1
|
Shen H, Liangpunsakul S, Iwakiri Y, Szabo G, Wang H. Immunological mechanisms and emerging therapeutic targets in alcohol-associated liver disease. Cell Mol Immunol 2025:10.1038/s41423-025-01291-w. [PMID: 40399593 DOI: 10.1038/s41423-025-01291-w] [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: 03/09/2025] [Accepted: 04/19/2025] [Indexed: 05/23/2025] Open
Abstract
Alcohol-associated liver disease (ALD) is a major global health challenge, with inflammation playing a central role in its progression. As inflammation emerges as a critical therapeutic target, ongoing research aims to unravel its underlying mechanisms. This review explores the immunological pathways of ALD, highlighting the roles of immune cells and their inflammatory mediators in disease onset and progression. We also examine the complex interactions between inflammatory cells and non-parenchymal liver cells, as well as their crosstalk with extra-hepatic organs, including the gut, adipose tissue, and nervous system. Furthermore, we summarize current clinical research on anti-inflammatory therapies and discuss promising therapeutic targets. Given the heterogeneity of ALD-associated inflammation, we emphasize the need for precision medicine to optimize treatment strategies and improve patient outcomes.
Collapse
Affiliation(s)
- Haiyuan Shen
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| | - Yasuko Iwakiri
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Gyongyi Szabo
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China.
| |
Collapse
|
2
|
Shi W, Chen D, Chen W, Chen Y, Fu X, Xiao Y, Duan W, Zhang J. Relationship of thyroid parameters with chronic inflammation in patients with euthyroid type 2 diabetes. Front Endocrinol (Lausanne) 2024; 15:1433782. [PMID: 39525855 PMCID: PMC11543494 DOI: 10.3389/fendo.2024.1433782] [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: 05/16/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Aims We evaluated the association of multiple thyroid parameters with the levels interleukin 6 (IL6) and interleukin 8 (IL8) in patients with euthyroid type 2 diabetes (T2D). Methods A total of 166 adults with euthyroid T2D were examined. Serum IL6, IL8, triiodothyronine (FT3), free thyroxin (FT4), thyroid-stimulating hormone (TSH), five thyroid sensitivity indexes (FT3/FT4, TSH index [TSHI], thyrotroph T4 resistance index [TT4RI], thyroid feedback quantile-based index by FT3 [TFQIFT3], and TFQIFT4), and biochemical parameters were determined. Results The median age was 64 years (IQR: 54.75,70) and the median duration of diabetes was 10 years (IQR: 3,18). Patients with high levels of IL6 (≥2.38 pg/mL) had lower levels of FT3 and TFQIFT3 (P<0.05). Patients with high levels of IL8 (≥18.1 pg/mL) had lower levels of TSH and higher levels of FT4 (P<0.05). IL6 was negatively correlated with FT3 (r=-0.359, P<0.001), TFQIFT3 (r=-0.273, P=0.009), and FT3/FT4 (r=-0.22, P=0.037). IL8 was negatively correlated with TSH (r=-0.256, P=0.01), TSHI (r=-0.226, P=0.033), and TT4RI (r=-0.244, P=0.021). Binary logistic regression analysis with multiple adjusted models showed that a high level of IL6 was negatively associated with FT3 (aOR: 0.529; 95%CI: 0.302, 0.926, P=0.026), and that a high level of IL8 was negatively associated with TSH (aOR: 0.343; 95%CI: 0.155, 0.759; P=0.008) and TT4RI (aOR: 0.398;95%CI: 0.191, 0.831; P=0.014). Conclusion Patients with euthyroid T2D who had lower levels of FT3 had significantly higher levels of IL6, and those with lower levels of TSH and TT4RI had significantly higher levels of IL8.
Collapse
Affiliation(s)
- Wei Shi
- Department of Endocrinology, Hubei Integrated Traditional Chinese and Western Medicine Hospital, Hubei University of Chinese Medicine, Wuhan, China
| | | | | | | | | | | | | | - Jing Zhang
- Department of Endocrinology, Hubei Integrated Traditional Chinese and Western Medicine Hospital, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
3
|
Feng D, Hwang S, Guillot A, Wang Y, Guan Y, Chen C, Maccioni L, Gao B. Inflammation in Alcohol-Associated Hepatitis: Pathogenesis and Therapeutic Targets. Cell Mol Gastroenterol Hepatol 2024; 18:101352. [PMID: 38697358 PMCID: PMC11234022 DOI: 10.1016/j.jcmgh.2024.04.009] [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: 01/09/2024] [Revised: 04/25/2024] [Accepted: 04/25/2024] [Indexed: 05/05/2024]
Abstract
Alcohol-associated hepatitis (AH) is an acute-on-chronic liver injury that occurs in patients with chronic alcohol-associated liver disease (ALD). Patients with severe AH have high short-term mortality and lack effective pharmacologic therapies. Inflammation is believed to be one of the key factors promoting AH progression and has been actively investigated as therapeutic targets over the last several decades, but no effective inflammatory targets have been identified so far. In this review, we discuss how inflammatory cells and the inflammatory mediators produced by these cells contribute to the development and progression of AH, with focus on neutrophils and macrophages. The crosstalk between inflammatory cells and liver nonparenchymal cells in the pathogenesis of AH is elaborated. We also deliberate the application of recent cutting-edge technologies in characterizing liver inflammation in AH. Finally, the potential therapeutic targets of inflammatory mediators for AH are briefly summarized.
Collapse
Affiliation(s)
- Dechun Feng
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland.
| | - Seonghwan Hwang
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Adrien Guillot
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Yang Wang
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Yukun Guan
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Cheng Chen
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Luca Maccioni
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
4
|
Martinelli G, Fumagalli M, Piazza S, Maranta N, Genova F, Sperandeo P, Sangiovanni E, Polissi A, Dell’Agli M, De Fabiani E. Investigating the Molecular Mechanisms Underlying Early Response to Inflammation and Helicobacter pylori Infection in Human Gastric Epithelial Cells. Int J Mol Sci 2023; 24:15147. [PMID: 37894827 PMCID: PMC10607124 DOI: 10.3390/ijms242015147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Helicobacter pylori is a leading cause of chronic gastric inflammation, generally associated with gastritis and adenocarcinoma. Activation of the NF-κB pathway mainly contributes to the inflammatory phenotype observed in H. pylori infection in humans and experimental models. Since the gastric epithelium undergoes rapid turnover, inflammation and pathogenicity of H. pylori result from early phase and chronically activated pathways. In the present study we investigated the early host response to H. pylori in non-tumoral human gastric epithelial cells (GES-1). To dissect the pathogen-specific mechanisms we also examined the response to tumor necrosis factor (TNF), a prototypical cytokine. By analyzing the activation state of NF-κB signaling, cytokine expression and secretion, and the transcriptome, we found that the inflammatory response of GES-1 cells to H. pylori and TNF results from activation of multiple pathways and transcription factors, e.g., NF-κB and CCAAT/enhancer-binding proteins (CEBPs). By comparing the transcriptomic profiles, we found that H. pylori infection induces a less potent inflammatory response than TNF but affects gene transcription to a greater extent by specifically inducing transcription factors such as CEBPβ and numerous zinc finger proteins. Our study provides insights on the cellular pathways modulated by H. pylori in non-tumoral human gastric cells unveiling new potential targets.
Collapse
Affiliation(s)
| | | | - Stefano Piazza
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, University of Milan, 20133 Milan, Italy; (G.M.); (M.F.); (N.M.); (F.G.); (P.S.); (E.S.); (A.P.); (M.D.); (E.D.F.)
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Niemelä O, Bloigu A, Bloigu R, Nivukoski U, Kultti J, Pohjasniemi H. Patterns of IgA Autoantibody Generation, Inflammatory Responses and Extracellular Matrix Metabolism in Patients with Alcohol Use Disorder. Int J Mol Sci 2023; 24:13124. [PMID: 37685930 PMCID: PMC10487441 DOI: 10.3390/ijms241713124] [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: 07/12/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Recent data have emphasized the role of inflammation and intestinal immunoglobulin A (IgA) responses in the pathogenesis of alcoholic liver disease (ALD). In order to further explore such associations, we compared IgA titers against antigens targeted to ethanol metabolites and tissue transglutaminase with pro- and anti-inflammatory mediators of inflammation, markers of liver status, transferrin protein desialylation and extracellular matrix metabolism in alcohol-dependent patients with or without liver disease and in healthy controls. Serum IgAs against protein adducts with acetaldehyde (HbAch-IgA), the first metabolite of ethanol, and tissue transglutaminase (tTG-IgA), desialylated transferrin (CDT), pro- and anti-inflammatory cytokines, markers of liver status (GT, ALP) and extracellular matrix metabolism (PIIINP, PINP, hyaluronic acid, ICTP and CTx) were measured in alcohol-dependent patients with (n = 83) or without (n = 105) liver disease and 88 healthy controls representing either moderate drinkers or abstainers. In ALD patients, both tTG-IgA and HbAch-IgA titers were significantly higher than those in the alcoholics without liver disease (p < 0.0005 for tTG-IgA, p = 0.006 for Hb-Ach-IgA) or in healthy controls (p < 0.0005 for both comparisons). The HbAch-IgA levels in the alcoholics without liver disease also exceeded those found in healthy controls (p = 0.0008). In ROC analyses, anti-tTG-antibodies showed an excellent discriminative value in differentiating between ALD patients and healthy controls (AUC = 0.95, p < 0.0005). Significant correlations emerged between tTG-IgAs and HbAch-IgAs (rs = 0.462, p < 0.0005), CDT (rs = 0.413, p < 0.0001), GT (rs = 0.487, p < 0.0001), alkaline phosphatase (rs = 0.466, p < 0.0001), serum markers of fibrogenesis: PIIINP (rs = 0.634, p < 0.0001), hyaluronic acid (rs = 0.575, p < 0.0001), ICTP (rs = 0.482, p < 0.0001), pro-inflammatory cytokines IL-6 (rs = 0.581, p < 0.0001), IL-8 (rs = 0.535, p < 0.0001) and TNF-α (rs = 0.591, p < 0.0001), whereas significant inverse correlations were observed with serum TGF-β (rs = -0.366, p < 0.0001) and CTx, a marker of collagen degradation (rs = -0.495, p < 0.0001). The data indicate that the induction of IgA immune responses toward ethanol metabolites and tissue transglutaminaseis a characteristic feature of patients with AUD and coincides with the activation of inflammation, extracellular matrix remodeling and the generation of aberrantly glycosylated proteins. These processes appear to work in concert in the sequence of events leading from heavy drinking to ALD.
Collapse
Affiliation(s)
- Onni Niemelä
- Department of Laboratory Medicine and Medical Research Unit, Seinäjoki Central Hospital, 60220 Seinäjoki, Finland; (U.N.); (J.K.); (H.P.)
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
| | - Aini Bloigu
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland;
| | - Risto Bloigu
- Infrastructure of Population Studies, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland;
| | - Ulla Nivukoski
- Department of Laboratory Medicine and Medical Research Unit, Seinäjoki Central Hospital, 60220 Seinäjoki, Finland; (U.N.); (J.K.); (H.P.)
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
| | - Johanna Kultti
- Department of Laboratory Medicine and Medical Research Unit, Seinäjoki Central Hospital, 60220 Seinäjoki, Finland; (U.N.); (J.K.); (H.P.)
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
| | - Heidi Pohjasniemi
- Department of Laboratory Medicine and Medical Research Unit, Seinäjoki Central Hospital, 60220 Seinäjoki, Finland; (U.N.); (J.K.); (H.P.)
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
| |
Collapse
|
6
|
Abstract
The medical disorders of alcoholism rank among the leading public health problems worldwide and the need for predictive and prognostic risk markers for assessing alcohol use disorders (AUD) has been widely acknowledged. Early-phase detection of problem drinking and associated tissue toxicity are important prerequisites for timely initiations of appropriate treatments and improving patient's committing to the objective of reducing drinking. Recent advances in clinical chemistry have provided novel approaches for a specific detection of heavy drinking through assays of unique ethanol metabolites, phosphatidylethanol (PEth) or ethyl glucuronide (EtG). Carbohydrate-deficient transferrin (CDT) measurements can be used to indicate severe alcohol problems. Hazardous drinking frequently manifests as heavy episodic drinking or in combinations with other unfavorable lifestyle factors, such as smoking, physical inactivity, poor diet or adiposity, which aggravate the metabolic consequences of alcohol intake in a supra-additive manner. Such interactions are also reflected in multiple disease outcomes and distinct abnormalities in biomarkers of liver function, inflammation and oxidative stress. Use of predictive biomarkers either alone or as part of specifically designed biological algorithms helps to predict both hepatic and extrahepatic morbidity in individuals with such risk factors. Novel approaches for assessing progression of fibrosis, a major determinant of prognosis in AUD, have also been made available. Predictive algorithms based on the combined use of biomarkers and clinical observations may prove to have a major impact on clinical decisions to detect AUD in early pre-symptomatic stages, stratify patients according to their substantially different disease risks and predict individual responses to treatment.
Collapse
Affiliation(s)
- Onni Niemelä
- Department of Laboratory Medicine and Medical Research Unit, Seinäjoki Central Hospital and Tampere University, Seinäjoki, Finland.
| |
Collapse
|
7
|
Li N, Liu H, Xue Y, Xu Z, Miao X, Guo Y, Li Z, Fan Z, Xu Y. Targetable Brg1-CXCL14 axis contributes to alcoholic liver injury by driving neutrophil trafficking. EMBO Mol Med 2023; 15:e16592. [PMID: 36722664 PMCID: PMC9994483 DOI: 10.15252/emmm.202216592] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 02/02/2023] Open
Abstract
Alcoholic liver disease (ALD) accounts for a large fraction of patients with cirrhosis and hepatocellular carcinoma. In the present study we investigated the involvement of Brahma-related gene 1 (Brg1) in ALD pathogenesis and implication in ALD intervention. We report that Brg1 expression was elevated in mouse models of ALD, in hepatocyte exposed to alcohol, and in human ALD specimens. Manipulation of Brg1 expression in hepatocytes influenced the development of ALD in mice. Flow cytometry showed that Brg1 deficiency specifically attenuated hepatic infiltration of Ly6G+ neutrophils in the ALD mice. RNA-seq identified C-X-C motif chemokine ligand 14 (CXCL14) as a potential target for Brg1. CXCL14 knockdown alleviated whereas CXCL14 over-expression enhanced ALD pathogenesis in mice. Importantly, pharmaceutical inhibition of Brg1 with a small-molecule compound PFI-3 or administration of an antagonist to the CXCL14 receptor ameliorated ALD pathogenesis in mice. Finally, a positive correlation between Brg1 expression, CXCL14 expression, and neutrophil infiltration was detected in ALD patients. In conclusion, our data provide proof-of-concept for targeting the Brg1-CXCL14 axis in ALD intervention.
Collapse
Affiliation(s)
- Nan Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of PathophysiologyNanjing Medical UniversityNanjingChina
| | - Hong Liu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of PathophysiologyNanjing Medical UniversityNanjingChina
| | - Yujia Xue
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of PathophysiologyNanjing Medical UniversityNanjingChina
| | - Zheng Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of PathophysiologyNanjing Medical UniversityNanjingChina
| | - Xiulian Miao
- Collage of Life Sciences and Institute of Biomedical Research, Liaocheng UniversityLiaochengChina
| | - Yan Guo
- Collage of Life Sciences and Institute of Biomedical Research, Liaocheng UniversityLiaochengChina
| | - Zilong Li
- State Key Laboratory of Natural Medicines, Department of PharmacologyChina Pharmaceutical UniversityNanjingChina
| | - Zhiwen Fan
- Department of PathologyNanjing Drum Tower Hospital Affiliated to Nanjing University Medical SchoolNanjingChina
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of PathophysiologyNanjing Medical UniversityNanjingChina
- Collage of Life Sciences and Institute of Biomedical Research, Liaocheng UniversityLiaochengChina
- State Key Laboratory of Natural Medicines, Department of PharmacologyChina Pharmaceutical UniversityNanjingChina
| |
Collapse
|
8
|
Drug Screening for Hepatitis A Virus (HAV): Nicotinamide Inhibits c-Jun Expression and HAV Replication. J Virol 2023; 97:e0198722. [PMID: 36728416 PMCID: PMC9973044 DOI: 10.1128/jvi.01987-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Hepatitis A virus (HAV) infection often causes acute hepatitis, which results in a case fatality rate of 0.2% and fulminant hepatitis in 0.5% of cases. However, no specific potent anti-HAV drug is available on the market to date. In the present study, we focused on inhibition of HAV internal ribosomal entry site (IRES)-mediated translation and investigated novel therapeutic drugs through drug repurposing by screening for inhibitors of HAV IRES-mediated translation and cell viability using a reporter assay and cell viability assay, respectively. The initial screening of 1,158 drugs resulted in 77 candidate drugs. Among them, nicotinamide significantly inhibited HAV HA11-1299 genotype IIIA replication in Huh7 cells. This promising drug also inhibited HAV HM175 genotype IB subgenomic replicon and HAV HA11-1299 genotype IIIA replication in a dose-dependent manner. In the present study, we found that nicotinamide inhibited the activation of activator protein 1 (AP-1) and that knockdown of c-Jun, which is one of the components of AP-1, inhibited HAV HM175 genotype IB IRES-mediated translation and HAV HA11-1299 genotype IIIA and HAV HM175 genotype IB replication. Taken together, the results showed that nicotinamide inhibited c-Jun, resulting in the suppression of HAV IRES-mediated translation and HAV replication, and therefore, it could be useful for the treatment of HAV infection. IMPORTANCE Drug screening methods targeting HAV IRES-mediated translation with reporter assays are attractive and useful for drug repurposing. Nicotinamide (vitamin B3, niacin) has been shown to effectively inhibit HAV replication. Transcription complex activator protein 1 (AP-1) plays an important role in the transcriptional regulation of cellular immunity or viral replication. The results of this study provide evidence that AP-1 is involved in HAV replication and plays a role in the HAV life cycle. In addition, nicotinamide was shown to suppress HAV replication partly by inhibiting AP-1 activity and HAV IRES-mediated translation. Nicotinamide may be useful for the control of acute HAV infection by inhibiting cellular AP-1 activity during HAV infection processes.
Collapse
|
9
|
Rohbeck E, Niersmann C, Köhrer K, Wachtmeister T, Roden M, Eckel J, Romacho T. Positive allosteric GABA A receptor modulation counteracts lipotoxicity-induced gene expression changes in hepatocytes in vitro. Front Physiol 2023; 14:1106075. [PMID: 36860523 PMCID: PMC9968943 DOI: 10.3389/fphys.2023.1106075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Introduction: We have previously shown that the novel positive allosteric modulator of the GABAA receptor, HK4, exerts hepatoprotective effects against lipotoxicity-induced apoptosis, DNA damage, inflammation and ER stress in vitro. This might be mediated by downregulated phosphorylation of the transcription factors NF-κB and STAT3. The current study aimed to investigate the effect of HK4 on lipotoxicity-induced hepatocyte injury at the transcriptional level. Methods: HepG2 cells were treated with palmitate (200 μM) in the presence or absence of HK4 (10 μM) for 7 h. Total RNA was isolated and the expression profiles of mRNAs were assessed. Differentially expressed genes were identified and subjected to the DAVID database and Ingenuity Pathway Analysis software for functional and pathway analysis, all under appropriate statistical testing. Results: Transcriptomic analysis showed substantial modifications in gene expression in response to palmitate as lipotoxic stimulus with 1,457 differentially expressed genes affecting lipid metabolism, oxidative phosphorylation, apoptosis, oxidative and ER stress among others. HK4 preincubation resulted in the prevention of palmitate-induced dysregulation by restoring initial gene expression pattern of untreated hepatocytes comprising 456 genes. Out of the 456 genes, 342 genes were upregulated and 114 downregulated by HK4. Enriched pathways analysis of those genes by Ingenuity Pathway Analysis, pointed towards oxidative phosphorylation, mitochondrial dysregulation, protein ubiquitination, apoptosis, and cell cycle regulation as affected pathways. These pathways are regulated by the key upstream regulators TP53, KDM5B, DDX5, CAB39 L and SYVN1, which orchestrate the metabolic and oxidative stress responses including modulation of DNA repair and degradation of ER stress-induced misfolded proteins in the presence or absence of HK4. Discussion: We conclude that HK4 specifically targets mitochondrial respiration, protein ubiquitination, apoptosis and cell cycle. This not only helps to counteract lipotoxic hepatocellular injury through modification of gene expression, but - by targeting transcription factors responsible for DNA repair, cell cycle progression and ER stress - might even prevent lipotoxic mechanisms. These findings suggest that HK4 has a great potential for the treatment of non-alcoholic fatty liver disease (NAFLD).
Collapse
Affiliation(s)
- Elisabeth Rohbeck
- German Diabetes Center, Institute for Clinical Diabetology, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany,German Center for Diabetes Research (DZD), Partner Düsseldorf, München-Neuherberg, Germany,CureDiab Metabolic Research GmbH, Düsseldorf, Germany
| | - Corinna Niersmann
- German Diabetes Center, Institute for Clinical Diabetology, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany,German Center for Diabetes Research (DZD), Partner Düsseldorf, München-Neuherberg, Germany,CureDiab Metabolic Research GmbH, Düsseldorf, Germany
| | - Karl Köhrer
- Biological and Medical Research Centre (BMFZ), Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Thorsten Wachtmeister
- Biological and Medical Research Centre (BMFZ), Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Michael Roden
- German Diabetes Center, Institute for Clinical Diabetology, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany,German Center for Diabetes Research (DZD), Partner Düsseldorf, München-Neuherberg, Germany,Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Jürgen Eckel
- German Diabetes Center, Institute for Clinical Diabetology, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany,CureDiab Metabolic Research GmbH, Düsseldorf, Germany
| | - Tania Romacho
- German Diabetes Center, Institute for Clinical Diabetology, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany,Chronic Complications of Diabetes Lab (ChroCoDiL), Department of Nursing Sciences, Physiotherapy and Medicine, Faculty of Health Sciences, University of Almería, Almería, Spain,*Correspondence: Tania Romacho,
| |
Collapse
|
10
|
Blood Cell Responses Following Heavy Alcohol Consumption Coincide with Changes in Acute Phase Reactants of Inflammation, Indices of Hemolysis and Immune Responses to Ethanol Metabolites. Int J Mol Sci 2022; 23:ijms232112738. [PMID: 36361528 PMCID: PMC9656529 DOI: 10.3390/ijms232112738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/06/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
Aberrations in blood cells are common among heavy alcohol drinkers. In order to shed further light on such responses, we compared blood cell status with markers of hemolysis, mediators of inflammation and immune responses to ethanol metabolites in alcohol-dependent patients at the time of admission for detoxification and after abstinence. Blood cell counts, indices of hemolysis (LDH, haptoglobin, bilirubin), calprotectin (a marker of neutrophil activation), suPAR, CD163, pro- and anti-inflammatory cytokines and autoantibodies against protein adducts with acetaldehyde, the first metabolite of ethanol, were measured from alcohol-dependent patients (73 men, 26 women, mean age 43.8 ± 10.4 years) at baseline and after 8 ± 1 days of abstinence. The assessments also included information on the quantities of alcohol drinking and assays for biomarkers of alcohol consumption (CDT), liver function (AST, ALT, ALP, GGT) and acute phase reactants of inflammation. At baseline, the patients showed elevated values of CDT and biomarkers of liver status, which decreased significantly during abstinence. A significant decrease also occurred in LDH, bilirubin, CD163 and IgA and IgM antibodies against acetaldehyde adducts, whereas a significant increase was noted in blood leukocytes, platelets, MCV and suPAR levels. The changes in blood leukocytes correlated with those in serum calprotectin (p < 0.001), haptoglobin (p < 0.001), IL-6 (p < 0.02) and suPAR (p < 0.02). The changes in MCV correlated with those in LDH (p < 0.02), MCH (p < 0.01), bilirubin (p < 0.001) and anti-adduct IgG (p < 0.01). The data indicates that ethanol-induced changes in blood leukocytes are related with acute phase reactants of inflammation and release of neutrophil calprotectin. The studies also highlight the role of hemolysis and immune responses to ethanol metabolites underlying erythrocyte abnormalities in alcohol abusers.
Collapse
|
11
|
Khodayari N, Oshins R, Mehrad B, Lascano JE, Qiang X, West JR, Holliday LS, Lee J, Wiesemann G, Eydgahi S, Brantly M. Cigarette smoke exposed airway epithelial cell-derived EVs promote pro-inflammatory macrophage activation in alpha-1 antitrypsin deficiency. Respir Res 2022; 23:232. [PMID: 36068572 PMCID: PMC9446525 DOI: 10.1186/s12931-022-02161-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 08/27/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alpha-1 antitrypsin deficiency (AATD) is a genetic disorder most commonly secondary to a single mutation in the SERPINA1 gene (PI*Z) that causes misfolding and accumulation of alpha-1 antitrypsin (AAT) in hepatocytes and mononuclear phagocytes which reduces plasma AAT and creates a toxic gain of function. This toxic gain of function promotes a pro-inflammatory phenotype in macrophages that contributes to lung inflammation and early-onset COPD, especially in individuals who smoke cigarettes. The aim of this study is to determine the role of cigarette exposed AATD macrophages and bronchial epithelial cells in AATD-mediated lung inflammation. METHODS Peripheral blood mononuclear cells from AATD and healthy individuals were differentiated into alveolar-like macrophages and exposed to air or cigarette smoke while in culture. Macrophage endoplasmic reticulum stress was quantified and secreted cytokines were measured using qPCR and cytokine ELISAs. To determine whether there is "cross talk" between epithelial cells and macrophages, macrophages were exposed to extracellular vesicles released by airway epithelial cells exposed to cigarette smoke and their inflammatory response was determined. RESULTS AATD macrophages spontaneously produce several-fold more pro-inflammatory cytokines as compared to normal macrophages. AATD macrophages have an enhanced inflammatory response when exposed to cigarette smoke-induced extracellular vesicles (EVs) released from airway epithelial cells. Cigarette smoke-induced EVs induce expression of GM-CSF and IL-8 in AATD macrophages but have no effect on normal macrophages. Release of AAT polymers, potent neutrophil chemo attractants, were also increased from AATD macrophages after exposure to cigarette smoke-induced EVs. CONCLUSIONS The expression of mutated AAT confers an inflammatory phenotype in AATD macrophages which disposes them to an exaggerated inflammatory response to cigarette smoke-induced EVs, and thus could contribute to progressive lung inflammation and damage in AATD individuals.
Collapse
Affiliation(s)
- Nazli Khodayari
- Division of Pulmonary, Critical Care, and Sleep Medicine, College of Medicine, University of Florida, 1600 SW Archer Rd Rm M453A, Gainesville, FL, 32610, USA.
| | - Regina Oshins
- Division of Pulmonary, Critical Care, and Sleep Medicine, College of Medicine, University of Florida, 1600 SW Archer Rd Rm M453A, Gainesville, FL, 32610, USA
| | - Borna Mehrad
- Division of Pulmonary, Critical Care, and Sleep Medicine, College of Medicine, University of Florida, 1600 SW Archer Rd Rm M453A, Gainesville, FL, 32610, USA
| | - Jorge E Lascano
- Division of Pulmonary, Critical Care, and Sleep Medicine, College of Medicine, University of Florida, 1600 SW Archer Rd Rm M453A, Gainesville, FL, 32610, USA
| | | | - Jesse R West
- Division of Pulmonary, Critical Care, and Sleep Medicine, College of Medicine, University of Florida, 1600 SW Archer Rd Rm M453A, Gainesville, FL, 32610, USA
| | - L Shannon Holliday
- Department of Orthodontics, College of Dentistry, University of Florida, Gainesville, FL, USA
| | - Jungnam Lee
- Division of Pulmonary, Critical Care, and Sleep Medicine, College of Medicine, University of Florida, 1600 SW Archer Rd Rm M453A, Gainesville, FL, 32610, USA
| | - Gayle Wiesemann
- College of Medicine, University of Florida, Gainesville, FL, USA
| | - Soroush Eydgahi
- Division of Pulmonary, Critical Care, and Sleep Medicine, College of Medicine, University of Florida, 1600 SW Archer Rd Rm M453A, Gainesville, FL, 32610, USA
| | - Mark Brantly
- Division of Pulmonary, Critical Care, and Sleep Medicine, College of Medicine, University of Florida, 1600 SW Archer Rd Rm M453A, Gainesville, FL, 32610, USA
| |
Collapse
|
12
|
Mohammed EAH, Peng Y, Wang Z, Qiang X, Zhao Q. Synthesis, Antiviral, and Antibacterial Activity of the Glycyrrhizic Acid and Glycyrrhetinic Acid Derivatives. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2022; 48:906-918. [PMID: 35919388 PMCID: PMC9333650 DOI: 10.1134/s1068162022050132] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 12/28/2021] [Accepted: 12/31/2021] [Indexed: 11/28/2022]
Abstract
Glycyrrhizic acid and its primary metabolite glycyrrhetinic acid, are the main active ingredients in the licorice roots (glycyrrhiza species), which are widely used in several countries of the world, especially in east asian countries (China, Japan). These ingredients and their derivatives play an important role in treating many diseases, especially infectious diseases such as COVID-19 and hepatic infections. This review aims to summarize the different ways of synthesising the amide derivatives of glycyrrhizic acid and the main ways to synthesize the glycyrrhitinic acid derivatives. Also, to determine the main biological and pharmacological activity for these compounds from the previous studies to provide essential data to researchers for future studies. Supplementary Information The online version contains supplementary material available at 10.1134/S1068162022050132.
Collapse
Affiliation(s)
- E. A. H. Mohammed
- Institute of Medicinal Chemistry, School of Pharmacy of Lanzhou University, 730000 Lanzhou, China
| | - Y. Peng
- Institute of Medicinal Chemistry, School of Pharmacy of Lanzhou University, 730000 Lanzhou, China
| | - Z. Wang
- Institute of Medicinal Chemistry, School of Pharmacy of Lanzhou University, 730000 Lanzhou, China
| | - X. Qiang
- Institute of Medicinal Chemistry, School of Pharmacy of Lanzhou University, 730000 Lanzhou, China
| | - Q. Zhao
- Institute of Medicinal Chemistry, School of Pharmacy of Lanzhou University, 730000 Lanzhou, China
| |
Collapse
|
13
|
Babaei P, Hoseini R. Exercise training modulates adipokine dysregulations in metabolic syndrome. SPORTS MEDICINE AND HEALTH SCIENCE 2022; 4:18-28. [PMID: 35782776 PMCID: PMC9219261 DOI: 10.1016/j.smhs.2022.01.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/01/2022] [Accepted: 01/07/2022] [Indexed: 12/16/2022] Open
Abstract
Metabolic syndrome (MetS) is a cluster of risk factors for various metabolic diseases, and it is characterized by central obesity, dyslipidemia, hypertension, and insulin resistance. The core component for MetS is adipose tissue, which releases adipokines and influences physical health. Adipokines consist of pro and anti-inflammatory cytokines and contribute to various physiological functions. Generally, a sedentary lifestyle promotes fat accumulation and secretion of pro-inflammatory adipokines. However, regular exercise has been known to exert various beneficial effects on metabolic and cognitive disorders. Although the mechanisms underlying exercise beneficial effects in MetS are not fully understood, changes in energy expenditure, fat accumulation, circulatory level of myokines, and adipokines might be involved. This review article focuses on some of the selected adipokines in MetS, and their responses to exercise training considering possible mechanisms.
Collapse
Affiliation(s)
- Parvin Babaei
- Cellular & Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Rastegar Hoseini
- Department of Sports Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| |
Collapse
|
14
|
Immunoproteasome Activity and Content Determine Hematopoietic Cell Sensitivity to ONX-0914 and to the Infection of Cells with Lentiviruses. Cells 2021; 10:cells10051185. [PMID: 34066177 PMCID: PMC8150886 DOI: 10.3390/cells10051185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/03/2021] [Accepted: 05/07/2021] [Indexed: 01/08/2023] Open
Abstract
Proteasomes are intracellular structures responsible for protein degradation. The 20S proteasome is a core catalytic element of the proteasome assembly. Variations of catalytic subunits generate different forms of 20S proteasomes including immunoproteasomes (iPs), which are present mostly in the immune cells. Certain cells of the immune system are primary targets of retroviruses. It has been shown that several viral proteins directly affect proteasome functionality, while inhibition of proteasome activity with broad specificity proteasome inhibitors stimulates viral transduction. Here we specifically addressed the role of the immunoproteasomes during early stages of viral transduction and investigated the effects of specific immunoproteasome inhibition and activation prior to infection using a panel of cell lines. Inhibition of iPs in hematopoietic cells with immunoproteasome-specific inhibitor ONX-0914 resulted in increased infection by VSV-G pseudotyped lentiviruses. Moreover, a tendency for increased infection of cloned cells with endogenously decreased proteasome activity was revealed. Conversely, activation of iPs by IFN-γ markedly reduced the viral infectivity, which was rescued upon simultaneous immunoproteasome inhibition. Our results indicate that immunoproteasome activity might be determinative for the cellular antiretroviral resistance at least for the cells with high iP content. Finally, therapeutic application of immunoproteasome inhibitors might promote retroviral infection of cells in vivo.
Collapse
|
15
|
Park G, Jung S, Wellen KE, Jang C. The interaction between the gut microbiota and dietary carbohydrates in nonalcoholic fatty liver disease. Exp Mol Med 2021; 53:809-822. [PMID: 34017059 PMCID: PMC8178320 DOI: 10.1038/s12276-021-00614-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/24/2021] [Indexed: 02/04/2023] Open
Abstract
Imbalance between fat production and consumption causes various metabolic disorders. Nonalcoholic fatty liver disease (NAFLD), one such pathology, is characterized by abnormally increased fat synthesis and subsequent fat accumulation in hepatocytes1,2. While often comorbid with obesity and insulin resistance, this disease can also be found in lean individuals, suggesting specific metabolic dysfunction2. NAFLD has become one of the most prevalent liver diseases in adults worldwide, but its incidence in both children and adolescents has also markedly increased in developed nations3,4. Progression of this disease into nonalcoholic steatohepatitis (NASH), cirrhosis, liver failure, and hepatocellular carcinoma in combination with its widespread incidence thus makes NAFLD and its related pathologies a significant public health concern. Here, we review our understanding of the roles of dietary carbohydrates (glucose, fructose, and fibers) and the gut microbiota, which provides essential carbon sources for hepatic fat synthesis during the development of NAFLD.
Collapse
Affiliation(s)
- Grace Park
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Sunhee Jung
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
16
|
Tsunoda M, Fukasawa M, Nishihara A, Takada L, Asano M. JunB can enhance the transcription of IL-8 in oral squamous cell carcinoma. J Cell Physiol 2020; 236:309-317. [PMID: 32510596 DOI: 10.1002/jcp.29843] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 05/25/2020] [Accepted: 05/25/2020] [Indexed: 11/09/2022]
Abstract
Proteasome inhibitor MG132 was shown to enhance the secretion of interleukin 8 (IL-8) by various cells. The enhancement is regulated by the transcription factor activator protein-1 (AP-1) at the transcriptional level. AP-1 is a dimer formed by AP-1 family proteins. The purpose of the present study was to explore the combinations of the AP-1 family proteins that contribute to MG132-driven IL-8 secretion. Oral squamous cell carcinoma-derived cell lines, Ca9-22 and HSC3, were used to demonstrate their response to MG132. IL-8 secretion was augmented by MG132 in both cell lines. c-Jun expression was detected in both the cell lines, whereas c-Fos expression was detected only in the HSC3. The influence of MG132 stimulation on c-Jun and c-Fos expression was further examined by western blot analysis. c-Jun expression was increased by MG132 stimulation, whereas c-Fos expression was not detected even after MG132 stimulation. As JunB is reported to inhibit the transcriptional activity of the AP-1 complex, we speculated that the c-Jun homodimer should contribute to IL-8 enhancement. Expression vectors encoding wild type and c-Jun mutants, M17 and M22-23, respectively, were constructed and transfected into the Ca9-22 cells. In contrast to our expectations, MG132-induced IL-8 secretion was significantly reduced in all the transfectants suggesting that other c-Jun members might form homodimers with c-Jun and contribute to IL-8 enhancement. Transfection of the cells with c-Jun or JunB small hairpin RNA (shRNA) reduced IL-8 secretion up to 50% and 65% of the control shRNA transfectant. Furthermore, cotransfection of both shRNA almost completely inhibited the IL-8 secretion. These results indicate that JunB not only inhibits but also enhances the transcription of c-Jun targets in combination with c-Jun.
Collapse
Affiliation(s)
- Mariko Tsunoda
- Department of Pathology, Nihon University School of Dentistry, Tokyo, Japan.,Division of Immunology and Pathobiology, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Mai Fukasawa
- Division of Applied Oral Sciences, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Anna Nishihara
- Division of Oral Structural and Functional Biology, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Leo Takada
- Division of Oral Health Sciences, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Masatake Asano
- Department of Pathology, Nihon University School of Dentistry, Tokyo, Japan.,Division of Immunology and Pathobiology, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| |
Collapse
|
17
|
Nerstedt A, Kurhe Y, Cansby E, Caputo M, Gao L, Vorontsov E, Ståhlman M, Nuñez-Durán E, Borén J, Marschall HU, Mashek DG, Saunders DN, Sihlbom C, Hoy AJ, Mahlapuu M. Lipid droplet-associated kinase STK25 regulates peroxisomal activity and metabolic stress response in steatotic liver. J Lipid Res 2019; 61:178-191. [PMID: 31857389 DOI: 10.1194/jlr.ra119000316] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/05/2019] [Indexed: 12/18/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) are emerging as leading causes of liver disease worldwide and have been recognized as one of the major unmet medical needs of the 21st century. Our recent translational studies in mouse models, human cell lines, and well-characterized patient cohorts have identified serine/threonine kinase (STK)25 as a protein that coats intrahepatocellular lipid droplets (LDs) and critically regulates liver lipid homeostasis and progression of NAFLD/NASH. Here, we studied the mechanism-of-action of STK25 in steatotic liver by relative quantification of the hepatic LD-associated phosphoproteome from high-fat diet-fed Stk25 knockout mice compared with their wild-type littermates. We observed a total of 131 proteins and 60 phosphoproteins that were differentially represented in STK25-deficient livers. Most notably, a number of proteins involved in peroxisomal function, ubiquitination-mediated proteolysis, and antioxidant defense were coordinately regulated in Stk25 -/- versus wild-type livers. We confirmed attenuated peroxisomal biogenesis and protection against oxidative and ER stress in STK25-deficient human liver cells, demonstrating the hepatocyte-autonomous manner of STK25's action. In summary, our results suggest that regulation of peroxisomal function and metabolic stress response may be important molecular mechanisms by which STK25 controls the development and progression of NAFLD/NASH.
Collapse
Affiliation(s)
- Annika Nerstedt
- Departments of Chemistry and Molecular Biology University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Yeshwant Kurhe
- Departments of Chemistry and Molecular Biology University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Emmelie Cansby
- Departments of Chemistry and Molecular Biology University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mara Caputo
- Departments of Chemistry and Molecular Biology University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lei Gao
- Departments of Chemistry and Molecular Biology University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Egor Vorontsov
- Proteomics Core Facility, University of Gothenburg, Gothenburg, Sweden
| | - Marcus Ståhlman
- Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Esther Nuñez-Durán
- Departments of Chemistry and Molecular Biology University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jan Borén
- Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Hanns-Ulrich Marschall
- Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Douglas G Mashek
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN
| | - Darren N Saunders
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Carina Sihlbom
- Proteomics Core Facility, University of Gothenburg, Gothenburg, Sweden
| | - Andrew J Hoy
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Margit Mahlapuu
- Departments of Chemistry and Molecular Biology University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
18
|
TNF-α in Combination with Palmitate Enhances IL-8 Production via The MyD88- Independent TLR4 Signaling Pathway: Potential Relevance to Metabolic Inflammation. Int J Mol Sci 2019; 20:ijms20174112. [PMID: 31443599 PMCID: PMC6747275 DOI: 10.3390/ijms20174112] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 08/16/2019] [Accepted: 08/17/2019] [Indexed: 01/08/2023] Open
Abstract
Elevated levels of IL-8 (CXCL8) in obesity have been linked with insulin resistance and type 2 diabetes (T2D). The mechanisms that lead to the profound production of IL-8 in obesity remains to be understood. TNF-α and saturated free fatty acids (FFAs) are increased in obese humans and correlate with insulin resistance. Hence, we sought to investigate whether the cooccurrence of TNF-α and FFAs led to increase the production of IL-8 by human monocytes. We found that co-stimulation of human monocytes with palmitate and TNF-α led to increased IL-8 production as compared to those stimulated with palmitate or TNF-α alone. The synergistic production of IL-8 by TNF-α/palmitate was suppressed by neutralizing anti- Toll like receptor 4 (TLR4) antibody and by genetic silencing of TLR4. Both MyD88-deficient and MyD88-competent cells responded comparably to TNF-α/Palmitate. However, TIR-domain-containing adapter-inducing interferon (TRIF) inhibition or interferon regulatory transcription factor 3 (IRF3) knockdown partly blocked the synergistic production of IL-8. Our human data show that increased adipose tissue TNF-α expression correlated positively with IL-8 expression (r = 0.49, P = 0.001). IL-8 and TNF-α correlated positively with macrophage markers including CD68, CD163 and CD86 in adipose tissue. These findings suggest that the signaling cross-talk between saturated fatty acid palmitate and TNF-α may be a key driver in obesity-associated chronic inflammation via an excessive production of IL-8.
Collapse
|
19
|
Chen P, Hu M, Liu F, Yu H, Chen C. S-allyl-l-cysteine (SAC) protects hepatocytes from alcohol-induced apoptosis. FEBS Open Bio 2019; 9:1327-1336. [PMID: 31161729 PMCID: PMC6609569 DOI: 10.1002/2211-5463.12684] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 05/19/2019] [Accepted: 05/31/2019] [Indexed: 01/06/2023] Open
Abstract
Hepatocyte apoptosis is frequently observed in alcohol‐related liver disease (ARLD), which ranks among the 30 leading causes of death worldwide. In the current study, we explored the impact of S‐allyl‐l‐cysteine (SAC), an organosulfur component of garlic, on hepatocyte apoptosis induced by alcohol. Rat liver (BRL‐3A) cells were challenged by ethanol with or without SAC treatment. Cell death/viability, reactive oxygen species (ROS) generation, mitochondrial Cytochrome C release, and caspase 3 activity were then examined. We found that ethanol remarkably induced apoptosis of hepatocytes, while SAC treatment rescued ethanol‐induced hepatocyte injury, as demonstrated by cell counting kit‐8 (CCK8) assay, TUNEL assay, and annexin V/PI staining assay. Ethanol evoked ROS generation in BRL‐3A cells, and this was abated by SAC pretreatment, as indicated by 2′,7′‐dichlorofluorescin diacetate (DCFDA) staining assay. Moreover, ethanol suppressed cellular anti‐apoptotic protein B‐cell lymphoma‐2 (Bcl‐2) expression, increased pro‐apoptotic protein Bcl‐2‐associated X protein (Bax) expression, induced mitochondrial Cytochrome C release, and activated the caspase 3‐dependent apoptosis pathway in BRL‐3A cells. SAC was sufficient to abolish all these changes induced by ethanol, thereby revealing the molecular mechanisms underlying its protective effects. In conclusion, SAC protects hepatocytes from ethanol‐induced apoptosis and may be suitable for use as a novel anti‐apoptotic agent for treating ARLD.
Collapse
Affiliation(s)
- Peng Chen
- Department of Hepatopancreatobiliary Surgery, the Second Affiliated Hospital of Kunming Medical University, China
| | - Mingdao Hu
- Department of Hepatopancreatobiliary Surgery, the Second Affiliated Hospital of Kunming Medical University, China
| | - Feng Liu
- Department of Hepatopancreatobiliary Surgery, the Second Affiliated Hospital of Kunming Medical University, China
| | - Henghai Yu
- Department of Hepatopancreatobiliary Surgery, the Second Affiliated Hospital of Kunming Medical University, China
| | - Chen Chen
- Department of Ophthalmology, the Second People's Hospital of Yunnan Province, Kunming, China
| |
Collapse
|
20
|
Cimini FA, Barchetta I, Porzia A, Mainiero F, Costantino C, Bertoccini L, Ceccarelli V, Morini S, Baroni MG, Lenzi A, Cavallo MG. Circulating IL-8 levels are increased in patients with type 2 diabetes and associated with worse inflammatory and cardiometabolic profile. Acta Diabetol 2017; 54:961-967. [PMID: 28836077 DOI: 10.1007/s00592-017-1039-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/04/2017] [Indexed: 01/20/2023]
Abstract
AIMS Interleukin-8 (IL-8) is a chemokine involved in systemic immunity, macrophages infiltration and activation in adipose tissue and may play a significant role in the pathogenesis of type 2 diabetes (T2D) and atherosclerosis. Aims of this study were to evaluate circulating IL-8 levels in adult patients with T2D in comparison with non-diabetic subjects and to describe clinical and biochemical correlates of IL-8 concentration. METHODS For this cross-sectional study, we enrolled 79 consecutive T2D individuals referring to our diabetes outpatient clinics at Sapienza University of Rome, and 37 sex, age and BMI comparable non-diabetic subjects as a control group. Clinical parameters and medical history were recorded; fasting blood sampling was performed for biochemistry and for measuring serum IL-8, IL-6, TNF-α, CRP, adiponectin and 25(OH)vitamin D [25(OH)D] levels. RESULTS Patients with T2D exhibited significantly higher serum IL-8 levels than non-diabetic subjects (69.27 ± 112.83 vs. 16.03 ± 24.27 pg/mL, p < 0.001). In diabetic patients, increased IL-8 concentration correlated with higher IL-6 (p < 0.001), TNF-α (p = 0.02), FBG (p = 0.035), HbA1c (p = 0.04) and LDL-C (p = 0.04) and with lower adiponectin (p = 0.02) and 25(OH)D (p = 0.003) concentrations. CONCLUSIONS Patients with T2D display a marked elevation of circulating IL-8 levels which identify subjects with worse inflammatory, glycometabolic and lipid profile and lower vitamin D levels. Further studies are warranted for evaluating a possible role of IL-8 as a novel marker for risk stratification in T2D patients.
Collapse
Affiliation(s)
- F A Cimini
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - I Barchetta
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - A Porzia
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - F Mainiero
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - C Costantino
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - L Bertoccini
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - V Ceccarelli
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - S Morini
- Microscopic and Ultrastructural Anatomy (CIR), University Campus Bio-Medico, Rome, Italy
| | - M G Baroni
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - A Lenzi
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - M G Cavallo
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy.
| |
Collapse
|
21
|
Bettermann K. NF-κB and Its Implication in Liver Health and Cancer Development. MECHANISMS OF MOLECULAR CARCINOGENESIS – VOLUME 1 2017:87-114. [DOI: 10.1007/978-3-319-53659-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
22
|
Abouzied MM, Eltahir HM, Taye A, Abdelrahman MS. Experimental evidence for the therapeutic potential of tempol in the treatment of acute liver injury. Mol Cell Biochem 2015; 411:107-15. [DOI: 10.1007/s11010-015-2572-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 09/26/2015] [Indexed: 01/04/2023]
|
23
|
Kondo Y, Kimura O, Tanaka Y, Ninomiya M, Iwata T, Kogure T, Inoue J, Sugiyama M, Morosawa T, Fujisaka Y, Shimosegawa T. Differential Expression of CX3CL1 in Hepatitis B Virus-Replicating Hepatoma Cells Can Affect the Migration Activity of CX3CR1+ Immune Cells. J Virol 2015; 89:7016-7027. [PMID: 25926643 PMCID: PMC4473591 DOI: 10.1128/jvi.00716-15] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 04/17/2015] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED In addition to stellate cells and immune cells, inflamed hepatocytes and hepatoma cells express various kinds of chemokines that attract various kinds of immune cells. Previously, we reported that hepatitis B virus (HBV) replication can induce physiological stress. The aim of this study was to analyze the effect of chemokines produced by HBV-infected hepatocytes and hepatoma cells. A real-time PCR array targeting genes related to chemokines and enzyme-linked immunosorbent assay (ELISA) were carried out to detect the specific chemokines produced by Huh7 cells and HepG2 cells infected with various HBV genotypes. A migration assay, flow cytometry analysis, and immunohistochemistry were carried out to analyze the candidate immune cells that can affect the immunopathogenesis of HBV infection. The expressions of CX3CL1 mRNA and protein were significantly different among HBV genotypes A, B, and C and control cells (mock) (P < 0.05). CD56(+) NK cells and CD8(+) T cells migrated to the hepatoma cells with HBV replication. Moreover, the migration activity of both immune cells was partially cancelled after the treatment of CX3CL1 neutralizing antibody. The expression level of NKG2D on CX3CR1(+) NK cells in HCC with HBV infection was significantly lower than that in hepatocellular carcinoma (HCC) with HCV infection and chronic hepatitis B and C patients (P < 0.05). On the other hand, the frequency of PD-1(high) CX3CR1(+) CD8(+) T cells in HCC with HBV infection was significantly higher than that in HCC with HCV infection and chronic hepatitis B and C (P < 0.05). The expression of CX3CL1 in HBV-replicating hepatocytes and hepatoma cells could contribute to the immunopathogenesis of HBV infection. IMPORTANCE The progressions of the disease are significantly different among HBV genotypes. However, it has not been clear that how different HBV genotypes could induce different inflammatory responses. Here, we first report that the levels of expression of CX3CL1 mRNA and protein were significantly different among HBV genotypes A, B, and C and mock. Not only the differential expression of CX3CL1 among the genotypes but also the phenotype of CX3CR1(+) NK cells and T cells were gradually changed during the progression of the disease status. In addition to in vitro study, the analysis of immunohistochemistry with human samples and NOG mice with human lymphocytes and hepatoma cells supports this phenomenon. The quantification of CX3CL1 could contribute to better understanding of the disease status of HBV infection. Moreover, modifying CX3CL1 might induce an immune response appropriate to the disease status of HBV infection.
Collapse
Affiliation(s)
- Yasuteru Kondo
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai City, Japan
| | - Osamu Kimura
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai City, Japan
| | - Yasuhito Tanaka
- Department of Virology and Liver Unit, Nagoya City University Graduate School of Medicine, Nagoya City, Japan
| | - Masashi Ninomiya
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai City, Japan
| | - Tomoaki Iwata
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai City, Japan
| | - Takayuki Kogure
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai City, Japan
| | - Jun Inoue
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai City, Japan
| | - Masaya Sugiyama
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| | - Tatsuki Morosawa
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai City, Japan
| | - Yasuyuki Fujisaka
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai City, Japan
| | - Tooru Shimosegawa
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai City, Japan
| |
Collapse
|
24
|
Ginsenoside Rg1, a potential JNK inhibitor, protects against ischemia/reperfusion-induced liver damage. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.04.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
25
|
Ceni E, Mello T, Galli A. Pathogenesis of alcoholic liver disease: Role of oxidative metabolism. World J Gastroenterol 2014; 20:17756-17772. [PMID: 25548474 PMCID: PMC4273126 DOI: 10.3748/wjg.v20.i47.17756] [Citation(s) in RCA: 348] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 05/22/2014] [Accepted: 08/28/2014] [Indexed: 02/06/2023] Open
Abstract
Alcohol consumption is a predominant etiological factor in the pathogenesis of chronic liver diseases, resulting in fatty liver, alcoholic hepatitis, fibrosis/cirrhosis, and hepatocellular carcinoma (HCC). Although the pathogenesis of alcoholic liver disease (ALD) involves complex and still unclear biological processes, the oxidative metabolites of ethanol such as acetaldehyde and reactive oxygen species (ROS) play a preeminent role in the clinical and pathological spectrum of ALD. Ethanol oxidative metabolism influences intracellular signaling pathways and deranges the transcriptional control of several genes, leading to fat accumulation, fibrogenesis and activation of innate and adaptive immunity. Acetaldehyde is known to be toxic to the liver and alters lipid homeostasis, decreasing peroxisome proliferator-activated receptors and increasing sterol regulatory element binding protein activity via an AMP-activated protein kinase (AMPK)-dependent mechanism. AMPK activation by ROS modulates autophagy, which has an important role in removing lipid droplets. Acetaldehyde and aldehydes generated from lipid peroxidation induce collagen synthesis by their ability to form protein adducts that activate transforming-growth-factor-β-dependent and independent profibrogenic pathways in activated hepatic stellate cells (HSCs). Furthermore, activation of innate and adaptive immunity in response to ethanol metabolism plays a key role in the development and progression of ALD. Acetaldehyde alters the intestinal barrier and promote lipopolysaccharide (LPS) translocation by disrupting tight and adherent junctions in human colonic mucosa. Acetaldehyde and LPS induce Kupffer cells to release ROS and proinflammatory cytokines and chemokines that contribute to neutrophils infiltration. In addition, alcohol consumption inhibits natural killer cells that are cytotoxic to HSCs and thus have an important antifibrotic function in the liver. Ethanol metabolism may also interfere with cell-mediated adaptive immunity by impairing proteasome function in macrophages and dendritic cells, and consequently alters allogenic antigen presentation. Finally, acetaldehyde and ROS have a role in alcohol-related carcinogenesis because they can form DNA adducts that are prone to mutagenesis, and they interfere with methylation, synthesis and repair of DNA, thereby increasing HCC susceptibility.
Collapse
|
26
|
Refsnes M, Skuland T, Låg M, Schwarze PE, Øvrevik J. Differential NF-κB and MAPK activation underlies fluoride- and TPA-mediated CXCL8 (IL-8) induction in lung epithelial cells. J Inflamm Res 2014; 7:169-85. [PMID: 25540590 PMCID: PMC4270361 DOI: 10.2147/jir.s69646] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Different toxic agents have a varying potential to induce the production of the proinflammatory chemokine, CXCL8 (interleukin [IL]-8), in lung cells. A critical question is which mechanisms determine the magnitude and persistence of the CXCL8 responses to different stimuli. To approach this, we compared the potential of the phorbol ester, 12-O-tetradecanoylphorbol-13-acetate (TPA), and sodium fluoride (NaF) to induce CXCL8 responses in A549 cells, with emphasis on the importance of nuclear factor kappa B (NF-κB)- and mitogen-activated protein kinase (MAPK) signaling. Notably, TPA induced a greater release of CXCL8 than did NaF. Furthermore, TPA induced a strong, rapid, but transient upregulation of CXCL8 messenger (m)RNA, whereas NaF induced a weaker, more delayed, but persistent upregulation. With respect to signaling, TPA led to an early, strong, and relatively transient extracellular signal-regulated kinase (ERK)1/2 phosphorylation, and a less marked and even more transient phosphorylation of c-jun-N-terminal kinases (JNK1/2) and p38. In contrast, NaF elicited a lower, but relatively sustained increase in phosphorylation of ERK1/2, and a marked phosphorylation of p38 and JNK1/2, with the JNK1/2 response as most transient. Only ERK1/2 inhibition affected the TPA response, whereas inhibition of all the three MAPK cascades reduced NaF-induced CXCL8 release. TPA also induced an early, marked phosphorylation/translocation of p65 (NF-κB), whereas NaF induced slower, less pronounced effects on p65. The CXCL8 responses by TPA and NaF were reduced by p65-siRNA. In conclusion, all MAPK cascades were involved in NaF-induced CXCL8 release, whereas only ERK1/2 activation was involved in response to TPA. Furthermore, NF-κB activation appeared to be indispensable for CXCL8 induction. The early response, magnitude, and persistency of MAPK and NF-κB signaling seemed to be critical determinants for the potential to induce CXCL8. These findings underscore that a strong, rapid, and relatively transient activation of ERK1/2 in combination with NF-kB may be sufficient for a strong induction of CXCL8, which may exceed the effects of a more moderate ERK1/2 activation in combination with activation of p38, JNK1/2, and NF-κB.
Collapse
Affiliation(s)
- Magne Refsnes
- Department of Air Pollution and Noise, Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway
| | - Tonje Skuland
- Department of Air Pollution and Noise, Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway
| | - Marit Låg
- Department of Air Pollution and Noise, Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway
| | - Per E Schwarze
- Department of Air Pollution and Noise, Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway
| | - Johan Øvrevik
- Department of Air Pollution and Noise, Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
27
|
Golbidi S, Laher I. Exercise induced adipokine changes and the metabolic syndrome. J Diabetes Res 2014; 2014:726861. [PMID: 24563869 PMCID: PMC3915640 DOI: 10.1155/2014/726861] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 10/18/2013] [Indexed: 12/25/2022] Open
Abstract
The lack of adequate physical activity and obesity created a worldwide pandemic. Obesity is characterized by the deposition of adipose tissue in various parts of the body; it is now evident that adipose tissue also acts as an endocrine organ capable of secreting many cytokines that are though to be involved in the pathophysiology of obesity, insulin resistance, and metabolic syndrome. Adipokines, or adipose tissue-derived proteins, play a pivotal role in this scenario. Increased secretion of proinflammatory adipokines leads to a chronic inflammatory state that is accompanied by insulin resistance and glucose intolerance. Lifestyle change in terms of increased physical activity and exercise is the best nonpharmacological treatment for obesity since these can reduce insulin resistance, counteract the inflammatory state, and improve the lipid profile. There is growing evidence that exercise exerts its beneficial effects partly through alterations in the adipokine profile; that is, exercise increases secretion of anti-inflammatory adipokines and reduces proinflammatory cytokines. In this paper we briefly describe the pathophysiologic role of four important adipokines (adiponectin, leptin, TNF-α, and IL-6) in the metabolic syndrome and review some of the clinical trials that monitored these adipokines as a clinical outcome before and after exercise.
Collapse
Affiliation(s)
- Saeid Golbidi
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada V6T 1Z3
| | - Ismail Laher
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada V6T 1Z3
- *Ismail Laher:
| |
Collapse
|
28
|
Ho LJ, Hung LF, Liu FC, Hou TY, Lin LC, Huang CY, Lai JH. Ginkgo biloba extract individually inhibits JNK activation and induces c-Jun degradation in human chondrocytes: potential therapeutics for osteoarthritis. PLoS One 2013; 8:e82033. [PMID: 24349175 PMCID: PMC3859542 DOI: 10.1371/journal.pone.0082033] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 10/20/2013] [Indexed: 12/15/2022] Open
Abstract
Osteoarthritis (OA) is a common joint disorder with varying degrees of inflammation. The ideal anti-OA drug should have immunomodulatory effects while at the same time having limited or no toxicity. We examined the anti-inflammatory effects of Ginkgo biloba extract (EGb) in interleukin-1 (IL-1)-stimulated human chondrocytes. Chondrocytes were prepared from cartilage specimens taken from patients with osteoarthritis who had received total hip or total knee replacement. The concentrations of chemokines and the degree of cell migration were determined by ELISA and chemotaxis assays, respectively. The activation of inducible nitric oxide synthase (iNOS), mitogen-activated protein kinases (MAPKs), activator protein-1 (AP-1), and nuclear factor-kappaB (NF-κB) was determined by immunoblotting, immunohistochemistry, and electrophoretic mobility shift assay. We found that EGb inhibited IL-1-induced production of chemokines, which in turn resulted in attenuation of THP-1 cell migration toward EGb-treated cell culture medium. EGb also suppressed IL-1-stimulated iNOS expression and release of nitric oxide (NO). The EGb-mediated suppression of the iNOS-NO pathway correlated with the attenuation of activator protein-1 (AP-1) but not nuclear factor-kappaB (NF-κB) DNA-binding activity. Of the mitogen-activated protein kinases (MAPKs), EGb inhibited only c-Jun N-terminal kinase (JNK). Unexpectedly, EGb selectively caused degradation of c-Jun protein. Further investigation revealed that EGb-mediated c-Jun degradation was preceded by ubiquitination of c-Jun and could be prevented by the proteosome inhibitor MG-132. The results imply that EGb protects against chondrocyte degeneration by inhibiting JNK activation and inducing ubiquitination-dependent c-Jun degradation. Although additional research is needed, our results suggest that EGb is a potential therapeutic agent for the treatment of OA.
Collapse
Affiliation(s)
- Ling-Jun Ho
- Institute of Cellular and System Medicine, National Health Research Institute, Zhunan, Taiwan, R.O.C.
- Graduate Institute of Basic Medical Science, PhD Program of Aging, China Medical University, Taichung, Taiwan, R.O.C.
| | - Li-Feng Hung
- Institute of Cellular and System Medicine, National Health Research Institute, Zhunan, Taiwan, R.O.C.
| | - Feng-Cheng Liu
- Rheumatology/Immunology and Allergy, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C.
| | - Tsung-Yun Hou
- Rheumatology/Immunology and Allergy, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C.
| | - Leou-Chyr Lin
- Department of Orthopaedics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C.
| | - Chuan-Yueh Huang
- Institute of Cellular and System Medicine, National Health Research Institute, Zhunan, Taiwan, R.O.C.
| | - Jenn-Haung Lai
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei, Taiwan, R.O.C.
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Tao-Yuan, Taiwan, R.O.C.
- * E-mail:
| |
Collapse
|
29
|
Venturelli S, Berger A, Weiland T, Essmann F, Waibel M, Nuebling T, Häcker S, Schenk M, Schulze-Osthoff K, Salih HR, Fulda S, Sipos B, Johnstone RW, Lauer UM, Bitzer M. Differential induction of apoptosis and senescence by the DNA methyltransferase inhibitors 5-azacytidine and 5-aza-2'-deoxycytidine in solid tumor cells. Mol Cancer Ther 2013; 12:2226-36. [PMID: 23924947 DOI: 10.1158/1535-7163.mct-13-0137] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Epigenetic alterations are a hallmark of cancer that govern the silencing of genes. Up to now, 5-azacytidine (5-aza-CR, Vidaza) and 5-aza-2'-deoxycytidine (5-aza-dC, Dacogen) are the only clinically approved DNA methyltransferase inhibitors (DNMTi). Current effort tries to exploit DNMTi application beyond acute leukemia or myelodysplastic syndrome, especially to solid tumors. Although both drugs only differ by a minimal structural difference, they trigger distinct molecular mechanisms that are highly relevant for a rational choice of new combination therapies. Therefore, we investigated cell death pathways in vitro in human hepatoma, colon, renal, and lung cancer cells and in vivo in chorioallantoic membrane and xenograft models. Real-time cancer cell monitoring and cytokine profiling revealed a profoundly distinct response pattern to both drugs. 5-aza-dC induced p53-dependent tumor cell senescence and a high number of DNA double-strand breaks. In contrast, 5-aza-CR downregulated p53, induced caspase activation and apoptosis. These individual response patterns of tumor cells could be verified in vivo in chorioallantoic membrane assays and in a hepatoma xenograft model. Although 5-aza-CR and 5-aza-dC are viewed as drugs with similar therapeutic activity, they induce a diverse molecular response in tumor cells. These findings together with other reported differences enable and facilitate a rational design of new combination strategies to further exploit the epigenetic mode of action of these two drugs in different areas of clinical oncology.
Collapse
Affiliation(s)
- Sascha Venturelli
- Corresponding Author: Michael Bitzer, Medical University Hospital, University of Tuebingen, Otfried-Mueller-Str. 10, Tuebingen D-72076, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Orman ES, Odena G, Bataller R. Alcoholic liver disease: pathogenesis, management, and novel targets for therapy. J Gastroenterol Hepatol 2013; 28 Suppl 1:77-84. [PMID: 23855300 PMCID: PMC4405238 DOI: 10.1111/jgh.12030] [Citation(s) in RCA: 179] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/20/2013] [Indexed: 02/06/2023]
Abstract
Alcohol use is a leading cause of preventable morbidity and mortality worldwide, with much of its negative impact as the result of alcoholic liver disease (ALD). ALD is a broad term that encompasses a spectrum of phenotypes ranging from simple steatosis to steatohepatitis, progressive fibrosis, cirrhosis, and hepatocellular carcinoma. The mechanisms underlying the development of these different disease stages are incompletely understood. Standard treatment of ALD, which includes abstinence, nutritional support, and corticosteroids, has not changed in the last 40 years despite continued poor outcomes. Novel therapies are therefore urgently needed. The development of such therapies has been hindered by inadequate resources for research and unsuitable animal models. However, recent developments in translational research have allowed for identification of new potential targets for therapy. These targets include: (i) CXC chemokines, (ii) IL-22/STAT3, (iii) TNF receptor superfamily, (iv) osteopontin, (v) gut microbiota and lipopolysaccharide (LPS), (vi) endocannabinoids, and (vii) inflammasomes. We review the natural history, risk factors, pathogenesis, and current treatments for ALD. We further discuss the findings of recent translational studies and potential therapeutic targets.
Collapse
Affiliation(s)
- Eric S Orman
- Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina, Chapel Hill, North Carolina 27599, USA.
| | | | | |
Collapse
|
31
|
Zhao Q, Qin CY, Zhao ZH, Fan YC, Wang K. Epigenetic modifications in hepatic stellate cells contribute to liver fibrosis. TOHOKU J EXP MED 2013; 229:35-43. [PMID: 23238615 DOI: 10.1620/tjem.229.35] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Liver fibrosis represents the final common pathway of virtually all types of chronic liver diseases, and it has been a major public health concern. Many genes have been demonstrated to be involved in the pathogenesis of liver fibrosis, while the mechanisms underlying gene regulation still needs further research. On the other hand, hepatic stellate cells (HSCs) are quiescent cells in the perisinusoidal space in liver. HSCs facilitate hepatocytes interactions via releasing soluble inflammatory factors and producing extracellular matrix. HSCs can be activated in response to liver injury, and they differentiate to myofibroblasts, which greatly contribute to the fibrogenesis process. Various epigenetic procedures, including DNA methylation, histone modification and formation of particular chromatin structure, play crucial roles in the gene transcriptional expression in HSCs, regulating various vital processes. For instance, epigenetic modulation on the peroxisome proliferator-activated receptor gamma (PPAR-γ) gene promoter accounts for HSC differentiation through interacting pathways. Aberrant expression of a series of histones and chemokines in activated HSCs can aggravate inflammation and oxidative stress, which in turn promotes differentiation of HSCs to myofibroblasts and enhances the whole fibrogenesis process. Degradation of extracellular matrix is also regulated through epigenetic modulation on matrix associated enzymes. Moreover, fibrosis-related epigenetic modifications in the parental generation may be inherited to their offspring. In this review, we firstly summarize the vital epigenetic modifications of fibrosis-related genes in HSCs, and highlight specific nucleic acid sequences and structures in gene promoters as important action sites, which may provide indicators for liver fibrosis diagnosis in the future.
Collapse
Affiliation(s)
- Qi Zhao
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, Shangdong, PR China.
| | | | | | | | | |
Collapse
|
32
|
Mohammad MK, Avila D, Zhang J, Barve S, Arteel G, McClain C, Joshi-Barve S. Acrolein cytotoxicity in hepatocytes involves endoplasmic reticulum stress, mitochondrial dysfunction and oxidative stress. Toxicol Appl Pharmacol 2012; 265:73-82. [PMID: 23026831 DOI: 10.1016/j.taap.2012.09.021] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 09/18/2012] [Accepted: 09/21/2012] [Indexed: 11/27/2022]
Abstract
Acrolein is a common environmental, food and water pollutant and a major component of cigarette smoke. Also, it is produced endogenously via lipid peroxidation and cellular metabolism of certain amino acids and drugs. Acrolein is cytotoxic to many cell types including hepatocytes; however the mechanisms are not fully understood. We examined the molecular mechanisms underlying acrolein hepatotoxicity in primary human hepatocytes and hepatoma cells. Acrolein, at pathophysiological concentrations, caused a dose-dependent loss of viability of hepatocytes. The death was apoptotic at moderate and necrotic at high concentrations of acrolein. Acrolein exposure rapidly and dramatically decreased intracellular glutathione and overall antioxidant capacity, and activated the stress-signaling MAP-kinases JNK, p42/44 and p38. Our data demonstrate for the first time in human hepatocytes, that acrolein triggered endoplasmic reticulum (ER) stress and activated eIF2α, ATF-3 and -4, and Gadd153/CHOP, resulting in cell death. Notably, the protective/adaptive component of ER stress was not activated, and acrolein failed to up-regulate the protective ER-chaperones, GRP78 and GRP94. Additionally, exposure to acrolein disrupted mitochondrial integrity/function, and led to the release of pro-apoptotic proteins and ATP depletion. Acrolein-induced cell death was attenuated by N-acetyl cysteine, phenyl-butyric acid, and caspase and JNK inhibitors. Our data demonstrate that exposure to acrolein induces a variety of stress responses in hepatocytes, including GSH depletion, oxidative stress, mitochondrial dysfunction and ER stress (without ER-protective responses) which together contribute to acrolein toxicity. Our study defines basic mechanisms underlying liver injury caused by reactive aldehyde pollutants such as acrolein.
Collapse
Affiliation(s)
- Mohammad K Mohammad
- Department of Medicine, University of Louisville, USA; Alcohol Research Center, University of Louisville, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Golbidi S, Mesdaghinia A, Laher I. Exercise in the metabolic syndrome. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2012; 2012:349710. [PMID: 22829955 PMCID: PMC3399489 DOI: 10.1155/2012/349710] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 05/13/2012] [Indexed: 02/06/2023]
Abstract
The metabolic syndrome is a clustering of obesity, diabetes, hyperlipidemia, and hypertension that is occurring in increasing frequency across the global population. Although there is some controversy about its diagnostic criteria, oxidative stress, which is defined as imbalance between the production and inactivation of reactive oxygen species, has a major pathophysiological role in all the components of this disease. Oxidative stress and consequent inflammation induce insulin resistance, which likely links the various components of this disease. We briefly review the role of oxidative stress as a major component of the metabolic syndrome and then discuss the impact of exercise on these pathophysiological pathways. Included in this paper is the effect of exercise in reducing fat-induced inflammation, blood pressure, and improving muscular metabolism.
Collapse
Affiliation(s)
- Saeid Golbidi
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada V6T 1Z3
| | - Azam Mesdaghinia
- Physiology Research Center, Kashan University of Medical Sciences and Health Services, Kashan 87155/111, Iran
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada V6T 1Z3
| |
Collapse
|
34
|
Abstract
Alcoholic liver disease (ALD) is associated with a spectrum of liver injury ranging from steatosis and steatohepatitis to fibrosis and cirrhosis. While multifactorial pathogenesis plays a role in the disease progression, enhanced inflammation in the liver during ethanol exposure is a major feature of ALD. Dysregulated cytokine metabolism and activity are crucial to the initiation of alcohol-induced liver injury. The pro-inflammatory cytokine tumor necrosis factor (TNF-α) has been demonstrated to be one of the key factors in the various aspects of pathophysiology of ALD. The immunomodulatory cytokines such as interleukin 10 and interleukin 6 play roles in exerting hepatic protective effects. Adiponectin is an adipose tissue-derived hormone, which displays protective actions on ethanol-induced liver injury. Treatment for mice with adiponectin decreases TNF-α expression, steatosis and prevents alcohol-induced liver injury. Adiponectin exerts its anti-inflammatory effects via suppression of TNF-α expression and induction of anti-inflammatory cytokines such as IL-10. Adiponectin attenuates alcoholic liver injury by the complex network of multiple signaling pathways in the liver, leading to enhanced fatty acid oxidation and reduced steatosis. Interactions between pro- and anti-inflammatory cytokines such as TNFα and adiponectin and other cytokines are likely to play important roles in the development and progression of alcoholic liver disease.
Collapse
|
35
|
GAO BIN, BATALLER RAMON. Alcoholic liver disease: pathogenesis and new therapeutic targets. Gastroenterology 2011; 141:1572-85. [PMID: 21920463 PMCID: PMC3214974 DOI: 10.1053/j.gastro.2011.09.002] [Citation(s) in RCA: 1479] [Impact Index Per Article: 105.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 09/08/2011] [Accepted: 09/08/2011] [Indexed: 02/07/2023]
Abstract
Alcoholic liver disease (ALD) is a major cause of chronic liver disease worldwide and can lead to fibrosis and cirrhosis. The latest surveillance report published by the National Institute on Alcohol Abuse and Alcoholism showed that liver cirrhosis was the 12th leading cause of death in the United States, with a total of 29,925 deaths in 2007, 48% of which were alcohol related. The spectrum of ALD includes simple steatosis, alcoholic hepatitis, fibrosis, cirrhosis, and superimposed hepatocellular carcinoma. Early work on the pathogenesis of the disease focused on ethanol metabolism-associated oxidative stress and glutathione depletion, abnormal methionine metabolism, malnutrition, and production of endotoxins that activate Kupffer cells. We review findings from recent studies that have characterized specific intracellular signaling pathways, transcriptional factors, aspects of innate immunity, chemokines, epigenetic features, microRNAs, and stem cells that are associated with ALD, improving our understanding of its pathogenesis. Despite this progress, no targeted therapies are available. The cornerstone of treatment for alcoholic hepatitis remains as it was 40 years ago: abstinence, nutritional support, and corticosteroids. There is an urgent need to develop new pathophysiology-oriented therapies. Recent translational studies of human samples and animal models have identified promising therapeutic targets.
Collapse
Affiliation(s)
- BIN GAO
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - RAMON BATALLER
- Liver Unit, Hospital Clínic, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona, Catalonia, Spain
| |
Collapse
|
36
|
Increases in β-amyloid protein in the hippocampus caused by diabetic metabolic disorder are blocked by minocycline through inhibition of NF-κB pathway activation. Pharmacol Rep 2011; 63:381-91. [PMID: 21602593 DOI: 10.1016/s1734-1140(11)70504-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Revised: 09/06/2010] [Indexed: 01/07/2023]
Abstract
Activation of the NF-κB pathway plays an important role in the pathophysiology of Alzheimer's disease (AD), and blocking NF-κB pathway activation has been shown to attenuate cognitive impairment. Diabetic metabolic disorder contributes to β-amyloid protein (Aβ) generation. The goal of this study was to determine the effect of minocycline on Aβ generation and the NF-κB pathway in the hippocampus of diabetic rats and to elucidate the neuroprotective mechanisms of minocycline for the treatment of diabetic metabolic disorder. The diabetic rat model was established using a high-fat diet and an intraperitoneal injection of streptozocin (STZ). Behavioral tests showed that the capacity of learning and memory was significantly lower in diabetic rats. The levels of NF-κB, COX-2, iNOS, IL-1β and TNF-α after the STZ injection were significantly increased in the hippocampus. Significant increases in Aβ, BACE1, NF-κB, COX-2, iNOS, IL-1β and TNF-α were found in diabetic rats. The levels of Aβ, NF-κB, COX-2, iNOS, IL-1β and TNF-α were significantly decreased after minocycline administration; however, minocycline had no effect on BACE1 expression. In sum, diabetes contributes to the activation of the NF-κB pathway and upregulates BACE1 and Aβ. Minocycline downregulates Aβ in the hippocampus by inhibiting NF-κB pathway activation.
Collapse
|
37
|
Zinc deprivation mediates alcohol-induced hepatocyte IL-8 analog expression in rodents via an epigenetic mechanism. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:693-702. [PMID: 21708112 DOI: 10.1016/j.ajpath.2011.04.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2010] [Revised: 04/04/2011] [Accepted: 04/25/2011] [Indexed: 12/20/2022]
Abstract
Neutrophil infiltration caused by IL-8 production is a central mechanism in alcohol-induced hepatitis. This study was performed to examine if an epigenetic mechanism is involved in alcohol-induced IL-8 production. Mice were pair-fed an alcohol-containing liquid diet for 4 weeks. Alcohol exposure induced hepatitis as indicated by increased expression of keratinocyte-derived cytokine (mouse IL-8) and neutrophil infiltration. Alcohol exposure induced histone 3 hyperacetylation owing to inhibition of histone deacetylase (HDAC) in association with NF-κB activation. Cell culture studies showed that alcohol exposure induced IL-8 and cytokine-induced neutrophil chemoattractant-1 (CINC-1, rat IL-8) production in human VL-17A cells and rat H4IIEC3 cells, respectively, dependent on acetaldehyde production, oxidative stress, and zinc release. Zinc deprivation alone induced CINC-1 production and acted synergistically with lipopolysaccharide or tumor necrosis factor-α on CINC-1 production. Zinc deprivation induced histone 3 hyperacetylation at lysine 9 through suppression of HDAC activity. Zinc deprivation caused nuclear translocation of NF-κB, and reduced HDAC binding to NF-κB. Chromatin immunoprecipitation (ChIP) showed that zinc deprivation caused histone 3 hyperacetylation as well as increased NF-κB binding to the CINC-1 promoter. In conclusion, inactivation of HDAC caused by zinc deprivation is a novel mechanism underlying IL-8 up-regulation in alcoholic hepatitis.
Collapse
|
38
|
Toll-like receptors in the pathogenesis of alcoholic liver disease. Gastroenterol Res Pract 2010; 2010. [PMID: 20827314 PMCID: PMC2933900 DOI: 10.1155/2010/710381] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Accepted: 07/20/2010] [Indexed: 12/15/2022] Open
Abstract
In the multifactorial pathophysiology of alcoholic liver disease (ALD), inflammatory cascade activation plays a central role. Recent studies demonstrated that Toll-like Receptors, the sensors of microbial and endogenous danger signals, are expressed and activated in innate immune cells as well as in parenchymal cells in the liver and thereby contribute to ALD. In this paper, we discuss the importance of gut-derived endotoxin and its recognition by TLR4. The significance of TLR-induced intracellular signaling pathways and cytokine production as well as the contribution of reactive oxygen radicals is evaluated. The contribution of TLR signaling to induction of liver fibrosis and hepatocellular cancer is reviewed in the context of alcohol-induced liver disease.
Collapse
|
39
|
Diesen DL, Kuo PC. Nitric oxide and redox regulation in the liver: Part I. General considerations and redox biology in hepatitis. J Surg Res 2010; 162:95-109. [PMID: 20444470 PMCID: PMC2885581 DOI: 10.1016/j.jss.2009.09.019] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Revised: 09/01/2009] [Accepted: 09/04/2009] [Indexed: 12/16/2022]
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are created in normal hepatocytes and are critical for normal physiologic processes, including oxidative respiration, growth, regeneration, apoptosis, and microsomal defense. When the levels of oxidation products exceed the capacity of normal antioxidant systems, oxidative stress occurs. This type of stress, in the form of ROS and RNS, can be damaging to all liver cells, including hepatocytes, Kupffer cells, stellate cells, and endothelial cells, through induction of inflammation, ischemia, fibrosis, necrosis, apoptosis, or through malignant transformation by damaging lipids, proteins, and/or DNA. In Part I of this review, we will discuss basic redox biology in the liver, including a review of ROS, RNS, and antioxidants, with a focus on nitric oxide as a common source of RNS. We will then review the evidence for oxidative stress as a mechanism of liver injury in hepatitis (alcoholic, viral, nonalcoholic). In Part II of this review, we will review oxidative stress in common pathophysiologic conditions, including ischemia/reperfusion injury, fibrosis, hepatocellular carcinoma, iron overload, Wilson's disease, sepsis, and acetaminophen overdose. Finally, biomarkers, proteomic, and antioxidant therapies will be discussed as areas for future therapeutic interventions.
Collapse
Affiliation(s)
- Diana L Diesen
- Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | |
Collapse
|
40
|
Wu LH, Shi BZ, Zhao QL, Wu XZ. Fucosylated glycan inhibition of human hepatocellular carcinoma cell migration through binding to chemokine receptors. Glycobiology 2009; 20:215-23. [PMID: 19884117 DOI: 10.1093/glycob/cwp168] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
SMMC-7721 hepatocellular carcinoma cells (HCC) were incubated with fucosylated glycoproteins that had been isolated from retinoic acid-treated cells by affinity chromatography. HCC migration was significantly inhibited by AAL- and LCA-glycoproteins. Glycopeptides, obtained by digestion of the glycoproteins with trypsin and papain, were found to have a similar inhibitory effect on HCC migration as the corresponding glycoproteins. The inhibitory actions of the glycoproteins were almost abolished after digestion with alpha-L-1,3/4- or alpha-L-1,2-fucosidase. Induction of HCC migration with chemokines including interleukin-8 (IL-8), lymphotactin, monocyte chemoattractant protein-1, and stroma cell-derived factor-1 was examined and IL-8 was found to be the most potent. Interestingly, the isolated glycoproteins significantly inhibited HCC migration and F-actin aggregation induced by IL-8, whereas the glycans themselves did not induce F-actin assembly. From receptor binding analysis AAL-glycan was found to bind IL-8 receptors especially CXCR2 directly and such binding could be blocked by 3'- or 2'-fucosyllactose. After CXCR2 silence by target RNAi, the cells almost lost the response to AAL-glycan inhibition. Our findings suggest that fucosylation plays an important role in the interaction between IL-8 and its receptors inducing HCC migration.
Collapse
Affiliation(s)
- Li Hui Wu
- Department of Biochemistry, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | | | | | | |
Collapse
|
41
|
Bousquet-Dubouch MP, Nguen S, Bouyssié D, Burlet-Schiltz O, French SW, Monsarrat B, Bardag-Gorce F. Chronic ethanol feeding affects proteasome-interacting proteins. Proteomics 2009; 9:3609-22. [PMID: 19609968 PMCID: PMC2766596 DOI: 10.1002/pmic.200800959] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Accepted: 04/05/2009] [Indexed: 12/20/2022]
Abstract
Studies on alcoholic liver injury mechanisms show a significant inhibition of the proteasome activity. To investigate this phenomenon, we isolated proteasome complexes from the liver of rats fed ethanol chronically, and from the liver of their pair-fed controls, using a non-denaturing multiple centrifugations procedure to preserve proteasome-interacting proteins (PIPs). ICAT and MS/MS spectral counting, further confirmed by Western blot, showed that the levels of several PIPs were significantly decreased in the isolated ethanol proteasome fractions. This was the case of PA28alpha/beta proteasome activator subunits, and of three proteasome-associated deubiquitinases, Rpn11, ubiquitin C-terminal hydrolase 14, and ubiquitin carboxyl-terminal hydrolase L5. Interestingly, Rpn13 C-terminal end was missing in the ethanol proteasome fraction, which probably altered the linking of ubiquitin carboxyl-terminal hydrolase L5 to the proteasome. 20S proteasome and most 19S subunits were however not changed but Ecm29, a protein known to stabilize the interactions between the 20S and its activators, was decreased in the isolated ethanol proteasome fractions. It is proposed that ethanol metabolism causes proteasome inhibition by several mechanisms, including by altering PIPs and proteasome regulatory complexes binding to the proteasome.
Collapse
Affiliation(s)
- Marie-Pierre Bousquet-Dubouch
- CNRS; IPBS (Institut de Pharmacologie et de Biologie Structurale); 205 route de Narbonne, F-31077 Toulouse, France
- Université de Toulouse; UPS; IPBS; F-31077 Toulouse, France
| | | | - David Bouyssié
- CNRS; IPBS (Institut de Pharmacologie et de Biologie Structurale); 205 route de Narbonne, F-31077 Toulouse, France
- Université de Toulouse; UPS; IPBS; F-31077 Toulouse, France
| | - Odile Burlet-Schiltz
- CNRS; IPBS (Institut de Pharmacologie et de Biologie Structurale); 205 route de Narbonne, F-31077 Toulouse, France
- Université de Toulouse; UPS; IPBS; F-31077 Toulouse, France
| | | | - Bernard Monsarrat
- CNRS; IPBS (Institut de Pharmacologie et de Biologie Structurale); 205 route de Narbonne, F-31077 Toulouse, France
- Université de Toulouse; UPS; IPBS; F-31077 Toulouse, France
| | | |
Collapse
|
42
|
Joshi-Barve S, Amancherla K, Patil M, Bhatnagar A, Mathews S, Gobejishvili L, Cave M, McClain C, Barve S. Acrolein, a ubiquitous pollutant and lipid hydroperoxide product, inhibits antiviral activity of interferon-alpha: relevance to hepatitis C. Free Radic Biol Med 2009; 47:47-54. [PMID: 19345260 PMCID: PMC3947765 DOI: 10.1016/j.freeradbiomed.2009.03.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2008] [Revised: 03/26/2009] [Accepted: 03/26/2009] [Indexed: 01/31/2023]
Abstract
Hepatitis C virus (HCV) infection is a major cause of chronic liver disease and can lead to hepatocellular carcinoma and end-stage liver disease. The current FDA-approved treatment for HCV (pegylated interferon-alpha (IFNalpha) with ribavirin) is effective in only about 50% of patients. Epidemiological evidence suggests that obesity, alcohol, smoking, and environmental pollutants may contribute to resistance to IFNalpha therapy in HCV. Acrolein, a ubiquitous environmental pollutant and major component of cigarette smoke, is also generated endogenously by cellular metabolism and lipid peroxidation. This study examines the effects of acrolein on (i) IFNalpha-mediated signaling and antiviral gene expression in cultured and primary human hepatocytes and (ii) HCV replication in an HCV-replicon system. Our data demonstrate that nontoxic concentrations of acrolein significantly inhibited IFNalpha-induced tyrosine phosphorylation of both cytoplasmic and nuclear STAT1 and STAT2, without altering the total levels. Also, acrolein down-regulated IFNalpha-stimulated gene transcription, resulting in reduced expression of antiviral genes. Importantly, acrolein abolished the IFNalpha-mediated down-regulation of HCV viral expression in the HCV-replicon system. This study defines mechanisms involved in resistance to IFNalpha and identifies the pathogenic role of acrolein, and potentially other environmental pollutants, in suppressing IFNalpha antiviral activity and establishes their adverse impact on HCV therapy.
Collapse
Affiliation(s)
- Swati Joshi-Barve
- Department of Medicine, University of Louisville Medical Center
- Department of Pharmacology & Toxicology, University of Louisville Medical Center
| | | | - Madhuvanti Patil
- Department of Pharmacology & Toxicology, University of Louisville Medical Center
| | - Aruni Bhatnagar
- Department of Medicine, University of Louisville Medical Center
- Department of Pharmacology & Toxicology, University of Louisville Medical Center
| | - Stephanie Mathews
- Department of Pharmacology & Toxicology, University of Louisville Medical Center
| | | | - Matthew Cave
- Department of Medicine, University of Louisville Medical Center
- Louisville VA Medical Center, Louisville, Kentucky
| | - Craig McClain
- Department of Medicine, University of Louisville Medical Center
- Department of Pharmacology & Toxicology, University of Louisville Medical Center
- Louisville VA Medical Center, Louisville, Kentucky
| | - Shirish Barve
- Department of Medicine, University of Louisville Medical Center
- Department of Pharmacology & Toxicology, University of Louisville Medical Center
| |
Collapse
|
43
|
Fernandes AF, Bian Q, Jiang JK, Thomas CJ, Taylor A, Pereira P, Shang F. Proteasome inactivation promotes p38 mitogen-activated protein kinase-dependent phosphatidylinositol 3-kinase activation and increases interleukin-8 production in retinal pigment epithelial cells. Mol Biol Cell 2009; 20:3690-9. [PMID: 19570915 DOI: 10.1091/mbc.e08-10-1068] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Oxidative stress and inflammation are implicated in the pathogenesis of many age-related diseases. We have demonstrated previously that oxidative inactivation of the proteasome is a molecular link between oxidative stress and overexpression of interleukin (IL)-8. Here, we elucidated a novel signaling cascade that leads to up-regulation of IL-8 in response to proteasome inactivation. The sequence of events in this cascade includes proteasome inactivation, activation of mitogen-activated protein kinase kinase (MKK)3/MKK6, activation of p38 mitogen-activated protein kinase (MAPK), epidermal growth factor receptor phosphorylation, phosphatidylinositol 3-kinase (PI3K) activation and increased IL-8 expression. Blocking any of these signaling pathways abolished the up-regulation of IL-8 induced by proteasome inhibition. Although Akt is also activated in response to proteasome inactivation, we found that the PI3K-dependent up-regulation of IL-8 is independent of 3-phosphoinositide-dependent protein kinase (PDK)1 and Akt. Inhibition of PDK1 and Akt with chemical inhibitors or expression of constitutive active Akt had little effects on IL-8 expression in response to proteasome inactivation. In contrast, inhibition of interleukin 2-inducible T cell kinase, a kinase downstream of PI3K, significantly reduced the expression and secretion of IL-8 in response to proteasome inactivation. Together, these data elucidate a novel signaling network that leads to increased IL-8 production in response to proteasome inactivation.
Collapse
Affiliation(s)
- Alexandre F Fernandes
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Alcohol ingestion causes alteration in several cellular mechanisms, and leads to inflammation, apoptosis, immunological response defects, and fibrosis. These phenomena are associated with significant changes in the epigenetic mechanisms, and subsequently, to liver cell memory. The ubiquitin-proteasome pathway is one of the vital pathways in the cell that becomes dysfunctionial as a result of chronic ethanol consumption. Inhibition of the proteasome activity in the nucleus causes changes in the turnover of transcriptional factors, histone modifying enzymes, and therefore, affects epigenetic mechanisms. Alcohol consumption has been associated with an increase in histone acetylation and a decrease in histone methylation, which leads to gene expression changes. DNA and histone modifications that result from ethanol-induced proteasome inhibition are key players in regulating gene expression, especially genes involved in the cell cycle, immunological responses, and metabolism of ethanol. The present review highlights the consequences of ethanol-induced proteasome inhibition in the nucleus of liver cells that are chronically exposed to ethanol.
Collapse
|
45
|
Ferré N, Martínez-Clemente M, López-Parra M, González-Périz A, Horrillo R, Planagumà A, Camps J, Joven J, Tres A, Guardiola F, Bataller R, Arroyo V, Clària J. Increased susceptibility to exacerbated liver injury in hypercholesterolemic ApoE-deficient mice: potential involvement of oxysterols. Am J Physiol Gastrointest Liver Physiol 2009; 296:G553-62. [PMID: 19136384 DOI: 10.1152/ajpgi.00547.2007] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The contribution of metabolic factors to the severity of liver disease is not completely understood. In this study, apolipoprotein E-deficient (ApoE-/-) mice were evaluated to define potential effects of hypercholesterolemia on the severity of carbon tetrachloride (CCl4)-induced liver injury. Under baseline conditions, hypercholesterolemic ApoE-/- mice showed increased hepatic oxidative stress (SOD activity/4-hydroxy-2-nonenal immunostaining) and higher hepatic TGF-beta1, MCP-1, and TIMP-1 expression than wild-type control mice. After CCl4 challenge, ApoE-/- mice exhibited exacerbated steatosis (Oil Red O staining), necroinflammation (hematoxylin-eosin staining), macrophage infiltration (F4/80 immunohistochemistry), and fibrosis (Sirius red staining and alpha-smooth muscle actin immunohistochemistry) and more severe liver injury [alanine aminotransferase (ALT) and aspartate aminotransferase] than wild-type controls. Direct correlations were identified between serum cholesterol and hepatic steatosis, fibrosis, and ALT levels. These changes did not reflect the usual progression of the disease in ApoE-/- mice, since exacerbated liver injury was not present in untreated age-paired ApoE-/- mice. Moreover, hepatic cytochrome P-450 expression was unchanged in ApoE-/- mice. To explore potential mechanisms, cell types relevant to liver pathophysiology were exposed to selected cholesterol-oxidized products. Incubation of hepatocytes with a mixture of oxysterols representative of those detected by GC-MS in livers from ApoE-/- mice resulted in a concentration-dependent increase in total lipoperoxides and SOD activity. In hepatic stellate cells, oxysterols increased IL-8 secretion through a NF-kappaB-independent mechanism and upregulated TIMP-1 expression. In macrophages, oxysterols increased TGF-beta1 secretion and MCP-1 expression in a concentration-dependent manner. Oxysterols did not compromise cell viability. Taken together, these findings demonstrate that hypercholesterolemic mice are sensitized to liver injury and that cholesterol-derived products (i.e., oxysterols) are able to induce proinflammatory and profibrogenic mechanisms in liver cells.
Collapse
Affiliation(s)
- Natàlia Ferré
- Department of Biochemistry and Molecular Genetics, Liver Unit, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Sadanari H, Tanaka J, Li Z, Yamada R, Matsubara K, Murayama T. Proteasome inhibitor differentially regulates expression of the major immediate early genes of human cytomegalovirus in human central nervous system-derived cell lines. Virus Res 2009; 142:68-77. [PMID: 19201384 DOI: 10.1016/j.virusres.2009.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Revised: 01/13/2009] [Accepted: 01/13/2009] [Indexed: 10/21/2022]
Abstract
Proteasome inhibitor, which inhibits NF-kappaB activation, has been reported to activate c-Jun N-terminal kinase (JNK)-c-Jun pathway. In this study, we investigated the effects of proteasome inhibitor on the human cytomegalovirus (HCMV) major immediate early (MIE) gene expression in human central nervous system (CNS)-derived cell lines. Treatment of HCMV-infected 118MGC glioma and U373-MG astrocytoma cells with three proteasome inhibitors, MG132, clasto-lactacystin beta-lactone, and epoxomicin, suppressed MIE protein expression. In contrast, in HCMV-infected IMR-32 neuroblastoma cells, the proteasome inhibitors increased MIE protein expression, even in the presence of NF-kappaB inhibitor SN-50. A luciferase reporter assay demonstrated that MG132 markedly elevated the MIE promoter/enhancer (MIEP) activity in IMR-32 cells, but down-regulated it in 118MGC and U373-MG cells. Mutation in five cAMP response elements (CREs) within the MIEP resulted in a loss of the ability to respond to MG132 in IMR-32 cells. Moreover, Western blotting analysis revealed that MG132 induced c-Jun phosphorylation in all three CNS-derived cell lines, whereas a high level of activating transcription factor-2 (ATF-2) phosphorylation was observed only in IMR-32 cells. Finally, MG132-induced MIE protein expression was suppressed by JNK inhibitor that reduced the phosphorylation levels of both c-Jun and ATF-2. Taken together, these results suggest that the proteasome inhibitors activate CRE binding proteins consisting of c-Jun and ATF-2 through activating the JNK-c-Jun pathway, thereby inducing MIE protein synthesis in IMR-32 cells under the condition where NF-kappaB activity is inhibited.
Collapse
Affiliation(s)
- Hidetaka Sadanari
- Department of Microbiology and Immunology, Faculty of Pharmaceutical Sciences, Hokuriku University, Ho-3 Kanagawa-machi, Kanazawa, Ishikawa 920-1181, Japan.
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
The NF-kappaB signaling pathway has particular relevance to several liver diseases including hepatitis (liver infection by Helicobacter, viral hepatitis induced by HBV and HCV), liver fibrosis and cirrhosis and hepatocellular carcinoma. Furthermore, the NF-kappaB signaling pathway is a potential target for development of hepatoprotective agents. Several types of drugs including: selective estrogen receptor modulators (SERMs), antioxidants, proteasome inhibitors, IKK inhibitors and nucleic acid-based decoys have been shown to interfere with NF-kappaB activity at different levels and may be useful for the treatment of liver diseases. However, NF-kappaB also plays an important hepatoprotective function that needs to be taken into consideration during development of new therapeutic regimens.
Collapse
Affiliation(s)
- B Sun
- Liver Transplantation Center of the First Affiliated Hospital and Cancer Center, Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | | |
Collapse
|
48
|
Fernandes AF, Zhou J, Zhang X, Bian Q, Sparrow J, Taylor A, Pereira P, Shang F. Oxidative inactivation of the proteasome in retinal pigment epithelial cells. A potential link between oxidative stress and up-regulation of interleukin-8. J Biol Chem 2008; 283:20745-53. [PMID: 18502748 PMCID: PMC2475710 DOI: 10.1074/jbc.m800268200] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 04/04/2008] [Indexed: 12/19/2022] Open
Abstract
Oxidative stress and inflammation are implicated in the pathogenesis of many age-related diseases. Stress-induced overproduction of inflammatory cytokines, such as interleukin-8 (IL-8), is one of the early events of inflammation. The objective of this study was to elucidate mechanistic links between oxidative stress and overproduction of IL-8 in retinal pigment epithelial (RPE) cells. We found that exposure of RPE cells to H(2)O(2), paraquat, or A2E-mediated photooxidation resulted in increased expression and secretion of IL-8. All of these oxidative stressors also inactivated the proteasome in RPE cells. In contrast, tert-butylhydroperoxide (TBH), a lipophilic oxidant that did not stimulate IL-8 production, also did not inactivate the proteasome. Moreover, prolonged treatment of RPE cells with proteasome-specific inhibitors recapitulated the stimulation of IL-8 production. These data suggest that oxidative inactivation of the proteasome is a potential mechanistic link between oxidative stress and up-regulation of the proinflammatory IL-8. The downstream signaling pathways that govern the production of IL-8 include NF-kappaB and p38 MAPK. Proteasome inhibition both attenuated the activation and delayed the turnoff of NF-kappaB, resulting in biphasic effects on the production of IL-8. Prolonged proteasome inhibition (>2 h) resulted in activation of p38 MAPK via activation of MKK3/6 and increased the production of IL-8. Chemically inhibiting the p38 MAPK blocked the proteasome inhibition-induced up-regulation of IL-8. Together, these data indicate that oxidative inactivation of the proteasome and the related activation of the p38 MAPK pathway provide a potential link between oxidative stress and overproduction of proinflammatory cytokines, such as IL-8.
Collapse
Affiliation(s)
- Alexandre F. Fernandes
- Jean Mayer United States Department of
Agriculture Human Nutrition Research Center on Aging, Tufts University,
Boston, Massachusetts 02111, the Center of
Ophthalmology, IBILI, Faculty of Medicine, University of Coimbra,
3000–354 Portugal, and the Department of
Ophthalmology, Columbia University, New York, New York 10032
| | - Jilin Zhou
- Jean Mayer United States Department of
Agriculture Human Nutrition Research Center on Aging, Tufts University,
Boston, Massachusetts 02111, the Center of
Ophthalmology, IBILI, Faculty of Medicine, University of Coimbra,
3000–354 Portugal, and the Department of
Ophthalmology, Columbia University, New York, New York 10032
| | - Xinyu Zhang
- Jean Mayer United States Department of
Agriculture Human Nutrition Research Center on Aging, Tufts University,
Boston, Massachusetts 02111, the Center of
Ophthalmology, IBILI, Faculty of Medicine, University of Coimbra,
3000–354 Portugal, and the Department of
Ophthalmology, Columbia University, New York, New York 10032
| | - Qingning Bian
- Jean Mayer United States Department of
Agriculture Human Nutrition Research Center on Aging, Tufts University,
Boston, Massachusetts 02111, the Center of
Ophthalmology, IBILI, Faculty of Medicine, University of Coimbra,
3000–354 Portugal, and the Department of
Ophthalmology, Columbia University, New York, New York 10032
| | - Janet Sparrow
- Jean Mayer United States Department of
Agriculture Human Nutrition Research Center on Aging, Tufts University,
Boston, Massachusetts 02111, the Center of
Ophthalmology, IBILI, Faculty of Medicine, University of Coimbra,
3000–354 Portugal, and the Department of
Ophthalmology, Columbia University, New York, New York 10032
| | - Allen Taylor
- Jean Mayer United States Department of
Agriculture Human Nutrition Research Center on Aging, Tufts University,
Boston, Massachusetts 02111, the Center of
Ophthalmology, IBILI, Faculty of Medicine, University of Coimbra,
3000–354 Portugal, and the Department of
Ophthalmology, Columbia University, New York, New York 10032
| | - Paulo Pereira
- Jean Mayer United States Department of
Agriculture Human Nutrition Research Center on Aging, Tufts University,
Boston, Massachusetts 02111, the Center of
Ophthalmology, IBILI, Faculty of Medicine, University of Coimbra,
3000–354 Portugal, and the Department of
Ophthalmology, Columbia University, New York, New York 10032
| | - Fu Shang
- Jean Mayer United States Department of
Agriculture Human Nutrition Research Center on Aging, Tufts University,
Boston, Massachusetts 02111, the Center of
Ophthalmology, IBILI, Faculty of Medicine, University of Coimbra,
3000–354 Portugal, and the Department of
Ophthalmology, Columbia University, New York, New York 10032
| |
Collapse
|
49
|
Lu Y, Cederbaum AI. CYP2E1 and oxidative liver injury by alcohol. Free Radic Biol Med 2008; 44:723-38. [PMID: 18078827 PMCID: PMC2268632 DOI: 10.1016/j.freeradbiomed.2007.11.004] [Citation(s) in RCA: 582] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2007] [Revised: 11/02/2007] [Accepted: 11/05/2007] [Indexed: 12/11/2022]
Abstract
Ethanol-induced oxidative stress seems to play a major role in mechanisms by which ethanol causes liver injury. Many pathways have been suggested to contribute to the ability of ethanol to induce a state of oxidative stress. One central pathway seems to be the induction of cytochrome P450 2E1 (CYP2E1) by ethanol. CYP2E1 metabolizes and activates many toxicological substrates, including ethanol, to more reactive, toxic products. Levels of CYP2E1 are elevated under a variety of physiological and pathophysiological conditions and after acute and chronic alcohol treatment. CYP2E1 is also an effective generator of reactive oxygen species such as the superoxide anion radical and hydrogen peroxide and, in the presence of iron catalysts, produces powerful oxidants such as the hydroxyl radical. This review article summarizes some of the biochemical and toxicological properties of CYP2E1 and briefly describes the use of cell lines developed to constitutively express CYP2E1 and CYP2E1 knockout mice in assessing the actions of CYP2E1. Possible therapeutic implications for treatment of alcoholic liver injury by inhibition of CYP2E1 or CYP2E1-dependent oxidative stress will be discussed, followed by some future directions which may help us to understand the actions of CYP2E1 and its role in alcoholic liver injury.
Collapse
Affiliation(s)
- Yongke Lu
- Department of Pharmacology and Systems Therapeutics, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | |
Collapse
|
50
|
Cytokine and Chemokine Expression Associated with Steatohepatitis and Hepatocyte Proliferation in Rats Fed Ethanol via Total Enteral Nutrition. Exp Biol Med (Maywood) 2008; 233:344-55. [DOI: 10.3181/0707-rm-203] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
To determine the temporal relationship between alcohol-induced changes in cytokines and chemokines, development of liver pathology and stimulation of hepatocyte proliferation, male Sprague-Dawley rats were intragastrically fed low carbohydrate-containing ethanol (EtOH) diets via total enteral nutrition (TEN) for up to 49 d. Induction of EtOH metabolism and appearance of steatosis preceded development of oxidative stress, inflammation, and cell death. A transitory peak of tumor necrosis factor (TNFα) and interferon gamma (IFNγ) was observed at 14 d followed by reduced expression of TNFα, IFNγ and another Th1 cytokine IL-12 accompanied by reduced expression of the Th1 regulators T-bet and STAT4. After 35–49 d of EtOH, at a time when hepatocyte proliferation was stimulated, IL-12 returned to control values and a second peak of TNFα occurred. The Th2 cytokine IL-4 remained suppressed throughout the study and was accompanied by reductions in the Th2 regulator GATA3. There was no temporal effect of EtOH on expression of IL-6 or TGFβ. IL-5 and IL-13 mRNA were undetectable. Chemokine CXCL-2 expression increased progressively up to 35 d and preceded the appearance of inflammatory infiltrates. These data suggest that steatosis, increased ethanol metabolism, a transient induction of the innate immune response and suppression of Th2 responses were acute consequences of ethanol treatment and were followed by suppression of Th1 responses. However, the majority of necrosis, apoptosis and a late peak of TNFα only occurred after 6–7 weeks of ethanol, coincided with the appearance of inflammatory infiltrates and were associated with stimulation of hepatocyte proliferation.
Collapse
|