Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 712
1.
J Neurosci Res ; 102(5): e25337, 2024 May.
Article En | MEDLINE | ID: mdl-38680084

Hepatic encephalopathy (HE) is defined as decline in neurological function during chronic liver disease (CLD). Alcohol is a major etiological factor in the pathogenesis of fibrosis/cirrhosis and has also been documented to directly impact the brain. However, the role of alcohol in the development of HE in CLD remains unclear. Here, we investigated the impact of excessive alcohol administration on neurological deterioration in rats with CLD. Starting day 7 post-BDL surgery, rats were administered alcohol twice daily (51% v/v ethanol, 3 g/kg, via gavage) for 4 weeks. Motor coordination was assessed weekly using rotarod and anxiety-like behavior was evaluated with open field and elevated plus maze at 5 weeks. Upon sacrifice, brains were collected for western blot and immunohistochemical analyses to investigate neuronal integrity and oxidative stress status. Alcohol worsened motor coordination performance and increased anxiety-like behavior in BDL rats. Impairments were associated with decreased neuronal markers of NeuN and SMI311, increased apoptotic markers of cleaved/pro-caspase-3 and Bax/Bcl2, increased necroptosis markers of pRIP3 and pMLKL, decreased total antioxidant capacity (TAC), and increased 4-hydroxynonenal (4-HNE)modified proteins in the cerebellum of BDL-alcohol rats when compared to respective controls. Immunofluorescence confirmed the colocalization of cleaved caspase-3 and pMLKL in the granular neurons of the cerebellum of BDL-alcohol rats. Excessive alcohol consumption exacerbates HE which leads to associated apoptotic and necroptotic neuronal loss in the cerebellum of BDL-alcohol rats. Additionally, higher levels of 4-HNE and decreased TAC in the cerebellum of BDL-alcohol rats suggest oxidative stress is the triggering factor of apoptotic and necroptotic neuronal loss/injury.


Ethanol , Hepatic Encephalopathy , Neurons , Oxidative Stress , Animals , Male , Hepatic Encephalopathy/pathology , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/metabolism , Ethanol/toxicity , Ethanol/adverse effects , Rats , Neurons/pathology , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Cell Death/drug effects , Rats, Sprague-Dawley , Apoptosis/drug effects , Anxiety/etiology
2.
Life Sci ; 344: 122559, 2024 May 01.
Article En | MEDLINE | ID: mdl-38479595

AIMS: Cinnamaldehyde (CA), the main active constituent of cinnamon oil, is reported to have neuroprotective effects. However, the potential benefits of CA for brain protection in hepatic encephalopathy (HE) are still not understood. Thus, the present study investigates the possible ameliorative effect of CA (70 mg/kg/day, I.P.) either alone or in combination with lactulose (Lac) (5.3 g/kg/day, oral) against thioacetamide (TAA)-induced hepatic encephalopathy in rats. MATERIALS AND METHODS: For induction of HE, TAA (200 mg/kg) was intraperitoneally administered for 1 week at alternative days. CA, Lac and Lac+CA were administered for 14 days prior to and for further 7 days together with TAA injection. KEY FINDINGS: CA, Lac and Lac+CA combination effectively attenuated TAA-induced HE; as indicated by the improvement in behavioral tests, mitigation of pathological abnormalities in both liver and brain, the significant reduction in serum hyperammonemia and amelioration in liver function biomarkers; ALT and AST. This was accompanied with a substantial restoration of redox state in liver and brain; MDA and GSH levels. Moreover, CA, Lac and Lac+CA combination reduced neuroinflammation as demonstrated by the notable attenuation of P2X7R, NLRP3, caspase-1, IL-1ß, GFAP and Iba1 brain levels, as well as the amelioration of brain edema as manifested by reduction in AQP4 levels in brain. SIGNIFICANCE: Our study has demonstrated that CA in combination with Lac possesses a superior neuroprotective effect over Lac alone against TAA-induced HE by attenuation of P2X7R/NLRP3 mediated neuroinflammation and relieving brain edema.


Acrolein/analogs & derivatives , Brain Edema , Hepatic Encephalopathy , Rats , Animals , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/drug therapy , Lactulose/adverse effects , Inflammasomes , Thioacetamide/pharmacology , Brain Edema/pathology , NLR Family, Pyrin Domain-Containing 3 Protein , Neuroinflammatory Diseases , Rats, Wistar , Liver
3.
J Ethnopharmacol ; 323: 117700, 2024 Apr 06.
Article En | MEDLINE | ID: mdl-38176666

ETHNOPHARMACOLOGICAL RELEVANCE: Tinospora cordifolia (TC) a potential medicinal herb, has been ethnobotanically used as an eco-friendly supplement to manage various diseases, including cerebral fever. Earlier studies have shown that TC exhibits diverse beneficial effects, including hepatoprotective and neuroprotective effects. However, the effects of TC remain unexplored in animal models of encephalopathy including hepatic encephalopathy (HE). AIM OF THE STUDY: To evaluate the effects of TC stem extract against thioacetamide (TAA)-induced behavioural and molecular alterations in HE rats. METHODS AND MATERIALS: The extract was preliminarily screened through phytochemical and HR-LC/MS analysis. Animals were pre-treated with TC extract at doses 30 and 100 mg/kg, orally. Following 7 days of TC pre-treatment, HE was induced by administering TAA (300 mg/kg, i. p. thrice). Behavioural assessments were performed after 56 h of TAA first dose. The animals were then sacrificed to assess biochemical parameters in serum, liver and brain. Liver tissue was used for immunoblotting and histological studies to evaluate inflammatory and fibrotic signalling. Moreover, brain tissue was used to evaluate brain edema, activation of glial cells (GFAP, IBA-1) and NF-κB/NLRP3 downstream signalling via immunoblotting and immunohistochemical analysis in cortex and hippocampus. RESULTS: The pre-treatment with TC extract effective mitigated TAA-induced behavioural alterations, lowered serum LFT (AST, ALT, ALP, bilirubin) and oxidative stress markers in liver and brain. TC treatment significantly modulated hyperammonemia, cerebral edema and preserved the integrity of BBB proteins in HE animals. TC treatment attenuated TAA-induced histological changes, tissue inflammation (pNF-κB (p65), TNF-α, NLRP3) and fibrosis (collagen, α-SMA) in liver. In addition, immunoblotting analysis revealed TC pre-treatment inhibited fibrotic proteins such as vimentin, TGF-ß1 and pSmad2/3 in the liver. Our study further showed that TC treatment downregulated the expression of MAPK/NF-κB inflammatory signalling, as well as GFAP and IBA-1 (glial cell markers) in cortex and hippocampus of TAA-intoxicated rats. Additionally, TC-treated animals exhibited reduced expression of caspase3/9 and BAX induced by TAA. CONCLUSION: This study revealed promising insights on the protective effects of TC against HE. The findings clearly demonstrated that the significant inhibition of MAPK/NF-κB signalling and glial cell activation could be responsible for the observed beneficial effects of TC in TAA-induced HE rats.


Hepatic Encephalopathy , Hyperammonemia , Tinospora , Rats , Animals , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/drug therapy , Hepatic Encephalopathy/prevention & control , Thioacetamide/toxicity , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NF-kappa B/metabolism , Hyperammonemia/metabolism , Hyperammonemia/pathology , Liver , Oxidative Stress , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Plant Extracts/chemistry
5.
Biochemistry (Mosc) ; 88(9): 1404-1415, 2023 Sep.
Article En | MEDLINE | ID: mdl-37770406

Hepatic encephalopathy (HE), a neuropsychiatric disorder developing in patients with severe hepatic dysfunction, has been known for more than a century. However, pathogenetic mechanisms of cerebral dysfunction associated with liver disease are still poorly understood. There is a consensus that the primary cause of HE is accumulation of ammonia in the brain as a result of impaired liver detoxification capacity or the portosystemic shunt. Current evidence suggests that ammonia toxicity is mediated by hyperactivation of glutamate receptors, mainly N-methyl-D-aspartate receptors (NMDARs), and affects brain aerobic metabolism, which provides energy for multiple specific functions and neuronal viability. Recent reports on the presence of functional NMDARs in erythrocytes and the data on the deviations of blood parameters from their normal ranges indicate impaired hemodynamics and reduced oxygen-carrying capacity of erythrocytes in most patients with HE, thus suggesting a relationship between erythrocyte damage and cerebral dysfunction. In order to understand how hyperammonemia (HA)-induced disturbances in the energy metabolism in the brain (which needs a constant supply of large amounts of oxygen in the blood) lead to encephalopathy, it is necessary to reveal ammonia-induced impairments in the energy metabolism and antioxidant defense system of erythrocytes and to explore a potential role of ammonia in reduced brain oxygenation. To identify the said missing link, the activities of antioxidant enzymes and concentrations of reduced glutathione (GSH), oxidized glutathione (GSSG), and H2O2 were measured in the erythrocytes of rats with HA that were injected with the noncompetitive NMDAR antagonist MK-801. We found that in rats with HA, ammonia was accumulated in erythrocytes (cells lacking ammonia removal enzymes), which made them more susceptible to the prooxidant environment created during oxidative stress. This effect was completely or partially inhibited by MK-801. The data obtained might help to identify the risk factors in cognitive disorders and facilitate prediction of unfavorable outcomes of hypoperfusion in patients with a blood elevated ammonia concentration.


Hepatic Encephalopathy , Receptors, N-Methyl-D-Aspartate , Humans , Rats , Animals , Receptors, N-Methyl-D-Aspartate/metabolism , Antioxidants , Ammonia/toxicity , Ammonia/metabolism , Dizocilpine Maleate/pharmacology , Hydrogen Peroxide/metabolism , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/metabolism , Erythrocytes/metabolism , Oxygen/metabolism
6.
BMC Complement Med Ther ; 23(1): 265, 2023 Jul 25.
Article En | MEDLINE | ID: mdl-37491245

BACKGROUND: Hepatic encephalopathy (HE) is a consequence of chronic or acute liver diseases. This study evaluates the combined effect of gallic acid (GA), and metformin (Met) on the liver and brain damage associated with HE. METHODS: Acute HE was induced by a single dose of thioacetamide (TAA) (300 mg/kg) as an I.P. injection. Treated groups received GA group (100 mg/kg/day, p.o), Met (200 mg/kg/day, p.o), or their combination for 25 consecutive days before TAA injection. RESULTS: The administration of TAA induced various biochemical and histopathological alterations. In contrast, treatment with GA either alone or combined with Met resulted in improved liver functions by the significant reduction in serum ALT, AST, and ALP activities, and ammonia levels. Inflammatory mediators; TNF-α, IL-6, and NFkß levels were decreased by these treatments as well as apoptotic cascade via down-regulation of FAS and caspase-3 (CASP-3) expression in hepatic tissues. Furthermore, GA and Met either alone or combined protected the liver and brain tissues from damage by increased glutathione concentration while decreasing malondialdehyde. In addition, it was accompanied by the improvement of the brain neurotransmitter profile via the restoration of norepinephrine, dopamine, and serotonin levels. Based on our data, this is the first study to report a novel combined hepatoprotective and cognitive enhancing effect of GA and Met against TAA-induced acute liver and brain injury. CONCLUSION: GA and Met combination resulted in a prominent improvement in HE complications, relative to monotherapy. Both agents potentiated the antioxidant, anti-inflammatory, and anti-apoptotic effects of each other.


Hepatic Encephalopathy , Metformin , Rats , Animals , Hepatic Encephalopathy/drug therapy , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/pathology , NF-kappa B/metabolism , Thioacetamide/adverse effects , Gallic Acid/pharmacology , Caspase 3/metabolism , Metformin/pharmacology , Rats, Wistar , Inflammation/drug therapy , Inflammation/metabolism , Oxidative Stress , Signal Transduction , Apoptosis
7.
Environ Sci Pollut Res Int ; 30(35): 84791-84804, 2023 Jul.
Article En | MEDLINE | ID: mdl-37378730

The aim of the present study was to investigate the impact of arginine (ARG), a nitric oxide (NO) precursor, on thioacetamide (TAA)-induced hepatic encephalopathy (HE) in rats by injection of TAA (100 mg/kg, i.p) three times per week for six consecutive weeks. TAA-injected rats were administered ARG (100 mg/kg; p.o.) concurrently with TAA for the six consecutive weeks. Blood samples were withdrawn, and rats were sacrificed; liver and brain tissues were isolated. Results of the present study demonstrated that ARG administration to TAA-injected rats revealed a restoration in the serum and brain ammonia levels as well as serum aspartate transaminase, alanine transaminase, and alkaline phosphatase and total bilirubin levels as well as behavioral alterations evidenced by restoration in locomotor activity, motor skill performance, and memory impairment. ARG showed also improvement in the hepatic and neuro-biochemical values, pro-inflammatory cytokines, and oxidative stress biomarkers. All these results were confirmed by histopathological evaluation as well as ultrastructural imaging of the cerebellum using a transmission electron microscope. Furthermore, treatment with ARG could ameliorate the immunological reactivity of nuclear factor erythroid-2-related factor 2 (Nrf2) and cleaved caspase-3 proteins in the cerebellum and hepatic tissues. From all the previous results, it can be fulfilled that ARG showed a beneficial role in modulating the adverse complications associated with TAA-induced HE in rats via reducing hyperammonemia and downregulating nuclear factor kappa B (NF-κB)-mediated apoptosis.


Hepatic Encephalopathy , Rats , Animals , Hepatic Encephalopathy/chemically induced , NF-kappa B/metabolism , Thioacetamide/toxicity , Nitric Oxide/metabolism , Down-Regulation , Liver/metabolism , Arginine/adverse effects , Arginine/metabolism , Oxidative Stress
8.
Toxicology ; 492: 153542, 2023 06 15.
Article En | MEDLINE | ID: mdl-37150287

Indole-3-carbinol (I3C) is reported to have hepatic and neuroprotective properties. However, the I3C role in the protection of the liver and brain in the pathological condition of hepatic encephalopathy has not been investigated. Therefore, in the present study, we have assessed the hepatic and neuroprotective roles of I3C against thioacetamide (TAA)- induced hepatic encephalopathy in Wistar rats. TAA (300 mg/kg) was intraperitoneally administered to Wistar rats to induce hepatic encephalopathy. The elevated levels of ammonia in the blood, liver, and brain were substantially lowered by I3C treatment (25, 50, and 100 mg/kg, oral, 7 days). I3C significantly ameliorated the TAA-induced liver dysfunction by decreasing the alanine transaminase, aspartate transaminase, and alkaline phosphatase enzymes and reduced the elevated cytochrome P4502E1 (CYP2E1) activity in the liver and brain. Further, I3C alleviated mitochondrial dysfunction and oxidative stress in the brain. I3C treatment improved the anti-inflammatory cytokine interleukin (IL)- 10 while reducing inflammatory cytokines such as tumor necrosis factor-1 and IL-6 in hepatic encephalopathy rats. I3C reduced the levels of apoptotic indicators mediated by the mitochondria, including cytochrome c, caspase 9, and caspase 3. Concurrently, I3C mitigated the liver and brain histological abnormalities in hepatic encephalopathy rats. Therefore, the present study concluded that the I3C protected the liver and brain from TAA-induced hepatic encephalopathy injury by inhibiting CYP2E1 enzyme activity and decreasing ammonia, oxidative stress, inflammation, and apoptosis. The present study provides preclinical validation of I3C use as hepatic and neuroprotective for hepatic encephalopathy management.


Hepatic Encephalopathy , Rats , Animals , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/drug therapy , Hepatic Encephalopathy/prevention & control , Thioacetamide/toxicity , Rats, Wistar , Ammonia/adverse effects , Ammonia/metabolism , Cytochrome P-450 CYP2E1/metabolism , Liver/metabolism , Oxidative Stress , Cytokines/metabolism
9.
Neuropharmacology ; 236: 109575, 2023 Sep 15.
Article En | MEDLINE | ID: mdl-37201650

Hepatic encephalopathy (HE) is a life-threatening disease caused by acute or chronic liver failure manifested by aberrant CNS changes. In the present study, we aimed to explore the neuroprotective effect of lactoferrin (LF) against thioacetamide (TAA)-induced HE in rats. Animals were divided into four groups, control, LF control, TAA-induced HE, and LF treatment, where LF was administered (300 mg/kg, p.o.) for 15 days in groups 2 and 4 meanwhile, TAA (200 mg/kg, i.p.) was given as two injections on days 13 and 15 for the 3rd and 4th groups. Pretreatment with LF significantly improved liver function observed as a marked decline in serum AST, ALT, and ammonia, together with lowering brain ammonia and enhancing motor coordination as well as cognitive performance. Restoration of brain oxidative status was also noted in the LF-treated group, where lipid peroxidation was hampered, and antioxidant parameters, Nrf2, HO-1, and GSH, were increased. Additionally, LF downregulated HMGB1, TLR-4, MyD88, and NF-κB signaling pathways, together with reducing inflammatory cytokine, TNF-α, and enhancing brain BDNF levels. Moreover, the histopathology of brain and liver tissues revealed that LF alleviated TAA-induced liver and brain deficits. In conclusion, the promising results of LF in attenuating HMGB1/TLR-4/MyD88 signaling highlight its neuroprotective role against HE associated with acute liver injury via ameliorating neuroinflammation, oxidative stress, and stimulating neurogenesis.


HMGB1 Protein , Hepatic Encephalopathy , Animals , Rats , Ammonia/metabolism , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/drug therapy , HMGB1 Protein/metabolism , Lactoferrin/metabolism , Liver , Myeloid Differentiation Factor 88/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Rats, Wistar , Thioacetamide/toxicity , Toll-Like Receptor 4/metabolism
10.
Int J Mol Sci ; 24(4)2023 Feb 11.
Article En | MEDLINE | ID: mdl-36835059

The delayed consequences of the influence of hepatic encephalopathy (HE) on the metabolism of animals have not been studied enough. We have previously shown that the development of acute HE under the influence of the thioacetamide (TAA) toxin is accompanied by pathological changes in the liver, an imbalance in CoA and acetyl CoA, as well as a number of metabolites of the TCA cycle. This paper discusses the change in the balance of amino acids (AAs) and related metabolites, as well as the activity of glutamine transaminase (GTK) and ω-amidase enzymes in the vital organs of animals 6 days after a single exposure to TAA. The balance of the main AAs in blood plasma, liver, kidney, and brain samples of control (n = 3) and TAA-induced groups (n = 13) of rats that received the toxin at doses of 200, 400, and 600 mg/kg was considered. Despite the apparent physiological recovery of the rats at the time of sampling, a residual imbalance in AA and associated enzymes persisted. The data obtained give an idea of the metabolic trends in the body of rats after their physiological recovery from TAA exposure and may be useful for prognostic purposes when choosing the necessary therapeutic agents.


Amino Acids , Hepatic Encephalopathy , Animals , Rats , Amino Acids/metabolism , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/pathology , Liver/metabolism , Liver/pathology , Rats, Wistar , Thioacetamide/adverse effects
11.
Int J Mol Sci ; 24(2)2023 Jan 10.
Article En | MEDLINE | ID: mdl-36674898

Exposure to the toxin thioacetamide (TAA) causes acute hepatic encephalopathy (HE), changes in the functioning of systemic organs, and an imbalance in a number of energy metabolites. The deferred effects after acute HE development are poorly understood. The study considers the balance of the tricarboxylic acid (TCA) cycle metabolites in the blood plasma, liver, kidneys, and brain tissues of rats in the post-rehabilitation period. The samples of the control (n = 3) and TAA-induced groups of rats (n = 13) were collected six days after the administration of a single intraperitoneal TAA injection at doses of 200, 400, and 600 mg/kg. Despite the complete physiological recovery of rats by this date, a residual imbalance of metabolites in all the vital organs was noted. The results obtained showed a trend of stabilizing processes in the main organs of the animals and permit the use of these data both for prognostic purposes and the choice of potential therapeutic agents.


Brain Diseases , Hepatic Encephalopathy , Liver Failure, Acute , Rats , Animals , Hepatic Encephalopathy/chemically induced , Thioacetamide/toxicity , Tricarboxylic Acids/metabolism , Liver/metabolism , Liver Failure, Acute/chemically induced , Brain Diseases/metabolism
12.
Life Sci ; 313: 121280, 2023 Jan 15.
Article En | MEDLINE | ID: mdl-36526046

Hepatic encephalopathy (HE) is a serious neurological disorder which might occur in both acute and chronic liver injury. AIMS: This study was carried out to explore the protective effects of hesperidin against experimentally induced HE. MAIN METHODS: Rats were sorted into four groups each of six; Normal group, TAA group: rats were administered 350 mg/kg of TAA i.p. from day 5 to day 7. TAA+ Hesp 100 group: rats were administered hesperidin 100 mg/kg/day orally for 7 days along with i.p TAA injection 350 mg/kg from day 5 to 7. TAA+ Hesp 200 group: rats were administered hesperidin 200 mg/kg/day orally for 7 days along with i.p TAA injection 350 mg/kg from day 5 to 7. Liver function, oxidative stress biomarkers, behavioral tests in addition to histopathological examination were assessed. KEY FINDINGS: Hesperidin efficiently mitigated TAA-induced HE as evidenced by significant reduction in liver enzymes, bile and ammonia levels in serum. Moreover, hesperidin restored oxidant/antioxidant balance as manifested by reduction in MDA content in both cerebral and hepatic tissues. Additionally, hesperidin improved motor and cognitive abilities besides tissues' architecture as demonstrated by behavioral tests and histopathology results, respectively. Hesperidin also decreased levels of NLRP3 and increased levels of Sirt1 and FOXO in both cerebral and hepatic tissues. Finally, hesperidin markedly decreased the expression of IL-1ß and caspase-1 as shown by immunohistochemical results. SIGNIFICANCE: Taken together, the hepatoprotective impact of hesperidin and its ameliorative effect on the progression of HE appear to be mediated by its modulatory influence on NLRP3/Sirt1/FOXO signaling.


Hepatic Encephalopathy , Hesperidin , Rats , Animals , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/drug therapy , Hepatic Encephalopathy/prevention & control , Hesperidin/pharmacology , Hesperidin/therapeutic use , Thioacetamide/toxicity , Sirtuin 1/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Rats, Wistar , Liver/metabolism , Oxidative Stress , Cognition
13.
Drug Deliv Transl Res ; 13(1): 252-274, 2023 01.
Article En | MEDLINE | ID: mdl-35672652

Ashwagandha (ASH), a vital herb in Ayurvedic medicine, demonstrated potent preclinical hepato- and neuroprotective effects. However, its efficacy is limited due to low oral bioavailability. Accordingly, we encapsulated ASH extract in chitosan-alginate bipolymeric nanocapsules (ASH-BPNCs) to enhance its physical stability and therapeutic effectiveness in the gastrointestinal tract. ASH-BPNC was prepared by emulsification followed by sonication. The NCs showed small particle size (< 220 nm), zeta-potential of 25.2 mV, relatively high entrapment efficiency (79%), physical stability at acidic and neutral pH, and in vitro release profile that extended over 48 h. ASH-BPNC was then investigated in a thioacetamide-induced hepatic encephalopathy (HE) rat model. Compared with free ASH, ASH-BPNC improved survival, neurological score, general motor activity, and cognitive task-performance. ASH-BPNC restored ALT, AST and ammonia serum levels, and maintained hepatic and brain architecture. ASH-BPNC also restored GSH, MDA, and glutathione synthetase levels, and Nrf2 and MAPK signaling pathways in liver and brain tissues. Moreover, ASH-BPNC downregulated hepatic NF-κB immunohistochemical expression. Moreover, the in vivo biodistribution studies demonstrated that most of the administered ASH-BPNC is accumulated in the brain and hepatic tissues. In conclusion, chitosan-alginate BPNCs enhanced the hepatoprotective and neuroprotective effects of ASH, thus providing a promising therapeutic approach for HE.


Chitosan , Hepatic Encephalopathy , Nanocapsules , Neuroprotective Agents , Animals , Rats , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/drug therapy , Tissue Distribution , Signal Transduction , Plant Extracts/pharmacology
14.
ACS Chem Neurosci ; 13(19): 2829-2841, 2022 10 05.
Article En | MEDLINE | ID: mdl-36112416

Growing evidence suggests an essential role of neuroinflammation in behavioral abnormalities associated with hepatic encephalopathy (HE). Here, we report the involvement of autotaxin-lysophosphatidic acid (LPA) signaling in HE's pathogenesis. We demonstrate that the autotaxin (ATX) inhibitor PF-8380 attenuates neuroinflammation and improves neurological dysfunction in the mouse model of HE. In the thioacetamide (TAA)-induced model of HE, we found a twofold increase in the levels of ammonia in the brain and in plasma along with a significant change in HE-related behavioral parameters. Mice with HE show an increased brain weight, increased levels of tumor necrosis factor-α (TNF-α), IL-1ß (interleukin-1ß), interleukin-6 (IL-6), and LPA 18:0 in the cerebral cortex and hippocampus, and increased levels of LPA 18:0 in plasma. Treatment with the autotaxin inhibitor (ATXi) did not affect liver injury, as we observed no change in liver function markers including aspartate aminotransferase (AST), alanine aminotransferase (ALT), and total bilirubin (TBIL) and no change in ammonia levels in the brain and plasma. However, ATXi treatment significantly ameliorated the neuroinflammation, reduced the levels of LPA 18:0 in the cerebral cortex and hippocampus in the brain and plasma, and reduced brain edema and the levels of IL1ß, IL-6, and TNF-α. The neurobehavioral symptoms for HE such as the cognitive and motor function deficit and overall clinical grading score were significantly improved in ATXi-treated mice. Mouse astrocytes and microglia stimulated with NH4CL with or without ATXi showed significant attenuation of oxidative stress and the neuroinflammatory effect of NH4CL in ATXi-treated cells.


Brain Diseases , Hepatic Encephalopathy , Alanine Transaminase/therapeutic use , Ammonia/adverse effects , Animals , Aspartate Aminotransferases/therapeutic use , Bilirubin/adverse effects , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/drug therapy , Hepatic Encephalopathy/pathology , Interleukin-1beta , Interleukin-6 , Lysophospholipids , Mice , Neuroinflammatory Diseases , Thioacetamide/adverse effects , Tumor Necrosis Factor-alpha
15.
Neurotoxicology ; 92: 110-121, 2022 09.
Article En | MEDLINE | ID: mdl-35961375

RATIONALE: Hepatic encephalopathy (HE) is a neuropsychiatric disorder that results from either acute or chronic liver failure. CXCR2 plays an essential role in the pathophysiology of liver and brain diseases. In the present study, the potential beneficial effects of SB332235, a selective inhibitor of CXCR2, against HE were evaluated. METHODS: HE was induced in male rats by thioacetamide injection (200 mg/kg, i.p.) at three alternative days. SB332235 was injected in rats 1 h before TAA at a dose of 1 and 3 mg/kg i.p. RESULTS: SB332235 alleviated oxidative stress as shown by the decreased serum NO and reduced MDA, elevated GSH and SOD levels, and reduced TNF-α and NF-κB levels in both brain and liver tissues of rats. Additionally, SB332235 suppressed brain ASK-1, JNK, IL-8, and caspase-3 expression, and activated PI3K/AKT expression in brain tissues. Markers of brain dysfunction, such as ammonia, and markers of hepatic injury, such as LDH, albumin, bilirubin, γGT, AST, ALT, and ALP, were significantly ameliorated. Also, the protective effect of SB332235 was confirmed by histological examination of both brain and liver tissues. CONCLUSIONS: Both doses (1 and 3 mg/kg) of SB332235 revealed significant hepatic/neuroprotective effects due to their anti-inflammatory, antioxidant, and antiapoptotic activities via activation of the PI3K/AKT pathway. Between the two, the 1 mg/kg dose provided significantly improved outcomes.


Hepatic Encephalopathy , Neuroprotective Agents , Albumins/pharmacology , Ammonia , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Bilirubin , Caspase 3/metabolism , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/drug therapy , Hepatic Encephalopathy/metabolism , Interleukin-8/metabolism , Liver/metabolism , Male , NF-kappa B/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Superoxide Dismutase/metabolism , Thioacetamide/metabolism , Thioacetamide/toxicity , Tumor Necrosis Factor-alpha/metabolism
16.
Int Immunopharmacol ; 111: 109119, 2022 Oct.
Article En | MEDLINE | ID: mdl-35933745

Brain injury is the most common and serious consequence of hepatic encephalopathy (HE), and its pathophysiology is poorly understood. Excessive inflammatory, oxidative and apoptotic responses are the major mechanisms involved in the progression of brain injury induced by HE. Carvedilol is an adrenergic receptor antagonist with pronouncedantioxidant and anti-inflammatory activity. The present study aimed to investigatethe effects and underlying mechanisms of carvedilol on HE-induced brain damage in mice. Experimental model of HE was induced by the injection of thioacetamide (200 mg/kg) for two consecutive days and then mice were treated with carvedilol (10 or 20 mg/kg/day, orally) for 3 days in treatment groups. After the behavioral test, animals were sacrificed and the brain tissues were collected for biochemical, real time PCR and immunohistochemical analysis. The results showed that carvedilol improved locomotor impairment and reduced mortality rate in mice with HE. Carvedilol treatment decreased the brain levels of oxidative stress markers and induced Nrf2/HO-1 pathway. Carvedilol inhibited the activity of nuclear factor kappa B (NF-κB) and the expression of pro-inflammatory cytokines TNF-α, IL1ß and IL-6 in the brain tissues. Treatment of mice with carvedilol caused a significant reduction in the brain levels of iNOS/NO, myeloperoxidase (MPO), cyclooxygenase (COX)-2 and chemokine MCP-1 as proinflammatory mediators in HE. Moreover, the ratio of Bcl2/Bax was increased and apoptotic cell death was decreased in the brain of mice treated with carvedilol. In conclusion, carvedilol exerted protective effect against HE-induced brain injury through increasing antioxidant defense mechanisms and inhibitionof inflammatory and apoptotic pathways.


Brain Injuries , Hepatic Encephalopathy , Animals , Brain , Brain Injuries/metabolism , Carvedilol/therapeutic use , Hepatic Encephalopathy/chemically induced , Mice , NF-kappa B/metabolism , Oxidative Stress
17.
Acta Gastroenterol Belg ; 85(3): 433-437, 2022.
Article En | MEDLINE | ID: mdl-35833904

Background and study aims: Advanced liver disease frequently culminates in hepatic encephalopathy (HE), which can be classified as covert or overt HE, with subtle or clinically obvious changes respectively. 30-40% of patients with cirrhosis develop overt HE, which negatively affects the patients' quality of life. Next to lactulose, rifaximin-α has been prescribed as a second line therapy to treat and reduce the risk of recurrence of overt HE. In this study, we aimed to evaluate the effect of rifaximin-α therapy, both on the number of occurring infections and on the evolution in hospital admissions of patients with overt HE. Patients and methods: A total of 66 cirrhotic patients, treated for at least 6 months with rifaximin-α at AZ Maria Middelares, between October 1st 2014 and January 1st 2020, were included in the study analysis. Medical records of all patients were evaluated over a period of 6 months prior and after initiation of rifaximin-α therapy. Results: Data analysis revealed that the included cirrhotic patients were severely ill, with a mean model for end-stage liver disease (MELD) score of 21, and a median Child Pugh score of 11. Among these patients, rifaximin-α treatment significantly downgraded the total number of infections, with a main effect on respiratory infections. Furthermore, rifaximin-α therapy led to a significant decrease in HE-related, as well as in other liver-related hospital admissions. Conclusions: This study confirms the potential value of rifaximin-aα in reducing the number of developing infections and hospital admissions in a severely ill cirrhotic patient population.


End Stage Liver Disease , Hepatic Encephalopathy , Rifamycins , Drug Therapy, Combination , Gastrointestinal Agents/therapeutic use , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/etiology , Hospitals , Humans , Lactulose/therapeutic use , Liver Cirrhosis/complications , Liver Cirrhosis/drug therapy , Quality of Life , Rifamycins/therapeutic use , Rifaximin/therapeutic use , Severity of Illness Index
18.
Biometals ; 35(5): 935-953, 2022 10.
Article En | MEDLINE | ID: mdl-35857253

To investigate the metabolic changes in type C hepatic encephalopathy (CHE) rats after reducing manganese (Mn) intake. A total of 80 Sprague-Dawley rats were divided into control group and CHE groups (induced by intraperitoneal injection of thioacetamide at a dose of 250 mg/kg of body weight twice a week for 6 weeks). CHE rats were subdivided into 1Mn group (fed a standard diet, with 10 mg Mn/kg feed), 0.5Mn group (half-Mn diet), 0.25Mn group (quarter-Mn diet) and 0Mn group (no-Mn diet) for 4 to 8 weeks. Morris water maze (MWM), Y maze and narrow beam test (NBT) were used to evaluate cognitive and motor functions. Blood ammonia, brain Mn content, the number of GS-positive cells, and glutamine synthetase (GS) activity were measured. The metabolic changes of CHE rats were investigated using hydrogen-nuclear magnetic resonance and mass spectrometry. Multivariate statistical analysis was used to analyze the results. Significantly decreased numbers of entries in target area of MWM and Y maze, longer NBT latency and total time, higher blood ammonia, brain Mn content and GS activity were found in CHE rats. After reducing Mn intake, CHE rats had better behavioral performance, significantly lower blood ammonia, brain Mn content and GS activity. The main up-regulated metabolites were Ala, GABA, Glu, Gln, Lac, Tyr, Phe in 1Mn rats. After reducing Mn intake, metabolites returned to normal level at different degrees. Reducing Mn intake could reduce brain Mn content and blood ammonia, regulate GS activity and amino acid metabolism, ultimately improve behavioral performance in CHE rats.


Hepatic Encephalopathy , Amino Acids/metabolism , Ammonia/metabolism , Animals , Glutamate-Ammonia Ligase/metabolism , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/metabolism , Hepatic Encephalopathy/psychology , Hydrogen , Magnetic Resonance Spectroscopy , Manganese/metabolism , Rats , Rats, Sprague-Dawley , Thioacetamide , gamma-Aminobutyric Acid
19.
Food Funct ; 13(11): 6180-6194, 2022 Jun 06.
Article En | MEDLINE | ID: mdl-35583008

In the present study, we aimed to delineate the neuroprotective potential of thymol (THY) against neurotoxicity and cognitive deterioration induced by thioacetamide (TAA) in an experimental model of hepatic encephalopathy (HE). Rats received TAA (100 mg kg-1, intraperitoneally injected, three times per week) for two weeks. THY (30 and 60 mg kg-1), and Vit E (100 mg k-1) were administered daily by oral gavage for 30 days after HE induction. Supplementation with THY significantly improved liver function, reduced serum ammonia level, and ameliorated the locomotor and cognitive deficits. THY effectively modulated the alteration in oxidative stress markers, neurotransmitters, and brain ATP content. Histopathology of liver and brain tissues showed that THY had ameliorated TAA-induced damage, astrocyte swelling and brain edema. Furthermore, THY downregulated NF-kB and upregulated GFAP protein expression. In addition, THY significantly promoted CREB and BDNF expression at both mRNA and protein levels, together with enhancing brain cAMP level. In conclusion, THY exerted hepato- and neuroprotective effects against HE by mitigating hepatotoxicity, hyperammonemia and brain ATP depletion via its antioxidant, anti-inflammatory effects in addition to activation of the CREB/BDNF signaling pathway.


Hepatic Encephalopathy , Neurotoxicity Syndromes , Adenosine Triphosphate/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cognition , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/drug therapy , Hepatic Encephalopathy/metabolism , Liver/metabolism , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Oxidative Stress , Rats , Rats, Wistar , Signal Transduction , Thioacetamide/toxicity , Thymol/pharmacology
20.
Biofactors ; 48(5): 1166-1178, 2022 Sep.
Article En | MEDLINE | ID: mdl-35332953

This study aimed to investigate the possible usefulness of morin flavonoid in comparison to silymarin as a hepatic/neuronal-supportive agent with similar effects and higher bioavailability in a rat model of hepatic encephalopathy (HE). Morin effects on rat liver and brain were evaluated post-induction of HE by thioacetamide (TAA; 200 mg/kg/day for 3 successive days). Then, the serum activities of aspartate transaminase (AST) and alanine transaminase (ALT) together with ammonia concentration were estimated to assess the liver function. Also, the degree of brain effects was evaluated via the assessment of brain contents of reduced glutathione (GSH), malondialdehyde (MDA), tumor necrosis factor-alpha (TNF-α), and interleukin (IL-1ß) together with glutathione peroxidase (GPx) activity. In addition, the apoptotic and inflammatory changes in brain and liver tissues were also assessed via immunohistochemical examination. Our findings revealed a promising effect of morin against HE complications; as it corrected the liver functions, attenuated the brain/liver tissue injuries, and reduced the apoptotic and inflammatory insults of HE on both organs. These effects are comparable to those of silymarin. Morin could be introduced as a promising hepato- and neuro-therapeutic adjuvant in HE-associated neuronal complications especially in cases like silymarin intolerance.


Hepatic Encephalopathy , Silymarin , Alanine Transaminase , Ammonia/metabolism , Ammonia/pharmacology , Animals , Antioxidants/pharmacology , Aspartate Aminotransferases , Flavones , Flavonoids/metabolism , Flavonoids/pharmacology , Flavonoids/therapeutic use , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/drug therapy , Hepatic Encephalopathy/metabolism , Liver , Malondialdehyde/metabolism , Oxidative Stress , Rats , Rats, Wistar , Silymarin/metabolism , Silymarin/pharmacology , Thioacetamide/metabolism , Thioacetamide/toxicity , Tumor Necrosis Factor-alpha/metabolism
...