1
|
Merrill AH. Don't Be Surprised When These Surprise You: Some Infrequently Studied Sphingoid Bases, Metabolites, and Factors That Should Be Kept in Mind During Sphingolipidomic Studies. Int J Mol Sci 2025; 26:650. [PMID: 39859363 PMCID: PMC11765627 DOI: 10.3390/ijms26020650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Sphingolipidomic mass spectrometry has provided valuable information-and surprises-about sphingolipid structures, metabolism, and functions in normal biological processes and disease. Nonetheless, many noteworthy compounds are not routinely determined, such as the following: most of the sphingoid bases that mammals biosynthesize de novo other than sphingosine (and sometimes sphinganine) or acquire from exogenous sources; infrequently considered metabolites of sphingoid bases, such as N-(methyl)n-derivatives; "ceramides" other than the most common N-acylsphingosines; and complex sphingolipids other than sphingomyelins and simple glycosphingolipids, including glucosyl- and galactosylceramides, which are usually reported as "monohexosylceramides". These and other subspecies are discussed, as well as some of the circumstances when they are likely to be seen (or present and missed) due to experimental conditions that can influence sphingolipid metabolism, uptake from the diet or from the microbiome, or as artifacts produced during extraction and analysis. If these compounds and factors are kept in mind during the design and interpretation of lipidomic studies, investigators are likely to be surprised by how often they appear and thereby advance knowledge about them.
Collapse
Affiliation(s)
- Alfred H Merrill
- School of Biological Sciences and The Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
2
|
Dubot P, Sabourdy F, Levade T. Human genetic defects of sphingolipid synthesis. J Inherit Metab Dis 2025; 48:e12745. [PMID: 38706107 PMCID: PMC11730260 DOI: 10.1002/jimd.12745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 05/07/2024]
Abstract
Sphingolipids are ubiquitous lipids, present in the membranes of all cell types, the stratum corneum and the circulating lipoproteins. Autosomal recessive as well as dominant diseases due to disturbed sphingolipid biosynthesis have been identified, including defects in the synthesis of ceramides, sphingomyelins and glycosphingolipids. In many instances, these gene variants result in the loss of catalytic function of the mutated enzymes. Additional gene defects implicate the subcellular localization of the sphingolipid-synthesizing enzyme, the regulation of its activity, or even the function of a sphingolipid-transporter protein. The resulting metabolic alterations lead to two major, non-exclusive types of clinical manifestations: a neurological disease, more or less rapidly progressive, associated or not with intellectual disability, and an ichthyotic-type skin disorder. These phenotypes highlight the critical importance of sphingolipids in brain and skin development and homeostasis. The present article reviews the clinical symptoms, genetic and biochemical alterations, pathophysiological mechanisms and therapeutic options of this relatively novel group of metabolic diseases.
Collapse
Affiliation(s)
- Patricia Dubot
- Unité Mixte de Recherche INSERM 1037, CNRS 5071, Université Toulouse III—Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse (CRCT)ToulouseFrance
- Laboratoire de BiochimieInstitut Fédératif de Biologie, CHU PurpanToulouseFrance
- Centre de RecherchesCHU Sainte‐Justine, Université de MontréalMontréalCanada
| | - Frédérique Sabourdy
- Unité Mixte de Recherche INSERM 1037, CNRS 5071, Université Toulouse III—Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse (CRCT)ToulouseFrance
- Laboratoire de BiochimieInstitut Fédératif de Biologie, CHU PurpanToulouseFrance
| | - Thierry Levade
- Unité Mixte de Recherche INSERM 1037, CNRS 5071, Université Toulouse III—Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse (CRCT)ToulouseFrance
- Laboratoire de BiochimieInstitut Fédératif de Biologie, CHU PurpanToulouseFrance
| |
Collapse
|
3
|
Srinivas N, Peiffer L, Horny K, Lei KC, Buus TB, Kubat L, Luo M, Yin M, Spassova I, Sucker A, Farahpour F, Kehrmann J, Ugurel S, Livingstone E, Gambichler T, Ødum N, Becker JC. Single-cell RNA and T-cell receptor sequencing unveil mycosis fungoides heterogeneity and a possible gene signature. Front Oncol 2024; 14:1408614. [PMID: 39169943 PMCID: PMC11337020 DOI: 10.3389/fonc.2024.1408614] [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: 03/28/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024] Open
Abstract
Background Mycosis fungoides (MF) is the most common subtype of cutaneous T-cell lymphoma (CTCL). Comprehensive analysis of MF cells in situ and ex vivo is complicated by the fact that is challenging to distinguish malignant from reactive T cells with certainty. Methods To overcome this limitation, we performed combined single-cell RNA (scRNAseq) and T-cell receptor TCR sequencing (scTCRseq) of skin lesions of cutaneous MF lesions from 12 patients. A sufficient quantity of living T cells was obtained from 9 patients, but 2 had to be excluded due to unclear diagnoses (coexisting CLL or revision to a fixed toxic drug eruption). Results From the remaining patients we established single-cell mRNA expression profiles and the corresponding TCR repertoire of 18,630 T cells. TCR clonality unequivocally identified 13,592 malignant T cells. Reactive T cells of all patients clustered together, while malignant cells of each patient formed a unique cluster expressing genes typical of naive/memory, such as CD27, CCR7 and IL7R, or cytotoxic T cells, e.g., GZMA, NKG7 and GNLY. Genes encoding classic CTCL markers were not detected in all clusters, consistent with the fact that mRNA expression does not correlate linearly with protein expression. Nevertheless, we successfully pinpointed distinctive gene signatures differentiating reactive malignant from malignant T cells: keratins (KRT81, KRT86), galectins (LGALS1, LGALS3) and S100 genes (S100A4, S100A6) being overexpressed in malignant cells. Conclusions Combined scRNAseq and scTCRseq not only allows unambiguous identification of MF cells, but also revealed marked heterogeneity between and within patients with unexpected functional phenotypes. While the correlation between mRNA and protein abundance was limited with respect to established MF markers, we were able to identify a single-cell gene expression signature that distinguishes malignant from reactive T cells.
Collapse
Affiliation(s)
- Nalini Srinivas
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Medicine Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, University Hospital Essen, and German Cancer Consortium (DKTK) partner site Essen/Düsseldorf, Essen, Germany
| | - Lukas Peiffer
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Medicine Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Kai Horny
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Medicine Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kuan Cheok Lei
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Medicine Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Terkild B. Buus
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Linda Kubat
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Medicine Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, University Hospital Essen, and German Cancer Consortium (DKTK) partner site Essen/Düsseldorf, Essen, Germany
| | - Meng Luo
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Medicine Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Menghong Yin
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Medicine Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ivelina Spassova
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Medicine Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, University Hospital Essen, and German Cancer Consortium (DKTK) partner site Essen/Düsseldorf, Essen, Germany
| | - Antje Sucker
- Department of Dermatology, University Hospital Essen, and German Cancer Consortium (DKTK) partner site Essen/Düsseldorf, Essen, Germany
| | - Farnoush Farahpour
- Bioinformatics and Computational Biophysics, University Duisburg-Essen, and Group of Molecular Cell Biology, Institute for Cell Biology (Cancer Research), University Hospital Essen, Essen, Germany
| | - Jan Kehrmann
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, and German Cancer Consortium (DKTK) partner site Essen/Düsseldorf, Essen, Germany
| | - Elisabeth Livingstone
- Department of Dermatology, University Hospital Essen, and German Cancer Consortium (DKTK) partner site Essen/Düsseldorf, Essen, Germany
| | - Thilo Gambichler
- Department of Dermatology, Ruhr-University Bochum, Bochum, Germany
- Department of Dermatology, Dortmund Hospital, University Witten/Herdecke, Dortmund, Germany
| | - Niels Ødum
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Jürgen C. Becker
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Medicine Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, University Hospital Essen, and German Cancer Consortium (DKTK) partner site Essen/Düsseldorf, Essen, Germany
| |
Collapse
|
4
|
Jamjoum R, Majumder S, Issleny B, Stiban J. Mysterious sphingolipids: metabolic interrelationships at the center of pathophysiology. Front Physiol 2024; 14:1229108. [PMID: 38235387 PMCID: PMC10791800 DOI: 10.3389/fphys.2023.1229108] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 11/27/2023] [Indexed: 01/19/2024] Open
Abstract
Metabolic pathways are complex and intertwined. Deficiencies in one or more enzymes in a given pathway are directly linked with genetic diseases, most of them having devastating manifestations. The metabolic pathways undertaken by sphingolipids are diverse and elaborate with ceramide species serving as the hubs of sphingolipid intermediary metabolism and function. Sphingolipids are bioactive lipids that serve a multitude of cellular functions. Being pleiotropic in function, deficiency or overproduction of certain sphingolipids is associated with many genetic and chronic diseases. In this up-to-date review article, we strive to gather recent scientific evidence about sphingolipid metabolism, its enzymes, and regulation. We shed light on the importance of sphingolipid metabolism in a variety of genetic diseases and in nervous and immune system ailments. This is a comprehensive review of the state of the field of sphingolipid biochemistry.
Collapse
Affiliation(s)
- Rama Jamjoum
- Department of Pharmacy, Birzeit University, West Bank, Palestine
| | - Saurav Majumder
- National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Rockville, MD, United States
| | - Batoul Issleny
- Department of Pharmacy, Birzeit University, West Bank, Palestine
| | - Johnny Stiban
- Department of Biology and Biochemistry, Birzeit University, West Bank, Palestine
| |
Collapse
|
5
|
Zhang X, Gong Z, Shen Y, Cai Z, Yang L, Zhang T, Li W, Zhao Y, Zhu S, Liu C, Wang J, Wang X, Qi R, Liu J, Lei X, Wang W, Jiang C, Fu Y, Kong W. Alkaline ceramidase 1-mediated platelet ceramide catabolism mitigates vascular inflammation and abdominal aortic aneurysm formation. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1173-1189. [PMID: 39196139 DOI: 10.1038/s44161-023-00364-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 10/12/2023] [Indexed: 08/29/2024]
Abstract
Abdominal aortic aneurysm (AAA) is a highly lethal vascular disease. The role of platelets in AAA remains incompletely understood. Here we show that platelet ceramides, rather than other phospholipids, were elevated in an angiotensin II (AngII)-induced AAA murine model and in patients with AAA by using targeted lipidomic analysis. Among key ceramide metabolism enzymes, alkaline ceramidase 1 (Acer1) hydrolyzing ceramides were exclusively downregulated in AAA platelets. Platelet-specific Acer1 knockout mice were more susceptible to AAA upon AngII infusion without affecting hemostasis and thrombosis. Mechanistically, Acer1 deficiency in platelets facilitated platelet pro-inflammatory cytokine secretion as well as P-selectin-mediated circulating platelet-leukocyte aggregation and infiltration in aortic walls via the ceramide-p38 MAPK signaling axis. Of note, AngII repressed Acer1 expression in platelets by decreasing HuR-dependent mRNA stability. In conclusion, Acer1-mediated ceramide degradation in platelets exhibited anti-inflammatory effects and ameliorated AAA formation, potentially serving as a therapeutic target for AAA and other inflammatory vascular diseases.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Ze Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yicong Shen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Zeyu Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Liu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Tao Zhang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, China
| | - Weihao Li
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, China
| | - Yang Zhao
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Shirong Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Cihang Liu
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jin Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Xian Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Ruomei Qi
- Key Laboratory of Geriatrics, Beijing Hospital and Beijing Institute of Geriatrics, Ministry of Health, Beijing, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoguang Lei
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Wengong Wang
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| |
Collapse
|
6
|
de Jonckheere B, Kollotzek F, Münzer P, Göb V, Fischer M, Mott K, Coman C, Troppmair NN, Manke MC, Zdanyte M, Harm T, Sigle M, Kopczynski D, Bileck A, Gerner C, Hoffmann N, Heinzmann D, Assinger A, Gawaz M, Stegner D, Schulze H, Borst O, Ahrends R. Critical shifts in lipid metabolism promote megakaryocyte differentiation and proplatelet formation. NATURE CARDIOVASCULAR RESEARCH 2023; 2:835-852. [PMID: 38075556 PMCID: PMC7615361 DOI: 10.1038/s44161-023-00325-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2024]
Abstract
During megakaryopoiesis, megakaryocytes (MK) undergo cellular morphological changes with strong modification of membrane composition and lipid signaling. Here we adopt a lipid-centric multiomics approach to create a quantitative map of the MK lipidome during maturation and proplatelet formation. Data reveal that MK differentiation is driven by an increased fatty acyl import and de novo lipid synthesis, resulting in an anionic membrane phenotype. Pharmacological perturbation of fatty acid import and phospholipid synthesis blocked membrane remodeling and directly reduced MK polyploidization and proplatelet formation resulting in thrombocytopenia. The anionic lipid shift during megakaryopoiesis was paralleled by lipid-dependent relocalization of the scaffold protein CKIP-1 and recruitment of the kinase CK2α to the plasma membrane, which seems to be essential for sufficient platelet biogenesis. Overall, this study provides a framework to understand how the MK lipidome is altered during maturation and the impact of MK membrane lipid remodeling on MK kinase signaling involved in thrombopoiesis.
Collapse
Affiliation(s)
- Bianca de Jonckheere
- Institute of Analytical Chemistry, University of Vienna, Austria
- Vienna Doctoral School in Chemistry, University of Vienna, Austria
| | - Ferdinand Kollotzek
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Germany
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Patrick Münzer
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Germany
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Vanessa Göb
- Institute for Experimental Biomedicine, University Hospital Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany
| | - Melina Fischer
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Germany
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Kristina Mott
- Institute for Experimental Biomedicine, University Hospital Würzburg, Germany
| | - Cristina Coman
- Institute of Analytical Chemistry, University of Vienna, Austria
| | - Nina Nicole Troppmair
- Institute of Analytical Chemistry, University of Vienna, Austria
- Vienna Doctoral School in Chemistry, University of Vienna, Austria
| | - Mailin-Christin Manke
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Germany
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Monika Zdanyte
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Germany
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Tobias Harm
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Manuel Sigle
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | | | - Andrea Bileck
- Institute of Analytical Chemistry, University of Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University of Vienna, Austria
| | - Christopher Gerner
- Institute of Analytical Chemistry, University of Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University of Vienna, Austria
| | - Nils Hoffmann
- Institute of Analytical Chemistry, University of Vienna, Austria
- Forschungszentrum Jülich GmbH, Institute for Bio- and Geosciences (IBG-5) Jülich, Germany
| | - David Heinzmann
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Alice Assinger
- Institute of Physiology, Centre of Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - David Stegner
- Institute for Experimental Biomedicine, University Hospital Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany
| | - Harald Schulze
- Institute for Experimental Biomedicine, University Hospital Würzburg, Germany
| | - Oliver Borst
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Germany
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Robert Ahrends
- Institute of Analytical Chemistry, University of Vienna, Austria
| |
Collapse
|
7
|
Zhao P, Zhuang Z, Guan X, Yang J, Wang W, Kuang Z. Crystal structure of the 3-ketodihydrosphingosine reductase TSC10 from Cryptococcus neoformans. Biochem Biophys Res Commun 2023; 670:73-78. [PMID: 37285720 DOI: 10.1016/j.bbrc.2023.05.109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 05/25/2023] [Indexed: 06/09/2023]
Abstract
The second step in the de novo sphingolipid biosynthesis is the reduction of 3-ketodihydrosphingosine by 3-ketodihydrosphingosine reductase (KDSR) to produce dihydrosphingosine (sphinganine). Fungal TSC10 and mammalian KDSR (also named FVT-1) proteins are the enzymes responsible for this process and they belong to the short-chain dehydrogenase/reductase (SDR) superfamily. Albeit that both fungal and mammalian 3-ketodihydrosphingosine reductases were identified more than a decade ago, no structure of these enzymes from any species has been experimentally determined. Here we report the crystal structure of the catalytic domain of TSC10 from Cryptococcus neoformans in complex with NADPH. cnTSC10 adopts a Rossmann fold with a central seven-stranded β-sheet flanked by α-helices on both sides. Several regions are disordered that include the segment connecting the serine and tyrosine residues of the catalytic triad, the so-called 'substrate loop', and the C-terminal region that often participates in homo-tetramerization in other SDRs. In addition, the cofactor NADPH is not fully ordered. These structural features indicate that the catalytic site of cnTSC10 possesses significant flexibility. cnTSC10 is predominantly dimeric in solution while a minor portion of the protein forms homo-tetramer. The crystal structure reveals that the homo-dimer interface involves both hydrophobic and hydrophilic interactions mediated by helices α4 and α5, as well as the loop connecting strand β4 and helix α4. Because residues forming hydrogen bonds and salt bridges in the dimer interface are not conserved between fungal TSC10 and mammalian KDSR proteins, it might be possible to develop inhibitors that selectively target fungal TSC10 dimerization.
Collapse
Affiliation(s)
- Panqi Zhao
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, 510632, China; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Guangzhou, 510632, China; National Engineering Research Center of Genetic Medicine, Guangzhou, 510632, China
| | - Zewen Zhuang
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, 510632, China; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Guangzhou, 510632, China; National Engineering Research Center of Genetic Medicine, Guangzhou, 510632, China
| | - Xueyan Guan
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, 510632, China; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Guangzhou, 510632, China; National Engineering Research Center of Genetic Medicine, Guangzhou, 510632, China
| | - Jinjin Yang
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, 510632, China; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Guangzhou, 510632, China; National Engineering Research Center of Genetic Medicine, Guangzhou, 510632, China
| | - Weiwei Wang
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhihe Kuang
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, 510632, China; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Guangzhou, 510632, China; National Engineering Research Center of Genetic Medicine, Guangzhou, 510632, China.
| |
Collapse
|
8
|
Wong GY, Hung ZGK, Ho KKH, Ling SC, Fung JLF, Lee M, Chau JFT, Ha SY, Chung BHY. Refractory thrombocytopenia and myelofibrosis in a novel KDSR mutation: Case report and literature review. Pediatr Blood Cancer 2023; 70:e30157. [PMID: 36546314 DOI: 10.1002/pbc.30157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/24/2022]
Affiliation(s)
- G Y Wong
- Department of Pediatrics and Adolescent Medicine, Hong Kong Children's Hospital, Kowloon City, Hong Kong
| | - Zita G K Hung
- Department of Pediatrics and Adolescent Medicine, Hong Kong Children's Hospital, Kowloon City, Hong Kong
| | - Karin K H Ho
- Department of Pediatrics and Adolescent Medicine, Princess Margaret Hospital, Lai Chi Kok, Hong Kong
| | - S C Ling
- Department of Pediatrics and Adolescent Medicine, Princess Margaret Hospital, Lai Chi Kok, Hong Kong
| | - Jasmine L F Fung
- Department of Pediatrics and Adolescent Medicine, University of Hong Kong, Hong Kong
| | - Mianne Lee
- Department of Pediatrics and Adolescent Medicine, University of Hong Kong, Hong Kong
| | - Jeffrey F T Chau
- Department of Pediatrics and Adolescent Medicine, University of Hong Kong, Hong Kong
| | - S Y Ha
- Department of Pediatrics and Adolescent Medicine, Hong Kong Children's Hospital, Kowloon City, Hong Kong
| | - Brian H Y Chung
- Department of Pediatrics and Adolescent Medicine, Hong Kong Children's Hospital, Kowloon City, Hong Kong.,Department of Pediatrics and Adolescent Medicine, University of Hong Kong, Hong Kong
| |
Collapse
|
9
|
GONG W, XU S, SONG Y, ZHOU Y, QIN X. Hepatic metabolomics combined with network pharmacology to reveal the correlation between the anti-depression effect and nourishing blood effect of Angelicae Sinensis Radix. Chin J Nat Med 2023; 21:197-213. [PMID: 37003642 DOI: 10.1016/s1875-5364(23)60421-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Indexed: 04/01/2023]
Abstract
Angelicae Sinensis Radix (AS) is reproted to exert anti-depression effect (ADE) and nourishing blood effect (NBE) in a rat model of depression. The correlation between the two therapeutic effects and its underlying mechanisms deserves further study. The current study is designed to explore the underlying mechanisms of correlation between the ADE and NBE of AS based on hepatic metabonomics, network pharmacology and molecular docking. According to metabolomics analysis, 30 metabolites involved in 11 metabolic pathways were identified as the potential metabolites for depression. Furthermore, principal component analysis and correlation analysis showed that glutathione, sphinganine, and ornithine were related to pharmacodynamics indicators including behavioral indicators and hematological indicators, indicating that metabolic pathways such as sphingolipid metabolism were involved in the ADE and NBE of AS. Then, a target-pathway network of depression and blood deficiency syndrome was constructed by network pharmacology analysis, where a total of 107 pathways were collected. Moreover, 37 active components obtained from Ultra Performance Liquid Chromatography-Triple-Time of Flight Mass Spectrometer (UPLC-Triple-TOF/MS) in AS extract that passed the filtering criteria were used for network pharmacology, where 46 targets were associated with the ADE and NBE of AS. Pathway enrichment analysis further indicated the involvement of sphingolipid metabolism in the ADE and NBE of AS. Molecular docking analysis indciated that E-ligustilide in AS extract exhibited strong binding activity with target proteins (PIK3CA and PIK3CD) in sphingolipid metabolism. Further analysis by Western blot verified that AS regulated the expression of PIK3CA and PIK3CD on sphingolipid metabolism. Our results demonstrated that sphingolipid metabolic pathway was the core mechanism of the correlation between the ADE and NBE of AS.
Collapse
|
10
|
Wijsmans D, Spanoudi-Kitrimi I. Variable skin findings in two siblings with KDSR mutations manifesting in PERIOPTER syndrome. Pediatr Dermatol 2022; 40:330-332. [PMID: 36263748 DOI: 10.1111/pde.15155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 09/10/2022] [Indexed: 11/28/2022]
Abstract
Pathogenic variants in the KDSR gene give rise to a Mendelian disorder called PERIOPTER syndrome. The disease is caused by a disruption in ceramide synthesis, with an impact on both skin and bone marrow. Patients with PERIOPTER syndrome show intermittent thrombocytopenia and/or associated anemia as well as disorders of keratinization. We present two siblings with identical compound heterozygous pathogenic variants in the KDSR gene, but with different clinical manifestations.
Collapse
Affiliation(s)
- Daphne Wijsmans
- Department of Dermatology, University Hospital Leuven, Leuven, Belgium
| | | |
Collapse
|
11
|
Bañuelos MM, Zavaleta YJA, Roldan A, Reyes RJ, Guardado M, Chavez Rojas B, Nyein T, Rodriguez Vega A, Santos M, Huerta-Sanchez E, Rohlfs RV. Associations between forensic loci and expression levels of neighboring genes may compromise medical privacy. Proc Natl Acad Sci U S A 2022; 119:e2121024119. [PMID: 36166477 PMCID: PMC9546536 DOI: 10.1073/pnas.2121024119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 08/29/2022] [Indexed: 11/18/2022] Open
Abstract
A set of 20 short tandem repeats (STRs) is used by the US criminal justice system to identify suspects and to maintain a database of genetic profiles for individuals who have been previously convicted or arrested. Some of these STRs were identified in the 1990s, with a preference for markers in putative gene deserts to avoid forensic profiles revealing protected medical information. We revisit that assumption, investigating whether forensic genetic profiles reveal information about gene-expression variation or potential medical information. We find six significant correlations (false discovery rate = 0.23) between the forensic STRs and the expression levels of neighboring genes in lymphoblastoid cell lines. We explore possible mechanisms for these associations, showing evidence compatible with forensic STRs causing expression variation or being in linkage disequilibrium with a causal locus in three cases and weaker or potentially spurious associations in the other three cases. Together, these results suggest that forensic genetic loci may reveal expression levels and, perhaps, medical information.
Collapse
Affiliation(s)
- Mayra M. Bañuelos
- Department of Mathematics, San Francisco State University, San Francisco, CA 94132
- Ecology, Evolution and Organismal Biology, Brown University, Providence, RI 02912
- Center for Computational and Molecular Biology, Brown University, Providence, RI 02912
| | | | - Alennie Roldan
- Department of Biology, San Francisco State University, San Francisco, CA 94132
| | - Rochelle-Jan Reyes
- Department of Biology, San Francisco State University, San Francisco, CA 94132
| | - Miguel Guardado
- Department of Mathematics, San Francisco State University, San Francisco, CA 94132
| | | | - Thet Nyein
- Department of Mathematics, San Francisco State University, San Francisco, CA 94132
| | - Ana Rodriguez Vega
- Department of Biology, San Francisco State University, San Francisco, CA 94132
| | - Maribel Santos
- Department of Biology, San Francisco State University, San Francisco, CA 94132
| | - Emilia Huerta-Sanchez
- Ecology, Evolution and Organismal Biology, Brown University, Providence, RI 02912
- Center for Computational and Molecular Biology, Brown University, Providence, RI 02912
| | - Rori V. Rohlfs
- Department of Biology, San Francisco State University, San Francisco, CA 94132
| |
Collapse
|
12
|
Spears ME, Lee N, Hwang S, Park SJ, Carlisle AE, Li R, Doshi MB, Armando AM, Gao J, Simin K, Zhu LJ, Greer PL, Quehenberger O, Torres EM, Kim D. De novo sphingolipid biosynthesis necessitates detoxification in cancer cells. Cell Rep 2022; 40:111415. [PMID: 36170811 PMCID: PMC9552870 DOI: 10.1016/j.celrep.2022.111415] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 07/21/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
Sphingolipids play important signaling and structural roles in cells. Here, we find that during de novo sphingolipid biosynthesis, a toxic metabolite is formed with critical implications for cancer cell survival. The enzyme catalyzing the first step in this pathway, serine palmitoyltransferase complex (SPT), is upregulated in breast and other cancers. SPT is dispensable for cancer cell proliferation, as sphingolipids can be salvaged from the environment. However, SPT activity introduces a liability as its product, 3-ketodihydrosphingosine (3KDS), is toxic and requires clearance via the downstream enzyme 3-ketodihydrosphingosine reductase (KDSR). In cancer cells, but not normal cells, targeting KDSR induces toxic 3KDS accumulation leading to endoplasmic reticulum (ER) dysfunction and loss of proteostasis. Furthermore, the antitumor effect of KDSR disruption can be enhanced by increasing metabolic input (via high-fat diet) to allow greater 3KDS production. Thus, de novo sphingolipid biosynthesis entails a detoxification requirement in cancer cells that can be therapeutically exploited.
Collapse
Affiliation(s)
- Meghan E Spears
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Namgyu Lee
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Sunyoung Hwang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Sung Jin Park
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Anne E Carlisle
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Rui Li
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Mihir B Doshi
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Aaron M Armando
- School of Medicine, Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Jenny Gao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Karl Simin
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Lihua Julie Zhu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Paul L Greer
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Oswald Quehenberger
- School of Medicine, Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Eduardo M Torres
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Dohoon Kim
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA.
| |
Collapse
|
13
|
Xiong L, Wang L, Zhang T, Ye X, Huang F, Huang Q, Huang X, Wu J, Zeng J. UHPLC/MS-Based Serum Metabolomics Reveals the Mechanism of Radiation-Induced Thrombocytopenia in Mice. Int J Mol Sci 2022; 23:7978. [PMID: 35887324 PMCID: PMC9319504 DOI: 10.3390/ijms23147978] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/16/2022] [Accepted: 07/18/2022] [Indexed: 11/16/2022] Open
Abstract
Radiation-induced thrombocytopenia is a common and life-threatening side effect of ionizing radiation (IR) therapy. However, the underlying pathological mechanisms remain unclear. In the present study, irradiation was demonstrated to significantly reduce platelet levels, inhibit megakaryocyte differentiation, and promote the apoptosis of bone marrow (BM) cells. A metabolomics approach and a UHPLC-QTOF MS system were subsequently employed for the comprehensive analysis of serum metabolic profiles of normal and irradiated mice. A total of 66 metabolites were significantly altered, of which 56 were up-regulated and 10 were down-regulated in irradiated mice compared to normal mice on day 11 after irradiation. Pathway analysis revealed that disorders in glycerophospholipid metabolism, nicotinate and nicotinamide metabolism, sphingolipid metabolism, inositol phosphate metabolism, and tryptophan metabolism were involved in radiation-induced thrombocytopenia. In addition, three important differential metabolites, namely L-tryptophan, LysoPC (17:0), and D-sphinganine, which were up-regulated in irradiated mice, significantly induced the apoptosis of K562 cells. L-tryptophan inhibited megakaryocyte differentiation of K562 cells. Finally, serum metabolomics was performed on day 30 (i.e., when the platelet levels in irradiated mice recovered to normal levels). The contents of L-tryptophan, LysoPC (17:0), and D-sphinganine in normal and irradiated mice did not significantly differ on day 30 after irradiation. In conclusion, radiation can cause metabolic disorders, which are highly correlated with the apoptosis of hematopoietic cells and inhibition of megakaryocyte differentiation, ultimately resulting in thrombocytopenia. Further, the metabolites, L-tryptophan, LysoPC (17:0), and D-sphinganine can serve as biomarkers for radiation-induced thrombocytopenia.
Collapse
Affiliation(s)
- Ling Xiong
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (L.X.); (L.W.); (T.Z.); (X.Y.); (F.H.); (Q.H.); (X.H.)
| | - Long Wang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (L.X.); (L.W.); (T.Z.); (X.Y.); (F.H.); (Q.H.); (X.H.)
| | - Ting Zhang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (L.X.); (L.W.); (T.Z.); (X.Y.); (F.H.); (Q.H.); (X.H.)
| | - Xinyuan Ye
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (L.X.); (L.W.); (T.Z.); (X.Y.); (F.H.); (Q.H.); (X.H.)
| | - Feihong Huang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (L.X.); (L.W.); (T.Z.); (X.Y.); (F.H.); (Q.H.); (X.H.)
- Education Ministry Key Laboratory of Medical Electrophysiology, Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou 646000, China
| | - Qianqian Huang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (L.X.); (L.W.); (T.Z.); (X.Y.); (F.H.); (Q.H.); (X.H.)
- Education Ministry Key Laboratory of Medical Electrophysiology, Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou 646000, China
| | - Xinwu Huang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (L.X.); (L.W.); (T.Z.); (X.Y.); (F.H.); (Q.H.); (X.H.)
- Education Ministry Key Laboratory of Medical Electrophysiology, Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou 646000, China
| | - Jianming Wu
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (L.X.); (L.W.); (T.Z.); (X.Y.); (F.H.); (Q.H.); (X.H.)
- Education Ministry Key Laboratory of Medical Electrophysiology, Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou 646000, China
| | - Jing Zeng
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China; (L.X.); (L.W.); (T.Z.); (X.Y.); (F.H.); (Q.H.); (X.H.)
| |
Collapse
|
14
|
Warren JT, Di Paola J. Genetics of inherited thrombocytopenias. Blood 2022; 139:3264-3277. [PMID: 35167650 PMCID: PMC9164741 DOI: 10.1182/blood.2020009300] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 02/04/2022] [Indexed: 01/19/2023] Open
Abstract
The inherited thrombocytopenia syndromes are a group of disorders characterized primarily by quantitative defects in platelet number, though with a variety demonstrating qualitative defects and/or extrahematopoietic findings. Through collaborative international efforts applying next-generation sequencing approaches, the list of genetic syndromes that cause thrombocytopenia has expanded significantly in recent years, now with over 40 genes implicated. In this review, we focus on what is known about the genetic etiology of inherited thrombocytopenia syndromes and how the field has worked to validate new genetic discoveries. We highlight the important role for the clinician in identifying a germline genetic diagnosis and strategies for identifying novel causes through research-based endeavors.
Collapse
Affiliation(s)
- Julia T Warren
- Division of Hematology-Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Jorge Di Paola
- Division of Hematology-Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
15
|
Nachmias B, Khan DH, Voisin V, Mer AS, Thomas GE, Segev N, St-Germain J, Hurren R, Gronda M, Botham A, Wang X, Maclean N, Seneviratne AK, Duong N, Xu C, Arruda A, Orouji E, Algouneh A, Hakem R, Shlush L, Minden MD, Raught B, Bader GD, Schimmer AD. IPO11 regulates the nuclear import of BZW1/2 and is necessary for AML cells and stem cells. Leukemia 2022; 36:1283-1295. [PMID: 35152270 PMCID: PMC9061300 DOI: 10.1038/s41375-022-01513-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 01/06/2022] [Accepted: 01/26/2022] [Indexed: 12/24/2022]
Abstract
AML cells are arranged in a hierarchy with stem/progenitor cells giving rise to more differentiated bulk cells. Despite the importance of stem/progenitors in the pathogenesis of AML, the determinants of the AML stem/progenitor state are not fully understood. Through a comparison of genes that are significant for growth and viability of AML cells by way of a CRISPR screen, with genes that are differentially expressed in leukemia stem cells (LSC), we identified importin 11 (IPO11) as a novel target in AML. Importin 11 (IPO11) is a member of the importin β family of proteins that mediate transport of proteins across the nuclear membrane. In AML, knockdown of IPO11 decreased growth, reduced engraftment potential of LSC, and induced differentiation. Mechanistically, we identified the transcription factors BZW1 and BZW2 as novel cargo of IPO11. We further show that BZW1/2 mediate a transcriptional signature that promotes stemness and survival of LSC. Thus, we demonstrate for the first time how specific cytoplasmic-nuclear regulation supports stem-like transcriptional signature in relapsed AML.
Collapse
|
16
|
3-Ketodihydrosphingosine reductase maintains ER homeostasis and unfolded protein response in leukemia. Leukemia 2022; 36:100-110. [PMID: 34373586 PMCID: PMC8732298 DOI: 10.1038/s41375-021-01378-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/24/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023]
Abstract
Sphingolipids and their metabolic pathways have been implicated in disease development and therapeutic response; however, the detailed mechanisms remain unclear. Using a sphingolipid network focused CRISPR/Cas9 library screen, we identified an endoplasmic reticulum (ER) enzyme, 3-Ketodihydrosphingosine reductase (KDSR), to be essential for leukemia cell maintenance. Loss of KDSR led to apoptosis, cell cycle arrest, and aberrant ER structure. Transcriptomic analysis revealed the indispensable role of KDSR in maintaining the unfolded protein response (UPR) in ER. High-density CRISPR tiling scan and sphingolipid mass spectrometry pinpointed the critical role of KDSR's catalytic function in leukemia. Mechanistically, depletion of KDSR resulted in accumulated 3-ketodihydrosphingosine (KDS) and dysregulated UPR checkpoint proteins PERK, ATF6, and ATF4. Finally, our study revealed the synergism between KDSR suppression and pharmacologically induced ER-stress, underscoring a therapeutic potential of combinatorial targeting sphingolipid metabolism and ER homeostasis in leukemia treatment.
Collapse
|
17
|
Wu L, Zhang Y, Zi J, Yan Y, Yu L, Lin D, Huang L, Lai X, Liao X, Yang L. Case report: Compound heterozygous mutations in the KDSR gene cause progressive keratodermia and thrombocytopenia. Front Pediatr 2022; 10:940618. [PMID: 35958175 PMCID: PMC9360485 DOI: 10.3389/fped.2022.940618] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/24/2022] [Indexed: 11/23/2022] Open
Abstract
KDSR (3-ketodihydrosphingosine reductase) is a short-chain dehydrogenase located in the endoplasmic reticulum. Mutations in KDSR cause defects in ceramides, which play a key role in the biological processes of the skin and other tissues. Herein, we report a case of compound heterozygous mutations in KDSR that caused progressive keratodermia and thrombocytopenia in a 2-year-old male patient.
Collapse
Affiliation(s)
- Li Wu
- Department of Pediatric Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yajie Zhang
- Department of Pediatric Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Juan Zi
- Department of Pediatric Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yinyan Yan
- Department of Pediatric Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lihua Yu
- Department of Pediatric Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Danna Lin
- Department of Pediatric Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lulu Huang
- Department of Pediatric Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaorong Lai
- Department of Pediatric Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xu Liao
- Department of Pediatric Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lihua Yang
- Department of Pediatric Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
18
|
Pilz R, Opálka L, Majcher A, Grimm E, Van Maldergem L, Mihalceanu S, Schäkel K, Enk A, Aubin F, Bursztejn AC, Brischoux-Boucher E, Fischer J, Sandhoff R. Formation of keto-type ceramides in palmoplantar keratoderma based on biallelic KDSR mutations in patients. Hum Mol Genet 2021; 31:1105-1114. [PMID: 34686882 DOI: 10.1093/hmg/ddab309] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 10/14/2021] [Accepted: 10/18/2021] [Indexed: 11/14/2022] Open
Abstract
Functional skin barrier requires sphingolipid homeostasis. 3-ketodihydrosphingosine reductase or KDSR is a key enzyme of sphingolipid anabolism catalyzing the reduction of 3-ketodihydrosphingosine to sphinganine. Biallelic mutations in the KDSR gene may cause erythrokeratoderma variabilis et progressive-4, later specified as PERIOPTER syndrome, emphasizing a characteristic periorifical and ptychotropic erythrokeratoderma. We report another patient with compound heterozygous mutations in KDSR, born with generalized harlequin ichthyosis, which progressed into palmoplantar keratoderma. To determine whether patient-associated KDSR mutations lead to KDSR substrate accumulation and/or unrecognized sphingolipid downstream products in stratum corneum we analyzed lipids of this and previously published patients with non-identical biallelic mutations in KDSR. In stratum corneum of both patients we identified hitherto unobserved skin ceramides with an unusual keto-type sphingoid base in lesional and non-lesional areas, which accounted for up to 10% of the measured ceramide species. Furthermore, an overall shorter mean chain length of free and bound sphingoid bases was observed-shorter mean chain length of free sphingoid bases was also observed in lesional psoriasis vulgaris SC, but not generally in lesional atopic dermatitis SC. Formation of keto-type ceramides is probably due to a bottle neck in metabolic flux through KDSR and a bypass by ceramide synthases, which highlights the importance of tight intermediate regulation during sphingolipid anabolism and reveals substrate deprivation as potential therapy.
Collapse
Affiliation(s)
- Robert Pilz
- Lipid Pathobiochemistry Group, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Lukáš Opálka
- Lipid Pathobiochemistry Group, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,Skin Barrier Research Group, Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, 500 05 Hradec Králové, Czech Republic
| | - Adam Majcher
- Lipid Pathobiochemistry Group, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,Skin Barrier Research Group, Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, 500 05 Hradec Králové, Czech Republic
| | - Elisabeth Grimm
- Lipid Pathobiochemistry Group, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Lionel Van Maldergem
- Centre de Génétique Humaine, Université de Franche-Comté, 25000, Besançon, France.,Clinical Investigation Center 1431, National Institute of Health and Medical Research (INSERM), University Hospital, 25000, Besançon, France
| | - Silvia Mihalceanu
- Department of Dermatology, Medical Faculty of the University of Heidelberg, 69120, Heidelberg, Germany
| | - Knut Schäkel
- Department of Dermatology, Medical Faculty of the University of Heidelberg, 69120, Heidelberg, Germany
| | - Alexander Enk
- Department of Dermatology, Medical Faculty of the University of Heidelberg, 69120, Heidelberg, Germany
| | - François Aubin
- Service de Dermatologie et INSERM 1098 RIGHT, CHU et UFR Santé, 25000, Besançon France
| | | | | | - Judith Fischer
- Institute of Human Genetics, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg im Breisgau, Germany
| | - Roger Sandhoff
- Lipid Pathobiochemistry Group, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| |
Collapse
|
19
|
Sphingolipids in Hematopoiesis: Exploring Their Role in Lineage Commitment. Cells 2021; 10:cells10102507. [PMID: 34685487 PMCID: PMC8534120 DOI: 10.3390/cells10102507] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/30/2021] [Accepted: 09/18/2021] [Indexed: 11/17/2022] Open
Abstract
Sphingolipids, associated enzymes, and the sphingolipid pathway are implicated in complex, multifaceted roles impacting several cell functions, such as cellular homeostasis, apoptosis, cell differentiation, and more through intrinsic and autocrine/paracrine mechanisms. Given this broad range of functions, it comes as no surprise that a large body of evidence points to important functions of sphingolipids in hematopoiesis. As the understanding of the processes that regulate hematopoiesis and of the specific characteristics that define each type of hematopoietic cells is being continuously refined, the understanding of the roles of sphingolipid metabolism in hematopoietic lineage commitment is also evolving. Recent findings indicate that sphingolipid alterations can modulate lineage commitment from stem cells all the way to megakaryocytic, erythroid, myeloid, and lymphoid cells. For instance, recent evidence points to the ability of de novo sphingolipids to regulate the stemness of hematopoietic stem cells while a substantial body of literature implicates various sphingolipids in specialized terminal differentiation, such as thrombopoiesis. This review provides a comprehensive discussion focused on the mechanisms that link sphingolipids to the commitment of hematopoietic cells to the different lineages, also highlighting yet to be resolved questions.
Collapse
|
20
|
Altawil L, Alshihry H, Alfaraidi H, Alhashem A, Alhumidi A, Alkuraya FS. Progressive symmetrical erythrokeratoderma manifesting as harlequin-like ichthyosis with severe thrombocytopenia secondary to a homozygous 3-ketodihydrosphingosine reductase mutation. JAAD Case Rep 2021; 14:55-58. [PMID: 34277909 PMCID: PMC8261470 DOI: 10.1016/j.jdcr.2021.06.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Lama Altawil
- Department of Dermatology and Dermatologic Surgery, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Hind Alshihry
- Department of Dermatology and Dermatologic Surgery, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Huda Alfaraidi
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Amal Alhashem
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Ahmed Alhumidi
- Department of Pathology, King Saud University, Riyadh, Saudi Arabia
| | - Fowzan S. Alkuraya
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
21
|
Inherited Platelet Disorders: An Updated Overview. Int J Mol Sci 2021; 22:ijms22094521. [PMID: 33926054 PMCID: PMC8123627 DOI: 10.3390/ijms22094521] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/17/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Platelets play a major role in hemostasis as ppwell as in many other physiological and pathological processes. Accordingly, production of about 1011 platelet per day as well as appropriate survival and functions are life essential events. Inherited platelet disorders (IPDs), affecting either platelet count or platelet functions, comprise a heterogenous group of about sixty rare diseases caused by molecular anomalies in many culprit genes. Their clinical relevance is highly variable according to the specific disease and even within the same type, ranging from almost negligible to life-threatening. Mucocutaneous bleeding diathesis (epistaxis, gum bleeding, purpura, menorrhagia), but also multisystemic disorders and/or malignancy comprise the clinical spectrum of IPDs. The early and accurate diagnosis of IPDs and a close patient medical follow-up is of great importance. A genotype-phenotype relationship in many IPDs makes a molecular diagnosis especially relevant to proper clinical management. Genetic diagnosis of IPDs has been greatly facilitated by the introduction of high throughput sequencing (HTS) techniques into mainstream investigation practice in these diseases. However, there are still unsolved ethical concerns on general genetic investigations. Patients should be informed and comprehend the potential implications of their genetic analysis. Unlike the progress in diagnosis, there have been no major advances in the clinical management of IPDs. Educational and preventive measures, few hemostatic drugs, platelet transfusions, thrombopoietin receptor agonists, and in life-threatening IPDs, allogeneic hematopoietic stem cell transplantation are therapeutic possibilities. Gene therapy may be a future option. Regular follow-up by a specialized hematology service with multidisciplinary support especially for syndromic IPDs is mandatory.
Collapse
|
22
|
Learning the Ropes of Platelet Count Regulation: Inherited Thrombocytopenias. J Clin Med 2021; 10:jcm10030533. [PMID: 33540538 PMCID: PMC7867147 DOI: 10.3390/jcm10030533] [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: 12/30/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 02/06/2023] Open
Abstract
Inherited thrombocytopenias (IT) are a group of hereditary disorders characterized by a reduced platelet count sometimes associated with abnormal platelet function, which can lead to bleeding but also to syndromic manifestations and predispositions to other disorders. Currently at least 41 disorders caused by mutations in 42 different genes have been described. The pathogenic mechanisms of many forms of IT have been identified as well as the gene variants implicated in megakaryocyte maturation or platelet formation and clearance, while for several of them the pathogenic mechanism is still unknown. A range of therapeutic approaches are now available to improve survival and quality of life of patients with IT; it is thus important to recognize an IT and establish a precise diagnosis. ITs may be difficult to diagnose and an initial accurate clinical evaluation is mandatory. A combination of clinical and traditional laboratory approaches together with advanced sequencing techniques provide the highest rate of diagnostic success. Despite advancement in the diagnosis of IT, around 50% of patients still do not receive a diagnosis, therefore further research in the field of ITs is warranted to further improve patient care.
Collapse
|
23
|
Pecci A, Balduini CL. Inherited thrombocytopenias: an updated guide for clinicians. Blood Rev 2020; 48:100784. [PMID: 33317862 DOI: 10.1016/j.blre.2020.100784] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/05/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023]
Abstract
The great advances in the knowledge of inherited thrombocytopenias (ITs) made since the turn of the century have significantly changed our view of these conditions. To date, ITs encompass 45 disorders with different degrees of complexity of the clinical picture and very wide variability in the prognosis. They include forms characterized by thrombocytopenia alone, forms that present with other congenital defects, and conditions that predispose to acquire additional diseases over the course of life. In this review, we recapitulate the clinical features of ITs with emphasis on the forms predisposing to additional diseases. We then discuss the key issues for a rational approach to the diagnosis of ITs in clinical practice. Finally, we aim to provide an updated and comprehensive guide to the treatment of ITs, including the management of hemostatic challenges, the treatment of severe forms, and the approach to the manifestations that add to thrombocytopenia.
Collapse
Affiliation(s)
- Alessandro Pecci
- Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation and University of Pavia, Pavia, Italy.
| | | |
Collapse
|
24
|
Downes K, Borry P, Ericson K, Gomez K, Greinacher A, Lambert M, Leinoe E, Noris P, Van Geet C, Freson K. Clinical management, ethics and informed consent related to multi-gene panel-based high throughput sequencing testing for platelet disorders: Communication from the SSC of the ISTH. J Thromb Haemost 2020; 18:2751-2758. [PMID: 33079472 PMCID: PMC7589386 DOI: 10.1111/jth.14993] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 12/22/2022]
Abstract
Molecular diagnostics of inherited platelet disorders (IPD) has been revolutionized by the implementation of high-throughput sequencing (HTS) approaches. A conclusive diagnosis using HTS tests can be obtained quickly and cost-effectively in many, but not all patients. The expanding use of HTS tests has raised concerns regarding complex variant interpretation and the ethical implications of detecting unsolicited findings such as variants in IPD genes RUNX1, ETV6, and ANKRD26, which are associated with increased leukemic risk. This guidance document has been developed and written by a multidisciplinary team of researchers and clinicians, with expertise in hematology, clinical and molecular genetics, and bioethics, alongside a RUNX1 patient advocacy representative. We recommend that for clinical diagnostics, HTS for IPD should use a multigene panel of curated diagnostic-grade genes. Critically, we advise that an HTS test for clinical diagnostics should only be ordered by a clinical expert that is: (a) fully aware of the complexity of genotype-phenotype correlations for IPD; (b) able to discuss these complexities with a patient and family members before the test is initiated; and (c) able to interpret and appropriately communicate the results of a HTS diagnostic report, including the implication of variants of uncertain clinical significance. Each patient should know what an HTS test could mean for his or her clinical management before initiating a test. We hereby propose an exemplified informed consent document that includes information on these ethical concerns and can be used by the community for implementation of HTS of IPD in a clinical diagnostic setting. This paper does not include recommendations for HTS of IPD in a research setting.
Collapse
Affiliation(s)
- Kate Downes
- East Genomic Laboratory HubCambridge University Hospitals NHS Foundation TrustCambridgeUK
- Department of HaematologyUniversity of CambridgeCambridge Biomedical CampusCambridgeUK
| | - Pascal Borry
- Department of Public Health and Primary CareKU LeuvenLeuvenBelgium
| | | | - Keith Gomez
- Haemophilia Centre and Thrombosis UnitRoyal Free London NHS Foundation TrustLondonUK
| | - Andreas Greinacher
- Institut für Immunologie und TransfusionsmedizinUniversitätsmedizin GreifswaldGreifswaldGermany
| | - Michele Lambert
- Division of HematologyThe Children’s Hospital of PhiladelphiaPhiladelphiaPAUSA
- Department of PediatricsPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPAUSA
| | - Eva Leinoe
- Department of HaematologyRigshospitaletNational University HospitalCopenhagenDenmark
| | - Patrizia Noris
- IRCCS Policlinico San Matteo Foundation and University of PaviaPaviaItaly
| | - Chris Van Geet
- Department of Cardiovascular SciencesCenter or Molecular and Vascular BiologyKU LeuvenLeuvenBelgium
| | - Kathleen Freson
- Department of Cardiovascular SciencesCenter or Molecular and Vascular BiologyKU LeuvenLeuvenBelgium
| | | |
Collapse
|
25
|
Huber M, Chiticariu E, Bachmann D, Flatz L, Hohl D. Palmoplantar Keratoderma with Leukokeratosis Anogenitalis Caused by KDSR Mutations. J Invest Dermatol 2020; 140:1662-1665.e1. [PMID: 31987885 DOI: 10.1016/j.jid.2019.11.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 11/01/2019] [Accepted: 11/11/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Marcel Huber
- Service of Dermatology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Elena Chiticariu
- Service of Dermatology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Daniel Bachmann
- Service of Dermatology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Lukas Flatz
- Institute for Immune Biology, St. Gallen Cantonal Hospital, St. Gallen, Switzerland
| | - Daniel Hohl
- Service of Dermatology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
26
|
The Link between Gaucher Disease and Parkinson's Disease Sheds Light on Old and Novel Disorders of Sphingolipid Metabolism. Int J Mol Sci 2019; 20:ijms20133304. [PMID: 31284408 PMCID: PMC6651136 DOI: 10.3390/ijms20133304] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 06/26/2019] [Accepted: 06/29/2019] [Indexed: 12/23/2022] Open
Abstract
Sphingolipid metabolism starts with the biosynthesis of ceramide, a bioactive lipid and the backbone for the biosynthesis of complex sphingolipids such as sphingomyelin and glycosphingolipids. These are degraded back to ceramide and then to sphingosine, which enters the ceramide–sphingosine-1-phosphate signaling pathway or is further degraded. Several enzymes with multiple catalytic properties and subcellular localizations are thus involved in such metabolism. Hereditary defects of lysosomal hydrolases have been known for several years to be the cause of lysosomal storage diseases such as gangliosidoses, Gaucher disease, Niemann–Pick disease, Krabbe disease, Fabry disease, and Farber disease. More recently, many other inborn errors of sphingolipid metabolism have been recognized, involving enzymes responsible for the biosynthesis of ceramide, sphingomyelin, and glycosphingolipids. Concurrently, epidemiologic and biochemical evidence has established a link between Gaucher disease and Parkinson’s disease, showing that glucocerebrosidase variants predispose individuals to α-synuclein accumulation and neurodegeneration even in the heterozygous status. This appears to be due not only to lysosomal overload of non-degraded glucosylceramide, but to the derangement of vesicle traffic and autophagy, including mitochondrial autophagy, triggered by both sphingolipid intermediates and misfolded proteins. In this review, old and novel disorders of sphingolipid metabolism, in particular those of ganglioside biosynthesis, are evaluated in light of recent investigations of the link between Gaucher disease and Parkinson’s disease, with the aim of better understanding their pathogenic mechanisms and addressing new potential therapeutic strategies.
Collapse
|