Next Article in Journal
Granulocytic MDSC with Deficient CCR5 Alleviates Lipogenesis and Inflammation in Nonalcoholic Fatty Liver Disease
Previous Article in Journal
Optimization and Application of CRISPR/Cas9 Genome Editing in a Cosmopolitan Pest, Diamondback Moth
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Linking Nonalcoholic Fatty Liver Disease and Brain Disease: Focusing on Bile Acid Signaling

1
School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
2
School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
3
Dongzhimen Hospital, Beijing University of Traditional Chinese Medicine, Beijing 100700, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(21), 13045; https://doi.org/10.3390/ijms232113045
Submission received: 14 September 2022 / Revised: 21 October 2022 / Accepted: 25 October 2022 / Published: 27 October 2022
(This article belongs to the Section Molecular Endocrinology and Metabolism)

Abstract

:
A metabolic illness known as non-alcoholic fatty liver disease (NAFLD), affects more than one-quarter of the world’s population. Bile acids (BAs), as detergents involved in lipid digestion, show an abnormal metabolism in patients with NAFLD. However, BAs can affect other organs as well, such as the brain, where it has a neuroprotective effect. According to a series of studies, brain disorders may be extrahepatic manifestations of NAFLD, such as depression, changes to the cerebrovascular system, and worsening cognitive ability. Consequently, we propose that NAFLD affects the development of brain disease, through the bile acid signaling pathway. Through direct or indirect channels, BAs can send messages to the brain. Some BAs may operate directly on the central Farnesoid X receptor (FXR) and the G protein bile acid-activated receptor 1 (GPBAR1) by overcoming the blood–brain barrier (BBB). Furthermore, glucagon-like peptide-1 (GLP-1) and the fibroblast growth factor (FGF) 19 are released from the intestine FXR and GPBAR1 receptors, upon activation, both of which send signals to the brain. Inflammatory, systemic metabolic disorders in the liver and brain are regulated by the bile acid-activated receptors FXR and GPBAR1, which are potential therapeutic targets. From a bile acid viewpoint, we examine the bile acid signaling changes in NAFLD and brain disease. We also recommend the development of dual GPBAR1/FXR ligands to reduce side effects and manage NAFLD and brain disease efficiently.

1. Introduction

One-quarter of the world’s population is now affected by NAFLD, the most widespread cause of chronic liver disease [1], which is known to be intimately related to the metabolic syndrome’s elements [2]. A more serious stage of NAFLD, non-alcoholic steatohepatitis (NASH), can lead to advanced liver disease, cirrhosis, and hepatocellular carcinoma [3]. The primary driver of NAFLD is overnutrition, and the pathogenic pathways of NAFLD are influenced by multiple metabolic, genetic, and microbiome-related factors [4]. Although the main long-term effect of the metabolic syndrome is thought to be cardiovascular disease, other organs, including the brain, may also be affected [5]. In addition to an elevated risk of stroke [6], NAFLD patients may also have modest or early cognitive impairment [7]. NAFLD is also an independent risk factor for depression [8], which affects patients’ quality of life [9].
As multipurpose signaling molecules, BAs have the capacity to control lipid, glucose, and energy metabolism [10,11], and their aberrant metabolism is a significant contributor to NAFLD. It is possible that bile acids can govern the brain because bile acid receptors are expressed in the brain as well [12,13]. As a result, we suggest that NAFLD influences the bile acid signaling system, which in turn influences the emergence of brain illness. The importance of bile acids in neurological illnesses has been mentioned in a number of recent reviews, but they have not explored, in detail, how the bile acid signaling pathways connect NAFLD and brain problems [14,15,16]. We highlight brain disease in this review as an extrahepatic manifestation of NAFLD, linking the two conditions via the bile acid signaling system.

2. Bile Acid of Liver Damage in NAFLD

In hepatocytes, the neutral/classic pathway is largely used to make BAs, whereas the acidic/alternative pathway is used in a smaller number of cases. Cholesterol 7a-hydroxylase (CYP7A1) is mostly rate-limiting and controls the classical route. The alternative method uses cytochrome P450 27A1 (CYP27A1) to transform cholesterol into (25R)-26-hydroxycholesterol [17]. The major Bas: cholic acid (CA) and chenodeoxycholic acid (CDCA), are produced via these two routes [14,15]. An increased bile acid ionization, amphipathic characteristics, and solubility, which are transported to and stored in the gallbladder, are produced when Bas, such as CA and CDCA are conjugated to glycine and taurine [18,19]. Cholecystokinin (CCK), a hormone secreted by the pancreas after eating, causes the gallbladder to contract and release BAs into the digestive system [18]. About 95% of bile acid (BA) molecules are reabsorbed in the terminal ileum and enter the liver via the portal vein, creating a pool of circulating BAs [20]. The intestinal flora’s action causes the remaining BAs to enter the colon and produce the primary secondary Bas: deoxycholic acid (DCA) and lithocholic acid (LCA) [19,20]. Other bile acids, including ursodeoxycholic acid (UDCA), can be formed through additional chemical changes [21]. Figure 1 summarizes the biosynthesis of bile acids.
Patients with NAFLD/NASH have been found to have altered total BA levels, as well as composition. Adults with liver biopsy-confirmed NAFLD and NASH had their BA levels tested, and these levels were dose-dependently linked with the histological characteristics of NAFLD/NASH [22,23]. As the stage of liver inflammation and fibrosis increased, the proportion of primary bound BAs increased [22,23], especially the primary BAs bound to glycine [22]. It has been shown that patients with NASH have elevated levels of BAs, mainly hydrophobic and more cytotoxic secondary BAs [24,25]. The ratio of conjugated BAs changed together with the absolute concentration of the BAs. In early chronic liver disease (non-alcoholic fatty liver [NAFL] + NASH), one investigation verified that levels of glycocholic acid (GCA)/taurocholic acid (TCA) and glycodeoxycholic acid (GDCA)/taurodeoxycholic acid (TDCA), changed with the severity of the disease [26]. NAFLD resulted in a significant elevation of certain circulating BAs, which appeared to be based on an activation of the oxysterol 7-α hydroxylase (CYP7B1)-dependent alternative BA synthesis pathway [27], and the downregulation of the levels of uptake and export transporters of the BAs in the liver [28]. Additional genomic studies have connected higher circulating BAs in NAFLD to the genetic variation in genes [22]. Furthermore, there are some potential medicinal uses for bile acids. In preclinical research, specific gene clusters regulating drug metabolism, lipid homeostasis, and BA were strongly influenced by CDCA [29]. In the meantime, the leptin-deficient obese mice with UDCA may control the hepatic energy balance and white adipose tissue macrophage polarization [30], as well as restore the gut microbiota and reduce liver inflammation in a non-alcoholic steatohepatitic mouse model [31]. The ability of UDCA to lower alanine aminotransferase (ALT) in NAFLD was demonstrated by a meta-analysis of randomized clinical studies [32]. Additionally, after six months of treatment, UDCA reduces women’s 10-year atherosclerotic cardiovascular disease (ASCVD) risk and carotid intima-media thickness (CIMT), in the overall sample [33].
The two metabolic processes, known as the classical/neutral and alternative/acidic pathways, are used in the liver to synthesis CA and CDCA from cholesterol. Following their secretion into the bile ducts, these two primary BAs are then transferred to the gut where the intestinal microbiota transforms them into the secondary bile acids DCA and LCA, or in their corresponding oxo- and dioxygenic derivatives.

3. Bile Acid Alterations and Their Function in Brain Disease

Extensive evidence obtained in recent years has revealed that NAFLD could be linked to a lower cognitive performance [34], mood imbalances [35] (in particular, depression and anxiety), cerebrovascular alterations [36], and a low total cerebral volume [37]. When NAFLD develops, a number of pro-inflammatory cytokines and chemokines are released into the bloodstream, starting the coagulation cascade pathway and causing the endothelial dysfunction and cerebral atherosclerotic disease [38,39,40,41]. Overnutrition contributes to BBB damage, the development of neuroinflammation, and finally memory and cognitive impairment [42]. Simultaneously, the neurotransmitter synthesis and signaling are disrupted, further promoting depression-like behavior [43]. Risk factors for NAFLD have been hypothesized to hasten cerebral small artery disease, resulting in white-matter lesions, cerebral microbleeds, and brain shrinkage [5]. BAs, as the main organic solute in bile, play a vital role in the liver and other tissues, which are essential for nutrients in the small intestine and for liver metabolism. We take BAs as an entry point to further link NAFLD and brain disease. Figure 2 summarizes the consequences of bile acid-mediated signaling for brain disorders.

3.1. Stroke

The most common cause of mortality and lasting disability in the world is stroke [44]. The lifetime risk of stroke is about 25% for both men and women worldwide, at age 25, with East Asia, Central Europe, and Eastern Europe having the highest rates [45]. An increasing series of clinical studies have explored the relationship between stroke and NAFLD. The incidence of NAFLD in stroke patients, ranged from 40 to 64 percent in the majority of studies, and hepatic steatosis is typically identified by imaging methods (ultrasound or transient elastography) or liver enzymes [46,47,48,49,50]. Only one study shows a substantially lower prevalence of hepatic steatosis, at about 8 percent [51]. More studies have revealed a link between NAFLD and a higher risk of stroke [6,52,53]; however, it is unclear if this link extends to more severe strokes and worse outcomes [47,48,51]. The variable definitions of NAFLD, the low sensitivity of the tests used to diagnose NAFLD, and the decision of whether to account for potential confounding variables and the number of NAFLD patients, may be the causes of these inconsistent results. Additionally, a study from the REasons for Geographic and Racial Differences in Stroke (REGARDS) that found a link between the baseline fatty liver biomarkers and future stroke in women, but not men, in the United States, also addressed sex differences [53]. Based on the REGARDS study, a different study revealed that advanced liver fibrosis may only be a risk factor for ischemic stroke in women [54]. Additionally, a system analysis revealed that NAFLD was linked to a higher incidence of cerebrovascular accidents (CVAs) in both Caucasians and Asians [55]. These inconsistent results could be the result of race and gender-related hormonal variations.
Primary bile acid levels in rat brain tissue, used in ischemia/reperfusion models, were considerably lower than those in the control group [56]. Two hours after a traumatic brain injury, the bile acid receptors and transporters were downregulated and the hepatic acute phase response was activated [57]. Young patients with stroke had a significantly different bile acid composition from the control group, according to a metabonomics analysis [58]. As a glycine-conjugated bile acid, the levels of GCDCA were significantly elevated in young ischemic stroke patients [58]. A component of bile acid, CA, controls neuroinflammation, oxidative damage, and growth factors, leading to the recovery of the BBB function and neuronal phenotype [59]. Additionally, TUDCA has been demonstrated to have neuroprotective effects in a rodent model of a hemorrhagic brain injury, by preserving the stability of the mitochondrial membranes or inhibiting the mitochondrial disturbance [60], reducing apoptosis [60], inhibiting endoplasmic reticulum stress, and reducing neuronal pyroptosis [61]. Similarly, TUDCA plays a role in ischemic brain injury by improving the mitochondrial function and reducing apoptosis [62]. Additionally, through negatively affecting the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) signaling pathway, TUDCA may reduce oxidative stress, the inflammatory response, and apoptosis in rats with an acute cerebral infarction [63]. TUDCA has been shown to diminish the expression of chemoattractants and vascular adhesion proteins, as well as the activation of glial cells and the ability of microglia to migrate [64]. NAFLD has been linked in clinical studies to an increased risk of stroke, and preclinical research has demonstrated the neuroprotective properties of several bile acids, including CA and TUDCA.

3.2. Depression

Depressive disorders, also referred to as mood disorders or anhedonia, are common and debilitating mental health conditions [65], which account for the majority of years spent in a disabled state, globally [66]. There is growing evidence that suggests a connection between depression and the existence and even severity of NAFLD [67,68,69,70]. NAFLD is also regarded as a standalone depression risk factor [8,71]. A meta-analysis from 2020 showed that the co-prevalence of depression was 18.21% in NAFLD patients and 40.68% in NASH patients [68]. Meanwhile, numerous investigations conducted in the community, revealed that people with depression had a twofold increased risk of a metabolic syndrome, compared to those without a history of melancholy [72,73]. Depression was linked to a more severe hepatocyte ballooning in NAFLD patients, and there was a dose-dependent relationship between the intensity of depressed symptoms and the grade of the hepatocyte ballooning [74].
Over-activation of the intestinal signaling pathways FXR receptor-FGF15 and hepatic apoptosis signal-regulating kinase1 (ASK1) in dextran sulphate sodium-induced depressive rats, suggests hepatic metabolic disturbances [75]. The chronic unpredictable mild stress (CUMS) model group had considerably higher serum bile acid levels [76]. Liver metabolomics studies showed that TUDCA increased [77,78], while TDCA and TCDCA decreased in the CUMS group [79]. The fecal metabolomics investigation revealed that stress significantly reduced the levels of CA, DCA, and CDCA [80]. The antidepressant mechanism may be connected to the reduction of neuroinflammation [81,82], oxidative/nitrosative stress [81,82], and endoplasmic reticulum stress [82] in the brain, and TUDCA proved successful in alleviating depression-like behavior in rats. In an experiment verifying that interferon alpha (IFN-α) causing depression, via the modulation of glucocorticoid and serotonin receptors, the effect of IFN-α on the glucocorticoid receptors was abolished when used in combination with TUDCA [83]. Interestingly, TUDCA did not have any effect on the expression level of the serotonin receptor 1A [83]. People who suffer from depression are more likely to develop a metabolic syndrome, and TUDCA, one of the bile acids, has been demonstrated, in animal experiments, to reduce depression-like behavior.

3.3. Alzheimer’s Disease

The most prevalent form of late-life dementia, Alzheimer’s disease (AD), is a neurodegenerative condition with etiologies related to the accumulation of beta-amyloid plaques and neurofibrillary tangles, synapse and neuronal loss, and cerebral shrinkage [84]. In comparison to the control group, both APP/PS1 and APPNL-G-F animals have significantly different amounts of conjugated and unconjugated primary bile acids, indicating that the liver’s overall metabolic balance is disturbed [85]. Hepatic metabolism has been discovered as a biological factor in the neuropathological resilience in Alzheimer’s disease by a broad genetic investigation of resilience [86]. Moreover, NAFLD might promote the pathological AD indications in an AD model and trigger AD signs in wild-type mice [87]. When compared to the control group, serum and brain tissue from AD patients had higher ratios of secondary to primary bile acids, such as DCA:CA or GCDCA:CA [88,89,90]. Surprisingly, LCA grew 3.2-fold within 8–9 years, while being converted to AD as a secondary bile acid [91]. Additionally, it was shown that the amount of the bile acid precursor 7α,25-dihydroxy-3-oxocholest-4-en-26-oic acid was reduced in the cerebrospinal fluid (CSF) of AD patients [92]. Lower serum levels of primary BAs and 7α-hydroxycholesterol (7α-OHC) were associated with a faster buildup of white-matter lesions, a faster brain atrophy, and a higher amyloid deposition in the brain, mostly in males [93], possibly as a result of estrogen’s protective effects [94]. In fibroblasts, UDCA has been found to reduce the number of long mitochondria and boost the mitochondrial membrane potential and respiration [95]. Additionally, when activated by β-amyloid peptide (Aβ), UDCA may block the production of the nuclear factor-κB (NF-κB)-dependent genes in microglia [96]. For the treatment of AD in mice, TUDCA, an endogenous bile acid created by the conjugation of UDCA with taurine, has demonstrated a significant therapeutic potential. TUDCA can improve the cognitive function in mice by inhibiting apoptosis [97,98], improving neuroinflammation [99], preventing the reduction in dendritic spine numbers [99], and enhancing glucose homeostasis [100]. Additionally, rats treated with aluminum chloride (AlCl3) have an improved hippocampal insulin sensitivity due to CDCA [101]. In conclusion, some bile acids have also been demonstrated to be useful in preclinical trials of AD, including UDCA, TUDCA, and CDCA, and liver metabolism plays a significant role in AD patients.

3.4. Parkinson’s Disease

Parkinson’s disease (PD) is a long-term, progressive neurodegenerative condition that manifests in both the motor and non-motor systems [102]. Patients with PD typically present with resting tremor, rigidity, bradykinesia, and stooping posture [102]. Lewy bodies and a loss of dopaminergic neurons in the substantia nigra are symptoms of PD [103]. Chronic inflammation outside of the brain, such as NAFLD, was sufficient to induce neurodegeneration without genetic predisposition [87]. While this is happening, BAs are a significant aberrant biochemical route in PD patients, whether they are seen in the plasma, cerebral fluid, or intestinal tissues [104,105,106,107,108,109]. PD patients had significantly elevated plasma CA/CDCA and decreased glycoursodeoxycholic acid (GUDCA) [104,107], and the intensity of motor complaints was linked with taurine-conjugated bile acids [106]. More carefully differentiating the different subtypes of PD patients, GDCA was significantly higher and TCA was significantly lower in leucine-rich repeat kinase 2 (LRRK2) PD patients, GCDCA was significantly lower in sporadic PD (sPD) patients, and TCA was significantly higher in LRRK2 PD compared to sPD patients [109]. However, an increase in unconjugated bile acids (CA, DCA, and LCA) has also been reported in patients with LRRK2 PD [110]. Additionally, an examination of the data from Parkinson’s disease genome-wide association studies using an empirical Bayesian Lasso showed three significant pathways, including the main pathway for bile acid production [111]. 7α,(25R)26-dihydroxycholesterol and a second oxysterol 7α,x,y-trihydroxycholest-4-en-3-one (7α,x,y-triHCO) were found to be considerably higher in PD CSF, suggesting that the acidic pathway of bile acid production has been activated [108]. Unexpectedly, the appendix and ileum of Parkinson’s patients had higher levels of DCA and LCA, and hydrophobic and secondary bile acids [105].
Several experiments have shown that TUDCA could prevent the dopaminergic neuronal damage and improve Parkinsonian dyskinesia by inhibiting neuroinflammation [112,113,114], resisting oxidative stress [114,115,116,117], reducing endoplasmic reticulum stress [118], improving mitochondrial dysfunction [113,115], reducing apoptosis [119], and preventing autophagy [114]. UDCA has been validated not only in experimental animal models of prodromal PD [120,121,122] but also in Parkinson’s patients for its pharmacokinetics and safety [123]. UDCA may enhance the mitochondrial activity in PD patients [123], and a phase II, two-center, double-blind, randomized, placebo-controlled trial of UDCA in 30 individuals with early PD, was concluded in July 2021 [124].
The changes in bile acids in brain disease are not exactly similar to those in NAFLD. Patients with NAFLD have an elevated proportion of primary bound bile acids, especially those bound to glycine. Similarly, bile acids bound to glycine, such as GCDCA, are elevated in patients with young ischemic strokes. However, GUDCA has decreased in PD patients. In preclinical studies of depression, it was shown that taurine bound bile acids were altered. Additionally, in both serum and brain tissue from AD patients, the ratio of secondary to primary bile acids increased. UDCA has a therapeutic effect on NAFLD and has a positive effect on the risk of ASCVD. Additionally, UDCA has been shown to be safe in PD patients and is crucial in the treatment of neurodegenerative illnesses. More clinical trials are required to confirm TUDCA’s efficacy; however, it has demonstrated considerable therapeutic potential for the treatment of cerebrovascular diseases, neurodegenerative diseases, and various mental diseases.

4. Bile Acid Signaling to the Central Nervous System

Among the class of nuclear and cell-surface receptors, known as bile acid-activated receptors (BARs), FXR and GPBAR1, are the most distinctive. The expression of BARs is most common in hepatocytes and ileum, as well as in the brain [12,13,125], implying that the ligands could directly bind to the receptor after crossing the BBB. Furthermore, the intestinal FXR and GPBAR1 activation causes the release of FGF19 and GLP-1, both of which communicate with the brain [126]. The role of additional receptors that might bind bile acids and are located in the brain, is summarized in Table 1.

4.1. Sending Bile Acid Signaling to the Central Nervous System via a Direct Pathway

BAs can reportedly pass the BBB, both conjugated and unconjugated [125,127,128]. The lipid layer of the BBB can be damaged by BAs at high concentrations (≥1.5 mM), while at low concentrations (0.2–1.5 mM), CDCA and DCA can damage the tight junction protein through the Rac1-dependent processes [129,130]. Additionally, the BA uptake by neurons is facilitated by bile acid transporters produced in the central nervous system, such as the apical sodium-dependent bile acid transporter (ASBT) [57,131]. Meanwhile, bile acid synthesis-related enzymes and intermediates have been observed, indicating that potentially a portion of the bile acid biosynthetic pathway occurs in the brain [132,133,134]. Numerous investigations have demonstrated that bile acid receptors are dispersed throughout the brain, for instance, GPBAR1 is expressed in cortical neurons, astrocytes, and microglia in the brain [135,136], and FXR is expressed in prefrontal cortical neurons and hippocampal neurons [137,138]. Thus, bile acid ligands, that have the ability to cross the BBB can bind directly to the receptor. Although their primary function is still the removal of cholesterol, BAs also have a range of effects on the physiology of the brain, neurogenesis, neurotransmission, and neuroendocrine responses in addition to acting as neurotrophic agents [14,16].
Table 1. Bile acid receptors found in the brain.
Table 1. Bile acid receptors found in the brain.
Receptor Cellular LocalizationBile Acid LigandsMain Function References
FXRPrefrontal cortical neurons, hippocampal neuronsCDCA, DCA, LCA, CAThe function of FXR in the brain is still paradoxical.
FXR knockdown attenuated neuronal apoptosis in ischemic brain injury.
FXR aggravated amyloid-β-triggered apoptosis by modulating the CREB-BDNF pathway in vitro.
By causing CRTC2 to translocate into the cytoplasm and disrupt the CREB-BDNF signaling pathway in the hippocampus nucleus, FXR played a role in the pathophysiology of depression.
In rats, the hippocampal BDNF expression is decreased and depression-like behavior caused by CUMS is reversed by the hippocampus FXR knockdown.
FXR signaling pathway was neuroprotective in mice with depression.
In the hippocampi of AlCl3-treated rats, CDCA was successful in enhancing insulin sensitivity.
[101,137,138,139,140,141]
GPBAR1Neurons, astrocytes, microgliaLCA, DCA, CDCA, CAIn a rodent model of subarachnoid hemorrhage, neuroinflammation, oxidative stress, and apoptosis, as well as BBB damage and neuroinflammation after MCAO, were all reduced by the GPBAR1 activation.
The locomotor activity of mice was increased and obesity, depression, and cognitive decline were all protected by the olive leaf extract, which contains a GPBAR1 agonist.
Through the hippocampal CA3 pyramidal neurons afferent to the dorsolateral septum, GPBAR1 altered depressive-like behaviors.
GPBAR1 activation alleviated inflammatory neurodegeneration in a mouse model of PD by regulating the mitochondrial dynamics in microglia.
[135,136,142,143,144,145,146,147]
PXRBrain endothelial cells, hippocampal neuronsLCANonylphenol had neurotoxic and apoptotic effects on mouse hippocampus cells, and PXR was involved in the spread of those effects.
Knockdown expression of PXR in the midbrain of Long–Evans rats lead to impaired mating behavior and reduced hippocampal BDNF levels.
[148,149]
VDRNeurons, gliaLCAIn rat primary astrocytes, the VDR activation controlled the quantities of glutathione and gamma-glutamyl transpeptidase produced.
VDR activation suppressed the inducible synthesis of nitric oxide and decreased the generation of pro-inflammatory cytokines by the activated microglia.
The pathophysiological process of depressive-like behaviors brought on by persistent stress may involve the 25(OH) D and VDR.
By increasing the hippocampus BDNF expression, the VDR signaling helped mice with post-stroke depressive symptoms.
In a rat model of traumatic brain damage, the VDR activation altered the NADPH oxidase 2 activity and prevented neurological impairments and apoptosis.
Brain endothelial P-glycoprotein levels were reduced in PD via a VDR-dependent pathway.
[150,151,152,153,154,155]
S1P2RCortical neurons, microglia, astrocytes, hippocampal pyramidal cells, retinal ganglion cellsGCA,
GDCA
TCA TDCA, TUDCA
S1P2R antagonists improved the neural progenitor cell migration near the brain infarction and reduced the hepatic encephalopathy-related neurological impairment.
By weakening the adherens junctions, S1P2R may control BBB permeability.
[156,157,158,159]
α5β1 integrinCortical neurons, brain endothelial cellsTUDCA, nor UDCA
(UDCA homolog)
Activation of integrin α5β1 promoted angiogenesis in brain endothelial cells under cerebral hypoxia, as well as the vascular endothelial growth factor secretion in MCAO rats.
α5β1 integrin influenced the BBB permeability following an ischemic stroke.
[160,161,162]
GRNeurons, microglia, cortical neuronsGCDCA
TCA, TUDCA, UDCA
Inhibiting NF-κB in a glucocorticoid-dependent way throughout the middle stage of depressive-like behavior, GR was beneficial.
Both too little and too much GR-mediated signaling hampered the neuronal migration.
In neuron-like cells, GR suppressed the production of the brain-derived neurotrophic factor.
GR-signaling in ginseng had an anti-inflammatory protective effect on neurodegenerative models.
[163,164,165,166]
Abbreviations: CREB, cyclic AMP (cAMP)-response element-binding protein; CRTC, CREB-regulated transcription coactivator; PXR, pregnane X receptor; VDR, vitamin D receptor; NADPH, nicotinamide adenine dinucleotide phosphate; S1P2R, sphingosine 1-phosphate receptor 2; GR, glucocorticoid receptor.

4.2. Sending Bile Acid Signaling to the Central Nervous System via an Indirect Pathway

4.2.1. FXR Signaling

Inhibiting bile acid synthesis in the liver, controlling bile acid circulation between the liver and the intestines, and maintaining a consistent level of bile acids in the body are all possible effects of FXR. Small heterodimer partner (SHP) levels rise and sterol regulatory element binding protein (SREBP)-1c expression decreases as the FXR activation reduces lipid levels [167]. The expression of the human peroxisome proliferator-activated receptor α gene is also encouraged by the FXR activation [168]. In contrast, it is still unknown how FXR affects cholesterol and lipoprotein production in clinical trials, and giving individuals with NASH an FXR agonist raises their cholesterol levels and negatively affects their lipoprotein levels [169]. The transcriptional activity of the protein that binds to the carbohydrate-response element-binding protein (ChREBP), controls the glucose metabolism when FXR is activated [170]. Additionally, in response to the FXR activation, the intestine secretes FGF19, the human homolog of mouse FGF15, which binds to the hepatocytes via the enterohepatic circulation and stimulates the glycogen production [171]. It is known that FGF15/19 inhibits the CREB-peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) pathway, which in turn reduces the production of the hepatic gluconeogenic genes [172]. By decreasing the CYP7A1 gene transcription through a jnk-dependent mechanism, FGF15/19 regulates the bile acid homeostasis in the liver by binding to the fibroblast growth factor receptor (FGFR) 4 [173,174].
In the absence of a heparin-binding domain linking it to cells, FGF15/19 could enter the circulation, cross the BBB and act in the brain, but with lower penetration rates, compared with FGF21 [175,176]. Similar results from in vitro investigations showed that middle cerebral artery occlusion therapy dramatically decreased the number of FGF19-positive cells in the cerebral cortex, compared to the sham group [177]. FGFR (including FGFR1c, FGFR2c, and FGFR3c) and the plasma-membrane bound β-klotho co-receptor are both necessary for FGF19 to bind to them [178,179,180]. Strikingly, FGFR4 is unique in that it binds to FGF19 in the presence or absence of β-klotho [181,182]. In contrast to FGFR4, which is only found in the medial habenula, sub-commissural organ, and lateral habenula of rodents, FGFR1–3 are strongly expressed throughout the entire brain [183]. The brain expresses FGFR broadly and abundantly, whereas the β-klotho expression is rather modest and discrete in distribution [183,184,185,186,187]. Along with the forebrain areas, such as the hypothalamus, hippocampus, or amygdala [176], β-klotho is also found in the midbrain areas, including the ventral tegmental area, medial vestibular nucleus, and medial trigeminal neurons [183,184], as well as in the area postrema and nucleus of the solitary tract [183]. The limited co-expression of FGFR1 and β-klotho has been found in cells of the hippocampus CA1-CA3 transition zone, the primary sensory nucleus of the trigeminal nerve, medial trigeminal neurons, and suprachiasmatic nucleus [183]. To our knowledge, however, no research has looked at the co-expression of FGFR2, FGFR3, and β-klotho. As a result, following the activation of the FGFR-β-klotho complex, FGF15/19 may be able to target specific brain regions and cause a signaling cascade response.

4.2.2. GPBAR1 Signaling

The membrane-bound G-protein-coupled bile acid receptor GPBAR1, also known as the Takeda G-protein-coupled bile acid receptor 5 (TGR5), is activated by bile acids in the following order: LCA > DCA > CDCA > CA [188]. Several non-parenchymal cells, such as the Kupffer cells and liver sinusoidal cells (LSC), express GPBAR1 in the liver [189]. In wild-type mice, GPBAR1 inhibits NF-κB, which negatively affects the hepatic inflammatory response [190]. Through a TGR5-cAMP-dependent mechanism, BAs prevent Kupffer cells from producing cytokines, in response to lipopolysaccharide (LPS) [191]. In the bowel, the GPBAR1 activation increases the colonic motility and secretion, decreases the gastric emptying, and encourages the production of GLP-1 from L cells, which in turn encourages the release of insulin from pancreatic β-cells [189,192]. In vascular endothelial cells, GPBAR1 is capable of vasodilatation through the release of nitric oxide (NO) and hydrogen sulfide (H2S), thereby reducing the vascular inflammation and decreasing the development of atherogenesis [192]. Additionally, BAs stimulate GPBAR1 in adipose and muscular tissue, boosting thermogenesis and energy expenditure [193].
In addition to direct GPBAR1 ligand binding, endogenous neurosteroids in the brain may also activate GPBAR1 [12]. Furthermore, the bile acid activation of GPBAR1 in the intestine, results in the release of the gut hormone GLP-1, which communicates bile acid signals to many body systems, such as the central nervous system [194,195]. By directly stimulating the GPBAR1 receptors on the basolateral side of the L cells, bile acids may change how much GLP-1 is released [195]. Dipeptidyl peptidase-IV (DPP-IV) breaks down GLP-1 so that only 1/4 of the released amount enters the portal circulation after it diffuses into the lamina propria and is absorbed by a capillary [196]. Another 50–60% is retained in the liver, leaving 10–15% to enter the systemic circulation [197]. DDP-IV is also present in the plasma, and much less GLP-1 may reach the brain via the endocrine pathway [196]. GLP-1 is able to access the brain by simple diffusion [198]. The GLP-1 receptor is widely present in the hypothalamic region of rodents and primates, particularly in the paraventricular nucleus, dorsomedial hypothalamus, and arcuate nucleus [199]. Additionally, the intestinal GLP-1 can communicate with the brain and central nervous system by activating vagal afferent fibers. Afferent sensory nerve fibers coming from the nodose ganglion, which provides impulses to the nucleus of the solitary tract and ultimately to the hypothalamus, may interact with GLP-1 before it enters the capillaries to be destroyed by DPP-IV [200]. The hepatic vagus has been shown to specifically recognize the GLP-1 expressions in the hepatoportal area [201]. Additionally, GLP-1 may communicate with the central nervous system through spinal and local sensory nerves [194]. The fact that GLP-1 is produced by preproglucagon neurons in the lower brainstem, is a key factor [202]. The difficulty of assessing the peripheral GLP-1 that is entering the central nervous system for action increases as GLP-1 is released from the brain.

5. Bile Acid Signaling in Brain Disease

5.1. FXR Signaling

The FXR expression was elevated in the nuclei of neurons, following cerebral ischemia, but not in microglia, astrocytes, or endothelial cells [139]. When Aβ-triggers neuronal death in differentiated SH-SY5Y cells and mice hippocampus neurons, the FXR expression is increased [137]. Proteomic and metabolomics studies of plasma and hypothalamus in depressed mice, indicated significant changes in the FXR/retinoid X receptor (RXR) activation [203,204]. Chen et al. [141] reported that CUMS dramatically increased the expression of the hippocampal FXR, a key player in the pathophysiology of depression. Furthermore, Hu et al. [140] observed that chronic unexpected stress worsens depressive-like behavior by overexpressing FXR in the hippocampal CA1, but not in the dentate gyrus (DG) or medial prefrontal cortex (mPFC). However, chronic social defeat stress (CSDS)-induced mice had a considerably less FXR expression in the prefrontal cortex (PFC), which increased the inflammasome activity, impaired the neuronal synaptic function, and elevated the caspase-1 activity [138]. Taken together, in disease states, the FXR expression in the brain is region-specific.
In several experimental disease models, the activation and inhibition of the FXR signaling could have counterintuitive or even opposing consequences. In mice, after brain ischemia, the FXR deletion might lower the calcium influx, encourage the neurobehavioral recovery, lessen the ischemic brain injury, reduce the inflammatory release, and attenuate neuronal death [139]. In differentiated SH-SY5Y cells, the FXR overexpression worsened the Aβ-triggered neuronal death, and the FXR agonist 6ECDCA treatment further improved this result [137]. Through a signaling cascade, including CREB/BDNF, FXR controls the Aβ-induced neuronal death in vitro [137]. Chen et al. [141] showed that the overexpression of the hippocampal FXR could reduce the expression of BDNF in naïve rats, Hu et al.’s [140] study found that the FXR–CREB interaction and the CRTC2 cytoplasmic translocation in the CA1 region were essential for depression, which was confirmed by the use of the hippocampus-specific FXR-null mice. However, the antidepressant effect of ganoderic acid A was mediated by the direct inhibition of NOD-, LRR- and pyrin domain-containing 3 (NLRP3) inflammasome activity and the restoration of the FXR expression in the PFC to activate the AMPA receptor phosphorylation and expression, which was completely abolished in mice injected with the FXR-specific inhibitor z-guggulsterone or in FXR-null mice [138]. A powerful FXR activator called CDCA may increase insulin sensitivity in the hippocampi of an AD rat model [101]. According to one study, the enhanced insulin sensitivity was achieved by the CDCA agonistic action to FXR, which was thought to be mediated via the FXR binding to the GLUT4-FXR response element [205]. When considered collectively, the downstream pathways regulated by FXR are diverse and tissue/cell specific, making it more scientific to use the tissue-specific FXR knockdown to validate and prevent altering the expression of FXR in other tissues. FXR is typically known as a ligand-activated nuclear receptor. Nevertheless, it has been demonstrated that FXR also has physical binding functions in the cytoplasm that are unrelated to its usual transcriptional activity [206]. These may be the reasons for the conflicting or even diametrically opposed outcomes that FXR signaling activation and inhibition may have. To fully comprehend the significance of the FXR expression in brain illnesses, more research on the FXR ligand-dependent and ligand-independent action in tissue/cell-specific FXR-null mice is required.

5.2. GPBAR1 Signaling

Preclinical studies of the middle cerebral artery occlusion and SAH have shown that the endogenous GPBAR1 expression is elevated in each case [135,142,143,144]. Intriguingly, the hippocampal CA3 pyramidal neurons’ GPBAR1 expression was reduced in the chronic restraint stress (CRS) and CSDS models of depression [145]. There was no discernible difference in the GPBAR1 expression between the mouse model of the 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced motor impairments and cognitive impairment and the control group [146]. However, the GPBAR1 expression is significantly decreased in mice with an Aβ1-42-induced cognitive impairment [207]. Although the expression of GPBAR1 was inconsistent in the above diseases, INT777, a specific agonist of GPBAR1, has been shown to have a partial alleviation in some brain diseases. Through the Pellino3 suppression of caspase-8/NLRP3, INT777 dramatically reduced brain damage and enhanced the neurobehavioral outcomes following MCAO [142]. Inhibition of GPBAR1 or Pellino3 rendered INT777’s anti-inflammatory actions ineffective [142]. Activating GPBAR1 could reduce damage to the BBB and improve the neural function through the BRCA1/sirtuin-1 (Sirt1) signaling pathway [143]. Additionally, via the TGR5/cAMP/PKA signaling pathway, the INT777 activation of GPBAR1 could decrease the activation of the NLRP3-apoptosis-associated speck-like protein containing a CARD (ASC) inflammasome in microglia, reduce brain edema, and enhance the short-term neurobehavioral function following SAH [144]. Similarly, INT777 reduced oxidative stress and neuronal death after SAH via the cAMP/PKCε/aldehyde dehydrogenase 2 (ALDH2) signaling pathway, which was blocked by the GPBAR1 and ALDH2 knockdown [135]. Alternately, INT777 dramatically decreased apoptosis, enhanced the synaptic dysfunction, and ameliorated the cognitive impairment brought on by Aβ1-42 [207]. By reducing the mitochondrial damage and autophagic dysfunction in microglia, INT777 reduced the tumor necrosis factor α (TNFα) release and relieved the PD neurodegeneration [146]. In addition, the genetic overexpression of GPBAR1 or the re-expression of GPBAR1 in the CA3 pyramidal neurons, and the intra-CA3 infusion of INT-777, all significantly improved the depression-like behavior of mice via the CA3 pyramidal neurons→somatostatin-GABAergic neurons of the dorsolateral septum transmission [145]. In addition to the synthetic agonists of GPBAR1, some natural agonists of GPBAR1 could also improve the cognitive function and alleviate depression. Working memory losses after consuming a high-fat diet and being inactive were avoided by using olive leaf extract, which contains oleanolic acid, a powerful GPBAR1 agonist [147]. TUDCA, a taurine conjugate of UDCA, together with GPBAR1/TGR5-boosted cAMP levels in microglia that triggered anti-inflammatory markers while reducing the pro-inflammatory ones [208]. It has been reported that TUDCA reduces neuronal apoptosis and improves the neurological function after SAH through the TGR5/sirtuin-3 (SIRT3) signaling pathway, and TGR5 small interfering RNA (siRNA), could eliminate the protective effect of TUDCA [209]. Whether this could explain the neuroprotective effect of TUDCA in animal models of several brain diseases mentioned above, needs further investigation.

6. Therapeutic Targeting of the Bile Acid Signaling

6.1. FXR Agonists

A number of FXR agonists have been developed and tested in clinical trials for the treatment of NASH/NAFLD, in light of the efficacy of the FXR ligands, as a therapy method. Synthetic agonists in clinical trials are summarized in Table 2. The strongest endogenous FXR agonist in humans is CDCA, which is also followed by DCA, LCA, and CA [210].
In a 72-week phase IIb experiment, obeticholic acid (OCA) was found to improve the histological characteristics of the NASH liver (such as hepatocyte ballooning, steatosis, and lobular inflammation) (NCT01265498). The homeostasis model assessment of insulin resistance (HOMA-IR) and insulin alterations were also significantly worsened by the OCA administration, along with dyslipidemia and pruritus. However, the lipoprotein levels improve after the drug discontinuation [169]. In a randomized global phase III study, OCA 25 mg significantly improved the NASH fibrosis levels, but the incidence of pruritus could reach more than half (NCT02548351) [211]. In pruritus, a phase I study of the interaction between Linerixibat and OCA, is being studied in healthy adults (NCT05133830). Additionally, for dyslipidemia, data from a small-scale phase II investigation showed that atorvastatin (10 mg/day) treatment in NASH patients following four weeks of OCA, led to LDL-C levels below the baseline (NCT02633956).
Clinical trials are currently favoring Cilofexor, a strong non-steroidal FXR agonist. In NASH patients, hepatic steatosis was dramatically reduced by 30 mg and 100 mg of Cilofexor; however, liver fibrosis was not significantly improved. In total, 14 percent of individuals on Cilofexor 100 mg, experienced moderate-to-severe pruritus, compared to 4 percent of those taking a placebo (NCT02854605). Additionally, Cilofixol was introduced in a combination strategy with Selonsortib and Firsocostat, in a completed phase II trial. It has been demonstrated that Cilofexor and Firsocostat together were well tolerated, improved NASH activity, and may have an antifibrotic effect (NCT03449446).
Tropifexor has been shown to be safe in healthy human subjects in a phase I trial, and in phase II trials in adult patients with NASH and hepatic fibrosis, the effectiveness of the combination of Tropifexor and Licogliflozin (NCT04065841) and Tropifexor and Cenicriviroc (NCT03517540) is being investigated. EDP305 was shown to be able to lower ALT serum levels and liver fat content in a randomized placebo-controlled experiment. Pruritus occurred in 50.9% and 9.1% of patients in the 2.5 mg and 1 mg groups (NCT03421431), respectively. Other FXR agonists now being tested in phase II clinical studies include Px-104, EYP001, MET409, and TERN-101 (Table 2). For the treatment of brain disorders, the FXR agonists are currently in the preclinical stages of research; their effectiveness in humans is not yet known.

6.2. FXR Antagonists

Natural FXR antagonists include muricholic acids (α-β-MCA) [212], glycine-β-MCA [213], guggulsterone [214], stigmasterol [215], and marine steroids [216]. Intestinal FXR antagonists have been shown in numerous studies to decrease the ceramide release to lessen the hepatic triglyceride buildup, and they are being investigated in preclinical disease models, as possible therapeutics for metabolic illnesses [217,218]. In brain disease, the knockdown of FXR or the silencing of the FXR expression in specific regions, improves symptoms and alleviates inflammation and neuronal apoptosis [139,140,141]. Notably, it is unclear if intestine FXR antagonists have a positive impact on brain illness and whether these modifications to the BA composition have negative effects, following the long-term FXR suppression.

6.3. GPBAR1 Agonists

GPBAR1 receptors are activated by bile acids and by natural substances, such as oleanolic acid, ursolic acid, and betulinic acid. Preclinical research has suggested that INT777 may be helpful in the treatment of NAFLD or other brain disorders. In a mouse model of steatohepatitis, BAR501, a specific GPBAR1 agonist, might repair liver and vascular damage [219]. A phase IIa trial showed that the glucose effects of SB-756050 + sitagliptin, were comparable to those of sitagliptin alone and SB-756050 exhibited nonlinear pharmacokinetics (NCT00733577).

6.4. Dual GPBAR1/FXR Ligands

Two dual GPBAR1/FXR agonists, BAR502 and INT767, both prevent liver injury in preclinical experiments in NAFLD [189] (Figure 3). In contrast to BAR502, which has a modest preference for GPBAR1, INT767 is a preferential FXR agonist [189]. INT767 significantly reduces the atherosclerosis formation in preclinical studies [220,221]. In addition, a phase I study on safety, tolerability, pharmacokinetics, and pharmacodynamics of BAR502 is to be conducted in healthy subjects in 2022 (NCT05203367). The FXR antagonistic/GPBAR1 agonistic compound is in development [222].

7. Conclusions and Perspectives

Brain disease may be an extrahepatic manifestation of NAFLD. For NAFLD, the therapy that focuses on hepatic steatosis and fibrosis is simplistic, and the targets of currently developed drugs may be limited. In contrast, anti-inflammatory and anti-fibrotic effects, and the improvement of systemic metabolic disorders may be more consistent with a multi-targeted treatment model for the disease. Additionally, BAs have neuroprotective qualities, and direct and indirect pathways are included in the BA signaling to the brain. The therapeutic implementation of BAs and targeting BA-mediated signaling in brain disease cannot be overemphasized. FXR and GPBAR1 are activated by ligands to regulate not only the liver but also the whole body. However, currently developed agonists of FXR have achieved some efficacy in improving the liver, while affecting the lipid and systemic metabolism. Dual GPBAR1/FXR ligands may be more advantageous than the single FXR or GPBAR1 ligands for systemic metabolism and dyslipidemias. The improvement of dual GPBAR1/FXR ligands for brain disease still needs to be proven in clinical trials. We acknowledge that bile acid signaling may not link all brain diseases and NAFLD, but the better understanding of the BA signaling and its role in the brain will improve our comprehension of this axis, which leads to the development of new therapeutic strategies to manage related disorders.

Author Contributions

Z.-L.R. and C.-X.L.: writing—original draft. F.-F.C., C.-Y.M. and X.-Q.W.: writing—review and editing. D.C. and J.-H.C.: drawing—editing of picture. W.-X.X. and C.-A.C.: writing—editing of table. All authors have read and agreed to the published version of the manuscript.

Funding

The present study was supported by grants from the National Natural Science Foundation of China (NSFC)(8197151084, 8200142456, 82004327, U21A20400) and Projects of Beijing University of Chinese Medicine (2022-JYB-JBZR-004).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Authors are thankful for the financial support from NSFC and Projects of Beijing University of Chinese Medicine.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

CYP8B1, sterol 12a-hydroxylase; GCDCA, glycochenodeoxycholic acid; TCDCA, taurochenodeoxycholic acid; BSH, bile salt hydrolase; HSDH, hydroxysteroid dehydrogenase; MCAO, middle cerebral artery occlusion; SAH, subarachnoid hemorrhage; CTX, cerebrotendinous xanthomatosis; TUDCA, tauroursodeoxycholic acid; HD, Huntington’s disease; ALS, amyotrophic lateral sclerosis; HE, hepatic encephalopathy; BDNF, brain-derived neurotrophic factor.

References

  1. Younossi, Z.M.; Koenig, A.B.; Abdelatif, D.; Fazel, Y.; Henry, L.; Wymer, M. Global Epidemiology of Nonalcoholic Fatty Liver Disease-Meta-Analytic Assessment of Prevalence, Incidence, and Outcomes. Hepatology 2016, 64, 73–84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Younossi, Z.; Anstee, Q.M.; Marietti, M.; Hardy, T.; Henry, L.; Eslam, M.; George, J.; Bugianesi, E. Global Burden of NAFLD and NASH: Trends, Predictions, Risk Factors and Prevention. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 11–20. [Google Scholar] [CrossRef] [PubMed]
  3. Anstee, Q.M.; Targher, G.; Day, C.P. Progression of NAFLD to Diabetes Mellitus, Cardiovascular Disease or Cirrhosis. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 330–344. [Google Scholar] [CrossRef]
  4. Powell, E.E.; Wong, V.W.-S.; Rinella, M. Non-Alcoholic Fatty Liver Disease. Lancet 2021, 397, 2212–2224. [Google Scholar] [CrossRef]
  5. Bizino, M.B.; Sala, M.L.; de Heer, P.; van der Tol, P.; Smit, J.W.A.; Webb, A.G.; de Roos, A.; Lamb, H.J. MR of Multi-Organ Involvement in the Metabolic Syndrome. Magn. Reson. Imaging Clin. N. Am. 2015, 23, 41–58. [Google Scholar] [CrossRef]
  6. Mahfood Haddad, T.; Hamdeh, S.; Kanmanthareddy, A.; Alla, V.M. Nonalcoholic Fatty Liver Disease and the Risk of Clinical Cardiovascular Events: A Systematic Review and Meta-Analysis. Diabetes Metab. Syndr. Clin. Res. Rev. 2017, 11, S209–S216. [Google Scholar] [CrossRef] [PubMed]
  7. Celikbilek, A.; Celikbilek, M.; Bozkurt, G. Cognitive Assessment of Patients with Nonalcoholic Fatty Liver Disease. Eur. J. Gastroenterol. Hepatol. 2018, 30, 944–950. [Google Scholar] [CrossRef] [PubMed]
  8. Labenz, C.; Huber, Y.; Michel, M.; Nagel, M.; Galle, P.R.; Kostev, K.; Schattenberg, J.M. Nonalcoholic Fatty Liver Disease Increases the Risk of Anxiety and Depression. Hepatol. Commun. 2020, 4, 1293–1301. [Google Scholar] [CrossRef] [PubMed]
  9. Huang, R.; Fan, J.-G.; Shi, J.-P.; Mao, Y.-M.; Wang, B.-Y.; Zhao, J.-M.; Lu, L.-G.; Zhong, B.-H.; Zou, Z.-S.; Xu, Y.-Q.; et al. Health-Related Quality of Life in Chinese Population with Non-Alcoholic Fatty Liver Disease: A National Multicenter Survey. Health Qual. Life Outcomes 2021, 19, 140. [Google Scholar] [CrossRef] [PubMed]
  10. Chiang, J.Y.L.; Ferrell, J.M. Bile Acids as Metabolic Regulators and Nutrient Sensors. Annu. Rev. Nutr. 2019, 39, 175–200. [Google Scholar] [CrossRef] [PubMed]
  11. Chiang, J.Y.L.; Ferrell, J.M. Bile Acid Receptors FXR and TGR5 Signaling in Fatty Liver Diseases and Therapy. Am. J. Physiol. Gastrointest. Liver Physiol. 2020, 318, G554–G573. [Google Scholar] [CrossRef]
  12. Keitel, V.; Görg, B.; Bidmon, H.J.; Zemtsova, I.; Spomer, L.; Zilles, K.; Häussinger, D. The Bile Acid Receptor TGR5 (Gpbar-1) Acts as a Neurosteroid Receptor in Brain. Glia 2010, 58, 1794–1805. [Google Scholar] [CrossRef]
  13. Huang, C.; Wang, J.; Hu, W.; Wang, C.; Lu, X.; Tong, L.; Wu, F.; Zhang, W. Identification of Functional Farnesoid X Receptors in Brain Neurons. FEBS Lett. 2016, 590, 3233–3242. [Google Scholar] [CrossRef]
  14. McMillin, M.; DeMorrow, S. Effects of Bile Acids on Neurological Function and Disease. FASEB J. 2016, 30, 3658–3668. [Google Scholar] [CrossRef] [Green Version]
  15. Kiriyama, Y.; Nochi, H. The Biosynthesis, Signaling, and Neurological Functions of Bile Acids. Biomolecules 2019, 9, 232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Ackerman, H.D.; Gerhard, G.S. Bile Acids in Neurodegenerative Disorders. Front. Aging Neurosci. 2016, 8, 263. [Google Scholar] [CrossRef] [Green Version]
  17. Fakheri, R.J.; Javitt, N.B. 27-Hydroxycholesterol, Does It Exist? On the Nomenclature and Stereochemistry of 26-Hydroxylated Sterols. Steroids 2012, 77, 575–577. [Google Scholar] [CrossRef] [PubMed]
  18. Chiang, J.Y.L.; Ferrell, J.M. Bile Acid Metabolism in Liver Pathobiology. Gene Expr. 2018, 18, 71–87. [Google Scholar] [CrossRef] [Green Version]
  19. Farooqui, N.; Elhence, A.; Shalimar. A Current Understanding of Bile Acids in Chronic Liver Disease. J. Clin. Exp. Hepatol. 2022, 12, 155–173. [Google Scholar] [CrossRef]
  20. Chow, M.D.; Lee, Y.-H.; Guo, G.L. The Role of Bile Acids in Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis. Mol. Asp. Med. 2017, 56, 34–44. [Google Scholar] [CrossRef]
  21. Lepercq, P.; Gérard, P.; Béguet, F.; Raibaud, P.; Grill, J.-P.; Relano, P.; Cayuela, C.; Juste, C. Epimerization of Chenodeoxycholic Acid to Ursodeoxycholic Acid by Clostridium Baratii Isolated from Human Feces. FEMS Microbiol. Lett. 2004, 235, 65–72. [Google Scholar] [CrossRef] [PubMed]
  22. Nimer, N.; Choucair, I.; Wang, Z.; Nemet, I.; Li, L.; Gukasyan, J.; Weeks, T.L.; Alkhouri, N.; Zein, N.; Tang, W.H.W.; et al. Bile Acids Profile, Histopathological Indices and Genetic Variants for Non-Alcoholic Fatty Liver Disease Progression. Metabolism 2021, 116, 154457. [Google Scholar] [CrossRef] [PubMed]
  23. Caussy, C.; Hsu, C.; Singh, S.; Bassirian, S.; Kolar, J.; Faulkner, C.; Sinha, N.; Bettencourt, R.; Gara, N.; Valasek, M.A.; et al. Serum Bile Acid Patterns Are Associated with the Presence of NAFLD in Twins, and Dose-Dependent Changes with Increase in Fibrosis Stage in Patients with Biopsy-Proven NAFLD. Aliment. Pharmacol. Ther. 2019, 49, 183–193. [Google Scholar] [CrossRef] [PubMed]
  24. Shao, M.; Ye, Z.; Qin, Y.; Wu, T. Abnormal Metabolic Processes Involved in the Pathogenesis of Non-alcoholic Fatty Liver Disease (Review). Exp. Ther. Med. 2020, 20, 26. [Google Scholar] [CrossRef]
  25. Ferslew, B.C.; Xie, G.; Johnston, C.K.; Su, M.; Stewart, P.W.; Jia, W.; Brouwer, K.L.R.; Sidney Barritt, A. Altered Bile Acid Metabolome in Patients with Nonalcoholic Steatohepatitis. Dig. Dis. Sci. 2015, 60, 3318–3328. [Google Scholar] [CrossRef] [Green Version]
  26. Chen, T.; Zhou, K.; Sun, T.; Sang, C.; Jia, W.; Xie, G. Altered Bile Acid Glycine: Taurine Ratio in the Progression of Chronic Liver Disease. J. Gastroenterol. Hepatol. 2022, 37, 208–215. [Google Scholar] [CrossRef]
  27. Lake, A.D.; Novak, P.; Shipkova, P.; Aranibar, N.; Robertson, D.; Reily, M.D.; Lu, Z.; Lehman-McKeeman, L.D.; Cherrington, N.J. Decreased Hepatotoxic Bile Acid Composition and Altered Synthesis in Progressive Human Nonalcoholic Fatty Liver Disease. Toxicol. Appl. Pharmacol. 2013, 268, 132–140. [Google Scholar] [CrossRef] [Green Version]
  28. Okushin, K.; Tsutsumi, T.; Enooku, K.; Fujinaga, H.; Kado, A.; Shibahara, J.; Fukayama, M.; Moriya, K.; Yotsuyanagi, H.; Koike, K. The Intrahepatic Expression Levels of Bile Acid Transporters Are Inversely Correlated with the Histological Progression of Nonalcoholic Fatty Liver Disease. J. Gastroenterol. 2016, 51, 808–818. [Google Scholar] [CrossRef]
  29. Krattinger, R.; Boström, A.; Lee, S.M.L.; Thasler, W.E.; Schiöth, H.B.; Kullak-Ublick, G.A.; Mwinyi, J. Chenodeoxycholic Acid Significantly Impacts the Expression of MiRNAs and Genes Involved in Lipid, Bile Acid and Drug Metabolism in Human Hepatocytes. Life Sci. 2016, 156, 47–56. [Google Scholar] [CrossRef]
  30. Chen, Y.-S.; Liu, H.-M.; Lee, T.-Y. Ursodeoxycholic Acid Regulates Hepatic Energy Homeostasis and White Adipose Tissue Macrophages Polarization in Leptin-Deficiency Obese Mice. Cells 2019, 8, 253. [Google Scholar] [CrossRef]
  31. Li, H.; Wang, Q.; Chen, P.; Zhou, C.; Zhang, X.; Chen, L. Ursodeoxycholic Acid Treatment Restores Gut Microbiota and Alleviates Liver Inflammation in Non-Alcoholic Steatohepatitic Mouse Model. Front. Pharmacol. 2021, 12, 788558. [Google Scholar] [CrossRef]
  32. Zhang, W.; Tang, Y.; Huang, J.; Ren, H.; Yang, Y.; Yang, Q.; Hu, H. Efficacy of Ursodeoxycholic Acid in Nonalcoholic Fatty Liver Disease: An Updated Meta-Analysis of Randomized Controlled Trials. Asia Pac. J. Clin. Nutr. 2019, 29, 696–705. [Google Scholar] [CrossRef]
  33. Nadinskaia, M.; Maevskaya, M.; Ivashkin, V.; Kodzoeva, K.; Pirogova, I.; Chesnokov, E.; Nersesov, A.; Kaibullayeva, J.; Konysbekova, A.; Raissova, A.; et al. Ursodeoxycholic Acid as a Means of Preventing Atherosclerosis, Steatosis and Liver Fibrosis in Patients with Nonalcoholic Fatty Liver Disease. World J. Gastroenterol. 2021, 27, 959–975. [Google Scholar] [CrossRef]
  34. George, E.S.; Sood, S.; Daly, R.M.; Tan, S.-Y. Is There an Association between Non-Alcoholic Fatty Liver Disease and Cognitive Function? A Systematic Review. BMC Geriatr. 2022, 22, 47. [Google Scholar] [CrossRef]
  35. Shea, S.; Lionis, C.; Kite, C.; Atkinson, L.; Chaggar, S.S.; Randeva, H.S.; Kyrou, I. Non-Alcoholic Fatty Liver Disease (NAFLD) and Potential Links to Depression, Anxiety, and Chronic Stress. Biomedicines 2021, 9, 1697. [Google Scholar] [CrossRef]
  36. Hadjihambi, A. Cerebrovascular Alterations in NAFLD: Is It Increasing Our Risk of Alzheimer’s Disease? Anal. Biochem. 2022, 636, 114387. [Google Scholar] [CrossRef]
  37. Filipović, B.; Marković, O.; Đurić, V.; Filipović, B. Cognitive Changes and Brain Volume Reduction in Patients with Nonalcoholic Fatty Liver Disease. Can. J. Gastroenterol. Hepatol. 2018, 2018, 69638797. [Google Scholar] [CrossRef]
  38. Li, W.; Liu, J.; Cai, J.; Zhang, X.; Zhang, P.; She, Z.; Chen, S.; Li, H. NAFLD as a Continuous Driver in the Whole Spectrum of Vascular Disease. J. Mol. Cell. Cardiol. 2022, 163, 118–132. [Google Scholar] [CrossRef]
  39. Saha, D.; Saha, S.; Sergeeva, E.G.; Ionova, Z.I.; Gorbach, A.V. Tissue Factor and Atherothrombosis. Curr. Pharm. Des. 2015, 21, 1152–1157. [Google Scholar] [CrossRef]
  40. Posadas-Sánchez, R.; Vargas-Alarcón, G. Innate Immunity in Coronary Disease. The Role of Interleukin-12 Cytokine Family in Atherosclerosis. Rev. Investig. Clín. 2018, 70, 130. [Google Scholar] [CrossRef]
  41. Khan, R.; Rheaume, E.; Tardif, J.-C. Examining the Role of and Treatment Directed at IL-1β in Atherosclerosis. Curr. Atheroscler. Rep. 2018, 20, 53. [Google Scholar] [CrossRef]
  42. Więckowska-Gacek, A.; Mietelska-Porowska, A.; Wydrych, M.; Wojda, U. Western Diet as a Trigger of Alzheimer’s Disease: From Metabolic Syndrome and Systemic Inflammation to Neuroinflammation and Neurodegeneration. Ageing Res. Rev. 2021, 70, 101397. [Google Scholar] [CrossRef]
  43. Chan, K.L.; Cathomas, F.; Russo, S.J. Central and Peripheral Inflammation Link Metabolic Syndrome and Major Depressive Disorder. Physiology 2019, 34, 123–133. [Google Scholar] [CrossRef]
  44. Chen, Y.; Wright, N.; Guo, Y.; Turnbull, I.; Kartsonaki, C.; Yang, L.; Bian, Z.; Pei, P.; Pan, D.; Zhang, Y.; et al. Mortality and Recurrent Vascular Events after First Incident Stroke: A 9-Year Community-Based Study of 0·5 Million Chinese Adults. Lancet Glob. Health 2020, 8, e580–e590. [Google Scholar] [CrossRef] [Green Version]
  45. The GBD 2016 Lifetime Risk of Stroke Collaborators. Global, Regional, and Country-Specific Lifetime Risks of Stroke, 1990 and 2016. N. Engl. J. Med. 2018, 379, 2429–2437. [Google Scholar] [CrossRef]
  46. Moshayedi, H.; Ahrabi, R.; Mardani, A.; Sadigetegad, S.; Farhudi, M. Association between Non-Alcoholic Fatty Liver Disease and Ischemic Stroke. Iran. J. Neurol. 2014, 13, 144–148. [Google Scholar]
  47. Abdeldyem, S.M.; Goda, T.; Khodeir, S.A.; Abou Saif, S.; Abd-Elsalam, S. Nonalcoholic Fatty Liver Disease in Patients with Acute Ischemic Stroke Is Associated with More Severe Stroke and Worse Outcome. J. Clin. Lipidol. 2017, 11, 915–919. [Google Scholar] [CrossRef]
  48. Baik, M.; Kim, S.U.; Nam, H.S.; Heo, J.H.; Kim, Y.D. The Paradoxical Protective Effect of Liver Steatosis on Severity and Functional Outcome of Ischemic Stroke. Front. Neurol. 2019, 10, 375. [Google Scholar] [CrossRef]
  49. Baik, M.; Kim, S.U.; Kang, S.; Park, H.J.; Nam, H.S.; Heo, J.H.; Kim, B.K.; Park, J.Y.; Kim, D.Y.; Ahn, S.H.; et al. Liver Fibrosis, Not Steatosis, Associates with Long-Term Outcomes in Ischaemic Stroke Patients. Cerebrovasc. Dis. 2019, 47, 32–39. [Google Scholar] [CrossRef]
  50. Mori, T.; Yoshioka, K.; Tanno, Y. Non-Alcoholic Fatty Liver Disease Frequency and Associated Factors at Admission of Acute Stroke. Hepatol. Int. 2021, 16, 81–88. [Google Scholar] [CrossRef]
  51. Tziomalos, K.; Giampatzis, V.; Bouziana, S.D.; Spanou, M.; Papadopoulou, M.; Pavlidis, A.; Kostaki, S.; Bozikas, A.; Savopoulos, C.; Hatzitolios, A.I. Association between Nonalcoholic Fatty Liver Disease and Acute Ischemic Stroke Severity and Outcome. World J. Hepatol. 2013, 5, 621. [Google Scholar] [CrossRef]
  52. Xu, J.; Dai, L.; Zhang, Y.; Wang, A.; Li, H.; Wang, Y.; Meng, X.; Wu, S.; Wang, Y. Severity of Nonalcoholic Fatty Liver Disease and Risk of Future Ischemic Stroke Events. Stroke 2021, 52, 103–110. [Google Scholar] [CrossRef] [PubMed]
  53. Alexander, K.S.; Zakai, N.A.; Lidofsky, S.D.; Callas, P.W.; Judd, S.E.; Tracy, R.P.; Cushman, M. Non-Alcoholic Fatty Liver Disease, Liver Biomarkers and Stroke Risk: The Reasons for Geographic and Racial Differences in Stroke Cohort. PLoS ONE 2018, 13, e0194153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Parikh, N.S.; Koh, I.; VanWagner, L.B.; Elkind, M.S.V.; Zakai, N.A.; Cushman, M. Liver Fibrosis Is Associated with Ischemic Stroke Risk in Women but Not Men: The REGARDS Study. J. Stroke Cerebrovasc. Dis. 2021, 30, 105788. [Google Scholar] [CrossRef] [PubMed]
  55. Hu, J.; Xu, Y.; He, Z.; Zhang, H.; Lian, X.; Zhu, T.; Liang, C.; Li, J. Increased Risk of Cerebrovascular Accident Related to Non-Alcoholic Fatty Liver Disease: A Meta-Analysis. Oncotarget 2018, 9, 2752–2760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Zhou, P.; Zhou, L.; Shi, Y.; Li, Z.; Liu, L.; Zuo, L.; Zhang, J.; Liang, S.; Kang, J.; Du, S.; et al. Neuroprotective Effects of Danshen Chuanxiongqin Injection Against Ischemic Stroke: Metabolomic Insights by UHPLC-Q-Orbitrap HRMS Analysis. Front. Mol. Biosci. 2021, 8, 630291. [Google Scholar] [CrossRef]
  57. Nizamutdinov, D.; DeMorrow, S.; McMillin, M.; Kain, J.; Mukherjee, S.; Zeitouni, S.; Frampton, G.; Bricker, P.C.S.; Hurst, J.; Shapiro, L.A. Hepatic Alterations Are Accompanied by Changes to Bile Acid Transporter-Expressing Neurons in the Hypothalamus after Traumatic Brain Injury. Sci. Rep. 2017, 7, 40112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Liu, J.; Yuan, J.; Zhao, J.; Zhang, L.; Wang, Q.; Wang, G. Serum Metabolomic Patterns in Young Patients with Ischemic Stroke: A Case Study. Metabolomics 2021, 17, 24. [Google Scholar] [CrossRef]
  59. Li, C.; Wang, X.; Yan, J.; Cheng, F.; Ma, X.; Chen, C.; Wang, W.; Wang, Q. Cholic Acid Protects In Vitro Neurovascular Units against Oxygen and Glucose Deprivation-Induced Injury through the BDNF-TrkB Signaling Pathway. Oxidative Med. Cell. Longev. 2020, 2020, 1201624. [Google Scholar] [CrossRef]
  60. Rodrigues, C.M.P.; Solá, S.; Nan, Z.; Castro, R.E.; Ribeiro, P.S.; Low, W.C.; Steer, C.J. Tauroursodeoxycholic Acid Reduces Apoptosis and Protects against Neurological Injury after Acute Hemorrhagic Stroke in Rats. Proc. Natl. Acad. Sci. USA 2003, 100, 6087–6092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Chen, G.; Gao, C.; Yan, Y.; Wang, T.; Luo, C.; Zhang, M.; Chen, X.; Tao, L. Inhibiting ER Stress Weakens Neuronal Pyroptosis in a Mouse Acute Hemorrhagic Stroke Model. Mol. Neurobiol. 2020, 57, 5324–5335. [Google Scholar] [CrossRef]
  62. Rodrigues, C.M.P.; Spellman, S.R.; Solá, S.; Grande, A.W.; Linehan-Stieers, C.; Low, W.C.; Steer, C.J. Neuroprotection by a Bile Acid in an Acute Stroke Model in the Rat. J. Cereb. Blood Flow Metab. 2002, 22, 463–471. [Google Scholar] [CrossRef] [Green Version]
  63. Bian, K.-Y.; Jin, H.-F.; Sun, W.; Sun, Y.-J. DCA Can Improve the ACI-Induced Neurological Impairment through Negative Regulation of Nrf2 Signaling Pathway. Eur. Rev. Med. Pharm. Sci. 2019, 23, 343–351. [Google Scholar] [CrossRef]
  64. Yanguas-Casás, N.; Barreda-Manso, M.A.; Nieto-Sampedro, M.; Romero-Ramírez, L. Tauroursodeoxycholic Acid Reduces Glial Cell Activation in an Animal Model of Acute Neuroinflammation. J. Neuroinflamm. 2014, 11, 50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Kaltenboeck, A.; Harmer, C. The Neuroscience of Depressive Disorders: A Brief Review of the Past and Some Considerations about the Future. Brain Neurosci. Adv. 2018, 2, 2398212818799269. [Google Scholar] [CrossRef] [Green Version]
  66. James, S.L.; Abate, D.; Abate, K.H.; Abay, S.M.; Abbafati, C.; Abbasi, N.; Abbastabar, H.; Abd-Allah, F.; Abdela, J.; Abdelalim, A.; et al. Global, Regional, and National Incidence, Prevalence, and Years Lived with Disability for 354 Diseases and Injuries for 195 Countries and Territories, 1990–2017: A Systematic Analysis for the Global Burden of Disease Study 2017. Lancet 2018, 392, 1789–1858. [Google Scholar] [CrossRef] [Green Version]
  67. Choi, J.M.; Chung, G.E.; Kang, S.J.; Kwak, M.-S.; Yang, J.I.; Park, B.; Yim, J.Y. Association Between Anxiety and Depression and Nonalcoholic Fatty Liver Disease. Front. Med. 2021, 7, 585618. [Google Scholar] [CrossRef] [PubMed]
  68. Xiao, J.; Lim, L.K.E.; Ng, C.H.; Tan, D.J.H.; Lim, W.H.; Ho, C.S.H.; Tan, E.X.X.; Sanyal, A.J.; Muthiah, M.D. Is Fatty Liver Associated With Depression? A Meta-Analysis and Systematic Review on the Prevalence, Risk Factors, and Outcomes of Depression and Non-Alcoholic Fatty Liver Disease. Front. Med. 2021, 8, 691696. [Google Scholar] [CrossRef] [PubMed]
  69. Jung, J.Y.; Park, S.K.; Oh, C.-M.; Chung, P.-W.; Ryoo, J.-H. Non-Alcoholic Fatty Liver Disease and Its Association with Depression in Korean General Population. J. Korean Med. Sci. 2019, 34, e199. [Google Scholar] [CrossRef] [PubMed]
  70. Kim, D.; Yoo, E.R.; Li, A.A.; Tighe, S.P.; Cholankeril, G.; Harrison, S.A.; Ahmed, A. Depression Is Associated with Non-Alcoholic Fatty Liver Disease among Adults in the United States. Aliment. Pharmacol. Ther. 2019, 50, 590–598. [Google Scholar] [CrossRef] [PubMed]
  71. Zelber-Sagi, S.; Toker, S.; Armon, G.; Melamed, S.; Berliner, S.; Shapira, I.; Halpern, Z.; Santo, E.; Shibolet, O. Elevated Alanine Aminotransferase Independently Predicts New Onset of Depression in Employees Undergoing Health Screening Examinations. Psychol. Med. 2013, 43, 2603–2613. [Google Scholar] [CrossRef]
  72. Kinder, L.S.; Carnethon, M.R.; Palaniappan, L.P.; King, A.C.; Fortmann, S.P. Depression and the Metabolic Syndrome in Young Adults: Findings from the Third National Health and Nutrition Examination Survey. Psychosom. Med. 2004, 66, 316–322. [Google Scholar] [CrossRef] [PubMed]
  73. Roriz-Cruz, M.; Rosset, I.; Wada, T.; Sakagami, T.; Ishine, M.; Roriz-Filho, J.S.; Cruz, T.R.S.; Rodrigues, R.P.; Resmini, I.; Sudoh, S.; et al. Stroke-Independent Association between Metabolic Syndrome and Functional Dependence, Depression, and Low Quality of Life in Elderly Community-Dwelling Brazilian People. J. Am. Geriatr. Soc. 2007, 55, 374–382. [Google Scholar] [CrossRef] [PubMed]
  74. Youssef, N.A.; Abdelmalek, M.F.; Binks, M.; Guy, C.D.; Omenetti, A.; Smith, A.D.; Diehl, A.M.E.; Suzuki, A. Associations of Depression, Anxiety and Antidepressants with Histological Severity of Nonalcoholic Fatty Liver Disease. Liver Int. 2013, 33, 1062–1070. [Google Scholar] [CrossRef] [PubMed]
  75. Lv, W.; Liu, C.; Yu, L.; Zhou, J.; Li, Y.; Xiong, Y.; Guo, A.; Chao, L.; Qu, Q.; Wei, G.; et al. Melatonin Alleviates Neuroinflammation and Metabolic Disorder in DSS-Induced Depression Rats. Oxidative Med. Cell. Longev. 2020, 2020, 1241894. [Google Scholar] [CrossRef]
  76. Xu, E.; Wang, B.; Lu, S.; Zhang, C.; Zhu, L.; Liu, X.; Bai, M.; Li, Y. Tandem Mass Tag-Based Quantitative Proteomic Analysis of the Liver Reveals Potential Protein Targets of Xiaochaihutang in CUMS Model of Depression. J. Chromatogr. B 2021, 1181, 122898. [Google Scholar] [CrossRef] [PubMed]
  77. Jia, H.; Yu, M.; Ma, L.-Y.; Zhang, H.; Zou, Z. Chaihu-Shu-Gan-San Regulates Phospholipids and Bile Acid Metabolism against Hepatic Injury Induced by Chronic Unpredictable Stress in Rat. J. Chromatogr. B 2017, 1064, 14–21. [Google Scholar] [CrossRef]
  78. Jia, H.; Li, Q.; Zhou, C.; Yu, M.; Yang, Y.; Zhang, H.; Ding, G.; Shang, H.; Zou, Z. Chronic Unpredictive Mild Stress Leads to Altered Hepatic Metabolic Profile and Gene Expression. Sci. Rep. 2016, 6, 23441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Wang, P.; Gao, X.; Liang, M.; Fang, Y.; Jia, J.; Tian, J.; Li, Z.; Qin, X. Dose-Effect/Toxicity of Bupleuri Radix on Chronic Unpredictable Mild Stress and Normal Rats Based on Liver Metabolomics. Front. Pharmacol. 2021, 12, 627451. [Google Scholar] [CrossRef] [PubMed]
  80. Zhao, J.; Jung, Y.-H.; Jin, Y.; Kang, S.; Jang, C.-G.; Lee, J. A Comprehensive Metabolomics Investigation of Hippocampus, Serum, and Feces Affected by Chronic Fluoxetine Treatment Using the Chronic Unpredictable Mild Stress Mouse Model of Depression. Sci. Rep. 2019, 9, 7566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Cheng, L.; Huang, C.; Chen, Z. Tauroursodeoxycholic Acid Ameliorates Lipopolysaccharide-Induced Depression Like Behavior in Mice via the Inhibition of Neuroinflammation and Oxido-Nitrosative Stress. Pharmacology 2019, 103, 93–100. [Google Scholar] [CrossRef]
  82. Lu, X.; Yang, R.-R.; Zhang, J.-L.; Wang, P.; Gong, Y.; Hu, W.-F.; Wu, Y.; Gao, M.-H.; Huang, C. Tauroursodeoxycholic Acid Produces Antidepressant-like Effects in a Chronic Unpredictable Stress Model of Depression via Attenuation of Neuroinflammation, Oxido-Nitrosative Stress, and Endoplasmic Reticulum Stress. Fundam. Clin. Pharm. 2018, 32, 363–377. [Google Scholar] [CrossRef] [PubMed]
  83. Cai, W.; Khaoustov, V.I.; Xie, Q.; Pan, T.; Le, W.; Yoffe, B. Interferon-Alpha-Induced Modulation of Glucocorticoid and Serotonin Receptors as a Mechanism of Depression. J. Hepatol. 2005, 42, 880–887. [Google Scholar] [CrossRef] [PubMed]
  84. Lane, C.A.; Hardy, J.; Schott, J.M. Alzheimer’s Disease. Eur. J. Neurol. 2018, 25, 59–70. [Google Scholar] [CrossRef] [PubMed]
  85. Kaur, H.; Seeger, D.; Golovko, S.; Golovko, M.; Combs, C.K. Liver Bile Acid Changes in Mouse Models of Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 7451. [Google Scholar] [CrossRef]
  86. Dumitrescu, L.; Mahoney, E.R.; Mukherjee, S.; Lee, M.L.; Bush, W.S.; Engelman, C.D.; Lu, Q.; Fardo, D.W.; Trittschuh, E.H.; Mez, J.; et al. Genetic Variants and Functional Pathways Associated with Resilience to Alzheimer’s Disease. Brain 2020, 143, 2561–2575. [Google Scholar] [CrossRef]
  87. Kim, D.-G.; Krenz, A.; Toussaint, L.E.; Maurer, K.J.; Robinson, S.-A.; Yan, A.; Torres, L.; Bynoe, M.S. Non-Alcoholic Fatty Liver Disease Induces Signs of Alzheimer’s Disease (AD) in Wild-Type Mice and Accelerates Pathological Signs of AD in an AD Model. J. Neuroinflamm. 2016, 13, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. MahmoudianDehkordi, S.; Arnold, M.; Nho, K.; Ahmad, S.; Jia, W.; Xie, G.; Louie, G.; Kueider-Paisley, A.; Moseley, M.A.; Thompson, J.W.; et al. Altered Bile Acid Profile Associates with Cognitive Impairment in Alzheimer’s Disease—An Emerging Role for Gut Microbiome. Alzheimer’s Dement. 2019, 15, 76–92. [Google Scholar] [CrossRef]
  89. Baloni, P.; Funk, C.C.; Yan, J.; Yurkovich, J.T.; Kueider-Paisley, A.; Nho, K.; Heinken, A.; Jia, W.; Mahmoudiandehkordi, S.; Louie, G.; et al. Metabolic Network Analysis Reveals Altered Bile Acid Synthesis and Metabolism in Alzheimer’s Disease. Cell Rep. Med. 2020, 1, 100138. [Google Scholar] [CrossRef] [PubMed]
  90. Wang, J.; Wei, R.; Xie, G.; Arnold, M.; Kueider-Paisley, A.; Louie, G.; Mahmoudian Dehkordi, S.; Blach, C.; Baillie, R.; Han, X.; et al. Peripheral Serum Metabolomic Profiles Inform Central Cognitive Impairment. Sci. Rep. 2020, 10, 14059. [Google Scholar] [CrossRef]
  91. Marksteiner, J.; Blasko, I.; Kemmler, G.; Koal, T.; Humpel, C. Bile Acid Quantification of 20 Plasma Metabolites Identifies Lithocholic Acid as a Putative Biomarker in Alzheimer’s Disease. Metabolomics 2018, 14, 1. [Google Scholar] [CrossRef] [PubMed]
  92. Griffiths, W.J.; Abdel-Khalik, J.; Yutuc, E.; Roman, G.; Warner, M.; Gustafsson, J.-Å.; Wang, Y. Concentrations of Bile Acid Precursors in Cerebrospinal Fluid of Alzheimer’s Disease Patients. Free Radic. Biol. Med. 2019, 134, 42–52. [Google Scholar] [CrossRef] [PubMed]
  93. Varma, V.R.; Wang, Y.; An, Y.; Varma, S.; Bilgel, M.; Doshi, J.; Legido-Quigley, C.; Delgado, J.C.; Oommen, A.M.; Roberts, J.A.; et al. Bile Acid Synthesis, Modulation, and Dementia: A Metabolomic, Transcriptomic, and Pharmacoepidemiologic Study. PLoS Med. 2021, 18, e1003615. [Google Scholar] [CrossRef] [PubMed]
  94. Ballestri, S.; Nascimbeni, F.; Baldelli, E.; Marrazzo, A.; Romagnoli, D.; Lonardo, A. NAFLD as a Sexual Dimorphic Disease: Role of Gender and Reproductive Status in the Development and Progression of Nonalcoholic Fatty Liver Disease and Inherent Cardiovascular Risk. Adv. Ther. 2017, 34, 1291–1326. [Google Scholar] [CrossRef]
  95. Bell, S.M.; Barnes, K.; Clemmens, H.; Al-Rafiah, A.R.; Al-Ofi, E.A.; Leech, V.; Bandmann, O.; Shaw, P.J.; Blackburn, D.J.; Ferraiuolo, L.; et al. Ursodeoxycholic Acid Improves Mitochondrial Function and Redistributes Drp1 in Fibroblasts from Patients with Either Sporadic or Familial Alzheimer’s Disease. J. Mol. Biol. 2018, 430, 3942–3953. [Google Scholar] [CrossRef]
  96. Joo, S.S.; Won, T.J.; Lee, D.I. Potential Role of Ursodeoxycholic Acid in Suppression of Nuclear Factor Kappa B in Microglial Cell Line (BV-2). Arch. Pharm. Res. 2004, 27, 954–960. [Google Scholar] [CrossRef]
  97. Solá, S.; Castro, R.E.; Laires, P.A.; Steer, C.J.; Rodrigues, C.M.P. Tauroursodeoxycholic Acid Prevents Amyloid-β Peptide-Induced Neuronal Death Via a Phosphatidylinositol 3-Kinase-Dependent Signaling Pathway. Mol. Med. 2003, 9, 226–234. [Google Scholar] [CrossRef]
  98. Nunes, A.F.; Amaral, J.D.; Lo, A.C.; Fonseca, M.B.; Viana, R.J.S.; Callaerts-Vegh, Z.; D’Hooge, R.; Rodrigues, C.M.P. TUDCA, a Bile Acid, Attenuates Amyloid Precursor Protein Processing and Amyloid-β Deposition in APP/PS1 Mice. Mol. Neurobiol. 2012, 45, 440–454. [Google Scholar] [CrossRef]
  99. Dionísio, P.A.; Amaral, J.D.; Ribeiro, M.F.; Lo, A.C.; D’Hooge, R.; Rodrigues, C.M.P. Amyloid-β Pathology Is Attenuated by Tauroursodeoxycholic Acid Treatment in APP/PS1 Mice after Disease Onset. Neurobiol. Aging 2015, 36, 228–240. [Google Scholar] [CrossRef] [Green Version]
  100. Zangerolamo, L.; Vettorazzi, J.F.; Solon, C.; Bronczek, G.A.; Engel, D.F.; Kurauti, M.A.; Soares, G.M.; Rodrigues, K.S.; Velloso, L.A.; Boschero, A.C.; et al. The Bile Acid TUDCA Improves Glucose Metabolism in Streptozotocin-Induced Alzheimer’s Disease Mice Model. Mol. Cell. Endocrinol. 2021, 521, 111116. [Google Scholar] [CrossRef]
  101. Bazzari, F.H.; Abdallah, D.M.; El-Abhar, H.S. Chenodeoxycholic Acid Ameliorates AlCl3-Induced Alzheimer’s Disease Neurotoxicity and Cognitive Deterioration via Enhanced Insulin Signaling in Rats. Molecules 2019, 24, 1992. [Google Scholar] [CrossRef] [PubMed]
  102. Beitz, J.M. Parkinson’s Disease: A Review. Front. Biosci. Sch. 2014, 6, 65–74. [Google Scholar] [CrossRef]
  103. Kalia, L.V.; Lang, A.E. Parkinson’s Disease. Lancet 2015, 386, 896–912. [Google Scholar] [CrossRef]
  104. Shao, Y.; Li, T.; Liu, Z.; Wang, X.; Xu, X.; Li, S.; Xu, G.; Le, W. Comprehensive Metabolic Profiling of Parkinson’s Disease by Liquid Chromatography-Mass Spectrometry. Mol. Neurodegener. 2021, 16, 4. [Google Scholar] [CrossRef] [PubMed]
  105. Li, P.; Killinger, B.A.; Ensink, E.; Beddows, I.; Yilmaz, A.; Lubben, N.; Lamp, J.; Schilthuis, M.; Vega, I.E.; Woltjer, R.; et al. Gut Microbiota Dysbiosis Is Associated with Elevated Bile Acids in Parkinson’s Disease. Metabolites 2021, 11, 29. [Google Scholar] [CrossRef] [PubMed]
  106. Hertel, J.; Harms, A.C.; Heinken, A.; Baldini, F.; Thinnes, C.C.; Glaab, E.; Vasco, D.A.; Pietzner, M.; Stewart, I.D.; Wareham, N.J.; et al. Integrated Analyses of Microbiome and Longitudinal Metabolome Data Reveal Microbial-Host Interactions on Sulfur Metabolism in Parkinson’s Disease. Cell Rep. 2019, 29, 1767–1777.e8. [Google Scholar] [CrossRef] [Green Version]
  107. Zhao, H.; Wang, C.; Zhao, N.; Li, W.; Yang, Z.; Liu, X.; Le, W.; Zhang, X. Potential Biomarkers of Parkinson’s Disease Revealed by Plasma Metabolic Profiling. J. Chromatogr. B 2018, 1081–1082, 101–108. [Google Scholar] [CrossRef]
  108. Griffiths, W.J.; Abdel-Khalik, J.; Moore, S.F.; Wijeyekoon, R.S.; Crick, P.J.; Yutuc, E.; Farrell, K.; Breen, D.P.; Williams-Gray, C.H.; Theofilopoulos, S.; et al. The Cerebrospinal Fluid Profile of Cholesterol Metabolites in Parkinson’s Disease and Their Association With Disease State and Clinical Features. Front. Aging Neurosci. 2021, 13, 685594. [Google Scholar] [CrossRef]
  109. Yilmaz, A.; Ugur, Z.; Ustun, I.; Akyol, S.; Bahado-Singh, R.O.; Maddens, M.; Aasly, J.O.; Graham, S.F. Metabolic Profiling of CSF from People Suffering from Sporadic and LRRK2 Parkinson’s Disease: A Pilot Study. Cells 2020, 9, 2394. [Google Scholar] [CrossRef]
  110. Yakhine-Diop, S.M.S.; Morales-García, J.A.; Niso-Santano, M.; González-Polo, R.A.; Uribe-Carretero, E.; Martinez-Chacon, G.; Durand, S.; Maiuri, M.C.; Aiastui, A.; Zulaica, M.; et al. Metabolic Alterations in Plasma from Patients with Familial and Idiopathic Parkinson’s Disease. Aging 2020, 12, 16690–16708. [Google Scholar] [CrossRef]
  111. Huang, A.; Martin, E.R.; Vance, J.M.; Cai, X. Detecting Genetic Interactions in Pathway-Based Genome-Wide Association Studies: Pathway-Based GWAS by the Group Empirical Bayesian Lasso. Genet. Epidemiol. 2014, 38, 300–309. [Google Scholar] [CrossRef]
  112. Mendes, M.O.; Rosa, A.I.; Carvalho, A.N.; Nunes, M.J.; Dionísio, P.; Rodrigues, E.; Costa, D.; Duarte-Silva, S.; Maciel, P.; Rodrigues, C.M.P.; et al. Neurotoxic Effects of MPTP on Mouse Cerebral Cortex: Modulation of Neuroinflammation as a Neuroprotective Strategy. Mol. Cell. Neurosci. 2019, 96, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Rosa, A.I.; Duarte-Silva, S.; Silva-Fernandes, A.; Nunes, M.J.; Carvalho, A.N.; Rodrigues, E.; Gama, M.J.; Rodrigues, C.M.P.; Maciel, P.; Castro-Caldas, M. Tauroursodeoxycholic Acid Improves Motor Symptoms in a Mouse Model of Parkinson’s Disease. Mol. Neurobiol. 2018, 55, 9139–9155. [Google Scholar] [CrossRef]
  114. Cuevas, E.; Burks, S.; Raymick, J.; Robinson, B.; Gómez-Crisóstomo, N.P.; Escudero-Lourdes, C.; Lopez, A.G.G.; Chigurupati, S.; Hanig, J.; Ferguson, S.A.; et al. Tauroursodeoxycholic Acid (TUDCA) Is Neuroprotective in a Chronic Mouse Model of Parkinson’s Disease. Nutr. Neurosci. 2020, 25, 1374–1391. [Google Scholar] [CrossRef] [PubMed]
  115. Rosa, A.I.; Fonseca, I.; Nunes, M.J.; Moreira, S.; Rodrigues, E.; Carvalho, A.N.; Rodrigues, C.M.P.; Gama, M.J.; Castro-Caldas, M. Novel Insights into the Antioxidant Role of Tauroursodeoxycholic Acid in Experimental Models of Parkinson’s Disease. Biochim. Biophys. Acta BBA—Mol. Basis Dis. 2017, 1863, 2171–2181. [Google Scholar] [CrossRef]
  116. Castro-Caldas, M.; Carvalho, A.N.; Rodrigues, E.; Henderson, C.J.; Wolf, C.R.; Rodrigues, C.M.P.; Gama, M.J. Tauroursodeoxycholic Acid Prevents MPTP-Induced Dopaminergic Cell Death in a Mouse Model of Parkinson’s Disease. Mol. Neurobiol. 2012, 46, 475–486. [Google Scholar] [CrossRef]
  117. Moreira, S.; Fonseca, I.; Nunes, M.J.; Rosa, A.; Lemos, L.; Rodrigues, E.; Carvalho, A.N.; Outeiro, T.F.; Rodrigues, C.M.P.; Gama, M.J.; et al. Nrf2 Activation by Tauroursodeoxycholic Acid in Experimental Models of Parkinson’s Disease. Exp. Neurol. 2017, 295, 77–87. [Google Scholar] [CrossRef]
  118. Launay, N.; Ruiz, M.; Grau, L.; Ortega, F.J.; Ilieva, E.V.; Martínez, J.J.; Galea, E.; Ferrer, I.; Knecht, E.; Pujol, A.; et al. Tauroursodeoxycholic Bile Acid Arrests Axonal Degeneration by Inhibiting the Unfolded Protein Response in X-Linked Adrenoleukodystrophy. Acta Neuropathol. 2017, 133, 283–301. [Google Scholar] [CrossRef] [Green Version]
  119. Duan, W.-M.; Rodrigures, C.M.P.; Zhao, L.-R.; Steer, C.J.; Low, W.C. Tauroursodeoxycholic Acid Improves the Survival and Function of Nigral Transplants in a Rat Model of Parkinson’s Disease. Cell Transpl. 2002, 11, 195–205. [Google Scholar] [CrossRef] [Green Version]
  120. Abdelkader, N.F.; Safar, M.M.; Salem, H.A. Ursodeoxycholic Acid Ameliorates Apoptotic Cascade in the Rotenone Model of Parkinson’s Disease: Modulation of Mitochondrial Perturbations. Mol. Neurobiol. 2016, 53, 810–817. [Google Scholar] [CrossRef]
  121. Chun, H.S.; Low, W.C. Ursodeoxycholic Acid Suppresses Mitochondria-Dependent Programmed Cell Death Induced by Sodium Nitroprusside in SH-SY5Y Cells. Toxicology 2012, 292, 105–112. [Google Scholar] [CrossRef] [PubMed]
  122. Qi, H.; Shen, D.; Jiang, C.; Wang, H.; Chang, M. Ursodeoxycholic Acid Protects Dopaminergic Neurons from Oxidative Stress via Regulating Mitochondrial Function, Autophagy, and Apoptosis in MPTP/MPP+-Induced Parkinson’s Disease. Neurosci. Lett. 2021, 741, 135493. [Google Scholar] [CrossRef] [PubMed]
  123. Sathe, A.G.; Tuite, P.; Chen, C.; Ma, Y.; Chen, W.; Cloyd, J.; Low, W.C.; Steer, C.J.; Lee, B.; Zhu, X.; et al. Pharmacokinetics, Safety, and Tolerability of Orally Administered Ursodeoxycholic Acid in Patients With Parkinson’s Disease—A Pilot Study. J. Clin. Pharmacol. 2020, 60, 744–750. [Google Scholar] [CrossRef]
  124. Payne, T.; Sassani, M.; Buckley, E.; Moll, S.; Anton, A.; Appleby, M.; Maru, S.; Taylor, R.; McNeill, A.; Hoggard, N.; et al. Ursodeoxycholic Acid as a Novel Disease-Modifying Treatment for Parkinson’s Disease: Protocol for a Two-Centre, Randomised, Double-Blind, Placebo-Controlled Trial, The “UP” Study. BMJ Open 2020, 10, e038911. [Google Scholar] [CrossRef] [PubMed]
  125. McMillin, M.; Frampton, G.; Quinn, M.; Ashfaq, S.; de los Santos, M.; Grant, S.; DeMorrow, S. Bile Acid Signaling Is Involved in the Neurological Decline in a Murine Model of Acute Liver Failure. Am. J. Pathol. 2016, 186, 312–323. [Google Scholar] [CrossRef]
  126. Mertens, K.L.; Kalsbeek, A.; Soeters, M.R.; Eggink, H.M. Bile Acid Signaling Pathways from the Enterohepatic Circulation to the Central Nervous System. Front. Neurosci. 2017, 11, 617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Keene, C.D.; Rodrigues, C.M.; Eich, T.; Linehan-Stieers, C.; Abt, A.; Kren, B.T.; Steer, C.J.; Low, W.C. A Bile Acid Protects against Motor and Cognitive Deficits and Reduces Striatal Degeneration in the 3-Nitropropionic Acid Model of Huntington’s Disease. Exp. Neurol. 2001, 171, 351–360. [Google Scholar] [CrossRef] [PubMed]
  128. Palmela, I.; Correia, L.; Silva, R.F.M.; Sasaki, H.; Kim, K.S.; Brites, D.; Brito, M.A. Hydrophilic Bile Acids Protect Human Blood-Brain Barrier Endothelial Cells from Disruption by Unconjugated Bilirubin: An in Vitro Study. Front. Neurosci. 2015, 9, 80. [Google Scholar] [CrossRef] [Green Version]
  129. Greenwood, J.; Adu, J.; Davey, A.J.; Abbott, N.J.; Bradbury, M.W. The Effect of Bile Salts on the Permeability and Ultrastructure of the Perfused, Energy-Depleted, Rat Blood-Brain Barrier. J. Cereb. Blood Flow Metab. 1991, 11, 644–654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Quinn, M.; McMillin, M.; Galindo, C.; Frampton, G.; Pae, H.Y.; DeMorrow, S. Bile Acids Permeabilize the Blood Brain Barrier after Bile Duct Ligation in Rats via Rac1-Dependent Mechanisms. Dig. Liver Dis. 2014, 46, 527–534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. McMillin, M.; Frampton, G.; Quinn, M.; Divan, A.; Grant, S.; Patel, N.; Newell-Rogers, K.; DeMorrow, S. Suppression of the HPA Axis During Cholestasis Can Be Attributed to Hypothalamic Bile Acid Signaling. Mol. Endocrinol. 2015, 29, 1720–1730. [Google Scholar] [CrossRef]
  132. Lund, E.G.; Guileyardo, J.M.; Russell, D.W. CDNA Cloning of Cholesterol 24-Hydroxylase, a Mediator of Cholesterol Homeostasis in the Brain. Proc. Natl. Acad. Sci. USA 1999, 96, 7238–7243. [Google Scholar] [CrossRef] [Green Version]
  133. Ogundare, M.; Theofilopoulos, S.; Lockhart, A.; Hall, L.J.; Arenas, E.; Sjövall, J.; Brenton, A.G.; Wang, Y.; Griffiths, W.J. Cerebrospinal Fluid Steroidomics: Are Bioactive Bile Acids Present in Brain? J. Biol. Chem. 2010, 285, 4666–4679. [Google Scholar] [CrossRef] [Green Version]
  134. Mano, N.; Goto, T.; Uchida, M.; Nishimura, K.; Ando, M.; Kobayashi, N.; Goto, J. Presence of Protein-Bound Unconjugated Bile Acids in the Cytoplasmic Fraction of Rat Brain. J. Lipid Res. 2004, 45, 295–300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Zuo, G.; Zhang, T.; Huang, L.; Araujo, C.; Peng, J.; Travis, Z.; Okada, T.; Ocak, U.; Zhang, G.; Tang, J.; et al. Activation of TGR5 with INT-777 Attenuates Oxidative Stress and Neuronal Apoptosis via CAMP/PKCε/ALDH2 Pathway after Subarachnoid Hemorrhage in Rats. Free Radic. Biol. Med. 2019, 143, 441–453. [Google Scholar] [CrossRef]
  136. McMillin, M.; Frampton, G.; Tobin, R.; Dusio, G.; Smith, J.; Shin, H.; Newell-Rogers, K.; Grant, S.; DeMorrow, S. TGR5 Signaling Reduces Neuroinflammation during Hepatic Encephalopathy. J. Neurochem. 2015, 135, 565–576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Chen, Q.; Ma, H.; Guo, X.; Liu, J.; Gui, T.; Gai, Z. Farnesoid X Receptor (FXR) Aggravates Amyloid-β-Triggered Apoptosis by Modulating the CAMP-Response Element-Binding Protein (CREB)/Brain-Derived Neurotrophic Factor (BDNF) Pathway In Vitro. Med. Sci. Monit. 2019, 25, 9335–9345. [Google Scholar] [CrossRef] [PubMed]
  138. Bao, H.; Li, H.; Jia, Y.; Xiao, Y.; Luo, S.; Zhang, D.; Han, L.; Dai, L.; Xiao, C.; Feng, L.; et al. Ganoderic Acid A Exerted Antidepressant-like Action through FXR Modulated NLRP3 Inflammasome and Synaptic Activity. Biochem. Pharmacol. 2021, 188, 114561. [Google Scholar] [CrossRef]
  139. Shan, H.-M.; Zang, M.; Zhang, Q.; Shi, R.-B.; Shi, X.-J.; Mamtilahun, M.; Liu, C.; Luo, L.; Tian, X.; Zhang, Z.; et al. Farnesoid X Receptor Knockout Protects Brain against Ischemic Injury through Reducing Neuronal Apoptosis in Mice. J. Neuroinflamm. 2020, 17, 164. [Google Scholar] [CrossRef]
  140. Hu, W.; Wu, J.; Ye, T.; Chen, Z.; Tao, J.; Tong, L.; Ma, K.; Wen, J.; Wang, H.; Huang, C. Farnesoid X Receptor-Mediated Cytoplasmic Translocation of CRTC2 Disrupts CREB-BDNF Signaling in Hippocampal CA1 and Leads to the Development of Depression-Like Behaviors in Mice. Int. J. Neuropsychopharmacol. 2020, 23, 673–686. [Google Scholar] [CrossRef] [PubMed]
  141. Chen, W.-G.; Zheng, J.-X.; Xu, X.; Hu, Y.-M.; Ma, Y.-M. Hippocampal FXR Plays a Role in the Pathogenesis of Depression: A Preliminary Study Based on Lentiviral Gene Modulation. Psychiatry Res. 2018, 264, 374–379. [Google Scholar] [CrossRef] [PubMed]
  142. Liang, H.; Matei, N.; McBride, D.W.; Xu, Y.; Zhou, Z.; Tang, J.; Luo, B.; Zhang, J.H. TGR5 Activation Attenuates Neuroinflammation via Pellino3 Inhibition of Caspase-8/NLRP3 after Middle Cerebral Artery Occlusion in Rats. J. Neuroinflamm. 2021, 18, 40. [Google Scholar] [CrossRef] [PubMed]
  143. Liang, H.; Matei, N.; McBride, D.W.; Xu, Y.; Tang, J.; Luo, B.; Zhang, J.H. Activation of TGR5 Protects Blood Brain Barrier via the BRCA1/Sirt1 Pathway after Middle Cerebral Artery Occlusion in Rats. J. Biomed. Sci. 2020, 27, 61. [Google Scholar] [CrossRef]
  144. Hu, X.; Yan, J.; Huang, L.; Araujo, C.; Peng, J.; Gao, L.; Liu, S.; Tang, J.; Zuo, G.; Zhang, J.H. INT-777 Attenuates NLRP3-ASC Inflammasome-Mediated Neuroinflammation via TGR5/CAMP/PKA Signaling Pathway after Subarachnoid Hemorrhage in Rats. Brain Behav. Immun. 2021, 91, 587–600. [Google Scholar] [CrossRef] [PubMed]
  145. Wang, H.; Tan, Y.-Z.; Mu, R.-H.; Tang, S.-S.; Liu, X.; Xing, S.-Y.; Long, Y.; Yuan, D.-H.; Hong, H. Takeda G Protein–Coupled Receptor 5 Modulates Depression-like Behaviors via Hippocampal CA3 Pyramidal Neurons Afferent to Dorsolateral Septum. Biol. Psychiatry 2021, 89, 1084–1095. [Google Scholar] [CrossRef] [PubMed]
  146. Huang, R.; Gao, Y.; Chen, J.; Duan, Q.; He, P.; Zhang, J.; Huang, H.; Zhang, Q.; Ma, G.; Zhang, Y.; et al. TGR5 Agonist INT-777 Alleviates Inflammatory Neurodegeneration in Parkinson’s Disease Mouse Model by Modulating Mitochondrial Dynamics in Microglia. Neuroscience 2022, 490, 100–119. [Google Scholar] [CrossRef] [PubMed]
  147. Mikami, T.; Kim, J.; Park, J.; Lee, H.; Yaicharoen, P.; Suidasari, S.; Yokozawa, M.; Yamauchi, K. Olive Leaf Extract Prevents Obesity, Cognitive Decline, and Depression and Improves Exercise Capacity in Mice. Sci. Rep. 2021, 11, 12495. [Google Scholar] [CrossRef] [PubMed]
  148. Frye, C.A.; Koonce, C.J.; Walf, A.A. Involvement of Pregnane Xenobiotic Receptor in Mating-Induced Allopregnanolone Formation in the Midbrain and Hippocampus and Brain-Derived Neurotrophic Factor in the Hippocampus among Female Rats. Psychopharmacology 2014, 231, 3375–3390. [Google Scholar] [CrossRef] [PubMed]
  149. Litwa, E.; Rzemieniec, J.; Wnuk, A.; Lason, W.; Krzeptowski, W.; Kajta, M. RXRα, PXR and CAR Xenobiotic Receptors Mediate the Apoptotic and Neurotoxic Actions of Nonylphenol in Mouse Hippocampal Cells. J. Steroid Biochem. Mol. Biol. 2016, 156, 43–52. [Google Scholar] [CrossRef] [PubMed]
  150. Garcion, E.; Sindji, L.; Leblondel, G.; Brachet, P.; Darcy, F. 1,25-Dihydroxyvitamin D3 Regulates the Synthesis of γ-Glutamyl Transpeptidase and Glutathione Levels in Rat Primary Astrocytes. J. Neurochem. 2002, 73, 859–866. [Google Scholar] [CrossRef] [PubMed]
  151. He, Y.; Wu, Z.; Lan, T.; Wang, Y.; Tian, Y.; Chen, X.; Li, Y.; Bai, M.; Liu, J.; Gong, X.; et al. The 25(OH)D/VDR Signaling May Play a Role in Major Depression. Biochem. Biophys. Res. Commun. 2020, 523, 405–410. [Google Scholar] [CrossRef] [PubMed]
  152. Garcion, E.; Sindji, L.; Montero-Menei, C.; Andre, C.; Brachet, P.; Darcy, F. Expression of Inducible Nitric Oxide Synthase during Rat Brain Inflammation: Regulation by 1,25-Dihydroxyvitamin D3. Glia 1998, 22, 282–294. [Google Scholar] [CrossRef]
  153. Xu, Y.; Liang, L. Vitamin D3/Vitamin D Receptor Signaling Mitigates Symptoms of Post-Stroke Depression in Mice by Upregulating Hippocampal BDNF Expression. Neurosci. Res. 2021, 170, 306–313. [Google Scholar] [CrossRef] [PubMed]
  154. Kim, H.; Shin, J.-Y.; Lee, Y.-S.; Yun, S.P.; Maeng, H.-J.; Lee, Y. Brain Endothelial P-Glycoprotein Level Is Reduced in Parkinson’s Disease via a Vitamin D Receptor-Dependent Pathway. Int. J. Mol. Sci. 2020, 21, 8538. [Google Scholar] [CrossRef]
  155. Cui, C.; Song, S.; Cui, J.; Feng, Y.; Gao, J.; Jiang, P. Vitamin D Receptor Activation Influences NADPH Oxidase (NOX2) Activity and Protects against Neurological Deficits and Apoptosis in a Rat Model of Traumatic Brain Injury. Oxidative Med. Cell. Longev. 2017, 2017, 9245702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Kimura, A.; Ohmori, T.; Kashiwakura, Y.; Ohkawa, R.; Madoiwa, S.; Mimuro, J.; Shimazaki, K.; Hoshino, Y.; Yatomi, Y.; Sakata, Y. Antagonism of Sphingosine 1-Phosphate Receptor-2 Enhances Migration of Neural Progenitor Cells Toward an Area of Brain Infarction. Stroke 2008, 39, 3411–3417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Cruz-Orengo, L.; Daniels, B.P.; Dorsey, D.; Basak, S.A.; Grajales-Reyes, J.G.; McCandless, E.E.; Piccio, L.; Schmidt, R.E.; Cross, A.H.; Crosby, S.D.; et al. Enhanced Sphingosine-1-Phosphate Receptor 2 Expression Underlies Female CNS Autoimmunity Susceptibility. J. Clin. Investig. 2014, 124, 2571–2584. [Google Scholar] [CrossRef] [Green Version]
  158. McMillin, M.; Frampton, G.; Grant, S.; Khan, S.; Diocares, J.; Petrescu, A.; Wyatt, A.; Kain, J.; Jefferson, B.; DeMorrow, S. Bile Acid-Mediated Sphingosine-1-Phosphate Receptor 2 Signaling Promotes Neuroinflammation during Hepatic Encephalopathy in Mice. Front. Cell. Neurosci. 2017, 11, 191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Chen, Z.; Doyle, T.M.; Luongo, L.; Largent-Milnes, T.M.; Giancotti, L.A.; Kolar, G.; Squillace, S.; Boccella, S.; Walker, J.K.; Pendleton, A.; et al. Sphingosine-1-Phosphate Receptor 1 Activation in Astrocytes Contributes to Neuropathic Pain. Proc. Natl. Acad. Sci. USA 2019, 116, 10557–10562. [Google Scholar] [CrossRef] [Green Version]
  160. Wu, C.-C.; Wang, L.-C.; Su, Y.-T.; Wei, W.-Y.; Tsai, K.-J. Synthetic A5β1 Integrin Ligand PHSRN Is Proangiogenic and Neuroprotective in Cerebral Ischemic Stroke. Biomaterials 2018, 185, 142–154. [Google Scholar] [CrossRef]
  161. Li, L.; Welser-Alves, J.; van der Flier, A.; Boroujerdi, A.; Hynes, R.O.; Milner, R. An Angiogenic Role for the A5β1 Integrin in Promoting Endothelial Cell Proliferation during Cerebral Hypoxia. Exp. Neurol. 2012, 237, 46–54. [Google Scholar] [CrossRef]
  162. Wang, L.; Zhang, X.; Liu, X.; Feng, G.; Fu, Y.; Milner, R.; Li, L. Overexpression of A5β1 Integrin and Angiopoietin-1 Co-Operatively Promote Blood-Brain Barrier Integrity and Angiogenesis Following Ischemic Stroke. Exp. Neurol. 2019, 321, 113042. [Google Scholar] [CrossRef] [PubMed]
  163. Han, Y.-M.; Kim, M.S.; Jo, J.; Shin, D.; Kwon, S.-H.; Seo, J.B.; Kang, D.; Lee, B.D.; Ryu, H.; Hwang, E.M.; et al. Decoding the Temporal Nature of Brain GR Activity in the NFκB Signal Transition Leading to Depressive-like Behavior. Mol. Psychiatry 2021, 26, 5087–5096. [Google Scholar] [CrossRef]
  164. Yu, H.; Guo, Y.; Zhao, Y.; Zhou, F.; Zhao, K.; Li, M.; Wen, J.; He, Z.; Zhu, X.; He, X. Both Insufficient and Excessive Glucocorticoid Receptor-Mediated Signaling Impair Neuronal Migration. J. Endocrinol. 2019, 242, 103–114. [Google Scholar] [CrossRef] [PubMed]
  165. Chen, H.; Lombès, M.; Le Menuet, D. Glucocorticoid Receptor Represses Brain-Derived Neurotrophic Factor Expression in Neuron-like Cells. Mol. Brain 2017, 10, 12. [Google Scholar] [CrossRef] [Green Version]
  166. Sun, X.-C.; Ren, X.-F.; Chen, L.; Gao, X.-Q.; Xie, J.-X.; Chen, W.-F. Glucocorticoid Receptor Is Involved in the Neuroprotective Effect of Ginsenoside Rg1 against Inflammation-Induced Dopaminergic Neuronal Degeneration in Substantia Nigra. J. Steroid Biochem. Mol. Biol. 2016, 155, 94–103. [Google Scholar] [CrossRef] [PubMed]
  167. Watanabe, M.; Houten, S.M.; Wang, L.; Moschetta, A.; Mangelsdorf, D.J.; Heyman, R.A.; Moore, D.D.; Auwerx, J. Bile Acids Lower Triglyceride Levels via a Pathway Involving FXR, SHP, and SREBP-1c. J. Clin. Investig. 2004, 113, 1408–1418. [Google Scholar] [CrossRef] [Green Version]
  168. Pineda Torra, I.; Claudel, T.; Duval, C.; Kosykh, V.; Fruchart, J.-C.; Staels, B. Bile Acids Induce the Expression of the Human Peroxisome Proliferator-Activated Receptor α Gene via Activation of the Farnesoid X Receptor. Mol. Endocrinol. 2003, 17, 259–272. [Google Scholar] [CrossRef] [Green Version]
  169. Siddiqui, M.S.; van Natta, M.L.; Connelly, M.A.; Vuppalanchi, R.; Neuschwander-Tetri, B.A.; Tonascia, J.; Guy, C.; Loomba, R.; Dasarathy, S.; Wattacheril, J.; et al. Impact of Obeticholic Acid on the Lipoprotein Profile in Patients with Non-Alcoholic Steatohepatitis. J. Hepatol. 2020, 72, 25–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Caron, S.; Huaman Samanez, C.; Dehondt, H.; Ploton, M.; Briand, O.; Lien, F.; Dorchies, E.; Dumont, J.; Postic, C.; Cariou, B.; et al. Farnesoid X Receptor Inhibits the Transcriptional Activity of Carbohydrate Response Element Binding Protein in Human Hepatocytes. Mol. Cell Biol. 2013, 33, 2202–2211. [Google Scholar] [CrossRef] [Green Version]
  171. Kir, S.; Beddow, S.A.; Samuel, V.T.; Miller, P.; Previs, S.F.; Suino-Powell, K.; Xu, H.E.; Shulman, G.I.; Kliewer, S.A.; Mangelsdorf, D.J. FGF19 as a Postprandial, Insulin-Independent Activator of Hepatic Protein and Glycogen Synthesis. Science 2011, 331, 1621–1624. [Google Scholar] [CrossRef] [PubMed]
  172. Potthoff, M.J.; Boney-Montoya, J.; Choi, M.; He, T.; Sunny, N.E.; Satapati, S.; Suino-Powell, K.; Xu, H.E.; Gerard, R.D.; Finck, B.N.; et al. FGF15/19 Regulates Hepatic Glucose Metabolism by Inhibiting the CREB-PGC-1α Pathway. Cell Metab. 2011, 13, 729–738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Holt, J.A.; Luo, G.; Billin, A.N.; Bisi, J.; McNeill, Y.Y.; Kozarsky, K.F.; Donahee, M.; Wang, D.Y.; Mansfield, T.A.; Kliewer, S.A.; et al. Definition of a Novel Growth Factor-Dependent Signal Cascade for the Suppression of Bile Acid Biosynthesis. Genes Dev. 2003, 17, 1581–1591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Inagaki, T.; Choi, M.; Moschetta, A.; Peng, L.; Cummins, C.L.; McDonald, J.G.; Luo, G.; Jones, S.A.; Goodwin, B.; Richardson, J.A.; et al. Fibroblast Growth Factor 15 Functions as an Enterohepatic Signal to Regulate Bile Acid Homeostasis. Cell Metab. 2005, 2, 217–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Hsuchou, H.; Pan, W.; Kastin, A.J. Fibroblast Growth Factor 19 Entry into Brain. Fluids Barriers CNS 2013, 10, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Bono, B.S.; Koziel Ly, N.K.; Miller, P.A.; Williams-Ikhenoba, J.; Dumiaty, Y.; Chee, M.J. Spatial Distribution of Beta-klotho MRNA in the Mouse Hypothalamus, Hippocampal Region, Subiculum, and Amygdala. J. Comp. Neurol. 2022, 530, 1634–1657. [Google Scholar] [CrossRef]
  177. Zhou, Y.; Yang, L.; Bo, C.; Zhang, X.; Zhang, J.; Li, Y. MicroRNA-9-3p Aggravates Cerebral Ischemia/Reperfusion Injury by Targeting Fibroblast Growth Factor 19 (FGF19) to Inactivate GSK-3β/Nrf2/ARE Signaling. Neuropsychiatr. Dis. Treat. 2021, 17, 1989–2002. [Google Scholar] [CrossRef]
  178. Goetz, R.; Beenken, A.; Ibrahimi, O.A.; Kalinina, J.; Olsen, S.K.; Eliseenkova, A.V.; Xu, C.; Neubert, T.A.; Zhang, F.; Linhardt, R.J.; et al. Molecular Insights into the Klotho-Dependent, Endocrine Mode of Action of Fibroblast Growth Factor 19 Subfamily Members. Mol. Cell Biol. 2007, 27, 3417–3428. [Google Scholar] [CrossRef] [Green Version]
  179. Asada, M.; Shinomiya, M.; Suzuki, M.; Honda, E.; Sugimoto, R.; Ikekita, M.; Imamura, T. Glycosaminoglycan Affinity of the Complete Fibroblast Growth Factor Family. Biochim. Biophys. Acta 2009, 1790, 40–48. [Google Scholar] [CrossRef]
  180. Harmer, N.J.; Pellegrini, L.; Chirgadze, D.; Fernandez-Recio, J.; Blundell, T.L. The Crystal Structure of Fibroblast Growth Factor (FGF) 19 Reveals Novel Features of the FGF Family and Offers a Structural Basis for Its Unusual Receptor Affinity. Biochemistry 2004, 43, 629–640. [Google Scholar] [CrossRef] [Green Version]
  181. Adams, A.C.; Coskun, T.; Rovira, A.R.I.; Schneider, M.A.; Raches, D.W.; Micanovic, R.; Bina, H.A.; Dunbar, J.D.; Kharitonenkov, A. Fundamentals of FGF19 & FGF21 Action in Vitro and in Vivo. PLoS ONE 2012, 7, e38438. [Google Scholar] [CrossRef]
  182. Wu, X.; Ge, H.; Lemon, B.; Vonderfecht, S.; Weiszmann, J.; Hecht, R.; Gupte, J.; Hager, T.; Wang, Z.; Lindberg, R.; et al. FGF19-Induced Hepatocyte Proliferation Is Mediated through FGFR4 Activation. J. Biol. Chem. 2010, 285, 5165–5170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Hultman, K.; Scarlett, J.M.; Baquero, A.F.; Cornea, A.; Zhang, Y.; Salinas, C.B.G.; Brown, J.; Morton, G.J.; Whalen, E.J.; Grove, K.L.; et al. The Central Fibroblast Growth Factor Receptor/Beta Klotho System: Comprehensive Mapping in Mus musculus and Comparisons to Nonhuman Primate and Human Samples Using an Automated in Situ Hybridization Platform. J. Comp. Neurol. 2019, 527, 2069–2085. [Google Scholar] [CrossRef] [PubMed]
  184. Talukdar, S.; Owen, B.M.; Song, P.; Hernandez, G.; Zhang, Y.; Zhou, Y.; Scott, W.T.; Paratala, B.; Turner, T.; Smith, A.; et al. FGF21 Regulates Sweet and Alcohol Preference. Cell Metab. 2016, 23, 344–349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Jensen-Cody, S.O.; Flippo, K.H.; Claflin, K.E.; Yavuz, Y.; Sapouckey, S.A.; Walters, G.C.; Usachev, Y.M.; Atasoy, D.; Gillum, M.P.; Potthoff, M.J. FGF21 Signals to Glutamatergic Neurons in the Ventromedial Hypothalamus to Suppress Carbohydrate Intake. Cell Metab. 2020, 32, 273–286.e6. [Google Scholar] [CrossRef] [PubMed]
  186. Bookout, A.L.; de Groot, M.H.M.; Owen, B.M.; Lee, S.; Gautron, L.; Lawrence, H.L.; Ding, X.; Elmquist, J.K.; Takahashi, J.S.; Mangelsdorf, D.J.; et al. FGF21 Regulates Metabolism and Circadian Behavior by Acting on the Nervous System. Nat. Med. 2013, 19, 1147–1152. [Google Scholar] [CrossRef]
  187. Fon Tacer, K.; Bookout, A.L.; Ding, X.; Kurosu, H.; John, G.B.; Wang, L.; Goetz, R.; Mohammadi, M.; Kuro-o, M.; Mangelsdorf, D.J.; et al. Research Resource: Comprehensive Expression Atlas of the Fibroblast Growth Factor System in Adult Mouse. Mol. Endocrinol. 2010, 24, 2050–2064. [Google Scholar] [CrossRef] [Green Version]
  188. Kawamata, Y.; Fujii, R.; Hosoya, M.; Harada, M.; Yoshida, H.; Miwa, M.; Fukusumi, S.; Habata, Y.; Itoh, T.; Shintani, Y.; et al. A G Protein-Coupled Receptor Responsive to Bile Acids. J. Biol. Chem. 2003, 278, 9435–9440. [Google Scholar] [CrossRef] [Green Version]
  189. Fiorucci, S.; Distrutti, E.; Carino, A.; Zampella, A.; Biagioli, M. Bile Acids and Their Receptors in Metabolic Disorders. Prog. Lipid Res. 2021, 82, 101094. [Google Scholar] [CrossRef]
  190. Wang, Y.-D.; Chen, W.-D.; Yu, D.; Forman, B.M.; Huang, W. The G-Protein-Coupled Bile Acid Receptor, Gpbar1 (TGR5), Negatively Regulates Hepatic Inflammatory Response through Antagonizing Nuclear Factor κ Light-Chain Enhancer of Activated B Cells (NF-ΚB) in Mice. Hepatology 2011, 54, 1421–1432. [Google Scholar] [CrossRef] [Green Version]
  191. Keitel, V.; Donner, M.; Winandy, S.; Kubitz, R.; Häussinger, D. Expression and Function of the Bile Acid Receptor TGR5 in Kupffer Cells. Biochem. Biophys. Res. Commun. 2008, 372, 78–84. [Google Scholar] [CrossRef]
  192. Fiorucci, S.; Distrutti, E. Linking Liver Metabolic and Vascular Disease via Bile Acid Signaling. Trends Mol. Med. 2022, 28, 51–66. [Google Scholar] [CrossRef]
  193. Arab, J.P.; Karpen, S.J.; Dawson, P.A.; Arrese, M.; Trauner, M. Bile Acids and Nonalcoholic Fatty Liver Disease: Molecular Insights and Therapeutic Perspectives. Hepatology 2017, 65, 350–362. [Google Scholar] [CrossRef] [PubMed]
  194. Richards, P.; Parker, H.E.; Adriaenssens, A.E.; Hodgson, J.M.; Cork, S.C.; Trapp, S.; Gribble, F.M.; Reimann, F. Identification and Characterization of GLP-1 Receptor–Expressing Cells Using a New Transgenic Mouse Model. Diabetes 2014, 63, 1224–1233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Ullmer, C.; Alvarez Sanchez, R.; Sprecher, U.; Raab, S.; Mattei, P.; Dehmlow, H.; Sewing, S.; Iglesias, A.; Beauchamp, J.; Conde-Knape, K. Systemic Bile Acid Sensing by G Protein-Coupled Bile Acid Receptor 1 (GPBAR1) Promotes PYY and GLP-1 Release: Mechanism of GPBAR1-Mediated PYY/GLP-1 Release. Br. J. Pharm. 2013, 169, 671–684. [Google Scholar] [CrossRef]
  196. Holst, J.J. The Physiology of Glucagon-like Peptide 1. Physiol. Rev. 2007, 87, 1409–1439. [Google Scholar] [CrossRef] [Green Version]
  197. Deacon, C.F.; Pridal, L.; Klarskov, L.; Olesen, M.; Holst, J.J. Glucagon-like Peptide 1 Undergoes Differential Tissue-Specific Metabolism in the Anesthetized Pig. Am. J. Physiol. 1996, 271, E458–E464. [Google Scholar] [CrossRef] [PubMed]
  198. Kastin, A.J.; Akerstrom, V.; Pan, W. Interactions of Glucagon-like Peptide-1 (GLP-1) with the Blood-Brain Barrier. J. Mol. Neurosci. 2002, 18, 7–14. [Google Scholar] [CrossRef]
  199. Cork, S.C.; Richards, J.E.; Holt, M.K.; Gribble, F.M.; Reimann, F.; Trapp, S. Distribution and Characterisation of Glucagon-like Peptide-1 Receptor Expressing Cells in the Mouse Brain. Mol. Metab. 2015, 4, 718–731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Holst, J.J.; Deacon, C.F. Glucagon-like Peptide-1 Mediates the Therapeutic Actions of DPP-IV Inhibitors. Diabetologia 2005, 48, 612–615. [Google Scholar] [CrossRef] [PubMed]
  201. Nishizawa, M.; Nakabayashi, H.; Uchida, K.; Nakagawa, A.; Niijima, A. The Hepatic Vagal Nerve Is Receptive to Incretin Hormone Glucagon-like Peptide-1, but Not to Glucose-Dependent Insulinotropic Polypeptide, in the Portal Vein. J. Auton. Nerv. Syst. 1996, 61, 149–154. [Google Scholar] [CrossRef]
  202. Llewellyn-Smith, I.J.; Reimann, F.; Gribble, F.M.; Trapp, S. Preproglucagon Neurons Project Widely to Autonomic Control Areas in the Mouse Brain. Neuroscience 2011, 180, 111–121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Wu, Y.; Wei, Z.; Li, Y.; Wei, C.; Li, Y.; Cheng, P.; Xu, H.; Li, Z.; Guo, R.; Qi, X.; et al. Perturbation of Ephrin Receptor Signaling and Glutamatergic Transmission in the Hypothalamus in Depression Using Proteomics Integrated With Metabolomics. Front. Neurosci. 2019, 13, 1359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Yang, C.; Zhou, C.; Li, J.; Chen, Z.; Shi, H.; Yang, W.; Qin, Y.; Lü, L.; Zhao, L.; Fang, L.; et al. Quantitative Proteomic Study of the Plasma Reveals Acute Phase Response and LXR/RXR and FXR/RXR Activation in the Chronic Unpredictable Mild Stress Mouse Model of Depression. Mol. Med. Rep. 2017, 17, 93–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Zhang, H.-M.; Wang, X.; Wu, Z.-H.; Liu, H.-L.; Chen, W.; Zhang, Z.-Z.; Chen, D.; Zeng, T.-S. Beneficial Effect of Farnesoid X Receptor Activation on Metabolism in a Diabetic Rat Model. Mol. Med. Rep. 2016, 13, 2135–2142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Wang, H.; Ge, C.; Zhou, J.; Guo, Y.; Cui, S.; Huang, N.; Yan, T.; Cao, L.; Che, Y.; Zheng, Q.; et al. Noncanonical Farnesoid X Receptor Signaling Inhibits Apoptosis and Impedes Liver Fibrosis. EBioMedicine 2018, 37, 322–333. [Google Scholar] [CrossRef] [Green Version]
  207. Wu, X.; Lv, Y.-G.; Du, Y.-F.; Chen, F.; Reed, M.N.; Hu, M.; Suppiramaniam, V.; Tang, S.-S.; Hong, H. Neuroprotective Effects of INT-777 against Aβ1–42-Induced Cognitive Impairment, Neuroinflammation, Apoptosis, and Synaptic Dysfunction in Mice. Brain Behav. Immun. 2018, 73, 533–545. [Google Scholar] [CrossRef]
  208. Yanguas-Casás, N.; Barreda-Manso, M.A.; Nieto-Sampedro, M.; Romero-Ramírez, L. TUDCA: An Agonist of the Bile Acid Receptor GPBAR1/TGR5 With Anti-Inflammatory Effects in Microglial Cells: Anti-Inflammatory Effect of Tudca in Microglia. J. Cell. Physiol. 2017, 232, 2231–2245. [Google Scholar] [CrossRef] [PubMed]
  209. Wu, H.; Yu, N.; Wang, X.; Yang, Y.; Liang, H. Tauroursodeoxycholic Acid Attenuates Neuronal Apoptosis via the TGR5/SIRT3 Pathway after Subarachnoid Hemorrhage in Rats. Biol. Res. 2020, 53, 56. [Google Scholar] [CrossRef] [PubMed]
  210. Wang, H.; Chen, J.; Hollister, K.; Sowers, L.C.; Forman, B.M. Endogenous Bile Acids Are Ligands for the Nuclear Receptor FXR/BAR. Mol. Cell 1999, 3, 543–553. [Google Scholar] [CrossRef]
  211. Younossi, Z.M.; Ratziu, V.; Loomba, R.; Rinella, M.; Anstee, Q.M.; Goodman, Z.; Bedossa, P.; Geier, A.; Beckebaum, S.; Newsome, P.N.; et al. Obeticholic Acid for the Treatment of Non-Alcoholic Steatohepatitis: Interim Analysis from a Multicentre, Randomised, Placebo-Controlled Phase 3 Trial. Lancet 2019, 394, 2184–2196. [Google Scholar] [CrossRef]
  212. Sayin, S.I.; Wahlström, A.; Felin, J.; Jäntti, S.; Marschall, H.-U.; Bamberg, K.; Angelin, B.; Hyötyläinen, T.; Orešič, M.; Bäckhed, F. Gut Microbiota Regulates Bile Acid Metabolism by Reducing the Levels of Tauro-Beta-Muricholic Acid, a Naturally Occurring FXR Antagonist. Cell Metab. 2013, 17, 225–235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Gonzalez, F.J.; Jiang, C.; Patterson, A.D. An Intestinal Microbiota-Farnesoid X Receptor Axis Modulates Metabolic Disease. Gastroenterology 2016, 151, 845–859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Yamada, T.; Sugimoto, K. Guggulsterone and Its Role in Chronic Diseases. Adv. Exp. Med. Biol. 2016, 929, 329–361. [Google Scholar] [CrossRef] [PubMed]
  215. Carter, B.A.; Taylor, O.A.; Prendergast, D.R.; Zimmerman, T.L.; Von Furstenberg, R.; Moore, D.D.; Karpen, S.J. Stigmasterol, a Soy Lipid-Derived Phytosterol, Is an Antagonist of the Bile Acid Nuclear Receptor FXR. Pediatr. Res. 2007, 62, 301–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Fiorucci, S.; Distrutti, E.; Bifulco, G.; D’Auria, M.V.; Zampella, A. Marine Sponge Steroids as Nuclear Receptor Ligands. Trends Pharm. Sci. 2012, 33, 591–601. [Google Scholar] [CrossRef] [PubMed]
  217. Gonzalez, F.J.; Jiang, C.; Xie, C.; Patterson, A.D. Intestinal Farnesoid X Receptor Signaling Modulates Metabolic Disease. Dig. Dis. 2017, 35, 178–184. [Google Scholar] [CrossRef] [PubMed]
  218. Jiang, C.; Xie, C.; Li, F.; Zhang, L.; Nichols, R.G.; Krausz, K.W.; Cai, J.; Qi, Y.; Fang, Z.-Z.; Takahashi, S.; et al. Intestinal Farnesoid X Receptor Signaling Promotes Nonalcoholic Fatty Liver Disease. J. Clin. Investig. 2015, 125, 386–402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Carino, A.; Marchianò, S.; Biagioli, M.; Bucci, M.; Vellecco, V.; Brancaleone, V.; Fiorucci, C.; Zampella, A.; Monti, M.C.; Distrutti, E.; et al. Agonism for the Bile Acid Receptor GPBAR1 Reverses Liver and Vascular Damage in a Mouse Model of Steatohepatitis. FASEB J. 2019, 33, 2809–2822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. Miyazaki-Anzai, S.; Masuda, M.; Kohno, S.; Levi, M.; Shiozaki, Y.; Keenan, A.L.; Miyazaki, M. Simultaneous Inhibition of FXR and TGR5 Exacerbates Atherosclerotic Formation. J. Lipid Res. 2018, 59, 1709–1713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  221. Miyazaki-Anzai, S.; Masuda, M.; Levi, M.; Keenan, A.L.; Miyazaki, M. Dual Activation of the Bile Acid Nuclear Receptor FXR and G-Protein-Coupled Receptor TGR5 Protects Mice against Atherosclerosis. PLoS ONE 2014, 9, e108270. [Google Scholar] [CrossRef] [PubMed]
  222. Lamers, C.; Merk, D.; Gabler, M.; Flesch, D.; Kaiser, A.; Schubert-Zsilavecz, M. SAR Studies on FXR Modulators Led to the Discovery of the First Combined FXR Antagonistic/TGR5 Agonistic Compound. Future Med. Chem. 2016, 8, 133–148. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Bile acid biosynthesis.
Figure 1. Bile acid biosynthesis.
Ijms 23 13045 g001
Figure 2. Summary of the effects of bile acid-mediated signaling on brain disease.
Figure 2. Summary of the effects of bile acid-mediated signaling on brain disease.
Ijms 23 13045 g002
Figure 3. Potential molecular targets of single and dual GPBAR1/FXR agonists in NAFLD and brain illnesses include FXR and GPBAR1. BAs in the liver trigger the activation of FXR, which then has a number of downstream effects, including the prevention of adipogenesis, the decrease in bile acid production, the encouragement of gluconeogenesis, the increase in gluconeogenesis, and anti-fibrosis. Numerous non-parenchymal cells in the liver, including Kupffer cells, express GPBAR1, which may lessen the inflammatory response in the liver. FXR increases bile acid absorption and cholesterol excretion in the terminal ileum. Additionally, FXR promotes the synthesis of FGF15/19, which functions by interacting with FGFR4 in hepatocytes. Additionally, activating GPBAR1 in the intestine increases the colonic motility and secretion, decreases stomach emptying, and stimulates the production of GLP-1 from the L cells, which in turn stimulates the release of insulin from the pancreatic β-cells. In vascular endothelial cells, GPBAR1 is capable of vasodilatation through the release of NO and H2S, reducing the vascular inflammation and decreasing the development of atherogenesis. Furthermore, FXR is capable of vasodilatation through the release of H2S. BAs could use both direct and indirect channels to communicate with the central nervous system (CNS). (A) BAs in the colon bypass the enterohepatic circulation and breach the BBB to reach the systemic circulation where they interact with brain receptors. (B) Only a small portion of FGF19 released by the enterocytes makes it to the portal vein and then into the body’s circulatory system. In order to interact with the brain’s FGF receptors (1–4) and bind β-klotho to create stable complexes, FGF19 must pass the BBB. (C) GPBAR1 activation results in the release of GLP-1 in the L-cells, which DPP-IV swiftly breaks down. The amount of intact GLP-1 that reaches the brain and interacts with the GLP-1 receptor (GLP-1R) is debatable. FXR plays a contradictory role in the brain by boosting insulin sensitivity, lowering inflammation, and decreasing BDNF, while also increasing cell death. In the brain, GPBAR1 reduces neuroinflammation, oxidative stress and apoptosis, decreases BBB injury, improves synaptic dysfunction, and reduces the mitochondrial damage and autophagy. Additionally, BAs stimulate GPBAR1 in adipose and muscular tissue, which raises thermogenesis and energy expenditure.
Figure 3. Potential molecular targets of single and dual GPBAR1/FXR agonists in NAFLD and brain illnesses include FXR and GPBAR1. BAs in the liver trigger the activation of FXR, which then has a number of downstream effects, including the prevention of adipogenesis, the decrease in bile acid production, the encouragement of gluconeogenesis, the increase in gluconeogenesis, and anti-fibrosis. Numerous non-parenchymal cells in the liver, including Kupffer cells, express GPBAR1, which may lessen the inflammatory response in the liver. FXR increases bile acid absorption and cholesterol excretion in the terminal ileum. Additionally, FXR promotes the synthesis of FGF15/19, which functions by interacting with FGFR4 in hepatocytes. Additionally, activating GPBAR1 in the intestine increases the colonic motility and secretion, decreases stomach emptying, and stimulates the production of GLP-1 from the L cells, which in turn stimulates the release of insulin from the pancreatic β-cells. In vascular endothelial cells, GPBAR1 is capable of vasodilatation through the release of NO and H2S, reducing the vascular inflammation and decreasing the development of atherogenesis. Furthermore, FXR is capable of vasodilatation through the release of H2S. BAs could use both direct and indirect channels to communicate with the central nervous system (CNS). (A) BAs in the colon bypass the enterohepatic circulation and breach the BBB to reach the systemic circulation where they interact with brain receptors. (B) Only a small portion of FGF19 released by the enterocytes makes it to the portal vein and then into the body’s circulatory system. In order to interact with the brain’s FGF receptors (1–4) and bind β-klotho to create stable complexes, FGF19 must pass the BBB. (C) GPBAR1 activation results in the release of GLP-1 in the L-cells, which DPP-IV swiftly breaks down. The amount of intact GLP-1 that reaches the brain and interacts with the GLP-1 receptor (GLP-1R) is debatable. FXR plays a contradictory role in the brain by boosting insulin sensitivity, lowering inflammation, and decreasing BDNF, while also increasing cell death. In the brain, GPBAR1 reduces neuroinflammation, oxidative stress and apoptosis, decreases BBB injury, improves synaptic dysfunction, and reduces the mitochondrial damage and autophagy. Additionally, BAs stimulate GPBAR1 in adipose and muscular tissue, which raises thermogenesis and energy expenditure.
Ijms 23 13045 g003
Table 2. FXR synthetic ligands and their clinical status.
Table 2. FXR synthetic ligands and their clinical status.
AgonistClinical TrialsStatus
Obeticholic acid
(6-ECDCA, INT747)
NCT01265498Phase IIb: OCA as a FXR ligand in a NASH treatment trial (FLINT)
NCT03836937The function of OCA in NAFLD patients with elevated ALT
NCT02548351Phase III: Randomized global study to assess the impact of OCA for fibrosis on NASH
NCT03439254Phase III: Evaluating the OCA’s safety and effectiveness in patients who have compensated cirrhosis, as a result of NASH
Tropifexor (LJN452)NCT03681457Phase I: Tropifexor pharmacokinetics in patients with mild, moderate, and severe hepatic impairment
NCT04065841Phase II: Patients with NASH and hepatic fibrosis: efficacy, safety, and tolerability of the combination of Tropifexor and Licogliflozin and each single agent
NCT03517540Phase II: Adult treatment for NASH and liver fibrosis using LJN452 and Cenicriviroc: a study of safety, tolerability, and effectiveness
Cilofexor (GS-9674)NCT02808312Phase I: Adults’ pharmacokinetics and pharmacodynamics of Cilofexor in those with normal and abnormal liver function
NCT02654002Phase I: Safety, tolerability, pharmacokinetics, and pharmacodynamics, as well as the impact of food on these factors of GS-9674 in healthy volunteers
NCT02854605Phase II: Safety, tolerance, and efficacy of GS-9674 in participants with NASH
NCT04971785Phase II: Semaglutide, Cilofexor, and Firsocostat fixed-dose combination safety and efficacy study in patients with compensated cirrhosis due to NASH
NCT02781584Phase II: Selonsertib, Firsocostat, and Cilofexor in adults with NASH: safety, tolerability, and efficacy
NCT03987074Phase II: In participants with NASH, the safety, tolerability, and the efficacy of monotherapy and combination regimens were examined
NCT03449446Phase IIb: In people with compensated cirrhosis or bridging fibrosis, the effectiveness and safety of the drugs Selonsertib, Firsocostat, Cilofexor, and combinations for NASH, are being evaluated
EDP-305NCT03748628Phase I: The AME (Absorption, Metabolism, and Excretion) study of [14C]EDP-305 in healthy male subjects
NCT02918929Phase I: EDP 305 research in subjects with and without presumed NAFLD
NCT03207425Phase I: Comparison of participants in the EDP-305 study with mild and moderate hepatic impairment versus healthy people
NCT03421431Phase II: Safety, tolerability, pharmacokinetics and effectiveness of EDP-305 in patients with NASH
NCT04378010Phase IIb: Safety and effectiveness of EDP-305 in patients with liver-biopsy-confirmed NASH
Px-102NCT01998672Phase I: Study using multiple ascending oral doses and Px-102
Px-104NCT01999101Phase II: FXR agonist safety pilot study in NAFLD patients
Vonafexor (EYP001)NCT03976687Phase I: Examination of the safety, tolerability, pharmacokinetics, and pharmacodynamics of EYP001a in NASH patients and healthy volunteers
NCT03812029Phase IIa: EYP001a’s effectiveness, tolerability, and pharmacokinetics in NASH patients
MET409NCT04702490Phase IIa: In patients with type 2 diabetes and NASH, MET409 may be used alone or in combination with empagliflozin
TERN-101 (LY2562175)NCT04328077Phase IIa: An investigation on the pharmacokinetics, efficacy, and safety of TERN-101 in people with NASH but without cirrhosis
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ren, Z.-L.; Li, C.-X.; Ma, C.-Y.; Chen, D.; Chen, J.-H.; Xu, W.-X.; Chen, C.-A.; Cheng, F.-F.; Wang, X.-Q. Linking Nonalcoholic Fatty Liver Disease and Brain Disease: Focusing on Bile Acid Signaling. Int. J. Mol. Sci. 2022, 23, 13045. https://doi.org/10.3390/ijms232113045

AMA Style

Ren Z-L, Li C-X, Ma C-Y, Chen D, Chen J-H, Xu W-X, Chen C-A, Cheng F-F, Wang X-Q. Linking Nonalcoholic Fatty Liver Disease and Brain Disease: Focusing on Bile Acid Signaling. International Journal of Molecular Sciences. 2022; 23(21):13045. https://doi.org/10.3390/ijms232113045

Chicago/Turabian Style

Ren, Zi-Lin, Chang-Xiang Li, Chong-Yang Ma, Dan Chen, Jia-Hui Chen, Wen-Xiu Xu, Cong-Ai Chen, Fa-Feng Cheng, and Xue-Qian Wang. 2022. "Linking Nonalcoholic Fatty Liver Disease and Brain Disease: Focusing on Bile Acid Signaling" International Journal of Molecular Sciences 23, no. 21: 13045. https://doi.org/10.3390/ijms232113045

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

Article Metrics

Back to TopTop