401
|
MRI-based texture analysis: a potential technique to assess protectors against induced-liver fibrosis in rats. Radiol Oncol 2009. [DOI: 10.2478/v10019-009-0006-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
402
|
Abstract
Hepatic fibrosis is a scarring process associated with an increased and altered deposition of extracellular matrix in the liver. It is caused by a variety of stimuli and if fibrosis continues unopposed, it would progress to cirrhosis which poses a significant health problem worldwide. At the cellular and molecular level, this progressive process is characterized by cellular activation of hepatic stellate cells and aberrant activity of transforming growth factor-beta with its downstream cellular mediators. Liver biopsy has been the reference test for assessment of hepatic fibrosis, but because of its limitations, noninvasive markers of liver fibrosis were developed. Liver fibrosis or cirrhosis was considered irreversible in the past but progress of research on the molecular pathogenesis of liver fibrosis has shown that hepatic cellular recovery is possible. Currently, no acceptable therapeutic strategies exist, other than removal of the fibrogenic stimulus, to treat this potentially devastating disease.
Collapse
Affiliation(s)
- Mona H Ismail
- Department of Internal Medicine, Division of Gastroenterology at King Fahad Hospital of the University, Al-Khobar, Saudi Arabia.
| | | |
Collapse
|
403
|
Noyan-Ashraf MH, Sadeghinejad Z, Davies GF, Ross AR, Saucier D, Harkness TAA, Juurlink BHJ. Phase 2 protein inducers in the diet promote healthier aging. J Gerontol A Biol Sci Med Sci 2008; 63:1168-76. [PMID: 19038831 DOI: 10.1093/gerona/63.11.1168] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Oxidative stress drives many aging-associated problems. Because oxidative stress can be decreased by induction of phase 2 proteins, we hypothesized that incorporating the phase 2 protein inducer 2(3)-tert-butyl-4-hydroxyanisole (tBHA) into the diet would result in healthier aging. C57BL/6 mice were placed either on control mouse chow diet or on chow containing tBHA and were examined at 6, 12, and 18 months. Dietary tBHA resulted in the antioxidant response activation, decreased both oxidative stress and pro-inflammatory gene expression in tissues examined, counteracted the decrease in the transcription factors peroxisome proliferator-activated receptor-gamma and increase in CCAAT/enhancer binding protein-alpha levels seen in liver with aging, and was associated with mice having less weight gain, despite having no differences in food consumption, and better locomotor function. We conclude that simple changes in the diet such as incorporation of phase 2 protein inducers can have a profound influence on health and, thereby, the aging process.
Collapse
|
404
|
Shyu MH, Kao TC, Yen GC. Hsian-tsao (Mesona procumbens Heml.) prevents against rat liver fibrosis induced by CCl(4) via inhibition of hepatic stellate cells activation. Food Chem Toxicol 2008; 46:3707-13. [PMID: 18929613 DOI: 10.1016/j.fct.2008.09.051] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2008] [Revised: 08/04/2008] [Accepted: 09/22/2008] [Indexed: 02/05/2023]
Abstract
In this study, the protective effect of extract of Hsian-tsao (Mesona procumbens) (EHT) against liver fibrogenesis in carbon tetrachloride (CCl(4))-injured rats was evaluated. The inhibitory effect of oleanolic acid (OA) and ursolic acid (UA), which are the active compounds in EHT, on the activation of hepatic stellate cells (HSC) was also determined. The results showed that EHT at a dosage of 1.2g/kg of b.w. significantly reduced the liver injury induced by CCl(4) in rats. It also decreased the activity of serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) and the deposition of collagen in the liver. Oral administration of EHT reduced the levels of alpha-smooth muscle actin (alpha-SMA) and the activity of metalloproteinases (MMPs) in rats injured by treatment with CCl(4). In addition, we performed experiments with the rat hepatic stellate cell line HSC-T6 in which we induced the expression of MMP-2 and alpha-SMA with phorbol-12-myristate-13-acetate (PMA). Treating these cells with OA (20microM) or UA (10microM) caused a decrease in the levels of both proteins. Taken together, our data indicate that EHT can efficiently inhibit CCl(4)-induced liver fibrosis in rats. EHT may therefore be a useful functional food for preventing liver fibrosis.
Collapse
Affiliation(s)
- Ming-Huan Shyu
- Department of Food Science and Biotechnology, National Chung Hsing University, 250, Kuokuang Road, Taiching 402, Taiwan
| | | | | |
Collapse
|
405
|
Rezaei A, Ardestani SK, Forouzandeh M, Tavangar SM, Khorramizadeh MR, Payabvash S, Nezami BG, Jahanshiri Z, Tavakoli Z, Shariftabrizi A, Dehpour AR. The effects of N-acetylcysteine on the expression of matrix metalloproteinase-2 and tissue inhibitor of matrix metalloproteinase-2 in hepatic fibrosis in bile duct ligated rats. Hepatol Res 2008; 38:1252-63. [PMID: 18713279 DOI: 10.1111/j.1872-034x.2008.00393.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AIM N-acetylcysteine can inhibit the formation of intracellular reactive oxygen intermediates. Cellular redox state plays a role in regulating the secretion of matrix metalloproteinase-2. We investigated the effects of N-acetylcysteine on the expression of matrix metalloproteinase-2 and tissue inhibitor of matrix metalloproteinase-2. METHODS Bile duct ligated rats were used as a model of hepatic fibrosis. We compared the level of gene expression (using real-time reverse transcription polymerase chain reaction [RT-PCR]), liver function parameters, hepatic reactive oxygen production, lipid peroxidation and glutathione state in experimental groups. RESULTS N-acetylcysteine treatment significantly improved liver function parameters including the plasma levels of aspartate aminotransferase, alkaline phosphatase, gamma-glutamyl transpeptidase and bilirubin. In addition, significant improvement of glutathione state and reactive oxygen production were observed. Hepatic lipid peroxidation was reversed by N-acetylcysteine treatment. Although N-acetylcysteine treatment did not completely normalize the increased matrix metalloproteinase-2 expression, it significantly decreased its level by 65%. N-acetylcysteine treatment also significantly decreased matrix metalloproteinase-2 activity and normalized tissue inhibitor of matrix metalloproteinase-2 expression. CONCLUSION Collectively, N-acetylcysteine showed inhibition of matrix metalloproteinase-2 expression and activity. In addition, administration of N-acetylcysteine was associated with downregulation of the expression of tissue inhibitor of matrix metalloproteinase-2 and amelioration of oxidative stress in the liver of bile duct ligated rats.
Collapse
Affiliation(s)
- Arezou Rezaei
- Institute of Biochemistry and Biophysics, Tehran University, Tehran, Iran
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
406
|
Abstract
Uncontrolled production of collagen I is the main feature of liver fibrosis. Following a fibrogenic stimulus such as alcohol, hepatic stellate cells (HSC) transform into an activated collagen-producing cell. In alcoholic liver disease, numerous changes in gene expression are associated with HSC activation, including the induction of several intracellular signaling cascades, which help maintain the activated phenotype and control the fibrogenic and proliferative state of the cell. Detailed analyses for understanding the molecular basis of the collagen I gene regulation have revealed a complex process involving reactive oxygen species (ROS) as key mediators. Less is known, however, about the contribution of reactive nitrogen species (RNS). In addition, a series of cytokines, growth factors, and chemokines, which activate extracellular matrix (ECM)-producing cells through paracrine and autocrine loops, contribute to the fibrogenic response.
Collapse
Affiliation(s)
- R. Urtasun
- Mount Sinai School of Medicine, Box 1123, Department of Medicine/Division of Liver Diseases, 1425 Madison Avenue, Room 11-76, New York, NY 10029, USA
| | - L. Conde de la Rosa
- Mount Sinai School of Medicine, Box 1123, Department of Medicine/Division of Liver Diseases, 1425 Madison Avenue, Room 11-76, New York, NY 10029, USA
| | - N. Nieto
- Mount Sinai School of Medicine, Box 1123, Department of Medicine/Division of Liver Diseases, 1425 Madison Avenue, Room 11-76, New York, NY 10029, USA
| |
Collapse
|
407
|
Novo E, Parola M. Redox mechanisms in hepatic chronic wound healing and fibrogenesis. FIBROGENESIS & TISSUE REPAIR 2008; 1:5. [PMID: 19014652 PMCID: PMC2584013 DOI: 10.1186/1755-1536-1-5] [Citation(s) in RCA: 243] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2008] [Accepted: 10/13/2008] [Indexed: 12/24/2022]
Abstract
Reactive oxygen species (ROS) generated within cells or, more generally, in a tissue environment, may easily turn into a source of cell and tissue injury. Aerobic organisms have developed evolutionarily conserved mechanisms and strategies to carefully control the generation of ROS and other oxidative stress-related radical or non-radical reactive intermediates (that is, to maintain redox homeostasis), as well as to 'make use' of these molecules under physiological conditions as tools to modulate signal transduction, gene expression and cellular functional responses (that is, redox signalling). However, a derangement in redox homeostasis, resulting in sustained levels of oxidative stress and related mediators, can play a significant role in the pathogenesis of major human diseases characterized by chronic inflammation, chronic activation of wound healing and tissue fibrogenesis. This review has been designed to first offer a critical introduction to current knowledge in the field of redox research in order to introduce readers to the complexity of redox signalling and redox homeostasis. This will include ready-to-use key information and concepts on ROS, free radicals and oxidative stress-related reactive intermediates and reactions, sources of ROS in mammalian cells and tissues, antioxidant defences, redox sensors and, more generally, the major principles of redox signalling and redox-dependent transcriptional regulation of mammalian cells. This information will serve as a basis of knowledge to introduce the role of ROS and other oxidative stress-related intermediates in contributing to essential events, such as the induction of cell death, the perpetuation of chronic inflammatory responses, fibrogenesis and much more, with a major focus on hepatic chronic wound healing and liver fibrogenesis.
Collapse
Affiliation(s)
- Erica Novo
- Dipartimento di Medicina e Oncologia Sperimentale and Centro Interuniversitario di Fisiopatologia Epatica, Università degli Studi di Torino, Corso Raffaello 30, 10125 Torino, Italy
| | | |
Collapse
|
408
|
The Nrf2 transcription factor protects from toxin-induced liver injury and fibrosis. J Transl Med 2008; 88:1068-78. [PMID: 18679376 DOI: 10.1038/labinvest.2008.75] [Citation(s) in RCA: 165] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The liver is frequently exposed to insults, including toxic chemicals and alcohol, viral infection or metabolic overload. Although it can fully regenerate after acute injury, chronic liver damage causes liver fibrosis and cirrhosis, which can result in complete liver failure. In this study, we demonstrate that the NF-E2-related factor 2 (Nrf2) transcription factor protects the liver from acute and chronic toxin-mediated damage. Repair of the liver injury that occurs after a single treatment with the hepatotoxin carbon tetrachloride (CCl(4)) was severely delayed in Nrf2-deficient mice. The defect in repair was accompanied by an enhanced and prolonged inflammatory and profibrotic response. After long-term CCl(4) treatment, liver fibrosis was strongly aggravated in the Nrf2 knockout mice and inflammation was enhanced. We demonstrate that these abnormalities are at least in part due to the reduced expression of known and novel Nrf2 target genes in hepatocytes, which encode enzymes involved in the detoxification of CCl(4) and its metabolites. These results suggest that activation of Nrf2 may be a novel strategy to prevent or ameliorate toxin-induced liver injury and fibrosis.
Collapse
|
409
|
Svegliati-Baroni G, De Minicis S, Marzioni M. Hepatic fibrogenesis in response to chronic liver injury: novel insights on the role of cell-to-cell interaction and transition. Liver Int 2008; 28:1052-64. [PMID: 18783548 DOI: 10.1111/j.1478-3231.2008.01825.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
Hepatic fibrosis represents the wound-healing response process of the liver to chronic injury, independently from aetiology. Advanced liver fibrosis results in cirrhosis that can lead to liver failure, portal hypertension and hepatocellular carcinoma. Currently, no effective therapies are available for hepatic fibrosis. After the definition of hepatic stellate cells (HSCs) as the main liver extracellular matrix-producing cells in the 1980s, the subsequent decade was dedicated to determine the role of specific cytokines and growth factors. Fibrotic progression of chronic liver diseases can be nowadays considered as a dynamic and highly integrated process of cellular response to chronic liver injury. The present review is dedicated to the novel mechanisms of cellular response to chronic liver injury leading to hepatic myofibroblasts' activation. The understanding of the cellular and molecular pathways regulating their function is crucial to counteract therapeutically the organ dysfunction caused by myofibroblasts' activation.
Collapse
Affiliation(s)
- Gianluca Svegliati-Baroni
- Department of Gastroenterology, Università Politecnica delle Marche and Ospedali Riuniti University Hospital, Ancona, Italy.
| | | | | |
Collapse
|
410
|
Piemonte F, Petrini S, Gaeta LM, Tozzi G, Bertini E, Devito R, Boldrini R, Marcellini M, Ciacco E, Nobili V. Protein glutathionylation increases in the liver of patients with non-alcoholic fatty liver disease. J Gastroenterol Hepatol 2008; 23:e457-64. [PMID: 17683488 DOI: 10.1111/j.1440-1746.2007.05070.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND AND AIM Oxidative stress is an important pathophysiological mechanism in non-alcoholic steatohepatitis, where hepatocyte apoptosis is significantly increased correlating with disease severity. Protein glutathionylation occurs as a response to oxidative stress, where an increased concentration of oxidized glutathione modifies post-translational proteins by thiol disulfide exchange. In this study, we analyzed the protein glutathionylation in non-alcoholic fatty liver disease (NAFLD) and evaluated a potential association between glutathionylation, fibrosis, and vitamin E treatment. METHODS Protein glutathionylation was studied in the livers of 36 children (mean age 12.5 years, range 4-16 years) subdivided into three groups according to their NAFLD activity score (NAS) by Western blot analysis and immunohistochemistry, using a specific monoclonal antibody. In addition, we identified the hepatocyte ultrastructures involved in glutathionylation by immunogold electron microscopy. RESULTS Our findings showed that protein glutathionylation increases in the livers of patients with NAFLD and it is correlated with steatohepatitis and liver fibrosis. Its increase appears mainly in nuclei and cytosol of hepatocytes, and it is reversed by antioxidant therapy with reduced fibrosis. CONCLUSION Protein glutathionylation significantly increases in livers with NAFLD, strongly suggesting that oxidative injury plays a crucial role in this disease. Furthermore, the marked increase of protein glutathionylation, in correlation with collagen VI immunoreactivity, suggests a link between the redox status of hepatic protein thiols and fibrosis.
Collapse
Affiliation(s)
- Fiorella Piemonte
- Molecular Medicine Unit, Chemistry Laboratory, Bambino Gesù Children's Hospital and Research Institute, Rome, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
411
|
Abstract
Steatosis is a common feature of many liver diseases, namely non-alcoholic steatohepatitis (NASH) and hepatitis C virus (HCV) infection, but the pathogenic mechanisms differ. Insulin resistance (IR), a key feature of metabolic syndrome, is crucial for NASH development, associated with many underlying genetically determined or acquired mitochondrial and metabolic defects and culminates to inflammation and progression to fibrosis. This may have potential implications for new drug therapy. In HCV-related disease, steatosis impacts both fibrosis progression and response to treatment. Steatosis in HCV-related disease relates to both viral factors (HCV genotype 3), and host factors (alcohol consumption, overweight, hyperlipidemia, diabetes). Among others, IR is a recognized factor. Hepatic steatosis is reported to be associated with disturbance in the signaling cascade of interferon and downregulation of its receptors. Thus, hepatic steatosis should not be considered a benign feature, but rather a silent killer.
Collapse
|
412
|
Aslan M, Horoz M, Nazligul Y, Bolukbas C, Bolukbas FF, Selek S, Aksoy N, Erel O. Serum paraoxonase and arylesterase activities for the evaluation of patients with chronic hepatitis. Int J Clin Pract 2008; 62:1050-5. [PMID: 17887991 DOI: 10.1111/j.1742-1241.2006.01206..x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
The sensitivity of standard biochemical tests for liver function is low and insufficient for a reliable determination of the presence or absence of liver disease. The aim of the present study was to investigate serum paraoxonase and arylesterase activities and lipid hydroperoxide (LOOH) levels, and to find out that whether the measurement of serum paraoxonase and arylesterase activities would be useful as an index of liver function status in chronic hepatitis (CH). Fourty-four patients with CH (24 CHB and 20 CHC) and 38 controls were enrolled. Serum paraoxonase and arylesterase activities were detected spectrophotometrically. LOOH levels were measured by the FOX-2 assay. Serum paraoxonase and arylesterase activities were significantly lower in patients with CH than controls (p < 0.001 for both), while LOOH levels were significantly higher (p < 0.001). Paraoxonase and arylesterase activities were inversely correlated with LOOH levels (r = -0.394, p < 0.05; r =-0.362, p < 0.05, respectively). Fibrosis scores of CH patients were significantly correlated with paraoxonase and arylesterase activities and LOOH levels (r =-0.276, p < 0.05; r = -0.583, p < 0.001 and r = 0.562, p < 0.001, respectively). Our results indicated that decrease in the activities paraoxonase and arylesterase may play a role in the pathogenesis of CH. In addition, serum paraoxonase and arylesterase activities measurement may add a significant contribution to the liver function tests.
Collapse
Affiliation(s)
- M Aslan
- Department of Internal Medicine, School of Medicine, Harran University, Sanliurfa, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|
413
|
Othman AA, Shoheib ZS, Abdel-Aleem GA, Shareef MM. Experimental schistosomal hepatitis: protective effect of coenzyme-Q10 against the state of oxidative stress. Exp Parasitol 2008; 120:147-55. [PMID: 18640114 DOI: 10.1016/j.exppara.2008.06.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Revised: 06/09/2008] [Accepted: 06/20/2008] [Indexed: 01/22/2023]
Abstract
Schistosoma mansoni (S. mansoni) eggs trapped in the host liver elicit a chain of oxidative processes that may be, at least in part, responsible for the pathology and progression of fibrosis associated with schistosomal hepatitis. This study was designed to assess the protective effect of the antioxidant coenzyme-Q10 (Co-Q10) against experimental S. mansoni-induced oxidative stress in the liver, and its potential role as an adjuvant to praziquantel (PZQ) therapy. The oxidative stress and overall liver function were improved under Co-Q10 therapy as evidenced by significant reduction in oxidative stress markers and preservation of antioxidant factors. Liver fibrosis was also reduced with a positive impact on liver function. Moreover, addition of Co-Q10 to PZQ therapy caused: significant reduction of liver egg load, significant improvement of the redox status, and lastly decreased liver fibrosis.
Collapse
Affiliation(s)
- Ahmad A Othman
- Department of Parasitology, Faculty of Medicine, Tanta University, Tanta, Egypt.
| | | | | | | |
Collapse
|
414
|
Yang YY, Lee KC, Huang YT, Wang YW, Hou MC, Lee FY, Lin HC, Lee SD. Effects of N-acetylcysteine administration in hepatic microcirculation of rats with biliary cirrhosis. J Hepatol 2008; 49:25-33. [PMID: 18490076 DOI: 10.1016/j.jhep.2008.02.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2007] [Revised: 01/16/2008] [Accepted: 02/06/2008] [Indexed: 12/17/2022]
Abstract
BACKGROUND/AIMS Increased intrahepatic resistance (IHR) in cirrhosis is due to fibrosis and hepatic endothelial dysfunction (HED). Besides producing fibrosis, increased reactive oxygen species (ROS) promotes ROS-related nitration of anti-oxidative enzymes in cirrhotic livers. Tyrosine nitration (nitrotyrosilation)-related inactivation of anti-oxidative enzymes is increased in cirrhotic livers. This study investigates effects of N-acetylcysteine (NAC) administrations in bile-duct-ligation (BDL) rats. METHODS This study measured portal venous pressure (PVP), IHR, hepatic endothelial function, hepatic levels of anti-oxidants and oxidants, type III procollagen (PIIIP), proteins expression of thromboxane synthase (TXS), nitrotyrosine, manganese superoxide dismutase (MnSOD), and hepatic NOx and thromboxane A(2) (TXA(2)) production in perfusates. RESULTS The improvement of HED was associated with decreased PVP and IHR, hepatic protein and mRNA levels of PIIIP, protein expression of TXS and nitrotyrosine, oxidants and production of TXA(2) in NAC-treated BDL rat livers. Conversely, hepatic NOx production, anti-oxidants, and protein expression of MnSOD were increased in NAC-treated BDL rat livers. CONCLUSIONS In NAC-treated cirrhotic rats, the decrease in IHR was mainly caused by its anti-oxidative effect-related prevention of hepatic fibrogenesis associated with the decrease of oxidants-related nitrotyrosilation and improvement of HED.
Collapse
Affiliation(s)
- Ying-Ying Yang
- Division of General Medicine, Department of Medicine, Taipei Veterans General Hospital Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
415
|
Seok YM, Kim J, Park MJ, Boo YC, Park YK, Park KM. Wen-pi-tang-Hab-Wu-ling-san attenuates kidney fibrosis induced by ischemia/reperfusion in mice. Phytother Res 2008; 22:1057-63. [DOI: 10.1002/ptr.2440] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
416
|
Lin YL, Hsu YC, Chiu YT, Huang YT. Antifibrotic effects of a herbal combination regimen on hepatic fibrotic rats. Phytother Res 2008; 22:69-76. [PMID: 17724770 DOI: 10.1002/ptr.2265] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Liver fibrosis has been characterized as chronic inflammatory processes involving multiple molecular pathogenetic pathways. This therapeutic study investigated whether a combination regimen of Salvia miltiorrhiza (S), Ligusticum chuanxiong (L) and Glycyrrhiza glabra (G) exerted in vivo antifibrotic effects on rats with hepatic fibrosis. Fibrosis was induced in rats by dimethylnitrosamine (DMN) administration for 4 weeks. Fibrotic rats were randomly assigned to one of the three groups: control, SLG (50 mg/kg) or silymarin (50 mg/kg), each given by gavage twice daily for 3 weeks starting 1 week after DMN injection. The results showed that fibrosis scores of livers from DMN-treated rats with SLG (1.13 +/- 0.13) were significantly reduced in comparison with DMN-treated rats receiving vehicle (1.63 +/- 0.18). Moreover, the hepatic collagen content of DMN rats was significantly reduced by either SLG or silymarin treatment. The double immunohistochemical staining results also showed that alpha-SMA positive cells with NF kappa B nuclear translocation were decreased in the fibrotic livers by SLG and silymarin treatments. The mRNA expression levels of TGF-beta1, alpha-SMA, collagen1 alpha 2, iNOS and ICAM-1 genes were attenuated by SLG and silymarin treatment. The results showed that SLG exerted antifibrotic effects in rats with DMN-induced hepatic fibrosis.
Collapse
Affiliation(s)
- Yun-Lian Lin
- National Research Institute of Chinese Medicine, Taipei, Taiwan
| | | | | | | |
Collapse
|
417
|
Luo J, Yang L. Role of nicotinamide adenine dinucleotide phosphate-oxidase family in liver fibrogenesis. Shijie Huaren Xiaohua Zazhi 2008; 16:1768-1773. [DOI: 10.11569/wcjd.v16.i16.1768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
NADPH oxidase (nicotinamide adenine dinucleotide phosphate-oxidase, NOX) is a multi-protein complex producing reactive oxygen species (ROS), present both in phagocytes, being essential in host defense and in non-phagocytic cells, regulating intracellular signaling. In liver, NADPH oxidase plays a central role in fibrogenesis. A functionally active form of NADPH oxidase is expressed not only in Kupffer cells (phagocytic cell type) but also in hepatic stellate cells (HSCs) (non-phagocytic cell type), suggestive of its role the non-phagocytic NADPH oxidase in HSCs activation. This paper reviewed effects of NOX in liver fibrogenesis.
Collapse
|
418
|
|
419
|
Langer DA, Das A, Semela D, Kang-Decker N, Hendrickson H, Bronk SF, Katusic ZS, Gores GJ, Shah VH. Nitric oxide promotes caspase-independent hepatic stellate cell apoptosis through the generation of reactive oxygen species. Hepatology 2008; 47:1983-93. [PMID: 18459124 PMCID: PMC2562502 DOI: 10.1002/hep.22285] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
UNLABELLED Hepatic stellate cells (HSCs) contribute to portal hypertension through multiple mechanisms that include collagen deposition, vasoconstriction, and regulation of sinusoidal structure. Under normal physiologic conditions, endothelial nitric oxide (NO) synthase-derived NO exerts paracrine effects on HSCs; however, in cirrhosis, NO generation is impaired in association with concomitant HSC activation and changes in sinusoidal structure, events that contribute significantly to the development of portal hypertension. These concepts, in combination with recent evidence that induction of HSC-selective apoptosis may represent a useful target for treatment of chronic liver disease, led us to examine if NO may further limit HSC function through apoptosis. Indeed, both NO donors and endothelial NO synthase overexpression promoted HSC apoptotic pathways. HSC death conferred by NO occurred through mitochondrial membrane depolarization and through a caspase-independent pathway. Furthermore, NO-induced apoptosis of HSC did not occur through the canonical pathways of soluble guanylate cyclase or protein nitration, but rather through the generation of superoxide and hydroxyl radical intermediates. Lastly, HSC isolated from rats after bile duct ligation were more susceptible to NO-induced apoptosis. These data indicate that NO promotes HSC apoptosis through a signaling mechanism that involves mitochondria, is mediated by reactive oxygen species, and occurs independent of caspase activation. CONCLUSION We postulate that NO-dependent apoptosis of HSCs may maintain sinusoidal homeostasis, and may represent an additional beneficial effect of NO donors for therapy of portal hypertension.
Collapse
Affiliation(s)
- Daniel A. Langer
- Gastrointestinal Research Unit and Fiterman Center for Digestive Disease, Mayo Clinic College of Medicine, Rochester, MN
| | - Amitava Das
- Gastrointestinal Research Unit and Fiterman Center for Digestive Disease, Mayo Clinic College of Medicine, Rochester, MN
| | - David Semela
- Gastrointestinal Research Unit and Fiterman Center for Digestive Disease, Mayo Clinic College of Medicine, Rochester, MN
| | - Ningling Kang-Decker
- Gastrointestinal Research Unit and Fiterman Center for Digestive Disease, Mayo Clinic College of Medicine, Rochester, MN
| | - Helen Hendrickson
- Gastrointestinal Research Unit and Fiterman Center for Digestive Disease, Mayo Clinic College of Medicine, Rochester, MN
| | - Steven F. Bronk
- Center for Basic Research in Digestive Diseases and Fiterman Center for Digestive Disease, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN
| | - Zvonimir S. Katusic
- Department of Anesthesiology and Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN
| | - Gregory J. Gores
- Center for Basic Research in Digestive Diseases and Fiterman Center for Digestive Disease, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN
| | - Vijay H. Shah
- Gastrointestinal Research Unit and Fiterman Center for Digestive Disease, Mayo Clinic College of Medicine, Rochester, MN
| |
Collapse
|
420
|
Liu Y, He J, Ji S, Wang Q, Pu H, Jiang T, Meng L, Yang X, Ji J. Comparative studies of early liver dysfunction in senescence-accelerated mouse using mitochondrial proteomics approaches. Mol Cell Proteomics 2008; 7:1737-47. [PMID: 18515266 DOI: 10.1074/mcp.m800109-mcp200] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The liver is a complex and unique organ responsible for a breadth of functions crucial to sustaining life, especially for various metabolic processes in its mitochondria. Senescence-accelerated mouse prone/8 (SAMP8), a widely used aging model, exhibits an oxidative stress-induced aging phenotype and severe mitochondria-related liver pathology that are not seen in senescence-accelerated mouse resistant/1 (SAMR1). Here we used both two-dimensional electrophoresis- and ICAT-based mitochondrial proteomics analysis to view the liver mitochondrial protein alterations between SAMP8 and SAMR1. Compared with SAMR1, decreased expression and activity of mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase were detected in SAMP8 at 6 months old (SAMP8-6m). As the key enzyme of ketogenesis, 3-hydroxy-3-methylglutaryl-CoA synthase is well known to be transcriptionally regulated by peroxisome proliferator-activated receptor alpha, which was also expressed at lower levels in SAMP8-6m livers. In addition, down-regulation of two peroxisome proliferator-activated receptor alpha target gene products (acyl-CoA oxidase and enoyl-CoA hydratase), elevation of triglyceride, and reduction of acetyl-CoA were observed, indicating abnormal fatty acid metabolism in SAMP8-6m livers. In addition eight proteins (NDUAA, NDUBA, NDUB7, NDUS1, NDUS3, NDUV1, ETFA, and UCRI) of mitochondrial complexes were down-regulated in SAMP8-6m, resulting in mitochondria-related liver dysfunction characterized by enhanced oxidative stress-induced molecular damage (lipid peroxide and oxidized protein) and depressed energy production (ATP). Glutamine synthetase and ornithine aminotransferase involved in glutamine synthesis were up-regulated in SAMP8 livers at both 1 and 6 months old that may be related to the accumulation of glutamate and glutamine. Our work provided useful clues to understanding the molecular mechanism underlying liver dysfunction in senescence-accelerated mouse.
Collapse
Affiliation(s)
- Yashu Liu
- The National Laboratory of Protein Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871, China
| | | | | | | | | | | | | | | | | |
Collapse
|
421
|
Abstract
Substantial improvements in the treatment of chronic liver disease have accelerated interest in uncovering the mechanisms underlying hepatic fibrosis and its resolution. Activation of resident hepatic stellate cells into proliferative, contractile, and fibrogenic cells in liver injury remains a dominant theme driving the field. However, several new areas of rapid progress in the past 5-10 years also have taken root, including: (1) identification of different fibrogenic populations apart from resident stellate cells, for example, portal fibroblasts, fibrocytes, and bone-marrow-derived cells, as well as cells derived from epithelial mesenchymal transition; (2) emergence of stellate cells as finely regulated determinants of hepatic inflammation and immunity; (3) elucidation of multiple pathways controlling gene expression during stellate cell activation including transcriptional, post-transcriptional, and epigenetic mechanisms; (4) recognition of disease-specific pathways of fibrogenesis; (5) re-emergence of hepatic macrophages as determinants of matrix degradation in fibrosis resolution and the importance of matrix cross-linking and scar maturation in determining reversibility; and (6) hints that hepatic stellate cells may contribute to hepatic stem cell behavior, cancer, and regeneration. Clinical and translational implications of these advances have become clear, and have begun to impact significantly on the management and outlook of patients with chronic liver disease.
Collapse
|
422
|
Strnad P, Stumptner C, Zatloukal K, Denk H. Intermediate filament cytoskeleton of the liver in health and disease. Histochem Cell Biol 2008; 129:735-49. [PMID: 18443813 PMCID: PMC2386529 DOI: 10.1007/s00418-008-0431-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2008] [Indexed: 02/06/2023]
Abstract
Intermediate filaments (IFs) represent the largest cytoskeletal gene family comprising approximately 70 genes expressed in tissue specific manner. In addition to scaffolding function, they form complex signaling platforms and interact with various kinases, adaptor, and apoptotic proteins. IFs are established cytoprotectants and IF variants are associated with >30 human diseases. Furthermore, IF-containing inclusion bodies are characteristic features of several neurodegenerative, muscular, and other disorders. Acidic (type I) and basic keratins (type II) build obligatory type I and type II heteropolymers and are expressed in epithelial cells. Adult hepatocytes contain K8 and K18 as their only cytoplasmic IF pair, whereas cholangiocytes express K7 and K19 in addition. K8/K18-deficient animals exhibit a marked susceptibility to various toxic agents and Fas-induced apoptosis. In humans, K8/K18 variants predispose to development of end-stage liver disease and acute liver failure (ALF). K8/K18 variants also associate with development of liver fibrosis in patients with chronic hepatitis C. Mallory-Denk bodies (MDBs) are protein aggregates consisting of ubiquitinated K8/K18, chaperones and sequestosome1/p62 (p62) as their major constituents. MDBs are found in various liver diseases including alcoholic and non-alcoholic steatohepatitis and can be formed in mice by feeding hepatotoxic substances griseofulvin and 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC). MDBs also arise in cell culture after transfection with K8/K18, ubiquitin, and p62. Major factors that determine MDB formation in vivo are the type of stress (with oxidative stress as a major player), the extent of stress-induced protein misfolding and resulting chaperone, proteasome and autophagy overload, keratin 8 excess, transglutaminase activation with transamidation of keratin 8 and p62 upregulation.
Collapse
Affiliation(s)
- P Strnad
- Department of Internal Medicine I, University of Ulm, Robert-Koch-Strabe 8, 89081, Ulm, Germany.
| | | | | | | |
Collapse
|
423
|
Mitsuyoshi H, Itoh Y, Sumida Y, Minami M, Yasui K, Nakashima T, Okanoue T. Evidence of oxidative stress as a cofactor in the development of insulin resistance in patients with chronic hepatitis C. Hepatol Res 2008; 38:348-53. [PMID: 18021228 DOI: 10.1111/j.1872-034x.2007.00280.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AIM The mechanisms by which metabolic disorders develop in patients with chronic hepatitis C are unknown. Our study aimed to test whether oxidative stress contributes to these mechanisms. METHODS The index of homeostasis model assessment-insulin resistance (HOMA-IR) and serum and hepatic levels of thioredoxin (Trx), which are markers of oxidative stress, were evaluated in 203 biopsy-proven chronic hepatitis C patients with hepatitis C virus (HCV) genotype 1 or 2 infection. HOMA-IR and Trx levels were compared with baseline values after phlebotomy in 23 patients. RESULTS HOMA-IR and serum Trx levels were significantly correlated with disease stage (HOMA-IR, P < 0.00001; Trx, P < 0.0001) and independently predicted fibrosis scores (HOMA-IR, P < 0.05; Trx, P < 0.005). Steatosis (%) was significantly correlated with HOMA-IR (P < 0.00005) and Trx (P < 0.001) stage (P < 0.00001). Serum Trx levels were significantly correlated with HOMA-IR (P < 0.05), even after adjustment for body mass index (P < 0.05). Furthermore, the mRNA levels of hepatic Trx were significantly correlated with HOMA-IR (P < 0.05) and independently-predicted HOMA-IR (P < 0.05). The alanine aminotransferase (P < 0.00001), Trx (P < 0.05), and HOMA-IR (P < 0.05) serum levels decreased significantly after phlebotomy; these effects were similar even in non-responders to interferon. CONCLUSION Oxidative stress contributed to the development of IR irrespective of obesity in patients with HCV genotype 1 or 2 infection. This study could contribute to our understanding of how metabolic disorders develop and how they should be treated in chronic hepatitis C patients.
Collapse
Affiliation(s)
- Hironori Mitsuyoshi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
424
|
Gracia-Sancho J, Laviña B, Rodríguez-Vilarrupla A, García-Calderó H, Fernández M, Bosch J, García-Pagán JC. Increased oxidative stress in cirrhotic rat livers: A potential mechanism contributing to reduced nitric oxide bioavailability. Hepatology 2008; 47:1248-56. [PMID: 18273863 DOI: 10.1002/hep.22166] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED In cirrhotic livers, decreased nitric oxide (NO) bioavailability is a major factor increasing intrahepatic vascular tone. In several vascular disorders, an increase in superoxide (O(2) (-)) has been shown to contribute to reduced NO bioavailability through its reaction with NO to form peroxynitrite. This study was aimed to test the hypothesis that, in cirrhotic livers, increased O(2) (-), by reacting with NO, reduces NO bioavailability. In control and cirrhotic rat livers, NO bioavailability was evaluated by the measurement of cyclic guanosine monophosphate in liver tissue and by 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate (DAF-FM-DA) fluorescence in isolated sinusoidal endothelial cells (SEC); the O(2) (-) content was determined by dihydroethidium staining in fresh liver sections. In addition, the role of endothelial nitric oxide synthase (eNOS), xanthine oxidase (XO), and cyclooxygenase (COX) as possible sources of O(2) (-) and the role of superoxide dismutase (SOD) enzymatic activity as an O(2) (-) scavenger were determined in liver homogenates. Protein-nitrotyrosination, a marker of the NO-O(2) (-) reaction, was evaluated in liver homogenates. Furthermore, in control SEC and bovine aortic endothelial cells, NO modulation by O(2) (-) was evaluated. Cirrhotic livers exhibited increased O(2) (-) levels. This was due, at least in part, to increased production by COX and XO but not eNOS and to reduced scavenging by SOD. Increased O(2) (-) was associated with a significant reduction in NO bioavailability and increased nitrotyrosinated proteins. In endothelial cells, an inverse relationship between O(2) (-) levels and NO bioavailability was observed. CONCLUSION Our data show that oxidative stress may contribute to reduced NO bioavailability in cirrhotic livers, supporting the evaluation of O(2) (-) reduction as a potential mechanism to restore NO content.
Collapse
Affiliation(s)
- Jorge Gracia-Sancho
- Hepatic Hemodynamic Laboratory, Liver Unit, Institut de Malalties Digestives i Metabòliques (IMDiM), Hospital Clínic, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, University of Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
425
|
Wang D, Wang LJ, Zhu FX, Zhu JY, Chen XD, Zou L, Saito M, Li LT. In vitro and in vivo studies on the antioxidant activities of the aqueous extracts of Douchi (a traditional Chinese salt-fermented soybean food). Food Chem 2008. [DOI: 10.1016/j.foodchem.2007.09.072] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
426
|
Kawashima A, Tsukamoto I, Koyabu T, Murakami Y, Kawakami T, Kakibuchi N, Takaguchi K, Kita K, Okita M. Eicosapentaenoic Acid Supplementation for Chronic Hepatitis C Patients During Combination Therapy of Pegylated Interferon α-2b and Ribavirin. Lipids 2008; 43:325-33. [DOI: 10.1007/s11745-008-3157-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Accepted: 01/25/2008] [Indexed: 02/05/2023]
|
427
|
De Minicis S, Brenner DA. Oxidative stress in alcoholic liver disease: role of NADPH oxidase complex. J Gastroenterol Hepatol 2008; 23 Suppl 1:S98-103. [PMID: 18336675 DOI: 10.1111/j.1440-1746.2007.05277.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Alcohol is a well-known risk factor for liver damage and is one of the major causes of liver disease worldwide. Chronic intake of alcohol, over a certain limit, inevitably leads to hepatic steatosis. If the injury persists, steatosis with concomitant tumor necrosis factor-alpha and other cytokines, progresses to steatohepatitis, fibrosis and finally cirrhosis. Among the multiple factors involved in the process of alcohol-induced liver injury, a crucial role is played by oxidative stress. Several mechanisms during ethanol metabolism result in reactive oxygen species (ROS) production. Although the main site of ethanol metabolism is hepatocytes, other mechanisms are involved in alcohol-induced liver injury. Specifically, in the ROS production activity, an important role is played by the NADPH oxidase complex. NADPH oxidase is expressed in hepatocytes, hepatic stellate cells and Kupffer cells in the liver. Studying NADPH oxidase gives new insights into alcohol-induced liver damage and provides new direction for future therapeutic strategies.
Collapse
Affiliation(s)
- Samuele De Minicis
- UCSD School of Medicine, Department of Medicine, La Jolla, California 92093-0602, USA
| | | |
Collapse
|
428
|
|
429
|
Abstract
Fibrosis is characterized by excessive accumulation of extracellular matrix (ECM) in basement membranes and interstitial tissues, resulting from increased synthesis or decreased degradation of ECM or both. The plasminogen activator/plasmin system plays an important role in ECM degradation, whereas the plasminogen activator inhibitor 1 (PAI-1) is a physiologic inhibitor of plasminogen activators. PAI-1 expression is increased in the lung fibrotic diseases and in experimental fibrosis models. The deletion of the PAI-1 gene reduces, whereas the overexpression of PAI-1 enhances, the susceptibility of animals to lung fibrosis induced by different stimuli, indicating an important role of PAI-1 in the development of lung fibrosis. Many growth factors, including transforming growth factor beta (TGF-beta) and tumor necrosis factor alpha (TNF-alpha), as well as other chemicals/agents, induce PAI-1 expression in cultured cells and in vivo. Reactive oxygen and nitrogen species (ROS/RNS) have been shown to mediate the induction of PAI-1 by many of these stimuli. This review summarizes some recent findings that help us to understand the role of PAI-1 in the development of lung fibrosis and ROS/RNS in the regulation of PAI-1 expression during fibrogenesis.
Collapse
Affiliation(s)
- Rui-Ming Liu
- Department of Environmental Health Sciences, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA.
| |
Collapse
|
430
|
Molecular basis and mechanisms of progression of non-alcoholic steatohepatitis. Trends Mol Med 2008; 14:72-81. [PMID: 18218340 DOI: 10.1016/j.molmed.2007.12.003] [Citation(s) in RCA: 324] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2007] [Revised: 12/03/2007] [Accepted: 12/03/2007] [Indexed: 12/11/2022]
Abstract
Non-alcoholic steatohepatitis (NASH), a cause of cirrhosis and hepatocellular carcinoma, is characterized by fatty infiltration of the liver, inflammation, hepatocellular damage and fibrosis. Progress has been made in understanding the molecular and cellular mechanisms implicated in the pathogenesis of this condition, therefore, we here review recent developments regarding the basic mechanisms of NASH development. Accumulation of triglycerides in the hepatocytes is the result of increased inflow of free fatty acids and de novo lipogenesis. Steatosis leads to lipotoxicity, which causes apoptosis, necrosis, generation of oxidative stress and inflammation. The resulting chronic injury activates a fibrogenic response that leads eventually to end-stage liver disease. A better understanding of these mechanisms is crucial for the design of novel diagnostic and therapeutic strategies.
Collapse
|
431
|
Siqueira C, Moura MCD, Pedro AJ, Rocha P. Elevated nitric oxide and 3’,5’ cyclic guanosine monophosphate levels in patients with alcoholic cirrhosis. World J Gastroenterol 2008; 14:236-42. [PMID: 18186561 PMCID: PMC2675120 DOI: 10.3748/wjg.14.236] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To evaluate whether serum levels of nitric oxide (NO•) and plasma levels of cyclic guanosine monophosphate (cGMP) and total glutathione (GSH) are altered in patients with alcoholic cirrhosis and to examine their correlation with the severity of liver disease.
METHODS: Twenty-six patients with alcoholic liver cirrhosis were studied. Serum levels of NO• and plasma levels of cGMP and GSH were measured in 7 patients with compensated alcoholic cirrhosis (Child-Pugh A) and 19 patients with advanced cirrhosis (Child-Pugh B and C). The model for end-stage liver disease (MELD) score was evaluated. Sixteen healthy volunteers served as controls. Liver enzymes and creatinine levels were also tested.
RESULTS: NO• and cGMP levels were higher in patients with Child-Pugh B and C cirrhosis than in Child-Pugh A cirrhosis or controls (NO•: 21.70 ± 8.07 vs 11.70 ± 2.74; 21.70 ± 8.07 vs 7.26 ± 2.47 &mgr;mol/L, respectively; P < 0.001) and (cGMP: 20.12 ± 6.62 vs 10.14 ± 2.78; 20.12 ± 6.62 vs 4.95 ± 1.21 pmol/L, respectively; P < 0.001). Total glutathione levels were lower in patients with Child-Pugh B and C cirrhosis than in patients with Child-Pugh A cirrhosis or controls (16.04 ± 6.06 vs 23.01 ± 4.38 or 16.04 ± 6.06 vs 66.57 ± 26.23 &mgr;mol/L, respectively; P < 0.001). There was a significant correlation between NO• and cGMP levels in all patients with alcoholic cirrhosis. A significant negative correlation between reduced glutathione/glutathione disulfide and the MELD score was found in all cirrhotic patients.
CONCLUSION: Our results suggest a role for oxidative stress in alcoholic liver cirrhosis, which is more significant in decompensated patients with higher levels of NO• and cGMP and lower GSH levels than in compensated and control patients. Altered mediator levels in decompensated patients may influence the hemodynamic changes in and progression of liver disease.
Collapse
|
432
|
Fu Y, Zheng S, Lin J, Ryerse J, Chen A. Curcumin protects the rat liver from CCl4-caused injury and fibrogenesis by attenuating oxidative stress and suppressing inflammation. Mol Pharmacol 2007; 73:399-409. [PMID: 18006644 DOI: 10.1124/mol.107.039818] [Citation(s) in RCA: 291] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We previously demonstrated that curcumin, a polyphenolic antioxidant purified from turmeric, up-regulated peroxisome proliferator-activated receptor (PPAR)-gamma gene expression and stimulated its signaling, leading to the inhibition of activation of hepatic stellate cells (HSC) in vitro. The current study evaluates the in vivo role of curcumin in protecting the liver against injury and fibrogenesis caused by carbon tetrachloride (CCl(4)) in rats and further explores the underlying mechanisms. We hypothesize that curcumin might protect the liver from CCl(4)-caused injury and fibrogenesis by attenuating oxidative stress, suppressing inflammation, and inhibiting activation of HSC. This report demonstrates that curcumin significantly protects the liver from injury by reducing the activities of serum aspartate aminotransferase, alanine aminotransferase, and alkaline phosphatase, and by improving the histological architecture of the liver. In addition, curcumin attenuates oxidative stress by increasing the content of hepatic glutathione, leading to the reduction in the level of lipid hydroperoxide. Curcumin dramatically suppresses inflammation by reducing levels of inflammatory cytokines, including interferon-gamma, tumor necrosis factor-alpha, and interleukin-6. Furthermore, curcumin inhibits HSC activation by elevating the level of PPARgamma and reducing the abundance of platelet-derived growth factor, transforming growth factor-beta, their receptors, and type I collagen. This study demonstrates that curcumin protects the rat liver from CCl(4)-caused injury and fibrogenesis by suppressing hepatic inflammation, attenuating hepatic oxidative stress and inhibiting HSC activation. These results confirm and extend our prior in vitro observations and provide novel insights into the mechanisms of curcumin in the protection of the liver. Our results suggest that curcumin might be a therapeutic antifibrotic agent for the treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Yumei Fu
- Department of Pathology, School of Medicine, Saint Louis University, 1402 S. Grand Blvd., St. Louis, MO 63104, USA
| | | | | | | | | |
Collapse
|
433
|
Mencin A, Seki E, Osawa Y, Kodama Y, De Minicis S, Knowles M, Brenner DA. Alpha-1 antitrypsin Z protein (PiZ) increases hepatic fibrosis in a murine model of cholestasis. Hepatology 2007; 46:1443-52. [PMID: 17668872 DOI: 10.1002/hep.21832] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
UNLABELLED Alpha-1 antitrypsin (alpha1-AT) deficiency is the most common genetic cause of liver disease in children. The homozygous alpha1-ATZ mutation (PiZZ) results in significant liver disease in 10% of all affected patients. The alpha1-ATZ mutation also may lead to worse liver injury in the setting of other liver diseases such as cystic fibrosis, nonalcoholic fatty liver disease, and hepatitis C. Although cholestatic injury is common to many forms of liver disease, its effect on the PiZZ phenotype is unknown. To elucidate the interplay of cholestasis and the PiZZ phenotype, we performed bile duct ligation (BDL) on C57BL/6 mice possessing a transgenic alpha1-ATZ mutation and littermate controls. PiZ transgenic mice undergoing BDL developed more liver fibrosis by quantification of Sirius red staining (P = 0.0003) and hydroxyproline (P = 0.007) than wild-type mice after BDL. More activated hepatic stellate cells (HSCs) and apoptotic cells also were observed in the PiZ BDL model. Quantitative real time polymerase chain reaction (PCR) of the endoplasmic reticulum (ER) stress markers CHOP and GRP78 were 4-fold and 2-fold more up-regulated, respectively, in PiZ BDL mice when compared with wild-type BDL mice (P = 0.02, P = 0.02). Increased apoptosis was also noted in PiZ BDL mice by terminal deoxynucleotidyl transferase-mediated nick-end labeling (TUNEL) and cleaved caspase-3 histological staining. CONCLUSION PiZ transgenic mice are more susceptible to liver fibrosis induced by cholestasis from BDL. Cholestasis therefore may lead to increased fibrosis in alpha1-AT deficiency, and the alpha1-ATZ mutation may act as a modifier gene in patients with concurrent cholestatic liver diseases such as cystic fibrosis.
Collapse
Affiliation(s)
- Ali Mencin
- Department of Pediatrics, Morgan Stanley Children's Hospital of New York, Columbia University, College of Physicians and Surgeons, New York, NY, USA.
| | | | | | | | | | | | | |
Collapse
|
434
|
Pérez de Obanos MP, López-Zabalza MJ, Arriazu E, Modol T, Prieto J, Herraiz MT, Iraburu MJ. Reactive oxygen species (ROS) mediate the effects of leucine on translation regulation and type I collagen production in hepatic stellate cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:1681-8. [PMID: 17707924 DOI: 10.1016/j.bbamcr.2007.07.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2007] [Revised: 06/29/2007] [Accepted: 07/13/2007] [Indexed: 01/23/2023]
Abstract
The amino acid leucine causes an increase of collagen alpha1(I) synthesis in hepatic stellate cells through the activation of translational regulatory mechanisms and PI3K/Akt/mTOR and ERK signaling pathways. The aim of the present study was to evaluate the role played by reactive oxygen species on these effects. Intracellular reactive oxygen species levels were increased in hepatic stellate cells incubated with leucine 5 mM at early time points, and this effect was abolished by pretreatment with the antioxidant glutathione. Preincubation with glutathione also prevented 4E-BP1, eIF4E and Mnk-1 phosphorylation induced by leucine, as well as enhancement of procollagen alpha1(I) protein levels. Inhibitors for MEK-1 (PD98059), PI3K (wortmannin) or mTOR (rapamycin) did not affect leucine-induced reactive oxygen species production. However, preincubation with glutathione prevented ERK, Akt and mTOR phosphorylation caused by treatment with leucine. The mitochondrial electron chain inhibitor rotenone and the NADPH oxidase inhibitor apocynin prevented reactive oxygen species production caused by leucine. Leucine also induced an increased phosphorylation of IR/IGF-R that was abolished by pretreatment with either rotenone or apocynin. Therefore, leucine exerts on hepatic stellate cells a prooxidant action through NADPH oxidase and mitochondrial Reactive oxygen species production and these effects mediate the activation of IR/IGF-IR and signaling pathways, finally leading to changes in translational regulation of collagen synthesis.
Collapse
Affiliation(s)
- María P Pérez de Obanos
- Departamento de Bioquímica y Biología Molecular, Universidad de Navarra, 31008 Pamplona, Spain
| | | | | | | | | | | | | |
Collapse
|
435
|
Dahiru D, Obidoa O. Evaluation of the antioxidant effects of Ziziphus mauritiana Lam. Leaf extracts against chronic ethanol-induced hepatotoxicity in rat liver. AFRICAN JOURNAL OF TRADITIONAL, COMPLEMENTARY, AND ALTERNATIVE MEDICINES : AJTCAM 2007; 5:39-45. [PMID: 20162053 PMCID: PMC2816599 DOI: 10.4314/ajtcam.v5i1.31254] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Chronic alcohol ingestion is known to increase the generation of reactive oxygen species (ROS), thereby leading to liver damage. Antioxidant enzymes act individually or in combination to reduce or counter the effect of these ROS. Chronic administration of alcohol at (40% v/v, 1 ml/100 g), for 6 weeks showed a significant (p<0.05) elevated levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), and total bilirubin (TB). There was also a significant (p<0.05) decreased levels of catalase, glutathione peroxidase, glutathione reductase and superoxide dismutase compared to control rats. Pre-treatment of rats with 200, 400 mg/kg body weight of aqueous leaf extract of Ziziphus mauritiana or 100 mg/kg silymarin resulted in a significant (p<0.05) decreased levels of ALT, AST, ALP, and TB with levels of catalase, glutathione peroxidase, glutathione reductase and superoxide dismutase showing a significant (p<0.05) increase compared to group administered alcohol only. Histopathology of rat liver administered with alcohol only resulted in severe necrosis, mononuclear cell aggregation and fatty degeneration in the central and mid zonal areas which was a characteristic of a damaged liver. Pre-treatment with the aqueous extract of Ziziphus mauritiana or silymarin reduced the morphological changes that are associated with chronic alcohol administration. The presence of tannins, saponins and phenolic compounds observed in the plant extract could be responsible for the observed effects of decreasing the levels of injured tissue marker and lipid peroxidation.
Collapse
Affiliation(s)
- D Dahiru
- Department of Biochemistry, Federal University of Technology, Yola P.M.B. 2076 Yola, Adamawa State, Nigeria. v
| | | |
Collapse
|
436
|
Avellini C, Baccarani U, Trevisan G, Cesaratto L, Vascotto C, D'Aurizio F, Pandolfi M, Adani GL, Tell G. Redox proteomics and immunohistology to study molecular events during ischemia-reperfusion in human liver. Transplant Proc 2007; 39:1755-60. [PMID: 17692604 DOI: 10.1016/j.transproceed.2007.05.082] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Oxidative stress is a condition occurring in liver disorders and causing liver damage due to ischemia-reperfusion (I/R) during liver transplantation. Several markers of chronic oxidative stress are well known; however, early protein targets of oxidative injury are not well defined. To identify them, we used a differential proteomics approach to HepG2 human liver cells that has been treated for 10 minutes with 500 micromol/L H(2)O(2). By differential proteomic analysis, using two-dimensional gel electrophoresis and MALDI-TOF mass spectrometry, we identified four proteins sensitive to H(2)O(2) treatment that underwent posttranslational modification of native polypeptides. Three of the proteins belong to the Peroxiredoxin family of hydroperoxide scavengers, PrxI, PrxII, and Prx VI, that showed changes in their pI as result of hyperoxidation. Mass mapping experiments demonstrated specific modification of the peroxiredoxins active site thiol into sulphinic and/or sulphonic acid, thus explaining an increased negative charge. The oxidation kinetics of all peroxiredoxins were extremely rapid and sensitive, occurring at H(2)O(2) doses unable to affect common markers of cellular oxidative stress. A differential proteomics approach was also applied to liver needle biopsies after cold (T(1)) and warm (T(2)) ischemia. Proteomic analysis of this material was related to histological changes and immunophenotypic expression of APE1/Ref-1. Hyperoxidation of PrxI occurring during I/R upon liver transplantation is dependent on the time of warm ischemia. Histological changes and APE1/Ref-1 expression parallel Peroxiredoxin changes. Our present data may be relevant to better graft preservation and evaluation for transplantation.
Collapse
Affiliation(s)
- C Avellini
- Department of Medical and Morphological Sciences, University of Udine, Udine, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
437
|
Albano E. Oxidative mechanisms in the pathogenesis of alcoholic liver disease. Mol Aspects Med 2007; 29:9-16. [PMID: 18045675 DOI: 10.1016/j.mam.2007.09.004] [Citation(s) in RCA: 213] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Accepted: 09/28/2007] [Indexed: 12/12/2022]
Abstract
Although the capacity of ethanol to induce oxidative stress in the liver is well established, the mechanisms by which oxidative damage contributes to the pathogenesis of alcoholic liver disease (ALD) is still incompletely understood. Recent reports have implicated oxidative mechanisms in the onset of alcoholic steatosis and in the formation of Mallory's bodies. Moreover, by inducing mitochondrial alterations, oxidative stress promotes hepatocyte necrosis and contributes to alcohol-induced sensitization of hepatocyte to the pro-apoptotic action of TNF-alpha. Oxidative mechanisms play also a role in the progression of liver fibrosis by triggering the release of pro-fibrotic cytokines and activating collagen gene expression in hepatic stellate cells. Finally, immune responses towards antigens originating from the reactions of lipid peroxidation products with hepatic proteins might represent one of the mechanisms that contribute to perpetuate chronic hepatic inflammation in ALD. Altogether these observations give a rationale to the possible clinical application of antioxidants in the therapy of ALD.
Collapse
Affiliation(s)
- Emanuele Albano
- Department of Medical Sciences, University Amedeo Avogadro of East Piedmont, Via Solaroli 17, 28100 Novara, Italy.
| |
Collapse
|
438
|
Parola M, Marra F, Pinzani M. Myofibroblast - like cells and liver fibrogenesis: Emerging concepts in a rapidly moving scenario. Mol Aspects Med 2007; 29:58-66. [PMID: 18022682 DOI: 10.1016/j.mam.2007.09.002] [Citation(s) in RCA: 132] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2007] [Accepted: 09/28/2007] [Indexed: 02/06/2023]
Abstract
Fibrotic progression of chronic liver diseases of different aetiology to the common advanced-stage of cirrhosis can be envisaged as a dynamic and highly integrated cellular response to chronic liver injury. Liver fibrosis is accompanied by perpetuation of liver injury, chronic hepatitis and persisting activation of tissue repair mechanisms, leading eventually to excess deposition of ECM components. Liver fibrogenesis (i.e., the process) is sustained by populations of highly proliferative, pro-fibrogenic and contractile MFs that, according to current literature, originate by a process of activation involving perisinusoidal HSC, portal fibroblasts and even bone marrow-derived MSC. In this short review emerging concepts in hepatic fibrogenesis and related molecular mechanisms, as provided by recent experimental and clinical studies, are presented.
Collapse
Affiliation(s)
- Maurizio Parola
- Dipartimento di Medicina e Oncologia Sperimentale, University of Torino, Corso Raffaello 30, 10125 Torino, Italy.
| | | | | |
Collapse
|
439
|
PKC signaling in oxidative hepatic damage. Mol Aspects Med 2007; 29:36-42. [PMID: 18035409 DOI: 10.1016/j.mam.2007.09.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2007] [Accepted: 09/28/2007] [Indexed: 12/16/2022]
Abstract
Protein kinase C (PKC) is a family of isoenzymes differently involved in cell response to injury and many studies describe their role as "stress sensors". Oxidative stress is strictly involved in the pathogenesis of chronic liver diseases including alcohol- or drug-induced hepatotoxicity, iron overload, hepatitis and hepatocarcinoma development, but molecular mechanisms are not really defined. A crucial role of PKC as a redox sensitive signaling molecule has been widely accepted.
Collapse
|
440
|
Abstract
Iron is an essential element involved in various biological pathways. When present in excess within the cell, iron can be toxic due to its ability to catalyse the formation of damaging radicals, which promote cellular injury and cell death. Within the liver, iron related oxidative stress can lead to fibrosis and ultimately to cirrhosis. Here we review the role of excessive iron in the pathologies associated with various chronic diseases of the liver. We also describe the molecular mechanism by which iron contributes to the development of hepatic fibrosis.
Collapse
Affiliation(s)
- Marie-A Philippe
- Hepatic Fibrosis Group, The Queensland Institute of Medical Research, PO Royal Brisbane and Women's Hospital, Brisbane 4029, Australia
| | | | | |
Collapse
|
441
|
Seronello S, Sheikh MY, Choi J. Redox regulation of hepatitis C in nonalcoholic and alcoholic liver. Free Radic Biol Med 2007; 43:869-82. [PMID: 17697932 DOI: 10.1016/j.freeradbiomed.2007.05.036] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2007] [Revised: 05/26/2007] [Accepted: 05/30/2007] [Indexed: 12/19/2022]
Abstract
Hepatitis C virus (HCV) is an RNA virus of the Flaviviridae family that is estimated to have infected 170 million people worldwide. HCV can cause serious liver disease in humans, such as cirrhosis, steatosis, and hepatocellular carcinoma. HCV induces a state of oxidative/nitrosative stress in patients through multiple mechanisms, and this redox perturbation has been recognized as a key player in HCV-induced pathogenesis. Studies have shown that alcohol synergizes with HCV in the pathogenesis of liver disease, and part of these effects may be mediated by reactive species that are generated during hepatic metabolism of alcohol. Furthermore, reactive species and alcohol may influence HCV replication and the outcome of interferon therapy. Alcohol consumption has also been associated with increased sequence heterogeneity of the HCV RNA sequences, suggesting multiple modes of interaction between alcohol and HCV. This review summarizes the current understanding of oxidative and nitrosative stress during HCV infection and possible combined effects of HCV, alcohol, and reactive species in the pathogenesis of liver disease.
Collapse
Affiliation(s)
- Scott Seronello
- School of Natural Sciences, University of California at Merced, Merced, CA 95344, USA
| | | | | |
Collapse
|
442
|
Sivaramakrishnan V, Shilpa PNM, Praveen Kumar VR, Niranjali Devaraj S. Attenuation of N-nitrosodiethylamine-induced hepatocellular carcinogenesis by a novel flavonol-Morin. Chem Biol Interact 2007; 171:79-88. [PMID: 17950263 DOI: 10.1016/j.cbi.2007.09.003] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2007] [Revised: 09/08/2007] [Accepted: 09/10/2007] [Indexed: 12/14/2022]
Abstract
Morin (3,5,7,2',4'-pentahydroxyflavone), a plant-derived flavonoid belonging to the subclass of flavonol is believed to play a role in chemoprevention and cancer chemotherapy. In this study, we found that the cotreatment of morin (500 ppm in diet) for 16 weeks to N-nitosodiethylamine-induced (200 mg/kg bodyweight in drinking water) rats provides protection against the oxidative stress caused by the carcinogen and thereby prevents hepatocellular carcinogenesis. On administration of the carcinogen, the level of lipid peroxidation increased markedly, but was found to be significantly lowered by morin treatment. On the contrary, the antioxidant levels in both liver and serum were decreased in carcinogen-administered animals, which was improved to normalcy upon morin administration. Cotreatment with morin prevented the elevation of marker enzymes induced by N-nitrosodiethylamine. The body weight of the animals decreased and their relative liver weight increased significantly on N-nitrosodiethylamine administration when compared to control group. However, cotreatment with morin significantly prevented the decrease of the body weight and increase in relative liver weight caused by DEN. Histological observations of liver tissue too correlated with the biochemical observations. In conclusion, these findings indicate that morin prevents lipid peroxidation, hepatic cell damage and protects the antioxidant system in N-nitrosodiethylamine-induced hepatocellular carcinogenesis.
Collapse
|
443
|
Son G, Iimuro Y, Seki E, Hirano T, Kaneda Y, Fujimoto J. Selective inactivation of NF-kappaB in the liver using NF-kappaB decoy suppresses CCl4-induced liver injury and fibrosis. Am J Physiol Gastrointest Liver Physiol 2007; 293:G631-9. [PMID: 17640975 DOI: 10.1152/ajpgi.00185.2007] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Sustained hepatic inflammation induced by various causes can lead to liver fibrosis. Transcription factor NF-kappaB is important in regulating inflammatory responses, especially in macrophages. We presently investigated whether an NF-kappaB decoy, a synthetic oligodeoxynucleotide (ODN) imitating the NF-kappaB binding site, inhibited the inflammatory response after CCl(4) intoxication to prevent CCl(4)-induced hepatic injury and fibrosis. The NF-kappaB decoy was introduced into livers by injecting the spleens of mice, using a hemagglutinating virus of Japan (HVJ)-liposome method. ODN was transferred mainly to macrophages in normal or fibrotic livers. Increases in serum transaminases and production of inflammatory cytokines after a single challenge with CCl(4) were inhibited by the NF-kappaB decoy, which suppressed nuclear translocation of NF-kappaB in liver macrophages. Liver fibrosis induced by CCl(4) administration for 8 wk was suppressed by the NF-kappaB decoy, accompanied by diminished mRNA expression for transforming growth factor (TGF)-beta, procollagen type 1 alpha(1), and alpha-smooth muscle actin (SMA). In vitro, isolated liver macrophages showed increased DNA binding activity of NF-kappaB and inflammatory cytokine production after hydrogen peroxide treatment; both increases were inhibited significantly by the NF-kappaB decoy. In contrast, NF-kappaB decoy transferred to isolated hepatic stellate cells (HSC) had no effect on their morphological activation or alpha-SMA expression, although the decoy accelerated tumor necrosis factor (TNF)-alpha-induced apoptosis in activated HSC. The effect of NF-kappaB decoy suppressing fibrosis probably results mainly from anti-inflammatory effects on liver macrophages, with a possible minor contribution from its direct proapoptotic effect on activated HSC.
Collapse
Affiliation(s)
- Gakuhei Son
- First Department of Surgery, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| | | | | | | | | | | |
Collapse
|
444
|
Clemente C, Elba S, Buongiorno G, Guerra V, D'Attoma B, Orlando A, Russo F. Manganese superoxide dismutase activity and incidence of hepatocellular carcinoma in patients with Child-Pugh class A liver cirrhosis: a 7-year follow-up study. Liver Int 2007; 27:791-7. [PMID: 17617122 DOI: 10.1111/j.1478-3231.2007.01485.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
AIMS To evaluate possible modifications in the manganese superoxide dismutase (MnSOD) activity during neoplastic transformation of a cirrhotic liver and to find out whether its assessment may have predictive value to identify cirrhotic patients at a higher risk of hepatocellular carcinoma (HCC). METHODS Seventy-one consecutive subjects with Child-Pugh class A liver cirrhosis were recruited. At the time of enrolment, HCC was diagnosed in 20 cirrhotic patients. The 51 cirrhotic patients without HCC were followed up for the occurrence of tumour by 6-monthly screening for 7 years. During follow-up, 16 patients developed HCC. Seventy healthy subjects formed the control group. MnSOD activity was assayed spectrophotometrically. RESULTS Serum MnSOD activity was significantly lower in 70 healthy subjects compared with 51 cirrhotic patients and 20 cirrhotic patients with HCC. Cirrhotic patients who developed HCC during follow-up showed significantly higher values of MnSOD activity than HCC-free patients. The best cut-off of MnSOD activity was 0.40 U/ml. At this cut-off, chi2 analysis revealed that MnSOD activity was significantly different between the HCC-free cirrhotic patients and cirrhotic patients who developed HCC. CONCLUSION The present findings suggest that during neoplastic transformation of cirrhotic liver, an increase in MnSOD activity may occur already during the precancerous phase, making this enzyme a probable malignancy-associated parameter.
Collapse
Affiliation(s)
- Caterina Clemente
- Laboratory of Experimental Biochemistry, National Institute for Digestive Sciences, IRCCS S. De Bellis, Castellana Grotte (Ba) Italy.
| | | | | | | | | | | | | |
Collapse
|
445
|
Herrema H, Smit GPA, Reijngoud DJ, Kuipers F. Does increased fatty acid oxidation enhance development of liver cirrhosis and progression to hepatocellular carcinoma in patients with glycogen storage disease type-III? J Hepatol 2007; 47:298-300; author reply 300-1. [PMID: 17570555 DOI: 10.1016/j.jhep.2007.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
446
|
Qureshi K, Abrams GA. Metabolic liver disease of obesity and role of adipose tissue in the pathogenesis of nonalcoholic fatty liver disease. World J Gastroenterol 2007; 13:3540-53. [PMID: 17659704 PMCID: PMC4146793 DOI: 10.3748/wjg.v13.i26.3540] [Citation(s) in RCA: 186] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is an increasingly recognized cause of liver-related morbidity and mortality. It can develop secondary to numerous causes but a great majority of NAFLD cases occur in patients who are obese or present with other components of metabolic syndrome (hypertension, dyslipidemia, diabetes). This is called primary NAFLD and insulin resistance plays a key role in its pathogenesis. Obesity is characterized by expanded adipose tissue, which is under a state of chronic inflammation. This disturbs the normal storage and endocrine functions of adipose tissue. In obesity, the secretome (adipokines, cytokines, free fatty acids and other lipid moieties) of fatty tissue is amplified, which through its autocrine, paracrine actions in fat tissue and systemic effects especially in the liver leads to an altered metabolic state with insulin resistance (IR). IR leads to hyperglycemia and reactive hyperinsulinemia, which stimulates lipid-accumulating processes and impairs hepatic lipid metabolism. IR enhances free fatty acid delivery to liver from the adipose tissue storage due to uninhibited lipolysis. These changes result in hepatic abnormal fat accumulation, which may initiate the hepatic IR and further aggravate the altered metabolic state of whole body. Hepatic steatosis can also be explained by the fact that there is enhanced dietary fat delivery and physical inactivity. IR and NAFLD are also seen in various lipodystrophic states in contrary to popular belief that these problems only occur due to excessive adiposity in obesity. Hence, altered physiology of adipose tissue is central to development of IR, metabolic syndrome and NAFLD.
Collapse
Affiliation(s)
- Kamran Qureshi
- Department of Medicine, University of Alabama at Birmingham, 1918 University Blvd 286 MCLM Birmingham, AL 35294, USA
| | | |
Collapse
|
447
|
Leclercq IA, Da Silva Morais A, Schroyen B, Van Hul N, Geerts A. Insulin resistance in hepatocytes and sinusoidal liver cells: mechanisms and consequences. J Hepatol 2007; 47:142-56. [PMID: 17512085 DOI: 10.1016/j.jhep.2007.04.002] [Citation(s) in RCA: 244] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hepatic insulin resistance is an important underlying cause of the metabolic syndrome that manifests itself in diseases such as diabetes type II, atherosclerosis or non-alcoholic fatty liver disease (NAFLD). In this paper, we summarize comprehensively the current state of knowledge pertaining to the molecular mechanisms that lead to insulin resistance in hepatocytes and sinusoidal liver cells. In hepatocytes, the insulin resistant state is brought about by at least one, but more likely by a combination, of the following pathological alterations: hyperglycaemia and hyperinsulinaemia, formation of advanced glycation end-products, increased free fatty acids and their metabolites, oxidative stress and altered profiles of adipocytokines. Insulin resistance in hepatocytes distorts directly glucose metabolism, especially the control over glucose output into the circulation and interferes with cell survival and proliferation, while hepatic fatty acid synthesis remains stimulated by compensatory hyperinsulinaemia, resulting in steatosis. Very few studies have addressed insulin resistance in sinusoidal liver cells. These cells are not simply bystanders and passive witnesses of the changes affecting the hepatocytes. They are target cells that will respond to the pathological alterations occurring in the insulin resistant state. They are also effector cells that may exacerbate insulin resistance in hepatocytes by increasing oxidative stress and by secreting cytokines such as TNF and IL-6. Moreover, activation of sinusoidal endothelial cells, Kupffer cells and stellate cells will lead to chemo-attraction of inflammatory cells. Finally, activation of stellate cells will set in motion a fibrogenic response that paves the way to cirrhosis.
Collapse
Affiliation(s)
- Isabelle A Leclercq
- Laboratory of Gastroenterology, Faculty of Medicine, Université catholique de Louvain, GAEN 53/79, Avenue Mounier, 53, B-1200 Brussels, Belgium.
| | | | | | | | | |
Collapse
|
448
|
Tasci I, Mas MR, Vural SA, Deveci S, Comert B, Alcigir G, Mas N, Akay C, Bozdayi M, Yurdaydin C, Bozkaya H, Uzunalimoglu O, Isik AT, Said HM. Pegylated interferon-alpha plus taurine in treatment of rat liver fibrosis. World J Gastroenterol 2007; 13:3237-44. [PMID: 17589904 PMCID: PMC4436611 DOI: 10.3748/wjg.v13.i23.3237] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the antifibrotic effects of peginterferon-alpha 2b and taurine on oxidative stress markers and hepatocellular apoptosis.
METHODS: Sixty rats with CCl4-induced liver fibrosis were divided into 4 groups (n = 15). Group 1 was left for spontaneous recovery (SR). Groups 2-4 received peginterferon-alpha 2b, taurine, and their combination, respectively, for four weeks. Histological fibrosis scores, histomorphometric analysis, tissue hydroxyproline, tissue MDA, GPx and SOD activities were determined. Activated stellate cells and hepatocellular apoptosis were also evaluated.
RESULTS: The degree of fibrosis decreased in all treatment groups compared to spontaneous recovery group. Taurine alone and in combination with peginterferon-alpha 2b reduced oxidative stress markers, but peginterferon-alpha 2b alone did not. Apoptotic hepatocytes and activated stellate cells were higher in groups 2-4 than in group 1. Combined taurine and peginterferon-alpha 2b further reduced fibrosis and increased activated stellate cell apoptosis, but could not improve oxidative stress more than taurine alone.
CONCLUSION: Peginterferon-alpha 2b exerts anti-fibrotic effects on rat liver fibrosis. It seems ineffective against oxidative stress in vivo. Peginterferon-alpha 2b in combination with taurine seems to be an antifibrotic strategy.
Collapse
Affiliation(s)
- Ilker Tasci
- Department of Internal Medicine, Gulhane School of Medicine Etlik 06018 Ankara, Turkey.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
449
|
Hirose A, Ono M, Saibara T, Nozaki Y, Masuda K, Yoshioka A, Takahashi M, Akisawa N, Iwasaki S, Oben JA, Onishi S. Angiotensin II type 1 receptor blocker inhibits fibrosis in rat nonalcoholic steatohepatitis. Hepatology 2007; 45:1375-81. [PMID: 17518368 DOI: 10.1002/hep.21638] [Citation(s) in RCA: 182] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
UNLABELLED Nonalcoholic steatohepatitis (NASH) is now the most frequent cause of chronic liver impairment in developed countries and is a suggested causative factor in the development of cryptogenic cirrhosis and hepatocellular carcinoma. At present there is no effective and accepted therapy for NASH. The renin-angiotensin system is involved in hepatic fibrosis through activation of hepatic stellate cells, major fibrogenic cells in the liver. Hepatic stellate cells are activated by liver injury to express excessive matrix proteins and profibrogenic cytokines such as transforming growth factor-beta 1. Medicines that inhibit this pathway may be of therapeutic potential in NASH. Using a methionine-choline-deficient rat model of NASH, we studied the potential utility of an angiotensin II type 1 receptor blocker (ARB), olmesartan, on biochemical, histologic, and antioxidant measures of disease activity. ARB significantly attenuated increases in aspartate aminotransferase, activation of hepatic stellate cells, oxidative stress, expression of transforming growth factor-beta 1, expression of collagen genes, and liver fibrosis. CONCLUSION Our observations strongly suggest a potential preventive role for ARB in the progression of nonalcoholic steatohepatitis.
Collapse
Affiliation(s)
- Akira Hirose
- Department of Gastroenterology and Hepatology, Kochi Medical School, Nankoku, Kochi 783-8505, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
450
|
De Minicis S, Brenner DA. NOX in liver fibrosis. Arch Biochem Biophys 2007; 462:266-72. [PMID: 17531188 PMCID: PMC2727549 DOI: 10.1016/j.abb.2007.04.016] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2007] [Revised: 04/10/2007] [Accepted: 04/11/2007] [Indexed: 12/13/2022]
Abstract
NADPH oxidase is a multi-protein complex producing reactive oxygen species (ROS) both in phagocytic cells, being essential in host defense, and in non-phagocytic cells, regulating intracellular signalling. In the liver, NADPH oxidase plays a central role in fibrogenesis. A functionally active form of the NADPH oxidase is expressed not only in Kupffer cells (phagocytic cell type) but also in hepatic stellate cells (HSCs) (non-phagocytic cell type), suggesting a role of the non-phagocytic NADPH oxidase in HSC activation. Consistent with this concept, profibrogenic agonists such as Angiotensin II (Ang II) and platelet derived growth factor (PDGF), or apoptotic bodies exert their activity through NADPH oxidase-activation in HSCs. Both pharmacological inhibition with DPI and genetic studies using p47(phox) knockout mice provided evidence for a central role of NADPH oxidase in the regulation of HSC-activity and liver fibrosis. In addition to the p47(phox) component, only Rac1 has been identified as a functional active component of the NADPH oxidase complex in HSCs.
Collapse
Affiliation(s)
- Samuele De Minicis
- Department of Medicine, Columbia University, College of Physicians and Surgeons, New York, NY 10026
| | - David A. Brenner
- Department of Medicine, Columbia University, College of Physicians and Surgeons, New York, NY 10026
| |
Collapse
|