451
|
Dangana EO, Michael OS, Omolekulo TE, Areola ED, Olatunji LA. Enhanced hepatic glycogen synthesis and suppressed adenosine deaminase activity by lithium attenuates hepatic triglyceride accumulation in nicotine-exposed rats. Biomed Pharmacother 2018; 109:1417-1427. [PMID: 30551393 DOI: 10.1016/j.biopha.2018.10.067] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/05/2018] [Accepted: 10/12/2018] [Indexed: 01/08/2023] Open
Abstract
Reduced liver glycogen synthesis might signify increased glucose flux towards fat synthesis and triggers hepatic triglyceride accumulation and dysmetabolism. Adenosine deaminase (ADA) reduces adenosine content which increases glycogenolysis. In the present study, we evaluate the effect of modulating glycogen synthesis and ADA by lithium chloride (LiCl) on nicotine-induced dysmetabolism. Twenty four male Wistar rats (n = 6/group) were allotted into four groups namely; vehicle-treated (po), nicotine-treated (1.0 mg/kg; po), LiCl-treated (5.0 mg/kg; po) and nicotine + LiCl-treated groups. The treatments lasted for 8 weeks. Nicotine exposure resulted in reduced body weight gain, liver weight, visceral adiposity, glycogen content and synthase. Along with increased insulin resistance (IR), fasting plasma glucose, lactate, plasma and hepatic ADA, XO, UA, and triglyceride (TG), total cholesterol (TC), free fatty acid, lipid peroxidation and liver injury markers. However, plasma and hepatic glucose-6-phosphate dehydrogenase-dependent antioxidant defenses were not affected by nicotine exposure. Concurrent treatment with LiCl normalizes all alterations with exception of hepatic TC. This result shows that enhancement of hepatic glycogen synthesis and suppression of ADA/XO/uric acid pathway by lithium can salvage the liver from nicotine-induced TG accumulation.
Collapse
Affiliation(s)
- Elizabeth O Dangana
- HOPE Cardiometabolic Research Team, Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Olugbenga S Michael
- HOPE Cardiometabolic Research Team, Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria; Cardiometabolic Research Unit, Department of Physiology, College of Health Sciences, Bowen University Iwo, Nigeria
| | - Tolulope E Omolekulo
- HOPE Cardiometabolic Research Team, Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Emmanuel D Areola
- HOPE Cardiometabolic Research Team, Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Lawrence A Olatunji
- HOPE Cardiometabolic Research Team, Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria.
| |
Collapse
|
452
|
Niture S, Gyamfi MA, Kedir H, Arthur E, Ressom H, Deep G, Kumar D. Serotonin induced hepatic steatosis is associated with modulation of autophagy and notch signaling pathway. Cell Commun Signal 2018; 16:78. [PMID: 30409162 PMCID: PMC6225666 DOI: 10.1186/s12964-018-0282-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/09/2018] [Indexed: 01/08/2023] Open
Abstract
Background Besides its neurotransmitter and vasoconstriction functions, serotonin is an important mediator of numerous biological processes in peripheral tissues including cell proliferation, steatosis, and fibrogenesis. Recent reports indicate that serotonin may promote tumor growth in liver cancer, however, the molecular mechanisms remain elusive. n this study, we investigated the role and molecular signaling mechanisms mediated by serotonin in liver cancer cell survival, drug resistance, and steatosis. Methods Effect of serotonin on modulation of cell survival/proliferation was determined by MTT/WST1 assay. Effect of serotonin on the regulation of autophagy biomarkers and lipid/fatty acid proteins expression, AKT/mTOR and Notch signaling was evaluated by immunoblotting. The role of serotonin in normal human hepatocytes and liver cancer cell steatosis was analyzed by Oil Red O staining. The mRNA expression levels of lipid/fatty acid proteins and serotonin receptors were validated by qRT-PCR. The important roles of autophagy, Notch signaling, serotonin receptors and serotonin re-uptake proteins on serotonin-mediated cell steatosis were investigated by using selective inhibitors or antagonists. The association of peripheral serotonin, autophagy, and hepatic steatosis was also investigated using chronic EtOH fed mouse model. Results Exposure of liver cancer cells to serotonin induced Notch signaling and autophagy, independent of AKT/mTOR pathway. Also, serotonin enhanced cancer cell proliferation/survival and drug resistance. Furthermore, serotonin treatment up-regulated the expression of lipogenic proteins and increased steatosis in liver cancer cells. Inhibition of autophagy or Notch signaling reduced serotonin-mediated cell steatosis. Treatment with serotonin receptor antagonists 5-HTr1B and 5-HTr2B reduced serotonin-mediated cell steatosis; in contrast, treatment with selective serotonin reuptake inhibitors (SSRIs) increased steatosis. In addition, mice fed with chronic EtOH resulted in increased serum serotonin levels which were associated with the induction of hepatic steatosis and autophagy. Conclusions Serotonin regulates liver cancer cell steatosis, cells survival, and may promote liver carcinogenesis by activation of Notch signaling and autophagy. Electronic supplementary material The online version of this article (10.1186/s12964-018-0282-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Suryakant Niture
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 1801 Fayetteville St, Durham, NC, 27707, USA
| | - Maxwell A Gyamfi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 1801 Fayetteville St, Durham, NC, 27707, USA
| | - Habib Kedir
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 1801 Fayetteville St, Durham, NC, 27707, USA
| | - Elena Arthur
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 1801 Fayetteville St, Durham, NC, 27707, USA
| | - Habtom Ressom
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 20008, USA
| | - Gagan Deep
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, 27109, USA
| | - Deepak Kumar
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 1801 Fayetteville St, Durham, NC, 27707, USA. .,Department of Pharmaceutical Sciences, North Carolina Central University, Durham, NC, 27707, USA. .,Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 20008, USA.
| |
Collapse
|
453
|
Mboma J, Leblanc N, Wan S, Jacobs RL, Tchernof A, Dubé P, Angers P, Jacques H. Liver and plasma lipid changes induced by cyclic fatty acid monomers from heated vegetable oil in the rat. Food Sci Nutr 2018; 6:2092-2103. [PMID: 30510710 PMCID: PMC6261175 DOI: 10.1002/fsn3.766] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 07/11/2018] [Accepted: 07/14/2018] [Indexed: 12/28/2022] Open
Abstract
Cyclic fatty acid monomers (CFAM) generated through domestic or industrial heating of vegetable oils may alter liver enzymes and induce hepatomegaly and steatosis, but the underlying mechanisms are not clearly understood. This study aimed to assess the effects of CFAM on liver and plasma lipids and to determine whether these effects are modulated by dietary lipids. Thirty-six (36) male Wistar rats were fed either of the four isoenergetic diets consisting of canola oil or soybean oil with/without 500 mg/100 g CFAM of total fat for 28 days. Rats fed CFAM had higher liver total lipids (p = 0.03) and triacylglycerols (TAG) (p = 0.02), but less hepatic phosphatidylcholine (p = 0.02) compared to those fed the non-CFAM diets. CFAM did not alter liver phosphatidylethanolamine N-methyltransferase (PEMT) activity and CTP: phosphocholine cytidylyltransferase (CT-α) protein levels. Rats fed CFAM diets had higher levels of plasma total cholesterol (TC), VLDL + LDL cholesterol, higher ratio of TC to HDL cholesterol, and lower levels of HDL cholesterol compared with rats fed non-CFAM diets (p < 0.05). Plasma alanine transaminase (ALT) was decreased with CFAM, but plasma insulin, glucose, and TAG did not vary among the four diet groups (p < 0.05). Rats fed canola oil and CFAM had higher plasma levels of aspartate transaminase (AST) and AST/ALT ratio compared with the other three diet groups. These results indicate that CFAM may provoke an accumulation of TAG in the liver related to a decrease in phosphatidylcholine (PC) levels, but the effect of CFAM on PC concentrations may not occur through impairment of the two main PC biosynthesis pathways.
Collapse
Affiliation(s)
- Jean Mboma
- School of NutritionLaval UniversityQuebec CityQuebecCanada
| | - Nadine Leblanc
- School of NutritionLaval UniversityQuebec CityQuebecCanada
- Institute of Nutrition and Functional FoodsLaval UniversityQuebec CityQuebecCanada
| | - Sereana Wan
- Department of BiochemistryUniversity of AlbertaEdmontonAlbertaCanada
| | - René L. Jacobs
- Department of BiochemistryUniversity of AlbertaEdmontonAlbertaCanada
- Department of Agricultural, Food and Nutritional Science4‐002 Li Ka Shing Centre for Health Research InnovationsUniversity of AlbertaEdmontonAlbertaCanada
| | - André Tchernof
- School of NutritionLaval UniversityQuebec CityQuebecCanada
- Institute of Nutrition and Functional FoodsLaval UniversityQuebec CityQuebecCanada
- Quebec Heart and Lung InstituteQuebec CityQuebecCanada
| | - Pascal Dubé
- Institute of Nutrition and Functional FoodsLaval UniversityQuebec CityQuebecCanada
| | - Paul Angers
- Institute of Nutrition and Functional FoodsLaval UniversityQuebec CityQuebecCanada
- Department of Food ScienceLaval UniversityQuebec CityQuebecCanada
| | - Hélène Jacques
- School of NutritionLaval UniversityQuebec CityQuebecCanada
- Institute of Nutrition and Functional FoodsLaval UniversityQuebec CityQuebecCanada
| |
Collapse
|
454
|
Sadeghpour A, Parada ML, Vieira J, Povey M, Rappolt M. Global Small-Angle X-ray Scattering Data Analysis of Triacylglycerols in the Molten State (Part I). J Phys Chem B 2018; 122:10320-10329. [PMID: 30351127 DOI: 10.1021/acs.jpcb.8b06704] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The study of triacylglycerols (TAGs) in their molten state is of fundamental importance for a deeper understanding of the TAG crystallization processes, being highly relevant for both manufacturing and medical applications. Although different models have been proposed to explain the nanostructured nature of the fluid state of TAGs, none of them are fully satisfactory. In this paper, we propose a new model consisting of positionally uncorrelated lamellar TAG assemblies embedded in an isotropic medium that assist as prenucleating structures. This model was validated by applying a novel global fitting method, resulting in an excellent agreement with the small-angle X-ray scattering data. A deeper analysis of the scattering patterns at different temperatures, both in cooling and heating directions, allowed us further to detect the crystalline traces of TAGs even after heating to 40 °C and record, on cooling, the onset of crystallization at 30-25 °C. The application of the presented novel model not only explains the outstandingly structured fluid of molten TAGs, but also lays the basis for analyzing first the crystallization steps in greater detail, which is outlined in our follow-up paper "Global Small-Angle X-ray Scattering Data Analysis of Triacylglycerols in the α-Phase (Part II)".
Collapse
Affiliation(s)
- Amin Sadeghpour
- School of Food Science and Nutrition , University of Leeds , Leeds LS2 9JT , U.K.,Department of Materials Meet Life, Empa , Swiss Federal Laboratories for Materials Science and Technology , 8600 St. Gallen , Switzerland
| | - Marjorie Ladd Parada
- School of Food Science and Nutrition , University of Leeds , Leeds LS2 9JT , U.K
| | | | - Megan Povey
- School of Food Science and Nutrition , University of Leeds , Leeds LS2 9JT , U.K
| | - Michael Rappolt
- School of Food Science and Nutrition , University of Leeds , Leeds LS2 9JT , U.K
| |
Collapse
|
455
|
Hoang NA, Richter F, Schubert M, Lorkowski S, Klotz LO, Steinbrenner H. Differential capability of metabolic substrates to promote hepatocellular lipid accumulation. Eur J Nutr 2018; 58:3023-3034. [PMID: 30368556 DOI: 10.1007/s00394-018-1847-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023]
Abstract
PURPOSE Excessive storage of triacylglycerides (TAGs) in lipid droplets within hepatocytes is a hallmark of non-alcoholic fatty liver disease (NAFLD), one of the most widespread metabolic disorders in Western societies. For the purpose of exploring molecular pathways in NAFLD development and testing potential drug candidates, well-characterised experimental models of ectopic TAG storage in hepatocytes are needed. METHODS Using an optimised Oil Red O assay, immunoblotting and real-time qRT-PCR, we compared the capability of dietary monosaccharides and fatty acids to promote lipid accumulation in HepG2 human hepatoma cells. RESULTS Both high glucose and high fructose resulted in intracellular lipid accumulation after 48 h, and this was further augmented (up to twofold, as compared to basal levels) by co-treatment with the lipogenesis-stimulating hormone insulin and the pro-inflammatory cytokine tumour necrosis factor alpha (TNF-α), respectively. The fatty acids palmitic and oleic acid were even more effective than these carbohydrates, inducing significantly elevated TAG storage already after 24 h of treatment. Highest (about threefold) increases in lipid accumulation were observed upon treatment with oleic acid, alone as well as in combinations with palmitic acid or with high glucose and insulin. Increases in protein levels of a major lipid droplet coat protein, perilipin-2 (PLIN2), mirrored intracellular lipid accumulation following different treatment regimens. CONCLUSIONS Several treatment regimens of excessive fat and sugar supply promoted lipid accumulation in HepG2 cells, albeit with differences in the extent and rapidity of steatogenesis. PLIN2 is a candidate molecular marker of sustained lipid accumulation in HepG2 cells.
Collapse
Affiliation(s)
- Ngoc Anh Hoang
- Institute of Nutritional Sciences, Nutrigenomics, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - Friederike Richter
- Institute of Nutritional Sciences, Nutrigenomics, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - Martin Schubert
- Department of Nutritional Biochemistry and Physiology, Institute of Nutritional Sciences, Friedrich-Schiller-Universität Jena, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Jena, Germany
| | - Stefan Lorkowski
- Department of Nutritional Biochemistry and Physiology, Institute of Nutritional Sciences, Friedrich-Schiller-Universität Jena, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Jena, Germany
| | - Lars-Oliver Klotz
- Institute of Nutritional Sciences, Nutrigenomics, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - Holger Steinbrenner
- Institute of Nutritional Sciences, Nutrigenomics, Friedrich-Schiller-Universität Jena, Jena, Germany.
| |
Collapse
|
456
|
Pei E, Liu Y, Jiang W, Lin S, Huang L, Lin M, Cai L. Sleeve gastrectomy attenuates high fat diet-induced non-alcoholic fatty liver disease. Lipids Health Dis 2018; 17:243. [PMID: 30355361 PMCID: PMC6201512 DOI: 10.1186/s12944-018-0875-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/26/2018] [Indexed: 02/08/2023] Open
Abstract
Background A high-fat diet (HFD) is known to lead to obesity, and contributes to the progression of non-alcoholic fatty liver disease. The objective of this study was to evaluate the effects of sleeve gastrectomy (SG) on the progression of HFD-induced hepatic steatosis. Methods Fifteen 4-week-old, male Wistar rats were randomly assigned into three groups: NC, HFD + SHAM and HFD + SG. Their body weight, glucose-lipid metabolism, inflammation indices, hepatic steatosis and fibroblast growth factor 21 (FGF21) levels were measured. Results Postoperatively, body weights in the HFD + SHAM and HFD + SG group rats decreased during the first week. Thereafter, HFD + SG rats regained their body weight. Differences in insulin, homeostasis model assessment of insulin resistance, triglyceride, free fatty acid, tumor necrosis factor-α and monocyte chemotactic protein-1 levels were statistically significant across the three groups (all P < 0.05). Interestingly, FGF21 levels in the HFD + SG group were markedly lower than in the HFD + SHAM group (P = 0.015), however, there were no differences in the NC group. Hematoxylin and eosin staining demonstrated that more vacuoles were present in the HFD + SHAM liver when compared to the HFD + SG liver. Oil-red O staining showed less red dots in the HFD + SG liver. Conclusions Despite eating, surgical re-routing of the gut may prevent weight accumulation, regulate glucose-lipid metabolism and insulin sensitivity, control a chronic inflammatory state, change the secretion pattern of FGF21 and alleviate the severity of fatty liver.
Collapse
Affiliation(s)
- Erli Pei
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Yang Liu
- Department of Gerontology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Weiqing Jiang
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Songruo Lin
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Lei Huang
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Moubin Lin
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| | - Li Cai
- Department of Science and Research, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| |
Collapse
|
457
|
Xu M, Wu H, Li M, Wen Y, Yu C, Xia L, Xia Q, Kong X. DJ-1 Deficiency Protects Hepatic Steatosis by Enhancing Fatty Acid Oxidation in Mice. Int J Biol Sci 2018; 14:1892-1900. [PMID: 30443192 PMCID: PMC6231226 DOI: 10.7150/ijbs.28620] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 09/07/2018] [Indexed: 01/16/2023] Open
Abstract
Our previous studies have shown that DJ-1 play important roles in progression of liver diseases through modulating hepatic ROS production and immune response, but its role in hepatic steatosis remains obscure. In the present study, by adopting a high-fat-diet (HFD) induced mice model, we found that DJ-1 knockout (DJ-1-/-) mice showing decreased HFD-induced obesity and visceral adipose accumulation. In line with these changes, there were also reduced liver weight and ameliorated hepatic triglyceride (TG) accumulation in DJ-1-/- mice compared to wild-type (WT) mice. And there were also decreased blood glucose levels and insulin resistance and reduced glucose metabolic disorder in DJ-1-/- mice, whereas there were no significant differences in total cholesterol (TC) and serum lipid in two groups of mice. Mechanistically, we found that there were no differences in food intake in these two genotypes of mice. Furthermore, there were no significant differences in fatty acid synthesis and glycolysis, but the expression of key enzymes in fatty acid oxidation and the tricarboxylic acid (TCA) cycle, such as Cpt1α, Pparα, Acox1, Cs, Idh1 and Idh2, was increased in DJ-1-/- mice liver, suggesting that there was enhanced fatty acids oxidation and TCA cycle in DJ-1-/- mice. Our data indicate that deletion of DJ-1 enhancing fatty acids oxidation resulting in lower hepatic TG accumulation in mice, which protecting mice hepatic steatosis.
Collapse
Affiliation(s)
- Min Xu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hailong Wu
- Shanghai Key Laboratory for Molecular Imaging, Collaborative Research Center, Shanghai University of Medicine & Health Science, Shanghai, China
| | - Meng Li
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yankai Wen
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chang Yu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoni Kong
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
458
|
Molecular profiling of nonalcoholic fatty liver disease-associated hepatocellular carcinoma using SB transposon mutagenesis. Proc Natl Acad Sci U S A 2018; 115:E10417-E10426. [PMID: 30327349 PMCID: PMC6217425 DOI: 10.1073/pnas.1808968115] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the fastest rising cause of hepatocellular carcinoma (HCC) in Western countries; however, the molecular mechanisms driving NAFLD-HCC remain elusive. Using Sleeping Beauty transposon mutagenesis in two mouse models of NAFLD-HCC, we identified hundreds of NAFLD-HCC candidate cancer genes that were enriched in pathways often associated with NAFLD and HCC. We also showed that Sav1, which functions in the Hippo signaling pathway and was the most frequently mutated gene identified by SB in both screens, prevents progression of steatohepatitis and subsequent HCC development in coordination with PI3K signaling via suppression of Yap, a downstream effector of the Hippo pathway. Our forward genetic screens have thus identified pathways and genes driving the development of NAFLD-HCC. Nonalcoholic fatty liver disease (NAFLD) is the fastest rising cause of hepatocellular carcinoma (HCC) in Western countries; however, the molecular mechanisms that cause NAFLD-HCC remain elusive. To identify molecular drivers of NAFLD-HCC, we performed Sleeping Beauty (SB) transposon mutagenesis screens in liver-specific Pten knockout and in high-fat diet-fed mice, which are murine models of NAFLD-HCC. SB mutagenesis accelerated liver tumor formation in both models and identified 588 and 376 candidate cancer genes (CCGs), respectively; 257 CCGs were common to both screens and were enriched in signaling pathways known to be important for human HCC. Comparison of these CCGs with those identified in a previous SB screen of hepatitis B virus-induced HCC identified a core set of 141 CCGs that were mutated in all screens. Forty-one CCGs appeared specific for NAFLD-HCC, including Sav1, a component of the Hippo signaling pathway and the most frequently mutated gene identified in both NAFLD-HCC screens. Liver-specific deletion of Sav1 was found to promote hepatic lipid accumulation, apoptosis, and fibrogenesis, leading to the acceleration of hepatocarcinogenesis in liver-specific Pten mutant mice. Sav1/Pten double-mutant livers also showed a striking up-regulation of markers of liver progenitor cells (LPCs), along with synergistic activation of Yap, which is a major downstream effector of Hippo signaling. Lastly, Yap activation, in combination with Pten inactivation, was found to accelerate cell growth and sphere formation of LPCs in vitro and induce their malignant transformation in allografts. Our forward genetic screens in mice have thus identified pathways and genes driving the development of NAFLD-HCC.
Collapse
|
459
|
Horst AK, Najjar SM, Wagener C, Tiegs G. CEACAM1 in Liver Injury, Metabolic and Immune Regulation. Int J Mol Sci 2018; 19:ijms19103110. [PMID: 30314283 PMCID: PMC6213298 DOI: 10.3390/ijms19103110] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/02/2018] [Accepted: 10/04/2018] [Indexed: 02/06/2023] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is a transmembrane glycoprotein that is expressed on epithelial, endothelial and immune cells. CEACAM1 is a differentiation antigen involved in the maintenance of epithelial polarity that is induced during hepatocyte differentiation and liver regeneration. CEACAM1 regulates insulin sensitivity by promoting hepatic insulin clearance, and controls liver tolerance and mucosal immunity. Obese insulin-resistant humans with non-alcoholic fatty liver disease manifest loss of hepatic CEACAM1. In mice, deletion or functional inactivation of CEACAM1 impairs insulin clearance and compromises metabolic homeostasis which initiates the development of obesity and hepatic steatosis and fibrosis with other features of non-alcoholic steatohepatitis, and adipogenesis in white adipose depot. This is followed by inflammation and endothelial and cardiovascular dysfunctions. In obstructive and inflammatory liver diseases, soluble CEACAM1 is shed into human bile where it can serve as an indicator of liver disease. On immune cells, CEACAM1 acts as an immune checkpoint regulator, and deletion of Ceacam1 gene in mice causes exacerbation of inflammation and hyperactivation of myeloid cells and lymphocytes. Hence, hepatic CEACAM1 resides at the central hub of immune and metabolic homeostasis in both humans and mice. This review focuses on the regulatory role of CEACAM1 in liver and biliary tract architecture in health and disease, and on its metabolic role and function as an immune checkpoint regulator of hepatic inflammation.
Collapse
Affiliation(s)
- Andrea Kristina Horst
- Institute of Experimental Immunology and Hepatology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany.
| | - Sonia M Najjar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Irvine Hall, 1 Ohio University, Athens, OH 45701-2979, USA.
- The Diabetes Institute, Heritage College of Osteopathic Medicine, Irvine Hall, 1 Ohio University, Athens, OH 45701-2979, USA.
| | - Christoph Wagener
- University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany.
| | - Gisa Tiegs
- Institute of Experimental Immunology and Hepatology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany.
| |
Collapse
|
460
|
McCommis KS, Finck BN. Treating Hepatic Steatosis and Fibrosis by Modulating Mitochondrial Pyruvate Metabolism. Cell Mol Gastroenterol Hepatol 2018; 7:275-284. [PMID: 30686780 PMCID: PMC6352854 DOI: 10.1016/j.jcmgh.2018.09.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 12/12/2022]
Abstract
A hepatic comorbidity of metabolic syndrome, known as nonalcoholic fatty liver disease (NAFLD), is increasing in prevalence in conjunction with the pandemics of obesity and diabetes. The spectrum of NAFLD ranges from simple hepatic fat accumulation to a more severe disease termed nonalcoholic steatohepatitis (NASH), involving inflammation, hepatocyte death, and fibrosis. Importantly, NASH is linked to a much higher risk of cirrhosis, liver failure, and hepatocellular carcinoma, as well as an increased risk for nonhepatic malignancies and cardiovascular disease. Interest in the understanding of the disease processes and search for treatments for the spectrum of NAFLD-NASH has increased exponentially, but there are no approved pharmacologic therapies. In this review, we discuss the existing literature supporting insulin-sensitizing thiazolidinedione compounds as potential drug candidates for the treatment of NASH. In addition, we put these results into new context by summarizing recent studies suggesting these compounds alter mitochondrial metabolism by binding and inhibiting the mitochondrial pyruvate carrier.
Collapse
Affiliation(s)
| | - Brian N. Finck
- Correspondence Address correspondence to: Brian N. Finck, 660 South Euclid Avenue, Campus Box 8031, St. Louis, Missouri 63110. fax: (314) 362-8230.
| |
Collapse
|
461
|
Fc Gamma Receptor IIb Expressed in Hepatocytes Promotes Lipid Accumulation and Gluconeogenesis. Int J Mol Sci 2018; 19:ijms19102932. [PMID: 30261661 PMCID: PMC6213401 DOI: 10.3390/ijms19102932] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/03/2018] [Accepted: 09/08/2018] [Indexed: 01/01/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is characterized by ectopic lipid accumulation in the liver, usually combined with hepatic insulin resistance. Fc-gamma receptor-IIb (FcγRIIb) and its ligand are reported to be associated with obesity and type 2 diabetes mellitus (T2DM). As knowledge about FcγRIIb in the literature is mostly generated from studies on skeletal muscle tissue, the expression and function of FcγRIIb in the liver and hepatocytes are largely unknown. In this study, we identified the expression of FcγRIIb in primary cultured mouse hepatocytes: FcγRIIb was upregulated in response to oleic acid (OA) in a dose dependent manner. FcγRIIb knockdown using shRNA suppressed the lipid and triglyceride accumulation, and mRNA expression of ACC1, FASn, CD36, MTTP, and ApoB in OA-treated HepG2 cells. FcγRIIb deficiency mice fed with high fat diet (HFD) had significantly lower liver weight and liver to body weight ratio, as well as less triglyceride accumulation in the livers. In glycometabolism, FcγRIIb hindered insulin-induced phosphorylation of AKT and FOXO1, and in turn upregulated G6Pase and PEPCK mRNA expression, suggesting that FcγRIIb promotes gluconeogenesis by suppressing the AKT/FOXO1/G6Pase/PEPCK pathway in hepatocytes. This study reveals a novel role for FcγRIIb in regulating lipid metabolism and glycometabolism, and provides a new therapeutic target to improve NAFLD.
Collapse
|
462
|
Effect of silibinin on CFLAR-JNK pathway in oleic acid-treated HepG2 cells. Biomed Pharmacother 2018; 108:716-723. [PMID: 30248539 DOI: 10.1016/j.biopha.2018.09.089] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 09/15/2018] [Accepted: 09/16/2018] [Indexed: 02/08/2023] Open
Abstract
AIMS Silibinin is a flavonolignan from milk thistle with many pharmacological activities including lipid-lowering and antioxidant. Caspase 8 and Fas-associated protein with death domain-like apoptosis regulator (CFLAR) is an important target gene in regulating non-alcoholic steatohepatitis (NASH). At present, the effect of silibinin on CFLAR-JNK pathway related to NASH was unknown. Here the effect of silibinin on CFLAR-JNK pathway and its downstream target genes involved in lipid metabolism, glucose uptake, oxidative stress and inflammatory response were studied in oleic acid (OA)-treated HepG2 cells. MAIN METHODS OA-treated HepG2 cells were employed as a in vitro model of steatosis, insulin resistance and oxidative stress. The model cells were then treated by silibinin (5, 20, 50, and 100 μM) for 24 h and detected for the related indicators as follows: (1) cellular triglycerides (TG), nitric oxide (NO) and glucose uptake; (2) the mRNA levels of the sterol regulatory element binding protein-1C (SREBP-1C), patatin-like phospholipase domain containing 3 (PNPLA3) and peroxisome proliferator activated receptor-α (PPARα); (3) the protein levels of PPARα, SREBP-1C, PNPLA3, CFLAR, phosphorylated c-Jun N-terminal kinase (pJNK), phosphatidylinositol 3-kinase (PI3K), phosphorylated serine-threonine protein kinase (pAKT), nuclear factor E2-related factor 2 (NRF2), cytochrome P450 2E1 (CYP2E1) and 4A (CYP4A). KEY FINDINGS Compared to the control, OA-treatment led to a result as follows: (1) increased the intracellular levels of TG and NO; (2) up-regulated the protein expression of SREBP-1C, PNPLA3, pJNK, CYP 2E1 and CYP 4A; (3) decreased the uptake of 2-NBDG; (4) down-regulated the protein expression of CFLAR, PPARα, PI3K, pAKT and NRF2. Compared to OA-treated HepG2 cells, silibinin treatment could improve the indicators as follows: (1) decreased the intracellular levels of TG and NO; (2) down-regulated the protein expression of SREBP-1C, PNPLA3, pJNK, CYP 2E1 and CYP 4A; (3) increased the uptake of 2-NBDG; (4) up-regulated the protein expression of CFLAR, PPARα, PI3K, pAKT and NRF2. SIGNIFICANCE Silibinin can ameliorate some metabolic alterations and induce some molecular changes by activating the CFLAR-JNK pathway and thereby regulating its downstream target genes involved in lipid metabolism (PPARα, SREBP-1C and PNPLA3), glucose uptake (PI3K-AKT), oxidative stress (NRF2, CYP2E1, CYP4A) and inflammatory response(NO) in OA-treated HepG2 cells demonstrating its possible use in ameliorating various symptoms of NASH.
Collapse
|
463
|
Fan R, Cui J, Ren F, Wang Q, Huang Y, Zhao B, Wei L, Qian X, Xiong X. Overexpression of NRK1 ameliorates diet- and age-induced hepatic steatosis and insulin resistance. Biochem Biophys Res Commun 2018; 500:476-483. [PMID: 29678570 DOI: 10.1016/j.bbrc.2018.04.107] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 04/13/2018] [Indexed: 01/18/2023]
Abstract
NAD+ is a co-enzyme in redox reactions and a substrate required for activity of various enzyme families, including sirtuins and poly(ADP-ribose) polymerases. Dietary supplementation of NAD+ precursors nicotinamide mononucleotide (NMN) or nicotinamide riboside (NR) protects against metabolic disease, neurodegenerative disorders and age-related physiological decline in mammals. Here we sought to identify the roles of nicotinamide riboside kinase 1 (NRK1) plays in regulating hepatic NAD+ biosynthesis and lipid metabolism. Using adenovirus mediated gene transduction to overexpress or knockdown NRK1 in mouse liver, we have demonstrated that NRK1 is critical for maintaining hepatic NAD+ levels and triglyceride content. We have further shown that the hepatic expression of Nmrk1 mRNA is significantly decreased either in mice treated with high-fat diet or in aged mice. However, adenoviral delivery of NRK1 in these diet- and age-induced mice elevates hepatic NAD+ levels, reduces hepatic steatosis, and improves glucose tolerance and insulin sensitivity. Our results provide important insights in targeting NRK1 for treating hepatic steatosis.
Collapse
Affiliation(s)
- Rui Fan
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jing Cui
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
| | - Feng Ren
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
| | - Qingzhi Wang
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Laboratory of Molecular Metabolism, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yanmei Huang
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Bin Zhao
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Laboratory of Molecular Metabolism, Xinxiang Medical University, Xinxiang, Henan, China
| | - Lai Wei
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Laboratory of Molecular Metabolism, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xinlai Qian
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
| | - Xiwen Xiong
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Laboratory of Molecular Metabolism, Xinxiang Medical University, Xinxiang, Henan, China.
| |
Collapse
|
464
|
Trouwborst I, Bowser SM, Goossens GH, Blaak EE. Ectopic Fat Accumulation in Distinct Insulin Resistant Phenotypes; Targets for Personalized Nutritional Interventions. Front Nutr 2018; 5:77. [PMID: 30234122 PMCID: PMC6131567 DOI: 10.3389/fnut.2018.00077] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 08/15/2018] [Indexed: 12/13/2022] Open
Abstract
Cardiometabolic diseases are one of the leading causes for disability and mortality in the Western world. The prevalence of these chronic diseases is expected to rise even further in the next decades. Insulin resistance (IR) and related metabolic disturbances are linked to ectopic fat deposition, which is the storage of excess lipids in metabolic organs such as liver and muscle. Notably, a vicious circle exists between IR and ectopic fat, together increasing the risk for the development of cardiometabolic diseases. Nutrition is a key-determining factor for both IR and ectopic fat deposition. The macronutrient composition of the diet may impact metabolic processes related to ectopic fat accumulation and IR. Interestingly, however, the metabolic phenotype of an individual may determine the response to a certain diet. Therefore, population-based nutritional interventions may not always lead to the most optimal (cardiometabolic) outcomes at the individual level, and differences in the metabolic phenotype may underlie conflicting findings related to IR and ectopic fat in dietary intervention studies. Detailed metabolic phenotyping will help to better understand the complex relationship between diet and metabolic regulation, and to optimize intervention outcomes. A subgroup-based approach that integrates, among others, tissue-specific IR, cardiometabolic parameters, anthropometrics, gut microbiota, age, sex, ethnicity, and psychological factors may thereby increase the efficacy of dietary interventions. Nevertheless, the implementation of more personalized nutrition may be complex, costly, and time consuming. Future studies are urgently warranted to obtain insight into a more personalized approach to nutritional interventions, taking into account the metabolic phenotype to ultimately improve insulin sensitivity and reduce the risk for cardiometabolic diseases.
Collapse
Affiliation(s)
- Inez Trouwborst
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Suzanne M Bowser
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| |
Collapse
|
465
|
Ipsen DH, Lykkesfeldt J, Tveden-Nyborg P. Molecular mechanisms of hepatic lipid accumulation in non-alcoholic fatty liver disease. Cell Mol Life Sci 2018; 75:3313-3327. [PMID: 29936596 PMCID: PMC6105174 DOI: 10.1007/s00018-018-2860-6] [Citation(s) in RCA: 905] [Impact Index Per Article: 129.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/18/2018] [Accepted: 06/20/2018] [Indexed: 12/17/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is currently the world's most common liver disease, estimated to affect up to one-fourth of the population. Hallmarked by hepatic steatosis, NAFLD is associated with a multitude of detrimental effects and increased mortality. This narrative review investigates the molecular mechanisms of hepatic steatosis in NAFLD, focusing on the four major pathways contributing to lipid homeostasis in the liver. Hepatic steatosis is a consequence of lipid acquisition exceeding lipid disposal, i.e., the uptake of fatty acids and de novo lipogenesis surpassing fatty acid oxidation and export. In NAFLD, hepatic uptake and de novo lipogenesis are increased, while a compensatory enhancement of fatty acid oxidation is insufficient in normalizing lipid levels and may even promote cellular damage and disease progression by inducing oxidative stress, especially with compromised mitochondrial function and increased oxidation in peroxisomes and cytochromes. While lipid export initially increases, it plateaus and may even decrease with disease progression, sustaining the accumulation of lipids. Fueled by lipo-apoptosis, hepatic steatosis leads to systemic metabolic disarray that adversely affects multiple organs, placing abnormal lipid metabolism associated with NAFLD in close relation to many of the current life-style-related diseases.
Collapse
Affiliation(s)
- David Højland Ipsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870, Frederiksberg C, Denmark
| | - Jens Lykkesfeldt
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870, Frederiksberg C, Denmark
| | - Pernille Tveden-Nyborg
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870, Frederiksberg C, Denmark.
| |
Collapse
|
466
|
Molecular Pathogenesis of Nonalcoholic Steatohepatitis- (NASH-) Related Hepatocellular Carcinoma. Can J Gastroenterol Hepatol 2018; 2018:8543763. [PMID: 30228976 PMCID: PMC6136489 DOI: 10.1155/2018/8543763] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/12/2018] [Indexed: 12/14/2022] Open
Abstract
The proportion of obese or diabetic population has been anticipated to increase in the upcoming decades, which rises the prevalence of nonalcoholic fatty liver disease (NAFLD) and its progression to nonalcoholic steatohepatitis (NASH). Recent evidence indicates that NASH is the main cause of chronic liver diseases and it is an important risk factor for development of hepatocellular carcinoma (HCC). Although the literature addressing NASH-HCC is growing rapidly, limited data is available about the etiology of NASH-related HCC. Experimental studies on the molecular mechanism of HCC development in NASH reveal that the carcinogenesis is relevant to complex changes in signaling pathways that mediate cell proliferation and energy metabolism. Genetic or epigenetic modifications and alterations in metabolic, immunologic, and endocrine pathways have been shown to be closely related to inflammation, liver injury, and fibrosis in NASH along with its subsequent progression to HCC. In this review, we provide an overview on the current knowledge of NASH-related HCC development and emphasize molecular signaling pathways regarding their mechanism of action in NASH-derived HCC.
Collapse
|
467
|
Guo WW, Wang X, Chen XQ, Ba YY, Zhang N, Xu RR, Zhao WW, Wu X. Flavonones from Penthorum chinense Ameliorate Hepatic Steatosis by Activating the SIRT1/AMPK Pathway in HepG2 Cells. Int J Mol Sci 2018; 19:ijms19092555. [PMID: 30154382 PMCID: PMC6165420 DOI: 10.3390/ijms19092555] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/04/2018] [Accepted: 08/22/2018] [Indexed: 12/13/2022] Open
Abstract
Pinocembrin-7-O-β-d-glucoside (PCBG), pinocembrin (PCB), and 5-methoxy-pinocembrin-7-O-β-d-glucoside (MPG) are three flavonones isolated from Penthorum chinense Pursh (P. chinense). The effects of the three flavonones on hepatic steatosis and their molecular mechanisms in HepG2 cells were investigated in this study for the first time. A model of hepatic steatosis in HepG2 cells was induced by free fatty acid (FFA), and co-treated with the three flavonones as mentioned. Intracellular lipid droplets were detected by Oil Red O staining. PCB, PCBG, and MPG suppressed oxidative stress by decreasing malondialdehyde (MDA) levels and increasing superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) activities. The levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) were ameliorated. Moreover, these flavonones enhanced the phosphorylation of AMP-activated protein kinase (AMPK) and the expression of silent mating type information regulation 2 homolog 1 (SIRT1) and peroxisome proliferator-activated receptor α (PPARα), and reduced the expression of sterol regulatory element binding protein-1c (SREBP1c) and the downstream targets fatty acid synthase (FAS), acetyl-CoA carboxylase (ACC), and stearoyl-CoA desaturase 1 (SCD1). Molecular docking was used to predict the interaction and combination patterns between the three flavonones and the enzymes above. The results revealed that the SIRT1/AMPK pathway is involved in the functions of the three flavonones, and the most effective flavonone against hepatic steatosis might be PCBG, followed by MPG and PCB. Therefore, the three flavonones from P. chinense were found to exert preventive effects against hepatic steatosis by regulating the SIRT1/AMPK pathway.
Collapse
Affiliation(s)
- Wei-Wei Guo
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 10 Youanmen, Xitoutiao, Beijing 100069, China.
| | - Xing Wang
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 10 Youanmen, Xitoutiao, Beijing 100069, China.
| | - Xiao-Qing Chen
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 10 Youanmen, Xitoutiao, Beijing 100069, China.
| | - Yin-Ying Ba
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 10 Youanmen, Xitoutiao, Beijing 100069, China.
| | - Nan Zhang
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 10 Youanmen, Xitoutiao, Beijing 100069, China.
| | - Rong-Rong Xu
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 10 Youanmen, Xitoutiao, Beijing 100069, China.
| | - Wen-Wen Zhao
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 10 Youanmen, Xitoutiao, Beijing 100069, China.
| | - Xia Wu
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, 10 Youanmen, Xitoutiao, Beijing 100069, China.
| |
Collapse
|
468
|
Wang D, Cong H, Wang X, Cao Y, Ikuyama S, Fan B, Gu J. Pycnogenol protects against diet-induced hepatic steatosis in apolipoprotein-E-deficient mice. Am J Physiol Endocrinol Metab 2018; 315:E218-E228. [PMID: 29462565 DOI: 10.1152/ajpendo.00009.2017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PycnogenolR (PYC), a combination of active flavonoids derived from French maritime pine bark, is a natural antioxidant that has various pharmacological activities. Here, we investigated the beneficial effect of PYC on diet-induced hepatic steatosis. Apolipoprotein E (ApoE)-deficient male mice were administered PYC at oral doses of 30 or 100 mg·kg-1·day-1 for 2 wk in advance and were then fed a high-cholesterol and -fat diet (HCD) for 8 wk. Biochemical, immunohistochemical, and gene expression analyses were conducted to explore the effect of PYC on lipid metabolism in ApoE-deficient mice on a HCD. Short-term treatment with HCD in ApoE-deficient mice induced hepatic injuries, such as lipid metabolism disorder and hepatic histopathological changes. We found that PYC reduced body weight and the increase of serum lipids that had been caused by HCD. Supplementation of PYC significantly reduced lipid deposition in the liver, as shown by the lowered hepatic lipid content and histopathological lesions. We subsequently detected genes related to lipid metabolism and inflammatory cytokines. The study showed that PYC markedly suppressed the expression of genes related to hepatic lipogenesis, fatty acid uptake, and lipid storage while increasing the lipolytic gene, which thus reduced hepatic lipid content. Furthermore, PYC mainly reduced the expression of inflammatory cytokines and the infiltration of inflammatory cells, which were resistant to the development of hepatic steatosis. These results demonstrate that PYC protects against the occurrence and development of hepatic steatosis and may provide a new prophylactic approach for nonalcoholic fatty liver disease (NAFLD).
Collapse
Affiliation(s)
- Difei Wang
- Department of Geriatric Endocrinology, The First Hospital of China Medical University , Shenyang , China
| | - Huiying Cong
- Department of Endocrinology and Metabolism, The Endocrine Institute and The Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University , Shenyang , China
| | - Xiaoli Wang
- Department of Endocrinology and Metabolism, The Endocrine Institute and The Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University , Shenyang , China
| | - Yanli Cao
- Department of Endocrinology and Metabolism, The Endocrine Institute and The Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University , Shenyang , China
| | - Shoichiro Ikuyama
- Department of Clinical Investigation, Department of Diabetes, Endocrine and Rheumatic Diseases Oita San-ai Medical Center, Ichi, Oita , Japan
| | - Bin Fan
- Department of Neurology, Shengjing Hospital, China Medical University , Shenyang , China
| | - Jianqiu Gu
- Department of Endocrinology and Metabolism, The Endocrine Institute and The Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University , Shenyang , China
| |
Collapse
|
469
|
Kwon M, Lim SJ, Joung EJ, Lee B, Oh CW, Kim HR. Meroterpenoid-rich fraction of an ethanolic extract from Sargassum serratifolium alleviates obesity and non-alcoholic fatty liver disease in high fat-fed C57BL/6J mice. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.05.063] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
470
|
Molina-Molina E, Baccetto RL, Wang DQH, de Bari O, Krawczyk M, Portincasa P. Exercising the hepatobiliary-gut axis. The impact of physical activity performance. Eur J Clin Invest 2018; 48:e12958. [PMID: 29797516 PMCID: PMC8118139 DOI: 10.1111/eci.12958] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/21/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Physical inactivity puts the populations at risk of several health problems, while regular physical activity brings beneficial effects on cardiovascular disease, mortality and other health outcomes, including obesity, glycaemic control and insulin resistance. The hepatobiliary tract is greatly involved in several metabolic aspects which include digestion and absorption of nutrients in concert with intestinal motility, bile acid secretion and flow across the enterohepatic circulation and intestinal microbiota. Several metabolic abnormalities, including nonalcoholic fatty liver as well as cholesterol cholelithiasis, represent two conditions explained by changes of the aforementioned pathways. MATERIALS AND METHODS This review defines different training modalities and discusses the effects of physical activity in two metabolic disorders, that is nonalcoholic fatty liver disease (NAFLD) and cholelithiasis. Emphasis is given to pathogenic mechanisms involving intestinal bile acids, microbiota and inflammatory status. RESULTS A full definition of physical activity includes the knowledge of aerobic and endurance exercise, metabolic equivalent tasks, duration, frequency and intensity, beneficial and harmful effects. Physical activity influences the hepatobiliary-gut axis at different levels and brings benefits to fat distribution, liver fat and gallbladder disease while interacting with bile acids as signalling molecules, intestinal microbiota and inflammatory changes in the body. CONCLUSIONS Several beneficial effects of physical activity are anticipated on metabolic disorders linking liver steatosis, gallstone disease, gut motility, enterohepatic circulation of signalling bile acids in relation to intestinal microbiota and inflammatory changes.
Collapse
Affiliation(s)
- Emilio Molina-Molina
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, Bari, Italy
| | - Raquel Lunardi Baccetto
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, Bari, Italy
| | - David Q.-H. Wang
- Department of Medicine, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ornella de Bari
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, Bari, Italy
| | - Marcin Krawczyk
- Department of Medicine II, Saarland University Medical Center, Homburg, Germany
- Laboratory of Metabolic Liver Diseases, Centre for Preclinical Research, Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, Bari, Italy
| |
Collapse
|
471
|
Observed changes in brown, white, hepatic and pancreatic fat after bariatric surgery: Evaluation with MRI. Eur Radiol 2018; 29:849-856. [PMID: 30062524 DOI: 10.1007/s00330-018-5611-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 05/29/2018] [Accepted: 06/15/2018] [Indexed: 02/06/2023]
Abstract
OBJECTIVES To study the change in brown and white adipose tissue (BAT and WAT), as well as fat content in the liver and pancreas, in patients with morbid obesity before and after bariatric surgery. METHODS Twelve patients with morbid obesity (F=8, M=4, age: 45.4 years (38.4-51.2), BMI: 35.2 kg/m2 (32.5-38.6)) underwent pre-op MRI at baseline and two post-op scans at 6-month and 12-month intervals after bariatric surgery. Co-registered water, fat, fat-fraction and T2* image series were acquired. Supraclavicular BAT and abdominal WAT were measured using in-house algorithms. Intrahepatic triglyceride (IHTG) was measured using MR spectroscopy and pancreatic fat was measured using a region-of-interest approach. Fat contents were compared between baseline and the first and second 6-month intervals using non-parametric analysis of Friedman's test and Wilcoxon's signed-rank test. Level of significance was selected at p=0.017 (0.05/3). Threshold of non-alcoholic fatty liver disease was set at 5.56%. RESULTS Results indicated that BMI (p=0.005), IHTG (p=0.005), and subcutaneous (p=0.005) and visceral adipose tissues (p=0.005) were significantly reduced 6 months after surgery. Pancreatic fat (p=0.009) was significantly reduced at 12 months. Most reduction became stable between the 6-month and 12-month interval. No significant difference was observed in BAT volume, fat-fraction and T2* values. CONCLUSION The results of this study suggest that bariatric surgery effectively reduced weight, mainly as a result of the reduction of abdominal WAT. Liver and pancreatic fat were deceased below the threshold possibly due to the reduction of free fatty acid. BAT volume, fat-fraction and T2* showed no significant changes, probably because surgery itself might not have altered the metabolic profile of the patients. KEY POINTS • No significant changes were observed in fat-fraction, T2* and volume of brown adipose tissue after bariatric surgery. • Non-alcoholic fatty liver disease was resolved after surgery. • Abdominal white fat and liver fat were significantly reduced 6 months after surgery and become stable between 6 and 12 months while pancreatic fat was significantly reduced between 0 and 12 months.
Collapse
|
472
|
Mazidi M, Katsiki N, Mikhailidis DP, Banach M. Link between plasma trans-fatty acid and fatty liver is moderated by adiposity. Int J Cardiol 2018; 272:316-322. [PMID: 30072152 DOI: 10.1016/j.ijcard.2018.07.061] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 06/16/2018] [Accepted: 07/11/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND The prevalence of non-alcoholic fatty liver disease (NAFLD) is rising. This increase may be associated with obesity. It has been suggested that trans-fatty acids (TFAs) play an important role in non-communicable diseases. AIM We examined the link between liver tests, fatty liver index (FLI) and plasma TFAs. Furthermore, we evaluated the impact of adiposity on this link. METHODS The National Health and Nutrition Examination Survey (NHANES) was used to obtain the data on TFAs and liver function biomarkers. We took account of complex NHANES data, masked variance and weighting methodology. RESULTS Of the 4252 participants, 46.4% were men. The mean age was 50.6 years overall; 51.3 years for men and 49.8 years for women (p = 0.206). In a fully adjusted model (demographic and clinical factors), FLI increased as trans-9-hexadecenoic acid and trans-11-octadecenoic acid levels increased; FLI was 38.1 and 42.3 for the first quarter (Q1) of trans-9-hexadecenoic acid and trans-11-octadecenoic acid, respectively, reaching 65.1 and 69.3 for the highest quarters (Q4) (p < 0.001 for all comparisons). Multivariable logistic regression showed for all four studied TFAs, the likelihood of NAFLD (determined by FLI) increased with increasing TFAs levels (quartiles) in a stepwise manner (p < 0.001 for all comparisons). Based on moderation analysis, a strong impact of body mass index (BMI) on the link between FLI and TFAs was observed. CONCLUSIONS Our results suggest a direct significant association between plasma TFAs, liver tests and NAFLD (assessed by FLI). Furthermore, BMI was shown to mediate this relationship. These findings highlight the importance of avoiding TFAs consumption in order to minimize cardiometabolic risk.
Collapse
Affiliation(s)
- Mohsen Mazidi
- Department of Biology and Biological Engineering, Food and Nutrition Science, Chalmers University of Technology, SE-41296 Gothenburg, Sweden.
| | - Niki Katsiki
- Second Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Hippokration Hospital, Thessaloniki, Greece
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Campus, University College London Medical School, University College London (UCL), London, UK
| | - Maciej Banach
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland; Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland
| |
Collapse
|
473
|
Abstract
Nonalcoholic fatty liver disease (NAFLD) is an important cause of chronic hepatic disease and liver transplant in Western societies. The increasing prevalence is related to dietary changes and sedentarism and follows the increasing frequency of obesity and type 2 diabetes mellitus. Growing evidence of association of NAFLD with cardiovascular diseases (CVD), independent of cardiovascular risk factors, has prompted the clarification of whether the liver is mainly a key-effector or a target-organ of the metabolic disarrangements in the metabolic syndrome. The therapeutic strategies able to alter liver disease progression and, through this, reduce the cardiovascular risk have also been tested in the last 2 decades. This review focus on the possible interactions between hepatic disease, metabolic syndrome, and CVD, and on their implications for clinical practice.
Collapse
Affiliation(s)
- Elisabete Martins
- Department of Medicine, Faculty of Medicine.,Instituto de Investigação e Inovação em Saúde (i3s), University of Porto.,Department of Cardiology
| | - Ana Oliveira
- Department of Nuclear Medicine, São João Hospital Center, Porto, Portugal
| |
Collapse
|
474
|
Liu J, Yang P, Zuo G, He S, Tan W, Zhang X, Su C, Zhao L, Wei L, Chen Y, Ruan X, Chen Y. Long-chain fatty acid activates hepatocytes through CD36 mediated oxidative stress. Lipids Health Dis 2018; 17:153. [PMID: 30016988 PMCID: PMC6050651 DOI: 10.1186/s12944-018-0790-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 05/28/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Accumulating evidence suggests that activated hepatocytes are involved in the deposition of the excess extracellular matrix during liver fibrosis via the epithelial to mesenchymal transition. Lipid accumulation in hepatocytes are implicated in the pathogenesis of chronic liver injury. CD36 is known to mediate long-chain fatty acid (LCFA) uptake and lipid metabolism. However, it is unclear whether LCFA directly promotes hepatocyte activation and the involved mechanisms have not been fully clarified. METHODS Mice were fed with a high fat diet (HFD) and normal hepatocyte cells (Chang liver cells) were treated with palmitic acid (PA) in vivo and in vitro. Real-time polymerase chain reaction (RT-PCR) and western blotting were used to examine the gene and protein expression of molecules involved in hepatic fibrogenesis and hepatocyte activation. CD36 was knocked down by transfecting CD36 siRNA into hepatocyte cells. Hydrogen peroxide (H2O2) and reactive oxygen species (ROS) levels were detected using commercial kits. RESULTS HFD induced a profibrogenic response and up-regulated CD36 expression in vivo. Analogously, PA increased lipid accumulation and induced human hepatocyte activation in vitro, which was also accompanied by increased CD36 expression. Interestingly, knockdown of CD36 resulted in a reduction of hepatocyte lipid deposition and decreased expression of Acta2 (34% decrease), Vimentin (29% decrease), Desmin (60% decrease), and TGF-β signaling pathway related genes. In addition, HFD and PA increased the production of H2O2 in vivo (48% increase) and in vitro (385% increase), and the antioxidant, NAC, ameliorated PA-induced hepatocyte activation. Furthermore, silencing of CD36 in vitro markedly attenuated PA-induced oxidative stress (H2O2: 41% decrease; ROS: 39% decrease), and the anti-activation effects of CD36 knockdown could be abolished by pretreatment with H2O2. CONCLUSIONS Our study demonstrated that LCFA facilitates hepatocyte activation by up-regulating oxidative stress through CD36, which could be an important mechanism in the development of hepatic fibrosis.
Collapse
Affiliation(s)
- Jun Liu
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China.,Department of Gastroenterology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Ping Yang
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China
| | - Guoqing Zuo
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, China.
| | - Song He
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, China
| | - Wei Tan
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoyu Zhang
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China
| | - Chunxiao Su
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China
| | - Lei Zhao
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China
| | - Li Wei
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China
| | - Yao Chen
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China
| | - Xiongzhong Ruan
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China.,John Moorhead Research Laboratory, Centre for Nephrology, University College London Medical School, Royal Free Campus, University College London, NW3 2PF, London, UK.,The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (CCID), Zhejiang University, Hangzhou, 310058, China
| | - Yaxi Chen
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
475
|
Lee J, Kim E, Kim Y, Yoo SH. Leucrose, a Sucrose Isomer, Suppresses Hepatic Fat Accumulation by Regulating Hepatic Lipogenesis and Fat Oxidation in High-fat Diet-induced Obese Mice. J Cancer Prev 2018; 23:99-106. [PMID: 30003071 PMCID: PMC6037208 DOI: 10.15430/jcp.2018.23.2.99] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 06/21/2018] [Accepted: 06/24/2018] [Indexed: 11/15/2022] Open
Abstract
Obesity is currently one of the most serious public health problems and it can lead to numerous metabolic diseases. Leucrose, d-glucopyranosyl-α-(1-5)-d-fructopyranose, is an isoform of sucrose and it is naturally found in pollen and honey. The aim of this study was to investigate the effect of leucrose on metabolic changes induced by a high-fat diet (HFD) that lead to obesity. C57BL/6 mice were fed a 60% HFD or a HFD with 25% (L25) or 50% (L50) of its total sucrose content replaced with leucrose for 12 weeks. Leucrose supplementation improved fasting blood glucose levels and hepatic triglyceride content. In addition, leucrose supplementation reduced mRNA levels of lipogenesis-related genes, including peroxisome proliferator-activated receptor γ, sterol regulatory element binding protein 1C, and fatty acid synthase in HFD mice. Conversely, mRNA levels of β oxidation-related genes, such as carnitine palmitoyltransferase 1A and acyl CoA oxidase, returned to control levels with leucrose supplementation. Taken together, these results demonstrated the therapeutic potential of leucrose to prevent metabolic abnormalities by mediating regulation of plasma glucose level and hepatic triglyceride accumulation.
Collapse
Affiliation(s)
- Jihye Lee
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Korea
| | - Eunju Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Korea
| | - Yuri Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Korea
| | - Sang-Ho Yoo
- Department of Food Science and Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, Seoul, Korea
| |
Collapse
|
476
|
Johnston LW, Liu Z, Retnakaran R, Zinman B, Giacca A, Harris SB, Bazinet RP, Hanley AJ. Clusters of fatty acids in the serum triacylglyceride fraction associate with the disorders of type 2 diabetes. J Lipid Res 2018; 59:1751-1762. [PMID: 29986954 DOI: 10.1194/jlr.p084970] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/05/2018] [Indexed: 11/20/2022] Open
Abstract
Our aim was to examine longitudinal associations of triacylglyceride fatty acid (TGFA) composition with insulin sensitivity (IS) and β-cell function. Adults at risk for T2D (n = 477) had glucose and insulin measured from a glucose challenge at three time points over 6 years. The outcome variables Matsuda insulin sensitivity index, homeostatic model of assessment 2-percent sensitivity (HOMA2-%S), Insulinogenic Index over HOMA-IR (IGI/IR), and Insulin Secretion-Sensitivity Index-2 were computed from the glucose challenge. Gas chromatography quantified TGFA composition from the baseline. We used adjusted generalized estimating equation (GEE) models and partial least squares (PLS) regression for the analysis. In adjusted GEE models, four TGFAs (14:0, 16:0, 14:1n-7, and 16:1n-7 as mol%) had strong negative associations with IS, whereas others (e.g., 18:1n-7, 18:1n-9, 20:2n-6, and 20:5n-3) had strong positive associations. Few associations were seen for β-cell function, except for 16:0, 18:1n-7, and 20:2n-6. PLS analysis indicated four TGFAs (14:0, 16:0, 14:1n-7, and 16:1n-7) that clustered together and strongly related with lower IS. These four TGFAs also correlated highly (r > 0.4) with clinically measured triacylglyceride. We found that higher proportions of a cluster of four TGFAs strongly related with lower IS as well as hypertriglyceridemia, suggesting that only a few FAs within the TGFA composition may primarily explain lipids' role in glucose dysregulation.
Collapse
Affiliation(s)
- Luke W Johnston
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Zhen Liu
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Ravi Retnakaran
- Leadership Sinai Centre for Diabetes, Division of Endocrinology, University of Toronto, Toronto, Ontario, Canada; Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Bernard Zinman
- Leadership Sinai Centre for Diabetes, Division of Endocrinology, University of Toronto, Toronto, Ontario, Canada; Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Adria Giacca
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Stewart B Harris
- Centre for Studies in Family Medicine, University of Western Ontario, London, Ontario, Canada
| | - Richard P Bazinet
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Anthony J Hanley
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada; Leadership Sinai Centre for Diabetes, Division of Endocrinology, University of Toronto, Toronto, Ontario, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
477
|
El-Shinnawy NA, Abd Elhalem SS, Haggag NZ, Badr G. Ameliorative role of camel whey protein and rosuvastatin on induced dyslipidemia in mice. Food Funct 2018; 9:1038-1047. [PMID: 29349446 DOI: 10.1039/c7fo01871a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The incidence of obesity is rapidly increasing throughout the world. Dyslipidemia is a major risk factor for a number of chronic diseases, including diabetes and cardiovascular diseases. This work presents a novel approach to study the activity of camel whey protein (WP) with antioxidant and anti-inflammatory properties as a cheap dietary protein substance extracted from camel milk to produce satiety and help in building muscles. Mice model suffering from dyslipidemia as a result of feeding on high fat-cholesterol diet for 8 weeks were administrated with either camel WP and/or rosuvastatin for 4 weeks. Dyslipidemia revealed significant increase in anthropometrical measurements, levels of glucose, insulin, cholesterol, triglycerides, low-density lipoprotein, total leucocyte count, inflammatory cytokines and reactive oxygen species, accompanied by a significant elevation in activating transcription factor-3 and inducible nitric oxide synthase expressions. These alterations were correlated with a profound reduction in high-density lipoprotein, peroxisome proliferator-activated receptor alpha and adiponectin along with a decrease in liver and muscle mitochondrial proteins. Rosuvastatin treatment to mice suffering from dyslipidemia in combination with camel WP for 4 weeks ameliorated these parameters. Notably, animals treated with both camel WP and rosuvastatin exhibited a remarkable decrease in the incidence of dyslipidemia. In addition, camel WP succeeded to overcome the therapeutic drawback posed from rosuvastatin therapy alone with minimal side effects.
Collapse
Affiliation(s)
- Nashwa Ahmed El-Shinnawy
- Zoology Department, Faculty of Women for Arts, Science and Education, Ain Shams University, 11757 Cairo, Egypt.
| | | | | | | |
Collapse
|
478
|
de Melo AF, Moreira CCL, Sales CF, Rentz T, Raposo HF, Garófalo MAR, Botion LM, Kettelhut IDC, de Oliveira HCF, Chaves VE. Increase in liver cytosolic lipases activities and VLDL-TAG secretion rate do not prevent the non-alcoholic fatty liver disease in cafeteria diet-fed rats. Biochimie 2018; 150:16-22. [DOI: 10.1016/j.biochi.2018.04.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 04/24/2018] [Indexed: 01/15/2023]
|
479
|
Yu Q, Liu Y, Wu Y, Chen Y. Dihydrocurcumin ameliorates the lipid accumulation, oxidative stress and insulin resistance in oleic acid-induced L02 and HepG2 cells. Biomed Pharmacother 2018; 103:1327-1336. [PMID: 29864915 DOI: 10.1016/j.biopha.2018.04.143] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/18/2018] [Accepted: 04/18/2018] [Indexed: 12/12/2022] Open
|
480
|
Lee J, Hong SW, Kwon H, Park SE, Rhee EJ, Park CY, Oh KW, Park SW, Lee WY. Exendin-4 improves ER stress-induced lipid accumulation and regulates lipin-1 signaling in HepG2 cells. Cell Stress Chaperones 2018; 23:629-638. [PMID: 29934713 PMCID: PMC6045528 DOI: 10.1007/s12192-017-0872-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 12/20/2017] [Accepted: 12/25/2017] [Indexed: 12/12/2022] Open
Abstract
Lipin-1 performs dual function during lipid metabolism, i.e., it functions as a transcriptional coactivator and as a phosphatidate phosphatase during triglyceride biosynthesis. We investigated whether exendin-4 prevented endoplasmic reticulum (ER) stress-induced hepatic steatosis and whether the protective effects of exendin-4 were associated with lipin-1 signaling. Tunicamycin and thapsigargin, ER stress inducers, increased triglycerides (TG) content and expression of genes encoding lipid droplet surface proteins. Exendin-4 decreased the expression of ER stress markers phosphorylated PKR like ER kinase (PERK), phosphorylated inositol-requiring enzyme 1 alpha (IRE1α), and glucose-regulated protein 78 kDa (GRP78) proteins and spliced X-box binding protein 1 (XBP-1s) mRNA and increased the expression of genes encoding lipolytic enzymes hormone-sensitive lipase (HSL) and monoacylglycerol lipase (MGL) and VLDL assembly-associated proteins microsomal triglyceride transfer protein (MTP) and apolipoprotein B (APOB) in tunicamycin-pretreated cells. Moreover, exendin-4 significantly decreased lipin-1β/α ratio by increasing SFRP10 and increased lipin-1 nuclear localization. The decrease in lipin-1β/α ratio was also observed in SIRT1 and AMPK agonist-treated cells. These data suggest that exendin-4 improves ER stress-induced hepatic lipid accumulation by increasing lipolysis and VLDL assembly, which is partially mediated by the regulation of lipin-1 signaling.
Collapse
Affiliation(s)
- Jinmi Lee
- Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Seok-Woo Hong
- Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Hyemi Kwon
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Se Eun Park
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Eun-Jung Rhee
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Cheol-Young Park
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Ki-Won Oh
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Sung-Woo Park
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Won-Young Lee
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea.
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea.
| |
Collapse
|
481
|
Association of nonalcoholic fatty liver disease grades with the plasma cell antigen-1 (PC-1) gene polymorphism. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2018. [DOI: 10.1016/j.ejmhg.2017.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
482
|
Chen J, Montagner A, Tan NS, Wahli W. Insights into the Role of PPARβ/δ in NAFLD. Int J Mol Sci 2018; 19:ijms19071893. [PMID: 29954129 PMCID: PMC6073272 DOI: 10.3390/ijms19071893] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/13/2018] [Accepted: 06/23/2018] [Indexed: 12/14/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major health issue in developed countries. Although usually associated with obesity, NAFLD is also diagnosed in individuals with low body mass index (BMI) values, especially in Asia. NAFLD can progress from steatosis to non-alcoholic steatohepatitis (NASH), which is characterized by liver damage and inflammation, leading to cirrhosis and hepatocellular carcinoma (HCC). NAFLD development can be induced by lipid metabolism alterations; imbalances of pro- and anti-inflammatory molecules; and changes in various other factors, such as gut nutrient-derived signals and adipokines. Obesity-related metabolic disorders may be improved by activation of the nuclear receptor peroxisome proliferator-activated receptor (PPAR)β/δ, which is involved in metabolic processes and other functions. This review is focused on research findings related to PPARβ/δ-mediated regulation of hepatic lipid and glucose metabolism and NAFLD development. It also discusses the potential use of pharmacological PPARβ/δ activation for NAFLD treatment.
Collapse
Affiliation(s)
- Jiapeng Chen
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore 308232, Singapore.
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| | - Alexandra Montagner
- ToxAlim, Research Center in Food Toxicology, National Institute for Agricultural Research (INRA), 180 Chemin de Tournefeuille, 31300 Toulouse, France.
- Institut National de La Santé et de La Recherche Médicale (INSERM), UMR1048, Institute of Metabolic and Cardiovascular Diseases, 31027 Toulouse, France.
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore 308232, Singapore.
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
- KK Research Centre, KK Women's and Children Hospital, 100 Bukit Timah Road, Singapore 229899, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science Technology & Research, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore.
| | - Walter Wahli
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore 308232, Singapore.
- ToxAlim, Research Center in Food Toxicology, National Institute for Agricultural Research (INRA), 180 Chemin de Tournefeuille, 31300 Toulouse, France.
- Center for Integrative Genomics, University of Lausanne, Génopode, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
483
|
Asghari S, Rafraf M, Farzin L, Asghari-Jafarabadi M, Ghavami SM, Somi MH. Effects of Pharmacologic Dose of Resveratrol Supplementation on Oxidative/Antioxidative Status Biomarkers in Nonalcoholic Fatty Liver Disease Patients: A Randomized, Double-Blind, Placebo-Controlled Trial. Adv Pharm Bull 2018; 8:307-317. [PMID: 30023333 PMCID: PMC6046420 DOI: 10.15171/apb.2018.036] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/12/2018] [Accepted: 05/19/2018] [Indexed: 12/11/2022] Open
Abstract
Purpose: Despite a proposed role for oxidative stress in the pathogenesis of nonalcoholic fatty liver disease (NAFLD), antioxidant approaches have not been sufficiently investigated in human NAFLD management. Resveratrol has been reported to possess a wide range of biological functions, including antioxidant activities. This study aimed to evaluate the effects of resveratrol supplementation on oxidative/anti-oxidative status in patients with NAFLD. Methods: This randomized, double-blind, placebo-controlled clinical trial was conducted on 60 patients with NAFLD (males and females) aged 20 to 60 years, and body mass index (BMI) of 25-35 kg/m2. Subjects were randomly assigned to receive a daily dose of 600 mg resveratrol (2×300 mg pure trans-resveratrol capsules; n=30) or placebo capsules (n=30) for 12 wk. Fasting blood samples, anthropometric measurements, and dietary intakes were collected for all patients at baseline and at the end of the trial. Oxidative stress was evaluated by measurement of serum malondialdehyde (MDA), oxidized low-density lipoprotein (ox-LDL), total antioxidant capacity (TAC), and erythrocyte superoxide dismutase (SOD) as well as glutathione peroxidase (GSH-Px) activities. Changes in the outcomes were analyzed using analysis of covariance (ANCOVA). Results: Resveratrol supplementation did not significantly affect neither serum MDA, ox-LDL, and TAC levels, nor erythrocyte SOD and GSH-Px activities, compared to placebo group (All P>0.05). Moreover, changes in serum levels of liver enzymes (ALT, AST, GGT, and ALP) were not significant in neither of the study groups (All P>0.05). Conclusion: Resveratrol supplementation did not modify oxidative/anti-oxidative status in patients with NAFLD.
Collapse
Affiliation(s)
- Somayyeh Asghari
- Students' Research Committee, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Rafraf
- Nutrition Research Center, Department of Community Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Laleh Farzin
- Students' Research Committee, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Asghari-Jafarabadi
- Road Traffic Injury Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Statistics and Epidemiology, Faculty of Health, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed-Mostafa Ghavami
- Department of Radiology, Paramedical school, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad-Hossein Somi
- Liver and Gastrointestinal Diseases Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
484
|
Mardinoglu A, Boren J, Smith U, Uhlen M, Nielsen J. Systems biology in hepatology: approaches and applications. Nat Rev Gastroenterol Hepatol 2018; 15:365-377. [PMID: 29686404 DOI: 10.1038/s41575-018-0007-8] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Detailed insights into the biological functions of the liver and an understanding of its crosstalk with other human tissues and the gut microbiota can be used to develop novel strategies for the prevention and treatment of liver-associated diseases, including fatty liver disease, cirrhosis, hepatocellular carcinoma and type 2 diabetes mellitus. Biological network models, including metabolic, transcriptional regulatory, protein-protein interaction, signalling and co-expression networks, can provide a scaffold for studying the biological pathways operating in the liver in connection with disease development in a systematic manner. Here, we review studies in which biological network models were used to integrate multiomics data to advance our understanding of the pathophysiological responses of complex liver diseases. We also discuss how this mechanistic approach can contribute to the discovery of potential biomarkers and novel drug targets, which might lead to the design of targeted and improved treatment strategies. Finally, we present a roadmap for the successful integration of models of the liver and other human tissues with the gut microbiota to simulate whole-body metabolic functions in health and disease.
Collapse
Affiliation(s)
- Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden. .,Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.
| | - Jan Boren
- Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ulf Smith
- Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mathias Uhlen
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Jens Nielsen
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden.,Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
485
|
Namkung J, Shong KE, Kim H, Oh CM, Park S, Kim H. Inhibition of Serotonin Synthesis Induces Negative Hepatic Lipid Balance. Diabetes Metab J 2018; 42:233-243. [PMID: 29885107 PMCID: PMC6015967 DOI: 10.4093/dmj.2017.0084] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/14/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Hepatic steatosis is caused by metabolic stress associated with a positive lipid balance, such as insulin resistance and obesity. Previously we have shown the anti-obesity effects of inhibiting serotonin synthesis, which eventually improved insulin sensitivity and hepatic steatosis. However, it is not clear whether serotonin has direct effect on hepatic lipid accumulation. Here, we showed the possibility of direct action of serotonin on hepatic steatosis. METHODS Mice were treated with para-chlorophenylalanine (PCPA) or LP-533401 to inhibit serotonin synthesis and fed with high fat diet (HFD) or high carbohydrate diet (HCD) to induce hepatic steatosis. Hepatic triglyceride content and gene expression profiles were analyzed. RESULTS Pharmacological and genetic inhibition of serotonin synthesis reduced HFD-induced hepatic lipid accumulation. Furthermore, short-term PCPA treatment prevented HCD-induced hepatic steatosis without affecting glucose tolerance and browning of subcutaneous adipose tissue. Gene expression analysis revealed that the expressions of genes involved in de novo lipogenesis and triacylglycerol synthesis were downregulated by short-term PCPA treatment as well as long-term PCPA treatment. CONCLUSION Short-term inhibition of serotonin synthesis prevented hepatic lipid accumulation without affecting systemic insulin sensitivity and energy expenditure, suggesting the direct steatogenic effect of serotonin in liver.
Collapse
Affiliation(s)
- Jun Namkung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
- Department of Biochemistry, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Ko Eun Shong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Hyeongseok Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Chang Myung Oh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Sangkyu Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
- Department of Biochemistry, Catholic Kwandong University College of Medicine, Gangneung, Korea.
| | - Hail Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon, Korea.
| |
Collapse
|
486
|
Schuster S, Cabrera D, Arrese M, Feldstein AE. Triggering and resolution of inflammation in NASH. Nat Rev Gastroenterol Hepatol 2018; 15:349-364. [PMID: 29740166 DOI: 10.1038/s41575-018-0009-6] [Citation(s) in RCA: 615] [Impact Index Per Article: 87.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is considered the progressive form of nonalcoholic fatty liver disease (NAFLD) and is characterized by liver steatosis, inflammation, hepatocellular injury and different degrees of fibrosis. A central issue in this field relates to the identification of those factors that trigger inflammation, thus fuelling the transition from nonalcoholic fatty liver to NASH. These triggers of liver inflammation might have their origins outside the liver (such as in adipose tissue or the gut) as well as inside the organ (for instance, lipotoxicity, innate immune responses, cell death pathways, mitochondrial dysfunction and endoplasmic reticulum stress), both of which contribute to NASH development. In this Review, we summarize the currently available information on the key upstream triggers of inflammation in NASH. We further delineate the mechanisms by which liver inflammation is resolved and the implications of a defective pro-resolution process. A better knowledge of these mechanisms should help to design targeted therapies able to halt or reverse disease progression.
Collapse
Affiliation(s)
- Susanne Schuster
- Department of Pediatrics, University of California, San Diego, CA, USA
| | - Daniel Cabrera
- Department of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Ciencias Químicas y Biológicas, Facultad de Salud, Universidad Bernardo O Higgins, Santiago, Chile
| | - Marco Arrese
- Department of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centre for Aging and Regeneration (CARE), Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ariel E Feldstein
- Department of Pediatrics, University of California, San Diego, CA, USA.
| |
Collapse
|
487
|
Oniciu DC, Hashiguchi T, Shibazaki Y, Bisgaier CL. Gemcabene downregulates inflammatory, lipid-altering and cell-signaling genes in the STAM™ model of NASH. PLoS One 2018; 13:e0194568. [PMID: 29847555 PMCID: PMC5976190 DOI: 10.1371/journal.pone.0194568] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 03/06/2018] [Indexed: 12/11/2022] Open
Abstract
Background and aims Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) can advance, if untreated, to liver fibrosis, cirrhosis, hepatocellular carcinoma, liver failure and liver-related death. In the United States, NASH affects approximately 2–5% of the population and an additional 10–30% have NAFLD. The number of drugs in development for NASH is growing steadily, along with nonclinical models to support prediction of clinical success. Here we evaluate gemcabene, a first-in-class clinical candidate for dyslipidemia, for its potential utility, based on its combined lipid-lowering and anti-inflammatory efficacy in clinical trials, in a preclinical model of NASH. Methods Gemcabene was evaluated in the STAM™ murine model of NASH. Gemcabene intervention in mice made diabetic with streptozotocin and fed a high fat high-caloric diet was assessed for changes in plasma, and hepatic histological and mRNA markers of lipid metabolism and inflammation. Results Gemcabene significantly downregulated hepatic mRNA markers of inflammation (TNF-α, MCP-1, MIP-1β, CCR5, CCR2, NF-κB), lipogenesis and lipid modulation (ApoC-III, ACC1, ADH-4, Sulf-2), and fibrosis (TIMP-1 and MMP-2). These effects are important for the prevention of steatosis, inflammation, and hepatocyte ballooning (i.e., the components of the NAFLD Activity Score or NAS), and inhibition of fibrosis progression, and were observed following treatment with gemcabene. Conclusions These non-clinical findings corroborate with existing clinical data to support the clinical evaluation of gemcabene as a potential new treatment for NASH.
Collapse
|
488
|
Rix I, Steen Pedersen J, Storgaard H, Gluud LL. Cardiometabolic effects of antidiabetic drugs in non-alcoholic fatty liver disease. Clin Physiol Funct Imaging 2018; 39:122-127. [PMID: 29808958 DOI: 10.1111/cpf.12526] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/04/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE Non-alcoholic fatty liver disease (NAFLD) affects about 25% of the population worldwide. NAFLD may be viewed as the hepatological manifestation of metabolic syndrome. Patients with metabolic syndrome due to diabetes or obesity have an increased risk of cardiovascular disease. This narrative review describes cardiometabolic effects of antidiabetic drugs in NAFLD. METHODS We conducted a systematic search in PubMed and manually scanned bibliographies in trial databases and reference lists in relevant articles. RESULTS Heart disease is the leading cause of death in NAFLD. Conversely, NAFLD is an independent cardiovascular risk factor in patients suffering from metabolic syndrome. NAFLD is associated with markers of atherosclerosis, and patients have increased risk of ischaemic heart disease. Additionally, patients with NAFLD have increased risk of cardiac dysfunction and heart failure. There are no randomized controlled trials showing clear effects of medical treatment on clinical outcomes in patients with NAFLD. However, based on evidence from small trials and extrapolation from trials evaluating patients with type 2 diabetes, some antidiabetic drugs may be beneficial on cardiovascular function in patients with NAFLD. CONCLUSION At present, there is promising evidence of a potential effect of antidiabetic drugs for patients with NAFLD. Future studies should address the treatment of NAFLD and the liver-related consequences but also aim at improving the cardiometabolic outcomes.
Collapse
Affiliation(s)
- Iben Rix
- Gastrounit, Medical Division, Hvidovre Hospital, University of Copenhagen, Hvidovre, Denmark
| | - Julie Steen Pedersen
- Gastrounit, Medical Division, Hvidovre Hospital, University of Copenhagen, Hvidovre, Denmark
| | - Heidi Storgaard
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Lise Lotte Gluud
- Gastrounit, Medical Division, Hvidovre Hospital, University of Copenhagen, Hvidovre, Denmark
| |
Collapse
|
489
|
Cai C, Yu H, Huang G, Du X, Yu X, Zhou Y, Shen W. Histone modifications in fatty acid synthase modulated by carbohydrate responsive element binding protein are associated with non‑alcoholic fatty liver disease. Int J Mol Med 2018; 42:1215-1228. [PMID: 29786745 PMCID: PMC6089769 DOI: 10.3892/ijmm.2018.3702] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 04/26/2018] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a manifestation of metabolic syndrome in the liver and is closely associated with diabetes; however, its pathogenesis remains to be elucidated. Carbohydrate responsive element binding protein (ChREBP), the hub of glucolipid metabolism, regulates the induction of fatty acid synthase (FASN), the key enzyme of de novo lipogenesis, by directly binding to carbohydrate response element (ChoRE) in its promoter. Investigations of histone modifications on NAFLD remain in their infancy. In the present study, by using ChIP, the association between histone modifications and FASN transcription was investigated and histone modifications in FASN modulated by ChREBP were measured. It was demonstrated that ChREBP induced FASN ChREBP-ChoRE binding to accelerate the expression of FASN, leading to hepatocellular steatosis by facilitating H3 and H4 acetylation, H3K4 trimethylation and the phosphorylation of H3S10, but inhibiting the trimethylation of H3K9 and H4K20 in FASN promoter regions of HepG2 and L02 cells. It was also found that ChREBP-ChoRE binding of FASN relied on histone acetylation and that the transcriptional activity of ChREBP on FASN is required, based on the premise that histone acetylation causes conformational changes in FASN chromatin. This indicated histone acetylation as a crucial mechanism involved in the transcription of FASN modulated by ChREBP. Consequently, the present study provides further insight into the pathophysiology and a novel therapeutic potential of NAFLD based on epigenetic mechanisms.
Collapse
Affiliation(s)
- Can Cai
- Department of Gastroenterology, 2nd Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Huihong Yu
- Department of Gastroenterology, 2nd Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Guangming Huang
- Department of Gastroenterology, 2nd Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Xuan Du
- Department of Gastroenterology, 2nd Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Xiaoqing Yu
- Department of Gastroenterology, 2nd Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Youping Zhou
- Department of Gastroenterology, 2nd Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Wei Shen
- Department of Gastroenterology, 2nd Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
490
|
CREBH Regulates Systemic Glucose and Lipid Metabolism. Int J Mol Sci 2018; 19:ijms19051396. [PMID: 29738435 PMCID: PMC5983805 DOI: 10.3390/ijms19051396] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/30/2018] [Accepted: 05/06/2018] [Indexed: 12/23/2022] Open
Abstract
The cyclic adenosine monophosphate (cAMP)-responsive element-binding protein H (CREBH, encoded by CREB3L3) is a membrane-bound transcriptional factor that primarily localizes in the liver and small intestine. CREBH governs triglyceride metabolism in the liver, which mediates the changes in gene expression governing fatty acid oxidation, ketogenesis, and apolipoproteins related to lipoprotein lipase (LPL) activation. CREBH in the small intestine reduces cholesterol transporter gene Npc1l1 and suppresses cholesterol absorption from diet. A deficiency of CREBH in mice leads to severe hypertriglyceridemia, fatty liver, and atherosclerosis. CREBH, in synergy with peroxisome proliferator-activated receptor α (PPARα), has a crucial role in upregulating Fgf21 expression, which is implicated in metabolic homeostasis including glucose and lipid metabolism. CREBH binds to and functions as a co-activator for both PPARα and liver X receptor alpha (LXRα) in regulating gene expression of lipid metabolism. Therefore, CREBH has a crucial role in glucose and lipid metabolism in the liver and small intestine.
Collapse
|
491
|
Mayo R, Crespo J, Martínez-Arranz I, Banales JM, Arias M, Mincholé I, Aller de la Fuente R, Jimenez-Agüero R, Alonso C, de Luis DA, Vitek L, Stritesky J, Caballería J, Romero-Gómez M, Martín-Duce A, Mugüerza Huguet JM, Busteros-Moraza JI, Idowu MO, Castro A, Martínez-Chantar ML, Ortiz P, Bruha R, Lu SC, Bedossa P, Noureddin M, Sanyal AJ, Mato JM. Metabolomic-based noninvasive serum test to diagnose nonalcoholic steatohepatitis: Results from discovery and validation cohorts. Hepatol Commun 2018; 2:807-820. [PMID: 30027139 PMCID: PMC6049064 DOI: 10.1002/hep4.1188] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 03/15/2018] [Accepted: 03/15/2018] [Indexed: 12/11/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common type of chronic liver disease worldwide and includes a broad spectrum of histologic phenotypes, ranging from simple hepatic steatosis or nonalcoholic fatty liver (NAFL) to nonalcoholic steatohepatitis (NASH). While liver biopsy is the reference gold standard for NAFLD diagnosis and staging, it has limitations due to its sampling variability, invasive nature, and high cost. Thus, there is a need for noninvasive biomarkers that are robust, reliable, and cost effective. In this study, we measured 540 lipids and amino acids in serum samples from biopsy‐proven subjects with normal liver (NL), NAFL, and NASH. Using logistic regression analysis, we identified two panels of triglycerides that could first discriminate between NAFLD and NL and second between NASH and NAFL. These noninvasive tests were compared to blinded histology as a reference standard. We performed these tests in an original cohort of 467 patients with NAFLD (90 NL, 246 NAFL, and 131 NASH) that was subsequently validated in a separate cohort of 192 patients (7 NL, 109 NAFL, 76 NASH). The diagnostic performances of the validated tests showed an area under the receiver operating characteristic curve, sensitivity, and specificity of 0.88 ± 0.05, 0.94, and 0.57, respectively, for the discrimination between NAFLD and NL and 0.79 ± 0.04, 0.70, and 0.81, respectively, for the discrimination between NASH and NAFL. When the analysis was performed excluding patients with glucose levels >136 mg/dL, the area under the receiver operating characteristic curve for the discrimination between NASH and NAFL increased to 0.81 ± 0.04 with sensitivity and specificity of 0.73 and 0.80, respectively. Conclusion: The assessed noninvasive lipidomic serum tests distinguish between NAFLD and NL and between NASH and NAFL with high accuracy. (Hepatology Communications 2018;2:807‐820)
Collapse
Affiliation(s)
- Rebeca Mayo
- OWL Metabolomics Bizkaia Technology Park Derio Spain
| | - Javier Crespo
- Department of Digestive Disease Marqués de Valdecilla University Hospital, Cantabaria University Research Institute Marqués de Valdecilla (IDIVAL) Santander Spain
| | | | - Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute Donostia University Hospital, University of the Basque Country (UPV-EHU) CIBERehd, IKERBASQUE Donostia Spain
| | - Mayte Arias
- Department of Digestive Disease Marqués de Valdecilla University Hospital, Cantabaria University Research Institute Marqués de Valdecilla (IDIVAL) Santander Spain
| | | | - Rocío Aller de la Fuente
- Department of Digestive Disease, Clinic University Hospital University of Valladolid Valladolid Spain
| | - Raúl Jimenez-Agüero
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute Donostia University Hospital, University of the Basque Country (UPV-EHU) CIBERehd, IKERBASQUE Donostia Spain
| | | | - Daniel A de Luis
- Center of Investigation of Endocrinology and Clinical Nutrition, Medicine School and Department of Endocrinology and Nutrition, Clinic University Hospital University of Valladolid Valladolid Spain
| | - Libor Vitek
- Faculty General Hospital and the First Faculty of Medicine Charles University Prague Czech Republic
| | - Jan Stritesky
- Faculty General Hospital and the First Faculty of Medicine Charles University Prague Czech Republic
| | - Joan Caballería
- Liver Unit, Hospital Clinic, CIBERehd IDIBAPS Barcelona Spain
| | - Manuel Romero-Gómez
- Unit for the Clinical Management of Digestive Diseases and CIBERehd Valme University Hospital Seville Spain
| | - Antonio Martín-Duce
- Faculty of Medicine and Health Science University Hospital Príncipe de Asturias, Alcalá University Madrid Spain
| | | | | | - Michael O Idowu
- Division of Surgical Pathology, Department of Pathology Virginia Commonwealth University Medical Center Richmond VA United States
| | | | | | - Pablo Ortiz
- OWL Metabolomics Bizkaia Technology Park Derio Spain
| | - Radan Bruha
- Faculty General Hospital and the First Faculty of Medicine Charles University Prague Czech Republic
| | - Shelly C Lu
- Division of Digestive and Liver Diseases Cedars-Sinai Medical Center Los Angeles CA United States
| | - Pierre Bedossa
- Department of Pathology University Hospitals Paris Nord Val de Seine, Beaujon, Hauts-de-Seine Clichy France
| | - Mazen Noureddin
- Division of Digestive and Liver Diseases Cedars-Sinai Medical Center Los Angeles CA United States
| | - Arun J Sanyal
- Division of Gastroenterology and Hepatology Virginia Commonwealth University Medical Center Richmond VA United States
| | - José M Mato
- CIC bioGUNE, CIBERehd Bizkaia Technology Park Derio Bizkaia Spain
| |
Collapse
|
492
|
Qiu Y, Wang S, Wan T, Ye M, Jiang R, Pei L, Yang L. Blood-based novel biomarkers for nonalcoholic steatohepatitis. Biomark Med 2018; 12:501-515. [PMID: 29712439 DOI: 10.2217/bmm-2017-0361] [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] [Indexed: 01/14/2023] Open
Abstract
Nonalcoholic fatty liver disease has become a social health challenge of global concern. The term nonalcoholic steatohepatitis (NASH) is a more severe condition than simple steatosis and distinguishing NASH from nonalcoholic fatty liver disease is particularly important. Liver biopsy remains a gold standard in diagnosing NASH. Meanwhile, radiological techniques such as ultrasonography and MRI are also applied widely. However, the invasive and expensive examination is not suitable for screening, and there is a great need for reliable and appropriate biomarkers to screen patients for NASH. Based on the current studies of blood-based novel biomarkers, we attempt to summarize the latest findings on biomarkers for NASH, including blood biomarkers encompassing proteins, lipids and miRNAs; the correlation between extracellular vesicles and NASH; and treatment strategies for NASH.
Collapse
Affiliation(s)
- Yun Qiu
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong 510080, PR China.,Guangdong Provincial Key Laboratory of Food, Nutrition & Health, Guangzhou, Guangdong 510080, PR China
| | - Sufan Wang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong 510080, PR China.,Guangdong Provincial Key Laboratory of Food, Nutrition & Health, Guangzhou, Guangdong 510080, PR China
| | - Ting Wan
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong 510080, PR China.,Guangdong Provincial Key Laboratory of Food, Nutrition & Health, Guangzhou, Guangdong 510080, PR China
| | - Mingtong Ye
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong 510080, PR China.,Guangdong Provincial Key Laboratory of Food, Nutrition & Health, Guangzhou, Guangdong 510080, PR China
| | - Rui Jiang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong 510080, PR China.,Guangdong Provincial Key Laboratory of Food, Nutrition & Health, Guangzhou, Guangdong 510080, PR China
| | - Lei Pei
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong 510080, PR China.,Guangdong Provincial Key Laboratory of Food, Nutrition & Health, Guangzhou, Guangdong 510080, PR China
| | - Lili Yang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong 510080, PR China.,Guangdong Provincial Key Laboratory of Food, Nutrition & Health, Guangzhou, Guangdong 510080, PR China
| |
Collapse
|
493
|
Chiu CY, Wang LP, Liu SH, Chiang MT. Fish Oil Supplementation Alleviates the Altered Lipid Homeostasis in Blood, Liver, and Adipose Tissues in High-Fat Diet-Fed Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:4118-4128. [PMID: 29627983 DOI: 10.1021/acs.jafc.8b00529] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
This study investigated the effects of dietary supplementation of fish oil on the signals of lipid metabolism involved in hepatic cholesterol and triglyceride influx and excretion in high-fat diet (HFD)-fed rats. Fish oil (FO) repressed body (HFD, 533 ± 18.2 g; HFD+FO, 488 ± 28.0 g, p < 0.05) and liver weights (HFD, 5.7 ± 0.6 g/100 g of body weight; HFD+FO, 4.8 ± 0.4 g/100 g of body weight, p < 0.05) in HFD-fed rats. Fish oil could also improve HFD-induced imbalance of lipid metabolism in blood, liver, and adipose tissues including the significant decreases in plasma and liver total cholesterol (TC) (plasma-HFD, 113 ± 33.6 mg/dL; HFD+FO, 50.0 ± 5.95 mg/dL, p < 0.05; liver-HFD, 102 ± 13.0 mg/g liver; [corrected] HFD+FO, 86.6 ± 7.81 mg/g liver, [corrected] p < 0.05), blood, liver, and adipose triglyceride (TG) (blood-HFD, 52.5 ± 20.4 mg/dL; HFD+FO, 29.8 ± 4.30 mg/dL, p < 0.05; liver-HFD, 56.2 ± 10.0 mg/g liver; [corrected] HFD+FO, 30.3 ± 5.28 mg/g liver, [corrected] p < 0.05; adipose-HFD, 614 ± 73.2 mg/g liver, [corrected] HFD+FO, 409 ± 334 mg/g of adipose tissue, [corrected] p < 0.05), and low density (HFD, 79.8 ± 40.9 mg/dL; HFD+FO, 16.6 ± 5.47 mg/dL, p < 0.05) and very-low-density (HFD, 49.7 ± 33.3 mg/dL; HFD+FO, 10.4 ± 3.45 mg/dL, p < 0.05) lipoprotein and the significant increases in fecal TC (HFD, 12.2 ± 0.67 mg/g feces; [corrected] HFD+FO, 16.3 ± 2.04 mg/g feces, [corrected] < 0.05) and TG (HFD, 2.09 ± 0.10 mg/g feces; [corrected] HFD+FO, 2.38 ± 0.22 mg/g feces, [corrected] p < 0.05) and lipoprotein lipase activity of adipose tissues (HFD, 16.6 ± 3.64 μM p-nitrophenol; HFD+FO, 24.5 ± 4.19 μM p-nitrophenol, p < 0.05). Moreover, fish oil significantly activated the protein expressions of hepatic lipid metabolism regulators (AMPKα and PPARα) and significantly regulated the lipid-transport-related signaling molecules (ApoE, MTTP, ApoB, Angptl4, ApoCIII, ACOX1, and SREBPF1) in blood or liver of HFD-fed rats. These results suggest that fish oil supplementation improves HFD-induced imbalance of lipid homeostasis in blood, liver, and adipose tissues in rats.
Collapse
Affiliation(s)
- Chen-Yuan Chiu
- Institute of Food Safety and Health, College of Public Health , National Taiwan University , Taipei 100 , Taiwan
| | - Lou-Pin Wang
- Department of Food Science, College of Life Science , National Taiwan Ocean University , Keelung 202 , Taiwan
| | - Shing-Hwa Liu
- Graduate Institute of Toxicology, College of Medicine , National Taiwan University , Taipei 100 , Taiwan
- Department of Pediatrics, College of Medicine and Hospital , National Taiwan University , Taipei 100 , Taiwan
- Department of Medical Research, China Medical University Hospital , China Medical University , Taichung 404 , Taiwan
| | - Meng-Tsan Chiang
- Department of Food Science, College of Life Science , National Taiwan Ocean University , Keelung 202 , Taiwan
| |
Collapse
|
494
|
Zhang S, Mao Y, Fan X. Inhibition of ghrelin o-acyltransferase attenuated lipotoxicity by inducing autophagy via AMPK-mTOR pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:873-885. [PMID: 29713145 PMCID: PMC5912383 DOI: 10.2147/dddt.s158985] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background Nonalcoholic fatty liver disease (NAFLD) has been considered the most commonly occurring chronic hepatopathy in the world. Ghrelin o-acyltransferase (GOAT) is an acylation enzyme which has an acylated position 3 serine on ghrelin. Recent investigation revealed that activated autophagy could attenuate liver steatosis. The aim of this study was to explore therapeutic roles that inhibit GOAT exerted in NAFLD, and its potential association with autophagy. Materials and methods Human LO2 cells were pretreated with siRNA-GOAT to induce liver steatosis using free fatty acids (FFAs). A chronic NAFLD model was established by feeding male mice C57bl/6 with high-fat diet (HFD) for 56 days with GO-CoA-Tat administrated subcutaneously. Lipid droplets were identified by Oil Red O stains. Body weight (BW) of mice was measured every week. Autophagy, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), serum biochemical indicators (glucose [Glu], total cholesterol [TC], triglyceride [TG], aspartate aminotransferase [AST], alanine aminotransferase [ALT]) and signaling pathway proteins of phosphorylated AMPK–mTOR were measured. Results The TG contents of the FFA and HFD groups were decreased by the inhibition of GOAT. Among mice treated with GO-CoA-Tat and siRNA-GOAT, IL-6 and TNF-α concentrations were remarkably decreased. Indicators of liver injury such as ALT and AST were also remarkably decreased among mice treated with GO-CoA-Tat. Likewise, GO-CoA-Tat significantly reduced the BW of mice and serum TG, TC and Glu. Autophagy was induced along with reduced lipids in the cells of the FFA and HFD groups. The inhibition of GOAT upregulated autophagy via AMPK–mTOR restoration. Conclusion These results indicate that the inhibition of GOAT attenuates lipotoxicity by autophagy stimulation via AMPK–mTOR restoration and offers innovative evidence for using GO-CoA-Tat or siRNA-GOAT in NAFLD clinically.
Collapse
Affiliation(s)
- Shaoren Zhang
- Department of Gastroenterology and Hepatology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Yuqing Mao
- Department of Gastroenterology and Hepatology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Xiaoming Fan
- Department of Gastroenterology and Hepatology, Jinshan Hospital of Fudan University, Shanghai, China
| |
Collapse
|
495
|
Mishra A, Arindkar S, Sahay P, Kumar JM, Upadhyay PK, Majumdar SS, Nagarajan P. Evaluation of high-fat high-fructose diet treatment in factor VIII (coagulation factor)-deficient mouse model. Int J Exp Pathol 2018; 99:46-53. [PMID: 29656466 DOI: 10.1111/iep.12264] [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: 09/12/2017] [Accepted: 02/03/2018] [Indexed: 12/21/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD)-like conditions enhance the production and action of clotting factors in humans. However, studies examining the effect of NAFLD due to high-fat high-fructose (HFHF) diet in factor VIII-deficient (haemophilia A) animals or patients have not been reported previously. In this study, we investigated the individual role of factor VIII in the progression of diet-induced NAFLD in the factor 8-/- (F8-/- ) mouse model system and its consequences on the haemophilic status of the mice. The F8-/- mice were fed with HFHF diet for 14 weeks. Physiological, biochemical, haematological, molecular, pathological, and immune histochemical analyses were performed to evaluate the effect of this diet. The F8-/- mice developed hepatic steatosis after 14 weeks HFHF diet and displayed lower energy metabolism, higher myeloid cell infiltration in the liver, decreased platelet count, upregulated de novo fatty acid synthesis, lipid accumulation, and collagen deposition. This study helps to understand the role of factor VIII in NAFLD pathogenesis and to analyse the severity and consequences of steatosis in haemophilic patients as compared to normal population. This study suggests that haemophilic animals (F8-/- mice) are highly prone to hepatic steatosis and thrombocytopenia.
Collapse
Affiliation(s)
| | | | - Preeti Sahay
- National Institute of Immunology, New Delhi, India
| | | | | | | | | |
Collapse
|
496
|
Ippagunta SM, Kharitonenkov A, Adams AC, Hillgartner FB. Cholic Acid Supplementation of a High-Fat Obesogenic Diet Suppresses Hepatic Triacylglycerol Accumulation in Mice via a Fibroblast Growth Factor 21-Dependent Mechanism. J Nutr 2018; 148:510-517. [PMID: 29659970 DOI: 10.1093/jn/nxy022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/25/2018] [Indexed: 12/13/2022] Open
Abstract
Background Supplementation of a high-fat obesogenic diet (HFD) with cholic acid (CA) suppresses the development of obesity, insulin resistance, and hepatic steatosis in mice. Objective We investigated the role of fibroblast growth factor 21 (FGF21) in mediating the beneficial actions of CA on metabolic syndrome. Methods Male 7-wk-old wild-type (WT) mice and FGF21 knockout (FGF21KO) mice were fed an HFD for 12 wk followed by a 4-wk period in which the mice were fed the HFD alone or supplemented with 0.5% CA. Body composition, gross energy efficiency, glucose tolerance, homeostasis model assessment of insulin resistance (HOMA-IR), and hepatic triacylglycerol (TG) concentrations were measured. Results CA administration improved glucose tolerance and decreased total body fat accretion, gross energy efficiency, fasting blood glucose concentrations, and HOMA-IR in both WT mice and FGF21KO mice. The extent of the effect of CA on glucose tolerance, fasting blood glucose concentrations, and HOMA-IR was similar in both mouse strains, whereas the extent of the effect of CA on total body fat accretion and gross energy efficiency was 4.2- to 4.4-fold greater in FGF21KO mice than in WT mice. Further analyses showed that CA decreased hepatic TG concentrations in WT mice (49%) but had no effect on hepatic TG concentrations in FGF21KO mice. CA decreased the activation state of hepatic acetyl-CoA carboxylase 1 (ACC1) and adipose tissue hormone-sensitive lipase (HSL) in WT mice but was not effective in decreasing the activation of ACC1 and HSL in FGF21KO mice. Conclusions FGF21 signaling is required for the beneficial effect of CA on hepatic TG accumulation in mice fed an HFD. We propose that FGF21 signaling potentiates the ability of CA to decrease the activation of ACC1 and HSL, key enzymes controlling the supply of long-chain fatty acid precursors for hepatic TG synthesis.
Collapse
Affiliation(s)
- Siri M Ippagunta
- Department of Biochemistry, West Virginia University, Morgantown, WV
| | | | - Andrew C Adams
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN
| | | |
Collapse
|
497
|
Zhao N, Yang S, Jia Y, Sun B, He B, Zhao R. Maternal betaine supplementation attenuates glucocorticoid-induced hepatic lipid accumulation through epigenetic modification in adult offspring rats. J Nutr Biochem 2018; 54:105-112. [DOI: 10.1016/j.jnutbio.2017.12.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 11/15/2017] [Accepted: 12/06/2017] [Indexed: 12/21/2022]
|
498
|
Rosales-Cruz P, Domínguez-Pérez M, Reyes-Zárate E, Bello-Monroy O, Enríquez-Cortina C, Miranda-Labra R, Bucio L, Gómez-Quiroz LE, Rojas-Del Castillo E, Gutiérrez-Ruíz MC, Souza-Arroyo V. Cadmium exposure exacerbates hyperlipidemia in cholesterol-overloaded hepatocytes via autophagy dysregulation. Toxicology 2018; 398-399:41-51. [PMID: 29486218 DOI: 10.1016/j.tox.2018.02.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/13/2018] [Accepted: 02/23/2018] [Indexed: 02/07/2023]
|
499
|
Mohr R, Boesecke C, Dold L, Schierwagen R, Schwarze-Zander C, Wasmuth JC, Weisensee I, Rockstroh JK, Trebicka J. Return-to-health effect of modern combined antiretroviral therapy potentially predisposes HIV patients to hepatic steatosis. Medicine (Baltimore) 2018; 97:e0462. [PMID: 29702998 PMCID: PMC5944472 DOI: 10.1097/md.0000000000010462] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 03/26/2018] [Accepted: 03/27/2018] [Indexed: 12/15/2022] Open
Abstract
Prevalence and risk factors for hepatic steatosis (HS) in the human immunodeficiency virus (HIV)-positive population of western countries are controversially discussed and potentially confounded by coinfection with viral hepatitis. Significant HS (more than 10% of hepatocytes) can be accurately assessed using controlled attenuation parameter (CAP) determination. Aim of this study was to assess prevalence and factors associated with significant HS in HIV monoinfected patients.A total of 364 HIV-infected patients (289 monoinfected) were included in this prospective, cross-sectional study. All patients underwent CAP determination. Steatosis was classified as S1 (significant steatosis) with CAP > 238 dB/m, S2 with CAP > 260 dB/m, and S3 with CAP > 292 dB/m. Multivariable logistic regression analyses were performed to assess the factors associated with HS in this cohort.Significant HS was detected in 118 monoinfected patients (149 in the total cohort). In the total cohort as well as in the monoinfected patients alone, HS grade distribution showed a similar pattern (S1:29%, S2:34%, and S3:37%). Interestingly, patients with HS had a longer history of HIV infection and combined antiretroviral therapy (cART). Interalia, age, gender, ethnicity, and metabolic factors were strongly associated with HS, while body mass index (BMI), triglyceride, and glycated hemoglobin (HbA1c) levels were independently associated with significant HS.HS is highly prevalent among HIV monoinfected patients. Although metabolic risk factors, such as obesity and poorly controlled diabetes, are independently associated with HS in HIV monoinfected patients, cART and control of HIV seem to play an indirect role in the development of HS, probably through the return-to-health effect.
Collapse
Affiliation(s)
- Raphael Mohr
- Department of Medicine I, University Hospital Bonn
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Christoph Boesecke
- Department of Medicine I, University Hospital Bonn
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Leona Dold
- Department of Medicine I, University Hospital Bonn
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | | | - Carolynne Schwarze-Zander
- Department of Medicine I, University Hospital Bonn
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Jan-Christian Wasmuth
- Department of Medicine I, University Hospital Bonn
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | | | - Jürgen Kurt Rockstroh
- Department of Medicine I, University Hospital Bonn
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Jonel Trebicka
- Department of Medicine I, University Hospital Bonn
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- European Foundation for the Study of Chronic Liver Failure, EF Clif
- Institute for Bioengineering of Catalonia, Barcelona, Spain
| |
Collapse
|
500
|
Lindquist C, Bjørndal B, Rossmann CR, Svardal A, Hallström S, Berge RK. A fatty acid analogue targeting mitochondria exerts a plasma triacylglycerol lowering effect in rats with impaired carnitine biosynthesis. PLoS One 2018; 13:e0194978. [PMID: 29590220 PMCID: PMC5874065 DOI: 10.1371/journal.pone.0194978] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 03/14/2018] [Indexed: 01/04/2023] Open
Abstract
L-carnitine is important for the catabolism of long-chain fatty acids in the mitochondria. We investigated how the triacylglycerol (TAG)-lowering drug 2-(tridec-12-yn-1-ylthio)acetic acid (1-triple TTA) influenced lipid metabolism in carnitine-depleted, 3-(2,2,2-trimethylhydrazinium)propionate dehydrate (Mildronate; meldonium)-treated male Wistar rats. As indicated, carnitine biosynthesis was impaired by Mildronate. However, TAG levels of both plasma and liver were decreased by 1-triple TTA in Mildronate-treated animals. This was accompanied by increased gene expression of proteins involved in mitochondrial activity and proliferation and reduced mRNA levels of Dgat2, ApoB and ApoCIII in liver. The hepatic energy state was reduced in the group of Mildronate and 1-triple TTA as reflected by increased AMP/ATP ratio, reduced energy charge and induced gene expression of uncoupling proteins 2 and 3. The increase in mitochondrial fatty acid oxidation was observed despite low plasma carnitine levels, and was linked to strongly induced gene expression of carnitine acetyltransferase, translocase and carnitine transporter, suggesting an efficient carnitine turnover. The present data suggest that the plasma TAG-lowering effect of 1-triple TTA in Mildronate-treated rats is not only due to increased mitochondrial fatty acid oxidation reflected by increased mitochondrial biogenesis, but also to changes in plasma clearance and reduced TAG biosynthesis.
Collapse
Affiliation(s)
- Carine Lindquist
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Bodil Bjørndal
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Asbjørn Svardal
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Seth Hallström
- Institute of Physiological Chemistry, Medical University of Graz, Graz, Austria
| | - Rolf Kristian Berge
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|