1
|
Sun Q, Xing X, Wang H, Wan K, Fan R, Liu C, Wang Y, Wu W, Wang Y, Wang R. SCD1 is the critical signaling hub to mediate metabolic diseases: Mechanism and the development of its inhibitors. Biomed Pharmacother 2024; 170:115586. [PMID: 38042113 DOI: 10.1016/j.biopha.2023.115586] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 12/04/2023] Open
Abstract
Metabolic diseases, featured with dysregulated energy homeostasis, have become major global health challenges. Patients with metabolic diseases have high probability to manifest multiple complications in lipid metabolism, e.g. obesity, insulin resistance and fatty liver. Therefore, targeting the hub genes in lipid metabolism may systemically ameliorate the metabolic diseases, along with the complications. Stearoyl-CoA desaturase 1(SCD1) is a key enzyme that desaturates the saturated fatty acids (SFAs) derived from de novo lipogenesis or diet to generate monounsaturated fatty acids (MUFAs). SCD1 maintains the metabolic and tissue homeostasis by responding to, and integrating the multiple layers of endogenous stimuli, which is mediated by the synthesized MUFAs. It critically regulates a myriad of physiological processes, including energy homeostasis, development, autophagy, tumorigenesis and inflammation. Aberrant transcriptional and epigenetic activation of SCD1 regulates AMPK/ACC, SIRT1/PGC1α, NcDase/Wnt, etc, and causes aberrant lipid accumulation, thereby promoting the progression of obesity, non-alcoholic fatty liver, diabetes and cancer. This review critically assesses the integrative mechanisms of the (patho)physiological functions of SCD1 in metabolic homeostasis, inflammation and autophagy. For translational perspective, potent SCD1 inhibitors have been developed to treat various types of cancer. We thus discuss the multidisciplinary advances that greatly accelerate the development of SCD1 new inhibitors. In conclusion, besides cancer treatment, SCD1 may serve as the promising target to combat multiple metabolic complications simultaneously.
Collapse
Affiliation(s)
- Qin Sun
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Xiaorui Xing
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Huanyu Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Kang Wan
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Ruobing Fan
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Cheng Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Yongjian Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Wenyi Wu
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Yibing Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China.
| | - Ru Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
2
|
Oh SJ, Hwang Y, Hur KY, Lee MS. Lysosomal Ca 2+ as a mediator of palmitate-induced lipotoxicity. Cell Death Discov 2023; 9:100. [PMID: 36944629 PMCID: PMC10030853 DOI: 10.1038/s41420-023-01379-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/23/2023] Open
Abstract
While the mechanism of lipotoxicity by palmitic acid (PA), an effector of metabolic stress in vitro and in vivo, has been extensively investigated, molecular details of lipotoxicity are still not fully characterized. Since recent studies reported that PA can exert lysosomal stress in addition to well-known ER and mitochondrial stress, we studied the role of lysosomal events in lipotoxicity by PA, focusing on lysosomal Ca2+. We found that PA induced accumulation of mitochondrial ROS and that mitochondrial ROS induced release of lysosomal Ca2+ due to lysosomal Ca2+ exit channel activation. Lysosomal Ca2+ release led to increased cytosolic Ca2+ which induced mitochondrial permeability transition (mPT). Chelation of cytoplasmic Ca2+ or blockade of mPT with olesoxime or decylubiquinone (DUB) suppressed lipotoxicity. Lysosomal Ca2+ release led to reduced lysosomal Ca2+ content which was replenished by ER Ca2+, the largest intracellular Ca2+ reservoir (ER → lysosome Ca2+ refilling), which in turn activated store-operated Ca2+ entry (SOCE). Inhibition of ER → lysosome Ca2+ refilling by blockade of ER Ca2+ exit channel using dantrolene or inhibition of SOCE using BTP2 inhibited lipotoxicity in vitro. Dantrolene or DUB also inhibited lipotoxic death of hepatocytes in vivo induced by administration of ethyl palmitate together with LPS. These results suggest a novel pathway of lipotoxicity characterized by mPT due to lysosomal Ca2+ release which was supplemented by ER → lysosome Ca2+ refilling and subsequent SOCE, and also suggest the potential role of modulation of ER → lysosome Ca2+ refilling by dantrolene or other blockers of ER Ca2+ exit channels in disease conditions characterized by lipotoxicity such as metabolic syndrome, diabetes, cardiomyopathy or nonalcoholic steatohepatitis.
Collapse
Affiliation(s)
- Soo-Jin Oh
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06355, Korea
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-bio Science and Division of Endocrinology, Department of Internal Medicine, Soonchunhyang Medical Center, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Yeseong Hwang
- Severance Biomedical Science Institute, Graduate school of Medical Science, BK21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Kyu Yeon Hur
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Myung-Shik Lee
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-bio Science and Division of Endocrinology, Department of Internal Medicine, Soonchunhyang Medical Center, Soonchunhyang University College of Medicine, Cheonan, Korea.
- Severance Biomedical Science Institute, Graduate school of Medical Science, BK21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea.
| |
Collapse
|
3
|
Different Metabolism and Toxicity of TRANS Fatty Acids, Elaidate and Vaccenate Compared to Cis-Oleate in HepG2 Cells. Int J Mol Sci 2022; 23:ijms23137298. [PMID: 35806300 PMCID: PMC9266973 DOI: 10.3390/ijms23137298] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/25/2022] [Accepted: 06/28/2022] [Indexed: 01/27/2023] Open
Abstract
Trans fatty acids (TFAs) are not synthesized in the human body but are generally ingested in substantial amounts. The widespread view that TFAs, particularly those of industrial origin, are unhealthy and contribute to obesity, cardiovascular diseases and diabetes is based mostly on in vivo studies, and the underlying molecular mechanisms remain to be elucidated. Here, we used a hepatoma model of palmitate-induced lipotoxicity to compare the metabolism and effects of the representative industrial and ruminant TFAs, elaidate and vaccenate, respectively, with those of cis-oleate. Cellular FAs, triacylglycerols, diacylglycerols and ceramides were quantitated using chromatography, markers of stress and apoptosis were assessed at mRNA and protein levels, ultrastructural changes were examined by electron microscopy and viability was evaluated by MTT assay. While TFAs were just slightly more damaging than oleate when applied alone, they were remarkably less protective against palmitate toxicity in cotreatments. These differences correlated with their diverse incorporation into the accumulating diacylglycerols and ceramides. Our results provide in vitro evidence for the unfavorable metabolic features and potent stress-inducing character of TFAs in comparison with oleate. These findings strengthen the reasoning against dietary trans fat intake, and they can also help us better understand the molecular mechanisms of lipotoxicity.
Collapse
|
4
|
Urso C, Zhou H. Palmitic acid-induced defects in cell cycle progression and cytokinesis in Neuro-2a cells. Cell Cycle 2022; 21:1048-1057. [PMID: 35171079 PMCID: PMC9037450 DOI: 10.1080/15384101.2022.2040769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Obesity is associated with elevated levels of free fatty acids (FFAs). Excessive saturated fatty acids (SFAs) exhibit significant deleterious cytotoxic effects in many types of cells. However, the effects of palmitic acid (PA), the most common circulating SFA, on cell cycle progression in neuronal cells have not been well-examined. The aim of this study was to examine whether PA affects the proliferation and cell cycle progression in mouse neuroblastoma Neuro-2a (N2a) cells. Our studies found that 200 µM PA significantly decreased DNA synthesis and mitotic index in N2a cells as early as 4 h following treatment. 24 h treatment with 200 µM PA significantly decreased the percentage of diploid (2 N) cells while dramatically increasing the percentage of tetraploid (4 N) cells as compared to the BSA control. Moreover, our studies found that 24 h treatment with 200 µM PA increased the percentage of binucleate cells as compared to the BSA control. Our studies also found that unsaturated fatty acids (UFAs), including linoleic acid, oleic acid, α-linolenic acid, and docosahexaenoic acid, were able to abolish PA-induced decrease of 2 N cells, increase of 4 N cells, and accumulation of binucleate cells. Taken together, these results suggest that PA may affect multiple aspects of the cell cycle progression in N2a cells, including decreased DNA synthesis, G2/M arrest, and cytokinetic failure, which could be abolished by UFAs.Abbreviations: 4-PBA, 4-Phenylbutyric Acid; ALA, α-linolenic acid; BrdU, 5-bromo-2'-deoxyuridine; DAPI, 4',6-diamidino-2-phenylindole; ER, endoplasmic reticulum; FFA, free fatty acids; FITC, fluorescein isothiocyanate; LA, linoleic acid; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; N2a, Neuro-2a; NAC, N-acetyl cysteine; OA, oleic acid; PA, palmitic acid; pHH3, Phosphorylation of histone H3; PI, propidium iodide; SFA, saturated fatty acids; PUFA, polyunsaturated fatty acids; TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling; UFA, unsaturated fatty acids.
Collapse
Affiliation(s)
- C.J. Urso
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, USA
| | - Heping Zhou
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, USA,CONTACT Heping Zhou Department of Biological Sciences, Seton Hall University, South Orange, NJ07079, USA
| |
Collapse
|
5
|
Elrayess MA, Cyprian FS, Abdallah AM, Emara MM, Diboun I, Anwardeen N, Schuchardt S, Yassine HM. Metabolic Signatures of Type 2 Diabetes Mellitus and Hypertension in COVID-19 Patients With Different Disease Severity. Front Med (Lausanne) 2022; 8:788687. [PMID: 35083246 PMCID: PMC8784560 DOI: 10.3389/fmed.2021.788687] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 12/15/2021] [Indexed: 01/08/2023] Open
Abstract
Introduction: Increased COVID-19 disease severity is higher among patients with type 2 diabetes mellitus and hypertension. However, the metabolic pathways underlying this association are not fully characterized. This study aims to identify the metabolic signature associated with increased COVID-19 severity in patients with diabetes mellitus and hypertension. Methods: One hundred and fifteen COVID-19 patients were divided based on disease severity, diabetes status, and hypertension status. Targeted metabolomics of serum samples from all patients was performed using tandem mass spectrometry followed by multivariate and univariate models. Results: Reduced levels of various triacylglycerols were observed with increased disease severity in the diabetic patients, including those containing palmitic (C16:0), docosapentaenoic (C22:5, DPA), and docosahexaenoic (C22:6, DHA) acids (FDR < 0.01). Functional enrichment analysis revealed triacylglycerols as the pathway exhibiting the most significant changes in severe COVID-19 in diabetic patients (FDR = 7.1 × 10-27). Similarly, reduced levels of various triacylglycerols were also observed in hypertensive patients corresponding with increased disease severity, including those containing palmitic, oleic (C18:1), and docosahexaenoic acids. Functional enrichment analysis revealed long-chain polyunsaturated fatty acids (n-3 and n-6) as the pathway exhibiting the most significant changes with increased disease severity in hypertensive patients (FDR = 0.07). Conclusions: Reduced levels of triacylglycerols containing specific long-chain unsaturated, monounsaturated, and polyunsaturated fatty acids are associated with increased COVID-19 severity in diabetic and hypertensive patients, offering potential novel diagnostic and therapeutic targets.
Collapse
Affiliation(s)
| | - Farhan S Cyprian
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Abdallah M Abdallah
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Mohamed M Emara
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Ilhame Diboun
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | | | - Sven Schuchardt
- Department of Bio- and Environmental Analytics, Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Hadi M Yassine
- Biomedical Research Center (BRC), Qatar University, Doha, Qatar.,College of Health Sciences, Qatar University, Doha, Qatar
| |
Collapse
|
6
|
Huang G, Li M, Li Y, Mao Y. OUP accepted manuscript. Lab Med 2022; 53:545-551. [PMID: 35748329 DOI: 10.1093/labmed/lmac041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Guoqing Huang
- Department of Endocrinology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
- School of Medicine, Ningbo University, Ningbo, China
| | - Mingcai Li
- School of Medicine, Ningbo University, Ningbo, China
| | - Yan Li
- Department of Endocrinology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
- School of Medicine, Ningbo University, Ningbo, China
| | - Yushan Mao
- Department of Endocrinology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| |
Collapse
|
7
|
Hartwick Bjorkman S, Oliveira Pereira R. The Interplay Between Mitochondrial Reactive Oxygen Species, Endoplasmic Reticulum Stress, and Nrf2 Signaling in Cardiometabolic Health. Antioxid Redox Signal 2021; 35:252-269. [PMID: 33599550 PMCID: PMC8262388 DOI: 10.1089/ars.2020.8220] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: Mitochondria-derived reactive oxygen species (mtROS) are by-products of normal physiology that may disrupt cellular redox homeostasis on a regular basis. Nonetheless, failure to resolve sustained mitochondrial stress to mitigate high levels of mtROS might contribute to the etiology of numerous pathological conditions, such as obesity, insulin resistance, and cardiovascular disease (CVD). Recent Advances: Notably, recent studies have demonstrated that moderate mitochondrial stress might result in the induction of different stress response pathways that ultimately improve the organism's ability to deal with subsequent stress, a process termed mitohormesis. mtROS have been shown to play a key role in regulating this adaptation. Critical Issue: mtROS regulate the convergence of different signaling pathways that, when disturbed, might impair cardiometabolic health. Conversely, mtROS seem to be required to mediate activation of prosurvival pathways, contributing to improved cardiometabolic fitness. In the present review, we will primarily focus on the role of mtROS in the activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) antioxidant pathway and examine the role of endoplasmic reticulum (ER) stress in coordinating the convergence of ER stress and oxidative stress signaling through activation of Nrf2 and activating transcription factor 4 (ATF4). Future Directions: The mechanisms underlying cardiometabolic protection in response to mitochondrial stress have only started to be investigated. Integrated understanding of how mtROS and ER stress cooperatively promote activation of prosurvival pathways might shed mechanistic insight into the role of mitohormesis in mediating cardiometabolic protection and might inform future therapeutic avenues for the treatment of metabolic diseases contributing to CVD. Antioxid. Redox Signal. 35, 252-269.
Collapse
Affiliation(s)
- Sarah Hartwick Bjorkman
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.,Department of Obstetrics and Gynecology, Reproductive Endocrinology and Infertility, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Renata Oliveira Pereira
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
8
|
Joshi C, Jadeja V, Zhou H. Molecular Mechanisms of Palmitic Acid Augmentation in COVID-19 Pathologies. Int J Mol Sci 2021; 22:7127. [PMID: 34281182 PMCID: PMC8269364 DOI: 10.3390/ijms22137127] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 02/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has claimed over 2.7 million lives globally. Obesity has been associated with increased severity and mortality of COVID-19. However, the molecular mechanisms by which obesity exacerbates COVID-19 pathologies are not well-defined. The levels of free fatty acids (FFAs) are elevated in obese subjects. This study was therefore designed to examine how excess levels of different FFAs may affect the progression of COVID-19. Biological molecules associated with palmitic acid (PA) and COVID-19 were retrieved from QIAGEN Knowledge Base, and Ingenuity Pathway Analysis tools were used to analyze these datasets and explore the potential pathways affected by different FFAs. Our study found that one of the top 10 canonical pathways affected by PA was the coronavirus pathogenesis pathway, mediated by key inflammatory mediators, including PTGS2; cytokines, including IL1β and IL6; chemokines, including CCL2 and CCL5; transcription factors, including NFκB; translation regulators, including EEF1A1; and apoptotic mediators, including BAX. In contrast, n-3 fatty acids may attenuate PA's activation of the coronavirus pathogenesis pathway by inhibiting the activity of such mediators as IL1β, CCL2, PTGS2, and BAX. Furthermore, PA may modulate the expression of ACE2, the main cell surface receptor for the SARS-CoV-2 spike protein.
Collapse
Affiliation(s)
| | | | - Heping Zhou
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07079, USA; (C.J.); (V.J.)
| |
Collapse
|
9
|
Šrámek J, Němcová-Fürstová V, Kovář J. Molecular Mechanisms of Apoptosis Induction and Its Regulation by Fatty Acids in Pancreatic β-Cells. Int J Mol Sci 2021; 22:4285. [PMID: 33924206 PMCID: PMC8074590 DOI: 10.3390/ijms22084285] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/09/2021] [Accepted: 04/16/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic β-cell failure and death contribute significantly to the pathogenesis of type 2 diabetes. One of the main factors responsible for β-cell dysfunction and subsequent cell death is chronic exposure to increased concentrations of FAs (fatty acids). The effect of FAs seems to depend particularly on the degree of their saturation. Saturated FAs induce apoptosis in pancreatic β-cells, whereas unsaturated FAs are well tolerated and are even capable of inhibiting the pro-apoptotic effect of saturated FAs. Molecular mechanisms of apoptosis induction by saturated FAs in β-cells are not completely elucidated. Saturated FAs induce ER stress, which in turn leads to activation of all ER stress pathways. When ER stress is severe or prolonged, apoptosis is induced. The main mediator seems to be the CHOP transcription factor. Via regulation of expression/activity of pro- and anti-apoptotic Bcl-2 family members, and potentially also through the increase in ROS production, CHOP switches on the mitochondrial pathway of apoptosis induction. ER stress signalling also possibly leads to autophagy signalling, which may activate caspase-8. Saturated FAs activate or inhibit various signalling pathways, i.e., p38 MAPK signalling, ERK signalling, ceramide signalling, Akt signalling and PKCδ signalling. This may lead to the activation of the mitochondrial pathway of apoptosis, as well. Particularly, the inhibition of the pro-survival Akt signalling seems to play an important role. This inhibition may be mediated by multiple pathways (e.g., ER stress signalling, PKCδ and ceramide) and could also consequence in autophagy signalling. Experimental evidence indicates the involvement of certain miRNAs in mechanisms of FA-induced β-cell apoptosis, as well. In the rather rare situations when unsaturated FAs are also shown to be pro-apoptotic, the mechanisms mediating this effect in β-cells seem to be the same as for saturated FAs. To conclude, FA-induced apoptosis rather appears to be preceded by complex cross talks of multiple signalling pathways. Some of these pathways may be regulated by decreased membrane fluidity due to saturated FA incorporation. Few data are available concerning molecular mechanisms mediating the protective effect of unsaturated FAs on the effect of saturated FAs. It seems that the main possible mechanism represents a rather inhibitory intervention into saturated FA-induced pro-apoptotic signalling than activation of some pro-survival signalling pathway(s) or metabolic interference in β-cells. This inhibitory intervention may be due to an increase of membrane fluidity.
Collapse
Affiliation(s)
- Jan Šrámek
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism and Nutrition, Third Faculty of Medicine, Charles University, Ruská 87, 100 00 Prague, Czech Republic;
| | - Vlasta Němcová-Fürstová
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism and Nutrition, Third Faculty of Medicine, Charles University, Ruská 87, 100 00 Prague, Czech Republic;
| | | |
Collapse
|
10
|
Xu J, Taubert S. Beyond Proteostasis: Lipid Metabolism as a New Player in ER Homeostasis. Metabolites 2021; 11:52. [PMID: 33466824 PMCID: PMC7830277 DOI: 10.3390/metabo11010052] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/04/2021] [Accepted: 01/11/2021] [Indexed: 12/12/2022] Open
Abstract
Biological membranes are not only essential barriers that separate cellular and subcellular structures, but also perform other critical functions such as the initiation and propagation of intra- and intercellular signals. Each membrane-delineated organelle has a tightly regulated and custom-made membrane lipid composition that is critical for its normal function. The endoplasmic reticulum (ER) consists of a dynamic membrane network that is required for the synthesis and modification of proteins and lipids. The accumulation of unfolded proteins in the ER lumen activates an adaptive stress response known as the unfolded protein response (UPR-ER). Interestingly, recent findings show that lipid perturbation is also a direct activator of the UPR-ER, independent of protein misfolding. Here, we review proteostasis-independent UPR-ER activation in the genetically tractable model organism Caenorhabditis elegans. We review the current knowledge on the membrane lipid composition of the ER, its impact on organelle function and UPR-ER activation, and its potential role in human metabolic diseases. Further, we summarize the bi-directional interplay between lipid metabolism and the UPR-ER. We discuss recent progress identifying the different respective mechanisms by which disturbed proteostasis and lipid bilayer stress activate the UPR-ER. Finally, we consider how genetic and metabolic disturbances may disrupt ER homeostasis and activate the UPR and discuss how using -omics-type analyses will lead to more comprehensive insights into these processes.
Collapse
Affiliation(s)
- Jiaming Xu
- Graduate Program in Cell and Developmental Biology, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada;
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, BC V5Z 4H4, Canada
- Healthy Starts Theme, British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Stefan Taubert
- Graduate Program in Cell and Developmental Biology, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada;
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, BC V5Z 4H4, Canada
- Healthy Starts Theme, British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Medical Genetics, The University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| |
Collapse
|
11
|
Buhl JP, Garten A, Kratzsch J, Kiess W, Penke M. How Reliable are Commercially Available Glypican4 ELISA Kits? Exp Clin Endocrinol Diabetes 2020; 130:110-114. [PMID: 33065741 DOI: 10.1055/a-1257-0774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Glypican4 is an interesting new adipokine, which seems to play an important role in developmental processes and is potentially associated with metabolic changes in obesity and type 2 diabetes mellitus. Currently, only a few studies examined glypican4 in human blood, mainly in adults. DESIGN, PATIENTS AND MEASUREMENTS The aim of our study was to investigate glypican4 serum levels in lean, overweight, and obese children and adolescents, to unravel a possible association between glypican4 serum levels and parameters of obesity and insulin resistance. In order to determine a suitable method for investigating glypican4 serum levels, we validated two commercially available human glypican4 ELISA kits, using serum and plasma samples of an obese, insulin-resistant patient, and a healthy control subject, a human recombinant glypican4 protein fragment and glypican4-overexpressing cell lysate. RESULTS Using ELISA kit #1 we were not able to detect values above background level, apart from standard curve values. ELISA kit #2 initially seemed suitable to measure glypican4, but further validation experiments showed non-linearity of serial dilutions, no recognition of a human recombinant glypican4 protein fragment and non-linearity in the recovery of glypican4-overexpressing cell lysate. In addition, there was a considerable decrease (approx. 68%) of measured values between two experiments, performed at different time points with aliquots of the same serum sample. Contrary to that, further experiments found sample stability not to be compromised. CONCLUSIONS Extensive evaluation of the performance of two commercially available ELISA kits led to the conclusion that none of them is applicable for the measurement of glypican4 in human blood samples.
Collapse
Affiliation(s)
- Joseph P Buhl
- Center for Pediatric Research Leipzig, University Hospital for Children and Adolescents, Leipzig
| | - Antje Garten
- Center for Pediatric Research Leipzig, University Hospital for Children and Adolescents, Leipzig.,Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Edgbaston, Birmingham
| | - Jürgen Kratzsch
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig
| | - Wieland Kiess
- Center for Pediatric Research Leipzig, University Hospital for Children and Adolescents, Leipzig
| | - Melanie Penke
- Center for Pediatric Research Leipzig, University Hospital for Children and Adolescents, Leipzig
| |
Collapse
|
12
|
Sarnyai F, Somogyi A, Gór-Nagy Z, Zámbó V, Szelényi P, Mátyási J, Simon-Szabó L, Kereszturi É, Tóth B, Csala M. Effect of cis- and trans-Monounsaturated Fatty Acids on Palmitate Toxicity and on Palmitate-induced Accumulation of Ceramides and Diglycerides. Int J Mol Sci 2020; 21:ijms21072626. [PMID: 32283839 PMCID: PMC7178055 DOI: 10.3390/ijms21072626] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/02/2020] [Accepted: 04/07/2020] [Indexed: 01/22/2023] Open
Abstract
Dietary trans fatty acids (TFAs) have been implicated in serious health risks, yet little is known about their cellular effects and metabolism. We aim to undertake an in vitro comparison of two representative TFAs (elaidate and vaccenate) to the best-characterized endogenous cis-unsaturated FA (oleate). The present study addresses the possible protective action of TFAs on palmitate-treated RINm5F insulinoma cells with special regards to apoptosis, endoplasmic reticulum stress and the underlying ceramide and diglyceride (DG) accumulation. Both TFAs significantly improved cell viability and reduced apoptosis in palmitate-treated cells. They mildly attenuated palmitate-induced XBP-1 mRNA cleavage and phosphorylation of eukaryotic initiation factor 2α (eIF2α) and stress-activated protein kinase (SAPK)/c-Jun N-terminal kinase (JNK), but they were markedly less potent than oleate. Accordingly, all the three unsaturated FAs markedly reduced cellular palmitate incorporation and prevented harmful ceramide and DG accumulation. However, more elaidate or vaccenate than oleate was inserted into ceramides and DGs. Our results revealed a protective effect of TFAs in short-term palmitate toxicity, yet they also provide important in vitro evidence and even a potential mechanism for unfavorable long-term health effects of TFAs compared to oleate.
Collapse
Affiliation(s)
- Farkas Sarnyai
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, H-1085 Budapest, Hungary
| | - Anna Somogyi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, H-1085 Budapest, Hungary
| | - Zsófia Gór-Nagy
- Department of Inorganic and Analytical Chemistry, Budapest University of Technology and Economics, H-1111 Budapest, Hungary
| | - Veronika Zámbó
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, H-1085 Budapest, Hungary
| | - Péter Szelényi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, H-1085 Budapest, Hungary
| | - Judit Mátyási
- Department of Inorganic and Analytical Chemistry, Budapest University of Technology and Economics, H-1111 Budapest, Hungary
| | - Laura Simon-Szabó
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, H-1085 Budapest, Hungary
| | - Éva Kereszturi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, H-1085 Budapest, Hungary
| | - Blanka Tóth
- Department of Inorganic and Analytical Chemistry, Budapest University of Technology and Economics, H-1111 Budapest, Hungary
- Correspondence: (B.T.); (M.C.)
| | - Miklós Csala
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, H-1085 Budapest, Hungary
- Correspondence: (B.T.); (M.C.)
| |
Collapse
|
13
|
Oshima M, Pechberty S, Bellini L, Göpel SO, Campana M, Rouch C, Dairou J, Cosentino C, Fantuzzi F, Toivonen S, Marchetti P, Magnan C, Cnop M, Le Stunff H, Scharfmann R. Stearoyl CoA desaturase is a gatekeeper that protects human beta cells against lipotoxicity and maintains their identity. Diabetologia 2020; 63:395-409. [PMID: 31796987 PMCID: PMC6946759 DOI: 10.1007/s00125-019-05046-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 10/14/2019] [Indexed: 01/02/2023]
Abstract
AIMS/HYPOTHESIS During the onset of type 2 diabetes, excessive dietary intake of saturated NEFA and fructose lead to impaired insulin production and secretion by insulin-producing pancreatic beta cells. The majority of data on the deleterious effects of lipids on functional beta cell mass were obtained either in vivo in rodent models or in vitro using rodent islets and beta cell lines. Translating data from rodent to human beta cells remains challenging. Here, we used the human beta cell line EndoC-βH1 and analysed its sensitivity to a lipotoxic and glucolipotoxic (high palmitate with or without high glucose) insult, as a way to model human beta cells in a type 2 diabetes environment. METHODS EndoC-βH1 cells were exposed to palmitate after knockdown of genes related to saturated NEFA metabolism. We analysed whether and how palmitate induces apoptosis, stress and inflammation and modulates beta cell identity. RESULTS EndoC-βH1 cells were insensitive to the deleterious effects of saturated NEFA (palmitate and stearate) unless stearoyl CoA desaturase (SCD) was silenced. SCD was abundantly expressed in EndoC-βH1 cells, as well as in human islets and human induced pluripotent stem cell-derived beta cells. SCD silencing induced markers of inflammation and endoplasmic reticulum stress and also IAPP mRNA. Treatment with the SCD products oleate or palmitoleate reversed inflammation and endoplasmic reticulum stress. Upon SCD knockdown, palmitate induced expression of dedifferentiation markers such as SOX9, MYC and HES1. Interestingly, SCD knockdown by itself disrupted beta cell identity with a decrease in mature beta cell markers INS, MAFA and SLC30A8 and decreased insulin content and glucose-stimulated insulin secretion. CONCLUSIONS/INTERPRETATION The present study delineates an important role for SCD in the protection against lipotoxicity and in the maintenance of human beta cell identity. DATA AVAILABILITY Microarray data and all experimental details that support the findings of this study have been deposited in in the GEO database with the GSE130208 accession code.
Collapse
Affiliation(s)
- Masaya Oshima
- Université Paris Descartes, Institut Cochin, Inserm U1016, 123 bd du Port-Royal, 75014, Paris, France
| | - Séverine Pechberty
- Université Paris Descartes, Institut Cochin, Inserm U1016, 123 bd du Port-Royal, 75014, Paris, France
| | - Lara Bellini
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, France
| | - Sven O Göpel
- Bioscience Metabolism, Research and Early Development Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Mélanie Campana
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, France
| | - Claude Rouch
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, France
| | - Julien Dairou
- Université Paris Descartes CNRS UMR 8601, Paris, France
| | - Cristina Cosentino
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Federica Fantuzzi
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Sanna Toivonen
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Piero Marchetti
- University of Pisa, Department of Clinical and Experimental Medicine, Pisa, Italy
| | - Christophe Magnan
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, France
| | - Miriam Cnop
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
- Division of Endocrinology, ULB Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Hervé Le Stunff
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, France
- Université Paris-Sud, CNRS UMR 9197, Institut des Neurosciences Paris-Saclay (Neuro-PSI) - CNRS UMR 9197, Orsay, France
| | - Raphaël Scharfmann
- Université Paris Descartes, Institut Cochin, Inserm U1016, 123 bd du Port-Royal, 75014, Paris, France.
| |
Collapse
|
14
|
Palmitate and oleate modify membrane fluidity and kinase activities of INS-1E β-cells alongside altered metabolism-secretion coupling. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118619. [DOI: 10.1016/j.bbamcr.2019.118619] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 11/14/2019] [Accepted: 12/02/2019] [Indexed: 01/10/2023]
|
15
|
Zhang Y, Wang Q, Liu A, Wu Y, Liu F, Wang H, Zhu T, Fan Y, Yang B. Erythropoietin Derived Peptide Improved Endoplasmic Reticulum Stress and Ischemia-Reperfusion Related Cellular and Renal Injury. Front Med (Lausanne) 2020; 7:5. [PMID: 32039224 PMCID: PMC6992600 DOI: 10.3389/fmed.2020.00005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/08/2020] [Indexed: 12/23/2022] Open
Abstract
Ischemia-reperfusion (IR) injury often affects transplant and native kidneys alike. IR injury is one of the main causes of acute kidney injury (AKI) and further associated with the progression of chronic kidney disease. Our previous study revealed the renoprotection of erythropoietin derived cyclic helix-B surface peptide (CHBP) against IR injury. However, the precise role and underlying mechanism of endoplasmic reticulum stress (ERS) in the injury and the renoprotection induced by IR or CHBP, respectively, have not been fully defined. This study using mouse kidney epithelial cells (TCMK-1) revealed that the level of CHOP (a key marker of ERS), PERK, and JNK (regulators of CHOP) was gradually increased by the prolonged time of hydrogen peroxide (H2O2) stimulation. In addition, CHOP mRNA and protein were significantly reduced by small interfering RNA (siRNA) target CHOP, as were apoptotic and inflammatory mediator caspase-3 and HMGB-1, and early apoptosis. Furthermore, CHOP mRNA was correlated positively with PERK protein, active caspase-3, HMGB-1 and apoptosis, but negatively with cell viability in vitro, while CHOP protein was also correlated positively with the level of tubulointerstitial damage and active caspase-3 protein in vivo. Finally, CHBP improved the viability of TCMK-1 cells subjected to H2O2 stimulation time-dependently, with reduced level of CHOP mRNA. CHBP also inhibited the increase of CHOP protein, not only in TCMK-1 cells, but also in the IR injury kidneys at 2 weeks. Moreover, CHBP reduced the expression of PERK mRNA and protein, JNK and HMGB-1 protein, as well as early and later apoptosis. In addition, raised CHOP at 12 h post IR injury might be an early time window for intervention. In conclusion, the differential role of ERS and CHBP in IR-related injury was proved in mouse TCMK-1 cells and kidneys, in which the mechanistic signaling pathway was associated with CHOP/PERK/JNK, HMGB-1/caspase-3, and apoptosis. CHOP might be a potential biomarker and CHBP might be therapeutic drug for IR-induced AKI.
Collapse
Affiliation(s)
- Yufang Zhang
- Renal Group, Basic Medical Research Centre, Medical College of Nantong University, Nantong, China
| | - Qian Wang
- Department of Nephrology, Nantong-Leicester Joint Institute of Kidney Science, Affiliated Hospital of Nantong University, Nantong, China
| | - Aifen Liu
- Renal Group, Basic Medical Research Centre, Medical College of Nantong University, Nantong, China
| | - Yuanyuan Wu
- Department of Cardiovascular Sciences, University of Leicester, University Hospitals of Leicester, Leicester, United Kingdom
| | - Feng Liu
- Department of Nephrology, Nantong-Leicester Joint Institute of Kidney Science, Affiliated Hospital of Nantong University, Nantong, China
| | - Hui Wang
- Department of Nephrology, Nantong-Leicester Joint Institute of Kidney Science, Affiliated Hospital of Nantong University, Nantong, China
| | - Tongyu Zhu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Yaping Fan
- Department of Nephrology, Nantong-Leicester Joint Institute of Kidney Science, Affiliated Hospital of Nantong University, Nantong, China
| | - Bin Yang
- Department of Nephrology, Nantong-Leicester Joint Institute of Kidney Science, Affiliated Hospital of Nantong University, Nantong, China.,Department of Cardiovascular Sciences, University of Leicester, University Hospitals of Leicester, Leicester, United Kingdom
| |
Collapse
|
16
|
Shaping of Innate Immune Response by Fatty Acid Metabolite Palmitate. Cells 2019; 8:cells8121633. [PMID: 31847240 PMCID: PMC6952933 DOI: 10.3390/cells8121633] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/07/2019] [Accepted: 12/12/2019] [Indexed: 12/19/2022] Open
Abstract
Innate immune cells monitor invading pathogens and pose the first-line inflammatory response to coordinate with adaptive immunity for infection removal. Innate immunity also plays pivotal roles in injury-induced tissue remodeling and the maintenance of tissue homeostasis in physiological and pathological conditions. Lipid metabolites are emerging as the key players in the regulation of innate immune responses, and recent work has highlighted the importance of the lipid metabolite palmitate as an essential component in this regulation. Palmitate modulates innate immunity not only by regulating the activation of pattern recognition receptors in local innate immune cells, but also via coordinating immunological activity in inflammatory tissues. Moreover, protein palmitoylation controls various cellular physiological processes. Herein, we review the updated evidence that palmitate catabolism contributes to innate immune cell-mediated inflammatory processes that result in immunometabolic disorders.
Collapse
|
17
|
Zámbó V, Simon-Szabó L, Sarnyai F, Mátyási J, Gór-Nagy Z, Somogyi A, Szelényi P, Kereszturi É, Tóth B, Csala M. Investigation of the putative rate-limiting role of electron transfer in fatty acid desaturation using transfected HEK293T cells. FEBS Lett 2019; 594:530-539. [PMID: 31557308 DOI: 10.1002/1873-3468.13622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/10/2019] [Accepted: 09/21/2019] [Indexed: 11/09/2022]
Abstract
Elevated fatty acid (FA) levels contribute to severe metabolic diseases. Unbalanced oversupply of saturated FAs is particularly damaging, which renders stearoyl-CoA desaturase (SCD1) activity an important factor of resistance. A SCD1-related oxidoreductase protects cells against palmitate toxicity, so we aimed to test whether desaturase activity is limited by SCD1 itself or by the associated electron supply. Unsaturated/saturated FA ratio was markedly elevated by SCD1 overexpression while it remained unaffected by the overexpression of SCD1-related electron transfer proteins in HEK293T cells. Electron supply was not rate-limiting either in palmitate-treated cells or in cells with enhanced SCD1 expression. Our findings indicate the rate-limiting role of SCD1 itself, and that FA desaturation cannot be facilitated by reinforcing the electron supply of the enzyme.
Collapse
Affiliation(s)
- Veronika Zámbó
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Laura Simon-Szabó
- Pathobiochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University (MTA-SE), Budapest, Hungary
| | - Farkas Sarnyai
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | | | - Zsófia Gór-Nagy
- Department of Inorganic and Analytical Chemistry, Budapest University of Technology and Economics, Hungary
| | - Anna Somogyi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Péter Szelényi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Éva Kereszturi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Blanka Tóth
- Department of Inorganic and Analytical Chemistry, Budapest University of Technology and Economics, Hungary
| | - Miklós Csala
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| |
Collapse
|
18
|
Oleic acid increases the transcriptional activity of FoxO1 by promoting its nuclear translocation and β-catenin binding in pancreatic β-cells. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2753-2764. [PMID: 31255704 DOI: 10.1016/j.bbadis.2019.06.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/31/2019] [Accepted: 06/25/2019] [Indexed: 01/08/2023]
Abstract
In the setting of metabolic overload, chronic elevations of free fatty acids in blood and tissues are associated with pancreatic β-cell lipotoxicity and failure. Ultimately, obesity combined with insulin resistance increases the dysfunctional demand of β-cells and contributes to the development of type 2 diabetes. Forkhead box O1 (FoxO1) is a potent transcriptional regulator of pancreatic β-cell function and tolerance to lipid stress. The present study examined the effects of stearoyl-CoA desaturase 1 (SCD1)-metabolized precursors and products, notably oleic acid, on the compensatory capacity of β-cells and their relationship with regulation of the FoxO1 and Wnt pathways. The trioleate-induced compromise of insulin sensitivity blunted the compensatory response of pancreatic β-cells in primary rat islets. These events were associated with increases in the nuclear accumulation and transcriptional activity of FoxO1. Such effects were also observed in INS-1E cells that were subjected to oleate treatment. The overexpression of human SCD1 that was accompanied by endogenously generated oleic acid also led to an increase in the nuclear abundance of FoxO1. The mechanism of the oleate-mediated subcellular localization of FoxO1 was independent of the fatty acid receptor GPR40. Instead, the mechanism involved diversion of the active β-catenin pool from an interaction with transcription factor 7-like 2 toward FoxO1-mediated transcription in β-cells. Our findings identify a unique role for oleic acid in the compensatory response of pancreatic β-cells and emphasize the importance of FoxO1 in β-cell failure in obesity-induced insulin resistance.
Collapse
|
19
|
Liu X, Zeng X, Chen X, Luo R, Li L, Wang C, Liu J, Cheng J, Lu Y, Chen Y. Oleic acid protects insulin-secreting INS-1E cells against palmitic acid-induced lipotoxicity along with an amelioration of ER stress. Endocrine 2019; 64:512-524. [PMID: 30778898 DOI: 10.1007/s12020-019-01867-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 02/08/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE It is demonstrated that unsaturated fatty acids can counteract saturated fatty acids-induced lipotoxicity, but the molecular mechanisms are unclear. In this study, we investigated the protective effects of monounsaturated oleic acid (OA) against saturated palmitic acid (PA)-induced cytotoxicity in rat β cells as well as islets, and mechanistically focused on its regulation on endoplasmic reticulum (ER) stress. METHODS Rat insulinoma cell line INS-1E cells and primary islets were treated with PA with or without OA for 24 h to determine the cell viability, apoptosis, and ER stress. SD rats were fed with high-fat diet (HFD) for 16 w, then, HFD was half replaced by olive oil to observe the protective effects of monounsaturated fatty acids rich diet. RESULTS We demonstrated that PA impaired cell viability and insulin secretion of INS-1E cells and rat islets, but OA robustly rescued cells from cell death. OA substantially alleviated either PA or chemical ER stressors (thapsigargin or tunicamycin)-induced ER stress. Importantly, OA attenuated the activity of PERK-eIF2α-ATF4-CHOP pathway and regulated the ER Ca2+ homeostasis. In vivo, only olive oil supplementation did not cause significant changes, while high-fat diet (HFD) for 32 w obviously induced islets ER stress and impaired insulin sensitivity in SD rats. Half replacement of HFD with olive oil (a mixed diet) has ameliorated this effect. CONCLUSION OA alleviated PA-induced lipotoxicity in INS-1E cells and improved insulin sensitivity in HFD rats. The amelioration of PA triggered ER stress may be responsible for its beneficial effects in β cells.
Collapse
Affiliation(s)
- Xiaohong Liu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center, Endocrinology Department, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xin Zeng
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center, Endocrinology Department, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xuanming Chen
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center, Endocrinology Department, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Ruixi Luo
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center, Endocrinology Department, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Linzhao Li
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center, Endocrinology Department, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Chengshi Wang
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center, Endocrinology Department, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Jingping Liu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center, Endocrinology Department, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Jingqiu Cheng
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center, Endocrinology Department, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Yanrong Lu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center, Endocrinology Department, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Younan Chen
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center, Endocrinology Department, West China Hospital, Sichuan University, Chengdu, P.R. China.
| |
Collapse
|
20
|
Oleic acid ameliorates palmitic acid-induced ER stress and inflammation markers in naive and cerulein-treated exocrine pancreas cells. Biosci Rep 2019; 39:BSR20190054. [PMID: 30992393 PMCID: PMC6522823 DOI: 10.1042/bsr20190054] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/06/2019] [Accepted: 04/12/2019] [Indexed: 12/13/2022] Open
Abstract
Dietary fat overload (typical to obesity) increases the risk of pancreatic pathologies through mechanisms yet to be defined. We previously showed that saturated dietary fat induces pancreatic acinar lipotoxicity and cellular stress. The endoplasmic reticulum (ER) of exocrine pancreas cells is highly developed and thus predisposed to stress. We studied the combination of saturated and unsaturated FAs in metabolic and pancreatitis like cerulein (CER)-induced stress states on cellular ER stress. Exocrine pancreas AR42J and rat primary exocrine acinar cells underwent acute (24 h) challenge with different FAs (saturated, monounsaturated) at different concentrations (250 and 500 µM) and in combination with acute CER-induced stress, and were analyzed for fat accumulation, ER stress unfolded protein response (UPR) and immune and enzyme markers. Acute exposure of AR42J and pancreatic acinar cells to different FAs and their combinations increased triglyceride accumulation. Palmitic acid significantly dose-dependently enhanced the UPR, immune factors and pancreatic lipase (PL) levels, as demonstrated by XBP1 splicing and elevation in UPR transcripts and protein levels (Xbp1,Atf6, Atf4, Chop, Tnfα, Tgfβ and Il-6). Exposure to high palmitic levels in a CER-induced stress state synergistically increased ER stress and inflammation marker levels. Exposure to oleic acid did not induce ER stress and PL levels and significantly decreased immune factors in an acute CER-induced stress state. Combination of oleic and palmitic acids significantly reduced the palmitic-induced ER stress, but did not affect the immune factor response. We show that combination of monounsaturated and saturated FAs protects from exocrine pancreatic cellular ER stress in both metabolic and CER-induced stress.
Collapse
|
21
|
Ntika S, Thombare K, Aryapoor M, Kristinsson H, Bergsten P, Krizhanovskii C. Oleate increase neutral lipid accumulation, cellular respiration and rescues palmitate-exposed GLP-1 secreting cells by reducing ceramide-induced ROS. Biochimie 2019; 159:23-35. [DOI: 10.1016/j.biochi.2018.11.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/28/2018] [Indexed: 12/13/2022]
|
22
|
Balaban S, Nassar ZD, Zhang AY, Hosseini-Beheshti E, Centenera MM, Schreuder M, Lin HM, Aishah A, Varney B, Liu-Fu F, Lee LS, Nagarajan SR, Shearer RF, Hardie RA, Raftopulos NL, Kakani MS, Saunders DN, Holst J, Horvath LG, Butler LM, Hoy AJ. Extracellular Fatty Acids Are the Major Contributor to Lipid Synthesis in Prostate Cancer. Mol Cancer Res 2019; 17:949-962. [PMID: 30647103 DOI: 10.1158/1541-7786.mcr-18-0347] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 09/22/2018] [Accepted: 01/07/2019] [Indexed: 02/06/2023]
Abstract
Prostate cancer cells exhibit altered cellular metabolism but, notably, not the hallmarks of Warburg metabolism. Prostate cancer cells exhibit increased de novo synthesis of fatty acids (FA); however, little is known about how extracellular FAs, such as those in the circulation, may support prostate cancer progression. Here, we show that increasing FA availability increased intracellular triacylglycerol content in cultured patient-derived tumor explants, LNCaP and C4-2B spheroids, a range of prostate cancer cells (LNCaP, C4-2B, 22Rv1, PC-3), and prostate epithelial cells (PNT1). Extracellular FAs are the major source (∼83%) of carbons to the total lipid pool in all cell lines, compared with glucose (∼13%) and glutamine (∼4%), and FA oxidation rates are greater in prostate cancer cells compared with PNT1 cells, which preferentially partitioned extracellular FAs into triacylglycerols. Because of the higher rates of FA oxidation in C4-2B cells, cells remained viable when challenged by the addition of palmitate to culture media and inhibition of mitochondrial FA oxidation sensitized C4-2B cells to palmitate-induced apoptosis. Whereas in PC-3 cells, palmitate induced apoptosis, which was prevented by pretreatment of PC-3 cells with FAs, and this protective effect required DGAT-1-mediated triacylglycerol synthesis. These outcomes highlight for the first-time heterogeneity of lipid metabolism in prostate cancer cells and the potential influence that obesity-associated dyslipidemia or host circulating has on prostate cancer progression. IMPLICATIONS: Extracellular-derived FAs are primary building blocks for complex lipids and heterogeneity in FA metabolism exists in prostate cancer that can influence tumor cell behavior.
Collapse
Affiliation(s)
- Seher Balaban
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Zeyad D Nassar
- Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia, Australia.,South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Alison Y Zhang
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute for Medical Research, Darlinghurst, New South Wales, Australia.,Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Chris O'Brien Lifehouse, Camperdown, New South Wales, Australia
| | - Elham Hosseini-Beheshti
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Margaret M Centenera
- Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia, Australia.,South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Mark Schreuder
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia.,Faculty of Medicine, University of Utrecht, Utrecht, the Netherlands
| | - Hui-Ming Lin
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute for Medical Research, Darlinghurst, New South Wales, Australia
| | - Atqiya Aishah
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Bianca Varney
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Frank Liu-Fu
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Lisa S Lee
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Shilpa R Nagarajan
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Robert F Shearer
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute for Medical Research, Darlinghurst, New South Wales, Australia
| | - Rae-Anne Hardie
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Origins of Cancer Program, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia
| | - Nikki L Raftopulos
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Meghna S Kakani
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Darren N Saunders
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Jeff Holst
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Origins of Cancer Program, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia
| | - Lisa G Horvath
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute for Medical Research, Darlinghurst, New South Wales, Australia.,Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Chris O'Brien Lifehouse, Camperdown, New South Wales, Australia.,School of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia.,Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Lisa M Butler
- Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia, Australia.,South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Andrew J Hoy
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia.
| |
Collapse
|
23
|
Balaban S, Lee LS, Varney B, Aishah A, Gao Q, Shearer RF, Saunders DN, Grewal T, Hoy AJ. Heterogeneity of fatty acid metabolism in breast cancer cells underlies differential sensitivity to palmitate-induced apoptosis. Mol Oncol 2018; 12:1623-1638. [PMID: 30099850 PMCID: PMC6120225 DOI: 10.1002/1878-0261.12368] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 07/18/2018] [Accepted: 08/03/2018] [Indexed: 01/02/2023] Open
Abstract
Breast cancer (BrCa) metabolism is geared toward biomass synthesis and maintenance of reductive capacity. Changes in glucose and glutamine metabolism in BrCa have been widely reported, yet the contribution of fatty acids (FAs) in BrCa biology remains to be determined. We recently reported that adipocyte coculture alters MCF-7 and MDA-MB-231 cell metabolism and promotes proliferation and migration. Since adipocytes are FA-rich, and these FAs are transferred to BrCa cells, we sought to elucidate the FA metabolism of BrCa cells and their response to FA-rich environments. MCF-7 and MDA-MB-231 cells incubated in serum-containing media supplemented with FAs accumulate extracellular FAs as intracellular triacylglycerols (TAG) in a dose-dependent manner, with MDA-MB-231 cells accumulating more TAG. The differences in TAG levels were a consequence of distinct differences in intracellular partitioning of FAs, and not due to differences in the rate of FA uptake. Specifically, MCF-7 cells preferentially partition FAs into mitochondrial oxidation, whereas MDA-MB-231 cells partition FAs into TAG synthesis. These differences in intracellular FA handling underpin differences in the sensitivity to palmitate-induced lipotoxicity, with MDA-MB-231 cells being highly sensitive, whereas MCF-7 cells are partially protected. The attenuation of palmitate-induced lipotoxicity in MCF-7 cells was reversed by inhibition of FA oxidation. Pretreatment of MDA-MB-231 cells with FAs increased TAG synthesis and reduced palmitate-induced apoptosis. Our results provide novel insight into the potential influences of obesity on BrCa biology, highlighting distinct differences in FA metabolism in MCF-7 and MDA-MB-231 cells and how lipid-rich environments modulate these effects.
Collapse
Affiliation(s)
- Seher Balaban
- Discipline of Physiology, Faculty of Medicine and Health, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, The University of Sydney, Australia
| | - Lisa S Lee
- Discipline of Physiology, Faculty of Medicine and Health, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, The University of Sydney, Australia
| | - Bianca Varney
- Discipline of Physiology, Faculty of Medicine and Health, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, The University of Sydney, Australia
| | - Atqiya Aishah
- Discipline of Physiology, Faculty of Medicine and Health, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, The University of Sydney, Australia
| | - Quanqing Gao
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Australia
| | - Robert F Shearer
- Kinghorn Cancer Center, Garvan Institute of Medical Research, Darlinghurst, Australia
| | | | - Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Australia
| | - Andrew J Hoy
- Discipline of Physiology, Faculty of Medicine and Health, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, The University of Sydney, Australia
| |
Collapse
|
24
|
Fatostatin induces pro- and anti-apoptotic lipid accumulation in breast cancer. Oncogenesis 2018; 7:66. [PMID: 30140005 PMCID: PMC6107643 DOI: 10.1038/s41389-018-0076-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/12/2018] [Accepted: 07/18/2018] [Indexed: 12/27/2022] Open
Abstract
Given the dependence of cancers on de novo lipogenesis, we tested the effect of fatostatin, a small molecule thought to target this pathway by blocking activation of SREBP transcription factors, in breast cancer cell lines and xenograft tumors. We found that estrogen receptor (ER) positive cells were more sensitive to fatostatin than ER negative cells and responded with cell cycle arrest and apoptosis. Surprisingly, we found that rather than inhibiting lipogenesis, fatostatin caused an accumulation of lipids as a response to endoplasmic reticulum stress rather than inhibition of SREBP activity. In particular, ceramide and dihydroceramide levels increased and contributed to the apoptotic effects of fatostatin. In addition, an accumulation of triacylglycerides (TAGs), particularly those containing polyunsaturated fatty acids (PUFAs), was also observed as a result of elevated diacylglycerol transferase activity. Blocking PUFA-TAG production enhanced the apoptotic effect of fatostatin, suggesting that these lipids play a protective role and limit fatostatin response. Together, these findings indicate that the ability of breast cancer cells to respond to fatostatin depends on induction of endoplasmic reticulum stress and subsequent ceramide accumulation, and that limiting production of PUFA-TAGs may be therapeutically beneficial in specific tumor subtypes.
Collapse
|
25
|
Fatty Acid-Stimulated Insulin Secretion vs. Lipotoxicity. Molecules 2018; 23:molecules23061483. [PMID: 29921789 PMCID: PMC6100479 DOI: 10.3390/molecules23061483] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/13/2018] [Accepted: 06/15/2018] [Indexed: 12/29/2022] Open
Abstract
Fatty acid (FA)-stimulated insulin secretion (FASIS) is reviewed here in contrast to type 2 diabetes etiology, resulting from FA overload, oxidative stress, intermediate hyperinsulinemia, and inflammation, all converging into insulin resistance. Focusing on pancreatic islet β-cells, we compare the physiological FA roles with the pathological ones. Considering FAs not as mere amplifiers of glucose-stimulated insulin secretion (GSIS), but as parallel insulin granule exocytosis inductors, partly independent of the KATP channel closure, we describe the FA initiating roles in the prediabetic state that is induced by retardations in the glycerol-3-phosphate (glucose)-promoted glycerol/FA cycle and by the impaired GPR40/FFA1 (free FA1) receptor pathway, specifically in its amplification by the redox-activated mitochondrial phospholipase, iPLA2γ. Also, excessive dietary FAs stimulate intestine enterocyte incretin secretion, further elevating GSIS, even at low glucose levels, thus contributing to diabetic hyperinsulinemia. With overnutrition and obesity, the FA overload causes impaired GSIS by metabolic dysbalance, paralleled by oxidative and metabolic stress, endoplasmic reticulum stress and numerous pro-apoptotic signaling, all leading to decreased β-cell survival. Lipotoxicity is exerted by saturated FAs, whereas ω-3 polyunsaturated FAs frequently exert antilipotoxic effects. FA-facilitated inflammation upon the recruitment of excess M1 macrophages into islets (over resolving M2 type), amplified by cytokine and chemokine secretion by β-cells, leads to an inevitable failure of pancreatic β-cells.
Collapse
|
26
|
Baumann J, Kokabee M, Wong J, Balasubramaniyam R, Sun Y, Conklin DS. Global metabolite profiling analysis of lipotoxicity in HER2/neu-positive breast cancer cells. Oncotarget 2018; 9:27133-27150. [PMID: 29930756 PMCID: PMC6007458 DOI: 10.18632/oncotarget.25500] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/10/2018] [Indexed: 12/12/2022] Open
Abstract
Recent work has shown that HER2/neu-positive breast cancer cells rely on a unique Warburg-like metabolism for survival and aggressive behavior. These cells are dependent on fatty acid (FA) synthesis, show markedly increased levels of stored fats and disruption of the synthetic process results in apoptosis. In this study, we used global metabolite profiling and a multi-omics network analysis approach to model the metabolic changes in this physiology under palmitate-supplemented growth conditions to gain insights into the molecular mechanism and its relevance to disease prevention and treatment. Computational analyses were used to define pathway enrichment based on the dataset of significantly altered metabolites and to integrate metabolomics and transcriptomics data in a multi-omics network analysis. Network-predicted changes and functional relationships were tested with cell assays in vitro. Palmitate-supplemented growth conditions induce distinct metabolic alterations. Growth of HER2-normal MCF7 cells is unaffected under these conditions whereas HER2/neu-positive cells display unchanged neutral lipid content, AMPK activation, inhibition of fatty acid synthesis and significantly altered glutamine, glucose and serine/glycine metabolism. The predominant upregulated lipid species is the novel bioactive lipid N-palmitoylglycine, which is non-toxic to these cells. Limiting the availability of glutamine significantly ameliorates the lipotoxic effects of palmitate, reduces CHOP and XBP1(s) induction and restores the expression levels of HER2 and HER3. The study shows that HER2/neu-positive breast cancer cells change their metabolic phenotype in the presence of palmitate. Palmitate induces AMPK activation and inhibition of fatty acid synthesis that feeds back into glycolysis as well as anaplerotic glutamine metabolism.
Collapse
Affiliation(s)
- Jan Baumann
- Cancer Research Center, Department of Biomedical Sciences, State University of New York, University at Albany, Rensselaer, NY 12144, USA
| | - Mostafa Kokabee
- Cancer Research Center, Department of Biomedical Sciences, State University of New York, University at Albany, Rensselaer, NY 12144, USA
| | - Jason Wong
- Cancer Research Center, Department of Biomedical Sciences, State University of New York, University at Albany, Rensselaer, NY 12144, USA
| | - Rakshika Balasubramaniyam
- Cancer Research Center, Department of Biomedical Sciences, State University of New York, University at Albany, Rensselaer, NY 12144, USA
| | - Yan Sun
- Cancer Research Center, Department of Biomedical Sciences, State University of New York, University at Albany, Rensselaer, NY 12144, USA
| | - Douglas S Conklin
- Cancer Research Center, Department of Biomedical Sciences, State University of New York, University at Albany, Rensselaer, NY 12144, USA
| |
Collapse
|
27
|
Palomer X, Pizarro-Delgado J, Barroso E, Vázquez-Carrera M. Palmitic and Oleic Acid: The Yin and Yang of Fatty Acids in Type 2 Diabetes Mellitus. Trends Endocrinol Metab 2018; 29:178-190. [PMID: 29290500 DOI: 10.1016/j.tem.2017.11.009] [Citation(s) in RCA: 331] [Impact Index Per Article: 55.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 11/22/2017] [Accepted: 11/30/2017] [Indexed: 12/20/2022]
Abstract
Increased plasma non-esterified fatty acids (NEFAs) link obesity with insulin resistance and type 2 diabetes mellitus (T2DM). However, in contrast to the saturated FA (SFA) palmitic acid, the monounsaturated FA (MUFA) oleic acid elicits beneficial effects on insulin sensitivity, and the dietary palmitic acid:oleic acid ratio impacts diabetes risk in humans. Here we review recent mechanistic insights into the beneficial effects of oleic acid compared with palmitic acid on insulin resistance and T2DM, including its anti-inflammatory actions, and its capacity to inhibit endoplasmic reticulum (ER) stress, prevent attenuation of the insulin signaling pathway, and improve β cell survival. Understanding the molecular mechanisms of the antidiabetic effects of oleic acid may contribute to understanding the benefits of this FA in the prevention or delay of T2DM.
Collapse
Affiliation(s)
- Xavier Palomer
- Department of Pharmacology, Toxicology, and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Institute of Biomedicine of the University of Barcelona (IBUB), Pediatric Research Institute-Hospital Sant Joan de Déu, and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Avinguda Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Javier Pizarro-Delgado
- Department of Pharmacology, Toxicology, and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Institute of Biomedicine of the University of Barcelona (IBUB), Pediatric Research Institute-Hospital Sant Joan de Déu, and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Avinguda Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Emma Barroso
- Department of Pharmacology, Toxicology, and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Institute of Biomedicine of the University of Barcelona (IBUB), Pediatric Research Institute-Hospital Sant Joan de Déu, and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Avinguda Joan XXIII 27-31, E-08028 Barcelona, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology, and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Institute of Biomedicine of the University of Barcelona (IBUB), Pediatric Research Institute-Hospital Sant Joan de Déu, and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Avinguda Joan XXIII 27-31, E-08028 Barcelona, Spain.
| |
Collapse
|
28
|
Ho N, Xu C, Thibault G. From the unfolded protein response to metabolic diseases - lipids under the spotlight. J Cell Sci 2018; 131:131/3/jcs199307. [PMID: 29439157 DOI: 10.1242/jcs.199307] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The unfolded protein response (UPR) is classically viewed as a stress response pathway to maintain protein homeostasis at the endoplasmic reticulum (ER). However, it has recently emerged that the UPR can be directly activated by lipid perturbation, independently of misfolded proteins. Comprising primarily phospholipids, sphingolipids and sterols, individual membranes can contain hundreds of distinct lipids. Even with such complexity, lipid distribution in a cell is tightly regulated by mechanisms that remain incompletely understood. It is therefore unsurprising that lipid dysregulation can be a key factor in disease development. Recent advances in analysis of lipids and their regulators have revealed remarkable mechanisms and connections to other cellular pathways including the UPR. In this Review, we summarize the current understanding in UPR transducers functioning as lipid sensors and the interplay between lipid metabolism and ER homeostasis in the context of metabolic diseases. We attempt to provide a framework consisting of a few key principles to integrate the different lines of evidence and explain this rather complicated mechanism.
Collapse
Affiliation(s)
- Nurulain Ho
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551
| | - Chengchao Xu
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142-1479, USA
| | - Guillaume Thibault
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551
| |
Collapse
|
29
|
ALJohani AM, Syed DN, Ntambi JM. Insights into Stearoyl-CoA Desaturase-1 Regulation of Systemic Metabolism. Trends Endocrinol Metab 2017; 28:831-842. [PMID: 29089222 PMCID: PMC5701860 DOI: 10.1016/j.tem.2017.10.003] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/09/2017] [Accepted: 10/10/2017] [Indexed: 12/15/2022]
Abstract
Stearoyl-coenzyme A desaturase 1 (SCD1) is a central regulator of fuel metabolism and may represent a therapeutic target to control obesity and the progression of related metabolic diseases including type 2 diabetes and hepatic steatosis. SCD1 catalyzes the synthesis of monounsaturated fatty acids (MUFAs), mainly oleate and palmitoleate, which are important in controlling weight gain in response to feeding high carbohydrate diets. In this review, we evaluate the role of SCD1 isoform in the regulation of lipid and glucose metabolism in metabolic tissues. These highlights of recent findings are aimed toward advancing our understanding of the role of SCD1 in the development of metabolic diseases, which may help evaluate the possible health outcomes of modulating MUFA levels through targeting SCD1 activity.
Collapse
Affiliation(s)
- Ahmed M ALJohani
- School of Medicine and Public Health, Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI 53706, USA; King Saud bin Abdulaziz University for Health Sciences, National Guard Health Affairs, Riyadh, 11426, Saudi Arabia
| | - Deeba N Syed
- School of Medicine and Public Health, Department of Dermatology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - James M Ntambi
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
30
|
Amanullah A, Upadhyay A, Joshi V, Mishra R, Jana NR, Mishra A. Progressing neurobiological strategies against proteostasis failure: Challenges in neurodegeneration. Prog Neurobiol 2017; 159:1-38. [DOI: 10.1016/j.pneurobio.2017.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 06/01/2017] [Accepted: 08/25/2017] [Indexed: 02/07/2023]
|
31
|
Effect of Saturated Stearic Acid on MAP Kinase and ER Stress Signaling Pathways during Apoptosis Induction in Human Pancreatic β-Cells Is Inhibited by Unsaturated Oleic Acid. Int J Mol Sci 2017; 18:ijms18112313. [PMID: 29099080 PMCID: PMC5713282 DOI: 10.3390/ijms18112313] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 10/27/2017] [Accepted: 10/31/2017] [Indexed: 12/13/2022] Open
Abstract
It has been shown that saturated fatty acids (FAs) have a detrimental effect on pancreatic β-cells function and survival, leading to apoptosis, whereas unsaturated FAs are well tolerated and are even capable of inhibiting the pro-apoptotic effect of saturated FAs. Molecular mechanisms of apoptosis induction and regulation by FAs in β-cells remain unclear; however, mitogen-activated protein (MAP) kinase and endoplasmic reticulum (ER) stress signaling pathways may be involved. In this study, we tested how unsaturated oleic acid (OA) affects the effect of saturated stearic acid (SA) on the p38 mitogen-activated protein kinase (MAPK) and extracellular signal-regulated kinase (ERK) pathways as well as the ER stress signaling pathways during apoptosis induction in the human pancreatic β-cells NES2Y. We demonstrated that OA is able to inhibit all effects of SA. OA alone has only minimal or no effects on tested signaling in NES2Y cells. The point of OA inhibitory intervention in SA-induced apoptotic signaling thus seems to be located upstream of the discussed signaling pathways.
Collapse
|
32
|
Colvin BN, Longtine MS, Chen B, Costa ML, Nelson DM. Oleate attenuates palmitate-induced endoplasmic reticulum stress and apoptosis in placental trophoblasts. Reproduction 2017; 153:369-380. [PMID: 28159805 DOI: 10.1530/rep-16-0576] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/02/2016] [Accepted: 01/03/2017] [Indexed: 12/24/2022]
Abstract
Pre-pregnancy obesity is increasingly common and predisposes pregnant women and offspring to gestational diabetes, pre-eclampsia, fetal growth abnormalities and stillbirth. Obese women exhibit elevated levels of the two most common dietary fatty acids, palmitate and oleate, and the maternal blood containing these nutrients bathes the surface of trophoblasts of placental villi in vivo We test the hypothesis that the composition and concentration of free fatty acids modulate viability and function of primary human villous trophoblasts in culture. We found that palmitate increases syncytiotrophoblast death, specifically by caspase-mediated apoptosis, whereas oleate does not cause enhanced cell death. Importantly, exposure to both fatty acids in equimolar amounts yielded no increase in death or apoptosis, suggesting that oleate can protect syncytiotrophoblasts from palmitate-induced death. We further found that palmitate, but not oleate or oleate with palmitate, increases endoplasmic reticulum (ER) stress, signaling through the unfolded protein response, and yielding CHOP-mediated induction of apoptosis. Finally, we show that oleate or oleate plus palmitate both lead to increased lipid droplets in syncytiotrophoblasts, whereas palmitate does not. The data show palmitate is toxic to human syncytiotrophoblasts, through the induction of ER stress and apoptosis mediated by CHOP, whereas oleate is not toxic, abrogates palmitate toxicity and induces fat accumulation. We speculate that our in vitro results offer pathways by which the metabolic milieu of the obese pregnant woman can yield villous trophoblast dysfunction and sub-optimal placental function.
Collapse
Affiliation(s)
| | - Mark S Longtine
- Department of Obstetrics and GynecologyWashington University School of Medicine, St Louis, Missouri, USA
| | - Baosheng Chen
- Department of Obstetrics and GynecologyWashington University School of Medicine, St Louis, Missouri, USA
| | - Maria Laura Costa
- Department of Obstetrics and GynecologyWashington University School of Medicine, St Louis, Missouri, USA.,Department of Obstetrics and GynecologyUniversidade Estadual de Campinas, Cidade Universitaria Zeferino Vaz, Campinas, Brazil
| | - D Michael Nelson
- Department of Obstetrics and GynecologyWashington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
33
|
Penke M, Schuster S, Gorski T, Gebhardt R, Kiess W, Garten A. Oleate ameliorates palmitate-induced reduction of NAMPT activity and NAD levels in primary human hepatocytes and hepatocarcinoma cells. Lipids Health Dis 2017; 16:191. [PMID: 28974242 PMCID: PMC5627432 DOI: 10.1186/s12944-017-0583-6] [Citation(s) in RCA: 15] [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: 05/10/2017] [Accepted: 09/26/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Nicotinamide phosphoribosyltransferase (NAMPT) and nicotinamide adenine dinucleotide (NAD) levels are crucial for liver function. The saturated fatty acid palmitate and the unsaturated fatty acid oleate are the main free fatty acids in adipose tissue and human diet. We asked how these fatty acids affect cell survival, NAMPT and NAD levels in HepG2 cells and primary human hepatocytes. METHODS HepG2 cells were stimulated with palmitate (0.5mM), oleate (1mM) or a combination of both (0.5mM/1mM) as well as nicotinamide mononucleotide (NMN) (0.5 mM) or the specific NAMPT inhibitor FK866 (10nM). Cell survival was measured by WST-1 assay and Annexin V/propidium iodide staining. NAD levels were determined by NAD/NADH Assay or HPLC. Protein and mRNA levels were analysed by Western blot analyses and qPCR, respectively. NAMPT enzyme activity was measured using radiolabelled 14C-nicotinamide. Lipids were stained by Oil red O staining. RESULTS Palmitate significantly reduced cell survival and induced apoptosis at physiological doses. NAMPT activity and NAD levels significantly declined after 48h of palmitate. In addition, NAMPT mRNA expression was enhanced which was associated with increased NAMPT release into the supernatant, while intracellular NAMPT protein levels remained stable. Oleate alone did not influence cell viability and NAMPT activity but ameliorated the negative impact of palmitate on cell survival, NAMPT activity and NAD levels, as well as the increased NAMPT mRNA expression and secretion. NMN was able to normalize intracellular NAD levels but did not ameliorate cell viability after co-stimulation with palmitate. FK866, a specific NAMPT inhibitor did not influence lipid accumulation after oleate-treatment. CONCLUSIONS Palmitate targets NAMPT activity with a consequent cellular depletion of NAD. Oleate protects from palmitate-induced apoptosis and variation of NAMPT and NAD levels. Palmitate-induced cell stress leads to an increase of NAMPT mRNA and accumulation in the supernatant. However, the proapoptotic action of palmitate seems not to be mediated by decreased NAD levels.
Collapse
Affiliation(s)
- Melanie Penke
- Center for Pediatric Research Leipzig (CPL), University Hospital for Children & Adolescents, University of Leipzig, Liebigstraße 21, 04103 Leipzig, Germany
| | - Susanne Schuster
- Center for Pediatric Research Leipzig (CPL), University Hospital for Children & Adolescents, University of Leipzig, Liebigstraße 21, 04103 Leipzig, Germany
| | - Theresa Gorski
- Center for Pediatric Research Leipzig (CPL), University Hospital for Children & Adolescents, University of Leipzig, Liebigstraße 21, 04103 Leipzig, Germany
| | - Rolf Gebhardt
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany
| | - Wieland Kiess
- Center for Pediatric Research Leipzig (CPL), University Hospital for Children & Adolescents, University of Leipzig, Liebigstraße 21, 04103 Leipzig, Germany
| | - Antje Garten
- Center for Pediatric Research Leipzig (CPL), University Hospital for Children & Adolescents, University of Leipzig, Liebigstraße 21, 04103 Leipzig, Germany
| |
Collapse
|
34
|
Sasson S. Nutrient overload, lipid peroxidation and pancreatic beta cell function. Free Radic Biol Med 2017; 111:102-109. [PMID: 27600453 DOI: 10.1016/j.freeradbiomed.2016.09.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 09/02/2016] [Indexed: 12/16/2022]
Abstract
Since the landmark discovery of α,β-unsaturated 4-hydroxyalkenals by Esterbauer and colleagues most studies have addressed the consequences of the tendency of these lipid peroxidation products to form covalent adducts with macromolecules and modify cellular functions. Many studies describe detrimental and cytotoxic effects of 4-hydroxy-2E-nonenal (4-HNE) in myriad tissues and organs and many pathologies. Other studies similarly assigned unfavorable effects to 4-hydroxy-2E-hexenal (4-HHE) and 4-hydroxy-2E,6Z-dodecadienal (4-HDDE). Nutrient overload (e.g., hyperglycemia, hyperlipidemia) modifies lipid metabolism in cells and promotes lipid peroxidation and the generation of α,β-unsaturated 4-hydroxyalkenals. Advances glycation- and lipoxidation end products (AGEs and ALEs) have been associated with the development of insulin resistance and pancreatic beta cell dysfunction and the etiology of type 2 diabetes and its peripheral complications. Less acknowledged are genuine signaling properties of 4-hydroxyalkenals in hormetic processes that provide defense against the consequences of nutrient overload. This review addresses recent findings on such lipohormetic mechanisms that are associated with lipid peroxidation in pancreatic beta cells. This article is part of a Special Issue entitled SI: LIPID OXIDATION PRODUCTS, edited by Giuseppe Poli.
Collapse
Affiliation(s)
- Shlomo Sasson
- Institute for Drug Research, Section of Pharmacology, Diabetes Research Unit, Hebrew University Faculty of Medicine, Jerusalem 9112001, Israel.
| |
Collapse
|
35
|
Protective effects of a G. lucidum proteoglycan on INS-1 cells against IAPP-induced apoptosis via attenuating endoplasmic reticulum stress and modulating CHOP/JNK pathways. Int J Biol Macromol 2017; 106:893-900. [PMID: 28893685 DOI: 10.1016/j.ijbiomac.2017.08.089] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 08/03/2017] [Accepted: 08/14/2017] [Indexed: 01/09/2023]
Abstract
Fudan-Yueyang-G. lucidum (FYGL) is a water-soluble macromolecular proteoglycan extracted from Ganoderma lucidum which has been used for health promotion for a long time in China. The aim of this study was to investigate the protective effects of FYGL on INS-1 rat insulinoma beta cells against IAPP-induced cell apoptosis, as well as the underlying mechanisms. The results showed that apoptotic cells were significantly increased when incubated with islet amyloid polypeptide (IAPP). However, cytotoxicity of IAPP was significantly attenuated by co-incubation of the cells with FYGL. The results of RT-PCR showed that mRNA expression of caspase-3, caspase-12 and C/EBP homologous protein (CHOP) in IAPP-treated cells were inhibited by FYGL. Moreover, FYGL significantly prevented the IAPP-induced abnormal expression of inositol-requiring protein-1α (IRE1α), protein kinase RNA (PKR)-like ER kinase (PERK), activating transcription factor 6 (ATF6), as well as suppressed the activation of CHOP and c-Jun N-terminal kinase (JNK). Taken together, our results suggest that FYGL protects INS-1 pancreatic beta cells against IAPP-induced apoptosis through attenuating endoplasmic reticulum stress (ERS) and modulating CHOP/JNK pathways.
Collapse
|
36
|
Lindholm D, Korhonen L, Eriksson O, Kõks S. Recent Insights into the Role of Unfolded Protein Response in ER Stress in Health and Disease. Front Cell Dev Biol 2017; 5:48. [PMID: 28540288 PMCID: PMC5423914 DOI: 10.3389/fcell.2017.00048] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 04/13/2017] [Indexed: 12/20/2022] Open
Abstract
Unfolded stress response (UPR) is a conserved cellular pathway involved in protein quality control to maintain homeostasis under different conditions and disease states characterized by cell stress. Although three general schemes of and genes induced by UPR are rather well-established, open questions remain including the precise role of UPR in human diseases and the interactions between different sensor systems during cell stress signaling. Particularly, the issue how the normally adaptive and pro-survival UPR pathway turns into a deleterious process causing sustained endoplasmic reticulum (ER) stress and cell death requires more studies. UPR is also named a friend with multiple personalities that we need to understand better to fully recognize its role in normal physiology and in disease pathology. UPR interacts with other organelles including mitochondria, and with cell stress signals and degradation pathways such as autophagy and the ubiquitin proteasome system. Here we review current concepts and mechanisms of UPR as studied in different cells and model systems and highlight the relevance of UPR and related stress signals in various human diseases.
Collapse
Affiliation(s)
- Dan Lindholm
- Medicum, Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of HelsinkiHelsinki, Finland.,Minerva Foundation Institute for Medical ResearchHelsinki, Finland
| | - Laura Korhonen
- Minerva Foundation Institute for Medical ResearchHelsinki, Finland.,Division of Child Psychiatry, Helsinki University Central HospitalHelsinki, Finland
| | - Ove Eriksson
- Medicum, Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of HelsinkiHelsinki, Finland
| | - Sulev Kõks
- Department of Pathophysiology, University of TartuTartu, Estonia.,Department of Reproductive Biology, Estonian University of Life SciencesTartu, Estonia
| |
Collapse
|
37
|
Debbabi M, Zarrouk A, Bezine M, Meddeb W, Nury T, Badreddine A, Karym EM, Sghaier R, Bretillon L, Guyot S, Samadi M, Cherkaoui-Malki M, Nasser B, Mejri M, Ben-Hammou S, Hammami M, Lizard G. Comparison of the effects of major fatty acids present in the Mediterranean diet (oleic acid, docosahexaenoic acid) and in hydrogenated oils (elaidic acid) on 7-ketocholesterol-induced oxiapoptophagy in microglial BV-2 cells. Chem Phys Lipids 2017; 207:151-170. [PMID: 28408132 DOI: 10.1016/j.chemphyslip.2017.04.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 04/05/2017] [Indexed: 01/12/2023]
Abstract
Increased levels of 7-ketocholesterol (7KC), which results mainly from cholesterol auto-oxidation, are often found in the plasma and/or cerebrospinal fluid of patients with neurodegenerative diseases and might contribute to activation of microglial cells involved in neurodegeneration. As major cellular dysfunctions are induced by 7KC, it is important to identify molecules able to impair its side effects. Since consumption of olive and argan oils, and fish is important in the Mediterranean diet, the aim of the study was to determine the ability of oleic acid (OA), a major compound of olive and argan oil, and docosahexaenoic acid (DHA) present in fatty fishes, such as sardines, to attenuate 7KC-induced cytotoxic effects. Since elaidic acid (EA), the trans isomer of OA, can be found in hydrogenated cooking oils and fried foods, its effects on 7KC-induced cytotoxicity were also determined. In murine microglial BV-2 cells, 7KC induces cell growth inhibition, mitochondrial dysfunctions, reactive oxygen species overproduction and lipid peroxidation, increased plasma membrane permeability and fluidity, nuclei condensation and/or fragmentation and caspase-3 activation, which are apoptotic characteristics, and an increased LC3-II/LC3-I ratio, which is a criterion of autophagy. 7KC is therefore a potent inducer of oxiapoptophagy (OXIdation+APOPTOsis+autoPHAGY) on BV-2 cells. OA and EA, but not DHA, also favor the accumulation of lipid droplets revealed with Masson's trichrome, Oil Red O, and Nile Red staining. The cytotoxicity of 7KC was strongly attenuated by OA and DHA. Protective effects were also observed with EA. However, 7KC-induced caspase-3 activation was less attenuated with EA. Different effects of OA and EA on autophagy were also observed. In addition, EA (but not OA) increased plasma membrane fluidity, and only OA (but not EA) was able to prevent the 7KC-induced increase in plasma membrane fluidity. Thus, in BV-2 microglial cells, the principal fatty acids of the Mediterranean diet (OA, DHA) were able to attenuate the major toxic effects of 7KC, thus reinforcing the interest of natural compounds present in the Mediterranean diet to prevent the development of neurodegenerative diseases.
Collapse
Affiliation(s)
- Meryam Debbabi
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France; Univ Monastir, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', Monastir, Tunisia
| | - Amira Zarrouk
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France; Univ Monastir, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', Monastir, Tunisia; Univ Sousse, Faculty of Medicine, Sousse, Tunisia
| | - Maryem Bezine
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France; Univ Tunis El Manar - Pasteur Institut, Lab. 'Venoms & Therapeutic Biomolecules', Tunis, Tunisia
| | - Wiem Meddeb
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France; Univ Carthage, Faculty of Sciences, Bizerte, Tunisia
| | - Thomas Nury
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France
| | - Asmaa Badreddine
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France; Lab of 'Biochemistry of Neuroscience', Univ. Hassan I, Settat, Morocco
| | - El Mostafa Karym
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France; Lab of 'Biochemistry of Neuroscience', Univ. Hassan I, Settat, Morocco
| | - Randa Sghaier
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France; Univ Monastir, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', Monastir, Tunisia; Univ Sousse, Faculty of Medicine, Sousse, Tunisia
| | - Lionel Bretillon
- Eye & Nutrition Research Group, CSGA, UMR 1324 INRA, 6265 CNRS, Univ. Bourgogne Franche-Comté, Dijon, France
| | | | - Mohammad Samadi
- LCPMC-A2, ICPM, Département de Chimie, Université de Lorraine, Metz, France
| | - Mustapha Cherkaoui-Malki
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France
| | - Boubker Nasser
- Lab of 'Biochemistry of Neuroscience', Univ. Hassan I, Settat, Morocco
| | - Mondher Mejri
- Univ Carthage, Faculty of Sciences, Bizerte, Tunisia
| | - Sofien Ben-Hammou
- Department of Neurology, University Hospital Sahloul, 4000 Sousse, Tunisia
| | - Mohamed Hammami
- Univ Monastir, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', Monastir, Tunisia
| | - Gérard Lizard
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France.
| |
Collapse
|
38
|
Clearing the outer mitochondrial membrane from harmful proteins via lipid droplets. Cell Death Discov 2017; 3:17016. [PMID: 28386457 PMCID: PMC5357670 DOI: 10.1038/cddiscovery.2017.16] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/27/2017] [Accepted: 02/10/2017] [Indexed: 01/25/2023] Open
Abstract
In recent years it turned out that there is not only extensive communication between the nucleus and mitochondria but also between mitochondria and lipid droplets (LDs) as well. We were able to demonstrate that a number of proteins shuttle between LDs and mitochondria and it depends on the metabolic state of the cell on which organelle these proteins are predominantly localized. Responsible for the localization of the particular proteins is a protein domain consisting of two α-helices, which we termed V-domain according to the predicted structure. So far we have detected this domain in the following proteins: mammalian BAX, BCL-XL, TCTP and yeast Mmi1p and Erg6p. According to our experiments there are two functions of this domain: (1) shuttling of proteins to mitochondria in times of stress and apoptosis; (2) clearing the outer mitochondrial membrane from pro- as well as anti-apoptotic proteins by moving them to LDs after the stress ceases. In this way the LDs are used by the cell to modulate stress response.
Collapse
|
39
|
Protective Effects of α-Tocopherol, γ-Tocopherol and Oleic Acid, Three Compounds of Olive Oils, and No Effect of Trolox, on 7-Ketocholesterol-Induced Mitochondrial and Peroxisomal Dysfunction in Microglial BV-2 Cells. Int J Mol Sci 2016; 17:ijms17121973. [PMID: 27897980 PMCID: PMC5187773 DOI: 10.3390/ijms17121973] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/08/2016] [Accepted: 11/17/2016] [Indexed: 01/18/2023] Open
Abstract
Lipid peroxidation products, such as 7-ketocholesterol (7KC), may be increased in the body fluids and tissues of patients with neurodegenerative diseases and trigger microglial dysfunction involved in neurodegeneration. It is therefore important to identify synthetic and natural molecules able to impair the toxic effects of 7KC. We determined the impact of 7KC on murine microglial BV-2 cells, especially its ability to trigger mitochondrial and peroxisomal dysfunction, and evaluated the protective effects of α- and γ-tocopherol, Trolox, and oleic acid (OA). Multiple complementary chemical assays, flow cytometric and biochemical methods were used to evaluate the antioxidant and cytoprotective properties of these molecules. According to various complementary assays to estimate antioxidant activity, only α-, and γ-tocopherol, and Trolox had antioxidant properties. However, only α-tocopherol, γ-tocopherol and OA were able to impair 7KC-induced loss of mitochondrial transmembrane potential, which is associated with increased permeability to propidium iodide, an indicator of cell death. In addition, α-and γ-tocopherol, and OA were able to prevent the decrease in Abcd3 protein levels, which allows the measurement of peroxisomal mass, and in mRNA levels of Abcd1 and Abcd2, which encode for two transporters involved in peroxisomal β-oxidation. Thus, 7KC-induced side effects are associated with mitochondrial and peroxisomal dysfunction which can be inversed by natural compounds, thus supporting the hypothesis that the composition of the diet can act on the function of organelles involved in neurodegenerative diseases.
Collapse
|
40
|
Baumann J, Wong J, Sun Y, Conklin DS. Palmitate-induced ER stress increases trastuzumab sensitivity in HER2/neu-positive breast cancer cells. BMC Cancer 2016; 16:551. [PMID: 27464732 PMCID: PMC4964104 DOI: 10.1186/s12885-016-2611-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 07/25/2016] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND HER2/neu-positive breast cancer cells have recently been shown to use a unique Warburg-like metabolism for survival and aggressive behavior. These cells exhibit increased fatty acid synthesis and storage compared to normal breast cells or other tumor cells. Disruption of this synthetic process results in apoptosis. Since the addition of physiological doses of exogenous palmitate induces cell death in HER2/neu-positive breast cancer cells, the pathway is likely operating at its limits in these cells. We have studied the response of HER2/neu-positive breast cancer cells to physiological concentrations of exogenous palmitate to identify lipotoxicity-associated consequences of this physiology. Since epidemiological data show that a diet rich in saturated fatty acids is negatively associated with the development of HER2/neu-positive cancer, this cellular physiology may be relevant to the etiology and treatment of the disease. We sought to identify signaling pathways that are regulated by physiological concentrations of exogenous palmitate specifically in HER2/neu-positive breast cancer cells and gain insights into the molecular mechanism and its relevance to disease prevention and treatment. METHODS Transcriptional profiling was performed to assess programs that are regulated in HER2-normal MCF7 and HER2/neu-positive SKBR3 breast cancer cells in response to exogenous palmitate. Computational analyses were used to define and predict functional relationships and identify networks that are differentially regulated in the two cell lines. These predictions were tested using reporter assays, fluorescence-based high content microscopy, flow cytometry and immunoblotting. Physiological effects were confirmed in HER2/neu-positive BT474 and HCC1569 breast cancer cell lines. RESULTS Exogenous palmitate induces functionally distinct transcriptional programs in HER2/neu-positive breast cancer cells. In the lipogenic HER2/neu-positive SKBR3 cell line, palmitate induces a G2 phase cell cycle delay and CHOP-dependent apoptosis as well as a partial activation of the ER stress response network via XBP1 and ATF6. This response appears to be a general feature of HER2/neu-positive breast cancer cells but not cells that overexpress only HER2/neu. Exogenous palmitate reduces HER2 and HER3 protein levels without changes in phosphorylation and sensitizes HER2/neu-positive breast cancer cells to treatment with the HER2-targeted therapy trastuzumab. CONCLUSIONS Several studies have shown that HER2, FASN and fatty acid synthesis are functionally linked. Exogenous palmitate exerts its toxic effects in part through inducing ER stress, reducing HER2 expression and thereby sensitizing cells to trastuzumab. These data provide further evidence that HER2 signaling and fatty acid metabolism are highly integrated processes that may be important for disease development and progression.
Collapse
Affiliation(s)
- Jan Baumann
- Department of Biomedical Sciences, Cancer Research Center, State University of New York, University at Albany, Rensselaer, NY, 12144, USA
| | - Jason Wong
- Department of Biomedical Sciences, Cancer Research Center, State University of New York, University at Albany, Rensselaer, NY, 12144, USA
| | - Yan Sun
- Department of Biomedical Sciences, Cancer Research Center, State University of New York, University at Albany, Rensselaer, NY, 12144, USA
| | - Douglas S Conklin
- Department of Biomedical Sciences, Cancer Research Center, State University of New York, University at Albany, Rensselaer, NY, 12144, USA.
| |
Collapse
|
41
|
Ochi T, Munekage K, Ono M, Higuchi T, Tsuda M, Hayashi Y, Okamoto N, Toda K, Sakamoto S, Oben JA, Saibara T. Patatin-like phospholipase domain-containing protein 3 is involved in hepatic fatty acid and triglyceride metabolism through X-box binding protein 1 and modulation of endoplasmic reticulum stress in mice. Hepatol Res 2016; 46:584-92. [PMID: 26347999 DOI: 10.1111/hepr.12587] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/02/2015] [Accepted: 09/02/2015] [Indexed: 12/13/2022]
Abstract
AIM Non-alcoholic steatohepatitis (NASH) is the major cause of chronic liver disease worldwide. Endoplasmic reticulum (ER) stress is considered to be an important pathological characteristic in NASH. A sequence variation (I148M) in the patatin-like phospholipase domain-containing protein 3/adiponutrin (PNPLA3) gene is known to be associated with the development of NASH. However, PNPLA3 deficiency has been considered to not be associated with fatty liver disease. To clarify, therefore, the role of PNPLA3 in liver, we established PNPLA3 knockout (KO) mice and investigated the phenotypes and involved factors under ER stress. METHODS ER stress was induced by i.p. injection with tunicamycin or with saline at 0 and 24 h in KO and C57BL/6 (wild-type [WT]) mice. At 48 h after the starting of treatment, blood and liver samples were studied. RESULTS Hepatic steatosis and triglyceride content were remarkably increased in WT mice than in KO mice under ER stress. The hepatic palmitate/oleate ratio was significantly higher originally in KO mice than in WT mice. Moreover, the expression of stearoyl-coenzyme A desaturase-1 (SCD1) in KO mice under ER stress was decreased further than that in WT mice. Expression of ER stress markers X-box binding protein 1 (XBP1) and ERdj4 was increased in WT mice but not in KO mice under ER stress. CONCLUSION We first demonstrated the hepatic phenotype of PNPLA3 deficiency under ER stress. Our observations would indicate that PNPLA3 has an important role in hepatic fatty acid metabolism and triglyceride accumulation through XBP1 under ER stress.
Collapse
Affiliation(s)
- Tsunehiro Ochi
- Departments of Gastroenterology and Hepatology, Kochi Medical School, Kochi, Japan
| | - Kensuke Munekage
- Departments of Gastroenterology and Hepatology, Kochi Medical School, Kochi, Japan
| | - Masafumi Ono
- Departments of Gastroenterology and Hepatology, Kochi Medical School, Kochi, Japan
| | - Takuma Higuchi
- Laboratory of Molecular Biology, Science Research Center, Kochi Medical School, Kochi, Japan
| | - Masayuki Tsuda
- The Division of Laboratory Animal Science, Science Research Center, Kochi Medical School, Kochi, Japan
| | | | - Nobuto Okamoto
- Departments of Gastroenterology and Hepatology, Kochi Medical School, Kochi, Japan
| | - Katsumi Toda
- Department of Biochemistry, Kochi Medical School, Kochi, Japan
| | - Shuji Sakamoto
- Laboratory of Molecular Biology, Science Research Center, Kochi Medical School, Kochi, Japan
| | - Jude A Oben
- Institute for Liver and Digestive Health, Royal Free Hospital, University College London.,Department of Gastroenterology & Hepatology, Guy's and St. Thomas' Hospital, London, UK
| | - Toshiji Saibara
- Departments of Gastroenterology and Hepatology, Kochi Medical School, Kochi, Japan
| |
Collapse
|
42
|
Fernández del Río L, Gutiérrez-Casado E, Varela-López A, Villalba JM. Olive Oil and the Hallmarks of Aging. Molecules 2016; 21:163. [PMID: 26840281 PMCID: PMC6273542 DOI: 10.3390/molecules21020163] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/20/2016] [Accepted: 01/22/2016] [Indexed: 12/30/2022] Open
Abstract
Aging is a multifactorial and tissue-specific process involving diverse alterations regarded as the "hallmarks of aging", which include genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion and altered intracellular communication. Virtually all these hallmarks are targeted by dietary olive oil, particularly by virgin olive oil, since many of its beneficial effects can be accounted not only for the monounsaturated nature of its predominant fatty acid (oleic acid), but also for the bioactivity of its minor compounds, which can act on cells though both direct and indirect mechanisms due to their ability to modulate gene expression. Among the minor constituents of virgin olive oil, secoiridoids stand out for their capacity to modulate many pathways that are relevant for the aging process. Attenuation of aging-related alterations by olive oil or its minor compounds has been observed in cellular, animal and human models. How olive oil targets the hallmarks of aging could explain the improvement of health, reduced risk of aging-associated diseases, and increased longevity which have been associated with consumption of a typical Mediterranean diet containing this edible oil as the predominant fat source.
Collapse
Affiliation(s)
- Lucía Fernández del Río
- Department of Cell Biology, Physiology and Immunology, Agrifood Campus of International Excellence ceiA3, University of Córdoba, Campus Rabanales, Severo Ochoa Building, 14014 Córdoba, Spain.
| | - Elena Gutiérrez-Casado
- Department of Cell Biology, Physiology and Immunology, Agrifood Campus of International Excellence ceiA3, University of Córdoba, Campus Rabanales, Severo Ochoa Building, 14014 Córdoba, Spain.
| | - Alfonso Varela-López
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix", Biomedical Research Center (CIBM), University of Granada, Avda. del Conocimiento s.n., Armilla, 18100 Granada, Spain.
| | - José M Villalba
- Department of Cell Biology, Physiology and Immunology, Agrifood Campus of International Excellence ceiA3, University of Córdoba, Campus Rabanales, Severo Ochoa Building, 14014 Córdoba, Spain.
| |
Collapse
|
43
|
Cataldo LR, Mizgier ML, Busso D, Olmos P, Galgani JE, Valenzuela R, Mezzano D, Aranda E, Cortés VA, Santos JL. Serotonin- and Dopamine-Related Gene Expression in db/db Mice Islets and in MIN6 β-Cells Treated with Palmitate and Oleate. J Diabetes Res 2016; 2016:3793781. [PMID: 27366756 PMCID: PMC4913013 DOI: 10.1155/2016/3793781] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/26/2016] [Accepted: 05/10/2016] [Indexed: 12/20/2022] Open
Abstract
High circulating nonesterified fatty acids (NEFAs) concentration, often reported in diabetes, leads to impaired glucose-stimulated insulin secretion (GSIS) through not yet well-defined mechanisms. Serotonin and dopamine might contribute to NEFA-dependent β-cell dysfunction, since extracellular signal of these monoamines decreases GSIS. Moreover, palmitate-treated β-cells may enhance the expression of the serotonin receptor Htr2c, affecting insulin secretion. Additionally, the expression of monoamine-oxidase type B (Maob) seems to be lower in islets from humans and mice with diabetes compared to nondiabetic islets, which may lead to increased monoamine concentrations. We assessed the expression of serotonin- and dopamine-related genes in islets from db/db and wild-type (WT) mice. In addition, the effect of palmitate and oleate on the expression of such genes, 5HT content, and GSIS in MIN6 β-cell was determined. Lower Maob expression was found in islets from db/db versus WT mice and in MIN6 β-cells in response to palmitate and oleate treatment compared to vehicle. Reduced 5HT content and impaired GSIS in response to palmitate (-25%; p < 0.0001) and oleate (-43%; p < 0.0001) were detected in MIN6 β-cells. In conclusion, known defects of GSIS in islets from db/db mice and MIN6 β-cells treated with NEFAs are accompanied by reduced Maob expression and reduced 5HT content.
Collapse
Affiliation(s)
- L. R. Cataldo
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
- Facultad de Medicina, Universidad de los Andes, 7620001 Santiago, Chile
| | - M. L. Mizgier
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - D. Busso
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - P. Olmos
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - J. E. Galgani
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
- UDA-Ciencias de la Salud, Carrera de Nutrición y Dietética, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - R. Valenzuela
- Departamento de Nutrición, Facultad de Medicina, Universidad de Chile, 7550367 Santiago, Chile
| | - D. Mezzano
- Laboratorio de Hemostasia, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - E. Aranda
- Laboratorio de Hemostasia, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - V. A. Cortés
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - J. L. Santos
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
- *J. L. Santos:
| |
Collapse
|
44
|
Salvadó L, Palomer X, Barroso E, Vázquez-Carrera M. Targeting endoplasmic reticulum stress in insulin resistance. Trends Endocrinol Metab 2015; 26:438-48. [PMID: 26078196 DOI: 10.1016/j.tem.2015.05.007] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 05/14/2015] [Accepted: 05/15/2015] [Indexed: 02/06/2023]
Abstract
The endoplasmic reticulum (ER) is involved in the development of insulin resistance and progression to type 2 diabetes mellitus (T2DM). Disruption of ER homeostasis leads to ER stress, which activates the unfolded protein response (UPR). This response is linked to different processes involved in the development of insulin resistance (IR) and T2DM, including inflammation, lipid accumulation, insulin biosynthesis, and β-cell apoptosis. Understanding the mechanisms by which disruption of ER homeostasis leads to IR and its progression to T2DM may offer new pharmacological targets for the treatment and prevention of these diseases. Here, we examine ER stress, the UPR, and downstream pathways in insulin sensitive tissues, and in IR, and offer insights towards therapeutic strategies.
Collapse
Affiliation(s)
- Laia Salvadó
- Department of Pharmacology and Therapeutic Chemistry, Faculty of Pharmacy, University of Barcelona, Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
| | - Xavier Palomer
- Department of Pharmacology and Therapeutic Chemistry, Faculty of Pharmacy, University of Barcelona, Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
| | - Emma Barroso
- Department of Pharmacology and Therapeutic Chemistry, Faculty of Pharmacy, University of Barcelona, Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology and Therapeutic Chemistry, Faculty of Pharmacy, University of Barcelona, Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
45
|
Iwao C, Shidoji Y. Polyunsaturated Branched-Chain Fatty Acid Geranylgeranoic Acid Induces Unfolded Protein Response in Human Hepatoma Cells. PLoS One 2015; 10:e0132761. [PMID: 26186544 PMCID: PMC4506074 DOI: 10.1371/journal.pone.0132761] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 06/17/2015] [Indexed: 01/22/2023] Open
Abstract
The acyclic diterpenoid acid geranylgeranoic acid (GGA) has been reported to induce autophagic cell death in several human hepatoma-derived cell lines; however, the molecular mechanism for this remains unknown. In the present study, several diterpenoids were examined for ability to induce XBP1 splicing and/or lipotoxicity for human hepatoma cell lines. Here we show that three groups of diterpenoids emerged: 1) GGA, 2,3-dihydro GGA and 9-cis retinoic acid induce cell death and XBP1 splicing; 2) all-trans retinoic acid induces XBP1 splicing but little cell death; and 3) phytanic acid, phytenic acid and geranylgeraniol induce neither cell death nor XBP1 splicing. GGA-induced ER stress/ unfolded protein response (UPR) and its lipotoxicity were both blocked by co-treatment with oleic acid. The blocking activity of oleic acid for GGA-induced XBP1 splicing was not attenuated by methylation of oleic acid. These findings strongly suggest that GGA at micromolar concentrations induces the so-called lipid-induced ER stress response/UPR, which is oleate-suppressive, and shows its lipotoxicity in human hepatoma cells.
Collapse
Affiliation(s)
- Chieko Iwao
- Molecular and Cellular Biology, Graduate School of Human Health Science, University of Nagasaki, Nagasaki, Japan
| | - Yoshihiro Shidoji
- Molecular and Cellular Biology, Graduate School of Human Health Science, University of Nagasaki, Nagasaki, Japan
- * E-mail:
| |
Collapse
|
46
|
Mihai AD, Schröder M. Glucose starvation and hypoxia, but not the saturated fatty acid palmitic acid or cholesterol, activate the unfolded protein response in 3T3-F442A and 3T3-L1 adipocytes. Adipocyte 2015; 4:188-202. [PMID: 26257992 DOI: 10.4161/21623945.2014.989728] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 11/08/2014] [Accepted: 11/14/2014] [Indexed: 12/26/2022] Open
Abstract
Obesity is associated with endoplasmic reticulum (ER) stress and activation of the unfolded protein response (UPR) in adipose tissue. In this study we identify physiological triggers of ER stress and of the UPR in adipocytes in vitro. We show that two markers of adipose tissue remodelling in obesity, glucose starvation and hypoxia, cause ER stress in 3T3-F442A and 3T3-L1 adipocytes. Both conditions induced molecular markers of the IRE1α and PERK branches of the UPR, such as splicing of XBP1 mRNA and CHOP, as well as transcription of the ER stress responsive gene BiP. Hypoxia also induced an increase in phosphorylation of the PERK substrate eIF2α. By contrast, physiological triggers of ER stress in many other cell types, such as the saturated fatty acid palmitic acid, cholesterol, or several inflammatory cytokines including TNF-α, IL-1β, and IL-6, do not cause ER stress in 3T3-F442A and 3T3-L1 adipocytes. Our data suggest that physiological changes associated with remodelling of adipose tissue in obesity, such as hypoxia and glucose starvation, are more likely physiological ER stressors of adipocytes than the lipid overload or hyperinsulinemia associated with obesity.
Collapse
|
47
|
Japtok L, Schmitz EI, Fayyaz S, Krämer S, Hsu LJ, Kleuser B. Sphingosine 1-phosphate counteracts insulin signaling in pancreatic β-cells via the sphingosine 1-phosphate receptor subtype 2. FASEB J 2015; 29:3357-69. [PMID: 25911610 DOI: 10.1096/fj.14-263194] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 04/16/2015] [Indexed: 01/04/2023]
Abstract
Glucolipotoxic stress has been identified as a key player in the progression of pancreatic β-cell dysfunction contributing to insulin resistance and the development of type 2 diabetes mellitus (T2D). It has been suggested that bioactive lipid intermediates, formed under lipotoxic conditions, are involved in these processes. Here, we show that sphingosine 1-phosphate (S1P) levels are not only increased in palmitate-stimulated pancreatic β-cells but also regulate β-cell homeostasis in a divergent manner. Although S1P possesses a prosurvival effect in β-cells, an enhanced level of the sphingolipid antagonizes insulin-mediated cell growth and survival via the sphingosine 1-phosphate receptor subtype 2 (S1P2) followed by an inhibition of Akt-signaling. In an attempt to investigate the role of the S1P/S1P2 axis in vivo, the New Zealand obese (NZO) diabetic mouse model, characterized by β-cell loss under high-fat diet (HFD) conditions, was used. The occurrence of T2D was accompanied by an increase of plasma S1P levels. To examine whether S1P contributes to the morphologic changes of islets via S1P2, the receptor antagonist JTE-013 was administered. Most interestingly, JTE-013 rescued β-cell damage clearly indicating an important role of the S1P2 in β-cell homeostasis. Therefore, the present study provides a new therapeutic strategy to diminish β-cell dysfunction and the development of T2D.
Collapse
Affiliation(s)
- Lukasz Japtok
- *Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany; German Institute of Human Nutrition, Max Rubner Laboratory, Nuthetal, Germany; and Lpath Incorporated, San Diego, California, USA
| | - Elisabeth I Schmitz
- *Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany; German Institute of Human Nutrition, Max Rubner Laboratory, Nuthetal, Germany; and Lpath Incorporated, San Diego, California, USA
| | - Susann Fayyaz
- *Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany; German Institute of Human Nutrition, Max Rubner Laboratory, Nuthetal, Germany; and Lpath Incorporated, San Diego, California, USA
| | - Stephanie Krämer
- *Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany; German Institute of Human Nutrition, Max Rubner Laboratory, Nuthetal, Germany; and Lpath Incorporated, San Diego, California, USA
| | - Leigh J Hsu
- *Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany; German Institute of Human Nutrition, Max Rubner Laboratory, Nuthetal, Germany; and Lpath Incorporated, San Diego, California, USA
| | - Burkhard Kleuser
- *Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany; German Institute of Human Nutrition, Max Rubner Laboratory, Nuthetal, Germany; and Lpath Incorporated, San Diego, California, USA
| |
Collapse
|
48
|
Yamauchi A, Itaya-Hironaka A, Sakuramoto-Tsuchida S, Takeda M, Yoshimoto K, Miyaoka T, Fujimura T, Tsujinaka H, Tsuchida C, Ota H, Takasawa S. Synergistic activations of REG I α and REG I β promoters by IL-6 and Glucocorticoids through JAK/STAT pathway in human pancreatic β cells. J Diabetes Res 2015; 2015:173058. [PMID: 25767811 PMCID: PMC4342170 DOI: 10.1155/2015/173058] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/26/2015] [Indexed: 12/31/2022] Open
Abstract
Reg (Regenerating gene) gene was originally isolated from rat regenerating islets and its encoding protein was revealed as an autocrine/paracrine growth factor for β cells. Rat Reg gene is activated in inflammatory conditions for β cell regeneration. In human, although five functional REG family genes (REG Iα, REG Iβ, REG III, HIP/PAP, and REG IV) were isolated, their expressions in β cells under inflammatory conditions remained unclear. In this study, we found that combined addition of IL-6 and dexamethasone (Dx) induced REG Iα and REG Iβ expression in human 1.1B4 β cells. Promoter assay revealed that a signal transducer and activator of transcription- (STAT-) binding site in each promoter of REG Iα (TGCCGGGAA) and REG Iβ (TGCCAGGAA) was essential for the IL-6+Dx-induced promoter activation. A Janus kinase 2 (JAK2) inhibitor significantly inhibited the IL-6+Dx-induced REG Iα and REG Iβ transcription. Electrophoretic mobility shift assay and chromatin immunoprecipitation revealed that IL-6+Dx stimulation increased STAT3 binding to the REG Iα promoter. Furthermore, small interfering RNA-mediated targeting of STAT3 blocked the IL-6+Dx-induced expression of REG Iα and REG Iβ. These results indicate that the expression of REG Iα and REG Iβ should be upregulated in human β cells under inflammatory conditions through the JAK/STAT pathway.
Collapse
Affiliation(s)
- Akiyo Yamauchi
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | | | | | - Maiko Takeda
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Kiyomi Yoshimoto
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Tomoko Miyaoka
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Takanori Fujimura
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Hiroki Tsujinaka
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Chikatsugu Tsuchida
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Hiroyo Ota
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Shin Takasawa
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| |
Collapse
|
49
|
Kanekura K, Ou J, Hara T, Zhu LJ, Urano F. Research resource: Monitoring endoplasmic reticulum membrane integrity in β-cells at the single-cell level. Mol Endocrinol 2015; 29:473-80. [PMID: 25584413 DOI: 10.1210/me.2014-1260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Endoplasmic reticulum (ER) membrane integrity is an emerging target for human chronic diseases associated with ER stress. Despite the underlying importance of compromised ER membrane integrity in disease states, the entire process leading to ER membrane permeabilization and cell death is still not clear due to technical limitations. Here we describe a novel method for monitoring ER membrane integrity at the single-cell level in real time. Using a β-cell line expressing ER-targeted redox sensitive green fluorescent protein, we could identify a β-cell population undergoing ER membrane permeabilization induced by palmitate and could monitor cell fate and ER stress of these cells at the single-cell level. Our method could be used to develop a novel therapeutic modality targeting the ER membrane for ER-associated disorders, including β-cell death in diabetes, neurodegeneration, and Wolfram syndrome.
Collapse
Affiliation(s)
- Kohsuke Kanekura
- Department of Medicine (K.K., T.H., F.U.), Division of Endocrinology, Metabolism, and Lipid Research, and Department of Pathology and Immunology (F.U.), Washington University School of Medicine, St Louis, Missouri 63110; and Programs in Gene Function and Expression, Molecular Medicine, and Bioinformatics and Integrative Biology (J.O., L.J.Z.), University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | | | | | | | | |
Collapse
|
50
|
Metformin attenuates palmitate-induced endoplasmic reticulum stress, serine phosphorylation of IRS-1 and apoptosis in rat insulinoma cells. PLoS One 2014; 9:e97868. [PMID: 24896641 PMCID: PMC4045581 DOI: 10.1371/journal.pone.0097868] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 04/25/2014] [Indexed: 12/25/2022] Open
Abstract
Lipotoxicity refers to cellular dysfunctions caused by elevated free fatty acid levels playing a central role in the development and progression of obesity related diseases. Saturated fatty acids cause insulin resistance and reduce insulin production in the pancreatic islets, thereby generating a vicious cycle, which potentially culminates in type 2 diabetes. The underlying endoplasmic reticulum (ER) stress response can lead to even β-cell death (lipoapoptosis). Since improvement of β-cell viability is a promising anti-diabetic strategy, the protective effect of metformin, a known insulin sensitizer was studied in rat insulinoma cells. Assessment of palmitate-induced lipoapoptosis by fluorescent microscopy and by detection of caspase-3 showed a significant decrease in metformin treated cells. Attenuation of β-cell lipotoxicity was also revealed by lower induction/activation of various ER stress markers, e.g. phosphorylation of eukaryotic initiation factor 2α (eIF2α), c-Jun N-terminal kinase (JNK), insulin receptor substrate-1 (IRS-1) and induction of CCAAT/enhancer binding protein homologous protein (CHOP). Our results indicate that the β-cell protective activity of metformin in lipotoxicity can be at least partly attributed to suppression of ER stress.
Collapse
|