1
|
Richter Gorey CL, St Louis AP, Chorna T, Brill JA, Dason JS. Differential functions of phosphatidylinositol 4-kinases in neurotransmission and synaptic development. Eur J Neurosci 2024; 60:5966-5979. [PMID: 39267207 DOI: 10.1111/ejn.16526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 08/07/2024] [Accepted: 08/16/2024] [Indexed: 09/14/2024]
Abstract
Phosphoinositides, such as PI(4,5)P2, are known to function as structural components of membranes, signalling molecules, markers of membrane identity, mediators of protein recruitment and regulators of neurotransmission and synaptic development. Phosphatidylinositol 4-kinases (PI4Ks) synthesize PI4P, which are precursors for PI(4,5)P2, but may also have independent functions. The roles of PI4Ks in neurotransmission and synaptic development have not been studied in detail. Previous studies on PI4KII and PI4KIIIβ at the Drosophila larval neuromuscular junction have suggested that PI4KII and PI4KIIIβ enzymes may serve redundant roles, where single PI4K mutants yielded mild or no synaptic phenotypes. However, the precise synaptic functions (neurotransmission and synaptic growth) of these PI4Ks have not been thoroughly studied. Here, we used PI4KII and PI4KIIIβ null mutants and presynaptic-specific knockdowns of these PI4Ks to investigate their roles in neurotransmission and synaptic growth. We found that PI4KII and PI4KIIIβ appear to have non-overlapping functions. Specifically, glial PI4KII functions to restrain synaptic growth, whereas presynaptic PI4KIIIβ promotes synaptic growth. Furthermore, loss of PI4KIIIβ or presynaptic PI4KII impairs neurotransmission. The data presented in this study uncover new roles for PI4K enzymes in neurotransmission and synaptic growth.
Collapse
Affiliation(s)
| | | | - Tetyana Chorna
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Julie A Brill
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Jeffrey S Dason
- Department of Biomedical Sciences, University of Windsor, Windsor, Ontario, Canada
| |
Collapse
|
2
|
Shaheen A, Richter Gorey CL, Sghaier A, Dason JS. Cholesterol is required for activity-dependent synaptic growth. J Cell Sci 2023; 136:jcs261563. [PMID: 37902091 DOI: 10.1242/jcs.261563] [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: 08/22/2023] [Accepted: 10/24/2023] [Indexed: 10/31/2023] Open
Abstract
Changes in cholesterol content of neuronal membranes occur during development and brain aging. Little is known about whether synaptic activity regulates cholesterol levels in neuronal membranes and whether these changes affect neuronal development and function. We generated transgenic flies that express the cholesterol-binding D4H domain of perfringolysin O toxin and found increased levels of cholesterol in presynaptic terminals of Drosophila larval neuromuscular junctions following increased synaptic activity. Reduced cholesterol impaired synaptic growth and largely prevented activity-dependent synaptic growth. Presynaptic knockdown of adenylyl cyclase phenocopied the impaired synaptic growth caused by reducing cholesterol. Furthermore, the effects of knocking down adenylyl cyclase and reducing cholesterol were not additive, suggesting that they function in the same pathway. Increasing cAMP levels using a dunce mutant with reduced phosphodiesterase activity failed to rescue this impaired synaptic growth, suggesting that cholesterol functions downstream of cAMP. We used a protein kinase A (PKA) sensor to show that reducing cholesterol levels reduced presynaptic PKA activity. Collectively, our results demonstrate that enhanced synaptic activity increased cholesterol levels in presynaptic terminals and that these changes likely activate the cAMP-PKA pathway during activity-dependent growth.
Collapse
Affiliation(s)
- Amber Shaheen
- Department of Biomedical Sciences, University of Windsor, Windsor, Ontario, N9B 3P4, Canada
| | - Claire L Richter Gorey
- Department of Biomedical Sciences, University of Windsor, Windsor, Ontario, N9B 3P4, Canada
| | - Adam Sghaier
- Department of Biomedical Sciences, University of Windsor, Windsor, Ontario, N9B 3P4, Canada
| | - Jeffrey S Dason
- Department of Biomedical Sciences, University of Windsor, Windsor, Ontario, N9B 3P4, Canada
| |
Collapse
|
3
|
Beccano-Kelly DA, Cherubini M, Mousba Y, Cramb KM, Giussani S, Caiazza MC, Rai P, Vingill S, Bengoa-Vergniory N, Ng B, Corda G, Banerjee A, Vowles J, Cowley S, Wade-Martins R. Calcium dysregulation combined with mitochondrial failure and electrophysiological maturity converge in Parkinson's iPSC-dopamine neurons. iScience 2023; 26:107044. [PMID: 37426342 PMCID: PMC10329047 DOI: 10.1016/j.isci.2023.107044] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/30/2022] [Accepted: 06/01/2023] [Indexed: 07/11/2023] Open
Abstract
Parkinson's disease (PD) is characterized by a progressive deterioration of motor and cognitive functions. Although death of dopamine neurons is the hallmark pathology of PD, this is a late-stage disease process preceded by neuronal dysfunction. Here we describe early physiological perturbations in patient-derived induced pluripotent stem cell (iPSC)-dopamine neurons carrying the GBA-N370S mutation, a strong genetic risk factor for PD. GBA-N370S iPSC-dopamine neurons show an early and persistent calcium dysregulation notably at the mitochondria, followed by reduced mitochondrial membrane potential and oxygen consumption rate, indicating mitochondrial failure. With increased neuronal maturity, we observed decreased synaptic function in PD iPSC-dopamine neurons, consistent with the requirement for ATP and calcium to support the increase in electrophysiological activity over time. Our work demonstrates that calcium dyshomeostasis and mitochondrial failure impair the higher electrophysiological activity of mature neurons and may underlie the vulnerability of dopamine neurons in PD.
Collapse
Affiliation(s)
- Dayne A. Beccano-Kelly
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Marta Cherubini
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Yassine Mousba
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Kaitlyn M.L. Cramb
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Stefania Giussani
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Maria Claudia Caiazza
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Pavandeep Rai
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Siv Vingill
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Nora Bengoa-Vergniory
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Bryan Ng
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Gabriele Corda
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Abhirup Banerjee
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford OX3 9DU, UK
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford OX3 7DQ, UK
| | - Jane Vowles
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- The James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Sally Cowley
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- The James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Richard Wade-Martins
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| |
Collapse
|
4
|
NCS1 overexpression restored mitochondrial activity and behavioral alterations in a zebrafish model of Wolfram syndrome. Mol Ther Methods Clin Dev 2022; 27:295-308. [PMID: 36320410 PMCID: PMC9594121 DOI: 10.1016/j.omtm.2022.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 10/04/2022] [Indexed: 11/28/2022]
Abstract
Wolfram syndrome (WS) is a rare neurodegenerative disease resulting in deafness, optic atrophy, diabetes, and neurological disorders. Currently, no treatment is available for patients. The mutated gene, WFS1, encodes an endoplasmic reticulum (ER) protein, Wolframin. We previously reported that Wolframin regulated the ER-mitochondria Ca2+ transfer and mitochondrial activity by protecting NCS1 from degradation in patients' fibroblasts. We relied on a zebrafish model of WS, the wfs1ab KO line, to analyze the functional and behavioral impact of NCS1 overexpression as a novel therapeutic strategy. The wfs1ab KO line showed an increased locomotion in the visual motor and touch-escape responses. The absence of wfs1 did not impair the cellular unfolded protein response, in basal or tunicamycin-induced ER stress conditions. In contrast, metabolic analysis showed an increase in mitochondrial respiration in wfs1ab KO larvae. Interestingly, overexpression of NCS1 using mRNA injection restored the alteration of mitochondrial respiration and hyperlocomotion. Taken together, these data validated the wfs1ab KO zebrafish line as a pertinent experimental model of WS and confirmed the therapeutic potential of NCS1. The wfs1ab KO line therefore appeared as an efficient model to identify novel therapeutic strategies, such as gene or pharmacological therapies targeting NCS1 that will correct or block WS symptoms.
Collapse
|
5
|
Mechanisms underlying paclitaxel-induced neuropathic pain: Channels, inflammation and immune regulations. Eur J Pharmacol 2022; 933:175288. [PMID: 36122757 DOI: 10.1016/j.ejphar.2022.175288] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/07/2022] [Accepted: 09/13/2022] [Indexed: 11/22/2022]
Abstract
Paclitaxel is a chemotherapeutic agent widely used for many types of malignancies. However, when paclitaxel is used to treat tumors, patients commonly experience severe neuropathic pain that is difficult to manage. The mechanism underlying paclitaxel-induced neuropathic pain remains unclear. Evidence demonstrates correlations between mechanisms of paclitaxel-mediated pain and associated actions of ion channels, neuroinflammation, mitochondrial damage, and other factors. This review provides a comprehensive analysis of paclitaxel-induced neuropathic pain mechanisms and suggestions for effective interventions.
Collapse
|
6
|
Sánchez JC, Ehrlich BE. Functional Interaction between Transient Receptor Potential V4 Channel and Neuronal Calcium Sensor 1 and the Effects of Paclitaxel. Mol Pharmacol 2021; 100:258-270. [PMID: 34321341 PMCID: PMC8626786 DOI: 10.1124/molpharm.121.000244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 06/08/2021] [Indexed: 11/22/2022] Open
Abstract
Neuronal calcium sensor 1 (NCS1), a calcium-binding protein, and transient receptor potential V4 (TRPV4), a plasma membrane calcium channel, are fundamental in the regulation of calcium homeostasis. The interactions of these proteins and their regulation by paclitaxel (PTX) were investigated using biochemical, pharmacological, and electrophysiological approaches in both a breast cancer epithelial cell model and a neuronal model. TRPV4 and NCS1 reciprocally immunoprecipitated each other, suggesting that they make up a signaling complex. The functional consequence of this physical association was that TRPV4 currents increased with increased NCS1 expression. Calcium fluxes through TRPV4 correlated with the magnitude of TRPV4 currents, and these calcium fluxes depended on NCS1 expression levels. Exposure to PTX amplified the acute effects of TRPV4 expression, currents, and calcium fluxes but decreased the expression of NCS1. These findings augment the understanding of the properties of TRPV4, the role of NCS1 in the regulation of TRPV4, and the cellular mechanisms of PTX-induced neuropathy. SIGNIFICANCE STATEMENT: TRPV4 and NCS1 physically and functionally interact. Increased expression of NCS1 enhances TRPV4-dependent currents, which are further amplified by treatment with the chemotherapeutic drug paclitaxel, an effect associated with adverse effects of chemotherapy, including neuropathy.
Collapse
Affiliation(s)
- Julio C Sánchez
- Laboratory of Cell Physiology, Faculty of Health Sciences, Universidad Tecnológica de Pereira, Pereira, Colombia (J.C.S.), and Departments of Pharmacology and Cellular and Molecular Physiology, Yale University, New Haven, Connecticut (B.E.E.)
| | - Barbara E Ehrlich
- Laboratory of Cell Physiology, Faculty of Health Sciences, Universidad Tecnológica de Pereira, Pereira, Colombia (J.C.S.), and Departments of Pharmacology and Cellular and Molecular Physiology, Yale University, New Haven, Connecticut (B.E.E.)
| |
Collapse
|
7
|
Dason JS, Sokolowski MB. A cGMP-dependent protein kinase, encoded by the Drosophila foraging gene, regulates neurotransmission through changes in synaptic structure and function. J Neurogenet 2021; 35:213-220. [PMID: 33998378 DOI: 10.1080/01677063.2021.1905639] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
A cGMP-dependent protein kinase (PKG) encoded by the Drosophila foraging (for) gene regulates both synaptic structure (nerve terminal growth) and function (neurotransmission) through independent mechanisms at the Drosophila larval neuromuscular junction (nmj). Glial for is known to restrict nerve terminal growth, whereas presynaptic for inhibits synaptic vesicle (SV) exocytosis during low frequency stimulation. Presynaptic for also facilitates SV endocytosis during high frequency stimulation. for's effects on neurotransmission can occur independent of any changes in nerve terminal growth. However, it remains unclear if for's effects on neurotransmission affect nerve terminal growth. Furthermore, it's possible that for's effects on synaptic structure contribute to changes in neurotransmission. In the present study, we examined these questions using RNA interference to selectively knockdown for in presynaptic neurons or glia at the Drosophila larval nmj. Consistent with our previous findings, presynaptic knockdown of for impaired SV endocytosis, whereas knockdown of glial for had no effect on SV endocytosis. Surprisingly, we found that knockdown of either presynaptic or glial for increased neurotransmitter release in response to low frequency stimulation. Knockdown of presynaptic for did not affect nerve terminal growth, demonstrating that for's effects on neurotransmission does not alter nerve terminal growth. In contrast, knockdown of glial for enhanced nerve terminal growth. This enhanced nerve terminal growth was likely the cause of the enhanced neurotransmitter release seen following knockdown of glial for. Overall, we show that for can affect neurotransmitter release by regulating both synaptic structure and function.
Collapse
Affiliation(s)
- Jeffrey S Dason
- Department of Cell & Systems Biology, University of Toronto, Toronto, Canada.,Department of Biomedical Sciences, University of Windsor, Windsor, Canada
| | - Marla B Sokolowski
- Department of Cell & Systems Biology, University of Toronto, Toronto, Canada.,Department of Ecology & Evolutionary Biology, University of Toronto, Toronto, Canada.,Child and Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Canada
| |
Collapse
|
8
|
Comprehensive somatosensory and neurological phenotyping of NCS1 knockout mice. Sci Rep 2021; 11:2372. [PMID: 33504822 PMCID: PMC7840744 DOI: 10.1038/s41598-021-81650-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
Neuronal calcium sensor 1 (NCS1) regulates a wide range of cellular functions throughout the mammalian nervous systems. Altered NCS1 expression is associated with neurodevelopmental and neurodegenerative diseases. Previous studies focused on affective and cognitive behaviors in NCS1 knockout (KO) mice, but little is known about the physiological and pathological states associated with the loss of NCS1 in the peripheral nervous system. We previously reported that NCS1 expression was reduced following paclitaxel-induced peripheral neuropathy. Here, we comprehensively investigated the phenotypes of NCS1-KO mice through a battery of behavioral tests examining both central and peripheral nervous systems. Generally, only mild differences were observed in thermal sensation and memory acquisition between NCS1-WT and -KO male mice, but not in female mice. No differences were observed in motor performance, affective behaviors, and hearing in both sexes. These results suggest that NCS1 plays a modulatory role in sensory perceptions and cognition, particularly in male mice. NCS1 has been proposed as a pharmacological target for various diseases. Therefore, the sex-specific effects of NCS1 loss may be of clinical interest. As we examined a constitutive KO model, future studies focusing on various conditional KO models will further elucidate the precise physiological significance of NCS1.
Collapse
|
9
|
Allen AM, Neville MC, Birtles S, Croset V, Treiber CD, Waddell S, Goodwin SF. A single-cell transcriptomic atlas of the adult Drosophila ventral nerve cord. eLife 2020; 9:e54074. [PMID: 32314735 PMCID: PMC7173974 DOI: 10.7554/elife.54074] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 04/03/2020] [Indexed: 02/07/2023] Open
Abstract
The Drosophila ventral nerve cord (VNC) receives and processes descending signals from the brain to produce a variety of coordinated locomotor outputs. It also integrates sensory information from the periphery and sends ascending signals to the brain. We used single-cell transcriptomics to generate an unbiased classification of cellular diversity in the VNC of five-day old adult flies. We produced an atlas of 26,000 high-quality cells, representing more than 100 transcriptionally distinct cell types. The predominant gene signatures defining neuronal cell types reflect shared developmental histories based on the neuroblast from which cells were derived, as well as their birth order. The relative position of cells along the anterior-posterior axis could also be assigned using adult Hox gene expression. This single-cell transcriptional atlas of the adult fly VNC will be a valuable resource for future studies of neurodevelopment and behavior.
Collapse
Affiliation(s)
- Aaron M Allen
- Centre for Neural Circuits and Behaviour, University of OxfordOxfordUnited Kingdom
| | - Megan C Neville
- Centre for Neural Circuits and Behaviour, University of OxfordOxfordUnited Kingdom
| | - Sebastian Birtles
- Centre for Neural Circuits and Behaviour, University of OxfordOxfordUnited Kingdom
| | - Vincent Croset
- Centre for Neural Circuits and Behaviour, University of OxfordOxfordUnited Kingdom
| | | | - Scott Waddell
- Centre for Neural Circuits and Behaviour, University of OxfordOxfordUnited Kingdom
| | - Stephen F Goodwin
- Centre for Neural Circuits and Behaviour, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
10
|
Ng E, Georgiou J, Avila A, Trought K, Mun HS, Hodgson M, Servinis P, Roder JC, Collingridge GL, Wong AHC. Mice lacking neuronal calcium sensor-1 show social and cognitive deficits. Behav Brain Res 2019; 381:112420. [PMID: 31821787 DOI: 10.1016/j.bbr.2019.112420] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 12/06/2019] [Accepted: 12/06/2019] [Indexed: 12/18/2022]
Abstract
Neuronal calcium sensor-1 or Frequenin is a calcium sensor widely expressed in the nervous system, with roles in neurotransmission, neurite outgrowth, synaptic plasticity, learning, and motivated behaviours. Neuronal calcium sensor-1 has been implicated in neuropsychiatric disorders including autism spectrum disorder, schizophrenia, and bipolar disorder. However, the role of neuronal calcium sensor-1 in behavioural phenotypes and brain changes relevant to autism spectrum disorder have not been evaluated. We show that neuronal calcium sensor-1 deletion in the mouse leads to a mild deficit in social approach and impaired displaced object recognition without affecting social interactions, behavioural flexibility, spatial reference memory, anxiety-like behaviour, or sensorimotor gating. Morphologically, neuronal calcium sensor-1 deletion leads to increased dendritic arbour complexity in the frontal cortex. At the level of hippocampal synaptic plasticity, neuronal calcium sensor-1 deletion leads to a reduction in long-term potentiation in the dentate gyrus, but not area Cornu Ammonis 1. Metabotropic glutamate receptor-induced long-term depression was unaffected in both dentate and Cornu Ammonis 1. These studies identify roles for neuronal calcium sensor-1 in specific subregions of the brain including a phenotype relevant to neuropsychiatric disorders.
Collapse
Affiliation(s)
- Enoch Ng
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - John Georgiou
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
| | - Ariel Avila
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada; Basic Science Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción (UCSC), Concepción, 4090541, Chile
| | - Kathleen Trought
- Institute of Medical Science, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Ho-Suk Mun
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Meggie Hodgson
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
| | - Panayiotis Servinis
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
| | - John C Roder
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, M5S 1A8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Graham L Collingridge
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada; Tanz Centre for Research in Neurodegenerative Diseases and Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Albert H C Wong
- Institute of Medical Science, University of Toronto, Toronto, Ontario, M5S 1A8, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, M5T 1R8, Canada.
| |
Collapse
|
11
|
The Great Escape: how phosphatidylinositol 4-kinases and PI4P promote vesicle exit from the Golgi (and drive cancer). Biochem J 2019; 476:2321-2346. [DOI: 10.1042/bcj20180622] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/06/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022]
Abstract
Abstract
Phosphatidylinositol 4-phosphate (PI4P) is a membrane glycerophospholipid and a major regulator of the characteristic appearance of the Golgi complex as well as its vesicular trafficking, signalling and metabolic functions. Phosphatidylinositol 4-kinases, and in particular the PI4KIIIβ isoform, act in concert with PI4P to recruit macromolecular complexes to initiate the biogenesis of trafficking vesicles for several Golgi exit routes. Dysregulation of Golgi PI4P metabolism and the PI4P protein interactome features in many cancers and is often associated with tumour progression and a poor prognosis. Increased expression of PI4P-binding proteins, such as GOLPH3 or PITPNC1, induces a malignant secretory phenotype and the release of proteins that can remodel the extracellular matrix, promote angiogenesis and enhance cell motility. Aberrant Golgi PI4P metabolism can also result in the impaired post-translational modification of proteins required for focal adhesion formation and cell–matrix interactions, thereby potentiating the development of aggressive metastatic and invasive tumours. Altered expression of the Golgi-targeted PI 4-kinases, PI4KIIIβ, PI4KIIα and PI4KIIβ, or the PI4P phosphate Sac1, can also modulate oncogenic signalling through effects on TGN-endosomal trafficking. A Golgi trafficking role for a PIP 5-kinase has been recently described, which indicates that PI4P is not the only functionally important phosphoinositide at this subcellular location. This review charts new developments in our understanding of phosphatidylinositol 4-kinase function at the Golgi and how PI4P-dependent trafficking can be deregulated in malignant disease.
Collapse
|
12
|
Insights into real-time chemical processes in a calcium sensor protein-directed dynamic library. Nat Commun 2019; 10:2798. [PMID: 31243268 PMCID: PMC6595003 DOI: 10.1038/s41467-019-10627-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 05/22/2019] [Indexed: 12/12/2022] Open
Abstract
Dynamic combinatorial chemistry (DCC) has proven its potential in drug discovery speeding the identification of modulators of biological targets. However, the exchange chemistries typically take place under specific reaction conditions, with limited tools capable of operating under physiological parameters. Here we report a catalyzed protein-directed DCC working at low temperatures that allows the calcium sensor NCS-1 to find the best ligands in situ. Ultrafast NMR identifies the reaction intermediates of the acylhydrazone exchange, tracing the molecular assemblies and getting a real-time insight into the essence of DCC processes at physiological pH. Additionally, NMR, X-ray crystallography and computational methods are employed to elucidate structural and mechanistic aspects of the molecular recognition event. The DCC approach leads us to the identification of a compound stabilizing the NCS-1/Ric8a complex and whose therapeutic potential is proven in a Drosophila model of disease with synaptic alterations. Dynamic combinatorial chemistry (DCC) is instrumental in the discovery of ligands for pharmaceutical targets. Here, the authors adapted DCC to work at 4 degrees Celsius and used it to identify a ligand for Neuronal Calcium Sensor-1 that promotes NCS-1/Ric8a protein-protein interaction.
Collapse
|
13
|
Burgoyne RD, Helassa N, McCue HV, Haynes LP. Calcium Sensors in Neuronal Function and Dysfunction. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a035154. [PMID: 30833454 DOI: 10.1101/cshperspect.a035154] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Calcium signaling in neurons as in other cell types can lead to varied changes in cellular function. Neuronal Ca2+ signaling processes have also become adapted to modulate the function of specific pathways over a wide variety of time domains and these can have effects on, for example, axon outgrowth, neuronal survival, and changes in synaptic strength. Ca2+ also plays a key role in synapses as the trigger for fast neurotransmitter release. Given its physiological importance, abnormalities in neuronal Ca2+ signaling potentially underlie many different neurological and neurodegenerative diseases. The mechanisms by which changes in intracellular Ca2+ concentration in neurons can bring about diverse responses is underpinned by the roles of ubiquitous or specialized neuronal Ca2+ sensors. It has been established that synaptotagmins have key functions in neurotransmitter release, and, in addition to calmodulin, other families of EF-hand-containing neuronal Ca2+ sensors, including the neuronal calcium sensor (NCS) and the calcium-binding protein (CaBP) families, play important physiological roles in neuronal Ca2+ signaling. It has become increasingly apparent that these various Ca2+ sensors may also be crucial for aspects of neuronal dysfunction and disease either indirectly or directly as a direct consequence of genetic variation or mutations. An understanding of the molecular basis for the regulation of the targets of the Ca2+ sensors and the physiological roles of each protein in identified neurons may contribute to future approaches to the development of treatments for a variety of human neuronal disorders.
Collapse
Affiliation(s)
- Robert D Burgoyne
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Nordine Helassa
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Hannah V McCue
- Centre for Genomic Research, University of Liverpool, Liverpool, United Kingdom
| | - Lee P Haynes
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
14
|
Nakamura TY, Nakao S, Wakabayashi S. Emerging Roles of Neuronal Ca 2+ Sensor-1 in Cardiac and Neuronal Tissues: A Mini Review. Front Mol Neurosci 2019; 12:56. [PMID: 30886571 PMCID: PMC6409499 DOI: 10.3389/fnmol.2019.00056] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 02/15/2019] [Indexed: 11/13/2022] Open
Abstract
The EF-hand calcium (Ca2+)-binding protein, neuronal Ca2+ sensor-1 (NCS-1/frequenin), is predominantly expressed in neuronal tissues and plays a crucial role in neuronal functions, including synaptic transmission and plasticity. NCS-1 has diverse functional roles, as elucidated in the past 15 years, which include the regulation of phosphatidylinositol 4-kinase IIIβ (PI-4K-β) and several ion channels such as voltage-gated K+ and Ca2+ channels, the D2 dopamine receptors, and inositol 1,4,5-trisphosphate receptors (InsP3Rs). Functional analyses demonstrated that NCS-1 enhances exocytosis and neuronal survival after injury, as well as promotes learning and memory in mice. NCS-1 is also expressed in the heart including the Purkinje fibers (PFs) of the conduction system. NCS-1 interacts with KV4 K+ channels together with dipeptidyl peptidase-like protein-6 (DPP-6), and this macromolecule then composes the transient outward current in PFs and contributes to the repolarization of PF action potential, thus being responsible for idiopathic arrhythmia. Moreover, NCS-1 expression was reported to be significantly high at the immature stage and at hypertrophy in adults. That report demonstrated that NCS-1 positively regulates cardiac contraction in immature hearts by increasing intracellular Ca2+ signals through interaction with InsP3Rs. With the related signals, NCS-1 activates nuclear Ca2+ signals, which would be a mechanism underlying hormone-induced cardiac hypertrophy. Furthermore, NCS-1 contributes to stress tolerance in cardiomyocytes by activating mitochondrial detoxification pathways, with a key role in Ca2+-dependent pathways. In this review, we will discuss recent findings supporting the functional significance of NCS-1 in the brain and heart and will address possible underlying molecular mechanisms.
Collapse
Affiliation(s)
- Tomoe Y Nakamura
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Shu Nakao
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan.,Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Japan
| | - Shigeo Wakabayashi
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan.,Department of Pharmacology, Osaka Medical College, Takatsuki, Japan
| |
Collapse
|
15
|
Dason JS, Allen AM, Vasquez OE, Sokolowski MB. Distinct functions of a cGMP-dependent protein kinase in nerve terminal growth and synaptic vesicle cycling. J Cell Sci 2019; 132:jcs.227165. [DOI: 10.1242/jcs.227165] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 02/26/2019] [Indexed: 01/20/2023] Open
Abstract
Sustained neurotransmission requires the tight coupling of synaptic vesicle (SV) exocytosis and endocytosis. The mechanisms underlying this coupling are poorly understood. We tested the hypothesis that a cGMP-dependent protein kinase (PKG), encoded by the foraging (for) gene in Drosophila melanogaster, is critical for this process using a for null mutant, genomic rescues, and tissue specific rescues. We uncoupled FOR's exocytic and endocytic functions in neurotransmission using a temperature-sensitive shibire mutant in conjunction with fluorescein-assisted light inactivation of FOR. We discovered a dual role for presynaptic FOR, where FOR inhibits SV exocytosis during low frequency stimulation by negatively regulating presynaptic Ca2+ levels and maintains neurotransmission during high frequency stimulation by facilitating SV endocytosis. Additionally, glial FOR negatively regulated nerve terminal growth through TGF-β signaling and this developmental effect was independent from FOR's effects on neurotransmission. Overall, FOR plays a critical role in coupling SV exocytosis and endocytosis, thereby balancing these two components to maintain sustained neurotransmission.
Collapse
Affiliation(s)
- Jeffrey S. Dason
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, M5S 3B2, Canada
- Department of Biological Sciences, University of Windsor, Windsor, Ontario, N9B 3P4, Canada
| | - Aaron M. Allen
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, M5S 3B2, Canada
- Present Address: Centre for Neural Circuits and Behaviour, University of Oxford, OX1 3SR Oxford, UK
| | - Oscar E. Vasquez
- Department of Ecology & Evolutionary Biology, University of Toronto, Toronto, Ontario, M5S 3B2, Canada
| | - Marla B. Sokolowski
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, M5S 3B2, Canada
- Department of Ecology & Evolutionary Biology, University of Toronto, Toronto, Ontario, M5S 3B2, Canada
- Child and Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, M5G 1M1, Canada
| |
Collapse
|
16
|
Dason JS, Sokolowski MB, Wu CF. A reductionist approach to understanding the nervous system: the Harold Atwood legacy. J Neurogenet 2018; 32:127-130. [PMID: 30484389 DOI: 10.1080/01677063.2018.1504044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Jeffrey S Dason
- a Department of Biological Sciences , University of Windsor , Windsor , Canada
| | - Marla B Sokolowski
- b Department of Cell & Systems Biology , University of Toronto , Toronto , Canada.,c Department of Ecology & Evolutionary Biology , University of Toronto , Toronto , Canada.,d Child and Brain Development Program , Canadian Institute for Advanced Research (CIFAR) , Toronto , Canada
| | - Chun-Fang Wu
- e Department of Biology , University of Iowa , Iowa City , IA , USA
| |
Collapse
|
17
|
Xing X, Wu CF. Inter-relationships among physical dimensions, distal-proximal rank orders, and basal GCaMP fluorescence levels in Ca 2+ imaging of functionally distinct synaptic boutons at Drosophila neuromuscular junctions. J Neurogenet 2018; 32:195-208. [PMID: 30322321 DOI: 10.1080/01677063.2018.1504043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
GCaMP imaging is widely employed for investigating neuronal Ca2+ dynamics. The Drosophila larval neuromuscular junction (NMJ) consists of three distinct types of motor terminals (type Ib, Is and II). We investigated whether variability in synaptic bouton sizes and GCaMP expression levels confound interpretations of GCaMP readouts, in inferring the intrinsic Ca2+ handling properties among these functionally distinct synapses. Analysis of large data sets accumulated over years established the wide ranges of bouton sizes and GCaMP baseline fluorescence, with large overlaps among synaptic categories. We showed that bouton size and GCaMP baseline fluorescence were not confounding factors in determining the characteristic frequency responses among type Ib, Is and II synapses. More importantly, the drastic phenotypes that hyperexcitability mutations manifest preferentially in particular synaptic categories, were not obscured by bouton heterogeneity in physical size and GCaMP expression level. Our data enabled an extensive analysis of the distal-proximal gradient of GCaMP responses upon genetic and pharmacological manipulations. The results illustrate the conditions that disrupt or enhance the distal-proximal gradients. For example, stimulus frequencies just above the threshold level produced the steepest gradient in low Ca2+ (0.1 mM) saline, while supra-threshold stimulation flattened the gradient. Moreover, membrane hyperexcitability mutations (eag1 Sh120 and parabss1) and mitochondrial inhibition by dinitrophenol (DNP) disrupted the gradient. However, a novel distal-proximal gradient of decay kinetics appeared after long-term DNP incubation. We performed focal recording to assess the failure rates in transmission at low Ca2+ levels, which yielded indications of a mild distal-proximal gradient in release probability.
Collapse
Affiliation(s)
- Xiaomin Xing
- a Department of Biology , University of Iowa , Iowa City , IA , USA
| | - Chun-Fang Wu
- a Department of Biology , University of Iowa , Iowa City , IA , USA
| |
Collapse
|
18
|
Mansilla A, Jordán-Álvarez S, Santana E, Jarabo P, Casas-Tintó S, Ferrús A. Molecular mechanisms that change synapse number. J Neurogenet 2018; 32:155-170. [DOI: 10.1080/01677063.2018.1506781] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
19
|
Cantarutti KC, Burgess J, Brill JA, Dason JS. Type II phosphatidylinositol 4-kinase regulates nerve terminal growth and synaptic vesicle recycling. J Neurogenet 2018; 32:230-235. [DOI: 10.1080/01677063.2018.1502762] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
| | - Jason Burgess
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Julie A. Brill
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Jeffrey S. Dason
- Department of Biological Sciences, University of Windsor, Windsor, Canada
| |
Collapse
|
20
|
Lowe SA, Hodge JJL, Usowicz MM. A third copy of the Down syndrome cell adhesion molecule (Dscam) causes synaptic and locomotor dysfunction in Drosophila. Neurobiol Dis 2017; 110:93-101. [PMID: 29196216 PMCID: PMC5773243 DOI: 10.1016/j.nbd.2017.11.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/13/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023] Open
Abstract
Down syndrome (DS) is caused by triplication of chromosome 21 (HSA21). It is characterised by intellectual disability and impaired motor coordination that arise from changes in brain volume, structure and function. However, the contribution of each HSA21 gene to these various phenotypes and to the causal alterations in neuronal and synaptic structure and function are largely unknown. Here we have investigated the effect of overexpression of the HSA21 gene DSCAM (Down syndrome cell adhesion molecule), on glutamatergic synaptic transmission and motor coordination, using Drosophila expressing three copies of Dscam1. Electrophysiological recordings of miniature and evoked excitatory junction potentials at the glutamatergic neuromuscular junction of Drosophila larvae showed that the extra copy of Dscam1 changed the properties of spontaneous and electrically-evoked transmitter release and strengthened short-term synaptic depression during high-frequency firing of the motor nerve. Behavioural analyses uncovered impaired locomotor coordination despite preserved gross motor function. This work identifies DSCAM as a candidate causative gene in DS that is sufficient to modify synaptic transmission and synaptic plasticity and cause a DS behavioural phenotype. Drosophila expressing a third copy of Dscam have altered neuromuscular transmission. Drosophila expressing a third copy of Dscam have deficits in locomotor coordination. Drosophila are a powerful system for studying single-gene effects in Down syndrome.
Collapse
Affiliation(s)
- Simon A Lowe
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - James J L Hodge
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol BS8 1TD, UK.
| | - Maria M Usowicz
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol BS8 1TD, UK.
| |
Collapse
|
21
|
Gorkhali R, Huang K, Kirberger M, Yang JJ. Defining potential roles of Pb(2+) in neurotoxicity from a calciomics approach. Metallomics 2017; 8:563-78. [PMID: 27108875 DOI: 10.1039/c6mt00038j] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Metal ions play crucial roles in numerous biological processes, facilitating biochemical reactions by binding to various proteins. An increasing body of evidence suggests that neurotoxicity associated with exposure to nonessential metals (e.g., Pb(2+)) involves disruption of synaptic activity, and these observed effects are associated with the ability of Pb(2+) to interfere with Zn(2+) and Ca(2+)-dependent functions. However, the molecular mechanism behind Pb(2+) toxicity remains a topic of debate. In this review, we first discuss potential neuronal Ca(2+) binding protein (CaBP) targets for Pb(2+) such as calmodulin (CaM), synaptotagmin, neuronal calcium sensor-1 (NCS-1), N-methyl-d-aspartate receptor (NMDAR) and family C of G-protein coupled receptors (cGPCRs), and their involvement in Ca(2+)-signalling pathways. We then compare metal binding properties between Ca(2+) and Pb(2+) to understand the structural implications of Pb(2+) binding to CaBPs. Statistical and biophysical studies (e.g., NMR and fluorescence spectroscopy) of Pb(2+) binding are discussed to investigate the molecular mechanism behind Pb(2+) toxicity. These studies identify an opportunistic, allosteric binding of Pb(2+) to CaM, which is distinct from ionic displacement. Together, these data suggest three potential modes of Pb(2+) activity related to molecular and/or neural toxicity: (i) Pb(2+) can occupy Ca(2+)-binding sites, inhibiting the activity of the protein by structural modulation, (ii) Pb(2+) can mimic Ca(2+) in the binding sites, falsely activating the protein and perturbing downstream activities, or (iii) Pb(2+) can bind outside of the Ca(2+)-binding sites, resulting in the allosteric modulation of the protein activity. Moreover, the data further suggest that even low concentrations of Pb(2+) can interfere at multiple points within the neuronal Ca(2+) signalling pathways to cause neurotoxicity.
Collapse
Affiliation(s)
- Rakshya Gorkhali
- Department of Chemistry, Center for Diagnostics and Therapeutics, and Drug Design and Biotechnology, Georgia State University, Atlanta, GA 3030, USA.
| | - Kenneth Huang
- Department of Chemistry, Center for Diagnostics and Therapeutics, and Drug Design and Biotechnology, Georgia State University, Atlanta, GA 3030, USA.
| | - Michael Kirberger
- Department of Chemistry and Physics, Clayton State University, Morrow, GA 30260, USA.
| | - Jenny J Yang
- Department of Chemistry, Center for Diagnostics and Therapeutics, and Drug Design and Biotechnology, Georgia State University, Atlanta, GA 3030, USA.
| |
Collapse
|
22
|
Davis JK, Broadie K. Multifarious Functions of the Fragile X Mental Retardation Protein. Trends Genet 2017; 33:703-714. [PMID: 28826631 PMCID: PMC5610095 DOI: 10.1016/j.tig.2017.07.008] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/20/2017] [Accepted: 07/24/2017] [Indexed: 01/01/2023]
Abstract
Fragile X syndrome (FXS), a heritable intellectual and autism spectrum disorder (ASD), results from the loss of Fragile X mental retardation protein (FMRP). This neurodevelopmental disease state exhibits neural circuit hyperconnectivity and hyperexcitability. Canonically, FMRP functions as an mRNA-binding translation suppressor, but recent findings have enormously expanded its proposed roles. Although connections between burgeoning FMRP functions remain unknown, recent advances have extended understanding of its involvement in RNA, channel, and protein binding that modulate calcium signaling, activity-dependent critical period development, and the excitation-inhibition (E/I) neural circuitry balance. In this review, we contextualize 3 years of FXS model research. Future directions extrapolated from recent advances focus on discovering links between FMRP roles to determine whether FMRP has a multitude of unrelated functions or whether combinatorial mechanisms can explain its multifaceted existence.
Collapse
Affiliation(s)
- Jenna K Davis
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, USA
| | - Kendal Broadie
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, USA.
| |
Collapse
|
23
|
Jackman SL, Regehr WG. The Mechanisms and Functions of Synaptic Facilitation. Neuron 2017; 94:447-464. [PMID: 28472650 DOI: 10.1016/j.neuron.2017.02.047] [Citation(s) in RCA: 255] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 02/23/2017] [Accepted: 02/28/2017] [Indexed: 12/22/2022]
Abstract
The ability of the brain to store and process information relies on changing the strength of connections between neurons. Synaptic facilitation is a form of short-term plasticity that enhances synaptic transmission for less than a second. Facilitation is a ubiquitous phenomenon thought to play critical roles in information transfer and neural processing. Yet our understanding of the function of facilitation remains largely theoretical. Here we review proposed roles for facilitation and discuss how recent progress in uncovering the underlying molecular mechanisms could enable experiments that elucidate how facilitation, and short-term plasticity in general, contributes to circuit function and animal behavior.
Collapse
Affiliation(s)
- Skyler L Jackman
- Department of Neurobiology, Harvard Medical School, Boston, MA 02118, USA
| | - Wade G Regehr
- Department of Neurobiology, Harvard Medical School, Boston, MA 02118, USA.
| |
Collapse
|
24
|
Mansilla A, Chaves-Sanjuan A, Campillo NE, Semelidou O, Martínez-González L, Infantes L, González-Rubio JM, Gil C, Conde S, Skoulakis EMC, Ferrús A, Martínez A, Sánchez-Barrena MJ. Interference of the complex between NCS-1 and Ric8a with phenothiazines regulates synaptic function and is an approach for fragile X syndrome. Proc Natl Acad Sci U S A 2017; 114:E999-E1008. [PMID: 28119500 PMCID: PMC5307446 DOI: 10.1073/pnas.1611089114] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The protein complex formed by the Ca2+ sensor neuronal calcium sensor 1 (NCS-1) and the guanine exchange factor protein Ric8a coregulates synapse number and probability of neurotransmitter release, emerging as a potential therapeutic target for diseases affecting synapses, such as fragile X syndrome (FXS), the most common heritable autism disorder. Using crystallographic data and the virtual screening of a chemical library, we identified a set of heterocyclic small molecules as potential inhibitors of the NCS-1/Ric8a interaction. The aminophenothiazine FD44 interferes with NCS-1/Ric8a binding, and it restores normal synapse number and associative learning in a Drosophila FXS model. The synaptic effects elicited by FD44 feeding are consistent with the genetic manipulation of NCS-1. The crystal structure of NCS-1 bound to FD44 and the structure-function studies performed with structurally close analogs explain the FD44 specificity and the mechanism of inhibition, in which the small molecule stabilizes a mobile C-terminal helix inside a hydrophobic crevice of NCS-1 to impede Ric8a interaction. Our study shows the drugability of the NCS-1/Ric8a interface and uncovers a suitable region in NCS-1 for development of additional drugs of potential use on FXS and related synaptic disorders.
Collapse
Affiliation(s)
- Alicia Mansilla
- Departamento de Neurobiología del Desarrollo, Instituto Cajal, Spanish National Research Council, 28002 Madrid, Spain
| | - Antonio Chaves-Sanjuan
- Departamento de Cristalografía y Biología Estructural, Instituto de Química Física Rocasolano, Spanish National Research Council, 28006 Madrid, Spain
| | - Nuria E Campillo
- Centro de Investigaciones Biológicas, Spanish National Research Council, 28040 Madrid, Spain
| | - Ourania Semelidou
- Division of Neuroscience, Biomedical Sciences Research Centre Alexander Fleming, 16672 Vari, Greece
| | | | - Lourdes Infantes
- Departamento de Cristalografía y Biología Estructural, Instituto de Química Física Rocasolano, Spanish National Research Council, 28006 Madrid, Spain
| | - Juana María González-Rubio
- Departamento de Cristalografía y Biología Estructural, Instituto de Química Física Rocasolano, Spanish National Research Council, 28006 Madrid, Spain
| | - Carmen Gil
- Centro de Investigaciones Biológicas, Spanish National Research Council, 28040 Madrid, Spain
| | - Santiago Conde
- Instituto de Química Médica, Spanish National Research Council, 28006 Madrid, Spain
| | - Efthimios M C Skoulakis
- Division of Neuroscience, Biomedical Sciences Research Centre Alexander Fleming, 16672 Vari, Greece
| | - Alberto Ferrús
- Departamento de Neurobiología del Desarrollo, Instituto Cajal, Spanish National Research Council, 28002 Madrid, Spain
| | - Ana Martínez
- Centro de Investigaciones Biológicas, Spanish National Research Council, 28040 Madrid, Spain
| | - María José Sánchez-Barrena
- Departamento de Cristalografía y Biología Estructural, Instituto de Química Física Rocasolano, Spanish National Research Council, 28006 Madrid, Spain;
| |
Collapse
|
25
|
Gong Y, Zhu Y, Zou Y, Ma B, Nussinov R, Zhang Q. Human Neuronal Calcium Sensor-1 Protein Avoids Histidine Residues To Decrease pH Sensitivity. J Phys Chem B 2017; 121:508-517. [PMID: 28030949 PMCID: PMC6413881 DOI: 10.1021/acs.jpcb.6b11094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
pH is highly regulated in mammalian central nervous systems. Neuronal calcium sensor-1 (NCS-1) can interact with numerous target proteins. Compared to that in the NCS-1 protein of Caenorhabditis elegans, evolution has avoided the placement of histidine residues at positions 102 and 83 in the NCS-1 protein of humans and Xenopus laevis, possibly to decrease the conformational sensitivity to pH gradients in synaptic processes. We used all-atom molecular dynamics simulations to investigate the effects of amino acid substitutions between species on human NCS-1 by substituting Arg102 and Ser83 for histidine at neutral (R102H and S83H) and acidic pHs (R102Hp and S83Hp). Our cumulative 5 μs simulations revealed that the R102H mutation slightly increases the structural flexibility of loop L2 and the R102Hp mutation decreases protein stability. Community network analysis illustrates that the R102H and S83H mutations weaken the interdomain and strengthen the intradomain communications. Secondary structure contents in the S83H and S83Hp mutants are similar to those in the wild type, whereas the global structural stabilities and salt-bridge probabilities decrease. This study highlights the conformational dynamics effects of the R102H and S83H mutations on the local structural flexibility and global stability of NCS-1, whereas protonated histidine decreases the stability of NCS-1. Thus, histidines at positions 102 and 83 may not be compatible with the function of NCS-1 whether in the neutral or protonated state.
Collapse
Affiliation(s)
- Yehong Gong
- College of Physical Education and Training, Shanghai University of Sport, 399 Chang Hai Road, Shanghai, 200438, China
| | - Yuzhen Zhu
- Shanghai Normal University Physical Education College, 100 Gui Lin Road, Shanghai, 200234, China
| | - Yu Zou
- College of Physical Education and Training, Shanghai University of Sport, 399 Chang Hai Road, Shanghai, 200438, China
| | - Buyong Ma
- Basic Science Program, Leidos Biomedical Research, Inc. Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Ruth Nussinov
- Basic Science Program, Leidos Biomedical Research, Inc. Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702, United States
- Department of Human Genetics and Molecular Medicine, Sackler Inst. of Molecular Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Qingwen Zhang
- College of Physical Education and Training, Shanghai University of Sport, 399 Chang Hai Road, Shanghai, 200438, China
| |
Collapse
|
26
|
Nakamura TY, Nakao S, Nakajo Y, Takahashi JC, Wakabayashi S, Yanamoto H. Possible Signaling Pathways Mediating Neuronal Calcium Sensor-1-Dependent Spatial Learning and Memory in Mice. PLoS One 2017; 12:e0170829. [PMID: 28122057 PMCID: PMC5266288 DOI: 10.1371/journal.pone.0170829] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 01/11/2017] [Indexed: 01/10/2023] Open
Abstract
Intracellular Ca2+ signaling regulates diverse functions of the nervous system. Many of these neuronal functions, including learning and memory, are regulated by neuronal calcium sensor-1 (NCS-1). However, the pathways by which NCS-1 regulates these functions remain poorly understood. Consistent with the findings of previous reports, we revealed that NCS-1 deficient (Ncs1-/-) mice exhibit impaired spatial learning and memory function in the Morris water maze test, although there was little change in their exercise activity, as determined via treadmill-analysis. Expression of brain-derived neurotrophic factor (BDNF; a key regulator of memory function) and dopamine was significantly reduced in the Ncs1-/- mouse brain, without changes in the levels of glial cell-line derived neurotrophic factor or nerve growth factor. Although there were no gross structural abnormalities in the hippocampi of Ncs1-/- mice, electron microscopy analysis revealed that the density of large dense core vesicles in CA1 presynaptic neurons, which release BDNF and dopamine, was decreased. Phosphorylation of Ca2+/calmodulin-dependent protein kinase II-α (CaMKII-α, which is known to trigger long-term potentiation and increase BDNF levels, was significantly reduced in the Ncs1-/- mouse brain. Furthermore, high voltage electric potential stimulation, which increases the levels of BDNF and promotes spatial learning, significantly increased the levels of NCS-1 concomitant with phosphorylated CaMKII-α in the hippocampus; suggesting a close relationship between NCS-1 and CaMKII-α. Our findings indicate that NCS-1 may regulate spatial learning and memory function at least in part through activation of CaMKII-α signaling, which may directly or indirectly increase BDNF production.
Collapse
Affiliation(s)
- Tomoe Y. Nakamura
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
- * E-mail:
| | - Shu Nakao
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Yukako Nakajo
- Laboratory of Neurology and Neurosurgery, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Jun C. Takahashi
- Department of Neurosurgery, National Cerebral and Cardiovascular Center Hospital, Suita, Osaka, Japan
| | - Shigeo Wakabayashi
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Hiroji Yanamoto
- Laboratory of Neurology and Neurosurgery, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| |
Collapse
|
27
|
Zhu Y, Ma B, Nussinov R, Zhang Q. Temperature-Dependent Conformational Properties of Human Neuronal Calcium Sensor-1 Protein Revealed by All-Atom Simulations. J Phys Chem B 2016; 120:3551-9. [PMID: 27007011 PMCID: PMC6415918 DOI: 10.1021/acs.jpcb.5b12299] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Neuronal calcium sensor-1 (NCS-1) protein has orthologues from Saccharomyces cerevisiae to human with highly conserved amino acid sequences. NCS-1 is an important factor controlling the animal's response to temperature change. This leads us to investigate the temperature effects on the conformational dynamics of human NCS-1 at 310 and 316 K by all-atom molecular dynamics (MD) simulations and dynamic community network analysis. Four independent 500 ns MD simulations show that secondary structure content at 316 K is similar to that at 310 K, whereas the global protein structure is expanded. Loop 3 (L3) adopts an extended state occuping the hydrophobic crevice, and the number of suboptimal communication paths between residue D176 and V190 is reduced at 316 K. The dynamic community network analysis suggests that the interdomain correlation is weakened, and the intradomain coupling is strengthened at 316 K. The elevated temperature reduces the number of the salt bridges, especially in C-domain. This study suggests that the elevated temperature affects the conformational dynamics of human NCS-1 protein. Comparison of the structural dynamics of R102Q mutant and Δ176-190 truncated NCS-1 suggests that the structural and dynamical response of NCS-1 protein to elevated temperature may be one of its intrinsic functional properties.
Collapse
Affiliation(s)
- Yuzhen Zhu
- College of Physical Education and Training, Shanghai University of Sport, 399 Chang Hai Road, Shanghai, 200438, China
| | - Buyong Ma
- Basic Science Program, Leidos Biomedical Research, Inc. Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Ruth Nussinov
- Basic Science Program, Leidos Biomedical Research, Inc. Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702, United States
- Sackler Inst. of Molecular Medicine Department of Human Genetics and Molecular Medicine Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Qingwen Zhang
- College of Physical Education and Training, Shanghai University of Sport, 399 Chang Hai Road, Shanghai, 200438, China
| |
Collapse
|
28
|
Neuronal calcium sensor-1 deletion in the mouse decreases motivation and dopamine release in the nucleus accumbens. Behav Brain Res 2016; 301:213-25. [DOI: 10.1016/j.bbr.2015.12.037] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 12/18/2015] [Accepted: 12/22/2015] [Indexed: 12/24/2022]
|
29
|
Mun HS, Saab BJ, Ng E, McGirr A, Lipina TV, Gondo Y, Georgiou J, Roder JC. Self-directed exploration provides a Ncs1-dependent learning bonus. Sci Rep 2015; 5:17697. [PMID: 26639399 PMCID: PMC4671055 DOI: 10.1038/srep17697] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 11/02/2015] [Indexed: 11/21/2022] Open
Abstract
Understanding the mechanisms of memory formation is fundamental to establishing optimal educational practices and restoring cognitive function in brain disease. Here, we show for the first time in a non-primate species, that spatial learning receives a special bonus from self-directed exploration. In contrast, when exploration is escape-oriented, or when the full repertoire of exploratory behaviors is reduced, no learning bonus occurs. These findings permitted the first molecular and cellular examinations into the coupling of exploration to learning. We found elevated expression of neuronal calcium sensor 1 (Ncs1) and dopamine type-2 receptors upon self-directed exploration, in concert with increased neuronal activity in the hippocampal dentate gyrus and area CA3, as well as the nucleus accumbens. We probed further into the learning bonus by developing a point mutant mouse (Ncs1P144S/P144S) harboring a destabilized NCS-1 protein, and found this line lacked the equivalent self-directed exploration learning bonus. Acute knock-down of Ncs1 in the hippocampus also decoupled exploration from efficient learning. These results are potentially relevant for augmenting learning and memory in health and disease, and provide the basis for further molecular and circuit analyses in this direction.
Collapse
Affiliation(s)
- Ho-Suk Mun
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Bechara J Saab
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Preclinical Laboratory for Translational Research into Affective Disorders, University of Zurich Hospital for Psychiatry, August-Forel-Str 7, CH-8008, Zurich, Switzerland.,Neuroscience Center Zurich, Zurich, CH-8057, Switzerland
| | - Enoch Ng
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Alexander McGirr
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada.,Department of Psychiatry, University of British Columbia, Vancouver, BC, V6T 2A1, Canada
| | - Tatiana V Lipina
- Federal State Budgetary Scientific Institution, Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, 630117, Russia
| | - Yoichi Gondo
- Mutagenesis and Genomics Team, RIKEN BioResource Center, Tsukuba 305-0074, Japan
| | - John Georgiou
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - John C Roder
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| |
Collapse
|
30
|
Sastre A, Campillo NE, Gil C, Martinez A. Therapeutic approaches for the future treatment of Fragile X. Curr Opin Behav Sci 2015. [DOI: 10.1016/j.cobeha.2015.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
31
|
Jia L, Zhang D, Xiang Z, He N. Nonfunctional ingestion of plant miRNAs in silkworm revealed by digital droplet PCR and transcriptome analysis. Sci Rep 2015; 5:12290. [PMID: 26195298 PMCID: PMC4508662 DOI: 10.1038/srep12290] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 06/19/2015] [Indexed: 01/06/2023] Open
Abstract
Since a plant miRNA (miR168) cross-regulating a mammalian transcript was reported, miRNA-mediated cross-kingdom communication has become one of the most compelling but controversial topics. In the present study, we used silkworm and mulberry, which is a model for studies on the interactions between the insect and its host plant, to address whether miRNA-mediated cross-kingdom communication is a common phenomenon. The results of TA clone, Sanger sequencing and droplet digital PCR demonstrated that several mulberry-derived miRNAs could enter to silkworm hemolymph and multiple tested tissues. Synthetic miR166b was also detected in hemolymph and fat body. However, the ingestion of synthetic miR166b did not play roles in silkworm physiological progress, which was revealed by RNA-seq analyses, RT-PCR, and phenotypic investigations. Mulberry miRNAs are convincingly transferred to the silkworm orally and no physiological process associated with the miRNAs was demonstrable. The results provided a new aspect of cross-kingdom miRNA transfer.
Collapse
Affiliation(s)
- Ling Jia
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Beibei, Chongqing 400715, P. R. China
| | - Dayan Zhang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Beibei, Chongqing 400715, P. R. China
| | - Zhonghuai Xiang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Beibei, Chongqing 400715, P. R. China
| | - Ningjia He
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Beibei, Chongqing 400715, P. R. China
| |
Collapse
|
32
|
Yan J, Leal K, Magupalli VG, Nanou E, Martinez GQ, Scheuer T, Catterall WA. Modulation of CaV2.1 channels by neuronal calcium sensor-1 induces short-term synaptic facilitation. Mol Cell Neurosci 2015; 63:124-31. [PMID: 25447945 DOI: 10.1016/j.mcn.2014.11.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 10/29/2014] [Accepted: 11/03/2014] [Indexed: 12/01/2022] Open
Abstract
Facilitation and inactivation of P/Q-type Ca2+ currents mediated by Ca2+/calmodulin binding to Ca(V)2.1 channels contribute to facilitation and rapid depression of synaptic transmission, respectively. Other calcium sensor proteins displace calmodulin from its binding site and differentially modulate P/Q-type Ca2 + currents, resulting in diverse patterns of short-term synaptic plasticity. Neuronal calcium sensor-1 (NCS-1, frequenin) has been shown to enhance synaptic facilitation, but the underlying mechanism is unclear. We report here that NCS-1 directly interacts with IQ-like motif and calmodulin-binding domain in the C-terminal domain of Ca(V)2.1 channel. NCS-1 reduces Ca2 +-dependent inactivation of P/Q-type Ca2+ current through interaction with the IQ-like motif and calmodulin-binding domain without affecting peak current or activation kinetics. Expression of NCS-1 in presynaptic superior cervical ganglion neurons has no effect on synaptic transmission, eliminating effects of this calcium sensor protein on endogenous N-type Ca2+ currents and the endogenous neurotransmitter release machinery. However, in superior cervical ganglion neurons expressing wild-type Ca(V)2.1 channels, co-expression of NCS-1 induces facilitation of synaptic transmission in response to paired pulses and trains of depolarizing stimuli, and this effect is lost in Ca(V)2.1 channels with mutations in the IQ-like motif and calmodulin-binding domain. These results reveal that NCS-1 directly modulates Ca(V)2.1 channels to induce short-term synaptic facilitation and further demonstrate that CaS proteins are crucial in fine-tuning short-term synaptic plasticity.
Collapse
|
33
|
Burgoyne RD, Haynes LP. Sense and specificity in neuronal calcium signalling. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:1921-32. [PMID: 25447549 PMCID: PMC4728190 DOI: 10.1016/j.bbamcr.2014.10.029] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 10/25/2014] [Accepted: 10/29/2014] [Indexed: 11/02/2022]
Abstract
Changes in the intracellular free calcium concentration ([Ca²⁺]i) in neurons regulate many and varied aspects of neuronal function over time scales from microseconds to days. The mystery is how a single signalling ion can lead to such diverse and specific changes in cell function. This is partly due to aspects of the Ca²⁺ signal itself, including its magnitude, duration, localisation and persistent or oscillatory nature. The transduction of the Ca²⁺ signal requires Ca²⁺binding to various Ca²⁺ sensor proteins. The different properties of these sensors are important for differential signal processing and determine the physiological specificity of Ca(2+) signalling pathways. A major factor underlying the specific roles of particular Ca²⁺ sensor proteins is the nature of their interaction with target proteins and how this mediates unique patterns of regulation. We review here recent progress from structural analyses and from functional analyses in model organisms that have begun to reveal the rules that underlie Ca²⁺ sensor protein specificity for target interaction. We discuss three case studies exemplifying different aspects of Ca²⁺ sensor/target interaction. This article is part of a special issue titled the 13th European Symposium on Calcium.
Collapse
Affiliation(s)
- Robert D Burgoyne
- Department of Cellular and Molecular Physiology, The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool, L69 3BX, United Kingdom.
| | - Lee P Haynes
- Department of Cellular and Molecular Physiology, The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool, L69 3BX, United Kingdom
| |
Collapse
|
34
|
Lian LY, Pandalaneni SR, Todd PAC, Martin VM, Burgoyne RD, Haynes LP. Demonstration of binding of neuronal calcium sensor-1 to the cav2.1 p/q-type calcium channel. Biochemistry 2014; 53:6052-62. [PMID: 25188201 PMCID: PMC4180279 DOI: 10.1021/bi500568v] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In neurons, entry of extracellular calcium (Ca(2+)) into synaptic terminals through Cav2.1 (P/Q-type) Ca(2+) channels is the driving force for exocytosis of neurotransmitter-containing synaptic vesicles. This class of Ca(2+) channel is, therefore, pivotal during normal neurotransmission in higher organisms. In response to channel opening and Ca(2+) influx, specific Ca(2+)-binding proteins associate with cytoplasmic regulatory domains of the P/Q channel to modulate subsequent channel opening. Channel modulation in this way influences synaptic plasticity with consequences for higher-level processes such as learning and memory acquisition. The ubiquitous Ca(2+)-sensing protein calmodulin (CaM) regulates the activity of all types of mammalian voltage-gated Ca(2+) channels, including the P/Q class, by direct binding to specific regulatory motifs. More recently, experimental evidence has highlighted a role for additional Ca(2+)-binding proteins, particularly of the CaBP and NCS families in the regulation of P/Q channels. NCS-1 is a protein found from yeast to humans and that regulates a diverse number of cellular functions. Physiological and genetic evidence indicates that NCS-1 regulates P/Q channel activity, including calcium-dependent facilitation, although a direct physical association between the proteins has yet to be demonstrated. In this study, we aimed to determine if there is a direct interaction between NCS-1 and the C-terminal cytoplasmic tail of the Cav2.1 α-subunit. Using distinct but complementary approaches, including in vitro binding of bacterially expressed recombinant proteins, fluorescence spectrophotometry, isothermal titration calorimetry, nuclear magnetic resonance, and expression of fluorescently tagged proteins in mammalian cells, we show direct binding and demonstrate that CaM can compete for it. We speculate about how NCS-1/Cav2.1 association might add to the complexity of calcium channel regulation mediated by other known calcium-sensing proteins and how this might help to fine-tune neurotransmission in the mammalian central nervous system.
Collapse
Affiliation(s)
- Lu-Yun Lian
- NMR Centre for Structural Biology, Institute of Integrative Biology, University of Liverpool , Liverpool L69 3BX, U.K
| | | | | | | | | | | |
Collapse
|
35
|
Brini M, Calì T, Ottolini D, Carafoli E. Neuronal calcium signaling: function and dysfunction. Cell Mol Life Sci 2014; 71:2787-814. [PMID: 24442513 PMCID: PMC11113927 DOI: 10.1007/s00018-013-1550-7] [Citation(s) in RCA: 499] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 12/15/2013] [Accepted: 12/30/2013] [Indexed: 01/07/2023]
Abstract
Calcium (Ca(2+)) is an universal second messenger that regulates the most important activities of all eukaryotic cells. It is of critical importance to neurons as it participates in the transmission of the depolarizing signal and contributes to synaptic activity. Neurons have thus developed extensive and intricate Ca(2+) signaling pathways to couple the Ca(2+) signal to their biochemical machinery. Ca(2+) influx into neurons occurs through plasma membrane receptors and voltage-dependent ion channels. The release of Ca(2+) from the intracellular stores, such as the endoplasmic reticulum, by intracellular channels also contributes to the elevation of cytosolic Ca(2+). Inside the cell, Ca(2+) is controlled by the buffering action of cytosolic Ca(2+)-binding proteins and by its uptake and release by mitochondria. The uptake of Ca(2+) in the mitochondrial matrix stimulates the citric acid cycle, thus enhancing ATP production and the removal of Ca(2+) from the cytosol by the ATP-driven pumps in the endoplasmic reticulum and the plasma membrane. A Na(+)/Ca(2+) exchanger in the plasma membrane also participates in the control of neuronal Ca(2+). The impaired ability of neurons to maintain an adequate energy level may impact Ca(2+) signaling: this occurs during aging and in neurodegenerative disease processes. The focus of this review is on neuronal Ca(2+) signaling and its involvement in synaptic signaling processes, neuronal energy metabolism, and neurotransmission. The contribution of altered Ca(2+) signaling in the most important neurological disorders will then be considered.
Collapse
Affiliation(s)
- Marisa Brini
- Department of Biology, University of Padova, Via U.Bassi, 58/b, 35131 Padua, Italy
| | - Tito Calì
- Department of Biology, University of Padova, Via U.Bassi, 58/b, 35131 Padua, Italy
| | - Denis Ottolini
- Department of Biology, University of Padova, Via U.Bassi, 58/b, 35131 Padua, Italy
| | - Ernesto Carafoli
- Venetian Institute for Molecular Medicine (VIMM), Via G.Orus, 2, 35129 Padua, Italy
| |
Collapse
|
36
|
Romero-Pozuelo J, Dason JS, Mansilla A, Baños-Mateos S, Sardina JL, Chaves-Sanjuán A, Jurado-Gómez J, Santana E, Atwood HL, Hernández-Hernández Á, Sánchez-Barrena MJ, Ferrús A. The guanine-exchange factor Ric8a binds to the Ca²⁺ sensor NCS-1 to regulate synapse number and neurotransmitter release. J Cell Sci 2014; 127:4246-59. [PMID: 25074811 DOI: 10.1242/jcs.152603] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The conserved Ca(2+)-binding protein Frequenin (homolog of the mammalian NCS-1, neural calcium sensor) is involved in pathologies that result from abnormal synapse number and probability of neurotransmitter release per synapse. Both synaptic features are likely to be co-regulated but the intervening mechanisms remain poorly understood. We show here that Drosophila Ric8a (a homolog of mammalian synembryn, which is also known as Ric8a), a receptor-independent activator of G protein complexes, binds to Frq2 but not to the virtually identical homolog Frq1. Based on crystallographic data on Frq2 and site-directed mutagenesis on Frq1, the differential amino acids R94 and T138 account for this specificity. Human NCS-1 and Ric8a reproduce the binding and maintain the structural requirements at these key positions. Drosophila Ric8a and Gαs regulate synapse number and neurotransmitter release, and both are functionally linked to Frq2. Frq2 negatively regulates Ric8a to control synapse number. However, the regulation of neurotransmitter release by Ric8a is independent of Frq2 binding. Thus, the antagonistic regulation of these two synaptic properties shares a common pathway, Frq2-Ric8a-Gαs, which diverges downstream. These mechanisms expose the Frq2-Ric8a interacting surface as a potential pharmacological target for NCS-1-related diseases and provide key data towards the corresponding drug design.
Collapse
Affiliation(s)
- Jesús Romero-Pozuelo
- Department of Molecular, Cellular and Developmental Neurobiology, Institute Cajal, CSIC, Avenida Dr. Arce 37, Madrid 28002, Spain
| | - Jeffrey S Dason
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Alicia Mansilla
- Department of Molecular, Cellular and Developmental Neurobiology, Institute Cajal, CSIC, Avenida Dr. Arce 37, Madrid 28002, Spain
| | - Soledad Baños-Mateos
- Department of Crystallography and Structural Biology, Institute of Physical-Chemistry 'Rocasolano', CSIC, Serrano 119, Madrid 28006, Spain
| | - José L Sardina
- Department of Biochemistry and Molecular Biology, University of Salamanca, Salamanca 37007, Spain
| | - Antonio Chaves-Sanjuán
- Department of Crystallography and Structural Biology, Institute of Physical-Chemistry 'Rocasolano', CSIC, Serrano 119, Madrid 28006, Spain
| | - Jaime Jurado-Gómez
- Department of Molecular, Cellular and Developmental Neurobiology, Institute Cajal, CSIC, Avenida Dr. Arce 37, Madrid 28002, Spain
| | - Elena Santana
- Department of Molecular, Cellular and Developmental Neurobiology, Institute Cajal, CSIC, Avenida Dr. Arce 37, Madrid 28002, Spain
| | - Harold L Atwood
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ángel Hernández-Hernández
- Department of Molecular, Cellular and Developmental Neurobiology, Institute Cajal, CSIC, Avenida Dr. Arce 37, Madrid 28002, Spain Department of Biochemistry and Molecular Biology, University of Salamanca, Salamanca 37007, Spain Institute for Biomedical Research (IBSAL), Salamanca 37007, Spain
| | - María-José Sánchez-Barrena
- Department of Crystallography and Structural Biology, Institute of Physical-Chemistry 'Rocasolano', CSIC, Serrano 119, Madrid 28006, Spain
| | - Alberto Ferrús
- Department of Molecular, Cellular and Developmental Neurobiology, Institute Cajal, CSIC, Avenida Dr. Arce 37, Madrid 28002, Spain
| |
Collapse
|
37
|
Baños-Mateos S, Chaves-Sanjuán A, Mansilla A, Ferrús A, Sánchez-Barrena MJ. Frq2 from Drosophila melanogaster: cloning, expression, purification, crystallization and preliminary X-ray analysis. Acta Crystallogr F Struct Biol Commun 2014; 70:530-4. [PMID: 24699756 PMCID: PMC3976080 DOI: 10.1107/s2053230x14005408] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Accepted: 03/05/2014] [Indexed: 12/20/2022] Open
Abstract
Drosophila melanogaster contains two calcium-binding proteins, Frq1 and Frq2, in the nervous system that control the number of synapses and the probability of release. To understand the differential function of the two proteins, whose sequence is only 5% dissimilar, the crystal structures of Frq1 and Frq2 are needed. Here, the cloning, expression, purification, crystallization and preliminary crystallographic analysis of Frq2 are presented. The full-length protein was purified using a two-step chromatographic procedure. Two different diffracting crystal forms were obtained using a progressive streak-seeding method and detergents.
Collapse
Affiliation(s)
- Soledad Baños-Mateos
- Department of Crystallography and Structural Biology, Institute of Physical Chemistry ‘Rocasolano’, CSIC, Serrano 119, 28006 Madrid, Spain
| | - Antonio Chaves-Sanjuán
- Department of Crystallography and Structural Biology, Institute of Physical Chemistry ‘Rocasolano’, CSIC, Serrano 119, 28006 Madrid, Spain
| | - Alicia Mansilla
- Department of Molecular, Cellular and Developmental Neurobiology, Institute Cajal, CSIC, Avenida Dr Arce 37, 28002 Madrid, Spain
| | - Alberto Ferrús
- Department of Molecular, Cellular and Developmental Neurobiology, Institute Cajal, CSIC, Avenida Dr Arce 37, 28002 Madrid, Spain
| | - María José Sánchez-Barrena
- Department of Crystallography and Structural Biology, Institute of Physical Chemistry ‘Rocasolano’, CSIC, Serrano 119, 28006 Madrid, Spain
| |
Collapse
|
38
|
Dason JS, Smith AJ, Marin L, Charlton MP. Cholesterol and F-actin are required for clustering of recycling synaptic vesicle proteins in the presynaptic plasma membrane. J Physiol 2013; 592:621-33. [PMID: 24297851 DOI: 10.1113/jphysiol.2013.265447] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Synaptic vesicles (SVs) and their proteins must be recycled for sustained synaptic transmission. We tested the hypothesis that SV cholesterol is required for proper sorting of SV proteins during recycling in live presynaptic terminals. We used the reversible block of endocytosis in the Drosophila temperature-sensitive dynamin mutant shibire-ts1 to trap exocytosed SV proteins, and then examined the effect of experimental treatments on the distribution of these proteins within the presynaptic plasma membrane by confocal microscopy. SV proteins synaptotagmin, vglut and csp were clustered following SV trapping in control experiments but dispersed in samples treated with the cholesterol chelator methyl-β-cyclodextrin to extract SV cholesterol. There was accumulation of phosphatidylinositol (4,5)-bisphosphate (PIP2) in presynaptic terminals following SV trapping and this was reduced following SV cholesterol extraction. Reduced PIP2 accumulation was associated with disrupted accumulation of actin in presynaptic terminals. Similar to vesicular cholesterol extraction, disruption of actin by latrunculin A after SV proteins had been trapped on the plasma membrane resulted in the dispersal of SV proteins and prevented recovery of synaptic transmission due to impaired endocytosis following relief of the endocytic block. Our results demonstrate that vesicular cholesterol is required for aggregation of exocytosed SV proteins in the presynaptic plasma membrane and are consistent with a mechanism involving regulation of PIP2 accumulation and local actin polymerization by cholesterol. Thus, alteration of membrane or SV lipids may affect the ability of synapses to undergo sustained synaptic transmission by compromising the recycling of SV proteins.
Collapse
Affiliation(s)
- Jeffrey S Dason
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada, M5S 1A8.
| | | | | | | |
Collapse
|
39
|
Martin VM, Johnson JR, Haynes LP, Barclay JW, Burgoyne RD. Identification of key structural elements for neuronal calcium sensor-1 function in the regulation of the temperature-dependency of locomotion in C. elegans. Mol Brain 2013; 6:39. [PMID: 23981466 PMCID: PMC3765893 DOI: 10.1186/1756-6606-6-39] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 08/24/2013] [Indexed: 11/10/2022] Open
Abstract
Background Intracellular Ca2+ regulates many aspects of neuronal function through Ca2+ binding to EF hand-containing Ca2+ sensors that in turn bind target proteins to regulate their function. Amongst the sensors are the neuronal calcium sensor (NCS) family of proteins that are involved in multiple neuronal signalling pathways. Each NCS protein has specific and overlapping targets and physiological functions and specificity is likely to be determined by structural features within the proteins. Common to the NCS proteins is the exposure of a hydrophobic groove, allowing target binding in the Ca2+-loaded form. Structural analysis of NCS protein complexes with target peptides has indicated common and distinct aspects of target protein interaction. Two key differences between NCS proteins are the size of the hydrophobic groove that is exposed for interaction and the role of their non-conserved C-terminal tails. Results We characterised the role of NCS-1 in a temperature-dependent locomotion assay in C. elegans and identified a distinct phenotype in the ncs-1 null in which the worms do not show reduced locomotion at actually elevated temperature. Using rescue of this phenotype we showed that NCS-1 functions in AIY neurons. Structure/function analysis introducing single or double mutations within the hydrophobic groove based on information from characterised target complexes established that both N- and C-terminal pockets of the groove are functionally important and that deletion of the C-terminal tail of NCS-1 did not impair its ability to rescue. Conclusions The current work has allowed physiological assessment of suggestions from structural studies on the key structural features that underlie the interaction of NCS-1 with its target proteins. The results are consistent with the notion that full length of the hydrophobic groove is required for the regulatory interactions underlying NCS-1 function whereas the C-terminal tail of NCS-1 is not essential. This has allowed discrimination between two potential modes of interaction of NCS-1 with its targets.
Collapse
Affiliation(s)
- Victoria M Martin
- Department of Cellular and Molecular Physiology, The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK.
| | | | | | | | | |
Collapse
|
40
|
Palma-Guerrero J, Hall CR, Kowbel D, Welch J, Taylor JW, Brem RB, Glass NL. Genome wide association identifies novel loci involved in fungal communication. PLoS Genet 2013; 9:e1003669. [PMID: 23935534 PMCID: PMC3731230 DOI: 10.1371/journal.pgen.1003669] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 06/10/2013] [Indexed: 01/25/2023] Open
Abstract
Understanding how genomes encode complex cellular and organismal behaviors has become the outstanding challenge of modern genetics. Unlike classical screening methods, analysis of genetic variation that occurs naturally in wild populations can enable rapid, genome-scale mapping of genotype to phenotype with a medium-throughput experimental design. Here we describe the results of the first genome-wide association study (GWAS) used to identify novel loci underlying trait variation in a microbial eukaryote, harnessing wild isolates of the filamentous fungus Neurospora crassa. We genotyped each of a population of wild Louisiana strains at 1 million genetic loci genome-wide, and we used these genotypes to map genetic determinants of microbial communication. In N. crassa, germinated asexual spores (germlings) sense the presence of other germlings, grow toward them in a coordinated fashion, and fuse. We evaluated germlings of each strain for their ability to chemically sense, chemotropically seek, and undergo cell fusion, and we subjected these trait measurements to GWAS. This analysis identified one gene, NCU04379 (cse-1, encoding a homolog of a neuronal calcium sensor), at which inheritance was strongly associated with the efficiency of germling communication. Deletion of cse-1 significantly impaired germling communication and fusion, and two genes encoding predicted interaction partners of CSE1 were also required for the communication trait. Additionally, mining our association results for signaling and secretion genes with a potential role in germling communication, we validated six more previously unknown molecular players, including a secreted protease and two other genes whose deletion conferred a novel phenotype of increased communication and multi-germling fusion. Our results establish protein secretion as a linchpin of germling communication in N. crassa and shed light on the regulation of communication molecules in this fungus. Our study demonstrates the power of population-genetic analyses for the rapid identification of genes contributing to complex traits in microbial species. Many phenotypes of interest are controlled by multiple loci, and in biological systems identifying determinants of such complex traits is challenging. Here, we genotyped 112 wild isolates of Neurospora crassa and used this resource to identify genes that mediate a fundamental but poorly-understood attribute of this filamentous fungus: the ability of germinating spores to sense each other at a distance, extend projections toward one another, and fuse. Inheritance at a secretion gene, cse-1, was associated strongly with germling communication across wild strains; this association was validated in experiments showing reduced communication in a cse-1 deletion strain. By testing interacting partners of CSE1, and by assessing additional secretion and signaling factors whose inheritance associated more modestly with germling communication in wild strains, we identified eight other novel determinants of this phenotype. Our population of genotyped wild isolates provides a flexible and powerful community resource for the rapid identification of any varying, complex phenotype in N. crassa. The success of our approach, which used a phenotyping scheme far more tractable than would be required in a screen of the entire N. crassa gene deletion collection, serves as a proof of concept for association studies of wild populations for any organism.
Collapse
Affiliation(s)
- Javier Palma-Guerrero
- Department of Plant and Microbial Biology, University of California, Berkeley, California, United States of America
| | - Charles R. Hall
- Department of Plant and Microbial Biology, University of California, Berkeley, California, United States of America
| | - David Kowbel
- Department of Plant and Microbial Biology, University of California, Berkeley, California, United States of America
| | - Juliet Welch
- Department of Plant and Microbial Biology, University of California, Berkeley, California, United States of America
| | - John W. Taylor
- Department of Plant and Microbial Biology, University of California, Berkeley, California, United States of America
| | - Rachel B. Brem
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
- * E-mail: (RBB); (NLG)
| | - N. Louise Glass
- Department of Plant and Microbial Biology, University of California, Berkeley, California, United States of America
- * E-mail: (RBB); (NLG)
| |
Collapse
|
41
|
Dason JS, Romero-Pozuelo J, Atwood HL, Ferrús A. Multiple roles for frequenin/NCS-1 in synaptic function and development. Mol Neurobiol 2012; 45:388-402. [PMID: 22396213 DOI: 10.1007/s12035-012-8250-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 02/20/2012] [Indexed: 11/26/2022]
Abstract
The calcium-binding protein frequenin (Frq), discovered in the fruit fly Drosophila, and its mammalian homologue neuronal calcium sensor 1 (NCS-1) have been reported to affect several aspects of synaptic transmission, including basal levels of neurotransmission and short- and long-term synaptic plasticities. However, discrepant reports leave doubts about the functional roles of these conserved proteins. In this review, we attempt to resolve some of these seemingly contradictory reports. We discuss how stimulation protocols, sources of calcium (voltage-gated channels versus internal stores), and expression patterns (presynaptic versus postsynaptic) of Frq may result in the activation of various protein targets, leading to different synaptic effects. In addition, the potential interactions of Frq's C-terminal and N-terminal domains with other proteins are discussed. Frq also has a role in regulating neurite outgrowth, axonal regeneration, and synaptic development. We examine whether the effects of Frq on neurotransmitter release and neurite outgrowth are distinct or interrelated through homeostatic mechanisms. Learning and memory are affected by manipulations of Frq probably through changes in synaptic transmission and neurite outgrowth, raising the possibility that Frq may be implicated in human pathological conditions, including schizophrenia, bipolar disorder, and X-linked mental retardation.
Collapse
Affiliation(s)
- Jeffrey S Dason
- Department of Physiology, University of Toronto, Toronto, ON, Canada, M5S 1A8.
| | | | | | | |
Collapse
|
42
|
Burgoyne RD, Haynes LP. Understanding the physiological roles of the neuronal calcium sensor proteins. Mol Brain 2012; 5:2. [PMID: 22269068 PMCID: PMC3271974 DOI: 10.1186/1756-6606-5-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 01/23/2012] [Indexed: 01/22/2023] Open
Abstract
Calcium signalling plays a crucial role in the control of neuronal function and plasticity. Changes in neuronal Ca2+ concentration are detected by Ca2+-binding proteins that can interact with and regulate target proteins to modify their function. Members of the neuronal calcium sensor (NCS) protein family have multiple non-redundant roles in the nervous system. Here we review recent advances in the understanding of the physiological roles of the NCS proteins and the molecular basis for their specificity.
Collapse
Affiliation(s)
- Robert D Burgoyne
- Department of Cellular and Molecular Physiology, The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | | |
Collapse
|
43
|
Lian LY, Pandalaneni SR, Patel P, McCue HV, Haynes LP, Burgoyne RD. Characterisation of the interaction of the C-terminus of the dopamine D2 receptor with neuronal calcium sensor-1. PLoS One 2011; 6:e27779. [PMID: 22114693 PMCID: PMC3218054 DOI: 10.1371/journal.pone.0027779] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 10/25/2011] [Indexed: 11/19/2022] Open
Abstract
NCS-1 is a member of the neuronal calcium sensor (NCS) family of EF-hand Ca(2+) binding proteins which has been implicated in several physiological functions including regulation of neurotransmitter release, membrane traffic, voltage gated Ca(2+) channels, neuronal development, synaptic plasticity, and learning. NCS-1 binds to the dopamine D2 receptor, potentially affecting its internalisation and controlling dopamine D2 receptor surface expression. The D2 receptor binds NCS-1 via a short 16-residue cytoplasmic C-terminal tail. We have used NMR and fluorescence spectroscopy to characterise the interactions between the NCS-1/Ca(2+) and D2 peptide. The data show that NCS-1 binds D2 peptide with a K(d) of ∼14.3 µM and stoichiometry of peptide binding to NCS-1 of 2:1. NMR chemical shift mapping confirms that D2 peptide binds to the large, solvent-exposed hydrophobic groove, on one face of the NCS-1 molecule, with residues affected by the presence of the peptide spanning both the N and C-terminal portions of the protein. The NMR and mutagenesis data further show that movement of the C-terminal helix 11 of NCS-1 to fully expose the hydrophobic groove is important for D2 peptide binding. Molecular docking using restraints derived from the NMR chemical shift data, together with the experimentally-derived stoichiometry, produced a model of the complex between NCS-1 and the dopamine receptor, in which two molecules of the receptor are able to simultaneously bind to the NCS-1 monomer.
Collapse
Affiliation(s)
- Lu-Yun Lian
- NMR Centre for Structural Biology, Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- * E-mail: (LYL); (RDB)
| | - Sravan R. Pandalaneni
- NMR Centre for Structural Biology, Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Pryank Patel
- NMR Centre for Structural Biology, Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Hannah V. McCue
- The Physiological Laboratory, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Lee P. Haynes
- The Physiological Laboratory, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Robert D. Burgoyne
- The Physiological Laboratory, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
- * E-mail: (LYL); (RDB)
| |
Collapse
|
44
|
Mikhaylova M, Hradsky J, Kreutz MR. Between promiscuity and specificity: novel roles of EF-hand calcium sensors in neuronal Ca2+ signalling. J Neurochem 2011; 118:695-713. [PMID: 21722133 DOI: 10.1111/j.1471-4159.2011.07372.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In recent years, substantial progress has been made towards an understanding of the physiological function of EF-hand calcium sensor proteins of the Calmodulin (CaM) superfamily in neurons. This deeper appreciation is based on the identification of novel target interactions, structural studies and the discovery of novel signalling mechanisms in protein trafficking and synaptic plasticity, in which CaM-like sensor proteins appear to play a role. However, not all interactions are of plausible physiological relevance and in many cases it is not yet clear how the CaM signaling network relates to the proposed function of other EF-hand sensors. In this review, we will summarize these findings and address some of the open questions on the functional role of EF-hand calcium binding proteins in neurons.
Collapse
Affiliation(s)
- Marina Mikhaylova
- PG Neuroplasticity, Leibniz-Institute for Neurobiology, Magdeburg, Germany
| | | | | |
Collapse
|
45
|
Nejatbakhsh N, Feng ZP. Calcium binding protein-mediated regulation of voltage-gated calcium channels linked to human diseases. Acta Pharmacol Sin 2011; 32:741-8. [PMID: 21642945 DOI: 10.1038/aps.2011.64] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Calcium ion entry through voltage-gated calcium channels is essential for cellular signalling in a wide variety of cells and multiple physiological processes. Perturbations of voltage-gated calcium channel function can lead to pathophysiological consequences. Calcium binding proteins serve as calcium sensors and regulate the calcium channel properties via feedback mechanisms. This review highlights the current evidences of calcium binding protein-mediated channel regulation in human diseases.
Collapse
|
46
|
Guan Z, Buhl LK, Quinn WG, Littleton JT. Altered gene regulation and synaptic morphology in Drosophila learning and memory mutants. Learn Mem 2011; 18:191-206. [PMID: 21422168 DOI: 10.1101/lm.2027111] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Genetic studies in Drosophila have revealed two separable long-term memory pathways defined as anesthesia-resistant memory (ARM) and long-lasting long-term memory (LLTM). ARM is disrupted in radish (rsh) mutants, whereas LLTM requires CREB-dependent protein synthesis. Although the downstream effectors of ARM and LLTM are distinct, pathways leading to these forms of memory may share the cAMP cascade critical for associative learning. Dunce, which encodes a cAMP-specific phosphodiesterase, and rutabaga, which encodes an adenylyl cyclase, both disrupt short-term memory. Amnesiac encodes a pituitary adenylyl cyclase-activating peptide homolog and is required for middle-term memory. Here, we demonstrate that the Radish protein localizes to the cytoplasm and nucleus and is a PKA phosphorylation target in vitro. To characterize how these plasticity pathways may manifest at the synaptic level, we assayed synaptic connectivity and performed an expression analysis to detect altered transcriptional networks in rutabaga, dunce, amnesiac, and radish mutants. All four mutants disrupt specific aspects of synaptic connectivity at larval neuromuscular junctions (NMJs). Genome-wide DNA microarray analysis revealed ∼375 transcripts that are altered in these mutants, suggesting defects in multiple neuronal signaling pathways. In particular, the transcriptional target Lapsyn, which encodes a leucine-rich repeat cell adhesion protein, localizes to synapses and regulates synaptic growth. This analysis provides insights into the Radish-dependent ARM pathway and novel transcriptional targets that may contribute to memory processing in Drosophila.
Collapse
Affiliation(s)
- Zhuo Guan
- Department of Biology, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | | | |
Collapse
|
47
|
Abstract
Synaptic vesicles have a high sterol content, but the importance of vesicular sterols during vesicle recycling is unclear. We used the Drosophila temperature-sensitive dynamin mutant, shibire-ts1, to block endocytosis of recycling synaptic vesicles and to trap them reversibly at the plasma membrane where they were accessible to sterol extraction. Depletion of sterols from trapped vesicles prevented recovery of synaptic transmission after removal of the endocytic block. Measurement of vesicle recycling with synaptopHluorin, FM1-43, and FM4-64 demonstrated impaired membrane retrieval after vesicular sterol depletion. When plasma membrane sterols were extracted before vesicle trapping, no vesicle recycling defects were observed. Ultrastructural analysis indicated accumulation of endosomes and a defect in the formation of synaptic vesicles in synaptic terminals subjected to vesicular sterol depletion. Our results demonstrate the importance of a high vesicular sterol concentration for endocytosis and suggest that vesicular and membrane sterol pools do not readily intermingle during vesicle recycling.
Collapse
|
48
|
Freeman A, Franciscovich A, Bowers M, Sandstrom DJ, Sanyal S. NFAT regulates pre-synaptic development and activity-dependent plasticity in Drosophila. Mol Cell Neurosci 2010; 46:535-47. [PMID: 21185939 DOI: 10.1016/j.mcn.2010.12.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Accepted: 12/14/2010] [Indexed: 11/18/2022] Open
Abstract
The calcium-regulated transcription factor NFAT is emerging as a key regulator of neuronal development and plasticity but precise cellular consequences of NFAT function remain poorly understood. Here, we report that the single Drosophila NFAT homolog is widely expressed in the nervous system including motor neurons and unexpectedly controls neural excitability. Likely due to this effect on excitability, NFAT regulates overall larval locomotion and both chronic and acute forms of activity-dependent plasticity at the larval glutamatergic neuro-muscular synapse. Specifically, NFAT-dependent synaptic phenotypes include changes in the number of pre-synaptic boutons, stable modifications in synaptic microtubule architecture and pre-synaptic transmitter release, while no evidence is found for synaptic retraction or alterations in the level of the synaptic cell adhesion molecule FasII. We propose that NFAT regulates pre-synaptic development and constrains long-term plasticity by dampening neuronal excitability.
Collapse
Affiliation(s)
- Amanda Freeman
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA
| | | | | | | | | |
Collapse
|
49
|
Peptide-induced modulation of synaptic transmission and escape response in Drosophila requires two G-protein-coupled receptors. J Neurosci 2010; 30:14724-34. [PMID: 21048131 DOI: 10.1523/jneurosci.3612-10.2010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neuropeptides are found in both mammals and invertebrates and can modulate neural function through activation of G-protein-coupled receptors (GPCRS). The precise mechanisms by which many of these GPCRs modulate specific signaling cascades to regulate neural function are not well defined. We used Drosophila melanogaster as a model to examine both the cellular and behavioral effects of DPKQDFMRFamide, the most abundant peptide encoded by the dFMRF gene. We show that DPKQDFMRFamide enhanced synaptic transmission through activation of two G-protein-coupled receptors, Fmrf Receptor (FR) and Dromyosupressin Receptor-2 (DmsR-2). The peptide increased both the presynaptic Ca(2+) response and the quantal content of released transmitter. Peptide-induced modulation of synaptic function could be abrogated by depleting intracellular Ca(2+) stores or by interfering with Ca(2+) release from the endoplasmic reticulum through disruption of either the ryanodine receptor or the inositol 1,4,5-trisphosphate receptor. The peptide also altered behavior. Exogenous DPKQDFMRFamide enhanced fictive locomotion; this required both the FR and DmsR-2. Likewise, both receptors were required for an escape response to intense light exposure. Thus, coincident detection of a peptide by two GPCRs modulates synaptic function through effects of Ca(2+)-induced Ca(2+) release, and we hypothesize that these mechanisms are involved in behavioral responses to environmental stress.
Collapse
|
50
|
Weiss JL, Hui H, Burgoyne RD. Neuronal calcium sensor-1 regulation of calcium channels, secretion, and neuronal outgrowth. Cell Mol Neurobiol 2010; 30:1283-92. [PMID: 21104311 PMCID: PMC11498851 DOI: 10.1007/s10571-010-9588-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 09/02/2010] [Indexed: 12/01/2022]
Abstract
Calcium (Ca(2+)) is an important intracellular messenger underlying cell physiology. Ca(2+) channels are the main entry route for Ca(2+) into excitable cells, and regulate processes such as neurotransmitter release and neuronal outgrowth. Neuronal Calcium Sensor-1 (NCS-1) is a member of the Calmodulin superfamily of EF-hand Ca(2+) sensing proteins residing in the subfamily of NCS proteins. NCS-1 was originally discovered in Drosophila as an overexpression mutant (Frequenin), having an increased frequency of Ca(2+)-evoked neurotransmission. NCS-1 is N-terminally myristoylated, can bind intracellular membranes, and has a Ca(2+) affinity of 0.3 μM. Over 10 years ago it was discovered that NCS-1 overexpression enhances Ca(2+)-evoked secretion in bovine adrenal chromaffin cells. The mechanism was unclear, but there was no apparent direct effect on the exocytotic machinery. It was revealed, again in chromaffin cells, that NCS-1 regulates voltage-gated Ca(2+) channels (Cavs) in G-Protein Coupled Receptor (GPCR) signaling pathways. This work in chromaffin cells highlighted NCS-1 as an important modulator of neurotransmission. NCS-1 has since been shown to regulate and/or directly interact with many proteins including Cavs (P/Q, N, and L), TRPC1/5 channels, GPCRs, IP3R, and PI4 kinase type IIIβ. NCS-1 also affects neuronal outgrowth having roles in learning and memory affecting both short- and long-term synaptic plasticity. It is not known if NCS-1 affects neurotransmission and synaptic plasticity via its effect on PIP2 levels, and/or via a direct interaction with Ca(2+) channels or their signaling complexes. This review gives a historical account of NCS-1 function, examining contributions from chromaffin cells, PC12 cells and other models, to describe how NCS-1's regulation of Ca(2+) channels allows it to exert its physiological effects.
Collapse
Affiliation(s)
- Jamie L Weiss
- Department of Biology, William Paterson University, 300 Pompton Road, Wayne, NJ 07470, USA.
| | | | | |
Collapse
|