1
|
Donaldson J, Jacek T, Wychowański P, Zaworski K, Szkopek D, Woliński J, Grujic D, Pierzynowski S, Pierzynowska K. Rat Model of Endogenous and Exogenous Hyperammonaemia Induced by Different Diets. Int J Mol Sci 2025; 26:1818. [PMID: 40076444 PMCID: PMC11899528 DOI: 10.3390/ijms26051818] [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: 01/08/2025] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Two different diets able to induce dietary hyperammonaemia (a methionine-choline-deficient (MCD) diet and a methionine-deficient diet enriched with ammonium acetate (MAD + 20% ammonium acetate)) were tested in a rat model. The diets were shown to have different modes of action, inducing significant hyperammonaemia (HA) and growth retardation in the rats, with different metabolic consequences. The MCD diet resulted in the development of endogenous HA, with a decrease in bilirubin levels and an increase in hepatic fat content. In contrast, the MAD + 20% ammonium acetate diet increased circulating ALP and haptoglobin levels and decreased liver mass. The above results suggest that the MCD diet deteriorated the liver function of the rats, resulting in the development of endogenous HA, while the MAD diet caused moderate changes in liver metabolism, resulting in the development of exogenous HA. Interestingly, the commonly used oral treatments Lactulose and Rifaximin did not ameliorate hyperammonaemia during or after the treatment period. In conclusion, even though the diets used in the current study caused somewhat similar hyperammonaemia, they seemed to provoke different metabolic consequences. The latter can have an impact on the severity of the resulting hyperammonaemia and thus on the hyperammonaemia-induced encephalopathy, resulting in the development of distinguishing cognitive and metabolic (liver) effects compared to other forms of encephalopathy. We hypothesized that these rat models, with significantly increased serum ammonia levels, along with different liver injuries, could serve as a suitable double animal model for the testing of new, oral enzyme therapies for hepatic encephalopathy in future studies.
Collapse
Affiliation(s)
- Janine Donaldson
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Parktown, Johannesburg 2193, South Africa
- Anara AB, 23132 Trelleborg, Sweden; (D.G.); (S.P.)
| | - Tomasz Jacek
- National Research Institute of Animal Production, 32-083 Balice, Poland;
| | - Piotr Wychowański
- Department of Head and Neck and Sensory Organs, Division of Oral Surgery and Implantology, Institute of Clinical Dentistry, Gemelli Foundation for the University Policlinic, Catholic University of the “Sacred Heart”, 00168 Rome, Italy;
- Department of Interventional Dentistry, Collegium Medicum, Nicolaus Copernicus University, 85-067 Bydgoszcz, Poland
- Specialized Private Implantology Clinic Wychowański Stomatologia, 02-517 Warsaw, Poland
| | - Kamil Zaworski
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland;
| | - Dominika Szkopek
- Large Animal Models Laboratory, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland; (D.S.); (J.W.)
| | - Jarosław Woliński
- Large Animal Models Laboratory, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland; (D.S.); (J.W.)
| | | | - Stefan Pierzynowski
- Anara AB, 23132 Trelleborg, Sweden; (D.G.); (S.P.)
- Department of Biology, Lund University, 223 62 Lund, Sweden
- Department of Medical Biology, Institute of Rural Health, 20-090 Lublin, Poland
| | - Kateryna Pierzynowska
- Anara AB, 23132 Trelleborg, Sweden; (D.G.); (S.P.)
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland;
- Department of Biology, Lund University, 223 62 Lund, Sweden
| |
Collapse
|
2
|
Gad ES, Aldossary SA, El-Ansary MR, Abd El-Galil MM, Abd-El-Hamid AH, El-Ansary AR, Hassan NF. Cilostazol counteracts mitochondrial dysfunction in hepatic encephalopathy rat model: Insights into the role of cAMP/AMPK/SIRT1/ PINK-1/parkin hub and p-CREB /BDNF/ TrkB neuroprotective trajectory. Eur J Pharmacol 2025; 987:177194. [PMID: 39667427 DOI: 10.1016/j.ejphar.2024.177194] [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: 06/26/2024] [Revised: 11/17/2024] [Accepted: 12/09/2024] [Indexed: 12/14/2024]
Abstract
A devasting stage of chronic hepatic dysfunction is strictly correlated with neurological impairment, signifying hepatic encephalopathy (HE). HE is a multifactorial condition; therefore, hyperammonemia, oxidative stress, neuroinflammation, and mitochondrial dysfunction interplay in HE's progressive development. Cilostazol (Cilo) has shown promising neuroprotective and hepatoprotective effectiveness in different neuronal and hepatic disorders; however, its efficiency against HE hasn't yet been explored. This study aimed to investigate the protective role of Cilo against thioacetamide (TAA)-induced HE in rats targeting mitochondrial dysfunction via modulation of Adenosine monophosphate-activated protein kinase (AMPK)/Silent information regulator 1 (SIRT1) dependent pathways. Rats were allocated into three groups: the normal control group, the TAA group received (100 mg/kg, three times per week, for six weeks) to induce HE, and the Cilo group received (Cilo 100 mg/kg/day for six weeks, oral gavage) concurrently with TAA. Cilo counteracted HE indicated in the enhancement of cognitive impairment and the motor performance of rats (P < 0.0001), modulation AMPK/SIRT1signaling pathway causing reduction of NF-kB p65 (P < 0.0001) evoked inflammation along with histopathological alterations and glial fibrillary acidic protein (GFAP) immunoreactivity (P < 0.0001), restoration nuclear factor E2-related factor 2 (Nrf2) (P < 0.0001) antioxidant effects, reduction of Bax and elevation of Bcl2 immunoreactivity (P < 0.0001) in addition to boosting mitochondrial biogenesis by upregulation of PTEN-induced kinase-1 (PINK-1)/Parkin (P < 0.0001)and restoration of Brain-derived neurotrophic factor (BDNF) (P = 0.0002)/tropomyosin-related kinase B (TrkB) (P < 0.0001)/cAMP response element-binding (CREB) (P < 0.0001) neuroprotective axis. Collectively, Cilo activates the SIRT1 trajectory to abridge mitochondrial dysfunction invigorated in the HE rat model via restoration of mitochondrial hemostasis.
Collapse
Affiliation(s)
- Enas S Gad
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, AL Ahsa, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University, Kantara Branch, Ismailia, Egypt
| | - Sara A Aldossary
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, AL Ahsa, Saudi Arabia
| | - Mona R El-Ansary
- Department of Biochemistry, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Mona M Abd El-Galil
- Department of Histology and Cell Biology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Asmaa Hassan Abd-El-Hamid
- Department of Histology and Cell Biology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Amira R El-Ansary
- Department of Internal Medicine, Faculty of Medicine, Misr University for Science and Technology, Cairo, Egypt
| | - Noha F Hassan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt.
| |
Collapse
|
3
|
Dey S, Sankaran S. Engineered bacterial therapeutics with material solutions. Trends Biotechnol 2024; 42:1663-1676. [PMID: 39030122 DOI: 10.1016/j.tibtech.2024.06.011] [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: 05/09/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/21/2024]
Abstract
Recent advances in engineered bacterial therapeutics underscore their potential in treating diseases via targeted, live interventions. Despite their promising performance in early clinical phases, no engineered therapeutic bacteria have yet received approval, primarily due to challenges in proving efficacy while ensuring biosafety. Material science innovations, particularly the encapsulation of bacteria within hydrogels, present a promising avenue to enhance bacterial survival, efficacy, and safety in therapeutic applications. This review discusses this interdisciplinary approach to develop living therapeutic materials. Hydrogels not only safeguard the bacteria from harsh physiological conditions but also enable controlled therapeutic release and prevent unintended bacterial dissemination. The strategic use of encapsulation materials could redefine the delivery and functionality of engineered bacterial therapeutics, facilitating their clinical translation.
Collapse
Affiliation(s)
- Sourik Dey
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany
| | | |
Collapse
|
4
|
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] [Grants] [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
|
5
|
Zhang H, Zong R, Wu H, Jiang J, Liu C, Liu S. Transcription factor ASCL1 targets SLC6A13 to inhibit the progression of hepatocellular carcinoma via the glycine-inflammasome signaling. BIOMOLECULES & BIOMEDICINE 2024; 24:1606-1619. [PMID: 38780447 PMCID: PMC11496862 DOI: 10.17305/bb.2024.10328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024]
Abstract
Hepatocellular carcinoma (HCC), the most common primary liver cancer, typically arises from chronic liver conditions such as hepatitis, cirrhosis, or other chronic liver diseases, and is characterized by its aggressive nature and poor prognosis. The purpose of this research was to clarify the function of achaete-scute family bHLH transcription factor 1 (ASCL1) and solute carrier family 6 member 13 (SLC6A13) in influencing tumor cell behavior, inflammatory responses, and the regulation of inflammasomes. We analyzed the differentially expressed genes (DEGs) in the Cancer Genome Atlas-Liver Hepatocellular Carcinoma (TCGA-LIHC) database, as well as in the GSE14520 and GSE67764 datasets, to identify the expression changes of SLC6A13 in liver cancer. The prognostic significance of SLC6A13 in LIHC was assessed through Kaplan-Meier survival curve analysis. Transcriptional regulation of SLC6A13 by ASCL1 was explored using the Joint Annotation of the Human Genome and other species by the Systematic Pipeline for the Annotation of Regulatory Regions (JASPAR) database and dual-luciferase assays. In vitro experiments investigated the impact of ASCL1 and SLC6A13 overexpression on HCC cell growth. Additionally, the effects of ethanol treatment and glycine modulation on the inflammatory response in HCC cell lines were evaluated. HCC samples showed reduced levels of SLC6A13, which correlates with a better prognosis for liver metastases. Elevated SLC6A13 expression correlated with improved overall survival (OS), progression-free survival (PFS), recurrence-free survival (RFS), and disease-specific survival (DSS). ASCL1 upregulated SLC6A13 and inhibited proliferation, migration, and invasion of HCC cells. Ethanol induced the production of inflammatory cytokines, which was enhanced by overexpression of SLC6A13 but counteracted by glycine. This study highlighted elevated expression of SLC6A13 in LIHC which has been correlated with improved OS, PFS, RFS, and DSS. Overexpression of SLC6A13 and ASCL1 in HCC cells enhanced inflammasome activation, which was exacerbated by ethanol and attenuated by glycine.
Collapse
Affiliation(s)
- Hongyan Zhang
- Department of Intensive Care Medicine, The Third Hospital Affiliated to Naval Medical University, Jiading District, Shanghai, China
| | - Ruiqing Zong
- Department of Intensive Care Medicine, The Third Hospital Affiliated to Naval Medical University, Jiading District, Shanghai, China
| | - Huiqi Wu
- Department of Intensive Care Medicine, The Third Hospital Affiliated to Naval Medical University, Jiading District, Shanghai, China
| | - Jun Jiang
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences, Fudan University, Shanghai, China
| | - Chuanyong Liu
- Department of Medical Services, The First Hospital Affiliated to Naval Medical University, Yangpu District, Shanghai, China
| | - Suiyi Liu
- Department of Medical Engineering, The Third Hospital Affiliated to Naval Medical University, Jiading District, Shanghai, China
| |
Collapse
|
6
|
Kleidonas D, Hilfiger L, Lenz M, Häussinger D, Vlachos A. Ammonium chloride reduces excitatory synaptic transmission onto CA1 pyramidal neurons of mouse organotypic slice cultures. Front Cell Neurosci 2024; 18:1410275. [PMID: 39411004 PMCID: PMC11473415 DOI: 10.3389/fncel.2024.1410275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 09/03/2024] [Indexed: 10/19/2024] Open
Abstract
Acute liver dysfunction commonly leads to rapid increases in ammonia concentrations in both the serum and the cerebrospinal fluid. These elevations primarily affect brain astrocytes, causing modifications in their structure and function. However, its impact on neurons is not yet fully understood. In this study, we investigated the impact of elevated ammonium chloride levels (NH4Cl, 5 mM) on synaptic transmission onto CA1 pyramidal neurons in mouse organotypic entorhino-hippocampal tissue cultures. We found that acute exposure to NH4Cl reversibly reduced excitatory synaptic transmission and affected CA3-CA1 synapses. Notably, NH4Cl modified astrocytic, but not CA1 pyramidal neuron, passive intrinsic properties. To further explore the role of astrocytes in NH4Cl-induced attenuation of synaptic transmission, we used methionine sulfoximine to target glutamine synthetase, a key astrocytic enzyme for ammonia clearance in the central nervous system. Inhibition of glutamine synthetase effectively prevented the downregulation of excitatory synaptic activity, underscoring the significant role of astrocytes in adjusting excitatory synapses during acute ammonia elevation.
Collapse
Affiliation(s)
- Dimitrios Kleidonas
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Louis Hilfiger
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
7
|
Eshkiki ZS, Gholami M, Kadkhodaei A, Shayesteh AA. Prognostic indicators and risk factors for the in-hospital mortality rate of patients with cirrhosis. INTERNATIONAL JOURNAL OF GASTROINTESTINAL INTERVENTION 2024; 13:91-97. [DOI: 10.18528/ijgii240032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/12/2024] [Accepted: 07/05/2024] [Indexed: 01/04/2025] Open
Affiliation(s)
- Zahra Shokati Eshkiki
- Alimentary Tract Research Center, Clinical Sciences Research Institute, Imam Khomeini Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mobin Gholami
- Alimentary Tract Research Center, Clinical Sciences Research Institute, Imam Khomeini Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ahmad Kadkhodaei
- Alimentary Tract Research Center, Clinical Sciences Research Institute, Imam Khomeini Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Akbar Shayesteh
- Alimentary Tract Research Center, Clinical Sciences Research Institute, Imam Khomeini Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
8
|
Sedik AA, Hussein DT, Fathy K, Mowaad NA. Neuroprotective and cognitive enhancing effects of herbecetin against thioacetamide induced hepatic encephalopathy in rats via upregulation of AMPK and SIRT1 signaling pathways. Sci Rep 2024; 14:11396. [PMID: 38762495 PMCID: PMC11102433 DOI: 10.1038/s41598-024-61639-6] [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: 02/16/2024] [Accepted: 05/08/2024] [Indexed: 05/20/2024] Open
Abstract
Acute liver injury, there is a risky neurological condition known as hepatic encephalopathy (HE). Herbacetin is a glycosylated flavonoid with many pharmacological characteristics. The purpose of this study was to assess the ability of herbacetin to protect against the cognitive deficits associated with thioacetamide (TAA) rat model and delineate the underlying behavioral and pharmacological mechanisms. Rats were pretreated with herbacetin (20 and 40 mg/kg) for 30days. On 30th day, the rats were injected with TAA (i.p. 350 mg/kg) in a single dose. In addition to a histpathological studies, ultra-structural architecture of the brain, liver functions, oxidative stress biomarkers, and behavioral tests were evaluated. Compared to the TAA-intoxicated group, herbacetin improved the locomotor and cognitive deficits, serum hepatotoxicity indices and ammonia levels. Herbacetin reduced brain levels of malodialdeyde, glutamine synthetase (GS), tumor necrosis factor- alpha (TNF-α), interleukin 1 B (IL-1β), annexin v, and increased brain GSH, Sirtuin 1 (SIRT1), and AMP-activated kinase (AMPK) expression levels. Also, herbacetin improve the histopathological changes and ultra- structure of brain tissue via attenuating the number of inflammatory and apoptotic cells. Herbacetin treatment significantly reduced the toxicity caused by TAA. These findings suggest that herbacetin might be taken into account as a possible neuroprotective and cognitive enhancing agent due to its ability to reduce oxidative stress, inflammation and apoptosis associated with TAA.
Collapse
Affiliation(s)
- Ahmed A Sedik
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, El-Buhouth St., Dokki, Cairo, 12622, Egypt
| | - Dalia T Hussein
- Fellow of Biochemistry, Children Hospital, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Khaled Fathy
- Electron Microscopy Unit, Mansoura University, El Mansoura, 35516, Egypt
| | - Noha A Mowaad
- Narcotics, Ergogenics and Poisons Department, Medical Research and Clinical Studies Institute, National Research Centre, El-Buhouth St., Dokki, Cairo, 12622, Egypt.
| |
Collapse
|
9
|
Nady R, Ahmed RR, Moustafa N, Abdul-Hamid M. TNF-α blockage by etanercept restores spatial learning and reduces cellular degeneration in the hippocampus during liver cirrhosis. Tissue Cell 2023; 85:102249. [PMID: 37865039 DOI: 10.1016/j.tice.2023.102249] [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: 05/11/2023] [Revised: 09/29/2023] [Accepted: 10/16/2023] [Indexed: 10/23/2023]
Abstract
Hepatic encephalopathy (HE) is one of the most debilitating cerebral complications of liver cirrhosis. The one-year survival of patients with liver cirrhosis and severe encephalopathy is less than 50%. Recent studies have indicated that neuroinflammation is a new player in the pathogenesis of HE, which seems to be involved in the development of cognitive impairment. In this study, we demonstrated neurobehavioral and neuropathological consequences of liver cirrhosis and tested the therapeutic potential of the tumor necrosis factor-α (TNF-α) inhibitor, etanercept. Sixty male adult Wistar albino rats (120-190 g) were allocated into four groups, where groups I and IV served as controls. Thioacetamide (TAA; 300 mg/kg) was intraperitoneally injected twice a week for five months to induce liver cirrhosis in group II (n = 20). Both TAA and etanercept (2 mg/kg) were administered to group III (n = 20). At the end of the experiment, spatial learning was assessed using Morris water maze. TNF-α was detected in both serum and hippocampus. The excised brains were also immunohistochemically stained with glial fibrillary acidic protein (GFAP) to estimate both the number and integrity of hippocampal astrocytes. Ultrastructural changes in the hippocampus were characterized by transmission electron microscopy. The results showed that blocking TNF-α by etanercept was accompanied by a lower TNF-α expression and a higher number of GFAP-positive astrocytes in the hippocampus. Etanercept intervention alleviated the neuronal and glial degenerative changes and impeded the deterioration of spatial learning ability. In conclusion, TNF-α is strongly involved in the development of liver cirrhosis and the associated encephalopathy. TNF-α blockers may be a promising approach for management of hepatic cirrhosis and its cerebral complications.
Collapse
Affiliation(s)
- Rehab Nady
- Cell Biology, Histology and Genetics Division, Department of Zoology, Faculty of Science, Beni-Suef University, Beni Suef 62511, Egypt
| | - Rasha R Ahmed
- Cell Biology, Histology and Genetics Division, Department of Zoology, Faculty of Science, Beni-Suef University, Beni Suef 62511, Egypt
| | - Nadia Moustafa
- Cell Biology, Histology and Genetics Division, Department of Zoology, Faculty of Science, Beni-Suef University, Beni Suef 62511, Egypt
| | - Manal Abdul-Hamid
- Cell Biology, Histology and Genetics Division, Department of Zoology, Faculty of Science, Beni-Suef University, Beni Suef 62511, Egypt.
| |
Collapse
|
10
|
Gilbert MC, Setayesh T, Wan YJY. The contributions of bacteria metabolites to the development of hepatic encephalopathy. LIVER RESEARCH 2023; 7:296-303. [PMID: 38221945 PMCID: PMC10786625 DOI: 10.1016/j.livres.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Over 20% of mortality during acute liver failure is associated with the development of hepatic encephalopathy (HE). Thus, HE is a complication of acute liver failure with a broad spectrum of neuropsychiatric abnormalities ranging from subclinical alterations to coma. HE is caused by the diversion of portal blood into systemic circulation through portosystemic collateral vessels. Thus, the brain is exposed to intestinal-derived toxic substances. Moreover, the strategies to prevent advancement and improve the prognosis of such a liver-brain disease rely on intestinal microbial modulation. This is supported by the findings that antibiotics such as rifaximin and laxative lactulose can alleviate hepatic cirrhosis and/or prevent HE. Together, the significance of the gut-liver-brain axis in human health warrants attention. This review paper focuses on the roles of bacteria metabolites, mainly ammonia and bile acids (BAs) as well as BA receptors in HE. The literature search conducted for this review included searches for phrases such as BA receptors, BAs, ammonia, farnesoid X receptor (FXR), G protein-coupled bile acid receptor 1 (GPBAR1 or TGR5), sphingosine-1-phosphate receptor 2 (S1PR2), and cirrhosis in conjunction with the phrase hepatic encephalopathy and portosystemic encephalopathy. PubMed, as well as Google Scholar, was the search engines used to find relevant publications.
Collapse
Affiliation(s)
- Miranda Claire Gilbert
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA
| | - Tahereh Setayesh
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA
| | - Yu-Jui Yvonne Wan
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
11
|
Giri S, Singh A, Angadi S, Kolhe K, Roy A. Prevalence of hepatic encephalopathy in patients with non-cirrhotic portal hypertension: A systematic review and meta-analysis. Indian J Gastroenterol 2023; 42:642-650. [PMID: 37589913 DOI: 10.1007/s12664-023-01412-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 06/02/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND Hepatic encephalopathy, (HE) although commonly associated with cirrhosis, has also been reported in non-cirrhotic portal hypertension (NCPH). The importance of identifying and treating HE in NCPH lies in the fact that many patients may be wrongly diagnosed as having psychiatric or neurologic disorders. Hence, we aimed to systematically review the prevalence of HE in NCPH. METHODS A comprehensive search of three databases (Medline, Embase and Scopus) was conducted from inception to November 2022 for studies reporting on the prevalence of minimal HE (MHE) and overt HE (OHE) in patients with NCPH. Results were presented as pooled proportions with their 95% confidence intervals (CI). RESULTS Total 25 studies (n = 1487) were included after screening 551 records. The pooled prevalence of MHE in NPCH was 32.9% (95% CI: 26.7-39.0) without any difference between adult (32.9%, 95% CI: 23.5-42.3) and pediatric patients (32.6%, 95% CI: 26.1-39.1) (p = 0.941). There was no significant difference in the prevalence between patients with NCPH and compensated cirrhosis with odds ratio of 1.06 (95% CI: 0.77-1.44). The pooled event rate for prior history of OHE in NCPH was 1.2% (95% CI: 0.3-2.1). CONCLUSION Around one-third of the patients with NCPH have MHE, irrespective of age group. OHE is extremely rare in NCPH and is usually associated with a precipitating factor.
Collapse
Affiliation(s)
- Suprabhat Giri
- Department of Gastroenterology, Nizam's Institute of Medical Sciences, Hyderabad, 500 082, India
| | - Ankita Singh
- Department of Gastroenterology, Seth G S Medical College and K E M Hospital, Mumbai, 400 012, India
| | - Sumaswi Angadi
- Department of Gastroenterology, Nizam's Institute of Medical Sciences, Hyderabad, 500 082, India
| | - Kailash Kolhe
- Department of Gastroenterology, Narayana Hospital, Nanded, 431 602, India
| | - Akash Roy
- Institute of Gastrosciences and Liver Transplantation, Apollo Multispecialty Hospital, Kolkata, 700 054, India.
| |
Collapse
|
12
|
Cheon SY, Kim MY, Kim J, Kim EJ, Kam EH, Cho I, Koo BN, Kim SY. Hyperammonemia induces microglial NLRP3 inflammasome activation via mitochondrial oxidative stress in hepatic encephalopathy. Biomed J 2023; 46:100593. [PMID: 37059364 PMCID: PMC10498413 DOI: 10.1016/j.bj.2023.04.001] [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: 01/08/2023] [Revised: 03/10/2023] [Accepted: 04/04/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND The role of nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome in the pathogenesis of hepatic encephalopathy (HE) is unclear. Mitochondrial reactive oxygen species (mtROS) is a signal for NLRP3 inflammasome activation. Therefore, we aimed to determine whether mtROS-dependent NLRP3 inflammasome activation is involved in HE, using in vivo and in vitro models. METHODS Bile duct ligation (BDL) in C57/BL6 mice was used as an in vivo HE model. NLRP3 activation was assessed in the hippocampus. Immunofluorescence staining was performed to determine the cellular source of NLRP3 in the hippocampal tissue. For the in vitro experiment, BV-2 microglial cells were primed with lipopolysaccharide (LPS), followed by ammonia treatment. NLRP3 activation and mitochondrial dysfunction were measured. Mito-TEMPO was used to suppress mtROS production. RESULTS BDL mice showed cognitive impairment with hyperammonemia. Both the priming and activation steps of NLRP3 inflammasome activation were processed in the hippocampus of BDL mice. Moreover, intracellular ROS levels increased in the hippocampus, and NLRP3 was mainly expressed in the microglia of the hippocampus. In LPS-primed BV-2 cells, ammonia treatment induced NLRP3 inflammasome activation and pyroptosis, with elevation of mtROS and altered mitochondrial membrane potential. Pretreatment with Mito-TEMPO suppressed mtROS production and the subsequent NLRP3 inflammasome activation and pyroptosis under LPS and ammonia treatment in BV-2 cells. CONCLUSIONS Hyperammonemia in HE may be involved in mtROS overproduction and subsequent NLRP3 inflammasome activation. Further studies using NLRP3-specific inhibitor or NLRP3 knockout mice are needed to elucidate the important role of NLRP3 inflammasome in HE development.
Collapse
Affiliation(s)
- So Yeong Cheon
- Department of Biotechnology, College of Biomedical & Health Science, Konkuk University, Chungju, Republic of Korea
| | - Min-Yu Kim
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jeongmin Kim
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Jung Kim
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Hee Kam
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Inja Cho
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Bon-Nyeo Koo
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - So Yeon Kim
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
13
|
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: 2] [Impact Index Per Article: 1.0] [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
|
14
|
Bahrami T, Yaghmaei P, Yousofvand N. The effects of Ibuprofen and 1, 8- cineol on anxiety and spatial memory in hyperammonemic rats. Metab Brain Dis 2023; 38:613-620. [PMID: 36346500 DOI: 10.1007/s11011-022-01093-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 09/29/2022] [Indexed: 11/11/2022]
Abstract
In hepatic encephalopathy, hyperammonemia (HA) causes cognitive impairment and anxiety by causing neuroinflammation. Ibuprofen and 1,8- cineol have anti-inflammatory and antioxidant properties, respectively. The aim of this study was to evaluate the effects of ibuprofen alone and in combination with 1,8- cineol on anxiety and oxidative stress in a HA rat animal model. For this purpose, 36 rats were divided into six groups (n = 6) including the HA (received intraperitoneally (IP) ammonium acetate 2.5 mg/kg for four week), ibuprofen (induced HA rats that received 15 mg/kg, IP), cineol (induced HA rats that received 5 and 10 mg/kg, IP), Ib + cineol (induced HA rats that received 15 and 10 mg/kg, respectively, IP), and the control groups (received normal saline, IP). Except the HA group, all other groups received the aforementioned treatment for two weeks.. The Morris water maze and elevated plus maze were used to assess cognitive function and anxiety in the animals, respectively. Superoxide dismutase (SOD) activity was measured to evaluate oxidative stress. The mRNA expression levels of interleukin (IL)-6 and IL-1β was assessed by real-time PCR in the animal's brain. The results showed a significant improvement in spatial memory and anxiety of the Ib group compared to the HA group (P < 0.01), but no significant change was observed in SOD activity (P > 0.05). There was a significant improvement in spatial memory and anxiety as well as a significant increase in SOD activity in the Ib + cineol group (P < 0.01) compared to the HA group. These results indicate that the Ib + cineol, not only improve cognitive function and reduce anxiety, also reduce oxidative stress, therefore, the simultaneous use of these two compounds may be useful in improving HA-induced cognitive disorders and anxiety.
Collapse
Affiliation(s)
- Tayebeh Bahrami
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Parichehreh Yaghmaei
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Namdar Yousofvand
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran.
| |
Collapse
|
15
|
Ashwagandha-loaded nanocapsules improved the behavioral alterations, and blocked MAPK and induced Nrf2 signaling pathways in a hepatic encephalopathy rat model. Drug Deliv Transl Res 2023; 13:252-274. [PMID: 35672652 PMCID: PMC9726678 DOI: 10.1007/s13346-022-01181-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2022] [Indexed: 12/14/2022]
Abstract
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.
Collapse
|
16
|
Abdelghffar EAR, El-Nashar HAS, Fayez S, Obaid WA, Eldahshan OA. Ameliorative effect of oregano (Origanum vulgare) versus silymarin in experimentally induced hepatic encephalopathy. Sci Rep 2022; 12:17854. [PMID: 36284120 PMCID: PMC9596437 DOI: 10.1038/s41598-022-20412-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 09/13/2022] [Indexed: 01/20/2023] Open
Abstract
Hepatic encephalopathy (HE) is a deterioration of brain function in patients suffering from chronic liver disease, cirrhosis as a result of elevated blood ammonia and the production of pseudo-neurotransmitters. Herein, we investigated the chemical composition of hexane extract from Origanum vulgare (O. vulgare) leaves as well as its possible protective effects against thioacetamide (TAA)-induced HE in rats. GC-MS analysis of the extract revealed tentative identification of twenty-five compounds (82.93%), predominated by cholesten-3-one (27.30%), followed by γ-tocopherol (13.52%), α-tocopherol (5.01%), β-amyrin (5.24%) and α-amyrin (4.89%). Albino rats were distributed into seven groups (n = 7). G1 served as negative control; G2 and G3 served as controls treated with O. vulgare (100 and 200 mg/kg/p.o b.w, respectively); G4 served as TAA-positive control group (100 mg/kg/day/i.p., three alternative days per week for six weeks); G5, G6, and G7 served as TAA -induced HE rat model that received O. vulgare 100, O. vulgare 200, and silymarin (100 mg/kg of SILY, as standard drug), respectively. TAA showed depressive and anxiety-like behaviors in forced swimming test (FST) and reduction of cognitive score in elevated plus-maze test (EPMT) as well as impairment of locomotor and exploratory activities in open-field test (OFT). TAA caused a significant decline in body weight gain; however, the relative liver weight and brain water content were statistically increased. TAA-intoxicated rats showed significant increase of serum biomarker enzymes, proinflammatory cytokines, blood ammonia levels, brain serotonin, acetyl cholinesterase and cellular lipid peroxidation with significant decrease of brain dopamine, norepinephrine, antioxidant status. The hepatoprotective/neuro-protective activities of O. vulgare was found to be comparable with that of SILY in HE rats model. Where, treatment of TAA-intoxicated rats with O. vulgare attenuated anxiety, depressive-related behaviors, and reduced the biochemical changes in HE-induced by TAA. Therefore, O. vulgare could be an excellent hepato-/neuroprotective against hepatic injury and HE via improving the oxidative/inflammatory status through its antioxidant and neuro-modulatory properties and its effect is equal to that of SILY.
Collapse
Affiliation(s)
- Eman A. R. Abdelghffar
- grid.7269.a0000 0004 0621 1570Department of Zoology, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Heba A. S. El-Nashar
- grid.7269.a0000 0004 0621 1570Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Cairo, Egypt ,grid.7269.a0000 0004 0621 1570Centre of Drug Discovery Research and Development, Ain Shams University, Cairo, Egypt
| | - Shaimaa Fayez
- grid.7269.a0000 0004 0621 1570Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Cairo, Egypt ,grid.7269.a0000 0004 0621 1570Centre of Drug Discovery Research and Development, Ain Shams University, Cairo, Egypt
| | - Wael A. Obaid
- grid.412892.40000 0004 1754 9358Department of Biology, College of Science, Taibah University, Al-Madīnah Al-Munawarah, Saudi Arabia
| | - Omayma A. Eldahshan
- grid.7269.a0000 0004 0621 1570Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Cairo, Egypt ,grid.7269.a0000 0004 0621 1570Centre of Drug Discovery Research and Development, Ain Shams University, Cairo, Egypt
| |
Collapse
|
17
|
Xu J, Hao J, Gao Y, Shang L, Qian L, Zhao H. Hepatic cortical blindness. Am J Med Sci 2022; 364:492-498. [PMID: 35595077 DOI: 10.1016/j.amjms.2022.04.034] [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: 07/08/2020] [Revised: 10/26/2021] [Accepted: 04/28/2022] [Indexed: 01/25/2023]
Abstract
Hepatic cortical blindness is an unusual clinical complication of hepatic encephalopathy, and its epidemiology has not been studied in detail. Herein, we present a case report of a 37-year-old male with liver cirrhosis and immunoglobulin A nephropathy who noted visual impairment after eating high-protein food. In addition to the case report, all previously documented cases of hepatic cortical blindness occurring across the globe published from inception until August 2019 were reviewed. The incidence of hepatic cortical blindness is low, and diagnoses are easily missed. The diagnosis of hepatic cortical blindness is dependent on symptoms, and patients may experience several attacks of vision loss. Most patients experience complete blindness, but vision loss recovers within 10 min to one month. Early detection and therapy for hepatic encephalopathy may improve patient outcome.
Collapse
Affiliation(s)
- Junrong Xu
- Department of Gastroenterology, The Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, China
| | - Jie Hao
- Emergency Department, Shaanxi Provincial Hospital of Chinese Medicine, Xi'an, Shaanxi, China
| | - Yi Gao
- Department of Nephropathy, The Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, China
| | - Lei Shang
- Department of Health Statistics, Air Force Medical University, No.169 Changle west road, Xi'an, Shaanxi, China.
| | - Lu Qian
- Department of General Medicine, The Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, No.10 Eastern section of the third Fengcheng road, Xi'an, Shaanxi, China.
| | - Hengfang Zhao
- Department of Gastroenterology, The Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
18
|
Abdelaziz RR, Abdelrahman RS, Abdelmageed ME. SB332235, a CXCR2 antagonist, ameliorates thioacetamide-induced hepatic encephalopathy through modulation of the PI3K/AKT pathways in rats. Neurotoxicology 2022; 92:110-121. [PMID: 35961375 DOI: 10.1016/j.neuro.2022.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 06/27/2022] [Accepted: 08/08/2022] [Indexed: 11/29/2022]
Abstract
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.
Collapse
Affiliation(s)
- Rania R Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt
| | - Rehab S Abdelrahman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Taibah University, Al-Madina Al-Munawwarah, 30001, Saudi Arabia
| | - Marwa E Abdelmageed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt.
| |
Collapse
|
19
|
Angelova PR, Kerbert AJ, Habtesion A, Hall A, Abramov AY, Jalan R. Hyperammonemia induces mitochondrial dysfunction and neuronal cell death. JHEP REPORTS : INNOVATION IN HEPATOLOGY 2022; 4:100510. [PMID: 35845295 PMCID: PMC9278080 DOI: 10.1016/j.jhepr.2022.100510] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/21/2022] [Accepted: 05/10/2022] [Indexed: 11/27/2022]
Abstract
Background & Aims In cirrhosis, astrocytic swelling is believed to be the principal mechanism of ammonia neurotoxicity leading to hepatic encephalopathy (HE). The role of neuronal dysfunction in HE is not clear. We aimed to explore the impact of hyperammonaemia on mitochondrial function in primary co-cultures of neurons and astrocytes and in acute brain slices of cirrhotic rats using live cell imaging. Methods To primary cocultures of astrocytes and neurons, low concentrations (1 and 5 μM) of NH4Cl were applied. In rats with bile duct ligation (BDL)-induced cirrhosis, a model known to induce hyperammonaemia and minimal HE, acute brain slices were studied. One group of BDL rats was treated twice daily with the ammonia scavenger ornithine phenylacetate (OP; 0.3 g/kg). Fluorescence measurements of changes in mitochondrial membrane potential (Δψm), cytosolic and mitochondrial reactive oxygen species (ROS) production, lipid peroxidation (LP) rates, and cell viability were performed using confocal microscopy. Results Neuronal cultures treated with NH4Cl exhibited mitochondrial dysfunction, ROS overproduction, and reduced cell viability (27.8 ± 2.3% and 41.5 ± 3.7%, respectively) compared with untreated cultures (15.7 ± 1.0%, both p <0.0001). BDL led to increased cerebral LP (p = 0.0003) and cytosolic ROS generation (p <0.0001), which was restored by OP (both p <0.0001). Mitochondrial function was severely compromised in BDL, resulting in hyperpolarisation of Δψm with consequent overconsumption of adenosine triphosphate and augmentation of mitochondrial ROS production. Administration of OP restored Δψm. In BDL animals, neuronal loss was observed in hippocampal areas, which was partially prevented by OP. Conclusions Our results elucidate that low-grade hyperammonaemia in cirrhosis can severely impact on brain mitochondrial function. Profound neuronal injury was observed in hyperammonaemic conditions, which was partially reversible by OP. This points towards a novel mechanism of HE development. Lay summary The impact of hyperammonaemia, a common finding in patients with liver cirrhosis, on brain mitochondrial function was investigated in this study. The results show that ammonia in concentrations commonly seen in patients induces severe mitochondrial dysfunction, overproduction of damaging oxygen molecules, and profound injury and death of neurons in rat brain cells. These findings point towards a novel mechanism of ammonia-induced brain injury in liver failure and potential novel therapeutic targets. Low concentrations of ammonia induce mitochondrial dysfunction, overproduction of ROS, and cell death in primary neurons. Hyperammonaemia in cirrhotic rats leads to ROS and LP overproduction, which was prevented by the ammonia scavenger OP. In neurons from cirrhotic rats, hyperpolarisation of Δψm was observed, which was restored by OP treatment. In a rat model of cirrhosis, profound neuronal loss was observed in the hippocampus.
Collapse
|
20
|
A critical review of bile acids and their receptors in hepatic encephalopathy. Anal Biochem 2022; 643:114436. [PMID: 34715070 PMCID: PMC9798441 DOI: 10.1016/j.ab.2021.114436] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/15/2021] [Accepted: 10/22/2021] [Indexed: 01/01/2023]
Abstract
Hepatic encephalopathy describes an array of neurological complications that arise due to liver insufficiency. The pathogenesis of hepatic encephalopathy shares a longstanding association with hyperammonemia and inflammation, and recently, aberrant bile acid signaling has been implicated in the development of key features of hepatic encephalopathy. These key features include neuronal dysfunction, neuroinflammation and blood-brain barrier permeability. This review summarizes the findings of recent studies demonstrating a role for bile acids in the pathogenesis of hepatic encephalopathy via one of three main bile acid receptors and speculates on the possible downstream consequences of aberrant bile acid signaling.
Collapse
|
21
|
Serrano-Castro PJ, Garzón-Maldonado FJ, Casado-Naranjo I, Ollero-Ortiz A, Mínguez-Castellanos A, Iglesias-Espinosa M, Baena-Palomino P, Sánchez-Sanchez V, Sánchez-Pérez RM, Rubi-Callejon J, Estévez-María JC, Galeano-Bilbao B, Romero-Imbroda J, Sobrino B, Arrabal-Gomez C, Oliver-Martos B, Muñoz-Becerra L, Requena N, González Álvarez de Sotomayor MDM, Estivill-Torrus G, Suarez J, Ciano-Petersen NL, Pons-Pons G, Reyes-Bueno JA, Cabezudo-Garcia P, Aguilar-Castillo MJ, De la Cruz Cosme C, Duque-Holguera M, Cuartero-Rodriguez E, Vilches-Carrillo RM, Carrera-Muñoz I, Carnero-Pardo C, Ramirez-Garcia T, Oropesa JM, Dominguez-Mayoral A, Pelaez-Viñas N, Valiente L, de Fonseca FR. The cognitive and psychiatric subacute impairment in severe Covid-19. Sci Rep 2022; 12:3563. [PMID: 35241761 PMCID: PMC8894467 DOI: 10.1038/s41598-022-07559-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 02/21/2022] [Indexed: 02/08/2023] Open
Abstract
Neurologic impairment persisting months after acute severe SARS-CoV-2 infection has been described because of several pathogenic mechanisms, including persistent systemic inflammation. The objective of this study is to analyze the selective involvement of the different cognitive domains and the existence of related biomarkers. Cross-sectional multicentric study of patients who survived severe infection with SARS-CoV-2 consecutively recruited between 90 and 120 days after hospital discharge. All patients underwent an exhaustive study of cognitive functions as well as plasma determination of pro-inflammatory, neurotrophic factors and light-chain neurofilaments. A principal component analysis extracted the main independent characteristics of the syndrome. 152 patients were recruited. The results of our study preferential involvement of episodic and working memory, executive functions, and attention and relatively less affectation of other cortical functions. In addition, anxiety and depression pictures are constant in our cohort. Several plasma chemokines concentrations were elevated compared with both, a non-SARS-Cov2 infected cohort of neurological outpatients or a control healthy general population. Severe Covid-19 patients can develop an amnesic and dysexecutive syndrome with neuropsychiatric manifestations. We do not know if the deficits detected can persist in the long term and if this can trigger or accelerate the onset of neurodegenerative diseases.
Collapse
Affiliation(s)
- Pedro J Serrano-Castro
- Neurology Service, Regional University Hospital of Malaga, Malaga, Spain.
- Neuroimmunology and Neuroinflamation Group, Institute of Biomedical Research of Malaga (IBIMA), Malaga, Spain.
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain.
- Neurology Department, Instituto IBIMA, Hospital Regional Universitario de Málaga (España), Avda Carlos-Haya S/N, 4a planta, Malaga, Spain.
| | - Francisco J Garzón-Maldonado
- Neurology Service, Virgen de la Victoria University Hospital, Malaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Ignacio Casado-Naranjo
- Neurology Service, University Hospital of Cáceres, Cáceres, Spain
- Biomedical Network Research Center for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- University Institute of Biosanitary Research of Extremadura (INUBE), Cáceres, Spain
| | - Angela Ollero-Ortiz
- Neurology Service, Valme University Hospital, Seville, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Adolfo Mínguez-Castellanos
- Neurology Service, Virgen de las Nieves University Hospital, Granada, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
- Institute of Biosanitary Research of Granada (Ibs. GRANADA), Granada, Spain
| | - Mar Iglesias-Espinosa
- Neurology Service, Torrecárdenas University Hospital, Almería, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Pablo Baena-Palomino
- Neurology Service, Juan Ramón Jiménez University Hospital, Huelva, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Violeta Sánchez-Sanchez
- Neurology Service, Virgen Macarena University Hospital, Seville, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | | | - José Rubi-Callejon
- Neurology Unit, Internal Medicine Service, Hospital del Poniente de Almería, El Ejido (Almería), Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - José Carlos Estévez-María
- Neurology Service, Reina Sofia University Hospital, Cordoba, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Benito Galeano-Bilbao
- Neurology Service, Puerta del Mar University Hospital, Cadiz, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Jesús Romero-Imbroda
- Neurology Service, Regional University Hospital of Malaga, Malaga, Spain
- Neurology Service, Hospital Quirón-Salud Málaga, Malaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Beatriz Sobrino
- Infectious Diseases Service, Regional University Hospital of Malaga, Malaga, Spain
| | - Carlos Arrabal-Gomez
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Begoña Oliver-Martos
- Neuroimmunology and Neuroinflamation Group, Institute of Biomedical Research of Malaga (IBIMA), Malaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Luis Muñoz-Becerra
- Neurology Service, Regional University Hospital of Malaga, Malaga, Spain
- Neuroimmunology and Neuroinflamation Group, Institute of Biomedical Research of Malaga (IBIMA), Malaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Nerea Requena
- Neurology Service, Virgen de la Victoria University Hospital, Malaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - María Del Mar González Álvarez de Sotomayor
- Neurology Service, Regional University Hospital of Malaga, Malaga, Spain
- Neuroimmunology and Neuroinflamation Group, Institute of Biomedical Research of Malaga (IBIMA), Malaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Guillermo Estivill-Torrus
- Neurology Service, Regional University Hospital of Malaga, Malaga, Spain
- Neuroimmunology and Neuroinflamation Group, Institute of Biomedical Research of Malaga (IBIMA), Malaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Juan Suarez
- Neuropsychopharmacology Group, Institute of Biomedical Research of Malaga (IBIMA), Malaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Nicolas Lundahl Ciano-Petersen
- Neurology Service, Regional University Hospital of Malaga, Malaga, Spain
- Neuroimmunology and Neuroinflamation Group, Institute of Biomedical Research of Malaga (IBIMA), Malaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Gracia Pons-Pons
- Neurology Service, Regional University Hospital of Malaga, Malaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Jose Antonio Reyes-Bueno
- Neurology Service, Regional University Hospital of Malaga, Malaga, Spain
- Neuroimmunology and Neuroinflamation Group, Institute of Biomedical Research of Malaga (IBIMA), Malaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Pablo Cabezudo-Garcia
- Neurology Service, Regional University Hospital of Malaga, Malaga, Spain
- Neuroimmunology and Neuroinflamation Group, Institute of Biomedical Research of Malaga (IBIMA), Malaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Maria José Aguilar-Castillo
- Biotechnology Unit, Regional University Hospital of Malaga, Malaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Carlos De la Cruz Cosme
- Neurology Service, Virgen de la Victoria University Hospital, Malaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | | | - Eva Cuartero-Rodriguez
- Neurology Service, Valme University Hospital, Seville, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Rosa María Vilches-Carrillo
- Neurology Service, Virgen de las Nieves University Hospital, Granada, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Ismael Carrera-Muñoz
- Neurology Service, Virgen de las Nieves University Hospital, Granada, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | | | - Teresa Ramirez-Garcia
- Neurology Service, Torrecárdenas University Hospital, Almería, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Juan Manuel Oropesa
- Neurology Service, Juan Ramón Jiménez University Hospital, Huelva, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Ana Dominguez-Mayoral
- Neurology Service, Virgen Macarena University Hospital, Seville, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Nazaret Pelaez-Viñas
- Neurology Service, Reina Sofia University Hospital, Cordoba, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain
| | - Lucia Valiente
- Infectious Diseases Service, Regional University Hospital of Malaga, Malaga, Spain
| | - Fernando Rodríguez de Fonseca
- Neuropsychopharmacology Group, Institute of Biomedical Research of Malaga (IBIMA), Malaga, Spain.
- Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), Malaga, Spain.
- Neuropsychopharmacology Group, Instituto IBIMA, Hospital Regional Universitario de Málaga (España), Avda Carlos-Haya S/N, Malaga, Spain.
| |
Collapse
|
22
|
Protocatechuic acid protects against thioacetamide-induced chronic liver injury and encephalopathy in mice via modulating mTOR, p53 and the IL-6/ IL-17/ IL-23 immunoinflammatory pathway. Toxicol Appl Pharmacol 2022; 440:115931. [PMID: 35202709 DOI: 10.1016/j.taap.2022.115931] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 01/28/2022] [Accepted: 02/17/2022] [Indexed: 02/07/2023]
Abstract
Protocatechuic acid (PCA), a natural phenolic acid, is known for antioxidant, anti-inflammatory, anti-apoptotic, and anti-fibrotic activities. However, the protective mechanisms of PCA on thioacetamide (TAA)-induced liver/brain injury are not well addressed. Chronic liver injury was induced in mice by intraperitoneal injection of TAA (200 mg/kg, 3 times/week) for 8 weeks. Simultaneously, PCA (100, 150 mg/kg/day, p.o.) was given daily from the 4th week. Protocatechuic acid ameliorated liver and brain damage indicated by the decrease in serum activities of aminotransferases, gamma-glutamyl transferase, alkaline phosphatase, lactate dehydrogenase, levels of bilirubin, and ammonia concomitant with restoration of normal albumin levels. Additionally, PCA treatment ameliorated oxidative stress in liver and brain, confirmed by the decrease in malondialdehyde and nitric oxide levels and the increase in antioxidant activities. Moreover, PCA showed anti-inflammatory actions through downregulation of TNF-α expression in the liver and IL-6/IL-17/IL-23 levels in the brain, which is confirmed by the decrease in CD4+ T brain cell numbers. Most importantly, PCA treatment showed a significant decrease in mTOR level and number of LC3 positive cells in both liver and brain tissues. Consequently, PCA could inhibit mTOR-induced apoptosis, as it showed anti-apoptotic actions through downregulation of caspase-3 expression in liver and p53 expression in liver and brain. Furthermore, liver and brain tissues of treated mice showed restoration of normal histology. It can be concluded that, several mechanisms, including: antioxidant, anti-inflammatory, anti-autophagic and anti-apoptotic activities can be implicated in the hepato- and neuroprotective potentials of PCA.
Collapse
|
23
|
El-Baz FK, Elgohary R, Salama A. Amelioration of Hepatic Encephalopathy Using Dunaliella salina Microalgae in Rats: Modulation of Hyperammonemia/TLR4. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8843218. [PMID: 33855084 PMCID: PMC8021475 DOI: 10.1155/2021/8843218] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 02/27/2021] [Accepted: 03/19/2021] [Indexed: 12/28/2022]
Abstract
Hepatic encephalopathy (HE) is a neuropsychiatric disease that is developed as a complication of both acute and chronic liver failure affecting psychomotor dysfunction, memory, and concentration. This study is aimed at evaluating the therapeutic effects of Dunaliella salina (D. salina) microalgae in thioacetamide- (TAA-) induced HE in rats. HE was induced by TAA (200 mg/kg; i.p.) for three successive days. Forty male Wister albino rats were divided into 4 groups; the first group was served as a normal, and the second group was injected with TAA and served as TAA control. The third and fourth groups were administered D. salina (100 and 200 mg/kg; p.o.), respectively, after TAA injection for 7 days. The behavioral and biochemical markers as well as histological aspects of HE were estimated. This study revealed that TAA caused behavioral changes, oxidative stress, neuroinflammation, nuclear pyknosis, and neurons degeneration. D. salina improved liver function and decreased oxidative stress and inflammatory mediator as TLR4 protein expression. Also, D. salina elevated HSP-25 and IGF-1 as well as improved brain histopathological alterations. In conclusion, D. salina exerted a therapeutic potential against HE via its antioxidant, antiinflammatory and cytoprotective effects.
Collapse
Affiliation(s)
- Farouk K. El-Baz
- Plant Biochemistry Department, National Research Centre (NRC), 33 El Buhouth St., Dokki, Cairo 12622, Egypt
| | - Rania Elgohary
- Narcotics, Ergogenics and Poisons Department, National Research Centre (NRC), 33 El Buhouth St., Dokki, Cairo 12622, Egypt
| | - Abeer Salama
- Pharmacology Department, National Research Centre (NRC), 33 El Buhouth St., Dokki, Cairo 12622, Egypt
| |
Collapse
|
24
|
Brain-derived neurotrophic factor as a potential diagnostic marker in minimal hepatic encephalopathy. Clin Exp Hepatol 2021; 7:117-124. [PMID: 34027124 PMCID: PMC8122095 DOI: 10.5114/ceh.2021.103242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/15/2021] [Indexed: 11/22/2022] Open
Abstract
Introduction Minimal hepatic encephalopathy (MHE) is a common complication of liver cirrhosis not only leading to a decrease in the quality of life, but also predicting development of overt encephalopathy. The diagnosis of MHE usually relies on a combination of neuropsychological tests, while robust serum biomarkers are lacking. We aimed to assess serum concentrations of brain-derived neurotrophic factor (BDNF) in MHE patients. Material and methods Serum BDNF was assessed in 78 patients with liver cirrhosis (53 male, median age 55 years) and 40 healthy individuals. 43 subjects underwent extensive evaluation for MHE by psychometric hepatic encephalopathy score (PHES) and inhibitory control test (ICT) or critical flicker frequency (CFF). Results Serum BDNF was twofold lower in liver cirrhosis compared to healthy subjects [13.6 (7.8-22.6) vs. 33.0 (24.1-40.7) ng/ml, p < 0.001] and its decrease reflected a degree of liver insufficiency assessed by model for end-stage liver disease (MELD). BDNF showed a negative correlation with bilirubin (R = –0.35, p = 0.005) and international normalized ratio (INR) (R = –0.37, p = 0.003), and positive with platelets (PLT) (R = 0.36, p = 0.004), while no associations with age, sex, body mass index (BMI), waist-hip ratio (WHR), creatinine and ammonia were noted. Importantly, subjects with a diagnosis of MHE by at least two modalities showed the lowest levels of BDNF [10.9 (2.5-14.4) vs. 19.9 (9.3-29.4) ng/ml, p < 0.01]. Patients with self-reported sleep disturbances had significantly lower serum BDNF [13.0 (2.5-23.4) vs. 20.0 (8.4-31.3) ng/ml, p = 0.04]. Conclusions The lowest serum BDNF concentration was noted in patients with MHE and sleep disturbances, which suggests a role in pathophysiology of hepatic encephalopathy but also as a potential biomarker.
Collapse
|
25
|
Farshad O, Keshavarz P, Heidari R, Farahmandnejad M, Azhdari S, Jamshidzadeh A. The Potential Neuroprotective Role of Citicoline in Hepatic Encephalopathy. J Exp Pharmacol 2020; 12:517-527. [PMID: 33235522 PMCID: PMC7678475 DOI: 10.2147/jep.s261986] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 11/03/2020] [Indexed: 12/29/2022] Open
Abstract
Purpose Hepatic encephalopathy (HE) is described as impaired brain function induced by liver failure. Ammonia is the most suspected chemical involved in brain injury during HE. Although the precise mechanism of HE is not clear, several studies mentioned the role of oxidative stress in ammonia neurotoxicity. In animal models, the use of some compounds with antioxidant properties was reported to reduce the neurotoxic effects of ammonia, improve energy metabolism, and ameliorate the HE symptoms. Citicoline is a principal intermediate in the biosynthesis pathway of phosphatidylcholine that acts as neurovascular protection and repair effects. Various studies mentioned the neuroprotective and antioxidative effects of citicoline in the central nervous system. This study aims to investigate the potential protective effects of citicoline therapeutic in an animal model of HE. Materials and Methods Mice received acetaminophen (APAP,1g/kg, i. p.) and then treated with citicoline (500 mg/kg, i.p) one and two hours after APAP. Animals were monitored for locomotor activity and blood and brain ammonia levels. Moreover, markers of oxidative stress were assessed in the brain tissue. Results The result of the study revealed that plasma and brain ammonia and the liver injury markers increased, and locomotor activity impaired in the APAP-treated animals. Besides, an increase in markers of oxidative stress was evident in the brain of the APAP-treated mice. It was found that citicoline supplementation enhanced the animal’s locomotor activity and improved brain tissue markers of oxidative stress. Conclusion These data propose citicoline as a potential protective agent in HE. The effects of citicoline on oxidative stress markers could play a fundamental role in its neuroprotective properties during HE.
Collapse
Affiliation(s)
- Omid Farshad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pedram Keshavarz
- Department of Radiology, Tbilisi State Medical University (TSMU), Tbilisi, Georgia
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mina Farahmandnejad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Azhdari
- Department of Anatomy and Embryology, School of Medicine, Bam University of Medical Sciences, Bam, Iran
| | - Akram Jamshidzadeh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
26
|
Czarnecka AM, Milewski K, Albrecht J, Zielińska M. The Status of Bile Acids and Farnesoid X Receptor in Brain and Liver of Rats with Thioacetamide-Induced Acute Liver Failure. Int J Mol Sci 2020; 21:ijms21207750. [PMID: 33092050 PMCID: PMC7589210 DOI: 10.3390/ijms21207750] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/16/2020] [Accepted: 10/19/2020] [Indexed: 12/14/2022] Open
Abstract
Acute liver failure (ALF) leads to neurological symptoms defined as hepatic encephalopathy (HE). Although accumulation of ammonia and neuroinflammation are generally accepted as main contributors to HE pathomechanism, a buildup of bile acids (BA) in the blood is a frequent component of liver injury in HE patients. Recent studies have identified the nuclear farnesoid X receptor (FXR) acting via small heterodimer partner (SHP) as a mediator of BA-induced effects in the brain of ALF animals. The present study investigated the status of the BA–FXR axis in the brain and the liver, including selective changes in pertinent genes in thioacetamide (TAA)-induced ALF in Sprague–Dawley rats. FXR was found in rat neurons, confirming earlier reports for mouse and human brain. BA accumulated in blood but not in the brain tissue. Expression of mRNAs coding for Fxr and Shp was reduced in the hippocampus and of Fxr mRNA also in the cerebellum. Changes in Fxr mRNA levels were not followed by changes in FXR protein. The results leave open the possibility that mobilization of the BA–FXR axis in the brain may not be necessarily pathognomonic to HE but may depend upon ALF-related confounding factors.
Collapse
Affiliation(s)
- Anna Maria Czarnecka
- Correspondence: (A.M.C.); (M.Z.); Tel.: +48-22-6086416 (A.M.C.); +48-22-6086470 (M.Z.)
| | | | | | - Magdalena Zielińska
- Correspondence: (A.M.C.); (M.Z.); Tel.: +48-22-6086416 (A.M.C.); +48-22-6086470 (M.Z.)
| |
Collapse
|
27
|
Mohammed SAA, Khan RA, El-Readi MZ, Emwas AH, Sioud S, Poulson BG, Jaremko M, Eldeeb HM, Al-Omar MS, Mohammed HA. Suaeda vermiculata Aqueous-Ethanolic Extract-Based Mitigation of CCl 4-Induced Hepatotoxicity in Rats, and HepG-2 and HepG-2/ADR Cell-Lines-Based Cytotoxicity Evaluations. PLANTS 2020; 9:plants9101291. [PMID: 33003604 PMCID: PMC7601535 DOI: 10.3390/plants9101291] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/22/2020] [Accepted: 09/22/2020] [Indexed: 12/16/2022]
Abstract
Suaeda vermiculata, an edible halophytic plant, used by desert nomads to treat jaundice, was investigated for its hepatoprotective bioactivity and safety profile on its mother liquor aqueous-ethanolic extract. Upon LC-MS (Liquid Chromatography-Mass Spectrometry) analysis, the presence of several constituents including three major flavonoids, namely quercetin, quercetin-3-O-rutinoside, and kaempferol-O-(acetyl)-hexoside-pentoside were confirmed. The aqueous-ethanolic extract, rich in antioxidants, quenched the DPPH (1,1-diphenyl-2-picrylhydrazyl) radicals, and also showed noticeable levels of radical scavenging capacity in ABTS (2,2'-azino-bis-3-ethylbenzthiazoline-6-sulphonic acid) assay. For the hepatoprotective activity confirmation, the male rat groups were fed daily, for 7 days (n = 8/group, p.o.), either carboxyl methylcellulose (CMC) 0.5%, silymarin 200 mg/kg, the aqueous-ethanolic extract of the plant Suaeda vermiculata (100, 250, and 500 mg/kg extract), or quercetin (100 mg/kg) alone, and on day 7 of the administrations, all the animal groups, excluding a naïve (250 mg/kg aqueous-ethanolic extract-fed), and an intact animal group were induced hepatotoxicity by intraperitoneally administering carbon tetrachloride (CCl4). All the animals were sacrificed after 24 h, and aspartate transaminase and alanine transaminase serum levels were observed, which were noted to be significantly decreased for the aqueous-ethanolic extract, silymarin, and quercetin-fed groups in comparison to the CMC-fed group (p < 0.0001). No noticeable adverse effects were observed on the liver, kidney, or heart's functions of the naïve (250 mg/kg) group. The aqueous-ethanolic extract was found to be safe in the acute toxicity (5 g/kg) test and showed hepatoprotection and safety at higher doses. Further upon, the cytotoxicity testings in HepG-2 and HepG-2/ADR (Adriamycin resistant) cell-lines were also investigated, and the IC50 values were recorded at 56.19±2.55 µg/mL, and 78.40±0.32 µg/mL (p < 0.001, Relative Resistance RR 1.39), respectively, while the doxorubicin (Adriamycin) IC50 values were found to be 1.3±0.064, and 4.77±1.05 µg/mL (p < 0.001, RR 3.67), respectively. The HepG-2/ADR cell-lines when tested in a combination of the aqueous-ethanolic extract with doxorubicin, a significant reversal in the doxorubicin's IC50 value by 2.77 folds (p < 0.001, CI = 0.56) was noted as compared to the cytotoxicity test where the extract was absent. The mode of action for the reversal was determined to be synergistic in nature indicating the role of the aqueous-ethanolic extract.
Collapse
Affiliation(s)
- Salman A. A. Mohammed
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia;
- Correspondence: (S.A.A.M.); (R.A.K.); (H.A.M.); Tel.: +966-(0)530309899 (S.A.A.M.); +966-(0)508384296 (R.A.K.); +966-(0)566176074 (H.A.M.)
| | - Riaz A. Khan
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia;
- Correspondence: (S.A.A.M.); (R.A.K.); (H.A.M.); Tel.: +966-(0)530309899 (S.A.A.M.); +966-(0)508384296 (R.A.K.); +966-(0)566176074 (H.A.M.)
| | - Mahmoud Z. El-Readi
- Department of Clinical Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
- Department of Biochemistry, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Abdul-Hamid Emwas
- King Abdullah University of Science and Technology (KAUST), Core Labs, Thuwal 23955-6900, Saudi Arabia; (A.-H.E.); (S.S.)
| | - Salim Sioud
- King Abdullah University of Science and Technology (KAUST), Core Labs, Thuwal 23955-6900, Saudi Arabia; (A.-H.E.); (S.S.)
| | - Benjamin G. Poulson
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Sciences and Engineering Division (BESE), Thuwal, 23955-6900, Saudi Arabia; (B.G.P); (M.J.)
| | - Mariusz Jaremko
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Sciences and Engineering Division (BESE), Thuwal, 23955-6900, Saudi Arabia; (B.G.P); (M.J.)
| | - Hussein M. Eldeeb
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia;
- Department of Biochemistry, Faculty of Medicine, Al-Azhar University, Assiut, 71524, Egypt
| | - Mohsen S. Al-Omar
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia;
- Medicinal Chemistry and Pharmacognosy Department, Faculty of Pharmacy, JUST, Irbid 22110, Jordan
| | - Hamdoon A. Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia;
- Department of Pharmacognosy, Faculty of Pharmacy, Al-Azhar University, Cairo, 11371, Egypt
- Correspondence: (S.A.A.M.); (R.A.K.); (H.A.M.); Tel.: +966-(0)530309899 (S.A.A.M.); +966-(0)508384296 (R.A.K.); +966-(0)566176074 (H.A.M.)
| |
Collapse
|
28
|
Multidecade Mortality and a Homolog of Hepatitis C Virus in Bald Eagles (Haliaeetus leucocephalus), the National Bird of the USA. Sci Rep 2019; 9:14953. [PMID: 31628350 PMCID: PMC6802099 DOI: 10.1038/s41598-019-50580-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 09/12/2019] [Indexed: 12/21/2022] Open
Abstract
The bald eagle (Haliaeetus leucocephalus) once experienced near-extinction but has since rebounded. For decades, bald eagles near the Wisconsin River, USA, have experienced a lethal syndrome with characteristic clinical and pathological features but unknown etiology. Here, we describe a novel hepacivirus-like virus (Flaviviridae: Hepacivirus) identified during an investigation of Wisconsin River eagle syndrome (WRES). Bald eagle hepacivirus (BeHV) belongs to a divergent clade of avian viruses that share features with members of the genera Hepacivirus and Pegivirus. BeHV infected 31.9% of eagles spanning 4,254 km of the coterminous USA, with negative strand viral RNA demonstrating active replication in liver tissues. Eagles from Wisconsin were approximately 10-fold more likely to be infected than eagles from elsewhere. Eagle mitochondrial DNA sequences were homogeneous and geographically unstructured, likely reflecting a recent population bottleneck, whereas BeHV envelope gene sequences showed strong population genetic substructure and isolation by distance, suggesting localized transmission. Cophylogenetic analyses showed no congruity between eagles and their viruses, supporting horizontal rather than vertical transmission. These results expand our knowledge of the Flaviviridae, reveal a striking pattern of decoupled host/virus coevolution on a continental scale, and highlight knowledge gaps about health and conservation in even the most iconic of wildlife species.
Collapse
|