1
|
Lodde V, Zarbo IR, Farina G, Masia A, Solla P, Campesi I, Delogu G, Muroni MR, Tsitsipatis D, Gorospe M, Floris M, Idda ML. Identification of hsa_circ_0018905 as a New Potential Biomarker for Multiple Sclerosis. Cells 2024; 13:1668. [PMID: 39404430 PMCID: PMC11475351 DOI: 10.3390/cells13191668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 09/27/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Multiple sclerosis (MS) is a demyelinating autoimmune disease characterized by early onset, for which the interaction of genetic and environmental factors is crucial. Dysregulation of the immune system as well as myelinization-de-myelinization has been shown to correlate with changes in RNA, including non-coding RNAs. Recently, circular RNAs (circRNAs) have emerged as a key player in the complex network of gene dysregulation associated with MS. Despite several efforts, the mechanisms driving circRNA regulation and dysregulation in MS still need to be properly elucidated. Here, we explore the panorama of circRNA expression in PBMCs purified from five newly diagnosed MS patients and five healthy controls (HCs) using the Arraystar Human circRNAs microarray. Experimental validation was then carried out in a validation cohort, and a possible correlation with disease severity was tested. We identified 64 differentially expressed circRNAs, 53 of which were downregulated in PBMCs purified from MS compared to the HCs. The discovery dataset was subsequently validated using qRT-PCR with an independent cohort of 20 RRMS patients and 20 HCs. We validated seven circRNAs differentially expressed in the RRMS group versus the HC group. hsa_circ_0000518, hsa_circ_0000517, hsa_circ_0000514, and hsa_circ_0000511 were significantly upregulated in the MS group, while hsa_circ_0018905, hsa_circ_0048764, and hsa_circ_0003445 were significantly downregulated; Among them, the expression level of hsa_circ_0018905 was significantly decreased in patients showing a higher level of disability and in progressive forms of MS. We described the circRNAs expression profile of PBMCs in newly diagnosed MS patients and proposed hsa_circ_0018905 as potential MS biomarker.
Collapse
Affiliation(s)
- Valeria Lodde
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy; (V.L.); (I.C.); (G.D.); (M.F.)
| | - Ignazio Roberto Zarbo
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari 07100, Italy; (I.R.Z.); (A.M.); (P.S.); (M.R.M.)
- Unit of Clinical Neurology, AOU, Sassari 07100, Italy;
| | | | - Aurora Masia
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari 07100, Italy; (I.R.Z.); (A.M.); (P.S.); (M.R.M.)
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari 09042, Italy
| | - Paolo Solla
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari 07100, Italy; (I.R.Z.); (A.M.); (P.S.); (M.R.M.)
- Unit of Clinical Neurology, AOU, Sassari 07100, Italy;
| | - Ilaria Campesi
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy; (V.L.); (I.C.); (G.D.); (M.F.)
| | - Giuseppe Delogu
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy; (V.L.); (I.C.); (G.D.); (M.F.)
| | - Maria Rosaria Muroni
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari 07100, Italy; (I.R.Z.); (A.M.); (P.S.); (M.R.M.)
| | - Dimitrios Tsitsipatis
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA; (D.T.); (M.G.)
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA; (D.T.); (M.G.)
| | - Matteo Floris
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy; (V.L.); (I.C.); (G.D.); (M.F.)
| | - Maria Laura Idda
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy; (V.L.); (I.C.); (G.D.); (M.F.)
| |
Collapse
|
2
|
Weber-Stout M, Nicholson RJ, Dumaguit CDC, Holland WL, Summers SA. Ceramide microdomains: the major influencers of the sphingolipid media platform. Biochem Soc Trans 2024; 52:1765-1776. [PMID: 39082976 PMCID: PMC11845337 DOI: 10.1042/bst20231395] [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] [Indexed: 08/29/2024]
Abstract
Like 'influencers' who achieve fame and power through social media, ceramides are low abundance members of communication platforms that have a mighty impact on their surroundings. Ceramide microdomains form within sphingolipid-laden lipid rafts that confer detergent resistance to cell membranes and serve as important signaling hubs. In cells exposed to excessive amounts of saturated fatty acids (e.g. in obesity), the abundance of ceramide-rich microdomains within these rafts increases, leading to concomitant alterations in cellular metabolism and survival that contribute to cardiometabolic disease. In this mini-review, we discuss the evidence supporting the formation of these ceramide microdomains and describe the spectrum of harmful ceramide-driven metabolic actions under the context of an evolutionary theory. Moreover, we discuss the proximal 'followers' of these ceramide media stars that account for the diverse intracellular actions that allow them to influence obesity-linked disease.
Collapse
Affiliation(s)
- Mariah Weber-Stout
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, U.S.A
| | - Rebekah J Nicholson
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, U.S.A
| | - Carlos Dave C Dumaguit
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, U.S.A
| | - William L Holland
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, U.S.A
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, U.S.A
| |
Collapse
|
3
|
Dingjan T, Futerman AH. Fine-tuned protein-lipid interactions in biological membranes: exploration and implications of the ORMDL-ceramide negative feedback loop in the endoplasmic reticulum. Front Cell Dev Biol 2024; 12:1457209. [PMID: 39170919 PMCID: PMC11335536 DOI: 10.3389/fcell.2024.1457209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Biological membranes consist of a lipid bilayer in which integral membrane proteins are embedded. Based on the compositional complexity of the lipid species found in membranes, and on their specific and selective interactions with membrane proteins, we recently suggested that membrane bilayers can be best described as "finely-tuned molecular machines." We now discuss one such set of lipid-protein interactions by describing a negative feedback mechanism operating in the de novo sphingolipid biosynthetic pathway, which occurs in the membrane of the endoplasmic reticulum, and describe the atomic interactions between the first enzyme in the pathway, namely serine palmitoyl transferase, and the product of the fourth enzyme in the pathway, ceramide. We explore how hydrogen-bonding and hydrophobic interactions formed between Asn13 and Phe63 in the serine palmitoyl transferase complex and ceramide can influence the ceramide content of the endoplasmic reticulum. This example of finely-tuned biochemical interactions raises intriguing mechanistic questions about how sphingolipids and their biosynthetic enzymes could have evolved, particularly in light of their metabolic co-dependence.
Collapse
Affiliation(s)
- Tamir Dingjan
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
4
|
Chen X, Li J, Gao Z, Yang Y, Kuang W, Dong Y, Chua GH, Huang X, Jiang B, Tian H, Wang Y, Huang X, Li Y, Lam SM, Shui G. Endogenous ceramide phosphoethanolamine modulates circadian rhythm via neural-glial coupling in Drosophila. Natl Sci Rev 2022; 9:nwac148. [PMID: 36713590 PMCID: PMC9875363 DOI: 10.1093/nsr/nwac148] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 06/08/2022] [Accepted: 07/13/2022] [Indexed: 02/01/2023] Open
Abstract
While endogenous lipids are known to exhibit rhythmic oscillations, less is known about how specific lipids modulate circadian behavior. Through a series of loss-of-function and gain-of-function experiments on ceramide phosphoethanolamine (CPE) synthase of Drosophila, we demonstrated that pan-glial-specific deficiency in membrane CPE, the structural analog of mammalian sphingomyelin (SM), leads to arrhythmic locomotor behavior and shortens lifespan, while the reverse is true for increasing CPE. Comparative proteomics uncovered dysregulated synaptic glutamate utilization and transport in CPE-deficient flies. An extensive genetic screen was conducted to verify the role of differentially expressed proteins in circadian regulation. Arrhythmic locomotion under cpes1 mutant background was rescued only by restoring endogenous CPE or SM through expressing their respective synthases. Our results underscore the essential role of CPE in maintaining synaptic glutamate homeostasis and modulating circadian behavior in Drosophila. The findings suggest that region-specific elevations of functional membrane lipids can benefit circadian regulation.
Collapse
Affiliation(s)
| | | | - Zhongbao Gao
- University of Chinese Academy of Sciences, Beijing 100049, China,State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yang Yang
- University of Chinese Academy of Sciences, Beijing 100049, China,State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Wenqing Kuang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing100101, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yue Dong
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing100101, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gek Huey Chua
- LipidALL Technologies Company Limited, Changzhou213022, China
| | - Xiahe Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing100101, China
| | - Binhua Jiang
- LipidALL Technologies Company Limited, Changzhou213022, China
| | - He Tian
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing100101, China
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing100101, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xun Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing100101, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Li
- University of Chinese Academy of Sciences, Beijing 100049, China,State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | | | | |
Collapse
|
5
|
Berton MP, de Lemos MVA, Stafuzza NB, Simielli Fonseca LF, Silva DBDS, Peripolli E, Pereira ASC, Magalhães AFB, Albuquerque LG, Baldi F. Integration analyses of structural variations and differential gene expression associated with beef fatty acid profile in Nellore cattle. Anim Genet 2022; 53:570-582. [PMID: 35811456 DOI: 10.1111/age.13242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 06/06/2022] [Accepted: 06/22/2022] [Indexed: 11/26/2022]
Abstract
This study aimed to integrate analyses of structural variations and differentially expressed genes (DEGs) associated with the beef fatty acid (FA) profile in Nellore cattle. Copy numbers variation (CNV) detection was performed using the penncnv algorithm and CNVRuler software in 3794 genotyped animals through the High-Density Bovine BeadChip. In order to perform the genomic wide association study (GWAS), a total of 963 genotyped animals were selected to obtain the intramuscular lipid concentration and quantify the beef FA profile. A total of 48 animals belonging to the same farm and management lot were extracted from the 963 genotyped and phenotyped animals to carry out the transcriptomic and differentially expressed gene analyses. The GWAS with extreme groups of FA profiles was performed using a logistic model. A total of 43, 42, 66 and 35 significant CNV regions (p < 0.05) for saturated, monounsaturated, polyunsaturated and omega 3 and 6 fatty acids were identified respectively. The paired-end sequencing of 48 samples was performed using the Illumina HiSeq2500 platform. Real-time quantitative PCR was used to validate the DEGs identified by RNA-seq analysis. The results showed several DEGs associated with the FA profile of Longissimus thoracis, such as BSCL2 and SAMD8. Enriched terms as the cellular response to corticosteroid (GO:0071384) and glucocorticoid stimulus (GO:0071385) could be highlighted. The identification of structural variations harboring candidate genes for beef FA must contribute to the elucidation of the genetic basis that determines the beef FA composition of intramuscular fat in Nellore cattle. Our results will contribute to the identification of potential biomarkers for complex phenotypes, such as the FA profile, to improve the reliability of the genomic predictions including pre-selected variants using differentiated weighting in the genomic models.
Collapse
Affiliation(s)
- Mariana Piatto Berton
- Departamento de Zootecnia, Universidade Estadual Paulista, Faculdade de Ciências Agrárias e Veterinárias, Jaboticabal, Brazil
| | | | | | | | | | - Elisa Peripolli
- Departamento de Zootecnia, Universidade Estadual Paulista, Faculdade de Ciências Agrárias e Veterinárias, Jaboticabal, Brazil
| | - Angélica S C Pereira
- Departamento de Nutrição e Produção Animal, Universidade de São Paulo, Faculdade de Medicina Veterinária e Zootecnia, Pirassununga, Brazil
| | - Ana Fabricia Braga Magalhães
- Departamento de Zootecnia, Universidade Estadual Paulista, Faculdade de Ciências Agrárias e Veterinárias, Jaboticabal, Brazil
| | - Lucia G Albuquerque
- Departamento de Zootecnia, Universidade Estadual Paulista, Faculdade de Ciências Agrárias e Veterinárias, Jaboticabal, Brazil
| | - Fernando Baldi
- Departamento de Zootecnia, Universidade Estadual Paulista, Faculdade de Ciências Agrárias e Veterinárias, Jaboticabal, Brazil
| |
Collapse
|
6
|
Shimasaki K, Kumagai K, Sakai S, Yamaji T, Hanada K. Hyperosmotic Stress Induces Phosphorylation of CERT and Enhances Its Tethering throughout the Endoplasmic Reticulum. Int J Mol Sci 2022; 23:ijms23074025. [PMID: 35409383 PMCID: PMC8999913 DOI: 10.3390/ijms23074025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 12/04/2022] Open
Abstract
The ceramide transport protein (CERT) delivers ceramide from the endoplasmic reticulum (ER) to the Golgi apparatus, where ceramide is converted to sphingomyelin (SM). The function of CERT is regulated in two distinct phosphorylation-dependent events: multiple phosphorylations in a serine-repeat motif (SRM) and phosphorylation of serine 315 residue (S315). Pharmacological inhibition of SM biosynthesis results in an increase in SRM-dephosphorylated CERT, which serves as an activated form, and an enhanced phosphorylation of S315, which augments the binding of CERT to ER-resident VAMP-associated protein (VAP), inducing the full activation of CERT to operate at the ER–Golgi membrane contact sites (MCSs). However, it remains unclear whether the two phosphorylation-dependent regulatory events always occur coordinately. Here, we describe that hyperosmotic stress induces S315 phosphorylation without affecting the SRM-phosphorylation state. Under hyperosmotic conditions, the binding of CERT with VAP-A is enhanced in an S315 phosphorylation-dependent manner, and this increased binding occurs throughout the ER rather than restrictedly at the ER–Golgi MCSs. Moreover, we found that de novo synthesis of SM with very-long acyl chains preferentially increases via a CERT-independent mechanism under hyperosmotic-stressed cells, providing an insight into a CERT-independent ceramide transport pathway for de novo synthesis of SM.
Collapse
Affiliation(s)
- Kentaro Shimasaki
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan; (K.S.); (S.S.); (T.Y.)
| | - Keigo Kumagai
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan; (K.S.); (S.S.); (T.Y.)
- Correspondence: (K.K.); (K.H.)
| | - Shota Sakai
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan; (K.S.); (S.S.); (T.Y.)
| | - Toshiyuki Yamaji
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan; (K.S.); (S.S.); (T.Y.)
| | - Kentaro Hanada
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan; (K.S.); (S.S.); (T.Y.)
- Department of Quality Assurance, Radiation Safety and Information System, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
- Correspondence: (K.K.); (K.H.)
| |
Collapse
|
7
|
Tippetts TS, Holland WL, Summers SA. Cholesterol - the devil you know; ceramide - the devil you don't. Trends Pharmacol Sci 2021; 42:1082-1095. [PMID: 34750017 PMCID: PMC8595778 DOI: 10.1016/j.tips.2021.10.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 01/20/2023]
Abstract
Ectopic lipids play a key role in numerous pathologies, including heart disease, stroke, and diabetes. Of all the lipids studied, perhaps the most well understood is cholesterol, a widely used clinical biomarker of cardiovascular disease and a target of pharmacological interventions (e.g., statins). Thousands of studies have interrogated the regulation and action of this disease-causing sterol. As a growing body of literature indicates, a new class of lipid-based therapies may be on the horizon. Ceramides are cholesterol-independent biomarkers of heart disease and diabetes in humans. Studies in rodents suggest that they are causative agents of disease, as lowering ceramides through genetic or pharmacological interventions prevents cardiovascular disease and diabetes. Herein, we discuss the evidence supporting the potential of therapeutics targeting ceramides to treat cardiometabolic disease, contrasting it with the robust datasets that drove the creation of cholesterol-lowering pharmaceuticals.
Collapse
Affiliation(s)
| | | | - Scott A. Summers
- Correspondence should be addressed to: Scott A. Summers, Department of Nutrition and Integrative Physiology, University of Utah College of Health, 15N, 2030 East, Rm 3110, Salt Lake City Utah 84112, , Tel: 801-585-9359
| |
Collapse
|
8
|
Arsenault EJ, McGill CM, Barth BM. Sphingolipids as Regulators of Neuro-Inflammation and NADPH Oxidase 2. Neuromolecular Med 2021; 23:25-46. [PMID: 33547562 PMCID: PMC9020407 DOI: 10.1007/s12017-021-08646-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/14/2021] [Indexed: 12/14/2022]
Abstract
Neuro-inflammation accompanies numerous neurological disorders and conditions where it can be associated with a progressive neurodegenerative pathology. In a similar manner, alterations in sphingolipid metabolism often accompany or are causative features in degenerative neurological conditions. These include dementias, motor disorders, autoimmune conditions, inherited metabolic disorders, viral infection, traumatic brain and spinal cord injury, psychiatric conditions, and more. Sphingolipids are major regulators of cellular fate and function in addition to being important structural components of membranes. Their metabolism and signaling pathways can also be regulated by inflammatory mediators. Therefore, as certain sphingolipids exert distinct and opposing cellular roles, alterations in their metabolism can have major consequences. Recently, regulation of bioactive sphingolipids by neuro-inflammatory mediators has been shown to activate a neuronal NADPH oxidase 2 (NOX2) that can provoke damaging oxidation. Therefore, the sphingolipid-regulated neuronal NOX2 serves as a mechanistic link between neuro-inflammation and neurodegeneration. Moreover, therapeutics directed at sphingolipid metabolism or the sphingolipid-regulated NOX2 have the potential to alleviate neurodegeneration arising out of neuro-inflammation.
Collapse
Affiliation(s)
- Emma J Arsenault
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH, 03824, USA
| | - Colin M McGill
- Department of Chemistry, University of Alaska Anchorage, Anchorage, AK, 99508, USA
| | - Brian M Barth
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH, 03824, USA.
| |
Collapse
|
9
|
Sphingomyelin synthase-related protein generates diacylglycerol via the hydrolysis of glycerophospholipids in the absence of ceramide. J Biol Chem 2021; 296:100454. [PMID: 33621517 PMCID: PMC7988496 DOI: 10.1016/j.jbc.2021.100454] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/16/2021] [Accepted: 02/19/2021] [Indexed: 01/20/2023] Open
Abstract
Diacylglycerol (DG) is a well-established lipid second messenger. Sphingomyelin synthase (SMS)-related protein (SMSr) produces DG and ceramide phosphoethanolamine (CPE) by the transfer of phosphoethanolamine from phosphatidylethanolamine (PE) to ceramide. We previously reported that human SMSr overexpressed in COS-7 cells significantly increased DG levels, particularly saturated and/or monounsaturated fatty acid-containing DG molecular species, and provided DG to DG kinase (DGK) δ, which regulates various pathophysiological events, including epidermal growth factor-dependent cell proliferation, type 2 diabetes, and obsessive-compulsive disorder. However, mammalian SMSr puzzlingly produces only trace amounts of CPE/DG. To clarify this discrepancy, we highly purified SMSr and examined its activities other than CPE synthase. Intriguingly, purified SMSr showed a DG-generating activity via hydrolysis of PE, phosphatidic acid (PA), phosphatidylinositol (PI), and phosphatidylcholine (PC) in the absence of ceramide. DG generation through the PA phosphatase (PAP) activity of SMSr was approximately 300-fold higher than that with PE and ceramide. SMSr hydrolyzed PI ten times stronger than PI(4,5)bisphosphate (PI(4,5)P2). The PAP and PC-phospholipase C (PLC) activities of SMSr were inhibited by propranolol, a PAP inhibitor, and by D609, an SMS/PC-PLC inhibitor. Moreover, SMSr showed substrate selectivity for saturated and/or monounsaturated fatty acid-containing PA molecular species, but not arachidonic-acid-containing PA, which is exclusively generated in the PI(4,5)P2 cycle. We confirmed that SMSr expressed in COS-7 cells showed PAP and PI-PLC activities. Taken together, our study indicated that SMSr possesses previously unrecognized enzyme activities, PAP and PI/PE/PC-PLC, and constitutes a novel DG/PA signaling pathway together with DGKδ, which is independent of the PI(4,5)P2 cycle.
Collapse
|
10
|
Jain A, Dadsena S, Holthuis JCM. A switchable ceramide transfer protein for dissecting the mechanism of ceramide‐induced mitochondrial apoptosis. FEBS Lett 2020; 594:3739-3750. [DOI: 10.1002/1873-3468.13956] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Amrita Jain
- Molecular Cell Biology Division Department of Biology/Chemistry and Center for Cellular Nanoanalytics Osnabrück University of Osnabrück Germany
| | - Shashank Dadsena
- Molecular Cell Biology Division Department of Biology/Chemistry and Center for Cellular Nanoanalytics Osnabrück University of Osnabrück Germany
| | - Joost C. M. Holthuis
- Molecular Cell Biology Division Department of Biology/Chemistry and Center for Cellular Nanoanalytics Osnabrück University of Osnabrück Germany
| |
Collapse
|
11
|
Lee SR, Park Y, Park JW. Curvature Effect of a Phosphatidylethanolamine-Included Membrane on the Behavior of Cinnamycin on the Membrane. J Phys Chem B 2020; 124:8984-8988. [DOI: 10.1021/acs.jpcb.0c06029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Sang-Ryong Lee
- Department of Biological and Environmental Science, Dongguk University, Ilsandong-gu, Goyang-si, Gyeonggido 10326, South Korea
| | - Yeseul Park
- Department of Chemical and Biomolecular Engineering, College of Energy and Biotechnology, Seoul National University of Science and Technology, 232 Gongneung-ro, Nowon-gu, Seoul 01811, Republic of Korea
| | - Jin-Won Park
- Department of Chemical and Biomolecular Engineering, College of Energy and Biotechnology, Seoul National University of Science and Technology, 232 Gongneung-ro, Nowon-gu, Seoul 01811, Republic of Korea
| |
Collapse
|
12
|
Lee SR, Park Y, Park JW. Kinetic and thermodynamic studies of cinnamycin specific-adsorption on PE-Included-Membranes using surface plasmon resonance. J Biotechnol 2020; 320:77-79. [PMID: 32593691 DOI: 10.1016/j.jbiotec.2020.06.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 01/16/2023]
Abstract
The binding of the cinnamycin on the biomimetic membrane was studied with respect to time using the surface plasmon resonance(SPR). The membrane was composed of the inner layer tethered on the gold surface and the outer layer formed on the inner layer, which was at the desired ratio of dioleoylphosphatidylethanolamine(DOPE) to dioleoylphosphatidyl- choline(DOPC). On the bilayer, the cinnamycin solution was injected and showed different behavior of the binding with respect to time up on its concentration. For kinetic analysis, the behavior was converted to the coverage fraction with respect to time, which was ratio to the saturated response of 5 μM cinnamycin solution. The fraction change with respect to time was function of the available-site, which was eventually the subtraction of the fraction from one. With the fitting of the first order of the available site, the rate constant was acquired into 6∼7 × 10-3 s-1. Furthermore, the reciprocals of the fraction and the concentration were fitted with the Langmuir adsorption isotherm. From the fitting, the equilibrium constant was between 1 × 107 and 5 × 107 M-1.
Collapse
Affiliation(s)
- Sang-Ryong Lee
- Department of Biological and Environmental Science, Dongguk University, Ilsandong-gu, Goyang-si, Gyeonggido 10326, Republic of Korea
| | - Yeseul Park
- Department of Chemical and Biomolecular Engineering, College of Energy and Biotechnology, Seoul National University of Science and Technology, 232 Gongneung-ro, Nowon-gu, Seoul, 01811, Republic of Korea
| | - Jin-Won Park
- Department of Chemical and Biomolecular Engineering, College of Energy and Biotechnology, Seoul National University of Science and Technology, 232 Gongneung-ro, Nowon-gu, Seoul, 01811, Republic of Korea.
| |
Collapse
|
13
|
Wrapping axons in mammals and Drosophila: Different lipids, same principle. Biochimie 2020; 178:39-48. [PMID: 32800899 DOI: 10.1016/j.biochi.2020.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/16/2022]
Abstract
Plasma membranes of axon-wrapping glial cells develop specific cylindrical bilayer membranes that surround thin individual axons or axon bundles. Axons are wrapped with single layered glial cells in lower organisms whereas in the mammalian nervous system, axons are surrounded with a characteristic complex multilamellar myelin structure. The high content of lipids in myelin suggests that lipids play crucial roles in the structure and function of myelin. The most striking feature of myelin lipids is the high content of galactosylceramide (GalCer). Serological and genetic studies indicate that GalCer plays a key role in the formation and function of the myelin sheath in mammals. In contrast to mammals, Drosophila lacks GalCer. Instead of GalCer, ceramide phosphoethanolamine (CPE) has an important role to ensheath axons with glial cells in Drosophila. GalCer and CPE share similar physical properties: both lipids have a high phase transition temperature and high packing, are immiscible with cholesterol and form helical liposomes. These properties are caused by both the strong headgroup interactions and the tight packing resulting from the small size of the headgroup and the hydrogen bonds between lipid molecules. These results suggest that mammals and Drosophila wrap axons using different lipids but the same conserved principle.
Collapse
|
14
|
Taniguchi M, Okazaki T. Ceramide/Sphingomyelin Rheostat Regulated by Sphingomyelin Synthases and Chronic Diseases in Murine Models. J Lipid Atheroscler 2020; 9:380-405. [PMID: 33024732 PMCID: PMC7521967 DOI: 10.12997/jla.2020.9.3.380] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/16/2020] [Accepted: 07/19/2020] [Indexed: 12/16/2022] Open
Abstract
Ceramide and sphingomyelin (SM) are major components of the double membrane-bound sphingolipids. Ceramide is an essential bioactive lipid involved in numerous cell processes including apoptosis, necrosis, and autophagy-dependent cell death. Inversely, SM regulates opposite cellular processes such as proliferation and migration by changing receptor-mediated signal transduction in the lipid microdomain. SM is generated through a transfer of phosphocholine from phosphatidylcholine to ceramide by SM synthases (SMSs). Research during the past several decades has revealed that the ceramide/SM balance in cellular membranes regulated by SMSs is important to decide the cell fate, survival, and proliferation. In addition, recent experimental studies utilizing SMS knockout mice and murine disease models provide evidence that SMS-regulated ceramide/SM balance is involved in human diseases. Here, we review the basic structural and functional characteristics of SMSs and focus on their cellular functions through the regulation of ceramide/SM balance in membrane microdomains. In addition, we present the pathological or physiological implications of SMSs by analyzing their role in SMS-knockout mice and human disease models. This review finally presents evidence indicating that the regulation of ceramide/SM balance through SMS could be a therapeutic target for human disorders.
Collapse
Affiliation(s)
- Makoto Taniguchi
- Department of Life Science, Medical Research Institute, Kanazawa Medical University, Kahoku, Japan
| | - Toshiro Okazaki
- Research Institute for Bioresources and Biotechnology, Kanazawa Prefectural University, Nonoichi, Japan
| |
Collapse
|
15
|
Kim SE, Park JW. Analysis of interactions between cinnamycin and biomimetic membranes. Colloids Surf B Biointerfaces 2020; 185:110595. [PMID: 31735419 DOI: 10.1016/j.colsurfb.2019.110595] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 09/07/2019] [Accepted: 10/16/2019] [Indexed: 01/07/2023]
Abstract
The interaction between the cinnamycin and the biomimetic membranes was studied using the atomic force microscope(AFM). The bilayer was composed of the monolayer tethered on the gold surface and the outer layer fused with the vesicles on the monolayer. The vesicles were prepared at the desired ratio of dioleoylphosphatidylethanolamine(DOPE) to dioleoylphosphatidylcholine(DOPC). On the bilayer, the surface force measurement was performed with the cinnamycin immobilized covalently on the tip surface. The immobilization led to the presence of the adhesion, which was found while the tip was retracted from the bilayer. In addition, the magnitude of the adhesive force was changed with respect to the composition of DOPE in the outer layer. The difference in the adhesion may be attributed to the mean-molecular-area of DOPE and the specific-binding density on the outer layer. Furthermore, the analysis of the rupture force with respect to the loading rate indicated that the rupture length was around 0.1∼0.13 nm, which was similar to that of a van der Waals bond.
Collapse
Affiliation(s)
- Seong-Eun Kim
- Human IT Convergence Research Center, Korea Electronics Technology Institute, Gyeonggi-do, 13509, Republic of Korea
| | - Jin-Won Park
- Department of Chemical and Biomolecular Engineering, College of Energy and Biotechnology, Seoul National University of Science and Technology, 232 Gongneung-ro, Nowon-gu, Seoul, 01811, Republic of Korea.
| |
Collapse
|
16
|
Naviaux RK, Naviaux JC, Li K, Wang L, Monk JM, Bright AT, Koslik HJ, Ritchie JB, Golomb BA. Metabolic features of Gulf War illness. PLoS One 2019; 14:e0219531. [PMID: 31348786 PMCID: PMC6660083 DOI: 10.1371/journal.pone.0219531] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 06/27/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND More than 230,000 veterans-about 1/3 of US personnel deployed in the 1990-1991 Persian Gulf War-developed chronic, multi-symptom health problems now called "Gulf War illness" (GWI), for which mechanisms and objective diagnostic signatures continue to be sought. METHODS Targeted, broad-spectrum serum metabolomics was used to gain insights into the biology of GWI. 40 male participants, included 20 veterans who met both Kansas and CDC diagnostic criteria for GWI and 20 nonveteran controls without similar symptoms that were 1:1 matched to GWI cases by age, sex, and ethnicity. Serum samples were collected and archived at -80° C prior to testing. 358 metabolites from 46 biochemical pathways were measured by hydrophilic interaction liquid chromatography and tandem mass spectrometry. RESULTS Veterans with GWI, compared to healthy controls, had abnormalities in 8 of 46 biochemical pathways interrogated. Lipid abnormalities accounted for 78% of the metabolic impact. Fifteen ceramides and sphingomyelins, and four phosphatidylcholine lipids were increased. Five of the 8 pathways were shared with the previously reported metabolic phenotype of males with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). However, 4 of the 5 shared pathways were regulated in opposite directions; key pathways that were up-regulated in GWI were down-regulated in ME/CFS. The single pathway regulated in the same direction was purines, which were decreased. CONCLUSIONS Our data show that despite heterogeneous exposure histories, a metabolic phenotype of GWI was clearly distinguished from controls. Metabolomic differences between GWI and ME/CFS show that common clinical symptoms like fatigue can have different chemical mechanisms and different diagnostic implications. Larger studies will be needed to validate these findings.
Collapse
Affiliation(s)
- Robert K. Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California San Diego School of Medicine, San Diego, California, United States of America
- Department of Medicine, Division of Medical Genetics, University of California San Diego School of Medicine, San Diego, California, United States of America
- Department of Pediatrics, Division of Genetics, University of California San Diego School of Medicine, San Diego, California, United States of America
- Department of Pathology, Division of Comparative Pathology, University of California San Diego School of Medicine, San Diego, California, United States of America
| | - Jane C. Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California San Diego School of Medicine, San Diego, California, United States of America
- Department of Neurosciences, Division of Pediatric Neurology, University of California San Diego School of Medicine, San Diego, California, United States of America
| | - Kefeng Li
- The Mitochondrial and Metabolic Disease Center, University of California San Diego School of Medicine, San Diego, California, United States of America
- Department of Medicine, Division of Medical Genetics, University of California San Diego School of Medicine, San Diego, California, United States of America
| | - Lin Wang
- The Mitochondrial and Metabolic Disease Center, University of California San Diego School of Medicine, San Diego, California, United States of America
- Department of Medicine, Division of Medical Genetics, University of California San Diego School of Medicine, San Diego, California, United States of America
| | - Jonathan M. Monk
- The Mitochondrial and Metabolic Disease Center, University of California San Diego School of Medicine, San Diego, California, United States of America
- Department of Medicine, Division of Medical Genetics, University of California San Diego School of Medicine, San Diego, California, United States of America
| | - A. Taylor Bright
- The Mitochondrial and Metabolic Disease Center, University of California San Diego School of Medicine, San Diego, California, United States of America
- Department of Medicine, Division of Medical Genetics, University of California San Diego School of Medicine, San Diego, California, United States of America
| | - Hayley J. Koslik
- Department of Medicine, Division of General Internal Medicine, University of California San Diego School of Medicine, San Diego, California, United States of America
| | - Janis B. Ritchie
- Department of Medicine, Division of General Internal Medicine, University of California San Diego School of Medicine, San Diego, California, United States of America
| | - Beatrice A. Golomb
- Department of Medicine, Division of General Internal Medicine, University of California San Diego School of Medicine, San Diego, California, United States of America
| |
Collapse
|
17
|
Zelnik ID, Ventura AE, Kim JL, Silva LC, Futerman AH. The role of ceramide in regulating endoplasmic reticulum function. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1865:158489. [PMID: 31233888 DOI: 10.1016/j.bbalip.2019.06.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/15/2019] [Accepted: 06/18/2019] [Indexed: 12/12/2022]
Abstract
Sphingolipids (SLs) are an important class of membrane lipids containing a long chain sphingoid base backbone. SL synthesis is compartmentalized between two major cell organelles, the endoplasmic reticulum (ER) and the Golgi apparatus. The initial steps of sphingolipid synthesis take place in the ER, where the simplest SL, ceramide, is synthesized. Although ceramide is a critical membrane component, an imbalance of ceramide levels can have significant deleterious effects on cell properties leading to events such as apoptosis. For this reason and others, ER ceramide levels must be tightly regulated. Here, we describe the biological and biophysical properties of ceramide and discuss how this might impact the ER membrane. This article is part of a special issue entitled: ER Platforms for Membrane Lipid Dynamics.
Collapse
Affiliation(s)
- Iris D Zelnik
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ana E Ventura
- iMed.UL, Research Institute for Medicines and Pharmaceutical Sciences, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Centro de Química-Física Molecular and IN-Institute of Nanoscience and Nanotechnology, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Jiyoon L Kim
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Liana C Silva
- iMed.UL, Research Institute for Medicines and Pharmaceutical Sciences, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Anthony H Futerman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
18
|
Formation of tubules and helical ribbons by ceramide phosphoethanolamine-containing membranes. Sci Rep 2019; 9:5812. [PMID: 30967612 PMCID: PMC6456502 DOI: 10.1038/s41598-019-42247-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 03/19/2019] [Indexed: 11/25/2022] Open
Abstract
Ceramide phosphoethanolamine (CPE), a major sphingolipid in invertebrates, is crucial for axonal ensheathment in Drosophila. Darkfield microscopy revealed that an equimolar mixture of bovine buttermilk CPE (milk CPE) and 1,2-dioleoyl-sn-glycero-3-phosphocholine (diC18:1 PC) tends to form tubules and helical ribbons, while pure milk CPE mainly exhibits amorphous aggregates and, at low frequency, straight needles. Negative staining electron microscopy indicated that helices and tubules were composed of multilayered 5–10 nm thick slab-like structures. Using different molecular species of PC and CPE, we demonstrated that the acyl chain length of CPE but not of PC is crucial for the formation of tubules and helices in equimolar mixtures. Incubation of the lipid suspensions at the respective phase transition temperature of CPE facilitated the formation of both tubules and helices, suggesting a dynamic lipid rearrangement during formation. Substituting diC18:1 PC with diC18:1 PE or diC18:1 PS failed to form tubules and helices. As hydrated galactosylceramide (GalCer), a major lipid in mammalian myelin, has been reported to spontaneously form tubules and helices, it is believed that the ensheathment of axons in mammals and Drosophila is based on similar physical processes with different lipids.
Collapse
|
19
|
Mo M, Yang J, Jiang XC, Cao Y, Fei J, Chen Y, Qi X, Chu Y, Zhou L, Ye D. Discovery of 4-Benzyloxybenzo[ d]isoxazole-3-amine Derivatives as Highly Selective and Orally Efficacious Human Sphingomyelin Synthase 2 Inhibitors that Reduce Chronic Inflammation in db/ db Mice. J Med Chem 2018; 61:8241-8254. [PMID: 30074791 DOI: 10.1021/acs.jmedchem.8b00727] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sphingomyelin synthase 2 (SMS2) is a promising therapeutic target for several chronic inflammation-associated diseases, including atherosclerosis, fatty liver, and insulin resistance. Herein, we report the identification of 4-benzyloxybenzo[ d]isoxazole-3-amine derivatives as potent and highly selective SMS2 inhibitors through a conformational restriction strategy. After systematic structural modifications, several compounds with high selectivity and good potency in vitro were selected for further evaluation. Compound 15w demonstrated good pharmacokinetics (oral bioavailability, F = 56%) in vivo and has an inhibitory potency against sphingomyelin synthase activity when Institute of Cancer Research mice are provided with an oral dose of this compound. In addition, compound 15w attenuated chronic inflammation significantly in db/ db mice after oral dosing for 6 weeks.
Collapse
Affiliation(s)
- Mingguang Mo
- School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Jintong Yang
- School of Pharmacy , Fudan University , Shanghai 201203 , China
| | | | - Yu Cao
- Institute of Precision Medicine , Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai 200125 , China
| | - Jinyu Fei
- School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Yang Chen
- Institute of Precision Medicine , Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai 200125 , China
| | - Xiangyu Qi
- School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Yong Chu
- School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Lu Zhou
- School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Deyong Ye
- School of Pharmacy , Fudan University , Shanghai 201203 , China
| |
Collapse
|
20
|
Michno W, Kaya I, Nyström S, Guerard L, Nilsson KPR, Hammarström P, Blennow K, Zetterberg H, Hanrieder J. Multimodal Chemical Imaging of Amyloid Plaque Polymorphism Reveals Aβ Aggregation Dependent Anionic Lipid Accumulations and Metabolism. Anal Chem 2018; 90:8130-8138. [DOI: 10.1021/acs.analchem.8b01361] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Wojciech Michno
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Ibrahim Kaya
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Sofie Nyström
- IFM-Department of Chemistry, Linköping University, Linköping, Sweden
| | - Laurent Guerard
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- IMCF Biozentrum, University of Basel, Basel, Switzerland
| | | | - Per Hammarström
- IFM-Department of Chemistry, Linköping University, Linköping, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, United Kingdom
| | - Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, United Kingdom
- Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
21
|
Ugarte-Uribe B, García-Sáez AJ. Apoptotic foci at mitochondria: in and around Bax pores. Philos Trans R Soc Lond B Biol Sci 2018. [PMID: 28630156 DOI: 10.1098/rstb.2016.0217] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The permeabilization of the mitochondrial outer membrane by Bax and Bak during apoptosis is considered a key step and a point of no return in the signalling pathway. It is always closely related to the reorganization of mitochondrial cristae that frees cytochrome c to the intermembrane space and to massive mitochondrial fragmentation mediated by the dynamin-like protein Drp1. Despite multiple evidence in favour of a functional link between these processes, the molecular mechanisms that connect them and their relevance for efficient apoptosis signalling remain obscure. In this review, we discuss recent progress on our understanding of how Bax forms pores in the context of Drp1-stabilized signalling platforms at apoptotic foci in mitochondria.This article is part of the themed issue 'Membrane pores: from structure and assembly, to medicine and technology'.
Collapse
Affiliation(s)
- Begoña Ugarte-Uribe
- Interfaculty Institute of Biochemistry, University of Tübingen, Hoppe Seyler Straße 4, 72076 Tübingen, Germany.,Biofisika Institute (UPV/EHU, CSIC), Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Ana J García-Sáez
- Interfaculty Institute of Biochemistry, University of Tübingen, Hoppe Seyler Straße 4, 72076 Tübingen, Germany .,Max Planck Institute for Intelligent Systems, Stuttgart, Germany
| |
Collapse
|
22
|
Mina JGM, Denny PW. Everybody needs sphingolipids, right! Mining for new drug targets in protozoan sphingolipid biosynthesis. Parasitology 2018; 145:134-147. [PMID: 28637533 PMCID: PMC5964470 DOI: 10.1017/s0031182017001081] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 05/15/2017] [Accepted: 05/18/2017] [Indexed: 12/18/2022]
Abstract
Sphingolipids (SLs) are an integral part of all eukaryotic cellular membranes. In addition, they have indispensable functions as signalling molecules controlling a myriad of cellular events. Disruption of either the de novo synthesis or the degradation pathways has been shown to have detrimental effects. The earlier identification of selective inhibitors of fungal SL biosynthesis promised potent broad-spectrum anti-fungal agents, which later encouraged testing some of those agents against protozoan parasites. In this review we focus on the key enzymes of the SL de novo biosynthetic pathway in protozoan parasites of the Apicomplexa and Kinetoplastidae, outlining the divergence and interconnection between host and pathogen metabolism. The druggability of the SL biosynthesis is considered, alongside recent technology advances that will enable the dissection and analyses of this pathway in the parasitic protozoa. The future impact of these advances for the development of new therapeutics for both globally threatening and neglected infectious diseases is potentially profound.
Collapse
Affiliation(s)
- John G M Mina
- Department of Biosciences,Lower Mountjoy,Stockton Road,Durham DH1 3LE,UK
| | - P W Denny
- Department of Biosciences,Lower Mountjoy,Stockton Road,Durham DH1 3LE,UK
| |
Collapse
|
23
|
Bockelmann S, Mina JGM, Korneev S, Hassan DG, Müller D, Hilderink A, Vlieg HC, Raijmakers R, Heck AJR, Haberkant P, Holthuis JCM. A search for ceramide binding proteins using bifunctional lipid analogs yields CERT-related protein StarD7. J Lipid Res 2018; 59:515-530. [PMID: 29343537 DOI: 10.1194/jlr.m082354] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 01/12/2018] [Indexed: 12/21/2022] Open
Abstract
Ceramides are central intermediates of sphingolipid metabolism with dual roles as mediators of cellular stress signaling and mitochondrial apoptosis. How ceramides exert their cytotoxic effects is unclear and their poor solubility in water hampers a search for specific protein interaction partners. Here, we report the application of a photoactivatable and clickable ceramide analog, pacCer, to identify ceramide binding proteins and unravel the structural basis by which these proteins recognize ceramide. Besides capturing ceramide transfer protein (CERT) from a complex proteome, our approach yielded CERT-related steroidogenic acute regulatory protein D7 (StarD7) as novel ceramide binding protein. Previous work revealed that StarD7 is required for efficient mitochondrial import of phosphatidylcholine (PC) and serves a critical role in mitochondrial function and morphology. Combining site-directed mutagenesis and photoaffinity labeling experiments, we demonstrate that the steroidogenic acute regulatory transfer domain of StarD7 harbors a common binding site for PC and ceramide. While StarD7 lacks robust ceramide transfer activity in vitro, we find that its ability to shuttle PC between model membranes is specifically affected by ceramides. Besides demonstrating the suitability of pacCer as a tool to hunt for ceramide binding proteins, our data point at StarD7 as a candidate effector protein by which ceramides may exert part of their mitochondria-mediated cytotoxic effects.
Collapse
Affiliation(s)
- Svenja Bockelmann
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, D-49076 Osnabrück, Germany
| | - John G M Mina
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, D-49076 Osnabrück, Germany.,School of Biological and Biomedical Sciences, Durham University, Durham DH1 3LE, United Kingdom
| | - Sergei Korneev
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, D-49076 Osnabrück, Germany
| | - Dina G Hassan
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, D-49076 Osnabrück, Germany
| | - Dagmar Müller
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, D-49076 Osnabrück, Germany
| | - Angelika Hilderink
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, D-49076 Osnabrück, Germany
| | - Hedwich C Vlieg
- Membrane Biochemistry and Biophysics Division , Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Reinout Raijmakers
- Biomoleular Mass Spectrometry and Proteomics Division, Bijvoet Center and Institute of Biomembranes, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Albert J R Heck
- Biomoleular Mass Spectrometry and Proteomics Division, Bijvoet Center and Institute of Biomembranes, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Per Haberkant
- European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Joost C M Holthuis
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, D-49076 Osnabrück, Germany .,Membrane Biochemistry and Biophysics Division , Utrecht University, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
24
|
Chen Y, Cao Y. The sphingomyelin synthase family: proteins, diseases, and inhibitors. Biol Chem 2017; 398:1319-1325. [PMID: 28742512 DOI: 10.1515/hsz-2017-0148] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 07/13/2017] [Indexed: 12/18/2022]
Abstract
Sphingomyelin (SM) is among the most important biomolecules in eukaryotes and acts as both constructive components and signal carrier in physiological processes. SM is catalyzed by a membrane protein family, sphingomyelin synthases (SMSs), consisting of three members, SMS1, SMS2 and SMSr. SMSs modulate sphingomyelin and other sphingolipids levels, thereby regulating membrane mobility, ceramide-dependent apoptosis and DAG-dependent signaling pathways. SMSs was found associated with various diseases. Downregulation of SMS2 activity results in protective effects against obesity, atherosclerosis and diabetes and makes SMS2 inhibitors potential medicines. Structural guided specific drug design could be the next breakthrough, discriminating SMS2 from other homologs.
Collapse
|
25
|
Peña‐Blanco A, García‐Sáez AJ. Bax, Bak and beyond — mitochondrial performance in apoptosis. FEBS J 2017; 285:416-431. [DOI: 10.1111/febs.14186] [Citation(s) in RCA: 628] [Impact Index Per Article: 78.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/12/2017] [Accepted: 07/26/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Aida Peña‐Blanco
- Interfaculty Institute of Biochemistry Tübingen University Germany
| | - Ana J. García‐Sáez
- Interfaculty Institute of Biochemistry Tübingen University Germany
- Max‐Planck Institute for Intelligent Systems Stuttgart Germany
| |
Collapse
|
26
|
Prudent J, McBride HM. The mitochondria–endoplasmic reticulum contact sites: a signalling platform for cell death. Curr Opin Cell Biol 2017; 47:52-63. [DOI: 10.1016/j.ceb.2017.03.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 02/22/2017] [Accepted: 03/01/2017] [Indexed: 01/23/2023]
|
27
|
Ceramide phosphoethanolamine synthase SMSr is a target of caspase-6 during apoptotic cell death. Biosci Rep 2017; 37:BSR20170867. [PMID: 28659495 PMCID: PMC5567093 DOI: 10.1042/bsr20170867] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/25/2017] [Accepted: 06/28/2017] [Indexed: 01/11/2023] Open
Abstract
Ceramides are essential precursors of sphingolipids with a dual role as mediators of apoptotic cell death. Previous work revealed that the ER-resident ceramide phosphoethanolamine (CPE) synthase SMSr/SAMD8 is a suppressor of ceramide-mediated apoptosis in cultured cells. Anti-apoptotic activity of SMSr requires a catalytically active enzyme but also relies on the enzyme’s N-terminal sterile α-motif or SAM domain. Here, we demonstrate that SMSr itself is a target of the apoptotic machinery. Treatment of cells with staurosporine or the death receptor ligand FasL triggers caspase-mediated cleavage of SMSr at a conserved aspartate located downstream of the enzyme’s SAM domain and upstream of its first membrane span. Taking advantage of reconstitution experiments with SMSr produced in a cell-free expression system, specific caspase-inhibitors and gene silencing approaches, we show that SMSr is a novel and specific substrate of caspase-6, a non-conventional effector caspase implicated in Huntington’s and Alzheimer’s diseases. Our findings underscore a role of SMSr as negative regulator of ceramide-induced cell death and, in view of a prominent expression of the enzyme in brain, raise questions regarding its potential involvement in neurodegenerative disorders.
Collapse
|
28
|
Jain A, Holthuis JCM. Membrane contact sites, ancient and central hubs of cellular lipid logistics. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1450-1458. [PMID: 28554771 DOI: 10.1016/j.bbamcr.2017.05.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 05/10/2017] [Accepted: 05/17/2017] [Indexed: 12/26/2022]
Abstract
Membrane contact sites (MCSs) are regions where two organelles are closely apposed to facilitate molecular communication and promote a functional integration of compartmentalized cellular processes. There is growing evidence that MCSs play key roles in controlling intracellular lipid flows and distributions. Strikingly, even organelles connected by vesicular trafficking exchange lipids en bulk via lipid transfer proteins that operate at MCSs. Herein, we describe how MCSs developed into central hubs of lipid logistics during the evolution of eukaryotic cells. We then focus on how modern eukaryotes exploit MCSs to help solve a major logistical problem, namely to preserve the unique lipid mixtures of their early and late secretory organelles in the face of extensive vesicular trafficking. This article is part of a Special Issue entitled: Membrane Contact Sites edited by Christian Ungermann and Benoit Kornmann.
Collapse
Affiliation(s)
- Amrita Jain
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, D-49076 Osnabrück, Germany
| | - Joost C M Holthuis
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, D-49076 Osnabrück, Germany; Membrane Biochemistry & Biophysics, Bijvoet Center and Institute of Biomembranes, Utrecht University, 3584 CH Utrecht, The Netherlands.
| |
Collapse
|
29
|
Adachi R, Ogawa K, Matsumoto SI, Satou T, Tanaka Y, Sakamoto J, Nakahata T, Okamoto R, Kamaura M, Kawamoto T. Discovery and characterization of selective human sphingomyelin synthase 2 inhibitors. Eur J Med Chem 2017; 136:283-293. [PMID: 28505533 DOI: 10.1016/j.ejmech.2017.04.067] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/10/2017] [Accepted: 04/24/2017] [Indexed: 10/19/2022]
Abstract
Sphingomyelin synthase (SMS) is a membrane enzyme that catalyzes the synthesis of sphingomyelin, is required for the maintenance of plasma membrane microdomain fluidity, and has two isoforms: SMS1 and SMS2. Although these isoforms exhibit the same SMS activity, they are different enzymes with distinguishable subcellular localizations. It was reported that SMS2 KO mice displayed lower inflammatory responses and anti-atherosclerotic effects, suggesting that inhibition of SMS2 would be a potential therapeutic approach for controlling inflammatory responses and atherosclerosis. This study aimed to discover a novel small-molecule compound that selectively inhibits SMS2 enzymatic activity. We developed a human SMS2 enzyme assay with a high-throughput mass spectrometry-based screening system. We characterized the enzymatic properties of SMS2 and established a high-throughput screening-compatible assay condition. To identify human SMS2 inhibitors, we conducted compound screening using the enzyme assay. We identified a 2-quinolone derivative as a SMS2 selective inhibitor with an IC50 of 950 nM and >100-fold selectivity for SMS2 over SMS1. The 2-quinolone exhibited efficacy in a cell-based engagement assay. We demonstrated that a more potent derivative directly bound to SMS2-expressing membrane fractions in an affinity selection mass spectrometry assay. Mutational analyses revealed that the interaction of the inhibitor with SMS2 required the presence of the amino acids S227 and H229, which are located in the catalytic domain of SMS2. In conclusion, we discovered novel SMS2-selective inhibitors. 2-Quinolone SMS2 inhibitors are considered applicable for leading optimization studies. Further investigations using these SMS2 inhibitors would provide validation tools for SMS2-relevant pathways in vitro and in vivo.
Collapse
Affiliation(s)
- Ryutaro Adachi
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan.
| | - Kazumasa Ogawa
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Shin-Ichi Matsumoto
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Takuya Satou
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Yukiya Tanaka
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Jyunichi Sakamoto
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Takashi Nakahata
- CVM Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Rei Okamoto
- CVM Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Masahiro Kamaura
- Medicinal Chemistry Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Tomohiro Kawamoto
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
30
|
Kol M, Panatala R, Nordmann M, Swart L, van Suijlekom L, Cabukusta B, Hilderink A, Grabietz T, Mina JGM, Somerharju P, Korneev S, Tafesse FG, Holthuis JCM. Switching head group selectivity in mammalian sphingolipid biosynthesis by active-site-engineering of sphingomyelin synthases. J Lipid Res 2017; 58:962-973. [PMID: 28336574 DOI: 10.1194/jlr.m076133] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 03/07/2017] [Indexed: 12/17/2022] Open
Abstract
SM is a fundamental component of mammalian cell membranes that contributes to mechanical stability, signaling, and sorting. Its production involves the transfer of phosphocholine from phosphatidylcholine onto ceramide, a reaction catalyzed by SM synthase (SMS)1 in the Golgi and SMS2 at the plasma membrane. Mammalian cells also synthesize trace amounts of the SM analog, ceramide phosphoethanolamine (CPE), but the physiological relevance of CPE production is unclear. Previous work revealed that SMS2 is a bifunctional enzyme producing both SM and CPE, whereas a closely related enzyme, SMS-related protein (SMSr)/SAMD8, acts as a monofunctional CPE synthase in the endoplasmic reticulum. Using domain swapping and site-directed mutagenesis on enzymes expressed in defined lipid environments, we here identified structural determinants that mediate the head group selectivity of SMS family members. Notably, a single residue adjacent to the catalytic histidine in the third exoplasmic loop profoundly influenced enzyme specificity, with Glu permitting SMS-catalyzed CPE production and Asp confining the enzyme to produce SM. An exchange of exoplasmic residues with SMSr proved sufficient to convert SMS1 into a bulk CPE synthase. This allowed us to establish mammalian cells that produce CPE rather than SM as the principal phosphosphingolipid and provide a model of the molecular interactions that impart catalytic specificity among SMS enzymes.
Collapse
Affiliation(s)
- Matthijs Kol
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany .,Membrane Biochemistry and Biophysics, Bijvoet Center and Institute of Biomembranes, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Radhakrishnan Panatala
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany.,Membrane Biochemistry and Biophysics, Bijvoet Center and Institute of Biomembranes, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Mirjana Nordmann
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Leoni Swart
- Membrane Biochemistry and Biophysics, Bijvoet Center and Institute of Biomembranes, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Leonie van Suijlekom
- Membrane Biochemistry and Biophysics, Bijvoet Center and Institute of Biomembranes, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Birol Cabukusta
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Angelika Hilderink
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Tanja Grabietz
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - John G M Mina
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Pentti Somerharju
- Medical Biochemistry, Institute of Biomedicine, University of Helsinki, Helsinki 00014, Finland
| | - Sergei Korneev
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Fikadu G Tafesse
- Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239
| | - Joost C M Holthuis
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany .,Membrane Biochemistry and Biophysics, Bijvoet Center and Institute of Biomembranes, Utrecht University, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
31
|
Chun-Mei H, Qin-Min G, Shu-Ming P, Xiang-Yang Z. Expression profiling and ontology analysis of circulating long non-coding RNAs in septic acute kidney injury patients. Clin Chem Lab Med 2017; 54:e395-e399. [PMID: 27341563 DOI: 10.1515/cclm-2015-1281] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/30/2016] [Indexed: 01/20/2023]
|
32
|
Cabukusta B, Kol M, Kneller L, Hilderink A, Bickert A, Mina JGM, Korneev S, Holthuis JCM. ER residency of the ceramide phosphoethanolamine synthase SMSr relies on homotypic oligomerization mediated by its SAM domain. Sci Rep 2017; 7:41290. [PMID: 28120887 PMCID: PMC5264588 DOI: 10.1038/srep41290] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 12/19/2016] [Indexed: 12/12/2022] Open
Abstract
SMSr/SAMD8 is an ER-resident ceramide phosphoethanolamine synthase with a critical role in controlling ER ceramides and suppressing ceramide-induced apoptosis in cultured cells. SMSr-mediated ceramide homeostasis relies on the enzyme's catalytic activity as well as on its N-terminal sterile α-motif or SAM domain. Here we report that SMSr-SAM is structurally and functionally related to the SAM domain of diacylglycerol kinase DGKδ, a central regulator of lipid signaling at the plasma membrane. Native gel electrophoresis indicates that both SAM domains form homotypic oligomers. Chemical crosslinking studies show that SMSr self-associates into ER-resident trimers and hexamers that resemble the helical oligomers formed by DGKδ-SAM. Residues critical for DGKδ-SAM oligomerization are conserved in SMSr-SAM and their substitution causes a dissociation of SMSr oligomers as well as a partial redistribution of the enzyme to the Golgi. Conversely, treatment of cells with curcumin, a drug disrupting ceramide and Ca2+ homeostasis in the ER, stabilizes SMSr oligomers and promotes retention of the enzyme in the ER. Our data provide first demonstration of a multi-pass membrane protein that undergoes homotypic oligomerization via its SAM domain and indicate that SAM-mediated self-assembly of SMSr is required for efficient retention of the enzyme in the ER.
Collapse
Affiliation(s)
- Birol Cabukusta
- Molecular Cell Biology Division, Faculty of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Matthijs Kol
- Molecular Cell Biology Division, Faculty of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Laura Kneller
- Molecular Cell Biology Division, Faculty of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Angelika Hilderink
- Molecular Cell Biology Division, Faculty of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Andreas Bickert
- Molecular Genetics, Life and Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - John G. M. Mina
- Molecular Cell Biology Division, Faculty of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Sergei Korneev
- Molecular Cell Biology Division, Faculty of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Joost C. M. Holthuis
- Molecular Cell Biology Division, Faculty of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| |
Collapse
|
33
|
Abstract
The sphingolipid family of lipids modulate several cellular processes, including proliferation, cell cycle regulation, inflammatory signaling pathways, and cell death. Several members of the sphingolipid pathway have opposing functions and thus imbalances in sphingolipid metabolism result in deregulated cellular processes, which cause or contribute to diseases and disorders in humans. A key cellular process regulated by sphingolipids is apoptosis, or programmed cell death. Sphingolipids play an important role in both extrinsic and intrinsic apoptotic pathways depending on the stimuli, cell type and cellular response to the stress. During mitochondrial-mediated apoptosis, multiple pathways converge on mitochondria and induce mitochondrial outer membrane permeabilization (MOMP). MOMP results in the release of intermembrane space proteins such as cytochrome c and Apaf1 into the cytosol where they activate the caspases and DNases that execute cell death. The precise molecular components of the pore(s) responsible for MOMP are unknown, but sphingolipids are thought to play a role. Here, we review evidence for a role of sphingolipids in the induction of mitochondrial-mediated apoptosis with a focus on potential underlying molecular mechanisms by which altered sphingolipid metabolism indirectly or directly induce MOMP. Data available on these mechanisms is reviewed, and the focus and limitations of previous and current studies are discussed to present important unanswered questions and potential future directions.
Collapse
Affiliation(s)
- Gauri A Patwardhan
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA
| | - Levi J Beverly
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA.,Department of Medicine, University of Louisville, Louisville, KY, 40202, USA.,James Graham Brown Cancer Center, University of Louisville, 505 South Hancock Street, Clinical and Translational Research Building, Room 203, Louisville, KY, 40202, USA
| | - Leah J Siskind
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA. .,James Graham Brown Cancer Center, University of Louisville, 505 South Hancock Street, Clinical and Translational Research Building, Room 203, Louisville, KY, 40202, USA.
| |
Collapse
|
34
|
Liu LK, Choudhary V, Toulmay A, Prinz WA. An inducible ER-Golgi tether facilitates ceramide transport to alleviate lipotoxicity. J Cell Biol 2016; 216:131-147. [PMID: 28011845 PMCID: PMC5223604 DOI: 10.1083/jcb.201606059] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 10/20/2016] [Accepted: 12/07/2016] [Indexed: 01/27/2023] Open
Abstract
Liu et al. show that ER–Golgi tethering increases during ER stress in yeast. The protein Nvj2p is required for this tethering, which promotes nonvesicular ceramide transport from the ER to the Golgi to alleviate ceramide toxicity. Ceramides are key intermediates in sphingolipid biosynthesis and potent signaling molecules. However, excess ceramide is toxic, causing growth arrest and apoptosis. In this study, we identify a novel mechanism by which cells prevent the toxic accumulation of ceramides; they facilitate nonvesicular ceramide transfer from the endoplasmic reticulum (ER) to the Golgi complex, where ceramides are converted to complex sphingolipids. We find that the yeast protein Nvj2p promotes the nonvesicular transfer of ceramides from the ER to the Golgi complex. The protein is a tether that generates close contacts between these compartments and may directly transport ceramide. Nvj2p normally resides at contacts between the ER and other organelles, but during ER stress, it relocalizes to and increases ER–Golgi contacts. ER–Golgi contacts fail to form during ER stress in cells lacking Nvj2p. Our findings demonstrate that cells regulate ER–Golgi contacts in response to stress and reveal that nonvesicular ceramide transfer out of the ER prevents the buildup of toxic amounts of ceramides.
Collapse
Affiliation(s)
- Li-Ka Liu
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Vineet Choudhary
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Alexandre Toulmay
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - William A Prinz
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
35
|
Jain A, Beutel O, Ebell K, Korneev S, Holthuis JCM. Diverting CERT-mediated ceramide transport to mitochondria triggers Bax-dependent apoptosis. J Cell Sci 2016; 130:360-371. [PMID: 27888218 DOI: 10.1242/jcs.194191] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 11/14/2016] [Indexed: 01/02/2023] Open
Abstract
A deregulation of ceramide biosynthesis in the endoplasmic reticulum (ER) is frequently linked to induction of mitochondrial apoptosis. Although in vitro studies suggest that ceramides might initiate cell death by acting directly on mitochondria, their actual contribution to the apoptotic response in living cells is unclear. Here, we have analyzed the consequences of targeting the biosynthetic flow of ceramides to mitochondria using a ceramide transfer protein (encoded by COL4A3BP) equipped with an OMM anchor, mitoCERT. Cells expressing mitoCERT import ceramides into mitochondria and undergo Bax-dependent apoptosis. Apoptosis induction by mitoCERT was abolished through (i) removal of its ceramide transfer domain, (ii) disruption of its interaction with VAMP-associated proteins (VAPs) in the ER, (iii) addition of antagonistic CERT inhibitor HPA12, (iv) blocking de novo ceramide synthesis and (v) targeting of a bacterial ceramidase to mitochondria. Our data provide the first demonstration that translocation of ER ceramides to mitochondria specifically commits cells to death and establish mitoCERT as a valuable new tool to unravel the molecular principles underlying ceramide-mediated apoptosis.
Collapse
Affiliation(s)
- Amrita Jain
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, Osnabrück D-49076, Germany
| | - Oliver Beutel
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, Osnabrück D-49076, Germany.,Max-Planck-Institute for Molecular Cell Biology and Genetics, Dresden D-01307, Germany
| | - Katharina Ebell
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, Osnabrück D-49076, Germany
| | - Sergey Korneev
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, Osnabrück D-49076, Germany
| | - Joost C M Holthuis
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, Osnabrück D-49076, Germany .,Membrane Biochemistry & Biophysics, Bijvoet Center and Institute of Biomembranes, Utrecht University, Utrecht 3584 CH, The Netherlands
| |
Collapse
|
36
|
Probing phosphoethanolamine-containing lipids in membranes with duramycin/cinnamycin and aegerolysin proteins. Biochimie 2016; 130:81-90. [DOI: 10.1016/j.biochi.2016.09.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 09/27/2016] [Indexed: 02/07/2023]
|
37
|
Cabukusta B, Köhlen JA, Richter CP, You C, Holthuis JCM. Monitoring Changes in the Oligomeric State of a Candidate Endoplasmic Reticulum (ER) Ceramide Sensor by Single-molecule Photobleaching. J Biol Chem 2016; 291:24735-24746. [PMID: 27729449 DOI: 10.1074/jbc.m116.749812] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 10/07/2016] [Indexed: 11/06/2022] Open
Abstract
Single-molecule photobleaching has emerged as a powerful non-invasive approach to extract the stoichiometry of multimeric membrane proteins in their native cellular environment. However, this method has mainly been used to determine the subunit composition of ion channels and receptors at the plasma membrane. Here, we applied single-molecule photobleaching to analyze the oligomeric state of an endoplasmic reticulum (ER) resident candidate ceramide sensor protein, SMSr/SAMD8. Co-immunoprecipitation and chemical cross-linking studies previously revealed that the N-terminal sterile alpha motif (or SAM) domain of SMSr drives self-assembly of the protein into oligomers and that SMSr oligomerization is promoted by curcumin, a drug known to perturb ER ceramide and calcium homeostasis. Application of cell spreading surface-active coating materials in combination with total internal reflection fluorescence (TIRF) microscopy allowed us to image GFP-tagged SMSr proteins as single fluorescent spots in the ER of HeLa cells in which expression of endogenous SMSr was abolished. In line with our biochemical analysis, we find that the number of bleaching steps in SMSr-GFP-positive spots displays a substantial drop after removal of the SAM domain. In contrast, treatment of cells with curcumin increased the number of bleaching steps. Our results document the first successful application of single-molecule photobleaching to resolve drug-induced and domain-dependent changes in the oligomeric state of an ER-resident membrane protein, hence establishing a complementary method to unravel the mechanism by which SMSr controls ceramide levels in the ER.
Collapse
Affiliation(s)
| | | | - Christian P Richter
- Biophysics Division, Faculty of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany and
| | - Changjiang You
- Biophysics Division, Faculty of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany and
| | - Joost C M Holthuis
- From the Molecular Cell Biology Division and; Membrane Biochemistry and Biophysics, Bijvoet Center and Institute of Biomembranes, Utrecht University, 3584 CH Utrecht, The Netherlands.
| |
Collapse
|
38
|
Kol M, Panatala R, Nordmann M, Swart L, van Suijlekom L, Cabukusta B, Hilderink A, Grabietz T, Mina JGM, Somerharju P, Korneev S, Tafesse FG, Holthuis JCM. Switching head group selectivity in mammalian sphingolipid biosynthesis by active-site engineering of sphingomyelin synthases. J Lipid Res 2016; 57:1273-85. [PMID: 27165857 DOI: 10.1194/jlr.m068692] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Indexed: 01/23/2023] Open
Abstract
SM is a fundamental component of mammalian cell membranes that contributes to mechanical stability, signaling, and sorting. Its production involves the transfer of phosphocholine from phosphatidylcholine onto ceramide, a reaction catalyzed by SM synthase (SMS) 1 in the Golgi and SMS2 at the plasma membrane. Mammalian cells also synthesize trace amounts of the SM analog ceramide phosphoethanolamine (CPE), but the physiological relevance of CPE production is unclear. Previous work revealed that SMS2 is a bifunctional enzyme producing both SM and CPE, whereas a closely related enzyme, sphingomyelin synthase-related protein (SMSr)/SAMD8, acts as a monofunctional CPE synthase in the endoplasmatic reticulum. Using domain swapping and site-directed mutagenesis on enzymes expressed in defined lipid environments, we here identified structural determinants that mediate head group selectivity of SMS family members. Notably, a single residue adjacent to the catalytic histidine in the third exoplasmic loop profoundly influenced enzyme specificity, with glutamic acid permitting SMS-catalyzed CPE production and aspartic acid confining the enzyme to produce SM. An exchange of exoplasmic residues with SMSr proved sufficient to convert SMS1 into a bulk CPE synthase. This allowed us to establish mammalian cells that produce CPE rather than SM as the principal phosphosphingolipid and provide a model of the molecular interactions that impart catalytic specificity among SMS enzymes.
Collapse
Affiliation(s)
- Matthijs Kol
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany Membrane Biochemistry & Biophysics, Bijvoet Center and Institute of Biomembranes, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Radhakrishnan Panatala
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany Membrane Biochemistry & Biophysics, Bijvoet Center and Institute of Biomembranes, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Mirjana Nordmann
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Leoni Swart
- Membrane Biochemistry & Biophysics, Bijvoet Center and Institute of Biomembranes, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Leonie van Suijlekom
- Membrane Biochemistry & Biophysics, Bijvoet Center and Institute of Biomembranes, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Birol Cabukusta
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Angelika Hilderink
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Tanja Grabietz
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - John G M Mina
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Pentti Somerharju
- Medical Biochemistry, Institute of Biomedicine, University of Helsinki, Helsinki 00014, Finland
| | - Sergei Korneev
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Fikadu G Tafesse
- Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239
| | - Joost C M Holthuis
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany Membrane Biochemistry & Biophysics, Bijvoet Center and Institute of Biomembranes, Utrecht University, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
39
|
Olson DK, Fröhlich F, Farese RV, Walther TC. Taming the sphinx: Mechanisms of cellular sphingolipid homeostasis. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1861:784-792. [PMID: 26747648 DOI: 10.1016/j.bbalip.2015.12.021] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 12/14/2015] [Accepted: 12/28/2015] [Indexed: 12/11/2022]
Abstract
Sphingolipids are important structural membrane components of eukaryotic cells, and potent signaling molecules. As such, their levels must be maintained to optimize cellular functions in different cellular membranes. Here, we review the current knowledge of homeostatic sphingolipid regulation. We describe recent studies in Saccharomyces cerevisiae that have provided insights into how cells sense changes in sphingolipid levels in the plasma membrane and acutely regulate sphingolipid biosynthesis by altering signaling pathways. We also discuss how cellular trafficking has emerged as an important determinant of sphingolipid homeostasis. Finally, we highlight areas where work is still needed to elucidate the mechanisms of sphingolipid regulation and the physiological functions of such regulatory networks, especially in mammalian cells. This article is part of a Special Issue entitled: The cellular lipid landscape edited by Tim P. Levine and Anant K. Menon.
Collapse
Affiliation(s)
- D K Olson
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, United States; Department of Cell Biology, Yale School of Medicine, United States
| | - F Fröhlich
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, United States
| | - R V Farese
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, United States; Department of Cell Biology, Harvard Medical School, United States; Broad Institute of Harvard and MIT, United States.
| | - T C Walther
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, United States; Department of Cell Biology, Harvard Medical School, United States; Broad Institute of Harvard and MIT, United States; Howard Hughes Medical Institute, United States.
| |
Collapse
|
40
|
Zhou K, Blom T. Trafficking and Functions of Bioactive Sphingolipids: Lessons from Cells and Model Membranes. Lipid Insights 2015; 8:11-20. [PMID: 26715852 PMCID: PMC4685176 DOI: 10.4137/lpi.s31615] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/08/2015] [Accepted: 11/10/2015] [Indexed: 12/15/2022] Open
Abstract
Ceramide and sphingosine and their phosphorylated counterparts are recognized as "bioactive sphingolipids" and modulate membrane integrity, the activity of enzymes, or act as ligands of G protein-coupled receptors. The subcellular distribution of the bioactive sphingolipids is central to their function as the same lipid can mediate diametrically opposite effects depending on its location. To ensure that these lipids are present in the right amount and in the appropriate organelles, cells employ selective lipid transport and compartmentalize sphingolipid-metabolizing enzymes to characteristic subcellular sites. Our knowledge of key mechanisms involved in sphingolipid signaling and trafficking has increased substantially in the past decades-thanks to advances in biochemical and cell biological methods. In this review, we focus on the bioactive sphingolipids and discuss how the combination of studies in cells and in model membranes have contributed to our understanding of how they behave and function in living organisms.
Collapse
Affiliation(s)
- Kecheng Zhou
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tomas Blom
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
41
|
Pore-forming toxins: Properties, diversity, and uses as tools to image sphingomyelin and ceramide phosphoethanolamine. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1858:576-92. [PMID: 26498396 DOI: 10.1016/j.bbamem.2015.10.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 09/30/2015] [Accepted: 10/14/2015] [Indexed: 12/27/2022]
Abstract
Pore-forming toxins (PFTs) represent a unique class of highly specific lipid-binding proteins. The cytotoxicity of these compounds has been overcome through crystallographic structure and mutation studies, facilitating the development of non-toxic lipid probes. As a consequence, non-toxic PFTs have been utilized as highly specific probes to visualize the diversity and dynamics of lipid nanostructures in living and fixed cells. This review is focused on the application of PFTs and their non-toxic analogs as tools to visualize sphingomyelin and ceramide phosphoethanolamine, two major phosphosphingolipids in mammalian and insect cells, respectively. This article is part of a Special Issue entitled: Pore-Forming Toxins edited by Mauro Dalla Serra and Franco Gambale.
Collapse
|
42
|
LAPTM4B facilitates late endosomal ceramide export to control cell death pathways. Nat Chem Biol 2015; 11:799-806. [DOI: 10.1038/nchembio.1889] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 07/09/2015] [Indexed: 12/31/2022]
|
43
|
Ungermann C. vCLAMPs—an intimate link between vacuoles and mitochondria. Curr Opin Cell Biol 2015; 35:30-6. [DOI: 10.1016/j.ceb.2015.03.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 03/20/2015] [Accepted: 03/26/2015] [Indexed: 11/26/2022]
|
44
|
Bickert A, Ginkel C, Kol M, vom Dorp K, Jastrow H, Degen J, Jacobs RL, Vance DE, Winterhager E, Jiang XC, Dörmann P, Somerharju P, Holthuis JCM, Willecke K. Functional characterization of enzymes catalyzing ceramide phosphoethanolamine biosynthesis in mice. J Lipid Res 2015; 56:821-35. [PMID: 25667419 DOI: 10.1194/jlr.m055269] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Besides bulk amounts of SM, mammalian cells produce small quantities of the SM analog ceramide phosphoethanolamine (CPE). Little is known about the biological role of CPE or enzymes responsible for CPE production. Heterologous expression studies revealed that SM synthase (SMS)2 is a bifunctional enzyme producing both SM and CPE, whereas SMS-related protein (SMSr) serves as monofunctional CPE synthase. Acute disruption of SMSr catalytic activity in cultured cells causes a rise in endoplasmic reticulum (ER) ceramides, fragmentation of ER exit sites, and induction of mitochondrial apoptosis. To address the relevance of CPE biosynthesis in vivo, we analyzed the tissue-specific distribution of CPE in mice and generated mouse lines lacking SMSr and SMS2 catalytic activity. We found that CPE levels were >300-fold lower than SM in all tissues examined. Unexpectedly, combined inactivation of SMSr and SMS2 significantly reduced, but did not eliminate, tissue-specific CPE pools and had no obvious impact on mouse development or fertility. While SMSr is widely expressed and serves as the principal CPE synthase in the brain, blocking its catalytic activity did not affect ceramide levels or secretory pathway integrity in the brain or any other tissue. Our data provide a first inventory of CPE species and CPE-biosynthetic enzymes in mammals.
Collapse
Affiliation(s)
- Andreas Bickert
- Molecular Genetics, Life, and Medical Sciences Institute University of Bonn, 53115 Bonn, Germany
| | - Christina Ginkel
- Molecular Genetics, Life, and Medical Sciences Institute University of Bonn, 53115 Bonn, Germany
| | - Matthijs Kol
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Katharina vom Dorp
- Institute of Molecular Physiology and Biotechnology of Plants, University of Bonn, 53115 Bonn, Germany
| | - Holger Jastrow
- Imaging Center Essen, Electron Microscopy Unit, University Hospital University of Duisburg-Essen, 45147 Essen, Germany
| | - Joachim Degen
- Molecular Genetics, Life, and Medical Sciences Institute University of Bonn, 53115 Bonn, Germany
| | - René L Jacobs
- Departments of Agricultural, Food, and Nutritional Science, Molecular and Cell Biology of Lipids, University of Alberta, T6G 2S2 Edmonton, Canada
| | - Dennis E Vance
- Biochemistry, University of Alberta, T6G 2S2 Edmonton, Canada
| | - Elke Winterhager
- Department of Molecular Biology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Xian-Cheng Jiang
- Department of Anatomy and Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY 11203
| | - Peter Dörmann
- Institute of Molecular Physiology and Biotechnology of Plants, University of Bonn, 53115 Bonn, Germany
| | - Pentti Somerharju
- Medical Biochemistry, Institute of Biomedicine, University of Helsinki, 00014 Helsinki, Finland
| | - Joost C M Holthuis
- Molecular Cell Biology Division, Department of Biology/Chemistry, University of Osnabrück, 49076 Osnabrück, Germany
| | - Klaus Willecke
- Molecular Genetics, Life, and Medical Sciences Institute University of Bonn, 53115 Bonn, Germany
| |
Collapse
|
45
|
Ding T, Kabir I, Li Y, Lou C, Yazdanyar A, Xu J, Dong J, Zhou H, Park T, Boutjdir M, Li Z, Jiang XC. All members in the sphingomyelin synthase gene family have ceramide phosphoethanolamine synthase activity. J Lipid Res 2015; 56:537-545. [PMID: 25605874 DOI: 10.1194/jlr.m054627] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sphingomyelin synthase-related protein (SMSr) synthesizes the sphingomyelin analog ceramide phosphoethanolamine (CPE) in cells. Previous cell studies indicated that SMSr is involved in ceramide homeostasis and is crucial for cell function. To further examine SMSr function in vivo, we generated Smsr KO mice that were fertile and had no obvious phenotypic alterations. Quantitative MS analyses of plasma, liver, and macrophages from the KO mice revealed only marginal changes in CPE and ceramide as well as other sphingolipid levels. Because SMS2 also has CPE synthase activity, we prepared Smsr/Sms2 double KO mice. We found that CPE levels were not significantly changed in macrophages, suggesting that CPE levels are not exclusively dependent on SMSr and SMS2 activities. We then measured CPE levels in Sms1 KO mice and found that Sms1 deficiency also reduced plasma CPE levels. Importantly, we found that expression of Sms1 or Sms2 in SF9 insect cells significantly increased not only SM but also CPE formation, indicating that SMS1 also has CPE synthase activity. Moreover, we measured CPE synthase Km and Vmax for SMS1, SMS2, and SMSr using different NBD ceramides. Our study reveals that all mouse SMS family members (SMSr, SMS1, and SMS2) have CPE synthase activity. However, neither CPE nor SMSr appears to be a critical regulator of ceramide levels in vivo.
Collapse
Affiliation(s)
- Tingbo Ding
- School of Pharmacy, Fudan University, China; Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY
| | - Inamul Kabir
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY
| | - Yue Li
- School of Pharmacy, Fudan University, China; Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY
| | - Caixia Lou
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY; Guangdong Medical Laboratory Animal Center, Foshan, China
| | | | - Jiachen Xu
- School of Pharmacy, Fudan University, China
| | - Jibin Dong
- School of Pharmacy, Fudan University, China
| | - Hongwen Zhou
- Department of Endocrinology, The First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Taesik Park
- Department of Life Science, Gachon University, Sungnam, 461-701, South Korea
| | | | - Zhiqiang Li
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY
| | - Xian-Cheng Jiang
- School of Pharmacy, Fudan University, China; Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY; Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, Brooklyn, NY.
| |
Collapse
|
46
|
Yamaji T, Hanada K. Sphingolipid metabolism and interorganellar transport: localization of sphingolipid enzymes and lipid transfer proteins. Traffic 2014; 16:101-22. [PMID: 25382749 DOI: 10.1111/tra.12239] [Citation(s) in RCA: 293] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 10/29/2014] [Accepted: 11/06/2014] [Indexed: 11/28/2022]
Abstract
In recent decades, many sphingolipid enzymes, sphingolipid-metabolism regulators and sphingolipid transfer proteins have been isolated and characterized. This review will provide an overview of the intracellular localization and topology of sphingolipid enzymes in mammalian cells to highlight the locations where respective sphingolipid species are produced. Interestingly, three sphingolipids that reside or are synthesized in cytosolic leaflets of membranes (ceramide, glucosylceramide and ceramide-1-phosphate) all have cytosolic lipid transfer proteins (LTPs). These LTPs consist of ceramide transfer protein (CERT), four-phosphate adaptor protein 2 (FAPP2) and ceramide-1-phosphate transfer protein (CPTP), respectively. These LTPs execute functions that affect both the location and metabolism of the lipids they bind. Molecular details describing the mechanisms of regulation of LTPs continue to emerge and reveal a number of critical processes, including competing phosphorylation and dephosphorylation reactions and binding interactions with regulatory proteins and lipids that influence the transport, organelle distribution and metabolism of sphingolipids.
Collapse
Affiliation(s)
- Toshiyuki Yamaji
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | | |
Collapse
|
47
|
Vance JE. Phospholipid Synthesis and Transport in Mammalian Cells. Traffic 2014; 16:1-18. [DOI: 10.1111/tra.12230] [Citation(s) in RCA: 376] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 09/17/2014] [Accepted: 09/18/2014] [Indexed: 01/18/2023]
Affiliation(s)
- Jean E. Vance
- Department of Medicine and Group on Molecular and Cell Biology of Lipids; University of Alberta; Edmonton AB Canada
| |
Collapse
|
48
|
Siow D, Sunkara M, Morris A, Wattenberg B. Regulation of de novo sphingolipid biosynthesis by the ORMDL proteins and sphingosine kinase-1. Adv Biol Regul 2014; 57:42-54. [PMID: 25319495 DOI: 10.1016/j.jbior.2014.09.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 09/03/2014] [Accepted: 09/04/2014] [Indexed: 12/11/2022]
Abstract
Sphingolipids are a diverse set of structurally and metabolically related lipids that have numerous functions in cell structure and signaling. The regulation of these lipids is critical for normal cell function and disregulation has been implicated in pathophysiological conditions such as cancer and inflammation. Here we examine control of the initiating, and rate limiting, enzyme in sphingolipid biosynthesis, serine palmitoyltransferase (SPT). We find that de novo synthesis of sphingolipid is stimulated by a number of cancer chemotherapeutics, suggesting that this may be an important aspect of their cytotoxic effects. The three ORMDL proteins are membrane proteins of the endoplasmic reticulum related to the yeast Orm proteins, which have been shown to be homeostatic regulators of SPT. We find that the ORMDL proteins are also negative regulators of SPT that transmit cellular levels of sphingolipids to SPT. The three isoforms have redundant functions in this system. The sphingosine kinases (sphingosine kinase-1 and -2) phosphorylate both sphingosine, which is released from ceramide, but also dihydrosphingosine, which is in the de novo biosynthetic pathway. We therefore examined the role of the sphingosine kinases in controlling de novo ceramide biosynthesis and find that sphingosine kinase-1 does indeed act as a negative regulator of this pathway. This establishes that sphingosine kinase, in addition to producing sphingosine-1-phosphate as a signaling molecule, also consumes dihydrosphingosine to regulate ceramide synthesis. Our studies demonstrate that there are multiple mechanisms of regulation of SPT and suggest that these regulators are important mediators of cell stress responses.
Collapse
Affiliation(s)
- Deanna Siow
- James Graham Brown Cancer Center, University of Louisville School of Medicine, 505 South Hancock St., Louisville, KY 40202, USA
| | - Manjula Sunkara
- Division of Cardiovascular Medicine, Gill Heart Institute, University of Kentucky Lexington, Lexington, KY 40536, USA; Department of Veterans Affairs Medical Center, Lexington, KY, USA
| | - Andrew Morris
- Division of Cardiovascular Medicine, Gill Heart Institute, University of Kentucky Lexington, Lexington, KY 40536, USA; Department of Veterans Affairs Medical Center, Lexington, KY, USA
| | - Binks Wattenberg
- James Graham Brown Cancer Center, University of Louisville School of Medicine, 505 South Hancock St., Louisville, KY 40202, USA; Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, 505 South Hancock St., Louisville, KY 40202, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, 505 South Hancock St., Louisville, KY 40202, USA.
| |
Collapse
|
49
|
Cosentino K, García-Sáez AJ. Mitochondrial alterations in apoptosis. Chem Phys Lipids 2014; 181:62-75. [PMID: 24732580 DOI: 10.1016/j.chemphyslip.2014.04.001] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 03/20/2014] [Accepted: 04/02/2014] [Indexed: 12/31/2022]
Abstract
Besides their conventional role as energy suppliers for the cell, mitochondria in vertebrates are active regulators of apoptosis. They release apoptotic factors from the intermembrane space into the cytosol through a mechanism that involves the Bcl-2 protein family, mediating permeabilization of the outer mitochondrial membrane. Associated with this event, a number of additional changes affect mitochondria during apoptosis. They include loss of important mitochondrial functions, such as the ability to maintain calcium homeostasis and to generate ATP, as well as mitochondrial fragmentation and cristae remodeling. Moreover, the lipidic component of mitochondrial membranes undergoes important alterations in composition and distribution, which have turned out to be relevant regulatory events for the proteins involved in apoptotic mitochondrial damage.
Collapse
Affiliation(s)
- Katia Cosentino
- German Cancer Research Center, Heidelberg, Germany; Max-Planck Institute for Intelligent Systems, Stuttgart, Germany
| | - Ana J García-Sáez
- German Cancer Research Center, Heidelberg, Germany; Max-Planck Institute for Intelligent Systems, Stuttgart, Germany; Interfaculty Institute of Biochemistry (IFIB), University of Tübingen, Tübingen, Germany.
| |
Collapse
|