101
|
Ahmadpour A, Christensen RG, Zarrin M, Farjood F, Ahmadpour A. Reporting temporal fluctuations of hepatic C16 and C18 fatty acids during late gestation and early lactation in dromedary camel. Trop Anim Health Prod 2019; 51:1651-1660. [PMID: 30864045 DOI: 10.1007/s11250-019-01860-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 02/28/2019] [Indexed: 02/02/2023]
Abstract
Based on current knowledge, C16 and C18 fatty acids (FA) are considered the most functional FA in hepatic metabolism. Although these FAs have been satisfyingly investigated in cattle, other species such as camel have been neglected. For this reason, the current study was designed to scrutinize changing patterns of C16 and C18 FAs in 10 dromedary camels from the last 2 months of gestation to the first months of lactation. Camels were grazed on natural pasture and supplemented with a balanced ration. Liver biopsies were obtained through blind biopsy technique at about 60, 45, 30, and 15-day antepartum (AP), and at 3, 15, 30, 45, and 60 post-partum (PP). Data were analyzed by the ANOVA procedure of SPSS with repeated measurements. From 15-day AP, saturated FA content of the liver declined (P < 0.01) and 15-day PP reached its peak (P = 0.02). At 30-day PP it went down (P < 0.01), and re-elevated at 45-day PP (P < 0.01) but remained at a steady state for the duration of the study. Mono-unsaturated and polyunsaturated FA content of hepatic tissue were constant throughout AP, albeit observed to peak at 15-day AP compared with 45 (P = 0.04) and 30-day AP (P < 0.01) for mono-unsaturated FAs, and with 60-, 45-, and 30-day AP (P ≤ 0.01) for polyunsaturated FAs. The palmitic acid content of the liver reached a nadir at 30-day AP (P < 0.01), increased sharply (P < 0.01) at the next sampling time-point, and had a trend to escalate until 3-day PP. Palmitoleic acid levels were unchanged from 60- to 30-day AP, decreased at 15 AP and 3-day PP, increased at 15-day PP, then remained constant until the end of the study period (P ≤ 0.04). Stearic acid content started to grow at 15-day AP and reached its peak at 15-day PP (P < 0.01). At 30-day PP, stearic level in liver dropped abruptly (P < 0.01), then intensified at 45-day PP and did not change after; hepatic content of stearic acid was lower during AP compared with PP time-points. Other C18 FAs changed significantly during the study period. These results suggest that parturition could have a profound effect on FA composition and other metabolites in camel liver. Further research is required to establish the metabolic mechanism behind these changes.
Collapse
Affiliation(s)
- Amir Ahmadpour
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, 4815 Old Main Hill, Logan, UT, 84322, USA.
| | - Rachael G Christensen
- USDA-ARS, Northern Great Plains Agricultural Research Laboratory, Mandan, ND, 58554, USA
| | - Mousa Zarrin
- Department of Animal Sciences, Yasouj University, Yasouj, I.R., Iran
| | - Farhad Farjood
- Department of Biological Engineering, Utah State University, Logan, UT, 84322, USA
| | | |
Collapse
|
102
|
Panserat S, Marandel L, Seiliez I, Skiba-Cassy S. New Insights on Intermediary Metabolism for a Better Understanding of Nutrition in Teleosts. Annu Rev Anim Biosci 2019; 7:195-220. [DOI: 10.1146/annurev-animal-020518-115250] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The rapid development of aquaculture production throughout the world over the past few decades has led to the emergence of new scientific challenges to improve fish nutrition. The diet formulations used for farmed fish have been largely modified in the past few years. However, bottlenecks still exist in being able to suppress totally marine resources (fish meal and fish oil) in diets without negatively affecting growth performance and flesh quality. A better understanding of fish metabolism and its regulation by nutrients is thus mandatory. In this review, we discuss four fields of research that are highly important for improving fish nutrition in the future: ( a) fish genome complexity and subsequent consequences for metabolism, ( b) microRNAs (miRNAs) as new actors in regulation of fish metabolism, ( c) the role of autophagy in regulation of fish metabolism, and ( d) the nutritional programming of metabolism linked to the early life of fish.
Collapse
Affiliation(s)
- S. Panserat
- INRA, University of Pau & Pays de l'Adour, E2S UPPA, UMR1419 Nutrition, Metabolisme, Aquaculture, Aquapôle, F-64310 Saint-Pée-sur-Nivelle, France
| | - L. Marandel
- INRA, University of Pau & Pays de l'Adour, E2S UPPA, UMR1419 Nutrition, Metabolisme, Aquaculture, Aquapôle, F-64310 Saint-Pée-sur-Nivelle, France
| | - I. Seiliez
- INRA, University of Pau & Pays de l'Adour, E2S UPPA, UMR1419 Nutrition, Metabolisme, Aquaculture, Aquapôle, F-64310 Saint-Pée-sur-Nivelle, France
| | - S. Skiba-Cassy
- INRA, University of Pau & Pays de l'Adour, E2S UPPA, UMR1419 Nutrition, Metabolisme, Aquaculture, Aquapôle, F-64310 Saint-Pée-sur-Nivelle, France
| |
Collapse
|
103
|
Gjorgjieva M, Mithieux G, Rajas F. Hepatic stress associated with pathologies characterized by disturbed glucose production. Cell Stress 2019; 3:86-99. [PMID: 31225503 PMCID: PMC6551742 DOI: 10.15698/cst2019.03.179] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The liver is an organ with many facets, including a role in energy production and metabolic balance, detoxification and extraordinary capacity of regeneration. Hepatic glucose production plays a crucial role in the maintenance of normal glucose levels in the organism i.e. between 0.7 to 1.1 g/l. The loss of this function leads to a rare genetic metabolic disease named glycogen storage disease type I (GSDI), characterized by severe hypoglycemia during short fasts. On the contrary, type 2 diabetes is characterized by chronic hyperglycemia, partly due to an overproduction of glucose by the liver. Indeed, diabetes is characterized by increased uptake/production of glucose by hepatocytes, leading to the activation of de novo lipogenesis and the development of a non-alcoholic fatty liver disease. In GSDI, the accumulation of glucose-6 phosphate, which cannot be hydrolyzed into glucose, leads to an increase of glycogen stores and the development of hepatic steatosis. Thus, in these pathologies, hepatocytes are subjected to cellular stress mainly induced by glucotoxicity and lipotoxicity. In this review, we have compared hepatic cellular stress induced in type 2 diabetes and GSDI, especially oxidative stress, autophagy deregulation, and ER-stress. In addition, both GSDI and diabetic patients are prone to the development of hepatocellular adenomas (HCA) that occur on a fatty liver in the absence of cirrhosis. These HCA can further acquire malignant traits and transform into hepatocellular carcinoma. This process of tumorigenesis highlights the importance of an optimal metabolic control in both GSDI and diabetic patients in order to prevent, or at least to restrain, tumorigenic activity during disturbed glucose metabolism pathologies.
Collapse
Affiliation(s)
- Monika Gjorgjieva
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France.,Université de Lyon, Lyon, F-69008 France.,Université Lyon I, Villeurbanne, F-69622 France
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France.,Université de Lyon, Lyon, F-69008 France.,Université Lyon I, Villeurbanne, F-69622 France
| | - Fabienne Rajas
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France.,Université de Lyon, Lyon, F-69008 France.,Université Lyon I, Villeurbanne, F-69622 France
| |
Collapse
|
104
|
Ke PY. Diverse Functions of Autophagy in Liver Physiology and Liver Diseases. Int J Mol Sci 2019; 20:E300. [PMID: 30642133 PMCID: PMC6358975 DOI: 10.3390/ijms20020300] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/05/2019] [Accepted: 01/08/2019] [Indexed: 01/09/2023] Open
Abstract
Autophagy is a catabolic process by which eukaryotic cells eliminate cytosolic materials through vacuole-mediated sequestration and subsequent delivery to lysosomes for degradation, thus maintaining cellular homeostasis and the integrity of organelles. Autophagy has emerged as playing a critical role in the regulation of liver physiology and the balancing of liver metabolism. Conversely, numerous recent studies have indicated that autophagy may disease-dependently participate in the pathogenesis of liver diseases, such as liver hepatitis, steatosis, fibrosis, cirrhosis, and hepatocellular carcinoma. This review summarizes the current knowledge on the functions of autophagy in hepatic metabolism and the contribution of autophagy to the pathophysiology of liver-related diseases. Moreover, the impacts of autophagy modulation on the amelioration of the development and progression of liver diseases are also discussed.
Collapse
Affiliation(s)
- Po-Yuan Ke
- Department of Biochemistry & Molecular Biology and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan.
- Division of Allergy, Immunology, and Rheumatology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan.
| |
Collapse
|
105
|
GPR120 protects lipotoxicity-induced pancreatic β-cell dysfunction through regulation of PDX1 expression and inhibition of islet inflammation. Clin Sci (Lond) 2019; 133:101-116. [PMID: 30523046 DOI: 10.1042/cs20180836] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/29/2018] [Accepted: 12/05/2018] [Indexed: 12/23/2022]
Abstract
G-protein coupled receptor 120 (GPR120) has been shown to act as an omega-3 unsaturated fatty acid sensor and is involved in insulin secretion. However, the underlying mechanism in pancreatic β cells remains unclear. To explore the potential link between GPR120 and β-cell function, its agonists docosahexaenoic acid (DHA) and GSK137647A were used in palmitic acid (PA)-induced pancreatic β-cell dysfunction, coupled with GPR120 knockdown (KD) in MIN6 cells and GPR120 knockout (KO) mice to identify the underlying signaling pathways. In vitro and ex vivo treatments of MIN6 cells and islets isolated from wild-type (WT) mice with DHA and GSK137647A restored pancreatic duodenal homeobox-1 (PDX1) expression levels and β-cell function via inhibiting PA-induced elevation of proinflammatory chemokines and activation of nuclear factor κB, c-Jun amino (N)-terminal kinases1/2 and p38MAPK signaling pathways. On the contrary, these GPR120 agonism-mediated protective effects were abolished in GPR120 KD cells and islets isolated from GPR120 KO mice. Furthermore, GPR120 KO mice displayed glucose intolerance and insulin resistance relative to WT littermates, and β-cell functional related genes were decreased while inflammation was exacerbated in islets with increased macrophages in pancreas from GPR120 KO mice. DHA and GSK137647A supplementation ameliorated glucose tolerance and insulin sensitivity, as well as improved Pdx1 expression and islet inflammation in diet-induced obese WT mice, but not in GPR120 KO mice. These findings indicate that GPR120 activation is protective against lipotoxicity-induced pancreatic β-cell dysfunction, via the mediation of PDX1 expression and inhibition of islet inflammation, and that GPR120 activation may serve as a preventative and therapeutic target for obesity and diabetes.
Collapse
|
106
|
Liangpunsakul S, Chalasani N. Lipid mediators of liver injury in nonalcoholic fatty liver disease. Am J Physiol Gastrointest Liver Physiol 2019; 316:G75-G81. [PMID: 30383414 PMCID: PMC6383373 DOI: 10.1152/ajpgi.00170.2018] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) comprises a spectrum of histopathological phenotypes ranging from simple steatosis to more severe liver disease associated with cell injury, including nonalcoholic steatohepatitis (NASH), advanced fibrosis, and cirrhosis. Only a subset of patients with NAFLD develop NASH from yet incompletely understood mechanisms. Emerging data suggest lipid species other than triglycerides as contributors to the pathogenesis of NASH. In this mini review, we focus on the recent data on the mechanisms of NASH, focusing on these lipid mediators and their potential as therapeutic targets in NASH.
Collapse
Affiliation(s)
- Suthat Liangpunsakul
- 1Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana,2Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana,3Roudebush Veterans Administration Medical Center, Indianapolis, Indiana
| | - Naga Chalasani
- 1Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
107
|
Mignolet A, Wood BR, Goormaghtigh E. Intracellular investigation on the differential effects of 4 polyphenols on MCF-7 breast cancer cells by Raman imaging. Analyst 2018; 143:258-269. [PMID: 29214243 DOI: 10.1039/c7an01460k] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The past decades have seen significant interest in the study of polyphenolic compounds as potential therapeutic agents in medicine because they display a vast array of cellular effects beneficial to treat or manage a plethora of chronic diseases including inflammatory diseases, cardiovascular abnormalities and several types of cancer. These compounds act at different stages of carcinogenesis but deciphering their mode of action is a complex task. Live MCF-7 breast cancer cells were investigated using Raman imaging to evaluate the perturbations induced after incubating cells with four different polyphenols: EGCG, gallic acid, resveratrol and tannic acid. First, clear spectral changes could be observed between the spectra of the cytoplasm and the nucleus of live MCF-7 cancer cells demonstrating a difference in their respective global chemical composition. The treatments induced significant modifications in the cells but no clear common pattern of modifications from the 4 drugs could be observed in the cell spectra in the 1800-600 cm-1 region. The high spatial resolution of Raman confocal microscopy enabled both the nucleus and cytoplasm to be independently targeted to study the impact of the polyphenols on the cell line. Positive spectral variations at 2851 cm-1 and 2920 cm-1 as well as in the 1460-1420 cm-1 and 1660-1650 cm-1 spectral regions inside cell cytoplasm reflected an increase of the lipid content after exposure to polyphenols. Lipid accumulation appears to be an early biomarker of drug-induced cell stress and subsequent apoptosis. Interestingly an increase of cytochrome c into the cytosol was also induced by EGCG. These multiple events are possibly associated with cell apoptosis. In conclusion, Raman micro-spectroscopy provides a complementary spectroscopic method to realize biological investigations on live cancer cells and to evaluate the effects of polyphenols at the subcellular level.
Collapse
Affiliation(s)
- A Mignolet
- Center for Structural Biology and Bioinformatics, Laboratory for the Structure and Function of Biological Membranes; Université Libre de Bruxelles, Campus Plaine, Bld du Triomphe 2, CP206/2, B1050 Brussels, Belgium
| | | | | |
Collapse
|
108
|
Kochan K, Kus E, Szafraniec E, Wislocka A, Chlopicki S, Baranska M. Changes induced by non-alcoholic fatty liver disease in liver sinusoidal endothelial cells and hepatocytes: spectroscopic imaging of single live cells at the subcellular level. Analyst 2018; 142:3948-3958. [PMID: 28944783 DOI: 10.1039/c7an00865a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) is the most prevalent liver disorder worldwide, involving pathogenic mechanisms of liver sinusoidal endothelial cells (LSECs), hepatocytes and other liver cells. Here, we used a novel approach of label-free Raman confocal imaging to study primary LSECs and hepatocytes freshly isolated from the livers of mice with NAFLD induced by a high fat diet (HFD), in comparison to healthy controls. Our aim was to characterize changes in the biochemical composition in LSECs and hepatocytes that occur in a single cell at the subcellular level. LSECs from NAFLD livers displayed a significant increase in the intensity of marker bands of nuclear DNA that was not associated with changes in LSEC nucleus size. A number of changes in the cytoplasm of hepatocytes were identified. However, the most prominent change in hepatocytes was a substantial increase in the degree of unsaturation of LBs' (lipid bodies) lipids in NAFLD, suggesting an increase in the de novo lipogenesis of unsaturated lipids. The confocal Raman imaging of single live cells isolated from the liver provided a unique tool to better understand disease-induced cell-specific changes in the biochemical phenotype of primary liver cells.
Collapse
Affiliation(s)
- Kamila Kochan
- Centre for Biospectroscopy and School of Chemistry, Monash University, Clayton, 3800, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
109
|
Chu KY, O'Reilly L, Mellet N, Meikle PJ, Bartley C, Biden TJ. Oleate disrupts cAMP signaling, contributing to potent stimulation of pancreatic β-cell autophagy. J Biol Chem 2018; 294:1218-1229. [PMID: 30518550 DOI: 10.1074/jbc.ra118.004833] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 11/19/2018] [Indexed: 12/12/2022] Open
Abstract
Autophagy is critical for maintaining cellular function via clearance of excess nutrients and damaged organelles. In pancreatic β-cells, it helps counter the endoplasmic reticulum (ER) stress that impairs insulin secretory capacity during Type 2 diabetes. Chronic exposure of β-cells to saturated fatty acids (FAs) such as palmitate stimulates ER stress and modulates autophagy, but the effects of unsaturated FAs such as oleate, which are also elevated during obesity, are less well understood. We therefore treated MIN6 cells and mouse islets for 8-48 h with either palmitate or oleate, and then monitored autophagic flux, signaling pathways, lysosomal biology, and phospholipid profiles. Compared with palmitate, oleate more effectively stimulated both autophagic flux and clearance of autophagosomes. The flux stimulation occurred independently of ER stress, nutrient-sensing (mTOR) and signaling pathways (protein kinases A, C, and D). Instead the mechanism involved the exchange factor directly activated by cAMP 2 (EPAC2). Oleate reduced cellular cAMP, and its effects on autophagic flux were reproduced or inhibited, respectively, by Epac2 knockdown or activation. Oleate also increased lysosomal acidity and increased phospholipid saturation, consistent with improved autophagosomal fusion with lysosomes. We conclude that a potent stimulation of autophagy might help explain the known benefits of unsaturated FAs in countering the toxicity of saturated FAs in β-cells during obesity and lipid loading.
Collapse
Affiliation(s)
- Kwan Yi Chu
- Division of Diabetes and Metabolism, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW 2052
| | - Liam O'Reilly
- Division of Diabetes and Metabolism, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010
| | - Natalie Mellet
- Baker IDI Heart and Diabetes Institute, Prahran, Victoria 3004, Australia
| | - Peter J Meikle
- Baker IDI Heart and Diabetes Institute, Prahran, Victoria 3004, Australia
| | - Clarissa Bartley
- Division of Diabetes and Metabolism, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010
| | - Trevor J Biden
- Division of Diabetes and Metabolism, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW 2052.
| |
Collapse
|
110
|
Cheng KP, Ou HY, Hung HC, Li CH, Fan KC, Wu JS, Wu HT, Chang CJ. Unsaturated Fatty Acids Increase the Expression of Hepassocin through a Signal Transducer and Activator of Transcription 3-Dependent Pathway in HepG2 Cells. Lipids 2018; 53:863-869. [DOI: 10.1002/lipd.12099] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 09/25/2018] [Accepted: 10/03/2018] [Indexed: 01/22/2023]
Affiliation(s)
- Kai-Pi Cheng
- Division of Endocrinology and Metabolism, Department of Internal Medicine; National Cheng Kung University Hospital, National Cheng Kung University, No. 138, Sheng-Li, Road, 70403; Tainan Taiwan
| | - Horng-Yih Ou
- Division of Endocrinology and Metabolism, Department of Internal Medicine; National Cheng Kung University Hospital, National Cheng Kung University, No. 138, Sheng-Li, Road, 70403; Tainan Taiwan
| | - Hao-Chang Hung
- Division of Endocrinology and Metabolism, Department of Internal Medicine; National Cheng Kung University Hospital, National Cheng Kung University, No. 138, Sheng-Li, Road, 70403; Tainan Taiwan
| | - Chung-Hao Li
- Department of Health Management Center; National Cheng Kung University Hospital, National Cheng Kung University, No. 138, Sheng-Li, Road, 70403; Tainan Taiwan
- Department of Family Medicine; National Cheng Kung University Hospital, National Cheng Kung University, No. 138, Sheng-Li, Road, 70403; Tainan Taiwan
| | - Kang-Chih Fan
- Division of Endocrinology and Metabolism, Department of Internal Medicine; National Taiwan University Hospital Hsin-Chu Branch, No. 25, Lane 442, Sec. 1, Jingguo Road, 30059; Hsinchu City Taiwan
| | - Jin-Shang Wu
- Department of Family Medicine; National Cheng Kung University Hospital, National Cheng Kung University, No. 138, Sheng-Li, Road, 70403; Tainan Taiwan
| | - Hung-Tsung Wu
- Graduate Institute of Metabolism and Obesity Sciences; Taipei Medical University, No. 250, Wuxing St., 11031; Taipei Taiwan
| | - Chih-Jen Chang
- Department of Family Medicine; National Cheng Kung University Hospital, National Cheng Kung University, No. 138, Sheng-Li, Road, 70403; Tainan Taiwan
| |
Collapse
|
111
|
Mignolet A, Mathieu V, Goormaghtigh E. HTS-FTIR spectroscopy allows the classification of polyphenols according to their differential effects on the MDA-MB-231 breast cancer cell line. Analyst 2018; 142:1244-1257. [PMID: 27924981 DOI: 10.1039/c6an02135b] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Breast cancer is a major public health issue among women in the world. Meanwhile new anticancer treatments struggle more and more to be accepted in the pharmaceutical market and research costs still increase. There is therefore a need to find new treatments and new screening methods to test them more quickly and efficiently. Among natural compounds, an increasing interest has been given to polyphenols as they can take action at the different stages of carcinogenesis, from tumour initiation to metastasis formation, by disturbing multiple cellular signalling pathways. They constitute one of the largest groups of plant metabolites and more than 8000 compounds have already been identified based on their chemical structure. Traditionally in pharmacology, new anticancer drugs are first evaluated for their potential to inhibit the proliferation of cancer cell lines. Numerous potential drugs are discarded at this stage even though they could show interesting modes of action. In turn, there is an increasing demand for more systemic approaches in order to obtain a global and accurate insight into the biochemical processes mediated by drugs. Recently, FTIR spectroscopy was demonstrated to be an innovative tool to obtain a unique fingerprint of the effects of anticancer drugs on cells in culture. While this spectral technique appears to have a definite potential to sort drugs according to their spectral fingerprints, characteristic of the metabolic modifications induced, the present challenge remains to evaluate the drug-induced spectral changes in cancer cells on a larger scale. This article presents the results obtained for a 24 h-exposure of the breast cancer cell line MDA-MB-231 to 15 compounds belonging to different classes of polyphenols using FTIR spectroscopy connected to a high throughput screening extension. Through unsupervised and supervised statistical analyses (PCA, MANOVA, Student's t-tests and HCA), a distinction between polyphenol treatments and controls could be well established.
Collapse
Affiliation(s)
- A Mignolet
- Center for Structural Biology and Bioinformatics, Laboratory for the Structure and Function of Biological Membranes; Université Libre de Bruxelles, Campus Plaine, Bld du Triomphe 2, CP206/2, B1050 Brussels, Belgium
| | | | | |
Collapse
|
112
|
Yang L, Guan G, Lei L, Lv Q, Liu S, Zhan X, Jiang Z, Gu X. Palmitic acid induces human osteoblast-like Saos-2 cell apoptosis via endoplasmic reticulum stress and autophagy. Cell Stress Chaperones 2018; 23:1283-1294. [PMID: 30194633 PMCID: PMC6237680 DOI: 10.1007/s12192-018-0936-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/27/2018] [Accepted: 09/01/2018] [Indexed: 12/30/2022] Open
Abstract
Palmitic acid (PA) is the most common saturated long-chain fatty acid in food that causes cell apoptosis. However, little is known about the molecular mechanisms of PA toxicity. In this study, we explore the effects of PA on proliferation and apoptosis in human osteoblast-like Saos-2 cells and uncover the signaling pathways involved in the process. Our study showed that endoplasmic reticulum (ER) stress and autophagy are involved in PA-induced Saos-2 cell apoptosis. We found that PA inhibited the viability of Saos-2 cells in a dose- and time-dependent manner. At the same time, PA induced the expression of ER stress marker genes (glucose-regulated protein 78 (GRP78) and CCAAT/enhancer binding protein homologous protein (CHOP)), altered autophagy-related gene expression (microtubule-associated protein 1 light chain 3 (LC3), ATG5, p62, and Beclin), promoted apoptosis-related gene expression (Caspase 3 and BAX), and affected autophagic flux. Inhibiting ER stress with 4-PBA diminished the PA-induced cell apoptosis, activated autophagy, and increased the expression of Caspase 3 and BAX. Inhibiting autophagy with 3-MA attenuated the PA and ER stress-induced cell apoptosis and the apoptosis-related gene expression (Caspase 3 and BAX), but seemed to have no obvious effects on ER stress, although the CHOP expression was downregulated. Taken together, our results suggest that PA-induced Saos-2 cell apoptosis is activated via ER stress and autophagy, and the activation of autophagy depends on the ER stress during this process.
Collapse
Affiliation(s)
- Lei Yang
- College of Basic Medical, Jiujiang University, Jiujiang, 332000, Jiangxi, China.
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, Jiangxi, China.
| | - Gaopeng Guan
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, Jiangxi, China
- Affiliated Hospital of Jiujiang University, Jiujiang University, Jiujiang, 332000, Jiangxi, China
- Medicine Graduate School, Nanchang University, Nanchang, 330006, China
| | - Lanjie Lei
- Affiliated Hospital of Jiujiang University, Jiujiang University, Jiujiang, 332000, Jiangxi, China
| | - Qizhuang Lv
- College of Biology & Pharmacy, Yulin Normal University, Yulin, 537000, Guangxi, China
| | - Shengyuan Liu
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, Jiangxi, China
- Affiliated Hospital of Jiujiang University, Jiujiang University, Jiujiang, 332000, Jiangxi, China
- Medicine Graduate School, Nanchang University, Nanchang, 330006, China
| | - Xiuwen Zhan
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, Jiangxi, China
- Affiliated Hospital of Jiujiang University, Jiujiang University, Jiujiang, 332000, Jiangxi, China
| | - Zhenzhen Jiang
- Medicine Graduate School, Nanchang University, Nanchang, 330006, China
| | - Xiang Gu
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, Jiangxi, China.
- Affiliated Hospital of Jiujiang University, Jiujiang University, Jiujiang, 332000, Jiangxi, China.
| |
Collapse
|
113
|
Li Y, Lu Z, Ru JH, Lopes-Virella MF, Lyons TJ, Huang Y. Saturated fatty acid combined with lipopolysaccharide stimulates a strong inflammatory response in hepatocytes in vivo and in vitro. Am J Physiol Endocrinol Metab 2018; 315:E745-E757. [PMID: 29989851 PMCID: PMC6293169 DOI: 10.1152/ajpendo.00015.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease and consumption of high-fat diet (HFD) is a risk factor for NAFLD. The HFD not only increases intake of saturated fatty acid (SFA) but also induces metabolic endotoxemia, an HFD-associated increase in circulating lipopolysaccharide (LPS). Although it is known that SFA or LPS promote hepatic inflammation, a hallmark of NAFLD, it remains unclear how SFA in combination with LPS stimulates host inflammatory response in hepatocytes. In this study, we performed both in vivo and in vitro experiments to investigate the effect of SFA in combination with LPS on proinflammatory gene expression in hepatocytes. Our animal study showed that feeding low-density lipoprotein-deficient mice HFD enriched with SFA and injection of low-dose LPS cooperatively stimulated IL-6 expression in livers. To understand how SFA and LPS interact to promote IL-6 expression, our in vitro studies showed that palmitic acid (PA), a major SFA, and LPS exerted synergistic effect on the expression of IL-6 in hepatocytes. Furthermore, coculture of hepatocytes with macrophages resulted in a greater IL-6 expression than culture of hepatocytes without macrophages in response to the combination of PA and LPS. Finally, we observed that LPS and PA increased ceramide production by cooperatively stimulating ceramide de novo synthesis, which played an essential role in the synergistic stimulation of proinflammatory gene expression by LPS and PA. Taken together, this study showed that SFA in combination with LPS stimulated a strong inflammatory response in hepatocytes in vivo and in vitro.
Collapse
Affiliation(s)
- Yanchun Li
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, College of Medicine Medical University of South Carolina, South Carolina
| | - Zhongyang Lu
- Ralph H. Johnson Veterans Affairs Medical Center , Charleston, South Carolina
| | - Ji Hyun Ru
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, College of Medicine Medical University of South Carolina, South Carolina
| | - Maria F Lopes-Virella
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, College of Medicine Medical University of South Carolina, South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center , Charleston, South Carolina
| | - Timothy J Lyons
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, College of Medicine Medical University of South Carolina, South Carolina
| | - Yan Huang
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, College of Medicine Medical University of South Carolina, South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center , Charleston, South Carolina
| |
Collapse
|
114
|
Hoang-Minh LB, Siebzehnrubl FA, Yang C, Suzuki-Hatano S, Dajac K, Loche T, Andrews N, Schmoll Massari M, Patel J, Amin K, Vuong A, Jimenez-Pascual A, Kubilis P, Garrett TJ, Moneypenny C, Pacak CA, Huang J, Sayour EJ, Mitchell DA, Sarkisian MR, Reynolds BA, Deleyrolle LP. Infiltrative and drug-resistant slow-cycling cells support metabolic heterogeneity in glioblastoma. EMBO J 2018; 37:embj.201798772. [PMID: 30322894 DOI: 10.15252/embj.201798772] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 08/23/2018] [Accepted: 08/24/2018] [Indexed: 01/01/2023] Open
Abstract
Metabolic reprogramming has been described in rapidly growing tumors, which are thought to mostly contain fast-cycling cells (FCCs) that have impaired mitochondrial function and rely on aerobic glycolysis. Here, we characterize the metabolic landscape of glioblastoma (GBM) and explore metabolic specificities as targetable vulnerabilities. Our studies highlight the metabolic heterogeneity in GBM, in which FCCs harness aerobic glycolysis, and slow-cycling cells (SCCs) preferentially utilize mitochondrial oxidative phosphorylation for their functions. SCCs display enhanced invasion and chemoresistance, suggesting their important role in tumor recurrence. SCCs also demonstrate increased lipid contents that are specifically metabolized under glucose-deprived conditions. Fatty acid transport in SCCs is targetable by pharmacological inhibition or genomic deletion of FABP7, both of which sensitize SCCs to metabolic stress. Furthermore, FABP7 inhibition, whether alone or in combination with glycolysis inhibition, leads to overall increased survival. Our studies reveal the existence of GBM cell subpopulations with distinct metabolic requirements and suggest that FABP7 is central to lipid metabolism in SCCs and that targeting FABP7-related metabolic pathways is a viable therapeutic strategy.
Collapse
Affiliation(s)
- Lan B Hoang-Minh
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA.,Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Florian A Siebzehnrubl
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff, UK
| | - Changlin Yang
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA.,Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Silveli Suzuki-Hatano
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Kyle Dajac
- Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Tyler Loche
- Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Nicholas Andrews
- Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Michael Schmoll Massari
- Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Jaimin Patel
- Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Krisha Amin
- Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Alvin Vuong
- Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Ana Jimenez-Pascual
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff, UK
| | - Paul Kubilis
- Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Timothy J Garrett
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Craig Moneypenny
- Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL, USA
| | - Christina A Pacak
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jianping Huang
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA.,Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Elias J Sayour
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA.,Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Duane A Mitchell
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA.,Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Matthew R Sarkisian
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA.,Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Brent A Reynolds
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA .,Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Loic P Deleyrolle
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA .,Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
115
|
Saturated fatty acids promote chondrocyte matrix remodeling through reprogramming of autophagy pathways. Nutrition 2018; 54:144-152. [DOI: 10.1016/j.nut.2018.02.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 02/21/2018] [Accepted: 02/26/2018] [Indexed: 11/20/2022]
|
116
|
Novel Approaches To Kill Toxoplasma gondii by Exploiting the Uncontrolled Uptake of Unsaturated Fatty Acids and Vulnerability to Lipid Storage Inhibition of the Parasite. Antimicrob Agents Chemother 2018; 62:AAC.00347-18. [PMID: 30061287 DOI: 10.1128/aac.00347-18] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 07/21/2018] [Indexed: 12/17/2022] Open
Abstract
Toxoplasma gondii, an obligate intracellular parasite replicating in mammalian cells within a parasitophorous vacuole (PV), is an avid scavenger of lipids retrieved from the host cell. Following lipid uptake, this parasite stores excess lipids in lipid droplets (LD). Here, we examined the lipid storage capacities of Toxoplasma upon supplementation of the culture medium with various fatty acids at physiological concentrations. Supplemental unsaturated fatty acids (oleate [OA], palmitoleate, linoleate) accumulate in large LD and impair parasite replication, whereas saturated fatty acids (palmitate, stearate) neither stimulate LD formation nor impact growth. Examination of parasite growth defects with 0.4 mM OA revealed massive lipid deposits outside LD, indicating enzymatic inadequacies for storing neutral lipids in LD in response to the copious salvage of OA. Toxoplasma exposure to 0.5 mM OA led to irreversible growth arrest and lipid-induced damage, confirming a major disconnect between fatty acid uptake and the parasite's cellular lipid requirements. The importance of neutral lipid synthesis and storage to avoid lipotoxicity was further highlighted by the selective vulnerability of Toxoplasma, both the proliferative and the encysted forms, to subtoxic concentrations of the acyl coenzyme A:diacylglycerol acyltransferase 1 (DGAT1) pharmacological inhibitor T863. T863-treated parasites did not form LD but instead built up large membranous structures within the cytoplasm, which suggests improper channeling and management of the excess lipid. Dual addition of OA and T863 to infected cells intensified the deterioration of the parasite. Overall, our data pinpoint Toxoplasma DGAT as a promising drug target for the treatment of toxoplasmosis that would not incur the risk of toxicity for mammalian cells.
Collapse
|
117
|
Zeinvand-Lorestani M, Kalantari H, Khodayar MJ, Teimoori A, Saki N, Ahangarpour A, Rahim F, Alboghobeish S. Autophagy upregulation as a possible mechanism of arsenic induced diabetes. Sci Rep 2018; 8:11960. [PMID: 30097599 PMCID: PMC6086829 DOI: 10.1038/s41598-018-30439-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 07/30/2018] [Indexed: 01/24/2023] Open
Abstract
The key features of type 2 diabetes mellitus (T2DM) caused by high fat diet (HFD) in combination with arsenic (As) exposure (pronounced glucose intolerance despite a significant decrease in insulin resistance) are different from those expected for T2DM. Autophagy has been considered as a possible link between insulin resistance and obesity. Therefore in this study, we utilized autophagy gene expression profiling via real-time RT-PCR array analysis in livers of NMRI mice exposed to an environmentally relevant and minimally cytotoxic concentration of arsenite (50 ppm) in drinking water while being fed with a HFD for 20 weeks. Out of 84 genes associated with autophagy under study, 21 genes were related to autophagy machinery components of which 13 genes were downregulated when HDF diet was applied. In this study, for the first time, it was shown that the exposure to arsenic in the livers of mice chronically fed with HFD along with increased oxidative stress resulted in the restoration of autophagy [upregulation of genes involved in the early phase of phagophore formation, phagophore expansion and autophagosome-lysosome linkage stages]. Considering the role of arsenic in the induction of autophagy; it can be argued that reduced insulin resistance in HFD - As induced diabetes may be mediated by autophagy upregulation.
Collapse
Affiliation(s)
| | - Heibatullah Kalantari
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Javad Khodayar
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Ali Teimoori
- Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Health Research Institute, Research Center of Thalassemia and Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Akram Ahangarpour
- Health Research Institute, Diabetes Research Center, Department of Physiology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fakher Rahim
- Health Research Institute, Research Center of Thalassemia and Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Soheila Alboghobeish
- Department of Pharmacology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
118
|
Lipotoxic Effects of Palmitic Acid on Astrocytes Are Associated with Autophagy Impairment. Mol Neurobiol 2018; 56:1665-1680. [DOI: 10.1007/s12035-018-1183-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/07/2018] [Indexed: 12/23/2022]
|
119
|
Zeng R, Fang Y, Zhang Y, Bai S. p62 is linked to mitophagy in oleic acid-induced adipogenesis in human adipose-derived stromal cells. Lipids Health Dis 2018; 17:133. [PMID: 29866118 PMCID: PMC5987550 DOI: 10.1186/s12944-018-0733-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 04/03/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Obesity is closely related to the abnormal differentiation of adipocytes, which are subjected to high plasma levels of free fatty acids (FFAs). As the most abundant FFA in the bloodstream, oleic acid (OA) has the ability to induce adipogenic differentiation in human adipose-derived stromal cells (hADSCs). Recently, p62, an autophagy mediator, has been shown to play a role in obesity and adipose tissue metabolism. Therefore, the aim of this study was to investigate the roles of autophagy and mitochondrial function at different stages of OA (in combination with insulin and dexamethasone)-induced adipogenesis in hADSCs. METHODS The hADSCs were incubated with OA, insulin, and dexamethasone after pretreatment with autophagy inhibitors or knockdown of p62 with shRNA. The adiposeness level was then analyzed by oil red O staining in the cells. The related proteins or mRNA levels were detected by western blot analysis or quantitative real-time polymerase chain reaction (PCR). RESULTS Treatment with 80 μM OA (substituted for isobutylmethylxantine; IBMX) for 10 days successfully induced hADSCs to adipocytes. During OA-induced adipogenesis, autophagy was induced, with an increased LC3II/I ratio on day 3 and a decreased protein level of p62 on and after day 3. Inhibition of autophagy with 3-methyladenine (3MA) at the early stage (day 0 to day 3) of differentiation, but not at the middle or late stage, significantly decreased OA-induced adipogenesis; while knockdown of p62 with shRNA significantly promoted adipogenesis in hADSCs. Moreover, the copy number of mtDNA (the ND1 gene) and the protein level of TOM20, a mitochondrial membrane protein, were increased following OA treatment, which was related to the stability of mitochondria. Interestingly, knockdown of p62 increased the mito-LC3II/I and cyto-LC3II/I ratios by 110.1% and 73.3%, respectively. The increase in the ratio of mito-LC3II/I was higher than that of cyto-LC3II/I. Furthermore, p62 knockdown-enhanced adipogenesis in hADSCs was abolished by inhibiting mitophagy with cyclosporine A. CONCLUSIONS These results suggested that p62 plays a protective role in adipogenesis of hADSCs through regulating mitophagy.
Collapse
Affiliation(s)
- Ruixia Zeng
- Department of Tissue Engineering, School of Fundamental Sciences, China Medical University, Shenyang, 110001, China
- Department of Anatomy, College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, 121001, China
- Liaoning Key Laboratory of Follicular Development and Reproductive Health, Jinzhou, 121001, China
| | - Yan Fang
- Department of Anatomy, College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, 121001, China
| | - Yibo Zhang
- Department of Pathogenic Biology, College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, 121001, China
| | - Shuling Bai
- Department of Tissue Engineering, School of Fundamental Sciences, China Medical University, Shenyang, 110001, China.
| |
Collapse
|
120
|
Impaired Fasting-Induced Adaptive Lipid Droplet Biogenesis in Liver-Specific Atg5-Deficient Mouse Liver Is Mediated by Persistent Nuclear Factor-Like 2 Activation. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1833-1846. [PMID: 29803835 DOI: 10.1016/j.ajpath.2018.04.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/08/2018] [Accepted: 04/19/2018] [Indexed: 12/22/2022]
Abstract
Lipid droplets (LDs) are intracellular organelles that store neutral lipids as energy reservoir. Recent studies suggest that autophagy is important in maintaining the homeostasis of intracellular LDs by either regulating the biogenesis of LDs, mobilization of fatty acids, or degradation of LDs in cultured cells. Increasing evidence also supports a role of autophagy in regulating glucose and lipid metabolism in vivo in mammals. In response to fasting/starvation, lipids are mobilized from the adipose tissue to the liver, which increases the number of intracellular LDs and stimulates fatty acid oxidation and ketogenesis. However, it is still controversial and unclear how impaired autophagy in hepatocytes affects the biogenesis of LDs in mouse livers. In the present study, it was demonstrated that hepatic autophagy-deficient (L-Atg)5 knockout mice had impaired adaptation to fasting-induced hepatic biogenesis of LDs. The maladaptation to fasting-induced hepatic biogenesis of LDs in L-Atg5 knockout mouse livers was not due to hepatic changes of de novo lipogenesis, secretion of very-low-density lipoprotein or fatty acid β-oxidation, but it was due to persistent nuclear factor-like 2 activation because biogenesis of LDs restored in L-Atg5/nuclear factor-like 2 double-knockout mice.
Collapse
|
121
|
Rovadoscki GA, Pertile SFN, Alvarenga AB, Cesar ASM, Pértille F, Petrini J, Franzo V, Soares WVB, Morota G, Spangler ML, Pinto LFB, Carvalho GGP, Lanna DPD, Coutinho LL, Mourão GB. Estimates of genomic heritability and genome-wide association study for fatty acids profile in Santa Inês sheep. BMC Genomics 2018; 19:375. [PMID: 29783944 PMCID: PMC5963081 DOI: 10.1186/s12864-018-4777-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 05/10/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Despite the health concerns and nutritional importance of fatty acids, there is a relative paucity of studies in the literature that report genetic or genomic parameters, especially in the case of sheep populations. To investigate the genetic architecture of fatty acid composition of sheep, we conducted genome-wide association studies (GWAS) and estimated genomic heritabilities for fatty acid profile in Longissimus dorsi muscle of 216 male sheep. RESULTS Genomic heritability estimates for fatty acid content ranged from 0.25 to 0.46, indicating that substantial genetic variation exists for the evaluated traits. Therefore, it is possible to alter fatty acid profiles through selection. Twenty-seven genomic regions of 10 adjacent SNPs associated with fatty acids composition were identified on chromosomes 1, 2, 3, 5, 8, 12, 14, 15, 16, 17, and 18, each explaining ≥0.30% of the additive genetic variance. Twenty-three genes supporting the understanding of genetic mechanisms of fat composition in sheep were identified in these regions, such as DGAT2, TRHDE, TPH2, ME1, C6, C7, UBE3D, PARP14, and MRPS30. CONCLUSIONS Estimates of genomic heritabilities and elucidating important genomic regions can contribute to a better understanding of the genetic control of fatty acid deposition and improve the selection strategies to enhance meat quality and health attributes.
Collapse
Affiliation(s)
- G A Rovadoscki
- Department of Animal Science, University of São Paulo (USP) / Luiz de Queiroz College of Agriculture (ESALQ), Av. Pádua Dias, 11, ESALQ/USP, Piracicaba, São Paulo, 13418-900, Brazil
| | - S F N Pertile
- Department of Animal Science, University of São Paulo (USP) / Luiz de Queiroz College of Agriculture (ESALQ), Av. Pádua Dias, 11, ESALQ/USP, Piracicaba, São Paulo, 13418-900, Brazil
| | - A B Alvarenga
- Department of Animal Science, University of São Paulo (USP) / Luiz de Queiroz College of Agriculture (ESALQ), Av. Pádua Dias, 11, ESALQ/USP, Piracicaba, São Paulo, 13418-900, Brazil
| | - A S M Cesar
- Department of Animal Science, University of São Paulo (USP) / Luiz de Queiroz College of Agriculture (ESALQ), Av. Pádua Dias, 11, ESALQ/USP, Piracicaba, São Paulo, 13418-900, Brazil
| | - F Pértille
- Department of Animal Science, University of São Paulo (USP) / Luiz de Queiroz College of Agriculture (ESALQ), Av. Pádua Dias, 11, ESALQ/USP, Piracicaba, São Paulo, 13418-900, Brazil
| | - J Petrini
- Department of Animal Science, University of São Paulo (USP) / Luiz de Queiroz College of Agriculture (ESALQ), Av. Pádua Dias, 11, ESALQ/USP, Piracicaba, São Paulo, 13418-900, Brazil
| | - V Franzo
- Department of Animal Science, University of São Paulo (USP) / Luiz de Queiroz College of Agriculture (ESALQ), Av. Pádua Dias, 11, ESALQ/USP, Piracicaba, São Paulo, 13418-900, Brazil
| | - W V B Soares
- Institute of Zootechny (IZ), Nova Odessa, SP, Brazil
| | - G Morota
- Department of Animal Science, University of Nebraska, Lincoln, NE, USA
| | - M L Spangler
- Department of Animal Science, University of Nebraska, Lincoln, NE, USA
| | - L F B Pinto
- Department of Animal Science, Federal University of Bahia (UFBA), Salvador, BA, Brazil
| | - G G P Carvalho
- Department of Animal Science, Federal University of Bahia (UFBA), Salvador, BA, Brazil
| | - D P D Lanna
- Department of Animal Science, University of São Paulo (USP) / Luiz de Queiroz College of Agriculture (ESALQ), Av. Pádua Dias, 11, ESALQ/USP, Piracicaba, São Paulo, 13418-900, Brazil
| | - L L Coutinho
- Department of Animal Science, University of São Paulo (USP) / Luiz de Queiroz College of Agriculture (ESALQ), Av. Pádua Dias, 11, ESALQ/USP, Piracicaba, São Paulo, 13418-900, Brazil
| | - G B Mourão
- Department of Animal Science, University of São Paulo (USP) / Luiz de Queiroz College of Agriculture (ESALQ), Av. Pádua Dias, 11, ESALQ/USP, Piracicaba, São Paulo, 13418-900, Brazil.
| |
Collapse
|
122
|
Nakagawa S, Ueno T, Manabe T, Kawasaki K. Imidazolines increase the levels of the autophagosomal marker LC3-II in macrophage-like RAW264.7 cells. Can J Physiol Pharmacol 2018; 96:845-849. [PMID: 29633624 DOI: 10.1139/cjpp-2018-0021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study evaluated whether imidazolines can induce autophagy in the murine macrophage-like cell line RAW264.7. Idazoxan increased the content of LC3-II, an autophagosomal marker, in RAW264.7 cells. To determine whether this effect was due to the induction of its synthesis or inhibition of its degradation, idazoxan treatment was performed in the presence of bafilomycin A1, which blocks autophagosome-lysosome fusion, as well as Pepstatin A and E-64d, both of which block protein degradation in autolysosomes. An increased content of LC3-II was observed in the presence of bafilomycin A1 as well as the protease inhibitors. Furthermore, an increased number of autophagosomes was observed following idazoxan treatment using an autophagosome-specific dye. This indicated that idazoxan induced autophagy. Other imidazolines, such as efaroxan, clonidine, and 2-(2-benzofuranyl)-2-imidazoline, also increased the LC3-II content in RAW264.7 cells in the presence of bafilomycin A1. Taken together, these results indicate that some imidazolines, including idazoxan, can induce autophagy in RAW264.7 cells.
Collapse
Affiliation(s)
- Shiori Nakagawa
- Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto 610-0395, Japan.,Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto 610-0395, Japan
| | - Takayuki Ueno
- Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto 610-0395, Japan.,Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto 610-0395, Japan
| | - Takayuki Manabe
- Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto 610-0395, Japan.,Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto 610-0395, Japan
| | - Kiyoshi Kawasaki
- Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto 610-0395, Japan.,Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto 610-0395, Japan
| |
Collapse
|
123
|
Zhang Y, Hao J, Zeng J, Li Q, Rao E, Sun Y, Liu L, Mandal A, Landers VD, Morris RJ, Cleary MP, Suttles J, Li B. Epidermal FABP Prevents Chemical-Induced Skin Tumorigenesis by Regulation of TPA-Induced IFN/p53/SOX2 Pathway in Keratinocytes. J Invest Dermatol 2018; 138:1925-1934. [PMID: 29559340 DOI: 10.1016/j.jid.2018.02.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/06/2018] [Accepted: 02/26/2018] [Indexed: 12/15/2022]
Abstract
Skin lipids (e.g., fatty acids) are essential for normal skin functions. Epidermal FABP (E-FABP) is the predominant FABP expressed in skin epidermis. However, the role of E-FABP in skin homeostasis and pathology remains largely unknown. Herein, we utilized the 7,12-dimethylbenz(a)anthracene and 12-O-tetradecanolyphorbol-13-acetate-induced skin tumorigenesis model to assess the role of E-FABP in chemical-induced skin tumorigenesis. Compared to their wild-type littermates, mice deficient in E-FABP, but not adipose FABP, developed more skin tumors with higher incidence. 12-O-tetradecanolyphorbol-13-acetate functioning as a tumor promoter induced E-FABP expression and initiated extensive flaring inflammation in skin. Interestingly, 12-O-tetradecanolyphorbol-13-acetate -induced production of IFN-β and IFN-λ in the skin tissue was dependent on E-FABP expression. Further protein and gene expression arrays demonstrated that E-FABP was critical in enhancing IFN-induced p53 responses and in suppressing SOX2 expression in keratinocytes. Thus, E-FABP expression in skin suppresses chemical-induced skin tumorigenesis through regulation of IFN/p53/SOX2 pathway. Collectively, our data suggest an unknown function of E-FABP in prevention of skin tumor development, and offer E-FABP as a therapeutic target for improving skin innate immunity in chemical-induced skin tumor prevention.
Collapse
Affiliation(s)
- Yuwen Zhang
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - Jiaqing Hao
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - Jun Zeng
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA; School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Qiang Li
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Enyu Rao
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - Yanwen Sun
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - Lianliang Liu
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - Anita Mandal
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - V Douglas Landers
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - Rebecca J Morris
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Margot P Cleary
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Jill Suttles
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - Bing Li
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA.
| |
Collapse
|
124
|
Palomer X, Pizarro-Delgado J, Barroso E, Vázquez-Carrera M. Palmitic and Oleic Acid: The Yin and Yang of Fatty Acids in Type 2 Diabetes Mellitus. Trends Endocrinol Metab 2018; 29:178-190. [PMID: 29290500 DOI: 10.1016/j.tem.2017.11.009] [Citation(s) in RCA: 331] [Impact Index Per Article: 55.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 11/22/2017] [Accepted: 11/30/2017] [Indexed: 12/20/2022]
Abstract
Increased plasma non-esterified fatty acids (NEFAs) link obesity with insulin resistance and type 2 diabetes mellitus (T2DM). However, in contrast to the saturated FA (SFA) palmitic acid, the monounsaturated FA (MUFA) oleic acid elicits beneficial effects on insulin sensitivity, and the dietary palmitic acid:oleic acid ratio impacts diabetes risk in humans. Here we review recent mechanistic insights into the beneficial effects of oleic acid compared with palmitic acid on insulin resistance and T2DM, including its anti-inflammatory actions, and its capacity to inhibit endoplasmic reticulum (ER) stress, prevent attenuation of the insulin signaling pathway, and improve β cell survival. Understanding the molecular mechanisms of the antidiabetic effects of oleic acid may contribute to understanding the benefits of this FA in the prevention or delay of T2DM.
Collapse
Affiliation(s)
- Xavier Palomer
- Department of Pharmacology, Toxicology, and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Institute of Biomedicine of the University of Barcelona (IBUB), Pediatric Research Institute-Hospital Sant Joan de Déu, and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Avinguda Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Javier Pizarro-Delgado
- Department of Pharmacology, Toxicology, and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Institute of Biomedicine of the University of Barcelona (IBUB), Pediatric Research Institute-Hospital Sant Joan de Déu, and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Avinguda Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Emma Barroso
- Department of Pharmacology, Toxicology, and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Institute of Biomedicine of the University of Barcelona (IBUB), Pediatric Research Institute-Hospital Sant Joan de Déu, and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Avinguda Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology, and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Institute of Biomedicine of the University of Barcelona (IBUB), Pediatric Research Institute-Hospital Sant Joan de Déu, and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Avinguda Joan XXIII 27-31, E-08028 Barcelona, Spain.
| |
Collapse
|
125
|
Zhu Q, Yang J, Zhu R, Jiang X, Li W, He S, Jin J. Dihydroceramide-desaturase-1-mediated caspase 9 activation through ceramide plays a pivotal role in palmitic acid-induced HepG2 cell apoptosis. Apoptosis 2018; 21:1033-44. [PMID: 27364952 DOI: 10.1007/s10495-016-1267-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this study, results showed that the inhibition of PA-induced HepG2 cell growth takes place in a time- and concentration-dependent manner, that activation of caspase 9 is necessary for PA-induced HepG2 cell apoptosis, that dihydroceramide desaturase 1 (DES1) plays a key role in PA-mediated caspase 9 and caspase 3 activation, and that palmitoleic acid (POA), an omega-7 monounsaturated fatty acid, reverses PA-induced apoptosis through DES1 → Ceramide → Caspase 9 → Caspase 3 signaling.
Collapse
Affiliation(s)
- Qun Zhu
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, 210011, Nanjing, Jiangsu, People's Republic of China
- China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China
| | - Jianjun Yang
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China
| | - Rongping Zhu
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China
| | - Xin Jiang
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China
| | - Wanlian Li
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China
| | - Songqing He
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China.
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China.
| | - Junfei Jin
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China.
- China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China.
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin, 541001, Guangxi, People's Republic of China.
| |
Collapse
|
126
|
Namkoong S, Cho CS, Semple I, Lee JH. Autophagy Dysregulation and Obesity-Associated Pathologies. Mol Cells 2018; 41:3-10. [PMID: 29370691 PMCID: PMC5792710 DOI: 10.14348/molcells.2018.2213] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/02/2017] [Accepted: 10/10/2017] [Indexed: 12/20/2022] Open
Abstract
Autophagy is one of the major degradative mechanisms that can eliminate excessive nutrients, toxic protein aggregates, damaged organelles and invading microorganisms. In response to obesity and obesity-associated lipotoxic, proteotoxic and oxidative stresses, autophagy plays an essential role in maintaining physiological homeostasis. However, obesity and its associated stress insults can often interfere with the autophagic process through various mechanisms, which result in further aggravation of obesity-related metabolic pathologies in multiple metabolic organs. Paradoxically, inhibition of autophagy, within specific contexts, indirectly produces beneficial effects that can alleviate several detrimental consequences of obesity. In this minireview, we will provide a brief discussion about our current understanding of the impact of obesity on autophagy and the role of autophagy dysregulation in modulating obesity-associated pathological outcomes.
Collapse
Affiliation(s)
- Sim Namkoong
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109-2200,
USA
| | - Chun-Seok Cho
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109-2200,
USA
| | - Ian Semple
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109-2200,
USA
| | - Jun Hee Lee
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109-2200,
USA
- Institute of Gerontology, University of Michigan, Ann Arbor, Michigan 48109-2200,
USA
| |
Collapse
|
127
|
Cheng C, Deng X, Xu K. Increased expression of sterol regulatory element binding protein‑2 alleviates autophagic dysfunction in NAFLD. Int J Mol Med 2018; 41:1877-1886. [PMID: 29336468 PMCID: PMC5810215 DOI: 10.3892/ijmm.2018.3389] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 01/10/2018] [Indexed: 12/25/2022] Open
Abstract
Sterol regulatory element binding protein-2 (SREBP-2) is an important transcription factor in lipid homeostasis. A previous study showed that SREBP-2 also activated autophagic genes during cell-sterol depletion. Alterations in autophagy are reported to be involved in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). However, whether the regulation of SREBP-2 restores dysfunctional autophagy in hepatocytes during NAFLD remains to be elucidated. In the present study, a steatosis model was established with palmitic acid (PA) treatment at the indicated times and concentrations. Autophagosomes in hepatocytes were visualized by confocal microscopy after transfection with a tandem GFP-mCherry-LC3 construct. Autophagy-associated protein levels were analyzed by western blot analysis. Loss- and gain-of-function studies were performed to examine the role of SREBP-2 in the regulation of hepatocyte autophagy. It was demonstrated that PA induced autophagy and enhanced autophagic flux at the early stage, whereas prolonged treatment with PA resulted in dysfunction of autophagy in the PA-induced steatotic hepatocytes. In addition, different cellular models presented with differing dysfunctional autophagy in response to fatty acid overload. It was also confirmed that SREBP-2 regulated autophagy-related gene expression in hepatocytes, and it was shown that the overexpression of SREBP-2 increased the expression of autophagy-related genes, but did not affect the inhibition of the autophagic flux in lipid-overloaded HL-7702 cells. By contrast, increased SREBP-2 partly restored the inhibited autophagic activity in lipid-overloaded hepatoma HepG2 cells. Taken together, the present study demonstrated that autophagic function was impaired in lipid-overloaded human hepatocytes, and the differential effect of PA on autophagy was associated with the duration of PA and the cell type. Under these conditions, the overexpression of SREBP-2 alleviated the inhibited autophagic activity rather than the inhibition of autophagic flux. Consequently, the results indicated that restoration of autophagy dysfunction via the regulation of SREBP-2 may be a potential therapeutic target for the treatment of NAFLD.
Collapse
Affiliation(s)
- Chunwei Cheng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiaoling Deng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Keshu Xu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
128
|
Huang CH, Huang CY, Huang MH. Unsaturated Squalene Content in Emulsion Vaccine Adjuvants Plays a Crucial Role in ROS-Mediated Antigen Uptake and Cellular Immunity. Mol Pharm 2017; 15:420-429. [PMID: 29237267 DOI: 10.1021/acs.molpharmaceut.7b00800] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Emulsion-based adjuvants have been demonstrated to be an effective tool in increasing vaccine efficacy. Here, we aimed to launch a mechanistic study on how emulsion adjuvants interact with immune cells and to elucidate the roles of the core oil in vaccine immunogenicity. Our results showed that treatment of dendritic cells (DCs) and splenocytes with a squalene-based emulsion (referred as SqE) induced reactive oxidative species (ROS) production and resulted in an increase in apoptotic and necrotic cells in a concentration- and time-dependent manner. Furthermore, DCs cocultured with cellular debris of SqE-pretreated splenocytes resulted in a higher level of ovalbumin (OVA) antigen uptake by DCs than those cocultured with untreated splenocytes. Interestingly, the potency was rather attenuated when splenocytes were pretreated with N-acetyl-cysteine, an antioxidant. Notably, SqE possesses a high impact on eliciting ROS-mediated antigen uptake compared with a squalane-based emulsion (SqA). Concordantly, immunogenicity studies have shown that SqE is better able than SqA to activate antigen-presenting cells, and to enhance antigen-specific T-cell immunity. Taken together, our results show that unsaturated squalene oil cored within emulsions plays a crucial role in ROS-mediated antigen uptake and cellular immunity, providing a basis for the design and development of vaccine adjuvant.
Collapse
Affiliation(s)
- Chung-Hsiung Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes , 35053 Miaoli, Taiwan
| | - Chiung-Yi Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes , 35053 Miaoli, Taiwan
| | - Ming-Hsi Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes , 35053 Miaoli, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University , 40402 Taichung, Taiwan
| |
Collapse
|
129
|
Hong YJ, Ahn HJ, Shin J, Lee JH, Kim JH, Park HW, Lee SK. Unsaturated fatty acids protect trophoblast cells from saturated fatty acid-induced autophagy defects. J Reprod Immunol 2017; 125:56-63. [PMID: 29253794 DOI: 10.1016/j.jri.2017.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 11/16/2017] [Accepted: 12/11/2017] [Indexed: 01/22/2023]
Abstract
Dysregulated serum fatty acids are associated with a lipotoxic placental environment, which contributes to increased pregnancy complications via altered trophoblast invasion. However, the role of saturated and unsaturated fatty acids in trophoblastic autophagy has yet to be explored. Here, we demonstrated that prolonged exposure of saturated fatty acids interferes with the invasiveness of human extravillous trophoblasts. Saturated fatty acids (but not unsaturated fatty acids) inhibited the fusion of autophagosomes and lysosomes, resulting in the formation of intracellular protein aggregates. Furthermore, when the trophoblast cells were exposed to saturated fatty acids, unsaturated fatty acids counteracted the effects of saturated fatty acids by increasing degradation of autophagic vacuoles. Saturated fatty acids reduced the levels of the matrix metalloproteinases (MMP)-2 and MMP-9, while unsaturated fatty acids maintained their levels. In conclusion, saturated fatty acids induced decreased trophoblast invasion, of which autophagy dysfunction plays a major role.
Collapse
Affiliation(s)
- Ye-Ji Hong
- Department of Obstetrics and Gynecology, Myunggok Medical Research Institute, Konyang University Hospital, Daejeon 35365, Korea
| | - Hyo-Ju Ahn
- Department of Cell Biology, Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon 35365, Korea
| | - Jongdae Shin
- Department of Cell Biology, Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon 35365, Korea
| | - Joon H Lee
- Department of Cell Biology, Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon 35365, Korea; Myunggok Eye Research Institute, Kim's Eye Hospital, Seoul 07301, Korea
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center (SRC), Konkuk University, Korea
| | - Hwan-Woo Park
- Department of Cell Biology, Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Sung Ki Lee
- Department of Obstetrics and Gynecology, Myunggok Medical Research Institute, Konyang University Hospital, Daejeon 35365, Korea.
| |
Collapse
|
130
|
Nolan SJ, Fu MS, Coppens I, Casadevall A. Lipids Affect the Cryptococcus neoformans-Macrophage Interaction and Promote Nonlytic Exocytosis. Infect Immun 2017; 85:e00564-17. [PMID: 28947642 PMCID: PMC5695111 DOI: 10.1128/iai.00564-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 09/15/2017] [Indexed: 12/19/2022] Open
Abstract
Many microbes exploit host cellular lipid droplets during the host-microbe interaction, but this phenomenon has not been extensively studied for fungal pathogens. In this study, we analyzed the role of lipid droplets during the interaction of Cryptococcus neoformans with macrophages in the presence and the absence of exogenous lipids, in particular, oleate. The addition of oleic acid increased the frequency of lipid droplets in both C. neoformans and macrophages. C. neoformans responded to oleic acid supplementation by faster growth inside and outside macrophages. Fungal cells were able to harvest lipids from macrophage lipid droplets. Supplementation of C. neoformans and macrophages with oleic acid significantly increased the rate of nonlytic exocytosis while having no effect on lytic exocytosis. The process for lipid modulation of nonlytic exocytosis was associated with actin changes in macrophages. In summary, C. neoformans harvests lipids from macrophages, and the C. neoformans-macrophage interaction is modulated by exogenous lipids, providing a new tool for studying nonlytic exocytosis.
Collapse
Affiliation(s)
- Sabrina J Nolan
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Man Shun Fu
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
131
|
Wang ME, Singh BK, Hsu MC, Huang C, Yen PM, Wu LS, Jong DS, Chiu CH. Increasing Dietary Medium-Chain Fatty Acid Ratio Mitigates High-fat Diet-Induced Non-Alcoholic Steatohepatitis by Regulating Autophagy. Sci Rep 2017; 7:13999. [PMID: 29070903 PMCID: PMC5656678 DOI: 10.1038/s41598-017-14376-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/09/2017] [Indexed: 12/12/2022] Open
Abstract
Previous studies have demonstrated that saturated fatty acids (SFAs) are more lipotoxic than unsaturated fatty acids (UFAs) in inhibiting hepatic autophagy and promoting non-alcoholic steatohepatitis (NASH). However, there have been few studies have investigated the effects of carbon chain length on SFA-induced autophagy impairment and lipotoxicity. To investigate whether SFAs with shorter carbon chain lengths have differential effects on hepatic autophagy and NASH development, we partially replaced lard with coconut oil to elevate the ratio of medium-chain fatty acids (MCFAs) to long-chain fatty acids (LCFAs) in a mouse high-fat diet (HFD) and fed mice for 16 weeks. In addition, we treated HepG2 cells with different combinations of fatty acids to study the mechanisms of MCFAs-mediated hepatic protections. Our results showed that increasing dietary MCFA/LCFA ratio mitigated HFD-induced Type 2 diabetes and NASH in mice. Importantly, we demonstrated that increased MCFA ratio exerted its protective effects by restoring Rubicon-suppressed autophagy. Our study suggests that the relative amount of LCFAs and MCFAs in the diet, in addition to the amount of SFAs, can significantly contribute to autophagy impairment and hepatic lipotoxicity. Collectively, we propose that increasing dietary MCFAs could be an alternative therapeutic and prevention strategy for Type 2 diabetes and NASH.
Collapse
Affiliation(s)
- Mu-En Wang
- Laboratory of Animal Physiology, Department of Animal Science and Technology, National Taiwan University, Taipei, 10617, Taiwan.,Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, 16987, Singapore
| | - Brijesh K Singh
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, 16987, Singapore
| | - Meng-Chieh Hsu
- Laboratory of Animal Physiology, Department of Animal Science and Technology, National Taiwan University, Taipei, 10617, Taiwan
| | - Chien Huang
- Laboratory of Animal Physiology, Department of Animal Science and Technology, National Taiwan University, Taipei, 10617, Taiwan
| | - Paul M Yen
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, 16987, Singapore
| | - Leang-Shin Wu
- Laboratory of Animal Physiology, Department of Animal Science and Technology, National Taiwan University, Taipei, 10617, Taiwan
| | - De-Shien Jong
- Laboratory of Animal Physiology, Department of Animal Science and Technology, National Taiwan University, Taipei, 10617, Taiwan
| | - Chih-Hsien Chiu
- Laboratory of Animal Physiology, Department of Animal Science and Technology, National Taiwan University, Taipei, 10617, Taiwan.
| |
Collapse
|
132
|
Abstract
Enlarged fat cells in obese adipose tissue diminish capacity to store fat and are resistant to the anti-lipolytic effect of insulin. Insulin resistance (IR)-associated S-nitrosylation of insulin-signaling proteins increases in obesity. In accordance with the inhibition of insulin-mediated anti-lipolytic action, plasma free fatty acid (FFA) levels increase. Additionally, endoplasmic reticulum stress stimuli induce lipolysis by activating cyclic adenosine monophosphate/Protein kinase A (cAMP/PKA) and extracellular signal-regulated kinase ½ (ERK1/2) signaling in adipocytes. Failure of packaging of excess lipid into lipid droplets causes chronic elevation of circulating fatty acids, which can reach to toxic levels within non-adipose tissues. Deleterious effects of lipid accumulation in non-adipose tissues are known as lipotoxicity. In fact, triglycerides may also serve a storage function for long-chain non-esterified fatty acids and their products such as ceramides and diacylglycerols (DAGs). Thus, excess DAG, ceramide and saturated fatty acids in obesity can induce chronic inflammation and have harmful effect on multiple organs and systems. In this context, chronic adipose tissue inflammation, mitochondrial dysfunction and IR have been discussed within the scope of lipotoxicity.
Collapse
|
133
|
Wallstab C, Eleftheriadou D, Schulz T, Damm G, Seehofer D, Borlak J, Holzhütter HG, Berndt N. A unifying mathematical model of lipid droplet metabolism reveals key molecular players in the development of hepatic steatosis. FEBS J 2017; 284:3245-3261. [PMID: 28763157 DOI: 10.1111/febs.14189] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 06/02/2017] [Accepted: 07/28/2017] [Indexed: 12/16/2022]
Abstract
The liver responds to elevated plasma concentrations of free fatty acids (FFAs) with an enhanced uptake of FFAs and their esterification to triacylglycerol (TAG). On the long term, this may result in massive hepatic TAG accumulation called steatosis hepatitis. In hepatocytes, the poor water-soluble TAG is packed in specialized organelles: Lipid droplets (LDs) serving as transient cellular deposit and lipoproteins (LPs) transporting TAG and cholesterol esters to extra-hepatic tissues. The dynamics of these organelles is controlled by a variety of regulatory surface proteins (RSPs). Assembly and export of VLDLs are mainly regulated by the microsomal transfer protein (MTP) and apoprotein B100. Formation and lipolysis of LDs are regulated by several RSPs. The best studied regulators belong to the PAT (Perilipin/Adipophilin/TIP47) and CIDE families. Knockdown or overexpression of SRPs may significantly affect the total number and size distribution of LDs. Intriguingly, a large cell-to-cell heterogeneity with respect to the number and size of LDs has been found in various cell types including hepatocytes. These findings suggest that the extent of cellular lipid accumulation is determined not only by the imbalance between lipid supply and utilization but also by variations in the expression of RSPs and metabolic enzymes. To better understand the relative regulatory impact of individual processes involved in the cellular TAG turnover, we developed a comprehensive kinetic model encompassing the pathways of the fatty acid and triglyceride metabolism and the main molecular processes governing the dynamics of LDs. The model was parametrized such that a large number of experimental in vitro and in vivo findings are correctly recapitulated. A control analysis of the model revealed that variations in the activity of FFA uptake, diacylglycerol acyltransferase (DGAT) 2, and adipose triglyceride lipase (ATGL) have the strongest influence on the cellular TAG level. We used the model to simulate LD size distributions in human hepatoma cells and hepatocytes exposed to a challenge with FFAs. A random fold change by a factor of about two in the activity of RSPs was sufficient to reproduce the large diversity of droplet size distributions observed in individual cells. Under the premise that the same extent of variability of RSPs holds for the intact organ, our model predicts variations in the TAG content of individual hepatocytes by a factor of about 3-6 depending on the nutritional regime. Taken together, our modeling approach integrates numerous experimental findings on individual processes in the cellular TAG metabolism and LD dynamics metabolism to a consistent state-of-the-art dynamic network model that can be used to study how changes in the external conditions or systemic parameters will affect the TAG content of hepatocytes.
Collapse
Affiliation(s)
- Christin Wallstab
- Institute of Biochemistry, Computational Systems Biochemistry Group, Charite - University Medicine Berlin, Germany
| | - Dimitra Eleftheriadou
- Institute of Biochemistry, Computational Systems Biochemistry Group, Charite - University Medicine Berlin, Germany
| | - Theresa Schulz
- Clinic for General-, Visceral- and Transplantation Surgery, Charite - University Medicine Berlin, Germany
| | - Georg Damm
- Clinic for General-, Visceral- and Transplantation Surgery, Charite - University Medicine Berlin, Germany.,Department of Hepatobiliary Surgery and Visceral Transplantation, University of Leipzig, Germany
| | - Daniel Seehofer
- Clinic for General-, Visceral- and Transplantation Surgery, Charite - University Medicine Berlin, Germany.,Department of Hepatobiliary Surgery and Visceral Transplantation, University of Leipzig, Germany
| | - Jürgen Borlak
- Centre for Pharmacology and Toxicology, Institute for Pharmaco- and Toxicogenomics, Hannover Medical School, Hannover, Germany
| | - Hermann-Georg Holzhütter
- Institute of Biochemistry, Computational Systems Biochemistry Group, Charite - University Medicine Berlin, Germany
| | - Nikolaus Berndt
- Institute of Biochemistry, Computational Systems Biochemistry Group, Charite - University Medicine Berlin, Germany
| |
Collapse
|
134
|
Wu D, Qiao K, Feng M, Fu Y, Cai J, Deng Y, Tachibana H, Cheng X. Apoptosis of Acanthamoeba castellanii Trophozoites Induced by Oleic Acid. J Eukaryot Microbiol 2017; 65:191-199. [PMID: 28787535 DOI: 10.1111/jeu.12454] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 08/02/2017] [Accepted: 08/02/2017] [Indexed: 12/20/2022]
Abstract
Acanthamoeba spp. can be parasitic in certain situations and are responsible for serious human infections, including Acanthamoeba keratitis, granulomatous amoebic encephalitis, and cutaneous acanthamoebiasis. We analyzed the fatty acid composition of Acanthamoeba castellanii trophozoites and tested the inhibitory activity of the main fatty acids, oleic acid and arachidonic acid, in vitro. Oleic acid markedly inhibited the growth of A. castellanii, with trophozoite viability of 57.4% at a concentration of 200 μM. Caspase-3 staining and annexin V assays showed that apoptotic death occurred in A. castellanii trophozoites. Quantitative PCR and dot blot analysis showed increased levels of metacaspase and interleukin-1β converting enzyme, which is also an indication of apoptosis. In contrast, arachidonic acid showed negligible inhibition of growth of A. castellanii trophozoites. Stimulated expression of Atg3, Atg8 and LC3A/B genes and monodansylcadaverine labeling suggested that oleic acid induces apoptosis by triggering autophagy of trophozoites.
Collapse
Affiliation(s)
- Duo Wu
- Department of Medical Microbiology and Parasitology, Fudan University School of Medicine, Shanghai, 200032, China
| | - Ke Qiao
- Department of Medical Microbiology and Parasitology, Fudan University School of Medicine, Shanghai, 200032, China
| | - Meng Feng
- Department of Medical Microbiology and Parasitology, Fudan University School of Medicine, Shanghai, 200032, China
| | - Yongfeng Fu
- Department of Medical Microbiology and Parasitology, Fudan University School of Medicine, Shanghai, 200032, China
| | - Junlong Cai
- Department of Medical Microbiology and Parasitology, Fudan University School of Medicine, Shanghai, 200032, China
| | - Yihong Deng
- Department of Medical Microbiology and Parasitology, Fudan University School of Medicine, Shanghai, 200032, China
| | - Hiroshi Tachibana
- Department of Infectious Diseases, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| | - Xunjia Cheng
- Department of Medical Microbiology and Parasitology, Fudan University School of Medicine, Shanghai, 200032, China
| |
Collapse
|
135
|
Li Y, Zhao F, Wu Q, Li M, Zhu Y, Song S, Zhu J, Ma Y, Li H, Shi X, Xu X, Zhou G, Li C. Fish oil diet may reduce inflammatory levels in the liver of middle-aged rats. Sci Rep 2017; 7:6241. [PMID: 28740245 PMCID: PMC5524965 DOI: 10.1038/s41598-017-06506-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 06/12/2017] [Indexed: 01/23/2023] Open
Abstract
The impact of dietary soybean oil, lard and fish oil on physiological responses in middle age is little studied. In this study, we investigated the changes of oxidative stress, inflammatory cytokines, telomere length, and age-related gene expression in the liver of middle-aged rats in response to the above three fat diets. Male Sprague Dawley rats (12 months old) were fed AIN-93M diets for 3 months, in which soybean oil was equivalently replaced by lard or fish oil. As compared to the lard diet, intake of fish oil diet significantly decreased body weight gain, white blood cell count, and levels of hepatic triacylglycerol, total cholesterol, fat accumulation, low-density lipoprotein, oxidative stress and inflammatory cytokines (P < 0.05), but increased telomere length (P < 0.05). On the other hand, lard diet and soybean oil diet showed great similarity in the above variables. PCR array analysis further indicated that fish oil diet significantly down-regulated gene expression related to inflammatory response, apoptosis, DNA binding, proteostasis and telomere attrition. Differentially expressed genes were enriched in the complement and coagulation cascades pathways. Such physiological and molecular responses could be due to different fatty acid composition in fish oil, lard and soybean oil.
Collapse
Affiliation(s)
- Yingqiu Li
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
- Guangxi Vocational College of Technology and Business, Nanning, 530008, Guangxi, P.R. China
| | - Fan Zhao
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - Qiayu Wu
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - Mengjie Li
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - Yingying Zhu
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - Shangxin Song
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
- School of Food Science, Nanjing Xiaozhuang University, Nanjing, 211171, P.R. China
| | - Jing Zhu
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - Yafang Ma
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - He Li
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - Xuebin Shi
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - Xinglian Xu
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - Guanghong Zhou
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China.
| | - Chunbao Li
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China.
| |
Collapse
|
136
|
Jung WH, Liu CC, Yu YL, Chang YC, Lien WY, Chao HC, Huang SY, Kuo CH, Ho HC, Chan CC. Lipophagy prevents activity-dependent neurodegeneration due to dihydroceramide accumulation in vivo. EMBO Rep 2017; 18:1150-1165. [PMID: 28507162 DOI: 10.15252/embr.201643480] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 03/30/2017] [Accepted: 04/06/2017] [Indexed: 12/18/2022] Open
Abstract
Dihydroceramide desaturases are evolutionarily conserved enzymes that convert dihydroceramide (dhCer) to ceramide (Cer). While elevated Cer levels cause neurodegenerative diseases, the neuronal activity of its direct precursor, dhCer, remains unclear. We show that knockout of the fly dhCer desaturase gene, infertile crescent (ifc), results in larval lethality with increased dhCer and decreased Cer levels. Light stimulation leads to ROS increase and apoptotic cell death in ifc-KO photoreceptors, resulting in activity-dependent neurodegeneration. Lipid-containing Atg8/LC3-positive puncta accumulate in ifc-KO photoreceptors, suggesting lipophagy activation. Further enhancing lipophagy reduces lipid droplet accumulation and rescues ifc-KO defects, indicating that lipophagy plays a protective role. Reducing dhCer synthesis prevents photoreceptor degeneration and rescues ifc-KO lethality, while supplementing downstream sphingolipids does not. These results pinpoint that dhCer accumulation is responsible for ifc-KO defects. Human dhCer desaturase rescues ifc-KO larval lethality, and rapamycin reverses defects caused by dhCer accumulation in human neuroblastoma cells, suggesting evolutionarily conserved functions. This study demonstrates a novel requirement for dhCer desaturase in neuronal maintenance in vivo and shows that lipophagy activation prevents activity-dependent degeneration caused by dhCer accumulation.
Collapse
Affiliation(s)
- Wei-Hung Jung
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chung-Chih Liu
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Lian Yu
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Chin Chang
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Yu Lien
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsi-Chun Chao
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shu-Yi Huang
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Ching-Hua Kuo
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Han-Chen Ho
- Department of Anatomy, Tzu-Chi University, Hualien, Taiwan
| | - Chih-Chiang Chan
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
137
|
Shen C, Dou X, Ma Y, Ma W, Li S, Song Z. Nicotinamide protects hepatocytes against palmitate-induced lipotoxicity via SIRT1-dependent autophagy induction. Nutr Res 2017; 40:40-47. [PMID: 28473059 DOI: 10.1016/j.nutres.2017.03.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/27/2017] [Accepted: 03/09/2017] [Indexed: 12/17/2022]
Abstract
Lipotoxicity induced by saturated fatty acids (SFAs) plays a pathological role in the development of non-alcoholic fatty liver disease (NAFLD); however, the exact mechanism remains to be clearly elucidated. Palmitate is the most abundant SFA in the circulation and major lipotoxic inducer. Accumulating evidence supports that autophagy induction is protective against palmitate-induced cell death in a variety of cell types, including hepatocytes. Nicotinamide is the amide form of nicotinic acid (vitamin B3, Niacin) and a dietary supplementation as a source of vitamin B3. We previously reported that nicotinamide endowed hepatocytes resistance to palmitate-induced ER stress via up-regulating SIRT1, with cAMP/PKA/CREB pathway activation being a fundamental mechanism. This study was undertaken to investigate the potential anti-lipotoxic effect of nicotinamide and to elucidate underlying mechanism(s). Our data demonstrated that nicotinamide supplementation protected hepatocytes against palmitate-induced cell death. Mechanistic investigations revealed that nicotinamide supplementation activated autophagy in hepatocytes. Importantly, we showed that the anti-lipotoxic property of nicotinamide was abolished by autophagy inhibitors, suggesting that autophagy induction plays a mechanistic role in nicotinamide's anti-lipotoxic effect. Furthermore, we showed that SIRT1 inhibition blunted autophagy induction in response to nicotinamide supplementation and similarly abrogated the anti-lipotoxic effect conferred by nicotinamide supplementation. In conclusion, our data suggest that nicotinamide protects against palmitate-induced hepatotoxicity via SIRT1-dependent autophagy induction and that nicotinamide supplementation may represent a therapeutic choice for NAFLD.
Collapse
Affiliation(s)
- Chen Shen
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612
| | - Xiaobing Dou
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Yue Ma
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Wang Ma
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Songtao Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, 150086, PR China
| | - Zhenyuan Song
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612; Department of Pathology, University of Illinois, Medical Center, Chicago, IL 60612.
| |
Collapse
|
138
|
Mukhopadhyay S, Schlaepfer IR, Bergman BC, Panda PK, Praharaj PP, Naik PP, Agarwal R, Bhutia SK. ATG14 facilitated lipophagy in cancer cells induce ER stress mediated mitoptosis through a ROS dependent pathway. Free Radic Biol Med 2017; 104:199-213. [PMID: 28069524 DOI: 10.1016/j.freeradbiomed.2017.01.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/01/2017] [Accepted: 01/05/2017] [Indexed: 01/13/2023]
Abstract
Understanding the dynamics of autophagy and apoptosis crosstalk in cancer progression remains a challenging task. Here, we reported how the autophagy protein ATG14 induces lipophagy-mediated mitochondrial apoptosis. The overexpression of ATG14 in HeLa cells inhibited cell viability and increased mitochondrial apoptosis and endoplasmic reticulum (ER) stress. Furthermore, inhibition of this ATG14-induced autophagy promoted apoptosis. ATG14 overexpression resulted in the accumulation of free fatty acids (FFA), with a concomitant decrease in the number of lipid droplets. Our data showed that ER stress induced by ATG14 was due to the lipophagy-mediated FFA accumulation, which resulted in ROS-dependent mitochondrial stress leading to apoptosis. Inhibition of lipophagy in HeLa-ATG14 cells enhanced the cellular viability and rescued them from lipotoxicity. Mechanistically, we found that ATG14 interacted with Ulk1 and LC3, and knock down of Ulk1 prevented the lipidation of LC3 and autophagy in HeLa-ATG14 cells. We also identified a phosphatidylethanolamine (PE) binding region in ATG14, and the addition of Ulk1 to Hela-ATG14 cells decreased the ATG14-PE interaction. Lastly, confocal microscopy studies showed that the decrease in ATG14-PE binding was concomitant with the increase in LC3 lipidation over time, confirming the importance of Ulk1 to sort PE to LC3 during ATG14 mediated lipophagy induction. In conclusion, ATG14 and Ulk1 interact to induce lipophagy resulting in FFA accumulation leading to ER stress-mediated apoptosis.
Collapse
Affiliation(s)
- Subhadip Mukhopadhyay
- Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Isabel R Schlaepfer
- Division of Medical Oncology, University of Colorado School of Medicine, United States
| | - Bryan C Bergman
- Division of Endocrinology Metabolism and Diabetes, University of Colorado School of Medicine, United States
| | - Prashanta Kumar Panda
- Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India
| | | | - Prajna Paramita Naik
- Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado University of Colorado Cancer Center, University of Colorado Denver, Aurora, CO, United States
| | - Sujit Kumar Bhutia
- Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India.
| |
Collapse
|
139
|
Rosa-Caldwell ME, Lee DE, Brown JL, Brown LA, Perry RA, Greene ES, Carvallo Chaigneau FR, Washington TA, Greene NP. Moderate physical activity promotes basal hepatic autophagy in diet-induced obese mice. Appl Physiol Nutr Metab 2017; 42:148-156. [DOI: 10.1139/apnm-2016-0280] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Obesity is a known risk factor for the development of hepatic disease; obesity-induced fatty liver can lead to inflammation, steatosis, and cirrhosis and is associated with degeneration of the mitochondria. Lifestyle interventions such as physical activity may ameliorate this condition. The purpose of this study was to investigate regulation of mitochondrial and autophagy quality control in liver following Western diet-induced obesity and voluntary physical activity. Eight-week-old C57BL/6J mice were fed a Western diet (WD) or normal chow (NC, control) for 4 weeks; afterwards, groups were divided into voluntary wheel running (VWR) or sedentary (SED) conditions for an additional 4 weeks. WD-SED animals had a median histology score of 2, whereas WD-VWR was not different from NC groups (median score 1). There was no difference in mRNA of inflammatory markers Il6 and Tnfa in WD animals. WD animals had 50% lower mitochondrial content (COX IV and Cytochrome C proteins), 50% lower Pgc1a mRNA content, and reduced content of mitochondrial fusion and fission markers. Markers of autophagy were increased in VWR animals, regardless of obesity, as measured by 50% greater LC3-II/I ratio and 40% lower p62 protein content. BNIP3 protein content was 30% less in WD animals compared with NC animals, regardless of physical activity. Diet-induced obesity results in derangements in mitochondrial quality control that appear to occur prior to the onset of hepatic inflammation. Moderate physical activity appears to enhance basal autophagy in the liver; increased autophagy may provide protection from hepatic fat accumulation.
Collapse
Affiliation(s)
- Megan E. Rosa-Caldwell
- Integrative Muscle Metabolism Laboratory, Human Performance Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR 72701, USA
| | - David E. Lee
- Integrative Muscle Metabolism Laboratory, Human Performance Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR 72701, USA
| | - Jacob L. Brown
- Integrative Muscle Metabolism Laboratory, Human Performance Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR 72701, USA
| | - Lemuel A. Brown
- Exercise Muscle Biology Laboratory, Human Performance Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR 72701, USA
| | - Richard A. Perry
- Exercise Muscle Biology Laboratory, Human Performance Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR 72701, USA
| | - Elizabeth S. Greene
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - Francisco R. Carvallo Chaigneau
- California Animal Health and Food Safety Laboratory, University of California-Davis School of Veterinary Medicine, Davis, CA 95616, USA
| | - Tyrone A. Washington
- Exercise Muscle Biology Laboratory, Human Performance Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR 72701, USA
| | - Nicholas P. Greene
- Integrative Muscle Metabolism Laboratory, Human Performance Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR 72701, USA
| |
Collapse
|
140
|
HADHA, the alpha subunit of the mitochondrial trifunctional protein, is involved in long-chain fatty acid-induced autophagy in intestinal epithelial cells. Biochem Biophys Res Commun 2017; 484:636-641. [PMID: 28153718 DOI: 10.1016/j.bbrc.2017.01.159] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 01/27/2017] [Indexed: 12/19/2022]
Abstract
Genome-wide association studies have identified autophagy-related susceptibility genes for inflammatory bowel disease (IBD); however, whether autophagy regulators can be utilized as therapeutic targets remains unclear. To identify novel microtubule-associated protein 1 light chain 3 (LC3)-interacting proteins in intestinal epithelial cells (IECs), we isolated primary IECs from green fluorescent protein (GFP)-LC3 mice. We performed immunoprecipitation with a GFP antibody and then analyzed co-immunoprecipitates by mass spectrometry. HADHA was identified as an LC3-interacting protein from primary IECs. The HADHA gene encodes the alpha subunit of the mitochondrial trifunctional protein. Given that HADHA catalyzes the last three steps of mitochondrial beta-oxidation of long-chain fatty acids, we investigated whether long-chain fatty acids induce autophagy in IECs. We found that palmitic acid induced autophagy in DLD-1, HT29, and HCT116 cells. HADHA was expressed in not only the mitochondria but also the cytosol. LC3 puncta co-localized with HADHA, which were enhanced by palmitic acid stimulation. However, LC3 puncta did not co-localize with Tom20, suggesting that HADHA was induced to associate with LC3 puncta at sites other than the mitochondria. Thus, HADHA may have extra-mitochondrial functions. Furthermore, we found that palmitic acid induced cell death in IECs, which was accelerated by bafilomycin A and chloroquine. These findings suggested that palmitic acid-induced autophagy supports the survival of IECs. Taken together, these results suggested that HADHA is involved in long-chain fatty acid-induced autophagy in IECs, thus providing new insights into the pathology of IBD and revealing novel therapeutic targets of IBD.
Collapse
|
141
|
Cheng CI, Lee YH, Chen PH, Lin YC, Chou MH, Kao YH. Free Fatty Acids Induce Autophagy and LOX-1 Upregulation in Cultured Aortic Vascular Smooth Muscle Cells. J Cell Biochem 2017; 118:1249-1261. [PMID: 28072480 DOI: 10.1002/jcb.25784] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 11/03/2016] [Indexed: 11/07/2022]
Abstract
Elevation of free fatty acids (FFAs) is known to affect microvascular function and contribute to obesity-associated insulin resistance, hypertension, and microangiopathy. Proliferative and synthetic vascular smooth muscle cells (VSMCs) increase intimal thickness and destabilize atheromatous plaques. This study aimed to investigate whether saturated palmitic acid (PA) and monounsaturated oleic acid (OA) modulate autophagy activity, cell proliferation, and vascular tissue remodeling in an aortic VSMC cell line. Exposure to PA and OA suppressed growth of VSMCs without apoptotic induction, but enhanced autophagy flux with elevation of Beclin-1, Atg5, and LC3I/II. Cotreatment with autophagy inhibitors potentiated the FFA-suppressed VSMC growth and showed differential actions of PA and OA in autophagy flux retardation. Both FFAs upregulated lectin-like oxidized low-density lipoprotein receptor 1 (LOX-1) but only OA increased LDL uptake by VSMCs. Mechanistically, FFAs induced hyperphosphorylation of Akt, ERK1/2, JNK1/2, and p38 MAPK. All pathways, except OA-activated PI3K/Akt cascade, were involved in the LOX-1 upregulation, whereas blockade of PI3K/Akt and MEK/ERK cascades ameliorated the FFA-induced growth suppression on VSMCs. Moreover, both FFAs exhibited tissue remodeling effect through increasing MMP-2 and MMP-9 expression and their gelatinolytic activities, whereas high-dose OA significantly suppressed collagen type I expression. Conversely, siRNA-mediated LOX-1 knockdown significantly attenuated the OA-induced tissue remodeling effects in VSMCs. In conclusion, OA and PA enhance autophagy flux, suppress aortic VSMC proliferation, and exhibit vascular remodeling effect, thereby leading to the loss of VSMCs and interstitial ECM in vascular walls and eventually the instability of atheromatous plaques. J. Cell. Biochem. 118: 1249-1261, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Cheng-I Cheng
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yueh-Hong Lee
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Po-Han Chen
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Yu-Chun Lin
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Ming-Huei Chou
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Kaohsiung, Taiwan
| | - Ying-Hsien Kao
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| |
Collapse
|
142
|
Zhang Y, Rao E, Zeng J, Hao J, Sun Y, Liu S, Sauter ER, Bernlohr DA, Cleary MP, Suttles J, Li B. Adipose Fatty Acid Binding Protein Promotes Saturated Fatty Acid-Induced Macrophage Cell Death through Enhancing Ceramide Production. THE JOURNAL OF IMMUNOLOGY 2016; 198:798-807. [PMID: 27920274 DOI: 10.4049/jimmunol.1601403] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/09/2016] [Indexed: 01/12/2023]
Abstract
Macrophages play a critical role in obesity-associated chronic inflammation and disorders. However, the molecular mechanisms underlying the response of macrophages to elevated fatty acids (FAs) and their contribution to metabolic inflammation in obesity remain to be fully elucidated. In this article, we report a new mechanism by which dietary FAs, in particular, saturated FAs (sFAs), are able to directly trigger macrophage cell death. We demonstrated that excess sFAs, but not unsaturated FAs, induced the production of cytotoxic ceramides (Cers) in macrophage cell lines. Most importantly, expression of adipose FA binding protein (A-FABP) in macrophages facilitated metabolism of excess sFAs for Cer synthesis. Inhibition or deficiency of A-FABP in macrophage cell lines decreased sFA-induced Cer production, thereby resulting in reduced cell death. Furthermore, we validated the role of A-FABP in promoting sFA-induced macrophage cell death with primary bone marrow-derived macrophages and high-fat diet-induced obese mice. Altogether, our data reveal that excess dietary sFAs may serve as direct triggers in induction of Cer production and macrophage cell death through elevated expression of A-FABP, thus establishing A-FABP as a new molecular sensor in triggering macrophage-associated sterile inflammation in obesity.
Collapse
Affiliation(s)
- Yuwen Zhang
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202
| | - Enyu Rao
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202
| | - Jun Zeng
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202
| | - Jiaqing Hao
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202
| | - Yanwen Sun
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202
| | - Shujun Liu
- The Hormel Institute, University of Minnesota, Austin, MN 55912
| | - Edward R Sauter
- Hartford Healthcare Cancer Institute, Hartford, CT 06103; and
| | - David A Bernlohr
- College of Biological Sciences, University of Minnesota, Minneapolis, MN 55455
| | - Margot P Cleary
- The Hormel Institute, University of Minnesota, Austin, MN 55912
| | - Jill Suttles
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202
| | - Bing Li
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202;
| |
Collapse
|
143
|
Tanaka S, Hikita H, Tatsumi T, Sakamori R, Nozaki Y, Sakane S, Shiode Y, Nakabori T, Saito Y, Hiramatsu N, Tabata K, Kawabata T, Hamasaki M, Eguchi H, Nagano H, Yoshimori T, Takehara T. Rubicon inhibits autophagy and accelerates hepatocyte apoptosis and lipid accumulation in nonalcoholic fatty liver disease in mice. Hepatology 2016; 64:1994-2014. [PMID: 27637015 DOI: 10.1002/hep.28820] [Citation(s) in RCA: 245] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 08/13/2016] [Indexed: 12/11/2022]
Abstract
UNLABELLED Nonalcoholic fatty liver disease (NAFLD) is the most prevalent liver disease worldwide. It encompasses a spectrum ranging from simple steatosis to fatty liver with hepatocellular injury, termed nonalcoholic steatohepatitis. Recent studies have demonstrated hepatic autophagy being impaired in NAFLD. In the present study, we investigated the impact of Rubicon, a Beclin1-interacting negative regulator for autophagosome-lysosome fusion, in the pathogenesis of NAFLD. In HepG2 cells, BNL-CL2 cells, and murine primary hepatocytes, Rubicon was posttranscriptionally up-regulated by supplementation with saturated fatty acid palmitate. Up-regulation of Rubicon was associated with suppression of the late stage of autophagy, as evidenced by accumulation of both LC3-II and p62 expression levels as well as decreased autophagy flux. Its blockade by small interfering RNA attenuated autophagy impairment and reduced palmitate-induced endoplasmic reticulum stress, apoptosis, and lipid accumulation. Rubicon was also up-regulated in association with autophagy impairment in livers of mice fed a high-fat diet (HFD). Hepatocyte-specific Rubicon knockout mice generated by crossing Rubicon floxed mice with albumin-Cre transgenic mice did not produce any phenotypes on a normal diet. In contrast, on an HFD, they displayed significant improvement of both liver steatosis and injury as well as attenuation of both endoplasmic reticulum stress and autophagy impairment in the liver. In humans, liver tissues obtained from patients with NAFLD expressed significantly higher levels of Rubicon than those without steatosis. CONCLUSION Rubicon is overexpressed and plays a pathogenic role in NAFLD by accelerating hepatocellular lipoapoptosis and lipid accumulation, as well as inhibiting autophagy. Rubicon may be a novel therapeutic target for regulating NAFLD development and progression. (Hepatology 2016;64:1994-2014).
Collapse
Affiliation(s)
- Satoshi Tanaka
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hayato Hikita
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomohide Tatsumi
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ryotaro Sakamori
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasutoshi Nozaki
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sadatsugu Sakane
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuto Shiode
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tasuku Nakabori
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshinobu Saito
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Naoki Hiramatsu
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Keisuke Tabata
- Department of Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tsuyoshi Kawabata
- Department of Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Maho Hamasaki
- Department of Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tamotsu Yoshimori
- Department of Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
144
|
Sinha RA, Singh BK, Zhou J, Xie S, Farah BL, Lesmana R, Ohba K, Tripathi M, Ghosh S, Hollenberg AN, Yen PM. Loss of ULK1 increases RPS6KB1-NCOR1 repression of NR1H/LXR-mediated Scd1 transcription and augments lipotoxicity in hepatic cells. Autophagy 2016; 13:169-186. [PMID: 27846372 PMCID: PMC5240836 DOI: 10.1080/15548627.2016.1235123] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Lipotoxicity caused by saturated fatty acids (SFAs) induces tissue damage and inflammation in metabolic disorders. SCD1 (stearoyl-coenzyme A desaturase 1) converts SFAs to mono-unsaturated fatty acids (MUFAs) that are incorporated into triglycerides and stored in lipid droplets. SCD1 thus helps protect hepatocytes from lipotoxicity and its reduced expression is associated with increased lipotoxic injury in cultured hepatic cells and mouse models. To further understand the role of SCD1 in lipotoxicity, we examined the regulation of Scd1 in hepatic cells treated with palmitate, and found that NR1H/LXR (nuclear receptor subfamily 1 group H) ligand, GW3965, induced Scd1 expression and lipid droplet formation to improve cell survival. Surprisingly, ULK1/ATG1 (unc-51 like kinase) played a critical role in protecting hepatic cells from SFA-induced lipotoxicity via a novel mechanism that did not involve macroautophagy/autophagy. Specific loss of Ulk1 blocked the induction of Scd1 gene transcription by GW3965, decreased lipid droplet formation, and increased apoptosis in hepatic cells exposed to palmitate. Knockdown of ULK1 increased RPS6KB1 (ribosomal protein S6 kinase, polypeptide 1) signaling that, in turn, induced NCOR1 (nuclear receptor co-repressor 1) nuclear uptake, interaction with NR1H/LXR, and recruitment to the Scd1 promoter. These events abrogated the stimulation of Scd1 gene expression by GW3965, and increased lipotoxicity in hepatic cells. In summary, we have identified a novel autophagy-independent role of ULK1 that regulates NR1H/LXR signaling, Scd1 expression, and intracellular lipid homeostasis in hepatic cells exposed to a lipotoxic environment.
Collapse
Affiliation(s)
- Rohit Anthony Sinha
- a Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School , Singapore
| | - Brijesh K Singh
- a Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School , Singapore
| | - Jin Zhou
- a Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School , Singapore
| | - Sherwin Xie
- a Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School , Singapore
| | - Benjamin L Farah
- a Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School , Singapore
| | - Ronny Lesmana
- a Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School , Singapore.,b Department of Physiology , Universitas Padjadjaran , Bandung , Indonesia
| | - Kenji Ohba
- a Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School , Singapore
| | - Madhulika Tripathi
- c Stroke Trial Unit, National Neuroscience Institute Singapore , Singapore
| | - Sujoy Ghosh
- a Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School , Singapore
| | - Anthony N Hollenberg
- d Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston , MA USA
| | - Paul M Yen
- a Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School , Singapore
| |
Collapse
|
145
|
Pillon NJ, Chan KL, Zhang S, Mejdani M, Jacobson MR, Ducos A, Bilan PJ, Niu W, Klip A. Saturated fatty acids activate caspase-4/5 in human monocytes, triggering IL-1β and IL-18 release. Am J Physiol Endocrinol Metab 2016; 311:E825-E835. [PMID: 27624102 DOI: 10.1152/ajpendo.00296.2016] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 09/11/2016] [Indexed: 12/14/2022]
Abstract
Obesity is associated with metabolic tissue infiltration by monocyte-derived macrophages. Saturated fatty acids contribute to proinflammatory gene induction in tissue-embedded immune cells. However, it is unknown how circulating monocytes, the macrophage precursors, react to high-fat environments. In macrophages, saturated fatty acids activate inflammatory pathways and, notably, prime caspase-associated inflammasomes. Inflammasome-activated IL-1β contributes to type 2 diabetes. We hypothesized that 1) human monocytes from obese patients show caspase activation, and 2) fatty acids trigger this response and consequent release of IL-1β/IL-18. Human peripheral blood monocytes were sorted by flow cytometry, and caspase activity was measured with a FLICA dye-based assay. Blood monocytes from obese individuals exhibited elevated caspase activity. To explore the nature and consequence of this activity, human THP1 monocytes were exposed to saturated or unsaturated fatty acids. Caspase activity was revealed by isoform-specific cleavage and enzymatic activity; cytokine expression/release was measured by qPCR and ELISA. Palmitate, but not palmitoleate, increased caspase activity in parallel to the release of IL-1β and IL-18. Palmitate induced eventual monocyte cell death with features of pyroptosis (an inflammation-linked cell death program involving caspase-4/5), scored through LDH release, vital dye influx, cell volume changes, and nuclear morphology. Notably, selective gene silencing or inhibition of caspase-4/5 reduced palmitate-induced release of IL-1β and IL-18. In summary, monocytes from obese individuals present elevated caspase activity. Mechanistically, palmitate activates a pyroptotic program in monocytes through caspase-4/5, causing inflammatory cytokine release, additional to inflammasomes. These caspases represent potential, novel, therapeutic targets to taper obesity-associated inflammation.
Collapse
Affiliation(s)
- Nicolas J Pillon
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kenny L Chan
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Shitian Zhang
- Department of Immunology, Key Laboratory of Immuno Microenvironment and Disease of the Educational Ministry of China, Tianjin Medical University, Tianjin, China; and
| | - Marios Mejdani
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Maya R Jacobson
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Alexandre Ducos
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Philip J Bilan
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Wenyan Niu
- Department of Immunology, Key Laboratory of Immuno Microenvironment and Disease of the Educational Ministry of China, Tianjin Medical University, Tianjin, China; and
- Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Amira Klip
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada;
| |
Collapse
|
146
|
Lauschke VM, Mkrtchian S, Ingelman-Sundberg M. The role of microRNAs in liver injury at the crossroad between hepatic cell death and regeneration. Biochem Biophys Res Commun 2016; 482:399-407. [PMID: 27789285 DOI: 10.1016/j.bbrc.2016.10.084] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/19/2016] [Accepted: 10/23/2016] [Indexed: 02/07/2023]
Abstract
The liver fulfills critical metabolic functions, such as controlling blood sugar and ammonia levels, and is of central importance for lipid metabolism and detoxification of environmental and chemical agents, including drugs. Liver injuries of different etiology can elicit a spectrum of responses. Some hepatocytes initiate molecular programs resulting in cell death, whereas others undergo cellular divisions to regenerate the damaged organ. Interestingly, recent research indicates that microRNAs serve as very rapid as well as long-term regulators in these processes. In this review, we discuss their importance in liver disease etiology and progression as well as for therapy with particular focus on metabolic and inflammatory conditions. Furthermore, we highlight the central role of microRNAs in controlling hepatocyte differentiation and plasticity, which are required for successful regeneration, but under certain conditions, such as chronic liver insults, can result in the formation of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Volker M Lauschke
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institute, Stockholm, Sweden.
| | - Souren Mkrtchian
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institute, Stockholm, Sweden
| | - Magnus Ingelman-Sundberg
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
147
|
Protective Effect of Psoralea corylifolia L. Seed Extract against Palmitate-Induced Neuronal Apoptosis in PC12 Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:5410419. [PMID: 27843479 PMCID: PMC5097809 DOI: 10.1155/2016/5410419] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/18/2016] [Indexed: 01/04/2023]
Abstract
The extract of Psoralea corylifolia seeds (PCE) has been widely used as a herbal medicine because of its beneficial effect on human health. In this study, we investigated the protective effects and molecular mechanisms of PCE on palmitate- (PA-) induced toxicity in PC12 cells, a neuron-like cell line. PCE significantly increased cell viability in PA-treated PC12 cells and showed antiapoptotic effects, as evidenced by decreased expression of cleaved caspase-3, cleaved poly(ADP-ribose) polymerase, and bax protein as well as increased expression of bcl-2 protein. In addition, PCE treatment reduced PA-induced reactive oxygen species production and upregulated mRNA levels of antioxidant genes such as nuclear factor (erythroid-derived 2)-like 2 and heme oxygenase 1. Moreover, PCE treatment recovered the expression of autophagy marker genes such as beclin-1 and p62, which was decreased by PA treatment. Treatment with isopsoralen, one of the major components of PCE extract, also recovered the expression of autophagy marker genes and reduced PA-induced apoptosis. In conclusion, PCE exerts protective effects against lipotoxicity via its antioxidant function, and this effect is mediated by activation of autophagy. PCE might be a potential pharmacological agent to protect against neuronal cell injury caused by oxidative stress or lipotoxicity.
Collapse
|
148
|
Huang CW, Chien YS, Chen YJ, Ajuwon KM, Mersmann HM, Ding ST. Role of n-3 Polyunsaturated Fatty Acids in Ameliorating the Obesity-Induced Metabolic Syndrome in Animal Models and Humans. Int J Mol Sci 2016; 17:ijms17101689. [PMID: 27735847 PMCID: PMC5085721 DOI: 10.3390/ijms17101689] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 09/05/2016] [Accepted: 09/27/2016] [Indexed: 02/07/2023] Open
Abstract
The incidence of obesity and its comorbidities, such as insulin resistance and type II diabetes, are increasing dramatically, perhaps caused by the change in the fatty acid composition of common human diets. Adipose tissue plays a role as the major energy reservoir in the body. An excess of adipose mass accumulation caused by chronic positive energy balance results in obesity. The n-3 polyunsaturated fatty acids (n-3 PUFA), DHA (docosahexaenoic acid) and EPA (eicosapentaenoic acid) exert numerous beneficial effects to maintain physiological homeostasis. In the current review, the physiology of n-3 PUFA effects in the body is delineated from studies conducted in both human and animal experiments. Although mechanistic studies in human are limited, numerous studies conducted in animals and models in vitro provide potential molecular mechanisms of the effects of these fatty acids. Three aspects of n-3 PUFA in adipocyte regulation are discussed: (1) lipid metabolism, including adipocyte differentiation, lipolysis and lipogenesis; (2) energy expenditure, such as mitochondrial and peroxisomal fatty acid β-oxidation; and (3) inflammation, including adipokines and specialized pro-resolving lipid mediators. Additionally, the mechanisms by which n-3 PUFA regulate gene expression are highlighted. The beneficial effects of n-3 PUFA may help to reduce the incidence of obesity and its comorbidities.
Collapse
Affiliation(s)
- Chao-Wei Huang
- Department of Animal Science and Technology, National Taiwan University, Taipei 106, Taiwan.
| | - Yi-Shan Chien
- Department of Animal Science and Technology, National Taiwan University, Taipei 106, Taiwan.
| | - Yu-Jen Chen
- Institute of Biotechnology, National Taiwan University, Taipei 106, Taiwan.
| | - Kolapo M Ajuwon
- Department of Animal Science, Purdue University, West Lafayette, IN 47907-2054, USA.
| | - Harry M Mersmann
- Department of Animal Science and Technology, National Taiwan University, Taipei 106, Taiwan.
| | - Shih-Torng Ding
- Department of Animal Science and Technology, National Taiwan University, Taipei 106, Taiwan.
- Institute of Biotechnology, National Taiwan University, Taipei 106, Taiwan.
| |
Collapse
|
149
|
Lee DH, Han DH, Nam KT, Park JS, Kim SH, Lee M, Kim G, Min BS, Cha BS, Lee YS, Sung SH, Jeong H, Ji HW, Lee MJ, Lee JS, Lee HY, Chun Y, Kim J, Komatsu M, Lee YH, Bae SH. Ezetimibe, an NPC1L1 inhibitor, is a potent Nrf2 activator that protects mice from diet-induced nonalcoholic steatohepatitis. Free Radic Biol Med 2016; 99:520-532. [PMID: 27634173 DOI: 10.1016/j.freeradbiomed.2016.09.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/24/2016] [Accepted: 09/09/2016] [Indexed: 12/13/2022]
Abstract
Oxidative stress is important for the pathogenesis of nonalcoholic fatty liver disease (NAFLD), a chronic disease that ranges from hepatic steatosis to nonalcoholic steatohepatitis (NASH). The nuclear factor erythroid 2-related factor 2-Kelch-like ECH associated protein 1 (Nrf2-Keap1) pathway is essential for cytoprotection against oxidative stress. In this study, we found that oxidative stress or inflammatory biomarkers and TUNEL positive cells were markedly increased in NASH patients compared to normal or simple steatosis. In addition, we identified that the hepatic mRNA levels of Nrf2 target genes such as Nqo-1 and GSTA-1 were significantly increased in NASH patients. Ezetimibe, a drug approved by the Food and Drug Administration for the treatment of hypercholesterolemia, improves NAFLD and alleviates oxidative stress. However, the precise mechanism of its antioxidant function remains largely unknown. We now demonstrate that ezetimibe activates Nrf2-Keap1 pathway which was dependent of autophagy adaptor protein p62, without causing cytotoxicity. Ezetimibe activates AMP-activated protein kinase (AMPK), which in turn phosphorylates p62 (p-S351) via their direct interaction. Correspondingly, Ezetimibe protected liver cells from saturated fatty acid-induced apoptotic cell death through p62-dependent Nrf2 activation. Furthermore, its role as an Nrf2 activator was supported by methione- and choline- deficient (MCD) diet-induced NASH mouse model, showing that ezetimibe decreased the susceptibility of the liver to oxidative injury. These data demonstrate that the molecular mechanisms underlying ezetimibe's antioxidant role in the pathogenesis of NASH.
Collapse
Affiliation(s)
- Da Hyun Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University; Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Dai Hoon Han
- Department of Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Ki Taek Nam
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University; Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jeong Su Park
- Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Soo Hyun Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Milim Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Gyuri Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Byung Soh Min
- Department of Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Bong-Soo Cha
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Yu Seol Lee
- Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Su Haeng Sung
- Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Haengdueng Jeong
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University; Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Hye Won Ji
- Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Moon Joo Lee
- Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jae Sung Lee
- Department of Molecular Medicine and Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, School of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Hui-Young Lee
- Department of Molecular Medicine and Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, School of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Yoomi Chun
- Department of Oral Biochemistry and Molecular Biology, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joungmok Kim
- Department of Oral Biochemistry and Molecular Biology, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Masaaki Komatsu
- Department of Biochemistry, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata 951-8510, Japan
| | - Yong-Ho Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| | - Soo Han Bae
- Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| |
Collapse
|
150
|
Identification of cytochrome CYP2E1 as critical mediator of synergistic effects of alcohol and cellular lipid accumulation in hepatocytes in vitro. Oncotarget 2016; 6:41464-78. [PMID: 26497211 PMCID: PMC4747167 DOI: 10.18632/oncotarget.6203] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 10/09/2015] [Indexed: 02/07/2023] Open
Abstract
Clinical studies propose a causative link between the consumption of alcohol and the development and progression of liver disease in obese individuals. However, it is incompletely understood how alcohol and obesity interact and whether the combined effects are additive or synergistic. In this study, we developed an in vitro model to address this question. Lipid accumulation in primary human hepatocytes was induced by incubation with oleic acid. Subsequently, steatotic and control hepatocytes were incubated with up to 50 mM alcohol. This alcohol concentration on its own revealed only minimal effects but significantly enhanced oleate-induced lipogenesis and cellular triglyceride content compared to control cells. Similarly, lipid peroxidation, oxidative stress and pro-inflammatory gene expression as well as CYP2E1 levels and activity were synergistically induced by alcohol and steatosis. CYP2E1 inhibition blunted these synergistic pathological effects. Notably, alcohol and cellular steatosis also induced autophagy in a synergistic manner, and also this was mediated via CYP2E1. Further induction of autophagy ameliorated the joint effects of alcohol and oleic acid on hepatocellular lipid accumulation and inflammatory gene expression while inhibition of autophagy further enhanced the dual pathological effects. Further analyses revealed that the joint synergistic effect of alcohol and steatosis on autophagy was mediated via activation of the JNK-pathway. In summary, our data indicate that alcohol induces not only pathological but also protective mechanisms in steatotic hepatocytes via CYP2E1. These findings may have important implications on the prognosis and treatment of alcoholic liver disease particularly in obese individuals.
Collapse
|