1
|
Wang LY, Derks RJE, Brewster KAJ, Prtvar D, Tahirovic S, Berghoff SA, Giera M. Label-free quantitative shotgun analysis of bis(monoacylglycero)phosphate lipids. Anal Bioanal Chem 2025:10.1007/s00216-025-05890-4. [PMID: 40343460 DOI: 10.1007/s00216-025-05890-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/31/2025] [Accepted: 04/17/2025] [Indexed: 05/11/2025]
Abstract
Interest in the role of bis(monoacylglycero)phosphate (BMP) lipids in lysosomal function has significantly grown in recent years. Emerging evidence highlights BMPs as critical players not only in Niemann-Pick disease type C (NPC) but also in other pathologies such as neurodegeneration, cardiovascular diseases, and cancers. However, the selective analysis of BMPs is significantly hindered by isomeric phosphatidylglycerol (PG) lipids. While this can be addressed by chromatographic separation, it poses a significant challenge for shotgun lipidomics approaches. Here, we present a shotgun lipidomics strategy to detect and separate BMPs from PGs using differential fragmentation of sodiated ions. This approach, including isotope correction, is integrated into an existing quantitative shotgun lipidomics workflow (Lipidyzer combined with Shotgun Lipidomics Assistant software) that simultaneously quantifies >1400 lipids. Validation using K-562 cell extracts demonstrated acceptable linearity, trueness, repeatability, and a limit of quantification of 0.12 µM, confirming robust analytical performance. Finally, characteristic accumulation of BMP lipids is shown in bone marrow-derived macrophages from NPC mice, demonstrating its applicability. Our method presents a quantitative, selective, rapid, and robust solution for shotgun-based BMP analysis without the need for extensive chromatographic separation or derivatization. The integration of BMP lipid detection into the Lipidyzer platform, alongside the recently launched iSODA data visualization tool, empowers chemists and biologists to gain deeper insights into BMP lipid biology.
Collapse
Affiliation(s)
- Lian Y Wang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333ZA, The Netherlands
| | - Rico J E Derks
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333ZA, The Netherlands
| | - Kevin A J Brewster
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333ZA, The Netherlands
| | - Danilo Prtvar
- German Center for Neurodegenerative Diseases (DZNE), Munich, 81377, Germany
| | - Sabina Tahirovic
- German Center for Neurodegenerative Diseases (DZNE), Munich, 81377, Germany
| | - Stefan A Berghoff
- German Center for Neurodegenerative Diseases (DZNE), Munich, 81377, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333ZA, The Netherlands.
| |
Collapse
|
2
|
Davidson JW, Jain R, Kizzar T, Geoghegan G, Nesbitt DJ, Cavanagh A, Abe A, Nyame K, Hunger A, Chao X, James I, Walesewicz H, Baldwin DA, Wade G, Michorowska S, Verma R, Schueler K, Hinkovska-Galcheva V, Shishkova E, Ding WX, Coon JJ, Shayman JA, Abu-Remaileh M, Simcox JA. Hepatic lipid remodeling in cold exposure uncovers direct regulation of bis(monoacylglycero)phosphate lipids by phospholipase A2 group XV. Cell Metab 2025:S1550-4131(25)00253-0. [PMID: 40373767 DOI: 10.1016/j.cmet.2025.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/13/2025] [Accepted: 04/21/2025] [Indexed: 05/17/2025]
Abstract
Cold exposure is a selective environmental stress that elicits a rapid metabolic shift to maintain energy homeostasis. In response to cold exposure, the liver rewires the metabolic state, shifting from glucose to lipid catabolism. By probing the liver lipids in cold exposure, we observed that the lysosomal bis(monoacylglycero)phosphate (BMP) lipids were rapidly increased during cold exposure. BMP lipid changes occurred independently of lysosomal abundance but were dependent on the lysosomal transcriptional regulator transcription factor EB (TFEB). Knockdown of Tfeb in hepatocytes decreased BMP lipid levels and led to cold intolerance in mice. We assessed TFEB-binding sites of lysosomal genes and determined that the phospholipase a2 group XV (PLA2G15) regulates BMP lipid catabolism. Decreasing Pla2g15 levels in mice increased BMP lipids, ablated the cold-induced rise in BMP lipids, and improved cold tolerance. Mutation of the catalytic site of PLA2G15 ablated the BMP lipid breakdown. Together, our studies uncover TFEB regulation of BMP lipids through PLA2G15 catabolism.
Collapse
Affiliation(s)
- Jessica W Davidson
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Raghav Jain
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Howard Hughes Medical Institute, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Thomas Kizzar
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Gisela Geoghegan
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Daniel J Nesbitt
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Amy Cavanagh
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Akira Abe
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kwamina Nyame
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA; The Institute for Chemistry, Engineering & Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
| | - Andrea Hunger
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Xiaojuan Chao
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Isabella James
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Helaina Walesewicz
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dominique A Baldwin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Howard Hughes Medical Institute, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Gina Wade
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sylwia Michorowska
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Drug Chemistry, Pharmaceutical and Biomedical Analysis, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | - Rakesh Verma
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kathryn Schueler
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | - Evgenia Shishkova
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Joshua J Coon
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Morgridge Institute for Research, Madison, WI 53715, USA
| | - James A Shayman
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Monther Abu-Remaileh
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA; The Institute for Chemistry, Engineering & Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
| | - Judith A Simcox
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Howard Hughes Medical Institute, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
3
|
Davidson JW, Jain R, Kizzar T, Geoghegan G, Nesbitt DJ, Cavanagh A, Abe A, Nyame K, Hunger A, Chao X, James I, Von Bank H, Baldwin DA, Wade G, Michorowska S, Verma R, Scheuler K, Hinkovska-Galcheva V, Shishkova E, Ding WX, Coon JJ, Shayman JA, Abu-Remaileh M, Simcox JA. Modulation of hepatic transcription factor EB activity during cold exposure uncovers direct regulation of bis(monoacylglycero)phosphate lipids by Pla2g15. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.11.03.565498. [PMID: 37986778 PMCID: PMC10659384 DOI: 10.1101/2023.11.03.565498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Cold exposure is a selective environmental stress that elicits a rapid metabolic shift to maintain energy homeostasis. In response to cold exposure, the liver rewires the metabolic state shifting from glucose to lipid catabolism. By probing the liver lipids in cold exposure, we observed that the lysosomal bis(monoacylglycero)phosphate (BMP) lipids were rapidly increased during cold exposure. BMP lipid changes occurred independently of lysosomal abundance but were dependent on the lysosomal transcriptional regulator transcription factor EB (TFEB). Knockdown of TFEB in hepatocytes decreased BMP lipid levels and led to cold intolerance in mice. We assessed TFEB binding sites of lysosomal genes and determined that the phospholipase Pla2g15 regulates BMP lipid catabolism. Knockdown of Pla2g15 in mice increased BMP lipid levels, ablated the cold-induced rise, and improved cold tolerance. Knockout of Pla2g15 in mice and hepatocytes led to increased BMP lipid levels, that were decreased with re-expression of Pla2g15. Mutation of the catalytic site of Pla2g15 ablated the BMP lipid breakdown. Together, our studies uncover TFEB regulation of BMP lipids through Pla2g15 catabolism.
Collapse
|
4
|
Lopez ME, Wendt D, Lawrence R, Gong K, Ong H, Yip B, Chen J, Mangini L, Handyside B, Giaramita A, Lamichhane A, Lo M, Agrawal V, Van Vleet J, Abolhesn A, Felix JB, Villalpando I, Bhat V, De Angelis R, Ru Y, Khan A, Fong S, Christianson T, Bullens S, Crawford BE, Bunting S, Aoyagi-Scharber M. Intracerebroventricular administration of a modified hexosaminidase ameliorates late-stage neurodegeneration in a GM2 mouse model. PLoS One 2025; 20:e0315005. [PMID: 39752451 PMCID: PMC11698352 DOI: 10.1371/journal.pone.0315005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 11/19/2024] [Indexed: 01/06/2025] Open
Abstract
The GM2 gangliosidoses, Tay-Sachs disease and Sandhoff disease, are devastating neurodegenerative disorders caused by β-hexosaminidase A (HexA) deficiency. In the Sandhoff disease mouse model, rescue potential was severely reduced when HexA was introduced after disease onset. Here, we assess the effect of recombinant HexA and HexD3, a newly engineered mimetic of HexA optimized for the treatment of Tay-Sachs disease and Sandhoff disease. Enzyme replacement therapy was administered by repeat intracerebroventricular injections in Sandhoff disease model mice with dosing beginning before and after signs of neurodegeneration. As previously observed, HexA effectively increased the lifespan of Sandhoff disease mice by 3.5-fold only when treatment was started before onset of neurodegeneration. In contrast, HexD3 halted motor decline and ameliorated late-stage disease severity even when dosing began late, after neurodegeneration onset. Additionally, HexD3 had advantages over HexA in enzyme stability, distribution potential, and homodimer activity. Overall, our data indicate that advanced therapeutics may widen the treatment window for neurodegenerative disorders.
Collapse
Affiliation(s)
- Manuel E. Lopez
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Daniel Wendt
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Roger Lawrence
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Kerui Gong
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Hoonsan Ong
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Bryan Yip
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Joseph Chen
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Linley Mangini
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Britta Handyside
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | | | | | - Melanie Lo
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Vishal Agrawal
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Jeremy Van Vleet
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Amanda Abolhesn
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Jessica B. Felix
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | | | - Vikas Bhat
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | | | - Yuanbin Ru
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Ayesha Khan
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Sylvia Fong
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | | | - Sherry Bullens
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | | | - Stuart Bunting
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | | |
Collapse
|
5
|
Bulfon D, Breithofer J, Grabner GF, Fawzy N, Pirchheim A, Wolinski H, Kolb D, Hartig L, Tischitz M, Zitta C, Bramerdorfer G, Lass A, Taschler U, Kratky D, Greimel P, Zimmermann R. Functionally overlapping intra- and extralysosomal pathways promote bis(monoacylglycero)phosphate synthesis in mammalian cells. Nat Commun 2024; 15:9937. [PMID: 39548099 PMCID: PMC11568333 DOI: 10.1038/s41467-024-54213-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 11/03/2024] [Indexed: 11/17/2024] Open
Abstract
Bis(monoacylglycero)phosphate (BMP) is a major phospholipid constituent of intralumenal membranes in late endosomes/lysosomes, where it regulates the degradation and sorting of lipid cargo. Recent observations suggest that the Batten disease-associated protein CLN5 functions as lysosomal BMP synthase. Here, we show that transacylation reactions catalyzed by cytosolic and secreted enzymes enhance BMP synthesis independently of CLN5. The transacylases identified in this study are capable of acylating the precursor lipid phosphatidylglycerol (PG), generating acyl-PG, which is subsequently hydrolyzed to BMP. Extracellularly, acyl-PG and BMP are generated by endothelial lipase in cooperation with other serum enzymes of the pancreatic lipase family. The intracellular acylation of PG is catalyzed by several members of the cytosolic phospholipase A2 group IV (PLA2G4) family. Overexpression of secreted or cytosolic transacylases was sufficient to correct BMP deficiency in HEK293 cells lacking CLN5. Collectively, our observations suggest that functionally overlapping pathways promote BMP synthesis in mammalian cells.
Collapse
Grants
- Funding: this work was supported by SFB Lipid hydrolysis (10.55776/F73, D.K., R.Z.), 10.55776/P28533 (R.Z.), 10.55776/P35532 (R.Z.), the doctoral program doc-fund “Molecular Metabolism” 10.55776/DOC50 funded by the Austrian Science Fund FWF, Field of Excellence BioHealth – University of Graz, Graz, Austria, Province of Styria, City of Graz, BioTechMed-Graz, and NAWI Graz, and the Glycolipidologue Program of RIKEN (P.G.). For open access purposes, the authors have applied a CC BY public copyright license to any author accepted manuscript version arising from this submission
Collapse
Affiliation(s)
- Dominik Bulfon
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | | - Gernot F Grabner
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Nermeen Fawzy
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Anita Pirchheim
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Heimo Wolinski
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Dagmar Kolb
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
- Core Facility Ultrastructure Analysis, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Lennart Hartig
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Martin Tischitz
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Clara Zitta
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | | - Achim Lass
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Ulrike Taschler
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Peter Greimel
- Laboratory for Cell Function Dynamics, Center for Brain Science, RIKEN, Wako, Saitama, Japan
| | - Robert Zimmermann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria.
- Field of Excellence BioHealth, University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
6
|
Li Z, Chen L, Qu L, Yu W, Liu T, Ning F, Li J, Guo X, Sun F, Sun B, Luo L. Potential implications of natural compounds on aging and metabolic regulation. Ageing Res Rev 2024; 101:102475. [PMID: 39222665 DOI: 10.1016/j.arr.2024.102475] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Aging is generally accompanied by a progressive loss of metabolic homeostasis. Targeting metabolic processes is an attractive strategy for healthy-aging. Numerous natural compounds have demonstrated strong anti-aging effects. This review summarizes recent findings on metabolic pathways involved in aging and explores the anti-aging effects of natural compounds by modulating these pathways. The potential anti-aging effects of natural extracts rich in biologically active compounds are also discussed. Regulating the metabolism of carbohydrates, proteins, lipids, and nicotinamide adenine dinucleotide is an important strategy for delaying aging. Furthermore, phenolic compounds, terpenoids, alkaloids, and nucleotide compounds have shown particularly promising effects on aging, especially with respect to metabolism regulation. Moreover, metabolomics is a valuable tool for uncovering potential targets against aging. Future research should focus on identifying novel natural compounds that regulate human metabolism and should delve deeper into the mechanisms of metabolic regulation using metabolomics methods, aiming to delay aging and extend lifespan.
Collapse
Affiliation(s)
- Zhuozhen Li
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Lili Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; School of Life Science, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Liangliang Qu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Wenjie Yu
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Tao Liu
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Fangjian Ning
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Jinwang Li
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Xiali Guo
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Fengjie Sun
- Department of Biological Sciences, School of Science and Technology, Georgia Gwinnett College, Lawrenceville, GA 30043, USA
| | - Baoguo Sun
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Liping Luo
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China.
| |
Collapse
|
7
|
Good CJ, Butrico CE, Colley ME, Emmerson LN, Gibson-Corley KN, Cassat JE, Spraggins JM, Caprioli RM. Uncovering lipid dynamics in Staphylococcus aureus osteomyelitis using multimodal imaging mass spectrometry. Cell Chem Biol 2024; 31:1852-1868.e5. [PMID: 39389064 PMCID: PMC11977171 DOI: 10.1016/j.chembiol.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 06/20/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024]
Abstract
Osteomyelitis occurs when Staphylococcus aureus invades the bone microenvironment, resulting in a bone marrow abscess with a spatially defined architecture of cells and biomolecules. Imaging mass spectrometry and microscopy are tools that can be employed to interrogate the lipidome of S. aureus-infected murine femurs and reveal metabolic and signaling consequences of infection. Here, nearly 250 lipids were spatially mapped to healthy and infection-associated morphological features throughout the femur, establishing composition profiles for tissue types. Ether lipids and arachidonoyl lipids were altered between cells and tissue structures in abscesses, suggesting their roles in abscess formation and inflammatory signaling. Sterols, triglycerides, bis(monoacylglycero)phosphates, and gangliosides possessed ring-like distributions throughout the abscess, suggesting a hypothesized dysregulation of lipid metabolism in a population of cells that cannot be discerned with traditional microscopy. These data provide insight into the signaling function and metabolism of cells in the fibrotic border of abscesses, likely characteristic of lipid-laden macrophages.
Collapse
Affiliation(s)
- Christopher J Good
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
| | - Casey E Butrico
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Madeline E Colley
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA
| | - Lauren N Emmerson
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA; Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN 37235, USA
| | - Katherine N Gibson-Corley
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - James E Cassat
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jeffrey M Spraggins
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA.
| | - Richard M Caprioli
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA; Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
8
|
Janssens GE, Molenaars M, Herzog K, Grevendonk L, Remie CME, Vervaart MAT, Elfrink HL, Wever EJM, Schomakers BV, Denis SW, Waterham HR, Pras-Raves ML, van Weeghel M, van Kampen AHC, Tammaro A, Butter LM, van der Rijt S, Florquin S, Jongejan A, Moerland PD, Hoeks J, Schrauwen P, Vaz FM, Houtkooper RH. A conserved complex lipid signature marks human muscle aging and responds to short-term exercise. NATURE AGING 2024; 4:681-693. [PMID: 38609524 DOI: 10.1038/s43587-024-00595-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/22/2024] [Indexed: 04/14/2024]
Abstract
Studies in preclinical models suggest that complex lipids, such as phospholipids, play a role in the regulation of longevity. However, identification of universally conserved complex lipid changes that occur during aging, and how these respond to interventions, is lacking. Here, to comprehensively map how complex lipids change during aging, we profiled ten tissues in young versus aged mice using a lipidomics platform. Strikingly, from >1,200 unique lipids, we found a tissue-wide accumulation of bis(monoacylglycero)phosphate (BMP) during mouse aging. To investigate translational value, we assessed muscle tissue of young and older people, and found a similar marked BMP accumulation in the human aging lipidome. Furthermore, we found that a healthy-aging intervention consisting of moderate-to-vigorous exercise was able to lower BMP levels in postmenopausal female research participants. Our work implicates complex lipid biology as central to aging, identifying a conserved aging lipid signature of BMP accumulation that is modifiable upon a short-term healthy-aging intervention.
Collapse
Affiliation(s)
- Georges E Janssens
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands.
- Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, the Netherlands.
| | - Marte Molenaars
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, the Netherlands
| | - Katharina Herzog
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, the Netherlands
| | - Lotte Grevendonk
- Department of Nutrition and Human Movement Sciences, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre, Maastricht, the Netherlands
- TI Food and Nutrition, Wageningen, the Netherlands
| | - Carlijn M E Remie
- Department of Nutrition and Human Movement Sciences, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Martin A T Vervaart
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Core Facility Metabolomics, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Hyung L Elfrink
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Core Facility Metabolomics, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Eric J M Wever
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Core Facility Metabolomics, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Epidemiology and Data Science, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Bauke V Schomakers
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Core Facility Metabolomics, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Simone W Denis
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Hans R Waterham
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, the Netherlands
| | - Mia L Pras-Raves
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Core Facility Metabolomics, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Epidemiology and Data Science, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Core Facility Metabolomics, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Antoine H C van Kampen
- Epidemiology and Data Science, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health Methodology, Amsterdam, the Netherlands
- Amsterdam Infection and Immunity, Inflammatory Diseases, Amsterdam, the Netherlands
| | - Alessandra Tammaro
- Pathology Department, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Infection and Immunity, Amsterdam, the Netherlands
| | - Loes M Butter
- Pathology Department, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Infection and Immunity, Amsterdam, the Netherlands
| | - Sanne van der Rijt
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, the Netherlands
- Pathology Department, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Sandrine Florquin
- Pathology Department, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Infection and Immunity, Amsterdam, the Netherlands
| | - Aldo Jongejan
- Epidemiology and Data Science, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health Methodology, Amsterdam, the Netherlands
- Amsterdam Infection and Immunity, Inflammatory Diseases, Amsterdam, the Netherlands
| | - Perry D Moerland
- Epidemiology and Data Science, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health Methodology, Amsterdam, the Netherlands
- Amsterdam Infection and Immunity, Inflammatory Diseases, Amsterdam, the Netherlands
| | - Joris Hoeks
- Department of Nutrition and Human Movement Sciences, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre, Maastricht, the Netherlands
- TI Food and Nutrition, Wageningen, the Netherlands
| | - Patrick Schrauwen
- Department of Nutrition and Human Movement Sciences, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre, Maastricht, the Netherlands
- TI Food and Nutrition, Wageningen, the Netherlands
| | - Frédéric M Vaz
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands.
- Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, the Netherlands.
- Core Facility Metabolomics, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands.
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands.
- Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, the Netherlands.
- Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands.
| |
Collapse
|
9
|
Birtles D, Abbas W, Lee J. Bis(Monoacylglycero)Phosphate Promotes Membrane Fusion Facilitated by the SARS-CoV-2 Fusion Domain. J Phys Chem B 2024; 128:2675-2683. [PMID: 38466655 DOI: 10.1021/acs.jpcb.3c07863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Membrane fusion is a critical component of the viral lifecycle. For SARS-CoV-2, fusion is facilitated by the spike glycoprotein and can take place via either the plasma membrane or the endocytic pathway. The fusion domain (FD), which is found within the spike glycoprotein, is primarily responsible for the initiation of fusion as it embeds itself within the target cell's membrane. A preference for SARS-CoV-2 to fuse at low pH akin to the environment of the endocytic pathway has already been established; however, the impact of the target cell's lipid composition on the FD has yet to be explored. Here, we have shown that the SARS-CoV-2 FD preferentially initiates fusion at the late endosomal membrane over the plasma membrane, on the basis of lipid composition alone. A positive, fusogenic relationship with anionic lipids from the plasma membrane (POPS: 1-palmitoyl-2-oleoyl-sn-glycero-3-phospho-l-serine) and endosomal membrane (BMP: bis(monoacylglycero)phosphate) was established, with a large preference demonstrated for the latter. When comparing the binding affinity and secondary structure of the FD in the presence of different anionic lipids, little deviation was evident while the charge was maintained. However, it was discovered that BMP had a subtle, negative impact on lipid packing in comparison to that of POPS. Furthermore, an inverse relationship between lipid packing and the fusogenecity of the SARS-CoV-2 FD was witnessed. In conclusion, the SARS-CoV-2 FD preferentially initiates fusion at a membrane resembling that of the late endosomal compartment, predominately due to the presence of BMP and its impact on lipid packing.
Collapse
Affiliation(s)
- Daniel Birtles
- Department of Chemistry and Biochemistry, University of Maryland, College Park 20742, Maryland, United States
| | - Wafa Abbas
- Department of Chemistry and Biochemistry, University of Maryland, College Park 20742, Maryland, United States
| | - Jinwoo Lee
- Department of Chemistry and Biochemistry, University of Maryland, College Park 20742, Maryland, United States
| |
Collapse
|
10
|
Good CJ, Butrico CE, Colley ME, Gibson-Corley KN, Cassat JE, Spraggins JM, Caprioli RM. In situ lipidomics of Staphylococcus aureus osteomyelitis using imaging mass spectrometry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.569690. [PMID: 38077019 PMCID: PMC10705574 DOI: 10.1101/2023.12.01.569690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Osteomyelitis occurs when Staphylococcus aureus invades the bone microenvironment, resulting in a bone marrow abscess with a spatially defined architecture of cells and biomolecules. Imaging mass spectrometry and microscopy are invaluable tools that can be employed to interrogate the lipidome of S. aureus-infected murine femurs to reveal metabolic and signaling consequences of infection. Here, nearly 250 lipids were spatially mapped to healthy and infection-associated morphological features throughout the femur, establishing composition profiles for tissue types. Ether lipids and arachidonoyl lipids were significantly altered between cells and tissue structures in abscesses, suggesting their roles in abscess formation and inflammatory signaling. Sterols, triglycerides, bis(monoacylglycero)phosphates, and gangliosides possessed ring-like distributions throughout the abscess, indicating dysregulated lipid metabolism in a subpopulation of leukocytes that cannot be discerned with traditional microscopy. These data provide chemical insight into the signaling function and metabolism of cells in the fibrotic border of abscesses, likely characteristic of lipid-laden macrophages.
Collapse
Affiliation(s)
- Christopher J. Good
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
| | - Casey E. Butrico
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Madeline E. Colley
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA
| | - Katherine N. Gibson-Corley
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - James E. Cassat
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jeffrey M. Spraggins
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - Richard M. Caprioli
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA
- Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
11
|
Huang R, Chen J, Zhou M, Xin H, Lam SM, Jiang X, Li J, Deng F, Shui G, Zhang Z, Li MD. Multi-omics profiling reveals rhythmic liver function shaped by meal timing. Nat Commun 2023; 14:6086. [PMID: 37773240 PMCID: PMC10541894 DOI: 10.1038/s41467-023-41759-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 09/06/2023] [Indexed: 10/01/2023] Open
Abstract
Post-translational modifications (PTMs) couple feed-fast cycles to diurnal rhythms. However, it remains largely uncharacterized whether and how meal timing organizes diurnal rhythms beyond the transcriptome. Here, we systematically profile the daily rhythms of the proteome, four PTMs (phosphorylation, ubiquitylation, succinylation and N-glycosylation) and the lipidome in the liver from young female mice subjected to either day/sleep time-restricted feeding (DRF) or night/wake time-restricted feeding (NRF). We detect robust daily rhythms among different layers of omics with phosphorylation the most nutrient-responsive and succinylation the least. Integrative analyses reveal that clock regulation of fatty acid metabolism represents a key diurnal feature that is reset by meal timing, as indicated by the rhythmic phosphorylation of the circadian repressor PERIOD2 at Ser971 (PER2-pSer971). We confirm that PER2-pSer971 is activated by nutrient availability in vivo. Together, this dataset represents a comprehensive resource detailing the proteomic and lipidomic responses by the liver to alterations in meal timing.
Collapse
Affiliation(s)
- Rongfeng Huang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Jianghui Chen
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Meiyu Zhou
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Haoran Xin
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- LipidALL Technologies Company Limited, Changzhou, Jiangsu Province, China
| | - Xiaoqing Jiang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Jie Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Fang Deng
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Zhihui Zhang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China.
| | - Min-Dian Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
12
|
Farnoodian M, Bose D, Barone F, Nelson LM, Boyle M, Jun B, Do K, Gordon W, Guerin MAK, Perera R, Ji JX, Cogliati T, Sharma R, Brooks BP, Bazan NG, Bharti K. Retina and RPE lipid profile changes linked with ABCA4 associated Stargardt's maculopathy. Pharmacol Ther 2023; 249:108482. [PMID: 37385300 PMCID: PMC10530239 DOI: 10.1016/j.pharmthera.2023.108482] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/20/2023] [Accepted: 06/26/2023] [Indexed: 07/01/2023]
Abstract
Stargardt maculopathy, caused predominantly by mutations in the ABCA4 gene, is characterized by an accumulation of non-degradable visual pigment derivative, lipofuscin, in the retinal pigment epithelium (RPE) - resulting in RPE atrophy. RPE is a monolayer tissue located adjacent to retinal photoreceptors and regulates their health and functioning; RPE atrophy triggers photoreceptor cell death and vision loss in Stargardt patients. Previously, ABCA4 mutations in photoreceptors were thought to be the major contributor to lipid homeostasis defects in the eye. Recently, we demonstrated that ABCA4 loss of function in the RPE leads to cell-autonomous lipid homeostasis defects. Our work underscores that an incomplete understanding of lipid metabolism and lipid-mediated signaling in the retina and RPE are potential causes for lacking treatments for this disease. Here we report altered lipidomic in mouse and human Stargardt models. This work provides the basis for therapeutics that aim to restore lipid homeostasis in the retina and the RPE.
Collapse
Affiliation(s)
- Mitra Farnoodian
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Devika Bose
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Francesca Barone
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Luke Mathew Nelson
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Marisa Boyle
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Bokkyoo Jun
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, USA
| | - Khanh Do
- Faculty of Medicine, Phenikaa University, Hanoi, Viet Nam
| | - William Gordon
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, USA
| | - Marie-Audrey Kautzmann Guerin
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, USA
| | - Rasangi Perera
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, USA
| | - Jeff X Ji
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, USA
| | - Tiziana Cogliati
- Division of Aging Biology, National Institute on Aging, National Institute of Health, Bethesda, MD, USA
| | - Ruchi Sharma
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Brian P Brooks
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, USA
| | - Kapil Bharti
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA.
| |
Collapse
|
13
|
Issleny BM, Jamjoum R, Majumder S, Stiban J. Sphingolipids: From structural components to signaling hubs. Enzymes 2023; 54:171-201. [PMID: 37945171 DOI: 10.1016/bs.enz.2023.07.003] [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: 11/12/2023]
Abstract
In late November 2019, Prof. Lina M. Obeid passed away from cancer, a disease she spent her life researching and studying its intricate molecular underpinnings. Along with her husband, Prof. Yusuf A. Hannun, Obeid laid down the foundations of sphingolipid biochemistry and oversaw its remarkable evolution over the years. Lipids are a class of macromolecules that are primarily associated with cellular architecture. In fact, lipids constitute the perimeter of the cell in such a way that without them, there cannot be cells. Hence, much of the early research on lipids identified the function of this class of biological molecules as merely structural. Nevertheless, unlike proteins, carbohydrates, and nucleic acids, lipids are elaborately diverse as they are not made up of monomers in polymeric forms. This diversity in structure is clearly mirrored by functional pleiotropy. In this chapter, we focus on a major subset of lipids, sphingolipids, and explore their historic rise from merely inert structural components of plasma membranes to lively and necessary signaling molecules that transmit various signals and control many cellular processes. We will emphasize the works of Lina Obeid since she was an integral pillar of the sphingolipid research world.
Collapse
Affiliation(s)
- Batoul M Issleny
- Department of Pharmacy, Birzeit University, West Bank, Palestine
| | - Rama Jamjoum
- Department of Pharmacy, Birzeit University, West Bank, Palestine
| | | | - Johnny Stiban
- Department of Biology and Biochemistry, Birzeit University, West Bank, Palestine.
| |
Collapse
|
14
|
Fan LC, McConn K, Plataki M, Kenny S, Williams NC, Kim K, Quirke JA, Chen Y, Sauler M, Möbius ME, Chung KP, Area Gomez E, Choi AM, Xu JF, Cloonan SM. Alveolar type II epithelial cell FASN maintains lipid homeostasis in experimental COPD. JCI Insight 2023; 8:e163403. [PMID: 37606038 PMCID: PMC10543729 DOI: 10.1172/jci.insight.163403] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 07/10/2023] [Indexed: 08/23/2023] Open
Abstract
Alveolar epithelial type II (AEC2) cells strictly regulate lipid metabolism to maintain surfactant synthesis. Loss of AEC2 cell function and surfactant production are implicated in the pathogenesis of the smoking-related lung disease chronic obstructive pulmonary disease (COPD). Whether smoking alters lipid synthesis in AEC2 cells and whether altering lipid metabolism in AEC2 cells contributes to COPD development are unclear. In this study, high-throughput lipidomic analysis revealed increased lipid biosynthesis in AEC2 cells isolated from mice chronically exposed to cigarette smoke (CS). Mice with a targeted deletion of the de novo lipogenesis enzyme, fatty acid synthase (FASN), in AEC2 cells (FasniΔAEC2) exposed to CS exhibited higher bronchoalveolar lavage fluid (BALF) neutrophils, higher BALF protein, and more severe airspace enlargement. FasniΔAEC2 mice exposed to CS had lower levels of key surfactant phospholipids but higher levels of BALF ether phospholipids, sphingomyelins, and polyunsaturated fatty acid-containing phospholipids, as well as increased BALF surface tension. FasniΔAEC2 mice exposed to CS also had higher levels of protective ferroptosis markers in the lung. These data suggest that AEC2 cell FASN modulates the response of the lung to smoke by regulating the composition of the surfactant phospholipidome.
Collapse
Affiliation(s)
- Li-Chao Fan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Keith McConn
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Maria Plataki
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, USA
| | - Sarah Kenny
- School of Medicine, Trinity Biomedical Sciences Institute, and
| | | | - Kihwan Kim
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | | | - Yan Chen
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Maor Sauler
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Kuei-Pin Chung
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- Department of Laboratory Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Estela Area Gomez
- Division of Neuromuscular Medicine, Department of Neurology, Columbia University Irving Medical Center, Neurological Institute, New York, New York, USA
- Center for Biological Research “Margarita Salas”, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Augustine M.K. Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, USA
| | - Jin-Fu Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Suzanne M. Cloonan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- School of Medicine, Trinity Biomedical Sciences Institute, and
| |
Collapse
|
15
|
Bogojevic O, Zhang Y, Wolff CD, Nygaard JV, Wiking L, Arevång C, Guo Z. A sustainable and regioselective synthesis of Hemi-bis(monoacylglycero)phosphates and bis(diacylglycero)phosphates. iScience 2023; 26:107075. [PMID: 37448559 PMCID: PMC10336169 DOI: 10.1016/j.isci.2023.107075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/06/2023] [Accepted: 06/06/2023] [Indexed: 07/15/2023] Open
Abstract
A sustainable and green approach was developed for the scalable synthesis of uncommon naturally occurring phospholipid species, Hemi-bis(monoacylglycero)phosphates (Hemi-BMPs) and bis(diacylglycero)phosphates (BDPs) via the phospholipase D (PLD) mediated transphosphatidylation. PLD from Streptomyces sp. showed great substrate promiscuity for both phospholipids from different biological sources, and alcohol donors with diverse regiochemistry; monoacylglycerols with diverse fatty acyl structures (C12-C22), affording 74-92 wt% yields in 2 h. Experimental results demonstrated that the reaction rate is rather independent of phosphatidyls but to a large extent governed by the size, shape and regiolocation of fatty acyls incorporated on the glycerol backbone, particularly for the regio-isomers of bulky diacylglycerols (Sn-1,3 or Sn-1,2), which displays great diversity. In addition, a plausible mechanism is proposed based on molecular simulations for an elaborated explanation of the reaction thermodynamic and kinetic favorability toward the synthesis of Hemi-BMPs and BDPs.
Collapse
Affiliation(s)
- Oliver Bogojevic
- Department of Biological and Chemical Engineering, Faculty of Technical Sciences, Aarhus University, Gustav Wieds Vej 10, 8000 Aarhus, Denmark
| | - Yan Zhang
- Department of Biological and Chemical Engineering, Faculty of Technical Sciences, Aarhus University, Gustav Wieds Vej 10, 8000 Aarhus, Denmark
| | - Christian Daugaard Wolff
- Department of Biological and Chemical Engineering, Faculty of Technical Sciences, Aarhus University, Gustav Wieds Vej 10, 8000 Aarhus, Denmark
| | - Jens Vinge Nygaard
- Department of Biological and Chemical Engineering, Faculty of Technical Sciences, Aarhus University, Gustav Wieds Vej 10, 8000 Aarhus, Denmark
| | - Lars Wiking
- Department of Food Science, CiFOOD, Aarhus University, Agro Food Park 48, 8200 Aarhus N, Denmark
| | | | - Zheng Guo
- Department of Biological and Chemical Engineering, Faculty of Technical Sciences, Aarhus University, Gustav Wieds Vej 10, 8000 Aarhus, Denmark
| |
Collapse
|
16
|
Meng X, Bi Q, Ma Q, Wei Y, Li Y, Liang M, Xu H. Dietary Cholesterol Differentially Regulates the Muscle Lipidomics of Farmed Turbot and Tiger Puffer. Animals (Basel) 2023; 13:ani13101632. [PMID: 37238062 DOI: 10.3390/ani13101632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/27/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Exogenous cholesterol has been supplemented into aqua-feeds due to the reduced proportions of fishmeal and fish oil. This study aimed to investigate the effects of dietary cholesterol supplementation on the muscle lipidomics of two marine fish species, turbot and tiger puffer. A 70-day feeding trial was conducted, where two low-fishmeal diets supplemented with 0 or 1% cholesterol were used. The lipidomic analysis with targeted tandem mass spectrometry showed that, in turbot, a total of 49 individual lipids exhibited significant differences in their abundance in response to dietary cholesterol, whereas the number was 30 for tiger puffer. Dietary cholesterol up-regulated the abundance of cholesterol and cholesterol ester in both species. In turbot, the dietary cholesterol also increased the abundance of triacylglycerol and acylcarnitine, whereas in tiger puffer, it primarily regulated the abundance of phospholipids and BMP. This was the first time the responses of marine fish muscle lipidomics to dietary cholesterol supplementation have been investigated.
Collapse
Affiliation(s)
- Xiaoxue Meng
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao 266071, China
- College of Fisheries, Guangdong Ocean University, 1 Haida Road, Zhanjiang 524008, China
| | - Qingzhu Bi
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao 266071, China
| | - Qiang Ma
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao 266071, China
| | - Yuliang Wei
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao 266071, China
| | - Yanlu Li
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao 266071, China
| | - Mengqing Liang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao 266071, China
| | - Houguo Xu
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao 266071, China
| |
Collapse
|
17
|
Zeng J, Hao J, Yang Z, Ma C, Gao L, Chen Y, Li G, Li J. Anti-Allergic Effect of Dietary Polyphenols Curcumin and Epigallocatechin Gallate via Anti-Degranulation in IgE/Antigen-Stimulated Mast Cell Model: A Lipidomics Perspective. Metabolites 2023; 13:metabo13050628. [PMID: 37233669 DOI: 10.3390/metabo13050628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/23/2023] [Accepted: 04/28/2023] [Indexed: 05/27/2023] Open
Abstract
Polyphenol-rich foods exhibit anti-allergic/-inflammatory properties. As major effector cells of allergies, mast cells undergo degranulation after activation and then initiate inflammatory responses. Key immune phenomena could be regulated by the production and metabolism of lipid mediators by mast cells. Here, we analyzed the antiallergic activities of two representative dietary polyphenols, curcumin and epigallocatechin gallate (EGCG), and traced their effects on cellular lipidome rewiring in the progression of degranulation. Both curcumin and EGCG significantly inhibited degranulation as they suppressed the release of β-hexosaminidase, interleukin-4, and tumor necrosis factor-α from the IgE/antigen-stimulated mast cell model. A comprehensive lipidomics study involving 957 identified lipid species revealed that although the lipidome remodeling patterns (lipid response and composition) of curcumin intervention were considerably similar to those of EGCG, lipid metabolism was more potently disturbed by curcumin. Seventy-eight percent of significant differential lipids upon IgE/antigen stimulation could be regulated by curcumin/EGCG. LPC-O 22:0 was defined as a potential biomarker for its sensitivity to IgE/antigen stimulation and curcumin/EGCG intervention. The key changes in diacylglycerols, fatty acids, and bismonoacylglycerophosphates provided clues that cell signaling disturbances could be associated with curcumin/EGCG intervention. Our work supplies a novel perspective for understanding curcumin/EGCG involvement in antianaphylaxis and helps guide future attempts to use dietary polyphenols.
Collapse
Affiliation(s)
- Jun Zeng
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
- Xiamen Key Laboratory of Marine Functional Food, Xiamen 361021, China
| | - Jingwen Hao
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Zhiqiang Yang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Chunyu Ma
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Longhua Gao
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Yue Chen
- The Affiliated Stomatology Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, China
| | - Guiling Li
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
- Xiamen Key Laboratory of Marine Functional Food, Xiamen 361021, China
| | - Jia Li
- Key Laboratory of Tea Biology and Resources Utilization, Ministry of Agriculture, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou 310008, China
| |
Collapse
|
18
|
Boland S, Swarup S, Ambaw YA, Malia PC, Richards RC, Fischer AW, Singh S, Aggarwal G, Spina S, Nana AL, Grinberg LT, Seeley WW, Surma MA, Klose C, Paulo JA, Nguyen AD, Harper JW, Walther TC, Farese RV. Deficiency of the frontotemporal dementia gene GRN results in gangliosidosis. Nat Commun 2022; 13:5924. [PMID: 36207292 PMCID: PMC9546883 DOI: 10.1038/s41467-022-33500-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 09/21/2022] [Indexed: 02/07/2023] Open
Abstract
Haploinsufficiency of GRN causes frontotemporal dementia (FTD). The GRN locus produces progranulin (PGRN), which is cleaved to lysosomal granulin polypeptides. The function of lysosomal granulins and why their absence causes neurodegeneration are unclear. Here we discover that PGRN-deficient human cells and murine brains, as well as human frontal lobes from GRN-mutation FTD patients have increased levels of gangliosides, glycosphingolipids that contain sialic acid. In these cells and tissues, levels of lysosomal enzymes that catabolize gangliosides were normal, but levels of bis(monoacylglycero)phosphates (BMP), lipids required for ganglioside catabolism, were reduced with PGRN deficiency. Our findings indicate that granulins are required to maintain BMP levels to support ganglioside catabolism, and that PGRN deficiency in lysosomes leads to gangliosidosis. Lysosomal ganglioside accumulation may contribute to neuroinflammation and neurodegeneration susceptibility observed in FTD due to PGRN deficiency and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Sebastian Boland
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Sharan Swarup
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Yohannes A Ambaw
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
- Center on Causes and Prevention of Cardiovascular Disease, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Pedro C Malia
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Ruth C Richards
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Alexander W Fischer
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Shubham Singh
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Geetika Aggarwal
- Department of Internal Medicine, Division of Geriatric Medicine, and Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA
| | - Salvatore Spina
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Alissa L Nana
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Lea T Grinberg
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Pathology, University of California at San Francisco, San Francisco, CA, USA
| | - William W Seeley
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Pathology, University of California at San Francisco, San Francisco, CA, USA
| | | | | | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Andrew D Nguyen
- Department of Internal Medicine, Division of Geriatric Medicine, and Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA
| | - J Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
| | - Tobias C Walther
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA.
- Center on Causes and Prevention of Cardiovascular Disease, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA.
- Howard Hughes Medical Institute, Boston, MA, 02115, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, 02124, USA.
| | - Robert V Farese
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA.
- Center on Causes and Prevention of Cardiovascular Disease, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, 02124, USA.
| |
Collapse
|
19
|
Can M, Sengül T, Demir SA, İnci OK, Basırlı H, Seyrantepe V. Analysis of Brain Lipids in the Early-Onset Tay–Sachs Disease Mouse Model With the Combined Deficiency of β-Hexosaminidase A and Neuraminidase 3. Front Mol Biosci 2022; 9:892248. [PMID: 36003081 PMCID: PMC9393265 DOI: 10.3389/fmolb.2022.892248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction: Tay–Sachs disease is an autosomal recessively inherited lysosomal storage disease that results from loss-of-function mutations in the HEXA gene coding β-hexosaminidase A. HEXA gene deficiency affects the central nervous system owing to GM2 ganglioside accumulation in lysosomes resulting in progressive neurodegeneration in patients. We recently generated a novel mice model with a combined deficiency of β-hexosaminidase A and neuraminidase 3 (Hexa−/−Neu3−/−) that mimics both the neuropathological and clinical abnormalities of early-onset Tay–Sachs disease. Here, we aimed to explore the secondary accumulation of lipids in the brain of Hexa−/−Neu3−/− mice.Materials and Methods: In the cortex and hippocampus of five-month-old WT, Hexa−/−, Neu3−/−, and Hexa−/−Neu3−/− mice, lipid levels belonging to glycerolipids, glycerophospholipids, and sterol lipids were evaluated using a shotgun lipidomics approach. The levels of myelin were also assessed by luxol fast blue staining and immunohistochemistry using antibodies against myelin basic protein. We further examined glycoconjugate and cholesterol levels by periodic acid–Schiff and filipin staining, respectively. Toluidine blue staining was also performed to display axonal degeneration.Results: Among glycerophospholipids, we demonstrated elevated levels of phosphatidylcholine-ether and lysophosphatidylcholine while decreased levels of phosphatidylcholine and phosphatidylserine in both cortex and hippocampus of Hexa−/−Neu3−/− mice. In the glycerolipid class, we showed an alleviated level of sphingomyelin in both cortex and hippocampus, but the higher levels of diacylglycerol and triacylglycerol were detected in only the hippocampus of Hexa−/−Neu3−/− mice. The lower level of sterol was also detected in the cortex of Hexa−/−Neu3−/− mice but not in the hippocampus.Histochemical studies showed a decrease in the myelin level and axonal degeneration indicating neuronal pathology in the brain of Hexa−/−Neu3−/− mice. Although glycoconjugate accumulation was evident both in the cortex and hippocampus, we did not detect any changes in the level of cholesterol.Conclusion: Our results indicate that alterations in lipid metabolism and neuropathology, such as demyelination and axonal degeneration, might be related to the dysfunctionality of lipid-related cellular pathways like autophagy. Understanding of brain-specific lipid alterations contributes to evaluating the effectiveness of treatments in Hexa−/−Neu3−/− mice in future studies.
Collapse
Affiliation(s)
- Melike Can
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkey
| | - Tugce Sengül
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkey
| | | | - Orhan K. İnci
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkey
| | - Hande Basırlı
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkey
| | - Volkan Seyrantepe
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkey
- Izmir Institute of Technology, IYTEDEHAM, Izmir, Turkey
- *Correspondence: Volkan Seyrantepe,
| |
Collapse
|
20
|
Designer phospholipids – structural retrieval, chemo-/bio- synthesis and isotopic labeling. Biotechnol Adv 2022; 60:108025. [DOI: 10.1016/j.biotechadv.2022.108025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 07/12/2022] [Accepted: 07/26/2022] [Indexed: 11/23/2022]
|
21
|
Zaretsky DV, Zaretskaia MV, Molkov YI. Membrane channel hypothesis of lysosomal permeabilization by beta-amyloid. Neurosci Lett 2021; 770:136338. [PMID: 34767924 DOI: 10.1016/j.neulet.2021.136338] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 09/29/2021] [Accepted: 11/03/2021] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia affecting millions of people. Neuronal death in AD is initiated by oligomeric amyloid-β (Aβ) peptides. Recently, we proposed the amyloid degradation toxicity hypothesis, which explains multiple major observations associated with AD including autophagy failure and a decreased metabolism. According to the hypothesis, the key event in the cellular toxicity of amyloid is the formation of non-selective membrane channels in lysosomal membranes by amyloid fragments that are produced by the digestion of Aβ previously absorbed by endocytosis. Electrophysiological data suggest that amyloid-formed channels have different sizes, which can be explained by the fact that channel creating barrel-shaped amyloid aggregates can consist of different number of monomers. To estimate the ability of channels to leak molecules of various molecular weights, we modeled the channels as saline-filled cylinders in non-conductive membranes that pass spheres with a density of average globular proteins. As a basis, we used the conductance distribution taken from the previously published experimental dataset, in which single channels with electrical conductance of up to one nanosiemens were registered. Our calculations show that channels with such a giant conductance can allow for passing macromolecules such as large as lysosomal cathepsins implicated in the activation of apoptosis. The formation of giant channels is disproportionally promoted in an acidic environment. Also, amyloid fragments leaking from permeabilized lysosomes can reach the internal leaflet of the plasma membrane and permeabilize it. We conclude that while dissipation of the proton gradient by any (even smallest) amyloid channels readily explains lysosomal failure, the relatively rare events of lysosomal permeabilization to large macromolecules can be an additional mechanism of cellular death induced by exposure to Aβ.
Collapse
Affiliation(s)
| | | | - Yaroslav I Molkov
- Department of Mathematics and Statistics and Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States
| |
Collapse
|
22
|
Sighinolfi G, Clark S, Blanc L, Cota D, Rhourri-Frih B. Mass spectrometry imaging of mice brain lipid profile changes over time under high fat diet. Sci Rep 2021; 11:19664. [PMID: 34608169 PMCID: PMC8490458 DOI: 10.1038/s41598-021-97201-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 07/19/2021] [Indexed: 12/13/2022] Open
Abstract
Overweight and obesity have been shown to significantly affect brain structures and size. Obesity has been associated with cerebral atrophy, alteration of brain functions, including cognitive impairement, and psychiatric diseases such as depression. Given the importance of lipids in the structure of the brain, here, by using 47 mice fed a high fat diet (HFD) with 60% calories from fat (40% saturated fatty acids) and 20% calories from carbohydrates and age-matched control animals on a normal chow diet, we examined the effects of HFD and diet-induced obesity on the brain lipidome. Using a targeted liquid chromatography mass spectrometry analysis and a non-targeted mass spectrometry MALDI imaging approach, we show that the relative concentration of most lipids, in particular brain phospholipids, is modified by diet-induced obesity (+ 40%of body weight). Use of a non-targeted MALDI-MS imaging approach further allowed define cerebral regions of interest (ROI) involved in eating behavior and changes in their lipid profile. Principal component analysis (PCA) of the obese/chow lipidome revealed persistence of some of the changes in the brain lipidome of obese animals even after their switch to chow feeding and associated weight loss. Altogether, these data reveal that HFD feeding rapidly modifies the murine brain lipidome. Some of these HFD-induced changes persist even after weight loss, implying that some brain sequelae caused by diet-induced obesity are irreversible.
Collapse
Affiliation(s)
| | - Samantha Clark
- Physiopathologie de la Plasticité Neuronale, U1215, Neurocentre Magendie, INSERM, 33000, Bordeaux, France
- Physiopathologie de la Plasticité Neuronale, U1215, Neurocentre Magendie, University of Bordeaux, 33000, Bordeaux, France
| | | | - Daniela Cota
- Physiopathologie de la Plasticité Neuronale, U1215, Neurocentre Magendie, INSERM, 33000, Bordeaux, France
- Physiopathologie de la Plasticité Neuronale, U1215, Neurocentre Magendie, University of Bordeaux, 33000, Bordeaux, France
| | | |
Collapse
|
23
|
Czolkoss S, Borgert P, Poppenga T, Hölzl G, Aktas M, Narberhaus F. Synthesis of the unusual lipid bis(monoacylglycero)phosphate in environmental bacteria. Environ Microbiol 2021; 23:6993-7008. [PMID: 34528360 DOI: 10.1111/1462-2920.15777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 01/05/2023]
Abstract
The bacterial membrane is constantly remodelled in response to environmental conditions and the external supply of precursor molecules. Some bacteria are able to acquire exogenous lyso-phospholipids and convert them to the corresponding phospholipids. Here, we report that some soil-dwelling bacteria have alternative options to metabolize lyso-phosphatidylglycerol (L-PG). We find that the plant-pathogen Agrobacterium tumefaciens takes up this mono-acylated phospholipid and converts it to two distinct isoforms of the non-canonical lipid bis(monoacylglycero)phosphate (BMP). Chromatographic separation and quadrupole-time-of-flight MS/MS analysis revealed the presence of two possible BMP stereo configurations acylated at either of the free hydroxyl groups of the glycerol head group. BMP accumulated in the inner membrane and did not visibly alter cell morphology and growth behaviour. The plant-associated bacterium Sinorhizobium meliloti was also able to convert externally provided L-PG to BMP. Other bacteria like Pseudomonas fluorescens and Escherichia coli metabolized L-PG after cell disruption, suggesting that BMP production in the natural habitat relies both on dedicated uptake systems and on head-group acylation enzymes. Overall, our study adds two previously overlooked phospholipids to the repertoire of bacterial membrane lipids and provides evidence for the remarkable condition-responsive adaptation of bacterial membranes.
Collapse
Affiliation(s)
- Simon Czolkoss
- Microbial Biology, Ruhr University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Pia Borgert
- Microbial Biology, Ruhr University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Tessa Poppenga
- Microbial Biology, Ruhr University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Georg Hölzl
- Institute of Molecular Physiology and Biotechnology of Plants (IMBIO), University of Bonn, Karlrobert-Kreiten-Straße 13, 53115 Bonn, Germany
| | - Meriyem Aktas
- Microbial Biology, Ruhr University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Franz Narberhaus
- Microbial Biology, Ruhr University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| |
Collapse
|
24
|
Zaretsky DV, Zaretskaia MV. Mini-review: Amyloid degradation toxicity hypothesis of Alzheimer's disease. Neurosci Lett 2021; 756:135959. [PMID: 34000347 DOI: 10.1016/j.neulet.2021.135959] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 04/28/2021] [Accepted: 05/10/2021] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia affecting millions of people. Neuronal death in AD is initiated by oligomeric amyloid-β (Aβ) peptides. The amyloid channel hypothesis readily explains the primary molecular damage but does not address major observations associated with AD such as autophagy failure and decreased metabolism. The amyloid degradation toxicity hypothesis provides the interpretation as a sequence of molecular events. Aβ enters a cell by endocytosis, and the endocytic vesicle is merged with a lysosome. Lysosomal peptidases degrade the peptide. Fragments form membrane channels in lysosomal membranes that have a significant negative charge due to the presence of acidic phospholipids. Amyloid channels can transfer various ions (including protons) and even relatively large compounds, which explains lysosomal permeabilization. The neutralization of lysosomal content inactivates degradation enzymes, results in an accumulation of undigested amyloid, and stalls autophagy. Inadequate quality control of mitochondria is associated with an increased production of reactive oxygen species and decreased energy production. Also, the passage of lysosomal proteases through rare extremely large channels results in cell death. Proposed hypothesis identifies biochemical pathways involved in the initiation and progression of cellular damage induced by beta-amyloid and provides new potential pharmacological targets to treat Alzheimer's disease.
Collapse
|
25
|
Gruenberg J. Life in the lumen: The multivesicular endosome. Traffic 2021; 21:76-93. [PMID: 31854087 PMCID: PMC7004041 DOI: 10.1111/tra.12715] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 12/12/2022]
Abstract
The late endosomes/endo‐lysosomes of vertebrates contain an atypical phospholipid, lysobisphosphatidic acid (LBPA) (also termed bis[monoacylglycero]phosphate [BMP]), which is not detected elsewhere in the cell. LBPA is abundant in the membrane system present in the lumen of this compartment, including intralumenal vesicles (ILVs). In this review, the current knowledge on LBPA and LBPA‐containing membranes will be summarized, and their role in the control of endosomal cholesterol will be outlined. Some speculations will also be made on how this system may be overwhelmed in the cholesterol storage disorder Niemann‐Pick C. Then, the roles of intralumenal membranes in endo‐lysosomal dynamics and functions will be discussed in broader terms. Likewise, the mechanisms that drive the biogenesis of intralumenal membranes, including ESCRTs, will also be discussed, as well as their diverse composition and fate, including degradation in lysosomes and secretion as exosomes. This review will also discuss how intralumenal membranes are hijacked by pathogenic agents during intoxication and infection, and what is the biochemical composition and function of the intra‐endosomal lumenal milieu. Finally, this review will allude to the size limitations imposed on intralumenal vesicle functions and speculate on the possible role of LBPA as calcium chelator in the acidic calcium stores of endo‐lysosomes.
Collapse
Affiliation(s)
- Jean Gruenberg
- Biochemistry Department, University of Geneva, Geneva, Switzerland
| |
Collapse
|
26
|
Decrease in Myelin-Associated Lipids Precedes Neuronal Loss and Glial Activation in the CNS of the Sandhoff Mouse as Determined by Metabolomics. Metabolites 2020; 11:metabo11010018. [PMID: 33396723 PMCID: PMC7823728 DOI: 10.3390/metabo11010018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 02/02/2023] Open
Abstract
Sandhoff disease (SD) is a lysosomal disease caused by mutations in the gene coding for the β subunit of β-hexosaminidase, leading to deficiency in the enzymes β-hexosaminidase (HEX) A and B. SD is characterised by an accumulation of gangliosides and related glycolipids, mainly in the central nervous system, and progressive neurodegeneration. The underlying cellular mechanisms leading to neurodegeneration and the contribution of inflammation in SD remain undefined. The aim of the present study was to measure global changes in metabolism over time that might reveal novel molecular pathways of disease. We used liquid chromatography-mass spectrometry and 1H Nuclear Magnetic Resonance spectroscopy to profile intact lipids and aqueous metabolites, respectively. We examined spinal cord and cerebrum from healthy and Hexb
-/- mice, a mouse model of SD, at ages one, two, three and four months. We report decreased concentrations in lipids typical of the myelin sheath, galactosylceramides and plasmalogen-phosphatidylethanolamines, suggesting that reduced synthesis of myelin lipids is an early event in the development of disease pathology. Reduction in neuronal density is progressive, as demonstrated by decreased concentrations of N-acetylaspartate and amino acid neurotransmitters. Finally, microglial activation, indicated by increased amounts of myo-inositol correlates closely with the late symptomatic phases of the disease.
Collapse
|
27
|
Cavender C, Mangini L, Van Vleet JL, Corado C, McCullagh E, Gray-Edwards HL, Martin DR, Crawford BE, Lawrence R. Natural history study of glycan accumulation in large animal models of GM2 gangliosidoses. PLoS One 2020; 15:e0243006. [PMID: 33259552 PMCID: PMC7707493 DOI: 10.1371/journal.pone.0243006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 11/12/2020] [Indexed: 11/19/2022] Open
Abstract
β-hexosaminidase is an enzyme responsible for the degradation of gangliosides, glycans, and other glycoconjugates containing β-linked hexosamines that enter the lysosome. GM2 gangliosidoses, such as Tay-Sachs and Sandhoff, are lysosomal storage disorders characterized by β-hexosaminidase deficiency and subsequent lysosomal accumulation of its substrate metabolites. These two diseases result in neurodegeneration and early mortality in children. A significant difference between these two disorders is the accumulation in Sandhoff disease of soluble oligosaccharide metabolites that derive from N- and O-linked glycans. In this paper we describe our results from a longitudinal biochemical study of a feline model of Sandhoff disease and an ovine model of Tay-Sachs disease to investigate the accumulation of GM2/GA2 gangliosides, a secondary biomarker for phospholipidosis, bis-(monoacylglycero)-phosphate, and soluble glycan metabolites in both tissue and fluid samples from both animal models. While both Sandhoff cats and Tay-Sachs sheep accumulated significant amounts of GM2 and GA2 gangliosides compared to age-matched unaffected controls, the Sandhoff cats having the more severe disease, accumulated larger amounts of gangliosides compared to Tay-Sachs sheep in their occipital lobes. For monitoring glycan metabolites, we developed a quantitative LC/MS assay for one of these free glycans in order to perform longitudinal analysis. The Sandhoff cats showed significant disease-related increases in this glycan in brain and in other matrices including urine which may provide a useful clinical tool for measuring disease severity and therapeutic efficacy. Finally, we observed age-dependent increasing accumulation for a number of analytes, especially in Sandhoff cats where glycosphingolipid, phospholipid, and glycan levels showed incremental increases at later time points without signs of peaking. This large animal natural history study for Sandhoff and Tay-Sachs is the first of its kind, providing insight into disease progression at the biochemical level. This report may help in the development and testing of new therapies to treat these disorders.
Collapse
Affiliation(s)
- Catlyn Cavender
- Research, BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Linley Mangini
- Research, BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Jeremy L. Van Vleet
- Research, BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Carley Corado
- Research, BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Emma McCullagh
- Research, BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | | | - Douglas R. Martin
- Scott-Ritchey Research Center and Department of Anatomy, Physiology, and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, United States of America
| | - Brett E. Crawford
- Research, BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Roger Lawrence
- Research, BioMarin Pharmaceutical Inc., Novato, CA, United States of America
- * E-mail:
| |
Collapse
|
28
|
Luquain-Costaz C, Rabia M, Hullin-Matsuda F, Delton I. Bis(monoacylglycero)phosphate, an important actor in the host endocytic machinery hijacked by SARS-CoV-2 and related viruses. Biochimie 2020; 179:247-256. [PMID: 33159981 PMCID: PMC7642752 DOI: 10.1016/j.biochi.2020.10.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022]
Abstract
Viruses, including the novel coronavirus SARS-CoV-2, redirect infected cell metabolism to their own purposes. After binding to its receptor angiotensin-converting enzyme 2 (ACE2) on the cell surface, the SARS-CoV-2 is taken up by receptor-mediated endocytosis ending in the acidic endolysosomal compartment. The virus hijacks the endosomal machinery leading to fusion of viral and endosomal membranes and release of the viral RNA into the cytosol. This mini-review specifically highlights the membrane lipid organization of the endosomal system focusing on the unconventional and late endosome/lysosome-specific phospholipid, bis(monoacylglycero)phosphate (BMP). BMP is enriched in alveolar macrophages of lung, one of the target tissue of SARS-CoV-2. This review details the BMP structure, its unsaturated fatty acid composition and fusogenic properties that are essential for the highly dynamic formation of the intraluminal vesicles inside the endosomes. Interestingly, BMP is necessary for infection and replication of enveloped RNA virus such as SARS-CoV-1 and Dengue virus. We also emphasize the role of BMP in lipid sorting and degradation, especially cholesterol transport in cooperation with Niemann Pick type C proteins (NPC 1 and 2) and with some oxysterol-binding protein (OSBP)-related proteins (ORPs) as well as in sphingolipid degradation. Interestingly, numerous virus infection required NPC1 as well as ORPs along the endocytic pathway. Furthermore, BMP content is increased during pathological endosomal lipid accumulation in various lysosomal storage disorders. This is particularly important knowing the high percentage of patients with metabolic disorders among the SARS-CoV-2 infected patients presenting severe forms of COVID-19.
Collapse
Affiliation(s)
- Céline Luquain-Costaz
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Maxence Rabia
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | | | - Isabelle Delton
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France.
| |
Collapse
|
29
|
Showalter MR, Berg AL, Nagourney A, Heil H, Carraway KL, Fiehn O. The Emerging and Diverse Roles of Bis(monoacylglycero) Phosphate Lipids in Cellular Physiology and Disease. Int J Mol Sci 2020; 21:ijms21218067. [PMID: 33137979 PMCID: PMC7663174 DOI: 10.3390/ijms21218067] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023] Open
Abstract
Although understudied relative to many phospholipids, accumulating evidence suggests that bis(monoacylglycero)phosphate (BMP) is an important class of regulatory lipid that plays key roles in lysosomal integrity and function. BMPs are rare in most mammalian tissues, comprising only a few percent of total cellular lipid content, but are elevated in cell types such as macrophages that rely heavily on lysosomal function. BMPs are markedly enriched in endosomal and lysosomal vesicles compared to other organelles and membranous structures, and their unique sn-1:sn-1′ stereoconfiguration may confer stability within the hydrolytic lysosomal environment. BMP-enriched vesicles serve in endosomal-lysosomal trafficking and function as docking structures for the activation of lysosomal hydrolytic enzymes, notably those involved in the catabolic breakdown of sphingolipids. BMP levels are dysregulated in lysosomal storage disorders, phospholipidosis, metabolic diseases, liver and kidney diseases and neurodegenerative disorders. However, whether BMP alteration is a mediator or simply a marker of pathological states is unclear. Likewise, although BMP acyl chain composition may be altered with disease states, the functional significance of specific BMP species remains to be resolved. Newly developed tools for untargeted lipidomic analysis, together with a deeper understanding of enzymes mediating BMP synthesis and degradation, will help shed further light on the functional significance of BMPs in cellular physiology and pathology.
Collapse
Affiliation(s)
- Megan R. Showalter
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA 95616, USA; (M.R.S.); (A.N.); (H.H.)
| | - Anastasia L. Berg
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (A.L.B.); (K.L.C.III)
| | - Alexander Nagourney
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA 95616, USA; (M.R.S.); (A.N.); (H.H.)
| | - Hailey Heil
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA 95616, USA; (M.R.S.); (A.N.); (H.H.)
| | - Kermit L. Carraway
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (A.L.B.); (K.L.C.III)
| | - Oliver Fiehn
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA 95616, USA; (M.R.S.); (A.N.); (H.H.)
- Correspondence:
| |
Collapse
|
30
|
Djambazova KV, Klein DR, Migas LG, Neumann EK, Rivera ES, Van de Plas R, Caprioli RM, Spraggins JM. Resolving the Complexity of Spatial Lipidomics Using MALDI TIMS Imaging Mass Spectrometry. Anal Chem 2020; 92:13290-13297. [PMID: 32808523 DOI: 10.1021/acs.analchem.0c02520] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Lipids are a structurally diverse class of molecules with important biological functions including cellular signaling and energy storage. Matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry (IMS) allows for direct mapping of biomolecules in tissues. Fully characterizing the structural diversity of lipids remains a challenge due to the presence of isobaric and isomeric species, which greatly complicates data interpretation when only m/z information is available. Integrating ion mobility separations aids in deconvoluting these complex mixtures and addressing the challenges of lipid IMS. Here, we demonstrate that a MALDI quadrupole time-of-flight (Q-TOF) mass spectrometer with trapped ion mobility spectrometry (TIMS) enables a >250% increase in the peak capacity during IMS experiments. MALDI TIMS-MS separation of lipid isomer standards, including sn backbone isomers, acyl chain isomers, and double-bond position and stereoisomers, is demonstrated. As a proof of concept, in situ separation and imaging of lipid isomers with distinct spatial distributions were performed using tissue sections from a whole-body mouse pup.
Collapse
Affiliation(s)
- Katerina V Djambazova
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Station B 351822, Nashville, Tennessee 37235, United States.,Mass Spectrometry Research Center, Vanderbilt University, 465 21st Avenue S #9160, Nashville, Tennessee 37235, United States
| | - Dustin R Klein
- Mass Spectrometry Research Center, Vanderbilt University, 465 21st Avenue S #9160, Nashville, Tennessee 37235, United States.,Department of Biochemistry, Vanderbilt University, 607 Light Hall, Nashville, Tennessee 37205, United States
| | - Lukasz G Migas
- Delft Center for Systems and Control, Delft University of Technology, 2628 CD Delft, The Netherlands
| | - Elizabeth K Neumann
- Mass Spectrometry Research Center, Vanderbilt University, 465 21st Avenue S #9160, Nashville, Tennessee 37235, United States.,Department of Biochemistry, Vanderbilt University, 607 Light Hall, Nashville, Tennessee 37205, United States
| | - Emilio S Rivera
- Mass Spectrometry Research Center, Vanderbilt University, 465 21st Avenue S #9160, Nashville, Tennessee 37235, United States.,Department of Biochemistry, Vanderbilt University, 607 Light Hall, Nashville, Tennessee 37205, United States
| | - Raf Van de Plas
- Delft Center for Systems and Control, Delft University of Technology, 2628 CD Delft, The Netherlands
| | - Richard M Caprioli
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Station B 351822, Nashville, Tennessee 37235, United States.,Mass Spectrometry Research Center, Vanderbilt University, 465 21st Avenue S #9160, Nashville, Tennessee 37235, United States.,Department of Biochemistry, Vanderbilt University, 607 Light Hall, Nashville, Tennessee 37205, United States.,Department of Pharmacology, Vanderbilt University, 2220 Pierce Avenue, Nashville, Tennessee 37232, United States.,Department of Medicine, Vanderbilt University, 465 21st Avenue S #9160, Nashville, Tennessee 37235, United States
| | - Jeffrey M Spraggins
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Station B 351822, Nashville, Tennessee 37235, United States.,Mass Spectrometry Research Center, Vanderbilt University, 465 21st Avenue S #9160, Nashville, Tennessee 37235, United States.,Department of Biochemistry, Vanderbilt University, 607 Light Hall, Nashville, Tennessee 37205, United States
| |
Collapse
|
31
|
Kloska A, Węsierska M, Malinowska M, Gabig-Cimińska M, Jakóbkiewicz-Banecka J. Lipophagy and Lipolysis Status in Lipid Storage and Lipid Metabolism Diseases. Int J Mol Sci 2020; 21:E6113. [PMID: 32854299 PMCID: PMC7504288 DOI: 10.3390/ijms21176113] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/12/2020] [Accepted: 08/21/2020] [Indexed: 12/15/2022] Open
Abstract
This review discusses how lipophagy and cytosolic lipolysis degrade cellular lipids, as well as how these pathway ys communicate, how they affect lipid metabolism and energy homeostasis in cells and how their dysfunction affects the pathogenesis of lipid storage and lipid metabolism diseases. Answers to these questions will likely uncover novel strategies for the treatment of aforementioned human diseases, but, above all, will avoid destructive effects of high concentrations of lipids-referred to as lipotoxicity-resulting in cellular dysfunction and cell death.
Collapse
Affiliation(s)
- Anna Kloska
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (A.K.); (M.W.); (M.M.)
| | - Magdalena Węsierska
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (A.K.); (M.W.); (M.M.)
| | - Marcelina Malinowska
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (A.K.); (M.W.); (M.M.)
| | - Magdalena Gabig-Cimińska
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (A.K.); (M.W.); (M.M.)
- Laboratory of Molecular Biology, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Kładki 24, 80-822 Gdańsk, Poland
| | - Joanna Jakóbkiewicz-Banecka
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (A.K.); (M.W.); (M.M.)
| |
Collapse
|
32
|
Rabia M, Leuzy V, Soulage C, Durand A, Fourmaux B, Errazuriz-Cerda E, Köffel R, Draeger A, Colosetti P, Jalabert A, Di Filippo M, Villard-Garon A, Bergerot C, Luquain-Costaz C, Moulin P, Rome S, Delton I, Hullin-Matsuda F. Bis(monoacylglycero)phosphate, a new lipid signature of endosome-derived extracellular vesicles. Biochimie 2020; 178:26-38. [PMID: 32659447 DOI: 10.1016/j.biochi.2020.07.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/29/2020] [Accepted: 07/08/2020] [Indexed: 01/05/2023]
Abstract
Bis(monoacylglycero)phosphate (BMP), also known as lysobisphosphatidic acid (LBPA), is a phospholipid specifically enriched in the late endosome-lysosome compartment playing a crucial role for the fate of endocytosed components. Due to its presence in extracellular fluids during diseases associated with endolysosomal dysfunction, it is considered as a possible biomarker of disorders such as genetic lysosomal storage diseases and cationic amphiphilic drug-induced phospholipidosis. However, there is no true validation of this biomarker in human studies, nor a clear identification of the carrier of this endolysosome-specific lipid in biofluids. The present study demonstrates that in absence of any sign of renal failure, BMP, especially all docosahexaenoyl containing species, are significantly increased in the urine of patients treated with the antiarrhythmic drug amiodarone. Such urinary BMP increase could reflect a generalized drug-induced perturbation of the endolysosome compartment as observed in vitro with amiodarone-treated human macrophages. Noteworthy, BMP was associated with extracellular vesicles (EVs) isolated from human urines and extracellular medium of human embryonic kidney HEK293 cells and co-localizing with classical EV protein markers CD63 and ALIX. In the context of drug-induced endolysosomal dysfunction, increased BMP-rich EV release could be useful to remove excess of undigested material. This first human pilot study not only reveals BMP as a urinary biomarker of amiodarone-induced endolysosomal dysfunction, but also highlights its utility to prove the endosomal origin of EVs, also named as exosomes. This peculiar lipid already known as a canonical late endosome-lysosome marker, may be thus considered as a new lipid marker of urinary exosomes.
Collapse
Affiliation(s)
- Maxence Rabia
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Valentin Leuzy
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Christophe Soulage
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Annie Durand
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Baptiste Fourmaux
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France; Functional Lipidomics Platform, CarMeN Laboratory / IMBL-INSA Lyon, 69621, Villeurbanne Cedex, France
| | | | - René Köffel
- Department of Cell Biology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Annette Draeger
- Department of Cell Biology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Pascal Colosetti
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Audrey Jalabert
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Mathilde Di Filippo
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France; Department of Biochemistry and Molecular Biology, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, Bron, France
| | - Audrey Villard-Garon
- Department of Endocrinology, Hôpital Cardiovasculaire Louis Pradel, Hospices Civils de Lyon, Lyon, Bron, France
| | - Cyrille Bergerot
- Department of Cardiology, Hôpital Cardiovasculaire Louis Pradel, Hospices Civils de Lyon, Lyon, Bron, France
| | - Céline Luquain-Costaz
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Philippe Moulin
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France; Department of Endocrinology, Hôpital Cardiovasculaire Louis Pradel, Hospices Civils de Lyon, Lyon, Bron, France
| | - Sophie Rome
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Isabelle Delton
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | | |
Collapse
|
33
|
Rivero-Ríos P, Romo-Lozano M, Fasiczka R, Naaldijk Y, Hilfiker S. LRRK2-Related Parkinson's Disease Due to Altered Endolysosomal Biology With Variable Lewy Body Pathology: A Hypothesis. Front Neurosci 2020; 14:556. [PMID: 32581693 PMCID: PMC7287096 DOI: 10.3389/fnins.2020.00556] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/06/2020] [Indexed: 12/14/2022] Open
Abstract
Mutations in the gene encoding for leucine-rich repeat kinase 2 (LRRK2) are associated with both familial and sporadic Parkinson's disease (PD). LRRK2 encodes a large protein comprised of a GTPase and a kinase domain. All pathogenic variants converge on enhancing LRRK2 kinase substrate phosphorylation, and distinct LRRK2 kinase inhibitors are currently in various stages of clinical trials. Although the precise pathophysiological functions of LRRK2 remain largely unknown, PD-associated mutants have been shown to alter various intracellular vesicular trafficking pathways, especially those related to endolysosomal protein degradation events. In addition, biochemical studies have identified a subset of Rab proteins, small GTPases required for all vesicular trafficking steps, as substrate proteins for the LRRK2 kinase activity in vitro and in vivo. Therefore, it is crucial to evaluate the impact of such phosphorylation on neurodegenerative mechanisms underlying LRRK2-related PD, especially with respect to deregulated Rab-mediated endolysosomal membrane trafficking and protein degradation events. Surprisingly, a significant proportion of PD patients due to LRRK2 mutations display neuronal cell loss in the substantia nigra pars compacta in the absence of any apparent α-synuclein-containing Lewy body neuropathology. These findings suggest that endolysosomal alterations mediated by pathogenic LRRK2 per se are not sufficient to cause α-synuclein aggregation. Here, we will review current knowledge about the link between pathogenic LRRK2, Rab protein phosphorylation and endolysosomal trafficking alterations, and we will propose a testable working model whereby LRRK2-related PD may present with variable LB pathology.
Collapse
Affiliation(s)
- Pilar Rivero-Ríos
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain.,Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
| | - María Romo-Lozano
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Rachel Fasiczka
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Yahaira Naaldijk
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Sabine Hilfiker
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
34
|
Hu C, Duan Q, Han X. Strategies to Improve/Eliminate the Limitations in Shotgun Lipidomics. Proteomics 2020; 20:e1900070. [PMID: 31291508 PMCID: PMC7394605 DOI: 10.1002/pmic.201900070] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/15/2019] [Indexed: 11/05/2022]
Abstract
Direct infusion-based shotgun lipidomics is one of the most powerful and useful tools in comprehensive analysis of lipid species from lipid extracts of various biological samples with high accuracy/precision. However, despite many advantages, the classical shotgun lipidomics suffers some general dogmas of limitations, such as ion suppression, ambiguous identification of isobaric/isomeric lipid species, and ion source-generated artifacts, restraining the applications in analysis of low-abundance lipid species, particularly those less ionizable or isomers that yield almost identical fragmentation patterns. This article reviews the strategies (such as modifier addition, prefractionation, chemical derivatization, charge feature utilization) that have been employed to improve/eliminate these limitations in modern shotgun lipidomics approaches (e.g., high mass resolution mass spectrometry-based and multidimensional mass spectrometry-based shotgun lipidomics). Therefore, with the enhancement of these strategies for shotgun lipidomics, comprehensive analysis of lipid species including isomeric/isobaric species is achieved in a more accurate and effective manner, greatly substantiating the aberrant lipid metabolism, signaling trafficking, and homeostasis under pathological conditions.
Collapse
Affiliation(s)
- Changfeng Hu
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, 548 Bingwen Road, Hangzhou, Zhejiang 310053, China
| | - Qiao Duan
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, 548 Bingwen Road, Hangzhou, Zhejiang 310053, China
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229 USA
- Department of Medicine – Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229 USA
| |
Collapse
|
35
|
Lawrence R, Van Vleet JL, Mangini L, Harris A, Martin N, Clark W, Chandriani S, LeBowitz JH, Giugliani R, d'Azzo A, Yogalingam G, Crawford BE. Characterization of glycan substrates accumulating in GM1 Gangliosidosis. Mol Genet Metab Rep 2019; 21:100524. [PMID: 31720227 PMCID: PMC6838976 DOI: 10.1016/j.ymgmr.2019.100524] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/10/2019] [Accepted: 09/13/2019] [Indexed: 10/28/2022] Open
Abstract
Introduction GM1 gangliosidosis is a rare autosomal recessive genetic disorder caused by the disruption of the GLB1 gene that encodes β-galactosidase, a lysosomal hydrolase that removes β-linked galactose from the non-reducing end of glycans. Deficiency of this catabolic enzyme leads to the lysosomal accumulation of GM1 and its asialo derivative GA1 in β-galactosidase deficient patients and animal models. In addition to GM1 and GA1, there are other glycoconjugates that contain β-linked galactose whose metabolites are substrates for β-galactosidase. For example, a number of N-linked glycan structures that have galactose at their non-reducing end have been shown to accumulate in GM1 gangliosidosis patient tissues and biological fluids. Objective In this study, we attempt to fully characterize the broad array of GLB1 substrates that require GLB1 for their lysosomal turnover. Results Using tandem mass spectrometry and glycan reductive isotope labeling with data-dependent mass spectrometry, we have confirmed the accumulation of glycolipids (GM1 and GA1) and N-linked glycans with terminal beta-linked galactose. We have also discovered a novel set of core 1 and 2 O-linked glycan metabolites, many of which are part of structurally-related isobaric series that accumulate in disease. In the brain of GLB1 null mice, the levels of these glycan metabolites increased along with those of both GM1 and GA1 as a function of age. In addition to brain tissue, we found elevated levels of both N-linked and O-linked glycan metabolites in a number of peripheral tissues and in urine. Both brain and urine samples from human GM1 gangliosidosis patients exhibited large increases in steady state levels for the same glycan metabolites, demonstrating their correlation with this disease in humans as well. Conclusions Our studies illustrate that GLB1 deficiency is not purely a ganglioside accumulation disorder, but instead a broad oligosaccharidosis that include representatives of many β-linked galactose containing glycans and glycoconjugates including glycolipids, N-linked glycans, and various O-linked glycans. Accounting for all β-galactosidase substrates that accumulate when this enzyme is deficient increases our understanding of this severe disorder by identifying metabolites that may drive certain aspects of the disease and may also serve as informative disease biomarkers to fully evaluate the efficacy of future therapies.
Collapse
Key Words
- A2G2, Oxford glycan naming designation for NA2 glycan
- BMP, Bis(monoacylglycero) phosphate
- Beta-galactosidase
- Disease biomarkers
- GLB1
- GLB1, β-galactosidase
- GM1 gangliosidosis
- GRIL-LC/MS, glycan reductive isotope labeling liquid chromatography mass spectrometry
- Gal, galactose
- GlcNAc, N-acetylglucosamine
- Glycan metabolites
- Glycoanalysis
- Hex, hexose
- HexNAc, N-acetylhexosamine
- KS, keratan sulfate
- MPS, mucopolysaccharidosis
- Man, mannose
- NRE, non-reducing end
- TIC, total ion current
- XIC, extracted ion current
- dp, degree of polymerization
- m/z, mass over charge
Collapse
Affiliation(s)
- Roger Lawrence
- Research, BioMarin Pharmaceutical Inc., Novato, CA 94949, USA
| | | | - Linley Mangini
- Research, BioMarin Pharmaceutical Inc., Novato, CA 94949, USA
| | - Adam Harris
- Research, BioMarin Pharmaceutical Inc., Novato, CA 94949, USA
| | - Nathan Martin
- Research, BioMarin Pharmaceutical Inc., Novato, CA 94949, USA
| | - Wyatt Clark
- Research, BioMarin Pharmaceutical Inc., Novato, CA 94949, USA
| | | | | | - Roberto Giugliani
- Medical Genetics Service, HCPA, Department of Genetics, UFRGS, and INAGEMP, Porto Alegre, Brazil
| | - Alessandra d'Azzo
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | | |
Collapse
|
36
|
Hu C, Wang C, He L, Han X. Novel strategies for enhancing shotgun lipidomics for comprehensive analysis of cellular lipidomes. Trends Analyt Chem 2019; 120:115330. [PMID: 32647401 PMCID: PMC7344273 DOI: 10.1016/j.trac.2018.11.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Shotgun lipidomics is one of the most powerful tools in analysis of cellular lipidomes in lipidomics, which directly analyzes lipids from lipid extracts of diverse biological samples with high accuracy/precision. However, despite its great advances in high throughput analysis of cellular lipidomes, low coverage of poorly ionized lipids, especially those species in very low abundance, and some types of isomers within complex lipid extracts by shotgun lipidomics remains a huge challenge. In the past few years, many strategies have been developed to enhance shotgun lipidomics for comprehensive analysis of lipid species. Chemical derivatization represents one of the most attractive and effective strategies, already receiving considerable attention. This review focuses on novel advanced derivatization strategies for enhancing shotgun lipidomics. It is anticipated that with the development of enhanced strategies, shotgun lipidomics can make greater contributions to biological and biomedical research.
Collapse
Affiliation(s)
- Changfeng Hu
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, 548 Bingwen Road, Hangzhou, Zhejiang 310053, China
| | - Chunyan Wang
- Barshop Institute for Longevity and Aging Research, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
| | - Lijiao He
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, 548 Bingwen Road, Hangzhou, Zhejiang 310053, China
| | - Xianlin Han
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, 548 Bingwen Road, Hangzhou, Zhejiang 310053, China
- Barshop Institute for Longevity and Aging Research, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
- Department of Medicine – Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
| |
Collapse
|
37
|
Naoe S, Tsugawa H, Takahashi M, Ikeda K, Arita M. Characterization of Lipid Profiles after Dietary Intake of Polyunsaturated Fatty Acids Using Integrated Untargeted and Targeted Lipidomics. Metabolites 2019; 9:E241. [PMID: 31640217 PMCID: PMC6836067 DOI: 10.3390/metabo9100241] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/17/2019] [Accepted: 10/17/2019] [Indexed: 12/25/2022] Open
Abstract
Illuminating the comprehensive lipid profiles after dietary supplementation of polyunsaturated fatty acids (PUFAs) is crucial to revealing the tissue distribution of PUFAs in living organisms, as well as to providing novel insights into lipid metabolism. Here, we performed lipidomic analyses on mouse plasma and nine tissues, including the liver, kidney, brain, white adipose, heart, lung, small intestine, skeletal muscle, and spleen, with the dietary intake conditions of arachidonic acid (ARA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA) as the ethyl ester form. We incorporated targeted and untargeted approaches for profiling oxylipins and complex lipids such as glycerol (phospho) lipids, sphingolipids, and sterols, respectively, which led to the characterization of 1026 lipid molecules from the mouse tissues. The lipidomic analysis indicated that the intake of PUFAs strongly impacted the lipid profiles of metabolic organs such as the liver and kidney, while causing less impact on the brain. Moreover, we revealed a unique lipid modulation in most tissues, where phospholipids containing linoleic acid were significantly decreased in mice on the ARA-supplemented diet, and bis(monoacylglycero)phosphate (BMP) selectively incorporated DHA over ARA and EPA. We comprehensively studied the lipid profiles after dietary intake of PUFAs, which gives insight into lipid metabolism and nutrition research on PUFA supplementation.
Collapse
Affiliation(s)
- Satoko Naoe
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan.
| | - Hiroshi Tsugawa
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan.
- Metabolome informatics research team, RIKEN Center for Sustainable Resource Science, Yokohama 230-0045, Japan.
| | - Mikiko Takahashi
- Metabolome informatics research team, RIKEN Center for Sustainable Resource Science, Yokohama 230-0045, Japan.
| | - Kazutaka Ikeda
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan.
- Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi, Yokohama 230-0045, Japan.
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan.
- Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi, Yokohama 230-0045, Japan.
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo 105-8512, Japan.
| |
Collapse
|
38
|
McCauliff LA, Langan A, Li R, Ilnytska O, Bose D, Waghalter M, Lai K, Kahn PC, Storch J. Intracellular cholesterol trafficking is dependent upon NPC2 interaction with lysobisphosphatidic acid. eLife 2019; 8:50832. [PMID: 31580258 PMCID: PMC6855803 DOI: 10.7554/elife.50832] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 10/02/2019] [Indexed: 12/12/2022] Open
Abstract
Unesterified cholesterol accumulation in the late endosomal/lysosomal (LE/LY) compartment is the cellular hallmark of Niemann-Pick C (NPC) disease, caused by defects in the genes encoding NPC1 or NPC2. We previously reported the dramatic stimulation of NPC2 cholesterol transport rates to and from model membranes by the LE/LY phospholipid lysobisphosphatidic acid (LBPA). It had been previously shown that enrichment of NPC1-deficient cells with LBPA results in cholesterol clearance. Here we demonstrate that LBPA enrichment in human NPC2-deficient cells, either directly or via its biosynthetic precursor phosphtidylglycerol (PG), is entirely ineffective, indicating an obligate functional interaction between NPC2 and LBPA in cholesterol trafficking. We further demonstrate that NPC2 interacts directly with LBPA and identify the NPC2 hydrophobic knob domain as the site of interaction. Together these studies reveal a heretofore unknown step of intracellular cholesterol trafficking which is critically dependent upon the interaction of LBPA with functional NPC2 protein. Cholesterol is a type of fat that is essential for many processes in the body, such as repairing damaged cells and producing certain hormones. Normally, cholesterol enters cells from the bloodstream and is then moved to the parts of the cell that need it via a process known as ‘trafficking’. When cholesterol trafficking goes wrong, abnormally large amounts of cholesterol and other fats accumulate within the cell. Over time, these fatty deposits become toxic to cells and eventually damage the affected tissues. Niemann-Pick type C disease (NPC) is a severe genetic disorder affecting cholesterol trafficking. It is characterized by cholesterol build-up in multiple tissues, including the brain, which ultimately causes degeneration and death of nerve cells. Two proteins, NPC1 and NPC2, are involved in NPC disease. Both proteins normally help move cholesterol out of important trafficking compartments (known as the endosomal and lysosomal compartments) to other areas of the cell where it is needed. Patients with the disease can have mutations in either the gene for NPC1 or the gene for NPC2. This means that cells from NPC1 patients do not make enough functional NPC1 protein (but contain working NPC2), and vice versa. Previous studies had shown that giving cells with NPC1 mutations large amounts of the small molecule lysobisphosphatidic acid (LBPA for short) could compensate for the loss of NPC1, and stop the toxic build-up of cholesterol. McCauliff, Langan, Li et al. therefore wanted to explore exactly how LBPA was doing this. They had shown that LBPA dramatically increased the ability of purified NPC2 protein to transport cholesterol, and wondered if the effect of LBPA in the cells without NPC1 depended on NPC2. They predicted that boosting LBPA levels would not work in cells lacking NPC2. Biochemical experiments using purified protein showed that LBPA and NPC2 did indeed interact directly with each other. Systematically changing different building blocks of NPC2 revealed that a single region of the protein is sensitive to LBPA, and when this region was altered, LBPA could no longer interact with NPC2. Since LBPA is naturally produced by cells, they then stimulated cells grown in the laboratory to generate more LBPA using its precursor phosphatidylglycerol. They used cells from patients with mutations in either NPC1 or NPC2 and demonstrated that LBPA’s ability to reverse the accumulation of cholesterol was dependent on its interaction with NPC2. Thus, increasing LBPA levels in cells from patients with NPC1 mutations was beneficial, but had no effect on cells from patients with NPC2 mutations. These results shed new light not only on how cells transport cholesterol, but also on potential methods to combat disorders of cellular cholesterol trafficking. In the future, LBPA could be developed as a genetically tailored, patient-specific therapy for diseases like NPC.
Collapse
Affiliation(s)
- Leslie A McCauliff
- Department of Nutritional Sciences, Rutgers University, New Brunswick, United States.,Rutgers Center for Lipid Research, Rutgers University, New Brunswick, United States
| | - Annette Langan
- Department of Nutritional Sciences, Rutgers University, New Brunswick, United States.,Rutgers Center for Lipid Research, Rutgers University, New Brunswick, United States
| | - Ran Li
- Department of Nutritional Sciences, Rutgers University, New Brunswick, United States.,Rutgers Center for Lipid Research, Rutgers University, New Brunswick, United States
| | - Olga Ilnytska
- Department of Nutritional Sciences, Rutgers University, New Brunswick, United States.,Rutgers Center for Lipid Research, Rutgers University, New Brunswick, United States
| | - Debosreeta Bose
- Department of Nutritional Sciences, Rutgers University, New Brunswick, United States.,Rutgers Center for Lipid Research, Rutgers University, New Brunswick, United States
| | - Miriam Waghalter
- Department of Nutritional Sciences, Rutgers University, New Brunswick, United States
| | - Kimberly Lai
- Department of Nutritional Sciences, Rutgers University, New Brunswick, United States
| | - Peter C Kahn
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, United States
| | - Judith Storch
- Department of Nutritional Sciences, Rutgers University, New Brunswick, United States.,Rutgers Center for Lipid Research, Rutgers University, New Brunswick, United States
| |
Collapse
|
39
|
Pergande MR, Serna‐Perez F, Mohsin SB, Hanek J, Cologna SM. Lipidomic Analysis Reveals Altered Fatty Acid Metabolism in the Liver of the Symptomatic Niemann–Pick, Type C1 Mouse Model. Proteomics 2019; 19:e1800285. [DOI: 10.1002/pmic.201800285] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/22/2019] [Indexed: 12/23/2022]
Affiliation(s)
- Melissa R. Pergande
- Department of ChemistryUniversity of Illinois at Chicago Chicago IL 60607 USA
| | - Fidel Serna‐Perez
- Department of ChemistryUniversity of Illinois at Chicago Chicago IL 60607 USA
| | | | - Jonathon Hanek
- Department of ChemistryUniversity of Illinois at Chicago Chicago IL 60607 USA
| | - Stephanie M. Cologna
- Department of ChemistryUniversity of Illinois at Chicago Chicago IL 60607 USA
- Department of ChemistryLaboratory for Integrative NeuroscienceUniversity of Illinois at Chicago Chicago IL 60607 USA
| |
Collapse
|
40
|
Wang X, Schmitt MV, Xu L, Jiao Y, Guo L, Lienau P, Reichel A, Liu X. Quantitative molecular tissue atlas of Bis(monoacylglycero)phosphate and phosphatidylglycerol membrane lipids in rodent organs generated by methylation assisted high resolution mass spectrometry. Anal Chim Acta 2019; 1084:60-70. [PMID: 31519235 DOI: 10.1016/j.aca.2019.07.060] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 07/28/2019] [Indexed: 11/25/2022]
Abstract
Bis(monoacylglycero)phosphate (BMP) and phosphatidylglycerol (PG) are structural isomeric phospholipids with very different properties and biological functions. Due to their isomeric nature, it has thus far been challenging to simultaneously quantify BMP and PG lipids in tissue samples by mass spectrometry. Therefore, we have developed a sensitive LC-MS/MS based approach with prior methylation derivatization that is able to handle large batches of samples. Using this high throughput platform, a simulated MS/MS database was established for confident lipid assignment. In this work, we have simultaneously identified and quantified BMP and PG lipid molecules in different body tissues of rats and mice. We report for the first time a quantitative molecular atlas of BMP and PG lipids for 14 different tissues and organs in Wistar rats, NMRI and CD1 mice. Organ- and species-specificity was analyzed and compared for both lipid molecule classes. A total of 34 BMP and 10 PG molecules were quantified, with PG concentrations being generally much higher across tissues than BMP, but BMP lipids showing a much higher molecular diversity between animal organs. The large diversity of the BMP lipids with regard to their abundance and molecular composition suggests distinct biological function(s) of the individual BMP molecules in different tissues and organs of body. Particularly high tissue levels of BMP were seen in spleen, lung, liver, kidney and small intestines, i.e. tissues that are known for their high abundance and/or activity level of lysosomes late and endosomes. Elevated BMP levels in brain tissue of APP/PSEN transgenic compared to age matched wild-type mice were also observed using this platform. This analytical methodology presented a high throughput LC-based approach incorporating simulated MS/MS database to identify and quantify BMP lipids as well as PG molecules.
Collapse
Affiliation(s)
- Xueying Wang
- School of Life Sciences, Tsinghua University, China; National Protein Science Facility, Tsinghua University, Beijing, China
| | | | - Lina Xu
- School of Life Sciences, Tsinghua University, China; National Protein Science Facility, Tsinghua University, Beijing, China
| | - Yupei Jiao
- School of Life Sciences, Tsinghua University, China; National Protein Science Facility, Tsinghua University, Beijing, China
| | - Lvjun Guo
- School of Life Sciences, Tsinghua University, China; National Protein Science Facility, Tsinghua University, Beijing, China
| | - Philip Lienau
- Research Pharmacokinetics, Pharma R&D, Bayer AG, Berlin, Germany
| | - Andreas Reichel
- Research Pharmacokinetics, Pharma R&D, Bayer AG, Berlin, Germany.
| | - Xiaohui Liu
- School of Life Sciences, Tsinghua University, China; National Protein Science Facility, Tsinghua University, Beijing, China.
| |
Collapse
|
41
|
Seyfried TN, Choi H, Chevalier A, Hogan D, Akgoc Z, Schneider JS. Sex-Related Abnormalities in Substantia Nigra Lipids in Parkinson's Disease. ASN Neuro 2019; 10:1759091418781889. [PMID: 29932343 PMCID: PMC6024349 DOI: 10.1177/1759091418781889] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative movement disorder involving the selective loss of dopamine-producing neurons in the substantia nigra (SN). Differences in disease presentation, prevalence, and age of onset have been reported between males and females with PD. The content and composition of the major glycosphingolipids, phospholipids, and cholesterol were evaluated in the SN from 12 PD subjects and in 18 age-matched, neurologically normal controls. Total SN ganglioside sialic acid content and water content (%) were significantly lower in the male PD subjects than in the male controls. The content of all major gangliosides were reduced in the male PD subjects to some degree, but the neuronal-enriched gangliosides, GD1a and GT1b, were most significantly reduced. The distribution of phosphatidylethanolamine, phosphatidylcholine, and phosphatidylinositol was also significantly lower in the male PD subjects than in the male controls. However, the distribution of myelin-enriched cerebrosides and sulfatides was significantly higher in the male PD subjects than in the male controls suggesting myelin sparing in the male PD subjects. No elevation was detected for astrocytosis-linked GD3. These neurochemical changes provide evidence of selective neuronal loss in SN of the males with PD without robust astrocytosis. In contrast to the SN lipid abnormalities found in the male PD subjects, no significant abnormalities were found in the female PD subjects for SN water content or for any major SN lipids. These data indicate sex-related differences in SN lipid abnormalities in PD.
Collapse
Affiliation(s)
- T N Seyfried
- 1 Department of Biology, Boston College, Chestnut Hill, MA, USA
| | - H Choi
- 1 Department of Biology, Boston College, Chestnut Hill, MA, USA
| | - A Chevalier
- 1 Department of Biology, Boston College, Chestnut Hill, MA, USA
| | - D Hogan
- 1 Department of Biology, Boston College, Chestnut Hill, MA, USA
| | - Z Akgoc
- 1 Department of Biology, Boston College, Chestnut Hill, MA, USA
| | - J S Schneider
- 2 Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
42
|
Grabner GF, Fawzy N, Pribasnig MA, Trieb M, Taschler U, Holzer M, Schweiger M, Wolinski H, Kolb D, Horvath A, Breinbauer R, Rülicke T, Rabl R, Lass A, Stadlbauer V, Hutter-Paier B, Stauber RE, Fickert P, Zechner R, Marsche G, Eichmann TO, Zimmermann R. Metabolic disease and ABHD6 alter the circulating bis(monoacylglycerol)phosphate profile in mice and humans. J Lipid Res 2019; 60:1020-1031. [PMID: 30894461 PMCID: PMC6495172 DOI: 10.1194/jlr.m093351] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/18/2019] [Indexed: 02/03/2023] Open
Abstract
Bis(monoacylglycerol)phosphate (BMP) is a phospholipid that is crucial for lipid degradation and sorting in acidic organelles. Genetic and drug-induced lysosomal storage disorders (LSDs) are associated with increased BMP concentrations in tissues and in the circulation. Data on BMP in disorders other than LSDs, however, are scarce, and key enzymes regulating BMP metabolism remain elusive. Here, we demonstrate that common metabolic disorders and the intracellular BMP hydrolase α/β-hydrolase domain-containing 6 (ABHD6) affect BMP metabolism in mice and humans. In mice, dietary lipid overload strongly affects BMP concentration and FA composition in the liver and plasma, similar to what has been observed in LSDs. Notably, distinct changes in the BMP FA profile enable a clear distinction between lipid overload and drug-induced LSDs. Global deletion of ABHD6 increases circulating BMP concentrations but does not cause LSDs. In humans, nonalcoholic fatty liver disease and liver cirrhosis affect the serum BMP FA composition and concentration. Furthermore, we identified a patient with a loss-of-function mutation in the ABHD6 gene, leading to an altered circulating BMP profile. In conclusion, our results suggest that common metabolic diseases and ABHD6 affect BMP metabolism in mice and humans.
Collapse
Affiliation(s)
- Gernot F Grabner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Nermeen Fawzy
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; BioTechMed-Graz Graz, Austria
| | - Maria A Pribasnig
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Markus Trieb
- Division of Pharmacology Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Ulrike Taschler
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Michael Holzer
- Division of Pharmacology Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Martina Schweiger
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Heimo Wolinski
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Dagmar Kolb
- Core Facility Ultrastructure Analysis Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Angela Horvath
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Rolf Breinbauer
- Institute of Organic Chemistry Graz University of Technology, Graz, Austria; BioTechMed-Graz Graz, Austria
| | - Thomas Rülicke
- Institute of Laboratory Animal Science University of Veterinary Medicine Vienna, Vienna, Austria
| | | | - Achim Lass
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; BioTechMed-Graz Graz, Austria
| | - Vanessa Stadlbauer
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | | | - Rudolf E Stauber
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Peter Fickert
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; BioTechMed-Graz Graz, Austria
| | - Gunther Marsche
- Division of Pharmacology Otto Loewi Research Center, Medical University of Graz, Graz, Austria; BioTechMed-Graz Graz, Austria
| | - Thomas O Eichmann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; Center for Explorative Lipidomics Graz, Austria; BioTechMed-Graz Graz, Austria.
| | - Robert Zimmermann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; BioTechMed-Graz Graz, Austria. robert.zimmermann@uni-graz
| |
Collapse
|
43
|
Fuller M, Futerman AH. The brain lipidome in neurodegenerative lysosomal storage disorders. Biochem Biophys Res Commun 2018. [PMID: 29524416 DOI: 10.1016/j.bbrc.2018.03.042] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Cholesterol, sphingolipids and glycerophospholipids are critical constituents of the brain, subserving neuronal membrane architecture and providing a platform for biochemical processes essential for proper neurodevelopment and function. When lysosomal defects arise in a lipid metabolic pathway, it is therefore easy to imagine that neurological decline will transpire, however for deficits in non-lipid pathways, this becomes harder to envisage. Here we suggest the working hypothesis that neurodegenerative lysosomal storage disorders might manifest as primary and/or secondary disorders of lipid metabolism. Evidence suggests that the mere process of lysosomal substrate accumulation, ubiquitous in all lysosomal storage disorders, impairs lysosome integrity resulting in secondary lipid accumulation. Impaired lysosomal degradation of a specific lipid defines a primary disorder of lipid metabolism and as these lysosomal storage disorders additionally show secondary lipid alterations, all primary disorders can also be considered secondary disorders of lipid metabolism. The outcome is a generalized cellular lipid dyshomeostasis and consequently, the physiological architecture of the lipid-enriched plasma membrane is perturbed, including the lipid composition of specialized membrane microdomains, often termed lipid rafts. Neurotoxicity results from the complex interplay of malfunctioning signaling and vesicular trafficking important for neuronal communication and synaptic plasticity-induced by the imbalance in physiological membrane lipid composition - together with compensatory mechanisms aimed at restoring lipid homeostasis.
Collapse
Affiliation(s)
- Maria Fuller
- Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, 72 King William Road, North Adelaide and School of Medicine, University of Adelaide, Adelaide, South Australia, 5005, Australia.
| | - Anthony H Futerman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
44
|
Anderson DMG, Ablonczy Z, Koutalos Y, Hanneken AM, Spraggins JM, Calcutt MW, Crouch RK, Caprioli RM, Schey KL. Bis(monoacylglycero)phosphate lipids in the retinal pigment epithelium implicate lysosomal/endosomal dysfunction in a model of Stargardt disease and human retinas. Sci Rep 2017; 7:17352. [PMID: 29229934 PMCID: PMC5725462 DOI: 10.1038/s41598-017-17402-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 11/22/2017] [Indexed: 01/10/2023] Open
Abstract
Stargardt disease is a juvenile onset retinal degeneration, associated with elevated levels of lipofuscin and its bis-retinoid components, such as N-retinylidene-N-retinylethanolamine (A2E). However, the pathogenesis of Stargardt is still poorly understood and targeted treatments are not available. Utilizing high spatial and high mass resolution matrix assisted laser desorption ionization (MALDI) imaging mass spectrometry (IMS), we determined alterations of lipid profiles specifically localized to the retinal pigment epithelium (RPE) in Abca4 -/- Stargardt model mice compared to their relevant background strain. Extensive analysis by LC-MS/MS in both positive and negative ion mode was required to accurately confirm the identity of one highly expressed lipid class, bis(monoacylgylercoro)phosphate (BMP) lipids, and to distinguish them from isobaric species. The same BMP lipids were also detected in the RPE of healthy human retina. BMP lipids have been previously associated with the endosomal/lysosomal storage diseases Niemann-Pick and neuronal ceroid lipofuscinosis and have been reported to regulate cholesterol levels in endosomes. These results suggest that perturbations in lipid metabolism associated with late endosomal/lysosomal dysfunction may play a role in the pathogenesis of Stargardt disease and is evidenced in human retinas.
Collapse
Affiliation(s)
- David M G Anderson
- Mass Spectrometry Research Center and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Zsolt Ablonczy
- Department of Ophthalmology, Storm Eye Institute, Medical University of South Carolina, Charleston, SC, USA
- Preclinical Department, Ora Inc, Andover, MA, USA
| | - Yiannis Koutalos
- Department of Ophthalmology, Storm Eye Institute, Medical University of South Carolina, Charleston, SC, USA
| | - Anne M Hanneken
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Jeffrey M Spraggins
- Mass Spectrometry Research Center and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Nashville, TN, USA
| | - M Wade Calcutt
- Mass Spectrometry Research Center and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Rosalie K Crouch
- Department of Ophthalmology, Storm Eye Institute, Medical University of South Carolina, Charleston, SC, USA
| | - Richard M Caprioli
- Mass Spectrometry Research Center and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Nashville, TN, USA
- Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN, USA
| | - Kevin L Schey
- Mass Spectrometry Research Center and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
45
|
Kitakaze K, Mizutani Y, Sugiyama E, Tasaki C, Tsuji D, Maita N, Hirokawa T, Asanuma D, Kamiya M, Sato K, Setou M, Urano Y, Togawa T, Otaka A, Sakuraba H, Itoh K. Protease-resistant modified human β-hexosaminidase B ameliorates symptoms in GM2 gangliosidosis model. J Clin Invest 2016; 126:1691-703. [PMID: 27018595 DOI: 10.1172/jci85300] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 02/09/2016] [Indexed: 01/24/2023] Open
Abstract
GM2 gangliosidoses, including Tay-Sachs and Sandhoff diseases, are neurodegenerative lysosomal storage diseases that are caused by deficiency of β-hexosaminidase A, which comprises an αβ heterodimer. There are no effective treatments for these diseases; however, various strategies aimed at restoring β-hexosaminidase A have been explored. Here, we produced a modified human hexosaminidase subunit β (HexB), which we have termed mod2B, composed of homodimeric β subunits that contain amino acid sequences from the α subunit that confer GM2 ganglioside-degrading activity and protease resistance. We also developed fluorescent probes that allow visualization of endocytosis of mod2B via mannose 6-phosphate receptors and delivery of mod2B to lysosomes in GM2 gangliosidosis models. In addition, we applied imaging mass spectrometry to monitor efficacy of this approach in Sandhoff disease model mice. Following i.c.v. administration, mod2B was widely distributed and reduced accumulation of GM2, asialo-GM2, and bis(monoacylglycero)phosphate in brain regions including the hypothalamus, hippocampus, and cerebellum. Moreover, mod2B administration markedly improved motor dysfunction and a prolonged lifespan in Sandhoff disease mice. Together, the results of our study indicate that mod2B has potential for intracerebrospinal fluid enzyme replacement therapy and should be further explored as a gene therapy for GM2 gangliosidoses.
Collapse
|
46
|
First-Generation Antipsychotic Haloperidol Alters the Functionality of the Late Endosomal/Lysosomal Compartment in Vitro. Int J Mol Sci 2016; 17:404. [PMID: 26999125 PMCID: PMC4813259 DOI: 10.3390/ijms17030404] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 03/07/2016] [Accepted: 03/14/2016] [Indexed: 01/18/2023] Open
Abstract
First- and second-generation antipsychotics (FGAs and SGAs, respectively), have the ability to inhibit cholesterol biosynthesis and also to interrupt the intracellular cholesterol trafficking, interfering with low-density lipoprotein (LDL)-derived cholesterol egress from late endosomes/lysosomes. In the present work, we examined the effects of FGA haloperidol on the functionality of late endosomes/lysosomes in vitro. In HepG2 hepatocarcinoma cells incubated in the presence of 1,1'-dioctadecyl-3,3,3,3'-tetramethylindocarbocyanineperchlorate (DiI)-LDL, treatment with haloperidol caused the enlargement of organelles positive for late endosome markers lysosome-associated membrane protein 2 (LAMP-2) and LBPA (lysobisphosphatidic acid), which also showed increased content of both free-cholesterol and DiI derived from LDL. This indicates the accumulation of LDL-lipids in the late endosomal/lysosomal compartment caused by haloperidol. In contrast, LDL traffic through early endosomes and the Golgi apparatus appeared to be unaffected by the antipsychotic as the distribution of both early endosome antigen 1 (EEA1) and coatomer subunit β (β-COP) were not perturbed. Notably, treatment with haloperidol significantly increased the lysosomal pH and decreased the activities of lysosomal protease and β-d-galactosidase in a dose-dependent manner. We conclude that the alkalinization of the lysosomes' internal milieu induced by haloperidol affects lysosomal functionality.
Collapse
|
47
|
Selective normalisation of regional brain bis(monoacylglycero)phosphate in the mucopolysaccharidosis 1 (Hurler) mouse. Exp Neurol 2016; 277:68-75. [DOI: 10.1016/j.expneurol.2015.12.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 12/17/2015] [Accepted: 12/18/2015] [Indexed: 11/20/2022]
|
48
|
Anheuser S, Breiden B, Schwarzmann G, Sandhoff K. Membrane lipids regulate ganglioside GM2 catabolism and GM2 activator protein activity. J Lipid Res 2015; 56:1747-61. [PMID: 26175473 PMCID: PMC4548779 DOI: 10.1194/jlr.m061036] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 07/14/2015] [Indexed: 01/19/2023] Open
Abstract
Ganglioside GM2 is the major lysosomal storage compound of Tay-Sachs disease. It also accumulates in Niemann-Pick disease types A and B with primary storage of SM and with cholesterol in type C. Reconstitution of GM2 catabolism with β-hexosaminidase A and GM2 activator protein (GM2AP) at uncharged liposomal surfaces carrying GM2 as substrate generated only a physiologically irrelevant catabolic rate, even at pH 4.2. However, incorporation of anionic phospholipids into the GM2 carrying liposomes stimulated GM2 hydrolysis more than 10-fold, while the incorporation of plasma membrane stabilizing lipids (SM and cholesterol) generated a strong inhibition of GM2 hydrolysis, even in the presence of anionic phospholipids. Mobilization of membrane lipids by GM2AP was also inhibited in the presence of cholesterol or SM, as revealed by surface plasmon resonance studies. These lipids also reduced the interliposomal transfer rate of 2-NBD-GM1 by GM2AP, as observed in assays using Förster resonance energy transfer. Our data raise major concerns about the usage of recombinant His-tagged GM2AP compared with untagged protein. The former binds more strongly to anionic GM2-carrying liposomal surfaces, increases GM2 hydrolysis, and accelerates intermembrane transfer of 2-NBD-GM1, but does not mobilize membrane lipids.
Collapse
Affiliation(s)
- Susi Anheuser
- LIMES Institute, Membrane Biology and Lipid Biochemistry Unit, Kekulé-Institut für Organische Chemie und Biochemie, Universität Bonn, D-53121 Bonn, Germany
| | - Bernadette Breiden
- LIMES Institute, Membrane Biology and Lipid Biochemistry Unit, Kekulé-Institut für Organische Chemie und Biochemie, Universität Bonn, D-53121 Bonn, Germany
| | - Günter Schwarzmann
- LIMES Institute, Membrane Biology and Lipid Biochemistry Unit, Kekulé-Institut für Organische Chemie und Biochemie, Universität Bonn, D-53121 Bonn, Germany
| | - Konrad Sandhoff
- LIMES Institute, Membrane Biology and Lipid Biochemistry Unit, Kekulé-Institut für Organische Chemie und Biochemie, Universität Bonn, D-53121 Bonn, Germany
| |
Collapse
|
49
|
Abstract
Bis(monoacylglycero)phosphate (BMP) is a structural isomer of phosphatidylglycerol (PtdGro) with an unusual sn-1:sn-1' fatty acyl configuration and is found almost exclusively in late endosomes/lysosomes. BMP comprises only about 1-2% of the total phospholipids in most mammalian cells, but accumulates in tissues of humans and animals with lysosomal storage disorders including the gangliosidoses. Total BMP content was significantly greater in cells of macrophage/microglial origin than in cells of macroglial origin. BMP composition was similar in tumorigenic/metastatic macrophages and non-tumorigenic macrophages/microglia. Finally, BMP fatty acid composition differed between cells grown in culture and obtained in vivo suggesting an influence from growth environment.
Collapse
Affiliation(s)
- Zeynep Akgoc
- Biology Department, Boston College, 140 Commonwealth Ave, MA, 02467, Chestnut Hill, USA,
| | | | | |
Collapse
|