1
|
Aryal B, Kwakye J, Ariyo OW, Ghareeb AFA, Milfort MC, Fuller AL, Khatiwada S, Rekaya R, Aggrey SE. Major Oxidative and Antioxidant Mechanisms During Heat Stress-Induced Oxidative Stress in Chickens. Antioxidants (Basel) 2025; 14:471. [PMID: 40298812 PMCID: PMC12023971 DOI: 10.3390/antiox14040471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
Heat stress (HS) is one of the most important stressors in chickens, and its adverse effects are primarily caused by disturbing the redox homeostasis. An increase in electron leakage from the mitochondrial electron transport chain is the major source of free radical production under HS, which triggers other enzymatic systems to generate more radicals. As a defense mechanism, cells have enzymatic and non-enzymatic antioxidant systems that work cooperatively against free radicals. The generation of free radicals, particularly the reactive oxygen species (ROS) and reactive nitrogen species (RNS), under HS condition outweighs the cellular antioxidant capacity, resulting in oxidative damage to macromolecules, including lipids, carbohydrates, proteins, and DNA. Understanding these detrimental oxidative processes and protective defense mechanisms is important in developing mitigation strategies against HS. This review summarizes the current understanding of major oxidative and antioxidant systems and their molecular mechanisms in generating or neutralizing the ROS/RNS. Importantly, this review explores the potential mechanisms that lead to the development of oxidative stress in heat-stressed chickens, highlighting their unique behavioral and physiological responses against thermal stress. Further, we summarize the major findings associated with these oxidative and antioxidant mechanisms in chickens.
Collapse
Affiliation(s)
- Bikash Aryal
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
- Department of Animal Sciences, The Ohio State University, Wooster, OH 44691, USA;
| | - Josephine Kwakye
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| | - Oluwatomide W. Ariyo
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| | - Ahmed F. A. Ghareeb
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
- Boehringer Ingelheim Animal Health (BIAH), Gainesville, GA 30501, USA
| | - Marie C. Milfort
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| | - Alberta L. Fuller
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| | - Saroj Khatiwada
- Department of Animal Sciences, The Ohio State University, Wooster, OH 44691, USA;
| | - Romdhane Rekaya
- Department of Animal and Dairy Science, The University of Georgia, Athens, GA 30602, USA;
| | - Samuel E. Aggrey
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| |
Collapse
|
2
|
Moliterni C, Vari F, Schifano E, Tacconi S, Stanca E, Friuli M, Longo S, Conte M, Salvioli S, Gnocchi D, Mazzocca A, Uccelletti D, Vergara D, Dini L, Giudetti AM. Lipotoxicity of palmitic acid is associated with DGAT1 downregulation and abolished by PPARα activation in liver cells. J Lipid Res 2024; 65:100692. [PMID: 39505261 DOI: 10.1016/j.jlr.2024.100692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 10/21/2024] [Accepted: 10/31/2024] [Indexed: 11/08/2024] Open
Abstract
Lipotoxicity refers to the harmful effects of excess fatty acids on metabolic health, and it can vary depending on the type of fatty acids involved. Saturated and unsaturated fatty acids exhibit distinct effects, though the precise mechanisms behind these differences remain unclear. Here, we investigated the lipotoxicity of palmitic acid (PA), a saturated fatty acid, compared with oleic acid (OA), a monounsaturated fatty acid, in the hepatic cell line HuH7. Our results demonstrated that PA, unlike OA, induces lipotoxicity, endoplasmic reticulum (ER) stress, and autophagy inhibition. Compared with OA, PA treatment leads to less lipid droplet (LD) accumulation and a significant reduction in the mRNA and protein level of diacylglycerol acyltransferase 1 (DGAT1), a key enzyme of triacylglycerol synthesis. Using modulators of ER stress and autophagy, we established that DGAT1 downregulation by PA is closely linked to these cellular pathways. Notably, the ER stress inhibitor 4-phenylbutyrate can suppress PA-induced DGAT1 downregulation. Furthermore, knockdown of DGAT1 by siRNA or with A922500, a specific DGAT1 inhibitor, resulted in cell death, even with OA. Both PA and OA increased the oxygen consumption rate; however, the increase associated with PA was only partially coupled to ATP synthesis. Importantly, treatment with GW7647 a specific PPARα agonist mitigated the lipotoxic effects of PA, restoring PA-induced ER stress, autophagy block, and DGAT1 suppression. In conclusion, our study highlights the crucial role of DGAT1 in PA-induced lipotoxicity, broadening the knowledge of the mechanisms underlying hepatic lipotoxicity and providing the basis for potential therapeutic interventions.
Collapse
Affiliation(s)
- Camilla Moliterni
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Francesco Vari
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy; Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Emily Schifano
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Stefano Tacconi
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Eleonora Stanca
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Marzia Friuli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Serena Longo
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Maria Conte
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Stefano Salvioli
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy; IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Davide Gnocchi
- Interdisciplinary Department of Medicine, University of Bari School of Medicine, Bari, Italy
| | - Antonio Mazzocca
- Interdisciplinary Department of Medicine, University of Bari School of Medicine, Bari, Italy
| | - Daniela Uccelletti
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Daniele Vergara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Luciana Dini
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Rome, Italy.
| | - Anna Maria Giudetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy.
| |
Collapse
|
3
|
Qiu X, Lan X, Li L, Chen H, Zhang N, Zheng X, Xie X. The role of perirenal adipose tissue deposition in chronic kidney disease progression: Mechanisms and therapeutic implications. Life Sci 2024; 352:122866. [PMID: 38936605 DOI: 10.1016/j.lfs.2024.122866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024]
Abstract
Chronic kidney disease (CKD) represents a significant and escalating global health challenge, with morbidity and mortality rates rising steadily. Evidence increasingly implicates perirenal adipose tissue (PRAT) deposition as a contributing factor in the pathogenesis of CKD. This review explores how PRAT deposition may exert deleterious effects on renal structure and function. The anatomical proximity of PRAT to the kidneys not only potentially causes mechanical compression but also leads to the dysregulated secretion of adipokines and inflammatory mediators, such as adiponectin, leptin, visfatin, tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and exosomes. Additionally, PRAT deposition may contribute to renal lipotoxicity through elevated levels of free fatty acids (FFA), triglycerides (TAG), diacylglycerol (DAG), and ceramides (Cer). PRAT deposition is also linked to the hyperactivation of the renin-angiotensin-aldosterone system (RAAS), which further exacerbates CKD progression. Recognizing PRAT deposition as an independent risk factor for CKD underscores the potential of targeting PRAT as a novel strategy for the prevention and management of CKD. This review further discusses interventions that could include measuring PRAT thickness to establish a baseline, managing metabolic risk factors that promote its deposition, and inhibiting key PRAT-induced signaling pathways.
Collapse
Affiliation(s)
- Xiang Qiu
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Xin Lan
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Langhui Li
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Huan Chen
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China; Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Ningjuan Zhang
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, China
| | - Xiaoli Zheng
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China.
| | - Xiang Xie
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China.
| |
Collapse
|
4
|
Safi R, Menéndez P, Pol A. Lipid droplets provide metabolic flexibility for cancer progression. FEBS Lett 2024; 598:1301-1327. [PMID: 38325881 DOI: 10.1002/1873-3468.14820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 02/09/2024]
Abstract
A hallmark of cancer cells is their remarkable ability to efficiently adapt to favorable and hostile environments. Due to a unique metabolic flexibility, tumor cells can grow even in the absence of extracellular nutrients or in stressful scenarios. To achieve this, cancer cells need large amounts of lipids to build membranes, synthesize lipid-derived molecules, and generate metabolic energy in the absence of other nutrients. Tumor cells potentiate strategies to obtain lipids from other cells, metabolic pathways to synthesize new lipids, and mechanisms for efficient storage, mobilization, and utilization of these lipids. Lipid droplets (LDs) are the organelles that collect and supply lipids in eukaryotes and it is increasingly recognized that the accumulation of LDs is a new hallmark of cancer cells. Furthermore, an active role of LD proteins in processes underlying tumorigenesis has been proposed. Here, by focusing on three major classes of LD-resident proteins (perilipins, lipases, and acyl-CoA synthetases), we provide an overview of the contribution of LDs to cancer progression and discuss the role of LD proteins during the proliferation, invasion, metastasis, apoptosis, and stemness of cancer cells.
Collapse
Affiliation(s)
- Rémi Safi
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Pablo Menéndez
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Spain
- Consorcio Investigación Biomédica en Red de Cancer, CIBER-ONC, ISCIII, Barcelona, Spain
- Spanish Network for Advanced Cell Therapies (TERAV), Barcelona, Spain
| | - Albert Pol
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Spain
| |
Collapse
|
5
|
Song Q, Wang J, Griffiths A, Lee SM, Iyamu ID, Huang R, Cordoba-Chacon J, Song Z. Nicotinamide N-methyltransferase upregulation contributes to palmitate-elicited peroxisome proliferator-activated receptor transactivation in hepatocytes. Am J Physiol Cell Physiol 2023; 325:C29-C41. [PMID: 37212549 PMCID: PMC10259858 DOI: 10.1152/ajpcell.00010.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/23/2023]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) plays a pivotal role in regulating lipid metabolism and hepatic PPARγ transactivation contributes to fatty liver development. Fatty acids (FAs) are well-known endogenous ligands for PPARγ. Palmitate, a 16-C saturated FA (SFA) and the most abundant SFA in human circulation, is a strong inducer of hepatic lipotoxicity, a central pathogenic factor for various fatty liver diseases. In this study, using both alpha mouse liver 12 (AML12) and primary mouse hepatocytes, we investigated the effects of palmitate on hepatic PPARγ transactivation and underlying mechanisms, as well as the role of PPARγ transactivation in palmitate-induced hepatic lipotoxicity, all of which remain ambiguous currently. Our data revealed that palmitate exposure was concomitant with both PPARγ transactivation and upregulation of nicotinamide N-methyltransferase (NNMT), a methyltransferase catalyzing the degradation of nicotinamide, the predominant precursor for cellular NAD+ biosynthesis. Importantly, we discovered that PPARγ transactivation by palmitate was blunted by NNMT inhibition, suggesting that NNMT upregulation plays a mechanistic role in PPARγ transactivation. Further investigations uncovered that palmitate exposure is associated with intracellular NAD+ decline and NAD+ replenishment with NAD+-enhancing agents, nicotinamide and nicotinamide riboside, obstructed palmitate-induced PPARγ transactivation, implying that cellular NAD+ decline resulted from NNMT upregulation represents a potential mechanism behind palmitate-elicited PPARγ transactivation. At last, our data showed that the PPARγ transactivation marginally ameliorated palmitate-induced intracellular triacylglycerol accumulation and cell death. Collectively, our data provided the first-line evidence supporting that NNMT upregulation plays a mechanistic role in palmitate-elicited PPARγ transactivation, potentially through reducing cellular NAD+ contents.NEW & NOTEWORTHY Hepatic PPARγ transactivation contributes to fatty liver development. Saturated fatty acids (SFAs) induce hepatic lipotoxicity. Here, we investigated whether and how palmitate, the most abundant SFA in the human blood, affects PPARγ transactivation in hepatocytes. We reported for the first time that upregulation of nicotinamide N-methyltransferase (NNMT), a methyltransferase catalyzing the degradation of nicotinamide, the predominant precursor for cellular NAD+ biosynthesis, plays a mechanistic role in regulating palmitate-elicited PPARγ transactivation through reducing intracellular NAD+ contents.
Collapse
Affiliation(s)
- Qing Song
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Jun Wang
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Alexandra Griffiths
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Samuel Man Lee
- Division of Endocrinology/Diabetes & Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Iredia D Iyamu
- Department of Medicinal Chemistry and Molecular Pharmacology, Institute for Drug Discovery, Purdue University, West Lafayette, Indiana, United States
| | - Rong Huang
- Department of Medicinal Chemistry and Molecular Pharmacology, Institute for Drug Discovery, Purdue University, West Lafayette, Indiana, United States
| | - Jose Cordoba-Chacon
- Division of Endocrinology/Diabetes & Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Zhenyuan Song
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois, United States
| |
Collapse
|
6
|
Bresgen N, Kovacs M, Lahnsteiner A, Felder TK, Rinnerthaler M. The Janus-Faced Role of Lipid Droplets in Aging: Insights from the Cellular Perspective. Biomolecules 2023; 13:912. [PMID: 37371492 PMCID: PMC10301655 DOI: 10.3390/biom13060912] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
It is widely accepted that nine hallmarks-including mitochondrial dysfunction, epigenetic alterations, and loss of proteostasis-exist that describe the cellular aging process. Adding to this, a well-described cell organelle in the metabolic context, namely, lipid droplets, also accumulates with increasing age, which can be regarded as a further aging-associated process. Independently of their essential role as fat stores, lipid droplets are also able to control cell integrity by mitigating lipotoxic and proteotoxic insults. As we will show in this review, numerous longevity interventions (such as mTOR inhibition) also lead to strong accumulation of lipid droplets in Saccharomyces cerevisiae, Caenorhabditis elegans, Drosophila melanogaster, and mammalian cells, just to name a few examples. In mammals, due to the variety of different cell types and tissues, the role of lipid droplets during the aging process is much more complex. Using selected diseases associated with aging, such as Alzheimer's disease, Parkinson's disease, type II diabetes, and cardiovascular disease, we show that lipid droplets are "Janus"-faced. In an early phase of the disease, lipid droplets mitigate the toxicity of lipid peroxidation and protein aggregates, but in a later phase of the disease, a strong accumulation of lipid droplets can cause problems for cells and tissues.
Collapse
Affiliation(s)
- Nikolaus Bresgen
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| | - Melanie Kovacs
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| | - Angelika Lahnsteiner
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| | - Thomas Klaus Felder
- Department of Laboratory Medicine, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Mark Rinnerthaler
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| |
Collapse
|
7
|
Karabulut G, Barlas N. Endocrine adverse effects of mono(2-ethylhexyl) phthalate and monobutyl phthalate in male pubertal rats. Arh Hig Rada Toksikol 2022; 73:285-296. [PMID: 36607728 PMCID: PMC9985344 DOI: 10.2478/aiht-2022-73-3617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/01/2022] [Accepted: 11/01/2022] [Indexed: 01/07/2023] Open
Abstract
Considering that research of adverse effects of mono(2-ethylhexyl) phthalate (MEHP) and monobutyl phthalate (MBP), two key metabolites of the most common phthalates used as plasticisers in various daily-life products, has been scattered and limited, the aim of our study was to provide a more comprehensive analysis by focusing on major organ systems, including blood, liver, kidney, and pancreas in 66 male pubertal rats randomised into eleven groups of six. The animals were receiving either metabolite at doses of 25, 50, 100, 200, or 400 mg/kg bw a day by gavage for 28 days. The control group was receiving corn oil. At the end of the experiment, blood samples were collected for biochemical, haematological, and immunological analyses. Samples of kidney, liver, and pancreas were dissected for histopathological analyses. Exposure to either compound resulted in increased liver and decreased pancreas weight, especially at the highest doses. Exposed rats had increased ALT, AST, glucose, and triglyceride levels and decreased total protein and albumin levels. Both compounds increased MCV and decreased haemoglobin levels compared to control. Although they also lowered the insulin level, exposed rats had negative islet cell and insulin antibodies, same as control. Treatment-related histopathological changes included sinusoidal degeneration in the liver, glomerular degeneration in the kidney, and degeneration of pancreatic islets. Our findings document toxic outcomes of MEHP and MBP on endocrine organs in male pubertal rats but also suggest the need for additional studies to better understand the mechanisms behind adverse effects in chronic exposure.
Collapse
Affiliation(s)
- Gözde Karabulut
- Dumlupınar University Faculty of Science, Department of Biology, Kütahya, Turkey
| | - Nurhayat Barlas
- Hacettepe University Faculty of Science, Department of Biology, Ankara, Turkey
| |
Collapse
|
8
|
Lipke K, Kubis-Kubiak A, Piwowar A. Molecular Mechanism of Lipotoxicity as an Interesting Aspect in the Development of Pathological States-Current View of Knowledge. Cells 2022; 11:cells11050844. [PMID: 35269467 PMCID: PMC8909283 DOI: 10.3390/cells11050844] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 02/06/2023] Open
Abstract
Free fatty acids (FFAs) play numerous vital roles in the organism, such as contribution to energy generation and reserve, serving as an essential component of the cell membrane, or as ligands for nuclear receptors. However, the disturbance in fatty acid homeostasis, such as inefficient metabolism or intensified release from the site of storage, may result in increased serum FFA levels and eventually result in ectopic fat deposition, which is unfavorable for the organism. The cells are adjusted for the accumulation of FFA to a limited extent and so prolonged exposure to elevated FFA levels results in deleterious effects referred to as lipotoxicity. Lipotoxicity contributes to the development of diseases such as insulin resistance, diabetes, cardiovascular diseases, metabolic syndrome, and inflammation. The nonobvious organs recognized as the main lipotoxic goal of action are the pancreas, liver, skeletal muscles, cardiac muscle, and kidneys. However, lipotoxic effects to a significant extent are not organ-specific but affect fundamental cellular processes occurring in most cells. Therefore, the wider perception of cellular lipotoxic mechanisms and their interrelation may be beneficial for a better understanding of various diseases’ pathogenesis and seeking new pharmacological treatment approaches.
Collapse
|
9
|
Wu ZS, Huang SM, Wang YC. Palmitate Enhances the Efficacy of Cisplatin and Doxorubicin against Human Endometrial Carcinoma Cells. Int J Mol Sci 2021; 23:ijms23010080. [PMID: 35008502 PMCID: PMC8744704 DOI: 10.3390/ijms23010080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 12/13/2022] Open
Abstract
Endometrial cancer is the most common gynecological cancer worldwide. At present there is no effective screening test for its early detection and no curative treatment for women with advanced-stage or recurrent disease. Overexpression of fatty acid synthase is a common molecular feature of a subgroup of sex steroid-related cancers associated with poor prognoses, including endometrial cancers. Disruption of this fatty acid synthesis leads to cell apoptosis, making it a potential therapeutic target. The saturated fatty acid palmitate reportedly induces lipotoxicity and cell death by inducing oxidative stress in many cell types. Here, we explored the effects of palmitate combined with doxorubicin or cisplatin in the HEC-1-A and RL95-2 human endometrial cancer cell lines. The results showed that physiological concentrations of exogenous palmitate significantly increased cell cycle arrest, DNA damage, autophagy, and apoptosis in both RL95-2 and HEC-1-A cells. It also increased the chemosensitivity of both cell types. Notably, we did not observe that palmitate lipotoxicity reflected increased levels of reactive oxygen species, suggesting palmitate acts via a different mechanism in endometrial cancer. This study thus provides a potential therapeutic strategy in which palmitate is used as an adjuvant in the treatment of endometrial cancer.
Collapse
Affiliation(s)
- Zih-Syuan Wu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei City 114, Taiwan; (Z.-S.W.); (S.-M.H.)
| | - Shih-Ming Huang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei City 114, Taiwan; (Z.-S.W.); (S.-M.H.)
- Department of Biochemistry, National Defense Medical Center, Taipei City 114, Taiwan
| | - Yu-Chi Wang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei City 114, Taiwan; (Z.-S.W.); (S.-M.H.)
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei City 114, Taiwan
- Correspondence:
| |
Collapse
|
10
|
Compared study of fucoidan from sea cucumber (Holothuria tubulosa) with different molecular weight on ameliorating β cell apoptosis. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
11
|
The association between dairy products consumption with risk of type 1 diabetes mellitus in children: a meta-analysis of observational studies. Int J Diabetes Dev Ctries 2021. [DOI: 10.1007/s13410-021-00923-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
12
|
Ahmed B, Sultana R, Greene MW. Adipose tissue and insulin resistance in obese. Biomed Pharmacother 2021; 137:111315. [PMID: 33561645 DOI: 10.1016/j.biopha.2021.111315] [Citation(s) in RCA: 413] [Impact Index Per Article: 103.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 02/08/2023] Open
Abstract
Currently, obesity has become a global health issue and is referred to as an epidemic. Dysfunctional obese adipose tissue plays a pivotal role in the development of insulin resistance. However, the mechanism of how dysfunctional obese-adipose tissue develops insulin-resistant circumstances remains poorly understood. Therefore, this review attempts to highlight the potential mechanisms behind obesity-associated insulin resistance. Multiple risk factors are directly or indirectly associated with the increased risk of obesity; among them, environmental factors, genetics, aging, gut microbiota, and diets are prominent. Once an individual becomes obese, adipocytes increase in their size; therefore, adipose tissues become larger and dysfunctional, recruit macrophages, and then these polarize to pro-inflammatory states. Enlarged adipose tissues release excess free fatty acids (FFAs), reactive oxygen species (ROS), and pro-inflammatory cytokines. Excess systemic FFAs and dietary lipids enter inside the cells of non-adipose organs such as the liver, muscle, and pancreas, and are deposited as ectopic fat, generating lipotoxicity. Toxic lipids dysregulate cellular organelles, e.g., mitochondria, endoplasmic reticulum, and lysosomes. Dysregulated organelles release excess ROS and pro-inflammation, resulting in systemic inflammation. Long term low-grade systemic inflammation prevents insulin from its action in the insulin signaling pathway, disrupts glucose homeostasis, and results in systemic dysregulation. Overall, long-term obesity and overnutrition develop into insulin resistance and chronic low-grade systemic inflammation through lipotoxicity, creating the circumstances to develop clinical conditions. This review also shows that the liver is the most sensitive organ undergoing insulin impairment faster than other organs, and thus, hepatic insulin resistance is the primary event that leads to the subsequent development of peripheral tissue insulin resistance.
Collapse
Affiliation(s)
- Bulbul Ahmed
- Department of Nutrition, Auburn University, Auburn, AL, 36849, United States.
| | - Rifat Sultana
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, 57007, United States
| | - Michael W Greene
- Department of Nutrition, Auburn University, Auburn, AL, 36849, United States
| |
Collapse
|
13
|
Zoni V, Khaddaj R, Campomanes P, Thiam AR, Schneiter R, Vanni S. Pre-existing bilayer stresses modulate triglyceride accumulation in the ER versus lipid droplets. eLife 2021; 10:e62886. [PMID: 33522484 PMCID: PMC7895522 DOI: 10.7554/elife.62886] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 01/31/2021] [Indexed: 12/13/2022] Open
Abstract
Cells store energy in the form of neutral lipids (NLs) packaged into micrometer-sized organelles named lipid droplets (LDs). These structures emerge from the endoplasmic reticulum (ER) at sites marked by the protein seipin, but the mechanisms regulating their biogenesis remain poorly understood. Using a combination of molecular simulations, yeast genetics, and fluorescence microscopy, we show that interactions between lipids' acyl-chains modulate the propensity of NLs to be stored in LDs, in turn preventing or promoting their accumulation in the ER membrane. Our data suggest that diacylglycerol, which is enriched at sites of LD formation, promotes the packaging of NLs into LDs, together with ER-abundant lipids, such as phosphatidylethanolamine. On the opposite end, short and saturated acyl-chains antagonize fat storage in LDs and promote accumulation of NLs in the ER. Our results provide a new conceptual understanding of LD biogenesis in the context of ER homeostasis and function.
Collapse
Affiliation(s)
- Valeria Zoni
- University of Fribourg, Department of BiologyFribourgSwitzerland
| | - Rasha Khaddaj
- University of Fribourg, Department of BiologyFribourgSwitzerland
| | - Pablo Campomanes
- University of Fribourg, Department of BiologyFribourgSwitzerland
| | - Abdou Rachid Thiam
- Laboratoire de Physique de l’École Normale Supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université de ParisParisFrance
| | - Roger Schneiter
- University of Fribourg, Department of BiologyFribourgSwitzerland
| | - Stefano Vanni
- University of Fribourg, Department of BiologyFribourgSwitzerland
| |
Collapse
|
14
|
Lytrivi M, Ghaddar K, Lopes M, Rosengren V, Piron A, Yi X, Johansson H, Lehtiö J, Igoillo-Esteve M, Cunha DA, Marselli L, Marchetti P, Ortsäter H, Eizirik DL, Cnop M. Combined transcriptome and proteome profiling of the pancreatic β-cell response to palmitate unveils key pathways of β-cell lipotoxicity. BMC Genomics 2020; 21:590. [PMID: 32847508 PMCID: PMC7448506 DOI: 10.1186/s12864-020-07003-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 08/19/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Prolonged exposure to elevated free fatty acids induces β-cell failure (lipotoxicity) and contributes to the pathogenesis of type 2 diabetes. In vitro exposure of β-cells to the saturated free fatty acid palmitate is a valuable model of lipotoxicity, reproducing features of β-cell failure observed in type 2 diabetes. In order to map the β-cell response to lipotoxicity, we combined RNA-sequencing of palmitate-treated human islets with iTRAQ proteomics of insulin-secreting INS-1E cells following a time course exposure to palmitate. RESULTS Crossing transcriptome and proteome of palmitate-treated β-cells revealed 85 upregulated and 122 downregulated genes at both transcript and protein level. Pathway analysis identified lipid metabolism, oxidative stress, amino-acid metabolism and cell cycle pathways among the most enriched palmitate-modified pathways. Palmitate induced gene expression changes compatible with increased free fatty acid mitochondrial import and β-oxidation, decreased lipogenesis and modified cholesterol transport. Palmitate modified genes regulating endoplasmic reticulum (ER) function, ER-to-Golgi transport and ER stress pathways. Furthermore, palmitate modulated cAMP/protein kinase A (PKA) signaling, inhibiting expression of PKA anchoring proteins and downregulating the GLP-1 receptor. SLC7 family amino-acid transporters were upregulated in response to palmitate but this induction did not contribute to β-cell demise. To unravel critical mediators of lipotoxicity upstream of the palmitate-modified genes, we identified overrepresented transcription factor binding sites and performed network inference analysis. These identified LXR, PPARα, FOXO1 and BACH1 as key transcription factors orchestrating the metabolic and oxidative stress responses to palmitate. CONCLUSIONS This is the first study to combine transcriptomic and sensitive time course proteomic profiling of palmitate-exposed β-cells. Our results provide comprehensive insight into gene and protein expression changes, corroborating and expanding beyond previous findings. The identification of critical drivers and pathways of the β-cell lipotoxic response points to novel therapeutic targets for type 2 diabetes.
Collapse
Affiliation(s)
- Maria Lytrivi
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium.,Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Kassem Ghaddar
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Miguel Lopes
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Victoria Rosengren
- Diabetes Research Unit, Department of Clinical Science and Education, Sodersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Anthony Piron
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Xiaoyan Yi
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Henrik Johansson
- Clinical Proteomics Mass Spectrometry, Department of Oncology-Pathology, Karolinska Institutet, Science for Life Laboratory, 171 21, Solna, Sweden
| | - Janne Lehtiö
- Clinical Proteomics Mass Spectrometry, Department of Oncology-Pathology, Karolinska Institutet, Science for Life Laboratory, 171 21, Solna, Sweden
| | - Mariana Igoillo-Esteve
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Daniel A Cunha
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Henrik Ortsäter
- Diabetes Research Unit, Department of Clinical Science and Education, Sodersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Miriam Cnop
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium. .,Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
15
|
Tripathi AK, Rai D, Kothari P, Kushwaha P, Sinha S, Sardar A, Sashidhara KV, Trivedi R. Benzofuran pyran compound rescues rat and human osteoblast from lipotoxic effect of palmitate by inhibiting lipid biosynthesis and promoting stabilization of RUNX2. Toxicol In Vitro 2020; 66:104872. [DOI: 10.1016/j.tiv.2020.104872] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 04/06/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023]
|
16
|
Cruz ALS, Barreto EDA, Fazolini NPB, Viola JPB, Bozza PT. Lipid droplets: platforms with multiple functions in cancer hallmarks. Cell Death Dis 2020; 11:105. [PMID: 32029741 PMCID: PMC7005265 DOI: 10.1038/s41419-020-2297-3] [Citation(s) in RCA: 308] [Impact Index Per Article: 61.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 01/16/2020] [Accepted: 01/20/2020] [Indexed: 02/06/2023]
Abstract
Lipid droplets (also known as lipid bodies) are lipid-rich, cytoplasmic organelles that play important roles in cell signaling, lipid metabolism, membrane trafficking, and the production of inflammatory mediators. Lipid droplet biogenesis is a regulated process, and accumulation of these organelles within leukocytes, epithelial cells, hepatocytes, and other nonadipocyte cells is a frequently observed phenotype in several physiologic or pathogenic situations and is thoroughly described during inflammatory conditions. Moreover, in recent years, several studies have described an increase in intracellular lipid accumulation in different neoplastic processes, although it is not clear whether lipid droplet accumulation is directly involved in the establishment of these different types of malignancies. This review discusses current evidence related to the biogenesis, composition and functions of lipid droplets related to the hallmarks of cancer: inflammation, cell metabolism, increased proliferation, escape from cell death, and hypoxia. Moreover, the potential of lipid droplets as markers of disease and targets for novel anti-inflammatory and antineoplastic therapies will be discussed.
Collapse
Affiliation(s)
- André L S Cruz
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Laboratory of Physiopathology, Polo Novo Cavaleiros, Federal University of Rio De Janeiro (UFRJ), Macaé, Brazil
| | - Ester de A Barreto
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Narayana P B Fazolini
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - João P B Viola
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil.
| | - Patricia T Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil.
| |
Collapse
|
17
|
Zhu Y, Zhang X, Zhang L, Zhang M, Li L, Luo D, Zhong Y. Perilipin5 protects against lipotoxicity and alleviates endoplasmic reticulum stress in pancreatic β-cells. Nutr Metab (Lond) 2019; 16:50. [PMID: 31384284 PMCID: PMC6668071 DOI: 10.1186/s12986-019-0375-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 07/19/2019] [Indexed: 12/27/2022] Open
Abstract
Background Chronic exposure of pancreatic β-cells to excess free fatty acids is thought to contribute to type 2 diabetes pathogenesis in obesity by impairing β-cell function and even leading to apoptosis. In β-cells, lipid droplet-associated protein perilipin 5 (PLIN5) has been shown to enhance insulin secretion by regulating intracellular lipid metabolism; the roles of PLIN5 in response to lipotoxicity remain poorly understood. Methods INS-1 β-cells were transfected with PLIN5-overexpression adenovirus (Ad-PLIN5) and treated with palmitate. C57BL/6 J male mice were fed with high fat diet and tail intravenous injected with adeno-associated virus overexpressing PLIN5 (AAV-PLIN5) in β-cells. Results Our data showed that palmitate and PPAR agonists including WY14643 (PPARα), GW501516 (PPARβ/δ), rosiglitazone (PPARγ) in vitro all induced PLIN5 expression in INS-1 cells. Under palmitate overload, although upregulating PLIN5 promoted lipid droplet storage, it alleviated lipotoxicity in INS-1 β-cells with improved cell viability, cell apoptosis and β-cell function. The protection role of PLIN5 in β-cell function observed in cell experiments were further verified in in vivo study indicated by mitigated glucose intolerance in high fat diet fed mice with β-cell-specific overexpression of PLIN5. Mechanistic experiments revealed that enhanced FAO induced by elevation of PLIN5, followed by decreased ER stress may be a major mechanism responsible for alleviation of lipotoxicity observed in the present study. Conclusions Our finding substantiated the important role of PLIN5 in protection against lipotoxicity in β-cells. Electronic supplementary material The online version of this article (10.1186/s12986-019-0375-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yunxia Zhu
- 1Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600, Yishan Road, Shanghai, 200233 China
| | - Xiaoyan Zhang
- 1Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600, Yishan Road, Shanghai, 200233 China
| | - Li Zhang
- 1Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600, Yishan Road, Shanghai, 200233 China
| | - Mingliang Zhang
- 2Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233 China
| | - Ling Li
- 3Department of Endocrinology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065 China
| | - Deng Luo
- 4Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| | - Yuan Zhong
- 1Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600, Yishan Road, Shanghai, 200233 China
| |
Collapse
|
18
|
Kim K, Bae GD, Lee M, Park EY, Baek DJ, Kim CY, Jun HS, Oh YS. Allomyrina dichotoma Larva Extract Ameliorates the Hepatic Insulin Resistance of High-Fat Diet-Induced Diabetic Mice. Nutrients 2019; 11:nu11071522. [PMID: 31277481 PMCID: PMC6683090 DOI: 10.3390/nu11071522] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 12/20/2022] Open
Abstract
Allomyrina dichotoma larva is a nutritional-worthy future food resource and it contributes to multiple pharmacological functions. However, its antidiabetic effect and molecular mechanisms are not yet fully understood. Therefore, we investigated the hypolipidemic effect of A. dichotoma larva extract (ADLE) in a high-fat diet (HFD)-induced C57BL/6J mice model. Glucose tolerance and insulin sensitivity in HFD-induced diabetic mice significantly improved after ADLE administration for six weeks. The levels of serum triglyceride (TG), aspartate aminotransferase (AST), alanine transferase (ALT) activity, and lipid accumulation were increased in the liver of HFD-fed mice, but the levels were significantly reduced by the ADLE treatment. Moreover, hepatic fibrosis and inflammatory gene expression in the liver from HFD-treated mice were ameliorated by the ADLE treatment. Dephosphorylation of AMP-activated protein kinase (AMPK) by palmitate was inhibited in the ADLE treated HepG2 cells, and subsequently reduced expression of lipogenic genes, such as SREPBP-1c, ACC, and FAS were observed. The reduced expression of lipogenic genes and an increased phosphorylation of AMPK was also observed in the liver from diabetic mice treated with ADLE. In conclusion, ADLE ameliorates hyperlipidemia through inhibition of hepatic lipogenesis via activating the AMPK signaling pathway. These findings suggest that ADLE and its constituent bioactive compounds are valuable to prevent or treat hepatic insulin resistance in type 2 diabetes.
Collapse
Affiliation(s)
- Kyong Kim
- Department of Food and Nutrition, Eulji University, Seongnam 13135, Korea
| | - Gong Deuk Bae
- Lee Gil Ya Cancer and Diabetes Institute, Department of molecular medicine, Gachon University, Incheon 21999, Korea
| | - Minho Lee
- Department of Food Technology and Services, Eulji University, Seongnam 13135, Korea
| | - Eun-Young Park
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Korea
| | - Dong Jae Baek
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Korea
| | - Chul Young Kim
- College of Pharmacy, Hanyang University, Ansan 15888, Korea
| | - Hee-Sook Jun
- Lee Gil Ya Cancer and Diabetes Institute, Department of molecular medicine, Gachon University, Incheon 21999, Korea
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon 21936, Korea
| | - Yoon Sin Oh
- Department of Food and Nutrition, Eulji University, Seongnam 13135, Korea.
| |
Collapse
|
19
|
Bellini L, Campana M, Rouch C, Chacinska M, Bugliani M, Meneyrol K, Hainault I, Lenoir V, Denom J, Véret J, Kassis N, Thorens B, Ibberson M, Marchetti P, Blachnio-Zabielska A, Cruciani-Guglielmacci C, Prip-Buus C, Magnan C, Le Stunff H. Protective role of the ELOVL2/docosahexaenoic acid axis in glucolipotoxicity-induced apoptosis in rodent beta cells and human islets. Diabetologia 2018; 61:1780-1793. [PMID: 29754287 DOI: 10.1007/s00125-018-4629-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 03/08/2018] [Indexed: 12/19/2022]
Abstract
AIMS/HYPOTHESIS Dietary n-3 polyunsaturated fatty acids, especially docosahexaenoic acid (DHA), are known to influence glucose homeostasis. We recently showed that Elovl2 expression in beta cells, which regulates synthesis of endogenous DHA, was associated with glucose tolerance and played a key role in insulin secretion. The present study aimed to examine the role of the very long chain fatty acid elongase 2 (ELOVL2)/DHA axis on the adverse effects of palmitate with high glucose, a condition defined as glucolipotoxicity, on beta cells. METHODS We detected ELOVL2 in INS-1 beta cells and mouse and human islets using quantitative PCR and western blotting. Downregulation and adenoviral overexpression of Elovl2 was carried out in beta cells. Ceramide and diacylglycerol levels were determined by radio-enzymatic assay and lipidomics. Apoptosis was quantified using caspase-3 assays and poly (ADP-ribose) polymerase cleavage. Palmitate oxidation and esterification were determined by [U-14C]palmitate labelling. RESULTS We found that glucolipotoxicity decreased ELOVL2 content in rodent and human beta cells. Downregulation of ELOVL2 drastically potentiated beta cell apoptosis induced by glucolipotoxicity, whereas adenoviral Elovl2 overexpression and supplementation with DHA partially inhibited glucolipotoxicity-induced cell death in rodent and human beta cells. Inhibition of beta cell apoptosis by the ELOVL2/DHA axis was associated with a decrease in ceramide accumulation. However, the ELOVL2/DHA axis was unable to directly alter ceramide synthesis or metabolism. By contrast, DHA increased palmitate oxidation but did not affect its esterification. Pharmacological inhibition of AMP-activated protein kinase and etomoxir, an inhibitor of carnitine palmitoyltransferase 1 (CPT1), the rate-limiting enzyme in fatty acid β-oxidation, attenuated the protective effect of the ELOVL2/DHA axis during glucolipotoxicity. Downregulation of CPT1 also counteracted the anti-apoptotic action of the ELOVL2/DHA axis. By contrast, a mutated active form of Cpt1 inhibited glucolipotoxicity-induced beta cell apoptosis when ELOVL2 was downregulated. CONCLUSIONS/INTERPRETATION Our results identify ELOVL2 as a critical pro-survival enzyme for preventing beta cell death and dysfunction induced by glucolipotoxicity, notably by favouring palmitate oxidation in mitochondria through a CPT1-dependent mechanism.
Collapse
Affiliation(s)
- Lara Bellini
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Équipe Régulation de la glycémie par le système nerveux central, Université Paris Diderot, 4 rue Marie-Andrée-Lagroua-Weill-Hallé, 75205, Paris CEDEX 13, France
| | - Mélanie Campana
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Équipe Régulation de la glycémie par le système nerveux central, Université Paris Diderot, 4 rue Marie-Andrée-Lagroua-Weill-Hallé, 75205, Paris CEDEX 13, France
| | - Claude Rouch
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Équipe Régulation de la glycémie par le système nerveux central, Université Paris Diderot, 4 rue Marie-Andrée-Lagroua-Weill-Hallé, 75205, Paris CEDEX 13, France
| | - Marta Chacinska
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Bialystok, Poland
| | - Marco Bugliani
- Department of Clinical and Experimental Medicine, Islet Laboratory, University of Pisa, Pisa, Italy
| | - Kelly Meneyrol
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Équipe Régulation de la glycémie par le système nerveux central, Université Paris Diderot, 4 rue Marie-Andrée-Lagroua-Weill-Hallé, 75205, Paris CEDEX 13, France
| | | | - Véronique Lenoir
- Inserm U1016, Institut Cochin, Paris, France
- CNRS UMR 8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jessica Denom
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Équipe Régulation de la glycémie par le système nerveux central, Université Paris Diderot, 4 rue Marie-Andrée-Lagroua-Weill-Hallé, 75205, Paris CEDEX 13, France
| | - Julien Véret
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Équipe Régulation de la glycémie par le système nerveux central, Université Paris Diderot, 4 rue Marie-Andrée-Lagroua-Weill-Hallé, 75205, Paris CEDEX 13, France
| | - Nadim Kassis
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Équipe Régulation de la glycémie par le système nerveux central, Université Paris Diderot, 4 rue Marie-Andrée-Lagroua-Weill-Hallé, 75205, Paris CEDEX 13, France
| | - Bernard Thorens
- Centre for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Mark Ibberson
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, Islet Laboratory, University of Pisa, Pisa, Italy
| | - Agnieszka Blachnio-Zabielska
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Bialystok, Poland
| | - Céline Cruciani-Guglielmacci
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Équipe Régulation de la glycémie par le système nerveux central, Université Paris Diderot, 4 rue Marie-Andrée-Lagroua-Weill-Hallé, 75205, Paris CEDEX 13, France
| | - Carina Prip-Buus
- Inserm U1016, Institut Cochin, Paris, France
- CNRS UMR 8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Christophe Magnan
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Équipe Régulation de la glycémie par le système nerveux central, Université Paris Diderot, 4 rue Marie-Andrée-Lagroua-Weill-Hallé, 75205, Paris CEDEX 13, France
| | - Hervé Le Stunff
- Unité Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Équipe Régulation de la glycémie par le système nerveux central, Université Paris Diderot, 4 rue Marie-Andrée-Lagroua-Weill-Hallé, 75205, Paris CEDEX 13, France.
- Université Paris-Sud, Paris-Saclay Institute of Neuroscience, CNRS UMR 9197, Orsay, France.
| |
Collapse
|
20
|
MicroRNA-132 Negatively Regulates Palmitate-Induced NLRP3 Inflammasome Activation through FOXO3 Down-Regulation in THP-1 Cells. Nutrients 2017; 9:nu9121370. [PMID: 29258239 PMCID: PMC5748820 DOI: 10.3390/nu9121370] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 12/10/2017] [Accepted: 12/14/2017] [Indexed: 01/22/2023] Open
Abstract
Saturated fatty acids were proposed to activate the NLRP3 inflammasome, a molecular platform that mediates the processing of interleukin (IL)-1β and IL-18. However, the mechanisms underlying the miRNA-mediated regulation of palmitate (PA)-induced inflammasome activation are unclear. We examined the role of miR-132 in PA-induced NLRP3 inflammasome activation in THP-1 cells. To understand the regulatory role of miR-132 in inflammasome activation, we either overexpressed or suppressed miR-132 in THP-1 cells that expressed the NLRP3 inflammasome in response to stimulation by PA. We analyzed the mRNA and protein levels of NLRP3, caspase-1 p10, IL-18, and IL-1β; caspase-1 activity; and IL-1β secretion. The presence of PA activated the NLRP3 inflammasome and increased miR-132 expression. Overexpression of miR-132 reduced caspase-1 p10, IL-18, and IL-1β, while the suppression of miR-132 enhanced inflammasome activation. In addition, miR-132 regulated the mRNA and protein expression of FOXO3, which is a potential target of miR-132 in these cells. FOXO3 suppression by small interfering RNA decreased NLRP3 inflammasome activity stimulated by PA. Knockdown of FOXO3 attenuated NLRP3 inflammasome activation by the miR-132 inhibitor. Based on these findings, we conclude that miR-132 negatively regulates PA-induced NLRP3 inflammasome activation through FOXO3 down-regulation in THP-1 cells.
Collapse
|
21
|
Abstract
Cellular lipid metabolism and homeostasis are controlled by sterol regulatory-element binding proteins (SREBPs). In addition to performing canonical functions in the transcriptional regulation of genes involved in the biosynthesis and uptake of lipids, genome-wide system analyses have revealed that these versatile transcription factors act as important nodes of convergence and divergence within biological signalling networks. Thus, they are involved in myriad physiological and pathophysiological processes, highlighting the importance of lipid metabolism in biology. Changes in cell metabolism and growth are reciprocally linked through SREBPs. Anabolic and growth signalling pathways branch off and connect to multiple steps of SREBP activation and form complex regulatory networks. In addition, SREBPs are implicated in numerous pathogenic processes such as endoplasmic reticulum stress, inflammation, autophagy and apoptosis, and in this way, they contribute to obesity, dyslipidaemia, diabetes mellitus, nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, chronic kidney disease, neurodegenerative diseases and cancers. This Review aims to provide a comprehensive understanding of the role of SREBPs in physiology and pathophysiology at the cell, organ and organism levels.
Collapse
Affiliation(s)
- Hitoshi Shimano
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Life Science Center, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Ryuichiro Sato
- AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo 100-0004, Japan
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo 113-8657, Japan
| |
Collapse
|
22
|
Frago LM, Canelles S, Freire-Regatillo A, Argente-Arizón P, Barrios V, Argente J, Garcia-Segura LM, Chowen JA. Estradiol Uses Different Mechanisms in Astrocytes from the Hippocampus of Male and Female Rats to Protect against Damage Induced by Palmitic Acid. Front Mol Neurosci 2017; 10:330. [PMID: 29114202 PMCID: PMC5660686 DOI: 10.3389/fnmol.2017.00330] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/29/2017] [Indexed: 01/22/2023] Open
Abstract
An excess of saturated fatty acids can be toxic for tissues, including the brain, and this has been associated with the progression of neurodegenerative diseases. Since palmitic acid (PA) is a free fatty acid that is abundant in the diet and circulation and can be harmful, we have investigated the effects of this fatty acid on lipotoxicity in hippocampal astrocytes and the mechanism involved. Moreover, as males and females have different susceptibilities to some neurodegenerative diseases, we accessed the responses of astrocytes from both sexes, as well as the possible involvement of estrogens in the protection against fatty acid toxicity. PA increased endoplasmic reticulum stress leading to cell death in astrocytes from both males and females. Estradiol (E2) increased the levels of protective factors, such as Hsp70 and the anti-inflammatory cytokine interleukin-10, in astrocytes from both sexes. In male astrocytes, E2 decreased pJNK, TNFα, and caspase-3 activation. In contrast, in female astrocytes E2 did not affect the activation of JNK or TNFα levels, but decreased apoptotic cell death. Hence, although E2 exerted protective effects against the detrimental effects of PA, the mechanisms involved appear to be different between male and female astrocytes. This sexually dimorphic difference in the protective mechanisms induced by E2 could be involved in the different susceptibilities of males and females to some neurodegenerative processes.
Collapse
Affiliation(s)
- Laura M Frago
- Departamento de Pediatría, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Canelles
- Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Alejandra Freire-Regatillo
- Departamento de Pediatría, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Pilar Argente-Arizón
- Departamento de Pediatría, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Vicente Barrios
- Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Argente
- Departamento de Pediatría, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain.,IMDEA Food Institute, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Luis M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,CIBER de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Julie A Chowen
- Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
23
|
Sodium fluorocitrate having protective effect on palmitate-induced beta cell death improves hyperglycemia in diabetic db/db mice. Sci Rep 2017; 7:12916. [PMID: 29018279 PMCID: PMC5635019 DOI: 10.1038/s41598-017-13365-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 09/21/2017] [Indexed: 02/06/2023] Open
Abstract
Beta cell loss and insulin resistance play roles in the pathogenesis of type 2 diabetes. Elevated levels of free fatty acids in plasma might contribute to the loss of beta cells. The objective of this study was to find a chemical that could protect against palmitate-induced beta cell death and investigate whether such chemical could improve hyperglycemia in mouse model of type 2 diabetes. Sodium fluorocitrate (SFC), an aconitase inhibitor, was found to be strongly and specifically protective against palmitate-induced INS-1 beta cell death. However, the protective effect of SFC on palmitate-induced cell death was not likely to be due to its inhibitory activity for aconitase since inhibition or knockdown of aconitase failed to protect against palmitate-induced cell death. Since SFC inhibited the uptake of palmitate into INS-1 cells, reduced metabolism of fatty acids was thought to be involved in SFC’s protective effect. Ten weeks of treatment with SFC in db/db diabetic mice reduced glucose level but remarkably increased insulin level in the plasma. SFC improved impairment of glucose-stimulated insulin release and also reduced the loss of beta cells in db/db mice. Conclusively, SFC possessed protective effect against palmitate-induced lipotoxicity and improved hyperglycemia in mouse model of type 2 diabetes.
Collapse
|
24
|
Oxidative stress and calcium dysregulation by palmitate in type 2 diabetes. Exp Mol Med 2017; 49:e291. [PMID: 28154371 PMCID: PMC5336562 DOI: 10.1038/emm.2016.157] [Citation(s) in RCA: 227] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 10/07/2016] [Accepted: 10/16/2016] [Indexed: 12/12/2022] Open
Abstract
Free fatty acids (FFAs) are important substrates for mitochondrial oxidative metabolism and ATP synthesis but also cause serious stress to various tissues, contributing to the development of metabolic diseases. CD36 is a major mediator of cellular FFA uptake. Inside the cell, saturated FFAs are able to induce the production of cytosolic and mitochondrial reactive oxygen species (ROS), which can be prevented by co-exposure to unsaturated FFAs. There are close connections between oxidative stress and organellar Ca2+ homeostasis. Highly oxidative conditions induced by palmitate trigger aberrant endoplasmic reticulum (ER) Ca2+ release and thereby deplete ER Ca2+ stores. The resulting ER Ca2+ deficiency impairs chaperones of the protein folding machinery, leading to the accumulation of misfolded proteins. This ER stress may further aggravate oxidative stress by augmenting ER ROS production. Secondary to ER Ca2+ release, cytosolic and mitochondrial matrix Ca2+ concentrations can also be altered. In addition, plasmalemmal ion channels operated by ER Ca2+ depletion mediate persistent Ca2+ influx, further impairing cytosolic and mitochondrial Ca2+ homeostasis. Mitochondrial Ca2+ overload causes superoxide production and functional impairment, culminating in apoptosis. This vicious cycle of lipotoxicity occurs in multiple tissues, resulting in β-cell failure and insulin resistance in target tissues, and further aggravates diabetic complications.
Collapse
|
25
|
Knockdown of triglyceride synthesis does not enhance palmitate lipotoxicity or prevent oleate-mediated rescue in rat hepatocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1005-1014. [PMID: 27249207 DOI: 10.1016/j.bbalip.2016.05.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 05/14/2016] [Accepted: 05/26/2016] [Indexed: 12/14/2022]
Abstract
Experiments in a variety of cell types, including hepatocytes, consistently demonstrate the acutely lipotoxic effects of saturated fatty acids, such as palmitate (PA), but not unsaturated fatty acids, such as oleate (OA). PA+OA co-treatment fully prevents PA lipotoxicity through mechanisms that are not well defined but which have been previously attributed to more efficient esterification and sequestration of PA into triglycerides (TGs) when OA is abundant. However, this hypothesis has never been directly tested by experimentally modulating the relative partitioning of PA/OA between TGs and other lipid fates in hepatocytes. In this study, we found that addition of OA to PA-treated hepatocytes enhanced TG synthesis, reduced total PA uptake and PA lipid incorporation, decreased phospholipid saturation and rescued PA-induced ER stress and lipoapoptosis. Knockdown of diacylglycerol acyltransferase (DGAT), the rate-limiting step in TG synthesis, significantly reduced TG accumulation without impairing OA-mediated rescue of PA lipotoxicity. In both wild-type and DGAT-knockdown hepatocytes, OA co-treatment significantly reduced PA lipid incorporation and overall phospholipid saturation compared to PA-treated hepatocytes. These data indicate that OA's protective effects do not require increased conversion of PA into inert TGs, but instead may be due to OA's ability to compete against PA for cellular uptake and/or esterification and, thereby, normalize the composition of cellular lipids in the presence of a toxic PA load.
Collapse
|
26
|
Ponziani FR, Pecere S, Gasbarrini A, Ojetti V. Physiology and pathophysiology of liver lipid metabolism. Expert Rev Gastroenterol Hepatol 2016; 9:1055-67. [PMID: 26070860 DOI: 10.1586/17474124.2015.1056156] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Liver lipid metabolism and its modulation are involved in many pathologic conditions, such as obesity, non-alcoholic fatty liver disease, diabetes mellitus, atherosclerosis and cardiovascular disease. Metabolic disorders seem to share a similar background of low-grade chronic inflammation, even if the pathophysiological mechanisms leading to tissue and organ damage have not been completely clarified yet. The accumulation of neutral lipids in the liver is now recognized as a beneficial and protective mechanism; on the other hand, lipoperoxidation is involved in the development and progression of non-alcoholic steatohepatitis. The role of the gut microbiota in liver lipid metabolism has been the object of recent scientific investigations. It is likely that the gut microbiota is involved in a complex metabolic modulation and the translocation of gut microflora may also contribute to maintaining the low-grade inflammatory status of metabolic syndrome. Therefore, lipid metabolism pathology has vague limits and complex mechanisms, and the knowledge of these is essential to guide diagnostic and therapeutic decisions.
Collapse
|
27
|
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: 164] [Impact Index Per Article: 16.4] [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
|
28
|
Jung IR, Choi SE, Jung JG, Lee SA, Han SJ, Kim HJ, Kim DJ, Lee KW, Kang Y. Involvement of iron depletion in palmitate-induced lipotoxicity of beta cells. Mol Cell Endocrinol 2015; 407:74-84. [PMID: 25779532 DOI: 10.1016/j.mce.2015.03.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 01/28/2015] [Accepted: 03/10/2015] [Indexed: 12/28/2022]
Abstract
High levels of plasma free fatty acid are thought to contribute to the loss of pancreatic beta-cells in type 2 diabetes. In particular, saturated fatty acid such as palmitate or stearate can induce apoptosis in cultured beta cells (lipotoxicity). Endoplasmic reticulum stress is a critical mediator of free fatty acid-induced lipotoxicity. Recently, disorders in mitochondrial respiratory metabolism have been linked to lipotoxicity. Since iron is a critical component of respiratory metabolism, this study is initiated to determine whether abnormal iron metabolism is involved in palmitate-induced beta cell death. Immunoblotting analysis showed that treatment of INS-1 beta cells with palmitate reduced the level of transferrin receptor 1 (TfR1), but increased the level of heavy chain ferritin (FTH). In addition, palmitate reduced intracellular labile iron pool. Whereas iron depletion through treatment with iron-chelators deferoxamine or deferasirox augmented palmitate-induced cell death, iron supplementation with ferric chloride, ferrous sulfate, or holo-transferrin significantly protected cells against palmitate-induced death. Furthermore, overexpression of TfR1 reduced palmitate-induced cell death, whereas knockdown of TfR1 augmented cell death. In particular, treatment with deferoxamine increased the level of endoplasmic reticulum (ER) stress markers phospho-PERK, phospho-eIF2α, CHOP and phospho-c-Jun N-terminal kinase. Treatment with chemical chaperone significantly protected cells against deferoxamine-induced apoptosis. Iron supplementation also protected cells against palmitate-induced primary islet death. These data suggest that iron depletion plays an important role in palmitate-induced beta cell death through inducing ER stress. Therefore, attempts to block iron depletion might be able to prevent beta cell loss in type 2 diabetes.
Collapse
Affiliation(s)
- Ik-Rak Jung
- Department of Physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea; Department of Biomedical Science, The Graduate School, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Sung-E Choi
- Department of Physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Jong-Gab Jung
- Department of Biomedical Science, The Graduate School, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea; Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Sang-A Lee
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Seung Jin Han
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Hae Jin Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Dae Jung Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Kwan-Woo Lee
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Yup Kang
- Department of Physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea; Department of Biomedical Science, The Graduate School, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea.
| |
Collapse
|
29
|
Wang D, Tian M, Qi Y, Chen G, Xu L, Zou X, Wang K, Dong H, Lu F. Jinlida granule inhibits palmitic acid induced-intracellular lipid accumulation and enhances autophagy in NIT-1 pancreatic β cells through AMPK activation. JOURNAL OF ETHNOPHARMACOLOGY 2015; 161:99-107. [PMID: 25510732 DOI: 10.1016/j.jep.2014.12.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/06/2014] [Accepted: 12/01/2014] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jinlida granule (JLDG), composed of seventeen Chinese medical herbs, is a widely used Chinese herbal prescription for treating diabetes mellitus. However, the mechanism underlying this effect remains unclear. To determine the main components in JLDG and to explore the effect of JLDG on autophagy and lipid accumulation in NIT-1 pancreatic β cells exposed to politic acid (PA) through AMP activated protein kinase (AMPK) signaling pathway. MATERIALS AND METHODS JLDG was prepared and the main components contained in the granules were identified by ultra performance liquid chromatography (UPLC) fingerprint. Intracellular lipid accumulation in NIT-1 cells was induced by culturing with medium containing PA. Intracellular lipid droplets were observed by Oil Red O staining and triglyceride (TG) content was measured by colorimetric assay. The formation of autophagosomes was observed under transmission electron microscope. The expression of AMPK and phospho-AMPK (pAMPK) proteins as well as its downstream fatty acid metabolism-related proteins (fatty acid synthase, FAS; acetyl-coA carboxylase, ACC; carnitine acyltransferase 1, CPT-1) and autophagy-related genes (mammal target of rapamycin, mTOR; tuberous sclerosis complex 1, TSC1; microtubule-associated protein 1 light chain 3, LC3-II) were determined by Western blot. The expression of sterol regulating element binding protein 1c (SREBP-1c) mRNA was examined by real time PCR (RT-PCR). RESULTS Our data showed that JLDG could significantly reduce PA-induced intracellular lipid accumulation in NIT-1 pancreatic β cells. This effect was associated with increased protein expression of pAMPK and AMPK in NIT-1 cells. Treatment with JLDG also decreased the expression of AMPK downstream lipogenic genes (SREBP-1c mRNA, FAS and ACC proteins) whereas increased the expression of fatty acid oxidation gene (CPT-1 protein). Additionally, JLDG-treated cells displayed a markedly increase in the number of autophagosomes which was accompanied by the down-regulation of mTOR and the up-regulation of TSC1 and LC3-II proteins expression. However, when AMPK phosphorylation was inhibited by Compound C, JLDG supplementation did not exhibit any effect on the expression of these AMPK downstream molecules in NIT-1 cells. CONCLUSIONS The results suggest that JLDG could reduce intracellular lipid accumulation and enhance the autophagy in NIT-1 pancreatic β cells cultured with PA. The mechanism is possibly mediated by AMPK activation.
Collapse
Affiliation(s)
- Dingkun Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Min Tian
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yuan Qi
- Shijiazhuang YiLing Pharmaceutical Co., Ltd., Shijiazhuang, Hebei 050035, China
| | - Guang Chen
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Lijun Xu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xin Zou
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Kaifu Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Hui Dong
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Fuer Lu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
30
|
Jiang P, Gan M, Lin WL, Yen SHC. Nutrient deprivation induces α-synuclein aggregation through endoplasmic reticulum stress response and SREBP2 pathway. Front Aging Neurosci 2014; 6:268. [PMID: 25339898 PMCID: PMC4189422 DOI: 10.3389/fnagi.2014.00268] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 09/16/2014] [Indexed: 12/25/2022] Open
Abstract
Abnormal accumulation of filamentous α-synuclein (α-syn) in neurons, regarded as Lewy bodies (LBs), are a hallmark of Parkinson disease (PD). Although the exact mechanism(s) underlying LBs formation remains unknown, autophagy and ER stress response have emerged as two important pathways affecting α-syn aggregation. In present study we tested whether cells with the tetracycline-off inducible overexpression of α-syn and accumulating α-syn aggregates can benefit from autophagy activation elicited by nutrient deprivation (ND), since this approach was reported to effectively clear cellular polyglutamine aggregates. We found that nutrient deprivation of non-induced cells did not affect cell viability, but significantly activated autophagy reflected by increasing the level of autophagy marker LC3-II and autophagic flux and decrease of endogenous α-syn. Cells with induced α-syn expression alone displayed autophagy activation in an α-syn dose-dependent manner to reach a level comparable to that found in non-induced, nutrient deprived counterparts. Nutrient deprivation also activated autophagy further in α-syn induced cells, but the extent was decreased with increase of α-syn dose, indicating α-syn overexpression reduces the responsiveness of cells to nutrient deprivation. Moreover, the nutrient deprivation enhanced α-syn aggregations concomitant with significant increase of apoptosis as well as ER stress response, SREBP2 activation and cholesterolgenesis. Importantly, α-syn aggregate accumulation and other effects caused by nutrient deprivation were counteracted by knockdown of SREBP2, treatment with cholesterol lowering agent-lovastatin, or by GRP78 overexpression, which also caused decrease of SREBP2 activity. Similar results were obtained from studies of primary neurons with α-syn overexpression under nutrient deprivation. Together our findings suggested that down-regulation of SREBP2 activity might be a means to prevent α-syn aggregation in PD via reducing cholesterol levels.
Collapse
Affiliation(s)
- Peizhou Jiang
- Department of Neuroscience, Mayo Clinic College of Medicine Jacksonville, FL, USA
| | - Ming Gan
- Department of Neuroscience, Mayo Clinic College of Medicine Jacksonville, FL, USA
| | - Wen-Lang Lin
- Department of Neuroscience, Mayo Clinic College of Medicine Jacksonville, FL, USA
| | - Shu-Hui C Yen
- Department of Neuroscience, Mayo Clinic College of Medicine Jacksonville, FL, USA
| |
Collapse
|
31
|
Abstract
Free fatty acids (FFAs) exert both positive and negative effects on beta cell survival and insulin secretory function, depending on concentration, duration, and glucose abundance. Lipid signals are mediated not only through metabolic pathways, but also through cell surface and nuclear receptors. Toxicity is modulated by positive signals arising from circulating factors such as hormones, growth factors and incretins, as well as negative signals such as inflammatory mediators and cytokines. Intracellular mechanisms of lipotoxicity include metabolic interference and cellular stress responses such as oxidative stress, endoplasmic reticulum (ER) stress, and possibly autophagy. New findings strengthen an old hypothesis that lipids may also impair compensatory beta cell proliferation. Clinical observations continue to support a role for lipid biology in the risk and progression of both type 1 (T1D) and type 2 diabetes (T2D). This review summarizes recent work in this important, rapidly evolving field.
Collapse
Affiliation(s)
- Rohit B Sharma
- Diabetes Center of Excellence, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA
| | | |
Collapse
|
32
|
Leamy AK, Egnatchik RA, Shiota M, Ivanova PT, Myers DS, Brown HA, Young JD. Enhanced synthesis of saturated phospholipids is associated with ER stress and lipotoxicity in palmitate treated hepatic cells. J Lipid Res 2014; 55:1478-88. [PMID: 24859739 DOI: 10.1194/jlr.m050237] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Indexed: 01/22/2023] Open
Abstract
High levels of saturated FAs (SFAs) are acutely toxic to a variety of cell types, including hepatocytes, and have been associated with diseases such as type 2 diabetes and nonalcoholic fatty liver disease. SFA accumulation has been previously shown to degrade endoplasmic reticulum (ER) function leading to other manifestations of the lipoapoptotic cascade. We hypothesized that dysfunctional phospholipid (PL) metabolism is an initiating factor in this ER stress response. Treatment of either primary hepatocytes or H4IIEC3 cells with the SFA palmitate resulted in dramatic dilation of the ER membrane, coinciding with other markers of organelle dysfunction. This was accompanied by increased de novo glycerolipid synthesis, significant elevation of dipalmitoyl phosphatidic acid, diacylglycerol, and total PL content in H4IIEC3 cells. Supplementation with oleate (OA) reversed these markers of palmitate (PA)-induced lipotoxicity. OA/PA cotreatment modulated the distribution of PA between lipid classes, increasing the flux toward triacylglycerols while reducing its incorporation into PLs. Similar trends were demonstrated in both primary hepatocytes and the H4IIEC3 hepatoma cell line. Overall, these findings suggest that modifying the FA composition of structural PLs can protect hepatocytes from PA-induced ER stress and associated lipotoxicity.
Collapse
Affiliation(s)
- Alexandra K Leamy
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235-1604
| | - Robert A Egnatchik
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235-1604
| | - Masakazu Shiota
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235-1604
| | - Pavlina T Ivanova
- Department of Pharmacology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37235-1604
| | - David S Myers
- Department of Pharmacology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37235-1604
| | - H Alex Brown
- Department of Pharmacology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37235-1604 Department of Biochemistry, Vanderbilt University, Nashville, TN 37235-1604 Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235-1604
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235-1604 Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235-1604
| |
Collapse
|
33
|
Lee JH, Jung IR, Choi SE, Lee SM, Lee SJ, Han SJ, Kim HJ, Kim DJ, Lee KW, Kang Y. Toxicity generated through inhibition of pyruvate carboxylase and carnitine palmitoyl transferase-1 is similar to high glucose/palmitate-induced glucolipotoxicity in INS-1 beta cells. Mol Cell Endocrinol 2014; 383:48-59. [PMID: 24333689 DOI: 10.1016/j.mce.2013.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Revised: 11/14/2013] [Accepted: 12/01/2013] [Indexed: 01/22/2023]
Abstract
This work was initiated to determine whether toxicity generated through inhibition of mitochondrial fuel metabolism is similar to high glucose/palmitate (HG/PA)-induced glucolipotoxicity. Influx of glucose and free fatty acids into the tricarboxylic acid (TCA) cycle was inhibited by treatment with the pyruvate carboxylase (PC) inhibitor phenylacetic acid (PAA) and carnitine palmitoyl transferase-1 (CPT-1) inhibitor etomoxir (Eto), or knockdown of PC and CPT-1. Treatment of PAA/Eto or knockdown of PC/CPT-1 induced apoptotic death in INS-1 beta cells. Similar to HG/PA treatment, PAA/Eto increased endoplasmic reticulum stress responses but decreased the Akt signal. JNK inhibitor or chemical chaperone was protective against both PAA/Eto- and HG/PA-induced cell death. All attempts to reduce [Ca²⁺](i), stimulate lipid metabolism, and increase the TCA cycle intermediate pool protected PAA/Eto-induced death as well as HG/PA-induced death. These data suggest that signals induced from impaired mitochondrial fuel metabolism play a critical role in HG/PA-induced glucolipotoxicity.
Collapse
Affiliation(s)
- Ji-Hyun Lee
- Department of physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea; Department of Life Science, Korea University Seoul 136-701, Republic of Korea
| | - Ik-Rak Jung
- Department of physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Sung-E Choi
- Department of physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Sung-Mi Lee
- Department of physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Soo-Jin Lee
- Department of physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Seung Jin Han
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Hae Jin Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Dae Jung Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Kwan-Woo Lee
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Yup Kang
- Department of physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea.
| |
Collapse
|
34
|
Saturated Fatty Acid-induced cytotoxicity in liver cells does not involve phosphatase and tensin homologue deleted on chromosome 10. J Nutr Metab 2013; 2013:514206. [PMID: 23691291 PMCID: PMC3649318 DOI: 10.1155/2013/514206] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 03/11/2013] [Accepted: 03/25/2013] [Indexed: 01/22/2023] Open
Abstract
Liver specific deletion of the tumor suppressor phosphatase and tensin homologue deleted on chromosome 10 (PTEN) induces steatosis and hypersensitivity to insulin. Saturated fatty acids, which induce endoplasmic reticulum stress and cell death, appear to increase PTEN, whereas unsaturated fatty acids which do not induce endoplasmic reticulum stress or cell death reduce this protein. In the present study, the role of PTEN in saturated fatty acid-induced cytotoxicity was examined in H4IIE and HepG2 liver cells. Palmitate and stearate increased the expression of PTEN, whereas the unsaturated fatty acids, oleate and linoleate, reduced PTEN expression in both cell types. SiRNA-mediated knockdown of PTEN did not increase liver cell triglyceride stores or reduce palmitate- or stearate-mediated ER stress or apoptosis. These results suggest that PTEN does not play a significant role in saturated fatty acid-induced cytotoxicity in these liver cell models and in the absence of insulin.
Collapse
|
35
|
Protective effect of nicotinamide on high glucose/palmitate-induced glucolipotoxicity to INS-1 beta cells is attributed to its inhibitory activity to sirtuins. Arch Biochem Biophys 2013; 535:187-96. [PMID: 23562377 DOI: 10.1016/j.abb.2013.03.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 03/14/2013] [Accepted: 03/17/2013] [Indexed: 11/20/2022]
Abstract
This study was initiated to determine whether the protective effect of nicotinamide (NAM) on high glucose/palmitate (HG/PA)-induced INS-1 beta cell death was due to its role as an anti-oxidant, nicotinamide dinucleotide (NAD+) precursor, or inhibitor of NAD+-consuming enzymes such as poly (ADP-ribose) polymerase (PARP) or sirtuins. All anti-oxidants tested were not protective against HG/PA-induced INS-1 cell death. Direct supplementation of NAD+ or indirect supplementation through NAD+ salvage or de novo pathway did not protect the death. Knockdown of the NAD+ salvage pathway enzymes such as nicotinamide phosphoribosyl transferase (NAMPT) or nicotinamide mononucleotide adenyltransferase (NMNAT) did not augment death. On the other hand, pharmacological inhibition or knockdown of PARP did not affect death. However, sirtinol as an inhibitor of NAD-dependant deacetylase or knockdown of SIRT3 or SIRT4 significantly reduced the HG/PA-induced death. These data suggest that protective effect of NAM on beta cell glucolipotoxicity is attributed to its inhibitory activity on sirtuins.
Collapse
|
36
|
Leamy AK, Egnatchik RA, Young JD. Molecular mechanisms and the role of saturated fatty acids in the progression of non-alcoholic fatty liver disease. Prog Lipid Res 2013; 52:165-74. [PMID: 23178552 PMCID: PMC3868987 DOI: 10.1016/j.plipres.2012.10.004] [Citation(s) in RCA: 312] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 10/19/2012] [Accepted: 10/30/2012] [Indexed: 02/06/2023]
Abstract
The steady rise in Western obesity rates has been closely linked to significant increases in a multitude of accompanying health problems including non-alcoholic fatty liver disease (NAFLD). NAFLD severity ranges from simple steatosis to acute steatohepatitis, but the molecular mechanisms controlling progression of this disease are poorly understood. Recent literature suggests that elevated free fatty acids (FFAs), especially saturated FFAs, may play an important role in lipotoxic mechanisms, both in experimental models and in NAFLD patients. This review highlights important cellular pathways involved in hepatic lipotoxicity and how the degree of intrahepatic lipid saturation controls cell fate in response to an elevated FFA load. Relevant cellular processes that have been causally linked to lipid-induced apoptosis, known as lipoapoptosis, include endoplasmic reticulum (ER) stress, oxidative stress, mitochondrial dysfunction, and Jun N-terminal kinase (JNK) signaling. In contrast, increased triglyceride synthesis has been shown to have a protective effect against lipotoxicity, despite being one of the hallmark traits of NAFLD. Developing a more nuanced understanding of the molecular mechanisms underlying NAFLD progression will lead to more targeted and effective therapeutics for this increasingly prevalent disease, which to date has no proven pharmacologic treatment to prevent or reverse its course.
Collapse
Affiliation(s)
| | | | - Jamey D. Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235
| |
Collapse
|
37
|
Bakke SS, Moro C, Nikolić N, Hessvik NP, Badin PM, Lauvhaug L, Fredriksson K, Hesselink MK, Boekschoten MV, Kersten S, Gaster M, Thoresen GH, Rustan AC. Palmitic acid follows a different metabolic pathway than oleic acid in human skeletal muscle cells; lower lipolysis rate despite an increased level of adipose triglyceride lipase. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1821:1323-33. [DOI: 10.1016/j.bbalip.2012.07.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2011] [Revised: 05/31/2012] [Accepted: 07/03/2012] [Indexed: 01/22/2023]
|
38
|
A compound (DW1182v) protecting high glucose/palmitate-induced glucolipotoxicity to INS-1 beta cells preserves islet integrity and improves hyperglycemia in obese db/db mouse. Eur J Pharmacol 2012; 696:187-93. [PMID: 23026370 DOI: 10.1016/j.ejphar.2012.09.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 07/30/2012] [Accepted: 09/06/2012] [Indexed: 11/23/2022]
Abstract
Loss of beta cells is a pathogenic cause for the development of type 2 diabetes. High glucose/free fatty acid (HG/FFA)-induced glucolipotoxicity was thought to play a role in the beta cell loss. Thus, application of small molecules capable of preventing HG/FFA-induced glucolipotoxicty to beta cells could be an avenue for a therapeutic intervention for the development of type 2 diabetes. We screened a representative library supplied from Korean Chemical Bank for prevention of high glucose/palmitate (HG/PA)-induced viability reduction of INS-1 beta cells and were able to identify a new small molecule (DW1182v) with a function to protect HG/PA-induced glucolipotoxicity. The protective effect was specific to HG/PA-induced beta cell death since DW1182v did not protect streptozotocin- or cytokine-induced INS-1 cell death. The protective effect by DW1182v was likely due to the reduction of death-promoting endoplasmic reticulum (ER) stress responses such as phospho-C-Jun N-terminal kinase (JNK) and C/EBP homologous protein (CHOP). Treatment of obese diabetic db/db mice with DW1182v preserved islet integrity and thus increased insulin secretion and lowered blood glucose after glucose infusion. These results suggest that a small molecule protecting HG/PA-induced glucolipotoxicity to beta cells can be a new therapeutic candidate to prevent the development of type 2 diabetes.
Collapse
|
39
|
Cao M, Tong Y, Lv Q, Chen X, Long Y, Jiang L, Wan J, Zhang Y, Zhang F, Tong N. PPARδ Activation Rescues Pancreatic β-Cell Line INS-1E from Palmitate-Induced Endoplasmic Reticulum Stress through Enhanced Fatty Acid Oxidation. PPAR Res 2012; 2012:680684. [PMID: 22792088 PMCID: PMC3388384 DOI: 10.1155/2012/680684] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 04/10/2012] [Accepted: 04/12/2012] [Indexed: 02/05/2023] Open
Abstract
One of the key factors responsible for the development of type 2 diabetes is the loss of functional pancreatic β cells. This occurs due to a chronic exposure to a high fatty acid environment. ER stress is caused by an accumulation of irreversible misfold or unfold protein: these trigger the death of functional pancreatic β cells. PPARδ is an orphan nuclear receptor. It plays a pivotal role in regulating the metabolism of dietary lipids and fats. However, the correlation between PPARδ of fatty acids and ER stress of pancreatic β cells is not quite clear till date. Here, we show that PPARδ attenuates palmitate-induced ER stress of pancreatic β cells. On the other hand, PPARδ agonist inhibits both abnormal changes in ER structure and activation of signaling cascade, which is downstream ER stress. Further, we illustrate that PPARδ attenuates palmitate-induced ER stress by promoting fatty acid oxidation through treatment with etomoxir, an inhibitor of fatty acid oxidation. It dramatically abolishes PPARδ-mediated inhibition of ER stress. Finally, we show that PPARδ could protect pancreatic β cells from palmitate-induced cell death and dysfunction of insulin secretion. Our work elucidates the protective effect of PPARδ on the fatty-acid-induced toxicity of pancreatic β cells.
Collapse
Affiliation(s)
- Mingming Cao
- Division of Endocrinology, West China Hospital, Sichuan University, 37 Guoxuexiang, Chengdu 610041, China
| | - Yuzhen Tong
- School of Clinical Medicine, West China Hospital, Sichuan University, 37 Guoxuexiang, Chengdu 610041, China
| | - Qingguo Lv
- Division of Endocrinology, West China Hospital, Sichuan University, 37 Guoxuexiang, Chengdu 610041, China
| | - Xiang Chen
- Research Laboratory of Endocrine and Metabolic Diseases, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yang Long
- Research Laboratory of Endocrine and Metabolic Diseases, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Li Jiang
- Division of Endocrinology, West China Hospital, Sichuan University, 37 Guoxuexiang, Chengdu 610041, China
| | - Jun Wan
- Division of Endocrinology, West China Hospital, Sichuan University, 37 Guoxuexiang, Chengdu 610041, China
| | - Yuwei Zhang
- Division of Endocrinology, West China Hospital, Sichuan University, 37 Guoxuexiang, Chengdu 610041, China
| | - Fang Zhang
- Division of Endocrinology, West China Hospital, Sichuan University, 37 Guoxuexiang, Chengdu 610041, China
| | - Nanwei Tong
- Division of Endocrinology, West China Hospital, Sichuan University, 37 Guoxuexiang, Chengdu 610041, China
| |
Collapse
|
40
|
Meng ZX, Yin Y, Lv JH, Sha M, Lin Y, Gao L, Zhu YX, Sun YJ, Han X. Aberrant activation of liver X receptors impairs pancreatic beta cell function through upregulation of sterol regulatory element-binding protein 1c in mouse islets and rodent cell lines. Diabetologia 2012; 55:1733-44. [PMID: 22415588 DOI: 10.1007/s00125-012-2516-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 02/10/2012] [Indexed: 10/28/2022]
Abstract
AIMS/HYPOTHESIS Liver X receptors (LXR) are important transcriptional regulators of lipid and glucose metabolism. Our previous report demonstrated that LXR activation inhibited pancreatic beta cell proliferation through cell cycle arrest. Here we explore the role of LXR activation in beta cell insulin secretion and the underlying mechanism that might be involved. METHODS Mouse pancreatic islets or insulin-secreting MIN6 cells were exposed to the LXR agonist, T0901317, and insulin secretion, glucose and fatty acid oxidation, and lipogenic gene expression were assessed. The unsaturated fatty acid eicosapentaenoic acid and the dominant negative sterol regulatory element binding protein 1c (SREBP1c) were used to inhibit endogenous SREBP1c and evaluate the involvement of SREBP1c in beta cell dysfunction induced by LXR activation. RESULTS Treatment with the LXR agonist decreased beta cell glucose sensitivity and impaired glucose-stimulated insulin secretion in vivo and in vitro. This was accompanied by derangements of beta cell glucose oxygen consumption, glucose oxidation, ATP production and intracellular voltage-gated calcium channel flux. LXR activation also regulated the expression of lipid metabolism-related genes such as Fas, Acc (also known as Acaca) and Cpt1a, and led to intracellular lipid accumulation. Further studies revealed that inhibition of SREBP1c abolished LXR activation-induced lipid accumulation and improved beta cell glucose metabolism, ATP production and insulin secretion. CONCLUSIONS/INTERPRETATION Our data reveal that aberrant activation of LXR reproduced the phenomenon of beta cell dysfunction in the development of type 2 diabetes in vitro and in vivo. Upregulation of SREBP1c production and the lipotoxicity mediated by it played a central role in this process.
Collapse
Affiliation(s)
- Z X Meng
- Department of Biochemistry and Molecular Biology, Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Prentki M, Madiraju SRM. Glycerolipid/free fatty acid cycle and islet β-cell function in health, obesity and diabetes. Mol Cell Endocrinol 2012; 353:88-100. [PMID: 22108437 DOI: 10.1016/j.mce.2011.11.004] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 11/07/2011] [Accepted: 11/07/2011] [Indexed: 12/16/2022]
Abstract
Pancreatic β-cells secrete insulin in response to fluctuations in blood fuel concentrations, in particular glucose and fatty acids. However, chronic fuel surfeit can overwhelm the metabolic, signaling and secretory capacity of the β-cell leading to its dysfunction and death - often referred to as glucolipotoxicity. In β-cells and many other cells, glucose and lipid metabolic pathways converge into a glycerolipid/free fatty acid (GL/FFA) cycle, which is driven by the substrates, glycerol-3-phosphate and fatty acyl-CoA, derived from glucose and fatty acids, respectively. Although the overall operation of GL/FFA cycle, consisting of lipolysis and lipogenesis, is "futile" in terms of energy expenditure, this metabolic cycle likely plays an indispensable role for various β-cell functions, in particular insulin secretion and excess fuel detoxification. In this review, we discuss the significance of GL/FFA cycle in the β-cell, its regulation and role in generating essential metabolic signals that participate in the lipid amplification arm of glucose stimulated insulin secretion and in β-cell growth. We propose the novel concept that the lipolytic segment of GL/FFA cycle is instrumental in producing signals for insulin secretion, whereas, the lipogenic segment generates signals relevant for β-cell survival/death and growth/proliferation.
Collapse
Affiliation(s)
- Marc Prentki
- Departments of Nutrition and Biochemistry, University of Montreal, Montreal Diabetes Research Center, CR-CHUM, Technopôle Angus, 2901, Montreal, Canada QC H1W 4A4.
| | | |
Collapse
|
42
|
Boren J, Brindle KM. Apoptosis-induced mitochondrial dysfunction causes cytoplasmic lipid droplet formation. Cell Death Differ 2012; 19:1561-70. [PMID: 22460322 PMCID: PMC3422477 DOI: 10.1038/cdd.2012.34] [Citation(s) in RCA: 196] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
A characteristic of apoptosis is the rapid accumulation of cytoplasmic lipid droplets, which are composed largely of neutral lipids. The proton signals from these lipids have been used for the non-invasive detection of cell death using magnetic resonance spectroscopy. We show here that despite an apoptosis-induced decrease in the levels and activities of enzymes involved in lipogenesis, which occurs downstream of p53 activation and inhibition of the mTOR signaling pathway, the increase in lipid accumulation is due to increased de novo lipid synthesis. This results from inhibition of mitochondrial fatty acid β-oxidation, which coupled with an increase in acyl-CoA synthetase activity, diverts fatty acids away from oxidation and into lipid synthesis. The inhibition of fatty acid oxidation can be explained by a rapid rise in mitochondrial membrane potential and an attendant increase in the levels of reactive oxygen species.
Collapse
Affiliation(s)
- J Boren
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Cambridge, UK
| | | |
Collapse
|
43
|
Abstract
Given the functional importance of the endoplasmic reticulum (ER), an organelle that performs folding, modification, and trafficking of secretory and membrane proteins to the Golgi compartment, the maintenance of ER homeostasis in insulin-secreting β-cells is very important. When ER homeostasis is disrupted, the ER generates adaptive signaling pathways, called the unfolded protein response (UPR), to maintain homeostasis of this organelle. However, if homeostasis fails to be restored, the ER initiates death signaling pathways. New observations suggest that both chronic hyperglycemia and hyperlipidemia, known as important causative factors of type 2 diabetes (T2D), disrupt ER homeostasis to induce unresolvable UPR activation and β-cell death. This review examines how the UPR pathways, induced by high glucose and free fatty acids (FFAs), interact to disrupt ER function and cause β-cell dysfunction and death.
Collapse
Affiliation(s)
- Sung Hoon Back
- School of Biological Sciences, University of Ulsan, Ulsan, Republic of Korea 680-749
| | - Randal J. Kaufman
- Degenerative Disease Research Program, Neuroscience, Aging, and Stem Cell Research Center, Sanford Burnham Medical Research Institute, La Jolla, California 92037
| |
Collapse
|
44
|
Virtanen SM, Nevalainen J, Kronberg-Kippilä C, Ahonen S, Tapanainen H, Uusitalo L, Takkinen HM, Niinistö S, Ovaskainen ML, Kenward MG, Veijola R, Ilonen J, Simell O, Knip M. Food consumption and advanced β cell autoimmunity in young children with HLA-conferred susceptibility to type 1 diabetes: a nested case-control design. Am J Clin Nutr 2012; 95:471-8. [PMID: 22237062 DOI: 10.3945/ajcn.111.018879] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Evidence for the role of food consumption during childhood in the development of β cell autoimmunity is scarce and fragmentary. OBJECTIVE We set out to study the associations of longitudinal food consumption in children with the development of advanced β cell autoimmunity. DESIGN Children with advanced β cell autoimmunity (n = 232) (ie, with repeated positivity for antibodies against islet cells) together with positivity for at least one of the other 3 antibodies analyzed or clinical type 1 diabetes were identified from a prospective birth cohort of 6069 infants with HLA-DQB1-conferred susceptibility to type 1 diabetes who were born in 1996-2004, with the longest follow-up to the age of 11 y. Repeated 3-d food records were completed by the families and daycare personnel. Diabetes-associated autoantibodies and diets were measured at 3-12-mo intervals. Four control subjects, who were matched for birth date, sex, area, and genetic risk, were randomly selected for each case. RESULTS In the main food groups, only intakes of cow-milk products (OR: 1.05; 95% CI: 1.00, 1.10) and fruit and berry juices (OR: 1.09; 95% CI: 1.02, 1.12) were significantly, although marginally, associated with advanced β cell autoimmunity. The consumption of fresh milk products and cow milk-based infant formulas was related to the endpoint, whereas no evidence was shown for consumption of sour milk products and cheese. The intake of fat from all milk products and protein from fresh milk products was associated with risk of advanced β cell autoimmunity. CONCLUSION Intakes of cow milk and fruit and berry juices could be related to the development of advanced β cell autoimmunity. This trial was registered at clinicaltrials.gov as number NCT00223613.
Collapse
Affiliation(s)
- Suvi M Virtanen
- Unit of Nutrition, Department of Lifestyle and Participation, National Institute for Health and Welfare, Helsinki, Finland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Protective effects of arachidonic acid against palmitic acid-mediated lipotoxicity in HIT-T15 cells. Mol Cell Biochem 2011; 364:19-28. [DOI: 10.1007/s11010-011-1200-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Accepted: 12/15/2011] [Indexed: 01/03/2023]
|
46
|
Sargsyan E, Sol ERM, Bergsten P. UPR in palmitate-treated pancreatic beta-cells is not affected by altering oxidation of the fatty acid. Nutr Metab (Lond) 2011; 8:70. [PMID: 21978671 PMCID: PMC3197479 DOI: 10.1186/1743-7075-8-70] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 10/06/2011] [Indexed: 12/27/2022] Open
Abstract
Background Elevated levels of lipids are detrimental for beta-cell function and mass. One of the mechanisms of how fatty acids induce apoptosis is development of the unfolded protein response (UPR). It is still far from understood how fatty acids activate the UPR, however. Methods We examined how palmitate-induced activation of the UPR was affected by altering the metabolism of the fatty acid in insulin-secreting INS-1E and MIN6 cell lines and intact human islets. To increase oxidation, we used low glucose (5.5 mM) or AICAR; and to reduce oxidation, we used high glucose (25 mM) or etomoxir. UPR was measured after 3, 24 and 48 hours of palmitate treatment. Results Modulation of palmitate oxidation by either glucose or the pharmacological agents did not affect palmitate-induced UPR activation. Conclusion Our finding suggests that other factors than oxidation of palmitate play a role in the activation of UPR in fatty acid-treated beta-cells.
Collapse
Affiliation(s)
- Ernest Sargsyan
- Department of Medical Cell Biology, Uppsala University, Box 571, SE-75123, Uppsala, Sweden.
| | | | | |
Collapse
|
47
|
|
48
|
Choi SE, Kim TH, Yi SA, Hwang YC, Hwang WS, Choe SJ, Han SJ, Kim HJ, Kim DJ, Kang Y, Lee KW. Capsaicin attenuates palmitate-induced expression of macrophage inflammatory protein 1 and interleukin 8 by increasing palmitate oxidation and reducing c-Jun activation in THP-1 (human acute monocytic leukemia cell) cells. Nutr Res 2011; 31:468-78. [PMID: 21745629 DOI: 10.1016/j.nutres.2011.05.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Revised: 04/30/2011] [Accepted: 05/09/2011] [Indexed: 10/18/2022]
Abstract
Capsaicin, a spicy component of hot peppers, has been shown to improve inflammatory disease and obesity. In this study, we tested the hypothesis that the anti-inflammatory activity of capsaicin can be used to improve free fatty acid (FFA)-induced inflammation by reducing gene expression of macrophage inflammatory protein 1 (MIP-1) and interleukin 8 (IL-8) in THP-1 (human acute monocytic leukemia cell) macrophages. To investigate whether capsaicin ameliorates palmitate-induced MIP-1 and IL-8 gene expressions, we treated THP-1 cells with palmitate in the presence or absence of capsaicin and measured MIP-1 and IL-8 by real-time polymerase chain reaction. To elucidate the mechanism by which capsaicin effects on palmitate-induced MIP-1 and IL-8 gene expressions, we performed immunoblotting with stress kinase-related antibodies and measured palmitate oxidation and palmitate oxidation-related gene expression. Palmitate and stearate but not the unsaturated FFA oleate significantly increased MIP-1 and IL-8 expressions in THP-1 macrophages. Treatment with capsaicin or FFA oxidation stimulators inhibited palmitate-induced MIP-1 and IL-8 expressions in THP-1 macrophages. Capsaicin increased the gene expression of carnitine palmitoyltransferase 1 and the β-oxidation of palmitate. Furthermore, capsaicin significantly reduced palmitate-stimulated activation of c-Jun N-terminal kinase, c-Jun, and p38. Our data suggest that the attenuation of palmitate-induced MIP-1 and IL-8 gene expressions by capsaicin is associated with reduced activation of c-Jun N-terminal kinase, c-Jun, and p38 and preserved β-oxidation activity.
Collapse
Affiliation(s)
- Sung-E Choi
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon 443-749, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|