1
|
Duffy HB, Byrnes C, Zhu H, Tuymetova G, Lee YT, Platt FM, Proia RL. Deletion of Gba in neurons, but not microglia, causes neurodegeneration in a Gaucher mouse model. JCI Insight 2024; 9:e179126. [PMID: 39312723 DOI: 10.1172/jci.insight.179126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 09/12/2024] [Indexed: 09/25/2024] Open
Abstract
Gaucher disease, the most prevalent lysosomal storage disease, is caused by homozygous mutations at the GBA gene, which is responsible for encoding the enzyme glucocerebrosidase. Neuronopathic Gaucher disease is associated with microgliosis, astrogliosis, and neurodegeneration. However, the role that microglia, astrocytes, and neurons play in the disease remains to be determined. In the current study, we developed inducible, cell-type-specific Gba-KO mice to better understand the individual impacts of Gba deficiencies on microglia and neurons. Gba was conditionally knocked out either exclusively in microglia or neurons or throughout the body. These mouse models were developed using a tamoxifen-inducible Cre system, with tamoxifen administration commencing at weaning. Microglia-specific Gba-KO mice showed no signs of disease. However, the neuron-specific Gba KO resulted in a shortened lifespan, severe weight loss, and ataxia. These mice also had significant neurodegeneration, microgliosis, and astrogliosis accompanied by the accumulation of glucosylceramide and glucosylsphingosine, recapitulating Gaucher disease-like symptoms. These surprising findings reveal that, unlike the neuron-specific Gba deficiency, microglia-specific Gba deficiency alone does not induce disease. The neuronal Gaucher disease mouse model, with a median survival of 16 weeks, may be useful for future studies of pathogenesis and the evaluation of therapies.
Collapse
Affiliation(s)
- Hannah Bd Duffy
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Colleen Byrnes
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Hongling Zhu
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Galina Tuymetova
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Y Terry Lee
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Frances M Platt
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Richard L Proia
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| |
Collapse
|
2
|
Becker AP, Biletch E, Kennelly JP, Julio AR, Villaneuva M, Nagari RT, Turner DW, Burton NR, Fukuta T, Cui L, Xiao X, Hong SG, Mack JJ, Tontonoz P, Backus KM. Lipid- and protein-directed photosensitizer proximity labeling captures the cholesterol interactome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.20.608660. [PMID: 39229057 PMCID: PMC11370482 DOI: 10.1101/2024.08.20.608660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The physical properties of cellular membranes, including fluidity and function, are influenced by protein and lipid interactions. In situ labeling chemistries, most notably proximity-labeling interactomics are well suited to characterize these dynamic and often fleeting interactions. Established methods require distinct chemistries for proteins and lipids, which limits the scope of such studies. Here we establish a singlet-oxygen-based photocatalytic proximity labeling platform (POCA) that reports intracellular interactomes for both proteins and lipids with tight spatiotemporal resolution using cell-penetrant photosensitizer reagents. Using both physiologically relevant lipoprotein-complexed probe delivery and genetic manipulation of cellular cholesterol handling machinery, cholesterol-directed POCA captured established and unprecedented cholesterol binding proteins, including protein complexes sensitive to intracellular cholesterol levels and proteins uniquely captured by lipoprotein uptake. Protein-directed POCA accurately mapped known intracellular membrane complexes, defined sterol-dependent changes to the non-vesicular cholesterol transport protein interactome, and captured state-dependent changes in the interactome of the cholesterol transport protein Aster-B. More broadly, we find that POCA is a versatile interactomics platform that is straightforward to implement, using the readily available HaloTag system, and fulfills unmet needs in intracellular singlet oxygen-based proximity labeling proteomics. Thus, we expect widespread utility for POCA across a range of interactome applications, spanning imaging to proteomics.
Collapse
Affiliation(s)
- Andrew P. Becker
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, California 90095, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, California 90095, USA
| | - Elijah Biletch
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, California 90095, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, California 90095, USA
| | - John Paul Kennelly
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, California 90095, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, Los Angeles, California 90095, USA
| | - Ashley R. Julio
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, California 90095, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, California 90095, USA
| | - Miranda Villaneuva
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, California 90095, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, California 90095, USA
- Molecular Biology Institute, UCLA, Los Angeles, California 90095, USA
| | - Rohith T. Nagari
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, California 90095, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, Los Angeles, California 90095, USA
- Molecular Biology Institute, UCLA, Los Angeles, California 90095, USA
| | - Daniel W. Turner
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, California 90095, USA
| | - Nikolas R. Burton
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, California 90095, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, California 90095, USA
| | - Tomoyuki Fukuta
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, California 90095, USA
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Liujuan Cui
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, California 90095, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, Los Angeles, California 90095, USA
| | - Xu Xiao
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, California 90095, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, Los Angeles, California 90095, USA
| | - Soon-Gook Hong
- Molecular Biology Institute, UCLA, Los Angeles, California 90095, USA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California 90095, USA
| | - Julia J. Mack
- Molecular Biology Institute, UCLA, Los Angeles, California 90095, USA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California 90095, USA
| | - Peter Tontonoz
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, California 90095, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, Los Angeles, California 90095, USA
| | - Keriann M. Backus
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, California 90095, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, California 90095, USA
- Molecular Biology Institute, UCLA, Los Angeles, California 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, California 90095, USA
- Jonsson Cancer Center, David Geffen School of Medicine, UCLA, Los Angeles, California 90095, USA
- UCLA-DOE Institute for Genomics and Proteomics, UCLA, Los Angeles, California 90095, USA
| |
Collapse
|
3
|
Yue C, Lu W, Fan S, Huang Z, Yang J, Dong H, Zhang X, Shang Y, Lai W, Li D, Dong T, Yuan A, Wu J, Kang L, Hu Y. Nanoparticles for inducing Gaucher disease-like damage in cancer cells. NATURE NANOTECHNOLOGY 2024; 19:1203-1215. [PMID: 38740934 DOI: 10.1038/s41565-024-01668-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 03/27/2024] [Indexed: 05/16/2024]
Abstract
Nutrient avidity is one of the most distinctive features of tumours. However, nutrient deprivation has yielded limited clinical benefits. In Gaucher disease, an inherited metabolic disorder, cells produce cholesteryl-glucoside which accumulates in lysosomes and causes cell damage. Here we develop a nanoparticle (AbCholB) to emulate natural-lipoprotein-carried cholesterol and initiate Gaucher disease-like damage in cancer cells. AbCholB is composed of a phenylboronic-acid-modified cholesterol (CholB) and albumin. Cancer cells uptake the nanoparticles into lysosomes, where CholB reacts with glucose and generates a cholesteryl-glucoside-like structure that resists degradation and aggregates into microscale crystals, causing Gaucher disease-like damage in a glucose-dependent manner. In addition, the nutrient-sensing function of mTOR is suppressed. It is observed that normal cells escape severe damage due to their inferior ability to compete for nutrients compared with cancer cells. This work provides a bioinspired strategy to selectively impede the metabolic action of cancer cells by taking advantage of their nutrient avidity.
Collapse
Affiliation(s)
- Chunyan Yue
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Wenjing Lu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Shuxin Fan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Zhusheng Huang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Jiaying Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Hong Dong
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Xiaojun Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Yuxin Shang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Wenjia Lai
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, China
| | - Dandan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Tiejun Dong
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Ahu Yuan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Lifeng Kang
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China.
- Institute of Drug R&D, School of Life Science, Nanjing University, Nanjing, China.
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China.
| |
Collapse
|
4
|
Borsato G, Carnio F, Lunardon S, Moletta M, Pavan G, Terrin F, Scarso A, Plotegher N, Fabris F. A β-Glucosyl Sterol Probe for in situ Fluorescent Labelling in Neuronal Cells to Investigate Neurodegenerative Diseases. Chemistry 2024; 30:e202400778. [PMID: 38770991 DOI: 10.1002/chem.202400778] [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: 02/26/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 05/22/2024]
Abstract
A β-glucosyl sterol probe bearing a terminal alkyne moiety for fluorescent tagging enables the investigation of the neuronal and intracellular localization of this class of compounds involved in neurodegenerative diseases. The compound showed localization in the neuronal cells, with marked differences in the uptake and metabolism leading to enhanced persistence with respect to the un-glycosylated sterol analogue. In addition, a different impact was observed towards lysosomes, with the simple sterol probe showing the enlargement of the lysosome structures, while the β-glucosyl sterol was less capable to alter the morphology of this specific organelle.
Collapse
Affiliation(s)
- Giuseppe Borsato
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari Venezia, via Torino 155, 30172, Mestre Venezia, Italy
| | - Francesco Carnio
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari Venezia, via Torino 155, 30172, Mestre Venezia, Italy
| | - Sara Lunardon
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari Venezia, via Torino 155, 30172, Mestre Venezia, Italy
| | - Mattia Moletta
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari Venezia, via Torino 155, 30172, Mestre Venezia, Italy
| | - Giulio Pavan
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari Venezia, via Torino 155, 30172, Mestre Venezia, Italy
| | - Francesca Terrin
- Dipartimento di Biologia, Università degli Studi di Padova, viale G. Colombo 3, 35131, Padova, Italy
| | - Alessandro Scarso
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari Venezia, via Torino 155, 30172, Mestre Venezia, Italy
| | - Nicoletta Plotegher
- Dipartimento di Biologia, Università degli Studi di Padova, viale G. Colombo 3, 35131, Padova, Italy
| | - Fabrizio Fabris
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari Venezia, via Torino 155, 30172, Mestre Venezia, Italy
| |
Collapse
|
5
|
Natraj P, Rajan P, Jeon YA, Kim SS, Lee YJ. Antiadipogenic Effect of Citrus Flavonoids: Evidence from RNA Sequencing Analysis and Activation of AMPK in 3T3-L1 Adipocytes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:17788-17800. [PMID: 37955544 DOI: 10.1021/acs.jafc.3c03559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Citrus fruits are rich in dietary flavonoids and have many health benefits, but their antiadipogenic mechanism of action and their impact on lipid metabolism remain unclear. In this study, we investigated the effect of citrus flavonoids, namely, hesperidin (HES), narirutin (NAR), nobiletin (NOB), sinensetin (SIN), and tangeretin (TAN), on preventing fat cell development by gene expression in 3T3-L1 adipocytes. Among the citrus flavonoids tested, HES and NAR significantly reduced fat storage and triglyceride levels and increased glucose uptake in 3T3-L1 adipocytes. Additionally, HES and NAR treatment increased the phosphorylation of AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC) while reducing the protein expression of 3-hydroxy-3-methylglutaryl CoA reductase (HMGCR). Furthermore, in silico docking revealed that flavonoids activate AMPK. RNA sequencing analysis demonstrated that citrus flavonoids normalized the expression of 40 genes, which were either upregulated by more than 2-fold or downregulated by less than 0.6-fold including Acadv1, Acly, Akr1d1, Awat1, Cyp27a1, Decr1, Dhrs4, Elovl3, Fasn, G6pc, Gba, Hmgcs1, Mogat2, Lrp5, Sptlc3, and Snca to levels comparable to the control group. Altogether, HES and NAR among five citrus flavonoids showed antiadipogenic effects by regulating the expression of specific lipid metabolism genes partially restored to control levels in 3T3-L1 cells.
Collapse
Affiliation(s)
- Premkumar Natraj
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Korea
- Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Korea
| | - Priyanka Rajan
- Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju 63243, Korea
| | - Yoon A Jeon
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Korea
- Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Korea
| | - Sang Suk Kim
- Citrus Research Institute, National Institute of Horticultural & Herbal Science, RDA, Jeju 63607, Korea
| | - Young Jae Lee
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Korea
- Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Korea
| |
Collapse
|
6
|
Muraleedharan A, Vanderperre B. The endo-lysosomal system in Parkinson's disease: expanding the horizon. J Mol Biol 2023:168140. [PMID: 37148997 DOI: 10.1016/j.jmb.2023.168140] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/08/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer's disease, and its prevalence is increasing with age. A wealth of genetic evidence indicates that the endo-lysosomal system is a major pathway driving PD pathogenesis with a growing number of genes encoding endo-lysosomal proteins identified as risk factors for PD, making it a promising target for therapeutic intervention. However, detailed knowledge and understanding of the molecular mechanisms linking these genes to the disease are available for only a handful of them (e.g. LRRK2, GBA1, VPS35). Taking on the challenge of studying poorly characterized genes and proteins can be daunting, due to the limited availability of tools and knowledge from previous literature. This review aims at providing a valuable source of molecular and cellular insights into the biology of lesser-studied PD-linked endo-lysosomal genes, to help and encourage researchers in filling the knowledge gap around these less popular genetic players. Specific endo-lysosomal pathways discussed range from endocytosis, sorting, and vesicular trafficking to the regulation of membrane lipids of these membrane-bound organelles and the specific enzymatic activities they contain. We also provide perspectives on future challenges that the community needs to tackle and propose approaches to move forward in our understanding of these poorly studied endo-lysosomal genes. This will help harness their potential in designing innovative and efficient treatments to ultimately re-establish neuronal homeostasis in PD but also other diseases involving endo-lysosomal dysfunction.
Collapse
Affiliation(s)
- Amitha Muraleedharan
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois and Biological Sciences Department, Université du Québec à Montréal
| | - Benoît Vanderperre
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois and Biological Sciences Department, Université du Québec à Montréal
| |
Collapse
|
7
|
The Consequences of GBA Deficiency in the Autophagy-Lysosome System in Parkinson's Disease Associated with GBA. Cells 2023; 12:cells12010191. [PMID: 36611984 PMCID: PMC9818455 DOI: 10.3390/cells12010191] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/27/2022] [Accepted: 12/31/2022] [Indexed: 01/05/2023] Open
Abstract
GBA gene variants were the first genetic risk factor for Parkinson's disease. GBA encodes the lysosomal enzyme glucocerebrosidase (GBA), which is involved in sphingolipid metabolism. GBA exhibits a complex physiological function that includes not only the degradation of its substrate glucosylceramide but also the metabolism of other sphingolipids and additional lipids such as cholesterol, particularly when glucocerebrosidase activity is deficient. In the context of Parkinson's disease associated with GBA, the loss of GBA activity has been associated with the accumulation of α-synuclein species. In recent years, several hypotheses have proposed alternative and complementary pathological mechanisms to explain why lysosomal enzyme mutations lead to α-synuclein accumulation and become important risk factors in Parkinson's disease etiology. Classically, loss of GBA activity has been linked to a dysfunctional autophagy-lysosome system and to a subsequent decrease in autophagy-dependent α-synuclein turnover; however, several other pathological mechanisms underlying GBA-associated parkinsonism have been proposed. This review summarizes and discusses the different hypotheses with a special focus on autophagy-dependent mechanisms, as well as autophagy-independent mechanisms, where the role of other players such as sphingolipids, cholesterol and other GBA-related proteins make important contributions to Parkinson's disease pathogenesis.
Collapse
|
8
|
Inokuchi JI, Go S, Hirabayashi Y. Synthesis of O-Linked Glycoconjugates in the Nervous System. ADVANCES IN NEUROBIOLOGY 2023; 29:95-116. [DOI: 10.1007/978-3-031-12390-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
9
|
Aureli M, Mauri L, Carsana EV, Dobi D, Breviario S, Lunghi G, Sonnino S. Gangliosides and Cell Surface Ganglioside Metabolic Enzymes in the Nervous System. ADVANCES IN NEUROBIOLOGY 2023; 29:305-332. [DOI: 10.1007/978-3-031-12390-0_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
10
|
Sandhoff R, Sandhoff K. Neuronal Ganglioside and Glycosphingolipid (GSL) Metabolism and Disease : Cascades of Secondary Metabolic Errors Can Generate Complex Pathologies (in LSDs). ADVANCES IN NEUROBIOLOGY 2023; 29:333-390. [PMID: 36255681 DOI: 10.1007/978-3-031-12390-0_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Glycosphingolipids (GSLs) are a diverse group of membrane components occurring mainly on the surfaces of mammalian cells. They and their metabolites have a role in intercellular communication, serving as versatile biochemical signals (Kaltner et al, Biochem J 476(18):2623-2655, 2019) and in many cellular pathways. Anionic GSLs, the sialic acid containing gangliosides (GGs), are essential constituents of neuronal cell surfaces, whereas anionic sulfatides are key components of myelin and myelin forming oligodendrocytes. The stepwise biosynthetic pathways of GSLs occur at and lead along the membranes of organellar surfaces of the secretory pathway. After formation of the hydrophobic ceramide membrane anchor of GSLs at the ER, membrane-spanning glycosyltransferases (GTs) of the Golgi and Trans-Golgi network generate cell type-specific GSL patterns for cellular surfaces. GSLs of the cellular plasma membrane can reach intra-lysosomal, i.e. luminal, vesicles (ILVs) by endocytic pathways for degradation. Soluble glycoproteins, the glycosidases, lipid binding and transfer proteins and acid ceramidase are needed for the lysosomal catabolism of GSLs at ILV-membrane surfaces. Inherited mutations triggering a functional loss of glycosylated lysosomal hydrolases and lipid binding proteins involved in GSL degradation cause a primary lysosomal accumulation of their non-degradable GSL substrates in lysosomal storage diseases (LSDs). Lipid binding proteins, the SAPs, and the various lipids of the ILV-membranes regulate GSL catabolism, but also primary storage compounds such as sphingomyelin (SM), cholesterol (Chol.), or chondroitin sulfate can effectively inhibit catabolic lysosomal pathways of GSLs. This causes cascades of metabolic errors, accumulating secondary lysosomal GSL- and GG- storage that can trigger a complex pathology (Breiden and Sandhoff, Int J Mol Sci 21(7):2566, 2020).
Collapse
Affiliation(s)
- Roger Sandhoff
- Lipid Pathobiochemistry Group, German Cancer Research Center, Heidelberg, Germany
| | - Konrad Sandhoff
- LIMES, c/o Kekule-Institute for Organic Chemistry and Biochemistry, University of Bonn, Bonn, Germany.
| |
Collapse
|
11
|
Galper J, Kim WS, Dzamko N. LRRK2 and Lipid Pathways: Implications for Parkinson's Disease. Biomolecules 2022; 12:1597. [PMID: 36358947 PMCID: PMC9687231 DOI: 10.3390/biom12111597] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 04/10/2024] Open
Abstract
Genetic alterations in the LRRK2 gene, encoding leucine-rich repeat kinase 2, are a common risk factor for Parkinson's disease. How LRRK2 alterations lead to cell pathology is an area of ongoing investigation, however, multiple lines of evidence suggest a role for LRRK2 in lipid pathways. It is increasingly recognized that in addition to being energy reservoirs and structural entities, some lipids, including neural lipids, participate in signaling cascades. Early investigations revealed that LRRK2 localized to membranous and vesicular structures, suggesting an interaction of LRRK2 and lipids or lipid-associated proteins. LRRK2 substrates from the Rab GTPase family play a critical role in vesicle trafficking, lipid metabolism and lipid storage, all processes which rely on lipid dynamics. In addition, LRRK2 is associated with the phosphorylation and activity of enzymes that catabolize plasma membrane and lysosomal lipids. Furthermore, LRRK2 knockout studies have revealed that blood, brain and urine exhibit lipid level changes, including alterations to sterols, sphingolipids and phospholipids, respectively. In human LRRK2 mutation carriers, changes to sterols, sphingolipids, phospholipids, fatty acyls and glycerolipids are reported in multiple tissues. This review summarizes the evidence regarding associations between LRRK2 and lipids, and the functional consequences of LRRK2-associated lipid changes are discussed.
Collapse
Affiliation(s)
- Jasmin Galper
- Charles Perkins Centre and Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, NSW 2050, Australia
| | - Woojin S Kim
- Brain and Mind Centre and Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, NSW 2050, Australia
| | - Nicolas Dzamko
- Charles Perkins Centre and Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, NSW 2050, Australia
- Brain and Mind Centre and Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, NSW 2050, Australia
| |
Collapse
|
12
|
Pereira de Sa N, Del Poeta M. Sterylglucosides in Fungi. J Fungi (Basel) 2022; 8:1130. [PMID: 36354897 PMCID: PMC9698648 DOI: 10.3390/jof8111130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/24/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
Sterylglucosides (SGs) are sterol conjugates widely distributed in nature. Although their universal presence in all living organisms suggests the importance of this kind of glycolipids, they are yet poorly understood. The glycosylation of sterols confers a more hydrophilic character, modifying biophysical properties of cell membranes and altering immunogenicity of the cells. In fungi, SGs regulate different cell pathways to help overcome oxygen and pH challenges, as well as help to accomplish cell recycling and other membrane functions. At the same time, the level of these lipids is highly controlled, especially in wild-type fungi. In addition, modulating SGs metabolism is becoming a novel tool for vaccine and antifungal development. In the present review, we bring together multiple observations to emphasize the underestimated importance of SGs for fungal cell functions.
Collapse
Affiliation(s)
- Nivea Pereira de Sa
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Maurizio Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY 11794, USA
- Institute of Chemical Biology and Drug Discovery (ICB&DD), Stony Brook, NY 11794, USA
- Division of Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
- Veterans Administration Medical Center, Northport, NY 11768, USA
| |
Collapse
|
13
|
Sanchez-Mirasierra I, Ghimire S, Hernandez-Diaz S, Soukup SF. Targeting Macroautophagy as a Therapeutic Opportunity to Treat Parkinson's Disease. Front Cell Dev Biol 2022; 10:921314. [PMID: 35874822 PMCID: PMC9298504 DOI: 10.3389/fcell.2022.921314] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/13/2022] [Indexed: 12/18/2022] Open
Abstract
Macroautophagy, an evolutionary conserved catabolic process in the eukaryotic cell, regulates cellular homeostasis and plays a decisive role in self-engulfing proteins, protein aggregates, dysfunctional or damaged organelles, and invading pathogens. Growing evidence from in vivo and in vitro models shows that autophagy dysfunction plays decisive role in the pathogenesis of various neurodegenerative diseases, including Parkinson's disease (PD). PD is an incurable and second most common neurodegenerative disease characterised by neurological and motor dysfunction accompanied of non-motor symptoms that can also reduce the life quality of patients. Despite the investment in research, the aetiology of the disease is still unknown and the therapies available are aimed mostly at ameliorating motor symptoms. Hence, therapeutics regulating the autophagy pathway might play an important role controlling the disease progression, reducing neuronal loss and even ameliorating non-motor symptoms. In this review, we highlight potential therapeutic opportunities involved in different targeting options like an initiation of autophagy, Leucine-rich repeat kinase 2 (LRRK2) inhibition, mitophagy, lysosomes, lipid metabolism, immune system, gene expression, biomarkers, and also non-pharmacological interventions. Thus, strategies to identify therapeutics targeting the pathways modulating autophagy might hold a future for therapy development against PD.
Collapse
Affiliation(s)
| | - Saurav Ghimire
- Universite Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France
| | | | | |
Collapse
|
14
|
Nie L, Quan L, Wu T, He R, Lyu Q. TransPPMP: predicting pathogenicity of frameshift and non-sense mutations by a Transformer based on protein features. Bioinformatics 2022; 38:2705-2711. [PMID: 35561183 DOI: 10.1093/bioinformatics/btac188] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 01/04/2022] [Accepted: 03/26/2022] [Indexed: 11/13/2022] Open
Abstract
MOTIVATION Protein structure can be severely disrupted by frameshift and non-sense mutations at specific positions in the protein sequence. Frameshift and non-sense mutation cases can also be found in healthy individuals. A method to distinguish neutral and potentially disease-associated frameshift and non-sense mutations is of practical and fundamental importance. It would allow researchers to rapidly screen out the potentially pathogenic sites from a large number of mutated genes and then use these sites as drug targets to speed up diagnosis and improve access to treatment. The problem of how to distinguish between neutral and potentially disease-associated frameshift and non-sense mutations remains under-researched. RESULTS We built a Transformer-based neural network model to predict the pathogenicity of frameshift and non-sense mutations on protein features and named it TransPPMP. The feature matrix of contextual sequences computed by the ESM pre-training model, type of mutation residue and the auxiliary features, including structure and function information, are combined as input features, and the focal loss function is designed to solve the sample imbalance problem during the training. In 10-fold cross-validation and independent blind test set, TransPPMP showed good robust performance and absolute advantages in all evaluation metrics compared with four other advanced methods, namely, ENTPRISE-X, VEST-indel, DDIG-in and CADD. In addition, we demonstrate the usefulness of the multi-head attention mechanism in Transformer to predict the pathogenicity of mutations-not only can multiple self-attention heads learn local and global interactions but also functional sites with a large influence on the mutated residue can be captured by attention focus. These could offer useful clues to study the pathogenicity mechanism of human complex diseases for which traditional machine learning methods fall short. AVAILABILITY AND IMPLEMENTATION TransPPMP is available at https://github.com/lennylv/TransPPMP. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Liangpeng Nie
- School of Computer Science and Technology, Soochow University, Suzhou 215006, China
| | - Lijun Quan
- School of Computer Science and Technology, Soochow University, Suzhou 215006, China
- Province Key Lab for Information Processing Technologies, Soochow University, Suzhou 215006, China
- Collaborative Innovation Center of Novel Software Technology and Industrialization, Nanjing 210000, China
| | - Tingfang Wu
- School of Computer Science and Technology, Soochow University, Suzhou 215006, China
- Province Key Lab for Information Processing Technologies, Soochow University, Suzhou 215006, China
- Collaborative Innovation Center of Novel Software Technology and Industrialization, Nanjing 210000, China
| | - Ruji He
- School of Computer Science and Technology, Soochow University, Suzhou 215006, China
| | - Qiang Lyu
- School of Computer Science and Technology, Soochow University, Suzhou 215006, China
- Province Key Lab for Information Processing Technologies, Soochow University, Suzhou 215006, China
- Collaborative Innovation Center of Novel Software Technology and Industrialization, Nanjing 210000, China
| |
Collapse
|
15
|
Kezic S, McAleer MA, Jakasa I, Goorden SMI, Ghauharali-van der Vlugt K, Beers-Stet FS, Meijer J, Roelofsen J, Nieman MM, van Kuilenburg ABP, Irvine AD. Children with atopic dermatitis show increased activity of β- glucocerebrosidase and stratum corneum levels of glucosylcholesterol that are strongly related to local cytokine milieu. Br J Dermatol 2022; 186:988-996. [PMID: 34993951 PMCID: PMC9325351 DOI: 10.1111/bjd.20979] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 12/20/2021] [Accepted: 12/30/2021] [Indexed: 12/01/2022]
Abstract
Background Atopic dermatitis (AD) is characterized by immune dysregulations and an impaired skin barrier, including abnormalities in lipid organization. In the stratum corneum (SC), β‐glucocerebrosidase (GBA) mediates transformation of glucosylceramide (GlcCER) into ceramide (CER) and cholesterol into glucosylcholesterol (GlcChol). Alteration in GBA activity might contribute to skin barrier defects in AD. Objectives To investigate GBA activity in the SC of children with AD before and after topical corticosteroid therapy and to compare it with healthy controls; to determine SC levels of GlcCER‐ and CER‐containing hydroxysphingosine base (GlcCER[H] and CER[H], respectively) and GlcChol; and to relate them to disease severity, skin barrier function and the local cytokine milieu. Methods Lipid markers and cytokines of innate, T helper 1 and T helper 2 immunity were determined in SC collected from healthy children and from clinically unaffected skin of children with AD, before and after 6 weeks of therapy with topical corticosteroids. AD severity was assessed by Scoring Atopic Dermatitis and skin barrier function by transepidermal water loss (TEWL). Results Baseline GBA activity and GlcChol levels were increased in children with AD but declined after therapy. CER[H] levels and the CER[H] to GlcCER[H] ratio were increased in AD. GBA activity and GlcChol correlated with TEWL and levels of multiple cytokines, especially interleukin‐1α and interleukin‐18. GlcChol was strongly associated with disease severity. Conclusions We show increased GBA activity and levels of GlcChol in AD. Our data suggest an important role of inflammation in disturbed lipid processing. GBA activity or GlcChol might be useful biomarkers in the monitoring of therapeutic responses in AD. What is already known about this topic?Patients with atopic dermatitis (AD) have a reduced skin barrier, mainly caused by altered lipid organization. The mechanisms underlying these lipid anomalies are not fully understood but likely reflect both genetic abnormalities in AD skin and the local cutaneous inflammatory environment.
What does this study add?We show increased activity of the ceramide‐generating enzyme β‐glucocerebrosidase in AD. Activity of this enzyme was correlated with the local cytokine milieu and declined after local corticosteroid therapy. We show that glucosylcholesterol levels in the stratum corneum are increased in AD. The function of glucosylcholesterol and the physiological consequences of increased levels are not clear yet; however, its levels were strongly correlated with skin barrier function: high transepidermal water loss strongly correlated with high levels of glucosylcholesterol.
What is the translational message?Correction of cutaneous inflammation largely restores alterations in lipid metabolism in the stratum corneum of infants with AD.
Collapse
Affiliation(s)
- Sanja Kezic
- Amsterdam UMC, University of Amsterdam, Department of Public and Occupational Health, Amsterdam Public Health research institute, Amsterdam, The Netherlands
| | - Maeve A McAleer
- Clinical Medicine, Trinity College Dublin, Dublin, Ireland.,Paediatric Dermatology, Children's Health Ireland at Crumlin, Dublin 12, Ireland
| | - Ivone Jakasa
- Laboratory for Analytical Chemistry, Department of Chemistry and Biochemistry, Faculty of Food Technology and Biotechnology, University of Zagreb, Zagreb, Croatia
| | - Susan M I Goorden
- Amsterdam UMC, University of Amsterdam, Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry Amsterdam UMC, Amsterdam, The Netherlands
| | - Karen Ghauharali-van der Vlugt
- Amsterdam UMC, University of Amsterdam, Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry Amsterdam UMC, Amsterdam, The Netherlands
| | - Femke S Beers-Stet
- Amsterdam UMC, University of Amsterdam, Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry Amsterdam UMC, Amsterdam, The Netherlands
| | - Judith Meijer
- Amsterdam UMC, University of Amsterdam, Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry Amsterdam UMC, Amsterdam, The Netherlands
| | - Jeroen Roelofsen
- Amsterdam UMC, University of Amsterdam, Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry Amsterdam UMC, Amsterdam, The Netherlands
| | - Monique M Nieman
- Amsterdam UMC, University of Amsterdam, Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry Amsterdam UMC, Amsterdam, The Netherlands
| | - André B P van Kuilenburg
- Amsterdam UMC, University of Amsterdam, Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry Amsterdam UMC, Amsterdam, The Netherlands
| | - Alan D Irvine
- Clinical Medicine, Trinity College Dublin, Dublin, Ireland.,Paediatric Dermatology, Children's Health Ireland at Crumlin, Dublin 12, Ireland
| |
Collapse
|
16
|
Su Q, Schröder SP, Lelieveld LT, Ferraz MJ, Verhoek M, Boot RG, Overkleeft HS, Aerts JMFG, Artola M, Kuo C. Xylose-Configured Cyclophellitols as Selective Inhibitors for Glucocerebrosidase. Chembiochem 2021; 22:3090-3098. [PMID: 34459538 PMCID: PMC8596838 DOI: 10.1002/cbic.202100396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/29/2021] [Indexed: 02/03/2023]
Abstract
Glucocerebrosidase (GBA), a lysosomal retaining β-d-glucosidase, has recently been shown to hydrolyze β-d-xylosides and to transxylosylate cholesterol. Genetic defects in GBA cause the lysosomal storage disorder Gaucher disease (GD), and also constitute a risk factor for developing Parkinson's disease. GBA and other retaining glycosidases can be selectively visualized by activity-based protein profiling (ABPP) using fluorescent probes composed of a cyclophellitol scaffold having a configuration tailored to the targeted glycosidase family. GBA processes β-d-xylosides in addition to β-d-glucosides, this in contrast to the other two mammalian cellular retaining β-d-glucosidases, GBA2 and GBA3. Here we show that the xylopyranose preference also holds up for covalent inhibitors: xylose-configured cyclophellitol and cyclophellitol aziridines selectively react with GBA over GBA2 and GBA3 in vitro and in vivo, and that the xylose-configured cyclophellitol is more potent and more selective for GBA than the classical GBA inhibitor, conduritol B-epoxide (CBE). Both xylose-configured cyclophellitol and cyclophellitol aziridine cause accumulation of glucosylsphingosine in zebrafish embryo, a characteristic hallmark of GD, and we conclude that these compounds are well suited for creating such chemically induced GD models.
Collapse
Affiliation(s)
- Qin Su
- Department of Medical BiochemistryLeiden Institute of ChemistryLeiden UniversityEinsteinweg 552333 CCLeidenThe Netherlands
| | - Sybrin P. Schröder
- Department of Bio-organic SynthesisLeiden Institute of ChemistryLeiden UniversityEinsteinweg 552333 CCLeidenThe Netherlands
| | - Lindsey T. Lelieveld
- Department of Medical BiochemistryLeiden Institute of ChemistryLeiden UniversityEinsteinweg 552333 CCLeidenThe Netherlands
| | - Maria J. Ferraz
- Department of Medical BiochemistryLeiden Institute of ChemistryLeiden UniversityEinsteinweg 552333 CCLeidenThe Netherlands
| | - Marri Verhoek
- Department of Medical BiochemistryLeiden Institute of ChemistryLeiden UniversityEinsteinweg 552333 CCLeidenThe Netherlands
| | - Rolf G. Boot
- Department of Medical BiochemistryLeiden Institute of ChemistryLeiden UniversityEinsteinweg 552333 CCLeidenThe Netherlands
| | - Herman S. Overkleeft
- Department of Bio-organic SynthesisLeiden Institute of ChemistryLeiden UniversityEinsteinweg 552333 CCLeidenThe Netherlands
| | - Johannes M. F. G. Aerts
- Department of Medical BiochemistryLeiden Institute of ChemistryLeiden UniversityEinsteinweg 552333 CCLeidenThe Netherlands
| | - Marta Artola
- Department of Medical BiochemistryLeiden Institute of ChemistryLeiden UniversityEinsteinweg 552333 CCLeidenThe Netherlands
| | - Chi‐Lin Kuo
- Department of Medical BiochemistryLeiden Institute of ChemistryLeiden UniversityEinsteinweg 552333 CCLeidenThe Netherlands
| |
Collapse
|
17
|
Fonseca Cabral G, Schaan AP, Cavalcante GC, Sena-dos-Santos C, de Souza TP, Souza Port’s NM, dos Santos Pinheiro JA, Ribeiro-dos-Santos Â, Vidal AF. Nuclear and Mitochondrial Genome, Epigenome and Gut Microbiome: Emerging Molecular Biomarkers for Parkinson's Disease. Int J Mol Sci 2021; 22:9839. [PMID: 34576000 PMCID: PMC8471599 DOI: 10.3390/ijms22189839] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/23/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is currently the second most common neurodegenerative disorder, burdening about 10 million elderly individuals worldwide. The multifactorial nature of PD poses a difficult obstacle for understanding the mechanisms involved in its onset and progression. Currently, diagnosis depends on the appearance of clinical signs, some of which are shared among various neurologic disorders, hindering early diagnosis. There are no effective tools to prevent PD onset, detect the disease in early stages or accurately report the risk of disease progression. Hence, there is an increasing demand for biomarkers that may identify disease onset and progression, as treatment-based medicine may not be the best approach for PD. Over the last few decades, the search for molecular markers to predict susceptibility, aid in accurate diagnosis and evaluate the progress of PD have intensified, but strategies aimed to improve individualized patient care have not yet been established. CONCLUSIONS Genomic variation, regulation by epigenomic mechanisms, as well as the influence of the host gut microbiome seem to have a crucial role in the onset and progress of PD, thus are considered potential biomarkers. As such, the human nuclear and mitochondrial genome, epigenome, and the host gut microbiome might be the key elements to the rise of personalized medicine for PD patients.
Collapse
Affiliation(s)
- Gleyce Fonseca Cabral
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
| | - Ana Paula Schaan
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
| | - Giovanna C. Cavalcante
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
| | - Camille Sena-dos-Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
| | - Tatiane Piedade de Souza
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
| | - Natacha M. Souza Port’s
- Laboratório de Neurofarmacologia Molecular, Universidade de São Paulo, São Paulo 05508-000, Brazil;
| | - Jhully Azevedo dos Santos Pinheiro
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
| | - Ândrea Ribeiro-dos-Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará–R. dos Mundurucus, Belém 66073-000, Brazil
- Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil
| | - Amanda F. Vidal
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
- Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil
- ITVDS—Instituto Tecnológico Vale Desenvolvimento Sustentável–R. Boaventura da Silva, Belém 66055-090, Brazil
| |
Collapse
|
18
|
Xu R, Zheng R, Wang Y, Ma R, Tong G, Wei X, Feng D, Hu K. Transcriptome analysis to elucidate the toxicity mechanisms of fenvalerate, sulfide gatifloxacin, and ridomil on the hepatopancreas of Procambarus clarkii. FISH & SHELLFISH IMMUNOLOGY 2021; 116:140-149. [PMID: 34256134 DOI: 10.1016/j.fsi.2021.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/15/2021] [Accepted: 07/09/2021] [Indexed: 06/13/2023]
Abstract
Most antibiotics, insecticides, and other chemicals used in agricultural and fishery production tend to persist in the environment. Fenvalerate, sulfide gatifloxacin, and ridomil are widely used in aquaculture as antibacterial, antifungal, and antiparasitic drugs; however, their toxicity mechanism remains unclear. Thus, we herein analyzed the effects of these three drugs on the hepatopancreas of Procambarus clarkii at the transcriptome level. Twelve normalized cDNA libraries were constructed using RNA extracted from P. clarkii after treatment with fenvalerate, sulfide gatifloxacin, or ridomil and from an untreated control group, followed by Kyoto Encyclopedia of Genes and Genomes pathway analysis. In the control vs fenvalerate and control vs sulfide gatifloxacin groups, 14 and seven pathways were significantly enriched, respectively. Further, the effects of fenvalerate and sulfide gatifloxacin were similar on the hepatopancreas of P. clarkii. We also found that the expression level of genes encoding senescence marker protein-30 and arylsulfatase A was downregulated in the sulfide gatifloxacin group, indicating that sulfide gatifloxacin accelerated the apoptosis of hepatopancreatocytes. The expression level of major facilitator superfamily domain containing 10 was downregulated, implying that it interferes with the ability of the hepatopancreas to metabolize drugs. Interestingly, we found that Niemann pick type C1 and glucosylceramidase-β potentially interact with each other, consequently decreasing the antioxidant capacity of P. clarkii hepatopancreas. In the fenvalerate group, the downregulation of the expression level of xanthine dehydrogenase indicated that fenvalerate affected the immune system of P. clarkii; moreover, the upregulation of the expression level of pancreatitis-associated protein-2 and cathepsin C indicated that fenvalerate caused possible inflammatory pathological injury to P. clarkii hepatopancreas. In the ridomil group, no pathway was significantly enriched. In total, 21 genes showed significant differences in all three groups. To conclude, although there appears to be some overlap in the toxicity mechanisms of fenvalerate, sulfide gatifloxacin, and ridomil, further studies are warranted.
Collapse
Affiliation(s)
- Ruze Xu
- National Pathogen Collection Center for Aquatic Animals, Shanghai Engineering Research Center of Aquaculture, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, PR China; National Fisheries Technical Extension Center, Beijing, 100125, PR China; Key Laboratory of East China Sea Fishery Resources Exploitation, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, 200090, PR China.
| | - Ruizhou Zheng
- National Pathogen Collection Center for Aquatic Animals, Shanghai Engineering Research Center of Aquaculture, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, PR China; National Fisheries Technical Extension Center, Beijing, 100125, PR China; Key Laboratory of East China Sea Fishery Resources Exploitation, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, 200090, PR China
| | - Yali Wang
- National Pathogen Collection Center for Aquatic Animals, Shanghai Engineering Research Center of Aquaculture, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, PR China; National Fisheries Technical Extension Center, Beijing, 100125, PR China; Key Laboratory of East China Sea Fishery Resources Exploitation, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, 200090, PR China
| | - Rongrong Ma
- National Pathogen Collection Center for Aquatic Animals, Shanghai Engineering Research Center of Aquaculture, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, PR China
| | - Guixiang Tong
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, 530021, PR China
| | - Xinxian Wei
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, 530021, PR China
| | - Dongyue Feng
- National Fisheries Technical Extension Center, Beijing, 100125, PR China.
| | - Kun Hu
- National Pathogen Collection Center for Aquatic Animals, Shanghai Engineering Research Center of Aquaculture, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, PR China; National Fisheries Technical Extension Center, Beijing, 100125, PR China; Key Laboratory of East China Sea Fishery Resources Exploitation, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, 200090, PR China.
| |
Collapse
|
19
|
Wild-type GBA1 increases the α-synuclein tetramer-monomer ratio, reduces lipid-rich aggregates, and attenuates motor and cognitive deficits in mice. Proc Natl Acad Sci U S A 2021; 118:2103425118. [PMID: 34326260 PMCID: PMC8346893 DOI: 10.1073/pnas.2103425118] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The mechanisms responsible for brain α-synuclein (αS) dyshomeostasis, caused by Gaucher’s GBA1 mutations that increase Parkinson’s disease (PD) risk, are largely unknown. We previously showed that abrogating physiological αS tetramers by a familial PD-E46K–amplified 3K mutation produces PD-like syndrome in mice and that treatment with stearoyl-CoA desaturase inhibitors increased a portion of the αS tetramers, benefitting the motor phenotypes. Here, we show that—similar to previous findings in GBA1-mutant PD culture—GCase elevation prolonged the stabilization of wild-type and 3K mutant αS tetramers in wtGBA1–transduced mouse brains, improving lysosomal integrity and motor and cognitive phenotypes. These data help elucidating lipid modulators that impact the αS physiological state in vivo and the development of PD therapeutic approaches. Loss-of-function mutations in acid beta-glucosidase 1 (GBA1) are among the strongest genetic risk factors for Lewy body disorders such as Parkinson’s disease (PD) and Lewy body dementia (DLB). Altered lipid metabolism in PD patient–derived neurons, carrying either GBA1 or PD αS mutations, can shift the physiological α-synuclein (αS) tetramer–monomer (T:M) equilibrium toward aggregation-prone monomers. A resultant increase in pSer129+ αS monomers provides a likely building block for αS aggregates. 3K αS mice, representing a neuropathological amplification of the E46K PD–causing mutation, have decreased αS T:M ratios and vesicle-rich αS+ aggregates in neurons, accompanied by a striking PD-like motor syndrome. We asked whether enhancing glucocerebrosidase (GCase) expression could benefit αS dyshomeostasis by delivering an adeno-associated virus (AAV)–human wild-type (wt) GBA1 vector into the brains of 3K neonates. Intracerebroventricular AAV-wtGBA1 at postnatal day 1 resulted in prominent forebrain neuronal GCase expression, sustained through 6 mo. GBA1 attenuated behavioral deficits both in working memory and fine motor performance tasks. Furthermore, wtGBA1 increased αS solubility and the T:M ratio in both 3K-GBA mice and control littermates and reduced pS129+ and lipid-rich aggregates in 3K-GBA. We observed GCase distribution in more finely dispersed lysosomes, in which there was increased GCase activity, lysosomal cathepsin D and B maturation, decreased perilipin-stabilized lipid droplets, and a normalized TFEB translocation to the nucleus, all indicative of improved lysosomal function and lipid turnover. Therefore, a prolonged increase of the αS T:M ratio by elevating GCase activity reduced the lipid- and vesicle-rich aggregates and ameliorated PD-like phenotypes in mice, further supporting lipid modulating therapies in PD.
Collapse
|
20
|
Endo I, Watanabe T, Miyamoto T, Monjusho-Goda H, Ohara J, Hayashi M, Hama Y, Ishibashi Y, Okino N, Ito M. C4-monomethylsterol β-glucoside and its synthase in Aurantiochytrium limacinum mh0186. Glycobiology 2021; 31:1350-1363. [PMID: 34224567 DOI: 10.1093/glycob/cwab070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 11/14/2022] Open
Abstract
Thraustochytrids, unicellular marine protists, synthesize polyunsaturated fatty acids (PUFAs) and PUFA-containing phospholipids; however, little is known about their glycolipids and their associated metabolism. Here, we report two glycolipids (GL-A, B) and their synthases in Aurantiochytrium limacinum mh0186. Two glycolipids were purified from A. limacinum mh0186, and they were determined by gas chromatography, mass spectrometry and two-dimensional nuclear magnetic resonance to be 3-O-β-D-glucopyranosyl-stigmasta-5,7,22-triene (GL-A) and 3-O-β-D-glucopyranosyl-4α-methyl-stigmasta-7,22-diene (GL-B), both of which are sterol β-glucosides (β-SGs); the structure of GL-B has not been reported thus far. Seven candidate genes responsible for the synthesis of these β-SGs were extracted from the draft genome database of A. limacinum using the yeast sterol β-glucosyltransferase (SGT; EC 2.4.1.173) sequence as a query. Expression analysis using Saccharomyces cerevisiae revealed that two gene products (AlSGT-1 and 2) catalyze the transfer of glucose from UDP-glucose to sterols, generating sterylglucosides (SGs). Compared to AlSGT-1, AlSGT-2 exhibited wide specificity for sterols and used C4-monomethylsterol to synthesize GL-B. The disruption of alsgt-2 but not alsgt-1 in strain mh0186 resulted in a decrease in total SG and almost complete loss of GL-B, indicating that AlSGT-2 is responsible for the synthesis of β-SGs in A. limacinum mh0186, especially GL-B, which possesses a unique sterol structure.
Collapse
Affiliation(s)
- Ikumi Endo
- Department of Bioscience and Biotechnology
| | | | - Tomofumi Miyamoto
- Graduate School of Pharmaceutical Science, Kyushu University, Fukuoka 812-8582, Japan
| | | | | | - Masahiro Hayashi
- Department of Marine Biology and Environmental Sciences, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan
| | - Yoichiro Hama
- Applied Biochemistry and Food Science Course, Faculty of Agriculture, Saga University, 1 Honjo, Saga 840-8502, Japan
| | | | | | - Makoto Ito
- Department of Bioscience and Biotechnology.,Innovative Bio-Architecture Center, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka 819-0395
| |
Collapse
|
21
|
Prasongsukarn K, Dechkhajorn W, Benjathummarak S, Maneerat Y. TRPM2, PDLIM5, BCL3, CD14, GBA Genes as Feasible Markers for Premature Coronary Heart Disease Risk. Front Genet 2021; 12:598296. [PMID: 34093636 PMCID: PMC8172979 DOI: 10.3389/fgene.2021.598296] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 04/19/2021] [Indexed: 12/22/2022] Open
Abstract
Background: Beyond non-genetic risk factors, familial hypercholesterolemia (FH) plays a major role in the development of CHD. FH is a genetic disorder characterized by heritable and severely elevated levels of low-density lipoprotein (LDL) cholesterol, which can lead to premature cardiovascular disease, particularly familial coronary heart disease (FH-CHD). Method: To explore genes indicating a risk of familial (premature) coronary heart disease (FH-CHD) development in FH, 30 Thai male volunteers were enrolled: 7 healthy controls (N), 6 patients with hypercholesterolemia (H), 4 with FH, 10 with CHD, and 3 with FH-CHD. Transcriptome data were investigated using next-generation sequencing analysis in whole blood (n = 3). Genes that were significantly expressed in both FH and FH-CHD, but not in N, H, and CHD groups, were selected and functionally analyzed. Results: The findings revealed that 55 intersecting genes were differentially expressed between FH and FH-CHD groups. Ten of the 55 genes (MAPK14, TRPM2, STARD8, PDLIM5, BCL3, BLOC1S5, GBA, RBMS1, CD14, and CD36 were selected for validation. These 10 genes play potential roles in chronic inflammation and are involved in pathways related to pathogenesis of CHD. Using quantitative real-time PCR, we evaluated the mRNA expression of the selected genes in all 30 volunteers. TRPM2, PDLIM5, BCL3 were significantly upregulated and GBA was significantly downregulated in both FH and FH-CHD compared with the N, H, and CHD groups. Conclusion: our preliminary investigation reveals that the TRPM2, PDLIM5, BCL3, and GBA genes may have potential for further development as predictive markers for FH-CHD.
Collapse
Affiliation(s)
| | - Wilanee Dechkhajorn
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Surachet Benjathummarak
- Center of Excellence for Antibody Research, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Yaowapa Maneerat
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
22
|
Structure, metabolism and biological functions of steryl glycosides in mammals. Biochem J 2021; 477:4243-4261. [PMID: 33186452 PMCID: PMC7666875 DOI: 10.1042/bcj20200532] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/09/2020] [Accepted: 10/21/2020] [Indexed: 12/20/2022]
Abstract
Steryl glycosides (SGs) are sterols glycosylated at their 3β-hydroxy group. They are widely distributed in plants, algae, and fungi, but are relatively rare in bacteria and animals. Glycosylation of sterols, resulting in important components of the cell membrane SGs, alters their biophysical properties and confers resistance against stress by freezing or heat shock to cells. Besides, many biological functions in animals have been suggested from the observations of SG administration. Recently, cholesteryl glucosides synthesized via the transglycosidation by glucocerebrosidases (GBAs) were found in the central nervous system of animals. Identification of patients with congenital mutations in GBA genes or availability of respective animal models will enable investigation of the function of such endogenously synthesized cholesteryl glycosides by genetic approaches. In addition, mechanisms of the host immune responses against pathogenic bacterial SGs have partially been resolved. This review is focused on the biological functions of SGs in mammals taking into consideration their therapeutic applications in the future.
Collapse
|
23
|
Boer DE, Mirzaian M, Ferraz MJ, Zwiers KC, Baks MV, Hazeu MD, Ottenhoff R, Marques ARA, Meijer R, Roos JCP, Cox TM, Boot RG, Pannu N, Overkleeft HS, Artola M, Aerts JM. Human glucocerebrosidase mediates formation of xylosyl-cholesterol by β-xylosidase and transxylosidase reactions. J Lipid Res 2021; 62:100018. [PMID: 33361282 PMCID: PMC7903134 DOI: 10.1194/jlr.ra120001043] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 12/14/2020] [Accepted: 12/23/2020] [Indexed: 11/20/2022] Open
Abstract
Deficiency of glucocerebrosidase (GBA), a lysosomal β-glucosidase, causes Gaucher disease. The enzyme hydrolyzes β-glucosidic substrates and transglucosylates cholesterol to cholesterol-β-glucoside. Here we show that recombinant human GBA also cleaves β-xylosides and transxylosylates cholesterol. The xylosyl-cholesterol formed acts as an acceptor for the subsequent formation of di-xylosyl-cholesterol. Common mutant forms of GBA from patients with Gaucher disease with reduced β-glucosidase activity were similarly impaired in β-xylosidase, transglucosidase, and transxylosidase activities, except for a slightly reduced xylosidase/glucosidase activity ratio of N370S GBA and a slightly reduced transglucosylation/glucosidase activity ratio of D409H GBA. XylChol was found to be reduced in spleen from patients with Gaucher disease. The origin of newly identified XylChol in mouse and human tissues was investigated. Cultured human cells exposed to exogenous β-xylosides generated XylChol in a manner dependent on active lysosomal GBA but not the cytosol-facing β-glucosidase GBA2. We later sought an endogenous β-xyloside acting as donor in transxylosylation reactions, identifying xylosylated ceramide (XylCer) in cells and tissues that serve as donor in the formation of XylChol. UDP-glucosylceramide synthase (GCS) was unable to synthesize XylChol but could catalyze the formation of XylCer. Thus, food-derived β-D-xyloside and XylCer are potential donors for the GBA-mediated formation of XylChol in cells. The enzyme GCS produces XylCer at a low rate. Our findings point to further catalytic versatility of GBA and prompt a systematic exploration of the distribution and role of xylosylated lipids.
Collapse
Affiliation(s)
- Daphne E Boer
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Mina Mirzaian
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Maria J Ferraz
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Kimberley C Zwiers
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Merel V Baks
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Marc D Hazeu
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Roelof Ottenhoff
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - André R A Marques
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Rianne Meijer
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Jonathan C P Roos
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Timothy M Cox
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Rolf G Boot
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Navraj Pannu
- Department of Biophysical Structural Chemistry, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Herman S Overkleeft
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Marta Artola
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Johannes M Aerts
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, The Netherlands.
| |
Collapse
|
24
|
Effect of Expression of Human Glucosylceramidase 2 Isoforms on Lipid Profiles in COS-7 Cells. Metabolites 2020; 10:metabo10120488. [PMID: 33261081 PMCID: PMC7761373 DOI: 10.3390/metabo10120488] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/20/2020] [Accepted: 11/25/2020] [Indexed: 02/08/2023] Open
Abstract
Glucosylceramide (GlcCer) is a major membrane lipid and the precursor of gangliosides. GlcCer is mainly degraded by two enzymes, lysosomal acid β-glucosidase (GBA) and nonlysosomal β-glucosidase (GBA2), which may have different isoforms because of alternative splicing. To understand which GBA2 isoforms are active and how they affect glycosphingolipid levels in cells, we expressed nine human GBA2 isoforms in COS-7 cells, confirmed their expression by qRT-PCR and Western blotting, and assayed their activity to hydrolyze 4-methylumbelliferyl-β-D-glucopyranoside (4MUG) in cell extracts. Human GBA2 isoform 1 showed high activity, while the other isoforms had activity similar to the background. Comparison of sphingolipid levels by ultra-high resolution/accurate mass spectrometry (UHRAMS) analysis showed that isoform 1 overexpression increased ceramide and decreased hexosylceramide levels. Comparison of ratios of glucosylceramides to the corresponding ceramides in the extracts indicated that GBA2 isoform 1 has broad specificity for the lipid component of glucosylceramide, suggesting that only one GBA2 isoform 1 is active and affects sphingolipid levels in the cell. Our study provides new insights into how increased breakdown of GlcCer affects cellular lipid metabolic networks.
Collapse
|
25
|
Hanashima S, Fukuda N, Malabed R, Murata M, Kinoshita M, Greimel P, Hirabayashi Y. β-Glucosylation of cholesterol reduces sterol-sphingomyelin interactions. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1863:183496. [PMID: 33130096 DOI: 10.1016/j.bbamem.2020.183496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/10/2020] [Accepted: 10/16/2020] [Indexed: 12/24/2022]
Abstract
Cholesteryl-β-D-glucoside (ChoGlc) is a mammalian glycolipid that is expressed in brain tissue. The effects of glucosylation on the ordering and lipid interactions of cholesterol (Cho) were examined in membranes composed of N-stearoyl sphingomyelin (SSM), which is abundant in the brain, and to investigate the possible molecular mechanism involved in these interactions. Differential scanning calorimetry revealed that ChoGlc was miscible with SSM in a similar extent of Cho. Solid-state 2H NMR of deuterated SSM and fluorescent anisotropy using 1,6-diphenylhexatriene demonstrated that the glucosylation of Cho significantly reduced the effect of the sterol tetracyclic core on the ordering of SSM chains. The orientation of the sterol core was further examined by solid-state NMR analysis of deuterated and fluorinated ChoGlc analogues. ChoGlc had a smaller tilt angle between the long molecular axis (C3-C17) and the membrane normal than Cho in SSM bilayers, and the fluctuations in the tilt angle were largely unaffected by temperature-dependent mobility changes of SSM acyl chains. This orientation of the sterol core of ChoGlc leads to reduce sterol-SSM interactions. The MD simulation results suggested that the Glc moiety perturbs the SSM-sterol interactions, which reduces the umbrella effect of the phosphocholine headgroup because the hydrophilic glucose moiety resides at the same depth as an SSM amide group. These differences between ChoGlc and Cho also weaken the SSM-ChoGlc interactions. Thus, the distribution and localization of Cho and ChoGlc possibly control the stability of sphingomyelin-based domains that transiently occur at specific locations in biological membranes.
Collapse
Affiliation(s)
- Shinya Hanashima
- Department of Chemistry, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan.
| | - Nanami Fukuda
- Department of Chemistry, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan
| | - Raymond Malabed
- Department of Chemistry, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan
| | - Michio Murata
- Department of Chemistry, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan.
| | - Msanao Kinoshita
- Department of Chemistry, Graduate School of Science, Kyushu University, Fukuoka, Fukuoka 819-0395, Japan
| | - Peter Greimel
- Laboratory for Cell Function Dynamics, Brain Science Institute, RIKEN Institute, Wako, Saitama 351-0198, Japan
| | - Yoshio Hirabayashi
- RIKEN Cluster for Pioneering Research, RIKEN, Wako, Saitama 351-0198, Japan; Institute for Environmental and Gender-Specific Medicine, Juntendo University Graduate School of Medicine, Urayasu, Chiba 279-0021, Japan
| |
Collapse
|
26
|
Boer DEC, Mirzaian M, Ferraz MJ, Nadaban A, Schreuder A, Hovnanian A, van Smeden J, Bouwstra JA, Aerts JMFG. Glucosylated cholesterol in skin: Synthetic role of extracellular glucocerebrosidase. Clin Chim Acta 2020; 510:707-710. [PMID: 32946792 DOI: 10.1016/j.cca.2020.09.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/20/2020] [Accepted: 09/11/2020] [Indexed: 11/25/2022]
Abstract
The existence of glucosylated cholesterol (GlcChol) in tissue has recently been recognized. GlcChol is generated from glucosylceramide (GlcCer) and cholesterol through transglucosylation by two retaining β-glucosidases, GBA and GBA2. Given the abundance of GBA, GlcCer and cholesterol in the skin's stratum corneum (SC), we studied the occurrence of GlcChol. A significant amount of GlcChol was detected in SC (6 pmol/mg weight). The ratio GlcChol/GlcCer is higher in SC than epidermis, 0.083 and 0.011, respectively. Examination of GlcChol in patients with Netherton syndrome revealed comparable levels (11 pmol/mg). Concluding, GlcChol was identified as a novel component in SC and is likely locally metabolized by GBA. The physiological function of GlcChol in the SC warrants future investigation.
Collapse
Affiliation(s)
- Daphne E C Boer
- Medical Biochemistry Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Mina Mirzaian
- Medical Biochemistry Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Maria J Ferraz
- Medical Biochemistry Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Andreea Nadaban
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Anne Schreuder
- Medical Biochemistry Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Alain Hovnanian
- INSERM UMR1163, Imagine Institute, Paris Descartes University, 75015 Paris, France; Department of Genetics, Necker-Enfants Malades Hospital, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
| | - Jeroen van Smeden
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands; Centre for Human Drug Research, Leiden, the Netherlands
| | - Joke A Bouwstra
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Johannes M F G Aerts
- Medical Biochemistry Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands.
| |
Collapse
|
27
|
Sultana S, Stewart J, van der Spoel AC. Truncated mutants of beta-glucosidase 2 (GBA2) are localized in the mitochondrial matrix and cause mitochondrial fragmentation. PLoS One 2020; 15:e0233856. [PMID: 32492073 PMCID: PMC7269613 DOI: 10.1371/journal.pone.0233856] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/13/2020] [Indexed: 12/22/2022] Open
Abstract
The enzyme β-glucosidase 2 (GBA2) is clinically relevant because it is targeted by the drug miglustat (Zavesca®) and because it is involved in inherited diseases. Mutations in the GBA2 gene are associated with two neurological diseases on the ataxia-spasticity spectrum, hereditary spastic paraplegia 46 (SPG46) and Marinesco-Sjögren-like syndrome (MSS). To establish how GBA2 mutations give rise to neurological pathology, we have begun to investigate mutant forms of GBA2 encoded by disease-associated GBA2 alleles. Previously, we found that five GBA2 missense mutants and five C-terminally truncated mutants lacked enzyme activity. Here we have examined the cellular locations of wild-type (WT) and mutant forms of GBA2 by confocal and electron microscopy, using transfected cells. Similar to GBA2-WT, the D594H and M510Vfs*17 GBA2 mutants were located at the plasma membrane, whereas the C-terminally truncated mutants terminating after amino acids 233 and 339 (GBA2-233 and -339) were present in the mitochondrial matrix, induced mitochondrial fragmentation and loss of mitochondrial transmembrane potential. Deletional mutagenesis indicated that residues 161–200 are critical for the mitochondrial fragmentation of GBA2-233 and -339. Considering that the mitochondrial fragmentation induced by GBA2-233 and -339 is consistently accompanied by their localization to the mitochondrial matrix, our deletional analysis raises the possibility that that GBA2 residues 161–200 harbor an internal targeting sequence for transport to the mitochondrial matrix. Altogether, our work provides new insights into the behaviour of GBA2-WT and disease-associated forms of GBA2.
Collapse
Affiliation(s)
- Saki Sultana
- The Atlantic Research Centre, Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jacklyn Stewart
- Biomedical Sciences Program, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Aarnoud C. van der Spoel
- The Atlantic Research Centre, Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
- The Atlantic Research Centre, Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
- * E-mail:
| |
Collapse
|
28
|
Akiyama H, Ide M, Nagatsuka Y, Sayano T, Nakanishi E, Uemura N, Yuyama K, Yamaguchi Y, Kamiguchi H, Takahashi R, Aerts JMFG, Greimel P, Hirabayashi Y. Glucocerebrosidases catalyze a transgalactosylation reaction that yields a newly-identified brain sterol metabolite, galactosylated cholesterol. J Biol Chem 2020; 295:5257-5277. [PMID: 32144204 PMCID: PMC7170530 DOI: 10.1074/jbc.ra119.012502] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/18/2020] [Indexed: 01/05/2023] Open
Abstract
β-Glucocerebrosidase (GBA) hydrolyzes glucosylceramide (GlcCer) to generate ceramide. Previously, we demonstrated that lysosomal GBA1 and nonlysosomal GBA2 possess not only GlcCer hydrolase activity, but also transglucosylation activity to transfer the glucose residue from GlcCer to cholesterol to form β-cholesterylglucoside (β-GlcChol) in vitro β-GlcChol is a member of sterylglycosides present in diverse species. How GBA1 and GBA2 mediate β-GlcChol metabolism in the brain is unknown. Here, we purified and characterized sterylglycosides from rodent and fish brains. Although glucose is thought to be the sole carbohydrate component of sterylglycosides in vertebrates, structural analysis of rat brain sterylglycosides revealed the presence of galactosylated cholesterol (β-GalChol), in addition to β-GlcChol. Analyses of brain tissues from GBA2-deficient mice and GBA1- and/or GBA2-deficient Japanese rice fish (Oryzias latipes) revealed that GBA1 and GBA2 are responsible for β-GlcChol degradation and formation, respectively, and that both GBA1 and GBA2 are responsible for β-GalChol formation. Liquid chromatography-tandem MS revealed that β-GlcChol and β-GalChol are present throughout development from embryo to adult in the mouse brain. We found that β-GalChol expression depends on galactosylceramide (GalCer), and developmental onset of β-GalChol biosynthesis appeared to be during myelination. We also found that β-GlcChol and β-GalChol are secreted from neurons and glial cells in association with exosomes. In vitro enzyme assays confirmed that GBA1 and GBA2 have transgalactosylation activity to transfer the galactose residue from GalCer to cholesterol to form β-GalChol. This is the first report of the existence of β-GalChol in vertebrates and how β-GlcChol and β-GalChol are formed in the brain.
Collapse
Affiliation(s)
- Hisako Akiyama
- RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan; RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Mitsuko Ide
- RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan; Cellular Informatics Laboratory, RIKEN, Wako, Saitama 351-0198, Japan
| | | | - Tomoko Sayano
- RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Etsuro Nakanishi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Norihito Uemura
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Kohei Yuyama
- Lipid Biofunction Section, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Hokkaido 001-0021, Japan
| | - Yoshiki Yamaguchi
- Laboratory of Pharmaceutical Physical Chemistry, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan
| | - Hiroyuki Kamiguchi
- RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan; RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Johannes M F G Aerts
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden 2333 CC, The Netherlands
| | - Peter Greimel
- RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan
| | - Yoshio Hirabayashi
- RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan; Cellular Informatics Laboratory, RIKEN, Wako, Saitama 351-0198, Japan.
| |
Collapse
|
29
|
Kytidou K, Artola M, Overkleeft HS, Aerts JMFG. Plant Glycosides and Glycosidases: A Treasure-Trove for Therapeutics. FRONTIERS IN PLANT SCIENCE 2020; 11:357. [PMID: 32318081 PMCID: PMC7154165 DOI: 10.3389/fpls.2020.00357] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/11/2020] [Indexed: 05/10/2023]
Abstract
Plants contain numerous glycoconjugates that are metabolized by specific glucosyltransferases and hydrolyzed by specific glycosidases, some also catalyzing synthetic transglycosylation reactions. The documented value of plant-derived glycoconjugates to beneficially modulate metabolism is first addressed. Next, focus is given to glycosidases, the central theme of the review. The therapeutic value of plant glycosidases is discussed as well as the present production in plant platforms of therapeutic human glycosidases used in enzyme replacement therapies. The increasing knowledge on glycosidases, including structure and catalytic mechanism, is described. The novel insights have allowed the design of functionalized highly specific suicide inhibitors of glycosidases. These so-called activity-based probes allow unprecedented visualization of glycosidases cross-species. Here, special attention is paid on the use of such probes in plant science that promote the discovery of novel enzymes and the identification of potential therapeutic inhibitors and chaperones.
Collapse
Affiliation(s)
- Kassiani Kytidou
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Marta Artola
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Herman S. Overkleeft
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Johannes M. F. G. Aerts
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| |
Collapse
|
30
|
Breiden B, Sandhoff K. Mechanism of Secondary Ganglioside and Lipid Accumulation in Lysosomal Disease. Int J Mol Sci 2020; 21:ijms21072566. [PMID: 32272755 PMCID: PMC7178057 DOI: 10.3390/ijms21072566] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/26/2020] [Accepted: 04/04/2020] [Indexed: 02/06/2023] Open
Abstract
Gangliosidoses are caused by monogenic defects of a specific hydrolase or an ancillary sphingolipid activator protein essential for a specific step in the catabolism of gangliosides. Such defects in lysosomal function cause a primary accumulation of multiple undegradable gangliosides and glycosphingolipids. In reality, however, predominantly small gangliosides also accumulate in many lysosomal diseases as secondary storage material without any known defect in their catabolic pathway. In recent reconstitution experiments, we identified primary storage materials like sphingomyelin, cholesterol, lysosphingolipids, and chondroitin sulfate as strong inhibitors of sphingolipid activator proteins (like GM2 activator protein, saposin A and B), essential for the catabolism of many gangliosides and glycosphingolipids, as well as inhibitors of specific catabolic steps in lysosomal ganglioside catabolism and cholesterol turnover. In particular, they trigger a secondary accumulation of ganglioside GM2, glucosylceramide and cholesterol in Niemann–Pick disease type A and B, and of GM2 and glucosylceramide in Niemann–Pick disease type C. Chondroitin sulfate effectively inhibits GM2 catabolism in mucopolysaccharidoses like Hurler, Hunter, Sanfilippo, and Sly syndrome and causes a secondary neuronal ganglioside GM2 accumulation, triggering neurodegeneration. Secondary ganglioside and lipid accumulation is furthermore known in many more lysosomal storage diseases, so far without known molecular basis.
Collapse
|
31
|
Meng Y, Heybrock S, Neculai D, Saftig P. Cholesterol Handling in Lysosomes and Beyond. Trends Cell Biol 2020; 30:452-466. [PMID: 32413315 DOI: 10.1016/j.tcb.2020.02.007] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/14/2020] [Accepted: 02/21/2020] [Indexed: 01/06/2023]
Abstract
Lysosomes are of major importance for the regulation of cellular cholesterol homeostasis. Food-derived cholesterol and cholesterol esters contained within lipoproteins are delivered to lysosomes by endocytosis. From the lysosomal lumen, cholesterol is transported to the inner surface of the lysosomal membrane through the glycocalyx; this shuttling requires Niemann-Pick C (NPC) 1 and NPC2 proteins. The lysosomal membrane proteins lysosomal-associated membrane protein (LAMP)-2 and lysosomal integral membrane protein (LIMP)-2/SCARB2 also bind cholesterol. LAMP-2 may serve as a cholesterol reservoir, whereas LIMP-2, like NPC1, is able to transport cholesterol through a transglycocalyx tunnel. Contact sites and fusion events between lysosomes and other organelles mediate the distribution of cholesterol. Lysosomal cholesterol content is sensed thereby regulating mammalian target of rapamycin complex (mTORC)-dependent signaling. This review summarizes our understanding of the major steps in cholesterol handling from the moment it enters the lysosome until it leaves this compartment.
Collapse
Affiliation(s)
- Ying Meng
- Department of Cell Biology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Saskia Heybrock
- Biochemisches Institut, Christian-Albrechts-Universität Kiel, Kiel, Germany
| | - Dante Neculai
- Department of Cell Biology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Paul Saftig
- Biochemisches Institut, Christian-Albrechts-Universität Kiel, Kiel, Germany.
| |
Collapse
|
32
|
Glucocerebrosidase: Functions in and Beyond the Lysosome. J Clin Med 2020; 9:jcm9030736. [PMID: 32182893 PMCID: PMC7141376 DOI: 10.3390/jcm9030736] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 02/07/2023] Open
Abstract
Glucocerebrosidase (GCase) is a retaining β-glucosidase with acid pH optimum metabolizing the glycosphingolipid glucosylceramide (GlcCer) to ceramide and glucose. Inherited deficiency of GCase causes the lysosomal storage disorder named Gaucher disease (GD). In GCase-deficient GD patients the accumulation of GlcCer in lysosomes of tissue macrophages is prominent. Based on the above, the key function of GCase as lysosomal hydrolase is well recognized, however it has become apparent that GCase fulfills in the human body at least one other key function beyond lysosomes. Crucially, GCase generates ceramides from GlcCer molecules in the outer part of the skin, a process essential for optimal skin barrier property and survival. This review covers the functions of GCase in and beyond lysosomes and also pays attention to the increasing insight in hitherto unexpected catalytic versatility of the enzyme.
Collapse
|
33
|
Are Glucosylceramide-Related Sphingolipids Involved in the Increased Risk for Cancer in Gaucher Disease Patients? Review and Hypotheses. Cancers (Basel) 2020; 12:cancers12020475. [PMID: 32085512 PMCID: PMC7072201 DOI: 10.3390/cancers12020475] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/31/2020] [Accepted: 02/14/2020] [Indexed: 01/19/2023] Open
Abstract
The roles of ceramide and its catabolites, i.e., sphingosine and sphingosine 1-phosphate, in the development of malignancies and the response to anticancer regimens have been extensively described. Moreover, an abundant literature points to the effects of glucosylceramide synthase, the mammalian enzyme that converts ceramide to β-glucosylceramide, in protecting tumor cells from chemotherapy. Much less is known about the contribution of β-glucosylceramide and its breakdown products in cancer progression. In this chapter, we first review published and personal clinical observations that report on the increased risk of developing cancers in patients affected with Gaucher disease, an inborn disorder characterized by defective lysosomal degradation of β-glucosylceramide. The previously described mechanistic links between lysosomal β-glucosylceramidase, β-glucosylceramide and/or β-glucosylphingosine, and various hallmarks of cancer are reviewed. We further show that melanoma tumor growth is facilitated in a Gaucher disease mouse model. Finally, the potential roles of the β-glucosylceramidase protein and its lipidic substrates and/or downstream products are discussed.
Collapse
|
34
|
Design, properties and applications of fluorinated and fluoroalkylated N-containing monosaccharides and their analogues. J Fluor Chem 2019. [DOI: 10.1016/j.jfluchem.2019.109364] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
35
|
Validation of anti-glucocerebrosidase antibodies for western blot analysis on protein lysates of murine and human cells. Biochem J 2019; 476:261-274. [DOI: 10.1042/bcj20180708] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 02/01/2023]
Abstract
Abstract
Gaucher disease (GD) is a rare lysosomal storage disorder caused by mutations in the GBA1 gene, encoding the lysosome-resident glucocerebrosidase enzyme involved in the hydrolysis of glucosylceramide. The discovery of an association between mutations in GBA1 and the development of synucleinopathies, including Parkinson disease, has directed attention to glucocerebrosidase as a potential therapeutic target for different synucleinopathies. These findings initiated an exponential growth in research and publications regarding the glucocerebrosidase enzyme. The use of various commercial and custom-made glucocerebrosidase antibodies has been reported, but standardized in-depth validation is still not available for many of these antibodies. This work details the evaluation of several previously reported glucocerebrosidase antibodies for western blot analysis, tested on protein lysates of murine gba+/+ and gba−/− immortalized neurons and primary human wild-type and type 2 GD fibroblasts.
Collapse
|
36
|
van der Lienden MJC, Gaspar P, Boot R, Aerts JMFG, van Eijk M. Glycoprotein Non-Metastatic Protein B: An Emerging Biomarker for Lysosomal Dysfunction in Macrophages. Int J Mol Sci 2018; 20:E66. [PMID: 30586924 PMCID: PMC6337583 DOI: 10.3390/ijms20010066] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/18/2022] Open
Abstract
Several diseases are caused by inherited defects in lysosomes, the so-called lysosomal storage disorders (LSDs). In some of these LSDs, tissue macrophages transform into prominent storage cells, as is the case in Gaucher disease. Here, macrophages become the characteristic Gaucher cells filled with lysosomes laden with glucosylceramide, because of their impaired enzymatic degradation. Biomarkers of Gaucher cells were actively searched, particularly after the development of costly therapies based on enzyme supplementation and substrate reduction. Proteins selectively expressed by storage macrophages and secreted into the circulation were identified, among which glycoprotein non-metastatic protein B (GPNMB). This review focusses on the emerging potential of GPNMB as a biomarker of stressed macrophages in LSDs as well as in acquired pathologies accompanied by an excessive lysosomal substrate load in macrophages.
Collapse
Affiliation(s)
| | - Paulo Gaspar
- Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands.
| | - Rolf Boot
- Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands.
| | - Johannes M F G Aerts
- Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands.
| | - Marco van Eijk
- Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands.
| |
Collapse
|
37
|
Ben Bdira F, Artola M, Overkleeft HS, Ubbink M, Aerts JMFG. Distinguishing the differences in β-glycosylceramidase folds, dynamics, and actions informs therapeutic uses. J Lipid Res 2018; 59:2262-2276. [PMID: 30279220 PMCID: PMC6277158 DOI: 10.1194/jlr.r086629] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 09/04/2018] [Indexed: 12/12/2022] Open
Abstract
Glycosyl hydrolases (GHs) are carbohydrate-active enzymes that hydrolyze a specific β-glycosidic bond in glycoconjugate substrates; β-glucosidases degrade glucosylceramide, a ubiquitous glycosphingolipid. GHs are grouped into structurally similar families that themselves can be grouped into clans. GH1, GH5, and GH30 glycosidases belong to clan A hydrolases with a catalytic (β/α)8 TIM barrel domain, whereas GH116 belongs to clan O with a catalytic (α/α)6 domain. In humans, GH abnormalities underlie metabolic diseases. The lysosomal enzyme glucocerebrosidase (family GH30), deficient in Gaucher disease and implicated in Parkinson disease etiology, and the cytosol-facing membrane-bound glucosylceramidase (family GH116) remove the terminal glucose from the ceramide lipid moiety. Here, we compare enzyme differences in fold, action, dynamics, and catalytic domain stabilization by binding site occupancy. We also explore other glycosidases with reported glycosylceramidase activity, including human cytosolic β-glucosidase, intestinal lactase-phlorizin hydrolase, and lysosomal galactosylceramidase. Last, we describe the successful translation of research to practice: recombinant glycosidases and glucosylceramide metabolism modulators are approved drug products (enzyme replacement therapies). Activity-based probes now facilitate the diagnosis of enzyme deficiency and screening for compounds that interact with the catalytic pocket of glycosidases. Future research may deepen the understanding of the functional variety of these enzymes and their therapeutic potential.
Collapse
Affiliation(s)
- Fredj Ben Bdira
- Departments of Macromolecular Biochemistry,Leiden Institute of Chemistry, Leiden, The Netherlands
| | - Marta Artola
- Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden, The Netherlands
| | - Herman S Overkleeft
- Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden, The Netherlands
| | - Marcellus Ubbink
- Departments of Macromolecular Biochemistry,Leiden Institute of Chemistry, Leiden, The Netherlands
| | | |
Collapse
|
38
|
Franco R, Sánchez-Arias JA, Navarro G, Lanciego JL. Glucocerebrosidase Mutations and Synucleinopathies. Potential Role of Sterylglucosides and Relevance of Studying Both GBA1 and GBA2 Genes. Front Neuroanat 2018; 12:52. [PMID: 30002620 PMCID: PMC6031742 DOI: 10.3389/fnana.2018.00052] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/31/2018] [Indexed: 12/21/2022] Open
Abstract
Gaucher's disease (GD) is the most prevalent lysosomal storage disorder. GD is caused by homozygous mutations of the GBA1 gene, which codes for beta-glucocerebrosidase (GCase). Although GD primarily affects peripheral tissues, the presence of neurological symptoms has been reported in several GD subtypes. GBA1 mutations have recently deserved increased attention upon the demonstration that both homo- and heterozygous GBA1 mutations represent the most important genetic risk factor for the appearance of synucleinopathies like Parkinson's disease (PD) and dementia with Lewy bodies (LBD). Although reduced GCase activity leads to alpha-synuclein aggregation, the mechanisms sustaining a role for GCase in alpha-synuclein homeostasis still remain largely unknown. Furthermore, the role to be played by impairment in the physiological function of endoplasmic reticulum, mitochondria and other subcellular membranous components is currently under investigation. Here we focus on the impact of GCase loss-of-function that impact on the levels of sterylglucosides, molecules that are known to trigger a PD-related synucleinopathy upon administration in rats. Moreover, the concurrence of another gene also coding for an enzyme with GCase activity (GBA2 gene) should also be taken into consideration, bearing in mind that in addition to a hydrolytic function, both GCases also share transglycosylation as a second catalytic activity. Accordingly, sterylglycoside levels should also be considered to further assess their impact on the neurodegenerative process. In this regard-and besides GBA1 genotyping-we suggest that screening for GBA2 mutations should be considered, together with analytical measurements of cholesterol glycosides in body fluids, as biomarkers for both PD risk and disease progression.
Collapse
Affiliation(s)
- Rafael Franco
- Department of Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan A Sánchez-Arias
- Department of Neuroscience, Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain
| | - Gemma Navarro
- Department of Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain.,Department of Biochemistry and Physiology, School of Pharmacy, University of Barcelona, Barcelona, Spain
| | - José L Lanciego
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain.,Department of Neuroscience, Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain.,Department of Neuroscience, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
39
|
Maruyama Y, Ueno S, Morita M, Hayashi F, Maekawa S. Inhibitory effect of several sphingolipid metabolites on calcineurin. Neurosci Lett 2018. [PMID: 29524645 DOI: 10.1016/j.neulet.2018.03.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Neurons have well-developed membrane microdomains called "rafts" that are recovered as a detergent-resistant membrane microdomain fraction (DRM). NAP-22 is one of the major protein components of neuronal DRM. In a previous study, we showed that DRM-derived NAP-22 binds ganglioside and the inhibitory effect of ganglioside to calcineurin (CaN), a neuron-enriched calmodulin-regulated phosphoprotein phosphatase. Considering the important roles of CaN in neurons, identification of other cellular regulators of CaN could be a good clue to understand the molecular background of neuronal function. In this study, we screened the effect of several membrane lipid-derived molecules on the CaN activity and found sphingosine and some sphingosine-derived metabolites such as sphingosylphosphorylcholine, galactosylsphingosine (psychosine), and glucosylsphingosine, have inhibitory effect on CaN through the interaction with calmodulin.
Collapse
Affiliation(s)
- Yoko Maruyama
- Department of Biology, Graduate School of Science, Kobe-University, Kobe 657-8501, Japan
| | - Satoko Ueno
- Department of Biology, Graduate School of Science, Kobe-University, Kobe 657-8501, Japan
| | - Mitsuhiro Morita
- Department of Biology, Graduate School of Science, Kobe-University, Kobe 657-8501, Japan
| | - Fumio Hayashi
- Department of Biology, Graduate School of Science, Kobe-University, Kobe 657-8501, Japan
| | - Shohei Maekawa
- Department of Biology, Graduate School of Science, Kobe-University, Kobe 657-8501, Japan.
| |
Collapse
|
40
|
García-Sanz P, Moratalla R. The importance of cholesterol in Parkinson's disease. Mov Disord 2018; 33:343-344. [PMID: 29315826 DOI: 10.1002/mds.27251] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 10/30/2017] [Indexed: 01/16/2023] Open
Affiliation(s)
- Patricia García-Sanz
- Instituto Cajal, Consejo Superior de Investigaciones Cientificas, CSIC, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Rosario Moratalla
- Instituto Cajal, Consejo Superior de Investigaciones Cientificas, CSIC, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| |
Collapse
|
41
|
Mashima R, Maekawa M. Lipid biomarkers for the peroxisomal and lysosomal disorders: their formation, metabolism and measurement. Biomark Med 2018; 12:83-95. [DOI: 10.2217/bmm-2017-0225] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Lipid biomarkers play important roles in the diagnosis of and monitoring of treatment in peroxisomal disorders and lysosomal storage disorders. Today, a variety of lipids, including very long chain fatty acids, glycolipids, bile acids and the oxidation products of cholesterol, have been considered as biomarkers for these disorders. In this brief review, the authors summarized the recent advances regarding these lipid biomarkers in terms of their formation, metabolism and measurement in these disorders. An understanding of these biomarkers will offer a key to the development of novel diagnoses and help create more effective therapies in the future.
Collapse
Affiliation(s)
- Ryuichi Mashima
- Department of Clinical Laboratory Medicine, National Center for Child Health & Development, 2–10–1 Okura, Setagaya-ku, Tokyo 157–8535, Japan
| | - Masamitsu Maekawa
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1–1 Seiryo-machi, Aoba-ku, Sendai 980–8574, Japan
| |
Collapse
|
42
|
García-Sanz P, Orgaz L, Bueno-Gil G, Espadas I, Rodríguez-Traver E, Kulisevsky J, Gutierrez A, Dávila JC, González-Polo RA, Fuentes JM, Mir P, Vicario C, Moratalla R. N370S-GBA1 mutation causes lysosomal cholesterol accumulation in Parkinson's disease. Mov Disord 2017; 32:1409-1422. [PMID: 28779532 DOI: 10.1002/mds.27119] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 06/29/2017] [Accepted: 06/29/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Heterozygous mutations in the GBA1 gene, which encodes the lysosomal enzyme β-glucocerebrosidase-1, increase the risk of developing Parkinson's disease, although the underlying mechanisms remain unclear. The aim of this study was to explore the impact of the N370S-GBA1 mutation on cellular homeostasis and vulnerability in a patient-specific cellular model of PD. METHODS We isolated fibroblasts from 4 PD patients carrying the N370S/wild type GBA1 mutation and 6 controls to study the autophagy-lysosome pathway, endoplasmic reticulum stress, and Golgi apparatus structure by Western blot, immunofluorescence, LysoTracker and Filipin stainings, mRNA analysis, and electron microscopy. We evaluated cell vulnerability by apoptosis, reactive oxygen species and mitochondrial membrane potential with flow cytometry. RESULTS The N370S mutation produced a significant reduction in β-glucocerebrosidase-1 protein and enzyme activity and β-glucocerebrosidase-1 retention within the endoplasmic reticulum, which interrupted its traffic to the lysosome. This led to endoplasmic reticulum stress activation and triggered unfolded protein response and Golgi apparatus fragmentation. Furthermore, these alterations resulted in autophagosome and p62/SQSTM1 accumulation. This impaired autophagy was a result of dysfunctional lysosomes, indicated by multilamellar body accumulation probably caused by increased cholesterol, enlarged lysosomal mass, and reduced enzyme activity. This phenotype impaired the removal of damaged mitochondria and reactive oxygen species production and enhanced cell death. CONCLUSIONS Our results support a connection between the loss of β-glucocerebrosidase-1 function, cholesterol accumulation, and the disruption of cellular homeostasis in GBA1-PD. Our work reveals new insights into the cellular pathways underlying PD pathogenesis, providing evidence that GBA1-PD shares common features with lipid-storage diseases. © 2017 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Lorena Orgaz
- Instituto Cajal, CSIC, Madrid, Spain.,CIBERNED, Madrid, Spain
| | | | - Isabel Espadas
- Instituto Cajal, CSIC, Madrid, Spain.,CIBERNED, Madrid, Spain
| | | | - Jaime Kulisevsky
- CIBERNED, Madrid, Spain.,Movement Disorders Unit, Neurology Dpt, Hospital Sant Pau (IIB-Sant Pau), Univ. Autònoma de Barcelona, Barcelona, Spain
| | - Antonia Gutierrez
- CIBERNED, Madrid, Spain.,Dpto. de Biología Celular, Genética y Fisiología, Facultad de Ciencias, IBIMA, Universidad de Málaga, Málaga, Spain
| | - José C Dávila
- CIBERNED, Madrid, Spain.,Dpto. de Biología Celular, Genética y Fisiología, Facultad de Ciencias, IBIMA, Universidad de Málaga, Málaga, Spain
| | - Rosa A González-Polo
- CIBERNED, Madrid, Spain.,Dpto. de Bioquímica, Biología Molecular y Genética F. Enfermería y T.O., Univ. de Extremadura, Cáceres, Spain
| | - José M Fuentes
- CIBERNED, Madrid, Spain.,Dpto. de Bioquímica, Biología Molecular y Genética F. Enfermería y T.O., Univ. de Extremadura, Cáceres, Spain
| | - Pablo Mir
- CIBERNED, Madrid, Spain.,Neurology Dpt, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Carlos Vicario
- Instituto Cajal, CSIC, Madrid, Spain.,CIBERNED, Madrid, Spain
| | | |
Collapse
|
43
|
A novel function for glucocerebrosidase as a regulator of sterylglucoside metabolism. Biochim Biophys Acta Gen Subj 2017; 1861:2507-2514. [PMID: 28596107 DOI: 10.1016/j.bbagen.2017.06.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 05/30/2017] [Accepted: 06/02/2017] [Indexed: 01/20/2023]
Abstract
BACKGROUND Sterols are major cell membrane lipids, and in many organisms they are modified with glucose to generate sterylglucosides. Glucosylation dramatically changes the functional properties of sterols. The formation of sterylglucosides from sterols in plants, fungi, and bacteria uses UDP-glucose as a glucose donor. By contrast, sterylglucoside biosynthesis in mammals is catalyzed by the transglucosylation activity of glucocerebrosidases, with glucosylceramide acting as the glucose donor. Recent success in isolation and structural determination of sterylglucosides in the vertebrate central nervous system shows that transglucosylation also occurs in vivo. These analyses also revealed that sterylglucoside aglycons are composed of several cholesterol-related metabolites, including a plant-type sitosteryl. SCOPE OF REVIEW In this review, we discuss the biological functions and metabolism of sterylglucosides. We also summarize new findings from studies on the metabolism of vertebrate sterylglucosides and review the circumstances underlying the recent discovery of sterylglucosides in vertebrate brain. Finally, we discuss the role of sterylglucosides in a variety of neurodegenerative disorders such as Gaucher disease and Parkinson's disease. MAJOR CONCLUSIONS The biological significance of UDP-glucose-independent sterol glucosylation is still unknown, but it is plausible that glucosylation may provide sterols with novel biological functions. Even though sterol glucosylation is a simple reaction, it can dramatically change the physical properties of sterols. GENERAL SIGNIFICANCE Sterylglucosides may play roles in various physiological processes and in the pathogenesis of different diseases. Arriving at a better understanding of them at the organ and cellular level may open up new approaches to developing therapeutics for a variety of diseases. This article is part of a Special Issue entitled Neuro-glycoscience, edited by Kenji Kadomatsu and Hiroshi Kitagawa.
Collapse
|
44
|
Akiyama H, Nakajima K, Itoh Y, Sayano T, Ohashi Y, Yamaguchi Y, Greimel P, Hirabayashi Y. Aglycon diversity of brain sterylglucosides: structure determination of cholesteryl- and sitosterylglucoside. J Lipid Res 2016; 57:2061-2072. [PMID: 27697915 DOI: 10.1194/jlr.m071480] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Indexed: 02/07/2023] Open
Abstract
To date, sterylglucosides have been reported to be present in various fungi, plants, and animals. In bacteria, such as Helicobacter pylori, proton NMR spectral analysis of isolated 1-O-cholesteryl-β-d-glucopyranoside (GlcChol) demonstrated the presence of an α-glucosidic linkage. By contrast, in animals, no detailed structural analysis of GlcChol has been reported, in part because animal-derived samples contain a high abundance of glucosylceramides (GlcCers)/galactosylceramides, which exhibit highly similar chromatographic behavior to GlcChol. A key step in vertebrate GlcChol biosynthesis is the transglucosylation reaction catalyzed by glucocerebrosidase (GBA)1 or GBA2, utilizing GlcCer as a glucose donor. These steps are expected to produce a β-glucosidic linkage. Impaired GBA1 and GBA2 function is associated with neurological disorders, such as cerebellar ataxia, spastic paraplegia, and Parkinson's disease. Utilizing a novel three-step chromatographic procedure, we prepared highly enriched GlcChol from embryonic chicken brain, allowing complete structural confirmation of the β-glucosidic linkage by 1H-NMR analysis. Unexpectedly, during purification, two additional sterylglucoside fractions were isolated. NMR and GC/MS analyses confirmed that the plant-type sitosterylglucoside in vertebrate brain is present throughout embryonic development. The aglycon structure of the remaining sterylglucoside (GSX-2) remains elusive due to its low abundance. Together, our results uncovered unexpected aglycon heterogeneity of sterylglucosides in vertebrate brain.
Collapse
Affiliation(s)
- Hisako Akiyama
- Laboratory for Molecular Membrane Neuroscience, RIKEN Brain Science Institute, RIKEN, Wako, Saitama 351-0198, Japan
| | - Kazuki Nakajima
- Laboratory for Molecular Membrane Neuroscience, RIKEN Brain Science Institute, RIKEN, Wako, Saitama 351-0198, Japan
| | | | - Tomoko Sayano
- Laboratory for Molecular Membrane Neuroscience, RIKEN Brain Science Institute, RIKEN, Wako, Saitama 351-0198, Japan
| | - Yoko Ohashi
- Laboratory for Molecular Membrane Neuroscience, RIKEN Brain Science Institute, RIKEN, Wako, Saitama 351-0198, Japan
| | - Yoshiki Yamaguchi
- Structural Glycobiology Team, Systems Glycobiology Research Group, RIKEN-Max Planck Joint Research Center, RIKEN, Wako, Saitama 351-0198, Japan
| | - Peter Greimel
- Lipid Biology Laboratory, RIKEN, Wako, Saitama 351-0198, Japan
| | - Yoshio Hirabayashi
- Laboratory for Molecular Membrane Neuroscience, RIKEN Brain Science Institute, RIKEN, Wako, Saitama 351-0198, Japan
| |
Collapse
|
45
|
Nakajima K, Akiyama H, Tanaka K, Kohyama-Koganeya A, Greimel P, Hirabayashi Y. Separation and analysis of mono-glucosylated lipids in brain and skin by hydrophilic interaction chromatography based on carbohydrate and lipid moiety. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1031:146-153. [DOI: 10.1016/j.jchromb.2016.07.047] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/23/2016] [Accepted: 07/26/2016] [Indexed: 10/21/2022]
|
46
|
Astudillo L, Therville N, Colacios C, Ségui B, Andrieu-Abadie N, Levade T. Glucosylceramidases and malignancies in mammals. Biochimie 2016; 125:267-80. [DOI: 10.1016/j.biochi.2015.11.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 11/09/2015] [Indexed: 01/11/2023]
|
47
|
Blanz J, Saftig P. Parkinson's disease: acid-glucocerebrosidase activity and alpha-synuclein clearance. J Neurochem 2016; 139 Suppl 1:198-215. [PMID: 26860955 DOI: 10.1111/jnc.13517] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/07/2015] [Accepted: 12/09/2015] [Indexed: 12/27/2022]
Abstract
The role of mutations in the gene GBA1 encoding the lysosomal hydrolase β-glucocerebrosidase for the development of synucleinopathies, such as Parkinson's disease and dementia with Lewy bodies, was only very recently uncovered. The knowledge obtained from the study of carriers or patients suffering from Gaucher disease (a common lysosomal storage disorder because of GBA1 mutations) is of particular importance for understanding the role of the enzyme and its catabolic pathway in the development of synucleinopathies. Decreased activity of β-glucocerebrosidase leads to lysosomal dysfunction and the accumulation of its substrate glucosylceramide and related lipid derivatives. Glucosylceramide is suggested to stabilize toxic oligomeric forms of α-synuclein that negatively influence the activity of β-glucocerebrosidase and to partially block export of newly synthesized β-glucocerebrosidase from the endoplasmic reticulum to late endocytic compartments, amplifying the pathological effects of α-synuclein and ultimately resulting in neuronal cell death. This pathogenic molecular feedback loop and most likely other factors (such as impaired endoplasmic reticulum-associated degradation, activation of the unfolded protein response and dysregulation of calcium homeostasis induced by misfolded GC mutants) are involved in shifting the cellular homeostasis from monomeric α-synuclein towards oligomeric neurotoxic and aggregated forms, which contribute to Parkinson's disease progression. From a therapeutic point of view, strategies aiming to increase either the expression, stability or delivery of the β-glucocerebrosidase to lysosomes are likely to decrease the α-synuclein burden and may be useful for an in depth evaluation at the organismal level. Lysosomes are critical for protein and lipid homeostasis. Recent research revealed that dysfunction of this organelle contributes to the development of neurodegenerative diseases such as Parkinson's disease (PD). Mutations in the lysosomal hydrolase β-glucocerebrosidase (GBA1) are a major risk factor for the development of PD and the molecular events linked to the reduced activity of GBA1 and the pathological accumulation of lipids and α-synuclein are just at the beginning to be understood. New therapeutic concepts in regards to how to increase the expression, stability, or delivery of β-glucocerebrosidase to lysosomes are currently developed. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Judith Blanz
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Paul Saftig
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany.
| |
Collapse
|
48
|
Marques ARA, Mirzaian M, Akiyama H, Wisse P, Ferraz MJ, Gaspar P, Ghauharali-van der Vlugt K, Meijer R, Giraldo P, Alfonso P, Irún P, Dahl M, Karlsson S, Pavlova EV, Cox TM, Scheij S, Verhoek M, Ottenhoff R, van Roomen CPAA, Pannu NS, van Eijk M, Dekker N, Boot RG, Overkleeft HS, Blommaart E, Hirabayashi Y, Aerts JM. Glucosylated cholesterol in mammalian cells and tissues: formation and degradation by multiple cellular β-glucosidases. J Lipid Res 2016; 57:451-63. [PMID: 26724485 PMCID: PMC4766994 DOI: 10.1194/jlr.m064923] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Indexed: 12/20/2022] Open
Abstract
The membrane lipid glucosylceramide (GlcCer) is continuously formed and degraded. Cells express two GlcCer-degrading β-glucosidases, glucocerebrosidase (GBA) and GBA2, located in and outside the lysosome, respectively. Here we demonstrate that through transglucosylation both GBA and GBA2 are able to catalyze in vitro the transfer of glucosyl-moieties from GlcCer to cholesterol, and vice versa. Furthermore, the natural occurrence of 1-O-cholesteryl-β-D-glucopyranoside (GlcChol) in mouse tissues and human plasma is demonstrated using LC-MS/MS and 13C6-labeled GlcChol as internal standard. In cells, the inhibition of GBA increases GlcChol, whereas inhibition of GBA2 decreases glucosylated sterol. Similarly, in GBA2-deficient mice, GlcChol is reduced. Depletion of GlcCer by inhibition of GlcCer synthase decreases GlcChol in cells and likewise in plasma of inhibitor-treated Gaucher disease patients. In tissues of mice with Niemann-Pick type C disease, a condition characterized by intralysosomal accumulation of cholesterol, marked elevations in GlcChol occur as well. When lysosomal accumulation of cholesterol is induced in cultured cells, GlcChol is formed via lysosomal GBA. This illustrates that reversible transglucosylation reactions are highly dependent on local availability of suitable acceptors. In conclusion, mammalian tissues contain GlcChol formed by transglucosylation through β-glucosidases using GlcCer as donor. Our findings reveal a novel metabolic function for GlcCer.
Collapse
Affiliation(s)
- André R A Marques
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Mina Mirzaian
- Departments of Medical Biochemistry Leiden Institute of Chemistry, Leiden, The Netherlands
| | | | - Patrick Wisse
- Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden, The Netherlands
| | - Maria J Ferraz
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Paulo Gaspar
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Rianne Meijer
- Departments of Medical Biochemistry Leiden Institute of Chemistry, Leiden, The Netherlands
| | - Pilar Giraldo
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad de Investigación Traslacional, Zaragoza, Spain
| | - Pilar Alfonso
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad de Investigación Traslacional, Zaragoza, Spain
| | - Pilar Irún
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Unidad de Investigación Traslacional, Zaragoza, Spain
| | - Maria Dahl
- Department of Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
| | - Stefan Karlsson
- Department of Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
| | - Elena V Pavlova
- Addenbrooke's Hospital, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Timothy M Cox
- Addenbrooke's Hospital, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Saskia Scheij
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Marri Verhoek
- Departments of Medical Biochemistry Leiden Institute of Chemistry, Leiden, The Netherlands
| | - Roelof Ottenhoff
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Navraj S Pannu
- Departments of Medical Biochemistry Leiden Institute of Chemistry, Leiden, The Netherlands
| | - Marco van Eijk
- Departments of Medical Biochemistry Leiden Institute of Chemistry, Leiden, The Netherlands
| | - Nick Dekker
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Rolf G Boot
- Departments of Medical Biochemistry Leiden Institute of Chemistry, Leiden, The Netherlands
| | - Herman S Overkleeft
- Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden, The Netherlands
| | - Edward Blommaart
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Johannes M Aerts
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands Departments of Medical Biochemistry Leiden Institute of Chemistry, Leiden, The Netherlands
| |
Collapse
|
49
|
Rella A, Mor V, Farnoud AM, Singh A, Shamseddine AA, Ivanova E, Carpino N, Montagna MT, Luberto C, Del Poeta M. Role of Sterylglucosidase 1 (Sgl1) on the pathogenicity of Cryptococcus neoformans: potential applications for vaccine development. Front Microbiol 2015; 6:836. [PMID: 26322039 PMCID: PMC4531891 DOI: 10.3389/fmicb.2015.00836] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 07/29/2015] [Indexed: 01/01/2023] Open
Abstract
Cryptococcosis caused by Cryptococcus neoformans and Cryptococcus gattii affects a large population and is a cause of significant morbidity and mortality. Despite its public health burden, there are currently no vaccines against cryptococcosis and new strategies against such infections are needed. In this study, we demonstrate that C. neoformans has the biochemical ability to metabolize sterylglucosides (SGs), a class of immunomodulatory glycolipids. Genetic manipulations that eliminate cryptococccal sterylglucosidase lead to the accumulation of SGs and generate a mutant strain (Δsgl1) that is non-pathogenic in the mouse models of cryptococcosis. Interestingly, this mutant strain acts as a vaccine strain and protects mice against cryptococcosis following infection with C. neoformans or C. gattii. The immunity induced by the Δsgl1 strain is not CD4+ T-cells dependent. Immunocompromised mice, which lack CD4+ T-cells, are able to control the infection by Δsgl1 and acquire immunity against the challenge by wild-type C. neoformans following vaccination with the Δsgl1 strain. These findings are particularly important in the context of HIV/AIDS immune deficiency and suggest that the Δsgl1 strain might provide a potential vaccination strategy against cryptococcosis.
Collapse
Affiliation(s)
- Antonella Rella
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook NY, USA
| | - Visesato Mor
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook NY, USA
| | - Amir M Farnoud
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook NY, USA
| | - Ashutosh Singh
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook NY, USA
| | | | - Elitza Ivanova
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook NY, USA
| | - Nicholas Carpino
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook NY, USA
| | - Maria T Montagna
- Department of Biomedical Science and Human Oncology, Hygiene Section, University of Bari Bari, Italy
| | - Chiara Luberto
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook NY, USA
| | - Maurizio Del Poeta
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook NY, USA
| |
Collapse
|
50
|
Dawson G. Measuring brain lipids. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1851:1026-39. [PMID: 25701718 PMCID: PMC4457555 DOI: 10.1016/j.bbalip.2015.02.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 02/05/2015] [Accepted: 02/08/2015] [Indexed: 12/27/2022]
Abstract
The rapid development of analytical technology has made lipidomics an exciting new area and this review will focus more on modern approaches to lipidomics than on earlier technology. Although not fully comprehensive for all possible brain lipids, the intent is to at least provide a reference for the analysis of classes of lipids found in brain and nervous tissue. We will discuss problems posed by the brain because of its structural and functional heterogeneity, the development changes it undergoes (myelination, aging, pathology etc.) and its cellular heterogeneity (neurons, glia etc.). Section 2 will discuss the various ways in which brain tissue can be extracted to yield lipids for analysis and section 3 will cover a wide range of techniques used to analyze brain lipids such as chromatography and mass-spectrometry. In Section 4 we will discuss ways of analyzing some of the specific biologically active brain lipids found in very small amounts except in pathological conditions and section 5 looks to the future of experimental lipidomic modification in the brain. This article is part of a Special Issue entitled Brain Lipids.
Collapse
Affiliation(s)
- Glyn Dawson
- Dept. Pediatrics, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|