1
|
Singh VV, Prasad SK, Acharjee A, Srivastava S, Acharjee P. Investigating cognitive impairments and hippocampal proteome alterations in aged male rats with TAA-Induced minimal hepatic encephalopathy. Biogerontology 2024; 26:30. [PMID: 39704865 DOI: 10.1007/s10522-024-10158-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/13/2024] [Indexed: 12/21/2024]
Abstract
The aging population faces a gradual decline in physical and mental capacities, with an increased risk of liver cirrhosis and chronic liver diseases leading to hepatic encephalopathy (HE). The intertwining of physiological manifestations of aging with the pathophysiology of HE significantly impairs cognitive ability, reduces quality of life, and increases mortality. Hence, effective therapeutic intervention is imperative. The present study investigated the impact of minimal HE (MHE) on cognitive impairment in an aged rat population by analyzing hippocampal proteome dynamics. For this purpose, an old MHE rat model was induced via thioacetamide. The label-free LC‒MS/MS method was employed to explore hippocampal proteomic changes and associated dysregulated biological pathways. A total of 1533 proteins were identified, and among these, 30 proteins were significantly differentially expressed (18 upregulated, and 12 downregulated). Three upregulated proteins, namely, fetuin-A, p23, and intersectin-1 were selected and validated for their increased expression via western blotting and immunofluorescence analysis, which confirmed the mass spectrometry results. These proteins have not been reported previously in MHE cases. We also identified the possible dysregulated biological pathways associated with the differentially expressed proteins via Metascape, a network analysis tool. We found that the differentially expressed proteins may be involved in the generation of precursor metabolites and energy, the neurotransmitter release cycle, positive regulation of dendritic spine development, chaperone-mediated protein folding and protein stabilization. This study highlights the potential mechanisms underlying neurological dysfunction in the aged population with MHE and identifies novel therapeutic targets for improved disease management.
Collapse
Affiliation(s)
- Vishal Vikram Singh
- Biochemistry and Molecular Biology Unit, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Shambhu Kumar Prasad
- Biochemistry and Molecular Biology Unit, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Arup Acharjee
- Molecular Omics Laboratory, Department of Zoology, University of Allahabad, Prayagraj, 211002, India
| | - Sanjeeva Srivastava
- Department of Bioscience and Bioengineering, IIT Bombay, Powai, Mumbai, 400076, India
| | - Papia Acharjee
- Biochemistry and Molecular Biology Unit, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
2
|
Abd Elrazik NA, Abd El Salam ASG. Diacerein ameliorates thioacetamide-induced hepatic encephalopathy in rats via modulation of TLR4/AQP4/MMP-9 axis. Metab Brain Dis 2024; 40:10. [PMID: 39556255 PMCID: PMC11573817 DOI: 10.1007/s11011-024-01457-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 09/27/2024] [Indexed: 11/19/2024]
Abstract
Astrocyte swelling, blood brain barrier (BBB) dissipation and the subsequent brain edema are serious consequences of persistent hyperammonemia in hepatic encephalopathy (HE) in which if inadequately controlled it will lead to brain death. The current study highlights the potential neuroprotective effect of diacerein against thioacetamide (TAA)-induced HE in acute liver failure rat model. HE was induced in male Sprague-Dawley rats via I.P. injection of TAA (200 mg/kg) for three alternative times/week at 3rd week of the experiment. Diacerein (50 mg/kg) was gavaged for 14 days prior to induction of HE and for further 7 days together with TAA injection for an overall period of 21 days. Diacerein attenuated TAA-induced HE in acute liver failure rat model; as proofed by significant lowering of serum and brain ammonia concentrations, serum AST and ALT activities and significant attenuation of both brain and hepatic MDA contents and IL-1β with marked increases in GSH contents (P < 0.0001). The neuroprotective effect of diacerein was demonstrated by marked improvement of motor and cognitive deficits, brain histopathological changes; hallmarks of HE. As shown by immunohistochemical results, diacerein markedly downregulated brain TLR4 expression which in turn significantly increased the GFAP expression, and significantly decreased AQP4 expression; the astrocytes swelling biomarkers (P < 0.0001). Moreover, diacerein preserved BBB integrity via downregulation of MMP-9 mediated digestion of tight junction proteins such as occludin (P < 0.0001). Collectively, diacerein ameliorated cerebral edema and maintained BBB integrity via modulation of TLR4/AQP4/MMP-9 axis thus may decrease the progression of HE induced in acute liver failure.
Collapse
Affiliation(s)
- Nesma A Abd Elrazik
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | | |
Collapse
|
3
|
El-Kashef DH, Abdel-Rahman N, Sharawy MH. Apocynin alleviates thioacetamide-induced acute liver injury: Role of NOX1/NOX4/NF-κB/NLRP3 pathways. Cytokine 2024; 183:156747. [PMID: 39236429 DOI: 10.1016/j.cyto.2024.156747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/01/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024]
Abstract
The liver has a distinctive capacity to regenerate, yet severe acute injury can be life-threatening if not treated appropriately. Inflammation and oxidative stress are central processes implicated in the pathophysiology of acute livery injury. NOX isoforms are important enzymes for ROS generation, NF-κB and NLRP3 activation, its inhibition could be vital in alleviating acute liver injury (ALI). Here in our study, we used apocynin, a natural occurring potent NOX inhibitor, to exploreits potential protective effect against thioacetamide (TAA)-induced ALI through modulating crucial oxidative and inflammatory pathways. Rats were injected once with TAA (500 mg/kg/i.p) and treated with apocynin (10 mg/kg/i.p) twice before TAA challenge. Sera and hepatic tissues were collected for biochemical, mRNA expression, western blot analysis and histopathological assessments. Pretreatment with apocynin improved liver dysfunction evidenced by decreased levels of aminotransferases, ALP, GGT and bilirubin. Apocynin reduced mRNA expression of NOX1 and NOX4 which in turn alleviated oxidative stress, as shown by reduction in MDA and NOx levels, and elevation in GSH levels andcatalase and SOD activities. Moreover, apocynin significantly reduced MPO gene expression. We also demonstrate that apocynin ameliorated inflammation through activating IκBα and suppressing IKKα, IKKβ, NF-κBp65 and p-NF-κBp65, IL-6 andTNF-α. Additionally, apocynin potentiated the gene expression of anti-inflammatory IL-10 and reduced levels of hepatic NLRP3, Caspase-1 and IL-1β. These results suggest that apocynin protects against ALI in association with the inhibition of NOX1 and NOX4 and regulating oxidative and inflammatory pathways.
Collapse
Affiliation(s)
- Dalia H El-Kashef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Noha Abdel-Rahman
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| | - Maha H Sharawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
4
|
Hassan NF, El-Ansary MR, El-Ansary AR, El-Saied MA, Zaki OS. Unveiling the protective potential of mirabegron against thioacetamide-induced hepatic encephalopathy in rats: Insights into cAMP/PPAR-γ/p-ERK1/2/p S536 NF-κB p 65 and p-CREB/BDNF/TrkB in parallel with oxidative and apoptotic trajectories. Biochem Pharmacol 2024; 229:116504. [PMID: 39179118 DOI: 10.1016/j.bcp.2024.116504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/12/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
Hepatic encephalopathy (HE) is one of the most prevalent and severe hepatic and brain disorders in which escalation of the oxidative, inflammatory and apoptotic trajectories pathologically connects acute liver injury with neurological impairment. Mirabegron (Mira) is a beta3 adrenergic receptor agonist with proven antioxidant and anti-inflammatory activities. The current research pointed to exploring Mira's hepato-and neuroprotective impacts against thioacetamide (TAA)-induced HE in rats. Rats were distributed into three experimental groups: the normal control group, the TAA group, received TAA (200 mg/kg/day for three consecutive days) and the Mira-treated group received Mira (10 mg/kg/day; oral gavage) for 15 consecutive days and intoxicated with TAA from the 13th to the 15th day of the experimental period. Mira counteracted hyperammonemia, enhanced rats' locomotor capability and motor coordination. It attenuated hepatic/neurological injuries by its antioxidant, anti-apoptotic as well as anti-inflammatory potentials. Mira predominantly targeted cyclic adenosine monophosphate (cAMP)/phosphorylated extracellular signal-regulated kinase (p-Erk1/2)/peroxisome proliferator-activated receptor gamma (PPARγ) dependent pathways via downregulation of p S536-nuclear factor kappa B p65 (p S536 NF-κB p 65)/tumor necrosis alpha (TNF-α) axis. Meanwhile, it attenuated nuclear factor erythroid 2-related factor (Nrf2) depletion in parallel with restoring of the neuroprotective defensive pathway by upregulation of cerebral cAMP/PPAR-γ/p-ERK1/2 and p-CREB/BDNF/TrkB besides reduction of GFAP immunoreactivity. Mira showed anti-apoptotic activity through inhibition of Bax immunoreactivity and elevation of Bcl2. To summarize, Mira exhibited a hepato-and neuroprotective effect against TAA-induced HE in rats via shielding antioxidant defense and mitigation of the pathological inflammatory and apoptotic axis besides upregulation of neuroprotective signaling pathways.
Collapse
Affiliation(s)
- Noha F Hassan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Mona R El-Ansary
- Department of Biochemistry, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Amira R El-Ansary
- Department of Internal Medicine, Faculty of Medicine, Misr University for Science and Technology, Giza, Egypt
| | - Mohamed A El-Saied
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Omnia S Zaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt.
| |
Collapse
|
5
|
Lien TS, Sun DS, Chang HH. Targeted Delivery to Dying Cells Through P-Selectin-PSGL-1 Axis: A Promising Strategy for Enhanced Drug Efficacy in Liver Injury Models. Cells 2024; 13:1778. [PMID: 39513885 PMCID: PMC11545035 DOI: 10.3390/cells13211778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/23/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
To minimize off-target adverse effects and improve drug efficacy, various tissue-specific drug delivery systems have been developed. However, even in diseased organs, both normal and stressed, dying cells coexist, and a targeted delivery system specifically for dying cells has yet to be explored to mitigate off-target effects within the same organ. This study aimed to establish such a system. By examining the surfaces of dying cells in vitro, we identified P-selectin glycoprotein ligand-1 (PSGL-1) as a universal marker for dying cells, positioning it as a potential target for selective drug delivery. We demonstrated that liposomes conjugated with the PSGL-1 binding protein P-selectin had significantly greater binding efficiency to dying cells compared to control proteins such as E-selectin, L-selectin, galectin-1, and C-type lectin-like receptor 2. Using thioacetamide (TAA) to induce hepatitis and hepatocyte damage in mice, we assessed the effectiveness of our P-selectin-based delivery system. In vivo, P-selectin-conjugated liposomes effectively delivered fluorescent dye and the apoptosis inhibitor z-DEVD to TAA-damaged livers in wild-type mice, but not in PSGL-1 knockout mice. In TAA-treated wild-type mice, unconjugated liposomes required a 100-fold higher z-DEVD dose compared to P-selectin-conjugated liposomes to achieve a comparable, albeit less effective, therapeutic outcome in lowering plasma alanine transaminase levels and alleviating thrombocytopenia. This emphasizes that P-selectin conjugation enhances drug delivery efficiency by approximately 100-fold in mice. These results suggest that P-selectin-based liposomes could be a promising strategy for targeted drug delivery, enabling both diagnosis and treatment by specifically delivering cell-labeling agents and rescue agents to dying cells via the P-selectin-PSGL-1 axis at the individual cell level.
Collapse
Grants
- 104-2320-B-320 -009 -MY3, 107-2311-B-320-002-MY3, 111-2320-B320-006-MY3, 112-2320-B-320-007 National Science and Technology Council, Taiwan
- TCMMP104-06, TCMMP108-04, TCMMP 111-01, TCAS111-02, TCAS-112-02, TCAS113-04, TCRD112-033, TCRD113-041 Tzu-Chi Medical Foundation
Collapse
Affiliation(s)
| | | | - Hsin-Hou Chang
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (T.-S.L.); (D.-S.S.)
| |
Collapse
|
6
|
Tong XY, Hussain H, Shamaladevi N, Norenberg MD, Fadel A, El Hiba O, Abdeljalil EG, Bilal EM, Kempuraj D, Natarajan S, Schally AV, Jaszberenyi M, Salgueiro L, Paidas MJ, Jayakumar AR. Age and Sex in the Development of Hepatic Encephalopathy: Role of Alcohol. BIOLOGY 2024; 13:228. [PMID: 38666840 PMCID: PMC11048384 DOI: 10.3390/biology13040228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024]
Abstract
Hepatic encephalopathy (HE) is a neurological condition linked to liver failure. Acute HE (Type A) occurs with acute liver failure, while chronic HE (Type C) is tied to cirrhosis and portal hypertension. HE treatments lag due to gaps in understanding its development by gender and age. We studied how sex and age impact HE and its severity with combined liver toxins. Our findings indicate that drug-induced (thioacetamide, TAA) brain edema was more severe in aged males than in young males or young/aged female rats. However, adding alcohol (ethanol, EtOH) worsens TAA's brain edema in both young and aged females, with females experiencing a more severe effect than males. These patterns also apply to Type A HE induced by azoxymethane (AZO) in mice. Similarly, TAA-induced behavioral deficits in Type C HE were milder in young and aged females than in males. Conversely, EtOH and TAA in young/aged males led to severe brain edema and fatality without noticeable behavioral changes. TAA metabolism was slower in aged males than in young or middle-aged rats. When TAA-treated aged male rats received EtOH, there was a slow and sustained plasma level of thioacetamide sulfoxide (TASO). This suggests that with EtOH, TAA-induced HE is more severe in aged males. TAA metabolism was similar in young, middle-aged, and aged female rats. However, with EtOH, young and aged females experience more severe drug-induced HE as compared to middle-aged adult rats. These findings strongly suggest that gender and age play a role in the severity of HE development and that the presence of one or more liver toxins may aggravate the severity of the disease progression.
Collapse
Affiliation(s)
- Xiao Y. Tong
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (X.Y.T.); (M.D.N.)
| | - Hussain Hussain
- Department of Internal Medicine and Infectious Disease, Larkin Community Hospital, Miami, FL 33143, USA;
| | | | - Michael D. Norenberg
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (X.Y.T.); (M.D.N.)
- General Medical Research, Neuropathology Section, R&D Service, Veterans Affairs Medical Center, Miami, FL 33125, USA; (A.V.S.); (M.J.); (L.S.)
- South Florida VA Foundation for Research and Education Inc., Veterans Affairs Medical Center, Miami, FL 33125, USA
| | - Aya Fadel
- Department of Internal Medicine, Ocean Medical Center-Hackensack Meridian Health, Brick, NJ 08724, USA;
| | - Omar El Hiba
- Laboratory of Anthropogenic, Biotechnology, Health, and Nutritional Physiopathologies, Neuroscience and Toxicology Team, Faculty of Sciences, Chouaib Doukkali University, Av. Des Facultés, El Jadida 24000, Morocco; (O.E.H.); (E.-M.B.)
- Hassan First University of Settat, Higher Institute of Health Sciences, Laboratory of Sciences and Health Technologies, Epidemiology and Biomedical Unit, Settat 26000, Morocco;
| | - El got Abdeljalil
- Hassan First University of Settat, Higher Institute of Health Sciences, Laboratory of Sciences and Health Technologies, Epidemiology and Biomedical Unit, Settat 26000, Morocco;
| | - El-Mansoury Bilal
- Laboratory of Anthropogenic, Biotechnology, Health, and Nutritional Physiopathologies, Neuroscience and Toxicology Team, Faculty of Sciences, Chouaib Doukkali University, Av. Des Facultés, El Jadida 24000, Morocco; (O.E.H.); (E.-M.B.)
- Hassan First University of Settat, Higher Institute of Health Sciences, Laboratory of Sciences and Health Technologies, Epidemiology and Biomedical Unit, Settat 26000, Morocco;
| | - Deepak Kempuraj
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO 65211, USA;
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veterans Hospital, Columbia, MO 65201, USA
| | - Sampath Natarajan
- Department of Chemistry, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613401, India;
| | - Andrew V. Schally
- General Medical Research, Neuropathology Section, R&D Service, Veterans Affairs Medical Center, Miami, FL 33125, USA; (A.V.S.); (M.J.); (L.S.)
- South Florida VA Foundation for Research and Education Inc., Veterans Affairs Medical Center, Miami, FL 33125, USA
- Pathology, Laboratory Medicine, Endocrine, Polypeptide and Cancer Institute, Department of Veterans Affairs, Miami, FL 33125, USA
| | - Miklos Jaszberenyi
- General Medical Research, Neuropathology Section, R&D Service, Veterans Affairs Medical Center, Miami, FL 33125, USA; (A.V.S.); (M.J.); (L.S.)
- South Florida VA Foundation for Research and Education Inc., Veterans Affairs Medical Center, Miami, FL 33125, USA
- Department of Pathophysiology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Luis Salgueiro
- General Medical Research, Neuropathology Section, R&D Service, Veterans Affairs Medical Center, Miami, FL 33125, USA; (A.V.S.); (M.J.); (L.S.)
- South Florida VA Foundation for Research and Education Inc., Veterans Affairs Medical Center, Miami, FL 33125, USA
| | - Michael J. Paidas
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Arumugam R. Jayakumar
- General Medical Research, Neuropathology Section, R&D Service, Veterans Affairs Medical Center, Miami, FL 33125, USA; (A.V.S.); (M.J.); (L.S.)
- South Florida VA Foundation for Research and Education Inc., Veterans Affairs Medical Center, Miami, FL 33125, USA
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| |
Collapse
|
7
|
Perazza F, Leoni L, Colosimo S, Musio A, Bocedi G, D’Avino M, Agnelli G, Nicastri A, Rossetti C, Sacilotto F, Marchesini G, Petroni ML, Ravaioli F. Metformin and the Liver: Unlocking the Full Therapeutic Potential. Metabolites 2024; 14:186. [PMID: 38668314 PMCID: PMC11052067 DOI: 10.3390/metabo14040186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Metformin is a highly effective medication for managing type 2 diabetes mellitus. Recent studies have shown that it has significant therapeutic benefits in various organ systems, particularly the liver. Although the effects of metformin on metabolic dysfunction-associated steatotic liver disease and metabolic dysfunction-associated steatohepatitis are still being debated, it has positive effects on cirrhosis and anti-tumoral properties, which can help prevent the development of hepatocellular carcinoma. Furthermore, it has been proven to improve insulin resistance and dyslipidaemia, commonly associated with liver diseases. While more studies are needed to fully determine the safety and effectiveness of metformin use in liver diseases, the results are highly promising. Indeed, metformin has a terrific potential for extending its full therapeutic properties beyond its traditional use in managing diabetes.
Collapse
Affiliation(s)
- Federica Perazza
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Laura Leoni
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Santo Colosimo
- Doctorate School of Nutrition Science, University of Milan, 20122 Milan, Italy;
| | | | - Giulia Bocedi
- U.O. Diabetologia, Ospedale C. Magati, Scandiano, 42019 Reggio Emilia, Italy;
| | - Michela D’Avino
- S.C. Endocrinologia Arcispedale Santa Maria Nuova, 42123 Reggio Emilia, Italy;
| | - Giulio Agnelli
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Alba Nicastri
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Chiara Rossetti
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Federica Sacilotto
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Giulio Marchesini
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Maria Letizia Petroni
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Federico Ravaioli
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
- Division of Hepatobiliary and Immunoallergic Diseases, Department of Internal Medicine, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
8
|
Babu S, Ranajit SK, Pattnaik G, Ghosh G, Rath G, Kar B. An Insight into Different Experimental Models used for Hepatoprotective Studies: A Review. Curr Drug Discov Technol 2024; 21:e191223224660. [PMID: 39206705 DOI: 10.2174/0115701638278844231214115102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/13/2023] [Accepted: 11/22/2023] [Indexed: 09/04/2024]
Abstract
Numerous factors, including exposure to harmful substances, drinking too much alcohol, contracting certain hepatitis serotypes, and using specific medicines, contribute to the development of liver illnesses. Lipid peroxidation and other forms of oxidative stress are the main mechanisms by which hepatotoxic substances harm liver cells. Pathological changes in the liver include a rise in the levels of blood serum, a decrease in antioxidant enzymes, as well as the formation of free radical radicals. It is necessary to find pharmaceutical alternatives to treat liver diseases to increase their efficacy and decrease their toxicity. For the development of new therapeutic medications, a greater knowledge of primary mechanisms is required. In order to mimic human liver diseases, animal models are developed. Animal models have been used for several decades to study the pathogenesis of liver disorders and related toxicities. For many years, animal models have been utilized to investigate the pathophysiology of liver illness and associated toxicity. The animal models are created to imitate human hepatic disorders. This review enlisted numerous hepatic damage in vitro and in vivo models using various toxicants, their probable biochemical pathways and numerous metabolic pathways via oxidative stressors, different serum biomarkers enzymes are discussed, which will help to identify the most accurate and suitable model to test any plant preparations to check and evaluate their hepatoprotective properties.
Collapse
Affiliation(s)
- Sucharita Babu
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar, 751050, India
| | - Santosh K Ranajit
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar, 751050, India
| | - Gurudutta Pattnaik
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar, 751050, India
| | - Goutam Ghosh
- School of Pharmaceutical Sciences, Siksha O Anusandhan Deemed to be University, Bhubaneswar, 751030, India
| | - Goutam Rath
- School of Pharmaceutical Sciences, Siksha O Anusandhan Deemed to be University, Bhubaneswar, 751030, India
| | - Biswakanth Kar
- School of Pharmaceutical Sciences, Siksha O Anusandhan Deemed to be University, Bhubaneswar, 751030, India
| |
Collapse
|
9
|
Zhang H, Xu J. Unveiling thioacetamide-induced toxicity: Multi-organ damage and omitted bone toxicity. Hum Exp Toxicol 2024; 43:9603271241241807. [PMID: 38531387 DOI: 10.1177/09603271241241807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
Thioacetamide (TAA), a widely employed hepatotoxic substance, has gained significant traction in the induction of liver failure disease models. Upon administration of TAA to experimental animals, the production of potent oxidative derivatives ensues, culminating in the activation of oxidative stress and subsequent infliction of severe damage upon multiple organs via dissemination through the bloodstream. This review summarized the various organ damages and corresponding mechanistic explanations observed in previous studies using TAA in toxicological animal experiments. The principal pathological consequences arising from TAA exposure encompass oxidative stress, inflammation, lipid peroxidation, fibrosis, apoptosis induction, DNA damage, and osteoclast formation. Recent in vivo and in vitro studies on TAA bone toxicity have confirmed that long-term high-dose use of TAA not only induces liver damage in experimental animals but also accompanies bone damage, which was neglected for a long time. By using TAA to model diseases in experimental animals and controlling TAA dosage, duration of use, and animal exposure environment, we can induce various organ injury models. It should be noted that TAA-induced injuries have a time-dependent effect. Finally, in our daily lives, especially for researchers, we should take precautions to minimize TAA exposure and reduce the probability of related organ injuries.
Collapse
Affiliation(s)
- Haodong Zhang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, P.R. China
| | - Jian Xu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, P.R. China
| |
Collapse
|
10
|
Sepehrinezhad A, Shahbazi A, Joghataei MT, Larsen FS, Sahab Negah S. Inhibition of autotaxin alleviates pathological features of hepatic encephalopathy at the level of gut-liver-brain axis: an experimental and bioinformatic study. Cell Death Dis 2023; 14:490. [PMID: 37528089 PMCID: PMC10394058 DOI: 10.1038/s41419-023-06022-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 07/19/2023] [Accepted: 07/26/2023] [Indexed: 08/03/2023]
Abstract
There is accumulating evidence that the circulatory levels of autotaxin (ATX) and lysophosphatidic acid (LPA) are increased in patients with severe liver disease. However, the potential role of the ATX-LPA axis in hepatic encephalopathy (HE) remains unclear. Our study aimed to investigate the role of the ATX-LPA signaling pathway in mice with thioacetamide (TAA) induced acute HE. To show the role of the ATX-LPA axis in the context of HE, we first measured the involvement of ATX-LPA in the pathogenesis of TAA-induced acute HE. Then, we compared the potential effects of ATX inhibitor (HA130) on astrocyte responses at in vitro and gut-liver-brain axis at in vivo levels. The inflammatory chemokine (C-C motif) ligand 3 was significantly increased in the hyperammonemic condition and could be prevented by ATX inhibition in astrocytes at in vitro level. Further statistical tests revealed that plasma and tissue pro-inflammatory cytokines were inhibited by HA130 in mice. Furthermore, the stage of HE was significantly improved by HA130. The most surprising result was that HA130 alleviated immune infiltrating cells in the liver and intestine and decreased mucus-secreting cells in the intestine. Further analysis showed that the levels of liver enzymes in serum were significantly decreased in response to ATX inhibition. Surprisingly, our data indicated that HA130 could recover permeabilization of the blood-brain barrier, neuroinflammation, and recognition memory. Besides that, we found that the changes of Interleukin-1 (IL-1) and aquaporin-4 (AQP4) in HE might have a connection with the glymphatic system based on bioinformatics analyses. Taken together, our data showed that the ATX-LPA axis contributes to the pathogenesis of HE and that inhibition of ATX improves HE.
Collapse
Affiliation(s)
- Ali Sepehrinezhad
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Shahbazi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | | | - Fin Stolze Larsen
- Department of Gastroenterology and Hepatology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sajad Sahab Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
| |
Collapse
|
11
|
Mohamed EK, Hafez DM. Gallic acid and metformin co-administration reduce oxidative stress, apoptosis and inflammation via Fas/caspase-3 and NF-κB signaling pathways in thioacetamide-induced acute hepatic encephalopathy in rats. BMC Complement Med Ther 2023; 23:265. [PMID: 37491245 PMCID: PMC10367384 DOI: 10.1186/s12906-023-04067-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/04/2023] [Indexed: 07/27/2023] Open
Abstract
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 NFkβ 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.
Collapse
Affiliation(s)
- Ehsan Khedre Mohamed
- Biochemistry department, Egyptian DRUG AUTHORITY (EDA), formerly National Organization of Drug Control and Research (NODCAR), Giza, Egypt.
| | - Dawlat Mohamed Hafez
- Histology department, Egyptian DRUG AUTHORITY (EDA), formerly National Organization of Drug Control and Research (NODCAR), Giza, Egypt
| |
Collapse
|
12
|
Santos RPC, Toscano ECDB, Rachid MA. Anti-inflammatory strategies for hepatic encephalopathy: preclinical studies. ARQUIVOS DE NEURO-PSIQUIATRIA 2023. [PMID: 37487550 PMCID: PMC10371400 DOI: 10.1055/s-0043-1767819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Hepatic encephalopathy (HE) is a potentially reversible neuropsychiatric syndrome. Often, HE causes cognitive and motor dysfunctions due to an acute or chronic insufficiency of the liver or a shunting between the hepatic portal vein and systemic vasculature. Liver damage induces peripheral changes, such as in the metabolism and peripheral inflammatory responses that trigger exacerbated neuroinflammation. In experimental models, anti-inflammatory strategies have demonstrated neuroprotective effects, leading to a reduction in HE-related cognitive and motor impairments. In this scenario, a growing body of evidence has shown that peripheral and central nervous system inflammation are promising preclinical targets. In this review, we performed an overview of FDA-approved drugs and natural compounds which are used in the treatment of other neurological and nonneurological diseases that have played a neuroprotective role in experimental HE, at least in part, through anti-inflammatory mechanisms. Despite the exciting results from animal models, the available data should be critically interpreted, highlighting the importance of translating the findings for clinical essays.
Collapse
Affiliation(s)
- Rafaela Pinto Coelho Santos
- Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Departamento de Patologia Geral, Laboratório de Patologia Celular e Molecular, Belo Horizonte MG, Brazil
| | - Eliana Cristina de Brito Toscano
- Universidade Federal de Juiz de Fora, Faculdade de Medicina, Departamento de Patologia, Laboratório Integrado de Pesquisa em Patologia, Juiz de Fora MG, Brazil
- Universidade Federal de Juiz e Fora, Faculdade de Medicina, Programa de Pós-Graduação em Saúde, Juiz de Fora MG, Brazil
| | - Milene Alvarenga Rachid
- Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Departamento de Patologia Geral, Laboratório de Patologia Celular e Molecular, Belo Horizonte MG, Brazil
| |
Collapse
|
13
|
Capiglioni AM, Capitani MC, Marrone J, Marinelli RA. Adenoviral Transfer of Human Aquaporin-8 Gene to Mouse Liver Improves Ammonia-Derived Ureagenesis. Cells 2023; 12:1535. [PMID: 37296655 PMCID: PMC10253139 DOI: 10.3390/cells12111535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/12/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
We previously reported that, in cultured hepatocytes, mitochondrial aquaporin-8 (AQP8) channels facilitate the conversion of ammonia to urea and that the expression of human AQP8 (hAQP8) enhances ammonia-derived ureagenesis. In this study, we evaluated whether hepatic gene transfer of hAQP8 improves detoxification of ammonia to urea in normal mice as well as in mice with impaired hepatocyte ammonia metabolism. A recombinant adenoviral (Ad) vector encoding hAQP8, AdhAQP8, or a control Ad vector was administered via retrograde infusion into the bile duct of the mice. Hepatocyte mitochondrial expression of hAQP8 was confirmed using confocal immunofluorescence and immunoblotting. The normal hAQP8-transduced mice showed decreased plasma ammonia and increased liver urea. Enhanced ureagenesis was confirmed via the NMR studies assessing the synthesis of 15N-labeled urea from 15N-labeled ammonia. In separate experiments, we made use of the model hepatotoxic agent, thioacetamide, to induce defective hepatic metabolism of ammonia in mice. The adenovirus-mediated mitochondrial expression of hAQP8 was able to restore normal ammonemia and ureagenesis in the liver of the mice. Our data suggest that hAQP8 gene transfer to mouse liver improves detoxification of ammonia to urea. This finding could help better understand and treat disorders with defective hepatic ammonia metabolism.
Collapse
Affiliation(s)
| | | | | | - Raúl A. Marinelli
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario 2000, Argentina
| |
Collapse
|
14
|
Essam RM, Saadawy MA, Gamal M, Abdelsalam RM, El-Sahar AE. Lactoferrin averts neurological and behavioral impairments of thioacetamide-induced hepatic encephalopathy in rats via modulating HGMB1/TLR-4/MyD88/Nrf2 pathway. Neuropharmacology 2023; 236:109575. [PMID: 37201650 DOI: 10.1016/j.neuropharm.2023.109575] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/20/2023] [Accepted: 05/05/2023] [Indexed: 05/20/2023]
Abstract
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.
Collapse
Affiliation(s)
- Reham M Essam
- Biology Department, School of Pharmacy, Newgiza University, Giza, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Mariam A Saadawy
- Biology Department, School of Pharmacy, Newgiza University, Giza, Egypt
| | - Mahitab Gamal
- Clinical Pharmacy Department, School of Pharmacy, Newgiza University, Giza, Egypt
| | - Rania M Abdelsalam
- Biology Department, School of Pharmacy, Newgiza University, Giza, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ayman E El-Sahar
- Biology Department, School of Pharmacy, Newgiza University, Giza, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
15
|
Ramakrishna K, Sinku S, Majumdar S, Singh N, Gajendra TA, Rani A, Krishnamurthy S. Indole-3-carbinol ameliorated the thioacetamide-induced hepatic encephalopathy in rats. Toxicology 2023; 492:153542. [PMID: 37150287 DOI: 10.1016/j.tox.2023.153542] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/09/2023]
Abstract
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 (300mg/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 100mg/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.
Collapse
Affiliation(s)
- Kakarla Ramakrishna
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University (IIT BHU), Varanasi, Uttar Pradesh, India; Department of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, Andhra Pradesh, India
| | - Sangeetha Sinku
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University (IIT BHU), Varanasi, Uttar Pradesh, India
| | - Shreyasi Majumdar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University (IIT BHU), Varanasi, Uttar Pradesh, India
| | - Neha Singh
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University (IIT BHU), Varanasi, Uttar Pradesh, India
| | - T A Gajendra
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University (IIT BHU), Varanasi, Uttar Pradesh, India
| | - Asha Rani
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University (IIT BHU), Varanasi, Uttar Pradesh, India
| | - Sairam Krishnamurthy
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University (IIT BHU), Varanasi, Uttar Pradesh, India.
| |
Collapse
|
16
|
Sepehrinezhad A, Stolze Larsen F, Ashayeri Ahmadabad R, Shahbazi A, Sahab Negah S. The Glymphatic System May Play a Vital Role in the Pathogenesis of Hepatic Encephalopathy: A Narrative Review. Cells 2023; 12:cells12070979. [PMID: 37048052 PMCID: PMC10093707 DOI: 10.3390/cells12070979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/20/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Hepatic encephalopathy (HE) is a neurological complication of liver disease resulting in cognitive, psychiatric, and motor symptoms. Although hyperammonemia is a key factor in the pathogenesis of HE, several other factors have recently been discovered. Among these, the impairment of a highly organized perivascular network known as the glymphatic pathway seems to be involved in the progression of some neurological complications due to the accumulation of misfolded proteins and waste substances in the brain interstitial fluids (ISF). The glymphatic system plays an important role in the clearance of brain metabolic derivatives and prevents aggregation of neurotoxic agents in the brain ISF. Impairment of it will result in aggravated accumulation of neurotoxic agents in the brain ISF. This could also be the case in patients with liver failure complicated by HE. Indeed, accumulation of some metabolic by-products and agents such as ammonia, glutamine, glutamate, and aromatic amino acids has been reported in the human brain ISF using microdialysis technique is attributed to worsening of HE and correlates with brain edema. Furthermore, it has been reported that the glymphatic system is impaired in the olfactory bulb, prefrontal cortex, and hippocampus in an experimental model of HE. In this review, we discuss different factors that may affect the function of the glymphatic pathways and how these changes may be involved in HE.
Collapse
Affiliation(s)
- Ali Sepehrinezhad
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad 9919191778, Iran
| | - Fin Stolze Larsen
- Department of Gastroenterology and Hepatology, Rigshospitalet, Copenhagen University Hospital, 999017 Copenhagen, Denmark
| | | | - Ali Shahbazi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Sajad Sahab Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad 9919191778, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran 1449614535, Iran
| |
Collapse
|
17
|
Hepatoprotective Effect of Kaempferol: A Review of the Dietary Sources, Bioavailability, Mechanisms of Action, and Safety. Adv Pharmacol Pharm Sci 2023; 2023:1387665. [PMID: 36891541 PMCID: PMC9988374 DOI: 10.1155/2023/1387665] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/27/2022] [Accepted: 02/03/2023] [Indexed: 03/02/2023] Open
Abstract
The liver is the body's most critical organ that performs vital functions. Hepatic disorders can affect the physiological and biochemical functions of the body. Hepatic disorder is a condition that describes the damage to cells, tissues, structures, and functions of the liver, which can cause fibrosis and ultimately result in cirrhosis. These diseases include hepatitis, ALD, NAFLD, liver fibrosis, liver cirrhosis, hepatic failure, and HCC. Hepatic diseases are caused by cell membrane rupture, immune response, altered drug metabolism, accumulation of reactive oxygen species, lipid peroxidation, and cell death. Despite the breakthrough in modern medicine, there is no drug that is effective in stimulating the liver function, offering complete protection, and aiding liver cell regeneration. Furthermore, some drugs can create adverse side effects, and natural medicines are carefully selected as new therapeutic strategies for managing liver disease. Kaempferol is a polyphenol contained in many vegetables, fruits, and herbal remedies. We use it to manage various diseases such as diabetes, cardiovascular disorders, and cancers. Kaempferol is a potent antioxidant and has anti-inflammatory effects, which therefore possesses hepatoprotective properties. The previous research has studied the hepatoprotective effect of kaempferol in various hepatotoxicity protocols, including acetaminophen (APAP)-induced hepatotoxicity, ALD, NAFLD, CCl4, HCC, and lipopolysaccharide (LPS)-induced acute liver injury. Therefore, this report aims to provide a recent brief overview of the literature concerning the hepatoprotective effect of kaempferol and its possible molecular mechanism of action. It also provides the most recent literature on kaempferol's chemical structure, natural source, bioavailability, and safety.
Collapse
|
18
|
Liu P, Li H, Xu H, Gong J, Jiang M, Qian J, Xu Z, Shi J. Chitooligosaccharides Attenuated Hepatic Encephalopathy in Mice through Stabilizing Gut-Liver-Brain Disturbance. Mol Nutr Food Res 2023; 67:e2200158. [PMID: 36281912 DOI: 10.1002/mnfr.202200158] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/10/2022] [Indexed: 01/19/2023]
Abstract
SCOPE Hepatic encephalopathy (HE) refers to neurological dysfunction associated with hepatic inadequacy and gut dysbiosis. Chitooligosaccharides (COS) possesses prominent biological activities including incalculable hepatoprotective, neuroprotective and prebiotic effects. This study evaluates the protective effects of COS on HE from the influence of gut-liver-brain axis in mice. METHODS AND RESULTS Hepatic injured mice show minimal symptoms of HE, reflecting in cognitive impairment, and learning and memory retardation, while they are reversed by COS following orally administrated. Furthermore, COS ameliorates brain function through inhibiting microglial and astrocyte activation in cerebral cortex and hippocampus, promoting neuronal regeneration characterized by the increase of neuron-specific marker (neuronal nuclear antigen, NeuN). Concurrently, neuroinflammation and hepatitis are restrained by COS through descending toll-like receptors 4/Nuclear factor kappa B (TLR4/NF-κB) pathway. Additionally, the dysbiosis of the composition and structure of gut microbiota is displayed in mice with HE, while it is modified by COS through decreasing the relative abundances of Muribaculaceae, Lactobacillus, and Enterorhabdus. The enhancement of blood ammonia is crucially slipped to basal levels by COS. CONCLUSION The present study shows that COS could prevent the pathological process of HE through regulating the gut-liver-brain cross-talk, which provids new insight into fundamental roles of COS.
Collapse
Affiliation(s)
- Peng Liu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Heng Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Hongyu Xu
- School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Jinsong Gong
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Min Jiang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Jianying Qian
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Zhenghong Xu
- School of Biotechnology, Jiangnan University, Wuxi, 214122, China.,National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Wuxi, 214122, China
| | - Jinsong Shi
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| |
Collapse
|
19
|
Sun T, Du H, Li Z, Xiong J, Liu Y, Li Y, Zhang W, Liang F, He J, Liu X, Xiang H. Decoding the contributions of gut microbiota and cerebral metabolism in acute liver injury mice with and without cognitive dysfunction. CNS Neurosci Ther 2022. [PMID: 36585803 DOI: 10.1111/cns.14069] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/23/2022] [Accepted: 12/02/2022] [Indexed: 01/01/2023] Open
Abstract
AIMS Patients with acute liver injury (ALI) can develop cognitive dysfunction (CD). The study investigated the role of gut microbiota and cerebral metabolism in ALI mice with and without CD. METHODS Male C57BL/6 mice that received thioacetamide were classified into ALI mice with (susceptible) or without (unsusceptible) CD-like phenotypes by hierarchical cluster analysis of behavior. The role of gut microbiota was investigated by 16S ribosomal RNA gene sequencing and feces microbiota transplantation (FMT). 1 H-[13 C] NMR and electrophysiology were used to detect the changes in cerebral neurotransmitter metabolic and synaptic transition in neurons or astrocytes. RESULTS Apromixlay 55% (11/20) of mice developed CD and FMT from the susceptible group transmitted CD to gut microbiota-depleted mice. Alloprevotella was enriched in the susceptible group. GABA production was decreased in the frontal cortex, while hippocampal glutamine was increased in the susceptible group. Altered Escherichia. Shigella and Alloprevotella were correlated with behaviors and cerebral metabolic kinetics and identified as good predictors of ALI-induced CD. The frequencies of both miniature inhibitory and excitatory postsynaptic currents in hippocampal CA1 and prefrontal cortex were decreased in the susceptible group. CONCLUSION Altered transmitter metabolism and synaptic transmission in the hippocampus and prefrontal cortex and gut microbiota disturbance may lead to ALI-induced CD.
Collapse
Affiliation(s)
- Tianning Sun
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongying Du
- Department of Food Science and Engineering, College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, China
| | - Zhen Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Xiong
- Hepatobiliary Surgery Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanbo Liu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yujuan Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wencui Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fangyuan Liang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Jingang He
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, China
| | - Xiaodong Liu
- Department of Anaesthesia and Intensive Care, Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Hongbing Xiang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
20
|
Huang L, Li X, Han R, Li Y, Xu L, Zeng Z, Wu K. Solubility measurement, correlation and mixing properties of thioacetamide in fifteen pure solvents. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.119847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
21
|
Adrenal histological and functional changes after hepatic encephalopathy: From mice model to an integrative bioinformatics analysis. Acta Histochem 2022; 124:151960. [DOI: 10.1016/j.acthis.2022.151960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/28/2022] [Indexed: 11/15/2022]
|
22
|
Elsherbini DMA, Ghoneim FM, El-Mancy EM, Ebrahim HA, El-Sherbiny M, El-Shafey M, Al-Serwi RH, Elsherbiny NM. Astrocytes profiling in acute hepatic encephalopathy: Possible enrolling of glial fibrillary acidic protein, tumor necrosis factor-alpha, inwardly rectifying potassium channel (Kir 4.1) and aquaporin-4 in rat cerebral cortex. Front Cell Neurosci 2022; 16:896172. [PMID: 36060277 PMCID: PMC9428715 DOI: 10.3389/fncel.2022.896172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Hepatic encephalopathy (HE) is a neurological disarray manifested as a sequel to chronic and acute liver failure (ALF). A potentially fatal consequence of ALF is brain edema with concomitant astrocyte enlargement. This study aims to outline the role of astrocytes in acute HE and shed light on the most critical mechanisms driving this role. Rats were allocated into two groups. Group 1, the control group, received the vehicle. Group 2, the TAA group, received TAA (300 mg/kg) for 3 days. Serum AST, ALT, and ammonia were determined. Liver and cerebral cortical sections were processed for hematoxylin and eosin staining. Additionally, mRNA expression and immunohistochemical staining of cortical GFAP, TNFα, Kir4.1, and AQP4 were performed. Cortical sections from the TAA group demonstrated neuropil vacuolation and astrocytes enlargement with focal gliosis. GFAP, TNFα, and AQP4 revealed increased mRNA expression, positive immunoreactivity, and a positive correlation to brain water content. In contrast, Kir 4.1 showed decreased mRNA expression and immunoreactivity and a negative correlation to brain water content. In conclusion, our findings revealed altered levels of TNFα, Kir 4.1, GFAP, and AQP4 in HE-associated brain edema. A more significant dysregulation of Kir 4.1 and TNFα was observed compared to AQP4 and GFAP.
Collapse
Affiliation(s)
- Dalia Mahmoud Abdelmonem Elsherbini
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
- Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- *Correspondence: Dalia Mahmoud Abdelmonem Elsherbini,
| | - Fatma M. Ghoneim
- Department of Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Eman Mohammed El-Mancy
- Deanship of Common First Year, Jouf University, Sakaka, Saudi Arabia
- Department of Zoology, Faculty of Women for Arts, Science and Education, Ain Shams University, Cairo, Egypt
| | - Hasnaa Ali Ebrahim
- Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
- Mohamed El-Sherbiny,
| | - Mohamed El-Shafey
- Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Department of Physiological Sciences, Fakeeh College for Medical Sciences, Jeddah, Saudi Arabia
| | - Rasha Hamed Al-Serwi
- Department of Basic Dental Sciences, College of Dentistry, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Nehal M. Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
23
|
Evaluation of the Effect of Wheat Germ Oil and Olmutinib on the Thioacetamide-Induced Liver and Kidney Toxicity in Mice. LIFE (BASEL, SWITZERLAND) 2022; 12:life12060900. [PMID: 35743930 PMCID: PMC9228264 DOI: 10.3390/life12060900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022]
Abstract
Thioacetamide (TAA) intoxication produces a reproducible standard animal model of induced liver and kidney injuries where free radicals are produced by phase I oxidation reactions, which eventually leads to liver and kidney failure. Wheat germ oil (WGO) is a unique food supplement with concentrated nutrient efficiency and has remarkable antioxidant functions. Olmutinib, on the other hand, is a chemotherapy drug considered safe for kidneys and the liver. Therefore, in this study, WGO and olmutinib were investigated for their effect on TAA-induced liver and kidney damage. Inflammatory markers; interleukin-1 beta (IL-1β); IL-6; and the levels of enzymatic markers ALT (Alanine aminotransferase), AST (Aspartate aminotransferase), LDH (Lactate dehydrogenase), and CK (creatinine kinase) in serum for liver and kidney were analyzed and evaluated along with histopathological changes in the tissue. Thirty male mice 4–6 weeks of age were grouped into five groups of six animals: the control group (saline) and the other groups (Groups II to V), which were given thioacetamide for two weeks. In addition, Group II continued with TAA; Group III was given olmutinib (30 mg/kg), Group IV was given the wheat germ oil (WGO) (1400 mg/kg), and Group V was given (olmutinib (30 mg/kg) + WGO (1400 mg/kg)) for five days. The results suggested that olmutinib treatment potentiated TAA-induced liver and kidney injury. At the same time, WGO efficiently alleviated TAA and TAA–olmutinib toxicity in Groups IV and V. The histological studies also showed reduced damage with WGO in the animal model. Hence, it was concluded that WGO could significantly reduce liver and kidney damage caused by TAA and olmutinib in mice.
Collapse
|
24
|
Hong W, Peng X, Zhou X, Li P, Ye Z, Liang W. FXR/ASS1 axis attenuates the TAA-induced liver injury through arginine metabolism. Biochem Biophys Res Commun 2022; 611:31-37. [PMID: 35477090 DOI: 10.1016/j.bbrc.2022.04.073] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/15/2022] [Indexed: 11/19/2022]
Abstract
Previous studies demonstrated that arginine biosynthesis was frequently impaired in acute liver injury. However, the underlying mechanisms remain elusive. In this study, we found that Argininosuccinate synthetase 1 (ASS1), a rate-limiting enzyme in arginine metabolism, was downregulated in the TAA-induced liver injury model. Single-cell RNA-seq data found that ASS1 was highly enriched in the hepatocytes. The reduction of ASS1 was attributed to the decreased expression of Farnesoid X receptor (FXR), which is a bile acid-activated nuclear hormone receptor with high expression in the liver. Subsequent studies demonstrated that activation of FXR by its agonist obeticholic acid (OCA) directly promoted ASS1 transcription and enhanced arginine synthesis, leading to the alleviation of TAA-mediated liver injury. Further experiments found that OCA, ASS1, and arginine supplement can rescue TAA-mediated hepatocytes apoptosis by decreasing the protein levels of Cyto C, PARP, and Caspase 3. Taken together, our study illustrated a protective role of the FXR/ASS1 axis in TAA-induced liver injury by targeting arginine metabolism, which might shed light on the development of novel therapeutic approaches for acute liver injury.
Collapse
Affiliation(s)
- Weilong Hong
- Department of Emergency, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Xuyun Peng
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China; Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Xue Zhou
- Department of Ultrasonic Medicine, Guangzhou Women and Children's Medical Center, Guangzhou, PR China
| | - Panlong Li
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China; Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Zhiqiang Ye
- Department of Emergency, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China.
| | - Weicheng Liang
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China; Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China; Vaccine Research Institute, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China.
| |
Collapse
|
25
|
Gao S, Yang Q, Wang X, Hu W, Lu Y, Yang K, Jiang Q, Li W, Song H, Sun F, Cheng H. Association Between Drug Treatments and the Incidence of Liver Injury in Hospitalized Patients With COVID-19. Front Pharmacol 2022; 13:799338. [PMID: 35387350 PMCID: PMC8978013 DOI: 10.3389/fphar.2022.799338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/28/2022] [Indexed: 01/08/2023] Open
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) has led to the emergence of global health care. In this study, we aimed to explore the association between drug treatments and the incidence of drug-induced liver injury (DILI) in hospitalized patients with COVID-19. A retrospective study was conducted on 5113 COVID-19 patients in Hubei province, among which 395 incurred liver injury. Hazard ratios (HRs) and 95% confidence intervals (CIs) were estimated by Cox proportional hazards models. The results showed that COVID-19 patients who received antibiotics (HR 1.97, 95% CI: 1.55–2.51, p < 0.001), antifungal agents (HR 3.10, 95% CI: 1.93–4.99, p < 0.001) and corticosteroids (HR 2.31, 95% CI: 1.80–2.96, p < 0.001) had a higher risk of DILI compared to non-users. Special attention was given to the use of parenteral nutrition (HR 1.82, 95% CI: 1.31–2.52, p < 0.001) and enteral nutrition (HR 2.71, 95% CI: 1.98–3.71, p < 0.001), which were the risk factors for liver injury. In conclusion, this study suggests that the development of DILI in hospitalized patients with COVID-19 needs to be closely monitored, and the above-mentioned drug treatments may contribute to the risk of DILI.
Collapse
Affiliation(s)
- Suyu Gao
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qingqing Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Xuanxuan Wang
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wen Hu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yun Lu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kun Yang
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qiaoli Jiang
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wenjing Li
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Haibo Song
- Key Laboratory for Research and Evaluation of Pharmacovigilance, National Medical Products Administration, Beijing, China.,Chinese Society of Toxicology, Beijing, China
| | - Feng Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Hong Cheng
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
26
|
Zhou C, Chen Y, Liu J, Shi Q, Xiong B. Molecular targeted therapy causes hepatic encephalopathy in patients after Transjugular intrahepatic portosystemic shunt (TIPS): A case report and literature review. J Interv Med 2022; 5:37-39. [PMID: 35586281 PMCID: PMC8947988 DOI: 10.1016/j.jimed.2021.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/25/2021] [Accepted: 12/29/2021] [Indexed: 11/26/2022] Open
Abstract
We report two cases of hepatic encephalopathy caused by molecular targeted drugs after the Transjugular intrahepatic portosystemic shunt (TIPS) procedure in our center. The liver toxicities and anti-angiogenic effects induced by targeted drugs may generate an imbalance in ammonia metabolism, elevating blood ammonia levels. TIPS diverts partial blood supply from the liver, aggravates liver impairment, and shunts ammonia-rich blood from the intestine into the systemic circulation. These may be the mechanisms leading to hepatic encephalopathy caused by molecular targeted drugs following TIPS. When clinicians choose molecular targeted therapy as the second or third targeted therapy for patients who have undergone TIPS, the consequence of drug-induced hepatic encephalopathy should also be considered.
Collapse
Affiliation(s)
- Chen Zhou
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, Hubei, China
| | - Yang Chen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, Hubei, China
| | - Jiacheng Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, Hubei, China
| | - Qin Shi
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, Hubei, China
| | - Bin Xiong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, Hubei, China
| |
Collapse
|
27
|
Shaker ME, Eisa NH, Elgaml A, El-Mesery A, El-Shafey M, El-Dosoky M, El-Mowafy M, El-Mesery M. Ingestion of mannose ameliorates thioacetamide-induced intrahepatic oxidative stress, inflammation and fibrosis in rats. Life Sci 2021; 286:120040. [PMID: 34637794 DOI: 10.1016/j.lfs.2021.120040] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS The monosaccharide mannose has gained recent interest for its beneficial effect against certain inflammatory disorders. Nevertheless, the influence of mannose on experimentally-induced liver fibrosis and the ensued inflammation is still not fully clear to date. MAIN METHODS The current study investigated the outcomes of treating rats with mannose (0.2 ml of 20% w/v, oral gavage) 30 min before the twice weekly intoxication with thioacetamide (TAA) (200 mg/kg, intraperitoneal) for a total period of 8 weeks. KEY FINDINGS The data indicated that mannose markedly dampened TAA-induced liver fibrosis, as indicated by lowering the fibrotic bridges shown by Masson's trichrome staining. This effect was consistent with reducing TAA-induced hepatocellular injury, as evidenced biochemically (serum ALT and AST activities) and pathologically (necroinflammation score). These hepatoprotective effects mediated by mannose were attributed to i) reversing TAA-induced rise in malondialdehyde (MDA) and decrease in reduced glutathione (GSH) expressions in the liver, ii) limiting TAA-induced release of the proinflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), iii) impairing TAA-induced activation of hepatic stellate cells by downregulating α-smooth muscle actin expression (α-SMA), and more importantly, iv) dampening TAA-induced fibrogenesis driven by transforming growth factor-β1 (TGF-β1) and connective tissue growth factor (CTGF). SIGNIFICANCE Mannose may be a valuable candidate for preventing oxidative stress, inflammation and fibrogenesis in the liver.
Collapse
Affiliation(s)
- Mohamed E Shaker
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Aljouf, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Nada H Eisa
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Abdelaziz Elgaml
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Department of Microbiology and Immunology, Faculty of Pharmacy, Horus University, New Damietta 34518, Egypt
| | - Ahmed El-Mesery
- Department of Tropical Medicine, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Mohamed El-Shafey
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Egypt; Physiological Sciences Department, Fakeeh College for Medical Sciences, Jeddah, Saudi Arabia
| | - Mohamed El-Dosoky
- Department of Neuroscience Technology, College of Applied Medical Science in Jubail, Imam Abdulalrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Mohammed El-Mowafy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Mohamed El-Mesery
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
28
|
Capiglioni AM, Müller GL, Marrone J, Alvarez ML, Marinelli RA. Enhanced ammonia detoxification to urea in hepatocytes transduced with human aquaporin-8 gene. Biotechnol Bioeng 2021; 118:4331-4337. [PMID: 34292591 DOI: 10.1002/bit.27901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 11/11/2022]
Abstract
Hepatic ammonia detoxification to urea is critical for the prevention of hyperammonemia and neurological damage. Hepatocyte mitochondrial aquaporin-8 (AQP8) channels have been involved in ammonia-derived ureagenesis. Herein, we studied whether the adenoviral gene transfer of human AQP8 (hAQP8) to hepatocyte mitochondria enhances ammonia conversion to urea. Using primary cultured rat hepatocytes, we first confirmed the mitochondrial expression of hAQP8 and then, using unlabeled or 15 N-labeled ammonia, we demonstrated that the urea synthesis was significantly enhanced in hAQP8-transduced hepatocytes. Studies using isolated hAQP8-expressing mitochondria also showed an increased ammonia metabolism. hAQP8 transduction was able to recover the impaired ammonia-derived ureagenesis in hepatotoxin-treated hepatocytes. Our data suggest that mitochondrially-expressed hAQP8 enhances and improves hepatocyte ammonia conversion to urea, a finding with potential therapeutic implications for liver disease with impaired ammonia detoxification.
Collapse
Affiliation(s)
- Alejo M Capiglioni
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Gabriela L Müller
- Centro de Estudios Fotosintéticos y Bioquímicos (CEFOBI-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Julieta Marrone
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Maria L Alvarez
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Raúl A Marinelli
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| |
Collapse
|