1
|
Staab TA, McIntyre G, Wang L, Radeny J, Bettcher L, Guillen M, Peck MP, Kalil AP, Bromley SP, Raftery D, Chan JP. The lipidomes of C. elegans with mutations in asm-3/acid sphingomyelinase and hyl-2/ceramide synthase show distinct lipid profiles during aging. Aging (Albany NY) 2023; 15:650-674. [PMID: 36787434 PMCID: PMC9970312 DOI: 10.18632/aging.204515] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 02/01/2023] [Indexed: 02/16/2023]
Abstract
Lipid metabolism affects cell and physiological functions that mediate animal healthspan and lifespan. Lipidomics approaches in model organisms have allowed us to better understand changes in lipid composition related to age and lifespan. Here, using the model C. elegans, we examine the lipidomes of mutants lacking enzymes critical for sphingolipid metabolism; specifically, we examine acid sphingomyelinase (asm-3), which breaks down sphingomyelin to ceramide, and ceramide synthase (hyl-2), which synthesizes ceramide from sphingosine. Worm asm-3 and hyl-2 mutants have been previously found to be long- and short-lived, respectively. We analyzed longitudinal lipid changes in wild type animals compared to mutants at 1-, 5-, and 10-days of age. We detected over 700 different lipids in several lipid classes. Results indicate that wildtype animals exhibit increased triacylglycerols (TAG) at 10-days compared to 1-day, and decreased lysophoshatidylcholines (LPC). We find that 10-day hyl-2 mutants have elevated total polyunsaturated fatty acids (PUFA) and increased LPCs compared to 10-day wildtype animals. These changes mirror another short-lived model, the daf-16/FOXO transcription factor that is downstream of the insulin-like signaling pathway. In addition, we find that hyl-2 mutants have poor oxidative stress response, supporting a model where mutants with elevated PUFAs may accumulate more oxidative damage. On the other hand, 10-day asm-3 mutants have fewer TAGs. Intriguingly, asm-3 mutants have a similar lipid composition as the long-lived, caloric restriction model eat-2/mAChR mutant. Together, these analyses highlight the utility of lipidomic analyses to characterize metabolic changes during aging in C. elegans.
Collapse
Affiliation(s)
- Trisha A. Staab
- Department of Biology, Marian University, Indianapolis, IN 46222, USA
| | - Grace McIntyre
- Department of Biology, Marian University, Indianapolis, IN 46222, USA
| | - Lu Wang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA
| | - Joycelyn Radeny
- Department of Biology, Juniata College, Huntingdon, PA 16652, USA
| | - Lisa Bettcher
- Northwest Metabolomics Research Center, University of Washington, Seattle, WA 98195, USA
| | - Melissa Guillen
- Department of Biology, Marian University, Indianapolis, IN 46222, USA
| | - Margaret P. Peck
- Department of Biology, Juniata College, Huntingdon, PA 16652, USA
| | - Azia P. Kalil
- Department of Biology, Juniata College, Huntingdon, PA 16652, USA
| | | | - Daniel Raftery
- Northwest Metabolomics Research Center, University of Washington, Seattle, WA 98195, USA
| | - Jason P. Chan
- Department of Biology, Marian University, Indianapolis, IN 46222, USA
| |
Collapse
|
2
|
Ceramide and Sphingosine-1-Phosphate in Neurodegenerative Disorders and Their Potential Involvement in Therapy. Int J Mol Sci 2022; 23:ijms23147806. [PMID: 35887154 PMCID: PMC9324343 DOI: 10.3390/ijms23147806] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 02/04/2023] Open
Abstract
Neurodegenerative disorders (ND) are progressive diseases of the nervous system, often without resolutive therapy. They are characterized by a progressive impairment and loss of specific brain regions and neuronal populations. Cellular and animal model studies have identified several molecular mechanisms that play an important role in the pathogenesis of ND. Among them are alterations of lipids, in particular sphingolipids, that play a crucial role in neurodegeneration. Overall, during ND, ceramide-dependent pro-apoptotic signalling is promoted, whereas levels of the neuroprotective spingosine-1-phosphate are reduced. Moreover, ND are characterized by alterations of the metabolism of complex sphingolipids. The finding that altered sphingolipid metabolism has a role in ND suggests that its modulation might provide a useful strategy to identify targets for possible therapies. In this review, based on the current literature, we will discuss how bioactive sphingolipids (spingosine-1-phosphate and ceramide) are involved in some ND (Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis) and their possible involvement in therapies.
Collapse
|
3
|
Ali T, Lei X, Barbour SE, Koizumi A, Chalfant CE, Ramanadham S. Alterations in β-Cell Sphingolipid Profile Associated with ER Stress and iPLA 2β: Another Contributor to β-Cell Apoptosis in Type 1 Diabetes. Molecules 2021; 26:molecules26216361. [PMID: 34770770 PMCID: PMC8587436 DOI: 10.3390/molecules26216361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/11/2021] [Accepted: 10/11/2021] [Indexed: 02/06/2023] Open
Abstract
Type 1 diabetes (T1D) development, in part, is due to ER stress-induced β-cell apoptosis. Activation of the Ca2+-independent phospholipase A2 beta (iPLA2β) leads to the generation of pro-inflammatory eicosanoids, which contribute to β-cell death and T1D. ER stress induces iPLA2β-mediated generation of pro-apoptotic ceramides via neutral sphingomyelinase (NSMase). To gain a better understanding of the impact of iPLA2β on sphingolipids (SLs), we characterized their profile in β-cells undergoing ER stress. ESI/MS/MS analyses followed by ANOVA/Student’s t-test were used to assess differences in sphingolipids molecular species in Vector (V) control and iPLA2β-overexpressing (OE) INS-1 and Akita (AK, spontaneous model of ER stress) and WT-littermate (AK-WT) β-cells. As expected, iPLA2β induction was greater in the OE and AK cells in comparison with V and WT cells. We report here that ER stress led to elevations in pro-apoptotic and decreases in pro-survival sphingolipids and that the inactivation of iPLA2β restores the sphingolipid species toward those that promote cell survival. In view of our recent finding that the SL profile in macrophages—the initiators of autoimmune responses leading to T1D—is not significantly altered during T1D development, we posit that the iPLA2β-mediated shift in the β-cell sphingolipid profile is an important contributor to β-cell death associated with T1D.
Collapse
Affiliation(s)
- Tomader Ali
- Research Department, Imperial College London Diabetes Center, Abu Dhabi 51133, United Arab Emirates;
| | - Xiaoyong Lei
- Department of Cell, Developmental, and Integrative Biology and Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Suzanne E. Barbour
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Akio Koizumi
- Department of Health and Environmental Sciences, Kyoto Graduate School of Medicine, Kyoto 606-8501, Japan;
| | - Charles E. Chalfant
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA;
| | - Sasanka Ramanadham
- Department of Cell, Developmental, and Integrative Biology and Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Correspondence: ; Tel.: +1-205-996-5973; Fax: +1-205-996-5220
| |
Collapse
|
4
|
Choi RH, Tatum SM, Symons JD, Summers SA, Holland WL. Ceramides and other sphingolipids as drivers of cardiovascular disease. Nat Rev Cardiol 2021; 18:701-711. [PMID: 33772258 PMCID: PMC8978615 DOI: 10.1038/s41569-021-00536-1] [Citation(s) in RCA: 186] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2021] [Indexed: 02/03/2023]
Abstract
Increases in calorie consumption and sedentary lifestyles are fuelling a global pandemic of cardiometabolic diseases, including coronary artery disease, diabetes mellitus, cardiomyopathy and heart failure. These lifestyle factors, when combined with genetic predispositions, increase the levels of circulating lipids, which can accumulate in non-adipose tissues, including blood vessel walls and the heart. The metabolism of these lipids produces bioactive intermediates that disrupt cellular function and survival. A compelling body of evidence suggests that sphingolipids, such as ceramides, account for much of the tissue damage in these cardiometabolic diseases. In humans, serum ceramide levels are proving to be accurate biomarkers of adverse cardiovascular disease outcomes. In mice and rats, pharmacological inhibition or depletion of enzymes driving de novo ceramide synthesis prevents the development of diabetes, atherosclerosis, hypertension and heart failure. In cultured cells and isolated tissues, ceramides perturb mitochondrial function, block fuel usage, disrupt vasodilatation and promote apoptosis. In this Review, we discuss the body of literature suggesting that ceramides are drivers - and not merely passengers - on the road to cardiovascular disease. Moreover, we explore the feasibility of therapeutic strategies to lower ceramide levels to improve cardiovascular health.
Collapse
Affiliation(s)
- Ran Hee Choi
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Sean M Tatum
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - J David Symons
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA.
| | - William L Holland
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
5
|
Crivelli SM, Giovagnoni C, Visseren L, Scheithauer AL, de Wit N, den Hoedt S, Losen M, Mulder MT, Walter J, de Vries HE, Bieberich E, Martinez-Martinez P. Sphingolipids in Alzheimer's disease, how can we target them? Adv Drug Deliv Rev 2020; 159:214-231. [PMID: 31911096 DOI: 10.1016/j.addr.2019.12.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 10/09/2019] [Accepted: 12/31/2019] [Indexed: 01/06/2023]
Abstract
Altered levels of sphingolipids and their metabolites in the brain, and the related downstream effects on neuronal homeostasis and the immune system, provide a framework for understanding mechanisms in neurodegenerative disorders and for developing new intervention strategies. In this review we will discuss: the metabolites of sphingolipids that function as second messengers; and functional aberrations of the pathway resulting in Alzheimer's disease (AD) pathophysiology. Focusing on the central product of the sphingolipid pathway ceramide, we describ approaches to pharmacologically decrease ceramide levels in the brain and we argue on how the sphingolipid pathway may represent a new framework for developing novel intervention strategies in AD. We also highlight the possible use of clinical and non-clinical drugs to modulate the sphingolipid pathway and sphingolipid-related biological cascades.
Collapse
|
6
|
The Role of Ceramide and Sphingosine-1-Phosphate in Alzheimer's Disease and Other Neurodegenerative Disorders. Mol Neurobiol 2019; 56:5436-5455. [PMID: 30612333 PMCID: PMC6614129 DOI: 10.1007/s12035-018-1448-3] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 12/06/2018] [Indexed: 12/11/2022]
Abstract
Bioactive sphingolipids-ceramide, sphingosine, and their respective 1-phosphates (C1P and S1P)-are signaling molecules serving as intracellular second messengers. Moreover, S1P acts through G protein-coupled receptors in the plasma membrane. Accumulating evidence points to sphingolipids' engagement in brain aging and in neurodegenerative disorders such as Alzheimer's, Parkinson's, and Huntington's diseases and amyotrophic lateral sclerosis. Metabolic alterations observed in the course of neurodegeneration favor ceramide-dependent pro-apoptotic signaling, while the levels of the neuroprotective S1P are reduced. These trends are observed early in the diseases' development, suggesting causal relationship. Mechanistic evidence has shown links between altered ceramide/S1P rheostat and the production, secretion, and aggregation of amyloid β/α-synuclein as well as signaling pathways of critical importance for the pathomechanism of protein conformation diseases. Sphingolipids influence multiple aspects of Akt/protein kinase B signaling, a pathway that regulates metabolism, stress response, and Bcl-2 family proteins. The cross-talk between sphingolipids and transcription factors including NF-κB, FOXOs, and AP-1 may be also important for immune regulation and cell survival/death. Sphingolipids regulate exosomes and other secretion mechanisms that can contribute to either the spread of neurotoxic proteins between brain cells, or their clearance. Recent discoveries also suggest the importance of intracellular and exosomal pools of small regulatory RNAs in the creation of disturbed signaling environment in the diseased brain. The identified interactions of bioactive sphingolipids urge for their evaluation as potential therapeutic targets. Moreover, the early disturbances in sphingolipid metabolism may deliver easily accessible biomarkers of neurodegenerative disorders.
Collapse
|
7
|
de Mello-Coelho V, Cutler RG, Bunbury A, Tammara A, Mattson MP, Taub DD. Age-associated alterations in the levels of cytotoxic lipid molecular species and oxidative stress in the murine thymus are reduced by growth hormone treatment. Mech Ageing Dev 2017; 167:46-55. [PMID: 28865931 DOI: 10.1016/j.mad.2017.08.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 07/24/2017] [Accepted: 08/22/2017] [Indexed: 12/13/2022]
Abstract
During age-associated thymic involution, thymocytes decrease and lipid-laden cells accumulate. However, if and how aging affects the thymic lipid profile is not well understood, nor is it known if the hormonal milieu modifies this process. Here we demonstrate a correlation between reduced thymocyte numbers and markers of inflammation and oxidative stress with age. Evaluating the lipidomics profile of the whole thymus, between the ages of 4 (young) and 18 months (old), we found increased amounts of triacylglycerides, free cholesterol, cholesterol ester and 4-hydroxynonenal (4-HNE) with age. Moreover, levels of C24:0 and C24:1 sphingomyelins and ceramide C16:0 were elevated in 12-14 month-old (middle-aged) mice while the levels of sulfatide ceramide and ganglioside GD1a increased in the old thymus. Evaluating isolated thymocytes, we found increased levels of cholesterol ester and 4-HNE adducts, as compared to young mice. Next, we treated middle-aged mice with growth hormone (GH), which has been considered a potent immunomodulator. GH reduced thymic levels of TNF-α and 4-HNE and increased the number of thymocytes as well as the thymic levels of dihydroceramide, a ceramide precursor and autophagic stimuli for cell survival. In conclusion, GH treatment attenuated inflammation and age-related increases in oxidative stress and lipotoxicity in the thymus.
Collapse
Affiliation(s)
- Valeria de Mello-Coelho
- Laboratory of Immunology, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224-6825, USA; Laboratory of Immunophysiology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Roy G Cutler
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224-6825, USA.
| | - Allyson Bunbury
- Laboratory of Immunology, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224-6825, USA.
| | - Anita Tammara
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224-6825, USA.
| | - Mark P Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224-6825, USA.
| | - Dennis D Taub
- Laboratory of Immunology, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224-6825, USA; Center for Translational Studies, Medical Service, VA Medical Center-DC, Washington DC, 20422, USA.
| |
Collapse
|
8
|
Voelzmann A, Wulf AL, Eckardt F, Thielisch M, Brondolin M, Pesch YY, Sociale M, Bauer R, Hoch M. NuclearDrosophilaCerS Schlank regulates lipid homeostasis via the homeodomain, independent of the lag1p motif. FEBS Lett 2016; 590:971-81. [DOI: 10.1002/1873-3468.12125] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 02/17/2016] [Accepted: 03/03/2016] [Indexed: 11/08/2022]
Affiliation(s)
- André Voelzmann
- Program Unit Development, Genetics & Molecular Physiology; Laboratory for Molecular Developmental Biology; LIMES-Institute; University of Bonn; Germany
| | - Anna-Lena Wulf
- Program Unit Development, Genetics & Molecular Physiology; Laboratory for Molecular Developmental Biology; LIMES-Institute; University of Bonn; Germany
| | - Franka Eckardt
- Program Unit Development, Genetics & Molecular Physiology; Laboratory for Molecular Developmental Biology; LIMES-Institute; University of Bonn; Germany
| | - Melanie Thielisch
- Program Unit Development, Genetics & Molecular Physiology; Laboratory for Molecular Developmental Biology; LIMES-Institute; University of Bonn; Germany
| | - Mirco Brondolin
- Program Unit Development, Genetics & Molecular Physiology; Laboratory for Molecular Developmental Biology; LIMES-Institute; University of Bonn; Germany
| | - Yanina-Yasmin Pesch
- Program Unit Development, Genetics & Molecular Physiology; Laboratory for Molecular Developmental Biology; LIMES-Institute; University of Bonn; Germany
| | - Mariangela Sociale
- Program Unit Development, Genetics & Molecular Physiology; Laboratory for Molecular Developmental Biology; LIMES-Institute; University of Bonn; Germany
| | - Reinhard Bauer
- Program Unit Development, Genetics & Molecular Physiology; Laboratory for Molecular Developmental Biology; LIMES-Institute; University of Bonn; Germany
| | - Michael Hoch
- Program Unit Development, Genetics & Molecular Physiology; Laboratory for Molecular Developmental Biology; LIMES-Institute; University of Bonn; Germany
| |
Collapse
|
9
|
Ranftler C, Meisslitzer-Ruppitsch C, Stangl H, Röhrl C, Fruhwürth S, Neumüller J, Pavelka M, Ellinger A. 2-Deoxy-D-glucose treatment changes the Golgi apparatus architecture without blocking synthesis of complex lipids. Histochem Cell Biol 2014; 143:369-80. [PMID: 25422148 DOI: 10.1007/s00418-014-1297-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2014] [Indexed: 11/29/2022]
Abstract
The classic Golgi apparatus organization, an arrangement of highly ordered cisternal stacks with tubular-vesicular membrane specializations on both sides, is the functional image of a continuous flow of contents and membranes with input, metabolization, and output in a dynamic steady state. In response to treatment with 2-deoxy-D-glucose (2-DG), which lowers the cellular ATP level by about 70% within minutes, this organization is rapidly replaced by tubular-glomerular membrane convolutes described as Golgi networks and bodies. 2-DG is a non-metabolizable glucose analogue and competitive inhibitor of glycolysis, which has become attractive in the context of therapeutic approaches for several kinds of tumors specifically targeting glycolysis in cancer. With the question of whether the functions of the Golgi apparatus in lipid synthesis would be influenced by the 2-DG-induced Golgi apparatus reorganization, we focused on lipid metabolism within the Golgi bodies. For this, we applied a fluorophore-labeled short-chain ceramide (BODIPY-Cer) in various combinations with 2-DG treatment to HepG2 cell cultures and followed uptake, enrichment and metabolization to higher ordered lipids. The cellular ATP status in each experiment was controlled with a bioluminescence assay, and the response of the Golgi apparatus was tracked by immunostaining of the trans-Golgi network protein TGN46. For electron microscopy, the fluorescent BODIPY-Cer signals were converted into electron-dense precipitates by photooxidation of diaminobenzidine (DAB); DAB precipitates labeled trans-Golgi areas in control cultures but also compartments at the periphery of the Golgi bodies formed in response to 2-DG treatment, thus indicating that concentration of ceramide takes place in spite of the Golgi apparatus reorganization. Lipid analyses by thin-layer chromatography (TLC) performed in parallel showed that BODIPY-Cer is not only concentrated in compartments of the 2-DG-induced Golgi bodies but is partly metabolized to BODIPY-sphingomyelin. Both, uptake and condensation of BODIPY-Cer and its conversion to complex lipids indicate that functions of the Golgi apparatus in the cellular lipid metabolism persist although the classic Golgi apparatus organization is abolished.
Collapse
Affiliation(s)
- Carmen Ranftler
- Department of Cell Biology and Ultrastructure Research, Center for Anatomy and Cell Biology, Medical University of Vienna, Schwarzspanierstr. 17, 1090, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Alexaki A, Gupta SD, Majumder S, Kono M, Tuymetova G, Harmon JM, Dunn TM, Proia RL. Autophagy regulates sphingolipid levels in the liver. J Lipid Res 2014; 55:2521-31. [PMID: 25332431 DOI: 10.1194/jlr.m051862] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Sphingolipid levels are tightly regulated to maintain cellular homeostasis. During pathologic conditions such as in aging, inflammation, and metabolic and neurodegenerative diseases, levels of some sphingolipids, including the bioactive metabolite ceramide, are elevated. Sphingolipid metabolism has been linked to autophagy, a critical catabolic process in both normal cell function and disease; however, the in vivo relevance of the interaction is not well-understood. Here, we show that blocking autophagy in the liver by deletion of the Atg7 gene, which is essential for autophagosome formation, causes an increase in sphingolipid metabolites including ceramide. We also show that overexpression of serine palmitoyltransferase to elevate de novo sphingolipid biosynthesis induces autophagy in the liver. The results reveal autophagy as a process that limits excessive ceramide levels and that is induced by excessive elevation of de novo sphingolipid synthesis in the liver. Dysfunctional autophagy may be an underlying mechanism causing elevations in ceramide that may contribute to pathogenesis in diseases.
Collapse
Affiliation(s)
- Aikaterini Alexaki
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Sita D Gupta
- Departments of Biochemistry Uniformed Services University of the Health Sciences, Bethesda, MD 20184
| | - Saurav Majumder
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Mari Kono
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Galina Tuymetova
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jeffrey M Harmon
- Molecular Biology and Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, MD 20184
| | - Teresa M Dunn
- Departments of Biochemistry Uniformed Services University of the Health Sciences, Bethesda, MD 20184
| | - Richard L Proia
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
11
|
Ohanian J, Liao A, Forman SP, Ohanian V. Age-related remodeling of small arteries is accompanied by increased sphingomyelinase activity and accumulation of long-chain ceramides. Physiol Rep 2014; 2:2/5/e12015. [PMID: 24872355 PMCID: PMC4098743 DOI: 10.14814/phy2.12015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The structure and function of large arteries alters with age leading to increased risk of cardiovascular disease. Age‐related large artery remodeling and arteriosclerosis is associated with increased collagen deposition, inflammation, and endothelial dysfunction. Bioactive sphingolipids are known to regulate these processes, and are also involved in aging and cellular senescence. However, less is known about age‐associated alterations in small artery morphology and function or whether changes in arterial sphingolipids occur in aging. We show that mesenteric small arteries from old sheep have increased lumen diameter and media thickness without a change in media to lumen ratio, indicative of outward hypertrophic remodeling. This remodeling occurred without overt changes in blood pressure or pulse pressure indicating it was a consequence of aging per se. There was no age‐associated change in mechanical properties of the arteries despite an increase in total collagen content and deposition of collagen in a thickened intima layer in arteries from old animals. Analysis of the sphingolipid profile showed an increase in long‐chain ceramide (C14–C20), but no change in the levels of sphingosine or sphingosine‐1‐phosphate in arteries from old compared to young animals. This was accompanied by a parallel increase in acid and neutral sphingomyelinase activity in old arteries compared to young. This study demonstrates remodeling of small arteries during aging that is accompanied by accumulation of long‐chain ceramides. This suggests that sphingolipids may be important mediators of vascular aging. In this study, we have investigated remodeling of small arteries in a large animal model of aging, the sheep. We show that there is age‐related formation of neointima and increased collagen deposition that is accompanied by changes in sphingolipid metabolism resulting in ceramide accumulation in the tissues. These are the first data implicating sphingolipids as important mediators of vascular aging in small arteries. Given that aging is a major risk factor for cardiovascular disease, our study opens a new area for further research into the mechanisms that underlie vascular remodeling in aging.
Collapse
Affiliation(s)
- Jacqueline Ohanian
- Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | - Aiyin Liao
- Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | - Simon P Forman
- Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | - Vasken Ohanian
- Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| |
Collapse
|
12
|
Rao RP, Scheffer L, Srideshikan SM, Parthibane V, Kosakowska-Cholody T, Masood MA, Nagashima K, Gudla P, Lockett S, Acharya U, Acharya JK. Ceramide transfer protein deficiency compromises organelle function and leads to senescence in primary cells. PLoS One 2014; 9:e92142. [PMID: 24642596 PMCID: PMC3958450 DOI: 10.1371/journal.pone.0092142] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 02/17/2014] [Indexed: 11/18/2022] Open
Abstract
Ceramide transfer protein (CERT) transfers ceramide from the endoplasmic reticulum (ER) to the Golgi complex. Its deficiency in mouse leads to embryonic death at E11.5. CERT deficient embryos die from cardiac failure due to defective organogenesis, but not due to ceramide induced apoptotic or necrotic cell death. In the current study we examined the effect of CERT deficiency in a primary cell line, namely, mouse embryonic fibroblasts (MEFs). We show that in MEFs, unlike in mutant embryos, lack of CERT does not lead to increased ceramide but causes an accumulation of hexosylceramides. Nevertheless, the defects due to defective sphingolipid metabolism that ensue, when ceramide fails to be trafficked from ER to the Golgi complex, compromise the viability of the cell. Therefore, MEFs display an incipient ER stress. While we observe that ceramide trafficking from ER to the Golgi complex is compromised, the forward transport of VSVG-GFP protein is unhindered from ER to Golgi complex to the plasma membrane. However, retrograde trafficking of the plasma membrane-associated cholera toxin B to the Golgi complex is reduced. The dysregulated sphingolipid metabolism also leads to increased mitochondrial hexosylceramide. The mitochondrial functions are also compromised in mutant MEFs since they have reduced ATP levels, have increased reactive oxygen species, and show increased glutathione reductase activity. Live-cell imaging shows that the mutant mitochondria exhibit reduced fission and fusion events. The mitochondrial dysfunction leads to an increased mitophagy in the CERT mutant MEFs. The compromised organelle function compromise cell viability and results in premature senescence of these MEFs.
Collapse
Affiliation(s)
- Raghavendra Pralhada Rao
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, Frederick, Maryland, United States of America
| | - Luana Scheffer
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, Frederick, Maryland, United States of America
| | - Sargur M. Srideshikan
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, Frederick, Maryland, United States of America
| | - Velayoudame Parthibane
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, Frederick, Maryland, United States of America
| | - Teresa Kosakowska-Cholody
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, Frederick, Maryland, United States of America
| | - M. Athar Masood
- Laboratory of Proteomics and Analytical Technologies, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Kunio Nagashima
- Electron Microscopy Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Prabhakar Gudla
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Stephen Lockett
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Usha Acharya
- Program in Gene Function and Expression, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Jairaj K. Acharya
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, Frederick, Maryland, United States of America
- * E-mail:
| |
Collapse
|
13
|
Embryonic expression of Drosophila ceramide synthase schlank in developing gut, CNS and PNS. Gene Expr Patterns 2011; 11:501-10. [PMID: 21907829 DOI: 10.1016/j.gep.2011.08.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 08/17/2011] [Accepted: 08/19/2011] [Indexed: 12/17/2022]
Abstract
Schlank is a member of the highly conserved ceramide synthase family and controls growth and body fat in Drosophila. Ceramide synthases are key enzymes in the sphingolipid de novo synthesis pathway. Ceramide synthase proteins and the (dihydro)ceramide produced are involved in a variety of biological processes among them apoptosis and neurodegeneration. The full extent of their involvement in these processes will require a precise analysis of the distribution and expression pattern of ceramide synthases. Paralogs of the ceramide synthase family have been found in all eukaryotes studied, however the mRNA and protein expression patterns have not yet been analysed systematically. In this study, we use antibodies that specifically recognize Schlank, a schlank mRNA probe and an endogenous schlank promoter driven LacZ reporter line to reveal the expression pattern of Schlank throughout embryogenesis. We found that Schlank is expressed in all embryonic epithelia during embryogenesis including the developing epidermis and the gastrointestinal tract. In addition, Schlank is upregulated in the developing central (CNS) and peripheral nervous system (PNS). Co-staining experiments with neuronal and glial markers revealed specific expression of Schlank in glial and neuronal cells of the CNS and PNS.
Collapse
|
14
|
Fu C, Wang T, Wang Y, Chen X, Jiao J, Ma F, Zhong M, Bi K. Metabonomics study of the protective effects of green tea polyphenols on aging rats induced by d-galactose. J Pharm Biomed Anal 2011; 55:1067-74. [DOI: 10.1016/j.jpba.2011.03.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 03/05/2011] [Accepted: 03/05/2011] [Indexed: 11/24/2022]
|
15
|
Ponnusamy S, Meyers-Needham M, Senkal CE, Saddoughi SA, Sentelle D, Selvam SP, Salas A, Ogretmen B. Sphingolipids and cancer: ceramide and sphingosine-1-phosphate in the regulation of cell death and drug resistance. Future Oncol 2011; 6:1603-24. [PMID: 21062159 DOI: 10.2217/fon.10.116] [Citation(s) in RCA: 238] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Sphingolipids have emerged as bioeffector molecules, controlling various aspects of cell growth and proliferation in cancer, which is becoming the deadliest disease in the world. These lipid molecules have also been implicated in the mechanism of action of cancer chemotherapeutics. Ceramide, the central molecule of sphingolipid metabolism, generally mediates antiproliferative responses, such as cell growth inhibition, apoptosis induction, senescence modulation, endoplasmic reticulum stress responses and/or autophagy. Interestingly, recent studies suggest de novo-generated ceramides may have distinct and opposing roles in the promotion/suppression of tumors, and that these activities are based on their fatty acid chain lengths, subcellular localization and/or direct downstream targets. For example, in head and neck cancer cells, ceramide synthase 6/C(16)-ceramide addiction was revealed, and this was associated with increased tumor growth, whereas downregulation of its synthesis resulted in ER stress-induced apoptosis. By contrast, ceramide synthase 1-generated C(18)-ceramide has been shown to suppress tumor growth in various cancer models, both in situ and in vivo. In addition, ceramide metabolism to generate sphingosine-1-phosphate (S1P) by sphingosine kinases 1 and 2 mediates, with or without the involvement of G-protein-coupled S1P receptor signaling, prosurvival, angiogenesis, metastasis and/or resistance to drug-induced apoptosis. Importantly, recent findings regarding the mechanisms by which sphingolipid metabolism and signaling regulate tumor growth and progression, such as identifying direct intracellular protein targets of sphingolipids, have been key for the development of new chemotherapeutic strategies. Thus, in this article, we will present conclusions of recent studies that describe opposing roles of de novo-generated ceramides by ceramide synthases and/or S1P in the regulation of cancer pathogenesis, as well as the development of sphingolipid-based cancer therapeutics and drug resistance.
Collapse
Affiliation(s)
- Suriyan Ponnusamy
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Rotstein NP, Miranda GE, Abrahan CE, German OL. Regulating survival and development in the retina: key roles for simple sphingolipids. J Lipid Res 2010; 51:1247-62. [PMID: 20100817 PMCID: PMC3035489 DOI: 10.1194/jlr.r003442] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Revised: 01/25/2010] [Indexed: 12/28/2022] Open
Abstract
Many sphingolipids have key functions in the regulation of crucial cellular processes. Ceramide (Cer) and sphingosine (Sph) induce growth arrest and cell death in multiple situations of cellular stress. On the contrary, sphingosine-1-phosphate (S1P), the product of Sph phosphorylation, promotes proliferation, differentiation, and survival in different cell systems. This review summarizes the roles of these simple sphingolipids in different tissues and then analyzes their possible functions in the retina. Alterations in proliferation, neovascularization, differentiation, and cell death are critical in major retina diseases and collective evidence points to a role for sphingolipids in these processes. Cer induces inflammation and apoptosis in endothelial and retinal pigmented epithelium cells, leading to several retinopathies. S1P can prevent this death but also promotes cell proliferation that might lead to neovascularization and fibrosis. Recent data support Cer and Sph as crucial mediators in the induction of photoreceptor apoptosis in diverse models of oxidative damage and neurodegeneration, and suggest that regulating their metabolism can prevent this death. New evidence proposes a central role for S1P controlling photoreceptor survival and differentiation. Finally, this review discusses the ability of trophic factors to regulate sphingolipid metabolism and transactivate S1P signaling pathways to control survival and development in retina photoreceptors.
Collapse
Affiliation(s)
- Nora P Rotstein
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional del Sur-CONICET, Bahía Blanca, Buenos Aires, Argentina.
| | | | | | | |
Collapse
|
17
|
Joseph JA, Bielinski DF, Fisher DR. Blueberry treatment antagonizes C-2 ceramide-induced stress signaling in muscarinic receptor-transfected COS-7 cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2010; 58:3380-3392. [PMID: 20178393 DOI: 10.1021/jf9039155] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Previous research has shown that muscarinic receptors (MAChRs) show loss of sensitivity in aging and AD and are selectively sensitive to oxidative stress (OS). Thus, COS-7 cells transfected (tn) with MAChR subtype M1 show > OS sensitivity [as reflected in the ability of the cell to extrude or sequester Ca(2+) following depolarization (recovery) by oxotremorine (oxo) and exposure to dopamine (DA) or amyloid beta (Abeta)] than M3-transfected COS-7 cells. Blueberry (BB) extract pretreatment prevented these deficits. Research has also indicated that C2 ceramide (Cer) has several age-related negative cellular effects (e.g., OS). When these cells were treated with Cer, the significant decrements in the ability of both types of tn cells to initially respond to oxo were antagonized by BB treatment. Present experiments assessed signaling mechanisms involved in BB protection in the presence or absence of DA, Abeta, and/or Cer in this model. Thus, control or BB-treated M1 and M3 tn COS-7 cells were exposed to DA or Abeta(42) in the presence or absence of Cer. Primarily, results showed that the effects of DA or Abeta(42) were to increase stress (e.g., PKCgamma, p38MAPK) and protective signals (e.g., pMAPK). Cer also appeared to raise several of the stress and protective signals in the absence of the other stressors, including PKCgamma, pJNK, pNfkappaB, p53, and p38MAPK, while not significantly altering MAPK, or Akt. pArc was, however, increased by Cer in both types of transfected cells. The protective effects of BB when combined with Cer generally showed greater protection when BB extract was applied prior to Cer, except for one protective signal (pArc) where a greater effect was seen in the M3 cells exposed to Abeta(42.) In the absence of the Abeta(42) or DA, for several of the stress signals (e.g., pNfkappaB, p53), BB lowered their Cer-induced increases in M1- and M3-transfected cells. We are exploring these interactions further, but it is clear that increases in ceramide, to the same levels as are seen in aging, can have profound effects on calcium clearance and signaling during oxidative stress.
Collapse
Affiliation(s)
- James A Joseph
- USDA-HNRCA at Tufts University, Boston, Massachusetts 02111, USA.
| | | | | |
Collapse
|
18
|
Effects of Fat-Enriched Diet on the Content of Sphingolipids in the Brain and on Cognitive Functions in Old Rats. NEUROPHYSIOLOGY+ 2010. [DOI: 10.1007/s11062-010-9101-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
19
|
Cerantola V, Guillas I, Roubaty C, Vionnet C, Uldry D, Knudsen J, Conzelmann A. Aureobasidin A arrests growth of yeast cells through both ceramide intoxication and deprivation of essential inositolphosphorylceramides. Mol Microbiol 2009; 71:1523-37. [PMID: 19210614 DOI: 10.1111/j.1365-2958.2009.06628.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
All mature Saccharomyces cerevisiae sphingolipids comprise inositolphosphorylceramides containing C26:0 or C24:0 fatty acids and either phytosphingosine or dihydrosphingosine. Here we analysed the lipid profile of lag1Delta lac1Delta mutants lacking acyl-CoA-dependent ceramide synthesis, which require the reverse ceramidase activity of overexpressed Ydc1p for sphingolipid biosynthesis and viability. These cells, termed 2Delta.YDC1, make sphingolipids containing exclusively dihydrosphingosine and an abnormally wide spectrum of fatty acids with between 18 and 26 carbon atoms. Like wild-type cells, 2Delta.YDC1 cells stop growing when exposed to Aureobasidin A (AbA), an inhibitor of the inositolphosphorylceramide synthase AUR1, yet their ceramide levels remain very low. This finding argues against a current hypothesis saying that yeast cells do not require inositolphosphorylceramides and die in the presence of AbA only because ceramides build up to toxic concentrations. Moreover, W303lag1Delta lac1Delta ypc1Delta ydc1Delta cells, reported to be AbA resistant, stop growing on AbA after a certain number of cell divisions, most likely because AbA blocks the biosynthesis of anomalous inositolphosphorylsphingosides. Thus, data argue that inositolphosphorylceramides of yeast, the equivalent of mammalian sphingomyelins, are essential for growth. Data also clearly confirm that wild-type strains, when exposed to AbA, immediately stop growing because of ceramide intoxication, long before inositolphosphorylceramide levels become subcritical.
Collapse
Affiliation(s)
- Vanessa Cerantola
- Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
In this chapter, roles of bioactive sphingolipids in the regulation of cancer pathogenesis and therapy will be reviewed. Sphingolipids have emerged as bioeffector molecules, which control various aspects of cell growth, proliferation, and anti-cancer therapeutics. Ceramide, the central molecule of sphingolipid metabolism, generally mediates anti-proliferative responses such as inhibition of cell growth, induction of apoptosis, and/or modulation of senescence. On the other hand, sphingosine 1-phosphate (S1P) plays opposing roles, and induces transformation, cancer cell growth, or angiogenesis. A network of metabolic enzymes regulates the generation of ceramide and S1P, and these enzymes serve as transducers of sphingolipid-mediated responses that are coupled to various exogenous or endogenous cellular signals. Consistent with their key roles in the regulation of cancer growth and therapy, attenuation of ceramide generation and/or increased S1P levels are implicated in the development of resistance to drug-induced apoptosis, and escape from cell death. These data strongly suggest that advances in the molecular and biochemical understanding of sphingolipid metabolism and function will lead to the development of novel therapeutic strategies against human cancers, which may also help overcome drug resistance.
Collapse
|
21
|
Ceramide starves cells to death by downregulating nutrient transporter proteins. Proc Natl Acad Sci U S A 2008; 105:17402-7. [PMID: 18981422 DOI: 10.1073/pnas.0802781105] [Citation(s) in RCA: 160] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Ceramide induces cell death in response to many stimuli. Its mechanism of action, however, is not completely understood. Ceramide induces autophagy in mammalian cells maintained in rich media and nutrient permease downregulation in yeast. These observations suggested to us that ceramide might kill mammalian cells by limiting cellular access to extracellular nutrients. Consistent with this proposal, physiologically relevant concentrations of ceramide produced a profound and specific downregulation of nutrient transporter proteins in mammalian cells. Blocking ceramide-induced nutrient transporter loss or supplementation with the cell-permeable nutrient, methyl pyruvate, reversed ceramide-dependent toxicity. Conversely, cells became more sensitive to ceramide when nutrient stress was increased by acutely limiting extracellular nutrients, inhibiting autophagy, or deleting AMP-activated protein kinase (AMPK). Observations that ceramide can trigger either apoptosis or caspase-independent cell death may be explained by this model. We found that methyl pyruvate (MP) also protected cells from ceramide-induced, nonapoptotic death consistent with the idea that severe bioenergetic stress was responsible. Taken together, these studies suggest that the cellular metabolic state is an important arbiter of the cellular response to ceramide. In fact, increasing nutrient demand by incubating cells in high levels of growth factor sensitized cells to ceramide. On the other hand, gradually adapting cells to tolerate low levels of extracellular nutrients completely blocked ceramide-induced death. In sum, these results support a model where ceramide kills cells by inducing intracellular nutrient limitation subsequent to nutrient transporter downregulation.
Collapse
|
22
|
Ponnusamy S, Alderson NL, Hama H, Bielawski J, Jiang JC, Bhandari R, Snyder SH, Jazwinski SM, Ogretmen B. Regulation of telomere length by fatty acid elongase 3 in yeast. Involvement of inositol phosphate metabolism and Ku70/80 function. J Biol Chem 2008; 283:27514-27524. [PMID: 18694931 DOI: 10.1074/jbc.m802980200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
In this study, we investigated the roles of very long-chain fatty acid (VLCFA) synthesis by fatty acid elongase 3 (ELO3) in the regulation of telomere length and life span in the yeast Saccharomyces cerevisiae. Loss of VLCFA synthesis via deletion of ELO3 reduced telomere length, and reconstitution of the expression of wild type ELO3, and not by its mutant with decreased catalytic activity, rescued telomere attrition. Further experiments revealed that alterations of phytoceramide seem to be dispensable for telomere shortening in response to loss of ELO3. Interestingly, telomere shortening in elo3Delta cells was almost completely prevented by deletion of IPK2 or KCS1, which are involved in the generation of inositol phosphates (IP4, IP5, and inositol pyrophosphates). Deletion of IPK1, which generates IP6, however, did not affect regulation of telomere length. Further data also suggested that elo3Delta cells exhibit accelerated chronologic aging, and reduced replicative life span compared with wild type cells, and deletion of KCS1 helped recover these biological defects. Importantly, to determine downstream mechanisms, epistasis experiments were performed, and data indicated that ELO3 and YKU70/80 share a common pathway for the regulation of telomere length. More specifically, chromatin immunoprecipitation assays revealed that the telomere binding and protective function of YKu80p in vivo was reduced in elo3Delta cells, whereas its non-homologues end-joining function was not altered. Deletion of KCS1 in elo3Delta cells recovered the telomere binding and protective function of Ku, consistent with the role of KCS1 mutation in the rescue of telomere length attrition. Thus, these findings provide initial evidence of a possible link between Elo3-dependent VLCFA synthesis, and IP metabolism by KCS1 and IPK2 in the regulation of telomeres, which play important physiological roles in the control of senescence and aging, via a mechanism involving alterations of the telomere-binding/protection function of Ku.
Collapse
Affiliation(s)
- Suriyan Ponnusamy
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425; Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Nathan L Alderson
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Hiroko Hama
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Jacek Bielawski
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425; Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - James C Jiang
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana 70118
| | - Rashna Bhandari
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Solomon H Snyder
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - S Michal Jazwinski
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana 70118
| | - Besim Ogretmen
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425; Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425.
| |
Collapse
|
23
|
Yabu T, Imamura S, Yamashita M, Okazaki T. Identification of Mg2+ -dependent neutral sphingomyelinase 1 as a mediator of heat stress-induced ceramide generation and apoptosis. J Biol Chem 2008; 283:29971-82. [PMID: 18678863 DOI: 10.1074/jbc.m805402200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Neutral sphingomyelinases (SMases) are involved in the induction of ceramide-mediated proapoptotic signaling under heat stress conditions. Although ceramide is an important mediator of apoptosis, the neutral SMase that is activated under heat stress has not been identified. In this study, we cloned an Mg(2+)-dependent neutral SMase from a zebrafish embryonic cell cDNA library using an Escherichia coli expression-cloning vector. Screening of the clones using an SMase activity assay with C(6)-7-nitro-2-1,3-benzoxadiazol-4-yl-sphingomyelin as the substrate resulted in the isolation of one neutral SMase cDNA clone. This cDNA encoded a polypeptide of 420 amino acids (putative molecular weight: 46,900) containing two predicted transmembrane domains in its C-terminal region. The cloned neutral SMase 1 acted as a mediator of stress-induced apoptosis. Bacterially expressed recombinant neutral SMase 1 hydrolyzed [choline-methyl-(14)C]sphingomyelin optimally at pH 7.5 in the presence of an Mg(2+) ion. In zebrafish embryonic cells, the endogenous SMase enzyme was localized in the microsomal fraction. In FLAG-tagged SMase-overexpressing cells, neutral SMase 1 colocalized with a Golgi marker in a cytochemical analysis. Inactivation of the enzyme by an antisense phosphorothioate oligonucleotide repressed the induction of ceramide generation, caspase-3 activation, and apoptotic cell death by heat stress. Thus, neutral SMase 1 participates in an inducible ceramide-mediating, proapoptotic signaling pathway that operates in heat-induced apoptosis in zebrafish embryonic cells.
Collapse
Affiliation(s)
- Takeshi Yabu
- National Research Institute of Fisheries Science, 2-12-4 Fukuura, Kanazawa, Yokohama, Kanagawa 236-8648, Japan.
| | | | | | | |
Collapse
|
24
|
Aronova S, Wedaman K, Aronov PA, Fontes K, Ramos K, Hammock BD, Powers T. Regulation of ceramide biosynthesis by TOR complex 2. Cell Metab 2008; 7:148-58. [PMID: 18249174 PMCID: PMC3882310 DOI: 10.1016/j.cmet.2007.11.015] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2007] [Revised: 11/02/2007] [Accepted: 11/30/2007] [Indexed: 01/08/2023]
Abstract
Ceramides and sphingoid long-chain bases (LCBs) are precursors to more complex sphingolipids and play distinct signaling roles crucial for cell growth and survival. Conserved reactions within the sphingolipid biosynthetic pathway are responsible for the formation of these intermediates. Components of target of rapamycin complex 2 (TORC2) have been implicated in the biosynthesis of sphingolipids in S. cerevisiae; however, the precise step regulated by this complex remains unknown. Here we demonstrate that yeast cells deficient in TORC2 activity are impaired for de novo ceramide biosynthesis both in vivo and in vitro. We find that TORC2 regulates this step in part by activating the AGC kinase Ypk2 and that this step is antagonized by the Ca2+/calmodulin-dependent phosphatase calcineurin. Because Ypk2 is activated independently by LCBs, the direct precursors to ceramides, our data suggest a model wherein TORC2 signaling is coupled with LCB levels to control Ypk2 activity and, ultimately, regulate ceramide formation.
Collapse
Affiliation(s)
- Sofia Aronova
- Section of Molecular and Cellular Biology, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Progress in aging research is now rapid, and surprisingly, studies in a single-celled eukaryote are a driving force. The genetic modulators of replicative life span in yeast are being identified, the molecular events that accompany aging are being discovered, and the extent to which longevity pathways are conserved between yeast and multicellular eukaryotes is being tested. In this review, we provide a brief retrospective view on the development of yeast as a model for aging and then turn to recent discoveries that have pushed aging research into novel directions and also linked aging in yeast to well-developed hypotheses in mammals. Although the question of what causes aging still cannot be answered definitively, that day may be rapidly approaching.
Collapse
Affiliation(s)
- K A Steinkraus
- Department of Pathology, University of Washington, Seattle, Washington 98195, USA.
| | | | | |
Collapse
|
26
|
Senkal CE, Ponnusamy S, Rossi MJ, Bialewski J, Sinha D, Jiang JC, Jazwinski SM, Hannun YA, Ogretmen B. Role of human longevity assurance gene 1 and C18-ceramide in chemotherapy-induced cell death in human head and neck squamous cell carcinomas. Mol Cancer Ther 2007; 6:712-22. [PMID: 17308067 DOI: 10.1158/1535-7163.mct-06-0558] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this study, quantitative isobologram studies showed that treatment with gemcitabine and doxorubicin, known inducers of ceramide generation, in combination, supra-additively inhibited the growth of human UM-SCC-22A cells in situ. Then, possible involvement of the human homologue of yeast longevity assurance gene 1 (LASS1)/C(18)-ceramide in chemotherapy-induced cell death in these cells was examined. Gemcitabine/doxorubicin combination treatment resulted in the elevation of mRNA and protein levels of LASS1 and not LASS2-6, which was consistent with a 3.5-fold increase in the endogenous (dihydro)ceramide synthase activity of LASS1 for the generation of C(18)-ceramide. Importantly, the overexpression of LASS1 (both human and mouse homologues) enhanced the growth-inhibitory effects of gemcitabine/doxorubicin with a concomitant induction of caspase-3 activation. In reciprocal experiments, partial inhibition of human LASS1 expression using small interfering RNA (siRNA) prevented cell death by about 50% in response to gemcitabine/doxorubicin. In addition, LASS1, and not LASS5, siRNA modulated the activation of caspase-3 and caspase-9, but not caspase-8, in response to this combination. Treatment with gemcitabine/doxorubicin in combination also resulted in a significant suppression of the head and neck squamous cell carcinoma (HNSCC) tumor growth in severe combined immunodeficiency mice bearing the UM-SCC-22A xenografts. More interestingly, analysis of endogenous ceramide levels in these tumors by liquid chromatography/mass spectroscopy showed that only the levels of C(18)-ceramide, the main product of LASS1, were elevated significantly (about 7-fold) in response to gemcitabine/doxorubicin when compared with controls. In conclusion, these data suggest an important role for LASS1/C(18)-ceramide in gemcitabine/doxorubicin-induced cell death via the activation of caspase-9/3 in HNSCC.
Collapse
Affiliation(s)
- Can E Senkal
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Aenlle KK, Kumar A, Cui L, Jackson TC, Foster TC. Estrogen effects on cognition and hippocampal transcription in middle-aged mice. Neurobiol Aging 2007; 30:932-45. [PMID: 17950954 DOI: 10.1016/j.neurobiolaging.2007.09.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2007] [Revised: 09/06/2007] [Accepted: 09/12/2007] [Indexed: 10/22/2022]
Abstract
Young and middle-aged female mice were ovariectomized and given cyclic injections of either estradiol or vehicle treatments. During the fifth week after surgery the Morris water maze was used to assess cognitive function. Age and treatment effects emerged over the course of spatial training such that middle-aged vehicle treated mice exhibited deficits in acquiring a spatial search strategy compared to younger vehicle treated mice and middle-age estradiol treated mice. Following behavioral characterization, mice were maintained on their injection schedule until week seven and hippocampi were collected 24h after the last injection. Hippocampal RNA was extracted and genes responsive to age and estrogen were identified using cDNA microarrays. Estradiol treatment in middle-aged mice altered the expression of genes related to transcriptional regulation, biosynthesis, growth, neuroprotection, and elements of cell signaling pathways. Expression profiles for representative genes were confirmed in a separate set of animals using oligonucleotide arrays and RT-PCR. Our results indicate that estrogen treatment in middle-aged animals may promote hippocampal health during the aging process.
Collapse
Affiliation(s)
- Kristina K Aenlle
- Department of Neuroscience, McKnight Brain Institute, University of Florida, P.O. Box 100244, Gainesville, FL 32610-0244, USA
| | | | | | | | | |
Collapse
|
28
|
Rutkute K, Karakashian AA, Giltiay NV, Dobierzewska A, Nikolova-Karakashian MN. Aging in rat causes hepatic hyperresposiveness to interleukin-1beta which is mediated by neutral sphingomyelinase-2. Hepatology 2007; 46:1166-76. [PMID: 17668873 DOI: 10.1002/hep.21777] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
UNLABELLED The process of aging has recently been shown to substantially affect the ability of cells to respond to inflammatory challenges. We demonstrate that aging leads to hepatic hyperresponsiveness to interleukin 1beta (IL-1beta), and we examine the factors that could be responsible for this phenomenon. IL-1beta-induced phosphorylation of c-jun N-terminal kinase (JNK) in hepatocytes isolated from aged rats was 3 times more potent than that in hepatocytes from young rats. Moreover, JNK was activated by substantially lower doses of IL-1beta. These age-related changes in JNK phosphorylation correlated with diminished IL-1beta-induced degradation of interleukin-1 receptor-associated kinase-1 (IRAK-1). Expression levels of IL1beta receptor I, total JNK, IRAK-1, and transforming growth factor-beta-activated kinase-1 (TAK-1) were not affected by aging. However, increased neutral sphingomyelinase activity was observed in hepatocytes from old animals, which we show is caused by induction of the plasma membrane localized neutral sphingomyelinase-2 (NSMase-2). We provide evidence that NSMase-2 is both required and sufficient for the onset of IL-1beta hyperresponsiveness during aging. Overexpression of NSMase-2 in hepatocytes from young rats leads both to a reduction in IRAK-1 degradation and potentiation of JNK phosphorylation, mimicking that seen in hepatocytes from old animals. More importantly, inhibition of NSMase activity in hepatocytes from aged rats using either scyphostatin or short interfering ribonucleic acid (siRNA) leads to reversion to the "young" phenotype of IL-1beta response. CONCLUSION These results show that the process of aging causes increased basal NSMase-2 activity in hepatocytes, which in turn leads to IRAK-1 stabilization, JNK potentiation, and ultimately IL-1beta hyperresponsiveness.
Collapse
Affiliation(s)
- Kristina Rutkute
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | | | | | | | | |
Collapse
|
29
|
Yoshida T, Tuder RM. Pathobiology of cigarette smoke-induced chronic obstructive pulmonary disease. Physiol Rev 2007; 87:1047-82. [PMID: 17615396 DOI: 10.1152/physrev.00048.2006] [Citation(s) in RCA: 375] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary diseases (COPD), comprised of pulmonary emphysema, chronic bronchitis, and structural and inflammatory changes of small airways, is a leading cause of morbidity and mortality in the world. A better understanding of the pathobiology of COPD is critical for the developing of novel therapies, as the majority of patients with the disease have little therapeutic options at the present time. The pathobiology of COPD encompasses multiple injurious processes including inflammation (excessive or inappropriate innate and adaptive immunity), cellular apoptosis, altered cellular and molecular alveolar maintenance program, abnormal cell repair, extracellular matrix destruction (protease and anti-protease imbalance), and oxidative stress (oxidant and antioxidant imbalance). These processes are triggered by urban and rural air pollutants and active and/or passive cigarette smoke and modified by cellular senescence and infection. A series of receptor-mediated signal transduction pathways are activated by reactive oxygen species and tobacco components, resulting in impairment of a variety of cell signaling and cytokine networks, subsequently leading to chronic airway responses with mucus production, airway remodeling, and alveolar destruction. The authors provide an updated insight into the molecular and cellular pathobiology of COPD based on human and/or animal data.
Collapse
Affiliation(s)
- Toshinori Yoshida
- Division of Cardiopulmonary Pathology, Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
30
|
Wang B, Shi G, Fu Y, Xu X. Cloning and characterization of a LASS1-GDF1 transcript in rat cerebral cortex: conservation of a bicistronic structure. DNA SEQUENCE : THE JOURNAL OF DNA SEQUENCING AND MAPPING 2007; 18:92-103. [PMID: 17364820 DOI: 10.1080/10425170601060947] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
LASS1 is the mammalian homologue of yeast longevity-assurance gene 1 (LAG1) which is differentially expressed during the yeast replicative life span and was shown to play a role in determining yeast longevity. Growth/differentiation factor 1 (GDF1) is a transforming growth factor-beta family member that was originally isolated from an mouse embryo cDNA library. GDF1 is expressed specifically in the nervous system and functions in left-right patterning of the mouse embryo. To explore the potential role of LASS1 in rat neuron aging, northern blot analysis for LASS1 mRNA was performed and detected a 2.7 kb transcript in adult rat cerebral cortex. Cloning and sequence analysis revealed that this transcript contains two nonoverlapping open reading frames (ORFs), LASS1 and GDF1, which are quite different to the predicted sequences in GenBank (accession number XM_224734 and XM_224733). The alignment with the rat genome database revealed this transcript matches the sequence of rat chromosome 16 genomic contig (GenBank accession number NW_047470) between nucleotide positions 1935060th and 1919439th, which contains eight exons and seven introns. Multiple sequence analysis revealed high conservation of the two ORFs. The phylogenetic analysis showed very close evolutionary relationship among rat, mouse and human. It raises the possibility that this mRNA may give rise to two different proteins and the conserved bicistronic structure might be of unknown significance.
Collapse
Affiliation(s)
- Baoheng Wang
- Laboratory of Cell Senescence, Shantou University Medical College, Shantou, Guangdong Province, 515041, People's Republic of China
| | | | | | | |
Collapse
|
31
|
Tuder RM, Yoshida T, Arap W, Pasqualini R, Petrache I. State of the art. Cellular and molecular mechanisms of alveolar destruction in emphysema: an evolutionary perspective. Ann Am Thorac Soc 2006; 3:503-10. [PMID: 16921129 PMCID: PMC2647641 DOI: 10.1513/pats.200603-054ms] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Emphysema consists of a unique pattern of alveolar destruction, resulting in marked airspace enlargement with reduction of alveolar capillary exchange area. Classical concepts of the pathogenesis of emphysema have relied on the paradigm set by the inflammation and protease/antiprotease imbalance. We propose herein that cigarette smoke constitutes an environmental hazard that causes alveolar destruction by the interaction of apoptosis, oxidative stress, and protease/antiprotease imbalance. We draw a parallel between organismal aging, organ structural maintenance, and the damage resulting from chronic cigarette smoke inhalation. The stochastic interaction between environmental hazards and the effort of an organism or a particular organ to fend off these hazards results in the accumulation of cellular damage and features characteristic of aging. Inflammation follows as the result of the multiplication of injuries. We highlight the importance of understanding the biology of the interaction of alveolar cells in homeostasis and in alveolar destruction, and the potential role of novel processes related to senescence and stress response. An evolutionary perspective of emphysema that incorporates mechanisms related to aging may lead to important advances in the understanding and therapeutic targeting of chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Rubin M Tuder
- Department of Pathology, Division of Cardiopulmonary Pathology, Ross Research Building, Ross 519, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, USA.
| | | | | | | | | |
Collapse
|
32
|
Ogretmen B. Sphingolipids in cancer: Regulation of pathogenesis and therapy. FEBS Lett 2006; 580:5467-76. [PMID: 16970943 DOI: 10.1016/j.febslet.2006.08.052] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2006] [Revised: 08/10/2006] [Accepted: 08/14/2006] [Indexed: 10/24/2022]
Abstract
Sphingolipids are known to play important roles in the regulation of cell proliferation, response to chemotherapeutic agents, and/or prevention of cancer. Recently, significant progress has been made in the identification of the enzymes and their biochemical functions involved in sphingolipid metabolism. In addition, development of new techniques for the quantitative analysis of sphingolipids at their physiological levels has facilitated studies to examine distinct functions of these bioactive sphingolipids in cancer pathogenesis and therapy. This review will focus on the recent developments regarding the roles of bioactive sphingolipids in the regulation of cell growth/proliferation, and anti-cancer therapeutics.
Collapse
Affiliation(s)
- Besim Ogretmen
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, 173 Ashley Avenue, Charleston, 29425, USA.
| |
Collapse
|
33
|
Specific protein kinase C isoforms as transducers and modulators of insulin signaling. Mol Genet Metab 2006; 89:32-47. [PMID: 16798038 DOI: 10.1016/j.ymgme.2006.04.017] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2006] [Revised: 04/23/2006] [Accepted: 04/23/2006] [Indexed: 12/14/2022]
Abstract
Recent studies implicate specific PKC isoforms in the insulin-signaling cascade. Insulin activates PKCs alpha, betaII, delta and zeta in several cell types. In addition, as will be documented in this review, certain members of the PKC family may also be activated and act upstream of PI3 and MAP kinases. Each of these isoforms has been shown one way or another either to mimic or to modify insulin-stimulated effects in one or all of the insulin-responsive tissues. Moreover, each of the isoforms has been shown to be activated by insulin stimulation or conditions important for effective insulin stimulation. Studies attempting to demonstrate a definitive role for any of the isoforms have been performed on different cells, ranging from appropriate model systems for skeletal muscle, liver and fat, such as primary cultures, and cell lines and even in vivo studies, including transgenic mice with selective deletion of specific PKC isoforms. In addition, studies have been done on certain expression systems such as CHO or HEK293 cells, which are far removed from the tissues themselves and serve mainly as vessels for potential protein-protein interactions. Thus, a clear picture for many of the isoforms remains elusive in spite of over two decades of intensive research. The recent intrusion of transgenic and precise molecular biology technologies into the research armamentarium has opened a wide range of additional possibilities for direct involvement of individual isoforms in the insulin signaling cascade. As we hope to discuss within the context of this review, whereas many of the long sought-after answers to specific questions are not yet clear, major advances have been made in our understanding of precise roles for individual PKC isoforms in mediation of insulin effects. In this review, in which we shall focus our attention on isoforms in the conventional and novel categories, a clear case will be made to show that these isoforms are not only expressed but are importantly involved in regulation of insulin metabolic effects.
Collapse
|
34
|
Abstract
The brain is enriched with sphingolipids, which are important membrane constituents and major lipid signaling molecules that have a role in motor and cognitive behavior. Vitamin K has been implicated in brain sphingolipid metabolism for more than 30 years. The in vitro and in vivo studies to date suggest a role of vitamin K in the regulation of multiple enzymes involved in sphingolipid metabolism within the myelin-rich regions in the brain. However, the precise mechanisms of action are not well understood. Further, the physiological consequences of the observed effects of vitamin K on sphingolipid metabolism have not been systematically studied.
Collapse
Affiliation(s)
- Natalia A Denisova
- Nutrition and Neurocognition Laboratory, Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts 02111, USA.
| | | |
Collapse
|
35
|
Persaud-Sawin DA, McNamara JO, Rylova S, Vandongen A, Boustany RMN. A galactosylceramide binding domain is involved in trafficking of CLN3 from Golgi to rafts via recycling endosomes. Pediatr Res 2004; 56:449-63. [PMID: 15240864 DOI: 10.1203/01.pdr.0000136152.54638.95] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Juvenile neuronal ceroid lipofuscinosis (JNCL) is due to mutations in the CLN3 gene. We previously determined that CLN3 protein harbors a highly conserved motif, VYFAE, necessary for its impact on cell growth and apoptosis. Using molecular modeling we demonstrated that this motif is embedded in a stretch of amino acids that is homologous to and structurally compatible with a galactosylceramide (GalCer) binding domain. This domain is present in the V3 loop of the HIV-1 gp120 envelope protein, beta-amyloid protein, and the infectious form of prionic protein, and defines a binding site for lipid rafts. We determined the subcellular localization of CLN3 in different cell systems including human neurons, primary rat hippocampal neurons, normal human fibroblasts, and JNCL fibroblasts homozygous for the 1.02 kb deletion in genomic DNA. Wild-type CLN3 protein was present within Golgi, lipid rafts in the plasma membrane, and early recycling endosomes, but not late endosomes/lysosomes. Wild-type CLN3 internalized from the plasma membrane to the Golgi via Rab4- and Rab11-positive recycling endosomes. Wild-type CLN3 co-localized with GalCer in the Golgi and in lipid rafts at the plasma membrane in normal cells. Neither mutant CLN3 protein nor GalCer were found at the plasma membrane in JNCL fibroblasts. Mutant CLN3p was retained within the Golgi and partially mis-localized to lysosomes, failing to reach recycling endosomes, plasma membrane, or lipid rafts. These studies identify a novel CLN3 domain that may dictate localization and function of CLN3.
Collapse
|
36
|
Affiliation(s)
- Besim Ogretmen
- Department of Biochemistry and Molecular Biology, and Hollings Cancer Center, 173 Ashley Avenue, Charleston, South Carolina 29425, USA
| | | |
Collapse
|