Open access peer-reviewed chapter

The Pathophysiology of Hepatic Encephalopathy at the Level of Gut-Liver-Brain Axis: The Role of Resident Innate Immune Cells

Written By

Ali Sepehrinezhad and Ali Shahbazi

Submitted: 16 October 2023 Reviewed: 15 November 2023 Published: 15 January 2024

DOI: 10.5772/intechopen.1004125

From the Edited Volume

Liver Cirrhosis and Its Complications - Advances in Diagnosis and Management

Ran Wang and Xingshun Qi

Chapter metrics overview

98 Chapter Downloads

View Full Metrics

Abstract

Hepatic encephalopathy (HE) reflects a wide spectrum of frequent and complex neurological complications that are associated with advanced liver diseases. It significantly impacts the quality of life and daily activities of those affected. Despite many investigations, the precise pathophysiology of HE is still under discussion. One contributing factor believed to be responsible for HE is the accumulation of neurotoxic substances in the brain such as ammonia, mercaptans, short-chain fatty acids, and lipopolysaccharides, originating from the dysfunctional liver. Strong data, however, suggests that HE is a complex symptom, and inflammation interacts synergistically with ammonia to worsen gliopathy and neuronal destruction. Recent data suggests that HE might come from the intestines. Increased activity of gut innate immune cells, especially macrophages and dendritic cells, can initiate inflammatory signals from the gut to systemic circulation, liver tissue, and finally the central nervous system. In this chapter, all inflammatory mechanisms at the levels of the gut-liver-brain axis following cirrhosis and HE are presented in detail. The chapter highlights the role of intestinal innate immune cells, liver Kupffer cells, and brain microglia in cirrhosis and the progression of HE.

Keywords

  • cirrhosis
  • hepatic encephalopathy
  • gut-liver-brain axis
  • innate immune cells
  • intestinal macrophages
  • Kupffer cells
  • microglia

1. Introduction

Hepatic encephalopathy (HE) is a frequent neurological disorder and a serious consequence of advanced liver disease, acute liver failure, or portal-systemic shunting. HE is defined as a wide range of neuropsychiatric disturbances, including changes in personality, a shortened attention span, intellectual disability, cognitive impairment, alterations in sleep patterns, delirium, asterixis, episodic confusion, seizures, decreased level of consciousness, and even coma [1, 2, 3]. Three varieties of HE are distinguished based on the underlying cause. Type A refers to brain dysfunction that arises as a result of acute liver failure. Type B is observed in cases where there is portosystemic shunting without liver insufficiency, and type C as a consequence of liver cirrhosis [3, 4]. According to the West Haven Criteria (WHC), HE is categorized into grades I to IV, ranging from a lack of attention (grade I) to coma (grade IV). Based on the severity of manifestations, the International Society of Hepatic Encephalopathy and Nitrogen Metabolism (ISHEN) classifies HE into two categories: covert HE (CHE) and overt HE (OHE). CHE refers to minimal HE (MHE) and grade I HE, which are characterized by neuropsychological alterations without clinical evidence of mental changes. On the other hand, OHE displays severe neurological manifestations [3, 5]. The exact mechanism behind brain dysfunction in patients with liver diseases is complex and not yet fully revealed. However, it has been agreed that ammonia plays a key role. It mainly originates from the degradation of nitrogen-containing compounds by the intestinal microbiome, as well as the hydrolysis of glutamine by gut glutaminase, which is then transported into the portal circulation. A large portion of circulating ammonia is metabolized in the urea cycle by intact hepatocytes and subsequently excreted by the kidneys as urea [6]. In patients with liver insufficiency, the injured hepatocytes are no longer able to effectively detoxify ammonia from the blood. Subsequently, the level of circulatory ammonia is increased, which can result in a condition called hyperammonemia. Increased levels of ammonia in systemic circulation can cause detrimental effects on the central nervous system (CNS) through various pathways [6, 7]. Findings from numerous animal and clinical studies have indicated that inflammation, synergistically with ammonia, can potentially link to the development of HE [8, 9, 10, 11, 12]. Recently, there has been a growing focus among researchers on finding out the pathogenesis of brain dysfunction that occurs following liver diseases in organs beyond the liver and CNS [13]. Evidence indicates a correlation between alteration of gut microbiota composition (gut dysbiosis) and its associated by-products with conditions such as intestinal and liver inflammation, systemic inflammation, hyperammonemia, and neuroinflammation in liver diseases and HE [14, 15, 16, 17]. The gut, liver, and brain have some strong associations. The portal system allows a considerable part of the blood that passes through the gastrointestinal tract and many gut-active metabolites to reach the liver parenchyma. Similarly, the liver can influence intestine processes by synthesizing and secreting bile acids. Furthermore, both the gut and the liver can control various cerebral processes via their metabolites and immune systems. Moreover, through a cascade of humoral, neuroendocrine, immunological, neural, and metabolic signals, the brain can alter the activities of the liver and intestines [18, 19, 20]. The gut-liver-brain axis is a fascinating concept that involves a complex and multidirectional communication pathway linking the gastrointestinal tract, hepatic system, and CNS. In this axis, the brain influences the activity of gut and hepatic innate immune cells, while the liver and intestine regulate mental processes through gut microbiota and immune interactions [21, 22]. Recent reports indicate that impairment of the gut-liver-brain axis may play a role in the pathophysiology of liver diseases and HE [23, 24, 25]. It has been reported that the components of the innate immune system are considered the main communication route between the gut, liver, and brain. Innate immune cells found in the enteric, hepatic, and central nervous systems may mediate complicated connections between these organs. Researchers would benefit from an accurate understanding of the inflammatory signals emanating from residual innate immune cells and their impact on the gut-liver-brain axis in advanced liver disease and HE.

Advertisement

2. Intestinal macrophages and dendritic cells produce gut inflammatory signals

Gut macrophages (Mϕ), like other tissue Mϕ, originate from pluripotent bone marrow stem cells that develop into circulatory monocytes in the presence of interleukin (IL)-1, IL-3, IL-6, and granulocyte-macrophage colony-stimulating factor (GM-CSF). Circulatory monocytes eventually extravasate into the intestinal lamina proparia, submucosal, and myentric plexus, where they come into contact with mucosal epithelium, luminal contents, and the enteric nervous system (ENS) [26, 27]. C-C motif chemokine receptor 2 (CCR2) signaling is totally required for blood monocyte penetration into the gut [28]. In mice, newly recruited monocytes are identified by the expression of profibrotic Ly6C and C-X3-C motif chemokine receptor 1 (CX3CR1), whereas mature resident intestinal Mϕ is identified by the expression of CD14, CD11b, MHCII, CX3CR1, and CD64 markers [28]. Furthermore, mediators, such as colony-stimulating factor 1 (CSF-1), transforming growth factor beta (TGFβ), IL-10, and CX3CR1, aid in the differentiation of monocytes into mature Mϕ [28, 29, 30, 31]. Intestinal Mϕ phagocytize pathogens as they express high levels of phagocytic markers such as Cd36, Gas6, Mertk, Axl, Itgav, Mrc1, Itgb5, Cd81, and C1qa-c [32]. They also express the efferocytosis receptor αvβ5, which facilitates the scavenging of apoptotic epithelial cells, hence maintaining the integrity of the mucosal epithelial barrier [33, 34]. The intestinal barrier and mucosal integrity are safeguarded by mediators produced by gut Mϕ, such as hepatocyte growth factor (HGF), Wnt signaling elements, IL-10, and prostaglandin E2 (PGE2) and their balloon-like protrusions [35, 36, 37, 38]. Additionally, intestinal Mϕ receives components of the luminal side through their projection processes into the mucosal layer. They play an important role in the presentation of antigens to dendritic cells (DCs) in the lamina proparia and the activation of T cells [39]. Gut Mϕ also interacts with both neural cells in ENS and enteric smooth muscle cells through the bone morphogenetic protein 2 (BMP2) and transient receptor potential cation channel subfamily V Member 4 (TRPV4)-PGE2-E-prostanoid receptor 1 or 3 (EP1/3) pathways, respectively [40, 41]. Colony stimulating factor 1 (CSF-1) is also secreted by ENS neurons, and it controls macrophage activity by binding to its membrane-bound CSF-1 receptors [42]. These multidirectional pathways allow Mϕ to regulate intestinal motility.

The homeostasis of the gut environment is maintained by an integrated epithelial mucosal barrier and intact tight junctions. Patients with cirrhosis and HE have been shown to have gut dysbiosis and a change in the composition of their gut microbiome from normal flora to pathogenic species such as Streptococcus salivarius, Proteobacteria, and Enterobacteriaceae [17, 43, 44]. Gut microbiota alteration was also revealed in several experimental animal models of liver disease and HE (Table 1) [63, 64, 65, 66, 67]. In addition, these patients had small bowel bacterial overgrowth, as determined by Qin et al. in a quantitative metagenomics analysis, which found differences in 75,245 genes between healthy people and cirrhotic patients [68]. Cirrhosis-related gut dysbiosis alters intestinal barrier permeability and epithelial tight junctions through the induction of oxidative stress, immune cell infiltration, a decrease in short-chain fatty acids, and a loss in the capacity of epithelial stem cells to self-renew [59, 67, 69, 70]. Portal hypertension also impairs the intestinal barrier via perturbation of intestinal circulation and cellular hypoxia in cirrhosis [71, 72]. In cirrhosis and liver disease, decrease in expression of some intestinal antimicrobial molecules, such as α-defensin5, α-defensin6, RegIII, and β-defensin 1, may exacerbate dysbiosis and bacterial translocation [73, 74, 75, 76]. In both rat models of acute liver failure and cirrhosis, a decrease in the expression of several intestine antimicrobial peptides was shown [77, 78]. In cirrhosis, bacterial translocation and endotoxin products, such as flagellin, peptidoglycan, lipopolysaccharide (LPS), bacterial DNA, and pathogen-associated molecular pattern molecules (PAMPs), were able to enter the lamina proparia and portal circulation due to the compromised intestinal barrier [79, 80]. These endotoxins and false metabolites stimulate Mϕ and DCs in Peyer’s patches of the lamina proparia. Activated Mϕ synthetize and secrete pro-inflammatory cytokines such as IL-1, IL-18, TGF-β, IL-6, interferon gamma (IFNγ), IL-1β, nitric oxide (NO), and tumor necrosis factor alpha (TNFα) (Figure 1). These metabolites also increase the phagocytosis capacity of DCs. Increasing levels of intestinal pro-inflammatory cytokines injure the mucosal epithelial barrier and enhance neutrophil infiltration into the lamina proparia. An electron microscopy analysis of biopsy samples from the duodenum revealed that patients with cirrhosis had an impaired intestinal barrier due to activated gut Mϕ and production of IL-6 and NO [61]. Fecal analysis in patients with acute decompensating cirrhosis indicated an increase in intestinal pro-inflammatory cytokines that were correlated with intestinal barrier disruption [81]. Furthermore, an increase in serum levels of LPS was associated with a decrease in the expression of intestinal tight junction proteins (i.e., occludin and claudin-1) in patients with cirrhosis [82]. IL-6 and TNFα can initiate downstream inflammatory signals by activating the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kβ) [83, 84, 85, 86, 87]. The number of NO-releasing duodenal CD33+-CD14+-Trem-1+ Mϕ increased in cirrhosis. Additionally, these patients had higher levels of blood pro-inflammatory cytokines and circulatory LPS, as well as overexpressed claudin-2, a permeable tight protein, in the duodenum [61]. Injured epithelial cells and activated Mϕ release damage-associated molecular patterns (DAMPs). These DAMPs, along with PAMPs, have the ability to activate pattern recognition receptors (PRRs), particularly toll-like receptors (TLRs) that are found in intestinal Mϕ and DCs [88, 89]. Among the TLR family, TLR1, TLR2, TLR4, TLR5, and TLR6 are known to localize on the cell membrane and recognize various pathogen membrane elements. On the other hand, intracellular TLR3, TLR7, TLR8, and TLR9 are able to detect nucleic acids [90]. LPS derived from altered microbiota triggers an inflammatory signaling pathway through TLR4 in Mϕ (Figure 1). This pathway involves the activation of signaling proteins, including TRAM, TRIF, RIP1, TRF3, or TRAF6, which subsequently lead to the overexpression of MAPK and NF-kβ proteins [91, 92]. The majority of PAMPs can trigger PRRs, predominantly TLR2 and TLR4, on DCs. This mediates the migration of DCs to mesenteric lymph nodes, where they subsequently differentiate the naïve T cells into Th1, Th2, and inflammatory effector T cells. Activated DCs by PAMPs-TLR also present their antigens to CD4+ T cells through the major histocompatibility complex II and T cell receptor interaction. Additionally, these activated DCs secrete pro-inflammatory cytokines that further activate CD4+ T cells [84, 88, 93]. Increased Th1 cell numbers and an imbalanced Th17/Treg ratio have been shown to worsen intestinal inflammation in cirrhosis and liver disease [67, 94]. Th1 cells produced several pro-inflammatory cytokines, such as IFN-γ, IL-2, and TNFα, that worsened intestinal barrier disruption [95, 96]. Basal secreting levels of IL-17 by Th17 cells maintain the intestinal barrier by increasing the expression of tight junction proteins [97, 98]. However, following gut dysbiosis, activated Mϕ and DCs by PAMPs and DAMPs increase the number of Th17 cells by secreting IL-23, IL-6, and IL-1β. A number of powerful pro-inflammatory cytokines are produced by activated Th17 cells, which aggravate intestinal inflammation [99, 100, 101, 102]. Additionally, Th17-derived cytokines stimulate the production of neutrophil chemotaxis factors such as IL-8, C-X-C motif chemokine ligand 8 (CXCL8), GM-CSF, and C-X-C motif chemokine ligand 10 (CXCL10) by Mϕ, epithelial cells, and DCs [103]. Moreover, cytokines produced by Th17 cells prevent naive T cells from differentiating into Treg cells. Treg cells produce and secrete anti-inflammatory cytokines in response to activated DCs [104].

StudyFindings (Intestinal tissue)References
Wang et al. (2017)⇧Intestinal permeability (Evans blue), ⇧mucosal barrier damage, ⇧alpha1-antitrypsin, ⇧calprotectin (neutrophil infiltration), ⇩claudin-1, ⇩claudin-6, ⇩occludin, ⇧TLR4, ⇧TLR9 (CCL4 rats)[45]
Wang et al. (2020)⇩Occludin, ⇩ZO-1, ⇧dysbiosis (CCL4 mice)[46]
Zhou et al. 2014⇩Occludin, ⇧mucosal injury, ⇧D-lactate and diamine oxidase (serum markers of intestinal barrier disruption) (BDL rats)[47]
Wang et al. (2021)⇩Villus height and width, ⇩crypt depth, ⇩Occludin, ⇩ZO-1, ⇩claudin-1 (CCL4 mice)[48]
Luyer et al. (2004)⇧Bacterial translocation (BDL rats)[49]
Liu et al. (2020)⇧Dysbiosis, (CCL4 mice)⇧Bacteroidaceae, ⇧Porphyromonadaceae, ⇧Marinillabiliaceae, ⇧Chloroplast, ⇧Methylobacteriaceae, ⇧Enterobacteriaceae, ⇧Streptococcaceae, ⇧Lactobacillaceae (small intestine)[14]
⇧Marinillabiliaceae, ⇧Prevotellaceae, ⇧Bacteroidales Incertae Sedis, ⇧Flammeovirgaceae, ⇧Streptococcaceae, ⇧Enterobacteriaceae, ⇧Methylobacteriaceae (colon)
Palma et al. (2007)⇧Bacterial translocation (CCL4 rats)[50]
Lei et al. (2022)⇩Occludin, ⇩claudin-1, ⇩ZO-1, ⇧NF-κB, (BDL rats)[51]
Fu et al. (2022)⇩Occludin mRNA and protein, ⇩ZO-1 mRNA and protein, ⇩claudin-1 mRNA and protein, ⇧endotoxemia (⇧LPS and ⇧MIP-1 in serum), ⇧dysbiosis (CCL4 mice)[52]
Sánchez et al. (2014)⇧Bacterial translocation (CCL4 rats)[53]
Cabrera-Rubio et al. (2019)⇧Gut dysbiosis (⇧Akkermansia, ⇧Bacteroides, ⇧Ruminococcaceae, ⇧Prevotella, ⇩Faecalibacterium prausnitzii) (BDL mice)[54]
Zhao et al. (2021)⇩Claudin-1 mRNA, ⇧HMGB1 mRNA, ⇧TLR4 mRNA, ⇧MyD88 mRNA, ⇧NF-kβ mRNA (CCL4 rats)[55]
Safari et al. (2023)⇩Occludin, ⇩claudin-1, ⇩farnesoid X receptor (FXR), ⇧claudin-2 (BDL rats)[56]
Assimakopoulos et al. (2003)⇧Endotoxemia, ⇧lipid peroxidation, ⇧protein oxidation, ⇧oxidized glutathione, ⇩glutathione (BDL rats)[57]
Chen et al. (2014)⇧Gut dysbiosis (patients with cirrhosis)[58]
Assimakopoulos et al. (2015)⇧Endotoxemia, ⇩mitotic cell count, ⇧apoptosis, ⇧lipid peroxidation, (patients with cirrhosis)[59]
Chen et al. (2011)⇧Gut dysbiosis (⇧Proteobacteria, ⇧Fusobacteria, ⇩Bacteroidetes) (patients with cirrhosis)[60]
Du Plessis et al. (2013)⇧LPS (serum), ⇧CD33+-CD14+-Trem-1+Mϕ, ⇧iNOS, ⇧NO, ⇧Claudin-2, ⇧intestinal barrier (patients with cirrhosis)[61]
Pascual et al. (2003)⇧Lactulose/mannitol ratio (marker of intestinal permeability) (patients with cirrhosis)[62]

Table 1.

Main intestinal changes following cirrhosis and HE.

Figure 1.

Gut dysbiosis and intestinal inflammation in pathophysiology of cirrhosis and HE, highlighting the role of intestinal macrophages and dendritic cells. CXCL8, C-X-C motif chemokine ligand 8; CXCL10, C-X-C motif chemokine ligand 10; DAMPs, damage-associated molecular pattern molecules; DC, dendritic cell; IFNγ, interferon gamma; IL-1β, Interleukin-1 beta; IL-6, Interleukin-6; IL-17, Interleukin-17; IL-21, Interleukin-21; IL-22, Interleukin-22; LPS, lipopolysaccharide; Mϕ, macrophages; NH4+, ammonia; NO, nitric oxide; PAMPs, pathogen-associated molecular patterns; PMN, polymorphonuclear; ROS, reactive oxygen species; TLR4, toll-like receptor 4; TNFα, tumor necrosis factor alpha. Created with BioRender.com.

Finally, all pro-inflammatory cytokines, chemokines, PAMPs, DAMPs, and gut microbiome-derived ammonia can reach the portal circulation and result in systemic inflammation and hyperammonemia. Gut dysbiosis, bacterial translocation, and elevated levels of circulating pro-inflammatory cytokines, as well as bacterial metabolites (i.e. methanol and threonine) were also observed in cirrhosis patients with HE [105]. Moreover, gut dysbiosis was associated with levels of circulatory pro-inflammatory cytokines, and circulatory bacterial products were positively associated with HE [105]. Likewise, elevated serum levels of pro-inflammatory cytokines, including ІFN-γ, ІL-1β, and ІL-6, were also correlated with grade of HE in patients with cirrhosis [106]. In cirrhosis patients with HE, the systemic inflammatory response score was also positively correlated with the grade of HE [107]. Furthermore, systemic inflammatory response and hyperammonemia were also reported in acute-on-chronic liver failure (ACLF) and HE patients. Also, in these patients, blood ammonia was correlated with grade of HE and mortality rate [108]. Furthermore, cirrhotic patients with history of previous HE presented higher levels of circulatory IL-6, IL-2, IL-1β, TNFα, and endotoxins [43]. Gut microbiome alteration was also correlated with systemic inflammation in these patients [43]. Increase the number of Th17 cells and reduce the population of Treg, resulting in decrease the Treg/Th17 ratio shown in the blood circulation of cirrhosis patients due to hepatitis C virus [94]. Soluble sCD163 is a good marker that represents monocyte expansion and macrophage activation [109]. The circulatory level of this peptide was significantly raised in cirrhotic patients with ACLF [110]. An increase in serum inflammatory mediators was also revealed in carbon tetrachloride (CCL4)-induced cirrhosis and HE in rats [106]. Furthermore, our group indicated that the levels of pro-inflammatory mediators and ammonia are increased in thioacetamide-induced HE mice [23]. Increased concentrations of pro-inflammatory cytokines, active monocytes, T helper CD34+ cells, and bacterial DNA were seen in circulation of CCL4-induced cirrhosis rats [85]. Moreover, the number of T helper CD34+ cells and monocytes as well as elevated levels of bacterial DNA were demonstrated in the mesenteric lymph nodes of these rats [85].

Advertisement

3. Activated Kupffer cells exacerbate hepatocyte injury and systemic inflammation

Active bacterial metabolites and products, DAMPs, gut-derived pro-inflammatory cytokines, ammonia, and activated immune cells finally reach the liver through the portal vein and are directly exposed to other types of body-resident Mϕ named Kupffer cells (KCs) [111]. They are localized to hepatic sinusoidal endothelial cells and have a great role in scavenging circulatory pathogenic agents (Figure 2). The KCs are amoeboid-shaped cells with several processes, including microvilli, lamellipodia, and pseudopodia, that let them move around and endocytosis pathogens, apoptotic cells, and endotoxins [112]. The liver KCs originate from the embryonic yolk sac and have self-renewing properties [113, 114]. However, it has been shown that bone marrow-derived circulatory monocytes can be differentiated into KCs [115]. The KCs are functionally divided into two distinct types: M1 (pro-inflammatory classically activated KCs) and M2 (anti-inflammatory alternatively activated KCs). M1 KCs are activated in response to LPS, bacterial DNA, and pathogens, as well as pro-inflammatory mediators, resulting in the induction of oxidative stress and the production of pro-inflammatory cytokines. On the other hand, M2 KCs are triggered in response to IL-10, IL-13, and IL4, which lead to the secretion of anti-inflammatory cytokines [116, 117, 118]. KCs express several surface receptors that distinguish them from other cell types in the liver parenchyma. These molecules include CD68, CD14, and TLR4 in humans and CD11b, CD68, F4/80, CD163, CLEC4F, TIM4, TLR4, TLR9, and CRIg in rodents [119].

Figure 2.

Activated Kupffer cells induce liver inflammation and exacerbate systemic inflammation in cirrhosis and HE. DAMPs, damage-associated molecular pattern molecules; CCL2, C-C motif chemokine ligand 2; CCL3, C-C motif chemokine ligand 2; CCL5, C-C motif chemokine ligand 5; DC, dendritic cell; FasL, Fas ligand; HSC, hepatic stellate cell; iNOS, inducible nitric oxide synthases; IFNα, interferon alpha; IFNβ, interferon beta; IFNγ, interferon gamma; IL-1β, Interleukin-1 beta; IL-6, Interleukin-6; IL-8, Interleukin-8; IL-12, Interleukin-12; IL-17, Interleukin-17; IL-21, Interleukin-21; IL-22, Interleukin-22; KC, Kupffer cell; LPS, lipopolysaccharide; MIP-1, macrophage inflammatory protein-1; NF-kβ, nuclear factor kappa-light-chain-enhancer of activated B cells; NH4+, ammonia; PAMPs, pathogen-associated molecular patterns; PRR, pattern recognition receptors; ROS, reactive oxygen species; TGFβ, transforming growth factor-β; TNFα, tumor necrosis factor alpha; TRAIL, TNF-related apoptosis-inducing ligand. Created with BioRender.com

In liver cirrhosis, hepatocyte injury produces a substantial amount of DAMPs such as apoptotic bodies, ATP, high mobility group protein B1 (HMGB1), ATP, and DNA in the perisinusoidal space (space of Disse), where hepatic stellate cells (HSCs) are located (Figure 2) [120, 121, 122]. These agents, along with PAMPs and other gut-derived metabolites, activate HSCs to secrete monocyte and neutrophil chemotactic agents, such as chemokine (C-C motif) ligand 2, as well as activate KCs [123]. Moreover, KCs, through some mediators such as TGFβ, CCL2, CCL3, and CCL5 activate HSCs [124]. The HSCs progress liver fibrosis through a signaling pathway dependent on TGFβ, NLRP3 inflammasome-Caspase1-IL1β, and WNT/β-catenin [125, 126, 127]. Like intestinal Mϕ, KCs express PRRs on their surface, including TLR, RIG-like receptors, C-type lectin receptors (CLR), and nucleotide-binding oligomerization domain (NOD)-like receptors that recognize DAMPs and PAMPs [128, 129, 130]. For instance, activation of TLR4 in response to circulatory LPS through activation of myeloid differentiation factor 88 and overexpression of NF-kβ induces the production of pro-inflammatory cytokines such as IL-1β, IL-6, IL-8, TNFα, intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1) [129, 131, 132, 133]. Moreover, activation of intracellular TLR3 in response to viral RNA through activation of TRIF and IRF3 produces IFNα and IFNβ in KCs [134]. In response to elevated levels of sinusoidal DAMPs and PAMPs, KCs are activated and release pro-inflammatory mediators (i.e. IL-12, IL-1β, iNOS, CCL2, and MIP-1) and induce oxidative stress in the liver parenchyma [118, 135]. Activated KCs also secrete Fas ligand (FasL), tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), and TNFα that trigger hepatocyte apoptosis [136]. Triggered KCs also exacerbate the recruitment of neutrophils and circulatory monocytes through production of several chemoattractant chemokines, such as CXCL1, CXCL2, CXCL8, CCL1, CCL2, and CCL25 in cirrhosis [137]. DCs are also triggered by activated KCs, DAMPs, PAMPs, and liver inflammation. According to function and surface receptors, five subsets of DCs are present in the nonlymphoid tissue of humans and mice. This includes Langerhans DCs (CD207+ and CD14, human), CD11b+ cells (CD207, CD14+, BDCA1+ and CD11chigh, human), CD8α+-like conventional cells (CD207, CD14, CD1a+, BDCA3+ and CD11chigh, human), monocyte-derived cells (Lyc6C+, MAC3+, CD11b+ and CD11c+, mouse), and plasmacytoid DCs (BDCA3+, CD11c-, human) [138, 139, 140, 141]. Two subtypes, including monocyte-derived and plasmacytoid DCs, are implicated in liver cirrhosis [142, 143]. The monocyte-derived DCs express TLR4, TLR8, and NOD-like receptors, while the plasmacytoid DCs express TLR1, TLR7, and TLR9 in both humans and mice [140, 144, 145]. In response to KCs-derived chemokines and liver inflammation, DCs recruit to the liver parenchyma. Liver DCs also express PRRs that mediate their responses to DAMPs and PAMPs. Activated DCs may trigger HSCs and CD8+ T cells through the secretion of IL-12 and programmed death ligand 1 (PD-L1), respectively [146, 147]. DCs also express MHC I and II peptides, which interact with CD8+ and CD4+ receptors and result in the activation of T cells. Moreover, activated DCs can trigger the differentiation of T cells through the production of IL-12 and IFNγ [148]. Severe infiltration of monocyte-derived and plasmacytoid DCs in the liver parenchyma along with increased production of IFNα have been reported in cirrhotic patients with ACLF [142]. In CCL4-induced cirrhosis and HE mouse models, liver inflammation along with increase in the number of CD11c+-MHCIIhigh-CD11b+ DCs have been shown in liver tissue [149]. Activated KCs and DCs have an impact on T cell activity. An increase in the infiltration of inflammatory Th17 cells in the liver parenchyma of patients with chronic hepatitis C has been shown [150]. Moreover, the recruitment of Th17 cells in liver tissue was correlated with grade of liver inflammation [150]. Likewise, the population of Th17 cells is increased in blood and liver tissue of patients with chronic hepatitis B and cirrhosis. Also, raise in the infiltration of Th17 cells into the liver parenchyma and an elevated level of IL-17 were associated with the severity of the disease and grade of liver fibrosis [151]. Furthermore, an increase in the Th17/Treg ratio along with overexpression of NF-kβ, TNFα, IL-1β, and IL-6 was revealed in patients with hepatitis B and cirrhosis [152]. In cirrhotic rats, an increase in the population of CD3+ T cells, T helper cells (CD4+-CD8), and CD134+ T cells were observed in hepatic lymph nodes [85]. Moreover, overexpression of IL-17 mRNA and protein was demonstrated in the liver tissue of bile duct ligation (BDL) rats as a model of chronic liver failure-induced HE [153]. In cirrhosis, the detoxification of ammonia, as well as endogenous agents, PAMPs, and DAMPs by injured hepatocytes is compromised, leading to liver inflammation (Table 2) and elevated levels of these compounds in the systemic circulation. Moreover, pro-inflammatory cytokines and chemokines produced by activated KCs, HSCs, DCs, and infiltrating neutrophils can be released into systemic blood flow and eventually make their way to the brain.

StudyFindings (Liver tissue)References
Luckey et al. (2001)⇧TNFα mRNA, ⇧IL-6 mRNA, ⇧IL-1β mRNA, ⇧cyclooxygenase 2 mRNA, ⇧CD14 mRNA, ⇧I kappa B alpha transcription (in activated Kupffer cells of CCL4 rats)[154]
Aoyama et al. (2010)⇧CX3CR1 mRNA, ⇧CX3CL1 mRNA, ⇧MIP-1α mRNA, ⇧MIP-1β mRNA, ⇧CD68 mRNA, ⇧IL-10, ⇧arginase-1 mRNA, ⇧TIMP Metallopeptidase Inhibitor 1 (TIMP-1) mRNA, ⇧alpha-smooth muscle actin (α-SMA; a marker for visualizing the activated hepatic stellate cells) (CCL4 mice)[155]
Dhanda et al. (2018)⇧IL-6 mRNA and protein, ⇧TNFα mRNA and protein, ⇧MCP-1 mRNA and protein (BDL rats)[156]
Tung et al. (2015)⇧Cells infiltration, ⇧bridging necrosis (BDL rats)[157]
Kang et al. (2014)⇧Swollen and necrotic hepatocytes (CCL4 mice)[158]
Moleset al. (2014)⇧α-SMA mRNA and protein, ⇧CXCL2 mRNA, ⇧TNFα mRNA, ⇧S100 calcium-binding protein A9 (S100a9) mRNA, ⇧neutrophil recruitment (CCL4 mice)[159]
Altaş et al. (2011)⇧Malondialdehyde, ⇧hepatocyte degeneration, ⇧vacuolization, ⇧ hepatocytes necrosis (CCL4 rats)[160]
Tag et al. (2015)⇧Portal fibrosis, ⇧perisinusoidal fibrosis, ⇧α-SMA, ⇧vimentin, ⇧CD45, ⇧infiltration of leukocytes (BDL rats)[161]
Canbay et al. (2004)⇧CXCL1 mRNA, ⇧MIP-2 mRNA, ⇧TGFβ mRNA, ⇧collagen 1 (BDL mice)[162]
Trebicka et al. (2010)⇧collagen accumulation, ⇧α-SMA, ⇧myofibroblasts accumulation, ⇧TGFβ1 mRNA, ⇧Platelet-derived growth factor (PDGF) mRNA, ⇧Connective tissue growth factor (CTGF) mRNA, ⇧Matrix Metallopeptidase 2 (MMP2) mRNA, ⇧inflammation (BDL rats)[163]
Gobejishvili et al. (2013)⇧Apoptosis, ⇧TNFα mRNA, ⇧TGFβ1, ⇧αSMA mRNA and reactivity (BDL rats)[164]
Weng et al. (2018)⇧Infiltration of inflammatory cells, ⇧biliary necrosis, ⇧TNFα mRNA, ⇧IL-6 mRNA, ⇧NF-kβ mRNA (BDL mice)[165]
Gehring et al. (2005)⇧periportal inflammation, ⇧bile duct proliferation, ⇧hepatocyte necrosis, ⇧neutrophil recruitment (BDL mice)[166]
Alric et al. (2000)⇧ED-1 (activated KCs and Mϕ marker), ⇧ED-2 (activated KCS marker) (CCL4 rats)[167]
Nagano et al. (1999)⇧IL-5 mRNA, ⇧-6 mRNA, ⇧IL-12 mRNA (primary biliary cirrhosis patients)[168]
Holland-Fischer et al. (2015)⇧Serum soluble CD163 (KCs activation), ⇧serum lipopolysaccharide binding protein (Cirrhosis patients)[169]
de Lalla et al. (2004)⇧IFNγ, ⇧IL-4, ⇧IL-13, ⇧CD1d + cells (infiltrating antigen presenting cells marker), ⇧periportal inflammation, ⇧fibrosis (cirrhosis patients with hepatitis B)[170]
Napoli et al. (1996)⇧IL-2 mRNA, ⇧IFNγ mRNA, ⇩IL-10 mRNA (Cirrhosis patients)[171]
Farinati et al. (2006)⇧IL-1β mRNA, ⇧c-Myc mRNA, ⇧8-hydroxydeoxyguanosine (a product of DAN oxidative damage) (patients with cirrhosis)[172]
Li et al. (2015)⇧Casphase 3, ⇧apoptosis, ⇧pigmented hepatocytes, ⇧ductular reaction, ⇧canalicular bilirubinostasis, ⇧TGFβ, ⇧IL-10 (patients with cirrhosis)[173]
Chuang et al. (2006)⇧CD56+ cells (natural killer cells) (patients with primary biliary cirrhosis)[174]

Table 2.

Significant hepatic alterations following cirrhosis and HE.

Advertisement

4. Activated brain microglia induce neuroinflammation

One subset of cerebral glial cells, known as brain microglia, makes up approximately 10–15% of the cells in the brain parenchyma. As resident macrophage cells, microglia differentiate from progenitors in the yolk sac and develop through CSF-1 and IL-34 signaling. During the development of the CNS, microglia have a crucial role in programmed cell death, neurogenesis, phagocytosis of neuronal cells, reforming synapses, and survival of neural progenitor cells [175, 176, 177]. Under normal conditions, microglia scavenge brain parenchyma from dead cells, apoptotic neurons, and misfolding proteins (via phagocytosis), maintain the activity of other brain cells (i.e. astrocytes, oligodendrocytes, and neurons) through extracellular signaling molecules, remodeling false synapses and presenting antigens in the CNS [178, 179, 180, 181]. Basically, the central nervous system is protected from circulatory neurotoxic substances through a well-organized and intact blood-brain barrier (BBB). The BBB is a multilayer semipermeable structure that consists of capillary single-layer endothelial cells, the basement membrane, pericytes, and the astrocyte end-feet [182, 183]. This barrier is restricted, and only lipophilic molecules (MW 500 Da) are permitted to passively cross through this structure. Moreover, carrier-mediated transport facilitates the transportation of glucose and amino acids. Other limit transport systems (i.e. restricted paracellular pathway, transcytosis transport) mediated the entry of hydrated ions, low-density lipoprotein, iron, and insulin-like growth factor into the brain tissue [17, 183]. Transport across the BBB is constrained by several proteins, such as claudin-5 and occludin, zonula occludens-1 (ZO-1), and junctional adhesion molecules between endothelial cells (paracellular pathway). Systemic inflammation and injuries negatively affect tight junction structure and functions, resulting in disruption of the integrity of the BBB and penetration of harmful substances into the brain tissue [17]. These agents activate microglia and astrocytes to produce pro-inflammatory cytokines, which cause neuroinflammation. While neuroinflammation is a normal physiological process that protects the brain environment from infection, toxic agents, and misfolding proteins, chronic cerebral inflammation exerts detrimental consequences in the CNS [184]. Microglia are identified by several surface receptors (CD11b, CD45, CX3CR1, CD16, CD40, CD80, CD68, CD115, and others) and intracellular proteins (IBA1, Pu.1, HexB, vimentin, and Sall1) in the CNS [185, 186, 187, 188, 189, 190, 191, 192]. Functionally, there are two distinct states of microglia. The resting or ramified microglia represent round cell bodies and vigorous branching processes, which constitute a large part of cerebral microglia in physiological conditions. Under pathological conditions (PAMPs, DAMPs, and toxic substances in the brain), amoeboid form or activated microglia are the dominant state. Microglia in this form are less branched and have round cell bodies with thick processes that make it easier to move toward injury sites [193, 194, 195, 196]. Just like liver KCs, microglia can be classified phenotypically into two types: M1 (pro-inflammatory phenotype) and M2 (anti-inflammatory). The M1 microglia (CD14+, CD16+, CD40+, CD32+, CD86+, and MHCII+) often triggered by pro-inflammatory factors such as TNFα, IFNγ, and LPS, which cause the production of pro-inflammatory mediators. M2 microglia (CD163+ and CD206+) in response to IL-13, TGFβ, IL-4, and IL-10 secrete anti-inflammatory mediators and neurotropic factors, including IL-13, CCL22, CCL17, IL-10, brain-derived neurotrophic factor (BDNF), CSF-1, glial cell-derived neurotrophic factor (GDNF), nerve growth factor (NGF), and TGFβ [197, 198].

Following cirrhosis, hyperammonemia, and systemic inflammation may interrupt the function of tight junction proteins and impair the integrity of the BBB (Table 3) [229, 230, 231]. Hyperammonemia, systemic inflammation, and enhanced BBB permeability were found in mice with BDL as a model for chronic liver disease-induced HE [8]. Similar findings were also revealed in CCL4-induced cirrhosis in rats [232]. Likewise, disruption of the BBB and decreased mRNA and protein expression of tight junction proteins (i.e. claudin-5, occludin, and ZO-1) were demonstrated in a BDL-rat model of HE [205]. Furthermore, systemic inflammation, hyperammonemia, and a decrease in the expression of thigh junction proteins were revealed in mice after the injection of CCL4 [233, 234]. Quinn et al. demonstrated elevated circulating bile acids increase the permeability of the BBB through a detrimental impact on tight junction proteins in BDL rats [235]. Elevated circulatory ammonia (unprotonated form; NH3) can passively diffuse into the brain parenchyma. Moreover, several transporting systems (i.e., aquaporins, Na+/H+ exchangers, Na+/K+-ATPase, and potassium channels) mediated ammonia transportation (ionic form; NH4+) across the BBB [236, 237, 238, 239, 240]. Disrupted integrity of the BBB and tight junction proteins leads to exposure of brain tissue to high levels of harmful circulatory substances, including LPS, pro-inflammatory cytokines, chemokines, ammonia, bacterial DNA, DAMPs, and bile acids (Figure 3). In addition to impaired BBB, several reported pathways were identified for the entry of systemic cytokines into the brain parenchyma: peripheral vagus nerve, circumventricular organs, recruitment of activated circulatory immune cells, and activated capillary endothelial cells that produce pro-inflammatory cytokines in the brain parenchyma [241, 242, 243, 244]. Elevated levels of DAMPs (i.e., HMGB1, ATP, bacterial DNA segments, and apoptotic bodies), pro-inflammatory cytokines, ammonia, LPS, bacterial metabolites, and their components in the brain parenchyma trigger microglia through activation of their TLR2, TLR4, TLR9, NOD2, RAGE, and purinergic receptors, resulting in change in their phenotypes from resting-branched to active-amoeboid state (Figure 3). The activated microglia produce and secrete pro-inflammatory mediators, including IL-1β, TNFα, IL-6, IL-12, CCL2, iNOS, IL-23, CXCL10, and PGE2, which progress the induction of oxidative stress and neuroinflammation (Figure 3). Chronic neuroinflammation in response to elevated cerebral levels of PAMPS, DAMPs, and ammonia induces neuronal loss and impairs neurogenesis, resulting in neurodegeneration and encephalopathy [199, 223, 224, 245, 246, 247, 248, 249, 250, 251, 252]. Interestingly, postmortem brain examinations of cirrhotic individuals with HE showed up-regulation of genes related to oxidative stress, inflammatory pathways, and microglial activation [224]. Hyperammonemia in BDL rats triggered microglial activation, increased cerebral levels of iNOS, PGE2, and IL-1β, and progressed motor dysfunction, as well as impaired cognitive functions [199]. Microglia activation and increased population of CD11+ cells, as well as an increase in the number of degenerative neurons demonstrated in the hippocampus and cerebellum of BDL rats [207]. Moreover, increased expression of microglia marker Iba1 and cerebral ammonia were seen in the prefrontal cortex of BDL mice [8]. Increased lipid peroxidation, decreased total antioxidant capacity, infiltrated leukocytes, and injured neuronal cells were observed in the brain tissues of CCL4-induced cirrhosis rats [253]. In addition, mouse models of liver fibrosis by CCL4 showed increased mRNA expression of IL-1β, TGF-β1, and matrix metalloproteinase-9 (MMP9) in the brain parenchyma [234]. Likewise, mRNA and protein levels of MMP9 and VCAM-1 were up-regulated in different brain regions of BDL rats [205]. Activated microglia also stimulate the recruitment of circulatory monocytes and neutrophils into brain tissue through their secretory chemoattractant agents, CCL2, MCP-1, CXCL3, and MMPs (Figure 3) [254, 255, 256]. In a mouse model of chronic liver failure due to bile duct resection, activated brain microglia with a mechanism dependent on TNFα-TNF receptor 1 (TNFR1)-produced CCL2 and MCP-1 stimulate monocyte chemotaxis into the brain [256]. Cerebral astrocytes (glial fibrillary acidic protein; GFAP) are directly exposed to ammonia and circulatory PAMPs and DAMPs. These agents changed the astrocyte state from resting to a reactive form that produced pro-inflammatory mediators and induced oxidative stress. Afterward, reactive astrocytes activate microglia through secretion of G-CSF and CCL11 [257, 258, 259, 260]. Moreover, activated microglia, via production of GM-CSF, TNFα, and IL-1β trigger astrocytes to produce pro-inflammatory cytokines that exacerbate neuroinflammation [231]. Hyperammonemic cerebral conditions induce astrocyte swelling and brain edema through mislocalization of aquaporin four water channels, inflammation, TLR4 activation, mitochondrial dysfunction, glutamine accumulation, and induction of oxidative stress [7, 257, 261]. Although cerebral edema often progresses in acute forms of HE, patients with cirrhosis were found to have minimal levels of brain edema using 1H-magnetic resonance (MR) spectroscopy [262]. Systemic inflammation, hyperammonemia, an abnormal phenotype of astrocytes (thin chromatin and enlarged nuclei), and increased brain water content (an indicator of cerebral edema) were demonstrated in BDL rats [263]. Moreover, in other experimental models of cirrhosis-induced HE, astrogliosis, gliopathy, neuroinflammation, and raise in brain water content were observed [8, 207, 233, 234, 248, 264].

StudyFindings (Brain tissue)References
Rodrigo et al. (2010)⇧MHCII, ⇧iNOS, ⇧IL-1β, ⇧PGE2, ⇧ammonia (BDL rats)[199]
Claeys et al. (2022)⇧Astrocyte reactivity (⇧GFAP), ⇧microglial activation, ⇧BBB permeabilization (BDL mice)[8]
Tamnanloo et al. (2023)⇧Cleaved caspase-3, ⇧IL-1β, ⇧TNFα, ⇧Bax/Bcl2 ratio, ⇧oxidative stress, ⇩total antioxidant capacity (BDL rats)[200]
Bosoi et al. (2012)⇧Brain water content, ⇧ammonia in cerebrospinal fluid (CSF) (BDL rats)[201]
Magen et al. (2009)⇧TNFR1 mRNA, ⇩BDNF mRNA, ⇩A2A adenosine receptor (BDL mice)[202]
André Clément et al. (2020)⇩NeuN, ⇧cleaved caspase-3, ⇧neuronal apoptosis[203]
Stojanović et al. (2023)⇩Catalase, ⇩superoxide dismutase activities, ⇩glutathione, ⇩peroxidase activity, ⇧lipid peroxidation, ⇧iNOS (CCL4 rats)[204]
Dhanda et al. (2018)⇧BBB permeabilization, ⇧brain water content, ⇧aquaporin-4 mRNA and protein, ⇧MMP-9 mRNA and protein, ⇩mRNA and protein occludin, ⇩mRNA and protein claudin-5, ⇩mRNA and protein ZO-1 (BDL rats)[205]
Yang et al. (2015)⇩BrdU (neurogenesis), ⇩NeuN (post-mitotic neurons) ⇩GFAP (astrocytes) (CCL4 rats)[206]
Golshani et al. (2019)⇧degenerative cerebellar Purkinje neurons, ⇧degenerative hippocampal pyramidal neurons, ⇧GFAP, ⇧CD11+ (BDL rats)[207]
Jung Hsu et al. (2021)⇧Iba1, ⇧IL-1β, ⇧IFNγ, ⇧TNFα, ⇧IGF-1, ⇧VEGFD (BDL rats)[208]
Chen et al. (2014)⇧Microglia activation, ⇧GFAP, ⇩dendritic spine density (BDL rats)[209]
Chen et al. 2012⇧NADPH-dependent superoxide anion, ⇧BBB disruption (BDL rats)[210]
Pierzchala et al. (2022)⇧8-Oxo-2′-deoxyguanosine, ⇧IL-6, ⇧glutathione peroxidase 1, ⇧superoxide dismutase, ⇧MnSOD (BDL rats)[211]
Ahmadi et al. (2020)⇧Jnk3 mRNA, ⇧JNK3 protein (BDL rats)[212]
Y Avraham et al. (2010)⇧TNFR1 mRNA, ⇩BDNF mRNA, ⇩5-HT1A receptor (BDL mice)[213]
M.V. Gee et al. (2023)⇩Astrocyte covering BBB, ⇧neuronal senescence, ⇩neuronal connectivity (BDL mice)[214]
Shabani et al. (2019)⇧Neuronal loss, ⇧neuronal apoptosis (BDL rats)[215]
Balasubramaniyan et al. (2012)⇧Brain water content, ⇧TNFα, ⇧NOX-1 protein, (BDL rats)[216]
Dhanda et al. (2018)⇧IL-6 mRNA and protein, ⇧TNFα mRNA and protein, ⇧MCP-1 mRNA and protein, ⇩Iba1 expression, ⇩GFAP expression, ⇩BDNF (BDL rats)[156]
Wright et al. (2010)⇧Ammonia, ⇧TNFα, ⇧IL-6, ⇧brain water content, ⇧iNOS, ⇧IL-1β, ⇧GFAP, ⇧TGFβ mRNA (BDL rats)[217]
Faropoulos et al. (2010)⇧Superoxide radical, ⇩occludin (BDL rats)[218]
Boer et al. (2009)⇩Activity of mitochondrial complexes I, II and IV (CCL4 rats)[219]
Liu et al. (2020)⇧IL1β mRNA, ⇧MCP-1 mRNA, ⇧IBA1 mRNA, ⇧GFAP mRNA, ⇧NeuN/Fox3 (CCL4 mice)[14]
Khan et al. (2019)⇧IL-1β, ⇧IL-6, ⇧TNFα, ⇩glutathione, ⇩catalase, ⇧malondialdehyde, ⇧nitrite, ⇩GFAP, ⇩BDNF, ⇩VEGF, ⇧apoptosis (CCL4 mice)[220]
Machado et al. (2015)⇧TNFα, ⇧IL-1β, ⇧IL-18, IL-6 (unchanged), IL-10 (unchanged) (CCL4 rats)[221]
Hadjihambi et al. (2018)⇩AQP4, ⇩glymphatic clearance (BDL rats)[222]
Jung Hsu et al. (2021)⇧NF-κβ mRNA, ⇧Iba1mRNA, ⇧TNFα mRNA (Cirrhosis patients with HE)[208]
Irina et al. (2021)⇧Iba1, cytokines unchanged (postmortem; cirrhosis patients with HE)[223]
Boris et al. (2013)Change in the expression patterns of genes related to oxidative stress, microglia activation, inflammatory signaling, cell proliferation, and apoptosis (postmortem; cirrhosis patients with HE)[224]
Grover et al. (2011)⇧PK11195 binding, a ligand for activated microglia (patients with chronic hepatitis C)[225]
Balzano et al. (2018)⇧Purkinje cell loss, ⇧neuronal loss in granular layer, ⇧microglial activation, ⇧reactive astrocytes, ⇧degeneration of Bergmann glia, ⇧lymphocytes infiltration (postmortem; cirrhosis patients with HE)[226]
Cagnin et al. (2006)⇧PK11195 binding, a ligand for activated glial cells (patients with cirrhosis)[227]
Butterworth et al. (1988)⇧Dysfunctional astrocytes (astrogliosis) (postmortem; cirrhosis patients with HE)[228]

Table 3.

Main cerebral changes following cirrhosis and HE.

Figure 3.

Hyperammonemia, systemic inflammation, and gut-derived compounds disrupt the integrity of the blood-brain barrier and induce neuroinflammation in cirrhosis, highlighting the role of brain-activated microglia. BBB, blood-brain barrier; DAMPs, damage-associated molecular pattern molecules; CCL2, C-C motif chemokine ligand 2; CCL11, C-C motif chemokine ligand 11; CXCL3, C-X-C motif chemokine ligand 3; CXCL10, C-X-C motif chemokine ligand 10; G-CSF, granulocyte colony-stimulating factor; iNOS, inducible nitric oxide synthases; IFNγ, interferon gamma; IL-1β, Interleukin-1 beta; IL-6, Interleukin-6; IL-12, Interleukin-12; IL-17, Interleukin-17; IL-22, Interleukin-22; IL-23, Interleukin-23; MCP-1, monocyte chemoattractant protein-1; MMPs, matrix metalloproteinases; NH4+, ammonia; PAMPs, pathogen-associated molecular patterns; PGE2, prostaglandin E2; PRR, pattern recognition receptors; ROS, reactive oxygen species; TNFα, tumor necrosis factor alpha. Created with BioRender.com.

Advertisement

5. Conclusion

While the pathophysiology of cirrhosis-induced HE remains to be elucidated, the progression of cirrhosis and HE is associated with innate immune disturbances at different levels of the gut-liver-brain axis. In cirrhosis, gut dysbiosis and bacterial overgrowth cause intestinal barrier disruption, leading to bacterial translocation and activation of intestinal Mϕ and DCs that initiate tissue inflammation. Bacterial metabolites, ammonia, and gut-derived inflammatory cytokines reach the hepatic sinusoids and activate HSCs and KCs, resulting in the production of liver inflammation and the induction of oxidative stress and exacerbate systemic inflammation. Also, injured hepatocytes cannot detoxify ammonia, resulting in hyperammonemia. Systemic inflammation, hyperammonemia, and endotoxemia induce the BBB injury and allow brain tissue to be exposed to all circulatory neurotoxic substances. These agents stimulate brain microglia to produce pro-inflammatory cytokines and induce oxidative stress in the CNS. Neuroinflammation and oxidative stress decrease neurogenesis and induce neuronal degeneration, resulting in neurological and psychological manifestations seen in HE. Due to the implication of Mϕ, immune cells and inflammation in different levels of gut-liver-brain axis, they can be potent therapeutic targets for future experimental and clinical research to find an all-encompassing treatment option that targets restoration of the immune system in multiple organs following cirrhosis and HE.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Rose CF, Amodio P, Bajaj JS, et al. Hepatic encephalopathy: Novel insights into classification, pathophysiology and therapy. Journal of Hepatology. 2020;73:1526-1547. DOI: 10.1016/j.jhep.2020.07.013
  2. 2. Ochoa-Sanchez R, Rose CF. Pathogenesis of hepatic encephalopathy in chronic liver disease. Journal of Clinical and Experimental Hepatology. 2018;8:262-271. DOI: 10.1016/j.jceh.2018.08.001
  3. 3. Jalan R, Rose CF. Heretical thoughts into hepatic encephalopathy. Journal of Hepatology. 2022;77:539-548. DOI: 10.1016/j.jhep.2022.03.014
  4. 4. Montagnese S, Russo FP, Amodio P, et al. Hepatic encephalopathy 2018: A clinical practice guideline by the Italian Association for the Study of the liver (AISF). Digestive and Liver Disease. 2019;51:190-205. DOI: 10.1016/j.dld.2018.11.035
  5. 5. Weissenborn K. Hepatic encephalopathy: Definition, clinical grading and diagnostic principles. Drugs. 2019;79:5-9. DOI: 10.1007/s40265-018-1018-z
  6. 6. Kroupina K, Bémeur C, Rose CF. Amino acids, ammonia, and hepatic encephalopathy. Analytical Biochemistry. 2022;649:114696
  7. 7. Sepehrinezhad A, Zarifkar A, Namvar G, et al. Astrocyte swelling in hepatic encephalopathy: Molecular perspective of cytotoxic edema. Metabolic Brain Disease. 2020;35:559-578. DOI: 10.1007/s11011-020-00549-8
  8. 8. Claeys W, Van Hoecke L, Geerts A, et al. A mouse model of hepatic encephalopathy: Bile duct ligation induces brain ammonia overload, glial cell activation and neuroinflammation. Scientific Reports. 2022;12:17558. DOI: 10.1038/s41598-022-22423-6
  9. 9. Shawcross D, Jalan R. The pathophysiologic basis of hepatic encephalopathy: Central role for ammonia and inflammation. Cellular and Molecular Life Sciences CMLS. 2005;62:2295-2304. DOI: 10.1007/s00018-005-5089-0
  10. 10. Shawcross DL, Wright G, Olde Damink SWM, et al. Role of ammonia and inflammation in minimal hepatic encephalopathy. Metabolic Brain Disease. 2007;22:125-138. DOI: 10.1007/s11011-006-9042-1
  11. 11. Aldridge DR, Tranah EJ, Shawcross DL. Pathogenesis of hepatic encephalopathy: Role of ammonia and systemic inflammation. Journal of Clinical and Experimental Hepatology. 2015;5:S7-S20. DOI: 10.1016/j.jceh.2014.06.004
  12. 12. Tranah TH, Vijay GK, Ryan JM, et al. Systemic inflammation and ammonia in hepatic encephalopathy. Metabolic Brain Disease. 2013;28:1-5. DOI: 10.1007/s11011-012-9370-2
  13. 13. Laguna, de la Vera A-L, Welsch C, Pfeilschifter W, et al. Chapter 8 - gut–liver–brain axis in chronic liver disease with a focus on hepatic encephalopathy. In: Stasi C, editor. The Complex Interplay between Gut-Brain, Gut-Liver, and Liver-Brain Axes. London, United Kingdom: Academic Press; 2021. pp. 159-185
  14. 14. Liu R, Kang JD, Sartor RB, et al. Neuroinflammation in murine cirrhosis is dependent on the gut microbiome and is attenuated by fecal transplant. Hepatology. 2020;71:611-626
  15. 15. Kang DJ, Betrapally NS, Ghosh SA, et al. Gut microbiota drive the development of neuroinflammatory response in cirrhosis in mice. Hepatology. 2016;64:1232-1248
  16. 16. Jain L, Sharma BC, Srivastava S, et al. Serum endotoxin, inflammatory mediators, and magnetic resonance spectroscopy before and after treatment in patients with minimal hepatic encephalopathy. Journal of Gastroenterology and Hepatology. 2013;28:1187-1193
  17. 17. Shahbazi A, Sepehrinezhad A, Vahdani E, et al. Gut dysbiosis and blood-brain barrier alteration in hepatic encephalopathy: From gut to brain. Biomedicine. 2023;11:1272
  18. 18. Wu H, Zhang Y, Yu J, et al. Gut-liver-brain axis: A complex network influences human health and diseases. Frontiers in Neuroscience. 2023;17:1241069
  19. 19. Nguyen HH, Swain MG. Avenues within the gut-liver-brain axis linking chronic liver disease and symptoms. Frontiers in Neuroscience. 2023;17:1171253
  20. 20. Beldowska A, Barszcz M, Dunislawska A. State of the art in research on the gut-liver and gut-brain axis in poultry. Journal of Animal Science and Biotechnology. 2023;14:37
  21. 21. Ding JH, Jin Z, Yang XX, et al. Role of gut microbiota via the gut-liver-brain axis in digestive diseases. World Journal of Gastroenterology. 2020;26:6141-6162. DOI: 10.3748/wjg.v26.i40.6141
  22. 22. Krakovski MA, Arora N, Jain S, et al. Diet-microbiome-gut-brain nexus in acute and chronic brain injury. Frontiers in Neuroscience. 2022;16:1002266. DOI: 10.3389/fnins.2022.1002266
  23. 23. Sepehrinezhad A, Shahbazi A, Joghataei MT, et al. Inhibition of autotaxin alleviates pathological features of hepatic encephalopathy at the level of gut–liver–brain axis: An experimental and bioinformatic study. Cell Death & Disease. 2023;14:490
  24. 24. Luo M, Xin R-J, Hu F-R, et al. Role of gut microbiota in the pathogenesis and therapeutics of minimal hepatic encephalopathy via the gut-liver-brain axis. World Journal of Gastroenterology. 2023;29:144
  25. 25. Ahluwalia V, Betrapally NS, Hylemon PB, et al. Impaired gut-liver-brain axis in patients with cirrhosis. Scientific Reports. 2016;6:26800. DOI: 10.1038/srep26800
  26. 26. Fogg DK, Sibon C, Miled C, et al. A clonogenic bone marrow progenitor specific for macrophages and dendritic cells. Science. 2006;311:83-87
  27. 27. van Furth R, Cohn ZA. The origin and kinetics of mononuclear phagocytes. The Journal of Experimental Medicine. 1968;128:415-435
  28. 28. Cerovic V, Bain CC, Mowat AM, et al. Intestinal macrophages and dendritic cells: what's the difference? Trends in Immunology. 2014;35:270-277
  29. 29. Zhang F, Wang H, Wang X, et al. TGF-β induces M2-like macrophage polarization via SNAIL-mediated suppression of a pro-inflammatory phenotype. Oncotarget. 2016;7:52294-52306. DOI: 10.18632/oncotarget.10561
  30. 30. Gong D, Shi W, Yi S-J, et al. TGFβ signaling plays a critical role in promoting alternative macrophage activation. BMC Immunology. 2012;13:31. DOI: 10.1186/1471-2172-13-31
  31. 31. Boulakirba S, Pfeifer A, Mhaidly R, et al. IL-34 and CSF-1 display an equivalent macrophage differentiation ability but a different polarization potential. Scientific Reports. 2018;8:256. DOI: 10.1038/s41598-017-18433-4
  32. 32. Schridde A, Bain CC, Mayer JU, et al. Tissue-specific differentiation of colonic macrophages requires TGFβ receptor-mediated signaling. Mucosal Immunology. 2017;10:1387-1399. DOI: 10.1038/mi.2016.142
  33. 33. Kumawat AK, Yu C, Mann EA, et al. Expression and characterization of αvβ5 integrin on intestinal macrophages. European Journal of Immunology. 2018;48:1181-1187
  34. 34. Henson PM. Cell removal: efferocytosis. Annual Review of Cell and Developmental Biology. 2017;33:127-144
  35. 35. Cosin-Roger J, Ortiz-Masià MD, Barrachina MD. Macrophages as an emerging source of Wnt ligands: Relevance in mucosal integrity. Frontiers in Immunology. 2019;10:2297. DOI: 10.3389/fimmu.2019.02297
  36. 36. Chikina AS, Nadalin F, Maurin M, et al. Macrophages maintain epithelium integrity by limiting fungal product absorption. Cell. 2020;183(411–428):e416
  37. 37. D'Angelo F, Bernasconi E, Schäfer M, et al. Macrophages promote epithelial repair through hepatocyte growth factor secretion. Clinical and Experimental Immunology. 2013;174:60-72. DOI: 10.1111/cei.12157
  38. 38. Na YR, Jung D, Stakenborg M, et al. Prostaglandin E(2) receptor PTGER4-expressing macrophages promote intestinal epithelial barrier regeneration upon inflammation. Gut. 2021;70:2249-2260. DOI: 10.1136/gutjnl-2020-322146
  39. 39. Viola MF, Boeckxstaens G. Niche-specific functional heterogeneity of intestinal resident macrophages. Gut. 2021;70:1383-1395
  40. 40. Muller PA, Koscsó B, Rajani GM, et al. Crosstalk between muscularis macrophages and enteric neurons regulates gastrointestinal motility. Cell. 2014;158:300-313. DOI: 10.1016/j.cell.2014.04.050
  41. 41. Luo J, Qian A, Oetjen LK, et al. TRPV4 channel signaling in macrophages promotes gastrointestinal motility via direct effects on smooth muscle cells. Immunity. 2018;49:107-119.e104. DOI: 10.1016/j.immuni.2018.04.021
  42. 42. Avetisyan M, Rood JE, Huerta Lopez S, et al. Muscularis macrophage development in the absence of an enteric nervous system. Proceedings of the National Academy of Sciences. 2018;115:4696-4701. DOI: 10.1073/pnas.1802490115
  43. 43. Bajaj JS, Betrapally NS, Hylemon PB, et al. Salivary microbiota reflects changes in gut microbiota in cirrhosis with hepatic encephalopathy. Hepatology. 2015;62:1260-1271. DOI: 10.1002/hep.27819
  44. 44. Zhang Z, Zhai H, Geng J, et al. Large-scale survey of gut microbiota associated with MHE via 16S rRNA-based pyrosequencing. American College of Gastroenterology. 2013;108:1601-1611
  45. 45. Wang WW, Zhang Y, Huang XB, et al. Fecal microbiota transplantation prevents hepatic encephalopathy in rats with carbon tetrachloride-induced acute hepatic dysfunction. World Journal of Gastroenterology. 2017;23:6983-6994. DOI: 10.3748/wjg.v23.i38.6983
  46. 46. Wang L, Cui H, Li Y, et al. Kang-Xian pills inhibit inflammatory response and decrease gut permeability to treat carbon tetrachloride-induced chronic hepatic injury through modulating gut microbiota. Evidence-based Complementary and Alternative Medicine. 2020;2020:8890182. DOI: 10.1155/2020/8890182
  47. 47. Zhou K, Jiang M, Liu Y, et al. Effect of bile pigments on the compromised gut barrier function in a rat model of bile duct ligation. PLoS One. 2014;9:e98905. DOI: 10.1371/journal.pone.0098905
  48. 48. Wang C, Ma C, Fu K, et al. Phillygenin attenuates carbon tetrachloride-induced liver fibrosis via modulating inflammation and gut microbiota. Frontiers in Pharmacology. 2021;12. DOI: 10.3389/fphar.2021.756924
  49. 49. Luyer MDP, Buurman WA, Hadfoune M, et al. High-fat enteral nutrition reduces endotoxin, tumor necrosis factor-alpha and gut permeability in bile duct-ligated rats subjected to hemorrhagic shock. Journal of Hepatology. 2004;41:377-383. DOI: 10.1016/j.jhep.2004.04.026
  50. 50. Palma P, Mihaljevic N, Hasenberg T, et al. Intestinal barrier dysfunction in developing liver cirrhosis: An in vivo analysis of bacterial translocation. Hepatology Research. 2007;37:6-12. DOI: 10.1111/j.1872-034X.2007.00004.x
  51. 51. Lei W, Zhao C, Sun J, et al. Electroacupuncture ameliorates intestinal barrier destruction in mice with bile duct ligation–induced liver injury by activating the cholinergic anti-inflammatory pathway. Neuromodulation. 2022;25:1122-1133. DOI: 10.1016/j.neurom.2022.02.001
  52. 52. Fu K, Ma C, Wang C, et al. Forsythiaside A alleviated carbon tetrachloride-induced liver fibrosis by modulating gut microbiota composition to increase short-chain fatty acids and restoring bile acids metabolism disorder. Biomedicine & Pharmacotherapy. 2022;151:113185. DOI: 10.1016/j.biopha.2022.113185
  53. 53. Sánchez E, Nieto JC, Boullosa A, et al. VSL#3 probiotic treatment decreases bacterial translocation in rats with carbon tetrachloride-induced cirrhosis. Liver International. 2015;35:735-745. DOI: 10.1111/liv.12566
  54. 54. Cabrera-Rubio R, Patterson AM, Cotter PD, et al. Cholestasis induced by bile duct ligation promotes changes in the intestinal microbiome in mice. Scientific Reports. 2019;9:12324. DOI: 10.1038/s41598-019-48784-z
  55. 55. Zhao T, Zhu Y, Yao L, et al. IGF-1 alleviates CCL4-induced hepatic cirrhosis and dysfunction of intestinal barrier through inhibition TLR4/NF-κB signaling mediated by down-regulation HMGB1. Annals of Hepatology. 2021;26:100560. DOI: 10.1016/j.aohep.2021.100560
  56. 56. Safari F, Sharifi M, Talebi A, et al. Alleviation of cholestatic liver injury and intestinal permeability by lubiprostone treatment in bile duct ligated rats: Role of intestinal FXR and tight junction proteins claudin-1, claudin-2, and occludin. Naunyn-Schmiedeberg's Archives of Pharmacology. 2023;396:2009-2022. DOI: 10.1007/s00210-023-02455-z
  57. 57. Assimakopoulos SF, Vagianos CE, Patsoukis N, et al. Evidence for intestinal oxidative stress in obstructive jaundice-induced gut barrier dysfunction in rats. Acta Physiologica Scandinavica. 2004;180:177-185. DOI: 10.1046/j.0001-6772.2003.01229.x
  58. 58. Chen Y, Qin N, Guo J, et al. Functional gene arrays-based analysis of fecal microbiomes in patients with liver cirrhosis. BMC Genomics. 2014;15:753. DOI: 10.1186/1471-2164-15-753
  59. 59. Assimakopoulos SF, Tsamandas AC, Tsiaoussis GI, et al. Intestinal mucosal proliferation, apoptosis and oxidative stress in patients with liver cirrhosis. Annals of Hepatology. 2015;12:301-307
  60. 60. Chen Y, Yang F, Lu H, et al. Characterization of fecal microbial communities in patients with liver cirrhosis. Hepatology. 2011;54:562-572. DOI: 10.1002/hep.24423
  61. 61. Du Plessis J, Vanheel H, Janssen CEI, et al. Activated intestinal macrophages in patients with cirrhosis release NO and IL-6 that may disrupt intestinal barrier function. Journal of Hepatology. 2013;58:1125-1132. DOI: 10.1016/j.jhep.2013.01.038
  62. 62. Pascual S, Such J, Esteban A, et al. Intestinal permeability is increased in patients with advanced cirrhosis. Hepato-Gastroenterology. 2003;50:1482-1486
  63. 63. Yang B, Sun T, Chen Y, et al. The role of gut microbiota in mice with bile duct ligation-evoked cholestatic liver disease-related cognitive dysfunction. Frontiers in Microbiology. 2022;13:909461
  64. 64. Đurašević S, Pejić S, Grigorov I, et al. Effects of C60 fullerene on thioacetamide-induced rat liver toxicity and gut microbiome changes. Antioxidants. 2021;10:911
  65. 65. De Minicis S, Rychlicki C, Agostinelli L, et al. Dysbiosis contributes to fibrogenesis in the course of chronic liver injury in mice. Hepatology. 2014;59:1738-1749
  66. 66. Li Y, Lv L, Ye J, et al. Bifidobacterium adolescentis CGMCC 15058 alleviates liver injury, enhances the intestinal barrier and modifies the gut microbiota in D-galactosamine-treated rats. Applied Microbiology and Biotechnology. 2019;103:375-393
  67. 67. Muñoz L, Borrero MJ, Úbeda M, et al. Intestinal immune dysregulation driven by dysbiosis promotes barrier disruption and bacterial translocation in rats with cirrhosis. Hepatology. 2019;70:925-938. DOI: 10.1002/hep.30349
  68. 68. Qin N, Yang F, Li A, et al. Alterations of the human gut microbiome in liver cirrhosis. Nature. 2014;513:59-64. DOI: 10.1038/nature13568
  69. 69. Úbeda M, Lario M, Muñoz L, et al. Obeticholic acid reduces bacterial translocation and inhibits intestinal inflammation in cirrhotic rats. Journal of Hepatology. 2016;64:1049-1057
  70. 70. Such J, Guardiola JV, de Juan J, et al. Ultrastructural characteristics of distal duodenum mucosa in patients with cirrhosis. European Journal of Gastroenterology & Hepatology. 2002;14:371-376
  71. 71. Ramachandran A, Prabhu R, Thomas S, et al. Intestinal mucosal alterations in experimental cirrhosis in the rat: Role of oxygen free radicals. Hepatology. 2002;35:622-629
  72. 72. Reiberger T, Ferlitsch A, Payer BA, et al. Non-selective betablocker therapy decreases intestinal permeability and serum levels of LBP and IL-6 in patients with cirrhosis. Journal of Hepatology. 2013;58:911-921
  73. 73. Kaliannan K. Compromise of α-defensin function in liver cirrhosis facilitates the toxic relationship between gut permeability and endotoxemia. Digestive Diseases and Sciences. 2018;63:2492-2494. DOI: 10.1007/s10620-018-5197-y
  74. 74. Wiest R, Lawson M, Geuking M. Pathological bacterial translocation in liver cirrhosis. Journal of Hepatology. 2014;60:197-209. DOI: 10.1016/j.jhep.2013.07.044
  75. 75. Yan AW, Fouts DE, Brandl J, et al. Enteric dysbiosis associated with a mouse model of alcoholic liver disease. Hepatology. 2011;53:96-105
  76. 76. Tsiaoussis GI, Assimakopoulos SF, Tsamandas AC, et al. Intestinal barrier dysfunction in cirrhosis: Current concepts in pathophysiology and clinical implications. World Journal of Hepatology. 2015;7:2058-2068. DOI: 10.4254/wjh.v7.i17.2058
  77. 77. Chen S, Li X, Li M, et al. Mucosal expression of defensin-5, soluble phospholipase A2 and lysozyme in the intestine in a rat model of acute liver failure and its relationship to intestinal bacterial translocation. Gastroenterología y Hepatología. 2020;43:293-300. DOI: 10.1016/j.gastrohep.2020.01.004
  78. 78. Teltschik Z, Wiest R, Beisner J, et al. Intestinal bacterial translocation in rats with cirrhosis is related to compromised paneth cell antimicrobial host defense. Hepatology. 2012;55:1154-1163. DOI: 10.1002/hep.24789
  79. 79. Irvine KM, Ratnasekera I, Powell EE, et al. Causes and consequences of innate immune dysfunction in cirrhosis. Frontiers in Immunology. 2019;10:293. DOI: 10.3389/fimmu.2019.00293
  80. 80. Wiest R, Garcia-Tsao G. Bacterial translocation (BT) in cirrhosis. Hepatology. 2005;41:422-433. DOI: 10.1002/hep.20632
  81. 81. Riva A, Gray EH, Azarian S, et al. Faecal cytokine profiling as a marker of intestinal inflammation in acutely decompensated cirrhosis. JHEP Reports. 2020;2:100151. DOI: 10.1016/j.jhepr.2020.100151
  82. 82. Assimakopoulos SF, Tsamandas AC, Tsiaoussis GI, et al. Altered intestinal tight junctions' expression in patients with liver cirrhosis: A pathogenetic mechanism of intestinal hyperpermeability. European Journal of Clinical Investigation. 2012;42:439-446. DOI: 10.1111/j.1365-2362.2011.02609.x
  83. 83. Muñoz L, Albillos A, Nieto M, et al. Mesenteric Th1 polarization and monocyte TNF-α production: First steps to systemic inflammation in rats with cirrhosis. Hepatology. 2005;42:411-419
  84. 84. Munoz L, Jose Borrero M, Ubeda M, et al. Interaction between intestinal dendritic cells and bacteria translocated from the gut in rats with cirrhosis. Hepatology. 2012;56:1861-1869
  85. 85. Úbeda M, Munoz L, Borrero MJ, et al. Critical role of the liver in the induction of systemic inflammation in rats with preascitic cirrhosis. Hepatology. 2010;52:2086-2095
  86. 86. Munoz L, Borrero M-J, Ubeda M, et al. Commensal gut flora drives the expansion of proinflammatory T cells in the small intestinal mucosa in rats with CCl4 cirrhosis. In: Hepatology. NJ, USA: Wiley-Blackwell; 2013. p. 985A
  87. 87. Albillos A, Lario M, Álvarez-Mon M. Cirrhosis-associated immune dysfunction: Distinctive features and clinical relevance. Journal of Hepatology. 2014;61:1385-1396. DOI: 10.1016/j.jhep.2014.08.010
  88. 88. Iwasaki A, Medzhitov R. Toll-like receptor control of the adaptive immune responses. Nature Immunology. 2004;5:987-995
  89. 89. McClure R, Massari P. TLR-dependent human mucosal epithelial cell responses to microbial pathogens. Frontiers in Immunology. 2014;5:386
  90. 90. Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: Update on toll-like receptors. Nature Immunology. 2010;11:373-384
  91. 91. Kawasaki T, Kawai T. Toll-like receptor signaling pathways. Frontiers in Immunology. 2014;5:461
  92. 92. Fitzgerald KA, Kagan JC. Toll-like receptors and the control of immunity. Cell. 2020;180:1044-1066
  93. 93. Schnare M, Barton GM, Holt AC, et al. Toll-like receptors control activation of adaptive immune responses. Nature Immunology. 2001;2:947-950
  94. 94. Lan YT, Wang ZL, Tian P, et al. Treg/Th17 imbalance and its clinical significance in patients with hepatitis B-associated liver cirrhosis. Diagnostic Pathology. 2019;14:114. DOI: 10.1186/s13000-019-0891-4
  95. 95. Leung S, Liu X, Fang L, et al. The cytokine milieu in the interplay of pathogenic Th1/Th17 cells and regulatory T cells in autoimmune disease. Cellular & Molecular Immunology. 2010;7:182-189. DOI: 10.1038/cmi.2010.22
  96. 96. Lee J, Lozano-Ruiz B, Yang FM, et al. The multifaceted role of Th1, Th9, and Th17 cells in immune checkpoint inhibition therapy. Frontiers in Immunology. 2021;12. DOI: 10.3389/fimmu.2021.625667
  97. 97. Blaschitz C, Raffatellu M. Th17 cytokines and the gut mucosal barrier. Journal of Clinical Immunology. 2010;30:196-203. DOI: 10.1007/s10875-010-9368-7
  98. 98. Lee Jacob S, Tato Cristina M, Joyce-Shaikh B, et al. Interleukin-23-independent IL-17 production regulates intestinal epithelial permeability. Immunity. 2015;43:727-738. DOI: 10.1016/j.immuni.2015.09.003
  99. 99. Song X. Intestinal microbiota mining: A Th17/Treg cell perspective. European Journal of BioMedical Research. 2015;1:28. DOI: 10.18088/ejbmr.1.4.2015.pp28-35
  100. 100. Cheng N-C, Guan CW, et al. The Th17/Treg cell balance: A gut microbiota-modulated story. Microorganisms. 2019;7:583. DOI: 10.3390/microorganisms7120583
  101. 101. Shao L, Li M, Zhang B, et al. Bacterial dysbiosis incites Th17 cell revolt in irradiated gut. Biomedicine & Pharmacotherapy. 2020;131:110674. DOI: 10.1016/j.biopha.2020.110674
  102. 102. Pandiyan P, Bhaskaran N, Zou M, et al. Microbiome dependent regulation of Tregs and Th17 cells in mucosa. Frontiers in Immunology. 2019:10:426. DOI: 10.3389/fimmu.2019.00426
  103. 103. Kang L, Fang X, Song YH, et al. Neutrophil-epithelial crosstalk during intestinal inflammation. Cellular and Molecular Gastroenterology and Hepatology. 2022;14:1257-1267. DOI: 10.1016/j.jcmgh.2022.09.002
  104. 104. Kimura A, Kishimoto T. IL-6: Regulator of Treg/Th17 balance. European Journal of Immunology. 2010;40:1830-1835. DOI: 10.1002/eji.201040391
  105. 105. Iebba V, Guerrieri F, Di Gregorio V, et al. Combining amplicon sequencing and metabolomics in cirrhotic patients highlights distinctive microbiota features involved in bacterial translocation, systemic inflammation and hepatic encephalopathy. Scientific Reports. 2018;8:8210. DOI: 10.1038/s41598-018-26509-y
  106. 106. Manzhalii E, Virchenko O, Falalyeyeva T, et al. Hepatic encephalopathy aggravated by systemic inflammation. Digestive Diseases. 2019;37:509-517. DOI: 10.1159/000500717
  107. 107. Shawcross DL, Sharifi Y, Canavan JB, et al. Infection and systemic inflammation, not ammonia, are associated with grade 3/4 hepatic encephalopathy, but not mortality in cirrhosis. Journal of Hepatology. 2011;54:640-649. DOI: 10.1016/j.jhep.2010.07.045
  108. 108. Verma N, Dhiman RK, Choudhury A, et al. Dynamic assessments of hepatic encephalopathy and ammonia levels predict mortality in acute-on-chronic liver failure. Hepatology International. 2021;15:970-982. DOI: 10.1007/s12072-021-10221-7
  109. 109. Nielsen MC, Andersen MN, Rittig N, et al. The macrophage-related biomarkers sCD163 and sCD206 are released by different shedding mechanisms. Journal of Leukocyte Biology. 2019;106:1129-1138. DOI: 10.1002/JLB.3A1218-500R
  110. 110. Grønbæk H, Rødgaard-Hansen S, Aagaard NK, et al. Macrophage activation markers predict mortality in patients with liver cirrhosis without or with acute-on-chronic liver failure (ACLF). Journal of Hepatology. 2016;64:813-822. DOI: 10.1016/j.jhep.2015.11.021
  111. 111. Dixon LJ, Barnes M, Tang H, et al. Kupffer cells in the liver. Comprehensive Physiology. 2013;3:785
  112. 112. Ma Y-y, Yang M-q, He Z-g, et al. The Biological Function of Kupffer Cells in Liver Disease. London, UK, London, UK: IntechOpen; 2017
  113. 113. Gomez Perdiguero E, Klapproth K, Schulz C, et al. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature. 2015;518:547-551
  114. 114. Naito M, Hasegawa G, Ebe Y, et al. Differentiation and function of Kupffer cells. Medical Electron Microscopy. 2004;37:16-28
  115. 115. Scott CL, Zheng F, De Baetselier P, et al. Bone marrow-derived monocytes give rise to self-renewing and fully differentiated Kupffer cells. Nature Communications. 2016;7:10321. DOI: 10.1038/ncomms10321
  116. 116. Laskin DL, Sunil VR, Gardner CR, et al. Macrophages and tissue injury: Agents of defense or destruction? Annual Review of Pharmacology and Toxicology. 2011;51:267-288
  117. 117. Sica A, Mantovani A. Macrophage plasticity and polarization: In vivo veritas. The Journal of Clinical Investigation. 2012;122:787-795
  118. 118. Sato K, Hall C, Glaser S, et al. Pathogenesis of Kupffer cells in cholestatic liver injury. The American Journal of Pathology. 2016;186:2238-2247. DOI: 10.1016/j.ajpath.2016.06.003
  119. 119. Weston CJ, Zimmermann HW, Adams DH. The role of myeloid-derived cells in the progression of liver disease. Frontiers in Immunology. 2019;10:893. DOI: 10.3389/fimmu.2019.00893
  120. 120. An P, Wei LL, Zhao S, et al. Hepatocyte mitochondria-derived danger signals directly activate hepatic stellate cells and drive progression of liver fibrosis. Nature Communications. 2020;11:2362. DOI: 10.1038/s41467-020-16092-0
  121. 121. Méndez-Sánchez N, Valencia-Rodríguez A, Coronel-Castillo C, et al. The cellular pathways of liver fibrosis in non-alcoholic steatohepatitis. Annals of Translational Medicine. 2020;8:400
  122. 122. Yan Y, Zeng J, Xing L, et al. Extra- and intra-cellular mechanisms of hepatic stellate cell activation. Biomedicine. 2021;9:1014. DOI: 10.3390/biomedicines9081014
  123. 123. Weiskirchen R, Tacke F. Cellular and molecular functions of hepatic stellate cells in inflammatory responses and liver immunology. Hepatobiliary Surgery and Nutrition. 2014;3:344-363
  124. 124. Brass A, Brenndörfer ED. The role of chemokines in hepatitis C virus-mediated liver disease. International Journal of Molecular Sciences. 2014;15:4747-4779. DOI: 10.3390/ijms15034747
  125. 125. Nishikawa K, Osawa Y, Kimura K. Wnt/β-catenin signaling as a potential target for the treatment of liver cirrhosis using antifibrotic drugs. International Journal of Molecular Sciences. 2018;19:3103
  126. 126. Xu F, Liu C, Zhou D, et al. TGF-β/SMAD pathway and its regulation in hepatic fibrosis. Journal of Histochemistry & Cytochemistry. 2016;64:157-167
  127. 127. Inzaugarat ME, Johnson CD, Holtmann TM, et al. NLR family pyrin domain-containing 3 inflammasome activation in hepatic stellate cells induces liver fibrosis in mice. Hepatology. 2019;69:845-859. DOI: 10.1002/hep.30252
  128. 128. Gao B, Jeong W-I, Tian Z. Liver: An organ with predominant innate immunity. Hepatology. 2008;47:729-736
  129. 129. Su GL, Klein RD, Aminlari A, et al. Kupffer cell activation by lipopolysaccharide in rats: Role for lipopolysaccharide binding protein and toll-like receptor 4. Hepatology. 2000;31:932-936
  130. 130. Jia F, Deng F, Xu P, et al. NOD1 agonist protects against lipopolysaccharide and D-galactosamine-induced fatal hepatitis through the upregulation of A20 expression in hepatocytes. Frontiers in Immunology. 2021;12:603192. DOI: 10.3389/fimmu.2021.603192
  131. 131. Su GL. Lipopolysaccharides in liver injury: Molecular mechanisms of Kupffer cell activation. American Journal of Physiology-Gastrointestinal and Liver Physiology. 2002;283:G256-G265
  132. 132. Fisher JE, McKenzie TJ, Lillegard JB, et al. Role of Kupffer cells and toll-like receptor 4 in acetaminophen-induced acute liver failure. The Journal of Surgical Research. 2013;180:147-155. DOI: 10.1016/j.jss.2012.11.051
  133. 133. Seki E, Tsutsui H, Nakano H, et al. Lipopolysaccharide-induced IL-18 secretion from murine Kupffer cells independently of myeloid differentiation factor 88 that is critically involved in induction of production of IL-12 and IL-1β. The Journal of Immunology. 2001;166:2651-2657
  134. 134. Werner M, Schefczyk S, Trippler M, et al. Antiviral toll-like receptor signaling in non-parenchymal liver cells is restricted to TLR3. Viruses. 2022;14:218
  135. 135. van der Heide D, Weiskirchen R, Bansal R. Therapeutic targeting of hepatic macrophages for the treatment of liver diseases. Frontiers in Immunology. 2019;10:2852. DOI: 10.3389/fimmu.2019.02852
  136. 136. Faruqui A. Alcohol induced gut microbiota modulation: The role of probiotics, pufas, and vitamin E in management of alcoholic liver disease, Japanese. Journal of Gastroenterology and Hepatology. 2021;5:1-9. DOI: 10.47829/JJGH.2021.51304
  137. 137. Dong X, Liu J, Xu Y, et al. Role of macrophages in experimental liver injury and repair in mice (review). Experimental and Therapeutic Medicine. 2019;17:3835-3847. DOI: 10.3892/etm.2019.7450
  138. 138. Heath WR, Carbone FR. Dendritic cell subsets in primary and secondary T cell responses at body surfaces. Nature Immunology. 2009;10:1237-1244
  139. 139. Henri S, Guilliams M, Poulin LF, et al. Disentangling the complexity of the skin dendritic cell network. Immunology & Cell Biology. 2010;88:366-375. DOI: 10.1038/icb.2010.34
  140. 140. Guilliams M, Henri S, Tamoutounour S, et al. From skin dendritic cells to a simplified classification of human and mouse dendritic cell subsets. European Journal of Immunology. 2010;40:2089-2094. DOI: 10.1002/eji.201040498
  141. 141. Manh TP, Alexandre Y, Baranek T, et al. Plasmacytoid, conventional, and monocyte-derived dendritic cells undergo a profound and convergent genetic reprogramming during their maturation. European Journal of Immunology. 2013;43:1706-1715. DOI: 10.1002/eji.201243106
  142. 142. Zhang Z, Zou Z-S, Fu J-L, et al. Severe dendritic cell perturbation is actively involved in the pathogenesis of acute-on-chronic hepatitis B liver failure. Journal of Hepatology. 2008;49:396-406. DOI: 10.1016/j.jhep.2008.05.017
  143. 143. Khanam A, Trehanpati N, Garg V, et al. Altered frequencies of dendritic cells and IFN-γ-secreting T cells with granulocyte colony-stimulating factor (G-CSF) therapy in acute-on-chronic liver failure. Liver International. 2014;34:505-513
  144. 144. Edwards AD, Diebold SS, Slack EM, et al. Toll-like receptor expression in murine DC subsets: Lack of TLR7 expression by CD8 alpha+ DC correlates with unresponsiveness to imidazoquinolines. European Journal of Immunology. 2003;33:827-833. DOI: 10.1002/eji.200323797
  145. 145. Cervantes JL, Weinerman B, Basole C, et al. TLR8: The forgotten relative revindicated. Cellular & Molecular Immunology. 2012;9:434-438. DOI: 10.1038/cmi.2012.38
  146. 146. Sundar R, Cho B-C, Brahmer J, et al. Nivolumab in NSCLC: Latest evidence and clinical potential. Therapeutic Advances in Medical Oncology. 2015;7:85-96. DOI: 10.1177/1758834014567470
  147. 147. Helft J, Ginhoux F, Bogunovic M, et al. Origin and functional heterogeneity of non-lymphoid tissue dendritic cells in mice. Immunological Reviews. 2010;234:55-75
  148. 148. Kumar J, Habib N, Huang K-W, et al. Recent advances: The imbalance of immune cells and cytokines in the pathogenesis of hepatocellular carcinoma. Diagnostics (Basel). 2020;10:338. DOI: 10.3390/diagnostics10050338
  149. 149. Sutti S, Bruzzì S, Heymann F, et al. CX3CR1 mediates the development of monocyte-derived dendritic cells during hepatic inflammation. Cell. 2019;8:1099. DOI: 10.3390/cells8091099
  150. 150. Chang Q, Wang Y-K, Zhao Q, et al. Th17 cells are increased with severity of liver inflammation in patients with chronic hepatitis C. Journal of Gastroenterology and Hepatology. 2012;27:273-278. DOI: 10.1111/j.1440-1746.2011.06782.x
  151. 151. Sun HQ, Zhang JY, Zhang H, et al. Increased Th17 cells contribute to disease progression in patients with HBV-associated liver cirrhosis. Journal of Viral Hepatitis. 2012;19:396-403. DOI: 10.1111/j.1365-2893.2011.01561.x
  152. 152. Mou H, Wu S, Zhao G, et al. Changes of Th17/Treg ratio in the transition of chronic hepatitis B to liver cirrhosis and correlations with liver function and inflammation. Experimental and Therapeutic Medicine. 2019;17:2963-2968. DOI: 10.3892/etm.2019.7299
  153. 153. Zepeda-Morales AS, Del Toro-Arreola S, García-Benavides L, et al. Liver fibrosis in bile duct-ligated rats correlates with increased hepatic IL-17 and TGF-β2 expression. Annals of Hepatology. 2016;15:418-426. DOI: 10.5604/16652681.1198820
  154. 154. Luckey SW, Petersen DR. Activation of Kupffer cells during the course of carbon tetrachloride-induced liver injury and fibrosis in rats. Experimental and Molecular Pathology. 2001;71:226-240. DOI: 10.1006/exmp.2001.2399
  155. 155. Aoyama T, Inokuchi S, Brenner DA, et al. CX3CL1-CX3CR1 interaction prevents carbon tetrachloride-induced liver inflammation and fibrosis in mice. Hepatology. 2010;52:1390-1400. DOI: 10.1002/hep.23795
  156. 156. Dhanda S, Gupta S, Halder A, et al. Systemic inflammation without gliosis mediates cognitive deficits through impaired BDNF expression in bile duct ligation model of hepatic encephalopathy. Brain, Behavior, and Immunity. 2018;70:214-232. DOI: 10.1016/j.bbi.2018.03.002
  157. 157. Tung HC, Lee FY, Wang SS, et al. The beneficial effects of P2X7 antagonism in rats with bile duct ligation-induced cirrhosis. PLoS One. 2015;10:e0124654. DOI: 10.1371/journal.pone.0124654
  158. 158. Kang H, Koppula S. Hepatoprotective effect of houttuynia cordata thunb extract against carbon tetrachloride-induced hepatic damage in mice. Indian Journal of Pharmaceutical Sciences. 2014;76:267-273
  159. 159. Moles A, Murphy L, Wilson CL, et al. A TLR2/S100A9/CXCL-2 signaling network is necessary for neutrophil recruitment in acute and chronic liver injury in the mouse. Journal of Hepatology. 2014;60:782-791. DOI: 10.1016/j.jhep.2013.12.005
  160. 160. Altaş S, Kızıl G, Kızıl M, et al. Protective effect of Diyarbakır watermelon juice on carbon tetrachloride-induced toxicity in rats. Food and Chemical Toxicology. 2011;49:2433-2438. DOI: 10.1016/j.fct.2011.06.064
  161. 161. Tag CG, Sauer-Lehnen S, Weiskirchen S, et al. Bile duct ligation in mice: Induction of inflammatory liver injury and fibrosis by obstructive cholestasis. Journal of Visualized Experiments. 2015;96:52438. DOI: 10.3791/52438
  162. 162. Canbay A, Feldstein A, Baskin-Bey E, et al. The caspase inhibitor IDN-6556 attenuates hepatic injury and fibrosis in the bile duct ligated mouse. Journal of Pharmacology and Experimental Therapeutics. 2004;308:1191-1196. DOI: 10.1124/jpet.103.060129
  163. 163. Trebicka J, Hennenberg M, Odenthal M, et al. Atorvastatin attenuates hepatic fibrosis in rats after bile duct ligation via decreased turnover of hepatic stellate cells. Journal of Hepatology. 2010;53:702-712. DOI: 10.1016/j.jhep.2010.04.025
  164. 164. Gobejishvili L, Barve S, Breitkopf-Heinlein K, et al. Rolipram attenuates bile duct ligation–induced liver injury in rats: A potential pathogenic role of PDE4. Journal of Pharmacology and Experimental Therapeutics. 2013;347:80-90. DOI: 10.1124/jpet.113.204933
  165. 165. Weng Z, Chi Y, Xie J, et al. Anti-inflammatory activity of dehydroandrographolide by TLR4/NF-κB signaling pathway inhibition in bile duct-ligated mice. Cellular Physiology and Biochemistry. 2018;49:1124-1137. DOI: 10.1159/000493292
  166. 166. Gehring S, Dickson EM, San Martin ME, et al. Kupffer cells abrogate cholestatic liver injury in mice. Gastroenterology. 2006;130:810-822. DOI: 10.1053/j.gastro.2005.11.015
  167. 167. Alric L, Orfila C, Carrere N, et al. Reactive oxygen intermediates and eicosanoid production by Kupffer cells and infiltrated macrophages in acute and chronic liver injury induced in rats by CCl4. Inflammation Research. 2000;49:700-707. DOI: 10.1007/s000110050649
  168. 168. Nagano T, Yamamoto K, Matsumoto S, et al. Cytokine profile in the liver of primary biliary cirrhosis. Journal of Clinical Immunology. 1999;19:422-427. DOI: 10.1023/A:1020511002025
  169. 169. Holland-Fischer P, Grønbæk H, Sandahl TD, et al. Kupffer cells are activated in cirrhotic portal hypertension and not normalised by TIPS. Gut. 2011;60:1389-1393. DOI: 10.1136/gut.2010.234542
  170. 170. de Lalla C, Galli G, Aldrighetti L, et al. Production of profibrotic cytokines by invariant NKT cells characterizes cirrhosis progression in chronic viral Hepatitis1. The Journal of Immunology. 2004;173:1417-1425. DOI: 10.4049/jimmunol.173.2.1417
  171. 171. Napoli J, Bishop GA, McGuinness PH, et al. Progressive liver injury in chronic hepatitis C infection correlates with increased intrahepatic expression of Th1-associated cytokines. Hepatology. 1996;24:759-765. DOI: 10.1002/hep.510240402
  172. 172. Farinati F, Cardin R, Bortolami M, et al. Oxidative damage, pro-inflammatory cytokines, TGF-alpha and c-myc in chronic HCV-related hepatitis and cirrhosis. World Journal of Gastroenterology. 2006;12:2065-2069. DOI: 10.3748/wjg.v12.i13.2065
  173. 173. Li H, Xia Q, Zeng B, et al. Submassive hepatic necrosis distinguishes HBV-associated acute on chronic liver failure from cirrhotic patients with acute decompensation. Journal of Hepatology. 2015;63:50-59. DOI: 10.1016/j.jhep.2015.01.029
  174. 174. Chuang Y-H, Lian Z-X, Tsuneyama K, et al. Increased killing activity and decreased cytokine production in NK cells in patients with primary biliary cirrhosis. Journal of Autoimmunity. 2006;26:232-240. DOI: 10.1016/j.jaut.2006.04.001
  175. 175. Schafer DP, Stevens B. Microglia function in central nervous system development and plasticity. Cold Spring Harbor Perspectives in Biology. 2015;7:a020545. DOI: 10.1101/cshperspect.a020545
  176. 176. Fujita Y, Yamashita T. Neuroprotective function of microglia in the developing brain. Neuronal Signaling. 2021;5:Ns20200024. DOI: 10.1042/ns20200024
  177. 177. Wolf SA, Boddeke HW, Kettenmann H. Microglia in physiology and disease. Annual Review of Physiology. 2017;79:619-643. DOI: 10.1146/annurev-physiol-022516-034406
  178. 178. Wlodarczyk A, Løbner M, Cédile O, et al. Comparison of microglia and infiltrating CD11c+ cells as antigen presenting cells for T cell proliferation and cytokine response. Journal of Neuroinflammation. 2014;11:1-9
  179. 179. Borst K, Dumas AA, Prinz M. Microglia: Immune and non-immune functions. Immunity. 2021;54:2194-2208
  180. 180. Hsieh CL, Koike M, Spusta SC, et al. A role for TREM2 ligands in the phagocytosis of apoptotic neuronal cells by microglia. Journal of Neurochemistry. 2009;109:1144-1156
  181. 181. Takahashi K, Rochford CD, Neumann H. Clearance of apoptotic neurons without inflammation by microglial triggering receptor expressed on myeloid cells-2. The Journal of Experimental Medicine. 2005;201:647-657
  182. 182. Wu D, Chen Q, Chen X, et al. The blood–brain barrier: Structure, regulation, and drug delivery. Signal Transduction and Targeted Therapy. 2023;8:217. DOI: 10.1038/s41392-023-01481-w
  183. 183. Lochhead JJ, Yang J, Ronaldson PT, et al. Structure, function, and regulation of the blood-brain barrier tight junction in central nervous system disorders. Frontiers in Physiology. 2020;11:914. DOI: 10.3389/fphys.2020.00914
  184. 184. Farooq RK, Alamoudi W, Alhibshi A, et al. Varied composition and underlying mechanisms of gut microbiome in neuroinflammation. Microorganisms. 2022;10:705
  185. 185. Buttgereit A, Lelios I, Yu X, et al. Sall1 is a transcriptional regulator defining microglia identity and function. Nature Immunology. 2016;17:1397-1406
  186. 186. Li K, Tan Y-H, Light AR, et al. Different peripheral tissue injury induces differential phenotypic changes of spinal activated microglia. Clinical and Developmental Immunology. 2013;2013:901420
  187. 187. Brockie S, Hong J, Fehlings MG. The role of microglia in modulating neuroinflammation after spinal cord injury. International Journal of Molecular Sciences. 2021;22:9706
  188. 188. Ohsawa K, Imai Y, Sasaki Y, et al. Microglia/macrophage-specific protein Iba1 binds to fimbrin and enhances its actin-bundling activity. Journal of Neurochemistry. 2004;88:844-856. DOI: 10.1046/j.1471-4159.2003.02213.x
  189. 189. Fadini GP, Cappellari R, Mazzucato M, et al. Monocyte–macrophage polarization balance in pre-diabetic individuals. Acta Diabetologica. 2013;50:977-982
  190. 190. Jones BA, Beamer M, Ahmed S. Fractalkine/CX3CL1: A potential new target for inflammatory diseases. Molecular Interventions. 2010;10:263
  191. 191. Jiang SX, Slinn J, Aylsworth A, et al. Vimentin participates in microglia activation and neurotoxicity in cerebral ischemia. Journal of Neurochemistry. 2012;122:764-774. DOI: 10.1111/j.1471-4159.2012.07823.x
  192. 192. Masuda T, Amann L, Sankowski R, et al. Novel Hexb-based tools for studying microglia in the CNS. Nature Immunology. 2020;21:802-815. DOI: 10.1038/s41590-020-0707-4
  193. 193. Leyh J, Paeschke S, Mages B, et al. Classification of microglial morphological phenotypes using machine learning. Frontiers in Cellular Neuroscience. 2021;15:701673. DOI: 10.3389/fncel.2021.701673
  194. 194. Vidal-Itriago A, Radford RAW, Aramideh JA, et al. Microglia morphophysiological diversity and its implications for the CNS. Frontiers in Immunology. 2022;13:997786. DOI: 10.3389/fimmu.2022.997786
  195. 195. Davalos D, Grutzendler J, Yang G, et al. ATP mediates rapid microglial response to local brain injury in vivo. Nature Neuroscience. 2005;8:752-758. DOI: 10.1038/nn1472
  196. 196. Das Sarma S, Chatterjee K, Dinda H, et al. Cytomorphological and cytochemical identification of microglia. International Scholarly Research Notices. 2013;2013:205431
  197. 197. Borch J, Haslund-Vinding J, Vilhardt F, et al. Meningioma–brain crosstalk: A scoping review. Cancers. 2021;13:4267. DOI: 10.3390/cancers13174267
  198. 198. Jurga AM, Paleczna M, Kuter KZ. Overview of general and discriminating markers of differential microglia phenotypes. Frontiers in Cellular Neuroscience. 2020;14:198. DOI: 10.3389/fncel.2020.00198
  199. 199. Rodrigo R, Cauli O, Gomez–Pinedo U et al. Hyperammonemia induces neuroinflammation that contributes to cognitive impairment in rats with hepatic encephalopathy, Gastroenterology 2010;139:675-684. doi:10.1053/j.gastro.2010.03.040.
  200. 200. Tamnanloo F, Ochoa-Sanchez R, Oliveira MM, et al. Multiple ammonia-induced episodes of hepatic encephalopathy provoke neuronal cell loss in bile-duct ligated rats. JHEP Reports. 2023;5:100904. DOI: 10.1016/j.jhepr.2023.100904
  201. 201. Bosoi CR, Yang X, Huynh J, et al. Systemic oxidative stress is implicated in the pathogenesis of brain edema in rats with chronic liver failure. Free Radical Biology and Medicine. 2012;52:1228-1235. DOI: 10.1016/j.freeradbiomed.2012.01.006
  202. 202. Magen I, Avraham Y, Ackerman Z, et al. Cannabidiol ameliorates cognitive and motor impairments in mice with bile duct ligation. Journal of Hepatology. 2009;51:528-534. DOI: 10.1016/j.jhep.2009.04.021
  203. 203. Clément M-A, Bosoi CR, Oliveira MM, et al. Bile-duct ligation renders the brain susceptible to hypotension-induced neuronal degeneration: Implications of ammonia. Journal of Neurochemistry. 2021;157:561-573. DOI: 10.1111/jnc.15290
  204. 204. Stojanović NM, Maslovarić A, Mihajlović I, et al. Melatonin treatment prevents carbon-tetrachloride induced rat brain injury. Toxicology Research. 2023;12(5):895-901. DOI: 10.1093/toxres/tfad083
  205. 205. Dhanda S, Sandhir R. Blood-brain barrier permeability is exacerbated in experimental model of hepatic encephalopathy via MMP-9 activation and downregulation of tight junction proteins. Molecular Neurobiology. 2018;55:3642-3659. DOI: 10.1007/s12035-017-0521-7
  206. 206. Yang N, Liu H, Jiang Y, et al. Lactulose enhances neuroplasticity to improve cognitive function in early hepatic encephalopathy. Neural Regeneration Research. 2015;10:1457-1462. DOI: 10.4103/1673-5374.165516
  207. 207. Golshani M, Basiri M, Shabani M, et al. Effects of erythropoietin on bile duct ligation-induced neuro-inflammation in male rats. AIMS Neuroscience. 2019;6:43-53. DOI: 10.3934/Neuroscience.2019.2.43
  208. 208. Hsu S-J, Zhang C, Jeong J, et al. Enhanced meningeal lymphatic drainage ameliorates neuroinflammation and hepatic encephalopathy in cirrhotic rats. Gastroenterology. 2021;160:1315-1329.e1313. DOI: 10.1053/j.gastro.2020.11.036
  209. 209. Chen J-R, Wang B-N, Tseng G-F, et al. Morphological changes of cortical pyramidal neurons in hepatic encephalopathy. BMC Neuroscience. 2014;15:15. DOI: 10.1186/1471-2202-15-15
  210. 210. Chen Y-C, Sheen J-M, Tain Y-L, et al. Alterations in NADPH oxidase expression and blood–brain barrier in bile duct ligation-treated young rats: Effects of melatonin. Neurochemistry International. 2012;60:751-758. DOI: 10.1016/j.neuint.2012.03.021
  211. 211. Pierzchala K, Simicic D, Sienkiewicz A, et al. Central nervous system and systemic oxidative stress interplay with inflammation in a bile duct ligation rat model of type C hepatic encephalopathy. Free Radical Biology and Medicine. 2022;178:295-307. DOI: 10.1016/j.freeradbiomed.2021.12.011
  212. 212. Ahmadi S, Khaledi S. Anxiety in rats with bile duct ligation is associated with activation of JNK3 mitogen-activated protein kinase in the hippocampus. Metabolic Brain Disease. 2020;35:579-588. DOI: 10.1007/s11011-020-00542-1
  213. 213. Magen I, Avraham Y, Ackerman Z, et al. Cannabidiol ameliorates cognitive and motor impairments in bile-duct ligated mice via 5-HT1A receptor activation. British Journal of Pharmacology. 2010;159:950-957. DOI: 10.1111/j.1476-5381.2009.00589.x
  214. 214. Gee LMV, Barron-Millar B, Leslie J, et al. Anti–cholestatic therapy with obeticholic acid improves short-term memory in bile duct–ligated mice. The American Journal of Pathology. 2023;193:11-26. DOI: 10.1016/j.ajpath.2022.09.005
  215. 215. Shabani M, Ebrahimpoor F, Firouzjaei MA, et al. Modulation of sphingosine-1-phosphate receptor by FTY720 contributes in improvement of hepatic encephalopathy induced by bile duct ligation. Brain Research Bulletin. 2019;146:253-269. DOI: 10.1016/j.brainresbull.2019.01.012
  216. 216. Balasubramaniyan V, Wright G, Sharma V, et al. Ammonia reduction with ornithine phenylacetate restores brain eNOS activity via the DDAH-ADMA pathway in bile duct-ligated cirrhotic rats. American Journal of Physiology-Gastrointestinal and Liver Physiology. 2012;302:G145-G152. DOI: 10.1152/ajpgi.00097.2011
  217. 217. Wright G, Wright G, Newman T, et al. OP08 evidence for early astrocyte activation, cellular stress and compensatory microglial related transforming growth factor-α responses in bile-duct ligated rats. Gut. 2010;59:A3-A4. DOI: 10.1136/gut.2010.223362.8
  218. 218. Faropoulos K, Chroni E, Assimakopoulos SF, et al. Altered occludin expression in brain capillaries induced by obstructive jaundice in rats. Brain Research. 2010;1325:121-127. DOI: 10.1016/j.brainres.2010.02.020
  219. 219. Boer LA, Panatto JP, Fagundes DA, et al. Inhibition of mitochondrial respiratory chain in the brain of rats after hepatic failure induced by carbon tetrachloride is reversed by antioxidants. Brain Research Bulletin. 2009;80:75-78. DOI: 10.1016/j.brainresbull.2009.04.009
  220. 220. Khan A, Shal B, Naveed M, et al. Matrine ameliorates anxiety and depression-like behaviour by targeting hyperammonemia-induced neuroinflammation and oxidative stress in CCl4 model of liver injury. Neurotoxicology. 2019;72:38-50. DOI: 10.1016/j.neuro.2019.02.002
  221. 221. de Souza MF, Marinho JP, Abujamra AL, et al. Carbon tetrachloride increases the pro-inflammatory cytokines levels in different brain areas of Wistar rats: The protective effect of acai frozen pulp. Neurochemical Research. 2015;40:1976-1983. DOI: 10.1007/s11064-015-1693-z
  222. 222. Hadjihambi A, Harrison IF, Costas-Rodríguez M, et al. Impaired brain glymphatic flow in experimental hepatic encephalopathy. Journal of Hepatology. 2019;70:40-49. DOI: 10.1016/j.jhep.2018.08.021
  223. 223. Zemtsova I, Görg B, Keitel V, et al. Microglia activation in hepatic encephalopathy in rats and humans. Hepatology. 2011;54:204-215. DOI: 10.1002/hep.24326
  224. 224. Görg B, Bidmon H-J, Häussinger D. Gene expression profiling in the cerebral cortex of patients with cirrhosis with and without hepatic encephalopathy. Hepatology. 2013;57:2436-2447
  225. 225. Grover VPB, Pavese N, Koh S-B, et al. Cerebral microglial activation in patients with hepatitis C: In vivo evidence of neuroinflammation. Journal of Viral Hepatitis. 2012;19:e89-e96. DOI: 10.1111/j.1365-2893.2011.01510.x
  226. 226. Balzano T, Forteza J, Molina P, et al. The cerebellum of patients with steatohepatitis shows lymphocyte infiltration, microglial activation and loss of Purkinje and granular neurons. Scientific Reports. 2018;8:3004. DOI: 10.1038/s41598-018-21399-6
  227. 227. Cagnin A, Taylor-Robinson SD, Forton DM, et al. In vivo imaging of cerebral “peripheral benzodiazepine binding sites” in patients with hepatic encephalopathy. Gut. 2006;55:547. DOI: 10.1136/gut.2005.075051
  228. 228. Butterworth RF, Lavoie J, Giguère J-F, et al. Affinities and densities of high-affinity [3H]muscimol (GABA-A) binding sites and of central benzodiazepine receptors are unchanged in autopsied brain tissue from cirrhotic patients with hepatic encephalopathy. Hepatology. 1988;8:1084-1088. DOI: 10.1002/hep.1840080517
  229. 229. Milewski K, Orzeł-Gajowik K, Zielińska M. Mitochondrial changes in rat brain endothelial cells associated with hepatic encephalopathy: Relation to the blood–brain barrier dysfunction. Neurochemical Research. 2022. pp.1-16. DOI: 10.1007/s11064-022-03698-7
  230. 230. Claeys W, Van Hoecke L, Lefere S, et al. The neurogliovascular unit in hepatic encephalopathy. JHEP Reports. 2021;3:100352. DOI: 10.1016/j.jhepr.2021.100352
  231. 231. Linnerbauer M, Wheeler MA, Quintana FJ. Astrocyte crosstalk in CNS inflammation. Neuron. 2020;108:608-622
  232. 232. Ede RJ, Zaki AEO, Silk DBA, et al. Experimental studies of blood-brain barrier permeability in chronic hepatic encephalopathy. In: Advances in Hepatic Encephalopathy and Urea Cycle Diseases: 5th International Symposium on Ammonia, Semmering, Austria, Basel, Switzerland. May 1984: Proceedings. S. Karger AG; 1985
  233. 233. Vairappan B, Sundhar M, Srinivas BH. Resveratrol restores neuronal tight junction proteins through correction of ammonia and inflammation in CCl(4)-induced cirrhotic mice. Molecular Neurobiology. 2019;56:4718-4729. DOI: 10.1007/s12035-018-1389-x
  234. 234. Baek SY, Lee EH, Oh TW, et al. Network pharmacology-based approaches of rheum undulatum linne and glycyrriza uralensis fischer imply their regulation of liver failure with hepatic encephalopathy in mice. Biomolecules. 2020;10:437
  235. 235. Quinn M, McMillin M, Galindo C, et al. Bile acids permeabilize the blood brain barrier after bile duct ligation in rats via Rac1-dependent mechanisms. Digestive and Liver Disease. 2014;46:527-534. DOI: 10.1016/j.dld.2014.01.159
  236. 236. Ott P, Larsen FS. Blood–brain barrier permeability to ammonia in liver failure: A critical reappraisal. Neurochemistry International. 2004;44:185-198. DOI: 10.1016/S0197-0186(03)00153-0
  237. 237. Chesler M. Regulation and modulation of pH in the brain. Physiological Reviews. 2003;83:1183-1221
  238. 238. Hertz L, Peng L, Song D. Ammonia, like K+, stimulates the Na+, K+, 2 Cl− cotransporter NKCC1 and the Na+, K+-ATPase and interacts with endogenous ouabain in astrocytes. Neurochemical Research. 2015;40:241-257
  239. 239. Larsen EH, Deaton LE, Onken H, et al. Osmoregulation and excretion. Comprehensive Physiology. 2011;4:405-573
  240. 240. Adlimoghaddam A, Ml B, Marini A-M, et al. Ammonia excretion in Caenorhabditis elegans: Mechanism and evidence of ammonia transport of the rhesus protein CeRhr-1. The Journal of Experimental Biology. 2015;218:675-683
  241. 241. Watkins LR, Maier SF, Goehler LE. Cytokine-to-brain communication: A review & analysis of alternative mechanisms. Life Sciences. 1995;57:1011-1026. DOI: 10.1016/0024-3205(95)02047-m
  242. 242. D'Mello C, Swain MG. Immune-to-brain communication pathways in inflammation-associated sickness and depression. Current Topics in Behavioral Neurosciences. 2017;31:73-94. DOI: 10.1007/7854_2016_37
  243. 243. Dantzer R, O'connor JC, Freund GG, et al. From inflammation to sickness and depression: When the immune system subjugates the brain. Nature Reviews Neuroscience. 2008;9:46-56
  244. 244. Ganong WF. Circumventricular organs: Definition and role in the regulation of endocrine and autonomic function. Clinical and Experimental Pharmacology & Physiology. 2000;27:422-427. DOI: 10.1046/j.1440-1681.2000.03259.x
  245. 245. Jaeger V, DeMorrow S, McMillin M. The direct contribution of astrocytes and microglia to the pathogenesis of hepatic encephalopathy. Journal of Clinical and Translational Hepatology. 2019;7:352-361. DOI: 10.14218/jcth.2019.00025
  246. 246. Garden GA, Möller T. Microglia biology in health and disease. Journal of Neuroimmune Pharmacology. 2006;1:127-137
  247. 247. Butterworth RF. Hepatic encephalopathy: A central neuroinflammatory disorder? Hepatology. 2011;53:1372-1376
  248. 248. Wright GA, Sharifi Y, Newman TA, et al. Characterisation of temporal microglia and astrocyte immune responses in bile duct-ligated rat models of cirrhosis. Liver International. 2014;34:1184-1191
  249. 249. Rao KVR, Brahmbhatt M, Norenberg MD. Microglia contribute to ammonia-induced astrocyte swelling in culture. Metabolic Brain Disease. 2013;28:139-143
  250. 250. Karababa A, Groos-Sahr K, Albrecht U, et al. Ammonia attenuates LPS-induced upregulation of pro-inflammatory cytokine mRNA in co-cultured astrocytes and microglia. Neurochemical Research. 2017;42:737-749
  251. 251. Fukushima S, Furube E, Itoh M, et al. Robust increase of microglia proliferation in the fornix of hippocampal axonal pathway after a single LPS stimulation. Journal of Neuroimmunology. 2015;285:31-40. DOI: 10.1016/j.jneuroim.2015.05.014
  252. 252. Li Y, Yin L, Fan Z, et al. Microglia: A potential therapeutic target for sepsis-associated encephalopathy and sepsis-associated chronic pain. Frontiers in Pharmacology. 2020;11:600421. DOI: 10.3389/fphar.2020.600421
  253. 253. Zargar S, Wani TA. Protective role of quercetin in carbon tetrachloride induced toxicity in rat brain: Biochemical, spectrophotometric assays and computational approach. Molecules. 2021;26:7526. DOI: 10.3390/molecules26247526
  254. 254. Kim E, Cho S. Microglia and monocyte-derived macrophages in stroke. Neurotherapeutics. 2016;13:702-718. DOI: 10.1007/s13311-016-0463-1
  255. 255. Somebang K, Rudolph J, Imhof I, et al. CCR2 deficiency alters activation of microglia subsets in traumatic brain injury. Cell Reports. 2021;36:109727. DOI: 10.1016/j.celrep.2021.109727
  256. 256. D'Mello C, Le T, Swain MG. Cerebral microglia recruit monocytes into the brain in response to tumor necrosis factorα signaling during peripheral organ inflammation. The Journal of Neuroscience. 2009;29:2089-2102. DOI: 10.1523/jneurosci.3567-08.2009
  257. 257. Carlos P-M, Aldo T-D. Astrocyte pathophysiology in liver disease. In: Maria Teresa G, Luca Colucci DA, editors. Astrocyte. Rijeka: IntechOpen; 2017, Ch. 10
  258. 258. Han W, Zhang H, Han Y, et al. Cognition-tracking-based strategies for diagnosis and treatment of minimal hepatic encephalopathy. Metabolic Brain Disease. 2020;35:869-881. DOI: 10.1007/s11011-020-00539-w
  259. 259. Chen Z, Ruan J, Li D, et al. The role of intestinal bacteria and gut–brain axis in hepatic encephalopathy. Frontiers in Cellular and Infection Microbiology. 2021;10:595759. DOI: 10.3389/fcimb.2020.595759
  260. 260. Sorge RE, Mapplebeck JC, Rosen S, et al. Different immune cells mediate mechanical pain hypersensitivity in male and female mice. Nature Neuroscience. 2015;18:1081-1083
  261. 261. Elsherbini DMA, Ghoneim FM, El-Mancy EM, et al. Astrocytes profiling in acute hepatic encephalopathy: Possible enrolling of glial fibrillary acidic protein, tumor necrosis factor-alpha, inwardly rectifying potassium channel (Kir 4.1) and aquaporin-4 in rat cerebral cortex. Frontiers in Cellular Neuroscience. 2022;16:896172. DOI: 10.3389/fncel.2022.896172
  262. 262. Córdoba J, Alonso J, Rovira A, et al. The development of low-grade cerebral edema in cirrhosis is supported by the evolution of (1)H-magnetic resonance abnormalities after liver transplantation. Journal of Hepatology. 2001;35:598-604. DOI: 10.1016/s0168-8278(01)00181-7
  263. 263. Jover R, Rodrigo R, Felipo V, et al. Brain edema and inflammatory activation in bile duct ligated rats with diet-induced hyperammonemia: A model of hepatic encephalopathy in cirrhosis. Hepatology. 2006;43:1257-1266. DOI: 10.1002/hep.21180
  264. 264. Sepehrinezhad A, Stolze Larsen F, Ashayeri Ahmadabad R, et al. The glymphatic system may play a vital role in the pathogenesis of hepatic encephalopathy: A narrative review. Cells. 2023;12:979. DOI: 10.3390/cells12070979

Written By

Ali Sepehrinezhad and Ali Shahbazi

Submitted: 16 October 2023 Reviewed: 15 November 2023 Published: 15 January 2024