1
|
Hong X, Huang S, Jiang H, Ma Q, Qiu J, Luo Q, Cao C, Xu Y, Chen F, Chen Y, Sun C, Fu H, Liu Y, Li C, Chen F, Qiu P. Alcohol-related liver disease (ALD): current perspectives on pathogenesis, therapeutic strategies, and animal models. Front Pharmacol 2024; 15:1432480. [PMID: 39669199 PMCID: PMC11635172 DOI: 10.3389/fphar.2024.1432480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/22/2024] [Indexed: 12/14/2024] Open
Abstract
Alcohol-related liver disease (ALD) is a major cause of morbidity and mortality worldwide. It encompasses conditions such as fatty liver, alcoholic hepatitis, chronic hepatitis with liver fibrosis or cirrhosis, and hepatocellular carcinoma. Numerous recent studies have demonstrated the critical role of oxidative stress, abnormal lipid metabolism, endoplasmic reticulum stress, various forms of cell death (including apoptosis, necroptosis, and ferroptosis), intestinal microbiota dysbiosis, liver immune response, cell autophagy, and epigenetic abnormalities in the pathogenesis of ALD. Currently, abstinence, corticosteroids, and nutritional therapy are the traditional therapeutic interventions for ALD. Emerging therapies for ALD mainly include the blockade of inflammatory pathways, the promotion of liver regeneration, and the restoration of normal microbiota. Summarizing the advances in animal models of ALD will facilitate a more systematic investigation of the pathogenesis of ALD and the exploration of therapeutic targets. This review summarizes the latest insight into the pathogenesis and molecular mechanisms of ALD, as well as the pros and cons of ALD rodent models, providing a basis for further research on therapeutic strategies for ALD.
Collapse
Affiliation(s)
- Xiao Hong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuo Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - He Jiang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qing Ma
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiang Qiu
- Department of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Qihan Luo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chunlu Cao
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiyang Xu
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fuzhe Chen
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yufan Chen
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chunfeng Sun
- The First People’s Hospital of Xiaoshan District, Xiaoshan Affiliated Hospital of Wenzhou Medical University, Hangzhou, China
| | - Haozhe Fu
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiming Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Changyu Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangming Chen
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ping Qiu
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
2
|
Bao Z, Chen X, Li Y, Jiang W, Pan D, Ma L, Wu Y, Chen Y, Chen C, Wang L, Zhao S, Wang T, Lu WY, Ma C, Wang S. The hepatic GABAergic system promotes liver macrophage M2 polarization and mediates HBV replication in mice. Antiviral Res 2023; 217:105680. [PMID: 37494980 DOI: 10.1016/j.antiviral.2023.105680] [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: 02/01/2023] [Revised: 07/17/2023] [Accepted: 07/23/2023] [Indexed: 07/28/2023]
Abstract
Macrophages display functional phenotypic plasticity. Hepatitis B virus (HBV) infection induces polarizations of liver macrophages either to M1-like pro-inflammatory phenotype or to M2-like anti-inflammatory phenotype. Gamma-aminobutyric acid (GABA) signaling exists in various non-neuronal cells including hepatocytes and some immune cells. Here we report that macrophages express functional GABAergic signaling components and activation of type A GABA receptors (GABAARs) promotes M2-polarization thus advancing HBV replication. Notably, intraperitoneal injection of GABA or the GABAAR agonist muscimol increased HBV replication in HBV-carrier mice that were generated by hydrodynamical injection of adeno-associated virus/HBV1.2 plasmids (pAAV/HBV1.2). The GABA-augmented HBV replication in HBV-carrier mice was significantly reduced by the GABAAR inhibitor picrotoxin although picrotoxin had no significant effect on serum HBsAg levels in control HBV-carrier mice. Depletion of liver macrophages by liposomal clodronate treatment also significantly reduced the GABA-augmented HBV replication. Yet adoptive transfer of liver macrophages isolated from GABA-treated donor HBV-carrier mice into the liposomal clodronate-pretreated recipient HBV-carrier mice restored HBV replication. Moreover, GABA or muscimol treatment increased the expression of "M2" cytokines in macrophages, but had no direct effect on HBV replication in the HepG2.2.15 cells, HBV1.3-transfected Huh7, HepG2, or HepaRG cells, or HBV-infected Huh7-NTCP cells. Taken together, these results suggest that increasing GABA signaling in the liver promotes HBV replication in HBV-carrier mice by suppressing the immunity of liver macrophages, but not by increasing the susceptibility of hepatocytes to HBV infection. Our study shows that a previously unknown GABAergic system in liver macrophage has an essential role in HBV replication.
Collapse
Affiliation(s)
- Ziyou Bao
- Department of Immunology, Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Xiaotong Chen
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China; Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, China
| | - Yan Li
- Translational Medical Research Centre, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Wenshan Jiang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Di Pan
- Department of Immunology, Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Science, Shandong University, Jinan, China; Department of Physiology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Lushun Ma
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China; Department of Paediatric Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yunxiao Wu
- Department of Physiology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Yunling Chen
- Department of Immunology, Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Science, Shandong University, Jinan, China; Department of Physiology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Chaojia Chen
- Department of Immunology, Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Liyuan Wang
- Department of Immunology, Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Songbo Zhao
- Department of Immunology, Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Tixiao Wang
- Department of Immunology, Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Wei-Yang Lu
- Department of Physiology and Pharmacology, Robarts Research Institute, University of Western Ontario, Canada.
| | - Chunhong Ma
- Department of Immunology, Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Science, Shandong University, Jinan, China.
| | - Shuanglian Wang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
3
|
Zhang ML, Zhao X, Li WX, Wang XY, Niu M, Zhang H, Chen YL, Kong DX, Gao Y, Guo YM, Bai ZF, Zhao YL, Tang JF, Xiao XH. Yin/Yang associated differential responses to Psoralea corylifolia Linn. In rat models: an integrated metabolomics and transcriptomics study. Chin Med 2023; 18:102. [PMID: 37592331 PMCID: PMC10433582 DOI: 10.1186/s13020-023-00793-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 06/28/2023] [Indexed: 08/19/2023] Open
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Psoralea corylifolia Linn. (BGZ) is a commonly used traditional Chinese medicine (TCM) for the treatment of kidney-yang deficiency syndrome (Yangsyn) with good curative effect and security. However, BGZ was also reported to induce liver injury in recent years. According to TCM theory, taking BGZ may induce a series of adverse reactions in patients with kidney-yin deficiency syndrome (Yinsyn), which suggests that BGZ-induced liver damage may be related to its unreasonable clinical use. AIM OF THE STUDY Liver injury caused by TCM is a rare but potentially serious adverse drug reaction, and the identification of predisposed individuals for drug-induced liver injury (DILI) remains challenging. The study aimed to investigate the differential responses to BGZ in Yangsyn and Yinsyn rat models and identify the corresponding characteristic biomarkers. MATERIALS AND METHODS The corresponding animal models of Yangsyn and Yinsyn were induced by hydrocortisone and thyroxine + reserpine respectively. Body weight, organ index, serum biochemistry, and Hematoxylin and Eosin (HE) staining were used to evaluate the liver toxicity effect of BGZ on rats with Yangsyn and Yinsyn. Transcriptomics and metabonomics were used to screen the representative biomarkers (including metabolites and differentially expressed genes (DEGs)) changed by BGZ in Yangsyn and Yinsyn rats, respectively. RESULTS The level changes of liver organ index, alanine aminotransferase (ALT), and aspartate aminotransferase (AST), suggested that BGZ has liver-protective and liver-damaging effects on Yangsyn and Yinsyn rats, respectively, and the results also were confirmed by the pathological changes of liver tissue. The results showed that 102 DEGs and 27 metabolites were significantly regulated related to BGZ's protective effect on Yangsyn, which is mainly associated with the glycerophospholipid metabolism, arachidonic acid metabolism, pantothenate, and coenzyme A (CoA) biosynthesis pathways. While 28 DEGs and 31 metabolites, related to the pathway of pantothenate and CoA biosynthesis, were significantly regulated for the BGZ-induced liver injury in Yinsyn. Furthermore, 4 DEGs (aldehyde dehydrogenase 1 family member B1 (Aldh1b1), solute carrier family 25 member 25 (Slc25a25), Pim-3 proto-oncogene, serine/threonine kinase (Pim3), out at first homolog (Oaf)) and 4 metabolites (phosphatidate, phosphatidylcholine, N-Acetylleucine, biliverdin) in the Yangsyn group and 1 DEG [galectin 5 (Lgals5)] and 1 metabolite (5-amino-1-(5-phospho-D-ribosyl)imidazole-4-carboxylate) in Yinsyn group were significantly correlated to the ALT and AST levels of BGZ treated and untreated groups (receiver operating characteristic (ROC) ≥ 0.9). CONCLUSIONS Yinsyn and Yangsyn are the predisposed syndromes for BGZ to exert liver damage and liver protection respectively, which are mainly related to the regulation of amino acid metabolism, lipid metabolism, energy metabolism, and metabolism of cofactors and vitamins. The results further suggest that attention should be paid to the selection of predisposed populations when using drugs related to the regulation of energy metabolism, and the Yinsyn/Yangsyn animal models based on the theory of TCM syndromes may be a feasible method for identifying the susceptible population to receive TCM.
Collapse
Affiliation(s)
- Ming-Liang Zhang
- Henan Province Engineering Laboratory for Clinical Evaluation Technology of Chinese Medicine, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Xu Zhao
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, China
- Military Institute of Chinese Materia, the Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Wei-Xia Li
- Henan Province Engineering Laboratory for Clinical Evaluation Technology of Chinese Medicine, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Xiao-Yan Wang
- Henan Province Engineering Laboratory for Clinical Evaluation Technology of Chinese Medicine, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Ming Niu
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, China
- Military Institute of Chinese Materia, the Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Hui Zhang
- Henan Province Engineering Laboratory for Clinical Evaluation Technology of Chinese Medicine, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yu-Long Chen
- Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - De-Xin Kong
- Henan Province Engineering Laboratory for Clinical Evaluation Technology of Chinese Medicine, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yuan Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yu-Ming Guo
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, China
- Military Institute of Chinese Materia, the Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Zhao-Fang Bai
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, China
- Military Institute of Chinese Materia, the Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Yan-Ling Zhao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Department of Pharmacy, The Fifth Medical Center of PLA General Hospital, Beijing, China.
| | - Jin-Fa Tang
- Henan Province Engineering Laboratory for Clinical Evaluation Technology of Chinese Medicine, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China.
| | - Xiao-He Xiao
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, China.
| |
Collapse
|
4
|
Mahdi WA, AlGhamdi SA, Alghamdi AM, Imam SS, Alshehri S, Almaniea MA, Hajjar BM, Al-Abbasi FA, Sayyed N, Kazmi I. Effect of Europinidin against Alcohol-Induced Liver Damage in Rats by Inhibiting the TNF-α/TGF-β/IFN-γ/NF-kB/Caspase-3 Signaling Pathway. ACS OMEGA 2023; 8:22656-22664. [PMID: 37396259 PMCID: PMC10308532 DOI: 10.1021/acsomega.3c01312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/12/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND The effect of europinidin on alcoholic liver damage in rats was examined in this research. METHODS A total of 24 Wistar rats were grouped in the same way into four groups: normal control (normal), ethanol control (EtOH), europinidin low dose (10 mg/kg), and europinidin higher dose (20 mg/kg). The test group rats were orally treated with europinidin-10 and europinidin-20 for 4 weeks, whereas 5 mL/kg distilled water was administered to control rats. In addition, 1 h after the last dose of the above-mentioned oral treatment, 5 mL/kg (i.p.) EtOH was injected to induce liver injury. After 5 h of EtOH treatment, samples of blood were withdrawn for biochemical estimations. RESULTS Administration of europinidin at both doses restored all of the estimated serum, i.e., liver function tests (ALT, AST, ALP), biochemical test (Creatinine, albumin, BUN, direct bilirubin, and LDH), lipid assessment (TC and TG), endogenous antioxidants (GSH-Px, SOD, and CAT), malondialdehyde (MDA), nitric oxide (NO), cytokines (TGF-β, TNF-α, IL-1β, IL-6, IFN-γ, and IL-12), caspase-3, and nuclear factor kappa B (NF-κB) associated with the EtOH group. CONCLUSION The results of the investigation showed that europinidin had favorable effects in rats given EtOH and may have hepatoprotective potential property.
Collapse
Affiliation(s)
- Wael A. Mahdi
- Department
of Pharmaceutics, College of Pharmacy, King
Saud University, Riyadh 11451, Saudi Arabia
| | - Shareefa A. AlGhamdi
- Department
of Biochemistry, Faculty of Sciences, King
Abdulaziz University, Jeddah 21589, Saudi Arabia
- Experimental
Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Amira M. Alghamdi
- Department
of Biochemistry, Faculty of Sciences, King
Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Syed Sarim Imam
- Department
of Pharmaceutics, College of Pharmacy, King
Saud University, Riyadh 11451, Saudi Arabia
| | - Sultan Alshehri
- Department
of Pharmaceutics, College of Pharmacy, King
Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad A. Almaniea
- Department
of Pharmaceutics, College of Pharmacy, King
Saud University, Riyadh 11451, Saudi Arabia
| | - Baraa Mohammed Hajjar
- Department
of Pharmaceutics, College of Pharmacy, King
Saud University, Riyadh 11451, Saudi Arabia
| | - Fahad A. Al-Abbasi
- Department
of Biochemistry, Faculty of Sciences, King
Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Nadeem Sayyed
- School
of Pharmacy, Glocal University, Saharanpur 247121, India
| | - Imran Kazmi
- Department
of Biochemistry, Faculty of Sciences, King
Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
5
|
Rohbeck E, Hasse B, Koopmans G, Romero A, Belgardt BF, Roden M, Eckel J, Romacho T. Positive allosteric γ-aminobutyric acid type A receptor modulation prevents lipotoxicity-induced injury in hepatocytes in vitro. Diabetes Obes Metab 2022; 24:1498-1508. [PMID: 35434888 DOI: 10.1111/dom.14719] [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: 02/16/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 11/29/2022]
Abstract
AIM To determine if a novel positive allosteric modulator of the γ-aminobutyric acid type A (GABAA ) receptor, the thioacrylamide-derivative HK4, which does not penetrate the blood-brain barrier, protects human hepatocytes against lipotoxicity-induced injury. MATERIALS AND METHODS Allosteric modulation of the GABAA receptor by HK4 was determined by patch clamp in HEK-293 cells, calcium influx in INS-1E cells and by using the specific GABAA channel blockers picrotoxin and tert-butylbicyclophosphorothionate (TBPS) in HepG2 cells. Apoptosis was analysed using caspase 3/7, terminal deoxynucleotidyl transferase-dUTP nick end labelling (TUNEL) and array assays in HepG2 cells and/or human primary hepatocytes. Phosphorylation of STAT3 and the NF-κB subunit p65, protein disulphide isomerase (PDI) and poly-ADP-ribose polymerase-1 (PARP-1) was detected by Western blotting. RESULTS Patch clamping, calcium influx measurements and apoptosis assays with the non-competitive GABAA channel blockers picrotoxin and TBPS proved HK4 as a selective positive allosteric modulator of the GABAA receptor. In HepG2 cells, which expressed the main GABAA receptor subunits, HK4 prevented palmitate-induced apoptosis. This protective effect was mediated by downregulation of caspase 3/7 activity and was additionally verified by TUNEL assay. HK4 effectively prevented palmitate-induced apoptosis in human primary hepatocytes. HK4 reduced STAT3 and NF-κB phosphorylation, reduced cleaved PARP-1 expression and upregulated the endoplasmic reticulum (ER) chaperone PDI. CONCLUSIONS HK4 reduced lipotoxic-induced apoptosis by preventing inflammation, DNA damage and ER stress. We propose that the effect of HK4 is mediated by STAT3 and NF-κB. It is suggested that thioacrylamide compounds represent an innovative pharmacological tool to treat or prevent non-alcoholic steatohepatitis as first-in-class drugs.
Collapse
Affiliation(s)
- Elisabeth Rohbeck
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | | | | | - Alejandra Romero
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Bengt-Frederik Belgardt
- Institute for Vascular and Islet Cell Biology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, München-Neuherberg, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, München-Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Jürgen Eckel
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Tania Romacho
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
6
|
Busquets O, Parcerisas A, Verdaguer E, Ettcheto M, Camins A, Beas-Zarate C, Castro-Torres RD, Auladell C. c-Jun N-Terminal Kinases in Alzheimer's Disease: A Possible Target for the Modulation of the Earliest Alterations. J Alzheimers Dis 2021; 82:S127-S139. [PMID: 33216036 DOI: 10.3233/jad-201053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Given the highly multifactorial origin of Alzheimer's disease (AD) neuropathology, disentangling and orderly knowing mechanisms involved in sporadic onset are arduous. Nevertheless, when the elements involved are dissected into smaller pieces, the task becomes more accessible. This review aimed to describe the link between c-Jun N-terminal Kinases (JNKs), master regulators of many cellular functions, and the early alterations of AD: synaptic loss and dysregulation of neuronal transport. Both processes have a role in the posterior cognitive decline observed in AD. The manuscript focuses on the molecular mechanisms of glutamatergic, GABA, and cholinergic synapses altered by the presence of amyloid-β aggregates and hyperphosphorylated tau, as well as on several consequences of the disruption of cellular processes linked to neuronal transport that is controlled by the JNK-JIP (c-jun NH2-terminal kinase (JNK)-interacting proteins (JIPs) complex, including the transport of AβPP or autophagosomes.
Collapse
Affiliation(s)
- Oriol Busquets
- Department of Pharmacology, Toxicology and Therapeutic Chemistry; Pharmacy and Food Sciences Faculty, Universitat de Barcelona, Barcelona, Spain.,Department of Biochemistry and Biotechnology, Medicine and Health Sciences Faculty, Universitat Rovira i Virgili, Reus, Spain.,Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Dominick P. Purpura Department of Neurosciences, Albert Einstein College of Medicine, New York City, NY, USA
| | - Antoni Parcerisas
- Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Biology Faculty, Universitat de Barcelona, Barcelona, Spain
| | - Ester Verdaguer
- Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Biology Faculty, Universitat de Barcelona, Barcelona, Spain
| | - Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry; Pharmacy and Food Sciences Faculty, Universitat de Barcelona, Barcelona, Spain.,Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry; Pharmacy and Food Sciences Faculty, Universitat de Barcelona, Barcelona, Spain.,Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Carlos Beas-Zarate
- Department of Cell and Molecular Biology, Laboratory of Neural Regeneration, C.U.C.B.A., Universidad de Guadalajara, Jalisco, Mexico
| | - Rubén Darío Castro-Torres
- Department of Cell and Molecular Biology, Laboratory of Biology of Neurotransmission, C.U.C.B.A., Universidad de Guadalajara, Jalisco, Mexico
| | - Carme Auladell
- Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Biology Faculty, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
7
|
Inhibition of the mTORC1/NF- κB Axis Alters Amino Acid Metabolism in Human Hepatocytes. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8621464. [PMID: 33542926 PMCID: PMC7843190 DOI: 10.1155/2021/8621464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/29/2020] [Accepted: 12/22/2020] [Indexed: 12/01/2022]
Abstract
In addition to serving as the building blocks for protein synthesis, amino acids can be used as an energy source, through catabolism. The transamination, oxidative deamination, and decarboxylation processes that occur during amino acid catabolism are catalyzed by specific enzymes, including aspartate aminotransferase (AST), glutamate dehydrogenase (GDH), glutamic acid decarboxylase (GAD), and ornithine decarboxylase (ODC); however, the overall molecular mechanisms through which amino acid catabolism occurs remain largely unknown. To examine the role of mechanistic target of rapamycin complex 1 (mTORC1) on amino acid catabolism, mTORC1 was inactivated by rapamycin or shRNA targeting Raptor, versus activated by overexpressing Rheb or amino acids in human hepatocytes. The expression of amino acid catabolic genes and related transcription factor was investigated by RT/real-time PCR and western blot analysis. A few types of amino acid metabolite were examined by ELISA and HPLC analysis. The data showed that inactivated mTORC1 resulted in inhibition of NF-κB and the expression of AST, GDH, GAD, and ODC, whereas activated mTORC1 enhanced NF-κB activation and the expression levels of the catabolism-associated genes. Further, inhibition of NF-κB reduced the expression levels of AST, GDH, GAD, and ODC. mTORC1 upregulated NF-κB activation and the expression of AST and ODC in response to glutamate and ornithine treatments, whereas rapamycin inhibited the utilization of glutamate and ornithine in hepatocytes. Taken together, these results indicated that the mTORC1/NF-κB axis modulates the rate of amino acid catabolism by regulating the expression of key catabolic enzymes in hepatocytes.
Collapse
|
8
|
Wang R, Sun F, Ren C, Zhai L, Xiong R, Yang Y, Yang W, Yi R, Li C, Zhao X. Hunan insect tea polyphenols provide protection against gastric injury induced by HCl/ethanol through an antioxidant mechanism in mice. Food Funct 2020; 12:747-760. [PMID: 33367402 DOI: 10.1039/d0fo02677h] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The purposes of this study were to explore the preventive and treatment effects of Hunan insect tea polyphenols (HITPs) on gastric injury in mice induced by HCl/ethanol and to investigate their molecular mechanisms of action. Both HITPs and ranitidine inhibited the formation and further deterioration of gastric mucosal lesions, reduced the secretion of gastric juice, and raised gastric juice pH compared to the control. The HITPs-H treated group had lower serum levels of motilin, substance P, and endothelin than the control group, but they had higher serum levels of vasoactive intestinal peptide and somatostatin. Mice treated with HITPs had lower serum levels of cytokines interleukin (IL)-6, IL-12, tumor necrosis factor-α (TNF-α), and interferon-γ than the control group. The activities of superoxide dismutase (SOD), nitric oxide, and glutathione peroxidase (GSH-Px) were higher in the gastric tissues of HITP-treated mice, but the malondialdehyde content was lower. Quantitative PCR analysis indicated that the mRNA expression of occludin, epidermal growth factor (EGF), EGF receptor (EGFR), vascular EGF (VEGF), inhibitor kappaB-α, cuprozinc-superoxide dismutase, manganese-superoxide dismutase, GSH-Px, neuronal nitric oxide synthase, and endothelial NOS increased significantly in the gastric tissues of HITP-treated mice. However, the activated B cell, inducible NOS, cyclooxygenase-2, TNF-α, IL-1 beta, and IL-6 mRNA expression levels in the HITPs group were lower than those in the control group. The protective effect of a high concentration (200 mg per kg bw) of HITPs on gastric injury induced by HCl/ethanol was stronger than that of a low concentration (100 mg per kg bw) of HITPs. High-performance liquid chromatography (HPLC) revealed that the HITPs contained cryptochlorogenic acid, (-)-epicatechin gallate, and isochlorogenic acid C. Taken together, our findings indicate that the HITPs played a role in the prevention of gastric damage. The antioxidant effect of the HITPs contributed to their potential value in the prevention and treatment of gastric injury. HITPs have broad prospects as biologically active substances for food development.
Collapse
Affiliation(s)
- Ranran Wang
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Siregar AS, Nyiramana MM, Kim EJ, Shin EJ, Woo MS, Kim JM, Kim JH, Lee DK, Hahm JR, Kim HJ, Kim CW, Kim NG, Park SH, Choi YJ, Kang SS, Hong SG, Han J, Kang D. Dipeptide YA is Responsible for the Positive Effect of Oyster Hydrolysates on Alcohol Metabolism in Single Ethanol Binge Rodent Models. Mar Drugs 2020; 18:md18100512. [PMID: 33050644 PMCID: PMC7601867 DOI: 10.3390/md18100512] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 12/15/2022] Open
Abstract
Accumulative alcohol hangovers cause liver damage through oxidative and inflammatory stress. Numerous antioxidant and anti-inflammatory reagents have been developed to reduce alcohol hangovers, but these reagents are still insignificant and have limitations in that they can cause liver toxicity. Oyster hydrolysate (OH), another reagent that has antioxidant and anti-inflammatory activity, is a product extracted through an enzymatic hydrolysis process from oysters (Crassostrea gigas), which can be easily eaten in meals. This study was aimed at determining the effects of OH on alcohol metabolism, using a single high dose of ethanol (EtOH) administered to rodents, by monitoring alcohol metabolic enzymes, oxidative stress signals, and inflammatory mediators. The effect of tyrosine-alanine (YA) peptide, a main component of OH, on EtOH metabolism was also identified. In vitro experiments showed that OH pretreatment inhibited EtOH-induced cell death, oxidative stress, and inflammation in liver cells and macrophages. In vivo experiments showed that OH and YA pre-administration increased alcohol dehydrogenase, aldehyde dehydrogenase, and catalase activity in EtOH binge treatment. In addition, OH pre-administration alleviated CYP2E1 activity, ROS production, apoptotic signals, and inflammatory mediators in liver tissues. These results showed that OH and YA enhanced EtOH metabolism and had a protective effect against acute alcohol liver damage. Our findings offer new insights into a single high dose of EtOH drinking and suggest that OH and YA could be used as potential marine functional foods to prevent acute alcohol-induced liver damage.
Collapse
Affiliation(s)
- Adrian S. Siregar
- Department of Physiology and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (A.S.S.); (M.M.N.); (E.-J.K.); (E.-J.S.); (M.S.W.); (D.K.L.); (S.-G.H.); (J.H.)
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea; (H.J.K.); (S.S.K.)
| | - Marie Merci Nyiramana
- Department of Physiology and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (A.S.S.); (M.M.N.); (E.-J.K.); (E.-J.S.); (M.S.W.); (D.K.L.); (S.-G.H.); (J.H.)
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea; (H.J.K.); (S.S.K.)
| | - Eun-Jin Kim
- Department of Physiology and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (A.S.S.); (M.M.N.); (E.-J.K.); (E.-J.S.); (M.S.W.); (D.K.L.); (S.-G.H.); (J.H.)
| | - Eui-Jung Shin
- Department of Physiology and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (A.S.S.); (M.M.N.); (E.-J.K.); (E.-J.S.); (M.S.W.); (D.K.L.); (S.-G.H.); (J.H.)
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea; (H.J.K.); (S.S.K.)
| | - Min Seok Woo
- Department of Physiology and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (A.S.S.); (M.M.N.); (E.-J.K.); (E.-J.S.); (M.S.W.); (D.K.L.); (S.-G.H.); (J.H.)
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea; (H.J.K.); (S.S.K.)
| | - Jin-Mok Kim
- Department of Clinical Laboratory Science, Masan University, Changwon 2640, Korea;
| | - Jung Hwan Kim
- Department of Premedicine, College of Medicine, Gyeongsang National University, Jinju 52727, Korea;
| | - Dong Kun Lee
- Department of Physiology and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (A.S.S.); (M.M.N.); (E.-J.K.); (E.-J.S.); (M.S.W.); (D.K.L.); (S.-G.H.); (J.H.)
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea; (H.J.K.); (S.S.K.)
| | - Jong Ryeal Hahm
- Department of Internal Medicine, Hospital and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea;
| | - Hyun Joon Kim
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea; (H.J.K.); (S.S.K.)
- Department of Anatomy and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea
| | - Chang-Woon Kim
- Department of Obstetrics and Gynecology, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon 51353, Korea;
| | - Nam-Gil Kim
- Department of Marine Biology and Aquaculture and Institute of Marine Industry, Gyeongsang National University, Tongyeong 53064, Korea;
| | - Si-Hyang Park
- Sunmarin Biotech, Jinju Bioindustry Foundation, Jinju 52839, Korea;
| | - Yeung Joon Choi
- Ocean-Pep, Jinju Bioindustry Foundation, Jinju 52839, Korea;
| | - Sang Soo Kang
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea; (H.J.K.); (S.S.K.)
- Department of Anatomy and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea
| | - Seong-Geun Hong
- Department of Physiology and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (A.S.S.); (M.M.N.); (E.-J.K.); (E.-J.S.); (M.S.W.); (D.K.L.); (S.-G.H.); (J.H.)
| | - Jaehee Han
- Department of Physiology and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (A.S.S.); (M.M.N.); (E.-J.K.); (E.-J.S.); (M.S.W.); (D.K.L.); (S.-G.H.); (J.H.)
| | - Dawon Kang
- Department of Physiology and Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (A.S.S.); (M.M.N.); (E.-J.K.); (E.-J.S.); (M.S.W.); (D.K.L.); (S.-G.H.); (J.H.)
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea; (H.J.K.); (S.S.K.)
- Correspondence:
| |
Collapse
|
10
|
Chemical Components and Hepatoprotective Mechanism of Xwak Granule in Mice Treated with Acute Alcohol. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:8538474. [PMID: 33062026 PMCID: PMC7539125 DOI: 10.1155/2020/8538474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 07/24/2020] [Accepted: 08/03/2020] [Indexed: 11/17/2022]
Abstract
Objective To evaluate the hepatoprotective mechanism of Xwak granule (Xwak) in treatment of mice with alcoholic liver injury via activating ERK/NF-κB and Nrf/HO-1 signaling pathways. Methods The chemical composition of Xwak was tested by liquid chromatography coupled with mass spectrometry (LC-MS). Herein, 1,1-diphenyl-2-picrylhydrazyl (DPPH) scavenging assay and 2,2-azino-bis (3-ethylbenzothiazoline-6-sulphonic acid (ABTS) radical tests were performed in vitro. The hepatoprotective effect of Xwak was assessed at different concentrations (1.5, 3, and 6 g/kg) in a mouse model of alcoholic liver injury. Results Totally, 48 compounds, including 16 flavonoids, 8 tannins, 9 chlorogenic acids, and 15 other compounds, were identified from Xwak. Xwak showed to have a satisfactory antioxidant activity in vitro. In a group of Xwak-treated mice, the serum levels of alanine transaminase (ALT), aspartate transaminase (AST), and alkaline phosphatase (ALP) were decreased compared with a group of the mouse model of alcoholic liver injury. In addition, the levels of antioxidant enzymes, such as glutathione peroxidase (GSH-PX), total superoxide dismutase (T-SOD), and catalase (CAT), were noticeably increased and the levels of malondialdehyde (MDA), tumor necrosis factor-α (TNF-α), transforming growth factor-β (TGF-β), and interleukin-6 (IL-6) were markedly reduced in the liver of mice. The state of oxidative stress in the mouse model of alcoholic liver injury was improved after treatment with Xwak. The improvement of inflammation-mediated disruption may conducive to the Xwak activity in the control of liver injury. The signals of p-ERK1/2, p-NF-κB, COX-2, iNOS, CYP2E1, Nrf, and HO-1 were significantly induced in the liver of mice after treatment with Xwak. Conclusions The abovementioned findings indicated that the hepatoprotective mechanism of Xwak could be achieved by activating ERK/NF-κB and Nrf/HO-1 signaling pathways to alleviate oxidative stress and inflammatory.
Collapse
|
11
|
Wang R, Zeng X, Liu B, Yi R, Zhou X, Mu J, Zhao X. Prophylactic effect of Lactobacillus plantarum KSFY06 on HCl/ethanol-induced gastric injury in mice. Food Funct 2020; 11:2679-2692. [PMID: 32162630 DOI: 10.1039/c9fo02474c] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
The present study was conducted to determine the prophylactic effect of Lactobacillus plantarum KSFY06 (LP-KSFY06) on HCl/ethanol-induced gastric injury in Kunming mice. The experimental mice were allocated into six groups: the normal group, HCl/ethanol treated group, HCl/ethanol + ranitidine treated group, HCl/ethanol + Lactobacillus delbrueckii subsp. Bulgaricus (LB) treated group, HCl/ethanol + low concentration of Lactobacillus plantans KSFY06 (LP-KSFY06-L) treated group, and HCl/ethanol + high concentration of Lactobacillus plantans KSFY06 (LP-KSFY06-H) treated group. The changes in daily body weight and food intake of the mice in the HCl/ethanol + LP-KSFY06-H treated group were the closest to those of the HCl/ethanol + ranitidine treated and normal groups. LP-KSFY06 significantly inhibited the formation of gastric mucosal lesions, reduced the area of gastric lesions, inhibited gastric-juice secretion, and increased pH compared with the HCl/ethanol treated group. After the treatment, the serum interleukin-6 (IL)-6, IL-12, tumor necrosis factor-α (TNF-α), and interferon-γ levels and the gastric-tissue IL-6 and IL-12 levels in the LP-KSFY06 (including LP-KSFY06-L and LP-KSFY06-H) group decreased compared with those in the HCl/ethanol treated group. The level of serum and gastric tissue malondialdehyde was lower and the nitric oxide, total superoxide dismutase, and glutathione activities in the LP-KSFY06 treated mice were higher than those in the HCl/ethanol treated mice. Quantitative polymerase chain reaction analysis and western blot analysis showed that LP-KSFY06 increased the mRNA and protein expression of the epidermal growth factor, epidermal growth factor receptor, vascular endothelial growth factor, inhibitor kappaB-α, neuronal nitric oxide synthase, and endothelial NOS and reduced the mRNA and protein expression of nuclear factor kappaB, inducible NOS, cyclooxygenase-2, TNF-α, and IL-1β in gastric tissues compared with the HCl/ethanol treated mice. These experimental results showed that a high concentration (1.0 × 109 CFU per kg B.W.) of LP-KSFY06 had a stronger effect on preventing gastric injury than a low concentration (1.0 × 108 CFU per kg B.W.) of LP-KSFY06. These results suggest that LP-KSFY06 has a potential probiotic effect in preventing gastric injury.
Collapse
Affiliation(s)
- Ranran Wang
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China. and Chongqing Engineering Research Center of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and College of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, P.R. China
| | - Xiaofei Zeng
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, P.R. China
| | - Bihui Liu
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China. and Chongqing Engineering Research Center of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and College of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, P.R. China
| | - Ruokun Yi
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China. and Chongqing Engineering Research Center of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and College of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, P.R. China
| | - Xianrong Zhou
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China. and Chongqing Engineering Research Center of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China
| | - Jianfei Mu
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China. and Chongqing Engineering Research Center of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China
| | - Xin Zhao
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China. and Chongqing Engineering Research Center of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing 400067, P.R. China and College of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, P.R. China
| |
Collapse
|
12
|
Park NH, Lee SJ, Mechesso AF, Boby N, Yixian Q, Yoon WK, Lee SP, Lee JS, Park SC. Hepatoprotective effects of gamma-aminobutyric acid-enriched fermented Hovenia dulcis extract on ethanol-induced liver injury in mice. BMC Complement Med Ther 2020; 20:75. [PMID: 32143613 PMCID: PMC7076742 DOI: 10.1186/s12906-020-2866-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 02/26/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Various extracts of Hovenia dulcis have been commonly used in Asia for cases of alcohol-related disorders. Fermentation is reported to enhance the level and biological activities of various bio-constituents of plant extracts. Therefore, this study was undertaken to evaluate the effects of fermented H. dulcis extract (FHDE) on ethanol-induced liver injury in mice. METHODS FHDE was prepared using Bacillus subtilis and Lactobacillus plantarum. The effects of FHDE on ethanol-induced liver injury were evaluated in C57BL/6 N CrSlc mice. A mixed feed preparation containing the fermented extract with and without ethanol was given to mice for 29 days, according to its group. At the end of the experiment, blood and liver samples were collected from all mice in the group. Plasma biochemical analysis and histopathological investigation were performed to evaluate the impacts of treatment on the biomarkers of hepatic damage and inflammatory changes. Besides, the expression of genes that regulate the activities of enzymes associated with alcohol metabolism, antioxidant activity, and fatty acid oxidation was assessed using a quantitative real-time polymerase chain reaction. Moreover, the amino acid contents and the active ingredients of the extract were evaluated before and after fermentation. RESULTS Fermentation resulted in a marked increase and decrease in the amount of Gamma-Amino-n-butyric acid (GABA) and glutamic acid, respectively. FHDE enhanced the body weight gain of mice compared to ethanol. Besides, plasma levels of triglyceride, low-density lipoprotein, the activities of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) were significantly (P < 0.05) reduced in the FHDE-treated groups relative to the ethanol-treated control. FHDE upregulated the expression of genes associated with enzymes involved in alcohol dehydrogenation (Adh1 and Aldh2), antioxidant activity (SOD and CAT), and fatty acid oxidation (PPAR-α and PGC-1α). However, the expressions of Cytochrome peroxidase Cyp2E1 and genes related to lipogenesis (SREBP-1c, FAS, SCD-1, and ACC) were significantly (P < 0.05) downregulated following treatment with the FHDE. Histopathological investigation demonstrated a slight degree of inflammatory cell infiltration and occasional fatty changes in the FHDE-treated groups. CONCLUSION The GABA-enriched fermented H. dulcis extract prevented ethanol-induced hepatic damage by enhancing the antioxidant defense system, fatty acid oxidation, and reducing lipogenesis.
Collapse
Affiliation(s)
- Na-Hye Park
- College of Veterinary Medicine, Kyungpook National University, 80, Daehak-ro, Buk-gu, 41566 Daegu, Republic of Korea
| | - Seung-Jin Lee
- College of Veterinary Medicine, Kyungpook National University, 80, Daehak-ro, Buk-gu, 41566 Daegu, Republic of Korea
| | - Abraham Fikru Mechesso
- College of Veterinary Medicine, Kyungpook National University, 80, Daehak-ro, Buk-gu, 41566 Daegu, Republic of Korea
| | - Naila Boby
- College of Veterinary Medicine, Kyungpook National University, 80, Daehak-ro, Buk-gu, 41566 Daegu, Republic of Korea
| | - Quah Yixian
- College of Veterinary Medicine, Kyungpook National University, 80, Daehak-ro, Buk-gu, 41566 Daegu, Republic of Korea
| | - Woong-Kyu Yoon
- Department of Food Science and Technology, Keimyung University, Daegu, 42601 Republic of Korea
| | - Sam-Pin Lee
- College of Veterinary Medicine, Kyungpook National University, 80, Daehak-ro, Buk-gu, 41566 Daegu, Republic of Korea
| | - Jong-Suk Lee
- Biocenter, Gyeonggido Business and Science Accelerator (GBSA), Suwon, Gyeonggi-do 16229 Republic of Korea
| | - Seung-Chun Park
- College of Veterinary Medicine, Kyungpook National University, 80, Daehak-ro, Buk-gu, 41566 Daegu, Republic of Korea
| |
Collapse
|