1
|
Wang Y, Ma Y, Tuo P, Naeem A, Tang Y, Su Q, Li H, Gao X, Wang X. Forsythoside A Alleviates Acute Alcoholic Liver Injury by Binding to TLR4 to Inhibit the Activation of the NF-κB Pathway. Phytother Res 2025. [PMID: 40342240 DOI: 10.1002/ptr.8485] [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: 12/04/2023] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 05/11/2025]
Abstract
Forsythoside A (FTA) is a key component found in the fruit and leaves of Forsythia suspensa, having anti-inflammatory and antioxidant properties. However, it is unclear whether FTA can have a protective effect against acute alcoholic liver injury (ALI) and how it may exert this effect. This research examined the potential protective effects of FTA against acute ALI using cell and animal models. The protective properties of FTA against acute ALI were attributed to its anti-inflammatory and antioxidant actions by detecting the markers of oxidative stress and inflammation. The underlying mechanism was explored through the utilization of Western blotting, Molecular Docking, and Microscale Thermophoresis techniques. The results showed that pretreatment with high doses of FTA had a significant protective effect on acute ALI in both cell and animal models. The pretreatment with high doses of FTA inhibited alcohol-induced oxidative stress and inflammation, raising antioxidative enzyme activity in both models. Furthermore, FTA has been shown to bind to TLR4, thereby inhibiting alcohol-induced activation of the NF-κB signaling pathway, leading to a decrease in cellular oxidative stress and inflammatory reactions. This interaction also facilitates the ubiquitination-mediated degradation of TLR4, ultimately diminishing its regulatory impact on the NF-κB signaling cascade. FTA has a significant protective effect on acute ALI. It binds to TLR4 to inhibit the activation of the NF-κB signaling pathway by alcohol, thereby reducing oxidative stress and inflammation and exerting a protective effect. The results of our study provide a theoretical basis for the development of FTA for the prevention and treatment of acute ALI.
Collapse
Affiliation(s)
- Yuehua Wang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Shiyan, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, College of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yuying Ma
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Shiyan, China
| | - Peng Tuo
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Shiyan, China
| | - Abid Naeem
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Yijun Tang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Shiyan, China
| | - Qianying Su
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Shiyan, China
| | - Huajian Li
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Shiyan, China
| | - Xiaokang Gao
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Shiyan, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, College of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Xiaoli Wang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Shiyan, China
| |
Collapse
|
2
|
Garcia-Moruno E, Blázquez AB, Jiménez de Oya N, Esteban A, Motta S, Costantini A, Sánchez-Cordón PJ, Pericuesta E, Martín-Acebes MA, Gutiérrez-Adán A, Saiz JC. Effect of long-term moderate red wine consumption in CD1 female mice. Food Funct 2025; 16:2656-2668. [PMID: 40025823 DOI: 10.1039/d5fo00435g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
In the early 80s, the so-called "French paradox" was proposed, that is, a correlation between wine consumption, a diet rich in fats, and low mortality from coronary disease. Conversely, it is well established that excessive alcohol consumption increases the risk of cirrhosis and cancer, but few studies have investigated the effects of moderate alcohol consumption. However, all these conclusions were derived from epidemiological population studies that may be subject to distortions due to multiple factors. Here, the effect of moderate consumption of red wine on health throughout life was examined in a murine model. Different variables were evaluated in groups of female animals that were fed a standard or a fat diet throughout their adult life and given water, or wine or alcohol diluted in water in proportions similar to what is considered moderate consumption in humans. Our results showed few differences in most of the analyzed variables (body weight, liver profile and survival rate) between the different female mouse groups. The most remarkable findings were observed in the fat-diet groups that showed more frequent and severe liver lesions and a lower average ovarian weight. Moreover, moderate and prolonged ethanol consumption significantly affected telomere length only when the diet was high in fat, whereas wine consumption showed no difference compared to water, pointing to a possible predominant role of the compounds, particularly polyphenols present in wine. On the other hand, wine-drinking mice fed a fat diet had more oocytes than those in the ethanol-drinking group. Overall, our data suggest that long-term moderate red wine consumption does not substantially influence the health of female mice.
Collapse
Affiliation(s)
- Emilia Garcia-Moruno
- CREA-Centro di Ricerca Viticoltura ed Enologia (CREA-VE), Via Pietro Micca 35, 14100 Asti, Italy.
| | - Ana-Belén Blázquez
- Dpt. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Ctra. Coruña Km. 7.5, Madrid, Spain.
| | - Nereida Jiménez de Oya
- Dpt. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Ctra. Coruña Km. 7.5, Madrid, Spain.
| | - Ana Esteban
- Dpt. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Ctra. Coruña Km. 7.5, Madrid, Spain.
| | - Silvia Motta
- CREA-Centro di Ricerca Viticoltura ed Enologia (CREA-VE), Via Pietro Micca 35, 14100 Asti, Italy.
| | - Antonella Costantini
- CREA-Centro di Ricerca Viticoltura ed Enologia (CREA-VE), Via Pietro Micca 35, 14100 Asti, Italy.
| | - Pedro J Sánchez-Cordón
- Unidad de Patología Veterinaria, Centro de Investigación en Sanidad Animal, CISA, (INIA-CSIC), 28130 Madrid, Spain
| | - Eva Pericuesta
- Dpt. Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Ctra. Coruña Km. 7.5, 28760 Madrid, Spain
| | - Miguel A Martín-Acebes
- Dpt. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Ctra. Coruña Km. 7.5, Madrid, Spain.
| | - Alfonso Gutiérrez-Adán
- Dpt. Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Ctra. Coruña Km. 7.5, 28760 Madrid, Spain
| | - Juan-Carlos Saiz
- Dpt. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Ctra. Coruña Km. 7.5, Madrid, Spain.
| |
Collapse
|
3
|
Ma S, Kang L, Gao Z, Pan Z, Huang L, Chen J, Liao Y, Guo J, Fu F. Hepatoprotective Effects of Citri reticulatae Pericarpium and Chaenomelese speciosa (Sweet) Nakai Extracts in Alcohol-Related Liver Injury: Modulation of Oxidative Stress, Lipid Metabolism, and Gut Microbiota. Antioxidants (Basel) 2025; 14:343. [PMID: 40227436 PMCID: PMC11939523 DOI: 10.3390/antiox14030343] [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: 02/08/2025] [Revised: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
Chronic and excessive alcohol consumption induces alcohol-related liver injury (ALI), characterized by oxidative stress (OS), disrupted lipid metabolism, and gut microbiota dysbiosis. Given the lack of effective pharmacological treatments, flavonoid-rich fruits have attracted growing attention as potential intervention strategies. This study investigated the independent and combined effects of extracts from Citri reticulatae pericarpium (CRPE) and Chaenomeles speciosa (Sweet) Nakai (CSPE), previously shown to possess hepatoprotective properties, in a mouse model of ethanol-induced chronic ALI. The flavonoid composition of CRPE and CSPE was characterized using LC-MS/MS, and their potential mechanisms of action were further elucidated through transcriptomic analysis. The results showed that CRPE and CSPE, whether administered individually or in combination, effectively alleviated alcohol-induced hepatic histological damage and inflammatory responses. Furthermore, both extracts significantly reduced OS and improved lipid metabolism. Notably, CRPE, CSPE, and their combination regulated the gut microbiota, as shown by increased abundances of beneficial bacteria such as Lactobacillus and Bifidobacterium, along with elevated levels of short-chain fatty acids (SCFAs). These findings highlight that combinations of multiple fruit extracts exhibit significant potential in alleviating ALI by modulating the gut microbiota, providing valuable insights for the development of functional foods.
Collapse
Affiliation(s)
- Shuangshuang Ma
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; (S.M.); (L.K.); (Y.L.)
- Dongting Laboratory, Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, China; (Z.P.); (L.H.); (J.C.)
| | - Lingtao Kang
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; (S.M.); (L.K.); (Y.L.)
- Dongting Laboratory, Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, China; (Z.P.); (L.H.); (J.C.)
| | - Zhipeng Gao
- College of Fisheries, Hunan Agricultural University, Changsha 410128, China;
| | - Zhaoping Pan
- Dongting Laboratory, Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, China; (Z.P.); (L.H.); (J.C.)
| | - Lvhong Huang
- Dongting Laboratory, Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, China; (Z.P.); (L.H.); (J.C.)
| | - Jiaxu Chen
- Dongting Laboratory, Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, China; (Z.P.); (L.H.); (J.C.)
| | - Yanfang Liao
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; (S.M.); (L.K.); (Y.L.)
- Dongting Laboratory, Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, China; (Z.P.); (L.H.); (J.C.)
| | - Jiajing Guo
- Dongting Laboratory, Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, China; (Z.P.); (L.H.); (J.C.)
| | - Fuhua Fu
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; (S.M.); (L.K.); (Y.L.)
- Dongting Laboratory, Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, China; (Z.P.); (L.H.); (J.C.)
| |
Collapse
|
4
|
Zyoud SH. Global scientific research landscape on binge drinking: a comprehensive bibliometric and visualization analysis of trends, collaborations, and future directions. Subst Abuse Treat Prev Policy 2025; 20:13. [PMID: 40065367 PMCID: PMC11895325 DOI: 10.1186/s13011-025-00641-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Binge drinking constitutes a significant public health concern. Defined as the consumption of five or more alcoholic beverages on a single occasion, binge drinking leads to acute cognitive and motor impairments and is associated with a multitude of detrimental health consequences. Therefore, the aim of this study was to analyse globally published peer-reviewed literature on binge drinking. METHODS A thorough search of the Scopus database was conducted to gather all the relevant research. Keywords related to binge drinking were used to locate a wide range of studies. Specific criteria were subsequently applied to narrow the results, ensuring the inclusion of only the most relevant articles. This process yielded a collection of 2,763 research papers. Finally, a software program called VOSviewer was utilized to analyse and visualize the connections between these studies. RESULTS A bibliometric analysis was performed to investigate trends in binge drinking research literature published between 1980 and 2024. The findings revealed a significant increase in publications (R²=0.916; p < 0.001), with a peak in 2018 (191 articles). The majority (89.65%, n = 2,477) were research articles, followed by review articles (4.74%, n = 131). Authors from 139 countries contributed to binge-drinking research, with the USA (n = 1,550; 56.1%) and the UK (n = 216; 7.82%) leading in the volume of publications. The National Institute on Alcohol Abuse and Alcoholism (n = 65; 2.35%) and the University of North Carolina at Chapel Hill (n = 63; 2.28%) emerged as the main institutional contributors. The National Institute on Alcohol Abuse and Alcoholism in the United States was the main funding source, supporting 599 articles (21.68%), followed by the National Institutes of Health in the United States, with 544 articles (19.69%). In particular, the post-2016 period witnessed a shift in research themes toward mechanistic investigations alongside studies on societal interventions, reflecting a growing focus on mitigating the broader social impact of binge drinking. CONCLUSIONS This study is the first comprehensive analysis of trends in binge drinking research. Over the past decade, binge drinking has increased dramatically, led by the United States, the UK, and Spain. Initially, focused on social and cultural factors, research shifted after 2016 to mechanistic and animal models, shaping future research directions and strategies.
Collapse
Affiliation(s)
- Sa'ed H Zyoud
- Poison Control and Drug Information Center (PCDIC), An-Najah National University, Nablus, 44839, Palestine.
- Department of Clinical and Community Pharmacy, College of Medicine and Health Sciences, An-Najah National University, Nablus, 44839, Palestine.
- Clinical Research Centre, An-Najah National University Hospital, Nablus, 44839, Palestine.
| |
Collapse
|
5
|
Kang J, Park SH, Khanam M, Park SB, Shin S, Seo W. Impact of binge drinking on alcoholic liver disease. Arch Pharm Res 2025; 48:212-223. [PMID: 40035998 DOI: 10.1007/s12272-025-01537-1] [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: 05/30/2024] [Accepted: 02/13/2025] [Indexed: 03/06/2025]
Abstract
Numerous studies have examined the pathophysiological changes induced by chronic alcohol (ethanol) consumption and the underlying mechanisms, while much less attention has been devoted to understanding the health impacts of binge drinking. Binge drinking is defined as the excessive consumption of alcohol within a single drinking episode, and is the typical consumption pattern among young people in Western countries. While most young binge drinkers are not clinically alcohol dependent, binge drinking has emerged as a significant social and public health concern. The circulating alcohol consumed during binge episodes permeates cellular membranes throughout the body, exerting profound effects on multiple organs, and signaling pathways. Regular binge drinking eventually induces hepatic steatosis (fatty liver), initiates acute inflammation, and accelerates neutrophil infiltration, de novo lipogenesis, adipocyte death/lipolysis, and the production of nonoxidative alcohol metabolites, processes that synergize to damage liver tissue and impair liver function. Metabolic abnormalities such as diabetes and obesity can also exacerbate the progression of alcohol-related liver disease among binge drinkers. Several animal models have been developed to evaluate the pathophysiological changes resulting from binge drinking; however, the pathogenesis of binge drinking is not fully understood due to differences in alcohol metabolism between animal models and humans. Thus, given the high prevalence and severe health implications of binge drinking, there is an urgent need for comprehensive experimental and clinical investigations to unravel the associated pathophysiological changes. This review summarizes recent research findings on the impact of binge drinking, specifically focusing on its contributions to alcoholic liver injury.
Collapse
Affiliation(s)
- Jisoo Kang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Seol Hee Park
- Department of Companion Animal Health, Hanyang Women's University, Seoul, 04763, Republic of Korea
| | - Mushira Khanam
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Seo Bhin Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Sumin Shin
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Wonhyo Seo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea.
- Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, 03760, Republic of Korea.
| |
Collapse
|
6
|
Wu Q, Yang D, Liu C, Xu T. Alcohol Plus Additional Risk Factors: Rodent Model of Liver Injury. Semin Liver Dis 2025; 45:81-98. [PMID: 39719149 DOI: 10.1055/a-2490-4278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
Alcohol-associated liver disease (ALD), primarily caused by chronic excessive alcohol consumption, is a leading cause of chronic liver disease worldwide. ALD includes alcohol-associated steatotic liver, alcohol-associated hepatitis (AH), fibrosis, cirrhosis, and can even progress to hepatocellular carcinoma (HCC). Existing research indicates that the risk factors of ALD are quite numerous. In addition to drinking patterns, factors such as aldehyde dehydrogenase 2 (ALDH2) deficiency, smoking, medication administration, high-fat diet (HFD), hepatitis virus infection, and disruption of circadian rhythms can also increase susceptibility to ALD. However, there is limited understanding regarding the exacerbation of liver injury by alcohol plus additional risk factors. This review presents rodent models of EtOH + "X," which simulate the synergistic effects of alcohol and additional risk factors in causing liver injury. These models offer a further exploration of the interactions between alcohol and additional risk factors, advancing the simulation of human ALD and providing a more reliable platform for studying disease mechanisms and exploring therapeutic interventions. We summarize the modeling methods, relevant indicators of liver injury, and focus on the targets of the synergistic effects as well as the associated mechanisms.
Collapse
Affiliation(s)
- Qixiang Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, P.R. China
- Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Dashuai Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, P.R. China
- Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Chixiang Liu
- Department of Blood Transfusion, Southern Medical University, Nanfang Hospital, Guangzhou, P.R. China
- School of Laboratory and Biotechnology, Institute of Antibody Engineering, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, P.R. China
- Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, P.R. China
| |
Collapse
|
7
|
Brigagão Pacheco da Silva C, Nascimento-Silva EA, Zaramela LS, da Costa BRB, Rodrigues VF, De Martinis BS, Carlos D, Tostes RC. Drinking pattern and sex modulate the impact of ethanol consumption on the mouse gut microbiome. Physiol Genomics 2025; 57:179-194. [PMID: 39918827 DOI: 10.1152/physiolgenomics.00031.2024] [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: 03/25/2024] [Revised: 04/17/2024] [Accepted: 01/28/2025] [Indexed: 03/04/2025] Open
Abstract
Gut microbiota impacts host homeostasis and diseases. Chronic plus binge ethanol consumption has been linked to increased injuries than chronic or binge ethanol intake alone. We hypothesized that distinct shapes in gut microbiota composition are induced by chronic, binge, and the association of these treatments, thereby affecting host functions and contributing to sex-based differences in alcohol use disorders. Male and female C57BL/6J mice were submitted to chronic, binge, or chronic plus binge ethanol feeding. DNA was extracted from fecal microbiota, followed by analysis of the V3-V4 region of the 16S rRNA gene and sequencing on an Illumina platform. Gut microbiome analysis was performed using QIIME v2022.2.0. Functional profiling of the gut microbiome was performed using PICRUSt2. Ethanol differentially affected the gut microbiota of female and male mice. Decreased α diversity was observed in male and female mice from the chronic plus binge and chronic groups, respectively. The genera Faecalibaculum, Lachnospiraceae, and Alistipes were identified as major potential biomarkers for gut dysbiosis induced by ethanol consumption. In addition, ethanol-induced gut dysbiosis altered several metabolic pathways. Ethanol consumption modifies the mouse gut microbiome in a drinking pattern- and sex-dependent manner, potentially leading to different susceptibility to ethanol-related diseases. Chronic plus binge ethanol intake induces a more pronounced gut dysbiosis in male mice. Conversely, chronic ethanol is linked to a greater degree of gut dysbiosis in female mice. The changed gut microbiome may be potentially targeted to prevent, mitigate, or treat alcohol use disorders.NEW & NOTEWORTHY Ethanol alters the mouse gut microbiome in a drinking pattern- and sex-dependent manner. Chronic plus binge ethanol intake induces a more severe gut dysbiosis in male mice, whereas chronic ethanol consumption appears to be a more potent inductor of gut dysbiosis in female mice. Ethanol-induced gut dysbiosis alters several pathways linked to metabolism, genetic and environmental information processing, cellular processes, organism systems, and neurological human diseases.
Collapse
Affiliation(s)
| | | | - Lívia Soares Zaramela
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Bruno Ruiz Brandão da Costa
- Department of Clinical, Toxicological and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Vanessa Fernandes Rodrigues
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Bruno Spinosa De Martinis
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Daniela Carlos
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rita C Tostes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
8
|
Schonfeld M, Nataraj K, Weinman S, Tikhanovich I. C/EBPβ transcription factor promotes alcohol-induced liver fibrosis in males via HDL remodeling. Hepatol Commun 2025; 9:e0645. [PMID: 39969482 PMCID: PMC11841851 DOI: 10.1097/hc9.0000000000000645] [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: 10/25/2024] [Accepted: 12/16/2024] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Alcohol-associated liver disease (ALD) is the main cause of alcohol-associated mortality. However, the mechanism of ALD development is poorly understood. Epigenetic changes are thought to play an important role in ALD. We aimed to define the epigenetic changes induced by alcohol and predict drivers of these changes. METHODS Mice were fed high-fat diet with or without 20% of alcohol in the drinking water for 20 weeks (WDA model). scATAC-seq data set was analyzed using Signac R package. To test the role of C/EBPβ, Cebpb-floxed mice were treated with AAV8-TBG-Cre or AAV8-control. RESULTS We analyzed differentially accessible regions in livers from control and alcohol-fed mice and found that activity of C/EBPβ transcription factor was associated with alcohol-induced epigenetic changes in hepatocytes. C/EBPβ protein levels were significantly upregulated in multiple models of ALD and human ALD samples. Using hepatocyte-specific Cebpb knockout mice we found that Cebpb loss protected male mice from alcohol-induced fibrosis development. We found no protection in female mice, suggesting that this mechanism is specific to male ALD. In vitro studies suggested that the protective effect of Cebpb loss was mediated by altered hepatocyte-macrophage cross talk. Cebpb knockout in hepatocytes reduced a profibrotic and promoted a pro-resolving phenotype in macrophages, thus modulating ALD development. We further identified the mediators of the cross talk. Cebpb knockout altered the expression of several HDL protein components, increasing APOA1 and apolipoprotein M and reducing apolipoprotein E and SAA levels in male mice. HDL secreted by Cebpb knockout hepatocytes was sufficient to confer anti-inflammatory and antifibrotic changes to macrophages. CONCLUSIONS Taken together, alcohol-induced C/EBPβ activation is a key driver of ALD fibrosis in males via C/EBPβ-dependent HDL remodeling.
Collapse
Affiliation(s)
- Michael Schonfeld
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Kruti Nataraj
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Steven Weinman
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
- Kansas City VA Medical Center, Kansas City, Missouri, USA
| | - Irina Tikhanovich
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
9
|
Zakaria S, Abass SA, Abdelatty M, Said S, Elsebaey S. Aescin ameliorates alcohol-induced liver injury. A possible implication of ROS / TNF-alpha / p38MAPK / caspase-3 signaling. Food Chem Toxicol 2025; 197:115270. [PMID: 39848460 DOI: 10.1016/j.fct.2025.115270] [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: 09/07/2024] [Revised: 12/18/2024] [Accepted: 01/19/2025] [Indexed: 01/25/2025]
Abstract
Alcoholic liver disease (ALD) is a commonly known liver disease mediated by prolonged alcohol consumption. Aescin is a triterpene saponin that can manage several conditions, including brain trauma, arthritis, venous congestion, stroke, and thrombophlebitis. Even so, studies illustrating the aescin role in ALD are scarce. Our study explored the potential effect of aescin in ALD in mice. In the current experiment, forty mice were utilized and sorted randomly into four groups: the control group received only vehicles, the alcohol group was given 5% alcohol in drinking water for four weeks, and the aescin-treated groups were given 5% alcohol in drinking water and aescin (10 and 20 mg/kg/day) for four weeks, then two doses of 60% alcohol (3g/kg) were given in the 29th and 30th day of the experiment. Our study revealed that aescin ameliorated alcohol-mediated liver damage, including reducing inflammatory cell infiltration and vascular dilatation. The serum concentrations of liver enzymes (ALT and AST) decreased in the aescin-treated groups. The apoptosis and oxidative stress also decreased, and the antioxidant enzyme activities were restored by aescin in alcohol-treated mice. Additionally, aescin decreased ethanol-induced inflammation by downregulating p38 MAPK and tumor necrosis factor-α (TNF-α), suggesting that aescin positively reduces alcohol-caused inflammation and oxidative stress. Consequently, aescin could ameliorate alcohol-induced hepatic damage by targeting the p38 MAPK/TNF-α signalling and could be developed as a novel health product that potentially ameliorates ALD.
Collapse
Affiliation(s)
- Sherin Zakaria
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kaferelsheikh University, Kaferelsheikh, Egypt.
| | - Shimaa A Abass
- Department of Biochemistry, Faculty of Pharmacy, Kaferelsheikh University, Kaferelsheikh, Egypt.
| | - Mona Abdelatty
- Department of Anatomy, Faculty of Medicine, Kaferelsheikh University, Kaferelsheikh, Egypt.
| | - Sama Said
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kaferelsheikh University, Kaferelsheikh, Egypt.
| | - Samar Elsebaey
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kaferelsheikh University, Kaferelsheikh, Egypt.
| |
Collapse
|
10
|
Leme RD, Marañón-Vásquez GA, Gonçalves JDL, de Carvalho FK, de Queiroz AM, de Paula-Silva FWG. Effect of prenatal alcohol consumption on dental enamel formation in offspring-An animal study protocol. PLoS One 2025; 20:e0317570. [PMID: 39951421 PMCID: PMC11828354 DOI: 10.1371/journal.pone.0317570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 12/30/2024] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND The etiology of developmental defects of enamel (DDE) remains incompletely understood. Prenatal alcohol exposure has been proposed as a potential risk factor for DDE. Animal studies suggest that in utero ethanol exposure can disrupt ameloblast function, leading to enamel abnormalities. This study aims to: (1) Assess the impact of prenatal alcohol consumption on the clinical and structural properties of dental enamel in offspring; and (2) Investigate the underlying mechanisms of these alterations through histological and molecular analyses. Pregnant Wistar rats will be assigned to two groups: one exposed to ethanol and a control group with no alcohol exposure. Ethanol exposure will follow a binge drinking model, with rats receiving 3 g/kg of ethanol (30% w/v) for 3 consecutive days, followed by 4 days of rest each week. This regimen will begin one week prior to conception and continue throughout pregnancy. The incisors and molars of offspring will be evaluated on the 10th (n = 22 per group) and 28th (n = 22 per group) days of life. Visible enamel changes will be documented through photographs. Enamel volume, thickness, and density will be assessed using micro-CT imaging. Mechanical properties will be evaluated using the Knoop microhardness test, while chemical composition will be analyzed through Scanning Electron Microscopy with Energy Dispersive X-ray (SEM-EDX) and Raman spectroscopy, respectively. The area of the organic enamel matrix will be quantified in histological sections. Genes Amelx, Enam, Ambn, Mmp2, Mmp9, Mmp20, Klk4, Cldn3, Cldn16, and Cldn19 will be evaluated in ameloblasts using real-time RT-PCR and protein synthesis will be confirmed by immunohistochemistry. Gelatinolytic activity in the ameloblast layer will be assessed by in situ zymography.
Collapse
Affiliation(s)
- Roberta Duarte Leme
- Department of Pediatric Dentistry, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Guido Artemio Marañón-Vásquez
- Department of Pediatric Dentistry, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Juliana de Lima Gonçalves
- Department of Pediatric Dentistry, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Fabrício Kitazono de Carvalho
- Department of Pediatric Dentistry, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Alexandra Mussolino de Queiroz
- Department of Pediatric Dentistry, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | |
Collapse
|
11
|
Liu M, Zhou M, Ren X, Xie Y. Establishment and application of murine models of alcoholic liver disease: A narrative review. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2025; 49:271-284. [PMID: 39715699 DOI: 10.1111/acer.15520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/29/2024] [Indexed: 12/25/2024]
Abstract
In recent years, there have been significant advances in pathological research on alcoholic liver disease (ALD), with suitable animal models making a significant contribution. However, the currently established animal ALD models still have some significant drawbacks, especially the inability to induce the entire human ALD lineage, which may be related to physiological differences between animals and humans. This review comprehensively summarized the most widely used experimental models of ALD, including voluntary drinking, Lieber-DeCarli, Meadows-Cook, Tsukamoto-French, NIAAA, and the "second hit" model. "Second hit" refers to an additional factor that damages the liver. There are various "second hit" models that fall into two main categories: particular diets and drugs. These models can either simulate human drinking patterns more accurately or produce varying degrees of ALD without significantly increasing animal mortality. We introduced the established method of the original models, discussed the advantages and disadvantages of the existing models from the aspects of operability and practicality, and provided existing improvement methods, hoping to provide a reference for future researchers.
Collapse
Affiliation(s)
- Mengsi Liu
- Peking University People's Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| | - Mingying Zhou
- Peking University People's Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| | - Xueyi Ren
- Peking University People's Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| | - Yandi Xie
- Peking University People's Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| |
Collapse
|
12
|
Cao P, Chao X, Ni HM, Ding WX. An Update on Animal Models of Alcohol-Associated Liver Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00032-X. [PMID: 39884572 DOI: 10.1016/j.ajpath.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/06/2024] [Accepted: 11/20/2024] [Indexed: 02/01/2025]
Abstract
Alcohol-associated liver disease (ALD) is a significant global health concern and a leading cause of liver disease-related deaths. However, the treatment options are limited due to the lack of animal models that accurately replicate ALD pathogenesis. An ideal ALD animal model should have pathological characteristics similar to those of human ALD, with a clear pathological process and ease of drug intervention. Over the years, researchers have focused on developing ideal ALD preclinical animal models by testing various methods, such as ad libitum drinking water with ethanol, acute, single large doses of ethanol gavage, multiple alcohol gavages in a short period, the Lieber-DeCarli liquid diet feeding model, the intragastric infusion model, and the Gao-binge model. With the increasing occurrence of obesity and metabolic dysfunction-associated steatotic liver disease, a new category of metabolic and alcohol-associated liver disease (MetALD) is also emerging. Studies have investigated the combined effects of a high-fat diet combined with binge alcohol or drinking water containing ethanol to mimic MetALD. In addition to mice, other species such as rats, guinea pigs, zebrafish, and non-human primates have also been tested to establish ALD preclinical models. This review aims to summarize current animal ALD models, particularly the emerging MetALD models, with the hope of providing a valuable reference for establishing more effective animal models in ALD studies in the future.
Collapse
Affiliation(s)
- Peng Cao
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Xiaojuan Chao
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas; Department of Internal Medicine, Division of Gastroenterology, Hepatology & Mobility, University of Kansas Medical Center, Kansas City, Kansas.
| |
Collapse
|
13
|
Radosavljevic T, Brankovic M, Djuretić J, Grujic-Milanovic J, Kovacic M, Jevtic J, Stankovic S, Samardzic J, Vucevic D, Jakovljevic V. Alpinetin Exhibits Antioxidant and Anti-Inflammatory Effects in C57BL/6 Mice with Alcoholic Liver Disease Induced by the Lieber-DeCarli Ethanol Liquid Diet. Int J Mol Sci 2024; 26:86. [PMID: 39795945 PMCID: PMC11720451 DOI: 10.3390/ijms26010086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 01/13/2025] Open
Abstract
Alcohol-associated liver disease (ALD) is a common non-communicable chronic liver disease characterized by a spectrum of conditions ranging from steatosis and alcohol-associated steatohepatitis (AH) to fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). The pathogenesis of ALD involves a complex interplay of various molecular, biochemical, genetic, epigenetic, and environmental factors. While the mechanisms are well studied, therapeutic options remain limited. Alpinetin, a natural flavonoid with antioxidant and anti-inflammatory properties, has shown potential hepatoprotective effects, though its efficacy in ALD remains unexplored. This study investigated the hepatoprotective effects of alpinetin using a Lieber-DeCarli ethanol liquid diet model of ALD in C57BL/6 mice. Mice were divided into three groups: the control group, the ethanol group, and the ethanol group treated with alpinetin. Serum activity of ALT, AST, γ-GT, and ALP was measured to assess liver function, along with antioxidative and oxidative/nitrosative stress markers in liver tissue. Pro-inflammatory cytokines and endoplasmic reticulum (ER) stress parameters in liver tissue were also evaluated. Histological assessment of disease activity was performed using the SALVE grading and staging system. Treatment with alpinetin significantly reduced serum levels of ALT, AST, γ-GT, and oxidative/nitrosative stress markers while increasing antioxidative markers. The levels of pro-inflammatory cytokines and ER stress parameters were significantly decreased. Histological analysis demonstrated reduced steatosis, hepatocyte ballooning, and inflammation. These findings suggest that alpinetin holds promise as a potential therapeutic agent for managing ALD.
Collapse
Affiliation(s)
- Tatjana Radosavljevic
- Institute of Pathophysiology “Ljubodrag Buba Mihailović”, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Milica Brankovic
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.B.); (J.S.)
| | - Jasmina Djuretić
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia;
| | - Jelica Grujic-Milanovic
- Institute for Medical Research, National Institute of the Republic of Serbia, Department of Cardiovascular Research, University of Belgrade, 11000 Belgrade, Serbia;
| | - Marijana Kovacic
- Group of Immunology, Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| | - Jovan Jevtic
- Institute of Pathology ‘Dr Đorđe Joannović’, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Sanja Stankovic
- Centre for Medical Biochemistry, University Clinical Centre of Serbia, 11000 Belgrade, Serbia;
- Department of Biochemistry, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Janko Samardzic
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.B.); (J.S.)
| | - Danijela Vucevic
- Institute of Pathophysiology “Ljubodrag Buba Mihailović”, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Vladimir Jakovljevic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
- Center of Excellence for the Study of Redox Balance in Cardiovascular and Metabolic Disorders, University of Kragujevac, 34000 Kragujevac, Serbia
- Department of Human Pathology, First Moscow State Medical University I.M. Sechenov, Trubetskaya Street 8, Str. 2, 119991 Moscow, Russia
| |
Collapse
|
14
|
Fu J, Zhang Z, Zhao Y, Li X, Jiang C, He H, Huo J, Xiao Q, Wu J, Zhu F, Chen J. Acetylcorynoline alleviates acute liver injury via inhibiting TLR4/JNK/NF-ĸB pathway Based on RNA-seq and molecular docking in vivo and in vitro. Int Immunopharmacol 2024; 143:113550. [PMID: 39522313 DOI: 10.1016/j.intimp.2024.113550] [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: 07/13/2024] [Revised: 10/19/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Acute liver injury is characterized by massive inflammatory cell infiltration, destruction of liver structure and abnormalities in liver function. Acetylcorynoline (AC) is one of the main chemical components of Corydalis bungeana Turcz. which has been shown to have a protective effect against acute liver injury. However, Whether AC is protective against acute liver injury remains unclear. This study aimed to explore the protective mechanism of AC against acute liver injury from in vivo as well as experiments in vitro. In experimental in vivo studies, AC pretreatment reduced the serum levels of ALT and AST and inhibited the expression of inflammatory factors in the liver of LPS/D-GalN-induced mice and alcohol liver disease mice. RNA-sequencing and molecular docking were used to predict that AC exerts its anti-inflammatory effects through the Toll-like receptor signaling pathway. Using RT-qPCR and Western blotting to detect expression levels of key genes and nodal proteins of the Toll-like receptor signaling pathway, AC was found to inhibit the phosphorylation of nuclear factor-kappaB (NF-ĸB) and c-Jun amino-terminal kinase (JNK). This finding was validated in cellular experiments. In conclusion, AC exerts its anti-hepatic injury effect by suppressing inflammation through inhibition of the TLR4/JNK/NF-ĸB pathway.
Collapse
Affiliation(s)
- Jun Fu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Key Laboratory of New Drug Delivery Systems of Chinese Materia Medica, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Zhenxu Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Key Laboratory of New Drug Delivery Systems of Chinese Materia Medica, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Yaning Zhao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Key Laboratory of New Drug Delivery Systems of Chinese Materia Medica, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Xin Li
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Key Laboratory of New Drug Delivery Systems of Chinese Materia Medica, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Cuihua Jiang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Key Laboratory of New Drug Delivery Systems of Chinese Materia Medica, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Haoran He
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Key Laboratory of New Drug Delivery Systems of Chinese Materia Medica, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Jiege Huo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Jiangsu Clinical Innovation Center of Digestive Cancer of Traditional Chinese Medicine, Nanjing 210028, PR China
| | - Qi Xiao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China
| | - Jie Wu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China
| | - Fenxia Zhu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Key Laboratory of New Drug Delivery Systems of Chinese Materia Medica, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, PR China.
| | - Jiaquan Chen
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing 211198, PR China.
| |
Collapse
|
15
|
Meng X, Ma Y, Li K, Ji M, Lin L, Xiao X, Zhao Y, Su G. Effect and Mechanism of Apple Polyphenols in Regulating Intestinal Flora and Inhibiting the TLR4/NF-κB/TGF-β Signaling Pathway to Alleviate Alcoholic Liver Fibrosis. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2024; 79:915-919. [PMID: 39298074 DOI: 10.1007/s11130-024-01235-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/31/2024] [Indexed: 09/21/2024]
Abstract
Apple polyphenols (APs) have gained attention for their various bioactivities, while no studies on anti-liver fibrosis activity are reported. This study evaluated the protective effect of APs on liver fibrosis using LPS-treated activated HSC-T6 cells and alcohol-treated liver fibrosis (ALF) mice. The results indicated that APs inhibited HSC-T6 cells activation in vitro and reduced the level of serum hyaluronic acid (HA) (p < 0.05), decreased fibrogenesis marker expression (p < 0.05), thereby alleviating ALF. In addition, APs (800 mg/kg b.w.) decreased the Firmicutes/Bacteroidetes ratio (p < 0.05) in ALF mice, inhibited LPS accumulation in the liver tissue and serum (p < 0.05), and significantly inhibited the TLR4/NF-κB/TGF-β signaling in mice liver. In conclusion, APs markedly ameliorated ALF, possibly by improving gut microbiota homeostasis, decreasing the translocation of bacterial endotoxins to the blood, and suppressing the TLR4/NF-κB/TGF-β signaling pathway, indicating its potential as lead compounds for functional foods and/or drugs against ALF.
Collapse
Affiliation(s)
- Xianyi Meng
- Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yunjie Ma
- Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Kaidi Li
- Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Maxin Ji
- Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Lizhen Lin
- Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xianyi Xiao
- Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yuqing Zhao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, 133002, China.
| | - Guangyue Su
- Shenyang Pharmaceutical University, Shenyang, 110016, China.
- Key Laboratory of Innovative Traditional Chinese Medicine for Major Chronic Diseases of Liaoning Province, Key Laboratory for TCM Material Basis Study and Innovative Drug Development of Shenyang City, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
16
|
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
|
17
|
Zhang L, Wang R, Nan Y, Kong L. Deciphering the role of LncRNA in alcoholic liver disease: Mechanisms and therapeutic potential. Medicine (Baltimore) 2024; 103:e40378. [PMID: 39533619 PMCID: PMC11557020 DOI: 10.1097/md.0000000000040378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Alcoholic liver disease (ALD) is a spectrum of liver damage caused by chronic alcohol consumption. The disease progresses in stages, starting with simple fatty liver, progressing to alcoholic hepatitis and potentially leading to fibrosis and cirrhosis. The pathophysiology of ALD is complex and involves several cellular and molecular mechanisms. Recent research has highlighted the role of long non-coding RNAs (LncRNAs) as critical regulators in the development and progression of ALD. This article reviews the current understanding of LncRNAs in ALD, focusing on their functions in key pathological processes and their potential as diagnostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Traditional and Western Medical Hepatology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Rongqi Wang
- Department of Traditional and Western Medical Hepatology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yuemin Nan
- Department of Traditional and Western Medical Hepatology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lingbo Kong
- Department of Traditional and Western Medical Hepatology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
18
|
Mittal A, Choudhary N, Chaudhary S, Kumari A, Rastogi A, Kumar G, Maras JS, Sarin SK, Sharma S. Ethanol with thioacetamide murine model of alcoholic liver disease identifies hepatic pathways as targets for the human disease. Ann Hepatol 2024; 30:101565. [PMID: 39276982 DOI: 10.1016/j.aohep.2024.101565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/21/2024] [Accepted: 07/17/2024] [Indexed: 09/17/2024]
Abstract
INTRODUCTION AND OBJECTIVES Hepatic proteome and gut microbiota alterations are known in alcohol-associated hepatitis (AAH). Current animal models sparsely mimic human AAH. We aimed to develop an murine model that closely resembled human AAH. MATERIALS AND METHODS Male C57BL/6N mice were pair-fed control/incremental ethanol Lieber-DeCarli diets and thioacetamide (TAA) for 12-weeks to induce AAH. Hepatic proteome was analyzed using LC-MS/MS. Gut-bacteria was determined using 16s-rRNA sequencing. RESULTS Mice exposed to EtOH+TAA displayed higher expression of liver triglycerides (1.5-fold, p = 0.001), pro-inflammatory (IL6, 1.5-fold, p = 0.002 and TNFα, 1.7-fold, p = 0.01), fibrotic (TGF-β, 2.7-fold, p = 0.01 and Col1α1, 2-fold, p = 0.01) and oxidative markers (GSH and SOD (-1.5 fold, p = 0.004 & 0.005 respectively)) as compared to EtOH alone. Histology of EtOH+TAA liver displayed pericellular liver fibrosis, increased steatosis, and neutrophil infiltration, which resembled human AAH. In the 12wk EtOH+TAA group, Desulfobacteria, Campylobacteria, and Patescibacteria increased by 2-fold (p = 0.02). Pathway combined score (CS, log10) in EtOH+TAA treatment showed upregulated hepatic ethanol oxidation (CS=1.93), fatty acid biosynthesis (CS=2.48), necrosis (CS=1.59), collagen formation (CS=1.28) and hypoxia (CS=0.68) and downregulated fatty acid beta-oxidation (CS=2.37), PPAR signaling (CS=1.35) fatty acid degradation (CS=2.35), bile acid metabolism (CS=1.87), and oxidative phosphorylation (CS=1.50), as observed in human disease. CONCLUSIONS Using an ethanol-thioacetamide combination in mice results in a faster establishment of AAH with fibrosis than previously known models. Differential protein expression strongly correlates with pathways found altered in human AAH, thus making the model mimic human disease better than other known models., respectively. Thioacetamide (TAA) was administered to enhance liver fibrosis and mimic human AAH.
Collapse
Affiliation(s)
- Ashi Mittal
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Nishu Choudhary
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Sudrishti Chaudhary
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Anupama Kumari
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Archana Rastogi
- Department of Histopathology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Guresh Kumar
- Department of Biostatistics, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Jaswinder Singh Maras
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Shiv K Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Shvetank Sharma
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India.
| |
Collapse
|
19
|
Jiang W, Zhang M, Cao R, Wang X, Zuo Y. Different ethanol exposure durations affect cytochrome P450 2E1-mediated sevoflurane metabolism in rat liver. BMC Anesthesiol 2024; 24:321. [PMID: 39256673 PMCID: PMC11384694 DOI: 10.1186/s12871-024-02704-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 08/27/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Chronic alcohol users often exhibit an increased minimum alveolar concentration (MAC) of sevoflurane, yet the specific mechanism remains unclear. It has been reported that ethanol exposure can upregulate the protein expression and enzyme activity of cytochrome P450 2E1 (CYP2E1). CYP2E1 is a key enzyme that converts 2-5% of sevoflurane into equimolar amounts of hexafluoroisopropanol (HFIP) and F-. This study aims to explore whether ethanol exposure could alter sevoflurane metabolism through CYP2E1 modulation, potentially explaining the increased MAC observed in alcohol users. METHODS Eighty adult male Sprague-Dawley (SD) rats were randomly divided into two groups and received either 50% ethanol (dose: 3 g/kg) or 0.9% saline twice daily by gavage. After 1, 2, 3, and 4 weeks of gavage, ten rats were randomly selected from each group to undergo 1-hour anesthesia with 2.3% sevoflurane. Blood samples were collected after anesthesia to measure the concentration of free HFIP using gas chromatography. Additionally, the left lobe tissue of the liver was collected for the analysis of CYP2E1 protein expression by Western blot and CYP2E1 enzyme activity by colorimetric assay. Correlations between these parameters were analyzed using Pearson's correlation. RESULTS In the ethanol group, CYP2E1 expression, activity, and the concentration of free HFIP were significantly higher at all time points compared to the control group (P < 0.05), except for protein expression in the first week (P > 0.05). Within-group comparisons indicated no significant changes in any of the parameters for the control group (P > 0.05). In the ethanol group, there was no difference in free HFIP concentration between the first and second weeks (P > 0.05), but a significant increase was observed in the third and fourth weeks (P < 0.01); protein expression and enzyme activity significantly varied over time, especially showing a notable increase from the first to the third and fourth weeks (P < 0.05). Correlation analysis revealed strong positive correlations between free HFIP concentration and CYP2E1 activity (r = 0.7898), free HFIP concentration and CYP2E1 expression (r = 0.8418), and CYP2E1 activity and expression (r = 0.8740), all with P < 0.001. CONCLUSIONS Ethanol exposure increased both the expression and enzymatic activity of CYP2E1, consequently enhancing the metabolism of sevoflurane.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Anesthesiology, School of Clinical Medicine of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Min Zhang
- Department of Anesthesiology, School of Clinical Medicine of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Rui Cao
- Department of Anesthesiology, School of Clinical Medicine of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Xinghao Wang
- Department of Anesthesiology, School of Clinical Medicine of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Youbo Zuo
- Department of Anesthesiology, School of Clinical Medicine of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
- Department of Anesthesiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| |
Collapse
|
20
|
Ock SA, Kim SY, Kim YI, Ju WS, Lee P. Enhanced In Vitro Recapitulation of In Vivo Liver Regeneration by Co-Culturing Hepatocyte Organoids with Adipose-Derived Mesenchymal Stem Cells, Alleviating Steatosis and Apoptosis in Acute Alcoholic Liver Injury. Cells 2024; 13:1303. [PMID: 39120332 PMCID: PMC11311897 DOI: 10.3390/cells13151303] [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: 06/26/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024] Open
Abstract
Hepatocyte organoids (HOs) have superior hepatic functions to cholangiocyte-derived organoids but suffer from shorter lifespans. To counteract this, we co-cultured pig HOs with adipose-derived mesenchymal stem cells (A-MSCs) and performed transcriptome analysis. The results revealed that A-MSCs enhanced the collagen synthesis pathways, which are crucial for maintaining the three-dimensional structure and extracellular matrix synthesis of the organoids. A-MSCs also increased the expression of liver progenitor cell markers (KRT7, SPP1, LGR5+, and TERT). To explore HOs as a liver disease model, we exposed them to alcohol to create an alcoholic liver injury (ALI) model. The co-culture of HOs with A-MSCs inhibited the apoptosis of hepatocytes and reduced lipid accumulation of HOs. Furthermore, varying ethanol concentrations (0-400 mM) and single-versus-daily exposure to HOs showed that daily exposure significantly increased the level of PLIN2, a lipid storage marker, while decreasing CYP2E1 and increasing CYP1A2 levels, suggesting that CYP1A2 may play a critical role in alcohol detoxification during short-term exposure. Moreover, daily alcohol exposure led to excessive lipid accumulation and nuclear fragmentation in HOs cultured alone. These findings indicate that HOs mimic in vivo liver regeneration, establishing them as a valuable model for studying liver diseases, such as ALI.
Collapse
Affiliation(s)
- Sun A Ock
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 1500 Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun 55365, Republic of Korea (P.L.)
| | | | | | | | | |
Collapse
|
21
|
Duan W, Liu F, Ren Y, Zhang X, Shi JS, Xue Y, Xu ZH, Geng Y. Differences in the Ability of Lactic Acid Bacteria To Prevent Acute Alcohol-Induced Liver Injury via the Gut Microbiota-Bile Acid-Liver Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:15265-15275. [PMID: 38918075 DOI: 10.1021/acs.jafc.4c01353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
Probiotics can regulate gut microbiota and protect against acute alcohol-induced liver injury through the gut-liver axis. However, efficacy is strain-dependent, and their mechanism remains unclear. This study investigated the effect of lactic acid bacteria (LAB), including Lacticaseibacillus paracasei E10 (E10), Lactiplantibacillus plantarum M (M), Lacticaseibacillus rhamnosus LGG (LGG), Lacticaseibacillus paracasei JN-1 (JN-1), and Lacticaseibacillus paracasei JN-8 (JN-8), on the prevention of acute alcoholic liver injury in mice. We found that LAB pretreatment reduced serum alanine transaminase (ALT) and aspartate transaminase (AST) and reduced hepatic total cholesterol (TC) and triglyceride (TG). JN-8 pretreatment exhibited superior efficacy in improving hepatic antioxidation. LGG and JN-8 pretreatment significantly attenuated hepatic and colonic inflammation by decreasing the expression of interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α) and increasing the expression of interleukin 10 (IL-10). JN-1 and JN-8 pretreatments have better preventive effects than other LAB pretreatment on intestinal barrier dysfunction. In addition, the LAB pretreatment improved gut microbial dysbiosis and bile acid (BA) metabolic abnormality. All of the strains were confirmed to have bile salt deconjugation capacities in vitro, where M and JN-8 displayed higher activities. This study provides new insights into the prevention and mechanism of LAB strains in preventing acute alcoholic liver injury.
Collapse
Affiliation(s)
- Wenhui Duan
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Fei Liu
- Wuxi Hospital of Traditional Chinese Medicine, Wuxi, Jiangsu 214122, People's Republic of China
| | - Yilin Ren
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Xiaojuan Zhang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Jin-Song Shi
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Yuzheng Xue
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Zheng-Hong Xu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
- Advanced Brewing Technology Innovation Center, Sichuan University, Chengdu, Sichuan 610065, People's Republic of China
| | - Yan Geng
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| |
Collapse
|
22
|
Han J, Lee C, Jeong H, Jeon S, Lee M, Lee H, Choi YH, Jung Y. Tumor necrosis factor-inducible gene 6 protein and its derived peptide ameliorate liver fibrosis by repressing CD44 activation in mice with alcohol-related liver disease. J Biomed Sci 2024; 31:54. [PMID: 38790021 PMCID: PMC11127441 DOI: 10.1186/s12929-024-01042-5] [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: 01/26/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Alcohol-related liver disease (ALD) is a major health concern worldwide, but effective therapeutics for ALD are still lacking. Tumor necrosis factor-inducible gene 6 protein (TSG-6), a cytokine released from mesenchymal stem cells, was shown to reduce liver fibrosis and promote successful liver repair in mice with chronically damaged livers. However, the effect of TSG-6 and the mechanism underlying its activity in ALD remain poorly understood. METHODS To investigate its function in ALD mice with fibrosis, male mice chronically fed an ethanol (EtOH)-containing diet for 9 weeks were treated with TSG-6 (EtOH + TSG-6) or PBS (EtOH + Veh) for an additional 3 weeks. RESULTS Severe hepatic injury in EtOH-treated mice was markedly decreased in TSG-6-treated mice fed EtOH. The EtOH + TSG-6 group had less fibrosis than the EtOH + Veh group. Activation of cluster of differentiation 44 (CD44) was reported to promote HSC activation. CD44 and nuclear CD44 intracellular domain (ICD), a CD44 activator which were upregulated in activated HSCs and ALD mice were significantly downregulated in TSG-6-exposed mice fed EtOH. TSG-6 interacted directly with the catalytic site of MMP14, a proteolytic enzyme that cleaves CD44, inhibited CD44 cleavage to CD44ICD, and reduced HSC activation and liver fibrosis in ALD mice. In addition, a novel peptide designed to include a region that binds to the catalytic site of MMP14 suppressed CD44 activation and attenuated alcohol-induced liver injury, including fibrosis, in mice. CONCLUSIONS These results demonstrate that TSG-6 attenuates alcohol-induced liver damage and fibrosis by blocking CD44 cleavage to CD44ICD and suggest that TSG-6 and TSG-6-mimicking peptide could be used as therapeutics for ALD with fibrosis.
Collapse
Affiliation(s)
- Jinsol Han
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Chanbin Lee
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
- Institute of Systems Biology, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Hayeong Jeong
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Seunghee Jeon
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Myunggyo Lee
- Department of Pharmacy, College of Pharmacy and Research Institute for Drug Development, Pusan National University, Pusan, 46241, Republic of Korea
| | - Haeseung Lee
- Department of Pharmacy, College of Pharmacy and Research Institute for Drug Development, Pusan National University, Pusan, 46241, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, Dong-Eui University College of Korean Medicine, Pusan, 47227, Republic of Korea
| | - Youngmi Jung
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea.
- Department of Biological Sciences, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea.
| |
Collapse
|
23
|
Peinado BRR, Frazão DR, Chemelo VS, Matos-Souza JM, Ferreira RDO, Bittencourt LO, Balbinot GDS, Collares FM, Fernandes LMP, Maia CSF, Lima RR. Physical training mitigates alveolar bone and blood enzymatic antioxidants defense impairment induced by binge ethanol consumption in rats. Biomed Pharmacother 2024; 174:116554. [PMID: 38636401 DOI: 10.1016/j.biopha.2024.116554] [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: 02/01/2024] [Revised: 03/26/2024] [Accepted: 04/04/2024] [Indexed: 04/20/2024] Open
Abstract
We aimed to investigate the effectiveness of physical training as a protective strategy to mitigate alveolar bone damage and blood antioxidant defense caused by ethanol (EtOH) consumption in a binge-drinking pattern. Male Wistar rats aged approximately 90 days were divided into four groups: control, training, EtOH, and training + EtOH. The physical training protocol was conducted on a treadmill for four consecutive weeks, while the animals in the EtOH group were administered EtOH via orogastric gavage for three consecutive days each week, following the binge drink pattern. After the training period, blood and mandibles were collected for plasma oxidative biochemistry analysis, and the alveolar bone was subjected to physicochemical composition analysis, tissue evaluation, and microtomography evaluation. Our results showed that EtOH induced oxidative stress and physical exercise promoted the recovery of antioxidant action. Physical training minimized the damage to the mineral/matrix composition of the alveolar bone due to EtOH consumption and increased the density of osteocytes in the trained group treated with EtOH than in those exposed only to EtOH. Furthermore, physical training reduced damage to the alveolar bone caused by EtOH consumption. Our findings suggest that physical training can serve as an effective strategy to reduce systemic enzymatic oxidative response damage and alleviate alveolar bone damage resulting from alcohol consumption. Further investigations are warranted to elucidate the underlying mechanisms and explore, in addition to physical training, the potential effects of other activities with varying intensities on managing alcohol-induced bone damage.
Collapse
Affiliation(s)
| | - Deborah Ribeiro Frazão
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém, PA, Brazil
| | - Victória Santos Chemelo
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém, PA, Brazil
| | - José Mario Matos-Souza
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém, PA, Brazil
| | - Railson de Oliveira Ferreira
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém, PA, Brazil
| | - Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém, PA, Brazil
| | - Gabriela de Souza Balbinot
- Dental Material Laboratory, School of Dentistry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Fabrício Mezzomo Collares
- Dental Material Laboratory, School of Dentistry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Luanna Melo Pereira Fernandes
- Laboratory of Neuropharmacology and Behavior, Center of Sciences Biological and Health, State University of Pará, Belém, PA, Brazil
| | - Cristiane Socorro Ferraz Maia
- Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará (UFPA), Belém, PA, Brazil
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém, PA, Brazil.
| |
Collapse
|
24
|
Tamnanloo F, Chen X, Oliveira MM, Tremblay M, Rose CF. Excessive intragastric alcohol administration exacerbates hepatic encephalopathy and provokes neuronal cell death in male rats with chronic liver disease. J Neurosci Res 2024; 102:e25337. [PMID: 38680084 DOI: 10.1002/jnr.25337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/11/2024] [Accepted: 04/07/2024] [Indexed: 05/01/2024]
Abstract
Hepatic encephalopathy (HE) is defined as decline in neurological function during chronic liver disease (CLD). Alcohol is a major etiological factor in the pathogenesis of fibrosis/cirrhosis and has also been documented to directly impact the brain. However, the role of alcohol in the development of HE in CLD remains unclear. Here, we investigated the impact of excessive alcohol administration on neurological deterioration in rats with CLD. Starting day 7 post-BDL surgery, rats were administered alcohol twice daily (51% v/v ethanol, 3 g/kg, via gavage) for 4 weeks. Motor coordination was assessed weekly using rotarod and anxiety-like behavior was evaluated with open field and elevated plus maze at 5 weeks. Upon sacrifice, brains were collected for western blot and immunohistochemical analyses to investigate neuronal integrity and oxidative stress status. Alcohol worsened motor coordination performance and increased anxiety-like behavior in BDL rats. Impairments were associated with decreased neuronal markers of NeuN and SMI311, increased apoptotic markers of cleaved/pro-caspase-3 and Bax/Bcl2, increased necroptosis markers of pRIP3 and pMLKL, decreased total antioxidant capacity (TAC), and increased 4-hydroxynonenal (4-HNE)modified proteins in the cerebellum of BDL-alcohol rats when compared to respective controls. Immunofluorescence confirmed the colocalization of cleaved caspase-3 and pMLKL in the granular neurons of the cerebellum of BDL-alcohol rats. Excessive alcohol consumption exacerbates HE which leads to associated apoptotic and necroptotic neuronal loss in the cerebellum of BDL-alcohol rats. Additionally, higher levels of 4-HNE and decreased TAC in the cerebellum of BDL-alcohol rats suggest oxidative stress is the triggering factor of apoptotic and necroptotic neuronal loss/injury.
Collapse
Affiliation(s)
- Farzaneh Tamnanloo
- Hepato-Neuro Lab, CRCHUM, Montréal, Québec, Canada
- Medicine Department, Université de Montréal, Montréal, Québec, Canada
| | - Xiaoru Chen
- Hepato-Neuro Lab, CRCHUM, Montréal, Québec, Canada
| | | | | | - Christopher F Rose
- Hepato-Neuro Lab, CRCHUM, Montréal, Québec, Canada
- Medicine Department, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
25
|
Wang D, Lv J, Fu Y, Shang Y, Liu J, Lyu Y, Wei M, Yu X. Optimization of Microwave-Assisted Extraction Process of Total Flavonoids from Salicornia bigelovii Torr. and Its Hepatoprotective Effect on Alcoholic Liver Injury Mice. Foods 2024; 13:647. [PMID: 38472759 DOI: 10.3390/foods13050647] [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: 12/15/2023] [Revised: 02/18/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
The objective of this study was to determine the optimal extraction conditions for total flavonoids from S. bigelovii using microwave-assisted extraction and to analyze the protective effect of total flavonoids from S. bigelovii on alcoholic liver injury in mice. The optimization of the process conditions for the microwave-assisted extraction of total flavonoids from S. bigelovii was performed using response surface methodology, and an alcohol-induced acute liver injury model in mice was used to investigate the effects of different doses of total flavonoids (100 mg/kg, 200 mg/kg, and 400 mg/kg) on the levels and activities of serum alanine aminotransferase kits (ALT), glutamic oxaloacetic transaminase kits (AST), superoxide dismutase kits (SOD), glutathione peroxidase kits (GSH-Px), and malondialdehyde (MDA). We performed hematoxylin-eosin (H&E) staining analysis on pathological sections of mouse liver tissue, and qRT-PCR technology was used to detect the expression levels of the inflammatory factors IL-1 β, IL-6, and TNF-α. The results revealed that the optimal extraction process conditions for total flavonoids in S. bigelovii were a material-to-liquid ratio of 1:30 (g/mL), an ethanol concentration of 60%, an extraction temperature of 50 °C, an ultrasound power of 250 W, and a yield of 5.71 ± 0.28 mg/g. Previous studies have demonstrated that the flavonoids of S. bigelovii can significantly inhibit the levels of ALT and AST in the serum (p < 0.001), reduce MDA levels (p < 0.001), increase the activity of the antioxidant enzymes SOD and GSH-Px (p < 0.001), and inhibit the IL-1 β, IL-6, and TNF-α gene expression levels (p < 0.001) of inflammatory factors. The total flavonoids of S. bigelovii exert a protective effect against alcoholic liver injury by reducing the levels of inflammation, oxidative stress, and lipid peroxidation caused by alcohol. The results of this study lay the foundation for the high-value utilization of S. bigelovii and provide new resources for the development of liver-protective drugs.
Collapse
Affiliation(s)
- Dujun Wang
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - Jing Lv
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - Yan Fu
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - Yueling Shang
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - Jinbin Liu
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - Yongmei Lyu
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - Ming Wei
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - Xiaohong Yu
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China
| |
Collapse
|
26
|
Sato F, Bhawal UK, Oikawa K, Muragaki Y. Loss of Dec1 inhibits alcohol-induced hepatic lipid accumulation and circadian rhythm disorder. BMC Mol Cell Biol 2024; 25:1. [PMID: 38166556 PMCID: PMC10763066 DOI: 10.1186/s12860-023-00497-y] [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: 06/28/2023] [Accepted: 12/18/2023] [Indexed: 01/04/2024] Open
Abstract
Chronic alcohol exposure increases liver damage such as lipid accumulation and hepatitis, resulting in hepatic cirrhosis. Chronic alcohol intake is known to disturb circadian rhythms in humans and animals. DEC1, a basic helix-loop-helix transcription factor, plays an important role in the circadian rhythm, inflammation, immune responses, and tumor progression. We have previously shown that Dec1 deficiency inhibits stresses such as periodontal inflammation and perivascular fibrosis of the heart. However, the significance of Dec1 deficiency in chronic alcohol consumption remains unclear. In the present study, we investigated whether the biological stress caused by chronic alcohol intake is inhibited in Dec1 knockout mice. We treated control and Dec1 knockout mice for three months by providing free access to 10% alcohol. The Dec1 knockout mice consumed more alcohol than control mice, however, we observed severe hepatic lipid accumulation and circadian rhythm disturbance in control mice. In contrast, Dec1 knockout mice exhibited little effect on these outcomes. We also investigated the expression of peroxisome proliferator-activated receptors (PPARs) and AMP-activated protein kinase (AMPK), which are involved in the regulation of fatty acid metabolism. Immunohistochemical analysis revealed increases of phosphorylation AMPK and PPARa but decreases PPARg in Dec1 knockout mice compared to that in control mice. This indicates a molecular basis for the inhibition of hepatic lipid accumulation in alcohol-treated Dec1 knockout mice. These results suggest a novel function of Dec1 in alcohol-induced hepatic lipid accumulation and circadian rhythm disorders.
Collapse
Affiliation(s)
- Fuyuki Sato
- Department of Diagnostic Pathology, Shizuoka Cancer Center, Sunto-gun, 411-8777, Japan.
- Department of Pathology, Wakayama Medical University School of Medicine, Wakayama, 641- 8509, Japan.
| | - Ujjal K Bhawal
- Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Chiba, 271-8587, Japan
- Center for Global Health Research , Saveetha Medical College and Hospitals , Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, India
| | - Kosuke Oikawa
- Department of Pathology, Wakayama Medical University School of Medicine, Wakayama, 641- 8509, Japan
| | - Yasuteru Muragaki
- Department of Pathology, Wakayama Medical University School of Medicine, Wakayama, 641- 8509, Japan
| |
Collapse
|
27
|
Yoladı FB, Burmaoğlu E, Palabıyık ŞS. Experimental In Vivo Toxicity Models for Alcohol Toxicity. Eurasian J Med 2023; 55:82-90. [PMID: 39109811 PMCID: PMC11075036 DOI: 10.5152/eurasianjmed.2023.23345] [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: 11/28/2023] [Accepted: 12/26/2023] [Indexed: 08/11/2024] Open
Abstract
Alcohol consumption poses a significant risk for the development of chronic illnesses, one of the leading causes of "preventable" disease and death worldwide. Harmful consumption of alcohol is thought to result in approximately 2.5-3 million deaths each year, the majority of which are caused by alcohol-related liver diseases. Hepatocellular carcinoma, cirrhosis, fibrosis, steatosis, and steatohepatitis are among the liver illnesses caused by alcohol. The mechanisms behind human diseases are often mimicked and understood through the use of animal models. Rodents are the ideal animals to study alcohol-related liver diseases. In these experimental models using rodents, the ethanol ratio, method of administration, and diet to be applied vary. Within the scope of this review, it is aimed at providing information about the experimental models used today for alcohol toxicity and the advantages and disadvantages of these models.
Collapse
Affiliation(s)
- Fatma Betül Yoladı
- Department of Pharmaceutical Toxicology, Atatürk University Faculty of Pharmacy, Erzurum, Turkey
- Department of Pharmaceutical Toxicology, Hacettepe University Faculty of Pharmacy, Ankara, Turkey
| | | | - Şaziye Sezin Palabıyık
- Department of Pharmaceutical Toxicology, Atatürk University Faculty of Pharmacy, Erzurum, Turkey
- Clinical Research, Development and Design Application and Research Center, Atatürk University, Erzurum, Turkey
| |
Collapse
|
28
|
Lin CY, Omoscharka E, Liu Y, Cheng K. Establishment of a Rat Model of Alcoholic Liver Fibrosis with Simulated Human Drinking Patterns and Low-Dose Chemical Stimulation. Biomolecules 2023; 13:1293. [PMID: 37759693 PMCID: PMC10526499 DOI: 10.3390/biom13091293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/16/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Although alcohol is a well-known causal factor associated with liver diseases, challenges remain in inducing liver fibrosis in experimental rodent models. These challenges include rodents' natural aversion to high concentrations of alcohol, rapid alcohol metabolism, the need for a prolonged duration of alcohol administration, and technical difficulties. Therefore, it is crucial to establish an experimental model that can replicate the features of alcoholic liver fibrosis. The objective of this study was to develop a feasible rat model of alcoholic liver fibrosis that emulates human drinking patterns and combines low-dose chemicals within a relatively short time frame. We successfully developed an 8-week rat model of alcoholic liver fibrosis that mimics chronic and heavy drinking patterns. Rats were fed with a control liquid diet, an alcohol liquid diet, or alcohol liquid diet combined with multiple binges via oral gavage. To accelerate the progression of alcoholic liver fibrosis, we introduced low-dose carbon tetrachloride (CCl4) through intraperitoneal injection. This model allows researchers to efficiently evaluate potential therapeutics in preclinical studies of alcoholic liver fibrosis within a reasonable time frame.
Collapse
Affiliation(s)
- Chien-Yu Lin
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Evanthia Omoscharka
- Department of Pathology, University Health/Truman Medical Center, School of Medicine, University of Missouri-Kansas City, 2301 Holmes Street, Kansas City, MO 64108, USA
| | - Yanli Liu
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Kun Cheng
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| |
Collapse
|
29
|
Kotulkar M, Robarts DR, Lin-Rahardja K, McQuillan T, Surgnier J, Tague SE, Czerwinski M, Dennis KL, Pritchard MT. Hyaluronan synthesis inhibition normalizes ethanol-enhanced hepatic stellate cell activation. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:1544-1559. [PMID: 37332093 DOI: 10.1111/acer.15127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 05/14/2023] [Accepted: 06/01/2023] [Indexed: 06/20/2023]
Abstract
BACKGROUND Chronic ethanol overconsumption promotes alcohol-associated liver disease (ALD), characterized by hepatocyte injury, inflammation, hepatic stellate cell (HSC) activation, and fibrosis. Hyaluronan (HA) concentration is greater in livers and blood from advanced ALD patients than patients with advanced non-ALD. In the liver, HSCs are the major HA producers. The relationship between ethanol, HA, and HSC activation is incompletely understood. Thus, here, we tested the hypothesis that ethanol enhances HSC activation in a HA-dependent manner. METHODS Liver tissue microarrays (TMAs) containing steatotic livers from donors with or without a history of alcohol consumption were used to measure HA and collagen content. Mice were fed a moderate (2%, v/v) ethanol-containing diet or pair-fed control diet for 2 days, after which they were given a single carbon tetrachloride (CCl4 ) injection. To inhibit HA synthesis, we provided 4-methylumbelliferone (4MU) daily. We used LX2 cells, a human HSC cell line, to determine the impact ethanol had on LPS responses, with or without concurrent 4MU exposure. RESULTS CCl4 induced liver injury, but it did not differ between ethanol or control diet fed mice with or without 4MU treatment. Ethanol feeding enhanced CCl4 -induced hepatic HA content, which was paralleled by HA synthase (Has)2 transcript abundance; 4MU treatment normalized both. Consistently, HSC activation, assessed by measuring αSMA mRNA and protein, was induced by CCl4 exposure, enhanced by ethanol feeding, and normalized by 4MU. Hepatic transcripts, but not protein, for Ccl2 were enhanced by ethanol feeding and normalized by 4MU exposure. Finally, ethanol-exposed LX2 cells made more LPS-stimulated CCL2 mRNA and protein than cells not exposed to ethanol; 4MU prevented this. CONCLUSION These data show that ethanol augments HSC activation through HA synthesis and enhances hepatic profibrogenic features. Therefore, targeting HSC HA production could potentially attenuate liver disease in ALD patients.
Collapse
Affiliation(s)
- Manasi Kotulkar
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Dakota R Robarts
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Kristi Lin-Rahardja
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Tara McQuillan
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jordan Surgnier
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Sarah E Tague
- Kansas Intellectual and Developmental Disabilities Research Center, University of Kansas Medical Center, Kansas City, Kansas, USA
| | | | - Katie L Dennis
- Department of Pathology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Michele T Pritchard
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
- The Liver Center, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
30
|
Wang H, Shen H, Seo W, Hwang S. Experimental models of fatty liver diseases: Status and appraisal. Hepatol Commun 2023; 7:e00200. [PMID: 37378635 DOI: 10.1097/hc9.0000000000000200] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/16/2023] [Indexed: 06/29/2023] Open
Abstract
Fatty liver diseases, including alcohol-associated liver disease (ALD) and nonalcoholic fatty liver disease nonalcoholic fatty liver disease (NAFLD), affect a large number of people worldwide and become one of the major causes of end-stage liver disease, such as liver cirrhosis and hepatocellular carcinoma (HCC). Unfortunately, there are currently no approved pharmacological treatments for ALD or NAFLD. This situation highlights the urgent need to explore new intervention targets and discover effective therapeutics for ALD and NAFLD. The lack of properly validated preclinical disease models is a major obstacle to the development of clinical therapies. ALD and NAFLD models have been in the development for decades, but there are still no models that recapitulate the full spectrum of ALD and NAFLD. Throughout this review, we summarize the current in vitro and in vivo models used for research on fatty liver diseases and discuss the advantages and limitations of these models.
Collapse
Affiliation(s)
- Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Haiyuan Shen
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Wonhyo Seo
- Laboratory of Hepatotoxicity, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Seonghwan Hwang
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
31
|
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
|
32
|
Andreu-Fernández V, Serra-Delgado M, Almeida-Toledano L, García-Meseguer À, Vieiros M, Ramos-Triguero A, Muñoz-Lozano C, Navarro-Tapia E, Martínez L, García-Algar Ó, Gómez-Roig MD. Effect of Postnatal Epigallocatechin-Gallate Treatment on Cardiac Function in Mice Prenatally Exposed to Alcohol. Antioxidants (Basel) 2023; 12:antiox12051067. [PMID: 37237934 DOI: 10.3390/antiox12051067] [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: 03/27/2023] [Revised: 04/24/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
Prenatal alcohol exposure affects the cardiovascular health of the offspring. Epigallocatechin-3-gallate (EGCG) may be a protective agent against it, but no data are available regarding its impact on cardiac dysfunction. We investigated the presence of cardiac alterations in mice prenatally exposed to alcohol and the effect of postnatal EGCG treatment on cardiac function and related biochemical pathways. C57BL/6J pregnant mice received 1.5 g/kg/day (Mediterranean pattern), 4.5 g/kg/day (binge pattern) of ethanol, or maltodextrin until Day 19 of pregnancy. Post-delivery, treatment groups received EGCG-supplemented water. At post-natal Day 60, functional echocardiographies were performed. Heart biomarkers of apoptosis, oxidative stress, and cardiac damage were analyzed by Western blot. BNP and Hif1α increased and Nrf2 decreased in mice prenatally exposed to the Mediterranean alcohol pattern. Bcl-2 was downregulated in the binge PAE drinking pattern. Troponin I, glutathione peroxidase, and Bax increased in both ethanol exposure patterns. Prenatal alcohol exposure led to cardiac dysfunction in exposed mice, evidenced by a reduced ejection fraction, left ventricle posterior wall thickness at diastole, and Tei index. EGCG postnatal therapy restored the physiological levels of these biomarkers and improved cardiac dysfunction. These findings suggest that postnatal EGCG treatment attenuates the cardiac damage caused by prenatal alcohol exposure in the offspring.
Collapse
Affiliation(s)
- Vicente Andreu-Fernández
- Grup de Recerca Infancia i Entorn (GRIE), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Biosanitary Research Institute, Valencian International University (VIU), 46002 Valencia, Spain
| | - Mariona Serra-Delgado
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- BCNatal, Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and Hospital Clínic, Universitat de Barcelona, 08950 Barcelona, Spain
| | - Laura Almeida-Toledano
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- BCNatal, Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and Hospital Clínic, Universitat de Barcelona, 08950 Barcelona, Spain
| | - Àgueda García-Meseguer
- Grup de Recerca Infancia i Entorn (GRIE), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, IDIBAPS, BCNatal, 08028 Barcelona, Spain
| | - Melina Vieiros
- Grup de Recerca Infancia i Entorn (GRIE), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, IDIBAPS, BCNatal, 08028 Barcelona, Spain
| | - Anna Ramos-Triguero
- Grup de Recerca Infancia i Entorn (GRIE), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, IDIBAPS, BCNatal, 08028 Barcelona, Spain
| | - Concha Muñoz-Lozano
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- BCNatal, Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and Hospital Clínic, Universitat de Barcelona, 08950 Barcelona, Spain
| | - Elisabet Navarro-Tapia
- Biosanitary Research Institute, Valencian International University (VIU), 46002 Valencia, Spain
| | - Leopoldo Martínez
- Department of Pediatric Surgery, Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Óscar García-Algar
- Grup de Recerca Infancia i Entorn (GRIE), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, IDIBAPS, BCNatal, 08028 Barcelona, Spain
| | - María D Gómez-Roig
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- BCNatal, Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and Hospital Clínic, Universitat de Barcelona, 08950 Barcelona, Spain
| |
Collapse
|
33
|
Jaber MA, Ghanim BY, Al-Natour M, Arqoub DA, Abdallah Q, Abdelrazig S, Alkrad JA, Kim DH, Qinna NA. Potential biomarkers and metabolomics of acetaminophen-induced liver injury during alcohol consumption: A preclinical investigation on C57/BL6 mice. Toxicol Appl Pharmacol 2023; 465:116451. [PMID: 36894070 DOI: 10.1016/j.taap.2023.116451] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023]
Abstract
The toxic effects of alcohol consumption on population health are significant worldwide and the synergistic toxic effects of concurrent intake of Acetaminophen and alcohol is of clinical concern. The understanding of molecular mechanisms beneath such synergism and acute toxicity may be enhanced through assessing underlying metabolomics changes. The molecular toxic activities of the model hereby, is assessed though metabolomics profile with a view to identifying metabolomics targets which could aid in the management of drug-alcohol interactions. In vivo exposure of C57/BL6 mice to APAP (70 mg/kg), single dose of ethanol (6 g/kg of 40%) and APAP after alcohol consumption was employed. Plasma samples were prepared and subjected to biphasic extraction for complete LC-MS profiling, and tandem mass MS2 analysis. Among the detected ions, 174 ions had significant (VIP scores >1 and FDR <0.05) changes between groups and were selected as potential biomarkers and significant variables. The presented metabolomics approach highlighted several affected metabolic pathways, including nucleotide and amino acid metabolism; aminoacyl-tRNA biosynthesis as well as bioenergetics of TCA and Krebs cycle. The impact of APAP on the concurrent administration of alcohol showed great biological interactions in the vital ATP and amino acid producing processes. The metabolomics changes show distinct metabolites which are altered to alcohol-APAP consumption while presenting several unneglectable risks on the vitality of metabolites and cellular molecules which shall be concerned.
Collapse
Affiliation(s)
- Malak A Jaber
- Department of Medicinal Chemistry and Analysis, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan
| | - Bayan Y Ghanim
- University of Petra Pharmaceutical Center (UPPC), Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan
| | - Mohammad Al-Natour
- Department of Pharmaceutical Technology, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan
| | - Duaa Abu Arqoub
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan
| | - Qasem Abdallah
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan
| | - Salah Abdelrazig
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Khartoum, Khartoum, Sudan
| | | | - Dong-Hyun Kim
- Centre for Analytical Bioscience, Advanced Material and Healthcare Technologies Division, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Nidal A Qinna
- University of Petra Pharmaceutical Center (UPPC), Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan; Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan.
| |
Collapse
|
34
|
Tien S, Zhou H, Zhou Q, Liu H, Wu B, Guo Y. PTTG1 alleviates acute alcoholic liver injury by inhibiting endoplasmic reticulum stress-induced hepatocyte pyroptosis. Liver Int 2023; 43:840-854. [PMID: 36737842 DOI: 10.1111/liv.15535] [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: 11/08/2022] [Revised: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND & AIMS Heavy drinking is a primary cause of alcoholic liver injury (ALI). Pituitary tumour transforming gene 1 (PTTG1) is involved in the occurrence and development of hepatocellular carcinoma (HCC), which is a well-known inflammation-related cancer with various aetiologies, including alcohol consumption. However, the role of PTTG1 in alcohol-induced liver injury and inflammation is not clear. METHODS Blood samples were collected from patients with acute alcohol intoxication (n = 20) and healthy controls (n = 20). PTTG1 knockout (KO) mice and PTTG1 transgenic (TG) mice were given a single gavage of alcohol (5 g/kg, 50%) to construct the alcohol-induced liver injury. RESULTS We found that serum PTTG1 levels were downregulated in acute ALI patients. In addition, acute alcohol administration significantly reduced PTTG1 levels in the serum and liver of mice. Compared to wild-type mice, PTTG1 KO mice had more serious liver injury, which was accompanied by worsened hepatic endoplasmic reticulum (ER) stress and hepatocyte pyroptosis induced by alcohol. Similarly, PTTG1 deficiency exacerbated alcohol-induced cell death in primary mouse hepatocytes and LO2 cells, by increasing hepatic ER stress and pyroptosis. Importantly, TUDCA, an ER stress inhibitor, could blocked alcohol-induced hepatic pyroptosis in PTTG1 knockdown LO2 cells. Finally, overexpression of PTTG1 substantially attenuated alcohol-induced liver injury by reducing ER stress and hepatic pyroptosis in mice. CONCLUSIONS We demonstrated that PTTG1 participates in ALI and has a protective effect against alcohol-induced hepatic ER stress and pyroptosis.
Collapse
Affiliation(s)
- Shiuan Tien
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, China
| | - Haoxiong Zhou
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, China
| | - Qi Zhou
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, China
| | - Huiling Liu
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, China
| | - Bin Wu
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, China
| | - Yunwei Guo
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, China
| |
Collapse
|
35
|
Ramadori P, Woitok MM, Estévez-Vázquez O, Benedé-Ubieto R, Leal-Lassalle H, Lamas-Paz A, Guo F, Fabre J, Otto J, Verwaayen A, Reissing J, Bruns T, Erschfeld S, Haas U, Paffen D, Nelson LJ, Vaquero J, Bañares R, Trautwein C, Cubero FJ, Liedtke C, Nevzorova YA. Lack of Cyclin E1 in hepatocytes aggravates ethanol-induced liver injury and hepatic steatosis in experimental murine model of acute and chronic alcohol-associated liver disease. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166646. [PMID: 36736843 DOI: 10.1016/j.bbadis.2023.166646] [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: 09/13/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND Cyclin E1 is the regulatory subunit of cyclin-dependent kinase 2 (Cdk2) and one of the central players in cell cycle progression. We recently showed its crucial role for initiation of liver fibrosis and hepatocarcinogenesis. In the present study, we investigated the role of Cyclin E1 in the development of alcohol-associated liver disease (ALD). METHODS Mice with constitutive (E1-/-), hepatocyte-specific (Cyclin E1Δhepa), or intestinal-epithelial-cell-specific (Cyclin E1ΔIEC) inactivation of Cyclin E1 and corresponding wild type littermate controls (WT) were administered either a Lieber-DeCarli ethanol diet (LDE) for 3 weeks or acute ethanol binges (6 g/kg) through oral gavage. Serum parameters of liver functionality were measured; hepatic tissues were collected for biochemical and histological analyses. RESULTS The administration of acute EtOH binge and chronic LDE diet to E1-/- mice enhanced hepatic steatosis, worsened liver damage and triggered body weight loss. Similarly, in the acute EtOH binge model, Cyclin E1Δhepa mice revealed a significantly worsened liver phenotype. In contrast, inactivation of Cyclin E1 only in intestinal epithelial cell (IECs)did not lead to any significant changes in comparison to WT mice after acute EtOH challenge. Remarkably, both acute and chronic EtOH administration in E1-/- animals resulted in increased levels of ADH and decreased expression of ALDH1/2. The additional application of a pan-Cdk inhibitor (S-CR8) further promoted liver damage in EtOH-treated WT mice. CONCLUSION Our data point to a novel unexpected role of Cyclin E1 in hepatocytes for alcohol metabolism, which seems to be independent of the canonical Cyclin E1/Cdk2 function as a cell cycle regulator.
Collapse
Affiliation(s)
- Pierluigi Ramadori
- Division of Chronic Inflammation and Cancer, German Cancer Research Center Heidelberg (DKFZ), Heidelberg, Germany
| | | | - Olga Estévez-Vázquez
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
| | - Raquel Benedé-Ubieto
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Department of Genetics, Physiology and Microbiology, Faculty of Biology, Complutense University, Madrid, Spain
| | - Hector Leal-Lassalle
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
| | - Arantza Lamas-Paz
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Feifei Guo
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Department of Obstetrics and Gynaecology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jeanne Fabre
- Polytech Angers, Département Génie Biologique et Santé, Angers, France
| | - Julia Otto
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany
| | - Anna Verwaayen
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany
| | - Johanna Reissing
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany
| | - Tony Bruns
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany
| | - Stephanie Erschfeld
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany
| | - Ute Haas
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany
| | - Daniela Paffen
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany
| | - Leonard J Nelson
- Institute for Bioengineering (IBioE), School of Engineering, The University of Edinburgh, Faraday Building, Edinburgh EH9 3 JL, UK
| | - Javier Vaquero
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain,; Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón, Madrid, Spain,; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Rafael Bañares
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain,; Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón, Madrid, Spain,; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain,; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Christian Liedtke
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany
| | - Yulia A Nevzorova
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany; Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain,; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.
| |
Collapse
|
36
|
Hong OK, Kim ES, Son JW, Kim SR, Yoo SJ, Kwon HS, Lee SS. Alcohol-induced increase in BMP levels promotes fatty liver disease in male prediabetic stage Otsuka Long-Evans Tokushima Fatty rats. J Cell Biochem 2023; 124:459-472. [PMID: 36791312 DOI: 10.1002/jcb.30385] [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: 05/20/2022] [Revised: 01/15/2023] [Accepted: 01/30/2023] [Indexed: 02/17/2023]
Abstract
Alcohol consumption exacerbates liver abnormalities in animal models, but whether it increases the severity of liver disease in early diabetic or prediabetic rats is unclear. To investigate the molecular mechanisms underlying alcohol-induced liver steatosis or hepatitis, we used a prediabetic animal model. Otsuka Long-Evans Tokushima Fatty (OLETF) and Long-Evans Tokushima Fatty (LETO) rats were pair-fed with an ethanol-containing liquid diet for 6 weeks. Compared with controls, OLETF and LETO rats displayed more pronounced liver steatosis and higher plasma levels of serum glutamic oxaloacetic transaminase (SGOT) and serum glutamate pyruvate transaminase (SPGT), indicating liver injury. Ethanol-fed LETO (Pd-L-E) rats showed mild liver steatosis and no inflammation compared with ethanol-fed OLETF (Pd-O-E) rats. Although precursor and active SREBP-1 levels in the liver of ethanol-fed OLETF rats significantly increased compared with control diet-fed OLETF rats (Pd-O-C), those of Pd-L-E rats did not. Bone morphogenetic protein (BMP) and TGF-β1 balance in Pd-O-E rats was significantly altered because BMP signaling was upregulated by inducing BMP2, BMP4, BMP7, BMP9, Smad1, and Smad4, whereas TGF-β1, Smad3, and Erk were downregulated. Activation of TGF-β/Smad signaling inhibited BMP2 and BMP9 expression and increased epithelial-mesenchymal transition (EMT) marker levels (Hepcidin, Snail, and Twist) in the liver of LETO rats. Livers of ethanol-fed OLETF rats showed increased levels of vimentin, FSP-1, α-SMA, MMP1, MMP13, and collagen III compared with rats of other groups, whereas EMT marker levels did not change. Thus, BMP exerted anti- and/or pro-fibrotic effects in ethanol-fed rats. Therefore, BMP and TGF-β, two key members of the TGF-β superfamily, play important but diverse roles in liver steatosis in young LETO and OLETF rats.
Collapse
Affiliation(s)
- Oak-Kee Hong
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Sook Kim
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jang-Won Son
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung-Rae Kim
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Soon Jib Yoo
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyuk-Sang Kwon
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seong-Su Lee
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
37
|
Thoen RU, Longo L, Leonhardt LC, Pereira MHM, Rampelotto PH, Cerski CTS, Álvares-da-Silva MR. Alcoholic liver disease and intestinal microbiota in an experimental model: Biochemical, inflammatory, and histologic parameters. Nutrition 2023; 106:111888. [PMID: 36436334 DOI: 10.1016/j.nut.2022.111888] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 10/03/2022] [Accepted: 10/11/2022] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Alcoholic liver disease (ALD) is the leading cause of alcohol-related deaths worldwide. Experimental ALD models are expensive and difficult to reproduce. A low-cost, reproducible ALD model was developed, and liver damage compared with the gut microbiota. The aims of this study were to develop an experimental model of ALD, through a high-fat diet, the chronic use of ethanol, and intragastric alcohol binge; and to evaluate the composition of the gut microbiota and its correlation with markers of inflammatory and liver disease progression in this model. METHODS Adult male Wistar rats were randomized (N = 24) to one of three groups: control (standard diet and water + 0.05% saccharin), ALC4 and ALC8 (sunflower seed, 10% ethanol + 0.05% saccharin for 4 and 8 wk, respectively). On the last day, ALC4/8 received alcoholic binge (5 g/kg). Clinical, nutritional, biochemical, inflammatory, pathologic, and gut microbiota data were analyzed. RESULTS ALC4/8 animals consumed more alcohol and lipids (P < 0.01) and less total energy, liquids, solids, carbohydrates, and proteins (P < 0.01), and gained less weight (P < 0.01) than controls. ALC8 had lower Lee index scores than controls and ALC4 (P < 0.01). Aminotransferases increased and albumin diminished in ALC4/8 but not in the control group (P < 0.03 for all). Glucose and aspartate transaminase/alanine aminotransaminase ratios were higher in the ALC8 rats than in the controls (P < 0.03). Cholesterol was higher in ALC4 and lower in ALC8 compared with controls (P < 0.03). Albumin and high-density lipoprotein cholesterol levels were lower in ALC8 (P < 0.03). Hepatic concentration of triacylglycerols was higher in ALC8 than in ALC4 and controls (P < 0.05). ALC4/8 presented microvesicular grade 2 and 3 steatosis, respectively, and macrovesicular grade 1. No change in the gene expression of inflammatory markers between groups was seen. ALC4/8 had lower fecal bacterial α-diversity and relative abundance of Firmicutes (P < 0.005) and greater Bacterioidetes (P < 0.0007) and Protobacteria (P < 0.001) than controls. Gut microbiota correlated with serum and liver lipids, steatosis, albumin, and aminotransferases (P < 0.01 for all). CONCLUSION The model induced nutritional, biochemical, histologic, and gut microbiota changes, and appears to be useful in the study of therapeutic targets.
Collapse
Affiliation(s)
- Rutiane Ullmann Thoen
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Experimental Hepatology and Gastroenterology Laboratory, Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Larisse Longo
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Experimental Hepatology and Gastroenterology Laboratory, Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Luiza Cecília Leonhardt
- Experimental Hepatology and Gastroenterology Laboratory, Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Matheus Henrique Mariano Pereira
- Experimental Hepatology and Gastroenterology Laboratory, Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Pabulo Henrique Rampelotto
- Experimental Hepatology and Gastroenterology Laboratory, Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Carlos Thadeu Schmidt Cerski
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Unit of Surgical Pathology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Mário Reis Álvares-da-Silva
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Experimental Hepatology and Gastroenterology Laboratory, Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Division of Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
| |
Collapse
|
38
|
Guo H, Yu X, Liu Y, Paik DT, Justesen JM, Chandy M, Jahng JWS, Zhang T, Wu W, Rwere F, Zhao SR, Pokhrel S, Shivnaraine RV, Mukherjee S, Simon DJ, Manhas A, Zhang A, Chen CH, Rivas MA, Gross ER, Mochly-Rosen D, Wu JC. SGLT2 inhibitor ameliorates endothelial dysfunction associated with the common ALDH2 alcohol flushing variant. Sci Transl Med 2023; 15:eabp9952. [PMID: 36696485 DOI: 10.1126/scitranslmed.abp9952] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The common aldehyde dehydrogenase 2 (ALDH2) alcohol flushing variant known as ALDH2*2 affects ∼8% of the world's population. Even in heterozygous carriers, this missense variant leads to a severe loss of ALDH2 enzymatic activity and has been linked to an increased risk of coronary artery disease (CAD). Endothelial cell (EC) dysfunction plays a determining role in all stages of CAD pathogenesis, including early-onset CAD. However, the contribution of ALDH2*2 to EC dysfunction and its relation to CAD are not fully understood. In a large genome-wide association study (GWAS) from Biobank Japan, ALDH2*2 was found to be one of the strongest single-nucleotide polymorphisms associated with CAD. Clinical assessment of endothelial function showed that human participants carrying ALDH2*2 exhibited impaired vasodilation after light alcohol drinking. Using human induced pluripotent stem cell-derived ECs (iPSC-ECs) and CRISPR-Cas9-corrected ALDH2*2 iPSC-ECs, we modeled ALDH2*2-induced EC dysfunction in vitro, demonstrating an increase in oxidative stress and inflammatory markers and a decrease in nitric oxide (NO) production and tube formation capacity, which was further exacerbated by ethanol exposure. We subsequently found that sodium-glucose cotransporter 2 inhibitors (SGLT2i) such as empagliflozin mitigated ALDH2*2-associated EC dysfunction. Studies in ALDH2*2 knock-in mice further demonstrated that empagliflozin attenuated ALDH2*2-mediated vascular dysfunction in vivo. Mechanistically, empagliflozin inhibited Na+/H+-exchanger 1 (NHE-1) and activated AKT kinase and endothelial NO synthase (eNOS) pathways to ameliorate ALDH2*2-induced EC dysfunction. Together, our results suggest that ALDH2*2 induces EC dysfunction and that SGLT2i may potentially be used as a preventative measure against CAD for ALDH2*2 carriers.
Collapse
Affiliation(s)
- Hongchao Guo
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xuan Yu
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yu Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David T Paik
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Johanne Marie Justesen
- Biomedical Data Science, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mark Chandy
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - James W S Jahng
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tiejun Zhang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Weijun Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Freeborn Rwere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shane Rui Zhao
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Suman Pokhrel
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | - Daniel J Simon
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amit Manhas
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Angela Zhang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Che-Hong Chen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Manuel A Rivas
- Biomedical Data Science, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Eric R Gross
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine (Division of Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
39
|
Zogona D, Zongo AWS, Elkhedir AE, Salah M, Tao M, Li R, Wu T, Xu X. Red raspberry supplementation mitigates alcohol-induced liver injury associated with gut microbiota alteration and intestinal barrier dysfunction in mice. Food Funct 2023; 14:1209-1226. [PMID: 36602148 DOI: 10.1039/d2fo03245g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alcoholic liver disease (ALD) is still a global health concern. Long-term alcohol intake alters the gut microbiota diversity and metabolic activity, and causes intestinal barrier dysfunction, leading to the development of ALD. This research explored the protective effects and underlying mechanisms of red raspberry (RR) on alcohol-related disorders in mice. Male C57BL/6J mice were fed a standard diet or a standard diet supplemented with 2%, 4%, and 8% weight/weight RR. Meanwhile, mice were administered 35% (v/v) ethanol (EtOH, 10 mL per kg body weight) intragastrically once daily for six weeks, except the control group mice. The results showed that RR supplementation decreased liver injury markers (alanine and aspartate transaminases) in the serum, reduced triglyceride level in the liver and downregulated hepatic cytochrome P450 2E1 mRNA expression in mice administered EtOH. In addition, EtOH-mediated oxidative stress in the liver was attenuated by RR supplementation through decreased hepatic malondialdehyde content and increased antioxidant (glutathione, glutathione peroxidase, and catalase) levels and activities in mice exposed to EtOH. Moreover, RR supplementation reversed EtOH-induced alteration in the cecal microbial composition at the phylum, order, genus, and species levels and improved the intestinal barrier function associated with the inhibition of the NF-κB/MLCK pathway, which was accompanied by upregulation of tight junctions (zonula occludens 1, occludin, claudin-1, and claudin-4) and E-cadherin mRNA and protein expressions. Accordingly, RR supplementation resulted in a decreased level of endotoxins in the serum and attenuation of the inflammatory response in the liver, illustrated by a significant decrease in tumor necrosis factor-alpha, interleukin (IL)-1β, and IL-6 levels. Overall, RR supplementation alleviated the adverse effects of chronic alcohol intake in C57BL/6J mice and could be a potential supplement for improving ALD.
Collapse
Affiliation(s)
- Daniel Zogona
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), Hubei Key Laboratory of Fruit & Vegetable Processing & Quality Control (Huazhong Agricultural University), College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China. .,Center for Research in Biological Sciences, Food and Nutrition, Department of Biochemistry and Microbiology, University Joseph Ki-Zerbo, BP 7021 Ouagadougou 03, Burkina Faso
| | - Abel Wend-Soo Zongo
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), Hubei Key Laboratory of Fruit & Vegetable Processing & Quality Control (Huazhong Agricultural University), College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China. .,Center for Research in Biological Sciences, Food and Nutrition, Department of Biochemistry and Microbiology, University Joseph Ki-Zerbo, BP 7021 Ouagadougou 03, Burkina Faso
| | - Abdeen E Elkhedir
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), Hubei Key Laboratory of Fruit & Vegetable Processing & Quality Control (Huazhong Agricultural University), College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| | - Mahmoud Salah
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), Hubei Key Laboratory of Fruit & Vegetable Processing & Quality Control (Huazhong Agricultural University), College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China. .,Department of Environmental Agricultural Science, Faculty of Graduate Studies and Environmental Research, Ain Shams University, Cairo 11566, Egypt
| | - Mingfang Tao
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), Hubei Key Laboratory of Fruit & Vegetable Processing & Quality Control (Huazhong Agricultural University), College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| | - Rong Li
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), Hubei Key Laboratory of Fruit & Vegetable Processing & Quality Control (Huazhong Agricultural University), College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| | - Ting Wu
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), Hubei Key Laboratory of Fruit & Vegetable Processing & Quality Control (Huazhong Agricultural University), College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| | - Xiaoyun Xu
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), Hubei Key Laboratory of Fruit & Vegetable Processing & Quality Control (Huazhong Agricultural University), College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
40
|
Gao J, Sun X, Zhou Q, Jiang S, Zhang Y, Ge H, Qin X. Circadian clock disruption aggravates alcohol liver disease in an acute mouse model. Chronobiol Int 2022; 39:1554-1566. [PMID: 36354126 DOI: 10.1080/07420528.2022.2132865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Circadian rhythms are important for organisms to adapt to the environment and maintain homeostasis. Disruptions of circadian rhythms contribute to the occurrence, progression, and exacerbation of diseases, such as cancer, psychiatric disorders, and metabolic disorders. Alcohol-induced liver disease (ALD) is one of the most prevalent liver diseases. Disruptions of the circadian clock enhance the ALD symptoms using chronic mice models or genetic manipulated mice. However, chronic models are time consuming and clock gene deletions interfere with metabolisms. Here, we report that constant light (LL) condition significantly disrupted the circadian clock in an acute ALD model, resulting in aggravated ALD phenotypes in wild type mice. Comparative transcriptome analysis revealed that the alcohol feeding affected the circadian pathway, as well as metabolic pathways. The acute alcohol feeding plus the LL condition further interfered with metabolic pathways and dysregulated canonical circadian gene expressions. These findings support the idea that disrupting the circadian clock could provide an improved ALD mouse model for further applications, such as facilitating identification of potential therapeutic targets for the prevention and treatment of ALD.Abbreviations: ALD, alcohol-induced liver disease; LD, 12 h light _ 12 h dark; LL, constant light; HF, high-fat liquid control diet; ETH, ethanol-containing diet; NIAAA, National Institute on Alcohol Abuse and Alcoholism; TTFLs, transcription-translation feedback loops; FDA, US Foods and Drug Administration; NAFLD, non-alcoholic fatty liver disease; RER, respiratory exchange rate; DEGs, differentially expressed genes; H&E, haematoxylin and eosin; ALT, alanine transaminase; AST, aspartate transaminase; TG, triglycerides.
Collapse
Affiliation(s)
- Jiajia Gao
- Institute of Health Sciences, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui Province, China
| | - Xianpu Sun
- Institute of Health Sciences, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui Province, China
| | - Qin Zhou
- Institute of Health Sciences, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui Province, China
| | - Shuo Jiang
- Institute of Health Sciences, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui Province, China
| | - Yunfei Zhang
- Modern Experiment Technology Center, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui Province, China
| | - Honghua Ge
- Institute of Health Sciences, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui Province, China
| | - Ximing Qin
- Institute of Health Sciences, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui Province, China
| |
Collapse
|
41
|
Mackowiak B, Xu M, Lin Y, Guan Y, Seo W, Ren R, Feng D, Jones JW, Wang H, Gao B. Hepatic CYP2B10 is highly induced by binge ethanol and contributes to acute-on-chronic alcohol-induced liver injury. Alcohol Clin Exp Res 2022; 46:2163-2176. [PMID: 36224745 PMCID: PMC9771974 DOI: 10.1111/acer.14954] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/10/2022] [Accepted: 10/05/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND The chronic-plus-binge model of ethanol consumption, where chronically (8-week) ethanol-fed mice are gavaged a single dose of ethanol (E8G1), is known to induce steatohepatitis in mice. However, how chronically ethanol-fed mice respond to multiple binges of ethanol remains unknown. METHODS We extended the E8G1 model to three gavages of ethanol (E8G3) spaced 24 h apart, sacrificed each group 9 h after the final gavage, analyzed liver injury, and examined gene expression changes using microarray analyses in each group to identify mechanisms contributing to liver responses to binge ethanol. RESULTS Surprisingly, E8G3 treatment induced lower levels of liver injury, steatosis, inflammation, and fibrosis as compared to mice after E8G1 treatment. Microarray analyses identified several pathways that may contribute to the reduced liver injury after E8G3 treatment compared to E8G1 treatment. The gene encoding cytochrome P450 2B10 (Cyp2b10) was one of the top upregulated genes in the E8G1 group and was further upregulated in the E8G3 group, but only moderately induced after chronic ethanol consumption, as confirmed by RT-qPCR and western blot analyses. Genetic disruption of Cyp2b10 worsened liver injury in E8G1 and E8G3 mice with higher blood ethanol levels compared to wild-type control mice, while in vitro experiments revealed that CYP2b10 did not directly promote ethanol metabolism. Metabolomic analyses revealed significant differences in hepatic metabolites from E8G1-treated Cyp2b10 knockout and WT mice, and these metabolic alterations may contribute to the reduced liver injury in Cyp2b10 knockout mice. CONCLUSION Hepatic Cyp2b10 expression is highly induced after ethanol binge, and such upregulation reduces acute-on-chronic ethanol-induced liver injury via the indirect modification of ethanol metabolism.
Collapse
Affiliation(s)
- Bryan Mackowiak
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mingjiang Xu
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yuhong Lin
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yukun Guan
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wonhyo Seo
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ruixue Ren
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dechun Feng
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jace W. Jones
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, USA
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, USA
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
42
|
Warner JB, Guenthner SC, Hardesty JE, McClain CJ, Warner DR, Kirpich IA. Liver-specific drug delivery platforms: Applications for the treatment of alcohol-associated liver disease. World J Gastroenterol 2022; 28:5280-5299. [PMID: 36185629 PMCID: PMC9521517 DOI: 10.3748/wjg.v28.i36.5280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/16/2022] [Accepted: 09/06/2022] [Indexed: 02/06/2023] Open
Abstract
Alcohol-associated liver disease (ALD) is a common chronic liver disease and major contributor to liver disease-related deaths worldwide. Despite its pre-valence, there are few effective pharmacological options for the severe stages of this disease. While much pre-clinical research attention is paid to drug development in ALD, many of these experimental therapeutics have limitations such as poor pharmacokinetics, poor efficacy, or off-target side effects due to systemic administration. One means of addressing these limitations is through liver-targeted drug delivery, which can be accomplished with different platforms including liposomes, polymeric nanoparticles, exosomes, bacteria, and adeno-associated viruses, among others. These platforms allow drugs to target the liver passively or actively, thereby reducing systemic circulation and increasing the ‘effective dose’ in the liver. While many studies, some clinical, have applied targeted delivery systems to other liver diseases such as viral hepatitis or hepatocellular carcinoma, only few have investigated their efficacy in ALD. This review provides basic information on these liver-targeting drug delivery platforms, including their benefits and limitations, and summarizes the current research efforts to apply them to the treatment of ALD in rodent models. We also discuss gaps in knowledge in the field, which when addressed, may help to increase the efficacy of novel therapies and better translate them to humans.
Collapse
Affiliation(s)
- Jeffrey Barr Warner
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, United States
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, United States
| | - Steven Corrigan Guenthner
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, United States
| | - Josiah Everett Hardesty
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, United States
| | - Craig James McClain
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, United States
- Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY 40202, United States
- Hepatobiology and Toxicology Center, University of Louisville School of Medicine, Louisville, KY 40202, United States
- Veterans Health Administration, Robley Rex Veterans Medical Center, Louisville, KY 40206, United States
| | - Dennis Ray Warner
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, United States
| | - Irina Andreyevna Kirpich
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, United States
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, United States
- Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY 40202, United States
- Hepatobiology and Toxicology Center, University of Louisville School of Medicine, Louisville, KY 40202, United States
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40202, United States
| |
Collapse
|
43
|
Moya M, Escudero B, Gómez-Blázquez E, Rebolledo-Poves AB, López-Gallardo M, Guerrero C, Marco EM, Orio L. Upregulation of TLR4/MyD88 pathway in alcohol-induced Wernicke’s encephalopathy: Findings in preclinical models and in a postmortem human case. Front Pharmacol 2022; 13:866574. [PMID: 36225571 PMCID: PMC9549119 DOI: 10.3389/fphar.2022.866574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Wernicke’s encephalopathy (WE) is a neurologic disease caused by vitamin B1 or thiamine deficiency (TD), being the alcohol use disorder its main risk factor. WE patients present limiting motor, cognitive, and emotional alterations related to a selective cerebral vulnerability. Neuroinflammation has been proposed to be one of the phenomena that contribute to brain damage. Our previous studies provide evidence for the involvement of the innate immune receptor Toll-like (TLR)4 in the inflammatory response induced in the frontal cortex and cerebellum in TD animal models (animals fed with TD diet [TDD] and receiving pyrithiamine). Nevertheless, the effects of the combination of chronic alcohol consumption and TD on TLR4 and their specific contribution to the pathogenesis of WE are currently unknown. In addition, no studies on TLR4 have been conducted on WE patients since brains from these patients are difficult to achieve. Here, we used rat models of chronic alcohol (CA; 9 months of forced consumption of 20% (w/v) alcohol), TD hit (TDD + daily 0.25 mg/kg i.p. pyrithiamine during 12 days), or combined treatment (CA + TDD) to check the activation of the proinflammatory TLR4/MyD88 pathway and related markers in the frontal cortex and the cerebellum. In addition, we characterized for the first time the TLR4 and its coreceptor MyD88 signature, along with other markers of this proinflammatory signaling such as phospo-NFκB p65 and IκBα, in the postmortem human frontal cortex and cerebellum (gray and white matter) of an alcohol-induced WE patient, comparing it with negative (no disease) and positive (aged brain with Alzheimer’s disease) control subjects for neuroinflammation. We found an increase in the cortical TLR4 and its adaptor molecule MyD88, together with an upregulation of the proinflammatory signaling molecules p-NF-ĸB and IĸBα in the CA + TDD animal model. In the patient diagnosed with alcohol-induced WE, we observed cortical and cerebellar upregulation of the TLR4/MyD88 pathway. Hence, our findings provide evidence, both in the animal model and the human postmortem brain, of the upregulation of the TLR4/MyD88 proinflammatory pathway in alcohol consumption–related WE.
Collapse
Affiliation(s)
- Marta Moya
- Department of Psychobiology and Methods in Behavioral Science, Faculty of Psychology, Complutense University of Madrid, Madrid, Spain
| | - Berta Escudero
- Department of Psychobiology and Methods in Behavioral Science, Faculty of Psychology, Complutense University of Madrid, Madrid, Spain
| | | | | | | | - Carmen Guerrero
- Biobanco of Hospital Universitario Fundación Alcorcón, Alcorcón, Spain
| | - Eva M. Marco
- Department of Genetics, Physiology and Microbiology, Faculty of Biology, Complutense University of Madrid, Madrid, Spain
| | - Laura Orio
- Department of Psychobiology and Methods in Behavioral Science, Faculty of Psychology, Complutense University of Madrid, Madrid, Spain
- Research Network in Primary Care in Addictions (Red de Investigación en Atención Primaria en Adicciones), Riapad, Spain
- *Correspondence: Laura Orio,
| |
Collapse
|
44
|
Brandão-Bezerra L, Aparecida da Rosa A, Figueiredo de Oliveira RM, Neves RH, Corrêa CL, Machado-Silva JR. Impact of acute schistosomiasis mansoni and long-term ethanol intake on mouse liver pathology. Exp Parasitol 2022; 242:108388. [PMID: 36174706 DOI: 10.1016/j.exppara.2022.108388] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 09/08/2022] [Accepted: 09/21/2022] [Indexed: 11/26/2022]
Abstract
While the effect of ethanol and schistosomiasis mansoni on liver injury has been well-documented, the influence of comorbidity on liver pathology remains unclear. To address this gap, schistosomiasis-infected mice were given one daily dose of 18% ethanol for 28 consecutive days, from day 35 post-infection. Mice were assigned to four groups: A. control; B. uninfected/ethanol gavage; C. infected; and D. infected/ethanol gavage. At day 64 post-infection, mice were euthanized by CO2 asphyxiation, livers were excised, fixed in 10% buffered formalin, paraffin embedded and cut into 5 μm sections. These were stained with hematoxylin and eosin (HE), Lennert's Giemsa and picrosirius red (for polarization microscopy) to assess histopathological and stereological changes. Group B showed alcoholic liver disease (ALD), including microsteatosis, hepatocyte karyopyknosis, karyorrhexis, karyolysis, increased frequency of Kupffer cells, hydropic degeneration of hepatocyte, thickened plasma membrane and binucleated hepatocytes. Infected mice showed typical exudative and exudative-productive hepatic granulomas, and destruction of the adjacent hepatic parenchyma, resulting in necrotic tissue and periovular leukocyte infiltrate. Group D showed hyperemia (parenchymal panlobular lesions), and liquefactive necrosis in hepatic abscess area. There was also reduced liver collagen deposition (-76%; p = 0.0001) and reduced microsteatosis (-80%, p = 0.0079) compared to group C and group B, respectively. In conclusion, comorbidity exacerbated liver damage.
Collapse
Affiliation(s)
- Luciana Brandão-Bezerra
- Romero Lascasas Porto Laboratory of Helminthology, Department of Microbiology, Immunology and Parasitology, Medical Sciences College (FCM), Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Aline Aparecida da Rosa
- Romero Lascasas Porto Laboratory of Helminthology, Department of Microbiology, Immunology and Parasitology, Medical Sciences College (FCM), Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Regina Maria Figueiredo de Oliveira
- Romero Lascasas Porto Laboratory of Helminthology, Department of Microbiology, Immunology and Parasitology, Medical Sciences College (FCM), Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Renata Heisler Neves
- Romero Lascasas Porto Laboratory of Helminthology, Department of Microbiology, Immunology and Parasitology, Medical Sciences College (FCM), Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Christiane Leal Corrêa
- Department of Pathology and Laboratories, Medical Sciences College (FCM), Rio de Janeiro State University (UERJ), Brazil. Medical College, Estácio de Sá University, Rio de Janeiro, Brazil
| | - José Roberto Machado-Silva
- Romero Lascasas Porto Laboratory of Helminthology, Department of Microbiology, Immunology and Parasitology, Medical Sciences College (FCM), Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil.
| |
Collapse
|
45
|
Zhu Q, Zhuo H, Yang L, Ouyang H, Chen J, Liu B, Huang H. A Peptide HEPFYGNEGALR from Apostichopus japonicus Alleviates Acute Alcoholic Liver Injury by Enhancing Antioxidant Response in Male C57BL/6J Mice. Molecules 2022; 27:molecules27185839. [PMID: 36144575 PMCID: PMC9503860 DOI: 10.3390/molecules27185839] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 12/08/2022] Open
Abstract
Liver-related disease caused by alcohol is a frequent disorder of the hepatic tract. Heavy consumption of alcohol in a short period causes oxidative damage to the liver. Sea cucumber is abundant in nutrients and its various extracts have been studied for antioxidant properties. One peptide was isolated and identified from Apostichopus japonicus in our recent study. We investigated the benefits of the peptide in a model of acute ethanol-induced male C57BL/6J mice. Dietary intake of the peptide could attenuate hepatomegaly, hepatitis and the accumulation of lipid droplets, and increase antioxidant enzyme activities in mice with acute alcoholic liver injury. The results indicated that a 20 mg/kg peptide supplement could activate the Nrf2/HO-1 pathway and block the nuclear translocation of NF-κB to alleviate oxidative stress and inflammation. In addition, the preventive effects of peptide supplementation may be related to autophagy. This study suggests that dietary supplementation with a sea cucumber-derived peptide is one of the potential candidates to alleviate acute alcoholic liver injury.
Collapse
Affiliation(s)
- Qiliang Zhu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Huiling Zhuo
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lamei Yang
- School of Nursing, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Haohong Ouyang
- School of Biosciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jun Chen
- School of Biosciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Bing Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510006, China
- Correspondence: (B.L.); (H.H.); Tel.: +86-135-8058-5585 (B.L.); +86-135-7090-8699 (H.H.)
| | - Hongliang Huang
- School of Biosciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Key Laboratory of New Drug Discovery and Evaluation, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Correspondence: (B.L.); (H.H.); Tel.: +86-135-8058-5585 (B.L.); +86-135-7090-8699 (H.H.)
| |
Collapse
|
46
|
Li DM, Wu YX, Hu ZQ, Wang TC, Zhang LL, Zhou Y, Tong X, Xu JY, Qin LQ. Lactoferrin Prevents Chronic Alcoholic Injury by Regulating Redox Balance and Lipid Metabolism in Female C57BL/6J Mice. Antioxidants (Basel) 2022; 11:antiox11081508. [PMID: 36009227 PMCID: PMC9405310 DOI: 10.3390/antiox11081508] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/19/2022] [Accepted: 07/29/2022] [Indexed: 02/01/2023] Open
Abstract
This study aimed to investigate the preventive effects of lactoferrin (Lf) on chronic alcoholic liver injury (ALI) in female mice. Female C57BL/6J mice were randomly divided into four groups: control group (CON), ethanol administration group (EtOH), low-dose Lf treatment group (LLf), and high-dose Lf group (HLf). In the last three groups, chronic ALI was induced by administering 20% ethanol ad libitum for 12 weeks. Mice in the CON and EtOH groups were fed with AIN-93G diet. Meanwhile, 0.4% and 4% casein in the AIN-93G diet were replaced by Lf as the diets of LLf and HLf groups, respectively. HLf significantly reduced hepatic triglyceride content and improved pathological morphology. HLf could inhibit cytochrome P450 2E1 overexpression and promote alcohol dehydrogenase-1 expression. HLf activated protein kinase B and AMP-activated protein kinase (AMPK), as well as upregulating nuclear-factor-erythroid-2-related factor-2 expression to elevate hepatic antioxidative enzyme activities. AMPK activation also benefited hepatic lipid metabolism. Meanwhile, HLf had no obvious beneficial effects on gut microbiota. In summary, Lf could alleviate chronic ALI in female mice, which was associated with redox balance and lipid metabolism regulation.
Collapse
Affiliation(s)
- De-Ming Li
- School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China; (D.-M.L.); (Y.-X.W.); (Z.-Q.H.); (T.-C.W.); (L.-L.Z.); (Y.Z.)
| | - Yun-Xuan Wu
- School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China; (D.-M.L.); (Y.-X.W.); (Z.-Q.H.); (T.-C.W.); (L.-L.Z.); (Y.Z.)
| | - Zhi-Qiang Hu
- School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China; (D.-M.L.); (Y.-X.W.); (Z.-Q.H.); (T.-C.W.); (L.-L.Z.); (Y.Z.)
| | - Tian-Ci Wang
- School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China; (D.-M.L.); (Y.-X.W.); (Z.-Q.H.); (T.-C.W.); (L.-L.Z.); (Y.Z.)
| | - Li-Li Zhang
- School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China; (D.-M.L.); (Y.-X.W.); (Z.-Q.H.); (T.-C.W.); (L.-L.Z.); (Y.Z.)
| | - Yan Zhou
- School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China; (D.-M.L.); (Y.-X.W.); (Z.-Q.H.); (T.-C.W.); (L.-L.Z.); (Y.Z.)
| | - Xing Tong
- Laboratory Center, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China;
| | - Jia-Ying Xu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China
- Correspondence: (J.-Y.X.); (L.-Q.Q.)
| | - Li-Qiang Qin
- School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China; (D.-M.L.); (Y.-X.W.); (Z.-Q.H.); (T.-C.W.); (L.-L.Z.); (Y.Z.)
- Correspondence: (J.-Y.X.); (L.-Q.Q.)
| |
Collapse
|
47
|
Zhu L, Li HD, Xu JJ, Li JJ, Cheng M, Meng XM, Huang C, Li J. Advancements in the Alcohol-Associated Liver Disease Model. Biomolecules 2022; 12:biom12081035. [PMID: 36008929 PMCID: PMC9406170 DOI: 10.3390/biom12081035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023] Open
Abstract
Alcohol-associated liver disease (ALD) is an intricate disease that results in a broad spectrum of liver damage. The presentation of ALD can include simple steatosis, steatohepatitis, liver fibrosis, cirrhosis, and even hepatocellular carcinoma (HCC). Effective prevention and treatment strategies are urgently required for ALD patients. In previous decades, numerous rodent models were established to investigate the mechanisms of alcohol-associated liver disease and explore therapeutic targets. This review provides a summary of the latest developments in rodent models, including those that involve EtOH administration, which will help us to understand the characteristics and causes of ALD at different stages. In addition, we discuss the pathogenesis of ALD and summarize the existing in vitro models. We analyse the pros and cons of these models and their translational relevance and summarize the insights that have been gained regarding the mechanisms of alcoholic liver injury.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiao-Ming Meng
- Correspondence: (X.-M.M.); (C.H.); (J.L.); Tel.: +86-551-65161001 (J.L.); Fax: +86-551-65161001 (J.L.)
| | - Cheng Huang
- Correspondence: (X.-M.M.); (C.H.); (J.L.); Tel.: +86-551-65161001 (J.L.); Fax: +86-551-65161001 (J.L.)
| | - Jun Li
- Correspondence: (X.-M.M.); (C.H.); (J.L.); Tel.: +86-551-65161001 (J.L.); Fax: +86-551-65161001 (J.L.)
| |
Collapse
|
48
|
Guo L, Guan Q, Duan W, Ren Y, Zhang XJ, Xu HY, Shi JS, Wang FZ, Lu R, Zhang HL, Xu ZH, Li H, Geng Y. Dietary Goji Shapes the Gut Microbiota to Prevent the Liver Injury Induced by Acute Alcohol Intake. Front Nutr 2022; 9:929776. [PMID: 35898713 PMCID: PMC9309278 DOI: 10.3389/fnut.2022.929776] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/08/2022] [Indexed: 11/25/2022] Open
Abstract
Diet is a major driver of the structure and function of the gut microbiota, which influences the host physiology. Alcohol abuse can induce liver disease and gut microbiota dysbiosis. Here, we aim to elucidate whether the well-known traditional health food Goji berry targets gut microbiota to prevent liver injury induced by acute alcohol intake. The results showed that Goji supplementation for 14 days alleviated acute liver injury as indicated by lowering serum aspartate aminotransferase, alanine aminotransferase, pro-inflammatory cytokines, as well as lipopolysaccharide content in the liver tissue. Goji maintained the integrity of the epithelial barrier and increased the levels of butyric acid in cecum contents. Furthermore, we established the causal relationship between gut microbiota and liver protection effects of Goji with the help of antibiotics treatment and fecal microbiota transplantation (FMT) experiments. Both Goji and FMT-Goji increased glutathione (GSH) in the liver and selectively enriched the butyric acid-producing gut bacterium Akkermansia and Ruminococcaceae by using 16S rRNA gene sequencing. Metabolomics analysis of cecum samples revealed that Goji and its trained microbiota could regulate retinoyl β-glucuronide, vanillic acid, and increase the level of glutamate and pyroglutamic acid, which are involved in GSH metabolism. Our study highlights the communication among Goji, gut microbiota, and liver homeostasis.
Collapse
Affiliation(s)
- Lin Guo
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Qijie Guan
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
- Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi, China
| | - Wenhui Duan
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Yilin Ren
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiao-Juan Zhang
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
- Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi, China
| | - Hong-Yu Xu
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
- Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi, China
| | - Jin-Song Shi
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | | | - Ran Lu
- Ningxia Red Power Goji Co., Ltd, Zhongwei, China
| | - Hui-Ling Zhang
- Ningxia Key Laboratory for Food Microbial-Applications Technology and Safety Control, Ningxia University, Yinchuan, China
| | - Zheng-Hong Xu
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
- Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi, China
| | - Huazhong Li
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- *Correspondence: Huazhong Li
| | - Yan Geng
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
- Yan Geng
| |
Collapse
|
49
|
Solopov PA, Colunga Biancatelli RML, Catravas JD. Alcohol Increases Lung Angiotensin-Converting Enzyme 2 Expression and Exacerbates Severe Acute Respiratory Syndrome Coronavirus 2 Spike Protein Subunit 1-Induced Acute Lung Injury in K18-hACE2 Transgenic Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:990-1000. [PMID: 35483427 PMCID: PMC9040477 DOI: 10.1016/j.ajpath.2022.03.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/14/2022] [Accepted: 03/31/2022] [Indexed: 12/18/2022]
Abstract
During the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic, alcohol consumption increased markedly. Nearly one in four adults reported drinking more alcohol to cope with stress. Chronic alcohol abuse is now recognized as a factor complicating the course of acute respiratory distress syndrome and increasing mortality. To investigate the mechanisms behind this interaction, a combined acute respiratory distress syndrome and chronic alcohol abuse mouse model was developed by intratracheally instilling the subunit 1 (S1) of SARS-CoV-2 spike protein (S1SP) in K18-human angiotensin-converting enzyme 2 (ACE2) transgenic mice that express the human ACE2 receptor for SARS-CoV-2 and were kept on an ethanol diet. Seventy-two hours after S1SP instillation, mice on an ethanol diet showed a strong decrease in body weight, a dramatic increase in white blood cell content of bronchoalveolar lavage fluid, and an augmented cytokine storm, compared with S1SP-treated mice on a control diet. Histologic examination of lung tissue showed abnormal recruitment of immune cells in the alveolar space, abnormal parenchymal architecture, and worsening Ashcroft score in S1SP- and alcohol-treated animals. Along with the activation of proinflammatory biomarkers [NF-κB, STAT3, NLR family pyrin domain-containing protein 3 (NLRP3) inflammasome], lung tissue homogenates from mice on an alcohol diet showed overexpression of ACE2 compared with mice on a control diet. This model could be useful for the development of therapeutic approaches against alcohol-exacerbated coronavirus disease 2019.
Collapse
Affiliation(s)
- Pavel A Solopov
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia.
| | | | - John D Catravas
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia; School of Medical Diagnostic and Translational Sciences, College of Health Sciences, Old Dominion University, Norfolk, Virginia
| |
Collapse
|
50
|
McGinnis CD, Jennings EQ, Harris PS, Galligan JJ, Fritz KS. Biochemical Mechanisms of Sirtuin-Directed Protein Acylation in Hepatic Pathologies of Mitochondrial Dysfunction. Cells 2022; 11:cells11132045. [PMID: 35805129 PMCID: PMC9266223 DOI: 10.3390/cells11132045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial protein acetylation is associated with a host of diseases including cancer, Alzheimer’s, and metabolic syndrome. Deciphering the mechanisms regarding how protein acetylation contributes to disease pathologies remains difficult due to the complex diversity of pathways targeted by lysine acetylation. Specifically, protein acetylation is thought to direct feedback from metabolism, whereby nutritional status influences mitochondrial pathways including beta-oxidation, the citric acid cycle, and the electron transport chain. Acetylation provides a crucial connection between hepatic metabolism and mitochondrial function. Dysregulation of protein acetylation throughout the cell can alter mitochondrial function and is associated with numerous liver diseases, including non-alcoholic and alcoholic fatty liver disease, steatohepatitis, and hepatocellular carcinoma. This review introduces biochemical mechanisms of protein acetylation in the regulation of mitochondrial function and hepatic diseases and offers a viewpoint on the potential for targeted therapies.
Collapse
Affiliation(s)
- Courtney D. McGinnis
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.D.M.); (P.S.H.)
| | - Erin Q. Jennings
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA; (E.Q.J.); (J.J.G.)
| | - Peter S. Harris
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.D.M.); (P.S.H.)
| | - James J. Galligan
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA; (E.Q.J.); (J.J.G.)
| | - Kristofer S. Fritz
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.D.M.); (P.S.H.)
- Correspondence:
| |
Collapse
|