151
|
Song YF, Hogstrand C, Wei CC, Wu K, Pan YX, Luo Z. Endoplasmic reticulum (ER) stress and cAMP/PKA pathway mediated Zn-induced hepatic lipolysis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2017; 228:256-264. [PMID: 28549333 DOI: 10.1016/j.envpol.2017.05.046] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 05/05/2017] [Accepted: 05/17/2017] [Indexed: 05/25/2023]
Abstract
The present study was performed to determine the effect of Zn exposure influencing endoplasmic reticulum (ER) stress, explore the underlying molecular mechanism of Zn-induced hepatic lipolysis in a fish species of significance for aquaculture, yellow catfish Pelteobagrus fulvidraco. We found that waterborne Zn exposure evoked ER stress and unfolded protein response (UPR), and activated cAMP/PKA pathway, and up-regulated hepatic lipolysis. The increase in ER stress and lipolysis were associated with activation of cAMP/PKA signaling pathway. Zn also induced an increase in intracellular Ca2+ level, which could be partially prevented by dantrolene (RyR receptor inhibitor) and 2-APB (IP3 receptor inhibitor), demonstrating that the disturbed Ca2+ homeostasis in ER contributed to ER stress and dysregulation of lipolysis. Inhibition of ER stress by PBA attenuated UPR, inhibited the activation of cAMP/PKA pathway and resulted in down-regulation of lipolysis. Inhibition of protein kinase RNA-activated-like ER kinase (PERK) by GSK2656157 and inositol-requiring enzyme (IRE) by STF-083010 differentially influenced Zn-induced changes of lipid metabolism, indicating that PERK and IRE pathways played different regulatory roles in Zn-induced lipolysis. Inhibition of PKA by H89 blocked the Zn-induced activation of cAMP/PKA pathway with a concomitant inhibition of ER stress-mediated lipolysis. Taken together, our findings highlight the importance of the ER stress-cAMP/PKA axis in Zn-induced lipolysis, which provides new insights into Zn toxicology in fish and probably in other vertebrates.
Collapse
Affiliation(s)
- Yu-Feng Song
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
| | - Christer Hogstrand
- Diabetes and Nutritional Sciences Division, School of Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
| | - Chuan-Chuan Wei
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
| | - Kun Wu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
| | - Ya-Xiong Pan
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhi Luo
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan 430070, China; Collaborative Innovation Center for Efficient and Health Production of Fisheries in Hunan Province, Changde 415000, China.
| |
Collapse
|
152
|
Mega C, Teixeira-de-Lemos E, Fernandes R, Reis F. Renoprotective Effects of the Dipeptidyl Peptidase-4 Inhibitor Sitagliptin: A Review in Type 2 Diabetes. J Diabetes Res 2017; 2017:5164292. [PMID: 29098166 PMCID: PMC5643039 DOI: 10.1155/2017/5164292] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 07/12/2017] [Indexed: 12/13/2022] Open
Abstract
Diabetic nephropathy (DN) is now the single commonest cause of end-stage renal disease (ESRD) worldwide and one of the main causes of death in diabetic patients. It is also acknowledged as an independent risk factor for cardiovascular disease (CVD). Since sitagliptin was approved, many studies have been carried out revealing its ability to not only improve metabolic control but also ameliorate dysfunction in various diabetes-targeted organs, especially the kidney, due to putative underlying cytoprotective properties, namely, its antiapoptotic, antioxidant, anti-inflammatory, and antifibrotic properties. Despite overall recommendations, many patients spend a long time well outside the recommended glycaemic range and, therefore, have an increased risk for developing micro- and macrovascular complications. Currently, it is becoming clearer that type 2 diabetes mellitus (T2DM) management must envision not only the improvement in glycaemic control but also, and particularly, the prevention of pancreatic deterioration and the evolution of complications, such as DN. This review aims to provide an overview of the current knowledge in the field of renoprotective actions of sitagliptin, namely, improvement in diabetic dysmetabolism, hemodynamic factors, renal function, diabetic kidney lesions, and cytoprotective properties.
Collapse
Affiliation(s)
- Cristina Mega
- Agrarian School of Viseu (ESAV), Polytechnic Institute of Viseu (IPV), 3500-606 Viseu, Portugal
- Centre for the Study of Education, Technologies and Health (CI&DETS), Polytechnic Institute of Viseu (IPV), 3500-606 Viseu, Portugal
- Institute of Pharmacology and Experimental Therapeutics and Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Edite Teixeira-de-Lemos
- Agrarian School of Viseu (ESAV), Polytechnic Institute of Viseu (IPV), 3500-606 Viseu, Portugal
- Centre for the Study of Education, Technologies and Health (CI&DETS), Polytechnic Institute of Viseu (IPV), 3500-606 Viseu, Portugal
| | - Rosa Fernandes
- Institute of Pharmacology and Experimental Therapeutics and Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CNC.IBILI Research Consortium, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Flávio Reis
- Institute of Pharmacology and Experimental Therapeutics and Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CNC.IBILI Research Consortium, University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
153
|
Choi WG, Han J, Kim JH, Kim MJ, Park JW, Song B, Cha HJ, Choi HS, Chung HT, Lee IK, Park TS, Hatzoglou M, Choi HS, Yoo HJ, Kaufman RJ, Back SH. eIF2α phosphorylation is required to prevent hepatocyte death and liver fibrosis in mice challenged with a high fructose diet. Nutr Metab (Lond) 2017; 14:48. [PMID: 28781602 PMCID: PMC5537942 DOI: 10.1186/s12986-017-0202-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 06/28/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Dietary fructose can rapidly cause fatty liver in animals through de novo lipogenesis (DNL) and contribute to the development and severity of nonalcoholic fatty liver disease (NAFLD). In response to diverse cellular insults including endoplasmic reticulum (ER) and oxidative stress, phosphorylation of the eukaryotic translation initiation factor 2 alpha subunit (eIF2α) attenuates general translation initiation, allowing cells to conserve resources and initiate adaptive gene expression to restore homeostasis. The present study aimed to investigate the role of eIF2α phosphorylation in protecting against NAFLD induced by high fructose ingestion in a hepatocyte-specific eIF2α-phosphorylation-deficient mouse model. METHODS Hepatocyte-specific non-phosphorylatable (S51A) eIF2α knock-in (A/A;fTg/0;CreHep/0, A/AHep ) mice were generated by crossing A/A;fTg/fTg mice with the floxed WT eIF2α transgene (fTg) with Alfp-Cre recombinase transgenic S/A;CreHep/0 (S/A-CreHep ) mice. Hepatocyte-specific eIF2α-phosphorylation-deficient 3-month-old mice or 12-month-old mice were fed a 60% high fructose diet (HFrD) for 16 or 5 wks, and the effects of eIF2α-phosphorylation deficiency on NADP/NADPH and GSSG/GSH levels, ROS-defense gene expression, oxidative damage, cell death, and fibrosis were observed. RESULTS Prolonged fructose feeding to mice caused dysregulation of the unfolded protein response (UPR) sensor activation and UPR gene expression, and then led to decreased expression of several ROS defense genes including glutathione biogenesis genes. Nonetheless, these changes were not sufficient to induce the death of eIF2α phosphorylation-sufficient hepatocytes. However, there was a substantial increase in hepatocyte death and liver fibrosis in fructose-fed middle-aged mice deficient in hepatocyte-specific eIF2α phosphorylation because of diminished antioxidant capacity due to reduced expression of antioxidant enzymes (GPX1 and HO-1) and lower NADPH and glutathione levels, as well as a possible increase in ROS-induced damage from infiltrating NOX2-expressing leukocytes; all this led to a vicious cycle of hepatocyte death and leukocyte infiltration. CONCLUSION Our findings suggest that eIF2α phosphorylation maintains NADPH and GSH levels and controls the expression of ROS-defense genes, thereby protecting hepatocytes from oxidative stresses induced by fructose metabolism.
Collapse
Affiliation(s)
- Woo-Gyun Choi
- School of Biological Sciences, University of Ulsan, Ulsan, 44610 Republic of Korea
| | - Jaeseok Han
- Soonchunhyang Institute of Med-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Choongchungnam-do, 31151 Republic of Korea.,Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Ji-Hyeon Kim
- School of Biological Sciences, University of Ulsan, Ulsan, 44610 Republic of Korea.,Biomedical Research Center, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, 05505 Republic of Korea
| | - Mi-Jeong Kim
- School of Biological Sciences, University of Ulsan, Ulsan, 44610 Republic of Korea
| | - Jae-Woo Park
- School of Biological Sciences, University of Ulsan, Ulsan, 44610 Republic of Korea
| | - Benbo Song
- NGM Biopharmaceuticals, Inc., 333 Oyster Point Blvd, South San Francisco, CA 94080 USA
| | - Hee-Jeong Cha
- Department of Pathology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, 44043 Republic of Korea
| | - Hye-Seon Choi
- School of Biological Sciences, University of Ulsan, Ulsan, 44610 Republic of Korea
| | - Hun-Taeg Chung
- School of Biological Sciences, University of Ulsan, Ulsan, 44610 Republic of Korea
| | - In-Kyu Lee
- Department of Internal Medicine and Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, 41944 Republic of Korea
| | - Tae-Sik Park
- Department of Life Science, Gachon University, Seongnam, Republic of Korea
| | - Maria Hatzoglou
- Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, OH 44106 USA
| | - Hueng-Sik Choi
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Hyun Ju Yoo
- Biomedical Research Center, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, 05505 Republic of Korea
| | - Randal J Kaufman
- Soonchunhyang Institute of Med-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Choongchungnam-do, 31151 Republic of Korea
| | - Sung Hoon Back
- School of Biological Sciences, University of Ulsan, Ulsan, 44610 Republic of Korea
| |
Collapse
|
154
|
Clinical and therapeutic potential of protein kinase PKR in cancer and metabolism. Expert Rev Mol Med 2017; 19:e9. [PMID: 28724458 DOI: 10.1017/erm.2017.11] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The protein kinase R (PKR, also called EIF2AK2) is an interferon-inducible double-stranded RNA protein kinase with multiple effects on cells that plays an active part in the cellular response to numerous types of stress. PKR has been extensively studied and documented for its relevance as an antiviral agent and a cell growth regulator. Recently, the role of PKR related to metabolism, inflammatory processes, cancer and neurodegenerative diseases has gained interest. In this review, we summarise and discuss the involvement of PKR in several cancer signalling pathways and the dual role that this kinase plays in cancer disease. We emphasise the importance of PKR as a molecular target for both conventional chemotherapeutics and emerging treatments based on novel drugs, and its potential as a biomarker and therapeutic target for several pathologies. Finally, we discuss the impact that the recent knowledge regarding PKR involvement in metabolism has in our understanding of the complex processes of cancer and metabolism pathologies, highlighting the translational research establishing the clinical and therapeutic potential of this pleiotropic kinase.
Collapse
|
155
|
Abstract
Background Formation of 43S and 48S preinitiation complexes plays an important role in muscle protein synthesis. There is no muscle-wasting mouse model caused by a repressed 43S preinitiation complex assembly. Objective The aim of the present study was to develop a convenient mouse model of skeletal muscle wasting with repressed 43S preinitiation complex assembly. Material and methods A ligand-activatable PERK derivative Fv2E-PERK causes the phosphorylation of eukaryotic initiation factor 2α (eIF2α), which inhibits 43S preinitiation complex assembly. Thus, muscle atrophic phenotypes, intracellular signaling pathways, and intracellular free amino acid profiles were investigated in human skeletal muscle α-actin (HSA) promoter-driven Fv2E-PERK transgenic (Tg) mice. Results HSA-Fv2E-PERK Tg mice treated with the artificial dimerizer AP20187 phosphorylates eIF2α in skeletal muscles and leads to severe muscle atrophy within a few days of ligand injection. Muscle atrophy was accompanied by a counter regulatory activation of mTORC1 signaling. Moreover, intracellular free amino acid levels were distinctively altered in the skeletal muscles of HSA-Fv2E-PERK Tg mice. Conclusions As a novel model of muscle wasting, HSA-Fv2E-PERK Tg mice provide a convenient tool for studying the pathogenesis of muscle loss and for assessing putative therapeutics.
Collapse
|
156
|
Overexpression of apolipoprotein A-I alleviates endoplasmic reticulum stress in hepatocytes. Lipids Health Dis 2017; 16:105. [PMID: 28577569 PMCID: PMC5455103 DOI: 10.1186/s12944-017-0497-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/24/2017] [Indexed: 11/10/2022] Open
Abstract
Background Abnormal lipid metabolism may contribute to an increase in endoplasmic reticulum (ER) stress, resulting in the pathogenesis of non-alcoholic steatohepatitis. Apolipoprotein A-I (apoA-I) accepts cellular free cholesterol and phospholipids transported by ATP-binding cassette transporter A1 to generate nascent high density lipoprotein particles. Previous studies have revealed that the overexpression of apoA-I alleviated hepatic lipid levels by modifying lipid transport. Here, we examined the effects of apoA-I overexpression on ER stress and genes involved in lipogenesis in both HepG2 cells and mouse hepatocytes. Methods Human apoA-I was overexpressed in HepG2 hepatocytes, which were then treated with 2 μg/mL tunicamycin or 500 μM palmitic acid. Eight-week-old male apoA-I transgenic or C57BL/6 wild-type mice were intraperitoneally injected with 1 mg/kg body weight tunicamycin or with saline. At 48 h after injecting, blood and liver samples were collected. Results The overexpression of apoA-I in the models above resulted in decreased protein levels of ER stress makers and lipogenic gene products, including sterol regulatory element binding protein 1, fatty acid synthase, and acetyl-CoA carboxylase 1. In addition, the cellular levels of triglycerides and free cholesterol also decreased. Some of gene products which are related to ER stress-associated apoptosis were also affected by apoA-I overexpression. These results suggested that apoA-I overexpression could reduce steatosis by decreasing lipid levels and by suppressing ER stress and lipogenesis in hepatocytes. Conclusion ApoA-I expression could significantly reduce hepatic ER stress and lipogenesis in hepatocytes.
Collapse
|
157
|
Abdulkarim B, Hernangomez M, Igoillo-Esteve M, Cunha DA, Marselli L, Marchetti P, Ladriere L, Cnop M. Guanabenz Sensitizes Pancreatic β Cells to Lipotoxic Endoplasmic Reticulum Stress and Apoptosis. Endocrinology 2017; 158:1659-1670. [PMID: 28323924 DOI: 10.1210/en.2016-1773] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 02/24/2017] [Indexed: 12/18/2022]
Abstract
Deficient as well as excessive/prolonged endoplasmic reticulum (ER) stress signaling can lead to pancreatic β cell failure and the development of diabetes. Saturated free fatty acids (FFAs) such as palmitate induce lipotoxic ER stress in pancreatic β cells. One of the main ER stress response pathways is under the control of the protein kinase R-like endoplasmic reticulum kinase (PERK), leading to phosphorylation of the eukaryotic translation initiation factor 2 (eIF2α). The antihypertensive drug guanabenz has been shown to inhibit eIF2α dephosphorylation and protect cells from ER stress. Here we examined whether guanabenz protects pancreatic β cells from lipotoxicity. Guanabenz induced β cell dysfunction in vitro and in vivo in rodents and led to impaired glucose tolerance. The drug significantly potentiated FFA-induced cell death in clonal rat β cells and in rat and human islets. Guanabenz enhanced FFA-induced eIF2α phosphorylation and expression of the downstream proapoptotic gene C/EBP homologous protein (CHOP), which mediated the sensitization to lipotoxicity. Thus, guanabenz does not protect β cells from ER stress; instead, it potentiates lipotoxic ER stress through PERK/eIF2α/CHOP signaling. These data demonstrate the crucial importance of the tight regulation of eIF2α phosphorylation for the normal function and survival of pancreatic β cells.
Collapse
Affiliation(s)
- Baroj Abdulkarim
- ULB Center for Diabetes Research, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Miriam Hernangomez
- ULB Center for Diabetes Research, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | | | - Daniel A Cunha
- ULB Center for Diabetes Research, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Lorella Marselli
- Department of Endocrinology and Metabolism, University of Pisa, 56126 Pisa, Italy
| | - Piero Marchetti
- Department of Endocrinology and Metabolism, University of Pisa, 56126 Pisa, Italy
| | - Laurence Ladriere
- ULB Center for Diabetes Research, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Miriam Cnop
- ULB Center for Diabetes Research, Université Libre de Bruxelles, 1070 Brussels, Belgium
- Division of Endocrinology, Erasmus Hospital, 1070 Brussels, Belgium
| |
Collapse
|
158
|
Protectin DX suppresses hepatic gluconeogenesis through AMPK-HO-1-mediated inhibition of ER stress. Cell Signal 2017; 34:133-140. [DOI: 10.1016/j.cellsig.2017.03.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/11/2017] [Accepted: 03/19/2017] [Indexed: 12/31/2022]
|
159
|
Horwath JA, Hurr C, Butler SD, Guruju M, Cassell MD, Mark AL, Davisson RL, Young CN. Obesity-induced hepatic steatosis is mediated by endoplasmic reticulum stress in the subfornical organ of the brain. JCI Insight 2017; 2:90170. [PMID: 28422749 PMCID: PMC5396512 DOI: 10.1172/jci.insight.90170] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 03/02/2017] [Indexed: 12/15/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), characterized by an excess accumulation of hepatic triglycerides, is a growing health epidemic. While ER stress in the liver has been implicated in the development of NAFLD, the role of brain ER stress - which is emerging as a key contributor to a number of chronic diseases including obesity - in NAFLD remains unclear. These studies reveal that chemical induction of ER stress in the brain caused hepatomegaly and hepatic steatosis in mice. Conversely, pharmacological reductions in brain ER stress in diet-induced obese mice rescued NAFLD independent of body weight, food intake, and adiposity. Evaluation of brain regions involved revealed robust activation of ER stress biomarkers and ER ultrastructural abnormalities in the circumventricular subfornical organ (SFO), a nucleus situated outside of the blood-brain-barrier, in response to high-fat diet. Targeted reductions in SFO-ER stress in obese mice via SFO-specific supplementation of the ER chaperone 78-kDa glucose-regulated protein ameliorated hepatomegaly and hepatic steatosis without altering body weight, food intake, adiposity, or obesity-induced hypertension. Overall, these findings indicate a novel role for brain ER stress, notably within the SFO, in the pathogenesis of NAFLD.
Collapse
Affiliation(s)
- Julie A. Horwath
- Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
- Cell and Developmental Biology, Weill Cornell Medical College, New York, New York, USA
| | - Chansol Hurr
- Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Scott D. Butler
- Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Mallikarjun Guruju
- Cell and Developmental Biology, Weill Cornell Medical College, New York, New York, USA
| | | | - Allyn L. Mark
- Cell and Developmental Biology, Weill Cornell Medical College, New York, New York, USA
- Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Robin L. Davisson
- Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
- Cell and Developmental Biology, Weill Cornell Medical College, New York, New York, USA
| | - Colin N. Young
- Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| |
Collapse
|
160
|
Tu HC, Hsiao YC, Yang WY, Tsai SL, Lin HK, Liao CY, Lu JW, Chou YT, Wang HD, Yuh CH. Up-regulation of golgi α-mannosidase IA and down-regulation of golgi α-mannosidase IC activates unfolded protein response during hepatocarcinogenesis. Hepatol Commun 2017; 1:230-247. [PMID: 29404456 PMCID: PMC5721452 DOI: 10.1002/hep4.1032] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/27/2017] [Accepted: 03/09/2017] [Indexed: 12/27/2022] Open
Abstract
α‐1,2 mannosidases, key enzymes in N‐glycosylation, are required for the formation of mature glycoproteins in eukaryotes. Aberrant regulation of α‐1,2 mannosidases can result in cancer, although the underlying mechanisms are unclear. Here, we report the distinct roles of α‐1,2 mannosidase subtypes (MAN1A, MAN1B, ERMAN1, MAN1C) in the formation of hepatocellular carcinoma (HCC). Clinicopathological analyses revealed that the clinical stage, tumor size, α‐fetoprotein level, and invasion status were positively correlated with the expression levels of MAN1A1, MAN1B1, and MAN1A2. In contrast, the expression of MAN1C1 was decreased as early as stage I of HCC. Survival analyses showed that high MAN1A1, MAN1A2, and MAN1B1 expression levels combined with low MAN1C1 expression levels were significantly correlated with shorter overall survival rates. Functionally, the overexpression of MAN1A1 promoted proliferation, migration, and transformation as well as in vivo migration in zebrafish. Conversely, overexpression of MAN1C1 reduced the migration ability both in vitro and in vivo, decreased the colony formation ability, and shortened the S phase of the cell cycle. Furthermore, the expression of genes involved in cell cycle/proliferation and migration was increased in MAN1A1‐overexpressing cells but decreased in MAN1C1‐overexpressing cells. MAN1A1 activated the expression of key regulators of the unfolded protein response (UPR), while treatment with endoplasmic reticulum stress inhibitors blocked the expression of MAN1A1‐activated genes. Using the MAN1A1 liver‐specific overexpression zebrafish model, we observed steatosis and inflammation at earlier stages and HCC formation at a later stage accompanied by the increased expression of the UPR modulator binding immunoglobulin protein (BiP). These data suggest that the up‐regulation of MAN1A1 activates the UPR and might initiate metastasis. Conclusion: MAN1A1 represents a novel oncogene while MAN1C1 plays a role in tumor suppression in hepatocarcinogenesis. (Hepatology Communications 2017;1:230‐247)
Collapse
Affiliation(s)
- Hsiao-Chen Tu
- Institute of Molecular and Genomic Medicine National Health Research Institutes Zhunan Miaoli Taiwan.,Institute of Biotechnology National Tsing-Hua University Hsinchu Taiwan
| | - Yung-Chun Hsiao
- Institute of Molecular and Genomic Medicine National Health Research Institutes Zhunan Miaoli Taiwan.,Institute of Biotechnology National Tsing-Hua University Hsinchu Taiwan
| | - Wan-Yu Yang
- Institute of Molecular and Genomic Medicine National Health Research Institutes Zhunan Miaoli Taiwan
| | - Shin-Lin Tsai
- Institute of Molecular and Genomic Medicine National Health Research Institutes Zhunan Miaoli Taiwan
| | - Hua-Kuo Lin
- Institute of Molecular and Genomic Medicine National Health Research Institutes Zhunan Miaoli Taiwan
| | - Chong-Yi Liao
- Institute of Molecular and Genomic Medicine National Health Research Institutes Zhunan Miaoli Taiwan.,Institute of Biotechnology National Tsing-Hua University Hsinchu Taiwan
| | - Jeng-Wei Lu
- Institute of Molecular and Genomic Medicine National Health Research Institutes Zhunan Miaoli Taiwan.,Department of Life Sciences National Central University Jhongli City Taoyuan Taiwan
| | - Yu-Ting Chou
- Institute of Molecular and Genomic Medicine National Health Research Institutes Zhunan Miaoli Taiwan.,Institute of Biotechnology National Tsing-Hua University Hsinchu Taiwan
| | - Horng-Dar Wang
- Institute of Biotechnology National Tsing-Hua University Hsinchu Taiwan
| | - Chiou-Hwa Yuh
- Institute of Molecular and Genomic Medicine National Health Research Institutes Zhunan Miaoli Taiwan.,Institute of Bioinformatics and Structural Biology National Tsing-Hua University Hsinchu Taiwan.,Department of Biological Science and Technology National Chiao Tung University Hsinchu Taiwan.,Kaohsiung Medical University Kaohsiung Taiwan
| |
Collapse
|
161
|
Modulation of glucose metabolism by a natural compound from Chloranthus japonicus via activation of AMP-activated protein kinase. Sci Rep 2017; 7:778. [PMID: 28396610 PMCID: PMC5429703 DOI: 10.1038/s41598-017-00925-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 03/17/2017] [Indexed: 12/25/2022] Open
Abstract
AMP-activated protein kinase (AMPK) is a key sensor and regulator of glucose metabolism. Here, we demonstrated that shizukaol F, a natural compound isolated from Chloranthus japonicus, can activate AMPK and modulate glucose metabolism both in vitro and in vivo. Shizukaol F increased glucose uptake in differentiated C2C12 myotubes by stimulating glucose transporter-4 (GLUT-4) membraned translocation. Treatment of primary mouse hepatocytes with shizukaol F decreased the expression of phosphoenolpyruvate carboxykinase 2 (PEPCK), glucose-6-phosphatase (G6Pase) and suppressed hepatic gluconeogenesis. Meanwhile, a single oral dose of shizukaol F reduced gluconeogenesis in C57BL/6 J mice. Further studies indicated that shizukaol F modulates glucose metabolism mainly by AMPKa phosphorylation activity. In addition, we also found that shizukaol F depolarizes the mitochondrial membrane and inhibits respiratory complex I, which may result in AMPK activation. Our results highlight the potential value of shizukaol F as a possible treatment of metabolic syndrome.
Collapse
|
162
|
Homocysteine Induces Hepatic Steatosis Involving ER Stress Response in High Methionine Diet-Fed Mice. Nutrients 2017; 9:nu9040346. [PMID: 28368295 PMCID: PMC5409685 DOI: 10.3390/nu9040346] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/16/2017] [Accepted: 03/28/2017] [Indexed: 12/23/2022] Open
Abstract
Elevated circulating homocysteine (Hcy) has been proposed to be associated with non-alcoholic fatty liver disease (NAFLD). It is also reported that Hcy causes protein misfolding in the endoplasmic reticulum (ER). In this study, we used a high methionine diet (HMD)-fed mouse model and cultured primary hepatocytes to investigate the effects of Hcy on hepatic lipids metabolism. C57BL/6J mice received either standard chow diet (CT, n = 10) or diet supplemented with 2% methionine (MET, n = 10) for 16 weeks. In in vitro experiments, cultured mouse primary hepatocytes were treated with Hcy, or Hcy combined with 4-phenylbutyric acid (4-PBA), or tunicamycin (TM), respectively. HMD-fed mice exhibited a mild increase in plasma Hcy level. There was no significant difference of body weight gain between the two groups. Nevertheless, HMD feeding increased epididymal fat/body weight ratio, elevated plasma triglyceride (TG) level, and decreased high-density lipoprotein cholesterol (HDL) level. Similarly, mice on HMD displayed higher liver/body weight ratio, plasma aspartate aminotransferase (AST) and its ratio to alanine aminotransferase (ALT), which was supported by the morphological observations of hepatic triglyceride accumulation in liver tissue as well as primary hepatocytes. Activation of the sterol response element-binding protein 1c (SREBP1c) in Hcy-treated hepatocytes with increased expression of genes involved in hepatic de novo lipogenesis was partially reduced by pretreatment of 4-PBA. Hcy-induced ER stress was also ameliorated by 4-PBA pretreatment, thus demonstrating an important role of Hcy-induced ER stress response in hepatic steatosis. These findings suggest that elevated Hcy was a critical factor in the pathogenesis of NAFLD. Activation of the ER stress response may be involved in Hcy-induced hepatic steatosis.
Collapse
|
163
|
Pereira CD, Passos E, Severo M, Vitó I, Wen X, Carneiro F, Gomes P, Monteiro R, Martins MJ. Ingestion of a natural mineral-rich water in an animal model of metabolic syndrome: effects in insulin signalling and endoplasmic reticulum stress. Horm Mol Biol Clin Investig 2017; 26:135-50. [PMID: 26741353 DOI: 10.1515/hmbci-2015-0033] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 11/18/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND High-fructose and/or low-mineral diets are relevant in metabolic syndrome (MS) development. Insulin resistance (IR) represents a central mechanism in MS development. Glucocorticoid signalling dysfunction and endoplasmic reticulum (ER) and oxidative stresses strongly contribute to IR and associate with MS. We have described that natural mineral-rich water ingestion delays fructose-induced MS development, modulates fructose effects on the redox state and glucocorticoid signalling and increases sirtuin 1 expression. Here, we investigated mineral-rich water ingestion effects on insulin signalling and ER homeostasis of fructose-fed rats. MATERIALS AND METHODS Adult male Sprague-Dawley rats had free access to standard-chow diet and different drinking solutions (8 weeks): tap water (CONT), 10%-fructose/tap water (FRUCT) or 10%-fructose/mineral-rich water (FRUCTMIN). Hepatic and adipose (visceral, VAT) insulin signalling and hepatic ER homeostasis (Western blot or PCR) as well as hepatic lipid accumulation were evaluated. RESULTS Hepatic p-IRS1Ser307/IRS1 (tendency), p-IRS1Ser307, total JNK and (activated IRE1α)/(activated JNK) decreased with fructose ingestion, while p-JNK tended to increase; mineral-rich water ingestion, totally or partially, reverted all these effects. Total PERK, p-eIF2α (tendency) and total IRS1 (tendency) decreased in both fructose-fed groups. p-ERK/ERK and total IRE1α increasing tendencies in FRUCT became significant in FRUCTMIN (similar pattern for lipid area). Additionally, unspliced-XBP1 increased with mineral-rich water. In VAT, total ERK fructose-induced increase was partially prevented in FRUCTMIN. CONCLUSIONS Mineral-rich water modulation of fructose-induced effects on insulin signalling and ER homeostasis matches the better metabolic profile previously reported. Increased p-ERK/ERK, adding to decreased IRE1α activation, and increased unspliced-XBP1 and lipid area may protect against oxidative stress and IR development in FRUCTMIN.
Collapse
|
164
|
Olexiková L, Pivko J, Makarevich AV, Kubovičová E, Ostró A, Živčák J, Chrenek P. Ultrastructure of Cell Organelles in Pre-implantation Embryos from Cows with Different Body Condition Score. Anat Histol Embryol 2017; 46:274-281. [PMID: 28239895 DOI: 10.1111/ahe.12267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Morphology of important cell organelles (mitochondria, lipid droplets, vacuoles, inclusion bodies and apoptotic bodies) in embryos derived from cows with different body condition score (BCS) was analysed by transmission electron microscopy (TEM). Embryos were recovered on 7th day after the insemination by a standard non-surgical flushing of the uterine horns from superovulated Holstein Friesian cows with BCS 2, 3, 4 and 5. Thereafter, the good quality blastocysts were processed for TEM. The electronograms were evaluated by stereological analysis. The relative volume of lipid droplets in BCS4 and BCS5 embryos increased significantly (18.53 and 22.40%) when compared to BCS3 embryos (5.46%). In the embryos from the BCS4 or BCS5 cows, we observed different morphological patterns of mitochondria, as well as the mitochondria containing vacuoles. BCS4 and BCS5 embryo cell nuclei showed the structure typical for low transcription activity (none or very few reticular nucleoli); also dilated inter-cellular spaces were often observed in these embryos. In conclusion, differences in the ultrastructural morphology of embryos from over-conditioned cows (BCS4 and BCS5), particularly the higher lipid content in the cytoplasm, can be a marker of their low quality, and this fact can be a contributing factor to subfertility in over-conditioned cows.
Collapse
Affiliation(s)
- L Olexiková
- Research Institute for Animal Production Nitra, National Agricultural and Food Centre, Hlohovecka 2, 95141, Lužianky-near-Nitra, Slovak Republic
| | - J Pivko
- Research Institute for Animal Production Nitra, National Agricultural and Food Centre, Hlohovecka 2, 95141, Lužianky-near-Nitra, Slovak Republic
| | - A V Makarevich
- Research Institute for Animal Production Nitra, National Agricultural and Food Centre, Hlohovecka 2, 95141, Lužianky-near-Nitra, Slovak Republic
| | - E Kubovičová
- Research Institute for Animal Production Nitra, National Agricultural and Food Centre, Hlohovecka 2, 95141, Lužianky-near-Nitra, Slovak Republic
| | - A Ostró
- Pavol Jozef Šafarik University in Košice, Clinics of Gynaecology and Obstetrics, Tr. SNP 1, 040 11, Košice, Slovak Republic
| | - J Živčák
- Technical University of Košice, Institute of Security and Biomedical Engineering, Letná 9, 042 00, Košice, Slovak Republic
| | - P Chrenek
- Research Institute for Animal Production Nitra, National Agricultural and Food Centre, Hlohovecka 2, 95141, Lužianky-near-Nitra, Slovak Republic.,Slovak University of Agriculture, Faculty of Biotechnology and Food Science, Tr.A.Hlinku 1, 94901, Nitra, Slovak Republic
| |
Collapse
|
165
|
Pinto AP, da Rocha AL, Oliveira LDC, Morais GP, de Vicente LG, Cintra DE, Pauli JR, Moura LP, Ropelle ER, da Silva ASR. Levels of Hepatic Activating Transcription Factor 6 and Caspase-3 Are Downregulated in Mice after Excessive Training. Front Endocrinol (Lausanne) 2017; 8:247. [PMID: 29018408 PMCID: PMC5622940 DOI: 10.3389/fendo.2017.00247] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/11/2017] [Indexed: 12/25/2022] Open
Abstract
Recently, we demonstrated that different running overtraining (OT) protocols with the same external load, but performed downhill (OTR/down), uphill (OTR/up), and without inclination (OTR), led to hepatic fat accumulation. As the disruption of endoplasmic reticulum (ER) homeostasis is linked to animal models of fatty liver disease, we investigated the effects of these OT models on the proteins related to ER stress (i.e., BiP, inositol-requiring enzyme 1, protein kinase RNA-like endoplasmic reticulum kinase, eIF2alpha, ATF6beta, and glucose-regulated protein 94) and apoptosis (C/EBP-homologous protein, Caspase-3, 4, and 12, Bax, and tumor necrosis factor receptor-associated factor 2) in livers of C57BL/6 mice. Also, aerobic training can attenuate cardiac ER stress and improve exercise capacity. Therefore, we investigated whether the decrease in performance induced by our OT protocols is linked to ER stress and apoptosis in mouse hearts. The rodents were divided into six groups: naïve (N, sedentary mice), control (CT, sedentary mice submitted to the performance evaluations), trained (TR), OTR/down, OTR/up, and OTR groups. Rotarod, incremental load, exhaustive, and grip force tests were used to evaluate performance. After the grip force test, the livers and cardiac muscles (i.e., left ventricle) were removed and used for immunoblotting. All of the OT protocols led to similar responses in the performance parameters and displayed significantly lower hepatic ATF6beta values compared to the N group. The OTR/down group exhibited lower liver cleaved caspase-3 values compared to the CT group. However, the other proteins related to ER stress and apoptosis were not modulated. Also, the cardiac proteins related to ER stress and apoptosis were not modulated in the experimental groups. In conclusion, the OT protocols decreased the levels of hepatic ATF6beta, and the OTR/down group decreased the levels of hepatic cleaved caspase-3. Also, the decrease in performance induced by our OT models is not associated with ER stress and apoptosis in mice hearts.
Collapse
Affiliation(s)
- Ana P. Pinto
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Alisson L. da Rocha
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Luciana da C. Oliveira
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Gustavo P. Morais
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Larissa G. de Vicente
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Dennys E. Cintra
- Sport Sciences Course, Faculty of Applied Sciences, State University of Campinas (UNICAMP), Limeira, Brazil
| | - José R. Pauli
- Sport Sciences Course, Faculty of Applied Sciences, State University of Campinas (UNICAMP), Limeira, Brazil
| | - Leandro P. Moura
- Sport Sciences Course, Faculty of Applied Sciences, State University of Campinas (UNICAMP), Limeira, Brazil
| | - Eduardo R. Ropelle
- Sport Sciences Course, Faculty of Applied Sciences, State University of Campinas (UNICAMP), Limeira, Brazil
| | - Adelino S. R. da Silva
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
- *Correspondence: Adelino S. R. da Silva,
| |
Collapse
|
166
|
Chandrahas VK, Han J, Kaufman RJ. Coordinating Organismal Metabolism During Protein Misfolding in the ER Through the Unfolded Protein Response. Curr Top Microbiol Immunol 2017; 414:103-130. [PMID: 28900680 DOI: 10.1007/82_2017_41] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The endoplasmic reticulum (ER) is a cellular organelle responsible for folding of secretory and membrane proteins. Perturbance in ER homeostasis caused by various intrinsic/extrinsic stimuli challenges the protein-folding capacity of the ER, leading to an ER dysfunction, called ER stress. Cells have developed a defensive response to adapt and/or survive in the face of ER stress that may be detrimental to cell function and survival. When exposed to ER stress, the cell activates a complex and elaborate signaling network that includes translational modulation and transcriptional induction of genes. In addition to these autonomous responses, recent studies suggest that the stressed tissue secretes peptides or unknown factors that transfer the signal to other cells in the same or different organs, leading the organism as a whole to cope with challenges in a non-autonomous manner. In this review, we discuss the mechanisms by which cells adapt to ER stress challenges autonomously and transfer the stress signal to non-stressed cells in different organs.
Collapse
Affiliation(s)
- Vishwanatha K Chandrahas
- Degenerative Diseases Program, Sanford_Burnham_Prebys Medical Discovery Institute, 92037, La Jolla, CA, USA
| | - Jaeseok Han
- Soonchunhyang Institute of Med-bio Science (SIMS), Soonchunhyang University, 31151, Cheonan-si, Chungcheongnam-do, Republic of Korea.
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford_Burnham_Prebys Medical Discovery Institute, 92037, La Jolla, CA, USA.
| |
Collapse
|
167
|
Gomez JA, Rutkowski DT. Experimental reconstitution of chronic ER stress in the liver reveals feedback suppression of BiP mRNA expression. eLife 2016; 5. [PMID: 27938665 PMCID: PMC5179193 DOI: 10.7554/elife.20390] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 12/09/2016] [Indexed: 12/13/2022] Open
Abstract
Endoplasmic reticulum (ER) stress is implicated in many chronic diseases, but very little is known about how the unfolded protein response (UPR) responds to persistent ER stress in vivo. Here, we experimentally reconstituted chronic ER stress in the mouse liver, using repeated injection of a low dose of the ER stressor tunicamycin. Paradoxically, this treatment led to feedback-mediated suppression of a select group of mRNAs, including those encoding the ER chaperones BiP and GRP94. This suppression was due to both silencing of the ATF6α pathway of UPR-dependent transcription and enhancement of mRNA degradation, possibly via regulated IRE1-dependent decay (RIDD). The suppression of mRNA encoding BiP was phenocopied by ectopic overexpression of BiP protein, and was also observed in obese mice. Our findings suggest that persistent cycles of UPR activation and deactivation create an altered, quasi-stable setpoint for UPR-dependent transcriptional regulation—an outcome that could be relevant to conditions such as metabolic syndrome. DOI:http://dx.doi.org/10.7554/eLife.20390.001 Toxic chemicals, extreme temperatures and other abnormal environmental conditions can cause the cells in our bodies to become stressed. Several kinds of stresses overwhelm a compartment in the cell called the endoplasmic reticulum, which is critical for processing new proteins so that they can work correctly. Endoplasmic reticulum stress has been linked to long-term diseases such as diabetes, cancer and neurodegenerative diseases. Most of what is known about how cells sense and respond to endoplasmic reticulum stress comes from studies on isolated cells that were subjected to harsh conditions that cells cannot tolerate for longer than a day or two. By contrast, little is known about how cells within whole organisms respond to milder but longer-lasting endoplasmic reticulum stress, which is closer to what occurs during disease. To investigate this issue, Gomez and Rutkowski treated mice repeatedly with a chemical that causes mild endoplasmic reticulum stress in the liver. The cells exposed to this persistent stress responded differently to those exposed to severe short-term stress. Whereas short-term stress causes liver cells to turn on genes that help the endoplasmic reticulum to process proteins more efficiently, long-term stress causes cells to turn off some of those genes. Further investigation revealed that cells in the livers of obese mice show similar patterns of gene activity as cells exposed to long-term endoplasmic reticulum stress. The findings presented by Gomez and Rutkowski could therefore also help us to understand more about the liver problems that often occur during obesity and diabetes. Further studies are now needed to examine exactly how long-lasting stress can shut off the cells’ protective mechanisms. Future experiments could also investigate whether other types of cells and organs respond to long-term endoplasmic reticulum stress in the same way as cells in the liver. DOI:http://dx.doi.org/10.7554/eLife.20390.002
Collapse
Affiliation(s)
- Javier A Gomez
- Graduate Program in Molecular and Cellular Biology, University of Iowa Carver College of Medicine, Iowa City, United States
| | - D Thomas Rutkowski
- Departments of Anatomy and Cell Biology and Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, United States
| |
Collapse
|
168
|
Abstract
Hepatic steatosis, the first step in the progression of nonalcoholic fatty liver disease, is characterized by triglyceride accumulation in hepatocytes and is highly prevalent in people with obesity. Although initially asymptomatic, hepatic steatosis is an important risk factor for the development of hepatic insulin resistance and type 2 diabetes mellitus and can also progress to more severe pathologies such as nonalcoholic steatohepatitis, liver fibrosis and cirrhosis; hepatic steatosis has, therefore, received considerable research interest in the past 20 years. The lipid accumulation that defines hepatic steatosis disturbs the function of the endoplasmic reticulum (ER) in hepatocytes, thereby generating chronic ER stress that interferes with normal cellular function. Although ubiquitous stress response mechanisms (namely, ER-associated degradation, unfolded protein response and autophagy) are the main processes for restoring cellular proteostasis, these mechanisms are unable to alleviate ER stress in the context of the fatty liver. Furthermore, ER stress and ER stress responses can promote lipid accumulation in hepatocytes in a counter-productive manner and could, therefore, be the origin of a vicious pathological cycle.
Collapse
Affiliation(s)
- Andrei Baiceanu
- Institut National de la Santé et de la Recherche Médicale, UMRS 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMRS 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMRS 1138, Centre de Recherche des Cordeliers, 15 rue de l'école de médecine, F-75006, Paris, France
- University of Medicine and Pharmacy Iuliu Hat¸ieganu, Faculty of Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania
| | - Pierre Mesdom
- Institut National de la Santé et de la Recherche Médicale, UMRS 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMRS 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMRS 1138, Centre de Recherche des Cordeliers, 15 rue de l'école de médecine, F-75006, Paris, France
| | - Marie Lagouge
- Institut National de la Santé et de la Recherche Médicale, UMRS 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMRS 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMRS 1138, Centre de Recherche des Cordeliers, 15 rue de l'école de médecine, F-75006, Paris, France
| | - Fabienne Foufelle
- Institut National de la Santé et de la Recherche Médicale, UMRS 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMRS 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMRS 1138, Centre de Recherche des Cordeliers, 15 rue de l'école de médecine, F-75006, Paris, France
| |
Collapse
|
169
|
Masuda M, Miyazaki-Anzai S, Keenan AL, Shiozaki Y, Okamura K, Chick WS, Williams K, Zhao X, Rahman SM, Tintut Y, Adams CM, Miyazaki M. Activating transcription factor-4 promotes mineralization in vascular smooth muscle cells. JCI Insight 2016; 1:e88646. [PMID: 27812542 DOI: 10.1172/jci.insight.88646] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Emerging evidence indicates that upregulation of the ER stress-induced pro-osteogenic transcription factor ATF4 plays an important role in vascular calcification, a common complication in patients with aging, diabetes, and chronic kidney disease (CKD). In this study, we demonstrated the pathophysiological role of ATF4 in vascular calcification using global Atf4 KO, smooth muscle cell-specific (SMC-specific) Atf4 KO, and transgenic (TG) mouse models. Reduced expression of ATF4 in global ATF4-haplodeficient and SMC-specific Atf4 KO mice reduced medial and atherosclerotic calcification under normal kidney and CKD conditions. In contrast, increased expression of ATF4 in SMC-specific Atf4 TG mice caused severe medial and atherosclerotic calcification. We further demonstrated that ATF4 transcriptionally upregulates the expression of type III sodium-dependent phosphate cotransporters (PiT1 and PiT2) by interacting with C/EBPβ. These results demonstrate that the ER stress effector ATF4 plays a critical role in the pathogenesis of vascular calcification through increased phosphate uptake in vascular SMCs.
Collapse
Affiliation(s)
- Masashi Masuda
- Division of Renal Diseases and Hypertension, Department of Medicine, and
| | | | - Audrey L Keenan
- Division of Renal Diseases and Hypertension, Department of Medicine, and
| | - Yuji Shiozaki
- Division of Renal Diseases and Hypertension, Department of Medicine, and
| | - Kayo Okamura
- Division of Renal Diseases and Hypertension, Department of Medicine, and
| | - Wallace S Chick
- Department of Cell and Developmental Biology, University of Colorado Denver, Aurora, Colorado, USA
| | - Kristina Williams
- Department of Cell and Developmental Biology, University of Colorado Denver, Aurora, Colorado, USA
| | - Xiaoyun Zhao
- Department of Cell and Developmental Biology, University of Colorado Denver, Aurora, Colorado, USA
| | | | - Yin Tintut
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, California, USA
| | | | - Makoto Miyazaki
- Division of Renal Diseases and Hypertension, Department of Medicine, and
| |
Collapse
|
170
|
Abstract
The burden of type 2 diabetes and its major complication cardiovascular disease is rapidly increasing worldwide. Understanding the underlying pathogenic mechanisms of these diseases is crucial to develop novel therapeutics. Recent work using genetic and biochemical methods in mouse models and human samples have identified disturbed calcium signalling and endoplasmic reticulum stress as emerging factors involved in the pathogenesis of many metabolic diseases. In this review, we will highlight the specific roles of calcium signalling and endoplasmic reticulum stress response in the development of insulin resistance and atherosclerosis.
Collapse
Affiliation(s)
- L Ozcan
- Department of Medicine, Columbia University, New York, NY, USA.
| | - I Tabas
- Department of Medicine, Columbia University, New York, NY, USA.,Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA.,Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| |
Collapse
|
171
|
Integrated stress response of vertebrates is regulated by four eIF2α kinases. Sci Rep 2016; 6:32886. [PMID: 27633668 PMCID: PMC5025754 DOI: 10.1038/srep32886] [Citation(s) in RCA: 193] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 08/16/2016] [Indexed: 12/17/2022] Open
Abstract
The integrated stress response (ISR) is a cytoprotective pathway initiated upon phosphorylation of the eukaryotic translation initiation factor 2 (eIF2α) residue designated serine-51, which is critical for translational control in response to various stress conditions. Four eIF2α kinases, namely heme-regulated inhibitor (HRI), protein kinase R (PKR), PKR-like endoplasmic reticulum kinase, (PERK) and general control non-depressible 2 (GCN2), have been identified thus far, and they are known to be activated by heme depletion, viral infection, endoplasmic reticulum stress, and amino acid starvation, respectively. Because eIF2α is phosphorylated under various stress conditions, the existence of an additional eIF2α kinase has been suggested. To validate the existence of the unidentified eIF2α kinase, we constructed an eIF2α kinase quadruple knockout cells (4KO cells) in which the four known eIF2α kinase genes were deleted using the CRISPR/Cas9-mediated genome editing. Phosphorylation of eIF2α was completely abolished in the 4KO cells by various stress stimulations. Our data suggests that the four known eIF2α kinases are sufficient for ISR and that there are no additional eIF2α kinases in vertebrates.
Collapse
|
172
|
DPP-4 inhibitors in diabetic complications: role of DPP-4 beyond glucose control. Arch Pharm Res 2016; 39:1114-28. [PMID: 27502601 DOI: 10.1007/s12272-016-0813-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 08/04/2016] [Indexed: 12/12/2022]
Abstract
Dipeptidyl peptidase-4 (DPP-4) inhibitors (gliptins) are an emerging class of antidiabetic drugs that constitutes approximately fifty percent of the market share of the oral hypoglycemic drugs. Its mechanism of action for lowering blood glucose is essentially via inhibition of the rapid degradation of incretin hormones, such as glucagon-like peptide (GLP)-1 and gastric inhibitory polypeptide (GIP), thus the plasma concentration of GLP-1 increases, which promotes insulin secretion from the pancreatic β cells and suppresses glucagon secretion from the α cells. In addition to the direct actions on the pancreas, GLP-1 exhibits diverse actions on different tissues through its action on GLP-1 receptor, which is expressed ubiquitously. Moreover, DPP-4 has multiple substrates besides GLP-1 and GIP, including cytokines, chemokines, neuropeptides, and growth factors, which are involved in many pathophysiological conditions. Recently, it was suggested that DPP-4 is a new adipokine secreted from the adipose tissue, which plays an important role in the regulation of the endocrine function in obesity-associated type 2 diabetes. Consequently, DPP-4 inhibitors have been reported to exhibit cytoprotective functions against various diabetic complications affecting the liver, heart, kidneys, retina, and neurons. This review outlines the current understanding of the effect of DPP-4 inhibitors on the complications associated with type 2 diabetes, such as liver steatosis and inflammation, dysfunction of the adipose tissue and pancreas, cardiovascular diseases, nephropathy, and neuropathy in preclinical and clinical studies.
Collapse
|
173
|
Mota M, Banini BA, Cazanave SC, Sanyal AJ. Molecular mechanisms of lipotoxicity and glucotoxicity in nonalcoholic fatty liver disease. Metabolism 2016; 65:1049-61. [PMID: 26997538 PMCID: PMC4931958 DOI: 10.1016/j.metabol.2016.02.014] [Citation(s) in RCA: 409] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 02/05/2016] [Accepted: 02/23/2016] [Indexed: 12/21/2022]
Abstract
The exposure of hepatocytes to high concentrations of lipids and carbohydrates and the ensuing hepatocellular injury are termed lipotoxicity and glucotoxicity, respectively. A common denominator is metabolic derangement, especially in regards to intracellular energy homeostasis, which is brought on by glucose intolerance and insulin resistance in tissues. In this review, we highlight the lipids and carbohydrates that provoke hepatocyte injury and the mechanisms involved in lipotoxicity and glucotoxicity, including endoplasmic reticulum stress, oxidative stress and mitochondrial impairment. Through upregulation of proteins involved in various pathways including PKR-like ER kinase (PERK), CCAAT/enhancer-binding homologous protein (CHOP), c-Jun NH2-terminal kinase-1 (JNK), Bcl-2 interacting mediator (BIM), p53 upregulated modulator of apoptosis (PUMA), and eventually caspases, hepatocytes in lipotoxic states ultimately undergo apoptosis. The protective role of certain lipids and possible targets for pharmacological therapy are explored. Finally, we discuss the role of high fructose and glucose diets in contributing to organelle impairment and poor glucose transport mechanisms, which perpetuate hyperglycemia and hyperlipidemia by shunting of excess carbohydrates into lipogenesis.
Collapse
Affiliation(s)
- Manoela Mota
- Department of Internal Medicine, Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| | - Bubu A Banini
- Department of Internal Medicine, Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Sophie C Cazanave
- Department of Internal Medicine, Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Arun J Sanyal
- Department of Internal Medicine, Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| |
Collapse
|
174
|
Rahman K, Liu Y, Kumar P, Smith T, Thorn NE, Farris AB, Anania FA. C/EBP homologous protein modulates liraglutide-mediated attenuation of non-alcoholic steatohepatitis. J Transl Med 2016; 96:895-908. [PMID: 27239734 PMCID: PMC4965279 DOI: 10.1038/labinvest.2016.61] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 04/15/2016] [Accepted: 04/26/2016] [Indexed: 02/06/2023] Open
Abstract
The CCAAT/enhancer-binding protein (C/EBP) homologous protein (CHOP), a major transcriptional regulator of endoplasmic reticulum (ER) stress-mediated apoptosis, is implicated in lipotoxicity-induced ER stress and hepatocyte apoptosis in non-alcoholic fatty liver disease (NAFLD). We have previously demonstrated that the glucagon-like peptide-1 (GLP-1) agonist, liraglutide, protects steatotic hepatocytes from lipotoxicity-induced apoptosis by improved handling of free fatty acid (FFA)-induced ER stress. In the present study, we investigated whether CHOP is critical for GLP-1-mediated restoration of ER homeostasis and mitigation of hepatocyte apoptosis in a murine model of NASH (non-alcoholic steatohepatitis). Our data show that despite similar caloric intake, CHOP KO (CHOP(-/-)) mice fed a diet high in fat, fructose, and cholesterol (HFCD) for 16 weeks developed more severe histological features of NASH compared with wild-type (WT) controls. Severity of NASH in HFCD-fed CHOP(-/-) mice correlated with significant decrease in peroxisomal β-oxidation, and increased de novo lipogenesis and ER stress-mediated hepatocyte apoptosis. Four weeks of liraglutide treatment markedly attenuated steatohepatitis in HFCD-fed WT mice by improving insulin sensitivity, and suppressing de novo lipogenesis and ER stress-mediated hepatocyte apoptosis. However, in the absence of CHOP, liraglutide did not improve insulin sensitivity, nor suppress peroxisomal β-oxidation or ER stress-mediated hepatocyte apoptosis. Taken together, these data indicate that CHOP protects hepatocytes from HFCD-induced ER stress, and has a significant role in the mechanism of liraglutide-mediated protection against NASH pathogenesis.
Collapse
Affiliation(s)
- Khalidur Rahman
- Division of Digestive Diseases, Emory University, Atlanta, GA,Atlanta VA Medical Center, Decatur, GA,Corresponding Author: Khalidur Rahman, PhD, Assistant Professor of Medicine, Emory University School of Medicine, 615 Michael Street, Suite 201, Atlanta, GA 30322, Phone: 404-712-2867 or 404-727-5638, Fax: 404-727-5767
| | | | - Pradeep Kumar
- Division of Digestive Diseases, Emory University, Atlanta, GA
| | - Tekla Smith
- Division of Digestive Diseases, Emory University, Atlanta, GA,Atlanta VA Medical Center, Decatur, GA
| | | | - Alton B. Farris
- Department of Pathology, Emory University Hospital, Atlanta, GA
| | - Frank A. Anania
- Division of Digestive Diseases, Emory University, Atlanta, GA,Atlanta VA Medical Center, Decatur, GA
| |
Collapse
|
175
|
Bozaykut P, Sahin A, Karademir B, Ozer NK. Endoplasmic reticulum stress related molecular mechanisms in nonalcoholic steatohepatitis. Mech Ageing Dev 2016; 157:17-29. [PMID: 27393639 DOI: 10.1016/j.mad.2016.07.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 06/23/2016] [Accepted: 07/02/2016] [Indexed: 12/18/2022]
|
176
|
Han J, Kaufman RJ. The role of ER stress in lipid metabolism and lipotoxicity. J Lipid Res 2016; 57:1329-38. [PMID: 27146479 DOI: 10.1194/jlr.r067595] [Citation(s) in RCA: 452] [Impact Index Per Article: 50.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Indexed: 12/23/2022] Open
Abstract
The endoplasmic reticulum (ER) is a cellular organelle important for regulating calcium homeostasis, lipid metabolism, protein synthesis, and posttranslational modification and trafficking. Numerous environmental, physiological, and pathological insults disturb ER homeostasis, referred to as ER stress, in which a collection of conserved intracellular signaling pathways, termed the unfolded protein response (UPR), are activated to maintain ER function for cell survival. However, excessive and/or prolonged UPR activation leads to initiation of self-destruction through apoptosis. Excessive accumulation of lipids and their intermediate products causes metabolic abnormalities and cell death, called lipotoxicity, in peripheral organs, including the pancreatic islets, liver, muscle, and heart. Because accumulating evidence links chronic ER stress and defects in UPR signaling to lipotoxicity in peripheral tissues, understanding the role of ER stress in cell physiology is a topic under intense investigation. In this review, we highlight recent findings that link ER stress and UPR signaling to the pathogenesis of peripheral organs due to lipotoxicity.
Collapse
Affiliation(s)
- Jaeseok Han
- Soonchunhyang Institute of Med-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Choongchungnam-do, 31151, Republic of Korea
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92307
| |
Collapse
|
177
|
Abstract
The aging process is characterized by tissue decline and the onset of age-associated disease. It is not, however, immutable, and aging can be modulated by various genetic and environmental means. One of the interventions that can modulate lifespan is the activation of cellular stress responses, including the unfolded protein response in the endoplasmic reticulum (UPRER). The ability to activate the UPRER declines with age, while its constitutive activation can extend longevity. It also plays complex roles in the onset and progression of many age-related diseases. Understanding how the UPRER changes with age, and how this impacts upon disease development, may open new therapeutic avenues for the treatment of a range of age-associated diseases. This article is part of a Special Issue entitled SI:ER stress.
Collapse
|
178
|
Lipopolysaccharide markedly changes glucose metabolism and mitochondrial function in the longissimus muscle of pigs. Animal 2016; 10:1204-12. [PMID: 26863995 DOI: 10.1017/s1751731116000100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Most previous studies on the effects of lipopolysaccharide (LPS) in pigs focused on the body's immune response, and few reports paid attention to body metabolism changes. To better understand the glucose metabolism changes in skeletal muscle following LPS challenge and to clarify the possible mechanism, 12 growing pigs were employed. Animals were treated with either 2 ml of saline or 15 µg/kg BW LPS, and samples were collected 6 h later. The glycolysis status and mitochondrial function in the longissimus dorsi (LD) muscle of pigs were analyzed. The results showed that serum lactate content and NADH content in LD muscle significantly increased compared with the control group. Most glycolysis-related genes expression, as well as hexokinase, pyruvate kinase and lactic dehydrogenase activity, in LD muscle was significantly higher compared with the control group. Mitochondrial complexes I and IV significantly increased, while mitochondrial ATP concentration markedly decreased. Significantly increased calcium content in the mitochondria was observed, and endoplasm reticulum (ER) stress has been demonstrated in the present study. The results showed that LPS treatment markedly changes glucose metabolism and mitochondrial function in the LD muscle of pigs, and increased calcium content induced by ER stress was possibly involved. The results provide new clues for clarifying metabolic diseases in muscle induced by LPS.
Collapse
|
179
|
Jung TW, Choi KM. Pharmacological Modulators of Endoplasmic Reticulum Stress in Metabolic Diseases. Int J Mol Sci 2016; 17:ijms17020192. [PMID: 26840310 PMCID: PMC4783926 DOI: 10.3390/ijms17020192] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Revised: 01/20/2016] [Accepted: 01/27/2016] [Indexed: 01/23/2023] Open
Abstract
The endoplasmic reticulum (ER) is the principal organelle responsible for correct protein folding, a step in protein synthesis that is critical for the functional conformation of proteins. ER stress is a primary feature of secretory cells and is involved in the pathogenesis of numerous human diseases, such as certain neurodegenerative and cardiometabolic disorders. The unfolded protein response (UPR) is a defense mechanism to attenuate ER stress and maintain the homeostasis of the organism. Two major degradation systems, including the proteasome and autophagy, are involved in this defense system. If ER stress overwhelms the capacity of the cell's defense mechanisms, apoptotic death may result. This review is focused on the various pharmacological modulators that can protect cells from damage induced by ER stress. The possible mechanisms for cytoprotection are also discussed.
Collapse
Affiliation(s)
- Tae Woo Jung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul 152-703, Korea.
| | - Kyung Mook Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul 152-703, Korea.
| |
Collapse
|
180
|
Song YF, Luo Z, Zhang LH, Hogstrand C, Pan YX. Endoplasmic reticulum stress and disturbed calcium homeostasis are involved in copper-induced alteration in hepatic lipid metabolism in yellow catfish Pelteobagrus fulvidraco. CHEMOSPHERE 2016; 144:2443-2453. [PMID: 26615493 DOI: 10.1016/j.chemosphere.2015.11.031] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/06/2015] [Accepted: 11/09/2015] [Indexed: 06/05/2023]
Abstract
The present study was conducted to investigate the effect of Cu exposure on ER stress and Ca(2+) homeostasis, and explore the underlying mechanism of the ER stress and disturbed Ca(2+) homeostasis in the regulation of hepatic lipid metabolism in yellow catfish Pelteobagrus fulvidraco. To this end, three experiments were conducted. In experiment 1, P. fulvidraco were exposed to three waterborne Cu concentrations for 56 days. Waterborne Cu exposure evoked ER stress and SREBP-1c activation and resulted in dysregulation of hepatic lipid metabolism in liver of P. fulvidraco in a time-dependent manner. In experiment 2, specific inhibitors 2-APB (IP3 receptor inhibitor) and dantrolene (RyR receptor inhibitor) were used to explore whether Ca(2+) release from ER was involved in the Cu-induced ER stress change. Dantrolene and 2-APB prevented Cu-induced intracellular Ca(2+) elevation, demonstrating that the release of Ca(2+) from the ER, mediated by both RyR and IP3R, contributed to dysregulation of lipid metabolism. In experiment 3, a chemical chaperone (PBA) was used to demonstrate whether Cu-induced alteration in lipid metabolism was suppressed through the attenuation of ER stress. PBA attenuated the Cu-induced elevation of mRNA expression of SREBP-1c, SCAP, ACC, FAS, GRP78/BiP, GRP94, CRT, eIF2α and XBP-1, and alleviated the Cu-induced downregulation of Insig-1. Based on these observations, these results reveal a link between ER stress and the change of lipid metabolism induced by Cu, which will help to understand the Cu-induced toxicity on cellular and molecular level, and provide some novel insights into the regulation of lipid metabolism in fish.
Collapse
Affiliation(s)
- Yu-Feng Song
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan, 430070, PR China; China and Freshwater Aquaculture Collaborative Innovative Center of Hubei Province, Wuhan, 430070, PR China
| | - Zhi Luo
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan, 430070, PR China; China and Freshwater Aquaculture Collaborative Innovative Center of Hubei Province, Wuhan, 430070, PR China.
| | - Li-Han Zhang
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan, 430070, PR China; China and Freshwater Aquaculture Collaborative Innovative Center of Hubei Province, Wuhan, 430070, PR China
| | - Christer Hogstrand
- Diabetes and Nutritional Sciences Division, School of Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
| | - Ya-Xiong Pan
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan, 430070, PR China; China and Freshwater Aquaculture Collaborative Innovative Center of Hubei Province, Wuhan, 430070, PR China
| |
Collapse
|
181
|
Kim OK, Jun W, Lee J. Effect of Cudrania tricuspidata and Kaempferol in Endoplasmic Reticulum Stress-Induced Inflammation and Hepatic Insulin Resistance in HepG2 Cells. Nutrients 2016; 8:nu8010060. [PMID: 26805878 PMCID: PMC4728671 DOI: 10.3390/nu8010060] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 01/11/2016] [Accepted: 01/13/2016] [Indexed: 02/06/2023] Open
Abstract
In this study, we quantitated kaempferol in water extract from Cudrania tricuspidata leaves (CTL) and investigated its effects on endoplasmic reticulum (ER) stress-induced inflammation and insulin resistance in HepG2 cells. The concentration of kaempferol in the CTL was 5.07 ± 0.08 mg/g. The HepG2 cells were treated with 300 µg/mL of CTL, 500 µg/mL of CTL, 1.5 µg/mL of kaempferol or 2.5 µg/mL of kaempferol, followed immediately by stimulation with 100 nM of thapsigargin for ER stress induction for 24 h. There was a marked increase in the activation of the ER stress and inflammation response in the thapsigargin-stimulated control group. The CTL treatment interrupted the ER stress response and ER stress-induced inflammation. Kaempferol partially inhibited the ER stress response and inflammation. There was a significant increase in serine phosphorylation of insulin receptor substrate (IRS)-1 and the expression of C/EBPα and gluconeogenic genes in the thapsigargin-stimulated control group compared to the normal control. Both CTL and kaempferol suppressed serine phosphorylation of IRS-1, and the treatments did not interrupt the C/EBPα/gluconeogenic gene pathway. These results suggest that kaempferol might be the active compound of CTL and that it might protect against ER stress-induced inflammation and hyperglycemia.
Collapse
Affiliation(s)
- Ok-Kyung Kim
- Department of Medical Nutrition, Kyung Hee University, Yongin 17104, Korea.
| | - Woojin Jun
- Division of Food and Nutritional Science, Chonnam National University, Gwangju 61186, Korea.
| | - Jeongmin Lee
- Department of Medical Nutrition, Kyung Hee University, Yongin 17104, Korea.
| |
Collapse
|
182
|
Zheng J, Peng C, Ai Y, Wang H, Xiao X, Li J. Docosahexaenoic Acid Ameliorates Fructose-Induced Hepatic Steatosis Involving ER Stress Response in Primary Mouse Hepatocytes. Nutrients 2016; 8:nu8010055. [PMID: 26805874 PMCID: PMC4728666 DOI: 10.3390/nu8010055] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 01/13/2016] [Accepted: 01/14/2016] [Indexed: 12/11/2022] Open
Abstract
The increase in fructose consumption is considered to be a risk factor for developing nonalcoholic fatty liver disease (NAFLD). We investigated the effects of docosahexaenoic acid (DHA) on hepatic lipid metabolism in fructose-treated primary mouse hepatocytes, and the changes of Endoplasmic reticulum (ER) stress pathways in response to DHA treatment. The hepatocytes were treated with fructose, DHA, fructose plus DHA, tunicamycin (TM) or fructose plus 4-phenylbutyric acid (PBA) for 24 h. Intracellular triglyceride (TG) accumulation was assessed by Oil Red O staining. The mRNA expression levels and protein levels related to lipid metabolism and ER stress response were determined by real-time PCR and Western blot. Fructose treatment led to obvious TG accumulation in primary hepatocytes through increasing expression of fatty acid synthase (FAS) and acetyl-CoA carboxylase (ACC), two key enzymes in hepatic de novo lipogenesis. DHA ameliorates fructose-induced TG accumulation by upregulating the expression of carnitine palmitoyltransferase 1A (CPT-1α) and acyl-CoA oxidase 1 (ACOX1). DHA treatment or pretreatment with the ER stress inhibitor PBA significantly decreased TG accumulation and reduced the expression of glucose-regulated protein 78 (GRP78), total inositol-requiring kinase 1 (IRE1α) and p-IRE1α. The present results suggest that DHA protects against high fructose-induced hepatocellular lipid accumulation. The current findings also suggest that alleviating the ER stress response seems to play a role in the prevention of fructose-induced hepatic steatosis by DHA.
Collapse
Affiliation(s)
- Jinying Zheng
- School of Public Health and Management, Chongqing Medical University, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing 400016, China.
| | - Chuan Peng
- Laboratory of Lipid & Glucose Metabolism, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Yanbiao Ai
- School of Public Health and Management, Chongqing Medical University, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing 400016, China.
| | - Heng Wang
- Laboratory of Lipid & Glucose Metabolism, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Xiaoqiu Xiao
- Laboratory of Lipid & Glucose Metabolism, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Jibin Li
- School of Public Health and Management, Chongqing Medical University, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing 400016, China.
| |
Collapse
|
183
|
Synergistic Effects of Cilostazol and Probucol on ER Stress-Induced Hepatic Steatosis via Heme Oxygenase-1-Dependent Activation of Mitochondrial Biogenesis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:3949813. [PMID: 27057275 PMCID: PMC4736599 DOI: 10.1155/2016/3949813] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 11/24/2015] [Accepted: 11/30/2015] [Indexed: 11/17/2022]
Abstract
The selective type-3 phosphodiesterase inhibitor cilostazol and the antihyperlipidemic agent probucol have antioxidative, anti-inflammatory, and antiatherogenic properties. Moreover, cilostazol and probucol can regulate mitochondrial biogenesis. However, the combinatorial effect of cilostazol and probucol on mitochondrial biogenesis remains unknown. Endoplasmic reticulum (ER) stress is a well-known causative factor of nonalcoholic fatty liver disease (NAFLD) which can impair mitochondrial function in hepatocytes. Here, we investigated the synergistic effects of cilostazol and probucol on mitochondrial biogenesis and ER stress-induced hepatic steatosis. A synergistic effect of cilostazol and probucol on HO-1 and mitochondrial biogenesis gene expression was found in human hepatocellular carcinoma cells (HepG2) and murine primary hepatocytes. Furthermore, in an animal model of ER stress involving tunicamycin, combinatorial treatment with cilostazol and probucol significantly increased the expression of HO-1 and mitochondrial biogenesis-related genes and proteins, whereas it downregulated serum ALT, eIF2 phosphorylation, and CHOP expression, as well as the lipogenesis-related genes SREBP-1c and FAS. Based on these results, we conclude that cilostazol and probucol exhibit a synergistic effect on the activation of mitochondrial biogenesis via upregulation of HO-1, which confers protection against ER stress-induced hepatic steatosis.
Collapse
|
184
|
Zheng Z, Wang G, Li L, Tseng J, Sun F, Chen X, Chang L, Heng H, Zhang K. Transcriptional signatures of unfolded protein response implicate the limitation of animal models in pathophysiological studies. ENVIRONMENTAL DISEASE 2016; 1:24-30. [PMID: 28265594 PMCID: PMC5336312 DOI: 10.4103/2468-5690.180333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background The unfolded protein response (UPR) refers to intracellular stress signaling pathways that protect cells from the stress caused by accumulation of unfolded or misfolded proteins in the endoplasmic reticulum (ER). The UPR signaling is crucially involved in the initiation and progression of a variety of human diseases by modulating transcriptional and translational programs of the stressed cells. In this study, we analyzed the gene expression signatures of primary stress sensors and major mediators of UPR pathways in a variety of tissues/organs of human and murine species. Methods We first analyzed protein sequence similarities of major UPR transducers and mediators of human and murine species, and then examined their gene expression profiles in 26 human and mouse common tissues based on the microarray datasets of public domains. The differential expression patterns of the UPR genes in human diseases were delineated. The involvements of the UPR genes in mouse pathology were also analyzed with mouse gene knockout models. Results The results indicated that expression patterns and pathophysiologic involvements of the major UPR stress sensors and mediators significantly differ in 26 common tissues/organs of human and murine species. Gene expression profiles suggest that the IRE1α/XBP1-mediated UPR pathway is induced in secretory and metabolic tissues or organs. While deletion of the UPR trans-activator XBP1 leads to pathological phenotypes in mice, alteration in XBP1 is less associated with human disease conditions. Conclusions Expression signatures of the major UPR genes differ among tissues or organs and among human and mouse species. The differential induction of the UPR pathways reflects the pathophysiologic differences of tissues or organs. The difference in UPR induction between human and mouse suggests the limitation of using animal models to study human pathophysiology or drugology associated with environmental stress.
Collapse
Affiliation(s)
- Ze Zheng
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201
| | - Guohui Wang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201; Department of Internal Medicine, The Affiliated Tumor Hospital of Zhengzhou University, Jinshui, Zhengzhou, Henan, China
| | - Li Li
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201
| | - Jeffery Tseng
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201
| | - Fei Sun
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Xuequn Chen
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Lin Chang
- Cardiovascular Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Henry Heng
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201; Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI 48201
| |
Collapse
|
185
|
White MF, Copps KD. The Mechanisms of Insulin Action. ENDOCRINOLOGY: ADULT AND PEDIATRIC 2016:556-585.e13. [DOI: 10.1016/b978-0-323-18907-1.00033-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
186
|
An inhibitor of HIV-1 protease modulates constitutive eIF2α dephosphorylation to trigger a specific integrated stress response. Proc Natl Acad Sci U S A 2015; 113:E117-26. [PMID: 26715744 DOI: 10.1073/pnas.1514076113] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Inhibitors of the HIV aspartyl protease [HIV protease inhibitors (HIV-PIs)] are the cornerstone of treatment for HIV. Beyond their well-defined antiretroviral activity, these drugs have additional effects that modulate cell viability and homeostasis. However, little is known about the virus-independent pathways engaged by these molecules. Here we show that the HIV-PI Nelfinavir decreases translation rates and promotes a transcriptional program characteristic of the integrated stress response (ISR). Mice treated with Nelfinavir display hallmarks of this stress response in the liver, including α subunit of translation initiation factor 2 (eIF2α) phosphorylation, activating transcription factor-4 (ATF4) induction, and increased expression of known downstream targets. Mechanistically, Nelfinavir-mediated ISR bypassed direct activation of the eIF2α stress kinases and instead relied on the inhibition of the constitutive eIF2α dephosphorylation and down-regulation of the phophatase cofactor CReP (Constitutive Repressor of eIF2α Phosphorylation; also known as PPP1R15B). These findings demonstrate that the modulation of eIF2α-specific phosphatase cofactor activity can be a rheostat of cellular homeostasis that initiates a functional ISR and suggest that the HIV-PIs could be repositioned as therapeutics in human diseases to modulate translation rates and stress responses.
Collapse
|
187
|
Undernourishment in utero Primes Hepatic Steatosis in Adult Mice Offspring on an Obesogenic Diet; Involvement of Endoplasmic Reticulum Stress. Sci Rep 2015; 5:16867. [PMID: 26581663 PMCID: PMC4652266 DOI: 10.1038/srep16867] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/21/2015] [Indexed: 01/29/2023] Open
Abstract
In order to investigate the possible involvement of endoplasmic reticulum (ER) stress in the developmental origins of hepatic steatosis associated with undernourishment in utero, we herein employed a fetal undernourishment mouse model by maternal caloric restriction in three cohorts; cohort 1) assessment of hepatic steatosis and the ER stress response at 9 weeks of age (wks) before a high fat diet (HFD), cohort 2) assessment of hepatic steatosis and the ER stress response on a HFD at 17 wks, cohort 3) assessment of hepatic steatosis and the ER stress response at 22 wks on a HFD after the alleviation of ER stress with a chemical chaperone, tauroursodeoxycholic acid (TUDCA), from 17 wks to 22 wks. Undernourishment in utero significantly deteriorated hepatic steatosis and led to the significant integration of the ER stress response on a HFD at 17 wks. The alleviation of ER stress by the TUDCA treatment significantly improved the parameters of hepatic steatosis in pups with undernourishment in utero, but not in those with normal nourishment in utero at 22 wks. These results suggest the pivotal involvement of the integration of ER stress in the developmental origins of hepatic steatosis in association with undernourishment in utero.
Collapse
|
188
|
The Role of Organelle Stresses in Diabetes Mellitus and Obesity: Implication for Treatment. Anal Cell Pathol (Amst) 2015; 2015:972891. [PMID: 26613076 PMCID: PMC4646985 DOI: 10.1155/2015/972891] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 10/08/2015] [Indexed: 12/17/2022] Open
Abstract
The type 2 diabetes pandemic in recent decades is a huge global health threat. This pandemic is primarily attributed to the surplus of nutrients and the increased prevalence of obesity worldwide. In contrast, calorie restriction and weight reduction can drastically prevent type 2 diabetes, indicating a central role of nutrient excess in the development of diabetes. Recently, the molecular links between excessive nutrients, organelle stress, and development of metabolic disease have been extensively studied. Specifically, excessive nutrients trigger endoplasmic reticulum stress and increase the production of mitochondrial reactive oxygen species, leading to activation of stress signaling pathway, inflammatory response, lipogenesis, and pancreatic beta-cell death. Autophagy is required for clearance of hepatic lipid clearance, alleviation of pancreatic beta-cell stress, and white adipocyte differentiation. ROS scavengers, chemical chaperones, and autophagy activators have demonstrated promising effects for the treatment of insulin resistance and diabetes in preclinical models. Further results from clinical trials are eagerly awaited.
Collapse
|
189
|
Kim OK, Nam DE, Jun W, Lee J. Cudrania tricuspidata water extract improved obesity-induced hepatic insulin resistance in db/db mice by suppressing ER stress and inflammation. Food Nutr Res 2015; 59:29165. [PMID: 26507490 PMCID: PMC4623289 DOI: 10.3402/fnr.v59.29165] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 10/04/2015] [Accepted: 10/05/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Obesity can play a role in the development of hepatic insulin resistance. Although the molecular mechanism of the association between obesity and hepatic insulin resistance is unclear, it has been reported that obesity leads to hepatic endoplasmic reticulum (ER) stress and inflammation, which can induce the development of insulin resistance in several tissues. OBJECTIVE In this study, we investigated the associations between hepatic insulin resistance, ER, and inflammation in obesity and the effect of water extract from Cudrania tricuspidata leaves (CTL) on hepatic insulin resistance induced by ER stress and inflammation in db/db mice. DESIGN The mice were randomly divided into four groups: a normal control group (C57BL/6J), a control group (C57BL/6J-db/db), a CTL 100 group (C57BL/6J-db/db mice fed a dietary supplement of 100 mg/kg of CTL), and a CTL 300 group (C57BL/6J-db/db mice fed a dietary supplement of 300 mg/kg of CTL). After 8 weeks, we performed an oral glucose tolerance test and the mice were sacrificed. RESULTS The C57BL/6J-db/db mice developed obesity and hyperglycemia, and the ER stress response and inflammation were activated in their livers. Interestingly, there was a marked decrease in the activation of the ER stress response and insulin resistance in the livers of the C57BL/6J-db/db mice treated with CTL due to decreases in the phosphorylation of eIF2α, IRE1α, and IRS-1 serine and decreases in the mRNA expression of ATF4, c-Jun N-terminal kinase, C/EBPα, and C/EBP homologous protein. Dietary supplementation with CTL also induced a statistically significant decrease in the expression of pro-inflammatory cytokines, C-reactive protein (CRP), and NF-κB phosphorylation. CONCLUSIONS Overall, these results suggest that CTL can improve hepatic insulin resistance and hyperglycemia by controlling obesity-induced ER stress and inflammation in the liver and that CTL may be a useful agent in treating hyperglycemia.
Collapse
Affiliation(s)
- Ok-Kyung Kim
- Department of Medical Nutrition, Graduate School of East-West Science, Kyung Hee University, Yongin, Republic of Korea
| | - Da-Eun Nam
- Department of Medical Nutrition, Graduate School of East-West Science, Kyung Hee University, Yongin, Republic of Korea
| | - Woojin Jun
- Division of Food and Nutrition, Chonnam National University, Gwangju, Republic of Korea
| | - Jeongmin Lee
- Department of Medical Nutrition, Graduate School of East-West Science, Kyung Hee University, Yongin, Republic of Korea;
| |
Collapse
|
190
|
Miyake M, Nomura A, Ogura A, Takehana K, Kitahara Y, Takahara K, Tsugawa K, Miyamoto C, Miura N, Sato R, Kurahashi K, Harding HP, Oyadomari M, Ron D, Oyadomari S. Skeletal muscle-specific eukaryotic translation initiation factor 2α phosphorylation controls amino acid metabolism and fibroblast growth factor 21-mediated non-cell-autonomous energy metabolism. FASEB J 2015; 30:798-812. [PMID: 26487695 PMCID: PMC4945323 DOI: 10.1096/fj.15-275990] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 10/13/2015] [Indexed: 01/02/2023]
Abstract
The eukaryotic translation initiation factor 2α (eIF2α) phosphorylation-dependent integrated stress response (ISR), a component of the unfolded protein response, has long been known to regulate intermediary metabolism, but the details are poorly worked out. We report that profiling of mRNAs of transgenic mice harboring a ligand-activated skeletal muscle-specific derivative of the eIF2α protein kinase R-like ER kinase revealed the expected up-regulation of genes involved in amino acid biosynthesis and transport but also uncovered the induced expression and secretion of a myokine, fibroblast growth factor 21 (FGF21), that stimulates energy consumption and prevents obesity. The link between the ISR and FGF21 expression was further reinforced by the identification of a small-molecule ISR activator that promoted Fgf21 expression in cell-based screens and by implication of the ISR-inducible activating transcription factor 4 in the process. Our findings establish that eIF2α phosphorylation regulates not only cell-autonomous proteostasis and amino acid metabolism, but also affects non-cell-autonomous metabolic regulation by induced expression of a potent myokine.
Collapse
Affiliation(s)
- Masato Miyake
- *Division of Molecular Biology, Institute for Genome Research, and Department of Molecular Research, Diabetes Therapeutics and Research Center, The University of Tokushima, Tokushima, Japan; Exploratory Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Company, Limited, Kanagawa, Japan; and Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Akitoshi Nomura
- *Division of Molecular Biology, Institute for Genome Research, and Department of Molecular Research, Diabetes Therapeutics and Research Center, The University of Tokushima, Tokushima, Japan; Exploratory Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Company, Limited, Kanagawa, Japan; and Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Atsushi Ogura
- *Division of Molecular Biology, Institute for Genome Research, and Department of Molecular Research, Diabetes Therapeutics and Research Center, The University of Tokushima, Tokushima, Japan; Exploratory Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Company, Limited, Kanagawa, Japan; and Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Kenji Takehana
- *Division of Molecular Biology, Institute for Genome Research, and Department of Molecular Research, Diabetes Therapeutics and Research Center, The University of Tokushima, Tokushima, Japan; Exploratory Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Company, Limited, Kanagawa, Japan; and Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Yoshihiro Kitahara
- *Division of Molecular Biology, Institute for Genome Research, and Department of Molecular Research, Diabetes Therapeutics and Research Center, The University of Tokushima, Tokushima, Japan; Exploratory Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Company, Limited, Kanagawa, Japan; and Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Kazuna Takahara
- *Division of Molecular Biology, Institute for Genome Research, and Department of Molecular Research, Diabetes Therapeutics and Research Center, The University of Tokushima, Tokushima, Japan; Exploratory Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Company, Limited, Kanagawa, Japan; and Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Kazue Tsugawa
- *Division of Molecular Biology, Institute for Genome Research, and Department of Molecular Research, Diabetes Therapeutics and Research Center, The University of Tokushima, Tokushima, Japan; Exploratory Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Company, Limited, Kanagawa, Japan; and Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Chinobu Miyamoto
- *Division of Molecular Biology, Institute for Genome Research, and Department of Molecular Research, Diabetes Therapeutics and Research Center, The University of Tokushima, Tokushima, Japan; Exploratory Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Company, Limited, Kanagawa, Japan; and Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Naoko Miura
- *Division of Molecular Biology, Institute for Genome Research, and Department of Molecular Research, Diabetes Therapeutics and Research Center, The University of Tokushima, Tokushima, Japan; Exploratory Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Company, Limited, Kanagawa, Japan; and Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Ryosuke Sato
- *Division of Molecular Biology, Institute for Genome Research, and Department of Molecular Research, Diabetes Therapeutics and Research Center, The University of Tokushima, Tokushima, Japan; Exploratory Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Company, Limited, Kanagawa, Japan; and Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Kiyoe Kurahashi
- *Division of Molecular Biology, Institute for Genome Research, and Department of Molecular Research, Diabetes Therapeutics and Research Center, The University of Tokushima, Tokushima, Japan; Exploratory Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Company, Limited, Kanagawa, Japan; and Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Heather P Harding
- *Division of Molecular Biology, Institute for Genome Research, and Department of Molecular Research, Diabetes Therapeutics and Research Center, The University of Tokushima, Tokushima, Japan; Exploratory Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Company, Limited, Kanagawa, Japan; and Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Miho Oyadomari
- *Division of Molecular Biology, Institute for Genome Research, and Department of Molecular Research, Diabetes Therapeutics and Research Center, The University of Tokushima, Tokushima, Japan; Exploratory Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Company, Limited, Kanagawa, Japan; and Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - David Ron
- *Division of Molecular Biology, Institute for Genome Research, and Department of Molecular Research, Diabetes Therapeutics and Research Center, The University of Tokushima, Tokushima, Japan; Exploratory Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Company, Limited, Kanagawa, Japan; and Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Seiichi Oyadomari
- *Division of Molecular Biology, Institute for Genome Research, and Department of Molecular Research, Diabetes Therapeutics and Research Center, The University of Tokushima, Tokushima, Japan; Exploratory Research Laboratories, Research Center, Ajinomoto Pharmaceuticals Company, Limited, Kanagawa, Japan; and Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
191
|
Lebeaupin C, Proics E, de Bieville CHD, Rousseau D, Bonnafous S, Patouraux S, Adam G, Lavallard VJ, Rovere C, Le Thuc O, Saint-Paul MC, Anty R, Schneck AS, Iannelli A, Gugenheim J, Tran A, Gual P, Bailly-Maitre B. ER stress induces NLRP3 inflammasome activation and hepatocyte death. Cell Death Dis 2015; 6:e1879. [PMID: 26355342 PMCID: PMC4650444 DOI: 10.1038/cddis.2015.248] [Citation(s) in RCA: 316] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 07/17/2015] [Accepted: 07/21/2015] [Indexed: 01/18/2023]
Abstract
The incidence of chronic liver disease is constantly increasing, owing to the obesity epidemic. However, the causes and mechanisms of inflammation-mediated liver damage remain poorly understood. Endoplasmic reticulum (ER) stress is an initiator of cell death and inflammatory mechanisms. Although obesity induces ER stress, the interplay between hepatic ER stress, NLRP3 inflammasome activation and hepatocyte death signaling has not yet been explored during the etiology of chronic liver diseases. Steatosis is a common disorder affecting obese patients; moreover, 25% of these patients develop steatohepatitis with an inherent risk for progression to hepatocarcinoma. Increased plasma LPS levels have been detected in the serum of patients with steatohepatitis. We hypothesized that, as a consequence of increased plasma LPS, ER stress could be induced and lead to NLRP3 inflammasome activation and hepatocyte death associated with steatohepatitis progression. In livers from obese mice, administration of LPS or tunicamycin results in IRE1α and PERK activation, leading to the overexpression of CHOP. This, in turn, activates the NLRP3 inflammasome, subsequently initiating hepatocyte pyroptosis (caspase-1, -11, interleukin-1β secretion) and apoptosis (caspase-3, BH3-only proteins). In contrast, the LPS challenge is blocked by the ER stress inhibitor TUDCA, resulting in: CHOP downregulation, reduced caspase-1, caspase-11, caspase-3 activities, lowered interleukin-1β secretion and rescue from cell death. The central role of CHOP in mediating the activation of proinflammatory caspases and cell death was characterized by performing knockdown experiments in primary mouse hepatocytes. Finally, the analysis of human steatohepatitis liver biopsies showed a correlation between the upregulation of inflammasome and ER stress markers, as well as liver injury. We demonstrate here that ER stress leads to hepatic NLRP3 inflammasome pyroptotic death, thus contributing as a novel mechanism of inflammation-mediated liver injury in chronic liver diseases. Inhibition of ER-dependent inflammasome activation and cell death pathways may represent a potential therapeutic approach in chronic liver diseases.
Collapse
Affiliation(s)
- C Lebeaupin
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France
| | - E Proics
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France
| | - C H D de Bieville
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France
| | - D Rousseau
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France
| | - S Bonnafous
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France.,Centre Hospitalier Universitaire Nice, Hôpital l'Archet, Département Digestif, Nice, France
| | - S Patouraux
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France.,Centre Hospitalier Universitaire Nice, Hôpital l'Archet, Département Biologie, Nice, France
| | - G Adam
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France
| | - V J Lavallard
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France
| | - C Rovere
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, UMR7275, Valbonne, France
| | - O Le Thuc
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, UMR7275, Valbonne, France
| | - M C Saint-Paul
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France.,Centre Hospitalier Universitaire Nice, Hôpital l'Archet, Département Digestif, Nice, France
| | - R Anty
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France.,Centre Hospitalier Universitaire Nice, Hôpital l'Archet, Département Digestif, Nice, France
| | - A S Schneck
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France.,Centre Hospitalier Universitaire Nice, Hôpital l'Archet, Département Digestif, Nice, France
| | - A Iannelli
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France.,Centre Hospitalier Universitaire Nice, Hôpital l'Archet, Département Digestif, Nice, France
| | - J Gugenheim
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France.,Centre Hospitalier Universitaire Nice, Hôpital l'Archet, Département Digestif, Nice, France
| | - A Tran
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France.,Centre Hospitalier Universitaire Nice, Hôpital l'Archet, Département Digestif, Nice, France
| | - P Gual
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France
| | - B Bailly-Maitre
- INSERM, U1065, Equipe 8 « Complications hépatiques de l'obésité », Bâtiment Universitaire ARCHIMED, Nice, France.,Université de Nice Sophia Antipolis, Faculté de Médecine, Nice, France
| |
Collapse
|
192
|
Nishio N, Isobe KI. GADD34-deficient mice develop obesity, nonalcoholic fatty liver disease, hepatic carcinoma and insulin resistance. Sci Rep 2015; 5:13519. [PMID: 26316333 PMCID: PMC4551985 DOI: 10.1038/srep13519] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 07/15/2015] [Indexed: 01/04/2023] Open
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) is increasing in parallel with the prevalence of obesity. DNA damage-inducible protein 34 (GADD34/Ppp1r15a), originally isolated from UV-inducible transcripts in Chinese hamster ovary (CHO) cells, dephosphorylates several kinases that function in important signaling cascades, including dephosphorylation of eIF2α. We examined the effects of GADD34 on natural life span by using GADD34-deficient mice. Here we observed for the first time that with age GADD34-deficient mice become obese, developing fatty liver followed by liver cirrhosis, hepatocellular carcinoma, and insulin resistance. We found that myofibroblasts and immune cells infiltrated the portal veins of aged GADD34-deficient mouse livers. A high-fat diet (HFD) induced a higher level of steatosis in young GADD34-deficient mice compared with WT mice. Differentiation into fat is dependent on insulin signaling. Insulin signaling in young GADD34-deficient mice was higher than that in WT mice, which explained the higher fat differentiation of mouse embryonic fibroblasts (MEFs) observed in GADD34-deficient mice. Through aging or a HFD, insulin signaling in GADD34-deficient liver converted to be down regulated compared with WT mice. We found that a HFD or palmitate treatment converted insulin signaling by up-regulating TNF-α and JNK.
Collapse
Affiliation(s)
- Naomi Nishio
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8550
| | - Ken-ichi Isobe
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi, 466-8550.,Department of Food Science and Nutrition, Nagoya woman's university, 3-40 Shioji-cho, Mizuho-ku, Nagoya, Aichi, 467-0003 Japan
| |
Collapse
|
193
|
Ashraf NU, Sheikh TA. Endoplasmic reticulum stress and Oxidative stress in the pathogenesis of Non-alcoholic fatty liver disease. Free Radic Res 2015. [PMID: 26223319 DOI: 10.3109/10715762.2015.1078461] [Citation(s) in RCA: 230] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the hepatic manifestation of metabolic syndrome. The underlying causes of the disease progression in NAFLD are unclear. Recent evidences suggest endoplasmic reticulum stress in the development of lipid droplets (steatosis) and subsequent generation of reactive oxygen species (ROS) in the progression to non-alcoholic steatohepatitis (NASH). The signalling pathway activated by disruption of endoplasmic reticulum (ER) homoeostasis, called as unfolded protein response, is linked with membrane biosynthesis, insulin action, inflammation and apoptosis. ROS are important mediators of inflammation. Protein folding in ER is linked to ROS. Therefore understanding the basic mechanisms that lead to ER stress and ROS in NAFLD have become the topics of immense interest. The present review focuses on the role of ER stress and ROS in the pathogenesis of NAFLD. We also highlight the cross talk between ER stress and oxidative stress which suggest and encourage the development of therapeutics for NAFLD. Further we have reviewed various strategies used for the management of NAFLD/NASH and limitations of such strategies. Our review therefore highlights the need for newer strategies with regards to ER stress and oxidative stress.
Collapse
Affiliation(s)
- N U Ashraf
- a Academy of Scientific and Innovative Research (AcSIR) , New Delhi , India.,b PK-PD and Toxicology Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu Tawi , Jammu and Kashmir , India
| | - T A Sheikh
- a Academy of Scientific and Innovative Research (AcSIR) , New Delhi , India.,b PK-PD and Toxicology Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu Tawi , Jammu and Kashmir , India
| |
Collapse
|
194
|
Endoplasmic Reticulum Stress-Related Genes in Yellow Catfish Pelteobagrus fulvidraco: Molecular Characterization, Tissue Expression, and Expression Responses to Dietary Copper Deficiency and Excess. G3-GENES GENOMES GENETICS 2015; 5:2091-104. [PMID: 26276384 PMCID: PMC4592992 DOI: 10.1534/g3.115.019950] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Two endoplasmic reticulum (ER) molecular chaperones [glucose-regulated protein 78 (grp78) and calreticulin (crt)] and three ER stress sensors [PKR-like ER kinase (perk), inositol requiring enzyme (ire)-1α, and activating transcription factor (atf)-6α] cDNAs were first characterized from yellow catfish, Pelteobagrus fulvidraco. The predicted amino acid sequences for the yellow catfish grp78, crt, perk, ire-1α, and atf-6α revealed that the proteins contained all of the structural features that were characteristic of the five genes in other species, including the KDEL motif, signal peptide, sensor domain, and effector domain. mRNAs of the five genes mentioned above were expressed in various tissues, but their mRNA levels varied among tissues. Dietary Cu excess, but not Cu deficiency, activated the chaperones (grp78 and crt) and folding sensors in ER, and the UPR signaling pathways (i.e., perk–eif2α and the ire1–xbp1) in a tissue-specific manner. For the first time, our study cloned grp78, crt, perk, ire-1α, and atf-6α genes in yellow catfish and demonstrated their differential expression among tissues. Moreover, the present study also indicated differential regulation of these ER stress–related genes by dietary Cu deficiency and excess, which will be beneficial for us to evaluate effects of dietary Cu levels in fish at the molecular level, based on the upstream pathway of lipid metabolism (the ER) and thus provide novel insights regarding the nutrition of Cu in fish.
Collapse
|
195
|
Screening a mouse liver gene expression compendium identifies modulators of the aryl hydrocarbon receptor (AhR). Toxicology 2015. [PMID: 26215100 DOI: 10.1016/j.tox.2015.07.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that mediates the biological and toxic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), dioxin-like compounds (DLC) as well as some drugs and endogenous tryptophan metabolites. Short-term activation of AhR can lead to hepatocellular steatosis, and chronic activation can lead to liver cancer in mice and rats. Analytical approaches were developed to identify biosets in a genomic database in which AhR activity was altered. A set of 63 genes was identified (the AhR gene expression biomarker) that was dependent on AhR for regulation after exposure to TCDD or benzo[a]pyrene and includes the known AhR targets Cyp1a1 and Cyp1b1. A fold-change rank-based test (Running Fisher's test; p-value ≤ 10(-4)) was used to evaluate the similarity between the AhR biomarker and a test set of 37 and 41 biosets positive or negative, respectively for AhR activation. The test resulted in a balanced accuracy of 95%. The rank-based test was used to identify factors that activate or suppress AhR in an annotated mouse liver/mouse primary hepatocyte gene expression database of ∼ 1850 comparisons. In addition to the expected activation of AhR by TCDD and DLC, AhR was activated by AP20189 and phenformin. AhR was suppressed by phenobarbital and 1,4-Bis[2-(3,5-dichloropyridyloxy)] benzene (TCPOBOP) in a constitutive activated receptor (CAR)-dependent manner and pregnenolone-16α-carbonitrile in a pregnane X receptor (PXR)-dependent manner. Inactivation of individual genes in nullizygous models led to AhR activation (Pxr, Ghrhr, Taf10) or suppression (Ahr, Ilst6st, Hnf1a). This study describes a novel screening strategy for identifying factors in mouse liver that perturb AhR in a gene expression compendium.
Collapse
|
196
|
Pan QR, Ren YL, Liu WX, Hu YJ, Zheng JS, Xu Y, Wang G. Resveratrol prevents hepatic steatosis and endoplasmic reticulum stress and regulates the expression of genes involved in lipid metabolism, insulin resistance, and inflammation in rats. Nutr Res 2015; 35:576-84. [PMID: 26055348 DOI: 10.1016/j.nutres.2015.05.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 05/04/2015] [Accepted: 05/14/2015] [Indexed: 02/08/2023]
Abstract
Previous research demonstrated that resveratrol possesses promising properties for preventing obesity. Endoplasmic reticulum (ER) stress was proposed to be involved in the pathophysiology of both obesity and hepatic steatosis. In the current study, we hypothesized that resveratrol could protect against high-fat diet (HFD)-induced hepatic steatosis and ER stress and regulate the expression of genes related to hepatic steatosis. Rats were fed either a control diet or a HFD for 12 weeks. After 4 weeks, HFD-fed rats were treated with either resveratrol or vehicle for 8 weeks. Body weight, serum metabolic parameters, hepatic histopathology, and hepatic ER stress markers were evaluated. Moreover, an RT2 Profiler Fatty Liver PCR Array was performed to investigate the mRNA expressions of 84 genes related to hepatic steatosis. Our work showed that resveratrol prevented dyslipidemia and hepatic steatosis induced by HFD. Resveratrol significantly decreased activating transcription factor 4, C/EBP-homologous protein and immunoglobulin binding protein levels, which were elevated by the HFD. Resveratrol also decreased PKR-like ER kinase phosphorylation, although it was not affected by the HFD. Furthermore, resveratrol increased the expression of peroxisome proliferator-activated receptor δ, while decreasing the expression of ATP citrate lyase, suppressor of cytokine signaling-3, and interleukin-1β. Our data suggest that resveratrol can prevent hepatic ER stress and regulate the expression of peroxisome proliferator-activated receptor δ, ATP citrate lyase, suppressor of cytokine signaling-3, tumor necrosis factor α, and interleukin-1β in diet-induced obese rats, and these effects likely contribute to resveratrol's protective function against excessive accumulation of fat in the liver.
Collapse
Affiliation(s)
- Qing-Rong Pan
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yan-Long Ren
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Wen-Xian Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Yan-Jin Hu
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jin-Su Zheng
- Department of Traditional Chinese medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yuan Xu
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Guang Wang
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
197
|
Nakamura T, Kunz RC, Zhang C, Kimura T, Yuan CL, Baccaro B, Namiki Y, Gygi SP, Hotamisligil GS. A critical role for PKR complexes with TRBP in Immunometabolic regulation and eIF2α phosphorylation in obesity. Cell Rep 2015; 11:295-307. [PMID: 25843719 DOI: 10.1016/j.celrep.2015.03.021] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 01/14/2015] [Accepted: 03/07/2015] [Indexed: 01/09/2023] Open
Abstract
Aberrant stress and inflammatory responses are key factors in the pathogenesis of obesity and metabolic dysfunction, and the double-stranded RNA-dependent kinase (PKR) has been proposed to play an important role in integrating these pathways. Here, we report the formation of a complex between PKR and TAR RNA-binding protein (TRBP) during metabolic and obesity-induced stress, which is critical for the regulation of eukaryotic translation initiation factor 2 alpha (eIF2α) phosphorylation and c-Jun N-terminal kinase (JNK) activation. We show that TRBP phosphorylation is induced in the setting of metabolic stress, leading to PKR activation. Suppression of hepatic TRBP reduced inflammation, JNK activity, and eIF2α phosphorylation and improved systemic insulin resistance and glucose metabolism, while TRBP overexpression exacerbated the impairment in glucose homeostasis in obese mice. These data indicate that the association between PKR and TRBP integrates metabolism with translational control and inflammatory signaling and plays important roles in metabolic homeostasis and disease.
Collapse
Affiliation(s)
- Takahisa Nakamura
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Division of Endocrinology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Ryan C Kunz
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Cai Zhang
- Division of Endocrinology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Taishi Kimura
- Division of Endocrinology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Celvie L Yuan
- Division of Endocrinology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Brenna Baccaro
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Yuka Namiki
- Biomedicinal Information Research Center, National Institute of Advanced Industrial Science and Technology, Tokyo 135-0063, Japan
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Gökhan S Hotamisligil
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Harvard-MIT Broad Institute, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.
| |
Collapse
|
198
|
Inaba Y, Furutani T, Kimura K, Watanabe H, Haga S, Kido Y, Matsumoto M, Yamamoto Y, Harada K, Kaneko S, Oyadomari S, Ozaki M, Kasuga M, Inoue H. Growth arrest and DNA damage-inducible 34 regulates liver regeneration in hepatic steatosis in mice. Hepatology 2015; 61:1343-56. [PMID: 25420998 DOI: 10.1002/hep.27619] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 11/19/2014] [Indexed: 12/24/2022]
Abstract
UNLABELLED The liver has robust regenerative potential in response to damage, but hepatic steatosis (HS) weakens this potential. We found that the enhanced integrated stress response (ISR) mediated by phosphorylation of the alpha subunit of eukaryotic initiation factor 2 (eIF2α) impairs regeneration in HS and that growth arrest and DNA damage-inducible 34 (Gadd34)-dependent suppression of ISR plays a crucial role in fatty liver regeneration. Although mice fed a high-fat diet for 2 weeks developed moderate fatty liver with no increase in eIF2α phosphorylation before 70% hepatectomy, they showed impaired liver regeneration as a result of reduced proliferation and increased death of hepatocytes with increased phosphorylation of eIF2α and ISR. An increased ISR through Gadd34 knockdown induced C/EBP homologous protein (CHOP)-dependent apoptosis and receptor-interacting protein kinase 3-dependent necrosis, resulting in increased hepatocyte death during fatty liver regeneration. Furthermore, Gadd34 knockdown and increased phosphorylation of eIF2α decreased cyclin D1 protein and reduced hepatocyte proliferation. In contrast, enhancement of Gadd34 suppressed phosphorylation of eIF2α and reduced CHOP expression and hepatocyte apoptosis without affecting hepatocyte proliferation, clearly improving fatty liver regeneration. In more severe fatty liver of leptin receptor-deficient db/db mice, forced expression of hepatic Gadd34 also promoted hepatic regeneration after hepatectomy. CONCLUSION Gadd34-mediated regulation of ISR acts as a physiological defense mechanism against impaired liver regeneration resulting from steatosis and is thus a possible therapeutic target for impaired regeneration in HS.
Collapse
Affiliation(s)
- Yuka Inaba
- Department of Physiology and Metabolism, Brain/Liver Interface Medicine Research Center, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
199
|
Giampietri C, Petrungaro S, Conti S, Facchiano A, Filippini A, Ziparo E. c-Flip KO fibroblasts display lipid accumulation associated with endoplasmic reticulum stress. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:929-36. [PMID: 25746012 DOI: 10.1016/j.bbalip.2015.02.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 02/04/2015] [Accepted: 02/26/2015] [Indexed: 12/01/2022]
Abstract
c-Flip proteins are well-known apoptosis modulators. They generally contribute to tissue homeostasis maintenance by inhibiting death-receptor-mediated cell death. In the present manuscript, we show that c-Flip knock-out (KO) mouse embryonic fibroblasts (MEFs) kept in culture under starvation conditions gradually modify their phenotype and accumulate vacuoles, becoming progressively larger according to the duration of starvation. Large vacuoles are present in KO MEFs though not in WT MEFs, and are Oil Red-O positive, which indicates that they represent lipid droplets. Western blot experiments reveal that, unlike WT MEFs, KO MEFs express high levels of the lipogenic transcription factor PPAR-γ. Lipid droplet accumulation was found to be associated with endoplasmic reticulum (ER) stress activation and autophagic modulation valuated by means of BIP increase, LC3 lipidation and AMP-activated protein kinase (AMPK) phosphorylation, and p62 accumulation. Interestingly, XBP-1, an ER stress-induced lipogenic transcription factor, was found to preferentially localize in the nucleus rather than in the cytoplasm of KO MEFs. These data demonstrate that, upon starvation, c-Flip affects lipid accumulation, ER stress and autophagy, thereby pointing to an important role of c-Flip in the adaptive response and ER stress response programs under both normal and pathological conditions.
Collapse
Affiliation(s)
- Claudia Giampietri
- Istituto Pasteur-Fondazione Cenci Bolognetti, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Section of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy.
| | - Simonetta Petrungaro
- Istituto Pasteur-Fondazione Cenci Bolognetti, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Section of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Silvia Conti
- Istituto Pasteur-Fondazione Cenci Bolognetti, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Section of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | | | - Antonio Filippini
- Istituto Pasteur-Fondazione Cenci Bolognetti, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Section of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Elio Ziparo
- Istituto Pasteur-Fondazione Cenci Bolognetti, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Section of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
200
|
Oshida K, Vasani N, Thomas RS, Applegate D, Rosen M, Abbott B, Lau C, Guo G, Aleksunes LM, Klaassen C, Corton JC. Identification of modulators of the nuclear receptor peroxisome proliferator-activated receptor α (PPARα) in a mouse liver gene expression compendium. PLoS One 2015; 10:e0112655. [PMID: 25689681 PMCID: PMC4331523 DOI: 10.1371/journal.pone.0112655] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 10/09/2014] [Indexed: 12/22/2022] Open
Abstract
The nuclear receptor family member peroxisome proliferator-activated receptor α (PPARα) is activated by therapeutic hypolipidemic drugs and environmentally-relevant chemicals to regulate genes involved in lipid transport and catabolism. Chronic activation of PPARα in rodents increases liver cancer incidence, whereas suppression of PPARα activity leads to hepatocellular steatosis. Analytical approaches were developed to identify biosets (i.e., gene expression differences between two conditions) in a genomic database in which PPARα activity was altered. A gene expression signature of 131 PPARα-dependent genes was built using microarray profiles from the livers of wild-type and PPARα-null mice after exposure to three structurally diverse PPARα activators (WY-14,643, fenofibrate and perfluorohexane sulfonate). A fold-change rank-based test (Running Fisher’s test (p-value ≤ 10-4)) was used to evaluate the similarity between the PPARα signature and a test set of 48 and 31 biosets positive or negative, respectively for PPARα activation; the test resulted in a balanced accuracy of 98%. The signature was then used to identify factors that activate or suppress PPARα in an annotated mouse liver/primary hepatocyte gene expression compendium of ~1850 biosets. In addition to the expected activation of PPARα by fibrate drugs, di(2-ethylhexyl) phthalate, and perfluorinated compounds, PPARα was activated by benzofuran, galactosamine, and TCDD and suppressed by hepatotoxins acetaminophen, lipopolysaccharide, silicon dioxide nanoparticles, and trovafloxacin. Additional factors that activate (fasting, caloric restriction) or suppress (infections) PPARα were also identified. This study 1) developed methods useful for future screening of environmental chemicals, 2) identified chemicals that activate or suppress PPARα, and 3) identified factors including diets and infections that modulate PPARα activity and would be hypothesized to affect chemical-induced PPARα activity.
Collapse
Affiliation(s)
- Keiyu Oshida
- National Health and Environmental Effects Research Lab, US-EPA, Research Triangle Park, North Carolina, United States of America
| | - Naresh Vasani
- National Health and Environmental Effects Research Lab, US-EPA, Research Triangle Park, North Carolina, United States of America
| | - Russell S. Thomas
- Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina, United States of America
| | - Dawn Applegate
- RegeneMed, San Diego, California, United States of America
| | - Mitch Rosen
- National Health and Environmental Effects Research Lab, US-EPA, Research Triangle Park, North Carolina, United States of America
| | - Barbara Abbott
- National Health and Environmental Effects Research Lab, US-EPA, Research Triangle Park, North Carolina, United States of America
| | - Christopher Lau
- National Health and Environmental Effects Research Lab, US-EPA, Research Triangle Park, North Carolina, United States of America
| | - Grace Guo
- Rutgers University, Ernest Mario School of Pharmacy, Department of Pharmacology and Toxicology, Piscataway, New Jersey, United States of America
| | - Lauren M. Aleksunes
- Rutgers University, Ernest Mario School of Pharmacy, Department of Pharmacology and Toxicology, Piscataway, New Jersey, United States of America
| | - Curtis Klaassen
- University of Washington, Seattle, Washington, United States of America
| | - J. Christopher Corton
- National Health and Environmental Effects Research Lab, US-EPA, Research Triangle Park, North Carolina, United States of America
- * E-mail:
| |
Collapse
|