeal lymphatics are sparsely distributed in the cranial vault and densely distributed in the cranial base. At the base of the skull, the meningeal lymphatics follow the distal cranial nerves, including the optic nerve, trigeminal nerve, glossopharyngeal nerve, paramedian nerve, and vagus nerve. Subsequently, it leaves the skull along with the cranial nerves through the neuroforamen of the skull base and eventually enters the deep cervical lymph nodes [7]. In addition to this, the meningeal lymphatic vessels are present in the sieve plate for distribution. It is able to leave the skull here and eventually enters the nasal mucosa [26].

Figure 1.

Figure 1.

Basic information about meningeal lymphatic vessels. Cerebrospinal fluid from the subarachnoid space enters the brain along the periarterial space, after which it leaves the periarterial space to exchange with the ISF in the brain parenchyma. It then exits along the perivenous space and returns to the subarachnoid space. A portion of this returns to the cerebrospinal fluid and a portion enters meningeal lymphatic vessels.

The specific functions of the meningeal lymphatic vessels differ depending on the location of their distribution. To date, several location-specific meningeal lymphatic systems have been studied in a targeted manner, such as basal MLVs, and dorsal MLVs [17, 27, 28]. The dorsal MLVs travel along the superior sagittal sinus (SSS) and the transverse sinus (TS). Their lumen diameter is small, most of them do not form branches, and they do not have valvular structures in their lumen. Their endothelial cells are interconnected in a discontinuous closed, loosely button-like fashion. In contrast, basal MLVs travel along the phosphoric sinus (PSS) and sigmoid sinus (SS). Their lumen diameter is large, with abundant capillary branches and rounded ends. Their endothelial cells have a typical oak leaf shape, and valves are present in the lumen. Moreover, in terms of their distribution, basal MLVs are closer to the subarachnoid space than dorsal MLVs. The anatomical differences between the basal MLVs and the dorsal MLVs determine that the former is more suitable for the drainage of large molecules in the cerebrospinal fluid. And the results of the study also proved that the basal MLVs are the main channels for cerebrospinal fluid drainage [27]. The study by Xue-ting Hu and her team showed that intracranial tumors were able to induce extensive remodeling of dorsal MLVs and were able to induce mild remodeling of basal MLVs in advanced stages [28]. This is a very valuable finding that the physiological functions of meningeal lymphatic vessels distributed in different sites may be somewhat different and require some differentiation in the study. For example, in studies addressing changes in meningeal lymphatic vessels with age, it was found that the function of dorsal MLVs consistently showed a trend of decay with increasing age in mice. In contrast, the basal MLVs changed less and even showed a compensatory enhancement in older mice. This fully demonstrates the functional differences between the two due to the different anatomical structures and distribution sites [27]. Therefore, targeted modulation of specific sites of meningeal lymphatics under pathological conditions may help us to greatly improve therapeutic efficiency and achieve better therapeutic benefits. It is also important to note that although the above studies mainly demonstrated the important role of basal MLVs in intracranial drainage, this does not mean that dorsal MLVs are less important. The two may simply have different functional emphases. At present, our understanding of the fine structure of meningeal lymphatics is still very limited, and a lot of relevant studies are still needed to explore it. The structural and compositional differences between different sites of meningeal lymphatics may inspire us to explore more potential targets, such as a specific receptor on the endothelial cells of a particular site of meningeal lymphatics. Another important issue is that the structure of the meningeal lymphatic vessels has been shown to change to some extent with age. And this alteration usually has a negative effect, which makes the risk of developing many types of diseases, such as neurodegenerative diseases [12, 29-31]. Therefore, after clarifying the meticulous changes in the structure of the meningeal lymphatic vessels with age, attempts to reverse this change during treatment might provide great therapeutic benefit.

Figure 2.

Figure 2.

Basic information about glymphatic system. The mass flow of interstitial solutes through the brain parenchyma through pressure gradients. In this way, metabolic waste products such as toxic proteins in the brain parenchyma can be effectively removed. This can effectively maintain brain homeostasis.

2.2. Perivascular spaces

Perivascular spaces are a particular class of structures that exist around the blood vessels of the brain, which are filled with interstitial fluid [32, 33]. The outer boundary of the perivascular spaces consists of a glial boundary membrane. It is a compact structure formed by astrocyte foot processes and the parenchymal basement membrane that covers them. The inner boundary of the perivascular spaces, on the other hand, consists of the basement membrane of the vascular endothelium. The inner boundary and the outer boundary together extend with the vascular tree all the way to the capillary network. Finally, the two fuse to form the blind end [34-37]. Depending on their location, perivascular spaces can be divided into three types: basal ganglia type, hemispheric type and midbrain type [38]. However, the specific structural and functional differences between these three types remain to be investigated. As an important component of the glymphatic system, the perivascular spaces may be deeply involved in fluid drainage and metabolic waste removal in the brain [39-41]. Rennels ML et al. showed that cerebrospinal fluid (CSF) first enters the brain parenchyma from the subarachnoid space via the periarterial spaces. It then exchanges fluid with interstitial fluid (ISF). Finally, the fluid flow returns through the perivenous spaces to the subarachnoid space [42]. In this process, wastes removal from the perivascular spaces rely primarily on microglia, pericytes and perivascular phagocytes [43-45]. It is important to note that there are also views that some perivascular spaces do not directly communicate with the subarachnoid space [33, 42, 46-48]. The specific processes and mechanisms by which the cerebrospinal fluid interacts with the interstitial fluid in the perivascular spaces of different types of vessels remain to be investigated. In addition, a drainage pathway called "intramural periarterial drainage" has also been identified. In this pathway, the basement membrane of the cerebral arteries or capillaries appears to play a similar role to that of the cerebral lymphatic system. The process is also highly dependent on sustained cardiac output [49, 50]. However, whether this particular pathway is also present in the perivascular spaces remains to be investigated further. Numerous studies have found that enlargement of the perivascular spaces may be present in several types of central nervous system diseases, such as Alzheimer's disease, neurofibromas and familial brain malformations. This may be related to factors such as damage to the cerebral microvasculature caused by the accumulation of metabolic waste [51-54]. In addition, Park CH's study revealed an age-dependent dilatation of the perivascular spaces, with a particularly marked dilatation of the basal ganglia type [55]. This demonstrates that ageing may be similarly strongly associated with the enlargement of the perivascular spaces [56-58]. Iliff JJ showed that ageing also causes a slowing of fluid flow within the perivascular spaces and hardening of the arterial wall [40]. However, there is still a lack of follow-up studies on the enlargement of the perivascular spaces in large samples over a long period of time. This remains to be further explored.

2.3. Astrocytic endfeet

The astrocyte endfeet are special subcellular structures emanating from astrocytes, which are distributed with a large amount of aquaporin-4 [59]. They play an important role in regulating brain homeostasis, maintaining the integrity of the blood-brain barrier and removing waste solutes [60, 61]. Numerous studies have shown that astrocytes are an important component of the cerebral lymphatic system [62, 63]. Astrocyte endfeet are involved in forming the outer wall of the perivascular spaces, which in turn form the glymphatic system [64].

Aquaporin-4 (AQP-4) is a tetrameric structure with a molecular weight of 30 kDa and a pore size of approximately 0.5 nm. AQP-4 is predominantly found in the endfeet of astrocytes around brain vessels and is usually polarized [65, 66]. Galina Yankova et al. showed that the endfeet of astrocytes have a wide coverage and that fluid and solutes must pass through the gap between the endfeet of AQP-4 or astrocytes in order to penetrate outside the perivascular spaces. Interestingly, the cellular gap between the end feet is often less than 20 nm, which implies that the fluid flow transfer pathway through AQP-4 may play a major role [67]. In fact, numerous studies have shown that AQP-4 on the astrocyte endfeet promotes fluid flow between the cerebrospinal fluid and interstitial fluid [3, 21, 65, 68, 69]. Yifan Wang et al. found that a decrease in AQP-4 may likewise disrupt the normal structure and function of astrocytes, leading to an impaired establishment of a reactive astrocytic network [21, 61, 65, 68, 70]. Nadia Aalling Jessen and Kazuhisa Ishida et al. demonstrated that the drainage of cerebrospinal fluid was severely impaired in AQP-4 knockout mice compared to wild group mice. This resulted in significantly increased levels of β-amyloid and tau proteins in the cerebrospinal fluid, which further exacerbated the associated neurodegenerative pathologies [21, 71]. However, the specific distribution of AQP-4 on astrocytes and the potential mechanisms by which it mediates the transport of fluids and solutes remain to be investigated [65, 69]. In addition, AQP-4 has an important functional feature of circadian rhythm [72]. Fluid flow and clearance of metabolic waste in the cerebral lymphatic system are both significantly enhanced and peak during deep sleep [73]. Lauren M. Hablitz et al. showed that the distribution of cerebrospinal fluid is controlled by circadian rhythms and regulated by AQP-4[74]. Therefore, studying the underlying mechanisms of the circadian rhythm of AQP-4 may help us to better explore the functional characteristics of AQP-4.

3. Functional characteristics of the cerebral lymphatic system

3.1. Role in regulating cerebrospinal fluid circulation

Cerebrospinal fluid is produced by the choroid plexus, brain interstitium, and meninges and is stored in the ventricular and subarachnoid spaces of the brain. It has a variety of functions including the transport of nutrients and metabolic waste, immunity and the regulation of intracranial pressure. The efficient circulation of cerebrospinal fluid is important for the maintenance of environmental homeostasis in the central nervous system [75, 76]. However, with further understanding of the cerebral lymphatic system, we are aware that the old hypothesis of cerebrospinal fluid circulation may be somewhat problematic. The presence of the cerebral lymphatic system has to some extent broadened the range of fluid and solute flow in the cerebrospinal fluid and enriched the pathways of cerebrospinal fluid circulation. This is mainly achieved by the glymphatic system [11, 12, 27, 77]. The current view is that substances in the CSF are able to enter the perivascular spaces of the arteries and enter the brain parenchyma via AQP-4 in the peduncle of astrocytes. The Bulat-Klarica-Oreˇskovi’c hypothesis proposes that the bidirectional exchange of solutes and fluids between cerebrospinal fluid and interstitial fluid via AQP-4 and the flow of fluid streams is continuous. Osmotic and hydrostatic pressures are the major drivers of this process [77-79]. Subsequently, the material follows the fluid flow in the interstitial fluid to the relatively hypertonic perivascular spaces. Thereafter, the fluid flow in the perivascular spaces can drain along the nerve sheaths of the cranial and spinal nerves, the arachnoid granules and the meningeal lymphatic vessels [3, 7, 80-86]. Many studies have shown that the cerebrospinal fluid outflow from the CNS through the dorsal and basal dural lymphatics in the cerebral lymphatic system accounts for 30-50% of the total cerebrospinal fluid drainage [8, 12, 87]. This demonstrates the importance of the cerebral lymphatic system in the process of cerebrospinal fluid circulation. Considering the immune function of the cerebral lymphatic system, by enhancing the function of the cerebral lymphatic system, we may be able to find more effective and complete solutions for the removal of harmful waste products from the central nervous system.

3.2. Macromolecule and immune cell transport

Recent studies have found that the cerebral lymphatic system plays an important role in the transport of macromolecular substances [88]. Conventional wisdom holds that the blood-brain barrier (BBB) to some extent impedes the entry of macromolecular substances into the brain [89]. However, the cerebral lymphatic system well bypasses the obstruction of the blood-brain barrier and provides a new pathway for the transport of macromolecular substances. On the one hand, the cerebral lymphatic system is able to transport and supply nutrients to the tissues and cells of the central nervous system [88], such as glucose [90], folic acid [91], lactate [92], and vitamins [91]. Notably, this pathway may be the only route to the CNS for some nutrients that cannot transit through the blood-brain barrier, such as glutamate [93]. On the other hand, the cerebral lymphatic system also takes on the important function of removing metabolic waste products produced by the CNS [94], such as soluble β-amyloid [3], tau protein [95] and α-synuclein [7]. However, further research is needed on the types of substances that are transported through the cerebral lymphatic system and the specific mechanisms of the transport process [96, 97]. In addition, the cerebral lymphatic system can also facilitate the movement of viruses into and out of the central nervous system. And this has a certain double-edged effect. On the one hand, peripheral viral infections may invade the CNS through the cerebral lymphatic system. This may be achieved through the blood-brain barrier, the nasal lymphatics or the meningeal lymphatic vessels [98]. On the other hand, meningeal lymphatic vessels may divert neurotropic viruses from the CNS to the deep cervical lymph nodes, thereby reducing CNS damage [99]. However, it is important to note that this process may also induce a peripheral immune response, which in turn may lead to inflammation and injury in the peripheral system [100]. Therefore, it is important to explore how to find a balance between these two effects.

Furthermore, the cerebral lymphatic system also has the function of transporting immune cells [11]. The cerebral lymphatic system is responsible for the transport of many types of immune cells, such as macrophages, dendritic cells, T cells, B cells, monocytes, neutrophils, etc. [11, 101-105]. Among these, the immune cells in the dura mater are more heterogeneous compared to those in other sites. This shows a certain peculiarity. After entering the cerebrospinal fluid, cytokines produced by T cells distributed in the dura mater may enter the brain parenchyma directly through the perivascular spaces to activate neurons. In addition, they may also indirectly regulate neuronal activity by inducing the secretion of other cytokines by glial cells [101, 106-111]. However, given that the dura mater is not in direct contact with the brain parenchyma, the exact pathways and mechanisms by which cytokines cross the arachnoid and enter the cerebrospinal fluid are still unclear. At the same time, the immune system of the central nervous system is able to interact with the peripheral immune system through the cerebral lymphatic system [112]. It is important to note that the immune response within the CNS differs somewhat from that in other tissues [113-116], but the underlying mechanisms remain to be explored. In addition, a recent study by Maiken Nedergaard's team identified a new structure in the brain, the subarachnoid lymphoid membrane (SLYM), which is a membrane structure composed of a layer of Prox1 protein-containing cells interwoven with collagen fibres. It is capable of dividing the subarachnoid space into an outer superficial chamber and an internal deep chamber [6]. Under physiological conditions, the SLYM not only mediates the controlled flow of cerebrospinal fluid and the exchange of substances, but it also contains immune cells that play an immunodetective role. When the homeostasis of the brain is disrupted, a large number of immune cells cross the SLMY and reach the brain parenchyma, causing neuroinflammation [117, 118]. Further studies of the SLMY may provide a better understanding of the flow of fluid in the brain and the mechanisms underlying the movement of immune cells in the brain.

3.3. Substance exchange between the central nervous system and peripheral system

Traditionally, the central nervous system is thought to be able to exchange substances with the periphery via the blood and cerebrospinal fluid, and this process is regulated primarily by the blood-brain and blood-cerebrospinal fluid barriers [119]. Studies have shown that 40% of Aβ in the brain can be transported to the peripheral system for clearance [3, 41, 43]. At the same time, peripheral injection of brain extracts containing large amounts of Aβ also induced excessive accumulation of Aβ in the brains of humans and mice [120-123]. This suggests that there may be a close exchange of substances between the central nervous system and the peripheral system. However, as we have learned more about the cerebral lymphatic system, we have discovered that it may also play an important role in the exchange of substances between the central nervous system and the peripheral system. Shinji Naganawa et al. found that when gadolinium-based contrast agents (GBCAs) were injected intravenously, GBCAs could bypass the blood-brain barrier and be transported through the cerebral lymphatic system into the brain parenchyma [124, 125]. However, the exact flow pathways and mechanisms of this process remain unclear. Notably, there may be a special link in the process of material interaction between the CNS and the peripheral system, i.e., a possible bidirectional traffic between the meningeal lymphatics and the deep cervical lymph nodes. This differs significantly from the familiar unidirectional transport of peripheral lymphatic vessels. The study showed that after injection of 99mTc-DX into the deep cervical lymph nodes, Fan Chen et al. detected radioactivity in the skull base dura, cerebrospinal fluid and brain tissue. Moreover, radioactivity could be detected in the skull base dura before brain tissue [126]. This may provide indirect evidence for a bidirectional interaction between the peripheral and cerebral lymphatic systems. Another possible route for the reentry of drainage fluid from the cervical lymph nodes into the brain may now be via blood circulation and finally across the blood-brain barrier. However, the exact pathway of how cervical lymph node drainage reenters the brain is currently unknown.

4. The critical role of the cerebral lymphatic system as a bridge between the peripheral system and the central nervous system

Numerous studies have shown that the cerebral lymphatic system is extensively and closely linked to the peripheral system. Some peripheral organs, such as the liver, gastrointestinal tract, and kidney, may be involved in the potential peripheral clearance pathways of toxic proteins from the central nervous system [127-131]. For example, as a classic neurodegenerative disease, Alzheimer's disease is a classic case of communication between the cerebral lymphatic system and the peripheral system [132]. The abnormal accumulation of toxic proteins such as amyloid-β and tau in the brain due to the imbalance between production and clearance is considered as the main causative factor [133-139]. And according to experimental results, 19% of pathological tau proteins [140] and 40% of Aβ in the brain are transported to the periphery for clearance [127]. Therefore, some alterations in the peripheral system may lead to abnormalities in the central nervous system through the cerebral lymphatic system as a mediator.

4.1. Communication and regulation between the central nervous system and the gastrointestinal tract via the cerebral lymphatic system

The main function of the gastrointestinal tract is the secretion of digestive enzymes and the absorption of ingested nutrients. This is mainly regulated by the central nervous system (CNS), the enteric nervous system (ENS) and enteroendocrine cells [141, 142]. Currently, several CNS disorders have been shown to be associated with the gastrointestinal tract and may even originate in the gastrointestinal system, such as Alzheimer's disease, Parkinson's disease, and ischemia/reperfusion injury [143-148]. Recent insights suggest that bidirectional effects exist between components of the microbial-brain-gut axis and are achieved through multiple neural, hormonal, metabolic, and immune mechanisms [149-153]. And in this process, the cerebral lymphatic system may be the key intermediate link between the brain and the gastrointestinal tract to communicate with each other. On the one hand, the gastrointestinal tract may have an impact on the function of the brain lymphatic system. It was found that in a mouse model of colitis induced with dextran sodium sulfate (DSS), enhanced NLRP3 inflammatory vesicle activity induced neuroinflammation and impaired astrocyte function. This leads to reduced clearance of the brain lymphatic system, which causes cognitive dysfunction in mice [14, 154-156]. Also, dysbiosis of gut microbes induces disturbances in cerebrospinal fluid flow, which triggers a decrease in the function of the cerebral lymphatic system [157]. In a mouse model of Porphyromonas gingivalis (Pg) gavage, we also observed a reduction in the rate of CSF-ISF exchange and ISF flow in the brain. This is thought to be related to the dysfunction of the cerebral lymphatic system [15].

Figure 3.

Figure 3.

Possible mechanisms of co-occurring neuroinflammatory response and brain edema in patients with hepatic encephalopathy and potential therapeutic targets for hepatic encephalopathy. Decreased detoxification of the liver in patients with cirrhosis leads to the accumulation of NH3 and inflammatory factors (IL-1β, IL-6, TNF-α) in their bodies. These substances can accumulate in the brain tissue with blood circulation and subsequently induce hepatic encephalopathy. (A) On the one hand, excess NH3 in the brain is converted to Gln by GS, causing a hyperosmotic environment in astrocytes. On the other hand, it can increase the activity of iNOS by activating NF-kB pathway, which makes the level of NO elevated. This can lead to astrocyte swelling and eventually brain edema. Meanwhile, targeted regulation of AQP-4 expression levels has a dual role. Increased AQP-4 expression not only promotes the drainage of metabolic waste from glymphatic system, but also mediates the entry of water into the astrocytes thus causing brain edema. (B) Increased inflammatory factors can activate microglia and astrocytes. Activated microglia and astrocytes in turn further promote the release of inflammatory factors. Positive feedback is formed between the two. Exogenous injection of VEGF-C may inhibit the NF-kB signaling pathway by upregulating the expression of Sik-1, which in turn reduces the inflammatory response. At the same time, VEGF-C may also promote the clearance of metabolic wastes such as NH3 by inducing the proliferation of meningeal lymphatic vessels.

In addition, metabolites of microorganisms may also have an effect on the cerebral lymphatic system. Some bacterial metabolites have been found to positively affect astrocytes, such as sodium butyrate that exacerbates MPTP-induced astrocyte activation in a mouse model of Parkinson's disease, thereby exacerbating neuro-inflammation and colitis [158]. However, some other bacterial metabolites have neuroprotective effects. For example, mucilaginous extracts of Archangium sp. UTMC 4070 and Cystobacter sp. UTMC 4073 strains could exert anti-oxidative stress effects to protect astrocytes. On the other hand, the cerebral lymphatic system may also provide a pathway for the transfer of bacteria and bacterial products, inflammatory factors and immune cells from the gastrointestinal tract to the center. Bostancıklıoğlu M suggested that the virus may enter the central nervous system via the lymphatic system based on the correlation between neurological and digestive symptoms in patients with novel coronavirus pneumonia [159, 160]. Similarly, Porphyromonas gingivalis may also cause brain inflammation through the cerebral lymphatic system [161, 162]. In dysbiosis of the gastrointestinal flora, the microbiota and its products stimulate the production of cytokines by gastrointestinal epithelial cells and macrophages. And the cerebral lymphatic system may provide a pathway for these cytokines to reach the brain [163]. On the other hand, during gastrointestinal flora dysbiosis, microbial-associated molecular patterns (MAMP) and pathogen-associated molecular patterns (PAMP) of the gut microbiota can be recognized by pattern recognition receptors on immune cells in the central nervous system. This may induce the release of inflammatory cytokines and affect the activity of astrocytes, which in turn may have an impact on the cerebral lymphatic system. Toll-like receptors recognize microbial nucleic acids, lipopolysaccharides, pepti-doglycans, and other components to signal to downstream inflammatory pathways, including NF-κB, MAPK, and/or interferon-regulatory factor signaling pathways [164]. In contrast, activation of NF-κB-related pathways mediated by NLRP3 inflammatory vesicles may reduce the polarity of AQP-4, which in turn leads to impaired normal function of the brain lymphatic system [14, 65, 165-168]. In addition, TREM receptors are potential mediators of the gastrointestinal tract affecting the brain through the cerebral lymphatic system [169]. TREM-2 is able to exert anti-inflammatory effects by negatively regulating Toll-like receptor 4 in astrocytes [170, 171].

Endocrine hormones, especially those involved in the hypothalamic-pituitary-adrenal axis, are important components of the brain-gut axis. They are able to influence the cerebral lymphatic system by participating in the regulation of astrocyte morphology and function, which may in turn [82, 151, 172-174]. Previously, the gut microbiota has been shown to influence the expression of genes associated with the HPA axis [175, 176]. Maria Meyer's study showed that adrenal glucocorticoids may trigger neuroinflammation by activating Toll-like receptor 4 on astrocytes, which subsequently triggers neuroinflammation. Adrenocorticotropin-releasing hormone can increase AQP-4 expression via the cAMP / PKA signaling pathway, which in turn causes astrocyte swelling [177, 178]. In addition, Yu-Xia Lou et al. found that adrenal glucocorticoids were able to impair the normal function of the gap junction between astrocytes [179]. This may likewise affect the drainage function of the cerebral lymphatic system.

When the ecology of the gastrointestinal tract is dysregulated, neural signals generated in the gastrointestinal tract are transmitted to the brain via afferent nerves. And the process produces certain neurotransmitters that may be involved in the regulation of astrocytes [172, 180]. For example, serotonin can be taken up by astrocytes and regulate their functions [181, 182]. And 90-95% of serotonin is produced in the gastrointestinal tract [183]. Some selective serotonin reuptake inhibitors (SSRI) have been found to inhibit the inflammatory response of astrocytes by inhibiting the NF-κB pathway, which in turn may have an effect on the cerebral lymphatic system [183-185].

4.2. Communication and regulation between the central nervous system and the liver via the cerebral lymphatic system

As a detoxification organ in the body, the liver plays an important role in the removal of toxins from the body [186, 187]. It plays an irreplaceable role in the metabolism and detoxification of NH3. When the liver is dysfunctional, this function can be significantly reduced, which can cause the accumulation of toxic products such as NH3 and cause serious damage to vital organs of the body, such as brain [188, 189]. Therefore, accelerating the clearance of toxic metabolites may be important for the treatment of this type of disease. As a new concept proposed in recent years, the cerebral lymphatic system has a role in drainage and removal of metabolic waste, immune monitoring, etc. This is highly similar to the functions of the liver. Numerous studies have also shown a solid link between the two.

4.2.1. The relationship between the cerebral lymphatic system and hepatic encephalopathy

Hepatic encephalopathy (HE) is a serious complication that occurs in the end-stage of various liver diseases such as cirrhosis. It is mainly caused by the weakened detoxification function of the liver and elevated blood ammonia due to portal-systemic shunting [190]. Besides, neuroinflammation is also an important causative factor of HE [191]. It has been shown that patients with cirrhosis are often accompanied by systemic inflammatory response syndrome [192]. These inflammatory factors activate microglia and astrocytes and prompt them to further release pro-inflammatory factors [193]. Positive feedback is formed between the two, which in turn exacerbates the neuroinflammatory response in the brain. Therefore, by promoting the excretion of NH3 and other metabolic wastes in the brain of patients with hepatic encephalopathy, it has some therapeutic significance for the neuroinflammatory response in the brain. The functional characteristics of the cerebral lymphatic system to remove and drain metabolic wastes make it a potential target for the treatment of hepatic encephalopathy.

Figure 4.

Figure 4.

The binding of CCL19/CCL21 to CCR7 activated signaling pathways in tumor cells, including PI3/Akt, MAPK/ERK and Jak/STAT, which enhanced the expression of VEGF-C and thus promoted lymphangiogenesis. Meanwhile, activation of the VEGF-C/VEGFR-3 signaling pathway can promote the release of CCL19 and CCL21 in lymph nodes. Furthermore, activation of the CCR7 chemokine axis can promote dendritic cell entry into the T cell compartment of lymph nodes, which enables specific recognition of CD80/CD86 on dendritic cells and CD28 on T cells. This activates T cells and promotes T cell infiltration into the tumour.

Aspelund et al. demonstrated that hypoplasia of the meningeal lymphatics leads to a significant reduction in the clearance of meningeal lymphatics in mice [7]. Shao-Jung Hsu and his team effectively promoted the drainage of metabolic waste from the brain by promoting the generation of meningeal lymphatics in a mouse model suffering from cirrhosis with hepatic encephalopathy. This helped alleviate the microglia activation and neuroinflammatory responses, thus ameliorating the symptoms of hepatic encephalopathy to some extent [13]. In addition to this, Pasquale Gallina et al. proposed a hydrodynamic hypothesis regarding the pathogenesis of glymphatic system damage in patients with hepatic encephalopathy [194]. Another study showed that defects in the glymphatic system led to reduced clearance of harmful substances in the brain of patients with cirrhosis. And its mechanism of action may be related to the decreased expression of AQP-4 [195]. Therefore, upregulation of AQP-4 may be able to promote the removal of toxic waste by the glymphatic system. This shows that the cerebral lymphatic system has great potential to become a treatment for hepatic encephalopathy.

4.2.2. Dual role of cerebral lymphatic system in brain metastasis from hepatocellular carcinoma

Brain metastasis from hepatocellular carcinoma is an important cause of death in patients with hepatocellular carcinoma. The current treatment options for it are relatively limited and the prognosis is generally poor [196, 197]. It is currently believed that most brain metastasis from tumors occur through hematogenous metastasis [198]. However, with the introduction of the concept of cerebral lymphatic system, we may be able to further improve the body of knowledge on brain metastasis from liver tumor. Numerous studies have shown that the CCL21/CCR7 axis plays an important role in the lymphatic metastasis of tumors [199, 200]. On the one hand, CCL21 interacts with CCR7 to control the transfer of immune cells such as DCs and T cells to lymphatic vessels and lymph nodes, which in turn enhances the anti-tumor immune response [201]. This is beneficial for tumor treatment. Xueting Hu and his team demonstrated that the combination of anti-PD-1/CTLA4 therapy showed better therapeutic effects after overexpression of VEGF-C in the intracranial tumor mouse model [28]. However, this effect was reversed after blocking the CCL21/CCR7 axis. This suggests that the effect of VEGF-C-enhanced antitumor immunotherapy is somewhat dependent on the CCL21/CCR7 axis. On the other hand, certain CCR7-expressing tumor cells can also use this to undergo lymphatic metastasis, leading to tumor spread and metastasis [202].

Figure 5.

Figure 5.

Communication between the gastrointestinal tract and cerebral lymphatic system is mediated directly and indirectly by immune, neural and endocrine pathways. (A) Changes in the gut microbiota occur when inflammatory damage occurs in the gut. Pathogenic microorganisms and their products, inflammatory factors, pathogen-associated molecular patterns (MAMPs, PAMPs), TLRs, and TREMs-positive activated macrophages may reach the brain through blood circulation and lymphatic circulation, which in turn affect the function of cerebral lymphatic system. (B) Some neurotransmitters (e.g., serotonin) also act on astrocytes and lead to activation of related pathways, which in turn affects the function of cerebral lymphatic system. (C) The gut microbiota can regulate hormones related to the hypothalamic-pituitary-adrenal axis, which in turn affects the function of cerebral lymphatic system.

Ding et al. found an association between high CCR7 expression and lymph node metastasis and poor prognosis in esophageal squamous cell carcinoma [203]. VEGF-C overexpression induces the formation of neo-lymphatic vessel [204], which undoubtedly facilitates the metastasis of hepatocellular carcinoma to the brain. However, a study showed that the incidence of brain metastasis in lung cancer patients showed a significant positive correlation with the high expression of VEGF-C [205]. So does it have similar results in patients with brain metastasis from hepatocellular carcinoma? However, there is still a lack of validated studies proving the relationship between brain metastasis of hepatocellular carcinoma and high expression of VEGF-C. Therefore, our team hypothesized that targeted regulation of VEGF-C and CCR7 may become a novel therapeutic option for the prevention and treatment of brain metastasis via lymphatic route in patients with hepatocellular carcinoma in the future. And it becomes crucial to master a balance between VEGF-C in enhancing anti-tumor immune response and promoting tumor metastasis.

4.3. Communication and regulation between the central nervous system and the kidney via the cerebral lymphatic system

The kidney is able to eliminate metabolic wastes from the body by producing urine, which in turn maintains the homeostasis of the body's internal environment [206]. In recent years, as the kidney-brain axis continues to receive widespread attention, the anatomical and patho-physiological connections between the kidney and the brain have been further revealed [207-213]. Interestingly, the cerebral lymphatic system is able to facilitate the removal of metabolic waste by deeply engaging in fluid circulation in the brain [13]. This is highly similar to the function of the kidney. Currently, numerous studies have shown that the cerebral lymphatic system is an important "bridge" between kidney and brain.

Figure 6.

Figure 6.

Mechanism of action of cerebral lymphatic system in brain diseases due to impaired renal function. (A) Indoxyl sulfate (IS) accumulates during impaired renal function and activates its receptor AhR (Aryl hydrocarbon Receptor), which in turn activates downstream oxidative stress, inflammation and apoptotic pathways. This ultimately leads to the damage of astrocytes. This is a common mechanism for the development of kidney-induced neurodegenerative and cerebrovascular diseases. (B) The accumulation of metabolic wastes in the brain is able to increase the expression of AQP-4 and widen the perivascular spaces. This induces the development of neurodegenerative diseases. (C) The accumulation of metabolic wastes in the brain can cause endothelial cell damage, increased AQP-4 expression, and the enlargement of perivascular spaces. This can induce the development of cerebrovascular diseases such as intracranial aneurysms.

4.3.1. The role of the cerebral lymphatic system as a bridge between renal disease and cerebrovascular disease

Numerous studies have shown that there are certain similarities in the anatomy and blood supply of kidney and the brain. As low resistance end organs that are also subject to high flow blood shock, both have a greater potential for microvascular injury [16, 207]. The cerebral lymphatic system plays an important role in the interplay of renal and cerebrovascular diseases. On the one hand, renal disease may promote the development of cerebrovascular disease. In the case of chronic kidney disease (CKD), its promotion of cerebrovascular disease is mainly attributed to the damaging effect on cerebrovascular endothelial cells caused by the accumulation of metabolic waste products (e.g., indolol sulfate) in the body [214, 215]. A study by Ashley A Penton and her team suggested that CKD may be a potential trigger for the enlargement of the perivascular gap in the brain in stroke patients. And the enlargement of the perivascular spaces may be highly associated with the dysfunction of the glymphatic system [216, 217]. Therefore, the cerebral lymphatic system may play a role in the induction of cerebrovascular disease by renal disease. On the other hand, cerebrovascular disease may promote the development of renal disease. Some studies have found that the incidence of CKD is much higher in stroke patients than in the general population [218]. However, the underlying mechanism by which cerebrovascular disease induces the development of renal disease is unclear. We speculate that this may be caused by the impaired function of the central nervous system leading to the normal physiological function of the kidney or by the existence of some potential common pathogenesis of cerebrovascular disease and renal disease [219-221]. And this still needs to be further explored.

4.3.2. The role of the cerebral lymphatic system in bridging the gap between renal disease and cognitive dysfunction

End-stage renal disease is usually associated with cognitive dysfunction, typically manifested by inattention, impaired inhibitory control, memory loss, language, visuospatial performance and executive dysfunction, and sleep disturbances [222]. The cerebral lymphatic system is also an important factor mediating the onset of cognitive dysfunction due to kidney disease.

Current studies on chronic kidney disease (CKD) have shown that the accumulation of uremic toxins in the brain is an important cause of cognitive dysfunction in patients [206]. On the one hand, patients with chronic kidney disease (CKD) may induce anemia due to reduced erythropoietin production, which causes a compensatory increase in cerebral blood flow. This leads to an increased distribution of uremic toxins in the body to the brain and eventually to cognitive dysfunction. On the other hand, reduced erythropoietin production in CKD patients may lead to a decrease in hemoglobin content to some extent. This may cause cerebral hypoxia, which may likewise induce cognitive dysfunction [223]. A study by Chang Min Heo and his team showed that patients with chronic kidney disease already have dysfunction of the glymphatic system in the early stages of the disease [222, 224]. This is mainly due to two factors. On the one hand, astrocytes may be damaged or apoptotic due to the inflammatory response and oxidative stress triggered by the accumulation of uremic toxins in the brain [225-227]. On the other hand, changes in blood osmolarity in uremic patients may lead to altered AQP-4 expression [228].

Neurological complications induced by uremic toxin accumulation in the brain of patients with acute kidney injury (AKI) are usually more severe and develop faster than in patients with CKD [229, 230]. This may be mainly related to the development of pro-inflammatory responses, the release of pro-inflammatory cytokines, and alterations in the blood-brain barrier [231-233]. AKI is able to induce damage and inflammatory responses in the brain. It has been shown that the number of cells presenting GFAP-positive in the CA1 region of the hippocampus is significantly elevated after ischemia-reperfusion injury of the kidney. This predicts a massive proliferation of astrocytes [234, 235]. And as an important component in the cerebral lymphatic system, the proliferation of astrocytes may affect the normal function of the glymphatic system to some extent. The specific mechanism remains to be further explored.

5. Potential implications for disease pathogenesis and therapeutic approaches

5.1. Potential targets for the diagnosis and treatment of liver disease

5.1.1. Hepatic encephalopathy

There is still a lack of specific indicators for the diagnosis of hepatic encephalopathy. However, Hajime Hirase and his team have done this by linking the gene for fluorescent protein to albumin and injecting it into C57BL/6JRj mice via a transgenic virus. They found that this induced the liver to produce modified albumin, which eventually produced a fluorescent tracer in the blood [236]. Since the liver can also produce lipoproteins, participate in processes such as glucose metabolism [237] and urea synthesis [238]. And these substances may also be potential targets for being modified. This may provide a novel idea for monitoring the progression of brain diseases in the future. However, it is worth thinking about the fact that the liver function of patients with hepatic encephalopathy has become severely impaired, resulting in its ability to synthesize proteins significantly diminished. Then, in order to achieve clear and precise tracing in different application scenarios, we should further determine the specific amount of fluorescent protein that should be induced in the liver and how to achieve this goal. This remains to be further explored.

Currently, the main clinical approach is to reduce blood ammonia concentrations by targeting the intestine and thus treating hepatic encephalopathy. However, this regimen has some limitations due to its long duration of action [189]. Therefore, considering that patients with hepatic encephalopathy already have a large accumulation of NH3 in the brain and the detoxification function of the liver is significantly reduced, accelerating the excretion of NH3 and other metabolic wastes in the brain of patients with hepatic encephalopathy may be another effective treatment for this disease. This may be achieved, for example, by promoting the production of new lymphatic vessels and increasing the flow of fluid. Vascular endothelial growth factor-C (VEGF-C) plays an important role in the formation of meningeal lymphatic vessels [7]. Its binding to VEGFR3 promotes the proliferation of lymphatic vessel endothelial cells and vascular endothelial cells, which in turn induces the generation of new lymphatic vessels and new blood vessels [204, 239]. For the brain, VEGF-C not only alleviates the neuroinflammatory symptoms in the mice with hepatic encephalopathy by increasing the drainage efficiency of the cerebral lymphatic system, but also suppresses microglia-induced inflammatory responses by upregulating Sik-1 expression and inhibiting the NF-kB signaling pathway [13, 240]. Therefore, we hypothesized that exogenous injection of VEGF-C might effectively reduce the accumulation of metabolic wastes in the brain and improve the symptoms of patients with hepatic encephalopathy by promoting the production of meningeal lymphatics and cerebral microvasculature. This has some promising applications. Meanwhile, for the liver, we speculate that upregulating VEGF-C in the liver may help establish effective collateral circulation in the liver by promoting hepatic angiogenesis. On the one hand, this could alleviate the symptoms of portal hypertension and liver ischemia and hypoxia in patients with cirrhosis. On the other hand, good collateral circulation could enable partial metabolism of NH3 in the liver and thus effectively relieve the neurological symptoms in patients with hepatic encephalopathy. However, the safety and efficacy of this potential treatment method still need to be explored and confirmed by numerous studies.

Aquaporin 4 (AQP-4) is the major water channel protein in the mammalian central nervous system. It is mainly found in the peduncle terminals of astrocytes and is important for maintaining normal water homeostasis in the CNS [165, 241, 242]. It also serves as a key hub for the glymphatic system to perform normal physiological functions and is closely related to fluid flow and substance exchange between the perivascular spaces and interstitial fluid [112]. On the one hand, enhanced AQP-4 expression can promote drainage of the glymphatic system, reduce the accumulation of metabolic waste in the brain, and thus improve the neurological symptoms in patients with hepatic encephalopathy [243]. But on the other hand, increased AQP-4 expression may also induce the formation of brain edema [244, 245]. From this perspective, down-regulation of AQP-4 expression could reduce brain edema and produce therapeutic effects in patients with hepatic encephalopathy complicated by brain edema. Therefore, it is important to find the balance between the two roles of AQP-4 in exerting enhanced lymphatic drainage and reducing cerebral edema. This still needs to be further explored.

5.1.2. Brain metastasis from hepatocellular carcinoma

5.1.2.1. Diagnosis

In addition to clarifying the potential mechanism of brain metastasis in hepatocellular carcinoma, the search for its early detection indicators has a key role. The most commonly used serological marker for diagnosing hepatocellular carcinoma in clinical practice is AFP. However, due to its lack of specificity, it is not possible to identify benign and malignant lesions in the liver by this indicator in some cases [246]. Therefore, it is clinically important to find specific diagnostic indicators for this disease. At present, the diagnosis of brain metastasis from hepatocellular carcinoma and its progression are mainly confirmed by clinical symptoms and imaging examinations. Patients with brain metastasis from hepatocellular carcinoma often have various clinical manifestations, such as headache, vomiting, hemianopia, disorientation, aphasia, hemiparesis, ataxia, mental deterioration, etc. Image examinations such as CT and MRI show tumors in the brain with edema. Laboratory tests often show significantly elevated levels of AFP, AFP- L3 and DCP in these patients [247].

In addition, Song et al. found that patients with esophageal cancer with positive expression of CCR7 mRNA and/or VEGF-C mRNA had a significantly higher recurrence rate of lymphatic metastases than those without expression of CCR7 mRNA and VEGF-C mRNA [248]. This suggests that VEGF-C and CCR7 may be reliable molecular indicators to mark lymphatic metastasis of tumors. Jianjun Zhao's team also found that serum levels of VEGF were higher in patients with hepatocellular carcinoma that had metastasized than in those without metastasis [249]. Therefore, VEGF-C may have some diagnostic value for brain metastasis from hepatocellular carcinoma. When patients develop brain metastasis from hepatocellular carcinoma, do the levels of VEGF-C and CCR7 in cerebrospinal fluid also change and become our molecular indicators to detect brain metastasis? Can the combination and indicators such as VEGF-C, CCR7 and tumor markers improve the accuracy of diagnosis? These questions remain to be further investigated.

5.1.2.2. Potential therapeutic targets

For patients with brain metastasis from hepatocellular carcinoma, although treatment modalities such as surgical resection, whole brain radiation therapy, and stereotactic radiation therapy have shown some therapeutic effects in some studies, the survival of patients with brain metastases from hepatocellular carcinoma is still not long at present [250-256]. Therefore, it is important to find potential therapeutic targets for brain metastasis from hepatocellular carcinoma. The current treatment of brain tumors mainly relies on radiotherapy and chemotherapy. However, the existence of multiple factors has led to the treatment effect of brain tumor has been unsatisfactory. On the one hand, due to the existence of the blood-brain barrier, the entry of therapeutic drugs into the central nervous system is hindered, which can affect the efficacy to a certain extent. On the other hand, in the central nervous system, the T cell-mediated immune response to tumor antigens is very limited, which can easily lead to uncontrolled tumor growth and thus worsen the disease. Currently, the emerging cancer therapy of "immune checkpoint blockade" has received a lot of attention and has been applied in the treatment of breast cancer [257], hepatocellular carcinoma [258] and other types of tumors. Recent findings suggest that vascular endothelial growth factor C (VEGF-C) may enhance immune checkpoint blockade, thereby enhancing the body's immune response to tumors in the brain as well as other immune-exempt sites [259-263]. CCL19 or CCL21 binding to CCR7 activates signaling pathways in tumor cells, including PI3/Akt [264-266], MAPK/ERK [267-269], and Jak/STAT [269, 270], thereby enhancing VEGF-C expression. It has been shown that VEGF-C is an important factor in the early development of lymphatic vessels [271]. It has been shown that ectopic expression of VEGF-C may enhance T cell-mediated antitumor responses by affecting the function of meningeal lymphatic vessels. This includes enhanced initiation of anti-tumor T cells in deep cervical lymph nodes, migration of T cells to tumors, rapid clearance of brain tumors, and establishment of long-lasting anti-tumor immune memory [272]. Eric Song and his team induced high expression of VEGF-C in CSF by injecting VEGF-C mRNA constructs into the large pool of mice, which in turn enhanced endogenous immune responses [261]. This confirms, to some extent, that increased VEGF-C expression levels can play a positive role in the treatment of brain tumor. The introduction of exogenous VEGF-C as a therapeutic option has some feasibility. However, the combination of local injection of VEGF-C and immune checkpoint inhibitors is not perfect at present, and a lot of basic and clinical studies are still needed to prove its effectiveness and safety [273].

However, the act of upregulating VEGF-C also has a double-edged sword effect [274]. In terms of its positive significance, numerous studies have shown that VEGF-C can promote the priming of cancer cell antigens, enhance the initiation of antitumor T cells in deep cervical lymph nodes, the migration of T cells to tumors, and the establishment of long-lasting antitumor immune memory [261, 274-276]. However, in terms of its negative impact, the ability of VEGF-C to promote the proliferation and expansion of meningeal lymphatic vessels may promote lymphatic metastasis of brain tumours. Mihaela Skobe et al., Yulong He et al. and Satoshi Hirakawa et al. all suggested that overexpression of VEGF-C could effectively promote lymphatic vasculature generation, which in turn promotes lymphatic metastasis of tumor cells [272, 277, 278]. The results of the study by N Khromova et al. similarly suggest that downregulation of VEGF-C can inhibit the growth and metastasis of tumours [279]. At the same time, we cannot ignore the background condition that tumor cells can evade immune surveillance by activating immune checkpoints. These factors expose the obvious drawbacks of the strategy of achieving therapeutic goals by simply increasing VEGF-C expression levels in vivo. In this regard, we may be able to inhibit the binding of VEGFR-2/VEGFR-3 to VEGF-C by using receptor antagonists of VEGFR-2/VEGFR-3, thereby inhibiting the production of blood vessels and lymphatic vessels. This may not only limit the metastatic pathway of the tumor, but also reduce the blood and oxygen supply to the tumor. Thus, tumor growth is limited to some extent [280]. By combining anti-lymphangiogenic therapy with anti-angiogenic therapy, it may provide a new therapeutic strategy for the treatment of advanced cancer, which deserves further investigation.

In conclusion, it is very important to grasp the balance between the positive and negative effects of rising VEGF-C levels. In the process of applying VEGF-C in the treatment of brain metastasis from hepatocellular carcinoma, the primary problem to be solved is to find the balance between its positive and negative effects, so as to achieve the best therapeutic effect. But this needs to be further investigated. This may also be another effective way to prevent brain metastasis from tumors.

Meanwhile, as a kind of nano-vesicles containing various contents such as nucleic acids, proteins and lipids, exosomes have low toxicity, low immunogenicity and good biocompatibility. Therefore, they are considered as a good carrier for targeted drug delivery [281-284]. The application of exosome-targeted drug delivery in cancer treatment is one of the research hotspots in the scientific community today, so what is its role in hepatocellular carcinoma? As the current first-line drug for the treatment of hepatocellular carcinoma, long-term application of sorafenib may produce certain side effects and drug resistance [285, 286]. And the discovery of exosomes has provided a new solution to this problem. It has been shown that miR-122 in exosomes produced by hepatocellular carcinoma may play an important role in improving drug resistance and inhibiting vascular regeneration [287-289]. By encapsulating miR-122 into exosomes secreted from adipose tissue-derived mesenchymal stem cells and targeting their expression to hepatocellular carcinoma cells, it was able to improve the sensitivity of the organism to chemotherapeutic drugs [290]. In addition, Wang et al. demonstrated that treatment of hepatocellular carcinoma cells with miR-744-rich exosomes similarly enhanced the sensitivity of the organism to chemotherapeutic drugs [291]. Thus, targeted delivery to hepatocellular carcinoma cells by loading mi-RNA into exosomes may become a new therapeutic modality for patients resistant to chemotherapeutic drugs. If we target delivery to the liver or brain of patients at risk of brain metastasis by loading monoclonal antibodies to VEGF-C into exosomes, thereby inhibiting lymphatic vessels and angiogenesis, could this effectively reduce the risk of brain metastasis from liver cancer? For patients with brain metastasis, can we load a miRNA or an effective drug into exosomes and target it to the brain through the cerebral lymphatic system? The above questions still need to be confirmed by further studies.

5.2. Possible therapeutic approaches based on gastrointestinal regulation of the cerebral lymphatic system

Treatment of brain disorders through the gut is a hot topic of current research. Based on the close relationship between the gastrointestinal tract and the cerebral lymphatic system, we may achieve modulation of the cerebral lymphatic system by interfering with the microorganisms in the gastrointestinal tract or with the help of the intestinal lymphatic system. Currently, the main therapeutic measures to regulate the intestinal flora include nanocapsules [292], photobiomodulation [293], electroacupuncture or acupuncture [294], diet [295], probiotics [296], antibiotics [297], fecal bacteria transplantation [298], etc.

5.2.1. Nanovesicles

Currently, various types of cell-derived nanovesicles and synthetic nanodrug carriers offer new possibilities to deliver drugs for the treatment of cerebral diseases through the cerebral lymphatic system [299, 300]. Compared with intravenous drug delivery, oral nanoparticle delivery has more advantages [301]. On the one hand, oral nanoparticle administration is relatively fast and convenient. On the other hand, oral nanoparticle administration is mainly absorbed and delivered through the lymphatic system. This can effectively avoid the first-pass elimination by the liver, thus solving the problem of poor pharmacokinetics that exists with the administration of nanovesicles through intravenous injection [302]. In addition, nanovesicles have good designability [299]. Therefore, can we design nanovesicles targeting astrocytes to achieve targeted therapy for the cerebral lymphatic system [303, 304]? For example, Xunwei Lai found that miR-146a-5p-modified exosomes derived from human umbilical cord mesenchymal stem cells could target neurotoxic astrocytes and attenuate their activation [292]. And this remains to be further explored.

5.2.2. Photobiomodulation (PBM)

Brain using transcranial and intranasal PBM therapy has been shown to be effective in reducing neuroinflammation and promoting neuronal repair and regeneration [305, 306]. It also significantly enhances the drainage of meningeal lymphatics [305-308]. The strong penetration of near infrared (NIR) allows it to reach deep into the skull and is non-invasive throughout the operation. This makes it a potential option that can be used to treat brain diseases such as neurodegenerative disorders [309, 310]. In addition, PBM has a direct effect on the gastrointestinal tract. On the one hand, PBM therapy targeting the abdominal region may also alter the gut microbiota and affect the central nervous system through the release of as yet unidentified circulating mediators [293]. On the other hand, by irradiating the abdominal region, PBM therapy may enhance the permeability of the mesenteric lymphatic vessels. This may help to enhance the intestinal absorption of oral nanovesicular drugs. By pairing nanoparticle engineering with biophotonics, we may be able to achieve better therapeutic benefits. However, the appropriate irradiation dose for PBM therapy still needs to be explored in further studies.

5.2.3. Acupuncture or electro-acupuncture

Numerous studies have demonstrated the unique role of acupuncture or electroacupuncture in regulating the gastrointestinal flora [311]. Electroacupuncture and acupuncture can cause an increase in the relative abundance of Lactobacillidae and elevated bacteroidetes/ Firmicutes in the gastrointestinal flora [312-314]. And acupuncture or electroacupuncture may be based on the regulation of intestinal flora, thus having some therapeutic effects on some central nervous system diseases, such as Alzheimer's disease [313], Parkinson's [315], and vascular dementia [316]. Currently, Yang B et al. found that acupuncture targeting APP/PS1 mice was effective in reducing the relative abundance of Porphyrionaceae and Helicobacter pylori family [317]. Porphyrionaceae [318] and H. pylori [319] have been shown to be associated with Alzheimer's disease. In particular, Porphyromonas gingivalis, a member of the Porphyromonas family, has been shown to cause dysfunction of the cerebral lymphatic system [15]. The specific acupuncture points vary for different diseases, including Baihui (DU20) [320, 321], Hegu (LI4) [322, 323], and Zusanli (ST36) [324, 325]. The application of acupuncture or electroacupuncture in the middle of the treatment process has good prospects, but still needs to be further explored.

5.2.4. Dietary Patterns

Dietary patterns are strongly associated with both gut microbes and the brain [326-328]. A high-fat, high-sugar diet affects both the relative abundance of gut microbes in female mice and leads to a reduced density of astrocytes in the hypothalamus [329-331]. By feeding triple transgenic 3xTg-AD (TG) female mice withcombination of dried nopal, soy, chia oil, and turmeric for 7 months, astrocyte activation was effectively reduced with attenuated neuroinflammation [295]. Interestingly, it was found that water channel proteins from corn, soy, spinach leaves, and tomatoes are homologous to AQP-4 on astrocytes in the human brain. Antibodies against aquaporins produced as a result of food may lead to autoimmune diseases under specific circumstances [332]. These all suggest that dietary patterns may have an impact on the cerebral lymphatic system. And special dietary patterns may be a new way to intervene in the cerebral lymphatic system.

5.2.5. Probiotics, prebiotics or synbiotics

Probiotics, prebiotics and synbiotics are able to reduce astrocyte activation and neuroinflammatory responses by regulating intestinal flora. Ultimately, it may affect the function of the cerebral lymphatic system [130, 333-338]. Lactobacillus rhamnosus administration attenuates microglia and astrocyte activation [339]. Jing Lu et al. found that by giving Lactobacillus acidophilus and Bifidobacterium infantis (LB) to pregnant C57/BL6J female mice, it promoted brain development and reduced astrocyte activation in their offspring [340]. However, due to the slow action of probiotics on intestinal flora and their colonization in the intestine is transient [333, 341]. Therefore, combining probiotics with nanovesicles may be a better strategy [342].

5.2.6. Antibiotics

Antibiotics can have some effects on the gastrointestinal flora [343]. Also, antibiotics can have some effects on the central nervous system [297, 344-348]. For example, combination antibiotic therapy was able to significantly affect the relative abundance of Lachnospiraceae in the gastrointestinal flora. However, it was also able to inhibit the activation of astrocytes [349, 350]. Therefore, we hypothesize that antibiotics may indirectly affect the CNS by affecting the gastrointestinal flora in addition to directly affecting it.

5.2.7. Fecal Materia Medica Transplantation (FMT)

Fecal transplantation is a method to rapidly alter the intestinal flora. It is mainly achieved by transferring intestinal flora from a healthy donor to the patient [351]. Fecal transplantation can reduce astrocyte activation and attenuate neuroinflammation by inhibiting NF-κB signaling pathway [352, 353]. Therefore, considering the importance of astrocytes for maintaining the normal function of the cerebral lymphatic system, fecal transplantation may also be a new idea to intervene in the cerebral lymphatic system.

5.3. Potential targets for the diagnosis and treatment of kidney disease

5.3.1. Diagnosis

Some metabolites in the central nervous system are first transported from the interior of the brain through the glymphatic system to the subarachnoid space and then drained to the peripheral lymphatic system through the meningeal lymphatic vessels. Eventually, metabolic wastes and toxins that are drained to the periphery via the cerebral lymphatic system are excreted through the kidney. Currently, tests for patients with cognitive dysfunction or cerebrovascular disease caused by renal disease are often not specific. In contrast, fluorescent protein tracer technique can help us to understand the basic situation of fluid flow in the body such as blood circulation and lymphatic circulation. And it can help us diagnose diseases by detecting changes in fluid flow [236]. Since both kidney and brain are high blood flow organs, there are more possible scenarios for the application of fluorescent protein tracing technique in the diagnosis of brain and kidney diseases. A recent study conducted by Xiaowen Wang et al. used a single intraperitoneal injection or intravenous injection of adeno-associated viral vector (AAV) to induce the hepatocytes to express fluorescent protein-tagged albumin. This enables stable labeling of blood for more than 3 months and can be progressively metabolized without long-term accumulation in the body [236]. Interestingly, there is a large observable fluid flow in the cerebral lymphatic system, circulatory system, and urinary system. Therefore, by monitoring the entire process of fluorescently labeled metabolic waste moving with fluid flow in the body, we can promptly detect dysfunctions in organs such as the brain and kidney, thus helping us to update our knowledge of many diseases and provide new diagnostic and treatment options.

5.3.2. Treatment

5.3.2.1. Current treatment status

The current main treatments for kidney diseases include dialysis, kidney transplantation, oral administration of activated charcoal with adsorptive properties, and hemoperfusion. Their main purpose is to remove metabolic waste to alleviate the cognitive dysfunction and cerebrovascular damage caused by them [354-357].

5.3.2.1.1. Dialysis

Dialysis is one of the main treatments for patients with end-stage renal disease (ESRD). After receiving hemodialysis treatment, the concentration of toxic proteins such as Aβ in the blood and cerebrospinal fluid of patients decreased significantly. This suggests that hemodialysis may inhibit the progression of neurodegenerative diseases [218, 358]. And the effect of hemodialysis on the cognitive function of ESRD patients may have a phase change. At the very beginning patients have a dramatic decline in cognitive function. Subsequently, cognitive function improves significantly in ESRD patients early in treatment, but deteriorates in ESRD patients in the intertreatment period [358, 359]. The dramatic decline in cognitive function during dialysis may be related to recurrent cerebral ischemia caused by blood pressure and hemodynamic instability during hemodialysis treatment [359-362]. In addition, patients often have concurrent cerebral edema during hemodialysis, which may also contribute to the dramatic decline in cognitive function during hemodialysis. During rapid dialysis, urea accumulates excessively among astrocytes due to the reduced expression of UT-B on astrocytes in CKD patients. At the same time, the flow of fluid into the brain interstitium increases due to the increased expression of AQP-4. Ultimately, the two together lead to the development of cerebral edema [363]. Therefore, the glymphatic system is important in the development and progression of cerebral edema in CKD patients receiving hemodialysis. In addition, newer methods of hemodialysis such as nocturnal hemodialysis or the use of cooled dialysate can better reduce cognitive dysfunction in patients compared to traditional hemodialysis methods [223]. It is important to note that different dialysis modalities can lead to changes in the incidence of cognitive dysfunction and cerebrovascular disease in ESRD patients. ESRD patients receiving peritoneal dialysis have significantly better cognitive function than those receiving hemodialysis [364]. As a continuous and gentle form of dialysis, peritoneal dialysis is effective in removing metabolic waste without causing dramatic fluctuations in blood pressure and hemodynamics. Therefore, peritoneal dialysis is more conducive to the recovery of cognitive function in ESRD patients. However, it is important to note that peritoneal dialysis improves cognitive function significantly slower than hemodialysis. Therefore, the mode of dialysis needs to be chosen according to the actual situation [365].

In a long-term perspective, dialysis can reduce the likelihood of cerebrovascular disease by reducing endothelial damage due to its filtering and removal of metabolic waste. However, in the short term, dialysis may induce the development of cerebrovascular disease. According to studies, the incidence of stroke is significantly higher in dialysis patients than in the healthy population and is dominated by hemorrhagic strokes. This may be due to the application of antithrombotic drugs during hemodialysis [218]. In addition, the vulnerability to stroke may be related to the dramatic changes in hemodynamics during dialysis and secondary factors such as blood pressure, vascular amyloidosis, and vascular calcification [215]. Therefore, maintaining hemodynamic and blood pressure stability during hemodialysis and applying antithrombotic drugs rationally are particularly important to prevent the occurrence of cerebrovascular diseases. In addition, peritoneal dialysis has a greater advantage in safety compared to hemodialysis because antithrombotic drugs are not required during the process [365].

5.3.2.1.2. Kidney transplantation

Kidney transplantation is the ideal treatment for patients with chronic kidney disease. Studies have shown that kidney transplantation can significantly improve cognitive function in patients. During the three months after kidney transplantation, the cognitive function of transplant patients does not differ from that of dialysis patients, but the concentration of uremic toxins in the blood of transplant patients changes significantly [355]. Three months after the kidney transplantation, the cognitive function of transplant patients begins to improve significantly [223]. In addition, the patient's health status can greatly affect the recovery of cognitive function after kidney transplantation. It has been noted that the cognitive function of patients with kidney transplantation within one year postoperatively does not show much difference depending on their health status. However, after one year postoperatively, cognitive function in patients with frail health status can show a significant decline [366].

5.3.2.1.3. Oral adsorption of activated carbon and blood perfusion

Both oral adsorptive activated charcoal and hemoperfusion apply adsorbents to adsorb uremic toxins produced by metabolism in the body and excrete them. The difference is that orally adsorbed activated charcoal such as AST-120 adsorbs uremic toxins and their precursors produced by microbial metabolism in the gastrointestinal tract and causes their excretion in the feces [367]. AST-120 reduces serum IS levels in a dose-dependent manner, and administration of AST-120 reduces the accumulation of IS and oxidative stress in rats with chronic kidney disease and subsequently improves endothelial dysfunction [367-369]. In contrast, hemoperfusion is the application of adsorbents to absorb small and medium molecular weight metabolic wastes and with protein-bound uremic toxins (PBUTs) in the purification system outside the body [370]. Thus, oral administration of adsorptive activated charcoal and blood perfusion can alleviate to some extent the cognitive dysfunction and cerebrovascular disease caused by the accumulation of uremic toxins.

5.3.2.2. Potential therapeutic approaches based on the cerebral lymphatic system
5.3.2.2.1. Treatment of cognitive dysfunction caused by kidney diseases based on cerebral lymphatic system

Current thinking on this aspect is focused on two perspectives by increasing the efficiency and working time of the cerebral lymphatic system. On the one hand, current studies have shown that AQP-4 facilitators, such as TGN-073, significantly increase the transport of water from cortical areas to the perivascular spaces, which in turn promotes the renewal of interstitial fluid and ultimately leads to enhanced clearance of the glymphatic system [243, 371]. However, at the same time, the use of TGN-073 may increase the incidence of cerebral edema during hemodialysis. And this remains to be further studied. It has been shown that MSC-derived exosomes can effectively inhibit the proliferation and inflammatory response of reactive astrocytes [372]. Therefore, we speculate that MSC-derived exosomes may enhance the clearance function of glymphatic system by repairing damaged astrocytes, and thus improve the cognitive dysfunction of the organism. This remains to be further explored by researchers. On the other hand, the clearance function of the glymphatic system is mainly active in the 3-4 phase of NREM [9]. And patients with chronic kidney disease are often accompanied by sleep disorders [373]. Therefore, by relieving patients' sleep disorders, i.e., prolonging stages 3-4 of NREM, it may provide new ideas for treating cognitive dysfunction in patients with chronic kidney disease.

5.3.2.2.2. Treatment of cognitive dysfunction due to kidney disease based on renal excretory function

Most neurodegenerative diseases usually manifest as cognitive dysfunction, and their pathogenesis is related to protein deposition with altered physicochemical properties [374]. Promoting the clearance of deposited proteins in the brain is important to prevent neurodegenerative diseases or to slow down the process of neurodegenerative diseases. The kidney, as the main organ for excreting metabolic wastes and toxins from the body [206], also plays an important role in the development of neurodegenerative diseases. It has been shown that the kidney has a strong ability to remove β-amyloid and plays a key role in maintaining normal levels of β-amyloid in the blood and brain. Furosemide was able to improve cognitive dysfunction in AD mice by enhancing renal clearance of β-amyloid [128]. Therefore, we speculate that the combination of AQP-4 promoter TGN-073 and diuretics (e.g., furosemide) may both promote the glymphatic system for the clearance of metabolic waste in the brain and enhance the clearance function of the kidney. And this needs to be further validated. In addition, mesenchymal stem cells (MSCs), as a multifunctional cell population with self-renewal and differentiation, have potential therapeutic effects in various renal diseases. MSCs can specifically localize to the injured region of the kidney and exert therapeutic significance by secreting trophic factors or delivering subcellular structures [375]. Numerous studies have shown that MSCs therapy can improve renal function through multiple mechanisms, including anti-inflammatory, anti-apoptotic, anti-fibrotic, and pro-angiogenic. For example, cell therapy with MSCs has been shown to significantly improve the symptoms of AKI patients [376]. Therefore, the improvement of renal excretory function may contribute to the drainage of the glymphatic system and the elimination of metabolic waste in the brain.

These findings suggest that transport between the brain and periphery is important for the clearance of metabolic waste and the maintenance of brain homeostasis. Reduced clearance by the peripheral system may impede circulatory processes and lead to the accumulation of pathological proteins in the central nervous system. Therefore, enhanced peripheral waste removal may alleviate the pathology associated with Aβ in the brain [127]. It may provide a new therapeutic approach for neurodegenerative diseases.

From the present perspective, the cerebral lymphatic system acts as a bridge between the brain and the peripheral system, allowing us to fully understand and appreciate the pathogenesis and potential therapeutic directions of neurodegenerative diseases from a systemic perspective. There are relatively few studies revealing the specific mechanism of the connection between the cerebral lymphatic system and peripheral tissues and organs, which may become a potential development direction and hot spot in this field in the future.

6. Conclusions and future directions

In this review, we first describe the structural components and functional properties of the cerebral lymphatic system. Secondly, we review the evidence for the involvement of the cerebral lymphatic system in some peripheral system diseases. Finally, we describe the role of the cerebral lymphatic system as a potential bridge between the central nervous system and the peripheral system and provide new ideas for the diagnosis and treatment of diseases of the peripheral system.

As a special lymphatic system existing in the central nervous system, the cerebral lymphatic system differs considerably from the peripheral lymphatic system known to us. This is mainly due to the unique flow patterns and pathways of the glymphatic system. This is mainly due to the unique way in which the glymphatic system flows and the pathways through which it flows. Further research on the structure and function of the cerebral lymphatic system is essential for a better understanding of the brain and for exploring further clinical applications. At this stage, there are a number of issues that need to be addressed in the study of the cerebral lymphatic system. First of all, we need to clarify whether there is a certain degree of limitation on the size and type of molecules that pass through the cerebral lymphatic system. This mainly refers to the fluid exchange that occurs at the AQP-4 of the astrocyte endfeet. In the second place, we need to clarify more about the specific locations and mechanisms of solute and immune cell exchange that occur between the meningeal lymphatic vessels and the cerebrospinal fluid at different sites. This will assist us to clarify the specific sites and methods of operation for therapeutic regimens which target the meningeal lymphatic vessels. Furthermore, the specific processes and mechanisms of interaction between the cerebral lymphatic system and the peripheral lymphatic system are still unclear.

As a bridge between the central nervous system and the peripheral system, the cerebral lymphatic system has also attracted our attention to the peripheral system at the same time. As more and more attention is now being paid to the impact of the peripheral system on central nervous system diseases, we have come to realise that the traditional concept of treating the central nervous system separately from the rest of the body is not justified. Diseases of the central nervous system should be treated as systemic diseases. Peripheral organs, tissues and cells such as the liver, kidneys, gastrointestinal tract, skin and mononuclear cells have gradually become new breakthroughs in the treatment of central system diseases. At the same time, the indirect aim of treating peripheral system diseases by improving the health of the central nervous system is also a highly viable potential idea. However, it is important to note that the peripheral system is closely linked to the immune function of the central nervous system. Intervention of peripheral immune cells for dual immunotherapy of the periphery and the centre is a promising area of research, but its efficacy remains to be investigated.

Venous drainage is the main drainage route for metabolic waste in most organs of the body. Thus, tests and analyses of blood in the veins are used as the main diagnostic and therapeutic target for various diseases. However, due to the presence of the blood-brain barrier, the main diagnostic and therapeutic target for central nervous system diseases have been replaced by the cerebrospinal fluid, which usually requires invasive procedures [377-379]. The cerebral lymphatic system, which has recently been shown to be closely associated with the flow of cerebrospinal fluid, has great potential to become a diagnostic and therapeutic target and may play a revolutionary role in the advancement of a non-invasive approach to CNS diagnosis and treatment. At the diagnostic level, the previously proposed method of assessing the flow of the glymphatic system by intrathecal injection of gadolinium contrast agent is an invasive procedure that is only suitable for a limited number of patients with clear clinical indications. Moreover, the tendency of gadolinium accumulating in the central nervous system renders this method as a potential health risk for the patients [380]. Therefore, non-invasive imaging methods that do not require contrast agents are preferred by clinicians. It has been shown that the "Diffusion Tensor Image-Analysis aLong the Perivascular Space (DTI-ALPS)" methods based on non-invasive MRI can effectively detect brain lymphatic system dysfunction and quantify the activity of fluid flow in patients with idiopathic normal pressure hydrocephalus [381]. This has amply demonstrated the feasibility of a non-invasive, effective and standardised clinical imaging method for the cerebral lymphatic system and has inspired further exploration [382-384]. Currently, various imaging modalities targeting the glymphatic system have shown relatively obvious drawbacks. The observation range of two-photon laser scanning microscopy is relatively limited. MRI coupled with intrathecal gadolinium-based contrast agents has poor resolution for delicate anatomical structures [3, 9, 40, 385-387]. Thus, the developing whole brain imaging techniques may enable us to systematically visualize the intact cerebral lymphatic system within the CNS and thus further reveal the specific pathways of fluid flow in the CNS. In the near future, this may become an indispensable basis for the clinical assessment of the function of the cerebral lymphatic system. In terms of treatment, non-invasive physical therapies are gaining more and more attention, such as photobiomodulation (PBM) therapy [388]. In addition, the use of nanoparticle engineering in conjunction with biophotonics during therapeutic procedures may provide better therapeutic benefits due to the high blood-brain barrier crossing properties and low toxicity of nanoparticles [389].

Current treatment options for central nervous system disorders usually consider stem cells as the primary treatment and rarely consider targeting immune cells. This is partly influenced by the traditional view of immune isolation of the brain. On the other hand, there is still a slight gap in research on the types of immune cells in the brain, their distribution, and when and how they function. However, as our understanding of the brain's lymphatic system improves, brain immunotherapy may become an important treatment tool for us in the future. According to current research, the immune response in the brain may be a double-edged sword. In the acute phase, the rapid and powerful immune response produces damage to brain tissue that is difficult to ignore. In the chronic phase, the immune response is able to remove potential pathogenic metabolic wastes from the brain and thus exerts a protective effect. This contributes to the recovery of damaged tissue. Therefore, in the future, we must select individualised treatment plans for different periods of the diseases with full consideration of the dual role of the immune system.

Efficient, low-damage drug delivery systems for the brain are currently receiving increasing attention. On the one hand, many attempts have been made to efficiently deliver drugs from the peripheral system into the central nervous system, such as improving the permeability of the blood-brain barrier, using lipid-soluble drugs, or employing emerging nanoparticle technologies. Rui Liu and his team systematically tracked nonreactive gold nanoclusters delivered to the brain and found that they could cross the blood-brain barrier through active transport by exosomes. At the same time, they can also exit the brain via the glymphatic system. The discovery of the cerebral lymphatic system may mean that we can no longer demand that the drugs cross the blood-brain barrier, but only that the drugs with the most suitable functional characteristics for our therapeutic needs are considered. On the other hand, by exploiting the ability of the glymphatic system to penetrate deeply into the brain parenchyma, we may be able to explore new delivery options for intrathecal or intracerebroventricular drug injections on this basis. Such innovative CNS drug delivery routes could be of great practical interest and research value. For example, it has recently been shown that antagonism of the α3 GABAA receptor enhances the activity of delta waves, thereby promoting deep sleep of animals [389]. So, could we later antagonise this receptor by feeding specific drugs into the cerebral lymphatic system to improve sleep quality or even meet the criteria for clinical sleep treatment? Further research in this area is likely to answer many similar questions.

The cerebral lymphatic system is expected to play a crucial role in the current development of treatments for various diseases, and there is growing evidence to support its potential as a diagnostic and therapeutic target for the prevention and treatment of diseases of the central nervous system and peripheral system. This remains to be further investigated and explored. In conclusion, this review contributes to a better understanding of the structure and function of the cerebral lymphatic system. As a bridge between the central nervous system and the peripheral system, it has enormous potential for new directions in the diagnosis and treatment of various diseases. We hope that this review will be helpful and enlightening for the study of the cerebral lymphatic system.

Acknowledgments

We are grateful to Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University and Department of neurosurgery, Tangdu hospital, Fourth Military Medical University for technical support. This work was supported by the Natural Science Foundation of Shandong Province [ZR2016HP06]; the Shandong Medical and Health Science and Technology Development Project [2019WS605, 202104110334]; the Weifang Medical University Students Innovation and Entrepreneurship Training Program Project [X2021517, X2021516, X2021514]; and the Weifang Science and Technology Development Project [2021YX028].

Funding Statement

We are grateful to Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University and Department of neurosurgery, Tangdu hospital, Fourth Military Medical University for technical support. This work was supported by the Natural Science Foundation of Shandong Province [ZR2016HP06]; the Shandong Medical and Health Science and Technology Development Project [2019WS605, 202104110334]; the Weifang Medical University Students Innovation and Entrepreneurship Training Program Project [X2021517, X2021516, X2021514]; and the Weifang Science and Technology Development Project [2021YX028].

Footnotes

Conflict of interest

All the authors declare that they do not have any conflict of interest.

Author contributions

Conceptualization, H.-X.Z. and D.-Y.F. ; methodology, H.-X.Z. , R.Z., X.-L.Z., Q.L. and Q.X. ; data curation, H.-X.Z. and W.-W.K. ; visualization, H.-X.Z. , W.-B.Z., X.-Y.Y. and L.-L.F. ; writing—original draft preparation, H.-X.Z. , Z.-Y.W. , B.-Y.C. and J.-X.D. ; writing—review and editing, H.-X.Z., F.-H.L., W.-B.Z., R.Z. and Y.Z. ; project administration, H.-X.Z. ,S.-Q.W. and X.-Y.Z. All authors have read and agreed to the published version of the manuscript.

References

  • [1].Sandrone S, Moreno-Zambrano D, Kipnis J, van Gijn J (2019). A (delayed) history of the brain lymphatic system. Nat Med, 25:538-40. [DOI] [PubMed] [Google Scholar]
  • [2].Louveau A, Smirnov I, Keyes TJ, Eccles JD, Rouhani SJ, Peske JD, et al. (2015). Structural and functional features of central nervous system lymphatic vessels. Nature, 523:337-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, et al. (2012). A paravascular pathway facilitates csf flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med, 4:111r-147r. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Taoka T, Masutani Y, Kawai H, Nakane T, Matsuoka K, Yasuno F, et al. (2017). Evaluation of glymphatic system activity with the diffusion mr technique: diffusion tensor image analysis along the perivascular space (dti-alps) in alzheimer's disease cases. Jpn J Radiol, 35:172-8. [DOI] [PubMed] [Google Scholar]
  • [5].Yokota H, Vijayasarathi A, Cekic M, Hirata Y, Linetsky M, Ho M, et al. (2019). Diagnostic performance of glymphatic system evaluation using diffusion tensor imaging in idiopathic normal pressure hydrocephalus and mimickers. Curr Gerontol Geriatr Res, 2019:5675014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Mollgard K, Beinlich F, Kusk P, Miyakoshi LM, Delle C, Pla V, et al. (2023). A mesothelium divides the subarachnoid space into functional compartments. Science, 379:84-8. [DOI] [PubMed] [Google Scholar]
  • [7].Aspelund A, Antila S, Proulx ST, Karlsen TV, Karaman S, Detmar M, et al. (2015). A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J Exp Med, 212:991-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Ma Q, Ineichen BV, Detmar M, Proulx ST (2017). Outflow of cerebrospinal fluid is predominantly through lymphatic vessels and is reduced in aged mice. Nat Commun, 8:1434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Plog BA, Nedergaard M (2018). The glymphatic system in central nervous system health and disease: past, present, and future. Annu Rev Pathol, 13:379-94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Li W, Chen D, Liu N, Luan Y, Zhu S, Wang H (2022). Modulation of lymphatic transport in the central nervous system. Theranostics, 12:1117-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Louveau A, Herz J, Alme MN, Salvador AF, Dong MQ, Viar KE, et al. (2018). Cns lymphatic drainage and neuroinflammation are regulated by meningeal lymphatic vasculature. Nat Neurosci, 21:1380-91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Da MS, Louveau A, Vaccari A, Smirnov I, Cornelison RC, Kingsmore KM, et al. (2018). Functional aspects of meningeal lymphatics in ageing and alzheimer's disease. Nature, 560:185-91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Hsu SJ, Zhang C, Jeong J, Lee SI, McConnell M, Utsumi T, et al. (2021). Enhanced meningeal lymphatic drainage ameliorates neuroinflammation and hepatic encephalopathy in cirrhotic rats. Gastroenterology, 160:1315-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].He XF, Li LL, Xian WB, Li MY, Zhang LY, Xu JH, et al. (2021). Chronic colitis exacerbates nlrp3-dependent neuroinflammation and cognitive impairment in middle-aged brain. J Neuroinflammation, 18:153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Chi L, Cheng X, Lin L, Yang T, Sun J, Feng Y, et al. (2021). Porphyromonas gingivalis-induced cognitive impairment is associated with gut dysbiosis, neuroinflammation, and glymphatic dysfunction. Front Cell Infect Microbiol, 11:755925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Penton AA, Lau H, Babikian VL, Shulman J, Cervantes-Arslanian A, Gangadhara S, et al. (2020). Chronic kidney disease as risk factor for enlarged perivascular spaces in patients with stroke and relation to racial group. Stroke, 51:3348-51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Hsu M, Rayasam A, Kijak JA, Choi YH, Harding JS, Marcus SA, et al. (2019). Neuroinflammation-induced lymphangiogenesis near the cribriform plate contributes to drainage of cns-derived antigens and immune cells. Nat Commun, 10:229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Lundgaard I, Li B, Xie L, Kang H, Sanggaard S, Haswell JD, et al. (2015). Direct neuronal glucose uptake heralds activity-dependent increases in cerebral metabolism. Nat Commun, 6:6807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Lundgaard I, Lu ML, Yang E, Peng W, Mestre H, Hitomi E, et al. (2017). Glymphatic clearance controls state-dependent changes in brain lactate concentration. J Cereb Blood Flow Metab, 37:2112-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Koh L, Zakharov A, Johnston M (2005). Integration of the subarachnoid space and lymphatics: is it time to embrace a new concept of cerebrospinal fluid absorption? Cerebrospinal Fluid Res, 2:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Jessen NA, Munk AS, Lundgaard I, Nedergaard M (2015). The glymphatic system: a beginner's guide. Neurochem Res, 40:2583-99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].He W, You J, Wan Q, Xiao K, Chen K, Lu Y, et al. (2020). The anatomy and metabolome of the lymphatic system in the brain in health and disease. Brain Pathol, 30:392-404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Raper D, Louveau A, Kipnis J (2016). How do meningeal lymphatic vessels drain the cns? Trends Neurosci, 39:581-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Da MS, Papadopoulos Z, Dykstra T, Brase L, Farias FG, Wall M, et al. (2021). Meningeal lymphatics affect microglia responses and anti-abeta immunotherapy. Nature, 593:255-60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Wu CH, Lirng JF, Ling YH, Wang YF, Wu HM, Fuh JL, et al. (2021). Noninvasive characterization of human glymphatics and meningeal lymphatics in an in vivo model of blood-brain barrier leakage. Ann Neurol, 89:111-24. [DOI] [PubMed] [Google Scholar]
  • [26].Kaliya-Perumal AK, Lu ML, Kao FC, Niu CC (2017). Delayed extensive lumbar sub-dural effusion following discectomy - clinical imaging and case report. Biomedicine (Taipei), 7:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Ahn JH, Cho H, Kim JH, Kim SH, Ham JS, Park I, et al. (2019). Meningeal lymphatic vessels at the skull base drain cerebrospinal fluid. Nature, 572:62-6. [DOI] [PubMed] [Google Scholar]
  • [28].Hu X, Deng Q, Ma L, Li Q, Chen Y, Liao Y, et al. (2020). Meningeal lymphatic vessels regulate brain tumor drainage and immunity. Cell Res, 30:229-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Kwon S, Moreno-Gonzalez I, Taylor-Presse K, Edwards IG, Gamez N, Calderon O, et al. (2019). Impaired peripheral lymphatic function and cerebrospinal fluid outflow in a mouse model of alzheimer's disease. J Alzheimers Dis, 69:585-93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Wang L, Zhang Y, Zhao Y, Marshall C, Wu T, Xiao M (2019). Deep cervical lymph node ligation aggravates ad-like pathology of app/ps1 mice. Brain Pathol, 29:176-92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Das NS, Delivanoglou N, Da MS (2021). Cns-draining meningeal lymphatic vasculature: roles, conundrums and future challenges. Front Pharmacol, 12:655052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Krahn V (1982). The pia mater at the site of the entry of blood vessels into the central nervous system. Anat Embryol (Berl), 164:257-63. [DOI] [PubMed] [Google Scholar]
  • [33].Zhang ET, Inman CB, Weller RO (1990). Interrelationships of the pia mater and the perivascular (virchow-robin) spaces in the human cerebrum. J Anat, 170:111-23. [PMC free article] [PubMed] [Google Scholar]
  • [34].Bacyinski A, Xu M, Wang W, Hu J (2017). The paravascular pathway for brain waste clearance: current understanding, significance and controversy. Front Neuroanat, 11:101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Hannocks MJ, Pizzo ME, Huppert J, Deshpande T, Abbott NJ, Thorne RG, et al. (2018). Molecular characterization of perivascular drainage pathways in the murine brain. J Cereb Blood Flow Metab, 38:669-86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Sixt M, Engelhardt B, Pausch F, Hallmann R, Wendler O, Sorokin LM (2001). Endothelial cell laminin isoforms, laminins 8 and 10, play decisive roles in t cell recruitment across the blood-brain barrier in experimental autoimmune encephalomyelitis. J Cell Biol, 153:933-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Ineichen BV, Okar SV, Proulx ST, Engelhardt B, Lassmann H, Reich DS (2022). Perivascular spaces and their role in neuroinflammation. Neuron, 110:3566-81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Lim AT, Chandra RV, Trost NM, McKelvie PA, Stuckey SL (2015). Large anterior temporal virchow-robin spaces: unique mr imaging features. Neuroradiology, 57:491-9. [DOI] [PubMed] [Google Scholar]
  • [39].Nedergaard M (2013). Neuroscience. Garbage truck of the brain. Science, 340:1529-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Iliff JJ, Wang M, Zeppenfeld DM, Venkataraman A, Plog BA, Liao Y, et al. (2013). Cerebral arterial pulsation drives paravascular csf-interstitial fluid exchange in the murine brain. J Neurosci, 33:18190-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Kress BT, Iliff JJ, Xia M, Wang M, Wei HS, Zeppenfeld D, et al. (2014). Impairment of paravascular clearance pathways in the aging brain. Ann Neurol, 76:845-61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Rennels ML, Gregory TF, Blaumanis OR, Fujimoto K, Grady PA (1985). Evidence for a 'paravascular' fluid circulation in the mammalian central nervous system, provided by the rapid distribution of tracer protein throughout the brain from the subarachnoid space. Brain Res, 326:47-63. [DOI] [PubMed] [Google Scholar]
  • [43].Shibata M, Yamada S, Kumar SR, Calero M, Bading J, Frangione B, et al. (2000). Clearance of alzheimer's amyloid-ss (1-40) peptide from brain by ldl receptor-related protein-1 at the blood-brain barrier. J Clin Invest, 106:1489-99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Perry VH, Teeling J (2013). Microglia and macrophages of the central nervous system: the contribution of microglia priming and systemic inflammation to chronic neurodegeneration. Semin Immunopathol, 35:601-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].London A, Cohen M, Schwartz M (2013). Microglia and monocyte-derived macrophages: functionally distinct populations that act in concert in cns plasticity and repair. Front Cell Neurosci, 7:34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Weller RO, Djuanda E, Yow HY, Carare RO (2009). Lymphatic drainage of the brain and the pathophysiology of neurological disease. Acta Neuropathol, 117:1-14. [DOI] [PubMed] [Google Scholar]
  • [47].Pollock H, Hutchings M, Weller RO, Zhang ET (1997). Perivascular spaces in the basal ganglia of the human brain: their relationship to lacunes. J Anat, 191(Pt 3):337-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Weller RO, Hawkes CA, Kalaria RN, Werring DJ, Carare RO (2015). White matter changes in dementia: role of impaired drainage of interstitial fluid. Brain Pathol, 25:63-78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Carare RO, Bernardes-Silva M, Newman TA, Page AM, Nicoll JA, Perry VH, et al. (2008). Solutes, but not cells, drain from the brain parenchyma along basement membranes of capillaries and arteries: significance for cerebral amyloid angiopathy and neuroimmunology. Neuropathol Appl Neurobiol, 34:131-44. [DOI] [PubMed] [Google Scholar]
  • [50].Schley D, Carare-Nnadi R, Please CP, Perry VH, Weller RO (2006). Mechanisms to explain the reverse perivascular transport of solutes out of the brain. J Theor Biol, 238:962-74. [DOI] [PubMed] [Google Scholar]
  • [51].Peng W, Achariyar TM, Li B, Liao Y, Mestre H, Hitomi E, et al. (2016). Suppression of glymphatic fluid transport in a mouse model of alzheimer’s disease. Neurobiol Dis, 93:215-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Li P, Zhao F, Liu P (2014). Multiple large dilated virchow-robin spaces in a 12-year-old with neurofibromatosis type 2. Pediatr Neurol, 51:856-7. [DOI] [PubMed] [Google Scholar]
  • [53].Hartel C, Bachmann S, Bonnemann C, Meinecke P, Sperner J (2005). Familial megalencephaly with dilated virchow-robin spaces in magnetic resonance imaging: an autosomal recessive trait? Clin Dysmorphol, 14:31-4. [PubMed] [Google Scholar]
  • [54].Passiak BS, Liu D, Kresge HA, Cambronero FE, Pechman KR, Osborn KE, et al. (2019). Perivascular spaces contribute to cognition beyond other small vessel disease markers. Neurology, 92:e1309-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Park CH, Shin NY, Nam Y, Yoon U, Ahn K, Lee SK (2023). Characteristics of perivascular space dilatation in normal aging. Hum Brain Mapp. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Mestre H, Verma N, Greene TD, Lin LA, Ladron-de-Guevara A, Sweeney AM, et al. (2022). Periarteriolar spaces modulate cerebrospinal fluid transport into brain and demonstrate altered morphology in aging and alzheimer's disease. Nat Commun, 13:3897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Bown CW, Carare RO, Schrag MS, Jefferson AL (2022). Physiology and clinical relevance of enlarged perivascular spaces in the aging brain. Neurology, 98:107-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Dubost F, Yilmaz P, Adams H, Bortsova G, Ikram MA, Niessen W, et al. (2019). Enlarged perivascular spaces in brain mri: automated quantification in four regions. Neuroimage, 185:534-44. [DOI] [PubMed] [Google Scholar]
  • [59].Diaz-Castro B, Robel S, Mishra A (2023). Astrocyte endfeet in brain function and pathology: open questions. Annu Rev Neurosci. [DOI] [PubMed] [Google Scholar]
  • [60].Clement T, Rodriguez-Grande B, Badaut J (2020). Aquaporins in brain edema. J Neurosci Res, 98:9-18. [DOI] [PubMed] [Google Scholar]
  • [61].Pati R, Palazzo C, Valente O, Abbrescia P, Messina R, Surdo NC, et al. (2022). The readthrough isoform AQP-4ex is constitutively phosphorylated in the perivascular astrocyte endfeet of human brain. Biomolecules, 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Acosta C, Anderson HD, Anderson CM (2017). Astrocyte dysfunction in alzheimer disease. J Neurosci Res, 95:2430-47. [DOI] [PubMed] [Google Scholar]
  • [63].Thomas JL, Jacob L, Boisserand L (2019). [lymphatic system in central nervous system]. Med Sci (Paris), 35:55-61. [DOI] [PubMed] [Google Scholar]
  • [64].Tice C, McDevitt J, Langford D (2020). Astrocytes, hiv and the glymphatic system: a disease of disrupted waste management? Front Cell Infect Microbiol, 10:523379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Silva I, Silva J, Ferreira R, Trigo D (2021). Glymphatic system, AQP-4, and their implications in alzheimer's disease. Neurol Res Pract, 3:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].He XF, Liu DX, Zhang Q, Liang FY, Dai GY, Zeng JS, et al. (2017). Voluntary exercise promotes glymphatic clearance of amyloid beta and reduces the activation of astrocytes and microglia in aged mice. Front Mol Neurosci, 10:144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Yankova G, Bogomyakova O, Tulupov A (2021). The glymphatic system and meningeal lymphatics of the brain: new understanding of brain clearance. Rev Neurosci, 32:693-705. [DOI] [PubMed] [Google Scholar]
  • [68].Tamura R, Yoshida K, Toda M (2020). Current understanding of lymphatic vessels in the central nervous system. Neurosurg Rev, 43:1055-64. [DOI] [PubMed] [Google Scholar]
  • [69].Reeves BC, Karimy JK, Kundishora AJ, Mestre H, Cerci HM, Matouk C, et al. (2020). Glymphatic system impairment in alzheimer's disease and idiopathic normal pressure hydrocephalus. Trends Mol Med, 26:285-95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Wang Y, Huang C, Guo Q, Chu H (2022). Aquaporin-4 and cognitive disorders. Aging Dis, 13:61-72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Ishida K, Yamada K, Nishiyama R, Hashimoto T, Nishida I, Abe Y, et al. (2022). Glymphatic system clears extracellular tau and protects from tau aggregation and neurodegeneration. J Exp Med, 219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Mestre H, Mori Y, Nedergaard M (2020). The brain's glymphatic system: current controversies. Trends Neurosci, 43:458-66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Ringstad G, Vatnehol S, Eide PK (2017). Glymphatic mri in idiopathic normal pressure hydrocephalus. Brain, 140:2691-705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Hablitz LM, Pla V, Giannetto M, Vinitsky HS, Staeger FF, Metcalfe T, et al. (2020). Circadian control of brain glymphatic and lymphatic fluid flow. Nat Commun, 11:4411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Brinker T, Stopa E, Morrison J, Klinge P (2014). A new look at cerebrospinal fluid circulation. Fluids Barriers CNS, 11:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Tumani H, Huss A, Bachhuber F (2017). The cerebrospinal fluid and barriers - anatomic and physiologic considerations. Handb Clin Neurol, 146:21-32. [DOI] [PubMed] [Google Scholar]
  • [77].Atchley TJ, Vukic B, Vukic M, Walters BC (2022). Review of cerebrospinal fluid physiology and dynamics: a call for medical education reform. Neurosurgery, 91:1-7. [DOI] [PubMed] [Google Scholar]
  • [78].Oreskovic D, Klarica M (2010). The formation of cerebrospinal fluid: nearly a hundred years of interpretations and misinterpretations. Brain Res Rev, 64:241-62. [DOI] [PubMed] [Google Scholar]
  • [79].Bulat M, Klarica M (2011). Recent insights into a new hydrodynamics of the cerebrospinal fluid. Brain Res Rev, 65:99-112. [DOI] [PubMed] [Google Scholar]
  • [80].Rasmussen MK, Mestre H, Nedergaard M (2018). The glymphatic pathway in neurological disorders. Lancet Neurol, 17:1016-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Oliver G, Kipnis J, Randolph GJ, Harvey NL (2020). The lymphatic vasculature in the 21(st) century: novel functional roles in homeostasis and disease. Cell, 182:270-96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Bohr T, Hjorth PG, Holst SC, Hrabetova S, Kiviniemi V, Lilius T, et al. (2022). The glymphatic system: current understanding and modeling. iScience, 25:104987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Mestre H, Hablitz LM, Xavier AL, Feng W, Zou W, Pu T, et al. (2018). Aquaporin-4-dependent glymphatic solute transport in the rodent brain. Elife, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Jiang H, Wei H, Zhou Y, Xiao X, Zhou C, Ji X (2022). Overview of the meningeal lymphatic vessels in aging and central nervous system disorders. Cell Biosci, 12:202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Davson H, Domer FR, Hollingsworth JR (1973). The mechanism of drainage of the cerebrospinal fluid. Brain, 96:329-36. [DOI] [PubMed] [Google Scholar]
  • [86].Bradbury MW, Westrop RJ (1983). Factors influencing exit of substances from cerebrospinal fluid into deep cervical lymph of the rabbit. J Physiol, 339:519-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Weller RO (1998). Pathology of cerebrospinal fluid and interstitial fluid of the cns: significance for alzheimer disease, prion disorders and multiple sclerosis. J Neuropathol Exp Neurol, 57:885-94. [DOI] [PubMed] [Google Scholar]
  • [88].Rasmussen MK, Mestre H, Nedergaard M (2022). Fluid transport in the brain. Physiol Rev, 102:1025-151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Liebner S, Dijkhuizen RM, Reiss Y, Plate KH, Agalliu D, Constantin G (2018). Functional morphology of the blood-brain barrier in health and disease. Acta Neuropathol, 135:311-36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Achariyar TM, Li B, Peng W, Verghese PB, Shi Y, McConnell E, et al. (2016). Glymphatic distribution of csf-derived apoe into brain is isoform specific and suppressed during sleep deprivation. Mol Neurodegener, 11:74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Grapp M, Wrede A, Schweizer M, Huwel S, Galla HJ, Snaidero N, et al. (2013). Choroid plexus transcytosis and exosome shuttling deliver folate into brain parenchyma. Nat Commun, 4:2123. [DOI] [PubMed] [Google Scholar]
  • [92].Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV (2019). Blood-brain barrier: from physiology to disease and back. Physiol Rev, 99:21-78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Cox MF, Hascup ER, Bartke A, Hascup KN (2022). Friend or foe? Defining the role of glutamate in aging and alzheimer's disease. Front Aging, 3:929474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Xu Z, Xiao N, Chen Y, Huang H, Marshall C, Gao J, et al. (2015). Deletion of aquaporin-4 in app/ps1 mice exacerbates brain abeta accumulation and memory deficits. Mol Neurodegener, 10:58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Keech CA, Gutierrez-Hartmann A (1989). Analysis of rat prolactin promoter sequences that mediate pituitary-specific and 3',5'-cyclic adenosine monophosphate-regulated gene expression in vivo. Mol Endocrinol, 3:832-9. [DOI] [PubMed] [Google Scholar]
  • [96].Foldi M (1977). Prelymphatic-lymphatic drainage of the brain. Am Heart J, 93:121-4. [DOI] [PubMed] [Google Scholar]
  • [97].Broadwell RD, Salcman M (1981). Expanding the definition of the blood-brain barrier to protein. Proc Natl Acad Sci U S A, 78:7820-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Sanmarti M, Ibanez L, Huertas S, Badenes D, Dalmau D, Slevin M, et al. (2014). Hiv-associated neurocognitive disorders. J Mol Psychiatry, 2:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Li X, Qi L, Yang D, Hao S, Zhang F, Zhu X, et al. (2022). Meningeal lymphatic vessels mediate neurotropic viral drainage from the central nervous system. Nat Neurosci, 25:577-87. [DOI] [PubMed] [Google Scholar]
  • [100].Cugurra A, Mamuladze T, Rustenhoven J, Dykstra T, Beroshvili G, Greenberg ZJ, et al. (2021). Skull and vertebral bone marrow are myeloid cell reservoirs for the meninges and cns parenchyma. Science, 373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Van Hove H, Martens L, Scheyltjens I, De Vlaminck K, Pombo AA, De Prijck S, et al. (2019). A single-cell atlas of mouse brain macrophages reveals unique transcriptional identities shaped by ontogeny and tissue environment. Nat Neurosci, 22:1021-35. [DOI] [PubMed] [Google Scholar]
  • [102].Derecki NC, Cardani AN, Yang CH, Quinnies KM, Crihfield A, Lynch KR, et al. (2010). Regulation of learning and memory by meningeal immunity: a key role for il-4. J Exp Med, 207:1067-80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Filiano AJ, Xu Y, Tustison NJ, Marsh RL, Baker W, Smirnov I, et al. (2016). Unexpected role of interferon-gamma in regulating neuronal connectivity and social behaviour. Nature, 535:425-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Brombacher TM, Nono JK, De Gouveia KS, Makena N, Darby M, Womersley J, et al. (2017). Il-13-mediated regulation of learning and memory. J Immunol, 198:2681-8. [DOI] [PubMed] [Google Scholar]
  • [105].Foldi M, Gellert A, Kozma M, Poberai M, Zoltan OT, Csanda E (1966). New contributions to the anatomical connections of the brain and the lymphatic system. Acta Anat (Basel), 64:498-505. [DOI] [PubMed] [Google Scholar]
  • [106].Kipnis J (2016). Multifaceted interactions between adaptive immunity and the central nervous system. Science, 353:766-71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Herz J, Filiano AJ, Wiltbank AT, Yogev N, Kipnis J (2017). Myeloid cells in the central nervous system. Immunity, 46:943-56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Norris GT, Kipnis J (2019). Immune cells and cns physiology: microglia and beyond. J Exp Med, 216:60-70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Choi GB, Yim YS, Wong H, Kim S, Kim H, Kim SV, et al. (2016). The maternal interleukin-17a pathway in mice promotes autism-like phenotypes in offspring. Science, 351:933-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Alves DLK, Rustenhoven J, Kipnis J (2020). Meningeal immunity and its function in maintenance of the central nervous system in health and disease. Annu Rev Immunol, 38:597-620. [DOI] [PubMed] [Google Scholar]
  • [111].Mrdjen D, Pavlovic A, Hartmann FJ, Schreiner B, Utz SG, Leung BP, et al. (2018). High-dimensional single-cell mapping of central nervous system immune cells reveals distinct myeloid subsets in health, aging, and disease. Immunity, 48:380-95. [DOI] [PubMed] [Google Scholar]
  • [112].Louveau A, Plog BA, Antila S, Alitalo K, Nedergaard M, Kipnis J (2017). Understanding the functions and relationships of the glymphatic system and meningeal lymphatics. J Clin Invest, 127:3210-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Planas AM, Gomez-Choco M, Urra X, Gorina R, Caballero M, Chamorro A (2012). Brain-derived antigens in lymphoid tissue of patients with acute stroke. J Immunol, 188:2156-63. [DOI] [PubMed] [Google Scholar]
  • [114].Chen J, Wang L, Xu H, Wang Y, Liang Q (2021). The lymphatic drainage system of the cns plays a role in lymphatic drainage, immunity, and neuroinflammation in stroke. J Leukoc Biol, 110:283-91. [DOI] [PubMed] [Google Scholar]
  • [115].Engelhardt B, Carare RO, Bechmann I, Flugel A, Laman JD, Weller RO (2016). Vascular, glial, and lymphatic immune gateways of the central nervous system. Acta Neuropathol, 132:317-38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Jian Z, Liu R, Zhu X, Smerin D, Zhong Y, Gu L, et al. (2019). The involvement and therapy target of immune cells after ischemic stroke. Front Immunol, 10:2167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Rustenhoven J, Drieu A, Mamuladze T, de Lima KA, Dykstra T, Wall M, et al. (2021). Functional characterization of the dural sinuses as a neuroimmune interface. Cell, 184:1000-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Brioschi S, Wang WL, Peng V, Wang M, Shchukina I, Greenberg ZJ, et al. (2021). Heterogeneity of meningeal b cells reveals a lymphopoietic niche at the cns borders. Science, 373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Abbott NJ (2005). Dynamics of cns barriers: evolution, differentiation, and modulation. Cell Mol Neurobiol, 25:5-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Jaunmuktane Z, Mead S, Ellis M, Wadsworth JD, Nicoll AJ, Kenny J, et al. (2015). Evidence for human transmission of amyloid-beta pathology and cerebral amyloid angiopathy. Nature, 525:247-50. [DOI] [PubMed] [Google Scholar]
  • [121].Eisele YS, Fritschi SK, Hamaguchi T, Obermuller U, Fuger P, Skodras A, et al. (2014). Multiple factors contribute to the peripheral induction of cerebral beta-amyloidosis. J Neurosci, 34:10264-73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Eisele YS, Obermuller U, Heilbronner G, Baumann F, Kaeser SA, Wolburg H, et al. (2010). Peripherally applied abeta-containing inoculates induce cerebral beta-amyloidosis. Science, 330:980-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Ritchie DL, Adlard P, Peden AH, Lowrie S, Le Grice M, Burns K, et al. (2017). Amyloid-beta accumulation in the cns in human growth hormone recipients in the uk. Acta Neuropathol, 134:221-40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Naganawa S, Nakane T, Kawai H, Taoka T (2017). Gd-based contrast enhancement of the perivascular spaces in the basal ganglia. Magn Reson Med Sci, 16:61-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Provenzano DA, Pellis Z, DeRiggi L (2019). Fatal gadolinium-induced encephalopathy following accidental intrathecal administration: a case report and a comprehensive evidence-based review. Reg Anesth Pain Med. [DOI] [PubMed] [Google Scholar]
  • [126].Gao YF, Yang ZQ (2005). Experimental study of cranial-cervical lymph return in rabbit. Zhonghua Er Bi Yan Hou Tou Jing Wai Ke Za Zhi, 40:182-5. [PubMed] [Google Scholar]
  • [127].Xiang Y, Bu XL, Liu YH, Zhu C, Shen LL, Jiao SS, et al. (2015). Physiological amyloid-beta clearance in the periphery and its therapeutic potential for alzheimer's disease. Acta Neuropathol, 130:487-99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Tian DY, Cheng Y, Zhuang ZQ, He CY, Pan QG, Tang MZ, et al. (2021). Physiological clearance of amyloid-beta by the kidney and its therapeutic potential for alzheimer's disease. Mol Psychiatry, 26:6074-82. [DOI] [PubMed] [Google Scholar]
  • [129].Guo M, Peng J, Huang X, Xiao L, Huang F, Zuo Z (2021). Gut microbiome features of chinese patients newly diagnosed with alzheimer's disease or mild cognitive impairment. J Alzheimers Dis, 80:299-310. [DOI] [PubMed] [Google Scholar]
  • [130].Wu S, Liu X, Jiang R, Yan X, Ling Z (2021). Roles and mechanisms of gut microbiota in patients with alzheimer's disease. Front Aging Neurosci, 13:650047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Tamaki C, Ohtsuki S, Iwatsubo T, Hashimoto T, Yamada K, Yabuki C, et al. (2006). Major involvement of low-density lipoprotein receptor-related protein 1 in the clearance of plasma free amyloid beta-peptide by the liver. Pharm Res, 23:1407-16. [DOI] [PubMed] [Google Scholar]
  • [132].Wang J, Gu BJ, Masters CL, Wang YJ (2017). A systemic view of alzheimer disease - insights from amyloid-beta metabolism beyond the brain. Nat Rev Neurol, 13:612-23. [DOI] [PubMed] [Google Scholar]
  • [133].Cheng Y, Wang YJ (2020). Meningeal lymphatic vessels: a drain of the brain involved in neurodegeneration? Neurosci Bull, 36:557-60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Brundin P, Dave KD, Kordower JH (2017). Therapeutic approaches to target alpha-synuclein pathology. Exp Neurol, 298:225-35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Gibbons GS, Lee V, Trojanowski JQ (2019). Mechanisms of cell-to-cell transmission of pathological tau: a review. JAMA Neurol, 76:101-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Goedert M, Eisenberg DS, Crowther RA (2017). Propagation of tau aggregates and neurodegeneration. Annu Rev Neurosci, 40:189-210. [DOI] [PubMed] [Google Scholar]
  • [137].Jagust W (2018). Imaging the evolution and pathophysiology of alzheimer disease. Nat Rev Neurosci, 19:687-700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Shankar GM, Li S, Mehta TH, Garcia-Munoz A, Shepardson NE, Smith I, et al. (2008). Amyloid-beta protein dimers isolated directly from alzheimer's brains impair synaptic plasticity and memory. Nat Med, 14:837-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Thal DR, Rub U, Orantes M, Braak H (2002). Phases of a beta-deposition in the human brain and its relevance for the development of ad. Neurology, 58:1791-800. [DOI] [PubMed] [Google Scholar]
  • [140].Wang J, Jin WS, Bu XL, Zeng F, Huang ZL, Li WW, et al. (2018). Physiological clearance of tau in the periphery and its therapeutic potential for tauopathies. Acta Neuropathol, 136:525-36. [DOI] [PubMed] [Google Scholar]
  • [141].Greenwood-Van MB, Johnson AC, Grundy D (2017). Gastrointestinal physiology and function. Handb Exp Pharmacol, 239:1-16. [DOI] [PubMed] [Google Scholar]
  • [142].Latorre R, Sternini C, De Giorgio R, Greenwood-Van MB (2016). Enteroendocrine cells: a review of their role in brain-gut communication. Neurogastroenterol Motil, 28:620-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Jiang C, Li G, Huang P, Liu Z, Zhao B (2017). The gut microbiota and alzheimer's disease. J Alzheimers Dis, 58:1-15. [DOI] [PubMed] [Google Scholar]
  • [144].Mulak A, Bonaz B (2015). Brain-gut-microbiota axis in parkinson's disease. World J Gastroenterol, 21:10609-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].Kalogeris T, Baines CP, Krenz M, Korthuis RJ (2016). Ischemia/reperfusion. Compr Physiol, 7:113-70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Horsager J, Andersen KB, Knudsen K, Skjaerbaek C, Fedorova TD, Okkels N, et al. (2020). Brain-first versus body-first parkinson's disease: a multimodal imaging case-control study. Brain, 143:3077-88. [DOI] [PubMed] [Google Scholar]
  • [147].Fu P, Gao M, Yung K (2020). Association of intestinal disorders with parkinson's disease and alzheimer's disease: a systematic review and meta-analysis. ACS Chem Neurosci, 11:395-405. [DOI] [PubMed] [Google Scholar]
  • [148].Wang X, Wan J, Wang M, Zhang Y, Wu K, Yang F (2022). Multiple sclerosis and inflammatory bowel disease: a systematic review and meta-analysis. Ann Clin Transl Neurol, 9:132-40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [149].Cryan JF, O'Riordan KJ, Cowan C, Sandhu KV, Bastiaanssen T, Boehme M, et al. (2019). The microbiota-gut-brain axis. Physiol Rev, 99:1877-2013. [DOI] [PubMed] [Google Scholar]
  • [150].Chen Z, Maqbool J, Sajid F, Hussain G, Sun T (2021). Human gut microbiota and its association with pathogenesis and treatments of neurodegenerative diseases. Microb Pathog, 150:104675. [DOI] [PubMed] [Google Scholar]
  • [151].Collins SM, Surette M, Bercik P (2012). The interplay between the intestinal microbiota and the brain. Nat Rev Microbiol, 10:735-42. [DOI] [PubMed] [Google Scholar]
  • [152].Sharkey KA, Beck PL, McKay DM (2018). Neuroimmunophysiology of the gut: advances and emerging concepts focusing on the epithelium. Nat Rev Gastroenterol Hepatol, 15:765-84. [DOI] [PubMed] [Google Scholar]
  • [153].Gunther C, Rothhammer V, Karow M, Neurath M, Winner B (2021). The gut-brain axis in inflammatory bowel disease-current and future perspectives. Int J Mol Sci, 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Carrascal L, Vazquez-Carretero MD, Garcia-Miranda P, Fontan-Lozano A, Calonge ML, Ilundain AA, et al. (2022). Acute colon inflammation triggers primary motor cortex glial activation, neuroinflammation, neuronal hyperexcitability, and motor coordination deficits. Int J Mol Sci, 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [155].Costa D, Bon-Frauches AC, Silva A, Lima-Junior R, Martins CS, Leitao R, et al. (2019). 5-fluorouracil induces enteric neuron death and glial activation during intestinal mucositis via a s100b-rage-nfkappab-dependent pathway. Sci Rep, 9:665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [156].Vitali R, Prioreschi C, Lorenzo RL, Colantoni E, Giovannini D, Frusciante S, et al. (2022). Gut-brain axis: insights from hippocampal neurogenesis and brain tumor development in a mouse model of experimental colitis induced by dextran sodium sulfate. Int J Mol Sci, 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [157].Rustenhoven J, Tanumihardja C, Kipnis J (2021). Cerebrovascular anomalies: perspectives from immunology and cerebrospinal fluid flow. Circ Res, 129:174-94. [DOI] [PubMed] [Google Scholar]
  • [158].Qiao CM, Sun MF, Jia XB, Li Y, Zhang BP, Zhao LP, et al. (2020). Sodium butyrate exacerbates parkinson's disease by aggravating neuroinflammation and colonic inflammation in mptp-induced mice model. Neurochem Res, 45:2128-42. [DOI] [PubMed] [Google Scholar]
  • [159].Bostanciklioglu M (2020). SARS-cov2 entry and spread in the lymphatic drainage system of the brain. Brain Behav Immun, 87:122-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Bostanciklioglu M (2020). Temporal correlation between neurological and gastrointestinal symptoms of SARS-cov-2. Inflamm Bowel Dis, 26:e89-91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [161].Iida Y, Honda K, Suzuki T, Matsukawa S, Kawai T, Shimahara T, et al. (2004). Brain abscess in which porphyromonas gingivalis was detected in cerebrospinal fluid. Br J Oral Maxillofac Surg, 42:180. [DOI] [PubMed] [Google Scholar]
  • [162].Hajishengallis G, Chavakis T (2021). Local and systemic mechanisms linking periodontal disease and inflammatory comorbidities. Nat Rev Immunol, 21:426-40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].Bai Z, Chen D, Wang L, Zhao Y, Liu T, Yu Y, et al. (2019). Cerebrospinal fluid and blood cytokines as biomarkers for multiple sclerosis: a systematic review and meta-analysis of 226 studies with 13,526 multiple sclerosis patients. Front Neurosci, 13:1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [164].Takeuchi O, Akira S (2010). Pattern recognition receptors and inflammation. Cell, 140:805-20. [DOI] [PubMed] [Google Scholar]
  • [165].Nielsen S, Nagelhus EA, Amiry-Moghaddam M, Bourque C, Agre P, Ottersen OP (1997). Specialized membrane domains for water transport in glial cells: high-resolution immunogold cytochemistry of aquaporin-4 in rat brain. J Neurosci, 17:171-80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [166].Bauernfeind FG, Horvath G, Stutz A, Alnemri ES, MacDonald K, Speert D, et al. (2009). Cutting edge: nf-kappab activating pattern recognition and cytokine receptors license nlrp3 inflammasome activation by regulating nlrp3 expression. J Immunol, 183:787-91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Willingham SB, Allen IC, Bergstralh DT, Brickey WJ, Huang MT, Taxman DJ, et al. (2009). Nlrp3 (nalp3, cryopyrin) facilitates in vivo caspase-1 activation, necrosis, and hmgb1 release via inflammasome-dependent and -independent pathways. J Immunol, 183:2008-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [168].Shao BZ, Xu ZQ, Han BZ, Su DF, Liu C (2015). Nlrp3 inflammasome and its inhibitors: a review. Front Pharmacol, 6:262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].Natale G, Biagioni F, Busceti CL, Gambardella S, Limanaqi F, Fornai F (2019). Trem receptors connecting bowel inflammation to neurodegenerative disorders. Cells, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [170].Zhang C, Kan X, Zhang B, Ni H, Shao J (2022). The role of triggering receptor expressed on myeloid cells-1 (trem-1) in central nervous system diseases. Mol Brain, 15:84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [171].Rosciszewski G, Cadena V, Murta V, Lukin J, Villarreal A, Roger T, et al. (2018). Toll-like receptor 4 (tlr4) and triggering receptor expressed on myeloid cells-2 (trem-2) activation balance astrocyte polarization into a proinflammatory phenotype. Mol Neurobiol, 55:3875-88. [DOI] [PubMed] [Google Scholar]
  • [172].Zhao YF, Wei DN, Tang Y (2021). Gut microbiota regulate astrocytic functions in the brain: possible therapeutic consequences. Curr Neuropharmacol, 19:1354-66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [173].Ha BK, Bishop GA, King JS, Burry RW (2000). Corticotropin releasing factor induces proliferation of cerebellar astrocytes. J Neurosci Res, 62:789-98. [DOI] [PubMed] [Google Scholar]
  • [174].Fried FE, Lamberti J, Sneed P (1977). Treatment of tension and migraine headaches with biofeedback techniques. Mo Med, 74:253-5. [PubMed] [Google Scholar]
  • [175].Luo Y, Zeng B, Zeng L, Du X, Li B, Huo R, et al. (2018). Gut microbiota regulates mouse behaviors through glucocorticoid receptor pathway genes in the hippocampus. Transl Psychiatry, 8:187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [176].Vodicka M, Ergang P, Hrncir T, Mikulecka A, Kvapilova P, Vagnerova K, et al. (2018). Microbiota affects the expression of genes involved in hpa axis regulation and local metabolism of glucocorticoids in chronic psychosocial stress. Brain Behav Immun, 73:615-24. [DOI] [PubMed] [Google Scholar]
  • [177].Takuma K, Matsuda T, Yoshikawa T, Kitanaka J, Gotoh M, Hayata K, et al. (1994). Corticotropin-releasing factor stimulates ca2+ influx in cultured rat astrocytes. Biochem Biophys Res Commun, 199:1103-7. [DOI] [PubMed] [Google Scholar]
  • [178].Chen SJ, Yang JF, Kong FP, Ren JL, Hao K, Li M, et al. (2014). Overactivation of corticotropin-releasing factor receptor type 1 and aquaporin-4 by hypoxia induces cerebral edema. Proc Natl Acad Sci U S A, 111:13199-204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Lou YX, Wang ZZ, Xia CY, Mou Z, Ren Q, Liu DD, et al. (2020). The protective effect of ginsenoside rg1 on depression may benefit from the gap junction function in hippocampal astrocytes. Eur J Pharmacol, 882:173309. [DOI] [PubMed] [Google Scholar]
  • [180].O'Leary OF, Ogbonnaya ES, Felice D, Levone BR, C CL, Fitzgerald P, et al. (2018). The vagus nerve modulates bdnf expression and neurogenesis in the hippocampus. Eur Neuropsychopharmacol, 28:307-16. [DOI] [PubMed] [Google Scholar]
  • [181].Dave V, Kimelberg HK (1994). Na(+)-dependent, fluoxetine-sensitive serotonin uptake by astrocytes tissue-printed from rat cerebral cortex. J Neurosci, 14:4972-86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [182].Miyazaki I, Asanuma M, Murakami S, Takeshima M, Torigoe N, Kitamura Y, et al. (2013). Targeting 5-ht(1a) receptors in astrocytes to protect dopaminergic neurons in parkinsonian models. Neurobiol Dis, 59:244-56. [DOI] [PubMed] [Google Scholar]
  • [183].Gershon MD, Tack J (2007). The serotonin signaling system: from basic understanding to drug development for functional gi disorders. Gastroenterology, 132:397-414. [DOI] [PubMed] [Google Scholar]
  • [184].Zhang X, Zhu LB, He JH, Zhang HQ, Ji SY, Zhang CN, et al. (2020). Paroxetine suppresses reactive microglia-mediated but not lipopolysaccharide-induced inflammatory responses in primary astrocytes. J Neuroinflammation, 17:50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [185].Li S, Sun Y, Song M, Song Y, Fang Y, Zhang Q, et al. (2021). Nlrp3/caspase-1/gsdmd-mediated pyroptosis exerts a crucial role in astrocyte pathological injury in mouse model of depression. JCI Insight, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Crispe IN (2009). The liver as a lymphoid organ. Annu Rev Immunol, 27:147-63. [DOI] [PubMed] [Google Scholar]
  • [187].Cheng ML, Nakib D, Perciani CT, MacParland SA (2021). The immune niche of the liver. Clin Sci (Lond), 135:2445-66. [DOI] [PubMed] [Google Scholar]
  • [188].Schousboe A, Waagepetersen HS, Leke R, Bak LK (2014). Effects of hyperammonemia on brain energy metabolism: controversial findings in vivo and in vitro. Metab Brain Dis, 29:913-7. [DOI] [PubMed] [Google Scholar]
  • [189].Rose CF, Amodio P, Bajaj JS, Dhiman RK, Montagnese S, Taylor-Robinson SD, et al. (2020). Hepatic encephalopathy: novel insights into classification, pathophysiology and therapy. J Hepatol, 73:1526-47. [DOI] [PubMed] [Google Scholar]
  • [190].Vilstrup H, Amodio P, Bajaj J, Cordoba J, Ferenci P, Mullen KD, et al. (2014). Hepatic encephalopathy in chronic liver disease: 2014 practice guideline by the american association for the study of liver diseases and the european association for the study of the liver. Hepatology, 60:715-35. [DOI] [PubMed] [Google Scholar]
  • [191].Seyan AS, Hughes RD, Shawcross DL (2010). Changing face of hepatic encephalopathy: role of inflammation and oxidative stress. World J Gastroenterol, 16:3347-57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [192].Abdel-Khalek EE, El-Fakhry A, Helaly M, Hamed M, Elbaz O (2011). Systemic inflammatory response syndrome in patients with liver cirrhosis. Arab J Gastroenterol, 12:173-7. [DOI] [PubMed] [Google Scholar]
  • [193].Sepehrinezhad A, Zarifkar A, Namvar G, Shahbazi A, Williams R (2020). Astrocyte swelling in hepatic encephalopathy: molecular perspective of cytotoxic edema. Metab Brain Dis, 35:559-78. [DOI] [PubMed] [Google Scholar]
  • [194].Gallina P, Gallo O, Nicoletti C, Romanelli RG (2019). A hydrodynamic hypothesis for the pathogenesis of glymphatic system impairment in hepatic encephalopathy. J Hepatol, 71:228-9. [DOI] [PubMed] [Google Scholar]
  • [195].Hadjihambi A, Harrison IF, Costas-Rodriguez M, Vanhaecke F, Arias N, Gallego-Duran R, et al. (2019). Impaired brain glymphatic flow in experimental hepatic encephalopathy. J Hepatol, 70:40-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [196].Nam HC, Sung PS, Song DS, Kwon JH, Nam SW, Yoon DJ, et al. (2019). Control of intracranial disease is associated with improved survival in patients with brain metastasis from hepatocellular carcinoma. Int J Clin Oncol, 24:666-76. [DOI] [PubMed] [Google Scholar]
  • [197].Sahin E, Ince V, Sahin TT, Kutlu R, Pasahan R, Yilmaz S (2021). Undetectable brain metastasis of hepatocellular carcinoma by pet-ct after liver transplantation: a case report. [J] Gastrointest Cancer. [DOI] [PubMed] [Google Scholar]
  • [198].Zhang X, Zhang W, Cao WD, Cheng G, Liu B, Cheng J (2012). A review of current management of brain metastases. Ann Surg Oncol, 19:1043-50. [DOI] [PubMed] [Google Scholar]
  • [199].An S, Tiruthani K, Wang Y, Xu L, Hu M, Li J, et al. (2019). Locally trapping the c-c chemokine receptor type 7 by gene delivery nanoparticle inhibits lymphatic metastasis prior to tumor resection. Small, 15:e1805182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [200].Jaeger K, Bruenle S, Weinert T, Guba W, Muehle J, Miyazaki T, et al. (2019). Structural basis for allosteric ligand recognition in the human cc chemokine receptor 7. Cell, 178:1222-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [201].Braun A, Worbs T, Moschovakis GL, Halle S, Hoffmann K, Bolter J, et al. (2011). Afferent lymph-derived t cells and dcs use different chemokine receptor ccr7-dependent routes for entry into the lymph node and intranodal migration. Nat Immunol, 12:879-87. [DOI] [PubMed] [Google Scholar]
  • [202].Zhang L, Wang F, Yao X, Ma S, Zhang L, Qin Z (2020). [progress in targeting therapy of cancer metastasis by ccl21/ccr7 axis]. Sheng Wu Gong Cheng Xue Bao, 36:2741-54. [DOI] [PubMed] [Google Scholar]
  • [203].Ding Y, Shimada Y, Maeda M, Kawabe A, Kaganoi J, Komoto I, et al. (2003). Association of cc chemokine receptor 7 with lymph node metastasis of esophageal squamous cell carcinoma. Clin Cancer Res, 9:3406-12. [PubMed] [Google Scholar]
  • [204].Tammela T, Alitalo K (2010). Lymphangiogenesis: molecular mechanisms and future promise. Cell, 140:460-76. [DOI] [PubMed] [Google Scholar]
  • [205].Chen G, Liu XY, Wang Z, Liu FY (2010). Vascular endothelial growth factor c: the predicator of early recurrence in patients with n2 non-small-cell lung cancer. Eur J Cardiothorac Surg, 37:546-51. [DOI] [PubMed] [Google Scholar]
  • [206].Hamed SA (2019). Neurologic conditions and disorders of uremic syndrome of chronic kidney disease: presentations, causes, and treatment strategies. Expert Rev Clin Pharmacol, 12:61-90. [DOI] [PubMed] [Google Scholar]
  • [207].Rajamani K (2021). The cerebro-renal system- anatomical and physiological considerations. J Stroke Cerebrovasc Dis, 30:105541. [DOI] [PubMed] [Google Scholar]
  • [208].Bugnicourt JM, Godefroy O, Chillon JM, Choukroun G, Massy ZA (2013). Cognitive disorders and dementia in ckd: the neglected kidney-brain axis. J Am Soc Nephrol, 24:353-63. [DOI] [PubMed] [Google Scholar]
  • [209].Cao W, Li A, Li J, Wu C, Cui S, Zhou Z, et al. (2017). Reno-cerebral reflex activates the renin-angiotensin system, promoting oxidative stress and renal damage after ischemia-reperfusion injury. Antioxid Redox Signal, 27:415-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [210].Miranda AS, Cordeiro TM, Dos SLST, Ferreira RN, Simoes ESA (2017). Kidney-brain axis inflammatory cross-talk: from bench to bedside. Clin Sci (Lond), 131:1093-105. [DOI] [PubMed] [Google Scholar]
  • [211].Chang TH, Chiu PF, Tsai CC, Chang CH, Wu CL, Kor CT, et al. (2019). Favourable renal outcomes after intravenous thrombolytic therapy for acute ischemic stroke: clinical implication of kidney-brain axis. Nephrology (Carlton), 24:896-903. [DOI] [PubMed] [Google Scholar]
  • [212].Mao H, Jiang C, Xu L, Chen D, Liu H, Xu Y, et al. (2020). Ginsenoside protects against aki via activation of hif-1alpha and vegf-a in the kidney-brain axis. Int J Mol Med, 45:939-46. [DOI] [PubMed] [Google Scholar]
  • [213].Ariton DM, Jimenez-Balado J, Maisterra O, Pujadas F, Soler MJ, Delgado P (2021). Diabetes, albuminuria and the kidney-brain axis. J Clin Med, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [214].Niwa T (2017). The role of carbon adsorbent in the conservative management of chronic kidney disease. Panminerva Med, 59:139-48. [DOI] [PubMed] [Google Scholar]
  • [215].Toyoda K, Ninomiya T (2014). Stroke and cerebrovascular diseases in patients with chronic kidney disease. Lancet Neurol, 13:823-33. [DOI] [PubMed] [Google Scholar]
  • [216].Mestre H, Kostrikov S, Mehta RI, Nedergaard M (2017). Perivascular spaces, glymphatic dysfunction, and small vessel disease. Clin Sci (Lond), 131:2257-74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [217].Jiaerken Y, Lian C, Huang P, Yu X, Zhang R, Wang S, et al. (2021). Dilated perivascular space is related to reduced free-water in surrounding white matter among healthy adults and elderlies but not in patients with severe cerebral small vessel disease. J Cereb Blood Flow Metab, 41:2561-70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [218].Naganuma T, Takemoto Y (2015). New aspects of cerebrovascular diseases in dialysis patients. Contrib Nephrol, 185:138-46. [DOI] [PubMed] [Google Scholar]
  • [219].Ovbiagele B, Wing JJ, Menon RS, Burgess RE, Gibbons MC, Sobotka I, et al. (2013). Association of chronic kidney disease with cerebral microbleeds in patients with primary intracerebral hemorrhage. Stroke, 44:2409-13. [DOI] [PubMed] [Google Scholar]
  • [220].Xiao L, Lan W, Sun W, Dai Q, Xiong Y, Li L, et al. (2015). Chronic kidney disease in patients with lacunar stroke: association with enlarged perivascular spaces and total magnetic resonance imaging burden of cerebral small vessel disease. Stroke, 46:2081-6. [DOI] [PubMed] [Google Scholar]
  • [221].Lee WJ, Jung KH, Ryu H, Oh KH, Kim JM, Lee ST, et al. (2021). Association of autosomal dominant polycystic kidney disease with cerebral small vessel disease. J Cereb Blood Flow Metab, 41:3365-77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [222].Viggiano D, Wagner CA, Martino G, Nedergaard M, Zoccali C, Unwin R, et al. (2020). Mechanisms of cognitive dysfunction in ckd. Nat Rev Nephrol, 16:452-69. [DOI] [PubMed] [Google Scholar]
  • [223].Pepin M, Ferreira AC, Arici M, Bachman M, Barbieri M, Bumblyte IA, et al. (2021). Cognitive disorders in patients with chronic kidney disease: specificities of clinical assessment. Nephrol Dial Transplant, 37:i23-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [224].Heo CM, Lee DA, Park KM, Lee YJ, Park S, Kim YW, et al. (2022). Glymphatic system dysfunction in patients with early chronic kidney disease. Front Neurol, 13:976089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [225].Schmidt R, Schmidt H, Curb JD, Masaki K, White LR, Launer LJ (2002). Early inflammation and dementia: a 25-year follow-up of the honolulu-asia aging study. Ann Neurol, 52:168-74. [DOI] [PubMed] [Google Scholar]
  • [226].Lin YT, Wu PH, Tsai YC, Hsu YL, Wang HY, Kuo MC, et al. (2019). Indoxyl sulfate induces apoptosis through oxidative stress and mitogen-activated protein kinase signaling pathway inhibition in human astrocytes. J Clin Med, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [227].Adesso S, Magnus T, Cuzzocrea S, Campolo M, Rissiek B, Paciello O, et al. (2017). Indoxyl sulfate affects glial function increasing oxidative stress and neuroinflammation in chronic kidney disease: interaction between astrocytes and microglia. Front Pharmacol, 8:370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [228].Badaut J, Lasbennes F, Magistretti PJ, Regli L (2002). Aquaporins in brain: distribution, physiology, and pathophysiology. J Cereb Blood Flow Metab, 22:367-78. [DOI] [PubMed] [Google Scholar]
  • [229].Grams ME, Rabb H (2012). The distant organ effects of acute kidney injury. Kidney Int, 81:942-8. [DOI] [PubMed] [Google Scholar]
  • [230].Burn DJ, Bates D (1998). Neurology and the kidney. J Neurol Neurosurg Psychiatry, 65:810-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [231].Vanholder R, De Deyn PP, Van Biesen W, Lameire N (2008). Marconi revisited: from kidney to brain--two organ systems communicating at long distance. J Am Soc Nephrol, 19:1253-5. [DOI] [PubMed] [Google Scholar]
  • [232].Konsman JP, Parnet P, Dantzer R (2002). Cytokine-induced sickness behaviour: mechanisms and implications. Trends Neurosci, 25:154-9. [DOI] [PubMed] [Google Scholar]
  • [233].Kuperberg SJ, Wadgaonkar R (2017). Sepsis-associated encephalopathy: the blood-brain barrier and the sphingolipid rheostat. Front Immunol, 8:597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [234].Tahamtan M, Kohlmeier KA, Faatehi M, Basiri M, Shabani M (2021). Electrophysiological and inflammatory changes of ca1 area in male rats exposed to acute kidney injury: neuroprotective effects of erythropoietin. Brain Res Bull, 171:25-34. [DOI] [PubMed] [Google Scholar]
  • [235].Liu M, Liang Y, Chigurupati S, Lathia JD, Pletnikov M, Sun Z, et al. (2008). Acute kidney injury leads to inflammation and functional changes in the brain. J Am Soc Nephrol, 19:1360-70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [236].Wang X, Delle C, Asiminas A, Akther S, Vittani M, Brogger P, et al. (2022). Liver-secreted fluorescent blood plasma markers enable chronic imaging of the microcirculation. Cell Rep Methods, 2:100302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [237].Trefts E, Gannon M, Wasserman DH (2017). The liver. Curr Biol, 27:R1147-51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [238].Wang H, Ran J, Jiang T (2014). Urea. Subcell Biochem, 73:7-29. [DOI] [PubMed] [Google Scholar]
  • [239].Hamada K, Oike Y, Takakura N, Ito Y, Jussila L, Dumont DJ, et al. (2000). Vegf-c signaling pathways through vegfr-2 and vegfr-3 in vasculoangiogenesis and hematopoiesis. Blood, 96:3793-800. [PubMed] [Google Scholar]
  • [240].Zhang Y, Gao W, Yang K, Tao H, Yang H (2018). Salt-inducible kinase 1 (sik1) is induced by alcohol and suppresses microglia inflammation via nf-kappab signaling. Cell Physiol Biochem, 47:1411-21. [DOI] [PubMed] [Google Scholar]
  • [241].Nagelhus EA, Mathiisen TM, Ottersen OP (2004). Aquaporin-4 in the central nervous system: cellular and subcellular distribution and coexpression with kir4.1. Neuroscience, 129:905-13. [DOI] [PubMed] [Google Scholar]
  • [242].Warth A, Mittelbronn M, Wolburg H (2005). Redistribution of the water channel protein aquaporin-4 and the k+ channel protein kir4.1 differs in low- and high-grade human brain tumors. Acta Neuropathol, 109:418-26. [DOI] [PubMed] [Google Scholar]
  • [243].Huber VJ, Igarashi H, Ueki S, Kwee IL, Nakada T (2018). Aquaporin-4 facilitator tgn-073 promotes interstitial fluid circulation within the blood-brain barrier: [17o]h2o jjvcpe mri study. Neuroreport, 29:697-703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [244].Eefsen M, Jelnes P, Schmidt LE, Vainer B, Bisgaard HC, Larsen FS (2010). Brain expression of the water channels aquaporin-1 and -4 in mice with acute liver injury, hyperammonemia and brain edema. Metab Brain Dis, 25:315-23. [DOI] [PubMed] [Google Scholar]
  • [245].Rama RK, Jayakumar AR, Tong X, Curtis KM, Norenberg MD (2010). Brain aquaporin-4 in experimental acute liver failure. J Neuropathol Exp Neurol, 69:869-79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [246].Gan W, Huang JL, Zhang MX, Fu YP, Yi Y, Jing CY, et al. (2018). New nomogram predicts the recurrence of hepatocellular carcinoma in patients with negative preoperative serum afp subjected to curative resection. J Surg Oncol, 117:1540-7. [DOI] [PubMed] [Google Scholar]
  • [247].Kamimura K, Kobayashi Y, Takahashi Y, Abe H, Kumaki D, Yokoo T, et al. (2017). Tumor markers for early diagnosis for brain metastasis of hepatocellular carcinoma: a case series and literature review for effective loco-regional treatment. Cancer Biol Ther, 18:79-84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [248].Song Y, Wang Z, Liu X, Jiang W, Shi M (2012). Ccr7 and vegf-c: molecular indicator of lymphatic metastatic recurrence in pn0 esophageal squamous cell carcinoma after ivor-lewis esophagectomy? Ann Surg Oncol, 19:3606-12. [DOI] [PubMed] [Google Scholar]
  • [249].Zhao J, Hu J, Cai J, Yang X, Yang Z (2003). Vascular endothelial growth factor expression in serum of patients with hepatocellular carcinoma. Chin Med J (Engl), 116:772-6. [PubMed] [Google Scholar]
  • [250].Han MS, Moon KS, Lee KH, Cho SB, Lim SH, Jang WY, et al. (2013). Brain metastasis from hepatocellular carcinoma: the role of surgery as a prognostic factor. BMC Cancer, 13:567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [251].Shao YY, Lu LC, Cheng AL, Hsu CH (2011). Increasing incidence of brain metastasis in patients with advanced hepatocellular carcinoma in the era of antiangiogenic targeted therapy. Oncologist, 16:82-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [252].Park Y, Kim KS, Kim K, Chie EK, Kim JH, Kim JS, et al. (2015). Nomogram prediction of survival in patients with brain metastases from hepatocellular carcinoma treated with whole-brain radiotherapy: a multicenter retrospective study. J Neurooncol, 125:377-83. [DOI] [PubMed] [Google Scholar]
  • [253].Imamura I (2003). Prognostic efficacy of treatment for extrahepatic metastasis after surgical treatment of hepatocellular carcinoma. Kurume Med J, 50:41-8. [DOI] [PubMed] [Google Scholar]
  • [254].Moriya H, Ohtani Y, Tsukui M, Tanaka Y, Tajima T, Makuuchi H, et al. (1999). A case report: tumorectomy for brain metastasis of hepatocellular carcinoma. Tokai J Exp Clin Med, 24:105-10. [PubMed] [Google Scholar]
  • [255].Hiraoka A, Horiike N, Koizumi Y, Tazuya N, Ichiryu M, Nakahara H, et al. (2008). Brain metastasis from hepatocellular carcinoma treated with a cyber-knife. Intern Med, 47:1993-6. [DOI] [PubMed] [Google Scholar]
  • [256].Xu Q, Wu P, Feng Y, Ye K, Tong Y, Zhou Y (2014). Gamma knife surgery for brain metastasis from hepatocellular carcinoma. PLoS One, 9:e88317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [257].Masoumi E, Tahaghoghi-Hajghorbani S, Jafarzadeh L, Sanaei MJ, Pourbagheri-Sigaroodi A, Bashash D (2021). The application of immune checkpoint blockade in breast cancer and the emerging role of nanoparticle. J Control Release, 340:168-87. [DOI] [PubMed] [Google Scholar]
  • [258].Cammarota A, Zanuso V, D'Alessio A, Pressiani T, Bozzarelli S, Personeni N, et al. (2022). The dual checkpoint blockade in unresectable hepatocellular carcinoma: opportunities emerging in clinical trials. Expert Opin Investig Drugs, 31:425-35. [DOI] [PubMed] [Google Scholar]
  • [259].Bordon Y (2020). Vegf-c shines a light on brain tumours. Nat Rev Immunol, 20:140-1. [DOI] [PubMed] [Google Scholar]
  • [260].Ju S, Xu C, Wang G, Zhang L (2019). Vegf-c induces alternative activation of microglia to promote recovery from traumatic brain injury. J Alzheimers Dis, 68:1687-97. [DOI] [PubMed] [Google Scholar]
  • [261].Song E, Mao T, Dong H, Boisserand L, Antila S, Bosenberg M, et al. (2020). Vegf-c-driven lymphatic drainage enables immunosurveillance of brain tumours. Nature, 577:689-94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [262].Michaelsen SR, Staberg M, Pedersen H, Jensen KE, Majewski W, Broholm H, et al. (2018). Vegf-c sustains vegfr2 activation under bevacizumab therapy and promotes glioblastoma maintenance. Neuro Oncol, 20:1462-74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [263].Jurisic G, Maby-El HH, Karaman S, Ochsenbein AM, Alitalo A, Siddiqui SS, et al. (2012). An unexpected role of semaphorin3a-neuropilin-1 signaling in lymphatic vessel maturation and valve formation. Circ Res, 111:426-36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [264].Wang J, Zhang X, Thomas SM, Grandis JR, Wells A, Chen ZG, et al. (2005). Chemokine receptor 7 activates phosphoinositide-3 kinase-mediated invasive and prosurvival pathways in head and neck cancer cells independent of egfr. Oncogene, 24:5897-904. [DOI] [PubMed] [Google Scholar]
  • [265].Liu FY, Zhao ZJ, Li P, Ding X, Guo N, Yang LL, et al. (2011). Nf-kappab participates in chemokine receptor 7-mediated cell survival in metastatic squamous cell carcinoma of the head and neck. Oncol Rep, 25:383-91. [DOI] [PubMed] [Google Scholar]
  • [266].Xu B, Zhou M, Qiu W, Ye J, Feng Q (2017). Ccr7 mediates human breast cancer cell invasion, migration by inducing epithelial-mesenchymal transition and suppressing apoptosis through akt pathway. Cancer Med, 6:1062-71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [267].Xiong Y, Huang F, Li X, Chen Z, Feng D, Jiang H, et al. (2017). Ccl21/ccr7 interaction promotes cellular migration and invasion via modulation of the mek/erk1/2 signaling pathway and correlates with lymphatic metastatic spread and poor prognosis in urinary bladder cancer. Int J Oncol, 51:75-90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [268].Shannon LA, Calloway PA, Welch TP, Vines CM (2010). Ccr7/ccl21 migration on fibronectin is mediated by phospholipase cgamma1 and erk1/2 in primary t lymphocytes. J Biol Chem, 285:38781-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [269].Liu FY, Safdar J, Li ZN, Fang QG, Zhang X, Xu ZF, et al. (2014). Ccr7 regulates cell migration and invasion through jak2/stat3 in metastatic squamous cell carcinoma of the head and neck. Biomed Res Int, 2014:415375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [270].Zhang Z, Liu F, Li Z, Wang D, Li R, Sun C (2017). Jak3 is involved in ccr7-dependent migration and invasion in metastatic squamous cell carcinoma of the head and neck. Oncol Lett, 13:3191-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [271].Karkkainen MJ, Haiko P, Sainio K, Partanen J, Taipale J, Petrova TV, et al. (2004). Vascular endothelial growth factor c is required for sprouting of the first lymphatic vessels from embryonic veins. Nat Immunol, 5:74-80. [DOI] [PubMed] [Google Scholar]
  • [272].Hirakawa S, Brown LF, Kodama S, Paavonen K, Alitalo K, Detmar M (2007). Vegf-c-induced lymphangiogenesis in sentinel lymph nodes promotes tumor metastasis to distant sites. Blood, 109:1010-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [273].Aslan K, Turco V, Blobner J, Sonner JK, Liuzzi AR, Nunez NG, et al. (2020). Heterogeneity of response to immune checkpoint blockade in hypermutated experimental gliomas. Nat Commun, 11:931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [274].Fankhauser M, Broggi M, Potin L, Bordry N, Jeanbart L, Lund AW, et al. (2017). Tumor lymphangiogenesis promotes t cell infiltration and potentiates immunotherapy in melanoma. Sci Transl Med, 9. [DOI] [PubMed] [Google Scholar]
  • [275].Sasso MS, Mitrousis N, Wang Y, Briquez PS, Hauert S, Ishihara J, et al. (2021). Lymphangiogenesis-inducing vaccines elicit potent and long-lasting t cell immunity against melanomas. Sci Adv, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [276].Farnsworth RH, Achen MG, Stacker SA (2018). The evolving role of lymphatics in cancer metastasis. Curr Opin Immunol, 53:64-73. [DOI] [PubMed] [Google Scholar]
  • [277].Skobe M, Hawighorst T, Jackson DG, Prevo R, Janes L, Velasco P, et al. (2001). Induction of tumor lymphangiogenesis by vegf-c promotes breast cancer metastasis. Nat Med, 7:192-8. [DOI] [PubMed] [Google Scholar]
  • [278].He Y, Rajantie I, Pajusola K, Jeltsch M, Holopainen T, Yla-Herttuala S, et al. (2005). Vascular endothelial cell growth factor receptor 3-mediated activation of lymphatic endothelium is crucial for tumor cell entry and spread via lymphatic vessels. Cancer Res, 65:4739-46. [DOI] [PubMed] [Google Scholar]
  • [279].Khromova N, Kopnin P, Rybko V, Kopnin BP (2012). Downregulation of vegf-c expression in lung and colon cancer cells decelerates tumor growth and inhibits metastasis via multiple mechanisms. Oncogene, 31:1389-97. [DOI] [PubMed] [Google Scholar]
  • [280].Chen JC, Chang YW, Hong CC, Yu YH, Su JL (2012). The role of the vegf-c/vegfrs axis in tumor progression and therapy. Int J Mol Sci, 14:88-107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [281].Colombo M, Raposo G, Thery C (2014). Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol, 30:255-89. [DOI] [PubMed] [Google Scholar]
  • [282].Sterzenbach U, Putz U, Low LH, Silke J, Tan SS, Howitt J (2017). Engineered exosomes as vehicles for biologically active proteins. Mol Ther, 25:1269-78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [283].EL AS, Mager I, Breakefield XO, Wood MJ (2013). Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov, 12:347-57. [DOI] [PubMed] [Google Scholar]
  • [284].Kooijmans SA, Vader P, van Dommelen SM, van Solinge WW, Schiffelers RM (2012). Exosome mimetics: a novel class of drug delivery systems. Int J Nanomedicine, 7:1525-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [285].Li Y, Gao ZH, Qu XJ (2015). The adverse effects of sorafenib in patients with advanced cancers. Basic Clin Pharmacol Toxicol, 116:216-21. [DOI] [PubMed] [Google Scholar]
  • [286].Xie B, Wang DH, Spechler SJ (2012). Sorafenib for treatment of hepatocellular carcinoma: a systematic review. Dig Dis Sci, 57:1122-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [287].Zeisel MB, Pfeffer S, Baumert TF (2013). Mir-122 acts as a tumor suppressor in hepatocarcinogenesis in vivo. J Hepatol, 58:821-3. [DOI] [PubMed] [Google Scholar]
  • [288].Zhang J, Song Q, Wu M, Zheng W (2021). The emerging roles of exosomes in the chemoresistance of hepatocellular carcinoma. Curr Med Chem, 28:93-109. [DOI] [PubMed] [Google Scholar]
  • [289].Li C, Deng M, Hu J, Li X, Chen L, Ju Y, et al. (2016). Chronic inflammation contributes to the development of hepatocellular carcinoma by decreasing mir-122 levels. Oncotarget, 7:17021-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [290].Lou G, Song X, Yang F, Wu S, Wang J, Chen Z, et al. (2015). Exosomes derived from mir-122-modified adipose tissue-derived mscs increase chemosensitivity of hepatocellular carcinoma. J Hematol Oncol, 8:122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [291].Wang G, Zhao W, Wang H, Qiu G, Jiang Z, Wei G, et al. (2019). Exosomal mir-744 inhibits proliferation and sorafenib chemoresistance in hepatocellular carcinoma by targeting pax2. Med Sci Monit, 25:7209-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [292].Lai X, Wang Y, Wang X, Liu B, Rong L (2022). Mir-146a-5p-modified hucmsc-derived exosomes facilitate spinal cord function recovery by targeting neurotoxic astrocytes. Stem Cell Res Ther, 13:487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [293].Blivet G, Meunier J, Roman FJ, Touchon J (2018). Neuroprotective effect of a new photobiomodulation technique against abeta (25-35) peptide-induced toxicity in mice: novel hypothesis for therapeutic approach of alzheimer's disease suggested. Alzheimers Dement (N Y), 4:54-63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [294].An J, Wang L, Song S, Tian L, Liu Q, Mei M, et al. (2022). Electroacupuncture reduces blood glucose by regulating intestinal flora in type 2 diabetic mice. J Diabetes, 14:695-710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [295].Syeda T, Sanchez-Tapia M, Pinedo-Vargas L, Granados O, Cuervo-Zanatta D, Rojas-Santiago E, et al. (2018). Bioactive food abates metabolic and synaptic alterations by modulation of gut microbiota in a mouse model of alzheimer's disease. J Alzheimers Dis, 66:1657-82. [DOI] [PubMed] [Google Scholar]
  • [296].Kim SK, Guevarra RB, Kim YT, Kwon J, Kim H, Cho JH, et al. (2019). Role of probiotics in human gut microbiome-associated diseases. J Microbiol Biotechnol, 29:1335-40. [DOI] [PubMed] [Google Scholar]
  • [297].Angelucci F, Cechova K, Amlerova J, Hort J (2019). Antibiotics, gut microbiota, and alzheimer's disease. J Neuroinflammation, 16:108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [298].Ruggiero M (2016). Fecal microbiota transplantation and the brain microbiota in neurological diseases. Clin Endosc, 49:579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [299].Liang Y, Iqbal Z, Lu J, Wang J, Zhang H, Chen X, et al. (2022). Cell-derived nanovesicle-mediated drug delivery to the brain: principles and strategies for vesicle engineering. Mol Ther. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [300].Wong KH, Riaz MK, Xie Y, Zhang X, Liu Q, Chen H, et al. (2019). Review of current strategies for delivering alzheimer's disease drugs across the blood-brain barrier. Int J Mol Sci, 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [301].Pardridge WM (2022). A historical review of brain drug delivery. Pharmaceutics, 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [302].Zhang Z, Lu Y, Qi J, Wu W (2021). An update on oral drug delivery via intestinal lymphatic transport. Acta Pharm Sin B, 11:2449-68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [303].Rufino-Ramos D, Albuquerque PR, Carmona V, Perfeito R, Nobre RJ, Pereira DAL (2017). Extracellular vesicles: novel promising delivery systems for therapy of brain diseases. J Control Release, 262:247-58. [DOI] [PubMed] [Google Scholar]
  • [304].Shahjin F, Chand S, Yelamanchili SV (2020). Extracellular vesicles as drug delivery vehicles to the central nervous system. J Neuroimmune Pharmacol, 15:443-58. [DOI] [PubMed] [Google Scholar]
  • [305].Salehpour F, Mahmoudi J, Kamari F, Sadigh-Eteghad S, Rasta SH, Hamblin MR (2018). Brain photobiomodulation therapy: a narrative review. Mol Neurobiol, 55:6601-36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [306].Hamblin MR (2016). Shining light on the head: photobiomodulation for brain disorders. BBA Clin, 6:113-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [307].Semyachkina-Glushkovskaya O, Abdurashitov A, Dubrovsky A, Klimova M, Agranovich I, Terskov A, et al. (2020). Photobiomodulation of lymphatic drainage and clearance: perspective strategy for augmentation of meningeal lymphatic functions. Biomed Opt Express, 11:725-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [308].Zinchenko E, Navolokin N, Shirokov A, Khlebtsov B, Dubrovsky A, Saranceva E, et al. (2019). Pilot study of transcranial photobiomodulation of lymphatic clearance of beta-amyloid from the mouse brain: breakthrough strategies for non-pharmacologic therapy of alzheimer's disease. Biomed Opt Express, 10:4003-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [309].Salehpour F, Khademi M, Hamblin MR (2021). Photobiomodulation therapy for dementia: a systematic review of pre-clinical and clinical studies. J Alzheimers Dis, 83:1431-52. [DOI] [PubMed] [Google Scholar]
  • [310].Berman MH, Nichols TW (2019). Treatment of neurodegeneration: integrating photobiomodulation and neurofeedback in alzheimer's dementia and parkinson's: a review. Photobiomodul Photomed Laser Surg, 37:623-34. [DOI] [PubMed] [Google Scholar]
  • [311].Li X, Wu D, Niu J, Sun Y, Wang Q, Yang B, et al. (2021). Intestinal flora: a pivotal role in investigation of traditional chinese medicine. Am J Chin Med, 49:237-68. [DOI] [PubMed] [Google Scholar]
  • [312].Wang L, An J, Song S, Mei M, Li W, Ding F, et al. (2020). Electroacupuncture preserves intestinal barrier integrity through modulating the gut microbiota in dss-induced chronic colitis. Life Sci, 261:118473. [DOI] [PubMed] [Google Scholar]
  • [313].Jiang J, Liu H, Wang Z, Tian H, Wang S, Yang J, et al. (2021). Electroacupuncture could balance the gut microbiota and improve the learning and memory abilities of alzheimer's disease animal model. PLoS One, 16:e259530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [314].Li P, Huang W, Yan YN, Cheng W, Liu S, Huang Y, et al. (2021). Acupuncture can play an antidepressant role by regulating the intestinal microbes and neurotransmitters in a rat model of depression. Med Sci Monit, 27:e929027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [315].Jang JH, Yeom MJ, Ahn S, Oh JY, Ji S, Kim TH, et al. (2020). Acupuncture inhibits neuroinflammation and gut microbial dysbiosis in a mouse model of parkinson's disease. Brain Behav Immun, 89:641-55. [DOI] [PubMed] [Google Scholar]
  • [316].Chen DF, Zhang H, Xie JY, Deng C, Qiu RR, Xu YY, et al. (2022). [effect of electroacupuncture on gut microbiota and serum il-1beta and il-18 in rats with vascular dementia based on principle of "curing brain disorders by treating intestines"]. Zhen Ci Yan Jiu, 47:216-23. [DOI] [PubMed] [Google Scholar]
  • [317].Yang B, He M, Chen X, Sun M, Pan T, Xu X, et al. (2022). Acupuncture effect assessment in app/ps1 transgenic mice: on regulating learning-memory abilities, gut microbiota, and microbial metabolites. Comput Math Methods Med, 2022:1527159. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • [318].Zhuang Z, Yang R, Wang W, Qi L, Huang T (2020). Associations between gut microbiota and alzheimer's disease, major depressive disorder, and schizophrenia. J Neuroinflammation, 17:288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [319].Shen L, Liu L, Ji HF (2017). Alzheimer's disease histological and behavioral manifestations in transgenic mice correlate with specific gut microbiome state. J Alzheimers Dis, 56:385-90. [DOI] [PubMed] [Google Scholar]
  • [320].Cao F, Xu Y, Zhang M, Li X, Chen Y, Zhi M, et al. (2022). Baihui (du20), shenmen (ht7) and sanyinjiao (sp6) target the camp/creb/bdnf and pi3k/akt pathways to reduce central nervous system apoptosis in rats with insomnia. Heliyon, 8:e12574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [321].Liu XY, Dai XH, Zou W, Yu XP, Teng W, Wang Y, et al. (2018). Acupuncture through baihui (du20) to qubin (gb7) mitigates neurological impairment after intracerebral hemorrhage. Neural Regen Res, 13:1425-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [322].Ji S, Zhang H, Qin W, Liu M, Zheng W, Han Y, et al. (2021). Effect of acupuncture stimulation of hegu (li4) and taichong (lr3) on the resting-state networks in alzheimer's disease: beyond the default mode network. Neural Plast, 2021:8876873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [323].Zhang L, Xu N (2017). [analysis on mechanism of acupuncture with three needles at hegu (li 4) and its clinical application]. Zhongguo Zhen Jiu, 37:51-3. [DOI] [PubMed] [Google Scholar]
  • [324].Oh JE, Kim SN (2021). Anti-inflammatory effects of acupuncture at st36 point: a literature review in animal studies. Front Immunol, 12:813748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [325].Wang M, Gao YH, Xu J, Chi Y, Wei XB, Lewith G, et al. (2015). Zusanli (st36) acupoint injection for preventing postoperative ileus: a systematic review and meta-analysis of randomized clinical trials. Complement Ther Med, 23:469-83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [326].Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, et al. (2011). Linking long-term dietary patterns with gut microbial enterotypes. Science, 334:105-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [327].Deshpande NG, Saxena J, Pesaresi TG, Carrell CD, Ashby GB, Liao MK, et al. (2019). High fat diet alters gut microbiota but not spatial working memory in early middle-aged sprague dawley rats. PLoS One, 14:e217553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [328].Ley RE, Backhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI (2005). Obesity alters gut microbial ecology. Proc Natl Acad Sci U S A, 102:11070-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [329].Manor O, Dai CL, Kornilov SA, Smith B, Price ND, Lovejoy JC, et al. (2020). Health and disease markers correlate with gut microbiome composition across thousands of people. Nat Commun, 11:5206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [330].Pickard JM, Zeng MY, Caruso R, Nunez G (2017). Gut microbiota: role in pathogen colonization, immune responses, and inflammatory disease. Immunol Rev, 279:70-89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [331].Daly CM, Saxena J, Singh J, Bullard MR, Bondy EO, Saxena A, et al. (2022). Sex differences in response to a high fat, high sucrose diet in both the gut microbiome and hypothalamic astrocytes and microglia. Nutr Neurosci, 25:321-35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [332].Lambert J, Mejia S, Vojdani A (2019). Plant and human aquaporins: pathogenesis from gut to brain. Immunol Res, 67:12-20. [DOI] [PubMed] [Google Scholar]
  • [333].Morkl S, Butler MI, Holl A, Cryan JF, Dinan TG (2020). Probiotics and the microbiota-gut-brain axis: focus on psychiatry. Curr Nutr Rep, 9:171-82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [334].Slavin J (2013). Fiber and prebiotics: mechanisms and health benefits. Nutrients, 5:1417-35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [335].Pandey KR, Naik SR, Vakil BV (2015). Probiotics, prebiotics and synbiotics- a review. J Food Sci Technol, 52:7577-87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [336].Azad M, Sarker M, Li T, Yin J (2018). Probiotic species in the modulation of gut microbiota: an overview. Biomed Res Int, 2018:9478630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [337].Fijan S (2014). Microorganisms with claimed probiotic properties: an overview of recent literature. Int J Environ Res Public Health, 11:4745-67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [338].Ma Y, Liu T, Fu J, Fu S, Hu C, Sun B, et al. (2019). Lactobacillus acidophilus exerts neuroprotective effects in mice with traumatic brain injury. J Nutr, 149:1543-52. [DOI] [PubMed] [Google Scholar]
  • [339].Chivero ET, Sil S, Singh S, Thangaraj A, Gordon L, Evah-Nzoughe GB, et al. (2022). Protective role of lactobacillus rhamnosus probiotic in reversing cocaine-induced oxidative stress, glial activation and locomotion in mice. J Neuroimmune Pharmacol, 17:62-75. [DOI] [PubMed] [Google Scholar]
  • [340].Lu J, Lu L, Yu Y, Baranowski J, Claud EC (2020). Maternal administration of probiotics promotes brain development and protects offspring's brain from postnatal inflammatory insults in c57/bl6j mice. Sci Rep, 10:8178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [341].Zmora N, Zilberman-Schapira G, Suez J, Mor U, Dori-Bachash M, Bashiardes S, et al. (2018). Personalized gut mucosal colonization resistance to empiric probiotics is associated with unique host and microbiome features. Cell, 174:1388-405. [DOI] [PubMed] [Google Scholar]
  • [342].Liu J, Wang Y, Heelan WJ, Chen Y, Li Z, Hu Q (2022). Mucoadhesive probiotic backpacks with ros nanoscavengers enhance the bacteriotherapy for inflammatory bowel diseases. Sci Adv, 8:p8798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [343].Ianiro G, Tilg H, Gasbarrini A (2016). Antibiotics as deep modulators of gut microbiota: between good and evil. Gut, 65:1906-15. [DOI] [PubMed] [Google Scholar]
  • [344].Frohlich EE, Farzi A, Mayerhofer R, Reichmann F, Jacan A, Wagner B, et al. (2016). Cognitive impairment by antibiotic-induced gut dysbiosis: analysis of gut microbiota-brain communication. Brain Behav Immun, 56:140-55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [345].Neuman H, Forsythe P, Uzan A, Avni O, Koren O (2018). Antibiotics in early life: dysbiosis and the damage done. FEMS Microbiol Rev, 42:489-99. [DOI] [PubMed] [Google Scholar]
  • [346].Neufeld NH, Mohamed NS, Grujich N, Shulman K (2017). Acute neuropsychiatric symptoms associated with antibiotic treatment of helicobacter pylori infections: a review. J Psychiatr Pract, 23:25-35. [DOI] [PubMed] [Google Scholar]
  • [347].Budni J, Garcez ML, de Medeiros J, Cassaro E, Bellettini-Santos T, Mina F, et al. (2016). The anti-inflammatory role of minocycline in alzheimer s disease. Curr Alzheimer Res, 13:1319-29. [DOI] [PubMed] [Google Scholar]
  • [348].Minter MR, Zhang C, Leone V, Ringus DL, Zhang X, Oyler-Castrillo P, et al. (2016). Antibiotic-induced perturbations in gut microbial diversity influences neuro-inflammation and amyloidosis in a murine model of alzheimer's disease. Sci Rep, 6:30028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [349].Minter MR, Hinterleitner R, Meisel M, Zhang C, Leone V, Zhang X, et al. (2017). Antibiotic-induced perturbations in microbial diversity during post-natal development alters amyloid pathology in an aged app(swe)/ps1(deltae9) murine model of Alzheimer's disease. Sci Rep, 7:10411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [350].Garwood CJ, Cooper JD, Hanger DP, Noble W (2010). Anti-inflammatory impact of minocycline in a mouse model of tauopathy. Front Psychiatry, 1:136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [351].Khoruts A, Sadowsky MJ (2016). Understanding the mechanisms of faecal microbiota transplantation. Nat Rev Gastroenterol Hepatol, 13:508-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [352].Sun MF, Zhu YL, Zhou ZL, Jia XB, Xu YD, Yang Q, et al. (2018). Neuroprotective effects of fecal microbiota transplantation on mptp-induced parkinson's disease mice: gut microbiota, glial reaction and tlr4/tnf-alpha signaling pathway. Brain Behav Immun, 70:48-60. [DOI] [PubMed] [Google Scholar]
  • [353].Rao J, Qiao Y, Xie R, Lin L, Jiang J, Wang C, et al. (2021). Fecal microbiota transplantation ameliorates stress-induced depression-like behaviors associated with the inhibition of glial and nlrp3 inflammasome in rat brain. J Psychiatr Res, 137:147-57. [DOI] [PubMed] [Google Scholar]
  • [354].Mistry K (2019). Dialysis disequilibrium syndrome prevention and management. Int J Nephrol Renovasc Dis, 12:69-77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [355].Te LE, van Roij C, Meijers B, De Loor H, Kessels R, Wetzels J (2020). Cognitive function and uremic toxins after kidney transplantation: an exploratory study. Kidney360, 1:1398-406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [356].Niwa T, Nomura T, Sugiyama S, Miyazaki T, Tsukushi S, Tsutsui S (1997). The protein metabolite hypothesis, a model for the progression of renal failure: an oral adsorbent lowers indoxyl sulfate levels in undialyzed uremic patients. Kidney Int Suppl (2011), 62:S23-8. [PubMed] [Google Scholar]
  • [357].Rocchetti MT, Cosola C, di Bari I, Magnani S, Galleggiante V, Scandiffio L, et al. (2020). Efficacy of divinylbenzenic resin in removing indoxyl sulfate and p-cresol sulfate in hemodialysis patients: results from an in vitro study and an in vivo pilot trial (xuanro4-nature 3.2). Toxins (Basel), 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [358].Hermann DM, Kribben A, Bruck H (2014). Cognitive impairment in chronic kidney disease: clinical findings, risk factors and consequences for patient care. J Neural Transm (Vienna), 121:627-32. [DOI] [PubMed] [Google Scholar]
  • [359].Murtaza A, Dasgupta I (2021). Chronic kidney disease and cognitive impairment. J Stroke Cerebrovasc Dis, 30:105529. [DOI] [PubMed] [Google Scholar]
  • [360].Drew DA, Weiner DE, Sarnak MJ (2019). Cognitive impairment in ckd: pathophysiology, management, and prevention. Am J Kidney Dis, 74:782-90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [361].Mizumasa T, Hirakata H, Yoshimitsu T, Hirakata E, Kubo M, Kashiwagi M, et al. (2004). Dialysis-related hypotension as a cause of progressive frontal lobe atrophy in chronic hemodialysis patients: a 3-year prospective study. Nephron Clin Pract, 97:c23-30. [DOI] [PubMed] [Google Scholar]
  • [362].McIntyre CW (2010). Recurrent circulatory stress: the dark side of dialysis. Semin Dial, 23:449-51. [DOI] [PubMed] [Google Scholar]
  • [363].Trinh-Trang-Tan MM, Cartron JP, Bankir L (2005). Molecular basis for the dialysis disequilibrium syndrome: altered aquaporin and urea transporter expression in the brain. Nephrol Dial Transplant, 20:1984-8. [DOI] [PubMed] [Google Scholar]
  • [364].Chuasuwan A, Pooripussarakul S, Thakkinstian A, Ingsathit A, Pattanaprateep O (2020). Comparisons of quality of life between patients underwent peritoneal dialysis and hemodialysis: a systematic review and meta-analysis. Health Qual Life Outcomes, 18:191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [365].Neumann D, Mau W, Wienke A, Girndt M (2018). Peritoneal dialysis is associated with better cognitive function than hemodialysis over a one-year course. Kidney Int, 93:430-8. [DOI] [PubMed] [Google Scholar]
  • [366].Chu NM, Gross AL, Shaffer AA, Haugen CE, Norman SP, Xue QL, et al. (2019). Frailty and changes in cognitive function after kidney transplantation. J Am Soc Nephrol, 30:336-45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [367].Asai M, Kumakura S, Kikuchi M (2019). Review of the efficacy of ast-120 (kremezin((r))) on renal function in chronic kidney disease patients. Ren Fail, 41:47-56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [368].Schulman G, Agarwal R, Acharya M, Berl T, Blumenthal S, Kopyt N (2006). A multicenter, randomized, double-blind, placebo-controlled, dose-ranging study of ast-120 (kremezin) in patients with moderate to severe ckd. Am J Kidney Dis, 47:565-77. [DOI] [PubMed] [Google Scholar]
  • [369].Namikoshi T, Tomita N, Satoh M, Sakuta T, Kuwabara A, Kobayashi S, et al. (2009). Oral adsorbent ast-120 ameliorates endothelial dysfunction independent of renal function in rats with subtotal nephrectomy. Hypertens Res, 32:194-200. [DOI] [PubMed] [Google Scholar]
  • [370].Magnani S, Atti M (2021). Uremic toxins and blood purification: a review of current evidence and future perspectives. Toxins (Basel), 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [371].Igarashi H, Tsujita M, Kwee IL, Nakada T (2014). Water influx into cerebrospinal fluid is primarily controlled by aquaporin-4, not by aquaporin-1: 17o jjvcpe mri study in knockout mice. Neuroreport, 25:39-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [372].Xian P, Hei Y, Wang R, Wang T, Yang J, Li J, et al. (2019). Mesenchymal stem cell-derived exosomes as a nanotherapeutic agent for amelioration of inflammation-induced astrocyte alterations in mice. Theranostics, 9:5956-75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [373].Natale P, Ruospo M, Saglimbene VM, Palmer SC, Strippoli GF (2019). Interventions for improving sleep quality in people with chronic kidney disease. Cochrane Database Syst Rev, 5:D12625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [374].Kovacs GG (2017). Concepts and classification of neurodegenerative diseases. Handb Clin Neurol, 145:301-7. [DOI] [PubMed] [Google Scholar]
  • [375].Huang Y, Yang L (2021). Mesenchymal stem cells and extracellular vesicles in therapy against kidney diseases. Stem Cell Res Ther, 12:219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [376].Savio-Silva C, Soinski-Sousa PE, Balby-Rocha M, Lira AO, Rangel EB (2020). Mesenchymal stem cell therapy in acute kidney injury (aki): review and perspectives. Rev Assoc Med Bras (1992), 66 Suppl 1:s45-54. [DOI] [PubMed] [Google Scholar]
  • [377].Begcevic I, Brinc D, Brown M, Martinez-Morillo E, Goldhardt O, Grimmer T, et al. (2018). Brain-related proteins as potential csf biomarkers of alzheimer's disease: a targeted mass spectrometry approach. J Proteomics, 182:12-20. [DOI] [PubMed] [Google Scholar]
  • [378].Koscova S, Zakova SD, Tomeckova I, Melicherova K, Stelzer M, Janakova A, et al. (2017). Cerebrospinal fluid biomarkers in the diagnosis of creutzfeldt-jakob disease in slovak patients: over 10-year period review. Mol Neurobiol, 54:5919-27. [DOI] [PubMed] [Google Scholar]
  • [379].Lattanzio F, Abu-Rumeileh S, Franceschini A, Kai H, Amore G, Poggiolini I, et al. (2017). Prion-specific and surrogate csf biomarkers in creutzfeldt-jakob disease: diagnostic accuracy in relation to molecular subtypes and analysis of neuropathological correlates of p-tau and abeta42 levels. Acta Neuropathol, 133:559-78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [380].Oner AY, Barutcu B, Aykol S, Tali ET (2017). Intrathecal contrast-enhanced magnetic resonance imaging-related brain signal changes: residual gadolinium deposition? Invest Radiol, 52:195-7. [DOI] [PubMed] [Google Scholar]
  • [381].Bae YJ, Choi BS, Kim JM, Choi JH, Cho SJ, Kim JH (2021). Altered glymphatic system in idiopathic normal pressure hydrocephalus. Parkinsonism Relat Disord, 82:56-60. [DOI] [PubMed] [Google Scholar]
  • [382].Kiviniemi V, Wang X, Korhonen V, Keinanen T, Tuovinen T, Autio J, et al. (2016). Ultra-fast magnetic resonance encephalography of physiological brain activity - glymphatic pulsation mechanisms? J Cereb Blood Flow Metab, 36:1033-45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [383].Harrison IF, Siow B, Akilo AB, Evans PG, Ismail O, Ohene Y, et al. (2018). Non-invasive imaging of csf-mediated brain clearance pathways via assessment of perivascular fluid movement with diffusion tensor mri. Elife, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [384].Fultz NE, Bonmassar G, Setsompop K, Stickgold RA, Rosen BR, Polimeni JR, et al. (2019). Coupled electrophysiological, hemodynamic, and cerebrospinal fluid oscillations in human sleep. Science, 366:628-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [385].Xie L, Kang H, Xu Q, Chen MJ, Liao Y, Thiyagarajan M, et al. (2013). Sleep drives metabolite clearance from the adult brain. Science, 342:373-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [386].Iliff JJ, Chen MJ, Plog BA, Zeppenfeld DM, Soltero M, Yang L, et al. (2014). Impairment of glymphatic pathway function promotes tau pathology after traumatic brain injury. J Neurosci, 34:16180-93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [387].Yang L, Kress BT, Weber HJ, Thiyagarajan M, Wang B, Deane R, et al. (2013). Evaluating glymphatic pathway function utilizing clinically relevant intrathecal infusion of csf tracer. J Transl Med, 11:107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [388].Zhou Y, Cai J, Zhang W, Gong X, Yan S, Zhang K, et al. (2020). Impairment of the glymphatic pathway and putative meningeal lymphatic vessels in the aging human. Ann Neurol, 87:357-69. [DOI] [PubMed] [Google Scholar]
  • [389].Uygun DS, Yang C, Tilli ER, Katsuki F, Hodges EL, McKenna JT, et al. (2022). Knockdown ofgaba(a) alpha3 subunits on thalamic reticularneurons enhances deep sleep in mice. NatCommun, 13:2246. [DOI] [PMC free article] [PubMed] [Google Scholar]

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

RESOURCES