1
|
Godoy P, Rezanezhad Dizaji B, Zardini Buzatto A, Sanchez L, Li L. The Lipid Composition of the Exo-Metabolome from Haemonchus contortus. Metabolites 2025; 15:193. [PMID: 40137157 PMCID: PMC11944095 DOI: 10.3390/metabo15030193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/24/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
Background/Objectives: Metabolomic studies of different parasite-derived biomolecules, such as lipids, are needed to broaden the discovery of novel targets and overcome anthelmintic resistance. Lipids are involved in diverse functions in biological systems, including parasitic helminths, but little is known about their role in the biology of these organisms and their impact on host-parasite interactions. This study aimed to characterize the lipid profile secreted by Haemonchus contortus, the major parasitic nematodes of farm ruminants. Methods: H. contortus adult worms were recovered from infected sheep and cultured ex vivo. Parasite medium was collected at different time points and samples were subjected to an untargeted global lipidomic analysis. Lipids were extracted and subjected to Liquid Chromatography-Mass Spectrometry (LC-MS/MS). Annotated lipids were normalized and subjected to statistical analysis. Lipid clusters' fold change (FC) and individual lipid features were compared at different time points. Lipids were also analyzed by structural composition and saturation bonding. Results: A total of 1057 H. contortus lipid features were annotated, including glycerophospholipids, fatty acyls, sphingolipids, glycerolipids, and sterols. Most of these compounds were unsaturated lipids. We found significant FC differences in the lipid profile in a time-dependent manner. Conclusions: We predict that many lipids found in our study act as signaling molecules for nematodes' physiological functions, such as adaptation to nutrient changes, life span and mating, and as modulators on the host immune responses.
Collapse
Affiliation(s)
- Pablo Godoy
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (B.R.D.); (L.S.)
- Independent Researcher and Animal Health Consultant, Montreal, QC H4A 2V2, Canada
| | - Behrouz Rezanezhad Dizaji
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (B.R.D.); (L.S.)
| | | | - Laura Sanchez
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (B.R.D.); (L.S.)
| | - Liang Li
- The Metabolomics Innovation Centre (TMIC), Edmonton, AB T6G 2E9, Canada
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2N4, Canada
| |
Collapse
|
2
|
Serafini S, O’Flaherty C. Sphingolipids modulate redox signalling during human sperm capacitation. Hum Reprod 2025; 40:210-225. [PMID: 39658334 PMCID: PMC11788196 DOI: 10.1093/humrep/deae268] [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: 07/02/2024] [Revised: 10/30/2024] [Indexed: 12/12/2024] Open
Abstract
STUDY QUESTION What role do sphingolipids have in mediating human sperm capacitation? SUMMARY ANSWER Sphingosine 1-phosphate (S1P) mediates the acquisition of fertilizing competency in human spermatozoa by engaging with its Gi-coupled receptor S1PR1 and promoting production of reactive oxygen species such as nitric oxide and superoxide anion. WHAT IS KNOWN ALREADY Bioactive sphingolipids, such as S1P, are fundamental for regulating numerous physiological domains and processes, such as cell membranes and signalling, cell death and proliferation, cell migration and invasiveness, inflammation, and central nervous system development. STUDY DESIGN, SIZE, DURATION Semen samples were obtained from a cohort of 10 healthy non-smoking volunteers (18-30 years old) to investigate the role of S1P in sperm. PARTICIPANTS/MATERIALS, SETTING, METHODS Percoll-selected human spermatozoa were incubated at 37°C for 3.5 h in BWW media with or without foetal cord serum ultrafiltrate (FCSu), sphingosine (Sph), or ceramide (Cer). Spermatozoa were also incubated with or without pharmacological inhibitors of sphingolipid metabolism. Protein tyrosine phosphorylation was determined by immunoblotting. The acrosome reaction was determined by PSA-FTIC labelling of the acrosome and analysed using fluorescence microscopy. Intracellular nitric oxide (NO•) production was determined using a DAF-2DA probe. Immunocytochemistry was performed to localize and assess the functional relationship of key components of lipid signalling in spermatozoa. Sperm viability and motility of the samples were evaluated by the hypo-osmotic swelling (HOS) test and computer-aided sperm analysis (CASA). Statistical differences between groups were determined using ANOVA and Tukey's test. Normal distribution of the data and variance homogeneity were assessed using Shapiro-Wilk and Levene's test, respectively. A difference was considered significant when the P-value was ≤0.05. MAIN RESULTS AND THE ROLE OF CHANCE S1P mediates the acquisition of fertilizing competency in human spermatozoa by engaging with its Gi-coupled receptor S1PR1. We found that S1PR1 redistributes to the post-acrosomal region upon induction of capacitation. S1P signalling promotes the activation of the PI3K-AKT pathway, leading to NO• production during sperm capacitation. L-NAME, an nitric oxide synthase inhibitor, impaired the Sph- and Cer-dependent capacitation. Additionally, Sph and Cer promote superoxide anion (O2•-) production, and the extracellular addition of superoxide dismutase (SOD) prevented Sph- and Cer-dependent capacitation, suggesting that Sph and Cer stimulate O2•- production during sperm capacitation. Protein kinase type R (PKR), ceramide kinase (CERK), and protein kinase C (PKC) are responsible for translocating and activating sphingosine kinase 1 (SphK1), which is necessary to promote S1P production for sperm capacitation. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION The utilization and actions of sphingolipids may differ in spermatozoa of different species. WIDER IMPLICATIONS OF THE FINDINGS Sphingolipid metabolites such as Sph, Cer, S1P, and ceramide 1-phosphate (C1P) play a crucial role in inducing human sperm capacitation. Our research has provided new insights into fundamental sphingolipid processes in human sperm, including the importance of C1P in translocating and activating SphK1 as well as the S1P signalling to regulate the PI3K/AKT/NOS pathway to generate NO• for sperm capacitation. We are the first to identify the presence of PKR in human spermatozoa and its role in the phosphorylation activities of SphK1 with the subsequent activation of S1P signalling. Furthermore, our study has identified that S1PR1 and S1PR3 are involved in capacitation and the acrosome reaction, respectively. These findings shed light on a novel mechanism by which sphingolipids drive capacitation in human sperm and pave the way for further exploration of the role of bioactive sphingolipid metabolites in this process. Lastly, our studies lay the foundation for examining the lipid profile of infertile males, as potential discrepancies can affect the functional capacity of spermatozoa to reach fertilizing potential. STUDY FUNDING/COMPETING INTEREST(S) This research was funded by the Canadian Institutes of Health Research (CIHR), grant number PJT-165962 to C.O.F. S.S. was awarded a Research Institute-MUHC Desjardins Studentship. There are no competing interests to report.
Collapse
Affiliation(s)
- Steven Serafini
- Experimental Medicine Division, Department of Medicine, McGill University, Montréal, QC, Canada
- Urology Division, Department of Surgery, McGill University, Montréal, QC, Canada
- The Research Institute, McGill University Health Centre, Montréal, QC, Canada
| | - Cristian O’Flaherty
- Experimental Medicine Division, Department of Medicine, McGill University, Montréal, QC, Canada
- Urology Division, Department of Surgery, McGill University, Montréal, QC, Canada
- The Research Institute, McGill University Health Centre, Montréal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University, Montréal, QC, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| |
Collapse
|
3
|
Shahi N, Thapliyal S, Babu K. Sensory modulation of neuropeptide signaling by CASY-1 gates cholinergic transmission at Caenorhabditis elegans neuromuscular junction. J Biosci 2025; 50:4. [PMID: 39912398 PMCID: PMC7617471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
The neuromuscular junction (NMJ) is crucial for understanding the fundamentals of synaptic transmission and activity. Various modulators operate within neuronal circuits, from sensory to motor neurons, to influence synaptic transmission at the NMJ. This study sheds light on the regulation of sensory-evoked cholinergic neurotransmission at motor neurons orchestrated by CASY-1, the mammalian calsyntenin orthologue. We report that the increased excitation-inhibition (E-I) ratio at the NMJ in casy-1 mutants is likely due to its interactions with neuromodulators in sensory neurons. We explored the intricate genetic interactions of CASY- 1 with the neuropeptide FLP-21 and its receptor, NPR-1, both of which display simultaneous alterations in cholinergic signaling at the NMJ. Through genetic, pharmacological, and bioimaging-based experiments, we proposed a mechanism by which CASY-1 potentially interacts with the neuropeptide-carrying vesicles to regulate synaptic transmission. The nematode Caenorhabditis elegans serves as an ideal model system for this study, enabling detailed insights into neuromodulatory mechanisms in the neuronal circuit.
Collapse
Affiliation(s)
- Navneet Shahi
- Centre for Neuroscience, Indian Institute of Science (IISc), Bangalore, Karnataka, India
| | - Shruti Thapliyal
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Mohali, Punjab, India
| | - Kavita Babu
- Centre for Neuroscience, Indian Institute of Science (IISc), Bangalore, Karnataka, India
| |
Collapse
|
4
|
Buckley M, Jacob WP, Bortey L, McClain ME, Ritter AL, Godfrey A, Munneke AS, Ramachandran S, Kenis S, Kolnik JC, Olofsson S, Nenadovich M, Kutoloski T, Rademacher L, Alva A, Heinecke O, Adkins R, Parkar S, Bhagat R, Lunato J, Beets I, Francis MM, Kowalski JR. Cell non-autonomous signaling through the conserved C. elegans glycoprotein hormone receptor FSHR-1 regulates cholinergic neurotransmission. PLoS Genet 2024; 20:e1011461. [PMID: 39561202 PMCID: PMC11614273 DOI: 10.1371/journal.pgen.1011461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 12/03/2024] [Accepted: 10/14/2024] [Indexed: 11/21/2024] Open
Abstract
Modulation of neurotransmission is key for organismal responses to varying physiological contexts such as during infection, injury, or other stresses, as well as in learning and memory and for sensory adaptation. Roles for cell autonomous neuromodulatory mechanisms in these processes have been well described. The importance of cell non-autonomous pathways for inter-tissue signaling, such as gut-to-brain or glia-to-neuron, has emerged more recently, but the cellular mechanisms mediating such regulation remain comparatively unexplored. Glycoproteins and their G protein-coupled receptors (GPCRs) are well-established orchestrators of multi-tissue signaling events that govern diverse physiological processes through both cell-autonomous and cell non-autonomous regulation. Here, we show that follicle stimulating hormone receptor, FSHR-1, the sole Caenorhabditis elegans ortholog of mammalian glycoprotein hormone GPCRs, is important for cell non-autonomous modulation of synaptic transmission. Inhibition of fshr-1 expression reduces muscle contraction and leads to synaptic vesicle accumulation in cholinergic motor neurons. The neuromuscular and locomotor defects in fshr-1 loss-of-function mutants are associated with an underlying accumulation of synaptic vesicles, build-up of the synaptic vesicle priming factor UNC-10/RIM, and decreased synaptic vesicle release from cholinergic motor neurons. Restoration of FSHR-1 to the intestine is sufficient to restore neuromuscular activity and synaptic vesicle localization to fshr-1-deficient animals. Intestine-specific knockdown of FSHR-1 reduces neuromuscular function, indicating FSHR-1 is both necessary and sufficient in the intestine for its neuromuscular effects. Re-expression of FSHR-1 in other sites of endogenous expression, including glial cells and neurons, also restored some neuromuscular deficits, indicating potential cross-tissue regulation from these tissues as well. Genetic interaction studies provide evidence that downstream effectors gsa-1/GαS, acy-1/adenylyl cyclase and sphk-1/sphingosine kinase and glycoprotein hormone subunit orthologs, GPLA-1/GPA2 and GPLB-1/GPB5, are important for intestinal FSHR-1 modulation of the NMJ. Together, our results demonstrate that FSHR-1 modulation directs inter-tissue signaling systems, which promote synaptic vesicle release at neuromuscular synapses.
Collapse
Affiliation(s)
- Morgan Buckley
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - William P. Jacob
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Letitia Bortey
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Makenzi E. McClain
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Alyssa L. Ritter
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Amy Godfrey
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Allyson S. Munneke
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Shankar Ramachandran
- Department of Neurobiology, University of Massachusetts Chan School of Medicine, Worcester, Massachusetts, United States of America
| | - Signe Kenis
- Neural Signaling and Circuit Plasticity Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Julie C. Kolnik
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Sarah Olofsson
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Milica Nenadovich
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Tanner Kutoloski
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Lillian Rademacher
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Alexandra Alva
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Olivia Heinecke
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Ryan Adkins
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Shums Parkar
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Reesha Bhagat
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Jaelin Lunato
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Isabel Beets
- Neural Signaling and Circuit Plasticity Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Michael M. Francis
- Department of Neurobiology, University of Massachusetts Chan School of Medicine, Worcester, Massachusetts, United States of America
| | - Jennifer R. Kowalski
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| |
Collapse
|
5
|
Buckley M, Jacob WP, Bortey L, McClain M, Ritter AL, Godfrey A, Munneke AS, Ramachandran S, Kenis S, Kolnik JC, Olofsson S, Adkins R, Kutoloski T, Rademacher L, Heinecke O, Alva A, Beets I, Francis MM, Kowalski JR. Cell non-autonomous signaling through the conserved C. elegans glycopeptide hormone receptor FSHR-1 regulates cholinergic neurotransmission. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.10.578699. [PMID: 38405708 PMCID: PMC10888917 DOI: 10.1101/2024.02.10.578699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Modulation of neurotransmission is key for organismal responses to varying physiological contexts such as during infection, injury, or other stresses, as well as in learning and memory and for sensory adaptation. Roles for cell autonomous neuromodulatory mechanisms in these processes have been well described. The importance of cell non-autonomous pathways for inter-tissue signaling, such as gut-to-brain or glia-to-neuron, has emerged more recently, but the cellular mechanisms mediating such regulation remain comparatively unexplored. Glycoproteins and their G protein-coupled receptors (GPCRs) are well-established orchestrators of multi-tissue signaling events that govern diverse physiological processes through both cell-autonomous and cell non-autonomous regulation. Here, we show that follicle stimulating hormone receptor, FSHR-1, the sole Caenorhabditis elegans ortholog of mammalian glycoprotein hormone GPCRs, is important for cell non-autonomous modulation of synaptic transmission. Inhibition of fshr-1 expression reduces muscle contraction and leads to synaptic vesicle accumulation in cholinergic motor neurons. The neuromuscular and locomotor defects in fshr-1 loss-of-function mutants are associated with an underlying accumulation of synaptic vesicles, build-up of the synaptic vesicle priming factor UNC-10/RIM, and decreased synaptic vesicle release from cholinergic motor neurons. Restoration of FSHR-1 to the intestine is sufficient to restore neuromuscular activity and synaptic vesicle localization to fshr-1- deficient animals. Intestine-specific knockdown of FSHR-1 reduces neuromuscular function, indicating FSHR-1 is both necessary and sufficient in the intestine for its neuromuscular effects. Re-expression of FSHR-1 in other sites of endogenous expression, including glial cells and neurons, also restored some neuromuscular deficits, indicating potential cross-tissue regulation from these tissues as well. Genetic interaction studies provide evidence that downstream effectors gsa-1 / Gα S , acy-1 /adenylyl cyclase and sphk-1/ sphingosine kinase and glycoprotein hormone subunit orthologs, GPLA-1/GPA2 and GPLB-1/GPB5, are important for FSHR-1 modulation of the NMJ. Together, our results demonstrate that FSHR-1 modulation directs inter-tissue signaling systems, which promote synaptic vesicle release at neuromuscular synapses.
Collapse
|
6
|
Tsentsevitsky AN, Gafurova CR, Mukhutdinova KA, Giniatullin AR, Fedorov NS, Malomouzh AI, Petrov AM. Sphingomyelinase modulates synaptic vesicle mobilization at the mice neuromuscular junctions. Life Sci 2023; 318:121507. [PMID: 36801470 DOI: 10.1016/j.lfs.2023.121507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/07/2023] [Accepted: 02/14/2023] [Indexed: 02/21/2023]
Abstract
AIMS Sphingomyelin is an abundant component of the presynaptic membrane and an organizer of lipid rafts. In several pathological conditions, sphingomyelin is hydrolyzed due to an upregulation and release of secretory sphingomyelinases (SMases). Herein, the effects of SMase on exocytotic neurotransmitter release were studied in the diaphragm neuromuscular junctions of mice. MAIN METHODS Microelectrode recordings of postsynaptic potentials and styryl (FM) dyes were used to estimate neuromuscular transmission. Membrane properties were assessed with fluorescent techniques. KEY FINDINGS Application of SMase at a low concentration (0.01 U ml-1) led to a disruption of lipid-packing in the synaptic membranes. Neither spontaneous exocytosis nor evoked neurotransmitter release (in response to single stimuli) were affected by SMase treatment. However, SMase significantly increased neurotransmitter release and the rate of fluorescent FM-dye loss from the synaptic vesicles at 10, 20 and 70 Hz stimulation of the motor nerve. In addition, SMase treatment prevented a shift of the exocytotic mode from "full-collapse" fusion to "kiss-and-run" during high-frequency (70 Hz) activity. The potentiating effects of SMase on neurotransmitter release and FM-dye unloading were suppressed when synaptic vesicle membranes were also exposed to this enzyme (i.e., stimulation occurred during SMase treatment). SIGNIFICANCE Thus, hydrolysis of the plasma membrane sphingomyelin can enhance mobilization of synaptic vesicles and facilitate full fusion mode of exocytosis, but SMase acting on vesicular membrane had a depressant effect on the neurotransmission. Partially, the effects of SMase can be related with the changes in synaptic membrane properties and intracellular signaling.
Collapse
Affiliation(s)
- Andrei N Tsentsevitsky
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky St, Box 30, Kazan, RT 420111, Russia
| | - Chulpan R Gafurova
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky St, Box 30, Kazan, RT 420111, Russia
| | - Kamilla A Mukhutdinova
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky St, Box 30, Kazan, RT 420111, Russia
| | - Arthur R Giniatullin
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky St, Box 30, Kazan, RT 420111, Russia; Kazan State Medial University, 49 Butlerova St., Kazan, RT 420012, Russia
| | - Nikita S Fedorov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky St, Box 30, Kazan, RT 420111, Russia
| | - Artem I Malomouzh
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky St, Box 30, Kazan, RT 420111, Russia
| | - Alexey M Petrov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky St, Box 30, Kazan, RT 420111, Russia; Kazan State Medial University, 49 Butlerova St., Kazan, RT 420012, Russia.
| |
Collapse
|
7
|
Cui M, Göbel V, Zhang H. Uncovering the 'sphinx' of sphingosine 1-phosphate signalling: from cellular events to organ morphogenesis. Biol Rev Camb Philos Soc 2021; 97:251-272. [PMID: 34585505 PMCID: PMC9292677 DOI: 10.1111/brv.12798] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 09/11/2021] [Accepted: 09/16/2021] [Indexed: 11/02/2022]
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid metabolite, functioning as a signalling molecule in diverse cellular processes. Over the past few decades, studies of S1P signalling have revealed that the physiological activity of S1P largely depends on S1P metabolizing enzymes, transporters and receptors on the plasma membrane, as well as on the intracellular proteins that S1P binds directly to. In addition to its roles in cancer signalling, immunity and inflammation, a large body of evidence has identified a close link of S1P signalling with organ morphogenesis. Here we discuss the vital role of S1P signalling in orchestrating various cellular events during organ morphogenesis through analysing each component along the extracellular and intracellular S1P signalling axes. For each component, we review advances in our understanding of S1P signalling and function from the upstream regulators to the downstream effectors and from cellular behaviours to tissue organization, primarily in the context of morphogenetic mechanisms. S1P-mediated vesicular trafficking is also discussed as a function independent of its signalling function. A picture emerges that reveals a multifaceted role of S1P-dependent pathways in the development and maintenance of organ structure and function.
Collapse
Affiliation(s)
- Mengqiao Cui
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China
| | - Verena Göbel
- Mucosal Immunology and Biology Research Center, Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, U.S.A
| | - Hongjie Zhang
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, 999078, China
| |
Collapse
|
8
|
Chemical and genetic rescue of in vivo progranulin-deficient lysosomal and autophagic defects. Proc Natl Acad Sci U S A 2021; 118:2022115118. [PMID: 34140407 DOI: 10.1073/pnas.2022115118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In 2006, GRN mutations were first linked to frontotemporal dementia (FTD), the leading cause of non-Alzheimer dementias. While much research has been dedicated to understanding the genetic causes of the disease, our understanding of the mechanistic impacts of GRN deficiency has only recently begun to take shape. With no known cure or treatment available for GRN-related FTD, there is a growing need to rapidly advance genetic and/or small-molecule therapeutics for this disease. This issue is complicated by the fact that, while lysosomal dysfunction seems to be a key driver of pathology, the mechanisms linking a loss of GRN to a pathogenic state remain unclear. In our attempt to address these key issues, we have turned to the nematode, Caenorhabditis elegans, to model, study, and find potential therapies for GRN-deficient FTD. First, we show that the loss of the nematode GRN ortholog, pgrn-1, results in several behavioral and molecular defects, including lysosomal dysfunction and defects in autophagic flux. Our investigations implicate the sphingolipid metabolic pathway in the regulation of many of the in vivo defects associated with pgrn-1 loss. Finally, we utilized these nematodes as an in vivo tool for high-throughput drug screening and identified two small molecules with potential therapeutic applications against GRN/pgrn-1 deficiency. These compounds reverse the biochemical, cellular, and functional phenotypes of GRN deficiency. Together, our results open avenues for mechanistic and therapeutic research into the outcomes of GRN-related neurodegeneration, both genetic and molecular.
Collapse
|
9
|
Lee K, Escobar I, Jang Y, Kim W, Ausubel FM, Mylonakis E. In the Model Host Caenorhabditis elegans, Sphingosine-1-Phosphate-Mediated Signaling Increases Immunity toward Human Opportunistic Bacteria. Int J Mol Sci 2020; 21:ijms21217813. [PMID: 33105563 PMCID: PMC7672543 DOI: 10.3390/ijms21217813] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 12/25/2022] Open
Abstract
Sphingosine-1-phophate (S1P) is a sphingolipid-derived signaling molecule that controls diverse cellular functions including cell growth, homeostasis, and stress responses. In a variety of metazoans, cytosolic S1P is transported into the extracellular space where it activates S1P receptors in a concentration-dependent manner. In the free-living nematode Caenorhabditis elegans, the spin-2 gene, which encodes a S1P transporter, is activated during Gram-positive or Gram-negative bacterial infection of the intestine. However, the role during infection of spin-2 and three additional genes in the C. elegans genome encoding other putative S1P transporters has not been elucidated. Here, we report an evolutionally conserved function for S1P and a non-canonical role for S1P transporters in the C. elegans immune response to bacterial pathogens. We found that mutations in the sphingosine kinase gene (sphk-1) or in the S1P transporter genes spin-2 or spin-3 decreased nematode survival after infection with Pseudomonas aeruginosa or Enterococcus faecalis. In contrast to spin-2 and spin-3, mutating spin-1 leads to an increase in resistance to P. aeruginosa. Consistent with these results, when wild-type C. elegans were supplemented with extracellular S1P, we found an increase in their lifespan when challenged with P. aeruginosa and E. faecalis. In comparison, spin-2 and spin-3 mutations suppressed the ability of S1P to rescue the worms from pathogen-mediated killing, whereas the spin-1 mutation had no effect on the immune-enhancing activity of S1P. S1P demonstrated no antimicrobial activity toward P. aeruginosa and Escherichia coli and only minimal activity against E. faecalis MMH594 (40 µM). These data suggest that spin-2 and spin-3, on the one hand, and spin-1, on the other hand, transport S1P across cellular membranes in opposite directions. Finally, the immune modulatory effect of S1P was diminished in C. eleganssek-1 and pmk-1 mutants, suggesting that the immunomodulatory effects of S1P are mediated by the p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Kiho Lee
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA; (K.L.); (I.E.); (Y.J.)
| | - Iliana Escobar
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA; (K.L.); (I.E.); (Y.J.)
| | - Yeeun Jang
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA; (K.L.); (I.E.); (Y.J.)
| | - Wooseong Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea;
| | - Frederick M. Ausubel
- Department of Genetics, Harvard Medical School and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA;
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA; (K.L.); (I.E.); (Y.J.)
- Correspondence: ; Tel.: +1-401-444-7856
| |
Collapse
|
10
|
McCulloch KA, Jin Y. The muscarinic agonist arecoline suppresses motor circuit hyperactivity in C. elegans. MICROPUBLICATION BIOLOGY 2020; 2020. [PMID: 32626844 PMCID: PMC7326331 DOI: 10.17912/micropub.biology.000272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Katherine A McCulloch
- Neurobiology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093
| | - Yishi Jin
- Neurobiology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093
| |
Collapse
|
11
|
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
|
12
|
Importin α5 Regulates Anxiety through MeCP2 and Sphingosine Kinase 1. Cell Rep 2019; 25:3169-3179.e7. [PMID: 30540948 PMCID: PMC6302549 DOI: 10.1016/j.celrep.2018.11.066] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 10/09/2018] [Accepted: 11/15/2018] [Indexed: 01/08/2023] Open
Abstract
Importins mediate transport from synapse to soma and from cytoplasm to nucleus, suggesting that perturbation of importin-dependent pathways should have significant neuronal consequences. A behavioral screen on five importin α knockout lines revealed that reduced expression of importin α5 (KPNA1) in hippocampal neurons specifically decreases anxiety in mice. Re-expression of importin α5 in ventral hippocampus of knockout animals increased anxiety behaviors to wild-type levels. Hippocampal neurons lacking importin α5 reveal changes in presynaptic plasticity and modified expression of MeCP2-regulated genes, including sphingosine kinase 1 (Sphk1). Knockout of importin α5, but not importin α3 or α4, reduces MeCP2 nuclear localization in hippocampal neurons. A Sphk1 blocker reverses anxiolysis in the importin α5 knockout mouse, while pharmacological activation of sphingosine signaling has robust anxiolytic effects in wild-type animals. Thus, importin α5 influences sphingosine-sensitive anxiety pathways by regulating MeCP2 nuclear import in hippocampal neurons. Reduced expression of importin α5 in hippocampal neurons decreases anxiety Importin α5 is required for nuclear localization of MeCP2 in hippocampal neurons Importin α5 knockout increases expression of Sphk1, an MeCP2-regulated gene Pharmacological modulation of Sphk1 and the S1P receptor affects anxiety
Collapse
|
13
|
Kim S, Sieburth D. Sphingosine Kinase Activates the Mitochondrial Unfolded Protein Response and Is Targeted to Mitochondria by Stress. Cell Rep 2019; 24:2932-2945.e4. [PMID: 30208318 PMCID: PMC6206875 DOI: 10.1016/j.celrep.2018.08.037] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 07/17/2018] [Accepted: 08/13/2018] [Indexed: 11/22/2022] Open
Abstract
The mitochondrial unfolded protein response (UPRmt) is critical for maintaining mitochondrial protein homeostasis in response to mitochondrial stress, but early steps in UPRmt activation are not well understood. Here, we report a function for SPHK-1 sphingosine kinase in activating the UPRmt in C. elegans. Genetic deficiency of sphk-1 in the intestine inhibits UPRmt activation, whereas selective SPHK-1 intestinal overexpression is sufficient to activate the UPRmt. Acute mitochondrial stress leads to rapid, reversible localization of SPHK-1::GFP fusion proteins with mitochondrial membranes before UPRmt activation. SPHK-1 variants lacking kinase activity or mitochondrial targeting fail to rescue the stress-induced UPRmt activation defects of sphk-1 mutants. Activation of the UPRmt by the nervous system requires sphk-1 and elicits SPHK-1 mitochondrial association in the intestine. We propose that stress-regulated mitochondrial recruitment of SPHK-1 and subsequent S1P production are critical early events for both cell autonomous and cell non-autonomous UPRmt activation. The mitochondrial unfolded protein response (UPRmt) maintains mitochondrial protein homeostasis in response to stress. Kim and Sieburth identify SPHK-1/sphingosine kinase as a positive regulator of the UPRmt that promotes UPRmt activation in response to a variety of mitochondrial stressors. SPHK-1 associates with mitochondria and SPHK-1 mitochondrial association is stress dependent, reversible, and necessary for the UPRmt, indicating that SPHK-1 mitochondrial targeting is an early step in UPRmt activation.
Collapse
Affiliation(s)
- Sungjin Kim
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Derek Sieburth
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
14
|
Cheng X, Jiang X, Tam KY, Li G, Zheng J, Zhang H. Sphingolipidomic Analysis of C. elegans reveals Development- and Environment-dependent Metabolic Features. Int J Biol Sci 2019; 15:2897-2910. [PMID: 31853226 PMCID: PMC6909964 DOI: 10.7150/ijbs.30499] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 10/02/2019] [Indexed: 01/12/2023] Open
Abstract
Sphingolipids (SLs) serve as structural and signaling molecules in regulating various cellular events and growth. Given that SLs contain various bioactive species possessing distinct roles, quantitative analysis of sphingolipidome is essential for elucidating their differential requirement during development. Herein we developed a comprehensive sphingolipidomic profiling approach using liquid chromatography-mass spectrometry coupled with multiple reaction monitoring mode (LC-MS-MRM). SL profiling of C. elegans revealed organism-specific, development-dependent and environment-driven metabolic features. We showed for the first time the presence of a series of sphingoid bases in C. elegans sphingolipid profiles, although only C17-sphingoid base is used for generating complex SLs. Moreover, we successfully resolved growth-, temperature- and nutrition-dependent SL profiles at both individual metabolite-level and network-level. Sphingolipidomic analysis uncovered significant SL composition changes throughout development, with SMs/GluCers ratios dramatically increasing from larva to adult stage whereas total sphingolipid levels exhibiting opposing trends. We also identified a temperature-dependent alteration in SMs/GluCers ratios, suggesting an organism-specific strategy for environmental adaptation. Finally, we found serine-biased GluCer increases between serine- versus alanine-supplemented worms. Our study builds a “reference” resource for future SL analysis in the worm, provides insights into natural variability and plasticity of eukaryotic multicellular sphingolipid composition and is highly valuable for investigating their functional significance.
Collapse
Affiliation(s)
- Xiaoxiang Cheng
- Faculty of Health Sciences, University of Macau, Taipa, Macau SAR 999078, China
| | - Xue Jiang
- Faculty of Health Sciences, University of Macau, Taipa, Macau SAR 999078, China
| | - Kin Yip Tam
- Faculty of Health Sciences, University of Macau, Taipa, Macau SAR 999078, China
| | - Gang Li
- Faculty of Health Sciences, University of Macau, Taipa, Macau SAR 999078, China
| | - Jun Zheng
- Faculty of Health Sciences, University of Macau, Taipa, Macau SAR 999078, China
| | - Hongjie Zhang
- Faculty of Health Sciences, University of Macau, Taipa, Macau SAR 999078, China.,Centre of Reproduction, Development and Ageing, University of Macau, Taipa, Macau SAR 999078, China
| |
Collapse
|
15
|
Sphingosine kinase and p38 MAP kinase signaling promote resistance to arsenite-induced lethality in Caenorhabditis elegan. Mol Cell Toxicol 2019. [DOI: 10.1007/s13273-019-0045-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
16
|
Mishra SK, Stephenson DJ, Chalfant CE, Brown RE. Upregulation of human glycolipid transfer protein (GLTP) induces necroptosis in colon carcinoma cells. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:158-167. [PMID: 30472325 PMCID: PMC6448591 DOI: 10.1016/j.bbalip.2018.11.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 11/03/2018] [Accepted: 11/10/2018] [Indexed: 02/06/2023]
Abstract
Human GLTP on chromosome 12 (locus 12q24.11) encodes a 24 kD amphitropic lipid transfer protein (GLTP) that mediates glycosphingolipid (GSL) intermembrane trafficking and regulates GSL homeostatic levels within cells. Herein, we provide evidence that GLTP overexpression inhibits the growth of human colon carcinoma cells (HT-29; HCT-116), but spares normal colonic cells (CCD-18Co). Mechanistic studies reveal that GLTP overexpression arrested the cell cycle at the G1/S checkpoint via upregulation of cyclin-dependent kinase inhibitor-1B (Kip1/p27) and cyclin-dependent kinase inhibitor 1A (Cip1/p21) at the protein and mRNA levels, and downregulation of cyclin-dependent kinase-2 (CDK2), cyclin-dependent kinase-4 (CDK4), cyclin E and cyclin D1 protein levels. Assessment of the biological fate of HCT-116 cells overexpressing GLTP indicated no increase in cell death suggesting induction of quiescence. However, HT-29 cells overexpressing GLTP underwent cell death by necroptosis as revealed by phosphorylation of human mixed lineage kinase domain-like protein (pMLKL) via receptor-interacting protein kinase-3 (RIPK-3), elevated cytosolic calcium, and plasma membrane permeabilization by pMLKL oligomerization. Overexpression of W96A-GLTP, an ablated GSL binding site mutant, failed to arrest the cell cycle or induce necroptosis. Sphingolipid assessment (ceramide, monohexosylceramide, sphingomyelin, ceramide-1-phosphate, sphingosine, and sphingosine-1-phosphate) of HT-29 cells overexpressing GLTP revealed large decreases (>5-fold) in sphingosine-1-phosphate with minimal change in 16:0-ceramide, tipping the 'sphingolipid rheostat' (S1P/16:0-Cer ratio) towards cell death. Depletion of RIPK-3 or MLKL abrogated necroptosis induced by GLTP overexpression. Our findings establish GLTP upregulation as a previously unknown suppressor of human colon carcinoma HT-29 cells via interference with cell cycle progression and induction of necroptosis.
Collapse
Affiliation(s)
| | - Daniel J Stephenson
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University Medical Center, Richmond, VA 23298-0614, USA; Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Charles E Chalfant
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA; Research Service, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; The Moffitt Cancer Center, Tampa, FL 33620, USA
| | | |
Collapse
|
17
|
West RJH, Briggs L, Perona Fjeldstad M, Ribchester RR, Sweeney ST. Sphingolipids regulate neuromuscular synapse structure and function in Drosophila. J Comp Neurol 2018; 526:1995-2009. [PMID: 29761896 PMCID: PMC6175220 DOI: 10.1002/cne.24466] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 04/10/2018] [Accepted: 04/10/2018] [Indexed: 12/27/2022]
Abstract
Sphingolipids are found in abundance at synapses and have been implicated in regulation of synapse structure, function, and degeneration. Their precise role in these processes, however, remains obscure. Serine Palmitoyl-transferase (SPT) is the first enzymatic step for synthesis of sphingolipids. Analysis of the Drosophila larval neuromuscular junction (NMJ) revealed mutations in the SPT enzyme subunit, lace/SPTLC2 resulted in deficits in synaptic structure and function. Although NMJ length is normal in lace mutants, the number of boutons per NMJ is reduced to ∼50% of the wild type number. Synaptic boutons in lace mutants are much larger but show little perturbation to the general ultrastructure. Electrophysiological analysis of lace mutant synapses revealed strong synaptic transmission coupled with predominance of depression over facilitation. The structural and functional phenotypes of lace mirrored aspects of Basigin (Bsg), a small Ig-domain adhesion molecule also known to regulate synaptic structure and function. Mutant combinations of lace and Bsg generated large synaptic boutons, while lace mutants showed abnormal accumulation of Bsg at synapses, suggesting that Bsg requires sphingolipid to regulate structure of the synapse. In support of this, we found Bsg to be enriched in lipid rafts. Our data points to a role for sphingolipids in the regulation and fine-tuning of synaptic structure and function while sphingolipid regulation of synaptic structure may be mediated via the activity of Bsg.
Collapse
Affiliation(s)
- Ryan J. H. West
- Department of Biology and Hull York Medical SchoolUniversity of YorkHeslingtonYork YO10 5DDUK
| | - Laura Briggs
- Department of Biology and Hull York Medical SchoolUniversity of YorkHeslingtonYork YO10 5DDUK
| | - Maria Perona Fjeldstad
- Euan MacDonald Centre for Motor Neurone Disease Research and Centre for Discovery Brain SciencesUniversity of EdinburghEdinburgh EH8 9JZUK
| | - Richard R. Ribchester
- Euan MacDonald Centre for Motor Neurone Disease Research and Centre for Discovery Brain SciencesUniversity of EdinburghEdinburgh EH8 9JZUK
| | - Sean T. Sweeney
- Department of Biology and Hull York Medical SchoolUniversity of YorkHeslingtonYork YO10 5DDUK
| |
Collapse
|
18
|
Sphingosine Kinase Regulates Neuropeptide Secretion During the Oxidative Stress-Response Through Intertissue Signaling. J Neurosci 2018; 38:8160-8176. [PMID: 30082417 DOI: 10.1523/jneurosci.0536-18.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 07/19/2018] [Accepted: 07/20/2018] [Indexed: 12/12/2022] Open
Abstract
The Nrf2 antioxidant transcription factor promotes redox homeostasis in part through reciprocal signaling between neurons and neighboring cells, but the signals involved in intertissue signaling in response to Nrf2 activation are not well defined. In Caenorhabditis elegans, activation of SKN-1/Nrf2 in the intestine negatively regulates neuropeptide secretion from motor neurons. Here, we show that sphingosine kinase (SPHK-1) functions downstream of SKN-1/Nrf2 in the intestine to regulate neuropeptide secretion from motor neurons during the oxidative stress response in C. elegans hermaphrodites. SPHK-1 localizes to mitochondria in the intestine and SPHK-1 mitochondrial localization and kinase activity are essential for its function in regulating motor neuron function. SPHK-1 is recruited to mitochondria from cytosolic pools and its mitochondrial abundance is negatively regulated by acute or chronic SKN-1 activation. Finally, the regulation of motor function by SKN-1 requires the activation of the p38 MAPK cascade in the intestine and occurs through controlling the biogenesis or maturation of dense core vesicles in motor neurons. These findings show that the inhibition of SPHK-1 in the intestine by SKN-1 negatively regulates neuropeptide secretion from motor neurons, revealing a new mechanism by which SPHK-1 signaling mediates its effects on neuronal function in response to oxidative stress.SIGNIFICANCE STATEMENT Neurons are highly susceptible to damage by oxidative stress, yet have limited capacity to activate the SKN-1/Nrf2 oxidative stress response, relying instead on astrocytes to provide redox homeostasis. In Caenorhabditis elegans, intertissue signaling from the intestine plays a key role in regulating neuronal function during the oxidative stress response. Here, through a combination of genetic, behavioral, and fluorescent imaging approaches, we found that sphingosine kinase functions in the SKN-1/Nrf2 pathway in the intestine to regulate neuropeptide biogenesis and secretion in motor neurons. These results implicate sphingolipid signaling as a new component of the oxidative stress response and suggest that C. elegans may be a genetically tractable model to study non-cell-autonomous oxidative stress signaling to neurons.
Collapse
|
19
|
McCammon JM, Blaker-Lee A, Chen X, Sive H. The 16p11.2 homologs fam57ba and doc2a generate certain brain and body phenotypes. Hum Mol Genet 2018; 26:3699-3712. [PMID: 28934389 PMCID: PMC5886277 DOI: 10.1093/hmg/ddx255] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 06/29/2017] [Indexed: 01/28/2023] Open
Abstract
Deletion of the 16p11.2 CNV affects 25 core genes and is associated with multiple symptoms affecting brain and body, including seizures, hyperactivity, macrocephaly, and obesity. Available data suggest that most symptoms are controlled by haploinsufficiency of two or more 16p11.2 genes. To identify interacting 16p11.2 genes, we used a pairwise partial loss of function antisense screen for embryonic brain morphology, using the accessible zebrafish model. fam57ba, encoding a ceramide synthase, was identified as interacting with the doc2a gene, encoding a calcium-sensitive exocytosis regulator, a genetic interaction not previously described. Using genetic mutants, we demonstrated that doc2a+/− fam57ba+/− double heterozygotes show hyperactivity and increased seizure susceptibility relative to wild-type or single doc2a−/− or fam57ba−/− mutants. Additionally, doc2a+/− fam57ba+/− double heterozygotes demonstrate the increased body length and head size. Single doc2a+/− and fam57ba+/− heterozygotes do not show a body size increase; however, fam57ba−/− homozygous mutants show a strongly increased head size and body length, suggesting a greater contribution from fam57ba to the haploinsufficient interaction between doc2a and fam57ba. The doc2a+/− fam57ba+/− interaction has not been reported before, nor has any 16p11.2 gene previously been linked to increased body size. These findings demonstrate that one pair of 16p11.2 homologs can regulate both brain and body phenotypes that are reflective of those in people with 16p11.2 deletion. Together, these findings suggest that dysregulation of ceramide pathways and calcium sensitive exocytosis underlies seizures and large body size associated with 16p11.2 homologs in zebrafish. The data inform consideration of mechanisms underlying human 16p11.2 deletion symptoms.
Collapse
Affiliation(s)
| | - Alicia Blaker-Lee
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Xiao Chen
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hazel Sive
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
20
|
Sphingosine Kinase 1 Cooperates with Autophagy to Maintain Endocytic Membrane Trafficking. Cell Rep 2017; 17:1532-1545. [PMID: 27806293 DOI: 10.1016/j.celrep.2016.10.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 09/15/2016] [Accepted: 10/05/2016] [Indexed: 11/21/2022] Open
Abstract
Sphingosine kinase 1 (Sphk1) associates with early endocytic membranes during endocytosis; however, the role of sphingosine or sphingosine-1-phosphate as the critical metabolite in endocytic trafficking has not been established. Here, we demonstrate that the recruitment of Sphk1 to sphingosine-enriched endocytic vesicles and the generation of sphingosine-1-phosphate facilitate membrane trafficking along the endosomal pathway. Exogenous sphingosine and sphingosine-based Sphk1 inhibitors induce the Sphk1-dependent fusion of endosomal membranes to accumulate enlarged late endosomes and amphisomes enriched in sphingolipids. Interestingly, Sphk1 also appears to facilitate endosomal fusion independent of its catalytic activity, given that catalytically inactive Sphk1G82D is recruited to endocytic membranes by sphingosine or sphingosine-based Sphk1 inhibitor and promotes membrane fusion. Furthermore, we reveal that the clearance of enlarged endosomes is dependent on the activity of ceramide synthase, lysosomal biogenesis, and the restoration of autophagic flux. Collectively, these studies uncover intersecting roles for Sphk1, sphingosine, and autophagic machinery in endocytic membrane trafficking.
Collapse
|
21
|
Chan JP, Brown J, Hark B, Nolan A, Servello D, Hrobuchak H, Staab TA. Loss of Sphingosine Kinase Alters Life History Traits and Locomotor Function in Caenorhabditis elegans. Front Genet 2017; 8:132. [PMID: 28983319 PMCID: PMC5613162 DOI: 10.3389/fgene.2017.00132] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/06/2017] [Indexed: 12/15/2022] Open
Abstract
Sphingolipid metabolism is important to balance the abundance of bioactive lipid molecules involved in cell signaling, neuronal function, and survival. Specifically, the sphingolipid sphingosine mediates cell death signaling, whereas its phosphorylated form, sphingosine-1-phosphate (S1P), mediates cell survival signaling. The enzyme sphingosine kinase produces S1P, and the activity of sphingosine kinase impacts the ability of cells to survive under stress and challenges. To examine the influence of sphingolipid metabolism, particularly enzymes regulating sphingosine and S1P, in mediating aging, neuronal function and stress response, we examined life history traits, locomotor capacities and heat stress responses of young and old animals using the model organism Caenorhabditis elegans. We found that C. elegans sphk-1 mutants, which lack sphingosine kinase, had shorter lifespans, reduced brood sizes, and smaller body sizes compared to wild type animals. By analyzing a panel of young and old animals with genetic mutations in the sphingolipid signaling pathway, we showed that aged sphk-1 mutants exhibited a greater decline in neuromuscular function and locomotor behavior. In addition, aged animals lacking sphk-1 were more susceptible to death induced by acute and prolonged heat exposure. On the other hand, older animals with loss of function mutations in ceramide synthase (hyl-1), which converts sphingosine to ceramide, showed improved neuromuscular function and stress response with age. This phenotype was dependent on sphk-1. Together, our data show that loss of sphingosine kinase contributes to poor animal health span, suggesting that sphingolipid signaling may be important for healthy neuronal function and animal stress response during aging.
Collapse
Affiliation(s)
- Jason P. Chan
- Department of Biology, Juniata CollegeHuntingdon, PA, United States
| | | | | | | | | | | | | |
Collapse
|
22
|
McCulloch KA, Qi YB, Takayanagi-Kiya S, Jin Y, Cherra SJ. Novel Mutations in Synaptic Transmission Genes Suppress Neuronal Hyperexcitation in Caenorhabditis elegans. G3 (BETHESDA, MD.) 2017; 7:2055-2063. [PMID: 28468816 PMCID: PMC5499116 DOI: 10.1534/g3.117.042598] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/22/2017] [Indexed: 01/29/2023]
Abstract
Acetylcholine (ACh) receptors (AChR) regulate neural circuit activity in multiple contexts. In humans, mutations in ionotropic acetylcholine receptor (iAChR) genes can cause neurological disorders, including myasthenia gravis and epilepsy. In Caenorhabditis elegans, iAChRs play multiple roles in the locomotor circuit. The cholinergic motor neurons express an ACR-2-containing pentameric AChR (ACR-2R) comprised of ACR-2, ACR-3, ACR-12, UNC-38, and UNC-63 subunits. A gain-of-function mutation in the non-α subunit gene acr-2 [acr-2(gf)] causes defective locomotion as well as spontaneous convulsions. Previous studies of genetic suppressors of acr-2(gf) have provided insights into ACR-2R composition and assembly. Here, to further understand how the ACR-2R regulates neuronal activity, we expanded the suppressor screen for acr-2(gf)-induced convulsions. The majority of these suppressor mutations affect genes that play critical roles in synaptic transmission, including two novel mutations in the vesicular ACh transporter unc-17 In addition, we identified a role for a conserved major facilitator superfamily domain (MFSD) protein, mfsd-6, in regulating neural circuit activity. We further defined a role for the sphingosine (SPH) kinase (Sphk) sphk-1 in cholinergic neuron activity, independent of previously known signaling pathways. Overall, the genes identified in our study suggest that optimal modulation of synaptic activity is balanced by the differential activities of multiple pathways, and the novel alleles provide valuable reagents to further dissect neuronal mechanisms regulating the locomotor circuit.
Collapse
Affiliation(s)
- Katherine A McCulloch
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, California 92093
| | - Yingchuan B Qi
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, California 92093
| | - Seika Takayanagi-Kiya
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, California 92093
| | - Yishi Jin
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, California 92093
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California 92093
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, California 92093
| | - Salvatore J Cherra
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, California 92093
| |
Collapse
|
23
|
Garcia‐Gil M, Pierucci F, Vestri A, Meacci E. Crosstalk between sphingolipids and vitamin D3: potential role in the nervous system. Br J Pharmacol 2017; 174:605-627. [PMID: 28127747 PMCID: PMC6398521 DOI: 10.1111/bph.13726] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/16/2016] [Accepted: 01/18/2017] [Indexed: 12/14/2022] Open
Abstract
Sphingolipids are both structural and bioactive compounds. In particular, ceramide and sphingosine 1-phosphate regulate cell fate, inflammation and excitability. 1-α,25-dihydroxyvitamin D3 (1,25(OH)2 D3 ) is known to play an important physiological role in growth and differentiation in a variety of cell types, including neural cells, through genomic actions mediated by its specific receptor, and non-genomic effects that result in the activation of specific signalling pathways. 1,25(OH)2 D3 and sphingolipids, in particular sphingosine 1-phosphate, share many common effectors, including calcium regulation, growth factors and inflammatory cytokines, but it is still not known whether they can act synergistically. Alterations in the signalling and concentrations of sphingolipids and 1,25(OH)2 D3 have been found in neurodegenerative diseases and fingolimod, a structural analogue of sphingosine, has been approved for the treatment of multiple sclerosis. This review, after a brief description of the role of sphingolipids and 1,25(OH)2 D3 , will focus on the potential crosstalk between sphingolipids and 1,25(OH)2 D3 in neural cells.
Collapse
Affiliation(s)
- Mercedes Garcia‐Gil
- Department of BiologyUniversity of PisaPisaItaly
- Interdepartmental Research Center Nutrafood ‘Nutraceuticals and Food for Health’University of PisaPisaItaly
| | - Federica Pierucci
- Department of Experimental and Clinical Biomedical Sciences ‘Mario Serio’, Molecular and Applied Biology Research UnitUniversity of FlorenceFlorenceItaly
- Interuniversitary Miology InstitutesItaly
| | - Ambra Vestri
- Department of Experimental and Clinical Biomedical Sciences ‘Mario Serio’, Molecular and Applied Biology Research UnitUniversity of FlorenceFlorenceItaly
- Interuniversitary Miology InstitutesItaly
| | - Elisabetta Meacci
- Department of Experimental and Clinical Biomedical Sciences ‘Mario Serio’, Molecular and Applied Biology Research UnitUniversity of FlorenceFlorenceItaly
- Interuniversitary Miology InstitutesItaly
| |
Collapse
|
24
|
Karunakaran I, van Echten-Deckert G. Sphingosine 1-phosphate - A double edged sword in the brain. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:1573-1582. [PMID: 28315304 DOI: 10.1016/j.bbamem.2017.03.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/28/2017] [Accepted: 03/13/2017] [Indexed: 12/31/2022]
Abstract
The physiological functions of sphingosine 1-phosphate (S1P) and its pathological roles in various diseases are increasingly being elucidated. Particularly, a growing body of literature has implicated S1P in the pathogenesis of brain related disorders. With the deciphering of more intricate aspects of S1P signalling, there is also a need to reconsider the notion of S1P only as a determinant of cell survival and proliferation. Further the concept of 'S1P-ceramide' balance as the controlling switch of cellular fate and functions needs to be refined. In this review, we focus on the brain related functions of S1P with special focus on its role in synaptic transmission, neuronal autophagy and neuroinflammation. The review also attempts to bring out the multi-faceted nature of S1P signalling aspects that makes it a 'double edged sword'. This article is part of a Special Issue entitled: Membrane Lipid Therapy: Drugs Targeting Biomembranes edited by Pablo V. Escribá.
Collapse
Affiliation(s)
- Indulekha Karunakaran
- LIMES Institute, Membrane Biology & Lipid Biochemistry, University of Bonn, Bonn, Germany
| | | |
Collapse
|
25
|
Lima S, Milstien S, Spiegel S. Sphingosine and Sphingosine Kinase 1 Involvement in Endocytic Membrane Trafficking. J Biol Chem 2017; 292:3074-3088. [PMID: 28049734 DOI: 10.1074/jbc.m116.762377] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/22/2016] [Indexed: 12/22/2022] Open
Abstract
The balance between cholesterol and sphingolipids within the plasma membrane has long been implicated in endocytic membrane trafficking. However, in contrast to cholesterol functions, little is still known about the roles of sphingolipids and their metabolites. Perturbing the cholesterol/sphingomyelin balance was shown to induce narrow tubular plasma membrane invaginations enriched with sphingosine kinase 1 (SphK1), the enzyme that converts the bioactive sphingolipid metabolite sphingosine to sphingosine-1-phosphate, and suggested a role for sphingosine phosphorylation in endocytic membrane trafficking. Here we show that sphingosine and sphingosine-like SphK1 inhibitors induced rapid and massive formation of vesicles in diverse cell types that accumulated as dilated late endosomes. However, much smaller vesicles were formed in SphK1-deficient cells. Moreover, inhibition or deletion of SphK1 prolonged the lifetime of sphingosine-induced vesicles. Perturbing the plasma membrane cholesterol/sphingomyelin balance abrogated vesicle formation. This massive endosomal influx was accompanied by dramatic recruitment of the intracellular SphK1 and Bin/Amphiphysin/Rvs domain-containing proteins endophilin-A2 and endophilin-B1 to enlarged endosomes and formation of highly dynamic filamentous networks containing endophilin-B1 and SphK1. Together, our results highlight the importance of sphingosine and its conversion to sphingosine-1-phosphate by SphK1 in endocytic membrane trafficking.
Collapse
Affiliation(s)
- Santiago Lima
- Department of Biochemistry and Molecular Biology and Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298.
| | - Sheldon Milstien
- Department of Biochemistry and Molecular Biology and Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology and Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298.
| |
Collapse
|
26
|
Sphingosine 1-phosphate lyase ablation disrupts presynaptic architecture and function via an ubiquitin- proteasome mediated mechanism. Sci Rep 2016; 6:37064. [PMID: 27883090 PMCID: PMC5121647 DOI: 10.1038/srep37064] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 10/24/2016] [Indexed: 01/28/2023] Open
Abstract
The bioactive lipid sphingosine 1-phosphate (S1P) is a degradation product of sphingolipids that are particularly abundant in neurons. We have shown previously that neuronal S1P accumulation is toxic leading to ER-stress and an increase in intracellular calcium. To clarify the neuronal function of S1P, we generated brain-specific knockout mouse models in which S1P-lyase (SPL), the enzyme responsible for irreversible S1P cleavage was inactivated. Constitutive ablation of SPL in the brain (SPLfl/fl/Nes) but not postnatal neuronal forebrain-restricted SPL deletion (SPLfl/fl/CaMK) caused marked accumulation of S1P. Hence, altered presynaptic architecture including a significant decrease in number and density of synaptic vesicles, decreased expression of several presynaptic proteins, and impaired synaptic short term plasticity were observed in hippocampal neurons from SPLfl/fl/Nes mice. Accordingly, these mice displayed cognitive deficits. At the molecular level, an activation of the ubiquitin-proteasome system (UPS) was detected which resulted in a decreased expression of the deubiquitinating enzyme USP14 and several presynaptic proteins. Upon inhibition of proteasomal activity, USP14 levels, expression of presynaptic proteins and synaptic function were restored. These findings identify S1P metabolism as a novel player in modulating synaptic architecture and plasticity.
Collapse
|
27
|
Sphingosine-1-Phosphate (S1P) Impacts Presynaptic Functions by Regulating Synapsin I Localization in the Presynaptic Compartment. J Neurosci 2016; 36:4624-34. [PMID: 27098703 DOI: 10.1523/jneurosci.3588-15.2016] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 03/16/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Growing evidence indicates that sphingosine-1-P (S1P) upregulates glutamate secretion in hippocampal neurons. However, the molecular mechanisms through which S1P enhances excitatory activity remain largely undefined. The aim of this study was to identify presynaptic targets of S1P action controlling exocytosis. Confocal analysis of rat hippocampal neurons showed that S1P applied at nanomolar concentration alters the distribution of Synapsin I (SynI), a presynaptic phosphoprotein that controls the availability of synaptic vesicles for exocytosis. S1P induced SynI relocation to extrasynaptic regions of mature neurons, as well as SynI dispersion from synaptic vesicle clusters present at axonal growth cones of developing neurons. S1P-induced SynI relocation occurred in a Ca(2+)-independent but ERK-dependent manner, likely through the activation of S1P3 receptors, as it was prevented by the S1P3 receptor selective antagonist CAY1044 and in neurons in which S1P3 receptor was silenced. Our recent evidence indicates that microvesicles (MVs) released by microglia enhance the metabolism of endogenous sphingolipids in neurons and stimulate excitatory transmission. We therefore investigated whether MVs affect SynI distribution and whether endogenous S1P could be involved in the process. Analysis of SynI immunoreactivity showed that exposure to microglial MVs induces SynI mobilization at presynaptic sites and growth cones, whereas the use of inhibitors of sphingolipid cascade identified S1P as the sphingolipid mediating SynI redistribution. Our data represent the first demonstration that S1P induces SynI mobilization from synapses, thereby indicating the phosphoprotein as a novel target through which S1P controls exocytosis. SIGNIFICANCE STATEMENT Growing evidence indicates that the bioactive lipid sphingosine and its metabolite sphingosine-1-P (S1P) stimulate excitatory transmission. While it has been recently clarified that sphingosine influences directly the exocytotic machinery by activating the synaptic vesicle protein VAMP2 to form SNARE fusion complexes, the molecular mechanism by which S1P promotes neurotransmission remained largely undefined. In this study, we identify Synapsin I, a presynaptic phosphoprotein involved in the control of availability of synaptic vesicles for exocytosis, as the key target of S1P action. In addition, we provide evidence that S1P can be produced at mature axon terminals as well as at immature growth cones in response to microglia-derived signals, which may be important to stabilize nascent synapses and to restore or potentiate transmission.
Collapse
|
28
|
Deng X, Kolesnick R. Caenorhabditis elegans as a model to study sphingolipid signaling. Biol Chem 2016; 396:767-73. [PMID: 25720116 DOI: 10.1515/hsz-2014-0298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 01/26/2015] [Indexed: 12/19/2022]
Abstract
Sphingolipid signaling in Caenorhabditis elegans is vital for sensing environmental change and effecting appropriate cellular response. Many molecular components in sphingolipid intermediary metabolism are conserved throughout evolution. Here we review use of C. elegans as a model system for conducting sphingolipid-based scientific investigation, which has helped us better understand vital roles these remarkable lipids play in human metabolism and disease.
Collapse
|
29
|
Tu-Sekine B, Goldschmidt H, Raben DM. Diacylglycerol, phosphatidic acid, and their metabolic enzymes in synaptic vesicle recycling. Adv Biol Regul 2014; 57:147-52. [PMID: 25446883 DOI: 10.1016/j.jbior.2014.09.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 09/03/2014] [Accepted: 09/03/2014] [Indexed: 01/10/2023]
Abstract
The synaptic vesicle (SV) cycle includes exocytosis of vesicles loaded with a neurotransmitter such as glutamate, coordinated recovery of SVs by endocytosis, refilling of vesicles, and subsequent release of the refilled vesicles from the presynaptic bouton. SV exocytosis is tightly linked with endocytosis, and variations in the number of vesicles, and/or defects in the refilling of SVs, will affect the amount of neurotransmitter available for release (Sudhof, 2004). There is increasing interest in the roles synaptic vesicle lipids and lipid metabolizing enzymes play in this recycling. Initial emphasis was placed on the role of polyphosphoinositides in SV cycling as outlined in a number of reviews (Lim and Wenk, 2009; Martin, 2012; Puchkov and Haucke, 2013; Rohrbough and Broadie, 2005). Other lipids are now recognized to also play critical roles. For example, PLD1 (Humeau et al., 2001; Rohrbough and Broadie, 2005) and some DGKs (Miller et al., 1999; Nurrish et al., 1999) play roles in neurotransmission which is consistent with the critical roles for phosphatidic acid (PtdOH) and diacylglycerol (DAG) in the regulation of SV exo/endocytosis (Cremona et al., 1999; Exton, 1994; Huttner and Schmidt, 2000; Lim and Wenk, 2009; Puchkov and Haucke, 2013; Rohrbough and Broadie, 2005). PLD generates phosphatidic acid by catalyzing the hydrolysis of phosphatidylcholine (PtdCho) and in some systems this PtdOH is de-phosphorylated to generate DAG. In contrast, DGK catalyzes the phosphorylation of DAG thereby converting it into PtdOH. While both enzymes are poised to regulate the levels of DAG and PtdOH, therefore, they both lead to the generation of PtdOH and could have opposite effects on DAG levels. This is particularly important for SV cycling as PtdOH and DAG are both needed for evoked exocytosis (Lim and Wenk, 2009; Puchkov and Haucke, 2013; Rohrbough and Broadie, 2005). Two lipids and their involved metabolic enzymes, two sphingolipids have also been implicated in exocytosis: sphingosine (Camoletto et al., 2009; Chan et al., 2012; Chan and Sieburth, 2012; Darios et al., 2009; Kanno et al., 2010; Rohrbough et al., 2004) and sphingosine-1-phosphate (Chan, Hu, 2012; Chan and Sieburth, 2012; Kanno et al., 2010). Finally a number of reports have focused on the somewhat less well studies roles of sphingolipids and cholesterol in SV cycling. In this report, we review the recent understanding of the roles PLDs, DGKs, and DAG lipases, as well as sphingolipids and cholesterol play in synaptic vesicle cycling.
Collapse
Affiliation(s)
- Becky Tu-Sekine
- The Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hana Goldschmidt
- The Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Daniel M Raben
- The Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
30
|
Shen H, Giordano F, Wu Y, Chan J, Zhu C, Milosevic I, Wu X, Yao K, Chen B, Baumgart T, Sieburth D, De Camilli P. Coupling between endocytosis and sphingosine kinase 1 recruitment. Nat Cell Biol 2014; 16:652-62. [PMID: 24929359 PMCID: PMC4230894 DOI: 10.1038/ncb2987] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 05/09/2014] [Indexed: 12/13/2022]
Abstract
Genetic studies have suggested a functional link between cholesterol/sphingolipid metabolism and endocytic membrane traffic. Here we show that perturbing the cholesterol/sphingomyelin balance in the plasma membrane results in the massive formation of clusters of narrow endocytic tubular invaginations positive for N-BAR proteins. These tubules are intensely positive for sphingosine kinase 1 (SPHK1). SPHK1 is also targeted to physiologically occurring early endocytic intermediates, and is highly enriched in nerve terminals, which are cellular compartments specialized for exo/endocytosis. Membrane recruitment of SPHK1 involves a direct, curvature-sensitive interaction with the lipid bilayer mediated by a hydrophobic patch on the enzyme's surface. The knockdown of SPHKs results in endocytic recycling defects, and a mutation that disrupts the hydrophobic patch of Caenorhabditis elegans SPHK fails to rescue the neurotransmission defects in loss-of-function mutants of this enzyme. Our studies support a role for sphingosine phosphorylation in endocytic membrane trafficking beyond the established function of sphingosine-1-phosphate in intercellular signalling.
Collapse
Affiliation(s)
- Hongying Shen
- 1] Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06510, USA [2] Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06510, USA [3]
| | - Francesca Giordano
- 1] Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06510, USA [2] Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06510, USA [3]
| | - Yumei Wu
- 1] Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06510, USA [2] Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Jason Chan
- 1] Zilkha Neurogenetic Institute, Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA [2]
| | - Chen Zhu
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Ira Milosevic
- 1] Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06510, USA [2] Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Xudong Wu
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Kai Yao
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Bo Chen
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Tobias Baumgart
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Derek Sieburth
- Zilkha Neurogenetic Institute, Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA
| | - Pietro De Camilli
- 1] Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06510, USA [2] Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| |
Collapse
|
31
|
Extrasynaptic muscarinic acetylcholine receptors on neuronal cell bodies regulate presynaptic function in Caenorhabditis elegans. J Neurosci 2013; 33:14146-59. [PMID: 23986249 DOI: 10.1523/jneurosci.1359-13.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Acetylcholine (ACh) is a potent neuromodulator in the brain, and its effects on cognition and memory formation are largely performed through muscarinic acetylcholine receptors (mAChRs). mAChRs are often preferentially distributed on specialized membrane regions in neurons, but the significance of mAChR localization in modulating neuronal function is not known. Here we show that the Caenorhabditis elegans homolog of the M1/M3/M5 family of mAChRs, gar-3, is expressed in cholinergic motor neurons, and GAR-3-GFP fusion proteins localize to cell bodies where they are enriched at extrasynaptic regions that are in contact with the basal lamina. The GAR-3 N-terminal extracellular domain is necessary and sufficient for this asymmetric distribution, and mutation of a predicted N-linked glycosylation site within the N-terminus disrupts GAR-3-GFP localization. In transgenic animals expressing GAR-3 variants that are no longer asymmetrically localized, synaptic transmission at neuromuscular junctions is impaired and there is a reduction in the abundance of the presynaptic protein sphingosine kinase at release sites. Finally, GAR-3 can be activated by endogenously produced ACh released from neurons that do not directly contact cholinergic motor neurons. Together, our results suggest that humoral activation of asymmetrically localized mAChRs by ACh is an evolutionarily conserved mechanism by which ACh modulates neuronal function.
Collapse
|
32
|
Puchkov D, Haucke V. Greasing the synaptic vesicle cycle by membrane lipids. Trends Cell Biol 2013; 23:493-503. [DOI: 10.1016/j.tcb.2013.05.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 04/30/2013] [Accepted: 05/02/2013] [Indexed: 12/18/2022]
|