Next Article in Journal
S1P1 Threonine 236 Phosphorylation Mediates the Invasiveness of Triple-Negative Breast Cancer and Sensitivity to FTY720
Next Article in Special Issue
Loss of Astrocytic µ Opioid Receptors Exacerbates Aversion Associated with Morphine Withdrawal in Mice: Role of Mitochondrial Respiration
Previous Article in Journal / Special Issue
STAT3 Drives GFAP Accumulation and Astrocyte Pathology in a Mouse Model of Alexander Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Glymphatic System May Play a Vital Role in the Pathogenesis of Hepatic Encephalopathy: A Narrative Review

by
Ali Sepehrinezhad
1,2,
Fin Stolze Larsen
3,
Rezan Ashayeri Ahmadabad
4,
Ali Shahbazi
1,5 and
Sajad Sahab Negah
2,4,*
1
Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
2
Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad 9919191778, Iran
3
Department of Gastroenterology and Hepatology, Rigshospitalet, Copenhagen University Hospital, 999017 Copenhagen, Denmark
4
Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran 1449614535, Iran
5
Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
*
Author to whom correspondence should be addressed.
Cells 2023, 12(7), 979; https://doi.org/10.3390/cells12070979
Submission received: 19 October 2022 / Revised: 20 February 2023 / Accepted: 21 March 2023 / Published: 23 March 2023
(This article belongs to the Special Issue Astrocytes in CNS Disorders)

Abstract

:
Hepatic encephalopathy (HE) is a neurological complication of liver disease resulting in cognitive, psychiatric, and motor symptoms. Although hyperammonemia is a key factor in the pathogenesis of HE, several other factors have recently been discovered. Among these, the impairment of a highly organized perivascular network known as the glymphatic pathway seems to be involved in the progression of some neurological complications due to the accumulation of misfolded proteins and waste substances in the brain interstitial fluids (ISF). The glymphatic system plays an important role in the clearance of brain metabolic derivatives and prevents aggregation of neurotoxic agents in the brain ISF. Impairment of it will result in aggravated accumulation of neurotoxic agents in the brain ISF. This could also be the case in patients with liver failure complicated by HE. Indeed, accumulation of some metabolic by-products and agents such as ammonia, glutamine, glutamate, and aromatic amino acids has been reported in the human brain ISF using microdialysis technique is attributed to worsening of HE and correlates with brain edema. Furthermore, it has been reported that the glymphatic system is impaired in the olfactory bulb, prefrontal cortex, and hippocampus in an experimental model of HE. In this review, we discuss different factors that may affect the function of the glymphatic pathways and how these changes may be involved in HE.

1. Introduction

Hepatic encephalopathy (HE) is a neurological and neuropsychiatric complication following acute liver failure (ALF) or chronic liver disease (CLD) [1,2,3]. The exact pathophysiological mechanisms of HE are not fully understood. However, numerous studies have demonstrated that HE has complex pathology at the cellular molecular levels, e.g., astrocyte dysfunction plays a critical role in the pathogenesis of both forms of HE (i.e., ALF and CLD) [4,5,6,7,8,9]. Furthermore, substances including ammonia, glutamine, glutamate, aromatic amino acids, lactate, alanine, bile acids, and neurosteroids accumulate in the brain parenchyma during HE [10,11,12] and are directly implicated in the pathogenesis. Intracellular molecules are normally removed by two major protein degradation systems including the ubiquitin-proteasome system and autophagy in the neural cells [13]. Thereafter, cytosolic proteins and wastes are released into the interstitial space. These waste proteins and metabolic products are released into the cerebrospinal fluid (CSF) and finally are drained away by the glymphatic system [14]. This pathway provides a unidirectional fluid current to transport glucose and other nutrients, but the system is particularly known for the clearance of waste substances from the brain interstitial space [15]. Impairment of the glymphatic system, and the subsequent accumulation of waste soluble proteins has been proposed for the development of neurological disorders, such as Alzheimer’s disease [16,17,18], and is implicated in stroke [19], and traumatic brain injury [20,21]. Recently, researchers have mentioned the disruption of the glymphatic system in an experimental model of HE [22,23]. In this review, we focus on the components of the glymphatic pathway that may be compromised in HE.

2. An Overview

The glymphatic system is a system for waste proteins and metabolic products clearance from interstitial fluids (ISF) in the brain parenchyma. The name “glymphatic” derives from the major role of astrocytes, the central nervous system’s most abundant glial cells [24] in this system that operates like a lymphatic system. Its unidirectional flow initiates from Virchow-Robin spaces or periarterial spaces, where CSF flows from the subarachnoid space to the brain tissue along the penetrating arterioles (Figure 1). The driving force for the influx of CSF to the ISF and movement of CSF-ISF into the interstitial space is arterial pulsation [25,26]. Astrocytic end-feet express aquaporin 4 (AQP4) water channels, where astrocytes and endothelial cells constitute barriers between CSF and ISF [27,28]. AQP4 channels are bidirectional and facilitate water flux between blood circulation, periarterial space, interstitial space, and perivenous space. These channels decrease the resistance of CSF flow from the periarterial space into the ISF and facilitate the movement of ISF toward the perivenous space due to the expression on astrocyte processes [29]. CSF-ISF fluid subsequently enters the perivenous space and releases interstitial metabolic agents, soluble proteins, and neurotoxic factors, including lactate, glutamine, glutamate, tau, and amyloid peptides. Finally, waste products are released into the meningeal lymphatic vessels in the dura mater and are transported to the deep cervical lymph nodes (Figure 1) [20]. Enhancing the drainage of CSF from brain tissue ensures the proper functioning of the glymphatic pathway as a powerful driving force for CSF-ISF flow.

3. Factors Affecting the Function of the Glymphatic System

The normal function of the glymphatic pathway is to eliminate misfolded proteins completely, maintain water and ions equilibrium, and regulate the CSF, ISF, and intracranial pressure. If any element of the glymphatic pathway is impaired, waste accumulates in the brain ISF. Such impairments occur in the development of neurological disorders. Several factors can affect the glymphatic function in the central nervous system (CNS), such as circadian cycle, body posture, aging, and autonomic innervation. The function of the glymphatic system decreases during wakefulness and becomes better during sleep and under anesthesia conditions [25]. Sleep and anesthesia lead to interstitial space expansion and promote the movement of CSF flow in the perivascular spaces in mice [25]. Another study of rodents showed that the glymphatic system is controlled under circadian rhythms, and AQP4 localization on perivascular astrocyte end-feet is highest in sleepy mice [26]. In a human study using intrathecal injection of gadobutrol as a CSF tracer, glymphatic function was also highest over-night compared to daytime, which indicates the role of sleep for maintenance of an efficient glymphatic pathway [30]. Anesthesia following injection of pentobarbital also improved glymphatic activity in mice [31]. In terms of other factors, the effect of body posture was also evaluated in that latter study. Humans and animals display different body postures at every moment and during sleep and wakefulness. Previous studies have shown that body position affects cerebral blood circulation and intracranial pressure [32,33]. In one study, dynamic-contrast-enhanced magnetic resonance imaging (MRI) and a radioactive CSF tracer revealed that CSF-ISF flow and waste clearance of ISF improved in lateral position in rats [34]. Aging is another factor that has negative effects on the function of the glymphatic system. Aging decreases vascular elasticity and, therefore, reduces the power of arterial pulsation [35,36]. Studies in mice have shown that aging suppresses CSF flow, CSF-ISF flow, and clearance of waste substances [37,38]. Decreasing the levels of collagen type IV and other basement membrane proteins is associated with vascular stiffness and suppression of the amplitude of arterial pulsation, which leads to impairment of CSF influx into the interstitial space [39]. Aging also affects the morphogenesis and cellular behavior of astrocytes, which are fundamental to the structure of glymphatic corridors [40,41]. Furthermore, the distribution of AQP4 throughout the brain was changed with aging [42,43]. The autonomic nervous system (ANS) is another factor that affects the glymphatic system. Activation of the sympathetic nervous system (SNS) can exert negative effects on the glymphatic system through the main neurotransmitter norepinephrine. Reciprocally, activation of the parasympathetic nervous system (PNS) through the vagus nerve positively regulates and improves glymphatic function [44]. Administration of dexmedetomidine, a suppressor of norepinephrine release from locus coeruleus neurons, enhanced glymphatic function in the hippocampus of lightly anesthetized rats [45]. Likewise, intracisternal injection of adrenergic receptor antagonists increased CSF influx into the ISF in conscious mice, as evaluated by a CSF tracer [25]. These studies suggest that norepinephrine and basal tone of the SNS are responsible for suppression of glymphatic transports during wakefulness. Stimulation of the vagus nerve through an implanted device enhanced CSF influx into the ISF and improved CSF-ISF flow in the brain of mice [44].

4. Direct Evidence of Glymphatic System Impairment in HE

The first evidence of a dysfunctional glymphatic system in HE was reported by Hadjihambi and her colleagues at University College London [22]. In this study, they measured the glymphatic function in brain parenchyma in bile duct ligation (BDL) rats as chronic liver disease-induced HE model, using dynamic contrast-enhanced MRI and mass-spectroscopy. Decreased penetration of the contrast agent gadolinium indicated reduced CSF-ISF flow in the olfactory bulb and prefrontal cortex of BDL rats. These findings correlated with reduced expression of AQP4 in these brain regions [22]. Thus, they concluded that reduced expression of AQP4 mediated glymphatic impairment and behavioral deficits in HE rats [22]. Furthermore, Jung Hsu et al. [46] demonstrated that the distribution of fluorescent ovalbumin dye as an indicator of glymphatic function was impaired in the brain parenchyma and later in deep cervical lymph nodes in BDL rats compared to the control animals. In addition, the BDL rats displayed microglia activation and neuroinflammation. This study indicated that dysfunctional glymphatic clearance was caused by impaired meningeal lymphatic drainage in HE [46]. A review paper by Claeys et al. [47] explained that impaired glymphatic clearance of waste substances may contribute to the pathophysiology of HE. An editorial letter by Gallina et al. [23] also hypothesized that several abnormalities (i.e., cardiomyopathy-heart failure, pulmonary hypertension, alternation of intracranial pressure, hyperdynamic circulation, and decreased venous outflow) might contribute to the impairment of glymphatic function following liver disease and HE. In this review we describe in detail the links between HE pathogenesis and malfunction of the glymphatic system. The presence of such links suggests areas for future research.

5. Factors That May Indirectly Affect the Efficacy of Glymphatic System in Hepatic Encephalopathy

5.1. Sleep Disturbances in HE

As mentioned earlier, the sleep–wake cycle can affect the glymphatic system. Sleep improves the function of glymphatic system, while wakefulness reduces it. Sleep disturbance is quite common in patients with liver insufficiency well before HE becomes clinically overt [48,49]. A neuroimaging study suggested that sleep disturbance is a valuable diagnostic sign for subclinical HE in cirrhotic patients [50]. Moreover, decreased sleep quality and daily drowsiness measured by polysomnography and sleep indexes have been reported in HE patients [48,51,52]. Long-lasting sleep abnormalities were also revealed in patients with portosystemic shunt encephalopathy [53]. In an experimental model of this type of HE, disruption of sleep patterns and significant reduction of both rapid eye movement sleep and non-rapid eye movement compared to control rats were shown [54]. Similar findings, such as increased total duration of wakefulness and decreased slow-wave sleep and paradoxical sleep times, have been reported in rats with hyperammonemia [55]. Therefore, manipulating the sleep–wake cycle and correcting sleep disturbances can be an effective effort for the recovery of glymphatic system function and consequently result in ameliorating HE.

5.2. Cardiomyopathy, Arterial Hypotension, and Impaired Cerebral Blood Flow in Cirrhosis and HE

Starling forces regulate the fluid flux across the cerebral microvessels, but are severely imbalanced in liver failure, especially if the normal function of the BBB is compromised [56,57]. The main driving forces for CSF influx into the interstitial space and CSF-ISF flow in the brain parenchyma are arterial pulsation induced by the amplitude of the cerebral perfusion pressure (mean arterial pressure (MAP) minus intracranial pressure) [58]. Decreased inotropic and chronotropic properties of heart muscles, reduced peripheral vascular resistance, and reduced systemic blood pressure are commonly seen in patients with insufficient liver function [59,60,61]. Ventricular diastolic dysfunction, left atrial enlargement [62], and both systolic and diastolic cardiac failures [63,64] have been reported in cirrhotic patients. Furthermore, an autopsy study has shown ventricular hypertrophy, cardiac valve calcification, interstitial edema, fibrosis in myocytes, and coronary artery atherosclerosis in cirrhotic patients [65]. Cardiomyopathy as systolic and diastolic abnormalities, inotropic and chronotropic dysfunction, increased plasma levels of myocardial enzymes, myocyte swelling and hypertrophy, and gross abnormalities in cardiomyocytes in histological examinations have also been reported in animal models of cirrhosis and HE [66,67,68,69]. Reduced response to vasoconstrictors and accumulation of circulatory vasodilators, such as nitric oxide, carbon monoxide, and endocannabinoids explain the suppression of systemic vascular resistance in liver failure [70,71,72,73]. In patients with acute liver failure and HE, arterial hypotension is very common [74]. Similar findings have also been shown in the experimental model of HE induced by bile duct ligation in rats, where the systolic blood pressure, measured by tail-cuff, significantly decreased compared to the sham group. Furthermore, injection of thioacetamide as a hepatotoxic agent for induction of HE resulted in a decrease in systolic blood pressure [75]. Dysregulation of cerebral blood flow (CBF) is another important contributing factor that may affect glymphatic transports. Normal regulation of CBF plays a critical role in the supply of vital nutrients to the neuronal cells and elimination of waste agents from brain parenchyma. The cerebral microcirculation is controlled by the partial carbon dioxide tension in the blood, the ANS and cerebral perfusion pressure; i.e., by the CBF autoregulation [76,77]. Numerous human and animal studies have reported that the normal regulation of CBF is impaired in liver failure and HE [12,78,79,80,81,82]. The intracranial pressure increases in severe HE due to astrocyte swelling [83,84,85,86,87,88,89]. Intracranial hypertension also leads to a reduction in cerebral perfusion pressure in patients with acute liver failure [86,90,91,92,93,94]. The combination of arterial hypotension, intracranial hypertension, and impaired regulation of CBF will tend to compromise all the transition pathways of the glymphatic system, aggravating the accumulation of waste metabolic products in the ISF as previously observed in patients with acute liver failure [95,96,97]. This further underscores the importance of the current clinical practice to promptly correct arterial hypotension and hyperammonemia to avoid brain edema and high intracranial pressure, thereby improving CSF distribution in para-arterial spaces and CSF-ISF flow in brain interstitial space.

5.3. Alteration in the Function of Autonomic Nervous System in HE

As mentioned in Section 3, ANS affects function of the glymphatic system. Hyperactivity of sympathetic tone and suppression of vagal tone is present in cirrhosis [98,99,100,101]. A recent study showed ANS imbalance in patients with chronic liver disease [102]. Activation of the SNS and elevated levels of norepinephrine in the systemic circulation of cirrhotic patients have been reported [98,103]. Furthermore, PNS activity, as the vagal tone, is strongly suppressed in patients with end-stage liver disease and candidates for liver transplantation [101]. Another study showed an increase in the tonicity of the SNS in patients with high grades of HE compared to healthy control individuals [104]. There was also a considerable change in the function of ANS in patients with HE in another research [105]. Hyperactivity of the SNS is also seen in experimental models of HE induced by thioacetamide in rats [106]. Plasma levels of norepinephrine were found to be increased in rat models of cirrhosis [107,108]. Regarding these findings, modulation of ANS function to restore parasympathetic balance could be of importance to preserve the normal functioning of the glymphatic system, but further study is needed.

5.4. Astrocyte Dysfunction and AQP4 Mislocalization in HE

The normal functioning of the glymphatic system in the clearance of ISF requires the presence of healthy astrocytes as a main component. Astrocytes are the most abundant brain glial cells, especially in the cerebral cortex, which regulate ion equilibrium in the extracellular space, provide nutrient agents for their surrounding cells, particularly neuronal cells, release neurotransmitters at the location of astrocyte-neuron and astrocyte-microglia interactions, repair injured regions of CNS, support cerebral microvessels endothelial cells, contribute to the BBB, and finally detoxify many neurotoxic substances (e.g., ammonia and osmotic agents) from the brain interstitial space [10,109,110,111]. The function and morphology of astrocytes are strongly impaired following HE [10]. Glial fibrillary acidic protein (GFAP) is an intermediate filament protein that is intensely expressed in astrocyte cells. GFAP plays an important role in maintaining cell structure, cell proliferation, astrocyte-neuron communication, and in the formation of glial scars following brain injury [112,113]. Postmortem analysis of brain samples from HE patients has revealed decreased protein levels of GFAP in the cerebral cortex and basal ganglia [114]. In the same way, experimental studies on animal models of HE have reported decreased expression of GFAP in several brain areas, such as substantia nigra, ventral tegmental area, hippocampus [115], sensorimotor cortex, thalamus [116], and subcortical white matter [117,118]. In hyperammonemic conditions, astrocytes are primarily the cells that detoxify ammonia from brain parenchyma through their glutamine synthetase (GS) enzyme [119]. The enzyme catalyzes the conversion of ammonia and glutamate to glutamine [119]. Glutamine, which is an active osmolyte, triggers the influx of water into the cell. In hyperammonemic conditions, accumulation of glutamine following ammonia detoxification leads to a hypertonic state, and the influx of water into the astrocytes results in astrocyte swelling [119,120,121,122]. A set of pathological agents and processes, such as inflammation, oxidative stress, ATP depletion, cell senescence, osmolyte accumulation, mitochondrial dysfunction, impaired GFAP, and AQP4 expression triggers and even exacerbates astrocyte swelling following hyperammonemic conditions [10]. Microglia activation-induced neuroinflammation is another factor by which production of proinflammatory cytokines and production of oxidative stress worsens astrocyte swelling following hyperammonemia [123,124,125]. Normal homeostasis of the internal environment of the brain tissue requires an intact BBB in the region of cerebral microvasculature [126]. The BBB is made up of several components: endothelial cells that are joined together with tight junctions, pericytes that are located on the abluminal side and incompletely cover the endothelial cells, and astrocyte end-feet that act as linkers between neuronal networks and blood vessels [127]. This barrier is semipermeable and allows some ions and crucial nutrients (i.e., glucose and amino acids) to transport between blood and brain parenchyma, which ensures the survival of neuronal cells. However, it should be mentioned that the BBB is strongly impermeable to substances for which it does not have a transporter due to tight junctions composed of occludin, claudins, and junctional adhesion molecules; thus, it prevents the entry of circulatory substances into the brain parenchyma [128]. Impairment of this barrier can make it permeable to many circulatory neurotoxic agents, such as ammonia, bile acids, proinflammatory cytokines, and bacterial toxins like lipopolysaccharides [129,130]. Disruption of the BBB may also cause penetration of inflammatory cells such as phagocytic cells, lymphocytes, and antigen-presenting cells into the brain tissue and result in the induction of neuroinflammation and production of reactive oxygen species [131,132,133,134,135]. Therefore, increasing the permeability of the BBB can exacerbate astrocyte damage/swelling, as the main component of glymphatic system, through induction of neuroinflammation and oxidative stress and activation of microglia cells. BBB disruption has been reported in the pathogenesis of HE from experimental to clinical studies. For example, treatment of the human cerebral microvessel endothelial cell (hCMEC/D3) line as an in vitro model of the BBB with plasma samples from cirrhotic patients with HE increases trans-endothelial migration of leukocytes [136]. Increased extravasation of Evans blue dye, as an indicator of BBB disruption, in mice models of HE has been reported [137,138]. Ultrastructural analysis of cerebral cortex, using transmission electron microscopy and [3H] inulin, in a rat model of HE has revealed increased leakage in cerebral capillaries [139]. Moreover, the protein level of S100 calcium-binding protein B, as a marker of BBB disruption, increased in serum samples of HE patients [140,141]. As a result, BBB dysfunction in HE is linked with astrocyte swelling and cerebral edema [139,142,143,144,145,146,147,148,149,150,151,152,153,154]. Microglia activation and neuroinflammation are triggered by astrogliosis and induction of the reactive inflammatory A1 phenotype subtype of astrocytes [155]. These reactive phenotypes release pro-inflammatory cytokines such as TNFα and IL-1β which induce BBB disruption, neurodegeneration, and gliopathy [156,157,158,159].
AQP4 water channels are integral membrane proteins that are localized on astrocytic end-feet surrounding the capillaries and facilitate the maintenance of cerebral water flow through their bidirectional properties [160]. However, AQP4 is expressed on plasma membrane instead of perivascular end-feet in swollen and reactive astrocytes [161,162,163,164,165,166]. This AQP4 mislocalization may be one of the main reasons for glymphatic impairment in several neurological diseases (Figure 2) [21,163,167,168]. For instance, the loss of perivascular AQP4 localization as a main part of the glymphatic system was assessed on clearance of amyloid β (Aβ) in human postmortem cases and the α-syntrophin knockout mouse model, which lacks perivascular AQP4 localization. Data indicated that the perivascular localization of AQP4 is significantly decreased in frontal cortical gray matter of AD cases compared to cognitively intact subjects. In the α-syntrophin knockout mouse, CSF influx, CSF-ISF flow, and perivenous interstitial fluids efflux were significantly decreased, while Aβ was increased [167]. Mislocalization of perivascular astrocyte AQP4 and impairment of the glymphatic system have been well identified in AD [16,169,170,171]. Postmortem cerebral cortex analyses from AD patients found that the loss of immunofluorescence for perivascular AQP4 was associated with misfolded amyloid-β [167,172]. The AQP4 channels were also decreased in perivascular reactive astrocytes of the APP/PS1 mice model of AD and aged mice, and the mislocalization was accompanied by a decrease in the transport of a fluorescent tracer as an indicator of lymphatic clearance in brain interstitial space [37,169]. Mislocalization of AQP4 channels on reactive perivascular astrocytes was also reported in mice models of traumatic brain injury (TBI) [173,174]. Moreover, localization of AQP4 on perivascular reactive astrocytes was impaired in hippocampus of TBI mice together with accumulation of hyperphosphorylated tau proteins in this region [163]. Immunofluorescence and tracer tracking techniques demonstrated that perivascular AQP4 mislocalized on reactive astrocytes was associated with a 60% decrease in glymphatic function in TBI mice [21]. Impairment of glymphatic clearance was also reported in the limbic system of TBI rats using dye tracing and MRI [175]. Astrocyte swelling and astrogliosis along with mislocalization of perivascular astrocytic AQP4 as well as glymphatic impairment were also revealed in stroke [162,176,177,178,179]. It is concluded that mislocalization of AQP4 from perivascular end-feet to the cell body in reactive astrocytes is associated with the reduction of efficacy of the glymphatic system and the onset of pathological cerebral findings following AD, TBI, and stroke (Figure 2). This raises the question, does the same mechanism occur in HE? In the context of liver diseases, it seems reasonable to consider similar mechanisms for HE onset. Astrocyte swelling, Alzheimer type II astrocytes, and reactive astrocytes are also predominant cerebral changes in HE that are associated with neurotoxicity [9,180,181,182,183]. These predominant phenotypes of astrocytes may negatively impact glymphatic function in HE as seen in AD, TBI, and stroke. Mislocalization of AQP4 channels in reactive perivascular astrocytes have been previously reported in liver diseases and HE [22,184,185,186,187]. A postmortem analysis of different brain regions (i.e., striatum, cerebellum, thalamus, hippocampus, and cortex) from cirrhotic patients indicated that the expression of AQP4 channels localized to the cell body’s plasma membrane was significantly increased [184]. Furthermore, decreased expression of AQP4 in the olfactory bulb and prefrontal cortex of BDL rats along with the impairment of the glymphatic system, using dynamic contrast-enhanced MRI and mass-spectroscopy, was also observed [22]. In these rats, penetration of gadolinium as an indicator of subarachnoid CSF-ISF flow as visualized by dynamic contrast-enhanced MRI was significantly decreased in prefrontal cortex and olfactory bulbs as compared to the control [22]. Moreover, microarray expression analyses of astrocytes isolated from hyperammonemic mice showed that the expression of the gap junction proteins connexin-43 and AQP4 was decreased compared to the control cells [187]. In addition to that, astrocytes exposed to ammonia in culture medium decreased the expression of AQP4 and changed the arrangement of AQP4 channels on the plasma membrane [186]. Based on the existing data, it is suggested that mislocalization of AQP4 water channels on astrocytes in HE may impair glymphatic clearance and waste aggregation (Figure 2; Table 1.). Further research is needed to confirm the role of the glymphatic system dysfunction in the context of HE.

6. Summary

The glymphatic system is an astrocyte-dependent unidirectional transport pathway that empties macromolecules and waste substances from the brain interstitial space. HE mainly evolves and progresses due to hyperammonemia and inflammation [217]. Accumulation of some neurotoxic and osmotic agents such as ammonia, glutamine, reactive oxygen species, glutamate, alanine, aromatic amino acids, lactate, and proinflammatory cytokines, into the brain parenchyma and interstitial space is considered a central issue in the development and progression of HE [10]. Therefore, an efficient glymphatic pathway that can help remove these waste agents from the brain parenchyma may be of potential clinical interest. For instance, it has been shown that an increase in meningeal lymphangiogenesis through injection of adeno-associated virus 8-vascular endothelial growth factor C (AAV8-VEGF-C) into the cisterna magna of bile duct ligation-induced rats with HE reduced microglial activation, improved the glymphatic function, decreased the expression of proinflammatory cytokines in the cerebral cortex, and improved the motor function [46]. However, there remains a paucity of evidence on the impairment of the glymphatic system HE, or on the effect of treatments such as liver transplantation on its function. Further systematic studies are thus warranted. For the first time, Hadjihambi et al. showed evidence of glymphatic system impairment in the BDL rats model of HE using neuroimaging and immunofluorescence techniques [22]. Here we discussed different levels of pathological changes in HE that can be related to the glymphatic pathway, including the sleep–wake cycle, arterial pulsation, astrocyte morphology and function, AQP4 expression patterns, and meningeal lymphangiogenesis, to draw attention to this topic for future studies (Figure 3).

Author Contributions

A.S. (Ali Sepehrinezhad) designed the study, performed the literature review, drafted the manuscript, and drew and prepared the figures. S.S.N. performed the literature review, supervised the work, corrected grammatical errors, and drafted the manuscript. F.S.L. drafted the manuscript, critically and scientifically edited the manuscript. R.A.A. clinically edited the whole manuscript, corrected grammatical errors, and added extra information where needed. A.S. (Ali Shahbazi) critically edited the final version. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AAV8-VEGF-C: adeno-associated virus 8-vascular endothelial growth factor C; AQP4: aquaporin 4; BBB: blood–brain barrier; CBF: cerebral blood flow; CNS: central nervous system; CSF: cerebrospinal fluid; GFAP: glial fibrillary acidic protein; hCMEC/D3: human cerebral microvessel endothelial cells; HE: hepatic encephalopathy; ICP: intracranial pressure; ISF: interstitial fluid; MAP: mean arterial pressure; mLVs: meningeal lymphatic vessels; MRI: magnetic resonance imaging.

References

  1. Wijdicks, E.F.M. Hepatic Encephalopathy. N. Engl. J. Med. 2016, 375, 1660–1670. [Google Scholar] [CrossRef] [PubMed]
  2. Stewart, C.A.; Malinchoc, M.; Kim, W.R.; Kamath, P.S. Hepatic encephalopathy as a predictor of survival in patients with end-stage liver disease. Liver Transplant. 2007, 13, 1366–1371. [Google Scholar] [CrossRef] [PubMed]
  3. Bloom, P.P.; Tapper, E.B.; Young, V.B.; Lok, A.S. Microbiome therapeutics for hepatic encephalopathy. J. Hepatol. 2021, 75, 1452–1464. [Google Scholar] [CrossRef] [PubMed]
  4. Vigo, M.B.; Pérez, M.J.; De Fino, F.; Gómez, G.; Martínez, S.A.; Bisagno, V.; Di Carlo, M.B.; Scazziota, A.; Manautou, J.E.; Ghanem, C.I. Acute acetaminophen intoxication induces direct neurotoxicity in rats manifested as astrogliosis and decreased dopaminergic markers in brain areas associated with locomotor regulation. Biochem. Pharmacol. 2019, 170, 113662. [Google Scholar] [CrossRef]
  5. Tevethia, H.V.; Choudhury, A.; Maiwall, R.; Mitra, L.; Saluja, V.; Singh, P.; Trigatia, A.; Kumar, G.; Thapar, S.; Sarin, S.K. Noninvasive diagnosis of cerebral edema in patients of acute liver failure. Hepatol. Int. 2018, 12, S614. [Google Scholar] [CrossRef]
  6. Tamnanloo, F.; Ochoa-Sanchez, R.; Tremblay, M.; Rose, C.F. Repeated ammonia-induced episodes of hepatic encephalopathy leads to neuronal cell loss in rats with chronic liver disease. Hepatology 2022, 76, S1168–S1169. [Google Scholar] [CrossRef]
  7. Claeys, W.; Van Hoecke, L.; Geerts, A.; Van Vlierberghe, H.; Lefere, S.; Van Imschoot, G.; Van Wonterghem, E.; Ghesquière, B.; Vandenbroucke, R.E.; Van Steenkiste, C. A mouse model of hepatic encephalopathy: Bile duct ligation induces brain ammonia overload, glial cell activation and neuroinflammation. Sci. Rep. 2022, 12, 17558. [Google Scholar] [CrossRef] [PubMed]
  8. Balzano, T.; Forteza, J.; Molina, P.; Giner, J.; Monzó, A.; Sancho-Jiménez, J.; Urios, A.; Montoliu, C.; Felipo, V. Lymphocyte infiltration, glial activation and neuronal loss in cerebellum of patients with different stages of chronic liver disease. Glia 2019, 67, E518–E519. [Google Scholar] [CrossRef] [Green Version]
  9. Agarwal, A.N.; Mais, D.D. Sensitivity and Specificity of Alzheimer Type II Astrocytes in Hepatic Encephalopathy. Arch. Pathol. Lab. Med. 2019, 143, 1256–1258. [Google Scholar] [CrossRef] [Green Version]
  10. Sepehrinezhad, A.; Zarifkar, A.; Namvar, G.; Shahbazi, A.; Williams, R. Astrocyte swelling in hepatic encephalopathy: Molecular perspective of cytotoxic edema. Metab. Brain Dis. 2020, 35, 559–578. [Google Scholar] [CrossRef]
  11. Weiss, N.; Barbier Saint Hilaire, P.; Colsch, B.; Isnard, F.; Attala, S.; Schaefer, A.; Amador, M.D.; Rudler, M.; Lamari, F.; Sedel, F.; et al. Cerebrospinal fluid metabolomics highlights dysregulation of energy metabolism in overt hepatic encephalopathy. J. Hepatol 2016, 65, 1120–1130. [Google Scholar] [CrossRef] [PubMed]
  12. Bjerring, P.N.; Gluud, L.L.; Larsen, F.S. Cerebral blood flow and metabolism in hepatic encephalopathy—A meta-analysis. J. Clin. Exp. Hepatol. 2018, 8, 286–293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Nedelsky, N.B.; Todd, P.K.; Taylor, J.P. Autophagy and the ubiquitin-proteasome system: Collaborators in neuroprotection. Biochim. Et Biophys. Acta 2008, 1782, 691–699. [Google Scholar] [CrossRef] [Green Version]
  14. Zhang, C.; Lin, J.; Wei, F.; Song, J.; Chen, W.; Shan, L.; Xue, R.; Wang, G.; Tao, J.; Zhang, G.; et al. Characterizing the glymphatic influx by utilizing intracisternal infusion of fluorescently conjugated cadaverine. Life Sci. 2018, 201, 150–160. [Google Scholar] [CrossRef] [PubMed]
  15. Abbott, N.J.; Pizzo, M.E.; Preston, J.E.; Janigro, D.; Thorne, R.G. The role of brain barriers in fluid movement in the CNS: Is there a ‘glymphatic’ system? Acta Neuropathol. 2018, 135, 387–407. [Google Scholar] [CrossRef] [Green Version]
  16. Reeves, B.C.; Karimy, J.K.; Kundishora, A.J.; Mestre, H.; Cerci, H.M.; Matouk, C.; Alper, S.L.; Lundgaard, I.; Nedergaard, M.; Kahle, K.T. Glymphatic System Impairment in Alzheimer’s Disease and Idiopathic Normal Pressure Hydrocephalus. Trends Mol. Med. 2020, 26, 285–295. [Google Scholar] [CrossRef]
  17. Silva, I.; Silva, J.; Ferreira, R.; Trigo, D. Glymphatic system, AQP4, and their implications in Alzheimer’s disease. Neurol. Res. Pract. 2021, 3, 5. [Google Scholar] [CrossRef]
  18. Harrison, I.F.; Ismail, O.; Machhada, A.; Colgan, N.; Ohene, Y.; Nahavandi, P.; Ahmed, Z.; Fisher, A.; Meftah, S.; Murray, T.K.; et al. Impaired glymphatic function and clearance of tau in an Alzheimer’s disease model. Brain 2020, 143, 2576–2593. [Google Scholar] [CrossRef]
  19. Goulay, R.; Flament, J.; Gauberti, M.; Naveau, M.; Pasquet, N.; Gakuba, C.; Emery, E.; Hantraye, P.; Vivien, D.; Aron-Badin, R.; et al. Subarachnoid Hemorrhage Severely Impairs Brain Parenchymal Cerebrospinal Fluid Circulation in Nonhuman Primate. Stroke 2017, 48, 2301–2305. [Google Scholar] [CrossRef]
  20. Plog, B.A.; Dashnaw, M.L.; Hitomi, E.; Peng, W.; Liao, Y.; Lou, N.; Deane, R.; Nedergaard, M. Biomarkers of traumatic injury are transported from brain to blood via the glymphatic system. J. Neurosci. 2015, 35, 518–526. [Google Scholar] [CrossRef] [Green Version]
  21. Iliff, J.J.; Chen, M.J.; Plog, B.A.; Zeppenfeld, D.M.; Soltero, M.; Yang, L.; Singh, I.; Deane, R.; Nedergaard, M. Impairment of glymphatic pathway function promotes tau pathology after traumatic brain injury. J. Neurosci. 2014, 34, 16180–16193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Hadjihambi, A.; Harrison, I.F.; Costas-Rodríguez, M.; Vanhaecke, F.; Arias, N.; Gallego-Durán, R.; Mastitskaya, S.; Hosford, P.S.; Olde Damink, S.W.M.; Davies, N.; et al. Impaired brain glymphatic flow in experimental hepatic encephalopathy. J. Hepatol. 2019, 70, 40–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Gallina, P.; Gallo, O.; Nicoletti, C.; Romanelli, R.G. A hydrodynamic hypothesis for the pathogenesis of glymphatic system impairment in hepatic encephalopathy. J. Hepatol. 2019, 71, 228–229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Valenza, M.; Facchinetti, R.; Steardo, L.; Scuderi, C. Altered waste disposal system in aging and Alzheimer’s disease: Focus on astrocytic aquaporin-4. Front. Pharmacol. 2020, 10, 1656. [Google Scholar] [CrossRef] [PubMed]
  25. Xie, L.; Kang, H.; Xu, Q.; Chen, M.J.; Liao, Y.; Thiyagarajan, M.; O’Donnell, J.; Christensen, D.J.; Nicholson, C.; Iliff, J.J. Sleep drives metabolite clearance from the adult brain. Science 2013, 342, 373–377. [Google Scholar] [CrossRef] [Green Version]
  26. Hablitz, L.M.; Plá, V.; Giannetto, M.; Vinitsky, H.S.; Stæger, F.F.; Metcalfe, T.; Nguyen, R.; Benrais, A.; Nedergaard, M. Circadian control of brain glymphatic and lymphatic fluid flow. Nat. Commun. 2020, 11, 4411. [Google Scholar] [CrossRef]
  27. Camassa, L.M.A.; Lunde, L.K.; Hoddevik, E.H.; Stensland, M.; Boldt, H.B.; De Souza, G.A.; Ottersen, O.P.; Amiry-Moghaddam, M. Mechanisms underlying AQP4 accumulation in astrocyte endfeet. Glia 2015, 63, 2073–2091. [Google Scholar] [CrossRef]
  28. De Bellis, M.; Pisani, F.; Mola, M.G.; Rosito, S.; Simone, L.; Buccoliero, C.; Trojano, M.; Nicchia, G.P.; Svelto, M.; Frigeri, A. Translational readthrough generates new astrocyte AQP4 isoforms that modulate supramolecular clustering, glial endfeet localization, and water transport. Glia 2017, 65, 790–803. [Google Scholar] [CrossRef]
  29. Mestre, H.; Hablitz, L.M.; Xavier, A.L.; Feng, W.; Zou, W.; Pu, T.; Monai, H.; Murlidharan, G.; Rivera, R.M.C.; Simon, M.J. Aquaporin-4-dependent glymphatic solute transport in the rodent brain. Elife 2018, 7, e40070. [Google Scholar] [CrossRef]
  30. Eide, P.K.; Ringstad, G. Delayed clearance of cerebrospinal fluid tracer from entorhinal cortex in idiopathic normal pressure hydrocephalus: A glymphatic magnetic resonance imaging study. J. Cereb. Blood Flow Metab. 2019, 39, 1355–1368. [Google Scholar] [CrossRef]
  31. Hablitz, L.M.; Vinitsky, H.S.; Sun, Q.; Stæger, F.F.; Sigurdsson, B.; Mortensen, K.N.; Lilius, T.O.; Nedergaard, M. Increased glymphatic influx is correlated with high EEG delta power and low heart rate in mice under anesthesia. Sci. Adv. 2019, 5, eaav5447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Brosnan, R.J.; Steffey, E.P.; LeCouteur, R.A.; Imai, A.; Farver, T.B.; Kortz, G.D. Effects of body position on intracranial and cerebral perfusion pressures in isoflurane-anesthetized horses. J. Appl. Physiol. 2002, 92, 2542–2546. [Google Scholar] [CrossRef] [Green Version]
  33. Kose, G.; Hatipoglu, S. Effect of head and body positioning on cerebral blood flow velocity in patients who underwent cranial surgery. J. Clin. Nurs. 2012, 21, 1859–1867. [Google Scholar] [CrossRef] [PubMed]
  34. Lee, H.; Xie, L.; Yu, M.; Kang, H.; Feng, T.; Deane, R.; Logan, J.; Nedergaard, M.; Benveniste, H. The Effect of Body Posture on Brain Glymphatic Transport. J. Neurosci. 2015, 35, 11034–11044. [Google Scholar] [CrossRef] [PubMed]
  35. Reddy, A.K.; Li, Y.-H.; Pham, T.T.; Ochoa, L.N.; Treviño, M.T.; Hartley, C.J.; Michael, L.H.; Entman, M.L.; Taffet, G.E. Measurement of aortic input impedance in mice: Effects of age on aortic stiffness. Am. J. Physiol.-Heart Circ. Physiol. 2003, 285, H1464–H1470. [Google Scholar] [CrossRef] [Green Version]
  36. Avolio, A.P.; Kuznetsova, T.; Heyndrickx, G.R.; Kerkhof, P.L.M.; Li, J.K. Arterial Flow, Pulse Pressure and Pulse Wave Velocity in Men and Women at Various Ages. Adv. Exp. Med. Biol. 2018, 1065, 153–168. [Google Scholar] [CrossRef]
  37. Kress, B.T.; Iliff, J.J.; Xia, M.; Wang, M.; Wei, H.S.; Zeppenfeld, D.; Xie, L.; Kang, H.; Xu, Q.; Liew, J.A.; et al. Impairment of paravascular clearance pathways in the aging brain. Ann. Neurol. 2014, 76, 845–861. [Google Scholar] [CrossRef] [Green Version]
  38. Ma, Q.; Ineichen, B.V.; Detmar, M.; Proulx, S.T. Outflow of cerebrospinal fluid is predominantly through lymphatic vessels and is reduced in aged mice. Nat. Commun. 2017, 8, 1434. [Google Scholar] [CrossRef] [Green Version]
  39. Chandra, P.K.; Cikic, S.; Rutkai, I.; Guidry, J.J.; Katakam, P.V.G.; Mostany, R.; Busija, D.W. Effects of aging on protein expression in mice brain microvessels: ROS scavengers, mRNA/protein stability, glycolytic enzymes, mitochondrial complexes, and basement membrane components. GeroScience 2022, 44, 371–388. [Google Scholar] [CrossRef]
  40. Bronzuoli, M.R.; Facchinetti, R.; Valenza, M.; Cassano, T.; Steardo, L.; Scuderi, C. Astrocyte Function Is Affected by Aging and Not Alzheimer’s Disease: A Preliminary Investigation in Hippocampi of 3xTg-AD Mice. Front. Pharmacol. 2019, 10, 644. [Google Scholar] [CrossRef] [Green Version]
  41. Palmer, A.L.; Ousman, S.S. Astrocytes and Aging. Front. Aging Neurosci. 2018, 10, 337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Shen, X.Q.; Miyajima, M.; Ogino, I.; Arai, H. Expression of the water-channel protein aquaporin 4 in the H-Tx rat: Possible compensatory role in spontaneously arrested hydrocephalus. J. Neurosurg. 2006, 105, 459–464. [Google Scholar] [CrossRef]
  43. Owasil, R.; O’Neill, R.; Keable, A.; Nimmo, J.; MacGregor Sharp, M.; Kelly, L.; Saito, S.; Simpson, J.E.; Weller, R.O.; Smith, C.; et al. The Pattern of AQP4 Expression in the Ageing Human Brain and in Cerebral Amyloid Angiopathy. Int. J. Mol. Sci. 2020, 21, 1225. [Google Scholar] [CrossRef] [Green Version]
  44. Cheng, K.P.; Brodnick, S.K.; Blanz, S.L.; Zeng, W.; Kegel, J.; Pisaniello, J.A.; Ness, J.P.; Ross, E.; Nicolai, E.N.; Settell, M.L.; et al. Clinically-derived vagus nerve stimulation enhances cerebrospinal fluid penetrance. Brain Stimul. 2020, 13, 1024–1030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Benveniste, H.; Lee, H.; Ding, F.; Sun, Q.; Al-Bizri, E.; Makaryus, R.; Probst, S.; Nedergaard, M.; Stein, E.A.; Lu, H. Anesthesia with Dexmedetomidine and Low-dose Isoflurane Increases Solute Transport via the Glymphatic Pathway in Rat Brain When Compared with High-dose Isoflurane. Anesthesiology 2017, 127, 976–988. [Google Scholar] [CrossRef]
  46. Hsu, S.J.; Zhang, C.; Jeong, J.; Lee, S.I.; McConnell, M.; Utsumi, T.; Iwakiri, Y. Enhanced Meningeal Lymphatic Drainage Ameliorates Neuroinflammation and Hepatic Encephalopathy in Cirrhotic Rats. Gastroenterology 2021, 160, 1315–1329.e13. [Google Scholar] [CrossRef] [PubMed]
  47. Claeys, W.; Van Hoecke, L.; Lefere, S.; Geerts, A.; Verhelst, X.; Van Vlierberghe, H.; Degroote, H.; Devisscher, L.; Vandenbroucke, R.E.; Van Steenkiste, C. The neurogliovascular unit in hepatic encephalopathy. JHEP Rep. 2021, 3, 100352. [Google Scholar] [CrossRef]
  48. Singh, J.; Sharma, B.C.; Puri, V.; Sachdeva, S.; Srivastava, S. Sleep disturbances in patients of liver cirrhosis with minimal hepatic encephalopathy before and after lactulose therapy. Metab. Brain Dis. 2017, 32, 595–605. [Google Scholar] [CrossRef]
  49. Agrawal, S.; Umapathy, S.; Dhiman, R.K. Minimal Hepatic Encephalopathy Impairs Quality of Life. J. Clin. Exp. Hepatol. 2015, 5, S42–S48. [Google Scholar] [CrossRef] [Green Version]
  50. Velissaris, D.; Solomou, E.; Kalogeropoulos, A.; Georgiopoulou, V.; Thomopoulos, C.; Karatza, C. Sleep disorders and brain MRI as early indicators of subclinical hepatic encephalopathy. Hepatogastroenterology 2006, 53, 51–54. [Google Scholar]
  51. Labenz, C.; Baron, J.S.; Toenges, G.; Schattenberg, J.M.; Nagel, M.; Sprinzl, M.F.; Nguyen-Tat, M.; Zimmermann, T.; Huber, Y.; Marquardt, J.U.; et al. Prospective evaluation of the impact of covert hepatic encephalopathy on quality of life and sleep in cirrhotic patients. Aliment. Pharmacol. Ther. 2018, 48, 313–321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Bruyneel, M.; Sersté, T.; Libert, W.; van den Broecke, S.; Ameye, L.; Dachy, B.; Mulkay, J.P.; Moreno, C.; Gustot, T. Improvement of sleep architecture parameters in cirrhotic patients with recurrent hepatic encephalopathy with the use of rifaximin. Eur. J. Gastroenterol. Hepatol. 2017, 29, 302–308. [Google Scholar] [CrossRef] [PubMed]
  53. Spahr, L.; Coeytaux, A.; Giostra, E.; Hadengue, A.; Annoni, J.M. Histamine H1 blocker hydroxyzine improves sleep in patients with cirrhosis and minimal hepatic encephalopathy: A randomized controlled pilot trial. Am. J. Gastroenterol. 2007, 102, 744–753. [Google Scholar] [CrossRef]
  54. Llansola, M.; Cantero, J.L.; Hita-Yañez, E.; Mirones-Maldonado, M.J.; Piedrafita, B.; Ahabrach, H.; Errami, M.; Agusti, A.; Felipo, V. Progressive reduction of sleep time and quality in rats with hepatic encephalopathy caused by portacaval shunts. Neuroscience 2012, 201, 199–208. [Google Scholar] [CrossRef] [PubMed]
  55. Beaubernard, C.; Salomon, F.; Bismuth, H. Experimental hepatic encephalopathy. Study of the organization of a diurnal sleep pattern in rats with portocaval anastomosis. Biomedicine 1980, 32, 76–80. [Google Scholar] [PubMed]
  56. Larsen, F.S. Cerebral blood flow in hyperammonemia: Heterogeneity and starling forces in capillaries. Metab. Brain Dis. 2002, 17, 229–235. [Google Scholar] [CrossRef] [PubMed]
  57. Larsen, F.S.; Wendon, J. Brain edema in liver failure: Basic physiologic principles and management. Liver Transplant. 2002, 8, 983–989. [Google Scholar] [CrossRef]
  58. Iliff, J.J.; Wang, M.; Zeppenfeld, D.M.; Venkataraman, A.; Plog, B.A.; Liao, Y.; Deane, R.; Nedergaard, M. Cerebral arterial pulsation drives paravascular CSF-interstitial fluid exchange in the murine brain. J. Neurosci. 2013, 33, 18190–18199. [Google Scholar] [CrossRef] [Green Version]
  59. Liu, H.; Gaskari, S.A.; Lee, S.S. Cardiac and vascular changes in cirrhosis: Pathogenic mechanisms. World J. Gastroenterol. 2006, 12, 837–842. [Google Scholar] [CrossRef] [Green Version]
  60. Pudil, R.; Pelouch, R.; Praus, R.; Vašatová, M.; Hůlek, P. Heart failure in patients with liver cirrhosis. Cor Et Vasa 2013, 55, e391–e396. [Google Scholar] [CrossRef] [Green Version]
  61. Berzigotti, A.; Bosch, J. Editorial: Increased cardiac output in cirrhosis—Non-invasive assessment of regional blood flow by magnetic resonance angiography. Aliment. Pharmacol. Ther. 2016, 43, 1340–1342. [Google Scholar] [CrossRef]
  62. Li, X.; Yu, S.; Li, L.; Han, D.; Dai, S.; Gao, Y. Cirrhosis-related changes in left ventricular function and correlation with the model for end-stage liver disease score. Int. J. Clin. Exp. Med. 2014, 7, 5751–5757. [Google Scholar]
  63. Sampaio, F.; Pimenta, J.; Bettencourt, N.; Fontes-Carvalho, R.; Silva, A.P.; Valente, J.; Bettencourt, P.; Fraga, J.; Gama, V. Systolic and diastolic dysfunction in cirrhosis: A tissue-Doppler and speckle tracking echocardiography study. Liver Int. 2013, 33, 1158–1165. [Google Scholar] [CrossRef] [PubMed]
  64. Cazzaniga, M.; Salerno, F.; Pagnozzi, G.; Dionigi, E.; Visentin, S.; Cirello, I.; Meregaglia, D.; Nicolini, A. Diastolic dysfunction is associated with poor survival in patients with cirrhosis with transjugular intrahepatic portosystemic shunt. Gut 2007, 56, 869–875. [Google Scholar] [CrossRef] [Green Version]
  65. Srinivasamurthy, B.C.; Saravanan, S.P.; Marak, F.K.; Manivel, P.; Bhat, R.V.; Mathiyazhagan, D. Morphological Cardiac Alterations in Liver Cirrhosis: An Autopsy Study. Heart Views Off. J. Gulf Heart Assoc. 2021, 22, 96–101. [Google Scholar] [CrossRef]
  66. Gregolin, C.S.; do Nascimento, M.; Borges de Souza, S.L.; Ferreira Mota, G.A.; Bomfim, G.F.; de Azevedo Melo Luvizotto, R.; Sugizaki, M.M.; Zanati Bazan, S.G.; Salomé de Campos, D.H.; Dias, M.C.; et al. Myocardial Dysfunction in Cirrhotic Cardiomyopathy is Associated with Alterations of Phospholamban Phosphorylation and IL-6 Levels. Arch. Med. Res. 2021, 52, 284–293. [Google Scholar] [CrossRef]
  67. Yu, S.; Sun, L.; Wang, H.; Jiang, J.; Zhou, Q. Autonomic regulation of imbalance-induced myocardial fibrosis and its mechanism in rats with cirrhosis. Exp. Ther. Med. 2021, 22, 1040. [Google Scholar] [CrossRef] [PubMed]
  68. Yang, C.H.; Ting, W.J.; Day, C.H.; Ju, D.T.; Yeh, Y.L.; Chung, L.C.; Tsai, F.J.; Tsai, C.H.; Tsai, Y.; Huang, C.Y. SHSST cyclodextrin complex prevents the fibrosis effect on CCl₄-induced cirrhotic cardiomyopathy in rats through TGF-β pathway inhibition effects. Int. J. Mol. Sci. 2014, 15, 8037–8048. [Google Scholar] [CrossRef] [Green Version]
  69. Farahnaz, J.; Razieh, A.; Alireza, A.; Giti, G.; Ahmad Reza, D. Evaluation of Chronic Losartan Treatment Effect on Cardiac Chronotropic Dysfunction in Biliary Cirrhotic Rats. Acta Med. Iran. 2018, 56, 4–13. [Google Scholar]
  70. Wiest, R.; Groszmann, R.J. The paradox of nitric oxide in cirrhosis and portal hypertension: Too much, not enough. Hepatology 2002, 35, 478–491. [Google Scholar] [CrossRef]
  71. Zardi, E.M.; Abbate, A.; Zardi, D.M.; Dobrina, A.; Margiotta, D.; Tassel, B.W.V.; Afeltra, A.; Sanyal, A.J. Cirrhotic Cardiomyopathy. J. Am. Coll. Cardiol. 2010, 56, 539–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Liu, H.; Ma, Z.; Lee, S.S. Contribution of nitric oxide to the pathogenesis of cirrhotic cardiomyopathy in bile duct-ligated rats. Gastroenterology 2000, 118, 937–944. [Google Scholar] [CrossRef] [PubMed]
  73. Bátkai, S.; Járai, Z.; Wagner, J.A.; Goparaju, S.K.; Varga, K.; Liu, J.; Wang, L.; Mirshahi, F.; Khanolkar, A.D.; Makriyannis, A.; et al. Endocannabinoids acting at vascular CB1 receptors mediate the vasodilated state in advanced liver cirrhosis. Nat. Med. 2001, 7, 827–832. [Google Scholar] [CrossRef]
  74. Trewby, P.N.; Williams, R. Pathophysiology of hypotension in patients with fulminant hepatic failure. Gut 1977, 18, 1021–1026. [Google Scholar] [CrossRef] [Green Version]
  75. Tsai, C.-Y.; Su, C.-H.; Chan, J.Y.H.; Chan, S.H.H. Nitrosative Stress-Induced Disruption of Baroreflex Neural Circuits in a Rat Model of Hepatic Encephalopathy: A DTI Study. Sci. Rep. 2017, 7, 40111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Tomiyama, Y.; Johnny, E.; Brian, J.; Todd, M.M. Plasma viscosity and cerebral blood flow. Am. J. Physiol.-Heart Circ. Physiol. 2000, 279, H1949–H1954. [Google Scholar] [CrossRef]
  77. Odunayo, A. Chapter 80–Traumatic Brain Injury. In August’s Consultations in Feline Internal Medicine; Little, S.E., Ed.; W.B. Saunders: St. Louis, PA, USA, 2016; Volume 7, pp. 799–802. [Google Scholar] [CrossRef]
  78. Bjerring, P.N.; Bjerrum, E.J.; Larsen, F.S. Impaired cerebral microcirculation induced by ammonium chloride in rats is due to cortical adenosine release. J. Hepatol. 2018, 68, 1137–1143. [Google Scholar] [CrossRef]
  79. Dam, G.; Keiding, S.; Munk, O.L.; Ott, P.; Vilstrup, H.; Bak, L.K.; Waagepetersen, H.S.; Schousboe, A.; Sørensen, M. Hepatic encephalopathy is associated with decreased cerebral oxygen metabolism and blood flow, not increased ammonia uptake. Hepatology 2013, 57, 258–265. [Google Scholar] [CrossRef]
  80. Zheng, G.; Zhang, L.J.; Cao, Y.; Lu, G.M. Venous blood ammonia can be associated with cerebral blood flow in hepatic encephalopathy. Hepatology 2013, 58, 832–833. [Google Scholar] [CrossRef]
  81. Strauss, G.; Hansen, B.A.; Kirkegaard, P.; Rasmussen, A.; Hjortrup, A.; Larsen, F.S. Liver function, cerebral blood flow autoregulation, and hepatic encephalopathy in fulminant hepatic failure. Hepatology 1997, 25, 837–839. [Google Scholar] [CrossRef]
  82. Larsen, F.S.; Knudsen, G.M.; Paulson, O.B.; Vilstrup, H. Cerebral blood flow autoregulation is absent in rats with thioacetamide-induced hepatic failure. J. Hepatol. 1994, 21, 491–495. [Google Scholar] [CrossRef]
  83. Donovan, J.P.; Schafer, D.F.; Shaw, B.W., Jr.; Sorrell, M.F. Cerebral oedema and increased intracranial pressure in chronic liver disease. Lancet 1998, 351, 719–721. [Google Scholar] [CrossRef] [PubMed]
  84. Crippin, J.S.; Gross, J.B., Jr.; Lindor, K.D. Increased intracranial pressure and hepatic encephalopathy in chronic liver disease. Am. J. Gastroenterol. 1992, 87, 879–882. [Google Scholar] [PubMed]
  85. Detry, O.; De Roover, A.; Honoré, P.; Meurisse, M. Brain edema and intracranial hypertension in fulminant hepatic failure: Pathophysiology and management. WJG 2006, 12, 7405. [Google Scholar] [CrossRef]
  86. Bernal, W.; Murphy, N.; Brown, S.; Whitehouse, T.; Bjerring, P.N.; Hauerberg, J.; Frederiksen, H.J.; Auzinger, G.; Wendon, J.; Larsen, F.S. A multicentre randomized controlled trial of moderate hypothermia to prevent intracranial hypertension in acute liver failure. J. Hepatol. 2016, 65, 273–279. [Google Scholar] [CrossRef]
  87. Bjerring, P.N.; Eefsen, M.; Hansen, B.A.; Larsen, F.S. The brain in acute liver failure. A tortuous path from hyperammonemia to cerebral edema. Metab. Brain Dis. 2009, 24, 5–14. [Google Scholar] [CrossRef]
  88. Blei, A.T.; Larsen, F.S. Pathophysiology of cerebral edema in fulminant hepatic failure. J. Hepatol. 1999, 31, 771–776. [Google Scholar] [CrossRef] [PubMed]
  89. Jalan, R.; Damink, S.W.O.; Deutz, N.E.; Hayes, P.C.; Lee, A. Moderate hypothermia in patients with acute liver failure and uncontrolled intracranial hypertension. Gastroenterology 2004, 127, 1338–1346. [Google Scholar] [CrossRef]
  90. Tofteng, F.; Larsen, F.S. The Effect of Indomethacin on Intracranial Pressure, Cerebral Perfusion and Extracellular Lactate and Glutamate Concentrations in Patients with Fulminant Hepatic Failure. J. Cereb. Blood Flow Metab. 2004, 24, 798–804. [Google Scholar] [CrossRef] [Green Version]
  91. Tofteng, F.; Hauerberg, J.; Hansen, B.A.; Pedersen, C.B.; Jørgensen, L.; Larsen, F.S. Persistent Arterial Hyperammonemia Increases the Concentration of Glutamine and Alanine in the Brain and Correlates with Intracranial Pressure in Patients with Fulminant Hepatic Failure. J. Cereb. Blood Flow Metab. 2006, 26, 21–27. [Google Scholar] [CrossRef] [Green Version]
  92. Ranjan, P.; Mishra, A.M.; Kale, R.; Saraswat, V.A.; Gupta, R.K. Cytotoxic Edema Is Responsible for Raised Intracranial Pressure in Fulminant Hepatic Failure: In Vivo Demonstration Using Diffusion-Weighted MRI in Human Subjects. Metab. Brain Dis. 2005, 20, 181–192. [Google Scholar] [CrossRef]
  93. Bjerring, P.N.; Hauerberg, J.; Jørgensen, L.; Frederiksen, H.-J.; Tofteng, F.; Hansen, B.A.; Larsen, F.S. Brain hypoxanthine concentration correlates to lactate/pyruvate ratio but not intracranial pressure in patients with acute liver failure. J. Hepatol. 2010, 53, 1054–1058. [Google Scholar] [CrossRef]
  94. Davenport, A.; Will, E.J.; Davison, A.M. Early Changes in Intracranial Pressure During Haemofiltration Treatment in Patients with Grade 4 Hepatic Encephalopathy and Acute Oliguric Renal Failure. Nephrol. Dial. Transplant. 1990, 5, 192–198. [Google Scholar] [CrossRef] [PubMed]
  95. Schmidt, L.E.; Tofteng, F.; Strauss, G.I.; Larsen, F.S. Effect of treatment with the Molecular Adsorbents Recirculating System on arterial amino acid levels and cerebral amino acid metabolism in patients with hepatic encephalopathy. Scand. J. Gastroenterol. 2004, 39, 974–980. [Google Scholar] [CrossRef] [PubMed]
  96. Bjerring, P.N.; Dale, N.; Larsen, F.S. Acute hyperammonemia and systemic inflammation is associated with increased extracellular brain adenosine in rats: A biosensor study. Neurochem. Res. 2015, 40, 258–264. [Google Scholar] [CrossRef] [PubMed]
  97. Bjerring, P.N.; Hauerberg, J.; Frederiksen, H.J.; Nielsen, H.B.; Clemmesen, J.O.; Larsen, F.S. The effect of fractionated plasma separation and adsorption on cerebral amino acid metabolism and oxidative metabolism during acute liver failure. J. Hepatol. 2012, 57, 774–779. [Google Scholar] [CrossRef]
  98. Lenaerts, A.; Codden, T.; Meunier, J.-C.; Henry, J.-P.; Ligny, G. Effects of clonidine on diuretic response in ascitic patients with cirrhosis and activation of sympathetic nervous system. Hepatology 2006, 44, 844–849. [Google Scholar] [CrossRef]
  99. Miyajima, H.; Nomura, M.; Muguruma, N.; Okahisa, T.; Shibata, H.; Okamura, S.; Honda, H.; Shimizu, I.; Harada, M.; Saito, K.; et al. Relationship among gastric motility, autonomic activity, and portal hemodynamics in patients with liver cirrhosis. J. Gastroenterol. Hepatol. 2001, 16, 647–659. [Google Scholar] [CrossRef]
  100. Henriksen, J.H.; Ring-Larsen, H.; Christensen, N.J. Sympathetic nervous activity in cirrhosis: A survey of plasma catecholamine studies. J. Hepatol. 1985, 1, 55–65. [Google Scholar] [CrossRef]
  101. Barron, H.V.; Alam, I.; Lesh, M.D.; Strunk, A.; Bass, N.M. Autonomic nervous system tone measured by baroreflex sensitivity is depressed in patients with end-stage liver disease. Am. J. Gastroenterol. 1999, 94, 986–989. [Google Scholar] [CrossRef]
  102. Lara, C.; Gajardo, A.; Giubergia, F.; Bustos, N.; Roblero, J.P.; Urzúa, A.; Poniachik, J.; Cattaneo, M. P-38 Utility of pupillary reactivity in the functional assessment of the autonomous nervous system in patients with chronic liver disease: Preliminary results. Ann. Hepatol. 2021, 24, 100402. [Google Scholar] [CrossRef]
  103. Esler, M.; Dudley, F.; Jennings, G.; Debinski, H.; Lambert, G.; Jones, P.; Crotty, B.; Colman, J.; Willett, I. Increased Sympathetic Nervous Activity and the Effects of Its Inhibition with Clonidine in Alcoholic Cirrhosis. Ann. Intern. Med. 1992, 116, 446–456. [Google Scholar] [CrossRef] [PubMed]
  104. Lenz, K.; Hörtnagl, H.; Magometschnigg, D.; Kleinberger, G.; Druml, W.; Laggner, A. Function of the autonomic nervous system in patients with hepatic encephalopathy. Hepatology 1985, 5, 831–836. [Google Scholar] [CrossRef]
  105. Rega, D.; Aiko, M.; Peñaranda, N.; Urios, A.; Gallego, J.J.; Giménez-Garzó, C.; Casanova, F.; Fiorillo, A.; Cabrera-Pastor, A.; San-Miguel, T.; et al. Patients with Minimal Hepatic Encephalopathy Show Altered Thermal Sensitivity and Autonomic Function. J. Clin. Med. 2021, 10, 239. [Google Scholar] [CrossRef] [PubMed]
  106. Zhang, Y.; Tan, S.-L.; Du, J.; Chen, Y.; Jia, J.; Feng, J.-G.; Liu, K.-X.; Zhou, J. Dexmedetomidine alleviates neuroinflammation, restores sleep disorders and neurobehavioral abnormalities in rats with minimal hepatic encephalopathy. Int. Immunopharmacol. 2021, 96, 107795. [Google Scholar] [CrossRef]
  107. Kirstetter, P.; Moreau, R.; Soupison, T.; Cailmail, S.; Hartleb, M.; Lebrec, D. Role of sympathetic cardiovascular tone in control of arterial pressure in rats with cirrhosis. Liver 1996, 16, 263–266. [Google Scholar] [CrossRef]
  108. Dietrich, P.; Moleda, L.; Kees, F.; Müller, M.; Straub, R.H.; Hellerbrand, C.; Wiest, R. Dysbalance in sympathetic neurotransmitter release and action in cirrhotic rats: Impact of exogenous neuropeptide Y. J. Hepatol. 2013, 58, 254–261. [Google Scholar] [CrossRef] [Green Version]
  109. Jaeger, V.; DeMorrow, S.; McMillin, M. The direct contribution of astrocytes and microglia to the pathogenesis of hepatic encephalopathy. J. Clin. Transl. Hepatol. 2019, 7, 352. [Google Scholar] [CrossRef] [Green Version]
  110. Ott, P.; Larsen, F.S. Blood-brain barrier permeability to ammonia in liver failure: A critical reappraisal. Neurochem. Int. 2004, 44, 185–198. [Google Scholar] [CrossRef]
  111. Siracusa, R.; Fusco, R.; Cuzzocrea, S. Astrocytes: Role and Functions in Brain Pathologies. Front. Pharmacol. 2019, 10, 1114. [Google Scholar] [CrossRef] [Green Version]
  112. Broadhead, M.J.; Miles, G.B. Bi-Directional Communication Between Neurons and Astrocytes Modulates Spinal Motor Circuits. Front. Cell. Neurosci. 2020, 14, 30. [Google Scholar] [CrossRef]
  113. Hol, E.M.; Pekny, M. Glial fibrillary acidic protein (GFAP) and the astrocyte intermediate filament system in diseases of the central nervous system. Curr. Opin. Cell Biol. 2015, 32, 121–130. [Google Scholar] [CrossRef]
  114. Kretzschmar, H.A.; DeArmond, S.J.; Forno, L.S. Measurement of GFAP in Hepatic Encephalopathy by ELISA and Transblots. J. Neuropathol. Exp. Neurol. 1985, 44, 459–471. [Google Scholar] [CrossRef]
  115. Hiba, O.E.; Elgot, A.; Ahboucha, S.; Gamrani, H. Differential regional responsiveness of astroglia in mild hepatic encephalopathy: An Immunohistochemical approach in bile duct ligated rat. Acta Histochem. 2016, 118, 338–346. [Google Scholar] [CrossRef] [PubMed]
  116. Shuliatnikova, T. Region-dependent heterogeneity in GFAP expression in acute hepatic encephalopathy. Interdiscip.Res. Sci. Horiz. Perspect. 2021, 2, 56–57. [Google Scholar]
  117. Chileski, G.S.; García, E.N.; Lértora, J.W.; Mussart, N.; Hernández, D.R.; Cholich, L.A. Hepatic encephalopathy in swine experimentally poisoned with Senna occidentalis seeds: Effects on astrocytes. Toxicon 2021, 201, 86–91. [Google Scholar] [CrossRef]
  118. Bélanger, M.; Desjardins, P.; Chatauret, N.; Butterworth, R.F. Loss of expression of glial fibrillary acidic protein in acute hyperammonemia. Neurochem. Int. 2002, 41, 155–160. [Google Scholar] [CrossRef]
  119. Jayakumar, A.; Rao, K.R.; Murthy, C.R.; Norenberg, M. Glutamine in the mechanism of ammonia-induced astrocyte swelling. Neurochem. Int. 2006, 48, 623–628. [Google Scholar] [CrossRef]
  120. Strauss, G.I.; Knudsen, G.M.; Kondrup, J.; Møller, K.; Larsen, F.S. Cerebral metabolism of ammonia and amino acids in patients with fulminant hepatic failure. Gastroenterology 2001, 121, 1109–1119. [Google Scholar] [CrossRef]
  121. Albrecht, J.; Norenberg, M.D. Glutamine: A Trojan horse in ammonia neurotoxicity. Hepatology 2006, 44, 788–794. [Google Scholar] [CrossRef] [PubMed]
  122. Tanigami, H.; Rebel, A.; Martin, L.J.; Chen, T.Y.; Brusilow, S.W.; Traystman, R.J.; Koehler, R.C. Effect of glutamine synthetase inhibition on astrocyte swelling and altered astroglial protein expression during hyperammonemia in rats. Neuroscience 2005, 131, 437–449. [Google Scholar] [CrossRef] [Green Version]
  123. Zemtsova, I.; Görg, B.; Keitel, V.; Bidmon, H.J.; Schrör, K.; Häussinger, D. Microglia activation in hepatic encephalopathy in rats and humans. Hepatology 2011, 54, 204–215. [Google Scholar] [CrossRef] [PubMed]
  124. McMillin, M.; Frampton, G.; Thompson, M.; Galindo, C.; Standeford, H.; Whittington, E.; Alpini, G.; DeMorrow, S. Neuronal CCL2 is upregulated during hepatic encephalopathy and contributes to microglia activation and neurological decline. J. Neuroinflammation 2014, 11, 121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Rao, K.V.R.; Brahmbhatt, M.; Norenberg, M.D. Microglia contribute to ammonia-induced astrocyte swelling in culture. Metab. Brain Dis. 2013, 28, 139–143. [Google Scholar] [CrossRef] [PubMed]
  126. Weiss, N.; Miller, F.; Cazaubon, S.; Couraud, P.O. The blood-brain barrier in brain homeostasis and neurological diseases. Biochim. Et Biophys. Acta 2009, 1788, 842–857. [Google Scholar] [CrossRef] [Green Version]
  127. Abbott, N.J.; Patabendige, A.A.; Dolman, D.E.; Yusof, S.R.; Begley, D.J. Structure and function of the blood–brain barrier. Neurobiol. Dis. 2010, 37, 13–25. [Google Scholar] [CrossRef]
  128. Hajal, C.; Le Roi, B.; Kamm, R.D.; Maoz, B.M. Biology and Models of the Blood–brain Barrier. Annu. Rev. Biomed. Eng. 2021, 23, 359–384. [Google Scholar] [CrossRef] [PubMed]
  129. Huang, X.; Hussain, B.; Chang, J. Peripheral inflammation and blood–brain barrier disruption: Effects and mechanisms. CNS Neurosci. Ther. 2021, 27, 36–47. [Google Scholar] [CrossRef]
  130. Galea, I. The blood–brain barrier in systemic infection and inflammation. Cell. Mol. Immunol. 2021, 18, 2489–2501. [Google Scholar] [CrossRef]
  131. Profaci, C.P.; Munji, R.N.; Pulido, R.S.; Daneman, R. The blood–brain barrier in health and disease: Important unanswered questions. J. Exp. Med. 2020, 217, e20190062. [Google Scholar] [CrossRef] [Green Version]
  132. Song, K.; Li, Y.; Zhang, H.; An, N.; Wei, Y.; Wang, L.; Tian, C.; Yuan, M.; Sun, Y.; Xing, Y. Oxidative stress-mediated blood-brain barrier (BBB) disruption in neurological diseases. Oxidative Med. Cell. Longev. 2020, 2020, 4356386. [Google Scholar] [CrossRef]
  133. Marchetti, L.; Engelhardt, B. Immune cell trafficking across the blood-brain barrier in the absence and presence of neuroinflammation. Vasc. Biol. 2020, 2, H1–H18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Jayakumar, A.R.; Rama Rao, K.V.; Norenberg, M.D. Neuroinflammation in hepatic encephalopathy: Mechanistic aspects. J. Clin. Exp. Hepatol. 2015, 5, S21–S28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Chen, Z.; Ruan, J.; Li, D.; Wang, M.; Han, Z.; Qiu, W.; Wu, G. The Role of Intestinal Bacteria and Gut–Brain Axis in Hepatic Encephalopathy. Front. Cell. Infect. Microbiol. 2021, 10, 595759. [Google Scholar] [CrossRef] [PubMed]
  136. Schaefer, A.; Journaux, M.; Mourabit, H.E.; Mouri, S.; Wendum, D.; Lasnier, E.; Couraud, P.-O.; Housset, C.; Thabut, D.; Rudler, M.; et al. A systemic mechanism of increased transendothelial migration of leukocytes through the blood-brain barrier in hepatic encephalopathy. Clin. Res. Hepatol. Gastroenterol. 2022, 46, 101801. [Google Scholar] [CrossRef]
  137. Masago, K.; Kihara, Y.; Yanagida, K.; Hamano, F.; Nakagawa, S.; Niwa, M.; Shimizu, T. Lysophosphatidic acid receptor, LPA6, regulates endothelial blood-brain barrier function: Implication for hepatic encephalopathy. Biochem. Biophys. Res. Commun. 2018, 501, 1048–1054. [Google Scholar] [CrossRef]
  138. McMillin, M.A.; Frampton, G.A.; Seiwell, A.P.; Patel, N.S.; Jacobs, A.N.; DeMorrow, S. TGFβ1 exacerbates blood–brain barrier permeability in a mouse model of hepatic encephalopathy via upregulation of MMP9 and downregulation of claudin-5. Lab. Investig. 2015, 95, 903–913. [Google Scholar] [CrossRef] [Green Version]
  139. Kato, M.; Sugihara, J.; Nakamura, T.; Muto, Y. Electron microscopic study of the blood-brain barrier in rats with brain edema and encephalopathy due to acute hepatic failure. Gastroenterol. Jpn. 1989, 24, 135–142. [Google Scholar] [CrossRef]
  140. Wiltfang, J.; Nolte, W.; Otto, M.; Wildberg, J.; Bahn, E.; Figulla, H.R.; Pralle, L.; Hartmann, H.; Rüther, E.; Ramadori, G. Elevated serum levels of astroglial S100beta in patients with liver cirrhosis indicate early and subclinical portal-systemic encephalopathy. Metab. Brain Dis. 1999, 14, 239–251. [Google Scholar] [CrossRef] [PubMed]
  141. Saleh, A.; Kamel, L.; Ghali, A.; Ismail, A.; El Khayat, H. Serum levels of astroglial S100-beta and neuron-specific enolase in hepatic encephalopathy patients. EMHJ-East. Mediterr. Health J. 2007, 13, 1114–1123. [Google Scholar] [CrossRef]
  142. Wright, G.; Davies, N.A.; Shawcross, D.L.; Hodges, S.J.; Zwingmann, C.; Brooks, H.F.; Mani, A.R.; Harry, D.; Stadlbauer, V.; Zou, Z.; et al. Endotoxemia produces coma and brain swelling in bile duct ligated rats. Hepatology 2007, 45, 1517–1526. [Google Scholar] [CrossRef] [PubMed]
  143. Cauli, O.; Llansola, M.; Agustí, A.; Rodrigo, R.; Hernández-Rabaza, V.; Rodrigues, T.B.; López-Larrubia, P.; Cerdán, S.; Felipo, V. Cerebral oedema is not responsible for motor or cognitive deficits in rats with hepatic encephalopathy. Liver Int. 2014, 34, 379–387. [Google Scholar] [CrossRef]
  144. Kale, R.A.; Gupta, R.K.; Saraswat, V.A.; Hasan, K.M.; Trivedi, R.; Mishra, A.M.; Ranjan, P.; Pandey, C.M.; Narayana, P.A. Demonstration of interstitial cerebral edema with diffusion tensor MR imaging in type C hepatic encephalopathy. Hepatology 2006, 43, 698–706. [Google Scholar] [CrossRef] [PubMed]
  145. Córdoba, J.; Sanpedro, F.; Alonso, J.; Rovira, A. 1H Magnetic Resonance in the Study of Hepatic Encephalopathy in Humans. Metab. Brain Dis. 2002, 17, 415–429. [Google Scholar] [CrossRef]
  146. Rai, R.; Ahuja, C.K.; Agrawal, S.; Kalra, N.; Duseja, A.; Khandelwal, N.; Chawla, Y.; Dhiman, R.K. Reversal of low-grade cerebral edema after lactulose/rifaximin therapy in patients with cirrhosis and minimal hepatic encephalopathy. Clin. Transl. Gastroenterol. 2015, 6, e111. [Google Scholar] [CrossRef] [PubMed]
  147. Grant, S.; McMillin, M.; Frampton, G.; Petrescu, A.D.; Williams, E.; Jaeger, V.; Kain, J.; DeMorrow, S. Direct Comparison of the Thioacetamide and Azoxymethane Models of Type A Hepatic Encephalopathy in Mice. Gene Expr. 2018, 18, 171–185. [Google Scholar] [CrossRef]
  148. Mardini, H.; Smith, F.E.; Record, C.O.; Blamire, A.M. Magnetic resonance quantification of water and metabolites in the brain of cirrhotics following induced hyperammonaemia. J. Hepatol. 2011, 54, 1154–1160. [Google Scholar] [CrossRef]
  149. Häussinger, D.; Kircheis, G.; Fischer, R.; Schliess, F.; vom Dahl, S. Hepatic encephalopathy in chronic liver disease: A clinical manifestation of astrocyte swelling and low-grade cerebral edema? J. Hepatol. 2000, 32, 1035–1038. [Google Scholar] [CrossRef]
  150. Martinez, A. Electron microscopy in human hepatic encephalopathy. Acta Neuropathol. 1968, 11, 82–86. [Google Scholar] [CrossRef]
  151. Jiang, W.; Desjardins, P.; Butterworth, R.F. Hypothermia attenuates oxidative/nitrosative stress, encephalopathy and brain edema in acute (ischemic) liver failure. Neurochem. Int. 2009, 55, 124–128. [Google Scholar] [CrossRef]
  152. Jayakumar, A.R.; Tong, X.Y.; Curtis, K.M.; Ruiz-Cordero, R.; Abreu, M.T.; Norenberg, M.D. Increased toll-like receptor 4 in cerebral endothelial cells contributes to the astrocyte swelling and brain edema in acute hepatic encephalopathy. J Neurochem. 2014, 128, 890–903. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Kato, M.; Hughes, R.D.; Keays, R.T.; Williams, R. Electron microscopic study of brain capillaries in cerebral edema from fulminant hepatic failure. Hepatology 1992, 15, 1060–1066. [Google Scholar] [CrossRef] [PubMed]
  154. Sepehrinezhad, A.; Shahbazi, A.; Sahab Negah, S.; Joghataei, M.T.; Larsen, F.S. Drug-induced-acute liver failure: A critical appraisal of the thioacetamide model for the study of hepatic encephalopathy. Toxicol. Rep. 2021, 8, 962–970. [Google Scholar] [CrossRef] [PubMed]
  155. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.-S.; Peterson, T.C.; et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Clark, D.P.Q.; Perreau, V.M.; Shultz, S.R.; Brady, R.D.; Lei, E.; Dixit, S.; Taylor, J.M.; Beart, P.M.; Boon, W.C. Inflammation in Traumatic Brain Injury: Roles for Toxic A1 Astrocytes and Microglial–Astrocytic Crosstalk. Neurochem. Res. 2019, 44, 1410–1424. [Google Scholar] [CrossRef]
  157. Zha, Z.; Liu, Y.-J.; Liu, S.-S.; Zhang, N.; Li, J.-L.; Qi, F.; Jin, L.-Y.; Xue, B.; Yang, T.; Fan, Y.-P.; et al. Bu Shen Yi Sui capsule promotes myelin repair by modulating the transformation of A1/A2 reactive astrocytes In Vivo and In Vitro. Oxidative Med. Cell. Longev. 2022, 2022, 3800004. [Google Scholar] [CrossRef]
  158. Zhang, Q.; Liu, C.; Shi, R.; Zhou, S.; Shan, H.; Deng, L.; Chen, T.; Guo, Y.; Zhang, Z.; Yang, G.Y.; et al. Blocking C3d(+)/GFAP(+) A1 Astrocyte Conversion with Semaglutide Attenuates Blood-Brain Barrier Disruption in Mice after Ischemic Stroke. Aging Dis. 2022, 13, 943–959. [Google Scholar] [CrossRef]
  159. Song, N.; Zhu, H.; Xu, R.; Liu, J.; Fang, Y.; Zhang, J.; Ding, J.; Hu, G.; Lu, M. Induced Expression of kir6.2 in A1 Astrocytes Propagates Inflammatory Neurodegeneration via Drp1-dependent Mitochondrial Fission. Front. Pharmacol. 2021, 11, 618992. [Google Scholar] [CrossRef]
  160. Manley, G.T.; Binder, D.K.; Papadopoulos, M.C.; Verkman, A.S. New insights into water transport and edema in the central nervous system from phenotype analysis of aquaporin-4 null mice. Neuroscience 2004, 129, 983–991. [Google Scholar] [CrossRef]
  161. Yi, M.-H.; Lee, Y.S.; Kang, J.W.; Kim, S.J.; Oh, S.-H.; Kim, Y.M.; Lee, Y.H.; Lee, S.D.; Kim, D.W. NFAT5-Dependent Expression of AQP4 in Astrocytes. Cell. Mol. Neurobiol. 2013, 33, 223–232. [Google Scholar] [CrossRef]
  162. Stokum, J.A.; Mehta, R.I.; Ivanova, S.; Yu, E.; Gerzanich, V.; Simard, J.M. Heterogeneity of aquaporin-4 localization and expression after focal cerebral ischemia underlies differences in white versus grey matter swelling. Acta Neuropathol. Commun. 2015, 3, 61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Zhao, Z.-A.; Li, P.; Ye, S.-Y.; Ning, Y.-L.; Wang, H.; Peng, Y.; Yang, N.; Zhao, Y.; Zhang, Z.-H.; Chen, J.-F.; et al. Perivascular AQP4 dysregulation in the hippocampal CA1 area after traumatic brain injury is alleviated by adenosine A2A receptor inactivation. Sci. Rep. 2017, 7, 2254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Binder, D.K. Astrocytes: Stars of the Sacred Disease. Epilepsy Curr. 2018, 18, 172–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Potokar, M.; Stenovec, M.; Jorgačevski, J.; Holen, T.; Kreft, M.; Ottersen, O.P.; Zorec, R. Regulation of AQP4 surface expression via vesicle mobility in astrocytes. Glia 2013, 61, 917–928. [Google Scholar] [CrossRef]
  166. Xu, Z.; Xiao, N.; Chen, Y.; Huang, H.; Marshall, C.; Gao, J.; Cai, Z.; Wu, T.; Hu, G.; Xiao, M. Deletion of aquaporin-4 in APP/PS1 mice exacerbates brain Aβ accumulation and memory deficits. Mol. Neurodegener. 2015, 10, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Simon, M.; Wang, M.X.; Ismail, O.; Braun, M.; Schindler, A.G.; Reemmer, J.; Wang, Z.; Haveliwala, M.A.; O’Boyle, R.P.; Han, W.Y.; et al. Loss of perivascular aquaporin-4 localization impairs glymphatic exchange and promotes amyloid β plaque formation in mice. Alzheimer’s Res. Ther. 2022, 14, 59. [Google Scholar] [CrossRef] [PubMed]
  168. Liu, E.; Sun, L.; Zhang, Y.; Wang, A.; Yan, J. Aquaporin4 Knockout Aggravates Early Brain Injury Following Subarachnoid Hemorrhage Through Impairment of the Glymphatic System in Rat Brain. Acta Neurochir. Suppl. 2020, 127, 59–64. [Google Scholar] [CrossRef] [PubMed]
  169. Wang, T.; Chen, Y.; Zou, Y.; Pang, Y.; He, X.; Chen, Y.; Liu, Y.; Feng, W.; Zhang, Y.; Li, Q.; et al. Locomotor Hyperactivity in the Early-Stage Alzheimer’s Disease-like Pathology of APP/PS1 Mice: Associated with Impaired Polarization of Astrocyte Aquaporin 4. Aging Dis. 2022, 13, 1504–1522. [Google Scholar] [CrossRef]
  170. Kamagata, K.; Andica, C.; Takabayashi, K.; Saito, Y.; Taoka, T.; Nozaki, H.; Kikuta, J.; Fujita, S.; Hagiwara, A.; Kamiya, K. Association of MRI indices of glymphatic system with amyloid deposition and cognition in mild cognitive impairment and Alzheimer disease. Neurology 2022, 99, e2648–e2660. [Google Scholar] [CrossRef]
  171. Taoka, T.; Masutani, Y.; Kawai, H.; Nakane, T.; Matsuoka, K.; Yasuno, F.; Kishimoto, T.; Naganawa, S. 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. 2017, 35, 172–178. [Google Scholar] [CrossRef]
  172. Zeppenfeld, D.M.; Simon, M.; Haswell, J.D.; D’Abreo, D.; Murchison, C.; Quinn, J.F.; Grafe, M.R.; Woltjer, R.L.; Kaye, J.; Iliff, J.J. Association of Perivascular Localization of Aquaporin-4 With Cognition and Alzheimer Disease in Aging Brains. JAMA Neurol. 2017, 74, 91–99. [Google Scholar] [CrossRef] [PubMed]
  173. Ren, Z.; Iliff, J.J.; Yang, L.; Yang, J.; Chen, X.; Chen, M.J.; Giese, R.N.; Wang, B.; Shi, X.; Nedergaard, M. ‘Hit & Run’ model of closed-skull traumatic brain injury (TBI) reveals complex patterns of post-traumatic AQP4 dysregulation. J. Cereb. Blood Flow Metab. 2013, 33, 834–845. [Google Scholar] [CrossRef] [PubMed]
  174. Lu, H.; Zhan, Y.; Ai, L.; Chen, H.; Chen, J. AQP4-siRNA alleviates traumatic brain edema by altering post-traumatic AQP4 polarity reversal in TBI rats. J. Clin. Neurosci. 2020, 81, 113–119. [Google Scholar] [CrossRef] [PubMed]
  175. Christensen, J.; Wright, D.K.; Yamakawa, G.R.; Shultz, S.R.; Mychasiuk, R. Repetitive Mild Traumatic Brain Injury Alters Glymphatic Clearance Rates in Limbic Structures of Adolescent Female Rats. Sci. Rep. 2020, 10, 6254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Banitalebi, S.; Skauli, N.; Geiseler, S.; Ottersen, O.P.; Amiry-Moghaddam, M. Disassembly and Mislocalization of AQP4 in Incipient Scar Formation after Experimental Stroke. Int. J. Mol. Sci. 2022, 23, 1117. [Google Scholar] [CrossRef] [PubMed]
  177. Migliati, E.R.; Amiry-Moghaddam, M.; Froehner, S.C.; Adams, M.E.; Ottersen, O.P.; Bhardwaj, A. Na+-K+-2Cl cotransport inhibitor attenuates cerebral edema following experimental stroke via the perivascular pool of aquaporin-4. Neurocritical Care 2010, 13, 123–131. [Google Scholar] [CrossRef]
  178. Steiner, E.; Enzmann, G.U.; Lin, S.; Ghavampour, S.; Hannocks, M.J.; Zuber, B.; Rüegg, M.A.; Sorokin, L.; Engelhardt, B. Loss of astrocyte polarization upon transient focal brain ischemia as a possible mechanism to counteract early edema formation. Glia 2012, 60, 1646–1659. [Google Scholar] [CrossRef]
  179. Gaberel, T.; Gakuba, C.; Goulay, R.; Martinez De Lizarrondo, S.; Hanouz, J.L.; Emery, E.; Touze, E.; Vivien, D.; Gauberti, M. Impaired glymphatic perfusion after strokes revealed by contrast-enhanced MRI: A new target for fibrinolysis? Stroke 2014, 45, 3092–3096. [Google Scholar] [CrossRef] [Green Version]
  180. García-Lezana, T.; Oria, M.; Romero-Giménez, J.; Bové, J.; Vila, M.; Genescà, J.; Chavarria, L.; Cordoba, J. Cerebellar neurodegeneration in a new rat model of episodic hepatic encephalopathy. J. Cereb. Blood Flow Metab. 2017, 37, 927–937. [Google Scholar] [CrossRef] [Green Version]
  181. Gelpi, E.; Rahimi, J.; Klotz, S.; Schmid, S.; Ricken, G.; Forcen-Vega, S.; Budka, H.; Kovacs, G.G. The autophagic marker p62 highlights Alzheimer type II astrocytes in metabolic/hepatic encephalopathy. Neuropathology 2020, 40, 358–366. [Google Scholar] [CrossRef]
  182. Shulyatnikova, T.; Shavrin, V. Mobilisation and redistribution of multivesicular bodies to the endfeet of reactive astrocytes in acute endogenous toxic encephalopathies. Brain Res. 2021, 1751, 147174. [Google Scholar] [CrossRef] [PubMed]
  183. Raviz, E.K.; Noormand, F.; Kermani, A.S.; Galedari, A.; Esmaeilpour, K.; Maneshian, M.; Kalantaripour, T.P.; Dabiri, S.; Asadi-Shekaari, M. Promising Effects of Naringenin and Melatonin against Hepatic Encephalopathy Impairments Induced by Bile Duct Ligation in Male Rats. Cent. Nerv. Syst. Agents Med. Chem. 2022, 22, 31–38. [Google Scholar] [CrossRef] [PubMed]
  184. Shuliatnikova, T.; Tumanskyi, V. Expression of the water channel protein aquaporin-4 in the brain during human liver cirrhosis. Grail Sci. 2022, 22, 287–288. [Google Scholar] [CrossRef]
  185. Eslimi Esfahani, D.; Oryan, S.; Nabiuni, M.; Hosseinynia, T.S. The effects of bile duct ligation on motor cortex region morphology and aquaporin 4 protein concentration in male Wistar rats. Nova Biol. Reper. 2019, 6, 1–9. [Google Scholar] [CrossRef] [Green Version]
  186. Bodega, G.; Suárez, I.; López-Fernández, L.A.; García, M.I.; Köber, M.; Penedo, M.; Luna, M.; Juárez, S.; Ciordia, S.; Oria, M.; et al. Ammonia induces aquaporin-4 rearrangement in the plasma membrane of cultured astrocytes. Neurochem. Int. 2012, 61, 1314–1324. [Google Scholar] [CrossRef] [Green Version]
  187. Lichter-Konecki, U.; Mangin, J.M.; Gordish-Dressman, H.; Hoffman, E.P.; Gallo, V. Gene expression profiling of astrocytes from hyperammonemic mice reveals altered pathways for water and potassium homeostasis in vivo. Glia 2008, 56, 365–377. [Google Scholar] [CrossRef] [Green Version]
  188. De Rui, M.; Schiff, S.; Aprile, D.; Angeli, P.; Bombonato, G.; Bolognesi, M.; Sacerdoti, D.; Gatta, A.; Merkel, C.; Amodio, P.; et al. Excessive daytime sleepiness and hepatic encephalopathy: It is worth asking. Metab. Brain Dis. 2013, 28, 245–248. [Google Scholar] [CrossRef]
  189. Velissaris, D.; Karamouzos, V.; Polychronopoulos, P.; Karanikolas, M. Chronotypology and melatonin alterations in minimal hepatic encephalopathy. J. Circadian Rhythm. 2009, 7, 6. [Google Scholar] [CrossRef] [Green Version]
  190. Lozeva, V.; Tuomisto, L.; Sola, D.; Plumed, C.; Hippeläinen, M.; Butterworth, R. Increased density of brain histamine H1 receptors in rats with portacaval anastomosis and in cirrhotic patients with chronic hepatic encephalopathy. Hepatology 2001, 33, 1370–1376. [Google Scholar] [CrossRef]
  191. Bajaj, J.S.; Saeian, K.; Schubert, C.M.; Franco, R.; Franco, J.; Heuman, D.M. Disruption of sleep architecture in minimal hepatic encephalopathy and ghrelin secretion. Aliment Pharm. 2011, 34, 103–105. [Google Scholar] [CrossRef]
  192. Vora, R.S.; Subramanian, R.M. Hypotension in Cirrhosis. Clin. Liver Dis. 2019, 13, 149–153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Kim, E.; Choi, D.; Lim, H.K.; Lim, J.H. Multiple infarcted regenerative nodules in liver cirrhosis after systemic hypotension due to septic shock: Radiologic findings. Abdom. Imaging 2004, 29, 208–210. [Google Scholar] [CrossRef] [PubMed]
  194. Sourianarayanane, A.; Barnes, D.S.; McCullough, A.J. Beneficial effect of midodrine in hypotensive cirrhotic patients with refractory ascites. Gastroenterol Hepatol 2011, 7, 132–134. [Google Scholar]
  195. Lozeva, V.; Valjakka, A.; Lecklin, A.; Olkkonen, H.; Hippeläinen, M.; Itkonen, M.; Plumed, C.; Tuomisto, L. Effects of the histamine H(1) receptor blocker, pyrilamine, on spontaneous locomotor activity of rats with long-term portacaval anastomosis. Hepatology 2000, 31, 336–344. [Google Scholar] [CrossRef] [PubMed]
  196. Ahabrach, H.; Piedrafita, B.; Ayad, A.; El Mlili, N.; Errami, M.; Felipo, V.; Llansola, M. Chronic hyperammonemia alters the circadian rhythms of corticosteroid hormone levels and of motor activity in rats. J. Neurosci. Res. 2010, 88, 1605–1614. [Google Scholar] [CrossRef] [PubMed]
  197. Bomzon, A.; Binh, O.; Blendis, L.M. Hypotension in experimental cirrhosis: Is loss of vascular responsiveness to norepinephrine the cause of hypotension in chronic bile-duct-ligated dogs? J. Hepatol. 1993, 17, 116–123. [Google Scholar] [CrossRef]
  198. Bomzon, A.; Rosenberg, M.; Gali, D.; Binah, O.; Mordechovitz, D.; Better, O.S.; Greig, P.D.; Blendis, L.M. Systemic hypotension and decreased pressor response in dogs with chronic bile duct ligation. Hepatology 1986, 6, 595–600. [Google Scholar] [CrossRef]
  199. Clària, J.; Jiménez, W.; Ros, J.; Asbert, M.; Castro, A.; Arroyo, V.; Rivera, F.; Rodés, J. Pathogenesis of arterial hypotension in cirrhotic rats with ascites: Role of endogenous nitric oxide. Hepatology 1992, 15, 343–349. [Google Scholar] [CrossRef]
  200. Caracuel, L.; Sastre, E.; Callejo, M.; Rodrigues-Díez, R.; García-Redondo, A.B.; Prieto, I.; Nieto, C.; Salaices, M.; Aller, M.Á.; Arias, J.; et al. Hepatic Encephalopathy-Associated Cerebral Vasculopathy in Acute-on-Chronic Liver Failure: Alterations on Endothelial Factor Release and Influence on Cerebrovascular Function. Front. Physiol. 2020, 11, 593371. [Google Scholar] [CrossRef] [PubMed]
  201. Trewby, P.N.; Hanid, M.A.; Mackenzie, R.L.; Mellon, P.J.; Williams, R. Effects of cerebral oedema and arterial hypotension on cerebral blood flow in an animal model of hepatic failure. Gut 1978, 19, 999–1005. [Google Scholar] [CrossRef]
  202. Estrela, H.F.G.; Damásio, E.S.; Fonseca, E.K.U.N.; Bergamaschi, C.T.; Campos, R.R. Differential Sympathetic Vasomotor Activation Induced by Liver Cirrhosis in Rats. PLoS ONE 2016, 11, e0152512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Rohde, J.; Pedersen, H.R.; Bjerring, P.N.; Larsen, F.S. Effects of Dexamethasone and Cox Inhibitors on Intracranial Pressure and Cerebral Perfusion in the Lipopolysaccharide Treated Rats with Hyperammonemia. PLoS ONE 2015, 10, e0117416. [Google Scholar] [CrossRef]
  204. Larsen, R.H.; Kjær, M.S.; Eefsen, M.; Larsen, F.S.; Bjerring, P.N. Ciclosporin does not attenuate intracranial hypertension in rats with acute hyperammonaemia. World J. Hepatol. 2013, 5, 513–520. [Google Scholar] [CrossRef] [PubMed]
  205. Master, S.; Gottstein, J.; Blei, A.T. Cerebral blood flow and the development of ammonia-induced brain edema in rats after portacaval anastomosis. Hepatology 1999, 30, 876–880. [Google Scholar] [CrossRef] [PubMed]
  206. Cauli, O.; López–Larrubia, P.; Rodrigo, R.; Agusti, A.; Boix, J.; Nieto–Charques, L.; Cerdán, S.; Felipo, V. Brain Region-Selective Mechanisms Contribute to the Progression of Cerebral Alterations in Acute Liver Failure in Rats. Gastroenterology 2011, 140, 638–645. [Google Scholar] [CrossRef] [PubMed]
  207. Lin, S.-Y.; Wang, Y.-Y.; Chuang, Y.-H.; Chen, C.-J. Skeletal muscle proteolysis is associated with sympathetic activation and TNF-α-ubiquitin-proteasome pathway in liver cirrhotic rats. J. Gastroenterol. Hepatol. 2016, 31, 890–896. [Google Scholar] [CrossRef]
  208. Jia, W.; Liu, J.; Hu, R.; Hu, A.; Tang, W.; Li, L.; Li, J. Xiaochaihutang improves the cortical astrocyte edema in thioacetamide-induced rat acute hepatic encephalopathy by activating NRF2 pathway. Front. Pharmacol. 2020, 11, 382. [Google Scholar] [CrossRef] [Green Version]
  209. Chen, J.-R.; Wang, B.-N.; Tseng, G.-F.; Wang, Y.-J.; Huang, Y.-S.; Wang, T.-J. Morphological changes of cortical pyramidal neurons in hepatic encephalopathy. BMC Neurosci. 2014, 15, 1–12. [Google Scholar] [CrossRef] [Green Version]
  210. Ismail, F.S.; Faustmann, T.J.; Corvace, F.; Tsvetanova, A.; Moinfar, Z.; Faustmann, P.M. Ammonia induced microglia activation was associated with limited effects on connexin 43 and aquaporin 4 expression in an astrocyte-microglia co-culture model. BMC Neurosci. 2021, 22, 21. [Google Scholar] [CrossRef]
  211. Rama Rao, K.V.; Chen, M.; Simard, J.M.; Norenberg, M.D. Increased aquaporin-4 expression in ammonia-treated cultured astrocytes. Neuroreport 2003, 14, 2379–2382. [Google Scholar] [CrossRef]
  212. Pan, C.-F. Ammonia induces upregulation of aquaporin-4 in neocortical astrocytes of rats through the p38 mitogen-activated protein kinase pathway. Chin. Med. J. 2010, 123, 1888–1892. [Google Scholar] [CrossRef] [PubMed]
  213. Stokum, J.A.; Kwon, M.S.; Woo, S.K.; Tsymbalyuk, O.; Vennekens, R.; Gerzanich, V.; Simard, J.M. SUR1-TRPM4 and AQP4 form a heteromultimeric complex that amplifies ion/water osmotic coupling and drives astrocyte swelling. Glia 2018, 66, 108–125. [Google Scholar] [CrossRef]
  214. Sinke, A.P.; Jayakumar, A.R.; Panickar, K.S.; Moriyama, M.; Reddy, P.V.B.; Norenberg, M.D. NFκB in the mechanism of ammonia-induced astrocyte swelling in culture. J. Neurochem. 2008, 106, 2302–2311. [Google Scholar] [CrossRef] [Green Version]
  215. Jayakumar, A.R.; Rama Rao, K.V.; Tong, X.Y.; Norenberg, M.D. Calcium in the mechanism of ammonia-induced astrocyte swelling. J. Neurochem. 2009, 109, 252–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Reinehr, R.; Görg, B.; Becker, S.; Qvartskhava, N.; Bidmon, H.J.; Selbach, O.; Haas, H.L.; Schliess, F.; Häussinger, D. Hypoosmotic swelling and ammonia increase oxidative stress by NADPH oxidase in cultured astrocytes and vital brain slices. Glia 2007, 55, 758–771. [Google Scholar] [CrossRef] [PubMed]
  217. Felipo, V.; Urios, A.; Montesinos, E.; Molina, I.; Garcia-Torres, M.L.; Civera, M.; Olmo, J.A.; Ortega, J.; Martinez-Valls, J.; Serra, M.A.; et al. Contribution of hyperammonemia and inflammatory factors to cognitive impairment in minimal hepatic encephalopathy. Metab. Brain Dis. 2012, 27, 51–58. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The components of the glymphatic pathway. In the region of Virchow-Robin spaces, CSF along with penetrating arterioles flow into the brain parenchyma. Arterial pulsation provides influx of CSF into the periarterial interstitial space. The system eliminates most neurotoxic agents and waste substances from ISF. AQP4 water channels that are localized in perivascular astrocyte end-feet mediate fluid movements throughout the system. AQP4: aquaporin 4; CSF: cerebrospinal fluid; ISF: interstitial fluid. Created with BioRender.com.
Figure 1. The components of the glymphatic pathway. In the region of Virchow-Robin spaces, CSF along with penetrating arterioles flow into the brain parenchyma. Arterial pulsation provides influx of CSF into the periarterial interstitial space. The system eliminates most neurotoxic agents and waste substances from ISF. AQP4 water channels that are localized in perivascular astrocyte end-feet mediate fluid movements throughout the system. AQP4: aquaporin 4; CSF: cerebrospinal fluid; ISF: interstitial fluid. Created with BioRender.com.
Cells 12 00979 g001
Figure 2. Injured astrocytes differentially express AQP4, impairing the glymphatic system. In several neurological disorders, AQP4 is primarily expressed on the cell body of swollen and reactive astrocytes (A1 phenotype). The AQP4 mislocalization disrupts the flow through the ISF and is responsible for impairment of the glymphatic system, resulting in accumulation of waste metabolites, microglial activation, neuroinflammation, and neuronal toxicity in the brain parenchyma. Similar astrogliosis and mislocalization of AQP4 have been observed in HE. These pathological changes in astrocytes, the main cellular component of the glymphatic pathway, may compromise clearance of ISF in HE. Created with BioRender.com.
Figure 2. Injured astrocytes differentially express AQP4, impairing the glymphatic system. In several neurological disorders, AQP4 is primarily expressed on the cell body of swollen and reactive astrocytes (A1 phenotype). The AQP4 mislocalization disrupts the flow through the ISF and is responsible for impairment of the glymphatic system, resulting in accumulation of waste metabolites, microglial activation, neuroinflammation, and neuronal toxicity in the brain parenchyma. Similar astrogliosis and mislocalization of AQP4 have been observed in HE. These pathological changes in astrocytes, the main cellular component of the glymphatic pathway, may compromise clearance of ISF in HE. Created with BioRender.com.
Cells 12 00979 g002
Figure 3. Schematic representations of some pathological processes which may affect normal function of the glymphatic system following HE. All these factors can impair the function of the glymphatic system throughout the cerebral tissue. This induces accumulation of waste substances in the brain interstitial space and triggers neuroinflammation, resulting in neuronal injury and progression of hepatic encephalopathy. CBF: cerebral blood flow; HE: hepatic encephalopathy. Created with BioRender.com.
Figure 3. Schematic representations of some pathological processes which may affect normal function of the glymphatic system following HE. All these factors can impair the function of the glymphatic system throughout the cerebral tissue. This induces accumulation of waste substances in the brain interstitial space and triggers neuroinflammation, resulting in neuronal injury and progression of hepatic encephalopathy. CBF: cerebral blood flow; HE: hepatic encephalopathy. Created with BioRender.com.
Cells 12 00979 g003
Table 1. Main effective factors on the normal function of glymphatic system following liver diseases and HE.
Table 1. Main effective factors on the normal function of glymphatic system following liver diseases and HE.
StudiesAffected FactorKey ResultsReferences
Clinical human casesSleep disordersSleep disturbances are defined as common features of liver diseases and indicators of subclinical HE in cirrhosis, evaluated by Pittsburgh Sleep Quality Index, Epworth Sleepiness Scale, and polysomnography.[48,50,53,188,189,190,191]
Arterial hypotensionPortosystemic shunt and accumulation of vasodilator mediators in systemic circulation, shock, decrease systemic vascular resistance, and blood pressure in HE and cirrhosis.[70,71,73,74,192,193,194]
Impaired cerebral blood flowDecrease in the cerebral perfusion pressure, intracranial hypertension, and impairment of CBF following liver failure and HE.[83,84,85,86,87,88,89,90,91,92,93,94]
Altered autonomic nervous systemGeneral imbalance in ANS tone, sympathetic tone hyperactivity, increase in plasma levels of norepinephrine and inhibition of vagal tone in cirrhosis, chronic liver disease and HE.[98,99,100,101,102,103,104,105]
Astrocyte dysfunctionDecrease in GFAP expression in the frontal cortex and basal ganglia in postmortem analysis of HE patients, solutes accumulation in astrocytes, brain edema, and astrocyte swelling in patients with fulminant hepatic failure.[114,144,150,153]
Dysregulation of AQP4Postmortem brain analysis of cirrhosis patients indicated an increase in the expression of AQP4 in cell body.[184]
In VivoSleep disordersParadoxical sleep, disruption of sleep patterns, suppression in the duration of rapid eye movement sleep, non-rapid eye movement sleep, and alternation of circadian rhythms in rat models of HE.[54,55,190,195,196]
CardiomyopathyCardiac hypertrophy, myocyte swelling, gross abnormalities in cardiomyocytes in histological examinations, increase plasma levels of myocardial enzymes, systolic and diastolic abnormalities, inotropic and chronotropic dysfunction, and QT interval prolongation in rat models of HE and cirrhosis.[66,67,68,69]
Arterial hypotensionDecrease in the systemic arterial blood pressure in animal models of HE and liver failure.[75,197,198,199,200,201,202]
Impaired cerebral blood flowRaising intracranial pressure, cerebral hyperemia, vasogenic edema, and impairment of CBF in hyperammonemic HE rats.[82,201,203,204,205,206]
Altered autonomic nervous systemSympathetic tone hyperactivity and increase in circulatory norepinephrine in rats model of cirrhosis and HE.[106,107,108,202,207]
Astrocyte dysfunctionDecrease in GFAP expression in substantia nigra, ventral tegmental area, hippocampus, sensorimotor cortex, thalamus, cerebral edema, astrogliosis, and astrocyte swelling in animal models of HE.[115,116,117,118,142,208,209]
Dysregulation of AQP4Decreased expression of AQP4 in olfactory bulb and prefrontal cortex accompanied with glymphatic impairment in BDL rats.[22]
Meningeal lymphatic drainageImprovement in the severity of HE in an experimental rat model following enhanced meningeal lymphatic drainage (requires further experiments)[46]
In VitroAstrocyte dysfunctionCell swelling and AQP4 rearrangement in ammonia-exposed astrocytes.[186,210,211,212,213,214,215,216]
Dysregulation of AQP4Decreased AQP4 expression in astrocytes isolated from hyperammonemic mice and AQP4 rearrangement in ammonia-exposed astrocytes.[187]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sepehrinezhad, A.; Stolze Larsen, F.; Ashayeri Ahmadabad, R.; Shahbazi, A.; Sahab Negah, S. The Glymphatic System May Play a Vital Role in the Pathogenesis of Hepatic Encephalopathy: A Narrative Review. Cells 2023, 12, 979. https://doi.org/10.3390/cells12070979

AMA Style

Sepehrinezhad A, Stolze Larsen F, Ashayeri Ahmadabad R, Shahbazi A, Sahab Negah S. The Glymphatic System May Play a Vital Role in the Pathogenesis of Hepatic Encephalopathy: A Narrative Review. Cells. 2023; 12(7):979. https://doi.org/10.3390/cells12070979

Chicago/Turabian Style

Sepehrinezhad, Ali, Fin Stolze Larsen, Rezan Ashayeri Ahmadabad, Ali Shahbazi, and Sajad Sahab Negah. 2023. "The Glymphatic System May Play a Vital Role in the Pathogenesis of Hepatic Encephalopathy: A Narrative Review" Cells 12, no. 7: 979. https://doi.org/10.3390/cells12070979

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop