1
|
Lee JA, Gu MJ, Lee YR, Kim Y, Choi I, Kim D, Ha SK. Lindera obtusiloba Blume Alleviates Non-Alcoholic Fatty Liver Disease Promoted by N ε-(carboxymethyl)lysine. Nutrients 2024; 16:2330. [PMID: 39064772 PMCID: PMC11280000 DOI: 10.3390/nu16142330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major issue because it is closely associated with metabolic diseases. Advanced glycation end products (AGEs) are implicated as risk factors for steatosis during NAFLD progression. AGEs influence NAFLD progression through a receptor-independent pathway involving AGE cross-link formation and a receptor-dependent pathway that binds to receptors like receptors for advanced glycation end products (RAGE). The objectives of this study are to examine the effect of Lindera obtusiloba Blume (LO) on NAFLD promoted by Nε-(carboxymethyl)lysine (CML), one of the most common dietary AGEs. The anti-glycation effects of LO were evaluated by inhibiting the AGEs formation and AGEs-collagen cross-links breaking. The efficacy of LO against NAFLD promoted by CML was assessed using both in vitro and in vivo models. NAFLD was induced in mice by feeding a high-fat diet and orally administering CML over a period of 12 weeks, and the effects of LO on lipid metabolism and its regulatory mechanisms were investigated. LO showed the effect of inhibited AGEs formation and breakage, and collagen cross-linking. Fed a high-fat diet with administered CML by gavage, LO administration resulted in a reduction in body weight, fat mass, serum triglycerides, total cholesterol, and low-density lipoprotein cholesterol levels. LO reduced hepatic CML accumulation and RAGE expression in mice fed a high-fat diet and orally administered CML. LO alleviated hepatic steatosis accompanied by lipid accumulation and histological damage by suppressing the expression of sterol regulatory element-binding protein 1c, carbohydrate response element binding protein, fatty acid synthase, stearoyl-CoA desaturase1, tumor necrosis factor-α, and interleukin-1β. LO alleviated the MAPK/NF-κB expression by attenuating CML and RAGE expression. Taken together, our results demonstrate that LO alleviates the progression of NAFLD by lowering the levels of AGEs by downregulating CML/RAGE expression.
Collapse
Affiliation(s)
- Jin-Ah Lee
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun 55365, Republic of Korea; (J.-A.L.); (M.J.G.); (Y.R.L.); (Y.K.); (I.C.)
| | - Min Ji Gu
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun 55365, Republic of Korea; (J.-A.L.); (M.J.G.); (Y.R.L.); (Y.K.); (I.C.)
| | - Yu Ra Lee
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun 55365, Republic of Korea; (J.-A.L.); (M.J.G.); (Y.R.L.); (Y.K.); (I.C.)
| | - Yoonsook Kim
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun 55365, Republic of Korea; (J.-A.L.); (M.J.G.); (Y.R.L.); (Y.K.); (I.C.)
| | - Inwook Choi
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun 55365, Republic of Korea; (J.-A.L.); (M.J.G.); (Y.R.L.); (Y.K.); (I.C.)
| | - Donghwan Kim
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun 55365, Republic of Korea; (J.-A.L.); (M.J.G.); (Y.R.L.); (Y.K.); (I.C.)
| | - Sang Keun Ha
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun 55365, Republic of Korea; (J.-A.L.); (M.J.G.); (Y.R.L.); (Y.K.); (I.C.)
- Division of Food Biotechnology, University of Science and Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
2
|
Almasri F, Collotta D, Aimaretti E, Sus N, Aragno M, Dal Bello F, Eva C, Mastrocola R, Landberg R, Frank J, Collino M. Dietary Intake of Fructooligosaccharides Protects against Metabolic Derangements Evoked by Chronic Exposure to Fructose or Galactose in Rats. Mol Nutr Food Res 2024; 68:e2300476. [PMID: 38158337 DOI: 10.1002/mnfr.202300476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/30/2023] [Indexed: 01/03/2024]
Abstract
SCOPE Diets rich in fat and sugars evoke chronic low-grade inflammation, leading to metabolic derangements. This study investigates the impact of fructose and galactose, two commonly consumed simple sugars, on exacerbation of the harmful effects caused by high fat intake. Additionally, the potential efficacy of fructooligosaccharides (FOS), a fermentable dietary fiber, in counteracting these effects is examined. METHODS AND RESULTS Male Sprague-Dawley rats (six/group) are fed 8 weeks as follows: control 5% fat diet (CNT), 20% fat diet (FAT), FAT+10% FOS diet (FAT+FOS), FAT+25% galactose diet (FAT+GAL), FAT+GAL+10% FOS diet (FAT+GAL+FOS), FAT+25% fructose diet (FAT+FRU), FAT+FRU+10% FOS diet (FAT+FRU+FOS). The dietary manipulations tested do not affect body weight gain, blood glucose, or markers of systemic inflammation whereas significant increases in plasma concentrations of triacylglycerols, cholesterol, aspartate aminotransferase, and alanine aminotrasferase are detected in both FAT+FRU and FAT+GAL compared to CNT. In the liver and skeletal muscle, both sugars induce significant accumulation of lipids and advanced glycation end-products (AGEs). FOS supplementation prevents these impairments. CONCLUSION This study extends the understanding of the deleterious effects of a chronic intake of simple sugars and demonstrates the beneficial role of the prebiotic FOS in dampening the sugar-induced metabolic impairments by prevention of lipid and AGEs accumulation.
Collapse
Affiliation(s)
- Fidèle Almasri
- Department of Food Biofunctionality, Institute of Nutritional Sciences, University of Hohenheim, Garbenstr. 28, 70599, Stuttgart, Germany
| | - Debora Collotta
- Department of Neurosciences "Rita Levi Montalcini", University of Turin, Corso Raffaello 30, Torino, 10125, Piemonte, Italy
| | - Eleonora Aimaretti
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, Turin, 10125, Piemonte, Italy
| | - Nadine Sus
- Department of Food Biofunctionality, Institute of Nutritional Sciences, University of Hohenheim, Garbenstr. 28, 70599, Stuttgart, Germany
| | - Manuela Aragno
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, Turin, 10125, Piemonte, Italy
| | - Federica Dal Bello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, Torino, 10126, Piemonte, Italy
| | - Carola Eva
- Department of Neurosciences "Rita Levi Montalcini", University of Turin, Corso Raffaello 30, Torino, 10125, Piemonte, Italy
| | - Raffaella Mastrocola
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, Turin, 10125, Piemonte, Italy
| | - Rikard Landberg
- Department of Life Sciences, Division of Food and Nutrition Science, Chalmers University of Technology, Gothenburg, 41296, Sweden
| | - Jan Frank
- Department of Food Biofunctionality, Institute of Nutritional Sciences, University of Hohenheim, Garbenstr. 28, 70599, Stuttgart, Germany
| | - Massimo Collino
- Department of Neurosciences "Rita Levi Montalcini", University of Turin, Corso Raffaello 30, Torino, 10125, Piemonte, Italy
| |
Collapse
|
3
|
Abouelezz HM, Shehatou GS, Shebl AM, Salem HA. A standardized pomegranate fruit extract ameliorates thioacetamide-induced liver fibrosis in rats via AGE-RAGE-ROS signaling. Heliyon 2023; 9:e14256. [PMID: 36938469 PMCID: PMC10015255 DOI: 10.1016/j.heliyon.2023.e14256] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 02/22/2023] [Accepted: 02/28/2023] [Indexed: 03/07/2023] Open
Abstract
This work aimed to investigate a possible mechanism that may mediate the hepatoprotective effects of pomegranate fruit extract (PFE) against thioacetamide (THIO)-induced liver fibrosis in rats. Male Sprague Dawley rats were randomly allocated into four groups (n = 8 each): control; PFE (150 mg/kg/day, orally); THIO (200 mg/kg, i.p, 3 times a week); and THIO and PFE-treated groups. Oral PFE treatment decreased liver/body weight ratio by 12.4%, diminished serum function levels of ALT, AST, ALP, LDH, and total bilirubin, increased serum albumin, boosted hepatic GSH (by 35.6%) and SOD (by 17.5%), and significantly reduced hepatic levels of ROS, MDA, 4-HNE, AGEs, and RAGE in THIO-fibrotic rats relative to untreated THIO group. Moreover, PFE administration downregulated the hepatic levels of profibrotic TGF-β1 (by 23.0%, P < 0.001) and TIMP-1 (by 41.5%, P < 0.001), attenuated α-SMA protein expression, decreased serum HA levels (by 41.3%), and reduced the hepatic levels of the fibrosis markers hydroxyproline (by 26.0%, P < 0.001), collagen type IV (by 44.3%, P < 0.001) and laminin (by 43.4%, P < 0.001) compared to the untreated THIO group. The histopathological examination has corroborated these findings, where PFE decreased hepatic nodule incidence, attenuated portal necroinflammation and reduced extent of fibrosis. These findings may suggest that oral PFE administration could slow the progression of hepatic fibrogenesis via reducing hepatic levels of AGEs, RAGE, ROS, TGF-β1, and TIMP-1.
Collapse
Affiliation(s)
- Hadeer M. Abouelezz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
- Corresponding author.
| | - George S.G. Shehatou
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa City, Egypt
| | - Abdelhadi M. Shebl
- Department of Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Hatem A. Salem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
4
|
Park JW, Kim MJ, Kim SE, Kim HJ, Jeon YC, Shin HY, Park SJ, Jang MK, Kim DJ, Park CK, Choi EK. Increased Expression of S100B and RAGE in a Mouse Model of Bile Duct Ligation-induced Liver Fibrosis. J Korean Med Sci 2021; 36:e90. [PMID: 33847081 PMCID: PMC8042478 DOI: 10.3346/jkms.2021.36.e90] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 01/27/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Liver fibrosis is defined as the accumulation of the extracellular matrix and scar formation. The receptor for advanced glycation end products (RAGE) has been demonstrated to participate in fibrogenesis. S100B is a ligand of RAGE and exerts extracellular functions by inducing a series of signal transduction cascades. However, the involvement of S100B and RAGE in cholestasis-induced liver fibrosis remains unclear. In this study, we investigated S100B and RAGE expression during liver fibrosis in mice that underwent common bile duct ligation (BDL). METHODS BDL was performed in 10-week-old male C57BL/6J mice with sham control (n = 26) and BDL (n = 26) groups. Expression levels of S100B, RAGE and fibrotic markers in the livers from both groups at week 1 and 3 after BDL were examined by western blot and quantitative real-time reverse transcription polymerase chain reaction analysis. Liver fibrotic changes were examined by histological and ultrastructural analysis. RESULTS Histological staining with Sirius Red and the evaluation of the messenger RNA expression of fibrotic markers showed noticeable periportal fibrosis and bile duct proliferation. S100B was mainly present in bile duct epithelial cells, and its expression was upregulated in proportion to the ductular reaction during fibrogenesis by BDL. RAGE expression was also increased, and interestingly, triple immunofluorescence staining and transmission electron microscopy showed that both S100B and RAGE were expressed in proliferating bile duct epithelial cells and activated hepatic stellate cells (HSCs) of the BDL livers. In addition, in rat HSCs (HSC-T6), treatment with recombinant S100B protein significantly increased fibrotic markers in a dose-dependent manner, and RAGE small interfering RNA (siRNA) suppressed S100B-stimulated upregulation of fibrotic markers compared with cells treated with scramble siRNA and S100B. CONCLUSION These findings suggest that the increased expression of S100B and RAGE and the interaction between S100B and RAGE may play an important role in ductular reaction and liver fibrosis induced by BDL.
Collapse
Affiliation(s)
- Ji Won Park
- Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, Korea
- Department of Internal Medicine, Hallym University Medical Center, Anyang, Korea
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Mo Jong Kim
- Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, Korea
- Ilsong Institute of Life Science, Hallym University, Anyang, Korea
| | - Sung Eun Kim
- Department of Internal Medicine, Hallym University Medical Center, Anyang, Korea
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Hee Jun Kim
- Ilsong Institute of Life Science, Hallym University, Anyang, Korea
| | - Yong Chul Jeon
- Ilsong Institute of Life Science, Hallym University, Anyang, Korea
| | - Hae Young Shin
- Ilsong Institute of Life Science, Hallym University, Anyang, Korea
| | - Se Jin Park
- Ilsong Institute of Life Science, Hallym University, Anyang, Korea
| | - Myoung Kuk Jang
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
- Department of Internal Medicine, Kangdong Sacred Heart Hospital of Hallym University Medical Center, Seoul, Korea
| | - Dong Joon Kim
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
- Department of Internal Medicine, Chuncheon Sacred Heart Hospital of Hallym University Medical Center, Chuncheon, Korea
| | - Choong Kee Park
- Department of Internal Medicine, Hallym University Medical Center, Anyang, Korea
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Eun Kyoung Choi
- Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, Korea
- Ilsong Institute of Life Science, Hallym University, Anyang, Korea.
| |
Collapse
|
5
|
Dehnad A, Fan W, Jiang JX, Fish SR, Li Y, Das S, Mozes G, Wong KA, Olson KA, Charville GW, Ali M, Török NJ. AGER1 downregulation associates with fibrosis in nonalcoholic steatohepatitis and type 2 diabetes. J Clin Invest 2021; 130:4320-4330. [PMID: 32657776 DOI: 10.1172/jci133051] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 05/05/2020] [Indexed: 12/25/2022] Open
Abstract
Type 2 diabetes is clinically associated with progressive necroinflammation and fibrosis in nonalcoholic steatohepatitis (NASH). Advanced glycation end-products (AGEs) accumulate during prolonged hyperglycemia, but the mechanistic pathways that lead to accelerated liver fibrosis have not been well defined. In this study, we show that the AGEs clearance receptor AGER1 was downregulated in patients with NASH and diabetes and in our NASH models, whereas the proinflammatory receptor RAGE was induced. These findings were associated with necroinflammatory, fibrogenic, and pro-oxidant activity via the NADPH oxidase 4. Inhibition of AGEs or RAGE deletion in hepatocytes in vivo reversed these effects. We demonstrate that dysregulation of NRF2 by neddylation of cullin 3 was linked to AGER1 downregulation and that induction of NRF2 using an adeno-associated virus-mediated approach in hepatocytes in vivo reversed AGER1 downregulation, lowered the level of AGEs, and improved proinflammatory and fibrogenic responses in mice on a high AGEs diet. In patients with NASH and diabetes or insulin resistance, low AGER1 levels were associated with hepatocyte ballooning degeneration and ductular reaction. Collectively, prolonged exposure to AGEs in the liver promotes an AGER1/RAGE imbalance and consequent redox, inflammatory, and fibrogenic activity in NASH.
Collapse
Affiliation(s)
- Ali Dehnad
- Gastroenterology and Hepatology, Stanford University, Stanford, and VA Palo Alto, California, USA
| | - Weiguo Fan
- Gastroenterology and Hepatology, Stanford University, Stanford, and VA Palo Alto, California, USA
| | | | | | - Yuan Li
- Gastroenterology and Hepatology, Stanford University, Stanford, and VA Palo Alto, California, USA
| | - Suvarthi Das
- Gastroenterology and Hepatology, Stanford University, Stanford, and VA Palo Alto, California, USA
| | - Gergely Mozes
- Gastroenterology and Hepatology, Stanford University, Stanford, and VA Palo Alto, California, USA
| | | | - Kristin A Olson
- Department of Pathology, UC Davis Medical Center, Sacramento, California, USA
| | | | - Mohammed Ali
- Department of Surgery, UC Davis Medical Center, Sacramento, California, USA
| | - Natalie J Török
- Gastroenterology and Hepatology, Stanford University, Stanford, and VA Palo Alto, California, USA
| |
Collapse
|
6
|
Abdel-Razik A, Mousa N, Zakaria S, Abdelsalam M, Eissa M, Abd El-Ghany MI, Hasan AS, Elhelaly R, Elzehery R, El-Wakeel N, Eldars W. Advanced Glycation End Products as a Predictor of Diabetes Mellitus in Chronic Hepatitis C-Related Cirrhosis. Front Med (Lausanne) 2020; 7:588519. [PMID: 33195350 PMCID: PMC7649387 DOI: 10.3389/fmed.2020.588519] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/25/2020] [Indexed: 12/16/2022] Open
Abstract
Background and Aims: Advanced glycation end products (AGEs) were found to be involved in the pathogenesis of various disorders. Chronic hepatitis C virus infection is the major cause of liver cirrhosis development and glucose metabolism alteration. We aimed to explore the association of AGEs with the development of diabetes mellitus (DM) in patients with cirrhosis in this study. Methods: Only 144 of the 165 non-diabetic patients with cirrhosis were consecutively included in this prospective cohort pilot study, in addition to 72 healthy control subjects. Clinical data and biochemical parameters including basal insulin secretion and insulin sensitivity indices together with AGEs were evaluated in all participants at baseline and every 1 year thereafter for 2 years. Multivariable Cox regression analysis was used to determine the parameters that could predict the development of DM within this period. Results: DM developed in 14 (10%) patients only. Univariate Cox regression analysis showed that AGEs (P = 0.004), Homeostatic Model Assessment-Insulin Resistance (HOMA-IR) (P = 0.018), HOMA-β (P = 0.015), and age (P = 0.012) were associated with DM. After adjusting multiple confounders, the multivariable Cox regression model showed that AGEs, HOMA-IR, and age were the strongest variables associated with DM (all P < 0.05). Using the receiver operating characteristic curve, AGEs at a cutoff value of more than 82.4 ng/ml had 99.23% specificity, 100% sensitivity, and 0.992 area under the curve (AUC) (all P < 0.001) for DM prediction. Conclusion: Our study suggests that AGEs are related to increased incidence of DM, especially in patients with cirrhosis, which is very promising in lowering the risk of DM in these patients.
Collapse
Affiliation(s)
- Ahmed Abdel-Razik
- Tropical Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Nasser Mousa
- Tropical Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Sahar Zakaria
- Tropical Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mostafa Abdelsalam
- Nephrology and Dialysis Unit, Internal Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed Eissa
- Internal Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohammed I. Abd El-Ghany
- Endocrinology and Diabetes Unit, Internal Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmad S. Hasan
- Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Rania Elhelaly
- Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Rasha Elzehery
- Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Niveen El-Wakeel
- Medical Microbiology and Immunology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Waleed Eldars
- Medical Microbiology and Immunology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
7
|
Pereira ENGDS, Silvares RR, Flores EEI, Rodrigues KL, Daliry A. Pyridoxamine improves metabolic and microcirculatory complications associated with nonalcoholic fatty liver disease. Microcirculation 2020; 27:e12603. [PMID: 31876010 DOI: 10.1111/micc.12603] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE We investigated the protective effects of pyridoxamine against metabolic and microcirculatory complications in nonalcoholic fatty liver disease. METHODS Nonalcoholic fatty liver disease was established by a high-fat diet administration over 28 weeks. Pyridoxamine was administered between weeks 20 and 28. The recruitment of leukocytes and the number of vitamin A-positive hepatic stellate cells were examined by in vivo microscopy. Laser speckle contrast imaging was used to evaluate microcirculatory hepatic perfusion. Thiobarbituric acid reactive substances measurement and RT-PCR were used for oxidative stress and inflammatory parameters. advanced glycation end products were evaluated by fluorescence spectroscopy. RESULTS The increase in body, liver, and fat weights, together with steatosis and impairment in glucose metabolism observed in the nonalcoholic fatty liver disease group were attenuated by pyridoxamine treatment. Regarding the hepatic microcirculatory parameters, rats with high-fat diet-induced nonalcoholic fatty liver disease showed increased rolling and adhesion of leukocytes, increased hepatic stellate cells activation, and decreased tissue perfusion, which were reverted by pyridoxamine. Pyridoxamine protected against the increased hepatic lipid peroxidation observed in the nonalcoholic fatty liver disease group. Pyridoxamine treatment was associated with increased levels of tumor necrosis factor alpha (TNF-α) mRNA transcripts in the liver. CONCLUSION Pyridoxamine modulates oxidative stress, advanced glycation end products, TNF-α transcripts levels, and metabolic disturbances, being a potential treatment for nonalcoholic fatty liver disease-associated microcirculatory and metabolic complications.
Collapse
Affiliation(s)
| | - Raquel Rangel Silvares
- Laboratory of Cardiovascular Investigation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | - Karine Lino Rodrigues
- Laboratory of Cardiovascular Investigation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Anissa Daliry
- Laboratory of Cardiovascular Investigation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
8
|
Zhao Z, Lin CY, Cheng K. siRNA- and miRNA-based therapeutics for liver fibrosis. Transl Res 2019; 214:17-29. [PMID: 31476281 PMCID: PMC6848786 DOI: 10.1016/j.trsl.2019.07.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/08/2019] [Accepted: 07/18/2019] [Indexed: 02/07/2023]
Abstract
Liver fibrosis is a wound-healing process induced by chronic liver injuries, such as nonalcoholic steatohepatitis, hepatitis, alcohol abuse, and metal poisoning. The accumulation of excessive extracellular matrix (ECM) in the liver is a key characteristic of liver fibrosis. Activated hepatic stellate cells (HSCs) are the major producers of ECM and therefore play irreplaceably important roles during the progression of liver fibrosis. Liver fibrogenesis is highly correlated with the activation of HSCs, which is regulated by numerous profibrotic cytokines. Using RNA interference to downregulate these cytokines in activated HSCs is a promising strategy to reverse liver fibrosis. Meanwhile, microRNAs (miRNAs) have also been exploited for the treatment of liver fibrosis. This review focuses on the current siRNA- and miRNA-based liver fibrosis treatment strategies by targeting activated HSCs in the liver.
Collapse
Affiliation(s)
- Zhen Zhao
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri
| | - Chien-Yu Lin
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri
| | - Kun Cheng
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri.
| |
Collapse
|
9
|
Gaskell H, Ge X, Nieto N. High-Mobility Group Box-1 and Liver Disease. Hepatol Commun 2018; 2:1005-1020. [PMID: 30202816 PMCID: PMC6128227 DOI: 10.1002/hep4.1223] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/03/2018] [Indexed: 12/12/2022] Open
Abstract
High‐mobility group box‐1 (HMGB1) is a ubiquitous protein. While initially thought to be simply an architectural protein due to its DNA‐binding ability, evidence from the last decade suggests that HMGB1 is a key protein participating in the pathogenesis of acute liver injury and chronic liver disease. When it is passively released or actively secreted after injury, HMGB1 acts as a damage‐associated molecular pattern that communicates injury and inflammation to neighboring cells by the receptor for advanced glycation end products or toll‐like receptor 4, among others. In the setting of acute liver injury, HMGB1 participates in ischemia/reperfusion, sepsis, and drug‐induced liver injury. In the context of chronic liver disease, it has been implicated in alcoholic liver disease, liver fibrosis, nonalcoholic steatohepatitis, and hepatocellular carcinoma. Recently, specific posttranslational modifications have been identified that could condition the effects of the protein in the liver. Here, we provide a detailed review of how HMGB1 signaling participates in acute liver injury and chronic liver disease.
Collapse
Affiliation(s)
- Harriet Gaskell
- Department of Pathology University of Illinois at Chicago Chicago IL
| | - Xiaodong Ge
- Department of Pathology University of Illinois at Chicago Chicago IL
| | - Natalia Nieto
- Department of Pathology University of Illinois at Chicago Chicago IL.,Department of Medicine University of Illinois at Chicago Chicago IL
| |
Collapse
|
10
|
Luo Z, Jegga AG, Bezerra JA. Gene-disease associations identify a connectome with shared molecular pathways in human cholangiopathies. Hepatology 2018; 67:676-689. [PMID: 28865156 PMCID: PMC5834359 DOI: 10.1002/hep.29504] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 08/29/2017] [Accepted: 08/29/2017] [Indexed: 12/21/2022]
Abstract
Cholangiopathies are a diverse group of progressive diseases whose primary cell targets are cholangiocytes. To identify shared pathogenesis and molecular connectivity among the three main human cholangiopathies (biliary atresia [BA], primary biliary cholangitis [PBC], and primary sclerosing cholangitis [PSC]), we built a comprehensive platform of published data on gene variants, gene expression, and functional studies and applied network-based analytics in the search for shared molecular circuits. Mining the data platform with largest connected component and interactome analyses, we validated previously reported associations and identified essential and hub genes. In addition to disease-specific modules, we found a substantial overlap of disease neighborhoods and uncovered a group of 34 core genes that are enriched for immune processes and abnormal intestine/hepatobiliary mouse phenotypes. Within this core, we identified a gene subcore containing signal transduction and activator of transcription 3, interleukin-6, tumor necrosis factor, and forkhead box P3 prominently placed in a regulatory connectome of genes related to cellular immunity and fibrosis. We also found substantial gene enrichment in the advanced glycation endproduct/receptor for advanced glycation endproducts (RAGE) pathway and showed that RAGE activation induced cholangiocyte proliferation. Conclusion: Human cholangiopathies share pathways enriched by immunity genes and a molecular connectome that links different pathogenic features of BA, PBC, and PSC. (Hepatology 2018;67:676-689).
Collapse
Affiliation(s)
- Zhenhua Luo
- The Liver Care Center and Divisions of Gastroenterology, Hepatology and Nutrition
| | - Anil G Jegga
- Biomedical Informatics of Cincinnati Children's Hospital Medical Center and the Department of Pediatrics of the University Of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jorge A Bezerra
- The Liver Care Center and Divisions of Gastroenterology, Hepatology and Nutrition
| |
Collapse
|
11
|
Hollenbach M. The Role of Glyoxalase-I (Glo-I), Advanced Glycation Endproducts (AGEs), and Their Receptor (RAGE) in Chronic Liver Disease and Hepatocellular Carcinoma (HCC). Int J Mol Sci 2017; 18:ijms18112466. [PMID: 29156655 PMCID: PMC5713432 DOI: 10.3390/ijms18112466] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/16/2017] [Accepted: 11/17/2017] [Indexed: 12/12/2022] Open
Abstract
Glyoxalase-I (Glo-I) and glyoxalase-II (Glo-II) comprise the glyoxalase system and are responsible for the detoxification of methylglyoxal (MGO). MGO is formed non-enzymatically as a by-product, mainly in glycolysis, and leads to the formation of advanced glycation endproducts (AGEs). AGEs bind to their receptor, RAGE, and activate intracellular transcription factors, resulting in the production of pro-inflammatory cytokines, oxidative stress, and inflammation. This review will focus on the implication of the Glo-I/AGE/RAGE system in liver injury and hepatocellular carcinoma (HCC). AGEs and RAGE are upregulated in liver fibrosis, and the silencing of RAGE reduced collagen deposition and the tumor growth of HCC. Nevertheless, data relating to Glo-I in fibrosis and cirrhosis are preliminary. Glo-I expression was found to be reduced in early and advanced cirrhosis with a subsequent increase of MGO-levels. On the other hand, pharmacological modulation of Glo-I resulted in the reduced activation of hepatic stellate cells and therefore reduced fibrosis in the CCl₄-model of cirrhosis. Thus, current research highlighted the Glo-I/AGE/RAGE system as an interesting therapeutic target in chronic liver diseases. These findings need further elucidation in preclinical and clinical studies.
Collapse
Affiliation(s)
- Marcus Hollenbach
- Department of Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Liebigstrasse 20, D-04103 Leipzig, Germany.
| |
Collapse
|
12
|
Fructose-human serum albumin interaction undergoes numerous biophysical and biochemical changes before forming AGEs and aggregates. Int J Biol Macromol 2017; 109:896-906. [PMID: 29133088 DOI: 10.1016/j.ijbiomac.2017.11.069] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/08/2017] [Accepted: 11/10/2017] [Indexed: 01/07/2023]
Abstract
Fructose is a reducing and highly lipogenic sugar that has unique metabolic effects in the liver. Non-enzymatic fructosylation of proteins generates advanced glycation end products (AGEs). Human serum albumin (HSA) may undergo fructosylation vis-à-vis AGEs formation. High fructose consumption may lead to structurally altered and functionally compromised fructosylated-HSA-AGEs, which can cause damage to hepatocytes resulting in hepatic macro- and microvesicular steatosis. In this study, HSA was incubated with varying concentrations of fructose for 10days and the induced changes were studied. Fructosylated-HSA exhibited hyperchromicity, increased AGE-specific fluorescence, quenching of tryptophan fluorescence and increased melting temperature. Nε-[carboxymethyl]-lysine (CML), was detected by liquid chromatography mass spectrometry (LC-MS). Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) results showed decreased mobility in fructosylated-HSA. Perturbations in secondary and tertiary structure were revealed by fourier transform-infrared spectroscopy (FT-IR), supported by far- and near-UV circular dichroism (CD). Dynamic light scattering (DLS) and Matrix-Assisted Laser Desorption Ionization-Time of Flight (MALDI-TOF) mass spectrometry studies suggested increase in molecular mass of fructosylated-HSA. Amyloidogenic aggregates were confirmed from Congo red, Thioflavin T assay and Scanning electron microscope (SEM). These investigations confirmed the structural alterations in fructosylated-HSA and warrants further study to probe the role of fructosylated-HSA-AGEs in hepatopathy vis-à-vis fatty liver diseases.
Collapse
|
13
|
Cannito S, Novo E, Parola M. Therapeutic pro-fibrogenic signaling pathways in fibroblasts. Adv Drug Deliv Rev 2017; 121:57-84. [PMID: 28578015 DOI: 10.1016/j.addr.2017.05.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/28/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023]
Abstract
Myofibroblasts (MFs) play a critical role in the progression of chronic inflammatory and fibroproliferative diseases in different tissues/organs, whatever the etiology. Fibrosis is preceded and sustained by persistent injury and inflammatory response in a profibrogenic scenario involving mutual interactions, operated by several mediators and pathways, of MFs and related precursor cells with innate immunity cells and virtually any cell type in a defined tissue. These interactions, mediators and related signaling pathways are critical in initiating and perpetuating the differentiation of precursor cells into MFs that in different tissues share peculiar traits and phenotypic responses, including the ability to proliferate, produce ECM components, migrate and contribute to the modulation of inflammatory response and tissue angiogenesis. Literature studies related to liver, lung and kidney fibrosis have outlined a number of MF-related core regulatory fibrogenic signaling pathways conserved across these different organs and potentially targetable in order to develop effective antifibrotic therapeutic strategies.
Collapse
|
14
|
Zhuang A, Yap FY, Bruce C, Leung C, Plan MR, Sullivan MA, Herath C, McCarthy D, Sourris KC, Kantharidis P, Coughlan MT, Febbraio MA, Hodson MP, Watt MJ, Angus P, Schulz BL, Forbes JM. Increased liver AGEs induce hepatic injury mediated through an OST48 pathway. Sci Rep 2017; 7:12292. [PMID: 28947796 PMCID: PMC5612946 DOI: 10.1038/s41598-017-12548-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/11/2017] [Indexed: 12/13/2022] Open
Abstract
The protein oligosaccharyltransferase-48 (OST48) is integral to protein N-glycosylation in the endoplasmic reticulum (ER) but is also postulated to act as a membrane localised clearance receptor for advanced glycation end-products (AGE). Hepatic ER stress and AGE accumulation are each implicated in liver injury. Hence the objective of this study was to increase the expression of OST48 and examine the effects on hepatic function and structure. Groups of 8 week old male mice (n = 10-12/group) over-expressing the gene for OST48, dolichyl-diphosphooligosaccharide-protein glycosyltransferase (DDOST+/-), were followed for 24 weeks, while randomised to diets either low or high in AGE content. By week 24 of the study, either increasing OST48 expression or consumption of high AGE diet impaired liver function and modestly increased hepatic fibrosis, but their combination significantly exacerbated liver injury in the absence of steatosis. DDOST+/- mice had increased both portal delivery and accumulation of hepatic AGEs leading to central adiposity, insulin secretory defects, shifted fuel usage to fatty and ketoacids, as well as hepatic glycogen accumulation causing hepatomegaly along with hepatic ER and oxidative stress. This study revealed a novel role of the OST48 and AGE axis in hepatic injury through ER stress, changes in fuel utilisation and glucose intolerance.
Collapse
Affiliation(s)
- Aowen Zhuang
- Glycation and Diabetes, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Australia
- School of Medicine, University of Queensland, St Lucia, Australia
| | - Felicia Yt Yap
- Diabetic Complications Group, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
- Department of Immunology and Medicine, Central and Eastern Clinical School, AMREP Precinct, Monash University, Clayton, Australia
| | - Clinton Bruce
- Institute for Physical Activity and Nutrition (IPAN), Deakin University, Burwood, Australia
| | - Chris Leung
- Department of Medicine, University of Melbourne, Austin Hospital, Heidelberg, Australia
| | - Manuel R Plan
- Metabolomics Australia, Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Australia
| | - Mitchell A Sullivan
- Centre for Nutrition and Food Science, Queensland Alliance for Agriculture and Food Innovation, University of Queensland, St Lucia, Australia
| | - Chandana Herath
- Department of Medicine, University of Melbourne, Austin Hospital, Heidelberg, Australia
| | - Domenica McCarthy
- Glycation and Diabetes, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Karly C Sourris
- Diabetic Complications Group, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
- Department of Immunology and Medicine, Central and Eastern Clinical School, AMREP Precinct, Monash University, Clayton, Australia
| | - Phillip Kantharidis
- Diabetic Complications Group, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Melinda T Coughlan
- Diabetic Complications Group, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
- Department of Immunology and Medicine, Central and Eastern Clinical School, AMREP Precinct, Monash University, Clayton, Australia
| | - Mark A Febbraio
- Diabetic Complications Group, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Mark P Hodson
- Metabolomics Australia, Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Australia
- School of Pharmacy, University of Queensland, Woolloongabba, Australia
| | - Matthew J Watt
- Biomedicine Discovery Program and the Department of Physiology, Monash University, Clayton, Australia
| | - Peter Angus
- Department of Medicine, University of Melbourne, Austin Hospital, Heidelberg, Australia
| | - Benjamin L Schulz
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, Australia
| | - Josephine M Forbes
- Glycation and Diabetes, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Australia.
- Department of Medicine, University of Melbourne, Austin Hospital, Heidelberg, Australia.
- Mater Clinical School, University of Queensland, St Lucia, Australia.
| |
Collapse
|
15
|
Khanjarsim V, Karimi J, Khodadadi I, Mohammadalipour A, Goodarzi MT, Solgi G, Hashemnia M. Ameliorative Effects of Nilotinib on CCl4 Induced Liver Fibrosis Via Attenuation of RAGE/HMGB1 Gene Expression and Oxidative Stress in Rat. Chonnam Med J 2017; 53:118-126. [PMID: 28584790 PMCID: PMC5457946 DOI: 10.4068/cmj.2017.53.2.118] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 04/02/2017] [Accepted: 04/10/2017] [Indexed: 01/01/2023] Open
Abstract
Nilotinib as a tyrosine kinase inhibitor has been recently used to improve the liver fibrosis process, but the exact mechanisms still require further clarification. In this study, we investigated the anti-fibrotic effects of Nilotinib via RAGE/HMGB1axis and antioxidant mechanisms. This experimental study was performed in the Hamadan University of Medical Sciences, Iran, from May 2015 to December 2016. Liver fibrosis was induced in Wistar male rats by CCL4. Rats were gavaged daily with Nilotinib (10 mg/kg). RAGE, HMGB1, TNF-α and TGF-β mRNA expression were evaluated by quantitative RT-PCR. TNF-α protein levels were measured using the immunoassay method. Thiol groups, carbonyl groups, nitric oxide levels and glutathione peroxidase activity were measured by spectrophotometric methods.The results showed that Nilotinib decreased TNF-α, TGF-β, RAGE and HMGB1 mRNA expression (p<0.001) in the liver tissues of the fibrosis group. Nilotinib also decreased carbonyl groups and nitric oxide levels and increased thiol groups and glutathione peroxidase activity in the fibrosis groups. The histopathological changes were found to be attenuated by Nilotinib. In conclusion, Nilotinib can improve liver fibrosis and open new mechanisms of the anti-fibrotic properties of Nilotinib.
Collapse
Affiliation(s)
- Vahid Khanjarsim
- Department of Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Jamshid Karimi
- Department of Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Khodadadi
- Department of Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Adel Mohammadalipour
- Department of Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Taghi Goodarzi
- Department of Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ghasem Solgi
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Hashemnia
- Department of Pathobiology, Veterinary Medicine Faculty, Razi University, Kermanshah, Iran
| |
Collapse
|
16
|
Zaman A, Arif Z, Alam K. Fructosylation induced structural changes in mammalian DNA examined by biophysical techniques. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2017; 174:171-176. [PMID: 27902958 DOI: 10.1016/j.saa.2016.11.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/15/2016] [Accepted: 11/16/2016] [Indexed: 06/06/2023]
Abstract
Glycosylation of DNA, proteins, lipids, etc. by reducing sugars, can lead to the formation of advanced glycation end products (AGEs). These products may accumulate and involve in the pathogenesis of a number of diseases, contributing to tissue injury via several mechanisms. In this study, fructosylation of calf thymus dsDNA was carried out with varying concentrations of fructose. The neo-structure of fructosylated-DNA was studied by various biophysical techniques and morphological characterization. Fructosylated-DNA showed hyperchromicity, increase in fluorescence intensity and decrease in melting temperature. The CD signal of modified-DNA shifted in the direction of higher wavelength indicative of structural changes in DNA. FTIR results indicated shift in specific band positions in fructosylated-DNA. Morphological characterization of fructosylated-DNA exhibited strand breakage and aggregation. The results suggest that the structure and conformation of DNA may be altered under high concentrations of fructose.
Collapse
Affiliation(s)
- Asif Zaman
- Department of Biochemistry, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, India
| | - Zarina Arif
- Department of Biochemistry, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, India
| | - Khursheed Alam
- Department of Biochemistry, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
17
|
Tai CJ, Choong CY, Lin YC, Shi YC, Tai CJ. The anti-hepatic fibrosis activity of ergosterol depended on upregulation of PPARgamma in HSC-T6 cells. Food Funct 2016; 7:1915-23. [PMID: 27040153 DOI: 10.1039/c6fo00117c] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Advanced glycation endproducts (AGEs) were shown to play an important role in metabolic syndrome and were suggested to contribute to the development of hepatic fibrosis. Evidence indicates that AGEs resulted in hepatic fibrosis coupled to the activation of the receptor for AGEs (RAGE) in hepatic stellate cells (HSCs). NADPH oxidase is downstream of the RAGE signaling pathway, resulting in an increase in reactive oxygen species (ROS), alpha-smooth muscle actin (alpha-SMA), RAGE, and matrix metalloproteinase-9 (MMP-9). This study was designed to evaluate the effects of ergosterol on RAGE signaling in HSC-T6 cells. Ergosterol suppressed the activation of HSC-T6 cells induced by AGEs, and attenuated overexpressions of alpha-SMA, MMP-9, and epithelial-mesenchymal transition (EMT) markers, including N-cadherin and vimentin. We also found that these inhibitory effects of ergosterol on the activation of HSCs were dependent on peroxisome proliferator-activated receptor-gamma (PPARgamma) confirmed by PPARgamma reporter assay and PPARgamma knockdown. In addition, ergosterol also showed an inhibitory effect on the generation of AGEs, fructosamine, and α-dicarbonyl compounds in this study. Our results show that ergosterol can be used as a protective agent against hepatic fibrosis caused by induction of AGEs.
Collapse
Affiliation(s)
- Chen-Jei Tai
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan and Department of Chinese Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan and Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Chen-Yen Choong
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Chun Lin
- Graduate Institute of Medical Sciences, Taipei Medical University Hospital, Taipei, Taipei 11031, Taiwan
| | - Yeu-Ching Shi
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medicine University Hospital, Taipei 11031, Taiwan. and Taiwan Indigena Botanica Co., Ltd, Taipei City 11494, Taiwan
| | - Cheng-Jeng Tai
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medicine University Hospital, Taipei 11031, Taiwan. and Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
18
|
Leung C, Herath CB, Jia Z, Andrikopoulos S, Brown BE, Davies MJ, Rivera LR, Furness JB, Forbes JM, Angus PW. Dietary advanced glycation end-products aggravate non-alcoholic fatty liver disease. World J Gastroenterol 2016; 22:8026-8040. [PMID: 27672297 PMCID: PMC5028816 DOI: 10.3748/wjg.v22.i35.8026] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/22/2016] [Accepted: 08/10/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To determine if manipulation of dietary advanced glycation end product (AGE), intake affects non-alcoholic fatty liver disease (NAFLD) progression and whether these effects are mediated via RAGE.
METHODS Male C57Bl6 mice were fed a high fat, high fructose, high cholesterol (HFHC) diet for 33 wk and compared with animals on normal chow. A third group were given a HFHC diet that was high in AGEs. Another group was given a HFHC diet that was marinated in vinegar to prevent the formation of AGEs. In a second experiment, RAGE KO animals were fed a HFHC diet or a high AGE HFHC diet and compared with wildtype controls. Hepatic biochemistry, histology, picrosirius red morphometry and hepatic mRNA were determined.
RESULTS Long-term consumption of the HFHC diet generated significant steatohepatitis and fibrosis after 33 wk. In this model, hepatic 4-hydroxynonenal content (a marker of chronic oxidative stress), hepatocyte ballooning, picrosirius red staining, α-smooth muscle actin and collagen type 1A gene expression were all significantly increased. Increasing the AGE content of the HFHC diet by baking further increased these markers of liver damage, but this was abrogated by pre-marination in acetic acid. In response to the HFHC diet, RAGE-/- animals developed NASH of similar severity to RAGE+/+ animals but were protected from the additional harmful effects of the high AGE containing diet. Studies in isolated Kupffer cells showed that AGEs increase cell proliferation and oxidative stress, providing a likely mechanism through which these compounds contribute to liver injury.
CONCLUSION In the HFHC model of NAFLD, manipulation of dietary AGEs modulates liver injury, inflammation, and liver fibrosis via a RAGE dependent pathway. This suggests that pharmacological and dietary strategies targeting the AGE/RAGE pathway could slow the progression of NAFLD.
Collapse
|
19
|
Xia P, Deng Q, Gao J, Yu X, Zhang Y, Li J, Guan W, Hu J, Tan Q, Zhou L, Han W, Yuan Y, Yu Y. Therapeutic effects of antigen affinity-purified polyclonal anti-receptor of advanced glycation end-product (RAGE) antibodies on cholestasis-induced liver injury in rats. Eur J Pharmacol 2016; 779:102-10. [PMID: 26970185 DOI: 10.1016/j.ejphar.2016.03.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/04/2016] [Accepted: 03/07/2016] [Indexed: 01/22/2023]
Abstract
Cholestasis leads to acute hepatic injury, fibrosis/cirrhosis, inflammation, and duct proliferation. We investigated whether blocking receptor of advanced glycation end-products (RAGE) with polyclonal anti-RAGE antibodies (anti-RAGE) could regulate acute liver injury and fibrosis in a rat bile duct ligation (BDL) model. Male Wister rats received 0.5mg/kg rabbit anti-RAGE or an equal amount of rabbit IgG by subcutaneous injection twice a week after BDL. Samples of liver tissue and peripheral blood were collected at 14 days after BDL. Serum biochemistry and histology were used to analyze the degree of liver injury. Quantitative real-time PCR (qPCR) and immunohistochemical staining were used to further analyze liver injury. Anti-RAGE improved the gross appearance of the liver and the rat survival rate. Liver tissue histology and relevant serum biochemistry indicated that anti-RAGE attenuated liver necrosis, inflammation, liver fibrosis, and duct proliferation in the BDL model. qPCR and western blotting showed significant reductions in interleukin-1β expression levels in the liver by treatment with anti-RAGE. Anti-RAGE also significantly reduced the mRNA levels of α1(1) collagen (Col1α1) and cholesterol 7α-hydroxylase, and the ratio of tissue inhibitor of matrix metalloproteinase-1 to matrix metalloproteinases (MMPs) in the liver. In addition, anti-RAGE regulated the transcriptional level of Col1α1 and MMP-9 in transforming growth factor-β-induced activated LX-2 cells in vitro. Anti-RAGE was found to inhibit hepatic stellate cell proliferation in vivo and in vitro. Therefore, anti-RAGE can protect the liver from injury induced by BDL in rats.
Collapse
Affiliation(s)
- Peng Xia
- Shanghai Municipality Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China
| | - Qing Deng
- Shanghai Municipality Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China
| | - Jin Gao
- Laboratory of Regenerative Medicine, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China
| | - Xiaolan Yu
- Shanghai Municipality Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China
| | - Yang Zhang
- Laboratory of Regenerative Medicine, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China
| | - Jingjing Li
- Laboratory of Regenerative Medicine, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China
| | - Wen Guan
- Shanghai Municipality Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China
| | - Jianjun Hu
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Rd., Xuhui, Shanghai 200233, China
| | - Quanhui Tan
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Rd., Xuhui, Shanghai 200233, China
| | - Liang Zhou
- Shanghai Municipality Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China
| | - Wei Han
- Laboratory of Regenerative Medicine, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China
| | - Yunsheng Yuan
- Laboratory of Regenerative Medicine, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China; Engineering Research Center of Cell and Therapeutic Antibody, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China.
| | - Yan Yu
- Shanghai Municipality Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China.
| |
Collapse
|
20
|
Al-Qattan KK, Mansour MH, Thomson M, Ali M. Garlic decreases liver and kidney receptor for advanced glycation end products expression in experimental diabetes. ACTA ACUST UNITED AC 2016; 23:135-45. [PMID: 26968224 DOI: 10.1016/j.pathophys.2016.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 02/18/2016] [Accepted: 02/29/2016] [Indexed: 12/19/2022]
Abstract
The up-regulation of the receptor for advanced glycation end products (RAGE) has been implicated as a major mediator in the development and progression of diabetic nephropathy and hepatic fibrogenesis. The present study was designed to investigate the potential of garlic (Allium sativum L.) to modulate the level of expression of RAGE in renal and hepatic tissues of diabetic rats. Three groups of rats were studied after 8 weeks following diabetes induction: normal, streptozotocin-induced diabetic (control diabetic), and garlic-treated diabetic rats. A polyclonal antibody of proven specificity to RAGE indicated in immunohistochemical assays that RAGE labeling was significantly increased in renal and hepatic tissues of control diabetic rats compared to the normal group. The increased RAGE labeling involved mesangial cells in glomeruli exhibiting signs of mesangial expansion, mesangial nodule formation and glomerulosclerosis. In the liver, a significant up-regulation of RAGE was observed in hepatocytes and bile ducts and vessels in portal tracts. In 2-dimensional Western blots, RAGE expression in both tissues was dominated by heterogeneous charge variants, represented by 46-50kDa isoforms with more basic pIs compared to their counterparts in normal rats. Compared to control diabetic rats, RAGE labeling in the garlic-treated diabetic group was significantly reduced throughout renal and hepatic regions and was marked by the expression of 43-50kDa acidic charge variants comparable to those observed in normal rats. The capacity of garlic to modulate diabetes-induced up-regulation of selective RAGE polymorphic variants may be implicated in attenuating the detrimental consequences of excessive RAGE signaling manifested by diabetes-associated disorders.
Collapse
Affiliation(s)
- Khaled K Al-Qattan
- Department of Biological Sciences, Faculty of Science, Kuwait University, P.O. Box 5969, Safat 13060, Kuwait.
| | - Mohamed H Mansour
- Department of Biological Sciences, Faculty of Science, Kuwait University, P.O. Box 5969, Safat 13060, Kuwait
| | - Martha Thomson
- Department of Biological Sciences, Faculty of Science, Kuwait University, P.O. Box 5969, Safat 13060, Kuwait
| | - Muslim Ali
- Department of Biological Sciences, Faculty of Science, Kuwait University, P.O. Box 5969, Safat 13060, Kuwait
| |
Collapse
|
21
|
Liu Y, Xia JR, Cai XG. Effect of siRNA-mediated down-regulation of receptor for advanced glycation end products on expression of matrix metalloproteinase-1 and tissue inhibitor of metalloproteinase-1 in rat hepatic stellate cells and hepatic fibrosis. Shijie Huaren Xiaohua Zazhi 2015; 23:3012-3021. [DOI: 10.11569/wcjd.v23.i19.3012] [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
AIM: To investigate the effect of small interfering RNA (siRNA)-mediated down-regulation of receptor for advanced glycation end products (RAGE) on the expression of matrix metalloproteinase-1 (MMP-1) and tissue inhibitor of metalloproteinase-1 (TIMP-1) in primary rat hepatic stellate cells (HSCs) and hepatic fibrosis (HF).
METHODS: In in vitro experiment, primary rat HSCs were cultured and isolated. The pAKD-GR126 vector carrying siRNA targeting RAGE was constructed and transfected to primary rat HSCs. Blank cells and cells transfected with unspecific siRNA vector pAKD-NC were used as controls. In in vivo experiment, liver fibrosis was induced in SD rats with CCl4. pAKD-GR126 was transfected to liver fibrosis rats at different doses via the tail vein. A blank group, a liver fibrosis model group and an unspecific siRNA vector pAKD-NC-transfected group were used as controls. Real-time PCR and Western blot were used to detect the expression of RAGE, MMP-1 and TIMP-1. The histological changes of the liver were observed by HE and Masson staining methods.
RESULTS: The mRNA and protein expression of RAGE and TIMP-1 in pAKD-GR126-transfected primary HSCs was significantly lower than that in the blank group and unspecific siRNA vector pAKD-NC-transfected group (P < 0.05 for all). However, the level of MMP-1 in pAKD-GR126-transfected primary HSCs was significantly higher than that in the blank group and pAKD-NC-transfected group (P < 0.05 for all). In vivo, the mRNA and protein expression of RAGE and TIMP-1 was significantly lower and that of MMP-1 was significantly higher in the low-, medium-, and high-dose RAGE siRNA groups than in the liver fibrosis model group (P < 0.05 for all). Compared with the liver fibrosis model group, liver fibrosis was significantly milder in the low-, medium-, and high-dose RAGE siRNA groups, especially the high-dose group.
CONCLUSION: RAGE specific siRNA could decrease the expression of RAGE and TIMP-1, increase the expression of MMP-1 in primary rat HSCs and HF rats, and reduce the degree of rat hepatic fibrosis.
Collapse
|
22
|
Wang XW, Li WD, Xia JR, Li Z, Cai XG. Small interfering RNA targeting receptor for advanced glycation end products suppresses the generation of proinflammatory cytokines. Exp Ther Med 2015; 10:584-590. [PMID: 26622358 DOI: 10.3892/etm.2015.2569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 03/10/2015] [Indexed: 12/17/2022] Open
Abstract
The aim of the present study was to investigate the effect of receptor for advanced glycation end products (RAGE)-specific small interfering (si)RNA on the generation of proinflammatory cytokines in primary rat hepatic stellate cells (HSCs) and hepatic fibrotic (HF) rats. The RAGE-specific siRNA expression vector pAKD-GR126 was constructed, and then transfected into primary rat HSCs. Reverse transcription-quantitative polymerase chain reaction and western blot analyses were conducted to determine the mRNA and protein expression levels, respectively, of RAGE, tumor necrosis factor (TNF)-α and interleukin (IL)-6 in the primary HSCs. In addition, a CCl4-induced Sprague Dawley (SD) rat model of hepatic fibrosis was established, and pAKD-GR126 was injected into the SD rats via the tail vein. Serum TNF-α and IL-6 concentrations were determined using radioimmunoassay. The mRNA and protein expression levels of RAGE (mRNA, F=7.791; protein, F=36.513), TNF-α (mRNA, F=474.568; protein, F=123.500) and IL-6 (mRNA, F=203.463; protein, F=320.555) in the pAKD-GR126-transfected primary HSCs were significantly reduced compared with those in the control and pAKD-NC groups (P<0.05). Serum TNF-α and IL-6 levels in the low-, medium- and high-dose pAKD-GR126 treatment groups were reduced compared with those in the fibrotic model group (TNF-α, F=416.397; IL-6, F=1,716.659; P<0.05). In summary, the RAGE-specific siRNA was able to effectively suppress the generation of the proinflammatory cytokines TNF-α and IL-6 in primary rat HSCs and HF rats.
Collapse
Affiliation(s)
- Xiao-Wei Wang
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Wei-Dong Li
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Jin-Rong Xia
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Zhan Li
- Department of Gastroenterology, Langxi Chinese Medicine Hospital, Langxi, Anhui 242100, P.R. China
| | - Xiao-Gang Cai
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
23
|
|
24
|
Xia JR, Chen TT, Li WD, Lu FL, Liu J, Cai XG, Lu Q, Yang CP. Inhibitory effect of receptor for advanced glycation end product‑specific small interfering RNAs on the development of hepatic fibrosis in primary rat hepatic stellate cells. Mol Med Rep 2015; 12:569-74. [PMID: 25673150 DOI: 10.3892/mmr.2015.3342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 12/19/2014] [Indexed: 11/06/2022] Open
Abstract
Specific small interfering RNAs (siRNAs) targeting receptor for advanced glycation end products (RAGE) inhibit the expression of RAGE, α-smooth muscle actin and type I collagen in the T6 hepatic stellate cells (HSCs), indicating that RAGE is important for the activation of HSCs and the expression of collagen. The present study aimed to investigate the effect of specific siRNAs targeting RAGE on the development of hepatic fibrosis (HF), using primary rat HSCs, which were isolated and cultured in vitro. The expression vectors for specific siRNAs targeting RAGE were constructed and transfected into primary rat HSCs. Untreated and nonspecific siRNA-transfected primary rat HSCs served as controls. The expression levels of RAGE, interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), transforming growth factor-β1 (TGF-β1), connective tissue growth factor (CTGF), laminin (LN), hyaluronic acid (HA) and N-terminal procollagen III propeptide (PIIINP) in primary HSCs were detected by reverse transcription quantitative polymerase chain reaction and western blotting. The mRNA and 42 kD protein expression of RAGE in the pAKD-GR126-transfected primary HSCs were significantly downregulated compared with those in the untreated and the pAKD-negative control (NC)-transfected controls. The mRNA and protein expression levels of IL-6, TNF-α, TGF-β1, CTGF, LN, HA and PIIINP in the pAKD-GR126-transfected primary HSCs were also markedly downregulated compared with those in the untreated and pAKD-NC-transfected controls. Therefore, RAGE-specific siRNAs inhibited the expression of RAGE in primary rat HSCs and inhibited the development of HF.
Collapse
Affiliation(s)
- Jin-Rong Xia
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Ting-Ting Chen
- Department of Gastroenterology, Binzhou People's Hospital, Binzhou, Shandong 256610, P.R. China
| | - Wei-Dong Li
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Feng-Lin Lu
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Juan Liu
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Xiao-Gang Cai
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Qin Lu
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Cui-Ping Yang
- Department of Gastroenterology, Shanghai First People's Hospital, Shanghai 200080, P.R. China
| |
Collapse
|
25
|
Takeuchi M, Takino JI, Sakasai-Sakai A, Takata T, Ueda T, Tsutsumi M, Hyogo H, Yamagishi SI. Involvement of the TAGE-RAGE system in non-alcoholic steatohepatitis: Novel treatment strategies. World J Hepatol 2014; 6:880-893. [PMID: 25544875 PMCID: PMC4269907 DOI: 10.4254/wjh.v6.i12.880] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 09/12/2014] [Accepted: 10/29/2014] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major cause of liver disease around the world. It includes a spectrum of conditions from simple steatosis to non-alcoholic steatohepatitis (NASH) and can lead to fibrosis, cirrhosis, liver failure, and/or hepatocellular carcinoma. NAFLD is also associated with other medical conditions such as obesity, diabetes mellitus (DM), metabolic syndrome, hypertension, insulin resistance, hyperlipidemia, and cardiovascular disease (CVD). In diabetes, chronic hyperglycemia contributes to the development of both macro- and microvascular conditions through a variety of metabolic pathways. Thus, it can cause a variety of metabolic and hemodynamic conditions, including upregulated advanced glycation end-products (AGEs) synthesis. In our previous study, the most abundant type of toxic AGEs (TAGE); i.e., glyceraldehyde-derived AGEs, were found to make a significant contribution to the pathogenesis of DM-induced angiopathy. Furthermore, accumulating evidence suggests that the binding of TAGE with their receptor (RAGE) induces oxidative damage, promotes inflammation, and causes changes in intracellular signaling and the expression levels of certain genes in various cell populations including hepatocytes and hepatic stellate cells. All of these effects could facilitate the pathogenesis of hypertension, cancer, diabetic vascular complications, CVD, dementia, and NASH. Thus, inhibiting TAGE synthesis, preventing TAGE from binding to RAGE, and downregulating RAGE expression and/or the expression of associated effector molecules all have potential as therapeutic strategies against NASH. Here, we examine the contributions of RAGE and TAGE to various conditions and novel treatments that target them in order to prevent the development and/or progression of NASH.
Collapse
|
26
|
Takeuchi M, Takino JI, Sakasai-Sakai A, Takata T, Ueda T, Tsutsumi M, Hyogo H, Yamagishi SI. Involvement of the TAGE-RAGE system in non-alcoholic steatohepatitis: Novel treatment strategies. World J Hepatol 2014. [PMID: 25544875 DOI: 10.4254/wjh.6.i12.880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major cause of liver disease around the world. It includes a spectrum of conditions from simple steatosis to non-alcoholic steatohepatitis (NASH) and can lead to fibrosis, cirrhosis, liver failure, and/or hepatocellular carcinoma. NAFLD is also associated with other medical conditions such as obesity, diabetes mellitus (DM), metabolic syndrome, hypertension, insulin resistance, hyperlipidemia, and cardiovascular disease (CVD). In diabetes, chronic hyperglycemia contributes to the development of both macro- and microvascular conditions through a variety of metabolic pathways. Thus, it can cause a variety of metabolic and hemodynamic conditions, including upregulated advanced glycation end-products (AGEs) synthesis. In our previous study, the most abundant type of toxic AGEs (TAGE); i.e., glyceraldehyde-derived AGEs, were found to make a significant contribution to the pathogenesis of DM-induced angiopathy. Furthermore, accumulating evidence suggests that the binding of TAGE with their receptor (RAGE) induces oxidative damage, promotes inflammation, and causes changes in intracellular signaling and the expression levels of certain genes in various cell populations including hepatocytes and hepatic stellate cells. All of these effects could facilitate the pathogenesis of hypertension, cancer, diabetic vascular complications, CVD, dementia, and NASH. Thus, inhibiting TAGE synthesis, preventing TAGE from binding to RAGE, and downregulating RAGE expression and/or the expression of associated effector molecules all have potential as therapeutic strategies against NASH. Here, we examine the contributions of RAGE and TAGE to various conditions and novel treatments that target them in order to prevent the development and/or progression of NASH.
Collapse
Affiliation(s)
- Masayoshi Takeuchi
- Masayoshi Takeuchi, Akiko Sakasai-Sakai, Takanobu Takata, Tadashi Ueda, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada-machi, Ishikawa 920-0293, Japan
| | - Jun-Ichi Takino
- Masayoshi Takeuchi, Akiko Sakasai-Sakai, Takanobu Takata, Tadashi Ueda, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada-machi, Ishikawa 920-0293, Japan
| | - Akiko Sakasai-Sakai
- Masayoshi Takeuchi, Akiko Sakasai-Sakai, Takanobu Takata, Tadashi Ueda, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada-machi, Ishikawa 920-0293, Japan
| | - Takanobu Takata
- Masayoshi Takeuchi, Akiko Sakasai-Sakai, Takanobu Takata, Tadashi Ueda, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada-machi, Ishikawa 920-0293, Japan
| | - Tadashi Ueda
- Masayoshi Takeuchi, Akiko Sakasai-Sakai, Takanobu Takata, Tadashi Ueda, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada-machi, Ishikawa 920-0293, Japan
| | - Mikihiro Tsutsumi
- Masayoshi Takeuchi, Akiko Sakasai-Sakai, Takanobu Takata, Tadashi Ueda, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada-machi, Ishikawa 920-0293, Japan
| | - Hideyuki Hyogo
- Masayoshi Takeuchi, Akiko Sakasai-Sakai, Takanobu Takata, Tadashi Ueda, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada-machi, Ishikawa 920-0293, Japan
| | - Sho-Ichi Yamagishi
- Masayoshi Takeuchi, Akiko Sakasai-Sakai, Takanobu Takata, Tadashi Ueda, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada-machi, Ishikawa 920-0293, Japan
| |
Collapse
|
27
|
Umadevi S, Gopi V, Vellaichamy E. Inhibitory effect of gallic acid on advanced glycation end products induced up-regulation of inflammatory cytokines and matrix proteins in H9C2 (2-1) cells. Cardiovasc Toxicol 2014; 13:396-405. [PMID: 24062022 DOI: 10.1007/s12012-013-9222-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Accumulating evidences have demonstrated that increased production of advanced glycation end products (AGEs) contributes to etiology of cardiac complications in diabetes. However, the underlying mechanism of AGE-induced effects is not well understood. Recent studies evince the beneficial role of phytochemicals in reducing the risk of cardiovascular morbidity and mortality in patients with cardiovascular diseases and diabetes mellitus. Hence, in the present study, the cardioprotective role of gallic acid (GA) against in vitro synthesized AGE in H9C2 (2-1) cells was elucidated. H9C2 (2-1) cells exposed to AGE (100 μg/ml) with/without GA pre-treatment (10 μM) and the release of reactive oxygen species (ROS), expression of oxidative stress markers, matrix proteins, and cytokines were analyzed. Cells exposed to AGE demonstrate a significant increase in ROS release with augmented expression (P < 0.01) of receptor for AGE (RAGE) and NOX-p47 phox (P < 0.001) proteins compared to untreated control cells. Moreover, an increased expression of matrix proteins and cytokines such as TNF-α (P < 0.01), TGF-β (P < 0.001), and iNOS (P < 0.001) was also found in AGE-treated cells, whereas, cells pre-treated with N-acetyl cysteine or RAGE neutralizing antibody notably (P < 0.01) impede the ROS release. Further, cells pre-treated with GA significantly attenuated the expression of NOX, RAGE, and other cytokines. In addition, the abnormal expressions of matrix proteins were also decreased especially in GA-treated cells. Thus, the results of the present study demonstrated the deleterious effect of AGEs that directly induce oxidative stress and matrix derangement and, on the other way, the "pleiotropic" activity of GA in reducing the risk of AGE-mediated cellular complications.
Collapse
Affiliation(s)
- Subramanian Umadevi
- Department of Biochemistry, University of Madras, Guindy Campus, Chennai, 600 025, Tamil Nadu, India
| | | | | |
Collapse
|
28
|
Kao YH, Lin YC, Tsai MS, Sun CK, Yuan SS, Chang CY, Jawan B, Lee PH. Involvement of the nuclear high mobility group B1 peptides released from injured hepatocytes in murine hepatic fibrogenesis. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1720-32. [PMID: 24970745 DOI: 10.1016/j.bbadis.2014.06.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Revised: 05/24/2014] [Accepted: 06/16/2014] [Indexed: 12/27/2022]
Abstract
This study investigated the pro-fibrogenic role of high mobility group box 1 (HMGB1) peptides in liver fibrogenesis. An animal model of carbon tetrachloride (CCl4)-induced liver fibrosis was used to examine the serum HMGB1 levels and its intrahepatic distribution. The increased serum HMGB1 levels were positively correlated with elevation of transforming growth factor-β1 (TGF-β1) and collagen deposition during fibrogenesis. The cytoplasmic distribution of HMGB1 was noted in the parenchymal hepatocytes of fibrotic livers. In vitro studies confirmed that exposure to hydrogen peroxide and CCl4 induced an intracellular mobilization and extracellular release of nuclear HMGB1 peptides in clone-9 and primary hepatocytes, respectively. An uptake of exogenous HMGB1 by hepatic stellate cells (HSCs) T6 cells indicated a possible paracrine action of hepatocytes on HSCs. Moreover, HMGB1 dose-dependently stimulated HSC proliferation, up-regulated de novo synthesis of collagen type I and α-smooth muscle actin (α-SMA), and triggered Smad2 phosphorylation and its nuclear translocation through a TGF-β1-independent mechanism. Blockade with neutralizing antibodies and gene silencing demonstrated the involvement of the receptor for advanced glycation end-products (RAGE), but not toll-like receptor 4, in cellular uptake of HMGB1 and the HMGB1-mediated Smad2 and ERK1/2 phosphorylation as well as α-SMA up-regulation in HSC-T6 cells. Furthermore, anti-RAGE treatment significantly ameliorated CCl4-induced liver fibrosis. In conclusion, the nuclear HMGB1 peptides released from parenchymal hepatocytes during liver injuries may directly activate HSCs through stimulating HSC proliferation and transformation, eventually leading to the fibrotic changes of livers. Blockade of HMGB1/RAGE signaling cascade may constitute a therapeutic strategy for treatment of liver fibrosis.
Collapse
Affiliation(s)
- Ying-Hsien Kao
- Department of Medical Research, E-DA Hospital, Kaohsiung, Taiwan
| | - Yu-Chun Lin
- Department of Surgery, E-DA Hospital, Kaohsiung, Taiwan
| | | | - Cheuk-Kwan Sun
- Department of Medical Education, E-DA Hospital, Kaohsiung, Taiwan
| | - Shyng-Shiou Yuan
- Translational Research Center and Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Yang Chang
- Department of Obstetrics and Gynecology, E-DA Hospital, Kaohsiung, Taiwan
| | - Bruno Jawan
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
| | - Po-Huang Lee
- Department of Surgery, E-DA Hospital, Kaohsiung, Taiwan; Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
29
|
Cai XG, Xia JR, Li WD, Lu FL, Liu J, Lu Q, Zhi H. Anti-fibrotic effects of specific-siRNA targeting of the receptor for advanced glycation end products in a rat model of experimental hepatic fibrosis. Mol Med Rep 2014; 10:306-14. [PMID: 24804792 DOI: 10.3892/mmr.2014.2207] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 03/18/2014] [Indexed: 12/31/2022] Open
Abstract
Since the receptor for advanced glycation end products (RAGE)-ligand axis has been demonstrated to be important in fibrogenesis, rat models may be used to assess whether specific small interfering RNAs (siRNAs) that target RAGE are able to reduce the progression of hepatic fibrosis. However, the effect of RAGE-targeted siRNA on established hepatic fibrosis remains to be elucidated. In the present study, RAGE-specific siRNA expression vectors were constructed prior to the animal experiment. Sprague-Dawley rats were treated initially with olive oil (2 ml/kg) or 50% CCl4 (2 ml/kg; CCl4/olive oil=1:1) twice per week for six weeks to generate the fibrosis model. The rats were then treated with phosphate‑buffered saline, a RAGE-specific siRNA expression vector, at different doses or a non-specific siRNA expression vector twice weekly via tail vein injection for up to six weeks, and were sacrificed at week two, four or six. Compared with the control groups, RAGE‑specific siRNA therapy significantly decreased RAGE mRNA and protein expression in rat livers (P<0.01). Following six weeks of RAGE gene-silencing treatment, the liver function, which was assessed by analyzing serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP) and total bilirubin (TBIL), improved to varying degrees (P<0.01). The expression of nuclear factor-κB (NF-κB) significantly decreased following RAGE gene‑silencing therapy (P<0.01). In addition, the serum levels of inflammatory cytokines, including tumor necrosis factor‑α (TNF-α) and interleukin-6 (IL-6), and extracellular matrix (ECM) components, including hyaluronic acid (HA), laminin (LN) and procollagen type III (PCIII) also decreased (P<0.01). Furthermore, the expression of α-smooth muscle actin (α-SMA) and collagen I, which indicate the activation of hepatic stellate cells (HSCs), were downregulated following RAGE gene-silencing therapy (P<0.01). Furthermore, the inflammatory activity grade and fibrosis stage of rat livers also significantly improved compared with the control groups following RAGE gene-silencing therapy. Specific targeting of RAGE using siRNA may inhibit RAGE gene expression effectively in the rat hepatic fibrosis model and attenuate the progression of established hepatic fibrosis. This therapeutic effect may be mediated via inhibition of the expression of NF-κB. These findings suggest that RAGE may be a new target to prevent hepatic fibrosis.
Collapse
Affiliation(s)
- Xiao-Gang Cai
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Jin-Rong Xia
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Wei-Dong Li
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Feng-Lin Lu
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Juan Liu
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Qin Lu
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Hong Zhi
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
30
|
Curcumin eliminates the effect of advanced glycation end-products (AGEs) on the divergent regulation of gene expression of receptors of AGEs by interrupting leptin signaling. J Transl Med 2014; 94:503-16. [PMID: 24614199 PMCID: PMC4006284 DOI: 10.1038/labinvest.2014.42] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 01/03/2014] [Accepted: 01/15/2014] [Indexed: 12/30/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a major risk factor for hepatic fibrogenesis. NASH is often found in diabetic patients with hyperglycemia. Hyperglycemia induces non-enzymatic glycation of proteins, yielding advanced glycation end-products (AGEs). Effects of AGEs are mainly mediated by two categories of cytoplasmic membrane receptors. Receptor for AGEs (RAGE) is associated with increased oxidative stress and inflammation, whereas AGE receptor-1 (AGE-R1) is involved in detoxification and clearance of AGEs. Activation of hepatic stellate cells (HSC) is crucial to the development of hepatic fibrosis. We recently reported that AGEs stimulated HSC activation likely by inhibiting gene expression of AGE-R1 and inducing gene expression of RAGE in HSC, which were eliminated by the antioxidant curcumin. This study is to test our hypothesis that curcumin eliminates the effects of AGEs on the divergent regulation of the two receptors of AGEs in HSC by interrupting the AGE-caused activation of leptin signaling, leading to the inhibition of HSC activation. We observed herein that AGEs activated leptin signaling by inducing gene expression of leptin and its receptor in HSC. Like AGEs, leptin differentially regulated gene expression of RAGE and AGE-R1. Curcumin eliminated the effects of AGEs in HSC by interrupting leptin signaling and activating transcription factor NF-E2 p45-related factor 2 (Nrf2), leading to the elevation of cellular glutathione and the attenuation of oxidative stress. In conclusions, curcumin eliminated the effects of AGEs on the divergent regulation of gene expression of RAGE and AGE-R1 in HSC by interrupting the AGE-caused activation of leptin signaling, leading to the inhibition of HSC activation.
Collapse
|
31
|
Leung C, Herath CB, Jia Z, Goodwin M, Mak KY, Watt MJ, Forbes JM, Angus PW. Dietary glycotoxins exacerbate progression of experimental fatty liver disease. J Hepatol 2014; 60:832-8. [PMID: 24316518 DOI: 10.1016/j.jhep.2013.11.033] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 11/05/2013] [Accepted: 11/25/2013] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Advanced glycation end-products (AGEs) levels are high in western diets and contribute to tissue injury via activation of RAGE (receptor for AGEs) and generation of reactive oxygen species (ROS). Here, we determined if high dietary AGE intake worsens progression of non-alcoholic fatty liver disease (NAFLD). METHODS Male Sprague Dawley rats were fed a methionine choline deficient (MCD) diet for 6 weeks before 6 weeks of a high AGE MCD diet through baking. They were compared with animals on MCD diet or a methionine choline replete (MCR) diet alone for 12 weeks. Hepatic ROS, triglycerides, biochemistry, picro-sirius morphometry, hepatic mRNA expression and immunohistochemistry were determined. Primary hepatic stellate cells (HSCs) from both MCR and MCD animals were exposed to AGEs. ROS, proliferation and mRNA expression were determined. RESULTS The high AGE MCD diet increased hepatic AGE content and elevated triglycerides, NADPH dependent superoxide production, HNE adducts, steatosis, steatohepatitis (CD43, IL-6, TNF-α) and fibrosis (α-SMA, CTGF, COL1A, picrosirius) compared to MCD alone. In HSCs, AGEs significantly increased ROS production, bromodeoxyuridine proliferation and MCP-1, IL-6, α-SMA, and RAGE expression in HSCs from MCD but not MCR animals. These effects were abrogated by RAGE or NADPH oxidase blockade. CONCLUSIONS In the MCD model of NAFLD, high dietary AGEs increases hepatic AGE content and exacerbates liver injury, inflammation, and liver fibrosis via oxidative stress and RAGE dependent profibrotic effects of AGEs on activated HSCs. This suggests that pharmacological and dietary strategies targeting the AGE/RAGE pathway could slow the progression of NAFLD.
Collapse
Affiliation(s)
- Christopher Leung
- Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria, Australia; Department of Gastroenterology and Hepatology, Austin Health, Austin Hospital, Heidelberg, Melbourne, Victoria, Australia; Glycation and Diabetes Group, Baker IDI Diabetes Institute, Melbourne, Victoria, Australia.
| | - Chandana B Herath
- Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria, Australia
| | - Zhiyuan Jia
- Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria, Australia
| | - Michelle Goodwin
- Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria, Australia; Department of Gastroenterology and Hepatology, Austin Health, Austin Hospital, Heidelberg, Melbourne, Victoria, Australia
| | - Kai Yan Mak
- Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria, Australia
| | - Matthew J Watt
- Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Josephine M Forbes
- Glycation and Diabetes Group, Baker IDI Diabetes Institute, Melbourne, Victoria, Australia; Glycation and Diabetes Complications Group, Mater Medical Research Institute, South Brisbane, Queensland, Australia
| | - Peter W Angus
- Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria, Australia; Department of Gastroenterology and Hepatology, Austin Health, Austin Hospital, Heidelberg, Melbourne, Victoria, Australia
| |
Collapse
|
32
|
Hsu WH, Lee BH, Hsu YW, Pan TM. Peroxisome proliferator-activated receptor-γ activators monascin and rosiglitazone attenuate carboxymethyllysine-induced fibrosis in hepatic stellate cells through regulating the oxidative stress pathway but independent of the receptor for advanced glycation end products signaling. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2013; 61:6873-6879. [PMID: 23796251 DOI: 10.1021/jf402082g] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Advanced glycation end products (AGEs) signaling through its receptors (RAGE) results in an increase in reactive oxygen species (ROS) and is thought to contribute to hepatic fibrosis via hyperglycemia. Carboxymethyllysine (CML) is a key AGE, with highly reactive dicarbonyl metabolites. We investigated the inhibitory effect of Monascus -fermented metabolite monascin and rosiglitazone on CML-induced RAGE signaling in hepatic stellate cells (HSCs) and its resulting antihepatic fibrosis activity. We found that monascin and rosiglitazone upregulated peroxisome proliferator-activated receptor-γ (PPAR-γ) to attenuate α-smooth muscle actin (SMA) and ROS generation in CML-treated HSCs in a RAGE activation-independent pathway. Therefore, monascin may delay or inhibit the progression of liver fibrosis through the activation of PPAR-γ and might prove to be a major antifibrotic mechanism to prevent liver disease.
Collapse
Affiliation(s)
- Wei-Hsuan Hsu
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | | | | | | |
Collapse
|
33
|
Wang FP, Li L, Li J, Wang JY, Wang LY, Jiang W. High mobility group box-1 promotes the proliferation and migration of hepatic stellate cells via TLR4-dependent signal pathways of PI3K/Akt and JNK. PLoS One 2013; 8:e64373. [PMID: 23696886 PMCID: PMC3655989 DOI: 10.1371/journal.pone.0064373] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 04/12/2013] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The migration of hepatic stellate cells (HSCs) is essential to the hepatic fibrotic response, and recently High-mobility group box 1 (HMGB1) has been shown up-regulated during liver fibrosis. Nevertheless, whether HMGB1 can modulate the proliferation and migration of HSCs is poorly understood, as well as the involved intracellular signaling. In this study, we examined the effect of HMGB1 on proliferation, migration, pro-fibrotic function of HSCs and investigated whether toll-like family of receptor 4 (TLR4) dependent signal pathway is involved in the intracellular signaling regulation. METHODOLOGY/PRINCIPAL FINDINGS Modified transwell chamber system to mimic the space of Disse was used to evaluate the migration of human primary HSCs, and the protein expressions of related signal factors were evaluated by western blot. Cell proliferation was analyzed by MTT assay, the pro-fibrotic functions of HSCs by qRT-PCR and ELISA respectively. Recombinant human HMGB1 could significantly promote migration of HSCs under both haptotactic and chemotactic stimulation, especially the latter. Human TLR4 neutralizing antibody could markedly inhibit HMGB1-induced migration of HSCs. HMGB1 could enhance the phosphorylation of JNK and PI3K/Akt, and TLR4 neutralizing antibody inhibited HMGB1-enhanced phosphorylation of JNK and PI3K/Akt and activation of NF-κB. JNK inhibitor (SP600125) and PI3K inhibitor (LY 294002) significantly inhibited HMGB1-induced proliferation and migration of HSCs, and also reduced HMGB1-enhanced related collagen expressions and pro-fibrotic cytokines production. CONCLUSIONS/SIGNIFICANCE HMGB1 could significantly enhance migration of HSCs in vitro, and TLR4-dependent JNK and PI3K/Akt signal pathways are involved in the HMGB1-induced proliferation, migration and pro-fibrotic effects of HSCs, which indicates HMGB1 might be an effective target to treat liver fibrosis.
Collapse
Affiliation(s)
- Fu-ping Wang
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lei Li
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Li
- Department of Gastroenterology, Tongji Hospital, Tongji University, Shanghai, China
| | - Ji-yao Wang
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ling-yan Wang
- Biomedical Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Jiang
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
34
|
Hamasaki MY, Hirata MH, Hirata RDC, Himelfarb ST, Campos LMG, Nogueira MI. [Analysis of the mRNA expression of the S100β protein in adipocytes of patients with diabetes mellitus, type 2]. ACTA ACUST UNITED AC 2013; 56:435-40. [PMID: 23108748 DOI: 10.1590/s0004-27302012000700005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 08/30/2012] [Indexed: 11/22/2022]
Abstract
OBJECTIVE This study aims to explore the possible relationship between the expression level of S100β protein mRNA with diabetes mellitus type 2 in adipocytes from patients with this disease in comparison with normoglycemic individuals. MATERIALS AND METHODS Samples of adipose tissue of eight patients from the coronary section of the Institute Dante Pazzanese of Cardiology (IDPC), four in Group Diabetes and four of Normoglycemic group, were evaluated by RT-PCR real time. RESULTS An increase around 15 times values, between the threshold cycle (ΔCt), of mRNA expression of S100β protein in adipocytes of the diabetes group was observed in comparison to the control group (p = 0.015). CONCLUSION Our results indicate, for the first time, that there is coexistence of increased expression of the S100β and the type 2 diabetes mellitus gene.
Collapse
Affiliation(s)
- Mike Yoshio Hamasaki
- Laboratório de Neurociências, Departamento de Anatomia, Instituto de Ciências Biomédicas III, Universidade de São Paulo, São Paulo, SP, Brasil
| | | | | | | | | | | |
Collapse
|
35
|
Goodwin M, Herath C, Jia Z, Leung C, Coughlan MT, Forbes J, Angus P. Advanced glycation end products augment experimental hepatic fibrosis. J Gastroenterol Hepatol 2013; 28:369-76. [PMID: 23173780 DOI: 10.1111/jgh.12042] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/02/2012] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND AIMS Advanced glycation end products (AGEs) are nonenzymatic modifications of proteins by reducing sugars. These compounds accumulate in a number of chronic disease states, contributing to tissue injury via several mechanisms, including activation of the receptor for advanced glycation end products (RAGE). We aimed to investigate whether AGEs can exacerbate chronic liver injury and contribute to hepatic fibrosis. METHODS We initially studied the effects of chronic hepatic exposure to high levels of AGEs given intraperitoneally as AGE-rat serum albumin. In a separate experiment, we examined the impact of high AGE exposure in rats following bile duct ligation (BDL). RESULTS In normal rats, chronic AGE-rat serum albumin administration induced significant increases in α-smooth muscle actin gene and protein expression but did not induce fibrosis or biochemical evidence of liver injury. However, in BDL animals, AGE-bovine serum albumin administration significantly increased hepatic fibrosis as evidenced by increased collagen content and α-smooth muscle actin expression, compared with BDL alone. Furthermore, AGEs increased hepatic oxidative stress and receptor for advanced glycation end products gene expression. CONCLUSIONS These findings suggest that AGEs may contribute to the pathogenesis of chronic liver injury and fibrosis.
Collapse
Affiliation(s)
- Michelle Goodwin
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia.
| | | | | | | | | | | | | |
Collapse
|
36
|
Honsawek S, Vejchapipat P, Payungporn S, Theamboonlers A, Chongsrisawat V, Poovorawan Y. Soluble receptor for advanced glycation end products and liver stiffness in postoperative biliary atresia. Clin Biochem 2013; 46:214-218. [PMID: 23195134 DOI: 10.1016/j.clinbiochem.2012.11.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 11/10/2012] [Accepted: 11/11/2012] [Indexed: 11/22/2022]
Abstract
OBJECTIVES Soluble receptor for advanced glycation end products (sRAGE) has emerged as a possible biomarker of several disease conditions, including liver injury. This study was aimed to assess serum sRAGE and liver stiffness in biliary atresia (BA). DESIGN AND METHODS Forty postoperative BA patients and 20 controls were enrolled. Serum sRAGE levels were analyzed by enzyme-linked immunosorbent assay. Liver stiffness scores were measured by transient elastography. RESULTS BA patients had higher serum sRAGE and liver stiffness values than controls (P<0.001). Serum sRAGE and liver stiffness values were significantly elevated in BA patients with jaundice compared to those without jaundice (P<0.001). Additionally, serum sRAGE was correlated with liver stiffness and serum total bilirubin (r=0.65, P<0.001 and r=0.71, P<0.001, respectively). CONCLUSION Serum sRAGE was associated with the severity of BA. Accordingly, serum sRAGE and liver stiffness may serve as indicators reflecting the severity and the development of hepatic fibrosis in postoperative BA.
Collapse
Affiliation(s)
- Sittisak Honsawek
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand.
| | | | | | | | | | | |
Collapse
|
37
|
Liu Y, Gardner CR, Laskin JD, Laskin DL. Classical and alternative activation of rat hepatic sinusoidal endothelial cells by inflammatory stimuli. Exp Mol Pathol 2012; 94:160-7. [PMID: 23103612 DOI: 10.1016/j.yexmp.2012.10.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 10/17/2012] [Accepted: 10/18/2012] [Indexed: 12/16/2022]
Abstract
The ability of rat hepatic sinusoidal endothelial cells (HSEC) to become activated in response to diverse inflammatory stimuli was analyzed. Whereas the classical macrophage activators, IFNγ and/or LPS upregulated expression of iNOS in HSEC, the alternative macrophage activators, IL-10 or IL-4+IL-13 upregulated arginase-1 and mannose receptor. Similar upregulation of iNOS and arginase-1 was observed in classically and alternatively activated Kupffer cells, respectively. Removal of inducing stimuli from the cells had no effect on expression of these markers, demonstrating that activation is persistent. Washing and incubation of IFNγ treated cells with IL-4+IL-13 resulted in decreased iNOS and increased arginase-1 expression, while washing and incubation of IL-4+IL-13 treated cells with IFNγ resulted in decreased arginase-1 and increased iNOS, indicating that classical and alternative activation of the cells is reversible. HSEC were more sensitive to phenotypic switching than Kupffer cells, suggesting greater functional plasticity. Hepatocyte viability and expression of PCNA, β-catenin and MMP-9 increased in the presence of alternatively activated HSEC. In contrast, the viability of hepatocytes pretreated for 2 h with 5 mM acetaminophen decreased in the presence of classically activated HSEC. These data demonstrate that activated HSEC can modulate hepatocyte responses following injury. The ability of hepatocytes to activate HSEC was also investigated. Co-culture of HSEC with acetaminophen-injured hepatocytes, but not control hepatocytes, increased the sensitivity of HSEC to classical and alternative activating stimuli. The capacity of HSEC to respond to phenotypic activators may represent an important mechanism by which they participate in inflammatory responses associated with hepatotoxicity.
Collapse
Affiliation(s)
- Yinglin Liu
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, 160 Frelinghuysen Rd., Piscataway, NJ 08854, USA
| | | | | | | |
Collapse
|
38
|
Arshad MI, Piquet-Pellorce C, Samson M. IL-33 and HMGB1 alarmins: sensors of cellular death and their involvement in liver pathology. Liver Int 2012; 32:1200-10. [PMID: 22530772 DOI: 10.1111/j.1478-3231.2012.02802.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 03/12/2012] [Indexed: 12/20/2022]
Abstract
'Alarmins' are a group of proteins or molecules that are released from cells during cellular demise to alert the host immune system. Two of them, Interleukin-33 (IL-33) and high-mobility group box-1 (HMGB1), share many similarities of cellular localization, functions and involvement in various inflammatory pathologies including hepatitis. The expressions of IL-33 and HMGB1, and their receptors ST2 and receptor for advanced glycation end products (RAGE), are substantially up-regulated during acute and chronic hepatitis. Recent data evidence a possible protective role of IL-33/ST2 axis during liver injury. A contrast in expression of IL-33 and HMGB1 alarmins were associated with type of hepatocellular death mediated by immune cells or hepato-toxic agents. The massive release of active form of IL-33 from hepatocytes may affect the recruitment and activation of its ST2-positive target immune cells in the liver to confer its alarmin functions. This review highlights the emerging roles of alarmin proteins in various liver pathologies, by focusing on classical HMGB1 and a newly discovered alarmin, the IL-33.
Collapse
Affiliation(s)
- Muhammad I Arshad
- Institut de Recherche Santé Environnement & Travail, Institut National de la Santé et de la Recherche Médicale (Inserm), U.1085, Rennes, France
| | | | | |
Collapse
|
39
|
Basta G, Navarra T, De Simone P, Del Turco S, Gastaldelli A, Filipponi F. What is the role of the receptor for advanced glycation end products-ligand axis in liver injury? Liver Transpl 2011; 17:633-40. [PMID: 21438128 DOI: 10.1002/lt.22306] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Multiligand receptor for advanced glycation end products (RAGE) is expressed in a wide variety of tissues, including the liver. Interactions with its ligands lead to cellular activation and thus prolonged inflammation and apoptosis. RAGE also exists in a soluble, truncated isoform called soluble RAGE, which has the same ligand-binding specificity as membrane-RAGE; acting as decoy, it can contribute to the removal/neutralization of circulating ligands and the resultant reduction of signaling pathway activation. Experimental and clinical studies have highlighted the idea that the RAGE-ligand axis is involved in the development of liver fibrosis, inflammation, and regeneration after a massive injury and in the setting of liver transplantation. The involvement of the RAGE-ligand axis in vascular disease, diabetes, cancer, and neurodegeneration is well established, but it still needs to be clarified in the setting of liver diseases. We present a review of the recent literature on this receptor in surgical and clinical settings involving the liver, and we highlight the open issues and possible directions of future research.
Collapse
Affiliation(s)
- Giuseppina Basta
- Institute of Clinical Physiology, National Research Council, Pisa, Italy.
| | | | | | | | | | | |
Collapse
|
40
|
Weston CJ, Adams DH. Hepatic consequences of vascular adhesion protein-1 expression. J Neural Transm (Vienna) 2011; 118:1055-64. [PMID: 21512782 DOI: 10.1007/s00702-011-0647-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Accepted: 04/10/2011] [Indexed: 01/09/2023]
|
41
|
Treatment with oligonol, a low-molecular polyphenol derived from lychee fruit, attenuates diabetes-induced hepatic damage through regulation of oxidative stress and lipid metabolism. Br J Nutr 2011; 106:1013-22. [PMID: 21477406 DOI: 10.1017/s0007114511001322] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We have identified the effects of oligonol, a low-molecular polyphenol derived from lychee fruit, on oxidative stress and lipid metabolism in a type 2 diabetic model. Oligonol was orally administered at 10 or 20 mg per kg body weight per d for 8 weeks to db/db mice, and its effects were compared with those of the vehicle in db/db and m/m mice. Serum and hepatic biochemical factors, and protein and mRNA expression related to lipid metabolism were measured. In the oligonol-administered group, there were significant reductions of reactive oxygen species (ROS), lipid peroxidation, and the TAG and total cholesterol concentrations in both the serum and liver. Additionally, oligonol attenuated oxidative stress through the inhibition of advanced glycation endproduct formation and its receptor expression. Furthermore, augmented expressions of NF-κBp65 and inducible NO synthase were down-regulated to the levels of m/m mice in the group treated with oligonol at 20 mg/kg. Regarding lipid metabolism, lower hepatic lipid resulted from the down-regulation of sterol regulatory element-binding protein-1 and its target gene of lipogenic enzymes in the liver of db/db mice. The present results suggest that oligonol has protective effects against ROS-related inflammation and excess lipid deposition in the type 2 diabetic liver.
Collapse
|