1
|
Misiti F, Diotaiuti P, Lombardo GE, Tellone E. Sphingosine-1-phosphate Decreases Erythrocyte Dysfunction Induced by β-Amyloid. Int J Mol Sci 2024; 25:5184. [PMID: 38791223 PMCID: PMC11121638 DOI: 10.3390/ijms25105184] [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: 03/15/2024] [Revised: 04/28/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Amyloid beta peptides (Aβ) have been identified as the main pathogenic agents in Alzheimer's disease (AD). Soluble Aβ oligomers, rather than monomer or insoluble amyloid fibrils, show red blood cell (RBC) membrane-binding capacity and trigger several morphological and functional alterations in RBCs that can result in impaired oxygen transport and delivery. Since bioactive lipids have been recently proposed as potent protective agents against Aβ toxicity, we investigated the role of sphingosine-1-phosphate (S1P) in signaling pathways involved in the mechanism underlying ATP release in Ab-treated RBCs. In RBCs following different treatments, the ATP, 2,3 DPG and cAMP levels and caspase 3 activity were determined by spectrophotometric and immunoassay. S1P rescued the inhibition of ATP release from RBCs triggered by Ab, through a mechanism involving caspase-3 and restoring 2,3 DPG and cAMP levels within the cell. These findings reveal the molecular basis of S1P protection against Aβ in RBCs and suggest new therapeutic avenues in AD.
Collapse
Affiliation(s)
- Francesco Misiti
- Human Sciences, Social and Health Department, University of Cassino and Lazio Meridionale, V. S. Angelo, Loc. Folcara, 03043 Cassino, Italy;
| | - Pierluigi Diotaiuti
- Human Sciences, Social and Health Department, University of Cassino and Lazio Meridionale, V. S. Angelo, Loc. Folcara, 03043 Cassino, Italy;
| | - Giovanni Enrico Lombardo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy; (G.E.L.); (E.T.)
| | - Ester Tellone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy; (G.E.L.); (E.T.)
| |
Collapse
|
2
|
Jiang ZJ, Gong LW. The SphK1/S1P Axis Regulates Synaptic Vesicle Endocytosis via TRPC5 Channels. J Neurosci 2023; 43:3807-3824. [PMID: 37185099 PMCID: PMC10217994 DOI: 10.1523/jneurosci.1494-22.2023] [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: 08/03/2022] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
Sphingosine-1-phosphate (S1P), a bioactive sphingolipid concentrated in the brain, is essential for normal brain functions, such as learning and memory and feeding behaviors. Sphingosine kinase 1 (SphK1), the primary kinase responsible for S1P production in the brain, is abundant within presynaptic terminals, indicating a potential role of the SphK1/S1P axis in presynaptic physiology. Altered S1P levels have been highlighted in many neurologic diseases with endocytic malfunctions. However, it remains unknown whether the SphK1/S1P axis may regulate synaptic vesicle endocytosis in neurons. The present study evaluates potential functions of the SphK1/S1P axis in synaptic vesicle endocytosis by determining effects of a dominant negative catalytically inactive SphK1. Our data for the first time identify a critical role of the SphK1/S1P axis in endocytosis in both neuroendocrine chromaffin cells and neurons from mice of both sexes. Furthermore, our Ca2+ imaging data indicate that the SphK1/S1P axis may be important for presynaptic Ca2+ increases during prolonged stimulations by regulating the Ca2+ permeable TRPC5 channels, which per se regulate synaptic vesicle endocytosis. Collectively, our data point out a critical role of the regulation of TRPC5 by the SphK1/S1P axis in synaptic vesicle endocytosis.SIGNIFICANCE STATEMENT Sphingosine kinase 1 (SphK1), the primary kinase responsible for brain sphingosine-1-phosphate (S1P) production, is abundant within presynaptic terminals. Altered SphK1/S1P metabolisms has been highlighted in many neurologic disorders with defective synaptic vesicle endocytosis. However, whether the SphK1/S1P axis may regulate synaptic vesicle endocytosis is unknown. Here, we identify that the SphK1/S1P axis regulates the kinetics of synaptic vesicle endocytosis in neurons, in addition to controlling fission-pore duration during single vesicle endocytosis in neuroendocrine chromaffin cells. The regulation of the SphK1/S1P axis in synaptic vesicle endocytosis is specific since it has a distinguished signaling pathway, which involves regulation of Ca2+ influx via TRPC5 channels. This discovery may provide novel mechanistic implications for the SphK1/S1P axis in brain functions under physiological and pathologic conditions.
Collapse
Affiliation(s)
- Zhong-Jiao Jiang
- Department of Biological Sciences, University of Illinois Chicago, Chicago, Illinois 60607
| | - Liang-Wei Gong
- Department of Biological Sciences, University of Illinois Chicago, Chicago, Illinois 60607
| |
Collapse
|
3
|
van Echten-Deckert G. The role of sphingosine 1-phosphate metabolism in brain health and disease. Pharmacol Ther 2023; 244:108381. [PMID: 36907249 DOI: 10.1016/j.pharmthera.2023.108381] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/13/2023]
Abstract
Lipids are essential structural and functional components of the central nervous system (CNS). Sphingolipids are ubiquitous membrane components which were discovered in the brain in the late 19th century. In mammals, the brain contains the highest concentration of sphingolipids in the body. Sphingosine 1-phosphate (S1P) derived from membrane sphingolipids evokes multiple cellular responses which, depending on its concentration and localization, make S1P a double-edged sword in the brain. In the present review we highlight the role of S1P in brain development and focus on the often contrasting findings regarding its contributions to the initiation, progression and potential recovery of different brain pathologies, including neurodegeneration, multiple sclerosis (MS), brain cancers, and psychiatric illnesses. A detailed understanding of the critical implications of S1P in brain health and disease may open the door for new therapeutic options. Thus, targeting S1P-metabolizing enzymes and/or signaling pathways might help overcome, or at least ameliorate, several brain illnesses.
Collapse
|
4
|
Dysregulation of sphingosine-1-phosphate (S1P) and S1P receptor 1 signaling in the 5xFAD mouse model of Alzheimer's disease. Brain Res 2023; 1799:148171. [PMID: 36410428 DOI: 10.1016/j.brainres.2022.148171] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/22/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022]
Abstract
Sphingolipid-1-phosphate (S1P) signaling through the activation S1P receptors (S1PRs) plays critical roles in cellular events in the brain. Aberrant S1P metabolism has been identified in the brains of Alzheimer's disease (AD) patients. Our recent studies have shown that treatment with fingolimod, an analog of sphingosine, provides neuroprotective effects in five familiar Alzheimer disease (5xFAD) transgenic mice, resulting in the reduction of amyloid-β (Aβ) neurotoxicity, inhibition of activation of microglia and astrocytes, increased hippocampal neurogenesis, and improved learning and memory. However, the pathways by which dysfunctional S1P and S1PR signaling may associate with the development of AD-like pathology remain unknown. In this study, we investigated the alteration of signaling of S1P/S1P receptor 1 (S1PR1), the most abundant S1PR subtype in the brain, in the cortex of 5xFAD transgenic mice at 3, 8, and 14 months of age. Compared to non-transgenic wildtype (WT) littermates, we found significant decreased levels of sphingosine kinases (SphKs), increased S1P lyase (S1PL), and increased S1PR1 in 8- and 14-month-old, but not in 3-month-old 5xFAD mice. Furthermore, we detected increased activation of the S1PR1 downstream pathway of Akt/mTor/Tau signaling in aging 5xFAD mice. Treatment with fingolimod from 1 to 8 months of age reversed the levels of SphKs, S1PL, and furthermore, those of S1PR1 and its downstream pathway of Akt/mTor/Tau signaling. Together the data reveal that dysregulation of S1P and S1PR signaling may associate with the development of AD-like pathology through Akt/mTor/Tau signaling.
Collapse
|
5
|
Bigi A, Cascella R, Fani G, Bernacchioni C, Cencetti F, Bruni P, Chiti F, Donati C, Cecchi C. Sphingosine 1-phosphate attenuates neuronal dysfunction induced by amyloid-β oligomers through endocytic internalization of NMDA receptors. FEBS J 2023; 290:112-133. [PMID: 35851748 PMCID: PMC10087929 DOI: 10.1111/febs.16579] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 05/24/2022] [Accepted: 07/18/2022] [Indexed: 01/14/2023]
Abstract
Soluble oligomers arising from the aggregation of the amyloid beta peptide (Aβ) have been identified as the main pathogenic agents in Alzheimer's disease (AD). Prefibrillar oligomers of the 42-residue form of Aβ (Aβ42 O) show membrane-binding capacity and trigger the disruption of Ca2+ homeostasis, a causative event in neuron degeneration. Since bioactive lipids have been recently proposed as potent protective agents against Aβ toxicity, we investigated the involvement of sphingosine 1-phosphate (S1P) signalling pathway in Ca2+ homeostasis in living neurons exposed to Aβ42 O. We show that both exogenous and endogenous S1P rescued neuronal Ca2+ dyshomeostasis induced by toxic Aβ42 O in primary rat cortical neurons and human neuroblastoma SH-SY5Y cells. Further analysis revealed a strong neuroprotective effect of S1P1 and S1P4 receptors, and to a lower extent of S1P3 and S1P5 receptors, which activate the Gi -dependent signalling pathways, thus resulting in the endocytic internalization of the extrasynaptic GluN2B-containing N-methyl-D-aspartate receptors (NMDARs). Notably, the S1P beneficial effect can be sustained over time by sphingosine kinase-1 overexpression, thus counteracting the down-regulation of the S1P signalling induced by Aβ42 O. Our findings disclose underlying mechanisms of S1P neuronal protection against harmful Aβ42 O, suggesting that S1P and its signalling axis can be considered promising targets for therapeutic approaches for AD.
Collapse
Affiliation(s)
- Alessandra Bigi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Roberta Cascella
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Giulia Fani
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Caterina Bernacchioni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Francesca Cencetti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Paola Bruni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Chiara Donati
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Cristina Cecchi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| |
Collapse
|
6
|
Custodia A, Romaus-Sanjurjo D, Aramburu-Núñez M, Álvarez-Rafael D, Vázquez-Vázquez L, Camino-Castiñeiras J, Leira Y, Pías-Peleteiro JM, Aldrey JM, Sobrino T, Ouro A. Ceramide/Sphingosine 1-Phosphate Axis as a Key Target for Diagnosis and Treatment in Alzheimer's Disease and Other Neurodegenerative Diseases. Int J Mol Sci 2022; 23:8082. [PMID: 35897658 PMCID: PMC9331765 DOI: 10.3390/ijms23158082] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 12/10/2022] Open
Abstract
Alzheimer's disease (AD) is considered the most prevalent neurodegenerative disease and the leading cause of dementia worldwide. Sphingolipids, such as ceramide or sphingosine 1-phosphate, are bioactive molecules implicated in structural and signaling functions. Metabolic dysfunction in the highly conserved pathways to produce sphingolipids may lead to or be a consequence of an underlying disease. Recent studies on transcriptomics and sphingolipidomics have observed alterations in sphingolipid metabolism of both enzymes and metabolites involved in their synthesis in several neurodegenerative diseases, including AD. In this review, we highlight the most relevant findings related to ceramide and neurodegeneration, with a special focus on AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Tomás Sobrino
- Neuro Aging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratories (LINCs), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (A.C.); (D.R.-S.); (M.A.-N.); (D.Á.-R.); (L.V.-V.); (J.C.-C.); (Y.L.); (J.M.P.-P.); (J.M.A.)
| | - Alberto Ouro
- Neuro Aging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratories (LINCs), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (A.C.); (D.R.-S.); (M.A.-N.); (D.Á.-R.); (L.V.-V.); (J.C.-C.); (Y.L.); (J.M.P.-P.); (J.M.A.)
| |
Collapse
|
7
|
Sphingolipid control of cognitive functions in health and disease. Prog Lipid Res 2022; 86:101162. [DOI: 10.1016/j.plipres.2022.101162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/10/2022] [Accepted: 03/12/2022] [Indexed: 12/14/2022]
|
8
|
Gao X, Li S, Cong C, Wang Y, Xu L. A Network Pharmacology Approach to Estimate Potential Targets of the Active Ingredients of Epimedium for Alleviating Mild Cognitive Impairment and Treating Alzheimer's Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:2302680. [PMID: 33574879 PMCID: PMC7861915 DOI: 10.1155/2021/2302680] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/21/2020] [Accepted: 01/15/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND The present study made use of a network pharmacological approach to evaluate the mechanisms and potential targets of the active ingredients of Epimedium for alleviating mild cognitive impairment (MCI) and treating Alzheimer's disease (AD). METHODS The active ingredients of Epimedium were acquired from the Traditional Chinese Medicine System Pharmacology database, and potential targets were predicted using the TCMSP target module, SwissTargetPrediction, and PharmMapper database. Target proteins correlating with MCI and AD were downloaded from the GeneCards, DisGeNet, and OMIM databases. The common targets of Epimedium, MCI, and AD were identified using the Jvenn online tool, and a protein-protein interaction (PPI) network was constructed using the String database and Cytoscape. Finally, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of the common targets was performed using DAVID, and molecular docking between active ingredients and target genes was modeled using AutoDock Vina. RESULTS A total of 20 active ingredients were analyzed, and 337 compound-related targets were identified for Epimedium. Out of 236 proteins associated with MCI and AD, 54 overlapped with the targets of Epimedium. The top 30 interacting proteins in this set were ranked by topological analysis. GO and KEGG enrichment analysis suggested that the common targets participated in diverse biological processes and pathways, including cell proliferation and apoptosis, inflammatory response, signal transduction, and protein phosphorylation through cancer pathway, MAPK signaling pathway, PI3K-Akt signaling pathway, HIF-1 signaling pathway, sphingolipid signaling pathway, FoxO signaling pathway, and TNF signaling pathway. Molecular docking analysis suggested that the 20 active ingredients could bind to the top 5 protein targets. CONCLUSIONS The present study provides theoretical evidence for in-depth analysis of the mechanisms and molecular targets by which Epimedium protects against MCI, AD, and other neurodegenerative diseases and lays the foundation for pragmatic clinical applications and potential new drug development.
Collapse
Affiliation(s)
- Xianwei Gao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shengnan Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Chao Cong
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yuejiao Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Lianwei Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| |
Collapse
|
9
|
Hao Y, Guo M, Feng Y, Dong Q, Cui M. Lysophospholipids and Their G-Coupled Protein Signaling in Alzheimer's Disease: From Physiological Performance to Pathological Impairment. Front Mol Neurosci 2020; 13:58. [PMID: 32351364 PMCID: PMC7174595 DOI: 10.3389/fnmol.2020.00058] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 03/24/2020] [Indexed: 12/21/2022] Open
Abstract
Lysophospholipids (LPLs) are bioactive signaling lipids that are generated from phospholipase-mediated hydrolyzation of membrane phospholipids (PLs) and sphingolipids (SLs). Lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) are two of the best-characterized LPLs which mediate a variety of cellular physiological responses via specific G-protein coupled receptor (GPCR) mediated signaling pathways. Considerable evidence now demonstrates the crucial role of LPA and S1P in neurodegenerative diseases, especially in Alzheimer’s disease (AD). Dysfunction of LPA and S1P metabolism can lead to aberrant accumulation of amyloid-β (Aβ) peptides, the formation of neurofibrillary tangles (NFTs), neuroinflammation and ultimately neuronal death. Summarizing LPA and S1P signaling profile may aid in profound health and pathological processes. In the current review, we will introduce the metabolism as well as the physiological roles of LPA and S1P in maintaining the normal functions of the nervous system. Given these pivotal functions, we will further discuss the role of dysregulation of LPA and S1P in promoting AD pathogenesis.
Collapse
Affiliation(s)
- Yining Hao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Min Guo
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yiwei Feng
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Grassi S, Mauri L, Prioni S, Cabitta L, Sonnino S, Prinetti A, Giussani P. Sphingosine 1-Phosphate Receptors and Metabolic Enzymes as Druggable Targets for Brain Diseases. Front Pharmacol 2019; 10:807. [PMID: 31427962 PMCID: PMC6689979 DOI: 10.3389/fphar.2019.00807] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/21/2019] [Indexed: 12/12/2022] Open
Abstract
The central nervous system is characterized by a high content of sphingolipids and by a high diversity in terms of different structures. Stage- and cell-specific sphingolipid metabolism and expression are crucial for brain development and maintenance toward adult age. On the other hand, deep dysregulation of sphingolipid metabolism, leading to altered sphingolipid pattern, is associated with the majority of neurological and neurodegenerative diseases, even those totally lacking a common etiological background. Thus, sphingolipid metabolism has always been regarded as a promising pharmacological target for the treatment of brain disorders. However, any therapeutic hypothesis applied to complex amphipathic sphingolipids, components of cellular membranes, has so far failed probably because of the high regional complexity and specificity of the different biological roles of these structures. Simpler sphingosine-based lipids, including ceramide and sphingosine 1-phosphate, are important regulators of brain homeostasis, and, thanks to the relative simplicity of their metabolic network, they seem a feasible druggable target for the treatment of brain diseases. The enzymes involved in the control of the levels of bioactive sphingoids, as well as the receptors engaged by these molecules, have increasingly allured pharmacologists and clinicians, and eventually fingolimod, a functional antagonist of sphingosine 1-phosphate receptors with immunomodulatory properties, was approved for the therapy of relapsing-remitting multiple sclerosis. Considering the importance of neuroinflammation in many other brain diseases, we would expect an extension of the use of such analogs for the treatment of other ailments in the future. Nevertheless, many aspects other than neuroinflammation are regulated by bioactive sphingoids in healthy brain and dysregulated in brain disease. In this review, we are addressing the multifaceted possibility to address the metabolism and biology of bioactive sphingosine 1-phosphate as novel targets for the development of therapeutic paradigms and the discovery of new drugs.
Collapse
Affiliation(s)
- Sara Grassi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Laura Mauri
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Simona Prioni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Livia Cabitta
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Sandro Sonnino
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Alessandro Prinetti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Paola Giussani
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
11
|
Angelopoulou E, Piperi C. Beneficial Effects of Fingolimod in Alzheimer's Disease: Molecular Mechanisms and Therapeutic Potential. Neuromolecular Med 2019; 21:227-238. [PMID: 31313064 DOI: 10.1007/s12017-019-08558-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/12/2019] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD), the most common cause of dementia remains of unclear etiology with current pharmacological therapies failing to halt disease progression. Several pathophysiological mechanisms have been implicated in AD pathogenesis including amyloid-β protein (Aβ) accumulation, tau hyperphosphorylation, neuroinflammation and alterations in bioactive lipid metabolism. Sphingolipids, such as sphingosine-1-phosphate (S1P) and intracellular ceramide/S1P balance are highly implicated in central nervous system physiology as well as in AD pathogenesis. FTY720/Fingolimod, a structural sphingosine analog and S1P receptor (S1PR) modulator that is currently used in the treatment of relapsing-remitting multiple sclerosis (RRMS) has been shown to exert beneficial effects on AD progression. Recent in vitro and in vivo evidence indicate that fingolimod may suppress Aβ secretion and deposition, inhibit apoptosis and enhance brain-derived neurotrophic factor (BDNF) production. Furthermore, it regulates neuroinflammation, protects against N-methyl-D-aspartate (NMDA)-excitotoxicity and modulates receptor for advanced glycation end products signaling axis that is highly implicated in AD pathogenesis. This review discusses the underlying molecular mechanisms of the emerging neuroprotective role of fingolimod in AD and its therapeutic potential, aiming to shed more light on AD pathogenesis as well as direct future treatment strategies.
Collapse
Affiliation(s)
- Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street - Bldg 16, 11527, Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street - Bldg 16, 11527, Athens, Greece.
| |
Collapse
|
12
|
Wu X, Wu Q, Zhou X, Huang J. SphK1 functions downstream of IGF-1 to modulate IGF-1-induced EMT, migration and paclitaxel resistance of A549 cells: A preliminary in vitro study. J Cancer 2019; 10:4264-4269. [PMID: 31413745 PMCID: PMC6691691 DOI: 10.7150/jca.32646] [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: 12/28/2018] [Accepted: 05/25/2019] [Indexed: 12/28/2022] Open
Abstract
Insulin-like growth factor-1 (IGF-1) -induced epithelial-mesenchymal transition (EMT) plays a key role in the metastasis and drug resistance of non-small cell lung cancer (NSCLC). Sphingosine kinase-1 (SphK1) is also involved in EMT of NSCLC. However, the interaction between SphK1 and IGF-1 in the EMT of NSCLC is largely unknown. To clarify this issue, we examined the involvement of SphK1 in IGF-1-induced EMT using human lung cancer cell line A549, and its paclitaxel-resistant subline. Cell viability was evaluated by cell counting kit-8 assay; Migratory ability was examined using scratch wound healing test; Protein expression levels of SphK1, vimentin, fibronectin, N-cadherin and E-cadherin were detected by western blot analysis, respectively. The results showed that, IGF-1 treatment of A549 cells stimulated the expression of SphK1, the activation of ERK and AKT, the cell migration, and the expression of EMT hallmark proteins, while inhibition of SphK1 by its specific inhibitor SKI-II suppressed all the above changes and increased the sensitivity of A549 cells to paclitaxel. Our data demonstrate that SphK1 acts as a downstream effector of IGF-1 and plays a critical role in IGF-1-induced EMT, cell migration and paclitaxel resistance of A549 cells, suggesting that SphK1 might be a potential therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Xingping Wu
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Jiangsu, P.R. China.,State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, P.R. China.,Department of Respirology, the First People's Hospital of Lianyungang, Jiangsu, P.R. China
| | - Qibiao Wu
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, P.R. China
| | - Xiqiao Zhou
- Department of Gastroenterology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu, P.R. China
| | - Jianan Huang
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Jiangsu, P.R. China
| |
Collapse
|
13
|
Jiang ZJ, Delaney TL, Zanin MP, Haberberger RV, Pitson SM, Huang J, Alford S, Cologna SM, Keating DJ, Gong LW. Extracellular and intracellular sphingosine-1-phosphate distinctly regulates exocytosis in chromaffin cells. J Neurochem 2019; 149:729-746. [PMID: 30963576 DOI: 10.1111/jnc.14703] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/28/2018] [Accepted: 03/27/2019] [Indexed: 01/18/2023]
Abstract
Sphingosine-1-phosphate (S1P) is an essential bioactive sphingosine lipid involved in many neurological disorders. Sphingosine kinase 1 (SphK1), a key enzyme for S1P production, is concentrated in presynaptic terminals. However, the role of S1P/SphK1 signaling in exocytosis remains elusive. By detecting catecholamine release from single vesicles in chromaffin cells, we show that a dominant negative SphK1 (SphK1DN ) reduces the number of amperometric spikes and increases the duration of foot, which reflects release through a fusion pore, implying critical roles for S1P in regulating the rate of exocytosis and fusion pore expansion. Similar phenotypes were observed in chromaffin cells obtained from SphK1 knockout mice compared to those from wild-type mice. In addition, extracellular S1P treatment increased the number of amperometric spikes, and this increase, in turn, was inhibited by a selective S1P3 receptor blocker, suggesting extracellular S1P may regulate the rate of exocytosis via activation of S1P3. Furthermore, intracellular S1P application induced a decrease in foot duration of amperometric spikes in control cells, indicating intracellular S1P may regulate fusion pore expansion during exocytosis. Taken together, our study represents the first demonstration that S1P regulates exocytosis through distinct mechanisms: extracellular S1P may modulate the rate of exocytosis via activation of S1P receptors while intracellular S1P may directly control fusion pore expansion during exocytosis. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Zhong-Jiao Jiang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Taylor L Delaney
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Mark P Zanin
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, P.R. China
| | - Rainer V Haberberger
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University, Adelaide, Australia
| | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Jian Huang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Simon Alford
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Stephanie M Cologna
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Damien J Keating
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University, Adelaide, Australia
| | - Liang-Wei Gong
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
14
|
The Role of Ceramide and Sphingosine-1-Phosphate in Alzheimer's Disease and Other Neurodegenerative Disorders. Mol Neurobiol 2019; 56:5436-5455. [PMID: 30612333 PMCID: PMC6614129 DOI: 10.1007/s12035-018-1448-3] [Citation(s) in RCA: 196] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 12/06/2018] [Indexed: 12/11/2022]
Abstract
Bioactive sphingolipids-ceramide, sphingosine, and their respective 1-phosphates (C1P and S1P)-are signaling molecules serving as intracellular second messengers. Moreover, S1P acts through G protein-coupled receptors in the plasma membrane. Accumulating evidence points to sphingolipids' engagement in brain aging and in neurodegenerative disorders such as Alzheimer's, Parkinson's, and Huntington's diseases and amyotrophic lateral sclerosis. Metabolic alterations observed in the course of neurodegeneration favor ceramide-dependent pro-apoptotic signaling, while the levels of the neuroprotective S1P are reduced. These trends are observed early in the diseases' development, suggesting causal relationship. Mechanistic evidence has shown links between altered ceramide/S1P rheostat and the production, secretion, and aggregation of amyloid β/α-synuclein as well as signaling pathways of critical importance for the pathomechanism of protein conformation diseases. Sphingolipids influence multiple aspects of Akt/protein kinase B signaling, a pathway that regulates metabolism, stress response, and Bcl-2 family proteins. The cross-talk between sphingolipids and transcription factors including NF-κB, FOXOs, and AP-1 may be also important for immune regulation and cell survival/death. Sphingolipids regulate exosomes and other secretion mechanisms that can contribute to either the spread of neurotoxic proteins between brain cells, or their clearance. Recent discoveries also suggest the importance of intracellular and exosomal pools of small regulatory RNAs in the creation of disturbed signaling environment in the diseased brain. The identified interactions of bioactive sphingolipids urge for their evaluation as potential therapeutic targets. Moreover, the early disturbances in sphingolipid metabolism may deliver easily accessible biomarkers of neurodegenerative disorders.
Collapse
|
15
|
Su L, Tian J, Sun J, Han N, Feng L, Yu B, Wang Y. Lentivirus-mediated siRNA knockdown of SPHK1 inhibits proliferation and tumorigenesis of neuroblastoma. Onco Targets Ther 2018; 11:7187-7196. [PMID: 30425511 PMCID: PMC6203087 DOI: 10.2147/ott.s180962] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The overexpression of sphingosine kinase 1 (SPHK1) is responsible for the progress of many cancers. However, the role of SPHK1 in the development and progression of neuroblastoma (NB) remain largely unknown. Here in this study, we explored whether silencing SPHK1 by lentivirus-mediated siRNA could be employed as a potential therapeutic target for NB. MATERIALS AND METHODS Lentivirus was adopted to load SPHK1 siRNA. The results were obtained using RT-qPCR, Western blot, cell proliferation assay, transwell cell migration/invasion assays as well as in vivo xenograft tumor models in nude mice. RESULTS Our results demonstrated that SPHK1 mRNA was upregulated in SH-SY5Y and SK-N-SH cells as well as in human NB tissues. SPHK1 knockdown by siRNA resulted in impaired proliferation, increased apoptosis, as well as impaired migration and invasion of SH-SY5Y and SK-N-SH cells. In addition, the in vivo study suggested that SPHK1 knockdown significantly reduced the tumorigenesis of SH-SY5Y xenograft model. Furthermore, intratumorally administered lentivirus-SPHK1 siRNA could significantly inhibit tumor growth in an SH-SY5Y xenograft mice model. Intensive investigations on mechanism revealed that these effects were achieved through the deactivation of STAT3 pathways. CONCLUSION These data suggest that SPHK1 inhibition via downregulation of STAT3 pathways by lentivirus-mediated siRNA knockdown can significantly suppress NB progression, which could be a promising target for future gene therapy of NB.
Collapse
Affiliation(s)
- Lin Su
- Department of Pediatric Surgery, Affiliated Hospital of Jining Medical University, Jining City 272029, Shandong Province, People's Republic of China,
| | - Junyan Tian
- Department of Pediatric Surgery, Affiliated Hospital of Jining Medical University, Jining City 272029, Shandong Province, People's Republic of China,
| | - Jinsong Sun
- Department of Pediatric Surgery, Affiliated Hospital of Jining Medical University, Jining City 272029, Shandong Province, People's Republic of China,
| | - Nuan Han
- Department of Pediatric Surgery, Affiliated Hospital of Jining Medical University, Jining City 272029, Shandong Province, People's Republic of China,
| | - Lin Feng
- Department of Pediatric Surgery, Affiliated Hospital of Jining Medical University, Jining City 272029, Shandong Province, People's Republic of China,
| | - Baohua Yu
- Department of Pediatric Surgery, Affiliated Hospital of Jining Medical University, Jining City 272029, Shandong Province, People's Republic of China,
| | - Yuepeng Wang
- Department of Pediatric Surgery, Affiliated Hospital of Jining Medical University, Jining City 272029, Shandong Province, People's Republic of China,
| |
Collapse
|
16
|
Tayebati SK. Phospholipid and Lipid Derivatives as Potential Neuroprotective Compounds. Molecules 2018; 23:molecules23092257. [PMID: 30189584 PMCID: PMC6225353 DOI: 10.3390/molecules23092257] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 08/22/2018] [Accepted: 09/04/2018] [Indexed: 12/13/2022] Open
Abstract
The worldwide demographical trend is changing towards a more elderly population. In particular, this phenomenon is increasing the number of neurodegenerative disease cases (e.g., Alzheimer’s disease) in advanced countries. Therefore, there is a fertile field for neuroprotective approaches to address this problem. A useful strategy to protect the membrane integrity of cells and reduce inflammatory processes. In this context, the neurons represent particularly vulnerable cells. Thus, a protection strategy should include their membrane preservation and improved anti-inflammatory processes. The contribution of phospholipid derivatives to this issue is crucial and many articles evidence their role in both health and disease. On the other hand, some lipids containing choline actively participate to increase the choline levels in the nervous system. It is acknowledged that the cholinergic system plays a pivotal role both in the central and in the peripheral nervous system. Neurons cannot synthesize choline, which is provided by the diet. The reuptake of ACh and its hydrolysis represent the principal source of choline. Therefore, to cover choline needs, choline-containing lipids may be used. There are different works which demonstrate their neuroprotective features This review article analyzes phospholipid and lipid derivatives that through different mechanisms are involved in these protective processes, although, sometimes the same molecules may behave as neurotoxic elements, therefore, their protective machinery should be detailed better.
Collapse
|
17
|
Modulatory Effects of Fingolimod (FTY720) on the Expression of Sphingolipid Metabolism-Related Genes in an Animal Model of Alzheimer's Disease. Mol Neurobiol 2018; 56:174-185. [PMID: 29687345 PMCID: PMC6334734 DOI: 10.1007/s12035-018-1040-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 03/27/2018] [Indexed: 01/30/2023]
Abstract
Sphingolipid signaling disturbances correlate with Alzheimer's disease (AD) progression. We examined the influence of FTY720/fingolimod, a sphingosine analog and sphingosine-1-phosphate (S1P) receptor modulator, on the expression of sphingolipid metabolism and signaling genes in a mouse transgenic AD model. Our results demonstrated that AβPP (V717I) transgene led with age to reduced mRNA expression of S1P receptors (S1PRs), sphingosine kinase SPHK2, ceramide kinase CERK, and the anti-apoptotic Bcl2 in the cerebral cortex and hippocampus, suggesting a pro-apoptotic shift in 12-month old mice. These changes largely emulated alterations we observed in the human sporadic AD hippocampus: reduced SPHK1, SPHK2, CERK, S1PR1, and BCL2. We observed that the responses to FTY720 treatment were modified by age and notably differed between control (APP-) and AD transgenic (APP+) animals. AβPP (V717I)-expressing 12-month-old animals reacted to fingolimod with wide changes in the gene expression program in cortex and hippocampus, including increased pro-survival SPHKs and CERK. Moreover, BCL2 was elevated by FTY720 in the cortex at all ages (3, 6, 12 months) while in hippocampus this increase was observed at 12 months only. In APP- mice, fingolimod did not induce any significant mRNA changes at 12 months. Our results indicate significant effect of FTY720 on the age-dependent transcription of genes involved in sphingolipid metabolism and pro-survival signaling, suggesting its neuroprotective role in AD animal model.
Collapse
|
18
|
Dominguez G, Maddelein ML, Pucelle M, Nicaise Y, Maurage CA, Duyckaerts C, Cuvillier O, Delisle MB. Neuronal sphingosine kinase 2 subcellular localization is altered in Alzheimer's disease brain. Acta Neuropathol Commun 2018; 6:25. [PMID: 29615132 PMCID: PMC5883421 DOI: 10.1186/s40478-018-0527-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/19/2018] [Indexed: 12/27/2022] Open
Abstract
Background Alzheimer’s disease (AD) is characterized by the accumulation of β-amyloid (Aβ) peptides and hyperphosphorylated tau protein accompanied by neuronal loss. Aβ accumulation has been associated with an impaired sphingosine 1-phosphate (S1P) metabolism. S1P is generated by sphingosine kinases (SphKs), of which there are two isoenzymes SphK1 and SphK2, and degraded by the sphingosine 1-phosphate lyase (SPL). We previously reported, that both a decrease in SphK1 expression and an increase in SPL expression, correlated with amyloid deposits in the entorhinal cortex of AD brains, suggesting a global loss of pro-survival S1P in AD neurons. SphK2 contribution has also been examined in AD yielding to conflicting results that may reflect the complexity of SphK2 regulation. The subcellular localization of SphK2, hence the compartmentalization of generated S1P, is recognized to play a crucial role in dictating either its pro-survival or pro-apoptotic functions. We therefore aimed at studying the expression of SphK2 and notably its subcellular localization in brain tissues from patients with AD. Results We report that a decrease in SphK2 protein cytosolic expression correlated with the density of amyloid deposits in a cohort of 25 post-mortem brains. Interestingly, we observed that the equilibrium between cytoplasmic and nuclear SphK2 is disrupted and showed that SphK2 is preferentially localized in the nucleus in AD brain extracts as compared to control extracts, with a marked increase of cleaved SphK2. Conclusions Our results suggest that a shift in the subcellular localization of the S1P generating SphK2 may compromise the well established pro-survival cytosolic S1P by favoring the production of nuclear S1P associated with adverse effects in AD pathogenesis. Electronic supplementary material The online version of this article (10.1186/s40478-018-0527-z) contains supplementary material, which is available to authorized users.
Collapse
|
19
|
Motyl J, Wencel PL, Cieślik M, Strosznajder RP, Strosznajder JB. Alpha-synuclein alters differently gene expression of Sirts, PARPs and other stress response proteins: implications for neurodegenerative disorders. Mol Neurobiol 2018; 55:727-740. [PMID: 28050792 PMCID: PMC5808059 DOI: 10.1007/s12035-016-0317-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 11/21/2016] [Indexed: 02/06/2023]
Abstract
Alpha-synuclein (ASN) is a presynaptic protein that can easily change its conformation under different types of stress. It's assumed that ASN plays an important role in the pathogenesis of Parkinson's and Alzheimer's disease. However, the molecular mechanism of ASN toxicity has not been elucidated. This study focused on the role of extracellular ASN (eASN) in regulation of transcription of sirtuins (Sirts) and DNA-bound poly(ADP-ribose) polymerases (PARPs) - proteins crucial for cells' survival/death. Our results indicate that eASN enhanced the free radicals level, decreased mitochondria membrane potential, cells viability and activated cells' death. Concomitantly eASN activated expression of antioxidative proteins (Sod2, Gpx4, Gadd45b) and DNA-bound Parp2 and Parp3. Moreover, eASN upregulated expression of Sirt3 and Sirt5, but downregulated of Sirt1, which plays an important role in cell metabolism including Aβ precursor protein (APP) processing. eASN downregulated gene expression of APP alpha secretase (Adam10) and metalloproteinases Mmp2, Mmp10 but upregulated Mmp11. Additionally, expression and activity of pro-survival sphingosine kinase 1 (Sphk1), Akt kinase and anti-apoptotic protein Bcl2 were inhibited. Moreover, higher expression of pro-apoptotic protein Bax and enhancement of apoptotic cells' death were observed. Summarizing, eASN significantly modulates transcription of Sirts and enzymes involved in APP/Aβ metabolism and through these mechanisms eASN toxicity may be enhanced. The inhibition of Sphk1 and Akt by eASN may lead to disturbances of survival pathways. These results suggest that eASN through alteration of transcription and by inhibition of pro-survival kinases may play important pathogenic role in neurodegenerative disorders.
Collapse
Affiliation(s)
- J Motyl
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Street, Warsaw, Poland
| | - P L Wencel
- Laboratory of Preclinical Research and Environmental Agents, Department of Neurosurgery, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Street, 02-106, Warsaw, Poland
| | - M Cieślik
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Street, Warsaw, Poland
| | - R P Strosznajder
- Laboratory of Preclinical Research and Environmental Agents, Department of Neurosurgery, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Street, 02-106, Warsaw, Poland.
| | - J B Strosznajder
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Street, Warsaw, Poland
| |
Collapse
|
20
|
Werth S, Müller-Fielitz H, Raasch W. Obesity-stimulated aldosterone release is not related to an S1P-dependent mechanism. J Endocrinol 2017; 235:251-265. [PMID: 28970286 DOI: 10.1530/joe-16-0550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 09/26/2017] [Indexed: 11/08/2022]
Abstract
Aldosterone has been identified as an important factor in obesity-associated hypertension. Here, we investigated whether sphingosine-1-phosphate (S1P), which has previously been linked to obesity, increases aldosterone release. S1P-induced aldosterone release was determined in NCI H295R cells in the presence of S1P receptor (S1PR) antagonists. In vivo release of S1P (100-300 µg/kgbw) was investigated in pithed, lean Sprague Dawley (SD) rats, diet-obese spontaneous hypertensive rats (SHRs), as well as in lean or obese Zucker rats. Aldosterone secretion was increased in NCI H295R cells by S1P, the selective S1PR1 agonist SEW2871 and the selective S1PR2 antagonist JTE013. Treatment with the S1PR1 antagonist W146 or fingolimod and the S1PR1/3 antagonist VPbib2319 decreased baseline and/or S1P-stimulated aldosterone release. Compared to saline-treated SD rats, plasma aldosterone increased by ~50 pg/mL after infusing S1P. Baseline levels of S1P and aldosterone were higher in obese than in lean SHRs. Adrenal S1PR expression did not differ between chow- or CD-fed rats that had the highest S1PR1 and lowest S1PR4 levels. S1P induced a short-lasting increase in plasma aldosterone in obese, but not in lean SHRs. However, 2-ANOVA did not demonstrate any difference between lean and obese rats. S1P-induced aldosterone release was also similar between obese and lean Zucker rats. We conclude that S1P is a local regulator of aldosterone production. S1PR1 agonism induces an increase in aldosterone secretion, while stimulating adrenal S1PR2 receptor suppresses aldosterone production. A significant role of S1P in influencing aldosterone secretion in states of obesity seems unlikely.
Collapse
Affiliation(s)
- Stephan Werth
- Institute of Experimental and Clinical Pharmacology and ToxicologyUniversity of Lübeck, Lübeck, Germany
| | - Helge Müller-Fielitz
- Institute of Experimental and Clinical Pharmacology and ToxicologyUniversity of Lübeck, Lübeck, Germany
- CBBM (Center of Brain, Behavior and Metabolism)Lübeck, Germany
| | - Walter Raasch
- Institute of Experimental and Clinical Pharmacology and ToxicologyUniversity of Lübeck, Lübeck, Germany
- CBBM (Center of Brain, Behavior and Metabolism)Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research)partner site Hamburg/Kiel/Lübeck, Lübeck, Germany
| |
Collapse
|
21
|
Grimm MOW, Mett J, Grimm HS, Hartmann T. APP Function and Lipids: A Bidirectional Link. Front Mol Neurosci 2017; 10:63. [PMID: 28344547 PMCID: PMC5344993 DOI: 10.3389/fnmol.2017.00063] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 02/24/2017] [Indexed: 12/14/2022] Open
Abstract
Extracellular neuritic plaques, composed of aggregated amyloid-β (Aβ) peptides, are one of the major histopathological hallmarks of Alzheimer's disease (AD), a progressive, irreversible neurodegenerative disorder and the most common cause of dementia in the elderly. One of the most prominent risk factor for sporadic AD, carrying one or two aberrant copies of the apolipoprotein E (ApoE) ε4 alleles, closely links AD to lipids. Further, several lipid classes and fatty acids have been reported to be changed in the brain of AD-affected individuals. Interestingly, the observed lipid changes in the brain seem not only to be a consequence of the disease but also modulate Aβ generation. In line with these observations, protective lipids being able to decrease Aβ generation and also potential negative lipids in respect to AD were identified. Mechanistically, Aβ peptides are generated by sequential proteolytic processing of the amyloid precursor protein (APP) by β- and γ-secretase. The α-secretase appears to compete with β-secretase for the initial cleavage of APP, preventing Aβ production. All APP-cleaving secretases as well as APP are transmembrane proteins, further illustrating the impact of lipids on Aβ generation. Beside the pathological impact of Aβ, accumulating evidence suggests that Aβ and the APP intracellular domain (AICD) play an important role in regulating lipid homeostasis, either by direct effects or by affecting gene expression or protein stability of enzymes involved in the de novo synthesis of different lipid classes. This review summarizes the current literature addressing the complex bidirectional link between lipids and AD and APP processing including lipid alterations found in AD post mortem brains, lipids that alter APP processing and the physiological functions of Aβ and AICD in the regulation of several lipid metabolism pathways.
Collapse
Affiliation(s)
- Marcus O. W. Grimm
- Experimental Neurology, Saarland UniversityHomburg/Saar, Germany
- Neurodegeneration and Neurobiology, Saarland UniversityHomburg/Saar, Germany
- Deutsches Institut für DemenzPrävention (DIDP), Saarland UniversityHomburg/Saar, Germany
| | - Janine Mett
- Experimental Neurology, Saarland UniversityHomburg/Saar, Germany
| | - Heike S. Grimm
- Experimental Neurology, Saarland UniversityHomburg/Saar, Germany
| | - Tobias Hartmann
- Experimental Neurology, Saarland UniversityHomburg/Saar, Germany
- Neurodegeneration and Neurobiology, Saarland UniversityHomburg/Saar, Germany
- Deutsches Institut für DemenzPrävention (DIDP), Saarland UniversityHomburg/Saar, Germany
| |
Collapse
|
22
|
Vitamin D 3 protects against Aβ peptide cytotoxicity in differentiated human neuroblastoma SH- SY5Y cells: A role for S1P1/p38MAPK/ATF4 axis. Neuropharmacology 2017; 116:328-342. [PMID: 28077289 DOI: 10.1016/j.neuropharm.2017.01.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 12/23/2016] [Accepted: 01/05/2017] [Indexed: 02/06/2023]
Abstract
Besides its classical function of bone metabolism regulation, 1alpha, 25-dihydroxyvitamin D3 (1,25(OH)2D3), acts on a variety of tissues including the nervous system, where the hormone plays an important role as neuroprotective, antiproliferating and differentiating agent. Sphingolipids are bioactive lipids that play critical and complex roles in regulating cell fate. In the present paper we have investigated whether sphingolipids are involved in the protective action of 1,25(OH)2D3. We have found that 1,25(OH)2D3 prevents amyloid-β peptide (Aβ(1-42)) cytotoxicity both in differentiated SH-SY5Y human neuroblastoma cells and in vivo. In differentiated SH-SY5Y cells, Aβ(1-42) strongly reduces the sphingosine-1-phosphate (S1P)/ceramide (Cer) ratio while 1,25(OH)2D3 partially reverts this effect. 1,25(OH)2D3 reverts also the Aβ(1-42)-induced reduction of sphingosine kinase activity. We have also studied the crosstalk between 1,25(OH)2D3 and S1P signaling pathways downstream to the activation of S1P receptor subtype S1P1. Notably, we found that 1,25(OH)2D3 prevents the reduction of S1P1 expression promoted by Aβ(1-42) and thereby it modulates the downstream signaling leading to ER stress damage (p38MAPK/ATF4). Similar effects were observed by using ZK191784. In addition, chronic treatment with 1,25(OH)2D3 protects from aggregated Aβ(1-42)-induced damage in the CA1 region of the rat hippocampus and promotes cell proliferation in the hippocampal dentate gyrus of adult mice. In conclusion, these results represent the first evidence of the role of 1,25(OH)2D3 and its structural analogue ZK191784 in counteracting the Aβ(1-42) peptide-induced toxicity through the modulation of S1P/S1P1/p38MAPK/ATF4 pathway in differentiated SH-SY5Y cells.
Collapse
|
23
|
Kitchen SA, Weis VM. The sphingosine rheostat is involved in the cnidarian heat stress response but not necessarily in bleaching. J Exp Biol 2017; 220:1709-1720. [DOI: 10.1242/jeb.153858] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 02/16/2017] [Indexed: 12/17/2022]
Abstract
Sphingolipids play important roles in mitigating cellular heat and oxidative stress by altering membrane fluidity, receptor clustering and gene expression. Accumulation of signaling sphingolipids that comprise the sphingosine rheostat, pro-apoptotic sphingosine (Sph) and pro-survival sphingosine-1-phosphate (S1P), is key to determining cell fate. Reef-building corals and other symbiotic cnidarians living in shallow tropical waters can experience elevated seawater temperature and high UV irradiance, two stressors that are increasing in frequency and severity with climate change. In symbiotic cnidarians, these stressors disrupt the photosynthetic machinery of the endosymbiont and ultimately result in the collapse of the partnership (dysbiosis), known as cnidarian bleaching. In a previous study, exogenously applied sphingolipids altered heat-induced bleaching in the symbiotic anemone Aiptasia pallida, but endogenous regulation of these lipids is unknown. Here, we characterized the role of the rheostat in the cnidarian heat stress response (HSR) and in dysbiosis. Gene expression of rheostat enzymes sphingosine kinase (AP-SPHK) and S1P phosphatase (AP-SGPP), and concentrations of sphingolipids were quantified from anemones incubated at elevated temperatures. We observed a biphasic HSR in A. pallida. At early exposure, rheostat gene expression and lipid levels were suppressed while gene expression of a heat stress biomarker increased and 40% of symbionts were lost. After longer incubations at the highest temperature, AP-SGPP and then Sph levels both increased. These results indicate that the sphingosine rheostat in A. pallida does not participate in initiation of dysbiosis, but instead functions in the chronic response to prolonged heat stress that promotes host survival.
Collapse
Affiliation(s)
- Sheila A. Kitchen
- Department of Integrative Biology, Oregon State University, 3029 Cordley Hall, Corvallis, OR 97331, USA
| | - Virginia M. Weis
- Department of Integrative Biology, Oregon State University, 3029 Cordley Hall, Corvallis, OR 97331, USA
| |
Collapse
|
24
|
Ghasemi R, Dargahi L, Ahmadiani A. Integrated sphingosine-1 phosphate signaling in the central nervous system: From physiological equilibrium to pathological damage. Pharmacol Res 2016; 104:156-64. [DOI: 10.1016/j.phrs.2015.11.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 11/14/2015] [Accepted: 11/15/2015] [Indexed: 01/09/2023]
|
25
|
TGFβ-Mediated induction of SphK1 as a potential determinant in human MDA-MB-231 breast cancer cell bone metastasis. BONEKEY REPORTS 2015; 4:719. [PMID: 26157579 DOI: 10.1038/bonekey.2015.88] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 05/08/2015] [Indexed: 12/17/2022]
Abstract
Mechanistic understanding of the preferential homing of circulating tumor cells to bone and their perturbation on bone metabolism within the tumor-bone microenvironment remains poorly understood. Alteration in both transforming growth factor β (TGFβ) signaling and sphingolipid metabolism results in the promotion of tumor growth and metastasis. Previous studies using MDA-MB-231 human breast cancer-derived cell lines of variable metastatic potential were queried for changes in sphingolipid metabolism genes to explore correlations between TGFβ dependence and bone metastatic behavior. Of these genes, only sphingosine kinase-1 (SPHK1) was identified to be significantly increased following TGFβ treatment. Induction of SPHK1 expression correlated to the degree of metastatic capacity in these MDA-MB-231-derived cell lines. We demonstrate that TGFβ mediates the regulation of SPHK1 gene expression, protein kinase activity and is critical to MDA-MB-231 cell viability. Furthermore, a bioinformatic analysis of human breast cancer gene expression supports SPHK1 as a hallmark TGFβ target gene that also bears the genetic fingerprint of the basal-like/triple-negative breast cancer molecular subtype. These data suggest a potential new signaling axis between TGFβ/SphK1 that may have a role in the development, prognosis or the clinical phenotype associated with tumor-bone metastasis.
Collapse
|
26
|
Ceramides in Alzheimer's Disease: Key Mediators of Neuronal Apoptosis Induced by Oxidative Stress and Aβ Accumulation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:346783. [PMID: 26090071 PMCID: PMC4458271 DOI: 10.1155/2015/346783] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 05/06/2015] [Accepted: 05/07/2015] [Indexed: 11/17/2022]
Abstract
Alzheimer's disease (AD), the most common chronic and progressive neurodegenerative disorder, is characterized by extracellular deposits of amyloid β-peptides (Aβ) and intracellular deposits of hyperphosphorylated tau (phospho-tau) protein. Ceramides, the major molecules of sphingolipid metabolism and lipid second messengers, have been associated with AD progression and pathology via Aβ generation. Enhanced levels of ceramides directly increase Aβ through stabilization of β-secretase, the key enzyme in the amyloidogenic processing of Aβ precursor protein (APP). As a positive feedback loop, the generated oligomeric and fibrillar Aβ induces a further increase in ceramide levels by activating sphingomyelinases that catalyze the catabolic breakdown of sphingomyelin to ceramide. Evidence also supports important role of ceramides in neuronal apoptosis. Ceramides may initiate a cascade of biochemical alterations, which ultimately leads to neuronal death by diverse mechanisms, including depolarization and permeabilization of mitochondria, increased production of reactive oxygen species (ROS), cytochrome c release, Bcl-2 depletion, and caspase-3 activation, mainly by modulating intracellular signalling, particularly along the pathways related to Akt/PKB kinase and mitogen-activated protein kinases (MAPKs). This review summarizes recent findings related to the role of ceramides in oxidative stress-driven neuronal apoptosis and interplay with Aβ in the cascade of events ending in neuronal degeneration.
Collapse
|
27
|
Muñoz-Sáez E, de Munck García E, Arahuetes Portero RM, Vicente F, Ortiz-López FJ, Cantizani J, Gómez Miguel B. Neuroprotective role of sphingosine-1-phosphate in L-BMAA treated neuroblastoma cells (SH-SY5Y). Neurosci Lett 2015; 593:83-9. [PMID: 25769802 DOI: 10.1016/j.neulet.2015.03.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 02/14/2015] [Accepted: 03/06/2015] [Indexed: 10/23/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive lipid which regulates proliferation, cell migration, survival and differentiation by specific receptors activation. We studied its effects on L-BMAA treated neuroblastoma cells (SH-SY5Y), an amino acid that can trigger neurodegenerative diseases such as amyotrophic lateral sclerosis/Parkinson dementia complex (ALS/PDC). We found that S1P protects from necrosis and prevents the GSK3 increasing as long as the PI3K/AKT pathway is active. Moreover, GSK3 inhibition protects against neuronal death caused by L-BMAA.
Collapse
Affiliation(s)
- Emma Muñoz-Sáez
- Departamento de Bioquímica y Biología Molecular-I, Universidad Complutense de Madrid, 28040-Madrid, Spain.
| | | | | | - Francisca Vicente
- Fundación MEDINA, Centro Excelencia Investigación Medicamentos Innovadores en Andalucía, Parque Tecnológico Ciencias de la Salud, 18016-Armilla-Granada, Spain
| | - Francisco Javier Ortiz-López
- Fundación MEDINA, Centro Excelencia Investigación Medicamentos Innovadores en Andalucía, Parque Tecnológico Ciencias de la Salud, 18016-Armilla-Granada, Spain
| | - Juan Cantizani
- Fundación MEDINA, Centro Excelencia Investigación Medicamentos Innovadores en Andalucía, Parque Tecnológico Ciencias de la Salud, 18016-Armilla-Granada, Spain
| | - Begoña Gómez Miguel
- Departamento de Bioquímica y Biología Molecular-I, Universidad Complutense de Madrid, 28040-Madrid, Spain
| |
Collapse
|
28
|
Sphingosine-1-phosphate and its effect on glucose deprivation/glucose reload stress: from gene expression to neuronal survival. Mol Neurobiol 2014; 51:1300-8. [PMID: 25056275 PMCID: PMC4434862 DOI: 10.1007/s12035-014-8807-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 07/08/2014] [Indexed: 12/14/2022]
Abstract
Sphingosine kinase-1 (Sphk1-1, EC 2.7.1.91) is a regulator of pro-survival signalling, and its alterations have been observed in Alzheimer’s disease, brain ischemia and other neurological disorders. In this study we addressed the question whether Sphk1 and its product, sphingosine-1-phosphate (S1P), play a significant role in glucose deprivation (GD)/glucose reload (GR) stress in hippocampal neuronal cells (HT22). It was found that GD (6 h) followed by 24 h of GR evoked enhancement of the free radical level and neuronal HT22 cell death. Moreover, the significantly stronger gene expression for the pro-apoptotic Bax protein and down-regulation of the anti-apoptotic Bcl-2 and Bcl-XL proteins were observed. Concomitantly, this stress up-regulated: gene expression, protein level and activity of Sphk1. Exogenous S1P at 1 μM concentration and the other agonists of the S1P1 receptor (SEW 2871 and P-FTY720) enhanced HT22 cell viability affected by GD/GR stress. This mechanism is mediated by S1P receptor(s) signalling and by the activation of gene expression for Bcl-2 and Bcl-XL. Summarising, our data suggest that sphingolipid metabolism may play an important role in the early events that take place in neuronal cell survival/death under GD/GR stress. Our data demonstrate that exogenous S1P, through the activation of specific receptors S1P1 and S1P3 signalling pathways, regulates the gene expression for anti-apoptotic proteins and enhances neuronal cell survival affected by GD/GR stress.
Collapse
|
29
|
Ceccom J, Delisle MB, Cuvillier O. [Sphingosine 1-phosphate as a biomarker for Alzheimer's disease?]. Med Sci (Paris) 2014; 30:493-5. [PMID: 24939530 DOI: 10.1051/medsci/20143005006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Affiliation(s)
- Johnatan Ceccom
- Service d'anatomie pathologique, CHU Toulouse Rangueil, faculté de médecine, 1, avenue Jean Poulhès, 31059 Toulouse Cedex 9, France
| | - Marie-Bernadette Delisle
- Service d'anatomie pathologique, CHU Toulouse Rangueil, faculté de médecine, 1, avenue Jean Poulhès, 31059 Toulouse Cedex 9, France - Inserm UMR 1037, France
| | - Olivier Cuvillier
- CNRS, institut de pharmacologie et de biologie structurale, Toulouse, France - Université de Toulouse, UPS, IPBS, Toulouse, France
| |
Collapse
|
30
|
Brizuela L, Martin C, Jeannot P, Ader I, Gstalder C, Andrieu G, Bocquet M, Laffosse JM, Gomez-Brouchet A, Malavaud B, Sabbadini RA, Cuvillier O. Osteoblast-derived sphingosine 1-phosphate to induce proliferation and confer resistance to therapeutics to bone metastasis-derived prostate cancer cells. Mol Oncol 2014; 8:1181-95. [PMID: 24768038 DOI: 10.1016/j.molonc.2014.04.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 03/27/2014] [Accepted: 04/02/2014] [Indexed: 02/07/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) plays important roles in cell proliferation, differentiation or survival mainly through its surface G-protein-coupled receptors S1P1-5. Bone represents the major site of metastasis for prostate cancer (CaP) cells, which rely on bone-derived factors to support their proliferation and resistance to therapeutics. In the present work we have found that conditioned medium (CM) from the MC3T3 osteoblastic cell line or primary murine and human osteoblast-like cells, as well as co-culture with MC3T3 stimulate proliferation of CaP lines in S1P-dependent manner. In addition, osteoblastic-derived S1P induces resistance of CaP cells to therapeutics including chemotherapy and radiotherapy. When S1P release from osteoblastic cells is decreased (inhibition of SphK1, knock-down of SphK1 or the S1P transporter, Spns2 by siRNA) or secreted S1P neutralized with anti-S1P antibody, the proliferative and survival effects of osteoblasts on CaP cells are abolished. Because of the paracrine nature of the signaling, we studied the role of the S1P receptors expressed on CaP cells in the communication with S1P secreted by osteoblasts. Strategies aimed at down-regulating S1P1, S1P2 or S1P3 (siRNA, antagonists), established the exclusive role of the S1P/S1P1 signaling between osteoblasts and CaP cells. Bone metastases from CaP are associated with osteoblastic differentiation resulting in abnormal bone formation. We show that the autocrine S1P/S1P3 signaling is central during differentiation to mature osteoblasts by regulating Runx2 level, a key transcription factor involved in osteoblastic maturation. Importantly, differentiated osteoblasts exhibited enhanced secretion of S1P and further stimulated CaP cell proliferation in a S1P-dependent manner. By establishing the dual role of osteoblast-borne S1P on both osteoblastic differentiation and CaP cell proliferation and survival, we uncover the importance of S1P in the bone metastatic microenvironment, which may open a novel area of study for the treatment of CaP bone metastasis by targeting S1P.
Collapse
Affiliation(s)
- Leyre Brizuela
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France; Université de Toulouse, UPS, IPBS, Toulouse, France; Equipe Labellisée Ligue contre le Cancer, France
| | - Claire Martin
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France; Université de Toulouse, UPS, IPBS, Toulouse, France; Equipe Labellisée Ligue contre le Cancer, France
| | - Pauline Jeannot
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France; Université de Toulouse, UPS, IPBS, Toulouse, France; Equipe Labellisée Ligue contre le Cancer, France
| | - Isabelle Ader
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France; Université de Toulouse, UPS, IPBS, Toulouse, France; Equipe Labellisée Ligue contre le Cancer, France
| | - Cécile Gstalder
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France; Université de Toulouse, UPS, IPBS, Toulouse, France; Equipe Labellisée Ligue contre le Cancer, France
| | - Guillaume Andrieu
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France; Université de Toulouse, UPS, IPBS, Toulouse, France; Equipe Labellisée Ligue contre le Cancer, France
| | - Magalie Bocquet
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France; Université de Toulouse, UPS, IPBS, Toulouse, France; Equipe Labellisée Ligue contre le Cancer, France
| | - Jean-Michel Laffosse
- Université de Toulouse, UPS, IPBS, Toulouse, France; CHU Toulouse, Service d'orthopédie et Traumatologie, Toulouse, France
| | - Anne Gomez-Brouchet
- Université de Toulouse, UPS, IPBS, Toulouse, France; CHU Toulouse, Service d'Anatomopathologie, Toulouse, France
| | - Bernard Malavaud
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France; Université de Toulouse, UPS, IPBS, Toulouse, France; Equipe Labellisée Ligue contre le Cancer, France; CHU Toulouse, Service d'Urologie et de Transplantation Rénale, Toulouse, France
| | | | - Olivier Cuvillier
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France; Université de Toulouse, UPS, IPBS, Toulouse, France; Equipe Labellisée Ligue contre le Cancer, France.
| |
Collapse
|
31
|
Yang Y, Wang M, Lv B, Ma R, Hu J, Dun Y, Sun S, Li G. Sphingosine Kinase-1 Protects Differentiated N2a Cells Against Beta-Amyloid25–35-Induced Neurotoxicity Via the Mitochondrial Pathway. Neurochem Res 2014; 39:932-40. [DOI: 10.1007/s11064-014-1290-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 03/09/2014] [Accepted: 03/20/2014] [Indexed: 12/14/2022]
|
32
|
Ceccom J, Loukh N, Lauwers-Cances V, Touriol C, Nicaise Y, Gentil C, Uro-Coste E, Pitson S, Maurage CA, Duyckaerts C, Cuvillier O, Delisle MB. Reduced sphingosine kinase-1 and enhanced sphingosine 1-phosphate lyase expression demonstrate deregulated sphingosine 1-phosphate signaling in Alzheimer's disease. Acta Neuropathol Commun 2014; 2:12. [PMID: 24468113 PMCID: PMC3912487 DOI: 10.1186/2051-5960-2-12] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 01/18/2014] [Indexed: 12/14/2022] Open
Abstract
Background The accumulation of beta amyloid (Aβ) peptides, a hallmark of Alzheimer’s disease (AD) is related to mechanisms leading to neurodegeneration. Among its pleiotropic cellular effects, Aβ accumulation has been associated with a deregulation of sphingolipid metabolism. Sphingosine 1-phosphate (S1P) derived from sphingosine is emerging as a critical lipid mediator regulating various biological activities including cell proliferation, survival, migration, inflammation, or angiogenesis. S1P tissue level is low and kept under control through equilibrium between its synthesis mostly governed by sphingosine kinase-1 (SphK1) and its degradation by sphingosine 1-phosphate lyase (SPL). We have previously reported that Aβ peptides were able to decrease the activity of SphK1 in cell culture models, an effect that could be blocked by the prosurvival IGF-1/IGF-1R signaling. Results Herein, we report for the first time the expression of both SphK1 and SPL by immunohistochemistry in frontal and entorhinal cortices from 56 human AD brains. Immunohistochemical analysis revealed a decreased expression of SphK1 and an increased expression of SPL both correlated to amyloid deposits in the entorhinal cortex. Otherwise, analysis of brain tissue extracts showed a decrease of SphK1 expression in AD brains whereas SPL expression was increased. The content of IGF-1R, an activator of SphK1, was found decreased in AD brains as well as S1P1, the major receptor for S1P. Conclusions Collectively, these results highlight the importance of S1P in AD suggesting the existence of a global deregulation of S1P signaling in this disease from its synthesis by SphK1 and degradation by SPL to its signaling by the S1P1 receptor.
Collapse
|
33
|
Gassowska M, Cieslik M, Wilkaniec A, Strosznajder JB. Sphingosine kinases/sphingosine-1-phosphate and death Signalling in APP-transfected cells. Neurochem Res 2014; 39:645-52. [PMID: 24452756 PMCID: PMC3962740 DOI: 10.1007/s11064-014-1240-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 12/07/2013] [Accepted: 01/09/2014] [Indexed: 02/06/2023]
Abstract
It has been postulated that disturbances in the sphingolipid metabolism play a key role in the pathogenesis of Alzheimer’s disease (AD). An alteration in sphingosine kinases 1, 2 (SphK1/2) and sphingosine-1-phosphate (S1P) was recently reported in AD. However, the effect of AD-related amyloid beta (Aβ) peptides on SphK1/2 and the role of S1P in Aβ toxicity have not been fully elucidated. In this study the relationship between the Aβ concentration and SphK1/2 expression/activity was analysed in PC12 cells transfected with the Aβ precursor protein, wild-type (APPwt) or bearing a double Swedish mutation (APPsw). The role of SphK(s)/S1P in cell survival and death was also investigated. Our results indicated that endogenously liberated Aβ significantly decreases expression and activity of SphK1/2. The SphK(s) inhibitor (SKI II, 10 μM) decreased the viability of APPwt, APPsw as well as empty vector-transfected PC12 control cells. Our data demonstrated that expression of S1P receptor-1 (S1P1) was significantly reduced in APP-transfected cells. The effect of S1P applied exogenously was cell type-dependent. In control and APPwt cells S1P reduced the effect of the SphK1 inhibitor on death signalling. Conversely, it decreased the survival of APPsw cells and had no protective effect on cells treated with SKI II. Using the S1P1 agonist (SEW2871, 5 μM) and antagonist (W123, 20 μM), we demonstrated that the cytoprotective effect of S1P was receptor-independent. Summarising, we showed that Aβ peptides evoke down-regulation of gene expression and activity for SphK(s) and S1P1. Inhibition of SphK(s) significantly decreased cell survival. The effect of exogenous S1P depended on the concentration of Aβ peptides.
Collapse
Affiliation(s)
- Magdalena Gassowska
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawinskiego 5, 02-106, Warsaw, Poland
| | | | | | | |
Collapse
|
34
|
Pyszko J, Strosznajder JB. Sphingosine kinase 1 and sphingosine-1-phosphate in oxidative stress evoked by 1-methyl-4-phenylpyridinium (MPP+) in human dopaminergic neuronal cells. Mol Neurobiol 2014; 50:38-48. [PMID: 24399507 DOI: 10.1007/s12035-013-8622-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 12/15/2013] [Indexed: 12/21/2022]
Abstract
Sphingosine kinases (Sphk1/2) are crucial enzymes in regulation of the biostat between sphingosine-1-phosphate (S1P) and ceramide and play an important role in the pathogenesis/pathomechanism of Alzheimer's disease (AD). These enzymes synthesise S1P, which regulates neurotransmission, synaptic function and neuron cell proliferation, by activating five G protein-coupled receptors (S1P1-5). However, S1P synthesised by Sphk2 could be involved in amyloid β (Aβ) release by stimulation of Aβ precursor protein degradation. The significance of this bioactive sphingolipid in the pathogenesis of Parkinson's disease (PD) is unknown. The aim of our study was to investigate the expression level of Sphk1 and its role in human dopaminergic neuronal cell (SH-SY5Y) viability under oxidative stress, evoked by 1-methyl-4-phenylpyridinium (MPP+). Moreover, the mechanism of S1P action on the death signalling pathway in these experimental conditions was evaluated. Our study indicated marked downregulation of Sphk1 expression in this cellular PD model. Inhibition of Sphk1 decreased SH-SY5Y cell viability and concomitantly enhanced the reactive oxygen species (ROS) level. It was found that exogenous S1P (1 μM) exerted the neuroprotective effect by activation of Sphk1 and S1P1 receptor gene expression. Moreover, S1P downregulated Bax and harakiri, death protein 5 (Hrk/DP5) expression and enhanced cell viability in MPP+-treated cells. The neuroprotective mechanism of S1P is mainly dependent on S1P1 receptor signalling, which was indicated by using specific agonists and antagonists of S1P1 receptor. The results show that S1P and S1P1 receptor agonists protected a significant population of neuronal cells against death.
Collapse
Affiliation(s)
- Joanna Pyszko
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | | |
Collapse
|
35
|
Hemmati F, Dargahi L, Nasoohi S, Omidbakhsh R, Mohamed Z, Chik Z, Naidu M, Ahmadiani A. Neurorestorative effect of FTY720 in a rat model of Alzheimer's disease: Comparison with Memantine. Behav Brain Res 2013; 252:415-21. [DOI: 10.1016/j.bbr.2013.06.016] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 06/03/2013] [Accepted: 06/07/2013] [Indexed: 12/14/2022]
|
36
|
Effects of small interfering RNA targeting sphingosine kinase-1 gene on the animal model of Alzheimer’s disease. ACTA ACUST UNITED AC 2013; 33:427-432. [DOI: 10.1007/s11596-013-1136-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Indexed: 12/21/2022]
|
37
|
Alshaker H, Sauer L, Monteil D, Ottaviani S, Srivats S, Böhler T, Pchejetski D. Therapeutic potential of targeting SK1 in human cancers. Adv Cancer Res 2013; 117:143-200. [PMID: 23290780 DOI: 10.1016/b978-0-12-394274-6.00006-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Sphingosine kinase 1 (SK1) is a lipid enzyme with oncogenic properties that converts the proapoptotic lipids ceramide and sphingosine into the antiapoptotic lipid sphingosine-1-phosphate and activates the signal transduction pathways that lead to cell proliferation, migration, the activation of the inflammatory response, and the impairment of apoptosis. There is compelling evidence that SK1 activation contributes to cancer progression leading to increased oncogenic transformation, tumor growth, resistance to therapies, tumor neovascularization, and metastatic spread. High levels of SK1 expression or activity have been associated with a poor prognosis in several human cancers. Recent studies using cancer cell and mouse models demonstrate a significant potential for SK1-targeting therapies to synergize with the effects of chemotherapy and radiotherapy; however, until recently the absence of clinically applicable SK1 inhibitors has limited the translation of these findings into patients. With the recent discovery of SK1 inhibiting properties of a clinically approved drug FTY720 (Fingolimod), SK1 has gained significant attention from both clinicians and the pharmaceutical industry and it is hoped that trials of newly developed SK1 inhibitors may follow soon. This review provides an overview of the SK1 signaling, its relevance to cancer progression, and the potential clinical significance of targeting SK1 for improved local or systemic control of human cancers.
Collapse
Affiliation(s)
- Heba Alshaker
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
The role of sphingolipids as bioactive signaling molecules that can regulate cell fate decisions puts them at center stage for cancer treatment and prevention. While ceramide and sphingosine have been established as antigrowth molecules, sphingosine-1-phosphate (S1P) offers a progrowth message to cells. The enzymes responsible for maintaining the balance between these "stop" or "go" signals are the sphingosine kinases (SK), SK1 and SK2. While the relative contribution of SK2 is still being elucidated and may involve an intranuclear role, a substantial amount of evidence suggests that regulation of sphingolipid levels by SK1 is an important component of carcinogenesis. Here, we review the literature regarding the role of SK1 as an oncogene that can function to enhance cancer cell viability and promote tumor growth and metastasis; highlighting the importance of developing specific SK1 inhibitors to supplement current cancer therapies.
Collapse
Affiliation(s)
- Linda A Heffernan-Stroud
- Molecular and Cellular Biology and Pathobiology Program, Medical University of South Carolina, Charleston, SC, USA
| | | |
Collapse
|
39
|
Administration of glucosylceramide ameliorated the memory impairment in aged mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:824120. [PMID: 23690856 PMCID: PMC3638615 DOI: 10.1155/2013/824120] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 03/06/2013] [Indexed: 12/27/2022]
Abstract
The function and the role of glucosylceramide have not been well studied in the central nervous system. This study was aimed to investigate the possible roles of glucosylceramide in memory function in aged mice. Glucosylceramide (50 mg/kg, p.o.) showed memory enhancing activity after 3-month treatment in the aged mice (C56BL/6, 18–20 months old) through Y-maze, novel objective test, and Morris water maze test. Long-term treatment of glucosylceramide decreased the expression of iNOS and COX-2 in the brain of aged mice. The LPS-induced mRNA level of iNOS, COX-2, IL-1β, and TNF-α was reduced by the acute treatment with glucosylceramide in adult mice. These results suggest that glucosylceramide plays an important role in anti-inflammatory and memory enhancement, and it could be a potential new therapeutic agent for the treatment of neurodegenerative diseases such as Alzheimer's disease.
Collapse
|
40
|
Asle-Rousta M, Kolahdooz Z, Oryan S, Ahmadiani A, Dargahi L. FTY720 (fingolimod) attenuates beta-amyloid peptide (Aβ42)-induced impairment of spatial learning and memory in rats. J Mol Neurosci 2013; 50:524-32. [PMID: 23435938 DOI: 10.1007/s12031-013-9979-6] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 02/08/2013] [Indexed: 12/29/2022]
Abstract
Imbalanced lipid metabolism and increase in the ceramide-to-S1P ratio in the brain have been postulated to play a role in amyloidogenesis, neuroinflammatory reactions, and neuronal apoptosis in Alzheimer's disease (AD) pathology. FTY720, the immunomodulatory sphingosine 1-phosphate (S1P) analog, has recently gained interest because of its CNS-directed effects. In addition to its immunomodulatory functions in multiple sclerosis, FTY720 possesses anti-inflammatory and neuroprotective roles in different cerebral ischemia models. In the present study, we examined the effects of FTY720 in a rat model of AD. Memory deficit was induced by bilateral intrahippocampus injection of beta-amyloid peptide (Aβ(42)) and examined through the Morris water maze test. The extent of histological injury in the hippocampus and the activation of caspase-3 were determined respectively by Nissl staining and Western blotting. Chronic daily administration of FTY720 (1 mg/kg, i.p., 14 days) significantly attenuated the Aβ(42)-induced learning and memory impairment and prevented the hippocampus neuronal damage as well as caspase-3 activation. These data show for the first time that FTY720 has a beneficial effect in restoring memory loss in Aβ(42)-induced neurotoxicity and also suggest that S1P receptors and signaling pathways may provide a potential target for the treatment of AD.
Collapse
Affiliation(s)
- Masoumeh Asle-Rousta
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Evin, Tehran, 19615-1178, Iran
| | | | | | | | | |
Collapse
|
41
|
Hyperphosphorylation of tau by GSK-3β in Alzheimer’s disease: The interaction of Aβ and sphingolipid mediators as a therapeutic target. Transl Neurosci 2013. [DOI: 10.2478/s13380-013-0144-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
AbstractAlzheimer’s disease (AD) is a progressive neurodegenerative disorder characterized by the extracellular deposits of β amyloid peptides (Aβ) in senile plaques, and intracellular aggregates of hyperphosphorylated tau in neurofibrillary tangles (NFT). Although accumulation of Aβ has been long considered a leading hypothesis in the disease pathology, it is increasingly evident that the role hyperphosphorylation of tau in destabilization of microtubule assembly and disturbance of axonal transport is equally detrimental in the neurodegenerative process. The main kinase involved in phosphorylation of tau is glycogen-synthase kinase 3-beta (GSK-3β). Intracellular accumulation of Aβ also likely induces increase in hyperphosphorylated tau by a mechanism dependent on GSK-3β. In addition, Aβ affects production of ceramides, the major sphingolipids in mammalian cells, by acting on sphingomyelinases, enzymes responsible for the catabolic formation of ceramides from the sphingomyelin. Generated ceramides in turn increase production of Aβ by acting on β-secretase, a key enzyme in the proteolytic processing of the amyloid precursor protein (APP), altogether leading to a ceramide-Aβ-hyperphosphorylated tau cascade that ends in neuronal death. Modulators and inhibitors acting on members of this devastating cascade are considered as potential targets for AD therapy. There is still no adequate treatment for AD patients. Novel therapeutic strategies increasingly consider the combination of multiple targets and interactions among the key members of implicated molecular pathways. This review summarizes recent findings and therapeutic perspectives in the pathology and treatment of AD, with the emphasis on the interplay between hyperphosphorylated tau, amyloid β, and sphingolipid mediators.
Collapse
|
42
|
The control of the balance between ceramide and sphingosine-1-phosphate by sphingosine kinase: Oxidative stress and the seesaw of cell survival and death. Comp Biochem Physiol B Biochem Mol Biol 2012; 163:26-36. [DOI: 10.1016/j.cbpb.2012.05.006] [Citation(s) in RCA: 158] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 05/09/2012] [Accepted: 05/12/2012] [Indexed: 12/19/2022]
|
43
|
Choi JW, Chun J. Lysophospholipids and their receptors in the central nervous system. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1831:20-32. [PMID: 22884303 DOI: 10.1016/j.bbalip.2012.07.015] [Citation(s) in RCA: 215] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 07/17/2012] [Accepted: 07/18/2012] [Indexed: 02/05/2023]
Abstract
Lysophosphatidic acid (LPA) and sphingosine 1-phosphate (S1P), two of the best-studied lysophospholipids, are known to influence diverse biological events, including organismal development as well as function and pathogenesis within multiple organ systems. These functional roles are due to a family of at least 11 G protein-coupled receptors (GPCRs), named LPA(1-6) and S1P(1-5), which are widely distributed throughout the body and that activate multiple effector pathways initiated by a range of heterotrimeric G proteins including G(i/o), G(12/13), G(q) and G(s), with actual activation dependent on receptor subtypes. In the central nervous system (CNS), a major locus for these signaling pathways, LPA and S1P have been shown to influence myriad responses in neurons and glial cell types through their cognate receptors. These receptor-mediated activities can contribute to disease pathogenesis and have therapeutic relevance to human CNS disorders as demonstrated for multiple sclerosis (MS) and possibly others that include congenital hydrocephalus, ischemic stroke, neurotrauma, neuropsychiatric disorders, developmental disorders, seizures, hearing loss, and Sandhoff disease, based upon the experimental literature. In particular, FTY720 (fingolimod, Gilenya, Novartis Pharma, AG) that becomes an analog of S1P upon phosphorylation, was approved by the FDA in 2010 as a first oral treatment for MS, validating this class of receptors as medicinal targets. This review will provide an overview and update on the biological functions of LPA and S1P signaling in the CNS, with a focus on results from studies using genetic null mutants for LPA and S1P receptors. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.
Collapse
Affiliation(s)
- Ji Woong Choi
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | |
Collapse
|
44
|
Annulus fibrosus cells interact with neuron-like cells to modulate production of growth factors and cytokines in symptomatic disc degeneration. Spine (Phila Pa 1976) 2012; 37:2-9. [PMID: 21386768 DOI: 10.1097/brs.0b013e31820cd2d8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN We hypothesized that AF/neuron interactions during annular injury were involved in neovascularization and nerve ingrowth, the pathologic hallmarks of symptomatic disc degeneration. OBJECTIVE To identify growth factors and inflammatory cytokines related to AF/neuron interactions using in vitro model. SUMMARY OF BACKGROUND DATA Discogenic pain is the chronic intractable pain initiated by tears in the outer annulus fibrosus (AF); this is a unique structure with free nerve endings at outer one-third, located beside dorsal root ganglia. The relationship between AF and neuron cells in annular injury has not been extensively investigated. METHODS Human AF cells were cocultured with a retinoic acid (RA)-treated SH-SY5Y human neuroblastoma cell line (neuron-like cells). Conditioned media from cells cultured alone or in coculture were assayed for growth factors and inflammatory cytokines using enzyme-linked immunosorbent assays. The responses of the neuron-like cells, the AF cells, and the cocultured group to IL-1β/TNF-α were compared using the same outcome measures. RESULTS RA-treated SH-SY5Y cells showed significant neurite outgrowth on the 7th day; this is a typical morphologic finding of neuron-like cells. Neuron-like cells produced vascular endothelial growth factor (VEGF) and IGF-1 under basal conditions and dose-dependently secreted small amounts of IL-8 in response to TNF-α. Coculturing enhanced the secretion of VEGF, TGF-β1, and β-NGF, and suppressed the production of IGF-1. VEGF in the coculture group and the AF cells was downregulated by IL-1β/TNF-α stimulation. IL-1β/TNF-α stimulation enhanced the production of large amounts of IL-6 and IL-8 from AF cells; IL-1β produced a greater response than TNF-α. The neuron-like cells did not produce detectable amounts of IL-6 or IL-8. CONCLUSION These studies suggest that AF cells are involved in an inflammatory reaction and that the interactions between AF and neuron-like cells enhance the production of growth factors responsible for neovascularization and nerve ingrowth. AF injury has the potential to initiate neovascularization/nerve ingrowth and an inflammatory reaction through the interactions of AF and neural tissues.
Collapse
|
45
|
RNAi screening in glioma stem-like cells identifies PFKFB4 as a key molecule important for cancer cell survival. Oncogene 2011; 31:3235-43. [PMID: 22056879 DOI: 10.1038/onc.2011.490] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The concept of cancer stem-like cells (CSCs) has gained considerable attention in various solid tumors including glioblastoma, the most common primary brain tumor. This sub-population of tumor cells has been intensively investigated and their role in therapy resistance as well as tumor recurrence has been demonstrated. In that respect, development of therapeutic strategies that target CSCs (and possibly also the tumor bulk) appears a promising approach in patients suffering from primary brain tumors. In the present study, we utilized RNA interference (RNAi) to screen the complete human kinome and phosphatome (682 and 180 targets, respectively) in order to identify genes and pathways relevant for the survival of brain CSCs and thereby potential therapeutical targets for glioblastoma. We report of 46 putative candidates including known survival-related kinases and phosphatases. Interestingly, a number of genes identified are involved in metabolism, especially glycolysis, such as PDK1 and PKM2 and, most prominently PFKFB4. In vitro studies confirmed an essential role of PFKFB4 in the maintenance of brain CSCs. Furthermore, high PFKFB4 expression was associated with shorter survival of primary glioblastoma patients. Our findings support the importance of the glycolytic pathway in the maintenance of malignant glioma cells and brain CSCs and imply tumor metabolism as a promising therapeutic target in glioblastoma.
Collapse
|
46
|
Jung JC, Lee Y, Moon S, Ryu JH, Oh S. Phytoceramide shows neuroprotection and ameliorates scopolamine-induced memory impairment. Molecules 2011; 16:9090-100. [PMID: 22037667 PMCID: PMC6264402 DOI: 10.3390/molecules16119090] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 10/22/2011] [Accepted: 10/26/2011] [Indexed: 01/03/2023] Open
Abstract
The function and the role phytoceramide (PCER) and phytosphingosine (PSO) in the central nervous system has not been well studied. This study was aimed at investigating the possible roles of PCER and PSO in glutamate-induced neurotoxicity in cultured neuronal cells and memory function in mice. Phytoceramide showed neuro-protective activity in the glutamate-induced toxicity in cultured cortical neuronal cells. Neither phytosphingosine nor tetraacetylphytosphingosine (TAPS) showed neuroproective effects in neuronal cells. PCER (50 mg/kg, p.o.) recovered the scopolamine-induced reduction in step-through latency in the passive avoidance test; however, PSO did not modulate memory function on this task. The ameliorating effects of PCER on spatial memory were confirmed by the Morris water maze test. In conclusion, through behavioral and neurochemical experimental results, it was demonstrated that central administration of PCER produces amelioration of memory impairment. These results suggest that PCER plays an important role in neuroprotection and memory enhancement and PCER could be a potential new therapeutic agent for the treatment of neurodegenerative diseases such as Alzheimer's disease.
Collapse
Affiliation(s)
- Jae-Chul Jung
- Institute of Life Science Research, Rexgene Biotech, Ochang, Chungbuk 368-831, Korea
| | - Yeonju Lee
- Department of Neuroscience and TIDRC, School of Medicine, Ewha Womans University, Seoul 158-710, Korea
| | - Sohyeon Moon
- Department of Neuroscience and TIDRC, School of Medicine, Ewha Womans University, Seoul 158-710, Korea
| | - Jong Hoon Ryu
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 130-701, Korea
| | - Seikwan Oh
- Department of Neuroscience and TIDRC, School of Medicine, Ewha Womans University, Seoul 158-710, Korea
| |
Collapse
|
47
|
Barth BM, Gustafson SJ, Kuhn TB. Neutral sphingomyelinase activation precedes NADPH oxidase-dependent damage in neurons exposed to the proinflammatory cytokine tumor necrosis factor-α. J Neurosci Res 2011; 90:229-42. [PMID: 21932365 DOI: 10.1002/jnr.22748] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Revised: 06/16/2011] [Accepted: 06/27/2011] [Indexed: 12/26/2022]
Abstract
Inflammation accompanied by severe oxidative stress plays a vital role in the orchestration and progression of neurodegeneration prevalent in chronic and acute central nervous system pathologies as well as in aging. The proinflammatory cytokine tumor necrosis factor-α (TNFα) elicits the formation of the bioactive ceramide by stimulating the hydrolysis of the membrane lipid sphingomyelin by sphingomyelinase activities. Ceramide stimulates the formation of reactive oxygen species (ROS) and apoptotic mechanisms in both neurons and nonneuronal cells, establishing a link between sphingolipid metabolism and oxidative stress. We demonstrated in SH-SY5Y human neuroblastoma cells and primary cortical neurons that TNFα is a potent stimulator of Mg(2+) -dependent neutral sphingomyelinase (Mg(2+) -nSMase) activity, and sphingomyelin hydrolysis, rather than de novo synthesis, was the predominant source of ceramide increases. Mg(2+) -nSMase activity preceded an accumulation of ROS by a neuronal NADPH oxidase (NOX). Notably, TNFα provoked an NOX-dependent oxidative damage to sphingosine kinase-1, which generates sphingosine-1-phosphate, a ceramide metabolite associated with neurite outgrowth. Indeed, ceramide and ROS inhibited neurite outgrowth of dorsal root ganglion neurons by disrupting growth cone motility. Blunting ceramide and ROS formation both rescued sphingosine kinase-1 activity and neurite outgrowth. Our studies suggest that TNFα-mediated activation of Mg(2+) -nSMase and NOX in neuronal cells not only produced the neurotoxic intermediates ceramide and ROS but also directly antagonized neuronal survival mechanisms, thus accelerating neurodegeneration.
Collapse
Affiliation(s)
- Brian M Barth
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, USA
| | | | | |
Collapse
|
48
|
Abstract
Sphingosine kinase 1 (SK1) is a lipid enzyme with oncogenic properties that converts the proapoptotic lipid sphingosine into the antiapoptotic lipid sphingosine-1-phosphate, which activates the signal transduction pathways that lead to cell proliferation, migration, activation of the inflammatory response and impairment of apoptosis. Compelling evidence suggests that SK1 activation contributes to cancer progression leading to increased oncogenic transformation, tumor growth, resistance to therapies, tumor neovascularization and metastatic spread. High levels of SK1 expression or activity have been associated with poor prognosis in several cancers, including those of the prostate. Recent studies using prostate cancer cell and mouse models demonstrate a significant potential for SK1-targeting therapies to synergize with the effects of docetaxel chemotherapy and radiotherapy. However, until recently the absence of clinically applicable SK1 inhibitors has limited the translation of these findings into patients. With the recent discovery that clinically approved drug fingolimod has SK1-inhibiting properties, SK1 has gained significant attention from both clinicians and the pharmaceutical industry and it is hoped that trials of newly developed SK1 inhibitors might follow soon.
Collapse
|
49
|
Lang F, Ullrich S, Gulbins E. Ceramide formation as a target in beta-cell survival and function. Expert Opin Ther Targets 2011; 15:1061-71. [PMID: 21635197 DOI: 10.1517/14728222.2011.588209] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Ceramide may be synthesized de novo or generated by sphingomyelinase-dependent hydrolysis of sphingomyelin. AREAS COVERED The role of ceramide, ceramide-sensitive signaling and ion channels in β-cell apoptosis, lipotoxicity and amyloid-induced β-cell death. EXPERT OPINION Ceramide participates in β-cell dysfunction and apoptosis after exposure to TNFα, IL-1β and IFN-γ, excessive amyloid and islet amyloid polypeptide or non-esterified fatty acids (lipotoxicity). Knockout of sphingomyelin synthase 1, which converts ceramide to sphingomyelin, leads to impairment of insulin secretion. Increased ceramidase activity or pharmacological inhibition of ceramide synthetase, inhibits β-cell apoptosis. Ceramide contributes to endoplasmatic reticulum (ER) stress, decreased mitochondrial membrane potential in insulin-secreting cells and mitochondrial release of cytochrome c into the cytosol, which are all triggers of apoptotic cell death. Ceramide-dependent signaling involves activation of extracellularly regulated kinases 1 and 2 (ERK1/2), downregulation of Period (Per)-aryl hydrocarbon receptor nuclear translocator (Arnt)-single-minded (Sim) kinase (PASK), activation of okadaic-acid-sensitive protein phosphatase 2A (PP2A) and stimulation of NADPH-oxidase with generation of superoxides and lipid peroxides. Ceramide reduces the activity of voltage gated potassium (Kv)-channels in insulin-secreting cells. The role of ceramide in β-cell survival and function may be therapeutically relevant, because ceramide formation can be suppressed by pharmacological inhibition of ceramide synthetase and/or sphingomyelinase.
Collapse
Affiliation(s)
- Florian Lang
- University of Tübingen, Institute of Physiology, Germany.
| | | | | |
Collapse
|
50
|
Perrone L, Mothes E, Vignes M, Mockel A, Figueroa C, Miquel MC, Maddelein ML, Faller P. Copper transfer from Cu-Abeta to human serum albumin inhibits aggregation, radical production and reduces Abeta toxicity. Chembiochem 2010; 11:110-8. [PMID: 19937895 DOI: 10.1002/cbic.200900474] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Amyloid-beta peptides (Abeta) and the protein human serum albumin (HSA) interact in vivo. They are both localised in the blood plasma and in the cerebrospinal fluid. Among other functions, HSA is involved in the transport of the essential metal copper. Complexes between Abeta and copper ions have been proposed to be an aberrant interaction implicated in the development of Alzheimer's disease, where Cu is involved in Abeta aggregation and production of reactive oxygen species (ROS). In the present work, we studied copper-exchange reaction between Abeta and HSA or the tetrapeptide DAHK (N-terminal Cu-binding domain of HSA) and the consequence of this exchange on Abeta-induced ROS production and cell toxicity. The following results were obtained: 1) HSA and DAHK removed Cu(II) from Abeta rapidly and stoichiometrically, 2) HSA and DAHK were able to decrease Cu-induced aggregation of Abeta, 3) HSA and DAHK suppressed the catalytic HO(.) production in vitro and ROS production in neuroblastoma cells generated by Cu-Abeta and ascorbate, 4) HSA and DAHK were able to rescue these cells from the toxicity of Cu-Abeta with ascorbate, 5) DAHK was more potent in ROS suppression and restoration of neuroblastoma cell viability than HSA, in correlation with an easier reduction of Cu(II)-HSA than Cu-DAHK by ascorbate, in vitro. Our data suggest that HSA is able to decrease aberrant Cu(II)-Abeta interaction. The repercussion of the competition between HSA and Abeta to bind Cu in the blood and brain and its relation to Alzheimer's disease are discussed.
Collapse
Affiliation(s)
- Lorena Perrone
- CNRS UMR 5089, Institut de Pharmacologie et de Biologie Structurale, 205 route de Narbonne, Toulouse, France
| | | | | | | | | | | | | | | |
Collapse
|