1
|
Yamada C, Akkaoui J, Morozov A, Movila A. Role of Canonical and Non-Canonical Sphingolipids and their Metabolic Enzymes in Bone Health. Curr Osteoporos Rep 2025; 23:21. [PMID: 40266422 PMCID: PMC12018623 DOI: 10.1007/s11914-025-00908-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/21/2025] [Indexed: 04/24/2025]
Abstract
PURPOSE OF REVIEW This review summarizes the recently published scientific evidence regarding the role of enzymes engaged in de novo anabolic biosynthesis, catabolic, and salvage pathways of ceramide bioactive sphingolipids in bone dynamics and skeletal health. RECENT FINDINGS Ceramides are precursors for bioactive sphingolipids, including sphingosine, sphingosine-1-phosphate, and others. Studies of bone metabolism and bone-related cells demonstrated that ceramide and sphingosine-1-phosphate control levels of bone remodeling and resorption generated by osteoblasts and osteoclasts. Multiple published studies demonstrated the critical role of enzymes in regulating the ceramide/sphingosine-1-phosphate ratio relative to bone physiology and the promotion of inflammatory osteolysis. Accordingly, emerging evidence suggests that targeting sphingolipid metabolism has the potential to alleviate inflammatory osteolysis and accelerate bone regeneration. Therefore, this study aimed to discuss current knowledge about crosstalk between sphingolipids and their metabolic enzymes within osteoclast and osteoblast coupling in bone remodeling and pathogenic osteolysis. This review highlights the complexity of de novo sphingolipid biosynthesis and knowledge gaps in bone physiology and pathology. We also discuss the importance of canonical and non-canonical mammalian and bacterial-derived sphingolipids relative to bone health.
Collapse
Affiliation(s)
- Chiaki Yamada
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
- Richard L. Roudebush Veterans' Administration Medical Center, Indianapolis, IN, USA
| | - Juliet Akkaoui
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Alexandr Morozov
- Institute of Zoology, Moldova State University, Chisinau, Republic of Moldova
- Medpark International Hospital, Chisinau, Republic of Moldova
| | - Alexandru Movila
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, USA.
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush Veterans' Administration Medical Center, Indianapolis, IN, USA.
| |
Collapse
|
2
|
Hornemann T. Sphingoid Base Diversity. Atherosclerosis 2025; 401:119091. [PMID: 39824719 DOI: 10.1016/j.atherosclerosis.2024.119091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 01/20/2025]
Abstract
Sphingolipids (SL) are crucial components of cellular membranes and play pivotal roles in various biological processes, including cell growth, differentiation, apoptosis, and stress responses. All SL contain a sphingoid base (SPB) backbone which is the shared and class-defining element. SPBs are heterogeneous in length and structure. This review summarizes our current understanding on minor SPBs and the role of the serine palmitoyltransferase (SPT) in particular of its subunits SPTLC3 and SPTSSA/B in forming a spectrum of structurally and metabolically distinct SPBs. Some minor SPBs, such as 1-deoxysphingolipids (1-deoxySL) are neurotoxic and associated with neurological disorders such as hereditary sensory neuropathy type 1 (HSAN1) and diabetic neuropathy. Furthermore, the review discusses the pathological implications of atypical SPBs in cardiometabolic conditions such as obesity, type 2 diabetes or cardiomyopathy, where the induction of the SPTLC3 subunit alters the SPB profile and contributes to disease progression. Understanding these, often neglected aspects of the sphingolipid metabolism provides potential therapeutic targets for metabolic and neurodegenerative diseases, emphasizing the need for continued research in this area.
Collapse
Affiliation(s)
- Thorsten Hornemann
- Institute for Clinical Chemistry, University Hospital and University Zurich, 8091, Zürich, Switzerland.
| |
Collapse
|
3
|
Merrill AH. Don't Be Surprised When These Surprise You: Some Infrequently Studied Sphingoid Bases, Metabolites, and Factors That Should Be Kept in Mind During Sphingolipidomic Studies. Int J Mol Sci 2025; 26:650. [PMID: 39859363 PMCID: PMC11765627 DOI: 10.3390/ijms26020650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Sphingolipidomic mass spectrometry has provided valuable information-and surprises-about sphingolipid structures, metabolism, and functions in normal biological processes and disease. Nonetheless, many noteworthy compounds are not routinely determined, such as the following: most of the sphingoid bases that mammals biosynthesize de novo other than sphingosine (and sometimes sphinganine) or acquire from exogenous sources; infrequently considered metabolites of sphingoid bases, such as N-(methyl)n-derivatives; "ceramides" other than the most common N-acylsphingosines; and complex sphingolipids other than sphingomyelins and simple glycosphingolipids, including glucosyl- and galactosylceramides, which are usually reported as "monohexosylceramides". These and other subspecies are discussed, as well as some of the circumstances when they are likely to be seen (or present and missed) due to experimental conditions that can influence sphingolipid metabolism, uptake from the diet or from the microbiome, or as artifacts produced during extraction and analysis. If these compounds and factors are kept in mind during the design and interpretation of lipidomic studies, investigators are likely to be surprised by how often they appear and thereby advance knowledge about them.
Collapse
Affiliation(s)
- Alfred H Merrill
- School of Biological Sciences and The Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
4
|
Wang J, Zhang X, Zhan S, Han F, Wang Q, Liu Y, Huang Z. Possible Metabolic Remodeling based on de novo Biosynthesis of L-serine in Se-Subtoxic or -Deficient Mammals. J Nutr 2025; 155:9-26. [PMID: 39477017 DOI: 10.1016/j.tjnut.2024.10.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/23/2024] [Accepted: 10/20/2024] [Indexed: 11/18/2024] Open
Abstract
Current research studies point to an increased risk of diabetes with selenium (Se) intake beyond the physiological requirement used to prevent cancers. The existing hypothesis of "selenoprotein overexpression leads to intracellular redox imbalance" cannot clearly explain the U-shaped dose-effect relationship between Se intake and the risk of diabetes. In this review, it is speculated that metabolic remodeling based on the de novo biosynthesis of L-serine may occur in mammals at supranutritional or subtoxic levels of Se. It is also speculated that a large amount of L-serine is consumed by the body during insufficient Se intake, thus resulting in similar metabolic reprogramming. The increase in atypical ceramide and its derivatives due to the lack of L-serine may also play a role in the development of diabetes.
Collapse
Affiliation(s)
- Jianrong Wang
- Department of Nutrition and Metabolism, National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, PR China
| | - Xue Zhang
- Department of Nutrition and Metabolism, National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, PR China
| | - Shuo Zhan
- Department of Nutrition and Metabolism, National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, PR China
| | - Feng Han
- Department of Nutrition and Metabolism, National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, PR China
| | - Qin Wang
- Department of Nutrition and Metabolism, National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, PR China
| | - Yiqun Liu
- Department of Nutrition and Metabolism, National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, PR China.
| | - Zhenwu Huang
- Department of Nutrition and Metabolism, National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, PR China; Key Laboratory of Public Nutrition and Health, National Health Commission, Beijing, PR China.
| |
Collapse
|
5
|
Hülsmeier AJ, Gunasegaram L, Wipfli F, Lone MA, Hornemann T. Long Chain Base Profiling with Multiple Reaction Monitoring Mass Spectrometry. Methods Mol Biol 2025; 2855:209-223. [PMID: 39354311 DOI: 10.1007/978-1-0716-4116-3_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Sphingolipids (SLs) are essential lipids with important functions in membrane formation and cell signaling. The presence of a long chain base (LCB) structure is common to all SLs. De novo SL synthesis is initiated by the enzyme serine-palmitoyltransferase (SPT), which forms an LCB by the conjugation from serine and fatty acyl-CoAs. SPT can metabolize a variety of acyl-CoA substrates, which form diverse LCB structures within and across species. The LCB then undergoes further metabolic modifications resulting in an extraordinarily diverse spectrum of sphingolipids formed. SL analysis, using liquid chromatography-mass spectrometry (LC-MS)-based methods, poses challenges due to the diverse range of frequently isobaric species. This complexity complicates the identification of underlying LCB structures using standard lipidomics approaches. Here, we describe a simplified method to analyze the LCB profile in cells, tissue, and blood. The procedure involves chemical hydrolysis to remove the conjugated headgroups and N-acyl chains, allowing to specifically resolve the underlying LCB structures by LC-MS. This method can also be combined with an isotope labeling approach to determine in vivo SPT activity and total SL de novo synthesis over time.
Collapse
Affiliation(s)
- Andreas J Hülsmeier
- Institute of Clinical Chemistry, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Lavanya Gunasegaram
- Institute of Clinical Chemistry, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Florine Wipfli
- Institute of Clinical Chemistry, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Museer A Lone
- Institute of Clinical Chemistry, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Thorsten Hornemann
- Institute of Clinical Chemistry, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
6
|
Byeon SK, Kim J, Wegwerth PJ, Zenka R, George JP, Pinto E Vairo F, Oglesbee D, Schultz MJ, Matern D, Pandey A. Development of a Multiplexed Sphingolipids Method for Diagnosis of Inborn Errors of Ceramide Metabolism. Clin Chem 2024; 70:1366-1374. [PMID: 39206579 DOI: 10.1093/clinchem/hvae115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 07/08/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Sphingolipids play a crucial role in cellular functions and are essential components of cell membranes, signaling molecules, and lipid metabolism. In particular, ceramide is a key intermediate in sphingolipid metabolism and defects in ceramide metabolism can lead to various inborn errors of metabolism, making ceramides important targets for clinical screening and diagnosis. Detecting altered concentration patterns of sphingolipids is desirable for distinguishing related inborn errors of metabolism for diagnosis and treatment monitoring. METHODS We developed a liquid chromatography-tandem mass spectrometry method with a pathway-oriented approach to focus on sphingolipids involved in ceramide metabolism. A total of 47 sphingolipids bearing different head groups and side chains were targeted. Precision/reproducibility, linearity, and spike recovery extraction efficiency tests were performed on plasma and serum samples from confirmed cases of sphingolipidosis. RESULTS Linearity of the method showed the coefficient of determination (r2) for all standards to be >0.99 with a slope of 1.00 ± 0.01. Intra- and interday reproducibility of standards spiked into plasma and serum revealed a coefficient of variation <20%. Spike and recovery assessment showed recovery values of 80%-120% for all standards. Altered levels of sphingolipids from patients with hereditary sensory and autonomic neuropathy caused by pathogenic variants in SPTLC2 and hypomyelinating leukodystrophy related to variants in DEGS1 were detected, in agreement with trends reported in earlier studies confirming the utility of this pathway-centric method. CONCLUSIONS This method can serve as a useful tool to simultaneously monitor sphingolipids, enabling screening and diagnosis of inborn errors of ceramide metabolism.
Collapse
Affiliation(s)
- Seul Kee Byeon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Jinyong Kim
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Peter Jared Wegwerth
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Roman Zenka
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - John P George
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
- Manipal Academy of Higher Education, Manipal, Karnataka, India
- Institute of Bioinformatics, International Technology Park, Bangalore, India
| | - Filippo Pinto E Vairo
- Center for Individualized Medicine, Mayo Clinic, Rochester, MNUnited States
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, United States
| | - Devin Oglesbee
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Matthew J Schultz
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Dietrich Matern
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
- Manipal Academy of Higher Education, Manipal, Karnataka, India
- Center for Individualized Medicine, Mayo Clinic, Rochester, MNUnited States
| |
Collapse
|
7
|
González-Ramírez EJ, García-Arribas AB, Artetxe I, Shaw WA, Goñi FM, Alonso A, Jiménez-Rojo N. (1-Deoxy)ceramides in bilayers containing sphingomyelin and cholesterol. Colloids Surf B Biointerfaces 2024; 243:114155. [PMID: 39137529 DOI: 10.1016/j.colsurfb.2024.114155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024]
Abstract
The discovery of a novel sphingolipid subclass, the (1-deoxy)sphingolipids, which lack the 1-hydroxy group, attracted considerable attention in the last decade, mainly due to their involvement in disease. They differed in their physico-chemical properties from the canonical (or 1-hydroxy) sphingolipids and they were more toxic when accumulated in cells, inducing neurodegeneration and other dysfunctions. (1-Deoxy)ceramides, (1-deoxy)dihydroceramides, and (1- deoxymethyl)dihydroceramides, the latter two containing a saturated sphingoid chain, have been studied in this work using differential scanning calorimetry, confocal fluorescence and atomic force microscopy, to evaluate their behavior in bilayers composed of mixtures of three or four lipids. When compared to canonical ceramides (Cer), a C16:0 (1-deoxy)Cer shows a lower miscibility in mixtures of the kind C16:0 sphingomyelin/cholesterol/XCer, where XCer is any (1-deoxy)ceramide, giving rise to the coexistence of a liquid-ordered phase and a gel phase. The latter resembles, in terms of thermotropic behavior and nanomechanical resistance, the gel phase of the C16:0 sphingomyelin/cholesterol/C16:0 Cer mixture [Busto et al., Biophys. J. 2014, 106, 621-630]. Differences are seen between the various C16:0 XCer under study in terms of nanomechanical resistance, bilayer thickness and bilayer topography. When examined in a more fluid environment (bilayers based on C24:1 SM), segregated gel phases are still present. Probably related to such lateral separation, XCer preserve the capacity for membrane permeation, but their effects are significantly lower than those of canonical ceramides. Moreover, C24:1 XCer show significantly lower membrane permeation capacity than their C16:0 counterparts. The above data may be relevant in the pathogenesis of certain sphingolipid-related diseases, including certain neuropathies, diabetes, and glycogen storage diseases.
Collapse
Affiliation(s)
- E J González-Ramírez
- Instituto Biofisika (CSIC, UPV/EHU) and Department of Biochemistry, University of the Basque Country, Leioa, 48940, Spain
| | - A B García-Arribas
- Instituto Biofisika (CSIC, UPV/EHU) and Department of Biochemistry, University of the Basque Country, Leioa, 48940, Spain
| | - I Artetxe
- Instituto Biofisika (CSIC, UPV/EHU) and Department of Biochemistry, University of the Basque Country, Leioa, 48940, Spain
| | - W A Shaw
- Avanti Polar Lipids, Alabaster, AL, USA
| | - F M Goñi
- Instituto Biofisika (CSIC, UPV/EHU) and Department of Biochemistry, University of the Basque Country, Leioa, 48940, Spain
| | - A Alonso
- Instituto Biofisika (CSIC, UPV/EHU) and Department of Biochemistry, University of the Basque Country, Leioa, 48940, Spain.
| | - N Jiménez-Rojo
- Instituto Biofisika (CSIC, UPV/EHU) and Department of Biochemistry, University of the Basque Country, Leioa, 48940, Spain.
| |
Collapse
|
8
|
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
|
9
|
Tzou FY, Hornemann T, Yeh JY, Huang SY. The pathophysiological role of dihydroceramide desaturase in the nervous system. Prog Lipid Res 2023; 91:101236. [PMID: 37187315 DOI: 10.1016/j.plipres.2023.101236] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 04/18/2023] [Accepted: 05/12/2023] [Indexed: 05/17/2023]
Abstract
Dihydroceramide desaturase 1 (DEGS1) converts dihydroceramide (dhCer) to ceramide (Cer) by inserting a C4-C5 trans (∆4E) double bond into the sphingoid backbone. Low DEGS activity causes accumulation of dhCer and other dihydrosphingolipid species. Although dhCer and Cer are structurally very similar, their imbalances can have major consequences both in vitro and in vivo. Mutations in the human DEGS1 gene are known to cause severe neurological defects, such as hypomyelinating leukodystrophy. Likewise, inhibition of DEGS1 activity in fly and zebrafish models causes dhCer accumulation and subsequent neuronal dysfunction, suggesting that DEGS1 activity plays a conserved and critical role in the nervous system. Dihydrosphingolipids and their desaturated counterparts are known to control various essential processes, including autophagy, exosome biogenesis, ER stress, cell proliferation, and cell death. Furthermore, model membranes with either dihydrosphingolipids or sphingolipids exhibit different biophysical properties, including membrane permeability and packing, thermal stability, and lipid diffusion. However, the links between molecular properties, in vivo functional data, and clinical manifestations that underlie impaired DEGS1 function remain largely unresolved. In this review, we summarize the known biological and pathophysiological roles of dhCer and its derivative dihydrosphingolipid species in the nervous system, and we highlight several possible disease mechanisms that warrant further investigation.
Collapse
Affiliation(s)
- Fei-Yang Tzou
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Thorsten Hornemann
- Institute for Clinical Chemistry, University Hospital and University Zurich, 8091 Zürich, Switzerland
| | - Jui-Yu Yeh
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shu-Yi Huang
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
10
|
Jojima K, Kihara A. Metabolism of sphingadiene and characterization of the sphingadiene-producing enzyme FADS3. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159335. [PMID: 37209771 DOI: 10.1016/j.bbalip.2023.159335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/22/2023]
Abstract
Of the long-chain bases (LCBs) that comprise the ceramides (CERs) present in mammals, only 4,14-sphingadiene (sphingadiene; SPD) has a cis double bond (at C14). Because of this unique structure, the metabolism of SPD may differ from that of other LCBs, but whether this is the case remains unclear. FADS3 is responsible for introducing the cis double bond in SPD. However, the substrate specificity of FADS3 and cofactors involved in the FADS3-catalyzed reaction are also unknown. In the present study, a cell-based assay using a ceramide synthase inhibitor and an in vitro experiment showed that FADS3 is active toward sphingosine (SPH)-containing CERs (SPH-CERs) but not toward free SPH. FADS3 exhibits specificity with respect to the chain length of the SPH moiety of SPH-CERs (active toward C16-20), but not that of the fatty acid moiety. Furthermore, FADS3 is active toward straight-chain and isobranched-chain SPH-containing CERs but not toward anteiso-branched forms. In addition to SPH-CERs, FADS3 also shows activity toward dihydrosphingosine-containing CERs, but this activity is approximately half of that toward SPH-CERs. It uses either NADH or NADPH as an electron donor, and the electron transfer is facilitated by cytochrome b5. The metabolic flow of SPD to sphingomyelin is predominant over that to glycosphingolipids. In the metabolic pathway from SPD to fatty acids, the chain length of the SPD is reduced by two carbons and the trans double bond at C4 is saturated. This study thus elucidates the enzymatic properties of FADS3 and the metabolism of SPD.
Collapse
Affiliation(s)
- Keisuke Jojima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Akio Kihara
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan.
| |
Collapse
|
11
|
Camunas-Alberca SM, Moran-Garrido M, Sáiz J, Gil-de-la-Fuente A, Barbas C, Gradillas A. Integrating the potential of ion mobility spectrometry-mass spectrometry in the separation and structural characterisation of lipid isomers. Front Mol Biosci 2023; 10:1112521. [PMID: 37006618 PMCID: PMC10060977 DOI: 10.3389/fmolb.2023.1112521] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/14/2023] [Indexed: 03/18/2023] Open
Abstract
It is increasingly evident that a more detailed molecular structure analysis of isomeric lipids is critical to better understand their roles in biological processes. The occurrence of isomeric interference complicates conventional tandem mass spectrometry (MS/MS)-based determination, necessitating the development of more specialised methodologies to separate lipid isomers. The present review examines and discusses recent lipidomic studies based on ion mobility spectrometry combined with mass spectrometry (IMS-MS). Selected examples of the separation and elucidation of structural and stereoisomers of lipids are described based on their ion mobility behaviour. These include fatty acyls, glycerolipids, glycerophospholipids, sphingolipids, and sterol lipids. Recent approaches for specific applications to improve isomeric lipid structural information using direct infusion, coupling imaging, or liquid chromatographic separation workflows prior to IMS-MS are also discussed, including: 1) strategies to improve ion mobility shifts; 2) advanced tandem MS methods based on activation of lipid ions with electrons or photons, or gas-phase ion-molecule reactions; and 3) the use of chemical derivatisation techniques for lipid characterisation.
Collapse
Affiliation(s)
- Sandra M. Camunas-Alberca
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - Maria Moran-Garrido
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - Jorge Sáiz
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - Alberto Gil-de-la-Fuente
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Departamento de Tecnologías de la Información, Escuela Politécnica Superior, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - Coral Barbas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - Ana Gradillas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- *Correspondence: Ana Gradillas,
| |
Collapse
|
12
|
Varshney R, Das S, Trahan GD, Farriester JW, Mullen GP, Kyere-Davies G, Presby DM, Houck JA, Webb PG, Dzieciatkowska M, Jones KL, Rodeheffer MS, Friedman JE, MacLean PS, Rudolph MC. Neonatal intake of Omega-3 fatty acids enhances lipid oxidation in adipocyte precursors. iScience 2023; 26:105750. [PMID: 36590177 PMCID: PMC9800552 DOI: 10.1016/j.isci.2022.105750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 09/26/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Establishing metabolic programming begins during fetal and postnatal development, and early-life lipid exposures play a critical role during neonatal adipogenesis. We define how neonatal consumption of a low omega-6 to -3 fatty acid ratio (n6/n3 FA ratio) establishes FA oxidation in adipocyte precursor cells (APCs) before they become adipocytes. In vivo, APCs isolated from mouse pups exposed to the low n6/n3 FA ratio had superior FA oxidation capacity, elevated beige adipocyte mRNAs Ppargc1α, Ucp2, and Runx1, and increased nuclear receptor NR2F2 protein. In vitro, APC treatment with NR2F2 ligand-induced beige adipocyte mRNAs and increased mitochondrial potential but not mass. Single-cell RNA-sequencing analysis revealed low n6/n3 FA ratio yielded more mitochondrial-high APCs and linked APC NR2F2 levels with beige adipocyte signatures and FA oxidation. Establishing beige adipogenesis is of clinical relevance, because fat depots with energetically active, smaller, and more numerous adipocytes improve metabolism and delay metabolic dysfunction.
Collapse
Affiliation(s)
- Rohan Varshney
- Harold Hamm Diabetes Center and Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Snehasis Das
- Harold Hamm Diabetes Center and Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - G. Devon Trahan
- Department of Pediatrics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Jacob W. Farriester
- Harold Hamm Diabetes Center and Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Gregory P. Mullen
- Harold Hamm Diabetes Center and Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Gertrude Kyere-Davies
- Harold Hamm Diabetes Center and Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - David M. Presby
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Julie A. Houck
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Patricia G. Webb
- Department of Reproductive Science, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry & Molecular Genetics, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Kenneth L. Jones
- Department of Cell Biology and Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Matthew S. Rodeheffer
- Department of Molecular, Cellular and Developmental Biology, Department of Comparative Medicine, Yale University, New Haven, CT, USA
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center and Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Paul S. MacLean
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Michael C. Rudolph
- Harold Hamm Diabetes Center and Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
13
|
Izquierdo E, López-Corrales M, Abad-Montero D, Rovira A, Fabriàs G, Bosch M, Abad JL, Marchán V. Fluorescently Labeled Ceramides and 1-Deoxyceramides: Synthesis, Characterization, and Cellular Distribution Studies. J Org Chem 2022; 87:16351-16367. [PMID: 36441972 PMCID: PMC9764360 DOI: 10.1021/acs.joc.2c02019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ceramides (Cer) are bioactive sphingolipids that have been proposed as potential disease biomarkers since they are involved in several cellular stress responses, including apoptosis and senescence. 1-Deoxyceramides (1-deoxyCer), a particular subtype of noncanonical sphingolipids, have been linked to the pathogenesis of type II diabetes. To investigate the metabolism of these bioactive lipids, as well as to have a better understanding of the signaling processes where they participate, it is essential to expand the toolbox of fluorescent sphingolipid probes exhibiting complementary subcellular localization. Herein, we describe a series of new sphingolipid probes tagged with two different organic fluorophores, a far-red/NIR-emitting coumarin derivative (COUPY) and a green-emitting BODIPY. The assembly of the probes involved a combination of olefin cross metathesis and click chemistry reactions as key steps, and these fluorescent ceramide analogues exhibited excellent emission quantum yields, being the Stokes' shifts of the COUPY derivatives much higher than those of the BODIPY counterparts. Confocal microscopy studies in HeLa cells confirmed an excellent cellular permeability for these sphingolipid probes and revealed that most of the vesicles stained by COUPY probes were either lysosomes or endosomes, whereas BODIPY probes accumulated either in Golgi apparatus or in nonlysosomal intracellular vesicles. The fact that the two sets of fluorescent Cer probes have such different staining patterns indicates that their subcellular distribution is not entirely defined by the sphingolipid moiety but rather influenced by the fluorophore.
Collapse
Affiliation(s)
- Eduardo Izquierdo
- Departament
de Química Inorgànica i Orgànica, Secció
de Química Orgànica, Universitat de Barcelona (UB), Martí i Franquès 1-11, 08028Barcelona, Spain
| | - Marta López-Corrales
- Departament
de Química Inorgànica i Orgànica, Secció
de Química Orgànica, Universitat de Barcelona (UB), Martí i Franquès 1-11, 08028Barcelona, Spain
| | - Diego Abad-Montero
- Departament
de Química Inorgànica i Orgànica, Secció
de Química Orgànica, Universitat de Barcelona (UB), Martí i Franquès 1-11, 08028Barcelona, Spain,Research
Unit on BioActive Molecules, Departament de Química Biològica, Institut de Química Avançada de Catalunya
(IQAC-CSIC), Jordi Girona
18-26, 08034Barcelona, Spain
| | - Anna Rovira
- Departament
de Química Inorgànica i Orgànica, Secció
de Química Orgànica, Universitat de Barcelona (UB), Martí i Franquès 1-11, 08028Barcelona, Spain
| | - Gemma Fabriàs
- Research
Unit on BioActive Molecules, Departament de Química Biològica, Institut de Química Avançada de Catalunya
(IQAC-CSIC), Jordi Girona
18-26, 08034Barcelona, Spain
| | - Manel Bosch
- Unitat
de Microscòpia Òptica Avanc̨ada, Centres Científics
i Tecnològics, Universitat de Barcelona
(UB), Av. Diagonal, 643, 08028Barcelona, Spain
| | - José Luís Abad
- Research
Unit on BioActive Molecules, Departament de Química Biològica, Institut de Química Avançada de Catalunya
(IQAC-CSIC), Jordi Girona
18-26, 08034Barcelona, Spain,
| | - Vicente Marchán
- Departament
de Química Inorgànica i Orgànica, Secció
de Química Orgànica, Universitat de Barcelona (UB), Martí i Franquès 1-11, 08028Barcelona, Spain,Institut
de Biomedicina de la Universitat de Barcelona (IBUB), 08028Barcelona, Spain,
| |
Collapse
|
14
|
Paglia G, Smith AJ, Astarita G. Ion mobility mass spectrometry in the omics era: Challenges and opportunities for metabolomics and lipidomics. MASS SPECTROMETRY REVIEWS 2022; 41:722-765. [PMID: 33522625 DOI: 10.1002/mas.21686] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 01/17/2021] [Accepted: 01/17/2021] [Indexed: 06/12/2023]
Abstract
Researchers worldwide are taking advantage of novel, commercially available, technologies, such as ion mobility mass spectrometry (IM-MS), for metabolomics and lipidomics applications in a variety of fields including life, biomedical, and food sciences. IM-MS provides three main technical advantages over traditional LC-MS workflows. Firstly, in addition to mass, IM-MS allows collision cross-section values to be measured for metabolites and lipids, a physicochemical identifier related to the chemical shape of an analyte that increases the confidence of identification. Second, IM-MS increases peak capacity and the signal-to-noise, improving fingerprinting as well as quantification, and better defining the spatial localization of metabolites and lipids in biological and food samples. Third, IM-MS can be coupled with various fragmentation modes, adding new tools to improve structural characterization and molecular annotation. Here, we review the state-of-the-art in IM-MS technologies and approaches utilized to support metabolomics and lipidomics applications and we assess the challenges and opportunities in this growing field.
Collapse
Affiliation(s)
- Giuseppe Paglia
- School of Medicine and Surgery, University of Milano-Bicocca, Vedano al Lambro (MB), Italy
| | - Andrew J Smith
- School of Medicine and Surgery, University of Milano-Bicocca, Vedano al Lambro (MB), Italy
| | - Giuseppe Astarita
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, District of Columbia, USA
| |
Collapse
|
15
|
Thomas JM, Sudhadevi T, Basa P, Ha AW, Natarajan V, Harijith A. The Role of Sphingolipid Signaling in Oxidative Lung Injury and Pathogenesis of Bronchopulmonary Dysplasia. Int J Mol Sci 2022; 23:ijms23031254. [PMID: 35163176 PMCID: PMC8835774 DOI: 10.3390/ijms23031254] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
Premature infants are born with developing lungs burdened by surfactant deficiency and a dearth of antioxidant defense systems. Survival rate of such infants has significantly improved due to advances in care involving mechanical ventilation and oxygen supplementation. However, a significant subset of such survivors develops the chronic lung disease, Bronchopulmonary dysplasia (BPD), characterized by enlarged, simplified alveoli and deformed airways. Among a host of factors contributing to the pathogenesis is oxidative damage induced by exposure of the developing lungs to hyperoxia. Recent data indicate that hyperoxia induces aberrant sphingolipid signaling, leading to mitochondrial dysfunction and abnormal reactive oxygen species (ROS) formation (ROS). The role of sphingolipids such as ceramides and sphingosine 1-phosphate (S1P), in the development of BPD emerged in the last decade. Both ceramide and S1P are elevated in tracheal aspirates of premature infants of <32 weeks gestational age developing BPD. This was faithfully reflected in the murine models of hyperoxia and BPD, where there is an increased expression of sphingolipid metabolites both in lung tissue and bronchoalveolar lavage. Treatment of neonatal pups with a sphingosine kinase1 specific inhibitor, PF543, resulted in protection against BPD as neonates, accompanied by improved lung function and reduced airway remodeling as adults. This was accompanied by reduced mitochondrial ROS formation. S1P receptor1 induced by hyperoxia also aggravates BPD, revealing another potential druggable target in this pathway for BPD. In this review we aim to provide a detailed description on the role played by sphingolipid signaling in hyperoxia induced lung injury and BPD.
Collapse
Affiliation(s)
- Jaya M. Thomas
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
| | - Tara Sudhadevi
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
| | - Prathima Basa
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
| | - Alison W. Ha
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Viswanathan Natarajan
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA;
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Anantha Harijith
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
- Correspondence: ; Tel.: +1-(216)-286-7038
| |
Collapse
|
16
|
Wang T, Wang Z, de Fabritus L, Tao J, Saied EM, Lee HJ, Ramazanov BR, Jackson B, Burkhardt D, Parker M, Gleinich AS, Wang Z, Seo DE, Zhou T, Xu S, Alecu I, Azadi P, Arenz C, Hornemann T, Krishnaswamy S, van de Pavert SA, Kaech SM, Ivanova NB, Santori FR. 1-deoxysphingolipids bind to COUP-TF to modulate lymphatic and cardiac cell development. Dev Cell 2021; 56:3128-3145.e15. [PMID: 34762852 PMCID: PMC8628544 DOI: 10.1016/j.devcel.2021.10.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 08/30/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022]
Abstract
Identification of physiological modulators of nuclear hormone receptor (NHR) activity is paramount for understanding the link between metabolism and transcriptional networks that orchestrate development and cellular physiology. Using libraries of metabolic enzymes alongside their substrates and products, we identify 1-deoxysphingosines as modulators of the activity of NR2F1 and 2 (COUP-TFs), which are orphan NHRs that are critical for development of the nervous system, heart, veins, and lymphatic vessels. We show that these non-canonical alanine-based sphingolipids bind to the NR2F1/2 ligand-binding domains (LBDs) and modulate their transcriptional activity in cell-based assays at physiological concentrations. Furthermore, inhibition of sphingolipid biosynthesis phenocopies NR2F1/2 deficiency in endothelium and cardiomyocytes, and increases in 1-deoxysphingosine levels activate NR2F1/2-dependent differentiation programs. Our findings suggest that 1-deoxysphingosines are physiological regulators of NR2F1/2-mediated transcription.
Collapse
Affiliation(s)
- Ting Wang
- Department of Immunobiology, Yale University, New Haven, CT 06520, USA; Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zheng Wang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, Shandong 266071, China; Department of Reproductive Medicine, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Lauriane de Fabritus
- Aix-Marseille Universite, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), 13288 Marseille Cedex 9, France
| | - Jinglian Tao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China; Center for Molecular Medicine, Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Essa M Saied
- Institut für Chemie, Humboldt Universität zu Berlin, Berlin 12489, Germany; Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| | - Ho-Joon Lee
- Department of Genetics, Yale University, New Haven, CT 06520, USA; Center for Genome Analysis, Yale University, New Haven, CT 06510, USA
| | - Bulat R Ramazanov
- Department of Cell Biology, Yale University, New Haven, CT 06520, USA
| | - Benjamin Jackson
- Center for Molecular Medicine, Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Daniel Burkhardt
- Department of Genetics, Yale University, New Haven, CT 06520, USA
| | - Mikhail Parker
- Center for Molecular Medicine, Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Anne S Gleinich
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Zhirui Wang
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Dong Eun Seo
- Center for Molecular Medicine, Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Ting Zhou
- Department of Immunobiology, Yale University, New Haven, CT 06520, USA
| | - Shihao Xu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, the Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Irina Alecu
- Neural Regeneration Laboratory, Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Christoph Arenz
- Institut für Chemie, Humboldt Universität zu Berlin, Berlin 12489, Germany
| | - Thorsten Hornemann
- Institute of Clinical Chemistry, University and University Hospital of Zurich, Zurich 8091, Switzerland
| | | | - Serge A van de Pavert
- Aix-Marseille Universite, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), 13288 Marseille Cedex 9, France
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, the Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| | - Natalia B Ivanova
- Center for Molecular Medicine, Department of Genetics, University of Georgia, Athens, GA 30602, USA.
| | - Fabio R Santori
- Department of Immunobiology, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
17
|
Karsai G, Steiner R, Kaech A, Lone MA, von Eckardstein A, Hornemann T. Metabolism of HSAN1- and T2DM-associated 1-deoxy-sphingolipids inhibits the migration of fibroblasts. J Lipid Res 2021; 62:100122. [PMID: 34563520 PMCID: PMC8521209 DOI: 10.1016/j.jlr.2021.100122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 11/03/2022] Open
Abstract
Hereditary sensory neuropathy type 1 (HSAN1) is a rare axonopathy, characterized by a progressive loss of sensation (pain, temperature, and vibration), neuropathic pain, and wound healing defects. HSAN1 is caused by several missense mutations in the serine palmitoyltransferase long-chain base subunit 1 and serine palmitoyltransferase long-chain base subunit 2 of the enzyme serine palmitoyltransferase-the key enzyme for the synthesis of sphingolipids. The mutations change the substrate specificity of serine palmitoyltransferase, which then forms an atypical class of 1-deoxy-sphinglipids (1-deoxySLs). Similarly, patients with type 2 diabetes mellitus also present with elevated 1-deoxySLs and a comparable clinical phenotype. The effect of 1-deoxySLs on neuronal cells was investigated in detail, but their impact on other cell types remains elusive. Here, we investigated the consequences of externally added 1-deoxySLs on the migration of fibroblasts in a scratch assay as a simplified cellular wound-healing model. We showed that 1-deoxy-sphinganine (1-deoxySA) inhibits the migration of NIH-3T3 fibroblasts in a dose- and time-dependent manner. This was not seen for a non-native, L-threo stereoisomer. Supplemented 1-deoxySA was metabolized to 1-deoxy-(dihydro)ceramide and downstream to 1-deoxy-sphingosine. Inhibiting downstream metabolism by blocking N-acylation rescued the migration phenotype. In contrast, adding 1-deoxy-sphingosine had a lesser effect on cell migration but caused the massive formation of intracellular vacuoles. Further experiments showed that the effect on cell migration was primarily mediated by 1-deoxy-dihydroceramides rather than by the free base or 1-deoxyceramides. Based on these findings, we suggest that limiting the N-acylation of 1-deoxySA could be a therapeutic approach to improve cell migration and wound healing in patients with HSAN1 and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Gergely Karsai
- Institute of Clinical Chemistry, University Hospital Zürich, Zürich, Switzerland
| | - Regula Steiner
- Institute of Clinical Chemistry, University Hospital Zürich, Zürich, Switzerland
| | - Andres Kaech
- Center for Microscopy and Image Analysis, University of Zürich, Zürich, Switzerland
| | - Museer A Lone
- Institute of Clinical Chemistry, University Hospital Zürich, Zürich, Switzerland
| | | | - Thorsten Hornemann
- Institute of Clinical Chemistry, University Hospital Zürich, Zürich, Switzerland.
| |
Collapse
|
18
|
Stereoselective Synthesis of Novel Sphingoid Bases Utilized for Exploring the Secrets of Sphinx. Int J Mol Sci 2021; 22:ijms22158171. [PMID: 34360937 PMCID: PMC8347175 DOI: 10.3390/ijms22158171] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/11/2022] Open
Abstract
Sphingolipids are ubiquitous in eukaryotic plasma membranes and play major roles in human and animal physiology and disease. This class of lipids is usually defined as being derivatives of sphingosine, a long-chain 1,3-dihydroxy-2-amino alcohol. Various pathological conditions such as diabetes or neuropathy have been associated with changes in the sphingolipidome and an increased biosynthesis of structurally altered non-canonical sphingolipid derivatives. These unusual or non-canonical sphingolipids hold great promise as potential diagnostic markers. However, due to their low concentrations and the unavailability of suitable standards, the research to explore the secret of this class of 'Sphinx' lipids is ultimately hampered. Therefore, the development of efficient and facile syntheses of standard compounds is a key endeavor. Here, we present various chemical approaches for stereoselective synthesis and in-depth chemical characterization of a set of novel sphingoid bases which were recently utilized as valuable tools to explore the metabolism and biophysical properties of sphingolipids, but also to develop efficient analytical methods for their detection and quantification.
Collapse
|
19
|
Ceramide Metabolism Enzymes-Therapeutic Targets against Cancer. ACTA ACUST UNITED AC 2021; 57:medicina57070729. [PMID: 34357010 PMCID: PMC8303233 DOI: 10.3390/medicina57070729] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022]
Abstract
Sphingolipids are both structural molecules that are essential for cell architecture and second messengers that are involved in numerous cell functions. Ceramide is the central hub of sphingolipid metabolism. In addition to being the precursor of complex sphingolipids, ceramides induce cell cycle arrest and promote cell death and inflammation. At least some of the enzymes involved in the regulation of sphingolipid metabolism are altered in carcinogenesis, and some are targets for anticancer drugs. A number of scientific reports have shown how alterations in sphingolipid pools can affect cell proliferation, survival and migration. Determination of sphingolipid levels and the regulation of the enzymes that are implicated in their metabolism is a key factor for developing novel therapeutic strategies or improving conventional therapies. The present review highlights the importance of bioactive sphingolipids and their regulatory enzymes as targets for therapeutic interventions with especial emphasis in carcinogenesis and cancer dissemination.
Collapse
|
20
|
Quinville BM, Deschenes NM, Ryckman AE, Walia JS. A Comprehensive Review: Sphingolipid Metabolism and Implications of Disruption in Sphingolipid Homeostasis. Int J Mol Sci 2021; 22:ijms22115793. [PMID: 34071409 PMCID: PMC8198874 DOI: 10.3390/ijms22115793] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 12/16/2022] Open
Abstract
Sphingolipids are a specialized group of lipids essential to the composition of the plasma membrane of many cell types; however, they are primarily localized within the nervous system. The amphipathic properties of sphingolipids enable their participation in a variety of intricate metabolic pathways. Sphingoid bases are the building blocks for all sphingolipid derivatives, comprising a complex class of lipids. The biosynthesis and catabolism of these lipids play an integral role in small- and large-scale body functions, including participation in membrane domains and signalling; cell proliferation, death, migration, and invasiveness; inflammation; and central nervous system development. Recently, sphingolipids have become the focus of several fields of research in the medical and biological sciences, as these bioactive lipids have been identified as potent signalling and messenger molecules. Sphingolipids are now being exploited as therapeutic targets for several pathologies. Here we present a comprehensive review of the structure and metabolism of sphingolipids and their many functional roles within the cell. In addition, we highlight the role of sphingolipids in several pathologies, including inflammatory disease, cystic fibrosis, cancer, Alzheimer’s and Parkinson’s disease, and lysosomal storage disorders.
Collapse
|
21
|
Santos TCB, Saied EM, Arenz C, Fedorov A, Prieto M, Silva LC. The long chain base unsaturation has a stronger impact on 1-deoxy(methyl)-sphingolipids biophysical properties than the structure of its C1 functional group. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183628. [PMID: 33915167 DOI: 10.1016/j.bbamem.2021.183628] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/13/2021] [Accepted: 04/17/2021] [Indexed: 12/22/2022]
Abstract
1-deoxy-sphingolipids, also known as atypical sphingolipids, are directly implicated in the development and progression of hereditary sensory and autonomic neuropathy type 1 and diabetes type 2. The mechanisms underlying their patho-physiological actions are yet to be elucidated. Accumulating evidence suggests that the biological actions of canonical sphingolipids are triggered by changes promoted on membrane organization and biophysical properties. However, little is known regarding the biophysical implications of atypical sphingolipids. In this study, we performed a comprehensive characterization of the effects of the naturally occurring 1-deoxy-dihydroceramide, 1-deoxy-ceramideΔ14Z and 1-deoxymethyl-ceramideΔ3E in the properties of a fluid membrane. In addition, to better define which structural features determine sphingolipid ability to form ordered domains, the synthetic 1-O-methyl-ceramideΔ4E and 1-deoxy-ceramideΔ4E were also studied. Our results show that natural and synthetic 1-deoxy(methyl)-sphingolipids fail to laterally segregate into ordered domains as efficiently as the canonical C16-ceramide. The impaired ability of atypical sphingolipids to form ordered domains was more dependent on the presence, position, and configuration of the sphingoid base double bond than on the structure of its C1 functional group, due to packing constraints introduced by an unsaturated backbone. Nonetheless, absence of a hydrogen bond donor and acceptor group at the C1 position strongly reduced the capacity of atypical sphingolipids to form gel domains. Altogether, the results showed that 1-deoxy(methyl)-sphingolipids induce unique changes on the biophysical properties of the membranes, suggesting that these alterations might, in part, trigger the patho-biological actions of these lipids.
Collapse
Affiliation(s)
- Tania C B Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Prof. Gama Pinto, Ed F, 1649-003 Lisbon, Portugal; iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Essa M Saied
- Humboldt Universität zu Berlin, Institute for Chemistry, Brook Taylor Str. 2, 12489 Berlin, Germany; Chemistry Department, Faculty of Science, Suez Canal University, The Ring Road km 4.5, Ismailia, Egypt
| | - Christoph Arenz
- Humboldt Universität zu Berlin, Institute for Chemistry, Brook Taylor Str. 2, 12489 Berlin, Germany
| | - Aleksander Fedorov
- iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Manuel Prieto
- iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Liana C Silva
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Prof. Gama Pinto, Ed F, 1649-003 Lisbon, Portugal.
| |
Collapse
|
22
|
Lam BWS, Yam TYA, Chen CP, Lai MKP, Ong WY, Herr DR. The noncanonical chronicles: Emerging roles of sphingolipid structural variants. Cell Signal 2020; 79:109890. [PMID: 33359087 DOI: 10.1016/j.cellsig.2020.109890] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 02/08/2023]
Abstract
Sphingolipids (SPs) are structurally diverse and represent one of the most quantitatively abundant classes of lipids in mammalian cells. In addition to their structural roles, many SP species are known to be bioactive mediators of essential cellular processes. Historically, studies have focused on SP species that contain the canonical 18‑carbon, mono-unsaturated sphingoid backbone. However, increasingly sensitive analytical technologies, driven by advances in mass spectrometry, have facilitated the identification of previously under-appreciated, molecularly distinct SP species. Many of these less abundant species contain noncanonical backbones. Interestingly, a growing number of studies have identified clinical associations between these noncanonical SPs and disease, suggesting that there is functional significance to the alteration of SP backbone structure. For example, associations have been found between SP chain length and cardiovascular disease, pain, diabetes, and dementia. This review will provide an overview of the processes that are known to regulate noncanonical SP accumulation, describe the clinical correlations reported for these molecules, and review the experimental evidence for the potential functional implications of their dysregulation. It is likely that further scrutiny of noncanonical SPs may provide new insight into pathophysiological processes, serve as useful biomarkers for disease, and lead to the design of novel therapeutic strategies.
Collapse
Affiliation(s)
- Brenda Wan Shing Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ting Yu Amelia Yam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Memory Aging and Cognition Centre, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Memory Aging and Cognition Centre, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wei-Yi Ong
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Deron R Herr
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Biology, San Diego State University, San Diego, CA, USA; American University of Health Sciences, Long Beach, CA, USA.
| |
Collapse
|
23
|
Muthusamy T, Cordes T, Handzlik MK, You L, Lim EW, Gengatharan J, Pinto AFM, Badur MG, Kolar MJ, Wallace M, Saghatelian A, Metallo CM. Serine restriction alters sphingolipid diversity to constrain tumour growth. Nature 2020; 586:790-795. [PMID: 32788725 PMCID: PMC7606299 DOI: 10.1038/s41586-020-2609-x] [Citation(s) in RCA: 200] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 05/11/2020] [Indexed: 11/22/2022]
Abstract
Serine, glycine and other nonessential amino acids are critical for tumour progression, and strategies to limit their availability are emerging as potential therapies for cancer1-3. However, the molecular mechanisms driving this response remain unclear and the effects on lipid metabolism are relatively unexplored. Serine palmitoyltransferase (SPT) catalyses the de novo biosynthesis of sphingolipids but also produces noncanonical 1-deoxysphingolipids when using alanine as a substrate4,5. Deoxysphingolipids accumulate in the context of mutations in SPTLC1 or SPTLC26,7-or in conditions of low serine availability8,9-to drive neuropathy, and deoxysphinganine has previously been investigated as an anti-cancer agent10. Here we exploit amino acid metabolism and the promiscuity of SPT to modulate the endogenous synthesis of toxic deoxysphingolipids and slow tumour progression. Anchorage-independent growth reprogrammes a metabolic network involving serine, alanine and pyruvate that drives the endogenous synthesis and accumulation of deoxysphingolipids. Targeting the mitochondrial pyruvate carrier promotes alanine oxidation to mitigate deoxysphingolipid synthesis and improve spheroid growth, similar to phenotypes observed with the direct inhibition of SPT or ceramide synthesis. Restriction of dietary serine and glycine potently induces the accumulation of deoxysphingolipids while decreasing tumour growth in xenograft models in mice. Pharmacological inhibition of SPT rescues xenograft growth in mice fed diets restricted in serine and glycine, and the reduction of circulating serine by inhibition of phosphoglycerate dehydrogenase (PHGDH) leads to the accumulation of deoxysphingolipids and mitigates tumour growth. The promiscuity of SPT therefore links serine and mitochondrial alanine metabolism to membrane lipid diversity, which further sensitizes tumours to metabolic stress.
Collapse
Affiliation(s)
| | - Thekla Cordes
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Michal K Handzlik
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Le You
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Esther W Lim
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Jivani Gengatharan
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Antonio F M Pinto
- Mass Spectrometry Core, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Mehmet G Badur
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Matthew J Kolar
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Martina Wallace
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Alan Saghatelian
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Christian M Metallo
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
24
|
Kirschbaum C, Saied EM, Greis K, Mucha E, Gewinner S, Schöllkopf W, Meijer G, Helden G, Poad BLJ, Blanksby SJ, Arenz C, Pagel K. Unterscheidung von isomeren Sphingolipiden mittels kryogener Infrarotspektroskopie. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202002459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Carla Kirschbaum
- Institut für Chemie und Biochemie Freie Universität Berlin Arnimallee 22 14195 Berlin Deutschland
- Abteilung Molekülphysik Fritz-Haber-Institut der Max-Planck-Gesellschaft Faradayweg 4–6 14195 Berlin Deutschland
| | - Essa M. Saied
- Institut für Chemie Humboldt-Universität zu Berlin Brook-Taylor-Straße 2 12489 Berlin Deutschland
- Chemistry Department Faculty of Science Suez Canal University Ismailia Ägypten
| | - Kim Greis
- Institut für Chemie und Biochemie Freie Universität Berlin Arnimallee 22 14195 Berlin Deutschland
- Abteilung Molekülphysik Fritz-Haber-Institut der Max-Planck-Gesellschaft Faradayweg 4–6 14195 Berlin Deutschland
| | - Eike Mucha
- Abteilung Molekülphysik Fritz-Haber-Institut der Max-Planck-Gesellschaft Faradayweg 4–6 14195 Berlin Deutschland
| | - Sandy Gewinner
- Abteilung Molekülphysik Fritz-Haber-Institut der Max-Planck-Gesellschaft Faradayweg 4–6 14195 Berlin Deutschland
| | - Wieland Schöllkopf
- Abteilung Molekülphysik Fritz-Haber-Institut der Max-Planck-Gesellschaft Faradayweg 4–6 14195 Berlin Deutschland
| | - Gerard Meijer
- Abteilung Molekülphysik Fritz-Haber-Institut der Max-Planck-Gesellschaft Faradayweg 4–6 14195 Berlin Deutschland
| | - Gert Helden
- Abteilung Molekülphysik Fritz-Haber-Institut der Max-Planck-Gesellschaft Faradayweg 4–6 14195 Berlin Deutschland
| | - Berwyck L. J. Poad
- Central Analytical Research Facility Institute for Future Environments Queensland University of Technology Brisbane QLD 4000 Australien
| | - Stephen J. Blanksby
- Central Analytical Research Facility Institute for Future Environments Queensland University of Technology Brisbane QLD 4000 Australien
| | - Christoph Arenz
- Institut für Chemie Humboldt-Universität zu Berlin Brook-Taylor-Straße 2 12489 Berlin Deutschland
| | - Kevin Pagel
- Institut für Chemie und Biochemie Freie Universität Berlin Arnimallee 22 14195 Berlin Deutschland
- Abteilung Molekülphysik Fritz-Haber-Institut der Max-Planck-Gesellschaft Faradayweg 4–6 14195 Berlin Deutschland
| |
Collapse
|
25
|
Santos TCB, Vaz A, Ventura AE, M Saied E, Arenz C, Fedorov A, Prieto M, Silva LC. Canonical and 1-Deoxy(methyl) Sphingoid Bases: Tackling the Effect of the Lipid Structure on Membrane Biophysical Properties. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:6007-6016. [PMID: 32369370 DOI: 10.1021/acs.langmuir.0c01000] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Compared to the canonical sphingoid backbone of sphingolipids (SLs), atypical long-chain bases (LCBs) lack C1-OH (1-deoxy-LCBs) or C1-CH2OH (1-deoxymethyl-LCBs). In addition, when unsaturated, they present a cis-double bond instead of the canonical Δ4-5 trans-double bond. These atypical LCBs are directly correlated with the development and progression of hereditary sensory and autonomic neuropathy type 1 and diabetes type II through yet unknown mechanisms. Changes in membrane properties have been linked to the biological actions of SLs. However, little is known about the influence of the LCB structure, particularly 1-deoxy(methyl)-LCB, on lipid-lipid interactions and their effect on membrane properties. To address this question, we used complementary fluorescence-based methodologies to study membrane model systems containing POPC and the different LCBs of interest. Our results show that 1-deoxymethyl-LCBs have the highest ability to reduce the fluidity of the membrane, while the intermolecular interactions of 1-deoxy-LCBs were found to be weaker, leading to the formation of less-ordered domains compared to their canonical counterparts-sphinganine and sphingosine. Furthermore, while the presence of a trans-double bond at the Δ4-5 position of the LCB increased the fluidity of the membrane compared to a saturated LCB, a cis-double bond completely disrupted the ability of the LCB to segregate into ordered domains. In conclusion, even small changes on the structure of the LCB, as seen in 1-deoxy(methyl)-LCBs, strongly affects lipid-lipid interactions and membrane fluidity. These results provide evidence that altered balance between species with different LCBs affect membrane properties and may contribute to the pathobiological role of these lipids.
Collapse
Affiliation(s)
- Tania C B Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon 1649-003, Portugal
- iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal
| | - Alexandra Vaz
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon 1649-003, Portugal
| | - Ana E Ventura
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon 1649-003, Portugal
- iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal
| | - Essa M Saied
- Institute for Chemistry, Humboldt Universität zu Berlin, Berlin 12489, Germany
- Faculty of Science, Chemistry Department, Suez Canal University, Ismailia 41522, Egypt
| | - Christoph Arenz
- Institute for Chemistry, Humboldt Universität zu Berlin, Berlin 12489, Germany
| | - Aleksander Fedorov
- iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal
| | - Manuel Prieto
- iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal
| | - Liana C Silva
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon 1649-003, Portugal
| |
Collapse
|
26
|
Kirschbaum C, Saied EM, Greis K, Mucha E, Gewinner S, Schöllkopf W, Meijer G, von Helden G, Poad BLJ, Blanksby SJ, Arenz C, Pagel K. Resolving Sphingolipid Isomers Using Cryogenic Infrared Spectroscopy. Angew Chem Int Ed Engl 2020; 59:13638-13642. [PMID: 32291895 PMCID: PMC7496694 DOI: 10.1002/anie.202002459] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/23/2020] [Indexed: 12/21/2022]
Abstract
1‐Deoxysphingolipids are a recently described class of sphingolipids that have been shown to be associated with several disease states including diabetic and hereditary neuropathy. The identification and characterization of 1‐deoxysphingolipids and their metabolites is therefore highly important. However, exact structure determination requires a combination of sophisticated analytical techniques due to the presence of various isomers, such as ketone/alkenol isomers, carbon–carbon double‐bond (C=C) isomers and hydroxylation regioisomers. Here we demonstrate that cryogenic gas‐phase infrared (IR) spectroscopy of ionized 1‐deoxysphingolipids enables the identification and differentiation of isomers by their unique spectroscopic fingerprints. In particular, C=C bond positions and stereochemical configurations can be distinguished by specific interactions between the charged amine and the double bond. The results demonstrate the power of gas‐phase IR spectroscopy to overcome the challenge of isomer resolution in conventional mass spectrometry and pave the way for deeper analysis of the lipidome.
Collapse
Affiliation(s)
- Carla Kirschbaum
- Institut für Chemie und Biochemie, Freie Universität Berlin, Arnimallee 22, 14195, Berlin, Germany.,Abteilung Molekülphysik, Fritz-Haber-Institut der Max-Planck-Gesellschaft, Faradayweg 4-6, 14195, Berlin, Germany
| | - Essa M Saied
- Institut für Chemie, Humboldt-Universität zu Berlin, Brook-Taylor-Straße 2, 12489, Berlin, Germany.,Chemistry Department, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Kim Greis
- Institut für Chemie und Biochemie, Freie Universität Berlin, Arnimallee 22, 14195, Berlin, Germany.,Abteilung Molekülphysik, Fritz-Haber-Institut der Max-Planck-Gesellschaft, Faradayweg 4-6, 14195, Berlin, Germany
| | - Eike Mucha
- Abteilung Molekülphysik, Fritz-Haber-Institut der Max-Planck-Gesellschaft, Faradayweg 4-6, 14195, Berlin, Germany
| | - Sandy Gewinner
- Abteilung Molekülphysik, Fritz-Haber-Institut der Max-Planck-Gesellschaft, Faradayweg 4-6, 14195, Berlin, Germany
| | - Wieland Schöllkopf
- Abteilung Molekülphysik, Fritz-Haber-Institut der Max-Planck-Gesellschaft, Faradayweg 4-6, 14195, Berlin, Germany
| | - Gerard Meijer
- Abteilung Molekülphysik, Fritz-Haber-Institut der Max-Planck-Gesellschaft, Faradayweg 4-6, 14195, Berlin, Germany
| | - Gert von Helden
- Abteilung Molekülphysik, Fritz-Haber-Institut der Max-Planck-Gesellschaft, Faradayweg 4-6, 14195, Berlin, Germany
| | - Berwyck L J Poad
- Central Analytical Research Facility, Institute for Future Environments, Queensland University of Technology, Brisbane, QLD, 4000, Australia
| | - Stephen J Blanksby
- Central Analytical Research Facility, Institute for Future Environments, Queensland University of Technology, Brisbane, QLD, 4000, Australia
| | - Christoph Arenz
- Institut für Chemie, Humboldt-Universität zu Berlin, Brook-Taylor-Straße 2, 12489, Berlin, Germany
| | - Kevin Pagel
- Institut für Chemie und Biochemie, Freie Universität Berlin, Arnimallee 22, 14195, Berlin, Germany.,Abteilung Molekülphysik, Fritz-Haber-Institut der Max-Planck-Gesellschaft, Faradayweg 4-6, 14195, Berlin, Germany
| |
Collapse
|
27
|
Karsai G, Lone M, Kutalik Z, Brenna JT, Li H, Pan D, von Eckardstein A, Hornemann T. FADS3 is a Δ14Z sphingoid base desaturase that contributes to gender differences in the human plasma sphingolipidome. J Biol Chem 2020; 295:1889-1897. [PMID: 31862735 PMCID: PMC7029104 DOI: 10.1074/jbc.ac119.011883] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/16/2019] [Indexed: 12/18/2022] Open
Abstract
Sphingolipids (SLs) are structurally diverse lipids that are defined by the presence of a long-chain base (LCB) backbone. Typically, LCBs contain a single Δ4E double bond (DB) (mostly d18:1), whereas the dienic LCB sphingadienine (d18:2) contains a second DB at the Δ14Z position. The enzyme introducing the Δ14Z DB is unknown. We analyzed the LCB plasma profile in a gender-, age-, and BMI-matched subgroup of the CoLaus cohort (n = 658). Sphingadienine levels showed a significant association with gender, being on average ∼30% higher in females. A genome-wide association study (GWAS) revealed variants in the fatty acid desaturase 3 (FADS3) gene to be significantly associated with the plasma d18:2/d18:1 ratio (p = -log 7.9). Metabolic labeling assays, FADS3 overexpression and knockdown approaches, and plasma LCB profiling in FADS3-deficient mice confirmed that FADS3 is a bona fide LCB desaturase and required for the introduction of the Δ14Z double bond. Moreover, we showed that FADS3 is required for the conversion of the atypical cytotoxic 1-deoxysphinganine (1-deoxySA, m18:0) to 1-deoxysphingosine (1-deoxySO, m18:1). HEK293 cells overexpressing FADS3 were more resistant to m18:0 toxicity than WT cells. In summary, using a combination of metabolic profiling and GWAS, we identified FADS3 to be essential for forming Δ14Z DB containing LCBs, such as d18:2 and m18:1. Our results unravel FADS3 as a Δ14Z LCB desaturase, thereby disclosing the last missing enzyme of the SL de novo synthesis pathway.
Collapse
Affiliation(s)
- Gergely Karsai
- Institute for Clinical Chemistry, University Hospital and University Zurich, 8091 Zürich, Switzerland
| | - Museer Lone
- Institute for Clinical Chemistry, University Hospital and University Zurich, 8091 Zürich, Switzerland
| | - Zoltán Kutalik
- University Center for Primary Care and Public Health, University of Lausanne, 1010 Lausanne, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - J Thomas Brenna
- Dell Pediatric Research Institute, Departments of Chemistry, Pediatrics, and Nutrition, University of Texas, Austin, Texas 78723
| | - Hongde Li
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Duojia Pan
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Arnold von Eckardstein
- Institute for Clinical Chemistry, University Hospital and University Zurich, 8091 Zürich, Switzerland
| | - Thorsten Hornemann
- Institute for Clinical Chemistry, University Hospital and University Zurich, 8091 Zürich, Switzerland.
| |
Collapse
|
28
|
Druggable Sphingolipid Pathways: Experimental Models and Clinical Opportunities. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1274:101-135. [PMID: 32894509 DOI: 10.1007/978-3-030-50621-6_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Intensive research in the field of sphingolipids has revealed diverse roles in cell biological responses and human health and disease. This immense molecular family is primarily represented by the bioactive molecules ceramide, sphingosine, and sphingosine 1-phosphate (S1P). The flux of sphingolipid metabolism at both the subcellular and extracellular levels provides multiple opportunities for pharmacological intervention. The caveat is that perturbation of any single node of this highly regulated flux may have effects that propagate throughout the metabolic network in a dramatic and sometimes unexpected manner. Beginning with S1P, the receptors for which have thus far been the most clinically tractable pharmacological targets, this review will describe recent advances in therapeutic modulators targeting sphingolipids, their chaperones, transporters, and metabolic enzymes.
Collapse
|
29
|
Hancock SE, Poad BLJ, Willcox MDP, Blanksby SJ, Mitchell TW. Analytical separations for lipids in complex, nonpolar lipidomes using differential mobility spectrometry. J Lipid Res 2019; 60:1968-1978. [PMID: 31511397 PMCID: PMC6824485 DOI: 10.1194/jlr.d094854] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 09/03/2019] [Indexed: 11/20/2022] Open
Abstract
Secretions from meibomian glands located within the eyelid (commonly known as meibum) are rich in nonpolar lipid classes incorporating very-long (22-30 carbons) and ultra-long (>30 carbons) acyl chains. The complex nature of the meibum lipidome and its preponderance of neutral, nonpolar lipid classes presents an analytical challenge, with typically poor chromatographic resolution, even between different lipid classes. To address this challenge, we have deployed differential mobility spectrometry (DMS)-MS to interrogate the human meibum lipidome and demonstrate near-baseline resolution of the two major nonpolar classes contained therein, namely wax esters and cholesteryl esters. Within these two lipid classes, we describe ion mobility behavior that is associated with the length of their acyl chains and location of unsaturation. This capability was exploited to profile the molecular speciation within each class and thus extend meibum lipidome coverage. Intriguingly, structure-mobility relationships in these nonpolar lipids show similar trends and inflections to those previously reported for other physicochemical properties of lipids (e.g., melting point and phase-transition temperatures). Taken together, these data demonstrate that differential ion mobility provides a powerful orthoganol separation technology for the analysis of neutral lipids in complex matrices, such as meibum, and may further provide a means to predict physicochemical properties of lipids that could assist in inferring their biological function(s).
Collapse
Affiliation(s)
- Sarah E Hancock
- School of Medicine and Molecular Horizons, University of Wollongong, Wollongong, Australia
- Illawarra Health and Medical Research Institute, Wollongong, Australia
| | - Berwyck L J Poad
- Central Analytical Research Facility, Institute for Future Environments, Queensland University of Technology, Brisbane, Australia
| | - Mark D P Willcox
- School of Optometry and Vision Science, University of New South Wales, Sydney, Australia
| | - Stephen J Blanksby
- Central Analytical Research Facility, Institute for Future Environments, Queensland University of Technology, Brisbane, Australia
| | - Todd W Mitchell
- School of Medicine and Molecular Horizons, University of Wollongong, Wollongong, Australia
- Illawarra Health and Medical Research Institute, Wollongong, Australia
| |
Collapse
|
30
|
Gerl MJ, Klose C, Surma MA, Fernandez C, Melander O, Männistö S, Borodulin K, Havulinna AS, Salomaa V, Ikonen E, Cannistraci CV, Simons K. Machine learning of human plasma lipidomes for obesity estimation in a large population cohort. PLoS Biol 2019; 17:e3000443. [PMID: 31626640 PMCID: PMC6799887 DOI: 10.1371/journal.pbio.3000443] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/04/2019] [Indexed: 01/05/2023] Open
Abstract
Obesity is associated with changes in the plasma lipids. Although simple lipid quantification is routinely used, plasma lipids are rarely investigated at the level of individual molecules. We aimed at predicting different measures of obesity based on the plasma lipidome in a large population cohort using advanced machine learning modeling. A total of 1,061 participants of the FINRISK 2012 population cohort were randomly chosen, and the levels of 183 plasma lipid species were measured in a novel mass spectrometric shotgun approach. Multiple machine intelligence models were trained to predict obesity estimates, i.e., body mass index (BMI), waist circumference (WC), waist-hip ratio (WHR), and body fat percentage (BFP), and validated in 250 randomly chosen participants of the Malmö Diet and Cancer Cardiovascular Cohort (MDC-CC). Comparison of the different models revealed that the lipidome predicted BFP the best (R2 = 0.73), based on a Lasso model. In this model, the strongest positive and the strongest negative predictor were sphingomyelin molecules, which differ by only 1 double bond, implying the involvement of an unknown desaturase in obesity-related aberrations of lipid metabolism. Moreover, we used this regression to probe the clinically relevant information contained in the plasma lipidome and found that the plasma lipidome also contains information about body fat distribution, because WHR (R2 = 0.65) was predicted more accurately than BMI (R2 = 0.47). These modeling results required full resolution of the lipidome to lipid species level, and the predicting set of biomarkers had to be sufficiently large. The power of the lipidomics association was demonstrated by the finding that the addition of routine clinical laboratory variables, e.g., high-density lipoprotein (HDL)- or low-density lipoprotein (LDL)- cholesterol did not improve the model further. Correlation analyses of the individual lipid species, controlled for age and separated by sex, underscores the multiparametric and lipid species-specific nature of the correlation with the BFP. Lipidomic measurements in combination with machine intelligence modeling contain rich information about body fat amount and distribution beyond traditional clinical assays.
Collapse
Affiliation(s)
| | | | - Michal A. Surma
- Lipotype GmbH, Dresden, Germany
- Łukasiewicz Research Network—PORT Polish Center for Technology Development, Wroclaw, Poland
| | | | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Emergency and Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Satu Männistö
- Public Health Promotion Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Katja Borodulin
- National Institute for Health and Welfare, Helsinki, Finland
| | - Aki S. Havulinna
- National Institute for Health and Welfare, Helsinki, Finland
- Institute for Molecular Medicine Finland (FIMM-HiLife), Helsinki, Finland
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - Elina Ikonen
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland
| | - Carlo V. Cannistraci
- Biomedical Cybernetics Group, Biotechnology Center (BIOTEC), Center for Molecular and Cellular Bioengineering (CMCB), Department of Physics, Technische Universität Dresden, Dresden, Germany
- Center for Systems Biology Dresden, Dresden, Germany
- Complex Network Intelligence Lab, Tsinghua Laboratory of Brain and Intelligence, Tsinghua University, Beijing, China
| | - Kai Simons
- Lipotype GmbH, Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
31
|
Marshall DL, Criscuolo A, Young RSE, Poad BLJ, Zeller M, Reid GE, Mitchell TW, Blanksby SJ. Mapping Unsaturation in Human Plasma Lipids by Data-Independent Ozone-Induced Dissociation. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2019; 30:1621-1630. [PMID: 31222675 DOI: 10.1007/s13361-019-02261-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/27/2019] [Accepted: 05/27/2019] [Indexed: 06/09/2023]
Abstract
Over 1500 different lipids have been reported in human plasma at the sum composition level. Yet the number of unique lipids present is surely higher, once isomeric contributions from double bond location(s) and fatty acyl regiochemistry are considered. In order to resolve this ambiguity, herein, we describe the incorporation of ozone-induced dissociation (OzID) into data-independent shotgun lipidomics workflows on a high-resolution hybrid quadrupole-Orbitrap platform. In this configuration, [M + Na]+ ions generated by electrospray ionization of a plasma lipid extract were transmitted through the quadrupole in 1 Da segments. Reaction of mass-selected lipid ions with ozone in the octopole collision cell yielded diagnostic ions for each double bond position. The increased ozone concentration in this region significantly improved ozonolysis efficiency compared with prior implementations on linear ion-trap devices. This advancement translates into increased lipidome coverage and improvements in duty cycle for data-independent MS/MS analysis using shotgun workflows. Grouping all precursor ions with a common OzID neutral loss enables straightforward classification of the lipidome by unsaturation position (with respect to the methyl terminus). Two-dimensional maps obtained from this analysis provide a powerful visualization of structurally related lipids and lipid isomer families within plasma. Global profiling of lipid unsaturation in plasma extracts reveals that most unsaturated lipids are present as isomeric mixtures. These new insights provide a unique picture of underlying metabolism that could in the future provide novel indicators of health and disease.
Collapse
Affiliation(s)
- David L Marshall
- Central Analytical Research Facility, Institute for Future Environments, Queensland University of Technology, Brisbane, QLD, 4000, Australia.
| | - Angela Criscuolo
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Universität Leipzig, Leipzig, Germany
- Center for Biotechnology and Biomedicine, Universität Leipzig, Leipzig, Germany
- Thermo Fisher Scientific (Bremen) GmbH, Hanna-Kunath Str. 11, 28199, Bremen, Germany
| | - Reuben S E Young
- School of Chemistry, Physics and Mechanical Engineering, Queensland University of Technology, Brisbane, Australia
| | - Berwyck L J Poad
- Central Analytical Research Facility, Institute for Future Environments, Queensland University of Technology, Brisbane, QLD, 4000, Australia
| | - Martin Zeller
- Thermo Fisher Scientific (Bremen) GmbH, Hanna-Kunath Str. 11, 28199, Bremen, Germany
| | - Gavin E Reid
- School of Chemistry, Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - Todd W Mitchell
- School of Medicine and Molecular Horizons, University of Wollongong, Wollongong, Australia
- Illawarra Health and Medical Research Institute, Wollongong, Australia
| | - Stephen J Blanksby
- Central Analytical Research Facility, Institute for Future Environments, Queensland University of Technology, Brisbane, QLD, 4000, Australia.
| |
Collapse
|
32
|
Carreira AC, Santos TC, Lone MA, Zupančič E, Lloyd-Evans E, de Almeida RFM, Hornemann T, Silva LC. Mammalian sphingoid bases: Biophysical, physiological and pathological properties. Prog Lipid Res 2019:100995. [PMID: 31445071 DOI: 10.1016/j.plipres.2019.100995] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 05/17/2019] [Accepted: 05/20/2019] [Indexed: 12/19/2022]
Abstract
Sphingoid bases encompass a group of long chain amino alcohols which form the essential structure of sphingolipids. Over the last years, these amphiphilic molecules were moving more and more into the focus of biomedical research due to their role as bioactive molecules. In fact, free sphingoid bases interact with specific receptors and target molecules and have been associated with numerous biological and physiological processes. In addition, they can modulate the biophysical properties of biological membranes. Several human diseases are related to pathological changes in the structure and metabolism of sphingoid bases. Yet, the mechanisms underlying their biological and pathophysiological actions remain elusive. Within this review, we aimed to summarize the current knowledge on the biochemical and biophysical properties of the most common sphingoid bases and to discuss their importance in health and disease.
Collapse
Affiliation(s)
- A C Carreira
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Centro de Química e Bioquímica (CQB) e Centro de Química Estrutural (CQE), Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, 1749-016 Lisboa, Portugal; Sir Martin Evans Building, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - T C Santos
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Centro de Química-Física Molecular - Institute of Nanoscience and Nanotechnology (CQFM-IN) and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Institute for Clinical Chemistry, University Hospital Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - M A Lone
- Institute for Clinical Chemistry, University Hospital Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - E Zupančič
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - E Lloyd-Evans
- Sir Martin Evans Building, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - R F M de Almeida
- Centro de Química e Bioquímica (CQB) e Centro de Química Estrutural (CQE), Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, 1749-016 Lisboa, Portugal
| | - T Hornemann
- Institute for Clinical Chemistry, University Hospital Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - L C Silva
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Centro de Química-Física Molecular - Institute of Nanoscience and Nanotechnology (CQFM-IN) and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
33
|
Carreira AC, Santos TC, Lone MA, Zupančič E, Lloyd-Evans E, de Almeida RFM, Hornemann T, Silva LC. Mammalian sphingoid bases: Biophysical, physiological and pathological properties. Prog Lipid Res 2019; 75:100988. [PMID: 31132366 DOI: 10.1016/j.plipres.2019.100988] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 05/17/2019] [Accepted: 05/21/2019] [Indexed: 12/11/2022]
Abstract
Sphingoid bases encompass a group of long chain amino alcohols which form the essential structure of sphingolipids. Over the last years, these amphiphilic molecules were moving more and more into the focus of biomedical research due to their role as bioactive molecules. In fact, free sphingoid bases interact with specific receptors and target molecules, and have been associated with numerous biological and physiological processes. In addition, they can modulate the biophysical properties of biological membranes. Several human diseases are related to pathological changes in the structure and metabolism of sphingoid bases. Yet, the mechanisms underlying their biological and pathophysiological actions remain elusive. Within this review, we aimed to summarize the current knowledge on the biochemical and biophysical properties of the most common sphingoid bases and to discuss their importance in health and disease.
Collapse
Affiliation(s)
- A C Carreira
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal; Centro de Química e Bioquímica (CQB) e Centro de Química Estrutural (CQE), Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, Lisboa 1749-016, Portugal; Sir Martin Evans Building, School of Biosciences, Cardiff University, Cardiff, UK
| | - T C Santos
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal; Centro de Química-Física Molecular - Institute of Nanoscience and Nanotechnology (CQFM-IN), IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Institute for Clinical Chemistry, University Hospital Zurich, Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - M A Lone
- Institute for Clinical Chemistry, University Hospital Zurich, Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - E Zupančič
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal
| | - E Lloyd-Evans
- Sir Martin Evans Building, School of Biosciences, Cardiff University, Cardiff, UK
| | - R F M de Almeida
- Centro de Química e Bioquímica (CQB) e Centro de Química Estrutural (CQE), Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, Lisboa 1749-016, Portugal
| | - T Hornemann
- Institute for Clinical Chemistry, University Hospital Zurich, Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - L C Silva
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal; Centro de Química-Física Molecular - Institute of Nanoscience and Nanotechnology (CQFM-IN), IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
34
|
Porta Siegel T, Ekroos K, Ellis SR. Reshaping Lipid Biochemistry by Pushing Barriers in Structural Lipidomics. Angew Chem Int Ed Engl 2019; 58:6492-6501. [PMID: 30601602 PMCID: PMC6563696 DOI: 10.1002/anie.201812698] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Indexed: 12/14/2022]
Abstract
Lipidomics is a rapidly growing field with numerous examples showing the importance of lipid molecules throughout biology. It has also shed light onto the vast and complex functions performed by many lipids that possess an immense diversity in molecular structures. Mass spectrometry (MS) is the tool of choice for analyzing lipids and has been the key catalyst driving the field forward. However, MS does not yet permit true molecular lipidomics wherein the identification and quantification of lipids having defined molecular structures can be routinely achieved. Here we describe recent advances in MS-based lipidomics that allow access to higher levels of molecular information in lipidomics experiments. These advances will form a key piece of the puzzle as the field moves towards systems characterization of lipids at the molecular level.
Collapse
Affiliation(s)
- Tiffany Porta Siegel
- Maastricht MultiModal Molecular Imaging (M4I) instituteDivision of Imaging Mass SpectrometryMaastricht UniversityUniversiteitssingel 506229 ERMaastrichtThe Netherlands
| | | | - Shane R. Ellis
- Maastricht MultiModal Molecular Imaging (M4I) instituteDivision of Imaging Mass SpectrometryMaastricht UniversityUniversiteitssingel 506229 ERMaastrichtThe Netherlands
| |
Collapse
|
35
|
New Frontiers in Lipidomics Analyses using Structurally Selective Ion Mobility-Mass Spectrometry. Trends Analyt Chem 2019; 116:316-323. [PMID: 31983792 DOI: 10.1016/j.trac.2019.03.031] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The growth of lipidomics and the high isomeric complexity of the lipidome has revealed a need for analytical techniques capable of structurally characterizing lipids with a high degree of specificity. Lipids are morphologically diverse molecules that can exist as any one of a large number of isomeric species, and as such are often indistinguishable by mass spectrometry without a complementary separation method. Recent developments in the field of lipidomics aim to address these challenges by utilizing a combination of multiple analytical techniques which are selective to lipid primary structure. This review summarizes two emerging strategies for lipidomic analysis, namely, ion mobility-mass spectrometry and ion fragmentation via ozonolysis.
Collapse
|
36
|
Schwartz NU, Mileva I, Gurevich M, Snider J, Hannun YA, Obeid LM. Quantifying 1-deoxydihydroceramides and 1-deoxyceramides in mouse nervous system tissue. Prostaglandins Other Lipid Mediat 2019; 141:40-48. [PMID: 30790665 PMCID: PMC6467697 DOI: 10.1016/j.prostaglandins.2019.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 02/08/2019] [Accepted: 02/11/2019] [Indexed: 02/06/2023]
Abstract
Accumulation of deoxysphingolipids (deoxySLs) has been implicated in many neural diseases, although mechanisms remain unclear. A major obstacle limiting understanding of deoxySLs has been the lack of a method easily defining measurement of deoxydihydroceramide (deoxydhCer) and deoxyceramide (deoxyCer) in neural tissues. Furthermore, it is poorly understood if deoxySLs accumulate in the nervous system with aging. To facilitate investigation of deoxydhCer and deoxyCer in nervous system tissue, we developed a method to evaluate levels of these lipids in mouse brain, spinal cord, and sciatic nerve. Many deoxydhCers and brain C24-deoxyCer were present at 1, 3, and 6 months of age. Furthermore, while ceramide levels decreased with age, deoxydhCers increased in sciatic nerve and spinal cord, suggesting they may accumulate in peripheral nerves. C22-deoxydhCer was the highest deoxydhCer species in all tissues, suggesting it may be important physiologically. The development of this method will facilitate straightforward profiling of deoxydhCers and deoxyCers and the study of their metabolism and function. These results also reveal that deoxydhCers accumulate in peripheral nerves with normal aging.
Collapse
Affiliation(s)
- Nicholas U Schwartz
- Health Science Center, L-4, 179, Stony Brook University Medical Center, Stony Brook, NY, 11794-8430, United States
| | - Izolda Mileva
- Health Science Center, L-4, 179, Stony Brook University Medical Center, Stony Brook, NY, 11794-8430, United States
| | - Mikhail Gurevich
- Health Science Center, L-4, 179, Stony Brook University Medical Center, Stony Brook, NY, 11794-8430, United States
| | - Justin Snider
- Health Science Center, L-4, 179, Stony Brook University Medical Center, Stony Brook, NY, 11794-8430, United States
| | - Yusuf A Hannun
- Health Science Center, L-4, 179, Stony Brook University Medical Center, Stony Brook, NY, 11794-8430, United States
| | - Lina M Obeid
- Health Science Center, L-4, 179, Stony Brook University Medical Center, Stony Brook, NY, 11794-8430, United States.
| |
Collapse
|
37
|
Porta Siegel T, Ekroos K, Ellis SR. Reshaping Lipid Biochemistry by Pushing Barriers in Structural Lipidomics. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201812698] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Tiffany Porta Siegel
- Maastricht MultiModal Molecular Imaging (M4I) instituteDivision of Imaging Mass SpectrometryMaastricht University Universiteitssingel 50 6229 ER Maastricht The Netherlands
| | | | - Shane R. Ellis
- Maastricht MultiModal Molecular Imaging (M4I) instituteDivision of Imaging Mass SpectrometryMaastricht University Universiteitssingel 50 6229 ER Maastricht The Netherlands
| |
Collapse
|
38
|
Karsai G, Kraft F, Haag N, Korenke GC, Hänisch B, Othman A, Suriyanarayanan S, Steiner R, Knopp C, Mull M, Bergmann M, Schröder JM, Weis J, Elbracht M, Begemann M, Hornemann T, Kurth I. DEGS1-associated aberrant sphingolipid metabolism impairs nervous system function in humans. J Clin Invest 2019; 129:1229-1239. [PMID: 30620338 DOI: 10.1172/jci124159] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 12/21/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Sphingolipids are important components of cellular membranes and functionally associated with fundamental processes such as cell differentiation, neuronal signaling, and myelin sheath formation. Defects in the synthesis or degradation of sphingolipids leads to various neurological pathologies; however, the entire spectrum of sphingolipid metabolism disorders remains elusive. METHODS A combined approach of genomics and lipidomics was applied to identify and characterize a human sphingolipid metabolism disorder. RESULTS By whole-exome sequencing in a patient with a multisystem neurological disorder of both the central and peripheral nervous systems, we identified a homozygous p.Ala280Val variant in DEGS1, which catalyzes the last step in the ceramide synthesis pathway. The blood sphingolipid profile in the patient showed a significant increase in dihydro sphingolipid species that was further recapitulated in patient-derived fibroblasts, in CRISPR/Cas9-derived DEGS1-knockout cells, and by pharmacological inhibition of DEGS1. The enzymatic activity in patient fibroblasts was reduced by 80% compared with wild-type cells, which was in line with a reduced expression of mutant DEGS1 protein. Moreover, an atypical and potentially neurotoxic sphingosine isomer was identified in patient plasma and in cells expressing mutant DEGS1. CONCLUSION We report DEGS1 dysfunction as the cause of a sphingolipid disorder with hypomyelination and degeneration of both the central and peripheral nervous systems. TRIAL REGISTRATION Not applicable. FUNDING Seventh Framework Program of the European Commission, Swiss National Foundation, Rare Disease Initiative Zurich.
Collapse
Affiliation(s)
- Gergely Karsai
- Center for Integrative Human Physiology, University of Zürich, Zürich, Switzerland.,Institute for Clinical Chemistry, University Hospital, Zürich, Switzerland
| | - Florian Kraft
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Natja Haag
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - G Christoph Korenke
- Clinic for Neuropediatrics and Congenital Metabolic Diseases, University Clinic for Paediatrics and Adolescent Medicine, Oldenburg, Germany
| | - Benjamin Hänisch
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Alaa Othman
- Center for Integrative Human Physiology, University of Zürich, Zürich, Switzerland.,Institute for Clinical Chemistry, University Hospital, Zürich, Switzerland
| | - Saranya Suriyanarayanan
- Center for Integrative Human Physiology, University of Zürich, Zürich, Switzerland.,Institute for Clinical Chemistry, University Hospital, Zürich, Switzerland
| | - Regula Steiner
- Center for Integrative Human Physiology, University of Zürich, Zürich, Switzerland.,Institute for Clinical Chemistry, University Hospital, Zürich, Switzerland
| | - Cordula Knopp
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Michael Mull
- Department of Diagnostic and Interventional Neuroradiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Markus Bergmann
- Institute for Neuropathology, Hospital Bremen-Mitte, Bremen, Germany
| | - J Michael Schröder
- Institute of Neuropathology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Joachim Weis
- Institute of Neuropathology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Miriam Elbracht
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Matthias Begemann
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Thorsten Hornemann
- Center for Integrative Human Physiology, University of Zürich, Zürich, Switzerland.,Institute for Clinical Chemistry, University Hospital, Zürich, Switzerland
| | - Ingo Kurth
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
39
|
Lone MA, Santos T, Alecu I, Silva LC, Hornemann T. 1-Deoxysphingolipids. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:512-521. [PMID: 30625374 DOI: 10.1016/j.bbalip.2018.12.013] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 12/14/2022]
Abstract
Sphingolipids (SLs) are fundamental components of eukaryotic cells. 1-Deoxysphingolipids differ structurally from canonical SLs as they lack the essential C1-OH group. Consequently, 1-deoxysphingolipids cannot be converted to complex sphingolipids and are not degraded over the canonical catabolic pathways. Pathologically elevated 1-deoxySLs are involved in several disease conditions. Within this review, we will provide an up-to-date overview on the metabolic, physiological and pathophysiological aspects of this enigmatic class of "headless" sphingolipids.
Collapse
Affiliation(s)
- M A Lone
- Institute for Clinical Chemistry, University Hospital Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - T Santos
- Institute for Clinical Chemistry, University Hospital Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland; iMed.ULisboa - Research Institute for Medicines, 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
| | - I Alecu
- Neural Regeneration Laboratory, India Taylor Lipidomic Research Platform, Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Ottawa Brain and Mind Research Institute, University of Ottawa, Canada
| | - L C Silva
- iMed.ULisboa - Research Institute for Medicines, 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
| | - T Hornemann
- Institute for Clinical Chemistry, University Hospital Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland.
| |
Collapse
|
40
|
Huynh K, Barlow CK, Jayawardana KS, Weir JM, Mellett NA, Cinel M, Magliano DJ, Shaw JE, Drew BG, Meikle PJ. High-Throughput Plasma Lipidomics: Detailed Mapping of the Associations with Cardiometabolic Risk Factors. Cell Chem Biol 2018; 26:71-84.e4. [PMID: 30415965 DOI: 10.1016/j.chembiol.2018.10.008] [Citation(s) in RCA: 229] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 04/06/2018] [Accepted: 10/05/2018] [Indexed: 12/26/2022]
Abstract
High-throughput targeted lipid profiling with liquid chromatography-mass spectrometry (LC-MS) has been used extensively to identify associations between plasma lipid species and disease states. Such methods, used to characterize larger clinical cohorts, often suffer from an inability to differentiate isomeric forms of glycerophospholipids that are typically reported as the sum fatty acid carbons and double bonds. Here we report a chromatography gradient coupled with a detailed characterization of the human plasma lipidome to provide improved resolution and identification of 636 lipid species, including previously unreported species, in a 15-min analysis. We have utilized this method on a subset of the Australian Diabetes, Obesity, and Lifestyle Study and have detailed associations of plasma lipid species with anthropometric and blood glucose measures. These results highlight the importance and power of high-throughput lipidomics coupled with a detailed characterization of the lipidome to better understand lipid biology in a population setting.
Collapse
Affiliation(s)
- Kevin Huynh
- Head Metabolomics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Christopher K Barlow
- Head Metabolomics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Kaushala S Jayawardana
- Head Metabolomics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Jacquelyn M Weir
- Head Metabolomics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Natalie A Mellett
- Head Metabolomics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Michelle Cinel
- Head Metabolomics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Dianna J Magliano
- Head Metabolomics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Jonathan E Shaw
- Head Metabolomics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Brian G Drew
- Head Metabolomics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Peter J Meikle
- Head Metabolomics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia.
| |
Collapse
|
41
|
Wilson ER, Kugathasan U, Abramov AY, Clark AJ, Bennett DLH, Reilly MM, Greensmith L, Kalmar B. Hereditary sensory neuropathy type 1-associated deoxysphingolipids cause neurotoxicity, acute calcium handling abnormalities and mitochondrial dysfunction in vitro. Neurobiol Dis 2018; 117:1-14. [PMID: 29778900 PMCID: PMC6060082 DOI: 10.1016/j.nbd.2018.05.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/23/2018] [Accepted: 05/16/2018] [Indexed: 01/03/2023] Open
Abstract
Hereditary sensory neuropathy type 1 (HSN-1) is a peripheral neuropathy most frequently caused by mutations in the SPTLC1 or SPTLC2 genes, which code for two subunits of the enzyme serine palmitoyltransferase (SPT). SPT catalyzes the first step of de novo sphingolipid synthesis. Mutations in SPT result in a change in enzyme substrate specificity, which causes the production of atypical deoxysphinganine and deoxymethylsphinganine, rather than the normal enzyme product, sphinganine. Levels of these abnormal compounds are elevated in blood of HSN-1 patients and this is thought to cause the peripheral motor and sensory nerve damage that is characteristic of the disease, by a largely unresolved mechanism. In this study, we show that exogenous application of these deoxysphingoid bases causes dose- and time-dependent neurotoxicity in primary mammalian neurons, as determined by analysis of cell survival and neurite length. Acutely, deoxysphingoid base neurotoxicity manifests in abnormal Ca2+ handling by the endoplasmic reticulum (ER) and mitochondria as well as dysregulation of cell membrane store-operated Ca2+ channels. The changes in intracellular Ca2+ handling are accompanied by an early loss of mitochondrial membrane potential in deoxysphingoid base-treated motor and sensory neurons. Thus, these results suggest that exogenous deoxysphingoid base application causes neuronal mitochondrial dysfunction and Ca2+ handling deficits, which may play a critical role in the pathogenesis of HSN-1.
Collapse
Affiliation(s)
- Emma R Wilson
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK; MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Umaiyal Kugathasan
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK; MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Andrey Y Abramov
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Alex J Clark
- Neural Injury Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - David L H Bennett
- Neural Injury Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Mary M Reilly
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK; Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Linda Greensmith
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK; MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Bernadett Kalmar
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK.
| |
Collapse
|
42
|
Hornemann T, Alecu I, Hagenbuch N, Zhakupova A, Cremonesi A, Gautschi M, Jung HH, Meienberg F, Bilz S, Christ E, Baumgartner MR, Hochuli M. Disturbed sphingolipid metabolism with elevated 1-deoxysphingolipids in glycogen storage disease type I - A link to metabolic control. Mol Genet Metab 2018; 125:73-78. [PMID: 30037504 DOI: 10.1016/j.ymgme.2018.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/10/2018] [Accepted: 07/10/2018] [Indexed: 11/26/2022]
Abstract
BACKGROUND 1-Deoxysphingolipids (1-deoxySLs) are atypical sphingolipids. They are formed during sphingolipid de novo synthesis by the enzyme serine palmitoyltransferase, due to the alternate use of alanine over its canonical substrate serine. Pathologically elevated 1-deoxySL are involved in several neurological and metabolic disorders. The objective of this study was to investigate the role of 1-deoxySL in glycogen storage disease type I (GSDI). METHODS In this prospective, longitudinal observational study (median follow-up 1.8y), the plasma 1-deoxySL profile was analyzed in 15 adult GSDI patients (12 GSDIa, 3 GSDIb), and 31 healthy controls, along with standard parameters for monitoring GSDI. RESULTS 1-Deoxysphinganine (1-deoxySA) concentrations were elevated in GSDI compared to controls (191 ± 129 vs 35 ± 14 nmol/l, p < 0.0001). Concordant with the mechanism of 1-deoxySL synthesis, plasma alanine was higher (625 ± 182 vs 398 ± 90 μmol/l, p < 0.0001), while serine was lower in GSDI than in controls (88 ± 22 vs 110 ± 18 μmol/l. p < 0.001). Accordingly, serine, alanine and triglycerides were determinants of 1-deoxySA in the longitudinal analysis of GSDIa. 1-deoxySA concentrations correlated with the occurrence of low blood glucose (area under the curve below 4 mmol/l) in continuous glucose monitoring. The 1-deoxySL profile in GSDIb was distinct from GSDIa, with a different ratio of saturated to unsaturated 1-deoxySL. CONCLUSION In addition to the known abnormalities of lipoproteins, GSDI patients also have a disturbed sphingolipid metabolism with elevated plasma 1-deoxySL concentrations. 1-DeoxySA relates to the occurrence of low blood glucose, and may constitute a potential new biomarker for assessing metabolic control. GSDIa and Ib have distinct 1-deoxySL profiles indicating that both GSD subtypes have diverse phenotypes regarding lipid metabolism.
Collapse
Affiliation(s)
- Thorsten Hornemann
- Institute of Clinical Chemistry, University Hospital Zurich, Zurich, Switzerland
| | - Irina Alecu
- Institute of Clinical Chemistry, University Hospital Zurich, Zurich, Switzerland.
| | - Niels Hagenbuch
- Institute of Biostatistics, University of Zurich, Zurich, Switzerland
| | - Assem Zhakupova
- Institute of Clinical Chemistry, University Hospital Zurich, Zurich, Switzerland
| | - Alessio Cremonesi
- Institute of Clinical Chemistry, University Children's Hospital, Zurich, Switzerland
| | - Matthias Gautschi
- Department of Pediatrics and Institute of Clinical Chemistry, University Hospital Bern, Inselspital, Bern, Switzerland
| | - Hans H Jung
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Fabian Meienberg
- Department of Endocrinology, Diabetes and Metabolism, University Hospital, Basel, Switzerland
| | - Stefan Bilz
- Division of Endocrinology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Emanuel Christ
- Department of Diabetes, Endocrinology, Nutritional medicine and Metabolism, University Hospital Bern, Inselspital, Bern, Switzerland
| | - Matthias R Baumgartner
- Division of Metabolism and Children's Research Center (CRC), University Children's Hospital, Zurich, Switzerland.; Radiz - Rare Disease Initiative Zurich, Clinical Research Priority Program for Rare Diseases, University of Zurich, Switzerland
| | - Michel Hochuli
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, Zurich, Switzerland; Radiz - Rare Disease Initiative Zurich, Clinical Research Priority Program for Rare Diseases, University of Zurich, Switzerland.
| |
Collapse
|
43
|
Saied EM, Le TLS, Hornemann T, Arenz C. Synthesis and characterization of some atypical sphingoid bases. Bioorg Med Chem 2018; 26:4047-4057. [PMID: 29960730 DOI: 10.1016/j.bmc.2018.06.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 06/19/2018] [Accepted: 06/23/2018] [Indexed: 11/18/2022]
Abstract
Sphingolipids are ubiquitous and abundant components of all eukaryotic and some prokaryotic organisms. Sphingolipids show a large structural variety not only between the different species, but also within an individual cell. This variety is not limited to alterations in the polar headgroups of e.g. glycosphingolipids, but also affects the lipophilic anchors comprised of different fatty acids on the one hand and different sphingoid bases on the other hand. The structural variations within different sphingoid bases e.g. in pathogens can be used to identify novel biomarkers and drug targets and the specific change in the profile of common and uncommon sphingolipids are associated with pathological conditions like diabetes or cancer. Therefore, the emerging field of sphingolipidomics is dedicated to collect data on the sphingolipidome of a cell and hence to assign changes therein to certain states of a cell or to pathological conditions. This powerful tool however is still limited by the availability of structural information about the individual lipid species as well as by the availability of appropriate internal standards for quantification. Herein we describe the synthesis of a variety of 1-deoxy-sphingoid bases. 1-DeoxySphingolipids have recently acquired significant attention due to its pathological role in the rare inherited neuropathy, HSAN1 but also as predictive biomarkers in diabetes type II. Some of the compounds synthesized and characterized herein, have been used and will be used to elucidate the correct structure of these disease-related lipids and their metabolites.
Collapse
Affiliation(s)
- Essa M Saied
- Institute for Chemistry, Humboldt Universität zu Berlin, Brook-Taylor-Str. 2, 12489 Berlin, Germany; Chemistry Department, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Thuy Linh-Stella Le
- Institute for Chemistry, Humboldt Universität zu Berlin, Brook-Taylor-Str. 2, 12489 Berlin, Germany
| | - T Hornemann
- University of Zurich; University Hospital of Zurich, Switzerland
| | - Christoph Arenz
- Institute for Chemistry, Humboldt Universität zu Berlin, Brook-Taylor-Str. 2, 12489 Berlin, Germany.
| |
Collapse
|
44
|
Poad BLJ, Maccarone AT, Yu H, Mitchell TW, Saied EM, Arenz C, Hornemann T, Bull JN, Bieske EJ, Blanksby SJ. Differential-Mobility Spectrometry of 1-Deoxysphingosine Isomers: New Insights into the Gas Phase Structures of Ionized Lipids. Anal Chem 2018; 90:5343-5351. [PMID: 29608293 DOI: 10.1021/acs.analchem.8b00469] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Separation and structural identification of lipids remain a major challenge for contemporary lipidomics. Regioisomeric lipids differing only in position(s) of unsaturation are often not differentiated by conventional liquid chromatography-mass spectrometry approaches leading to the incomplete, or sometimes incorrect, assignation of molecular structure. Here we describe an investigation of the gas phase separations by differential-mobility spectrometry (DMS) of a series of synthetic analogues of the recently described 1-deoxysphingosine. The dependence of the DMS behavior on the position of the carbon-carbon double bond within the ionized lipid is systematically explored and compared to trends from complementary investigations, including collision cross-sections measured by drift tube ion mobility, reaction efficiency with ozone, and molecular dynamics simulations. Consistent trends across these modes of interrogation point to the importance of direct, through-space interactions between the charge site and the carbon-carbon double bond. Differences in the geometry and energetics of this intramolecular interaction underpin DMS separations and influence reactivity trends between regioisomers. Importantly, the disruption and reformation of these intramolecular solvation interactions during DMS are proposed to be the causative factor in the observed separations of ionized lipids which are shown to have otherwise identical collision cross-sections. These findings provide key insights into the strengths and limitations of current ion-mobility technologies for lipid isomer separations and can thus guide a more systematic approach to improved analytical separations in lipidomics.
Collapse
Affiliation(s)
- Berwyck L J Poad
- Central Analytical Research Facility, Institute for Future Environments , Queensland University of Technology , Brisbane , Queensland 4001 , Australia
| | - Alan T Maccarone
- Mass Spectrometry User Resource and Research Facility , University of Wollongong , Wollongong , New South Wales 2522 , Australia.,School of Chemistry , University of Wollongong , Wollongong , New South Wales 2522 , Australia
| | - Haibo Yu
- School of Chemistry , University of Wollongong , Wollongong , New South Wales 2522 , Australia
| | - Todd W Mitchell
- School of Medicine , University of Wollongong , Wollongong , New South Wales 2522 , Australia
| | - Essa M Saied
- Institute for Chemistry , Humboldt Universität zu Berlin , 12489 Berlin , Germany.,Chemistry Department, Faculty of Science , Suez Canal University , 41522 Ismailia , Egypt
| | - Christoph Arenz
- Institute for Chemistry , Humboldt Universität zu Berlin , 12489 Berlin , Germany
| | - Thorsten Hornemann
- Institute of Clinical Chemistry , University Hospital of Zurich , CH-8091 Zurich , Switzerland
| | - James N Bull
- School of Chemistry , University of Melbourne , Parkville , Victoria 3010 , Australia
| | - Evan J Bieske
- School of Chemistry , University of Melbourne , Parkville , Victoria 3010 , Australia
| | - Stephen J Blanksby
- Central Analytical Research Facility, Institute for Future Environments , Queensland University of Technology , Brisbane , Queensland 4001 , Australia
| |
Collapse
|
45
|
Poad BLJ, Zheng X, Mitchell TW, Smith RD, Baker ES, Blanksby SJ. Online Ozonolysis Combined with Ion Mobility-Mass Spectrometry Provides a New Platform for Lipid Isomer Analyses. Anal Chem 2018; 90:1292-1300. [PMID: 29220163 PMCID: PMC5771865 DOI: 10.1021/acs.analchem.7b04091] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
One of the most significant challenges in contemporary lipidomics lies in the separation and identification of lipid isomers that differ only in site(s) of unsaturation or geometric configuration of the carbon-carbon double bonds. While analytical separation techniques including ion mobility spectrometry (IMS) and liquid chromatography (LC) can separate isomeric lipids under appropriate conditions, conventional tandem mass spectrometry cannot provide unequivocal identification. To address this challenge, we have implemented ozone-induced dissociation (OzID) in-line with LC, IMS, and high resolution mass spectrometry. Modification of an IMS-capable quadrupole time-of-flight mass spectrometer was undertaken to allow the introduction of ozone into the high-pressure trapping ion funnel region preceding the IMS cell. This enabled the novel LC-OzID-IMS-MS configuration where ozonolysis of ionized lipids occurred rapidly (10 ms) without prior mass-selection. LC-elution time alignment combined with accurate mass and arrival time extraction of ozonolysis products facilitated correlation of precursor and product ions without mass-selection (and associated reductions in duty cycle). Unsaturated lipids across 11 classes were examined using this workflow in both positive and negative ion modalities, and in all cases, the positions of carbon-carbon double bonds were unequivocally assigned based on predictable OzID transitions. Under these conditions, geometric isomers exhibited different IMS arrival time distributions and distinct OzID product ion ratios providing a means for discrimination of cis/trans double bonds in complex lipids. The combination of OzID with multidimensional separations shows significant promise for facile profiling of unsaturation patterns within complex lipidomes including human plasma.
Collapse
Affiliation(s)
- Berwyck L J Poad
- Central Analytical Research Facility, Insitutue for Future Environments, Queensland University of Technology , Brisbane, Queensland 4000, Australia
| | - Xueyun Zheng
- Biological Sciences Division, Pacific Northwest National Laboratory , Richland, Washington 99354, United States
| | - Todd W Mitchell
- School of Medicine, University of Wollongong , Wollongong, New South Wales 2522, Australia
| | - Richard D Smith
- Biological Sciences Division, Pacific Northwest National Laboratory , Richland, Washington 99354, United States
| | - Erin S Baker
- Biological Sciences Division, Pacific Northwest National Laboratory , Richland, Washington 99354, United States
| | - Stephen J Blanksby
- Central Analytical Research Facility, Insitutue for Future Environments, Queensland University of Technology , Brisbane, Queensland 4000, Australia
| |
Collapse
|
46
|
D'Atri V, Causon T, Hernandez-Alba O, Mutabazi A, Veuthey JL, Cianferani S, Guillarme D. Adding a new separation dimension to MS and LC-MS: What is the utility of ion mobility spectrometry? J Sep Sci 2017; 41:20-67. [PMID: 29024509 DOI: 10.1002/jssc.201700919] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 09/19/2017] [Accepted: 09/19/2017] [Indexed: 12/12/2022]
Abstract
Ion mobility spectrometry is an analytical technique known for more than 100 years, which entails separating ions in the gas phase based on their size, shape, and charge. While ion mobility spectrometry alone can be useful for some applications (mostly security analysis for detecting certain classes of narcotics and explosives), it becomes even more powerful in combination with mass spectrometry and high-performance liquid chromatography. Indeed, the limited resolving power of ion mobility spectrometry alone can be tackled when combining this analytical strategy with mass spectrometry or liquid chromatography with mass spectrometry. Over the last few years, the hyphenation of ion mobility spectrometry to mass spectrometry or liquid chromatography with mass spectrometry has attracted more and more interest, with significant progresses in both technical advances and pioneering applications. This review describes the theoretical background, available technologies, and future capabilities of these techniques. It also highlights a wide range of applications, from small molecules (natural products, metabolites, glycans, lipids) to large biomolecules (proteins, protein complexes, biopharmaceuticals, oligonucleotides).
Collapse
Affiliation(s)
- Valentina D'Atri
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Tim Causon
- Division of Analytical Chemistry, Department of Chemistry, University of Natural Resources and Life Sciences (BOKU Vienna), Vienna, Austria
| | - Oscar Hernandez-Alba
- BioOrganic Mass Spectrometry Laboratory (LSMBO), IPHC, Université de Strasbourg, CNRS, Strasbourg, France
| | - Aline Mutabazi
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Jean-Luc Veuthey
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Sarah Cianferani
- BioOrganic Mass Spectrometry Laboratory (LSMBO), IPHC, Université de Strasbourg, CNRS, Strasbourg, France
| | - Davy Guillarme
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| |
Collapse
|
47
|
Hancock SE, Poad BL, Batarseh A, Abbott SK, Mitchell TW. Advances and unresolved challenges in the structural characterization of isomeric lipids. Anal Biochem 2017; 524:45-55. [DOI: 10.1016/j.ab.2016.09.014] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 09/11/2016] [Accepted: 09/16/2016] [Indexed: 12/25/2022]
|
48
|
Alecu I, Tedeschi A, Behler N, Wunderling K, Lamberz C, Lauterbach MAR, Gaebler A, Ernst D, Van Veldhoven PP, Al-Amoudi A, Latz E, Othman A, Kuerschner L, Hornemann T, Bradke F, Thiele C, Penno A. Localization of 1-deoxysphingolipids to mitochondria induces mitochondrial dysfunction. J Lipid Res 2016; 58:42-59. [PMID: 27881717 DOI: 10.1194/jlr.m068676] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 10/27/2016] [Indexed: 12/20/2022] Open
Abstract
1-Deoxysphingolipids (deoxySLs) are atypical sphingolipids that are elevated in the plasma of patients with type 2 diabetes and hereditary sensory and autonomic neuropathy type 1 (HSAN1). Clinically, diabetic neuropathy and HSAN1 are very similar, suggesting the involvement of deoxySLs in the pathology of both diseases. However, very little is known about the biology of these lipids and the underlying pathomechanism. We synthesized an alkyne analog of 1-deoxysphinganine (doxSA), the metabolic precursor of all deoxySLs, to trace the metabolism and localization of deoxySLs. Our results indicate that the metabolism of these lipids is restricted to only some lipid species and that they are not converted to canonical sphingolipids or fatty acids. Furthermore, exogenously added alkyne-doxSA [(2S,3R)-2-aminooctadec-17-yn-3-ol] localized to mitochondria, causing mitochondrial fragmentation and dysfunction. The induced mitochondrial toxicity was also shown for natural doxSA, but not for sphinganine, and was rescued by inhibition of ceramide synthase activity. Our findings therefore indicate that mitochondrial enrichment of an N-acylated doxSA metabolite may contribute to the neurotoxicity seen in diabetic neuropathy and HSAN1. Hence, we provide a potential explanation for the characteristic vulnerability of peripheral nerves to elevated levels of deoxySLs.
Collapse
Affiliation(s)
- Irina Alecu
- Institute for Clinical Chemistry, University of Zurich, Zurich, Switzerland.,Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Andrea Tedeschi
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Natascha Behler
- LIMES Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Klaus Wunderling
- LIMES Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Christian Lamberz
- Cyro-Electron Microscopy and Tomography, German Center for Neurodegenerative Diseases, Bonn, Germany
| | | | - Anne Gaebler
- LIMES Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Daniela Ernst
- Institute for Clinical Chemistry, University of Zurich, Zurich, Switzerland
| | - Paul P Van Veldhoven
- Laboratory for Lipid Biochemistry and Protein Interactions, Campus Gasthuisberg, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Ashraf Al-Amoudi
- Cyro-Electron Microscopy and Tomography, German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Alaa Othman
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Lars Kuerschner
- LIMES Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Thorsten Hornemann
- Institute for Clinical Chemistry, University of Zurich, Zurich, Switzerland.,Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Frank Bradke
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Christoph Thiele
- LIMES Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Anke Penno
- LIMES Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
49
|
Alecu I, Othman A, Penno A, Saied EM, Arenz C, von Eckardstein A, Hornemann T. Cytotoxic 1-deoxysphingolipids are metabolized by a cytochrome P450-dependent pathway. J Lipid Res 2016; 58:60-71. [PMID: 27872144 DOI: 10.1194/jlr.m072421] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 11/11/2016] [Indexed: 12/14/2022] Open
Abstract
The 1-deoxysphingolipids (1-deoxySLs) are atypical sphingolipids (SLs) that are formed when serine palmitoyltransferase condenses palmitoyl-CoA with alanine instead of serine during SL synthesis. The 1-deoxySLs are toxic to neurons and pancreatic β-cells. Pathologically elevated 1-deoxySLs cause the inherited neuropathy, hereditary sensory autonomic neuropathy type 1 (HSAN1), and are also found in T2D. Diabetic sensory polyneuropathy (DSN) and HSAN1 are clinically very similar, suggesting that 1-deoxySLs may be implicated in both pathologies. The 1-deoxySLs are considered to be dead-end metabolites, as they lack the C1-hydroxyl group, which is essential for the canonical degradation of SLs. Here, we report a previously unknown metabolic pathway, which is capable of degrading 1-deoxySLs. Using a variety of metabolic labeling approaches and high-resolution high-accuracy MS, we identified eight 1-deoxySL downstream metabolites, which appear to be formed by cytochrome P450 (CYP)4F enzymes. Comprehensive inhibition and induction of CYP4F enzymes blocked and stimulated, respectively, the formation of the downstream metabolites. Consequently, CYP4F enzymes might be novel therapeutic targets for the treatment of HSAN1 and DSN, as well as for the prevention of T2D.
Collapse
Affiliation(s)
- Irina Alecu
- Institute for Clinical Chemistry, University Hospital Zurich, Zurich 8091, Switzerland.,Center for Integrative Human Physiology, University of Zurich, Zurich 8057, Switzerland
| | - Alaa Othman
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck D-23562, Germany
| | - Anke Penno
- LIMES Life and Medical Sciences Institute, University of Bonn, Bonn 53115, Germany
| | - Essa M Saied
- Institute for Chemistry, Humboldt Universität zu Berlin, Berlin D-12489, Germany.,Chemistry Department, Suez Canal University, Ismailia 41522, Egypt
| | - Christoph Arenz
- Institute for Chemistry, Humboldt Universität zu Berlin, Berlin D-12489, Germany
| | - Arnold von Eckardstein
- Institute for Clinical Chemistry, University Hospital Zurich, Zurich 8091, Switzerland.,Center for Integrative Human Physiology, University of Zurich, Zurich 8057, Switzerland
| | - Thorsten Hornemann
- Institute for Clinical Chemistry, University Hospital Zurich, Zurich 8091, Switzerland .,Center for Integrative Human Physiology, University of Zurich, Zurich 8057, Switzerland
| |
Collapse
|