1
|
Thierer JH, Foresti O, Yadav PK, Wilson MH, Moll TOC, Shen MC, Busch-Nentwich EM, Morash M, Mohlke KL, Rawls JF, Malhotra V, Hussain MM, Farber SA. Pla2g12b drives expansion of triglyceride-rich lipoproteins. Nat Commun 2024; 15:2095. [PMID: 38453914 PMCID: PMC10920679 DOI: 10.1038/s41467-024-46102-4] [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: 03/09/2023] [Accepted: 02/14/2024] [Indexed: 03/09/2024] Open
Abstract
Vertebrates transport hydrophobic triglycerides through the circulatory system by packaging them within amphipathic particles called Triglyceride-Rich Lipoproteins. Yet, it remains largely unknown how triglycerides are loaded onto these particles. Mutations in Phospholipase A2 group 12B (PLA2G12B) are known to disrupt lipoprotein homeostasis, but its mechanistic role in this process remains unclear. Here we report that PLA2G12B channels lipids within the lumen of the endoplasmic reticulum into nascent lipoproteins. This activity promotes efficient lipid secretion while preventing excess accumulation of intracellular lipids. We characterize the functional domains, subcellular localization, and interacting partners of PLA2G12B, demonstrating that PLA2G12B is calcium-dependent and tightly associated with the membrane of the endoplasmic reticulum. We also detect profound resistance to atherosclerosis in PLA2G12B mutant mice, suggesting an evolutionary tradeoff between triglyceride transport and cardiovascular disease risk. Here we identify PLA2G12B as a key driver of triglyceride incorporation into vertebrate lipoproteins.
Collapse
Affiliation(s)
- James H Thierer
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, 21218, USA
- Johns Hopkins University in Baltimore, Maryland Department of biology, Baltimore, MD, 21218, USA
| | - Ombretta Foresti
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, 08003, ES, Spain
| | - Pradeep Kumar Yadav
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY, 11501, USA
- Department of Botany, Faculty of Science, University of Allahabad, Prayagraj, India
| | - Meredith H Wilson
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, 21218, USA
- Johns Hopkins University in Baltimore, Maryland Department of biology, Baltimore, MD, 21218, USA
| | - Tabea O C Moll
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, 21218, USA
- Johns Hopkins University in Baltimore, Maryland Department of biology, Baltimore, MD, 21218, USA
| | - Meng-Chieh Shen
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, 21218, USA
| | | | - Margaret Morash
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, 27708, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, 27708, USA
| | - Vivek Malhotra
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, 08003, ES, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - M Mahmood Hussain
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY, 11501, USA
| | - Steven A Farber
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, 21218, USA.
- Johns Hopkins University in Baltimore, Maryland Department of biology, Baltimore, MD, 21218, USA.
| |
Collapse
|
2
|
Sharma B, Koren DT, Ghosh S. Nitric oxide modulates NMDA receptor through a negative feedback mechanism and regulates the dynamical behavior of neuronal postsynaptic components. Biophys Chem 2023; 303:107114. [PMID: 37832215 DOI: 10.1016/j.bpc.2023.107114] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/22/2023] [Accepted: 09/24/2023] [Indexed: 10/15/2023]
Abstract
Nitric oxide (NO) is known to be an important regulator of neurological processes in the central nervous system which acts directly on the presynaptic neuron and enhances the release of neurotransmitters like glutamate into the synaptic cleft. Calcium influx activates a cascade of biochemical reactions to influence the production of nitric oxide in the postsynaptic neuron. This has been modeled in the present work as a system of ordinary differential equations, to explore the dynamics of the interacting components and predict the dynamical behavior of the postsynaptic neuron. It has been hypothesized that nitric oxide modulates the NMDA receptor via a feedback mechanism and regulates the dynamic behavior of postsynaptic components. Results obtained by numerical analyses indicate that the biochemical system is stimulus-dependent and shows oscillations of calcium and other components within a limited range of concentration. Some of the parameters such as stimulus strength, extracellular calcium concentration, and rate of nitric oxide feedback are crucial for the dynamics of the components in the postsynaptic neuron.
Collapse
Affiliation(s)
- Bhanu Sharma
- Department of Biophysics, University of Delhi South Campus, New Delhi 110021, India
| | | | - Subhendu Ghosh
- Department of Biophysics, University of Delhi South Campus, New Delhi 110021, India.
| |
Collapse
|
3
|
Meng L. Chromatin-modifying enzymes as modulators of nuclear size during lineage differentiation. Cell Death Discov 2023; 9:384. [PMID: 37863956 PMCID: PMC10589317 DOI: 10.1038/s41420-023-01639-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/17/2023] [Accepted: 08/31/2023] [Indexed: 10/22/2023] Open
Abstract
The mechanism of nuclear size determination and alteration during normal lineage development and cancer pathologies which is not fully understood. As recently reported, chromatin modification can change nuclear morphology. Therefore, we screened a range of pharmacological chemical compounds that impact the activity of chromatin-modifying enzymes, in order to get a clue of the specific types of chromatin-modifying enzymes that remarkably effect nuclear size and shape. We found that interrupted activity of chromatin-modifying enzymes is associated with nuclear shape abnormalities. Furthermore, the activity of chromatin-modifying enzymes perturbs cell fate determination in cellular maintenance and lineage commitment. Our results indicated that chromatin-modifying enzyme regulates cell fate decision during lineage differentiation and is associate with nuclear size alteration.
Collapse
Affiliation(s)
- Lingjun Meng
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
4
|
Bittencourt LO, Dionizio A, Ferreira MKM, Aragão WAB, de Carvalho Cartágenes S, Puty B, do Socorro Ferraz Maia C, Zohoori FV, Buzalaf MAR, Lima RR. Prolonged exposure to high fluoride levels during adolescence to adulthood elicits molecular, morphological, and functional impairments in the hippocampus. Sci Rep 2023; 13:11083. [PMID: 37422569 PMCID: PMC10329641 DOI: 10.1038/s41598-023-38096-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 07/03/2023] [Indexed: 07/10/2023] Open
Abstract
Fluoride is added to water due to its anticariogenic activity. However, due to its natural presence in soils and reservoirs at high levels, it could be a potential environmental toxicant. This study investigated whether prolonged exposure to fluoride from adolescence to adulthood-at concentrations commonly found in artificially fluoridated water and in fluorosis endemic areas-is associated with memory and learning impairments in mice, and assessed the molecular and morphological aspects involved. For this endeavor, 21-days-old mice received 10 or 50 mg/L of fluoride in drinking water for 60 days and the results indicated that the increased plasma fluoride bioavailability was associated with the triggering of short- and long-term memory impairments after high F concentration levels. These changes were associated with modulation of the hippocampal proteomic profile, especially of proteins related to synaptic communication, and a neurodegenerative pattern in the CA3 and DG. From a translational perspective, our data provide evidence of potential molecular targets of fluoride neurotoxicity in the hippocampus at levels much higher than that in artificially fluoridated water and reinforce the safety of exposure to low concentrations of fluoride. In conclusion, prolonged exposure to the optimum fluoride level of artificially fluoridated water was not associated with cognitive impairments, while a higher concentration associated with fluorosis triggered memory and learning deficits, associated with a neuronal density reduction in the hippocampus.
Collapse
Affiliation(s)
- Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Augusto Corrêa street n. 01, Guamá, Belém, Pará, 66075-110, Brazil
| | - Aline Dionizio
- Department of Biological Sciences, Bauru Dental School, University of São Paulo, Bauru, São Paulo, Brazil
| | - Maria Karolina Martins Ferreira
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Augusto Corrêa street n. 01, Guamá, Belém, Pará, 66075-110, Brazil
| | - Walessa Alana Bragança Aragão
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Augusto Corrêa street n. 01, Guamá, Belém, Pará, 66075-110, Brazil
| | - Sabrina de Carvalho Cartágenes
- Laboratory of Inflammation and Behavior Pharmacology, Faculty of Pharmacy, Institute of Health Sciences, Federal University of Pará, Belém, Pará, Brazil
| | - Bruna Puty
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Augusto Corrêa street n. 01, Guamá, Belém, Pará, 66075-110, Brazil
| | - Cristiane do Socorro Ferraz Maia
- Laboratory of Inflammation and Behavior Pharmacology, Faculty of Pharmacy, Institute of Health Sciences, Federal University of Pará, Belém, Pará, Brazil
| | - Fatemeh Vida Zohoori
- School of Health and Life Sciences, Teesside University, Middlesbrough, United Kingdom
| | | | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Augusto Corrêa street n. 01, Guamá, Belém, Pará, 66075-110, Brazil.
| |
Collapse
|
5
|
Wojnacki J, Lujan AL, Brouwers N, Aranda-Vallejo C, Bigliani G, Rodriguez MP, Foresti O, Malhotra V. Tetraspanin-8 sequesters syntaxin-2 to control biphasic release propensity of mucin granules. Nat Commun 2023; 14:3710. [PMID: 37349283 PMCID: PMC10287693 DOI: 10.1038/s41467-023-39277-9] [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: 09/24/2022] [Accepted: 06/06/2023] [Indexed: 06/24/2023] Open
Abstract
Agonist-mediated stimulated pathway of mucin and insulin release are biphasic in which rapid fusion of pre-docked granules is followed by slow docking and fusion of granules from the reserve pool. Here, based on a cell-culture system, we show that plasma membrane-located tetraspanin-8 sequesters syntaxin-2 to control mucin release. Tetraspanin-8 affects fusion of granules during the second phase of stimulated mucin release. The tetraspanin-8/syntaxin-2 complex does not contain VAMP-8, which functions with syntaxin-2 to mediate granule fusion. We suggest that by sequestering syntaxin-2, tetraspanin-8 prevents docking of granules from the reserve pool. In the absence of tetraspanin-8, more syntaxin-2 is available for docking and fusion of granules and thus doubles the quantities of mucins secreted. This principle also applies to insulin release and we suggest a cell type specific Tetraspanin/Syntaxin combination is a general mechanism regulating the fusion of dense core granules.
Collapse
Affiliation(s)
- José Wojnacki
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Agustin Leonardo Lujan
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Nathalie Brouwers
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Carla Aranda-Vallejo
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Gonzalo Bigliani
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Maria Pena Rodriguez
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Ombretta Foresti
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Vivek Malhotra
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain.
- Universitat Pompeu Fabra (UPF), Barcelona, Spain.
- ICREA, Barcelona, Spain.
| |
Collapse
|
6
|
Tocotrienol-Rich Fraction and Levodopa Regulate Proteins Involved in Parkinson’s Disease-Associated Pathways in Differentiated Neuroblastoma Cells: Insights from Quantitative Proteomic Analysis. Nutrients 2022; 14:nu14214632. [DOI: 10.3390/nu14214632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
Abstract
Tocotrienol-rich fraction (TRF), a palm oil-derived vitamin E fraction, is reported to possess potent neuroprotective effects. However, the modulation of proteomes in differentiated human neuroblastoma SH-SY5Y cells (diff-neural cells) by TRF has not yet been reported. This study aims to investigate the proteomic changes implicated by TRF in human neural cells using a label-free liquid-chromatography-double mass spectrometry (LC-MS/MS) approach. Levodopa, a drug used in the treatment of Parkinson’s disease (PD), was used as a drug control. The human SH-SY5Y neuroblastoma cells were differentiated for six days and treated with TRF or levodopa for 24 h prior to quantitative proteomic analysis. A total of 81 and 57 proteins were differentially expressed in diff-neural cells following treatment with TRF or levodopa, respectively. Among these proteins, 32 similar proteins were detected in both TRF and levodopa-treated neural cells, with 30 of these proteins showing similar expression pattern. The pathway enrichment analysis revealed that most of the proteins regulated by TRF and levodopa are key players in the ubiquitin-proteasome, calcium signalling, protein processing in the endoplasmic reticulum, mitochondrial pathway and axonal transport system. In conclusion, TRF is an essential functional food that affects differential protein expression in human neuronal cells at the cellular and molecular levels.
Collapse
|
7
|
Knodel MM, Dutta Roy R, Wittum G. Influence of T-Bar on Calcium Concentration Impacting Release Probability. Front Comput Neurosci 2022; 16:855746. [PMID: 35586479 PMCID: PMC9108211 DOI: 10.3389/fncom.2022.855746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/09/2022] [Indexed: 11/25/2022] Open
Abstract
The relation of form and function, namely the impact of the synaptic anatomy on calcium dynamics in the presynaptic bouton, is a major challenge of present (computational) neuroscience at a cellular level. The Drosophila larval neuromuscular junction (NMJ) is a simple model system, which allows studying basic effects in a rather simple way. This synapse harbors several special structures. In particular, in opposite to standard vertebrate synapses, the presynaptic boutons are rather large, and they have several presynaptic zones. In these zones, different types of anatomical structures are present. Some of the zones bear a so-called T-bar, a particular anatomical structure. The geometric form of the T-bar resembles the shape of the letter “T” or a table with one leg. When an action potential arises, calcium influx is triggered. The probability of vesicle docking and neurotransmitter release is superlinearly proportional to the concentration of calcium close to the vesicular release site. It is tempting to assume that the T-bar causes some sort of calcium accumulation and hence triggers a higher release probability and thus enhances neurotransmitter exocytosis. In order to study this influence in a quantitative manner, we constructed a typical T-bar geometry and compared the calcium concentration close to the active zones (AZs). We compared the case of synapses with and without T-bars. Indeed, we found a substantial influence of the T-bar structure on the presynaptic calcium concentrations close to the AZs, indicating that this anatomical structure increases vesicle release probability. Therefore, our study reveals how the T-bar zone implies a strong relation between form and function. Our study answers the question of experimental studies (namely “Wichmann and Sigrist, Journal of neurogenetics 2010”) concerning the sense of the anatomical structure of the T-bar.
Collapse
Affiliation(s)
- Markus M. Knodel
- Goethe Center for Scientific Computing (GCSC), Goethe Universität Frankfurt, Frankfurt, Germany
- *Correspondence: Markus M. Knodel ; orcid.org/0000-0001-8739-0803
| | | | - Gabriel Wittum
- Goethe Center for Scientific Computing (GCSC), Goethe Universität Frankfurt, Frankfurt, Germany
- Applied Mathematics and Computational Science, Computer, Electrical and Mathematical Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| |
Collapse
|
8
|
Xue R, Meng H, Yin J, Xia J, Hu Z, Liu H. The Role of Calmodulin vs. Synaptotagmin in Exocytosis. Front Mol Neurosci 2021; 14:691363. [PMID: 34421537 PMCID: PMC8375295 DOI: 10.3389/fnmol.2021.691363] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/19/2021] [Indexed: 12/04/2022] Open
Abstract
Exocytosis is a Ca2+-regulated process that requires the participation of Ca2+ sensors. In the 1980s, two classes of Ca2+-binding proteins were proposed as putative Ca2+ sensors: EF-hand protein calmodulin, and the C2 domain protein synaptotagmin. In the next few decades, numerous studies determined that in the final stage of membrane fusion triggered by a micromolar boost in the level of Ca2+, the low affinity Ca2+-binding protein synaptotagmin, especially synaptotagmin 1 and 2, acts as the primary Ca2+ sensor, whereas calmodulin is unlikely to be functional due to its high Ca2+ affinity. However, in the meantime emerging evidence has revealed that calmodulin is involved in the earlier exocytotic steps prior to fusion, such as vesicle trafficking, docking and priming by acting as a high affinity Ca2+ sensor activated at submicromolar level of Ca2+. Calmodulin directly interacts with multiple regulatory proteins involved in the regulation of exocytosis, including VAMP, myosin V, Munc13, synapsin, GAP43 and Rab3, and switches on key kinases, such as type II Ca2+/calmodulin-dependent protein kinase, to phosphorylate a series of exocytosis regulators, including syntaxin, synapsin, RIM and Ca2+ channels. Moreover, calmodulin interacts with synaptotagmin through either direct binding or indirect phosphorylation. In summary, calmodulin and synaptotagmin are Ca2+ sensors that play complementary roles throughout the process of exocytosis. In this review, we discuss the complementary roles that calmodulin and synaptotagmin play as Ca2+ sensors during exocytosis.
Collapse
Affiliation(s)
- Renhao Xue
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Hao Meng
- Guangzhou Laboratory, Guangzhou, China.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Jiaxiang Yin
- Guangzhou Laboratory, Guangzhou, China.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Jingyao Xia
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Zhitao Hu
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Huisheng Liu
- Guangzhou Laboratory, Guangzhou, China.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| |
Collapse
|
9
|
SRC3 acetylates calmodulin in the mouse brain to regulate synaptic plasticity and fear learning. J Biol Chem 2021; 297:101044. [PMID: 34358562 PMCID: PMC8390517 DOI: 10.1016/j.jbc.2021.101044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/20/2021] [Accepted: 08/02/2021] [Indexed: 11/23/2022] Open
Abstract
Protein acetylation is a reversible posttranslational modification, which is regulated by lysine acetyltransferase (KAT) and lysine deacetyltransferase (KDAC). Although protein acetylation has been shown to regulate synaptic plasticity, this was mainly for histone protein acetylation. The function and regulation of nonhistone protein acetylation in synaptic plasticity and learning remain largely unknown. Calmodulin (CaM), a ubiquitous Ca2+ sensor, plays critical roles in synaptic plasticity such as long-term potentiation (LTP). During LTP induction, activation of NMDA receptor triggers Ca2+ influx, and the Ca2+ binds with CaM and activates calcium/calmodulin-dependent protein kinase IIα (CaMKIIα). In our previous study, we demonstrated that acetylation of CaM was important for synaptic plasticity and fear learning in mice. However, the KAT responsible for CaM acetylation is currently unknown. Here, following an HEK293 cell-based screen of candidate KATs, steroid receptor coactivator 3 (SRC3) is identified as the most active KAT for CaM. We further demonstrate that SRC3 interacts with and acetylates CaM in a Ca2+ and NMDA receptor-dependent manner. We also show that pharmacological inhibition or genetic downregulation of SRC3 impairs CaM acetylation, synaptic plasticity, and contextual fear learning in mice. Moreover, the effects of SRC3 inhibition on synaptic plasticity and fear learning could be rescued by 3KQ-CaM, a mutant form of CaM, which mimics acetylation. Together, these observations demonstrate that SRC3 acetylates CaM and regulates synaptic plasticity and learning in mice.
Collapse
|
10
|
Zhang HL, Zhao B, Han W, Sun YB, Yang P, Chen Y, Ni D, Zhang J, Yin DM. Acetylation of calmodulin regulates synaptic plasticity and fear learning. J Biol Chem 2021; 297:101034. [PMID: 34339735 PMCID: PMC8383114 DOI: 10.1016/j.jbc.2021.101034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/20/2021] [Accepted: 07/29/2021] [Indexed: 11/26/2022] Open
Abstract
Synaptic plasticity is critical for brain function, including learning and memory. It is regulated by gene transcription and protein synthesis as well as posttranslational modifications at synapses. Although protein acetylation has been shown to be involved in the regulation of synaptic plasticity, this was mainly for histone protein acetylation. To investigate whether acetylation of nonhistone proteins is important for synaptic plasticity, we analyzed mouse brain acetylome and found that calmodulin (CaM), a ubiquitous Ca2+ sensor, was acetylated on three lysine residues, which were conserved across species. NMDA receptor-dependent long-term potentiation (LTP) is considered the most compelling form of synaptic plasticity. During LTP induction, activation of NMDA receptor triggers Ca2+ influx, and the Ca2+ binds with CaM and activates calcium/calmodulin-dependent protein kinase IIα (CaMKIIα), which is essential for LTP induction. By using home-generated and site-specific antibodies against acetylated CaM, we show that CaM acetylation is upregulated by neural activities in an NMDA receptor-dependent manner. Moreover, mutation of acetyllysines in CaM1 proteins disrupts synaptic plasticity and fear learning in a mouse model. We further demonstrate that acetylation of CaM reduces the binding free energy and increases the binding affinity toward CaMKIIα, a protein kinase pivotal to synaptic plasticity and learning. Taken together, our results demonstrate importance of CaM acetylation in regulating synaptic plasticity and learning.
Collapse
Affiliation(s)
- Hai-Long Zhang
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Bing Zhao
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, China
| | - Wei Han
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, China
| | - Yi-Bei Sun
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, China
| | - Pin Yang
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, China
| | - Yongjun Chen
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Duan Ni
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pharmacy, Clinical and Fundamental Research Center, Renji Hospital, Shanghai Jiao-Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Jian Zhang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pharmacy, Clinical and Fundamental Research Center, Renji Hospital, Shanghai Jiao-Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Dong-Min Yin
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, China.
| |
Collapse
|
11
|
Zhou H, Guo F, Luo J, Zhang Y, Liu J, Zhang Y, Zheng X, Wan F, Ding W. Functional analysis of an upregulated calmodulin gene related to the acaricidal activity of curcumin against Tetranychus cinnabarinus (Boisduval). PEST MANAGEMENT SCIENCE 2021; 77:719-730. [PMID: 32865312 DOI: 10.1002/ps.6066] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 08/01/2020] [Accepted: 08/31/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Curcumin is a promising botanical acaricidal compound with activity against Tetranychus cinnabarinus. Calmodulin (CaM) is a key calcium ion (Ca2+ ) sensor that plays a vital role in calcium signaling. Overexpression of the CaM gene with inducible character occurs in curcumin-treated mites, but its functional role remains to be further analyzed by RNA interference (RNAi) and protein expression. RESULTS A CaM gene was cloned from T. cinnabarinus (designated TcCaM). TcCaM was upregulated and the protein was activated in mites by curcumin. The susceptibility of mites to curcumin was decreased after inhibiting CaM function with anti-CaM drug trifluoperazine (TFP) and silencing CaM transcription with RNAi, suggesting that the CaM gene is involved in the acaricidal activity of curcumin against mites. Moreover, the TFP pre-treated Sf9 cells were resistant to curcumin-mediated increase in [Ca2+ ]i levels, indicating that CaM-mediated Ca2+ homeostasis was disturbed by curcumin. TcCaM was then re-engineered for heterologous expression in Escherichia coli. Strikingly, our results showed that the recombinant CaM protein was directly activated by curcumin via inducing its conformational changes, its half-maximal effective concentration (EC50 ) value is 0.3 μmol L-1 in vitro, which is similar to curcumin against CaM-expressing Sf9 cells (0.76 μmol L-1 ) in vivo. CONCLUSION These results confirm that the overexpressed CaM gene is involved in the acaricidal activity of curcumin, and the mode of action of curcumin may be via activating CaM function, and thereby disrupting Ca2+ homeostasis in T. cinnabarinus. This study highlights the novel target mechanism of new acaricides, promoting our understanding of the molecular mechanism of CaM-mediated acaricide targets in mites.
Collapse
Affiliation(s)
- Hong Zhou
- Institute of Pesticide Science, College of Plant Protection, Southwest University, Chongqing, P. R. China
| | - Fuyou Guo
- Institute of Pesticide Science, College of Plant Protection, Southwest University, Chongqing, P. R. China
| | - Jinxiang Luo
- Institute of Pesticide Science, College of Plant Protection, Southwest University, Chongqing, P. R. China
| | - Yongqiang Zhang
- Institute of Pesticide Science, College of Plant Protection, Southwest University, Chongqing, P. R. China
| | - Jinlin Liu
- Institute of Pesticide Science, College of Plant Protection, Southwest University, Chongqing, P. R. China
| | - Yanchun Zhang
- Institute of Pesticide Science, College of Plant Protection, Southwest University, Chongqing, P. R. China
| | - Xinyu Zheng
- Institute of Pesticide Science, College of Plant Protection, Southwest University, Chongqing, P. R. China
| | - Fenglin Wan
- Institute of Pesticide Science, College of Plant Protection, Southwest University, Chongqing, P. R. China
| | - Wei Ding
- Institute of Pesticide Science, College of Plant Protection, Southwest University, Chongqing, P. R. China
| |
Collapse
|
12
|
Calmodulin Bidirectionally Regulates Evoked and Spontaneous Neurotransmitter Release at Retinal Ribbon Synapses. eNeuro 2021; 8:ENEURO.0257-20.2020. [PMID: 33293457 PMCID: PMC7808332 DOI: 10.1523/eneuro.0257-20.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 11/17/2020] [Accepted: 11/21/2020] [Indexed: 11/21/2022] Open
Abstract
For decades, a role for the Ca2+-binding protein calmodulin (CaM) in Ca2+-dependent presynaptic modulation of synaptic transmission has been recognized. Here, we investigated the influence of CaM on evoked and spontaneous neurotransmission at rod bipolar (RB) cell→AII amacrine cell synapses in the mouse retina. Our work was motivated by the observations that expression of CaM in RB axon terminals is extremely high and that [Ca2+] in RB terminals normally rises sufficiently to saturate endogenous buffers, making tonic CaM activation likely. Taking advantage of a model in which RBs can be stimulated by expressed channelrhodopsin-2 (ChR2) to avoid dialysis of the presynaptic terminal, we found that inhibition of CaM dramatically decreased evoked release by inhibition of presynaptic Ca channels while at the same time potentiating both Ca2+-dependent and Ca2+-independent spontaneous release. Remarkably, inhibition of myosin light chain kinase (MLCK), but not other CaM-dependent targets, mimicked the effects of CaM inhibition on evoked and spontaneous release. Importantly, initial antagonism of CaM occluded the effect of subsequent inhibition of MLCK on spontaneous release. We conclude that CaM, by acting through MLCK, bidirectionally regulates evoked and spontaneous release at retinal ribbon synapses.
Collapse
|
13
|
Alcalde J, Munk M, González-Muñoz M, Panina S, Berchtold MW, Villalobo A. Calmodulin downregulation in conditional knockout HeLa cells inhibits cell migration. Arch Biochem Biophys 2020; 697:108680. [PMID: 33220265 DOI: 10.1016/j.abb.2020.108680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/05/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022]
Abstract
The study of calmodulin (CaM) functions in living cells has been tackled up to date using cell-permeant CaM inhibitors or interference-RNA methods. CaM inhibitors may lack specificity and the siRNA interference approach is challenging, as all three CaM genes expressing an identical protein in mammals have to be blocked. Therefore, we recently introduced a novel genetic system using CRISPR/Cas9-mediated gene deletion and conditional CaM expression to study the function of CaM in HeLa cells. Here, we describe the effect of CaM downregulation on the basal and epidermal growth factor (EGF)-dependent 2D- and 3D-migration in HeLa cells. CaM downregulation inhibited cell migration on a 2D-surface in the absence but not in the presence of EGF. In contrast, CaM downregulation led to inhibition of 3D-migration across a porous membrane both in the absence and presence of EGF. CaM downregulation decreased the expression of Rac1, Cdc42 and RhoA, all known to play crucial roles in cell migration. These results show that EGF-dependent 2D- and 3D-migration utilize distinct CaM-regulated systems and identify several essential migratory proteins directly or indirectly regulated by CaM.
Collapse
Affiliation(s)
- Juan Alcalde
- Department of Biology, University of Copenhagen, 13 Universitetsparken, DK-2100, Copenhagen Ø, Denmark; Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Mads Munk
- Department of Biology, University of Copenhagen, 13 Universitetsparken, DK-2100, Copenhagen Ø, Denmark
| | - María González-Muñoz
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Svetlana Panina
- Department of Biology, University of Copenhagen, 13 Universitetsparken, DK-2100, Copenhagen Ø, Denmark
| | - Martin W Berchtold
- Department of Biology, University of Copenhagen, 13 Universitetsparken, DK-2100, Copenhagen Ø, Denmark.
| | - Antonio Villalobo
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain; Cancer and Human Molecular Genetics Area - Oto-Neurosurgery Research Group, University Hospital La Paz Research Institute (IdiPAZ), Paseo de la Castellana 261, E-28046, Madrid, Spain.
| |
Collapse
|
14
|
Sharma A, Hasan G. Modulation of flight and feeding behaviours requires presynaptic IP 3Rs in dopaminergic neurons. eLife 2020; 9:e62297. [PMID: 33155978 PMCID: PMC7647402 DOI: 10.7554/elife.62297] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 10/25/2020] [Indexed: 12/17/2022] Open
Abstract
Innate behaviours, although robust and hard wired, rely on modulation of neuronal circuits, for eliciting an appropriate response according to internal states and external cues. Drosophila flight is one such innate behaviour that is modulated by intracellular calcium release through inositol 1,4,5-trisphosphate receptors (IP3Rs). Cellular mechanism(s) by which IP3Rs modulate neuronal function for specific behaviours remain speculative, in vertebrates and invertebrates. To address this, we generated an inducible dominant negative form of the IP3R (IP3RDN). Flies with neuronal expression of IP3RDN exhibit flight deficits. Expression of IP3RDN helped identify key flight-modulating dopaminergic neurons with axonal projections in the mushroom body. Flies with attenuated IP3Rs in these presynaptic dopaminergic neurons exhibit shortened flight bouts and a disinterest in seeking food, accompanied by reduced excitability and dopamine release upon cholinergic stimulation. Our findings suggest that the same neural circuit modulates the drive for food search and for undertaking longer flight bouts.
Collapse
Affiliation(s)
- Anamika Sharma
- National Centre for Biological Sciences, TIFRBangaloreIndia
| | - Gaiti Hasan
- National Centre for Biological Sciences, TIFRBangaloreIndia
| |
Collapse
|
15
|
Bae B, Gruner HN, Lynch M, Feng T, So K, Oliver D, Mastick GS, Yan W, Pieraut S, Miura P. Elimination of Calm1 long 3'-UTR mRNA isoform by CRISPR-Cas9 gene editing impairs dorsal root ganglion development and hippocampal neuron activation in mice. RNA (NEW YORK, N.Y.) 2020; 26:1414-1430. [PMID: 32522888 PMCID: PMC7491327 DOI: 10.1261/rna.076430.120] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 06/06/2020] [Indexed: 05/04/2023]
Abstract
The majority of mouse and human genes are subject to alternative cleavage and polyadenylation (APA), which most often leads to the expression of two or more alternative length 3' untranslated region (3'-UTR) mRNA isoforms. In neural tissues, there is enhanced expression of APA isoforms with longer 3'-UTRs on a global scale, but the physiological relevance of these alternative 3'-UTR isoforms is poorly understood. Calmodulin 1 (Calm1) is a key integrator of calcium signaling that generates short (Calm1-S) and long (Calm1-L) 3'-UTR mRNA isoforms via APA. We found Calm1-L expression to be largely restricted to neural tissues in mice including the dorsal root ganglion (DRG) and hippocampus, whereas Calm1-S was more broadly expressed. smFISH revealed that both Calm1-S and Calm1-L were subcellularly localized to neural processes of primary hippocampal neurons. In contrast, cultured DRG showed restriction of Calm1-L to soma. To investigate the in vivo functions of Calm1-L, we implemented a CRISPR-Cas9 gene editing strategy to delete a small region encompassing the Calm1 distal poly(A) site. This eliminated Calm1-L expression while maintaining expression of Calm1-S Mice lacking Calm1-L (Calm1ΔL/ΔL ) exhibited disorganized DRG migration in embryos, and reduced experience-induced neuronal activation in the adult hippocampus. These data indicate that Calm1-L plays functional roles in the central and peripheral nervous systems.
Collapse
Affiliation(s)
- Bongmin Bae
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
| | - Hannah N Gruner
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
| | - Maebh Lynch
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
| | - Ting Feng
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
| | - Kevin So
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
| | - Daniel Oliver
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada 89557, USA
| | - Grant S Mastick
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
| | - Wei Yan
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada 89557, USA
| | - Simon Pieraut
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
| | - Pedro Miura
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
| |
Collapse
|
16
|
Chung D, Shum A, Caraveo G. GAP-43 and BASP1 in Axon Regeneration: Implications for the Treatment of Neurodegenerative Diseases. Front Cell Dev Biol 2020; 8:567537. [PMID: 33015061 PMCID: PMC7494789 DOI: 10.3389/fcell.2020.567537] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/14/2020] [Indexed: 01/06/2023] Open
Abstract
Growth-associated protein-43 (GAP-43) and brain acid-soluble protein 1 (BASP1) regulate actin dynamics and presynaptic vesicle cycling at axon terminals, thereby facilitating axonal growth, regeneration, and plasticity. These functions highly depend on changes in GAP-43 and BASP1 expression levels and post-translational modifications such as phosphorylation. Interestingly, examinations of GAP-43 and BASP1 in neurodegenerative diseases reveal alterations in their expression and phosphorylation profiles. This review provides an overview of the structural properties, regulations, and functions of GAP-43 and BASP1, highlighting their involvement in neural injury response and regeneration. By discussing GAP-43 and BASP1 in the context of neurodegenerative diseases, we also explore the therapeutic potential of modulating their activities to compensate for neuron loss in neurodegenerative diseases.
Collapse
Affiliation(s)
- Daayun Chung
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Andrew Shum
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Gabriela Caraveo
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
17
|
Synaptotagmin 1 oligomers clamp and regulate different modes of neurotransmitter release. Proc Natl Acad Sci U S A 2020; 117:3819-3827. [PMID: 32015138 PMCID: PMC7035618 DOI: 10.1073/pnas.1920403117] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Release of neurotransmitters relies on submillisecond coupling of synaptic vesicle fusion to the triggering signal: AP-evoked presynaptic Ca2+ influx. The key player that controls exocytosis of the synaptic vesicle is the Ca2+ sensor synaptotagmin 1 (Syt1). While the Ca2+ activation of Syt1 has been extensively characterized, how Syt1 reversibly clamps vesicular fusion remains enigmatic. Here, using a targeted mutation combined with fluorescence imaging and electrophysiology, we show that the structural feature of Syt1 to self-oligomerize provides the molecular basis for clamping of spontaneous and asynchronous release but is not required for triggering of synchronous release. Our findings propose a mechanistic model that explains how Syt1 oligomers regulate different modes of transmitter release in neuronal synapses. Synaptotagmin 1 (Syt1) synchronizes neurotransmitter release to action potentials (APs) acting as the fast Ca2+ release sensor and as the inhibitor (clamp) of spontaneous and delayed asynchronous release. While the Syt1 Ca2+ activation mechanism has been well-characterized, how Syt1 clamps transmitter release remains enigmatic. Here we show that C2B domain-dependent oligomerization provides the molecular basis for the Syt1 clamping function. This follows from the investigation of a designed mutation (F349A), which selectively destabilizes Syt1 oligomerization. Using a combination of fluorescence imaging and electrophysiology in neocortical synapses, we show that Syt1F349A is more efficient than wild-type Syt1 (Syt1WT) in triggering synchronous transmitter release but fails to clamp spontaneous and synaptotagmin 7 (Syt7)-mediated asynchronous release components both in rescue (Syt1−/− knockout background) and dominant-interference (Syt1+/+ background) conditions. Thus, we conclude that Ca2+-sensitive Syt1 oligomers, acting as an exocytosis clamp, are critical for maintaining the balance among the different modes of neurotransmitter release.
Collapse
|
18
|
CAMK2-Dependent Signaling in Neurons Is Essential for Survival. J Neurosci 2019; 39:5424-5439. [PMID: 31064859 DOI: 10.1523/jneurosci.1341-18.2019] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 03/29/2019] [Accepted: 03/29/2019] [Indexed: 01/09/2023] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CAMK2) is a key player in synaptic plasticity and memory formation. Mutations in Camk2a or Camk2b cause intellectual disability in humans, and severe plasticity and learning deficits in mice, indicating unique functions for each isoform. However, considering the high homology between CAMK2A and CAMK2B, it is conceivable that for critical functions, one isoform compensates for the absence of the other, and that the full functional spectrum of neuronal CAMK2 remains to be revealed.Here we show that germline as well as adult deletion of both CAMK2 isoforms in male or female mice is lethal. Moreover, Ca2+-dependent activity as well as autonomous activity of CAMK2 is essential for survival. Loss of both CAMK2 isoforms abolished LTP, whereas synaptic transmission remained intact. The double-mutants showed no gross morphological changes of the brain, and in contrast to the long-considered role for CAMK2 in the structural organization of the postsynaptic density (PSD), deletion of both CAMK2 isoforms did not affect the biochemical composition of the PSD. Together, these results reveal an essential role for CAMK2 signaling in early postnatal development as well as the mature brain, and indicate that the full spectrum of CAMK2 requirements cannot be revealed in the single mutants because of partial overlapping functions of CAMK2A and CAMK2B.SIGNIFICANCE STATEMENT CAMK2A and CAMK2B have been studied for over 30 years for their role in neuronal functioning. However, most studies were performed using single knock-out mice. Because the two isoforms show high homology with respect to structure and function, it is likely that some redundancy exists between the two isoforms, meaning that for critical functions CAMK2B compensates for the absence of CAMK2A and vice versa, leaving these functions to uncover. In this study, we generated Camk2a/Camk2b double-mutant mice, and observed that loss of CAMK2, as well as the loss of Ca2+-dependent and Ca2+-independent activity of CAMK2 is lethal. These results indicate that despite 30 years of research the full spectrum of CAMK2 functioning in neurons remains to be unraveled.
Collapse
|
19
|
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: 50] [Impact Index Per Article: 8.3] [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
|
20
|
Clarke SE. Analog Signaling With the "Digital" Molecular Switch CaMKII. Front Comput Neurosci 2018; 12:92. [PMID: 30524260 PMCID: PMC6262075 DOI: 10.3389/fncom.2018.00092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/31/2018] [Indexed: 11/13/2022] Open
Abstract
Molecular switches, such as the protein kinase CaMKII, play a fundamental role in cell signaling by decoding inputs into either high or low states of activity; because the high activation state can be turned on and persist after the input ceases, these switches have earned a reputation as "digital." Although this on/off, binary perspective has been valuable for understanding long timescale synaptic plasticity, accumulating experimental evidence suggests that the CaMKII switch can also control plasticity on short timescales. To investigate this idea further, a non-autonomous, nonlinear ordinary differential equation, representative of a general bistable molecular switch, is analyzed. The results suggest that switch activity in regions surrounding either the high- or low-stable states of activation could act as a reliable analog signal, whose short timescale fluctuations relative to equilibrium track instantaneous input frequency. The model makes intriguing predictions and is validated against previous work demonstrating its suitability as a minimal representation of switch dynamics; in combination with existing experimental evidence, the theory suggests a multiplexed encoding of instantaneous frequency information over short timescales, with integration of total activity over longer timescales.
Collapse
Affiliation(s)
- Stephen E Clarke
- Department of Bioengineering, Department of Neurosurgery, Stanford University, Stanford, CA, United States
| |
Collapse
|
21
|
McGowan H, Mirabella VR, Hamod A, Karakhanyan A, Mlynaryk N, Moore JC, Tischfield JA, Hart RP, Pang ZP. hsa-let-7c miRNA Regulates Synaptic and Neuronal Function in Human Neurons. Front Synaptic Neurosci 2018; 10:19. [PMID: 30065644 PMCID: PMC6056636 DOI: 10.3389/fnsyn.2018.00019] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/18/2018] [Indexed: 12/26/2022] Open
Abstract
Non-coding RNA, including microRNA (miRNA) serves critical regulatory functions in the developing brain. The let-7 family of miRNAs has been shown to regulate neuronal differentiation, neural subtype specification, and synapse formation in animal models. However, the regulatory role of human let-7c (hsa-let-7c) in human neuronal development has yet to be examined. Let-7c is encoded on chromosome 21 in humans and therefore may be overexpressed in human brains in Trisomy 21 (T21), a complex neurodevelopmental disorder. Here, we employ recent developments in stem cell biology to show that hsa-let-7c mediates important regulatory epigenetic functions that control the development and functional activity of human induced neuronal cells (iNs). We show that overexpression of hsa-let-7c in human iNs derived from induced pluripotent stem (iPS), as well as embryonic stem (ES), cells leads to morphological as well as functional deficits including impaired neuronal morphologic development, synapse formation and synaptic strength, as well as a marked reduction of neuronal excitability. Importantly, we have assessed these findings over three independent genetic backgrounds, showing that some of these effects are subject to influence by background genetic variability with the most robust and reproducible effect being a striking reduction in spontaneous neural firing. Collectively, these results suggest an important function for let-7 family miRNAs in regulation of human neuronal development and raise implications for understanding the complex molecular etiology of neurodevelopmental disorders, such as T21, where let-7c gene dosage is increased.
Collapse
Affiliation(s)
- Heather McGowan
- Child Health Institute of New Jersey, New Brunswick, NJ, United States
- Department of Neuroscience and Cell Biology, Rutgers University, Piscataway, NJ, United States
| | - Vincent R. Mirabella
- Child Health Institute of New Jersey, New Brunswick, NJ, United States
- Department of Neuroscience and Cell Biology, Rutgers University, Piscataway, NJ, United States
| | - Aula Hamod
- Child Health Institute of New Jersey, New Brunswick, NJ, United States
- Department of Neuroscience and Cell Biology, Rutgers University, Piscataway, NJ, United States
| | - Aziz Karakhanyan
- Child Health Institute of New Jersey, New Brunswick, NJ, United States
- Department of Neuroscience and Cell Biology, Rutgers University, Piscataway, NJ, United States
| | - Nicole Mlynaryk
- Child Health Institute of New Jersey, New Brunswick, NJ, United States
- Department of Neuroscience and Cell Biology, Rutgers University, Piscataway, NJ, United States
| | - Jennifer C. Moore
- Department of Genetics, Rutgers University, Piscataway, NJ, United States
| | - Jay A. Tischfield
- Department of Genetics, Rutgers University, Piscataway, NJ, United States
| | - Ronald P. Hart
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Zhiping P. Pang
- Child Health Institute of New Jersey, New Brunswick, NJ, United States
- Department of Neuroscience and Cell Biology, Rutgers University, Piscataway, NJ, United States
| |
Collapse
|
22
|
Singh M, Lujan B, Renden R. Presynaptic GCaMP expression decreases vesicle release probability at the calyx of Held. Synapse 2018; 72:e22040. [PMID: 29935099 PMCID: PMC6186185 DOI: 10.1002/syn.22040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/04/2018] [Accepted: 06/05/2018] [Indexed: 12/15/2022]
Abstract
Synaptic vesicle (SV) exocytosis is intimately dependent on free local Ca2+ near active zones. Genetically encoded calcium indicators (GECIs) have become an indispensable tool to monitor calcium dynamics during physiological responses, and they are widely used as a proxy to monitor activity in neuronal ensembles and at synaptic terminals. However, GECIs’ ability to bind Ca2+ at physiologically relevant concentration makes them strong candidates to affect calcium homeostasis and alter synaptic transmission by exogenously increasing Ca2+ buffering. In the present study, we show that genetically expressed GCaMP6m modulates SV release probability at the mouse calyx of Held synapse. GCaMP6m expression for approximately three weeks decreased initial SV release for both low‐frequency stimulation and high‐frequency stimulation trains, and slowed presynaptic short‐term depression. However, GCaMP6m does not affect quantal events during spontaneous activity at this synapse. This study emphasizes the careful use of GECIs as monitors of neuronal activity and inspects the role of these transgenic indicators which may alter calcium‐dependent physiological responses.
Collapse
Affiliation(s)
- Mahendra Singh
- Department of Physiology and Cell Biology, University of Nevada, Reno, Nevada, 89557
| | - Brendan Lujan
- Department of Physiology and Cell Biology, University of Nevada, Reno, Nevada, 89557.,Currently at Vollum Institute, Oregon Health and Science University, Portland, Oregon
| | - Robert Renden
- Department of Physiology and Cell Biology, University of Nevada, Reno, Nevada, 89557
| |
Collapse
|
23
|
PTPσ Drives Excitatory Presynaptic Assembly via Various Extracellular and Intracellular Mechanisms. J Neurosci 2018; 38:6700-6721. [PMID: 29934346 DOI: 10.1523/jneurosci.0672-18.2018] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/22/2018] [Accepted: 06/14/2018] [Indexed: 11/21/2022] Open
Abstract
Leukocyte common antigen-receptor protein tyrosine phosphatases (LAR-RPTPs) are hub proteins that organize excitatory and inhibitory synapse development through binding to various extracellular ligands. Here, we report that knockdown (KD) of the LAR-RPTP family member PTPσ reduced excitatory synapse number and transmission in cultured rat hippocampal neurons, whereas KD of PTPδ produced comparable decreases at inhibitory synapses, in both cases without altering expression levels of interacting proteins. An extensive series of rescue experiments revealed that extracellular interactions of PTPσ with Slitrks are important for excitatory synapse development. These experiments further showed that the intracellular D2 domain of PTPσ is required for induction of heterologous synapse formation by Slitrk1 or TrkC, suggesting that interaction of LAR-RPTPs with distinct intracellular presynaptic proteins, drives presynaptic machinery assembly. Consistent with this, double-KD of liprin-α2 and -α3 or KD of PTPσ substrates (N-cadherin and p250RhoGAP) in neurons inhibited Slitrk6-induced, PTPσ-mediated heterologous synapse formation activity. We propose a synaptogenesis model in presynaptic neurons involving LAR-RPTP-organized retrograde signaling cascades, in which both extracellular and intracellular mechanisms are critical in orchestrating distinct synapse types.SIGNIFICANCE STATEMENT In this study, we sought to test the unproven hypothesis that PTPσ and PTPδ are required for excitatory and inhibitory synapse formation/transmission, respectively, in cultured hippocampal neurons, using knockdown-based loss-of-function analyses. We further performed extensive structure-function analyses, focusing on PTPσ-mediated actions, to address the mechanisms of presynaptic assembly at excitatory synaptic sites. Using interdisciplinary approaches, we systematically applied a varied set of PTPσ deletion variants, point mutants, and splice variants to demonstrate that both extracellular and intracellular mechanisms are involved in organizing presynaptic assembly. Strikingly, extracellular interactions of PTPσ with heparan sulfates and Slitrks, intracellular interactions of PTPσ with liprin-α and its associated proteins through the D2 domain, as well as distinct substrates are all critical.
Collapse
|
24
|
Wang C, Kang X, Zhou L, Chai Z, Wu Q, Huang R, Xu H, Hu M, Sun X, Sun S, Li J, Jiao R, Zuo P, Zheng L, Yue Z, Zhou Z. Synaptotagmin-11 is a critical mediator of parkin-linked neurotoxicity and Parkinson's disease-like pathology. Nat Commun 2018; 9:81. [PMID: 29311685 PMCID: PMC5758517 DOI: 10.1038/s41467-017-02593-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 12/09/2017] [Indexed: 11/17/2022] Open
Abstract
Loss-of-function mutations in Parkin are the most common causes of autosomal recessive Parkinson’s disease (PD). Many putative substrates of parkin have been reported; their pathogenic roles, however, remain obscure due to poor characterization, particularly in vivo. Here, we show that synaptotagmin-11, encoded by a PD-risk gene SYT11, is a physiological substrate of parkin and plays critical roles in mediating parkin-linked neurotoxicity. Unilateral overexpression of full-length, but not C2B-truncated, synaptotagmin-11 in the substantia nigra pars compacta (SNpc) impairs ipsilateral striatal dopamine release, causes late-onset degeneration of dopaminergic neurons, and induces progressive contralateral motor abnormalities. Mechanistically, synaptotagmin-11 impairs vesicle pool replenishment and thus dopamine release by inhibiting endocytosis. Furthermore, parkin deficiency induces synaptotagmin-11 accumulation and PD-like neurotoxicity in mouse models, which is reversed by SYT11 knockdown in the SNpc or knockout of SYT11 restricted to dopaminergic neurons. Thus, PD-like neurotoxicity induced by parkin dysfunction requires synaptotagmin-11 accumulation in SNpc dopaminergic neurons. Mutations in the parkin, an ubiquitin ligase, are linked to Parkinson’s disease. Here the authors show that synaptotagmin-11 is a parkin substrate and that its upregulation affects dopamine release, triggers degeneration, and causes motor impairment.
Collapse
Affiliation(s)
- Changhe Wang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.,Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xinjiang Kang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.,College of Life Sciences, Liaocheng University, Liaocheng, 252059, China.,Key Lab of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Li Zhou
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Zuying Chai
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Qihui Wu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Rong Huang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Huadong Xu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Meiqin Hu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Xiaoxuan Sun
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Suhua Sun
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Jie Li
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Ruiying Jiao
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Panli Zuo
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Lianghong Zheng
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Zhenyu Yue
- Departments of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Zhuan Zhou
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
| |
Collapse
|
25
|
Chemin J, Taiakina V, Monteil A, Piazza M, Guan W, Stephens RF, Kitmitto A, Pang ZP, Dolphin AC, Perez-Reyes E, Dieckmann T, Guillemette JG, Spafford JD. Calmodulin regulates Ca v3 T-type channels at their gating brake. J Biol Chem 2017; 292:20010-20031. [PMID: 28972185 PMCID: PMC5723990 DOI: 10.1074/jbc.m117.807925] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/19/2017] [Indexed: 01/10/2023] Open
Abstract
Calcium (Cav1 and Cav2) and sodium channels possess homologous CaM-binding motifs, known as IQ motifs in their C termini, which associate with calmodulin (CaM), a universal calcium sensor. Cav3 T-type channels, which serve as pacemakers of the mammalian brain and heart, lack a C-terminal IQ motif. We illustrate that T-type channels associate with CaM using co-immunoprecipitation experiments and single particle cryo-electron microscopy. We demonstrate that protostome invertebrate (LCav3) and human Cav3.1, Cav3.2, and Cav3.3 T-type channels specifically associate with CaM at helix 2 of the gating brake in the I-II linker of the channels. Isothermal titration calorimetry results revealed that the gating brake and CaM bind each other with high-nanomolar affinity. We show that the gating brake assumes a helical conformation upon binding CaM, with associated conformational changes to both CaM lobes as indicated by amide chemical shifts of the amino acids of CaM in 1H-15N HSQC NMR spectra. Intact Ca2+-binding sites on CaM and an intact gating brake sequence (first 39 amino acids of the I-II linker) were required in Cav3.2 channels to prevent the runaway gating phenotype, a hyperpolarizing shift in voltage sensitivities and faster gating kinetics. We conclude that the presence of high-nanomolar affinity binding sites for CaM at its universal gating brake and its unique form of regulation via the tuning of the voltage range of activity could influence the participation of Cav3 T-type channels in heart and brain rhythms. Our findings may have implications for arrhythmia disorders arising from mutations in the gating brake or CaM.
Collapse
Affiliation(s)
- Jean Chemin
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, Montpellier F-34094, France
| | | | - Arnaud Monteil
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, Montpellier F-34094, France
| | - Michael Piazza
- Departments of Chemistry, Waterloo, Ontario N2L 3G1, Canada
| | - Wendy Guan
- Biology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | | | - Ashraf Kitmitto
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, United Kingdom
| | - Zhiping P Pang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey 08901
| | - Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Edward Perez-Reyes
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908
| | | | | | - J David Spafford
- Biology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada.
| |
Collapse
|
26
|
Intellicount: High-Throughput Quantification of Fluorescent Synaptic Protein Puncta by Machine Learning. eNeuro 2017; 4:eN-MNT-0219-17. [PMID: 29218324 PMCID: PMC5718246 DOI: 10.1523/eneuro.0219-17.2017] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 10/18/2017] [Accepted: 11/06/2017] [Indexed: 01/07/2023] Open
Abstract
Synapse formation analyses can be performed by imaging and quantifying fluorescent signals of synaptic markers. Traditionally, these analyses are done using simple or multiple thresholding and segmentation approaches or by labor-intensive manual analysis by a human observer. Here, we describe Intellicount, a high-throughput, fully-automated synapse quantification program which applies a novel machine learning (ML)-based image processing algorithm to systematically improve region of interest (ROI) identification over simple thresholding techniques. Through processing large datasets from both human and mouse neurons, we demonstrate that this approach allows image processing to proceed independently of carefully set thresholds, thus reducing the need for human intervention. As a result, this method can efficiently and accurately process large image datasets with minimal interaction by the experimenter, making it less prone to bias and less liable to human error. Furthermore, Intellicount is integrated into an intuitive graphical user interface (GUI) that provides a set of valuable features, including automated and multifunctional figure generation, routine statistical analyses, and the ability to run full datasets through nested folders, greatly expediting the data analysis process.
Collapse
|
27
|
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
|
28
|
Li J, Han W, Pelkey KA, Duan J, Mao X, Wang YX, Craig MT, Dong L, Petralia RS, McBain CJ, Lu W. Molecular Dissection of Neuroligin 2 and Slitrk3 Reveals an Essential Framework for GABAergic Synapse Development. Neuron 2017; 96:808-826.e8. [PMID: 29107521 PMCID: PMC5957482 DOI: 10.1016/j.neuron.2017.10.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/18/2017] [Accepted: 10/02/2017] [Indexed: 10/18/2022]
Abstract
In the brain, many types of interneurons make functionally diverse inhibitory synapses onto principal neurons. Although numerous molecules have been identified to function in inhibitory synapse development, it remains unknown whether there is a unifying mechanism for development of diverse inhibitory synapses. Here we report a general molecular mechanism underlying hippocampal inhibitory synapse development. In developing neurons, the establishment of GABAergic transmission depends on Neuroligin 2 (NL2), a synaptic cell adhesion molecule (CAM). During maturation, inhibitory synapse development requires both NL2 and Slitrk3 (ST3), another CAM. Importantly, NL2 and ST3 interact with nanomolar affinity through their extracellular domains to synergistically promote synapse development. Selective perturbation of the NL2-ST3 interaction impairs inhibitory synapse development with consequent disruptions in hippocampal network activity and increased seizure susceptibility. Our findings reveal how unique postsynaptic CAMs work in concert to control synaptogenesis and establish a general framework for GABAergic synapse development.
Collapse
Affiliation(s)
- Jun Li
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wenyan Han
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kenneth A Pelkey
- Program in Developmental Neuroscience, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jingjing Duan
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xia Mao
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ya-Xian Wang
- Advanced Imaging Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael T Craig
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Prince of Wales Road, Exeter EX4 4PS, UK
| | - Lijin Dong
- Genetic Engineering Core, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ronald S Petralia
- Advanced Imaging Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chris J McBain
- Program in Developmental Neuroscience, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Lu
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
29
|
Chowdhury D, Turner M, Patriarchi T, Hergarden AC, Anderson D, Zhang Y, Sun J, Chen CY, Ames JB, Hell JW. Ca 2+/calmodulin binding to PSD-95 mediates homeostatic synaptic scaling down. EMBO J 2017; 37:122-138. [PMID: 29118000 DOI: 10.15252/embj.201695829] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 09/07/2017] [Accepted: 10/02/2017] [Indexed: 11/09/2022] Open
Abstract
Postsynaptic density protein-95 (PSD-95) localizes AMPA-type glutamate receptors (AMPARs) to postsynaptic sites of glutamatergic synapses. Its postsynaptic displacement is necessary for loss of AMPARs during homeostatic scaling down of synapses. Here, we demonstrate that upon Ca2+ influx, Ca2+/calmodulin (Ca2+/CaM) binding to the N-terminus of PSD-95 mediates postsynaptic loss of PSD-95 and AMPARs during homeostatic scaling down. Our NMR structural analysis identified E17 within the PSD-95 N-terminus as important for binding to Ca2+/CaM by interacting with R126 on CaM. Mutating E17 to R prevented homeostatic scaling down in primary hippocampal neurons, which is rescued via charge inversion by ectopic expression of CaMR126E, as determined by analysis of miniature excitatory postsynaptic currents. Accordingly, increased binding of Ca2+/CaM to PSD-95 induced by a chronic increase in Ca2+ influx is a critical molecular event in homeostatic downscaling of glutamatergic synaptic transmission.
Collapse
Affiliation(s)
| | - Matthew Turner
- Department of Chemistry, University of California, Davis, CA, USA
| | | | - Anne C Hergarden
- Department of Pharmacology, University of California, Davis, CA, USA
| | - David Anderson
- Department of Chemistry, University of California, Davis, CA, USA
| | - Yonghong Zhang
- Department of Chemistry, University of Texas, Edinburgh, TX, USA
| | - Junqing Sun
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Chao-Yin Chen
- Department of Pharmacology, University of California, Davis, CA, USA
| | - James B Ames
- Department of Chemistry, University of California, Davis, CA, USA
| | - Johannes W Hell
- Department of Pharmacology, University of California, Davis, CA, USA
| |
Collapse
|
30
|
Velásquez E, Nogueira FCS, Velásquez I, Schmitt A, Falkai P, Domont GB, Martins-de-Souza D. Synaptosomal Proteome of the Orbitofrontal Cortex from Schizophrenia Patients Using Quantitative Label-Free and iTRAQ-Based Shotgun Proteomics. J Proteome Res 2017; 16:4481-4494. [PMID: 28949146 DOI: 10.1021/acs.jproteome.7b00422] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Schizophrenia is a chronic and incurable neuropsychiatric disorder that affects about one percent of the world population. The proteomic characterization of the synaptosome fraction of the orbitofrontal cortex is useful for providing valuable information about the molecular mechanisms of synaptic functions in these patients. Quantitative analyses of synaptic proteins were made with eight paranoid schizophrenia patients and a pool of eight healthy controls free of mental diseases. Label-free and iTRAQ labeling identified a total of 2018 protein groups. Statistical analyses revealed 12 and 55 significantly dysregulated proteins by iTRAQ and label-free, respectively. Quantitative proteome analyses showed an imbalance in the calcium signaling pathway and proteins such as reticulon-1 and cytochrome c, related to endoplasmic reticulum stress and programmed cell death. Also, it was found that there is a significant increase in limbic-system-associated membrane protein and α-calcium/calmodulin-dependent protein kinase II, associated with the regulation of human behavior. Our data contribute to a better understanding about apoptosis as a possible pathophysiological mechanism of this disease as well as neural systems supporting social behavior in schizophrenia. This study also is a joint effort of the Chr 15 C-HPP team and the Human Brain Proteome Project of B/D-HPP. All MS proteomics data are deposited in the ProteomeXchange Repository under PXD006798.
Collapse
Affiliation(s)
- Erika Velásquez
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro , Rio de Janeiro, 21941-909 Rio de Janeiro, Brazil
| | - Fabio C S Nogueira
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro , Rio de Janeiro, 21941-909 Rio de Janeiro, Brazil.,Laboratory of Proteomics, LADETEC, Institute of Chemistry, Federal University of Rio de Janeiro , Rio de Janeiro, 21941-598 Rio de Janeiro, Brazil
| | | | - Andrea Schmitt
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University of Munich (LMU) , 80336 Munich, Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University of Munich (LMU) , 80336 Munich, Germany
| | - Gilberto B Domont
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro , Rio de Janeiro, 21941-909 Rio de Janeiro, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry, Institute of Biology, University of Campinas (UNICAMP) , Campinas, 13083-862 São Paulo, Brazil.,UNICAMP's Neurobiology Center , Campinas, 13083-888 São Paulo, Brazil.,Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico , São Paulo, 01060-970 São Paulo, Brazil
| |
Collapse
|
31
|
Swiatkowski P, Nikolaeva I, Kumar G, Zucco A, Akum BF, Patel MV, D'Arcangelo G, Firestein BL. Role of Akt-independent mTORC1 and GSK3β signaling in sublethal NMDA-induced injury and the recovery of neuronal electrophysiology and survival. Sci Rep 2017; 7:1539. [PMID: 28484273 PMCID: PMC5431483 DOI: 10.1038/s41598-017-01826-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 04/03/2017] [Indexed: 01/02/2023] Open
Abstract
Glutamate-induced excitotoxicity, mediated by overstimulation of N-methyl-D-aspartate (NMDA) receptors, is a mechanism that causes secondary damage to neurons. The early phase of injury causes loss of dendritic spines and changes to synaptic activity. The phosphatidylinositol-4,5-bisphosphate 3-kinase/Akt/ mammalian target of rapamycin (PI3K/Akt/mTOR) pathway has been implicated in the modulation and regulation of synaptic strength, activity, maturation, and axonal regeneration. The present study focuses on the physiology and survival of neurons following manipulation of Akt and several downstream targets, such as GSK3β, FOXO1, and mTORC1, prior to NMDA-induced injury. Our analysis reveals that exposure to sublethal levels of NMDA does not alter phosphorylation of Akt, S6, and GSK3β at two and twenty four hours following injury. Electrophysiological recordings show that NMDA-induced injury causes a significant decrease in spontaneous excitatory postsynaptic currents at both two and twenty four hours, and this phenotype can be prevented by inhibiting mTORC1 or GSK3β, but not Akt. Additionally, inhibition of mTORC1 or GSK3β promotes neuronal survival following NMDA-induced injury. Thus, NMDA-induced excitotoxicity involves a mechanism that requires the permissive activity of mTORC1 and GSK3β, demonstrating the importance of these kinases in the neuronal response to injury.
Collapse
Affiliation(s)
- Przemyslaw Swiatkowski
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, New Jersey, 08854-8082, USA.,Graduate Program in Molecular Biosciences, Rutgers University, 604 Allison Road, Piscataway, New Jersey, 08854-8082, USA
| | - Ina Nikolaeva
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, New Jersey, 08854-8082, USA.,Graduate Program in Molecular Biosciences, Rutgers University, 604 Allison Road, Piscataway, New Jersey, 08854-8082, USA
| | - Gaurav Kumar
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, New Jersey, 08854-8082, USA
| | - Avery Zucco
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, New Jersey, 08854-8082, USA.,Graduate Program in Neurosciences, Rutgers University, 604 Allison Road, Piscataway, New Jersey, 08854-8082, USA
| | - Barbara F Akum
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, New Jersey, 08854-8082, USA
| | - Mihir V Patel
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, New Jersey, 08854-8082, USA.,Graduate Program in Neurosciences, Rutgers University, 604 Allison Road, Piscataway, New Jersey, 08854-8082, USA
| | - Gabriella D'Arcangelo
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, New Jersey, 08854-8082, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, New Jersey, 08854-8082, USA.
| |
Collapse
|
32
|
Wu D, Bacaj T, Morishita W, Goswami D, Arendt KL, Xu W, Chen L, Malenka RC, Südhof TC. Postsynaptic synaptotagmins mediate AMPA receptor exocytosis during LTP. Nature 2017; 544:316-321. [PMID: 28355182 PMCID: PMC5734942 DOI: 10.1038/nature21720] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 02/22/2017] [Indexed: 12/19/2022]
Abstract
Strengthening of synaptic connections by NMDA-receptor-dependent long-term potentiation (LTP) shapes neural circuits and mediates learning and memory. During NMDA-receptor-dependent LTP induction, Ca2+-influx stimulates recruitment of synaptic AMPA-receptors, thereby strengthening synapses. How Ca2+ induces AMPA-receptor recruitment, however, remains unclear. Here we show that, in pyramidal neurons of the hippocampal CA1-region, blocking postsynaptic expression of both synaptotagmin-1 and synaptotagmin-7, but not of synaptotagmin-1 or synaptotagmin-7 alone, abolished LTP. LTP was rescued by wild-type but not by Ca2+-binding-deficient mutant synaptotagmin-7. Blocking postsynaptic synaptotagmin-1/7 expression did not impair basal synaptic transmission, synaptic or extrasynaptic AMPA-receptor levels, or other AMPA-receptor trafficking events. Moreover, expression of dominant-negative mutant synaptotagmin-1 that inhibited Ca2+-dependent presynaptic vesicle exocytosis also blocked Ca2+-dependent postsynaptic AMPA-receptor exocytosis, thereby abolishing LTP. Our results suggest that postsynaptic synaptotagmin-1 and synaptotagmin-7 act as redundant Ca2+-sensors for Ca2+-dependent exocytosis of AMPA-receptors during LTP, thus delineating a simple mechanism for the recruitment of AMPA-receptors that mediates LTP.
Collapse
Affiliation(s)
- Dick Wu
- Department of Molecular &Cellular Physiology and Howard Hughes Medical Institute, Stanford University Medical School, Stanford, California 94305, USA.,Nancy Pritzker Laboratory, Stanford University Medical School, Stanford, California 94305, USA.,Department of Psychiatry &Behavioral Sciences, Stanford University Medical School, Stanford, California 94305, USA
| | - Taulant Bacaj
- Department of Molecular &Cellular Physiology and Howard Hughes Medical Institute, Stanford University Medical School, Stanford, California 94305, USA
| | - Wade Morishita
- Nancy Pritzker Laboratory, Stanford University Medical School, Stanford, California 94305, USA.,Department of Psychiatry &Behavioral Sciences, Stanford University Medical School, Stanford, California 94305, USA
| | - Debanjan Goswami
- Nancy Pritzker Laboratory, Stanford University Medical School, Stanford, California 94305, USA.,Department of Psychiatry &Behavioral Sciences, Stanford University Medical School, Stanford, California 94305, USA
| | - Kristin L Arendt
- Department of Neurosurgery, Stanford University Medical School, Stanford, California 94305, USA
| | - Wei Xu
- Department of Molecular &Cellular Physiology and Howard Hughes Medical Institute, Stanford University Medical School, Stanford, California 94305, USA
| | - Lu Chen
- Department of Neurosurgery, Stanford University Medical School, Stanford, California 94305, USA
| | - Robert C Malenka
- Nancy Pritzker Laboratory, Stanford University Medical School, Stanford, California 94305, USA.,Department of Psychiatry &Behavioral Sciences, Stanford University Medical School, Stanford, California 94305, USA
| | - Thomas C Südhof
- Department of Molecular &Cellular Physiology and Howard Hughes Medical Institute, Stanford University Medical School, Stanford, California 94305, USA
| |
Collapse
|
33
|
Lipstein N, Göth M, Piotrowski C, Pagel K, Sinz A, Jahn O. Presynaptic Calmodulin targets: lessons from structural proteomics. Expert Rev Proteomics 2017; 14:223-242. [DOI: 10.1080/14789450.2017.1275966] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Noa Lipstein
- Department of Molecular Neurobiology, Max-Planck-Institute of Experimental Medicine, Göttingen, Germany
| | - Melanie Göth
- Institute of Chemistry and Biochemistry, Free University Berlin, Berlin & Fritz Haber Institute of the Max-Planck-Society, Berlin, Germany
| | - Christine Piotrowski
- Department of Pharmaceutical Chemistry & Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Kevin Pagel
- Institute of Chemistry and Biochemistry, Free University Berlin, Berlin & Fritz Haber Institute of the Max-Planck-Society, Berlin, Germany
| | - Andrea Sinz
- Department of Pharmaceutical Chemistry & Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Olaf Jahn
- Proteomics Group, Max-Planck-Institute of Experimental Medicine, Göttingen, Germany
| |
Collapse
|
34
|
Presynaptic Neuronal Pentraxin Receptor Organizes Excitatory and Inhibitory Synapses. J Neurosci 2016; 37:1062-1080. [PMID: 27986928 DOI: 10.1523/jneurosci.2768-16.2016] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/23/2016] [Accepted: 12/12/2016] [Indexed: 01/05/2023] Open
Abstract
Three neuronal pentraxins are expressed in brain, the membrane-bound "neuronal pentraxin receptor" (NPR) and the secreted proteins NP1 and NARP (i.e., NP2). Neuronal pentraxins bind to AMPARs at excitatory synapses and play important, well-documented roles in the activity-dependent regulation of neural circuits via this binding activity. However, it is unknown whether neuronal pentraxins perform roles in synapses beyond modulating postsynaptic AMPAR-dependent plasticity, and whether they may even act in inhibitory synapses. Here, we show that NPR expressed in non-neuronal cells potently induces formation of both excitatory and inhibitory postsynaptic specializations in cocultured hippocampal neurons. Knockdown of NPR in hippocampal neurons, conversely, dramatically decreased assembly and function of both excitatory and inhibitory postsynaptic specializations. Overexpression of NPR rescued the NPR knockdown phenotype but did not in itself change synapse numbers or properties. However, the NPR knockdown decreased the levels of NARP, whereas NPR overexpression produced a dramatic increase in the levels of NP1 and NARP, suggesting that NPR recruits and stabilizes NP1 and NARP on the presynaptic plasma membrane. Mechanistically, NPR acted in excitatory synapse assembly by binding to the N-terminal domain of AMPARs; antagonists of AMPA and GABA receptors selectively inhibited NPR-induced heterologous excitatory and inhibitory synapse assembly, respectively, but did not affect neurexin-1β-induced synapse assembly as a control. Our data suggest that neuronal pentraxins act as signaling complexes that function as general trans-synaptic organizers of both excitatory and inhibitory synapses by a mechanism that depends, at least in part, on the activity of the neurotransmitter receptors at these synapses. SIGNIFICANCE STATEMENT Neuronal pentraxins comprise three neuronal proteins, neuronal pentraxin receptor (NPR) which is a type-II transmembrane protein on the neuronal surface, and secreted neuronal pentraxin-1 and NARP. The general functions of neuronal pentraxins at synapses have not been explored, except for their basic AMPAR binding properties. Here, we examined the functional role of NPR at synapses because it is the only neuronal pentraxin that is anchored to the neuronal cell-surface membrane. We find that NPR is a potent inducer of both excitatory and inhibitory heterologous synapses, and that knockdown of NPR in cultured neurons decreases the density of both excitatory and inhibitory synapses. Our data suggest that NPR performs a general, previously unrecognized function as a universal organizer of synapses.
Collapse
|
35
|
Yi F, Danko T, Botelho SC, Patzke C, Pak C, Wernig M, Südhof TC. Autism-associated SHANK3 haploinsufficiency causes Ih channelopathy in human neurons. Science 2016; 352:aaf2669. [PMID: 26966193 DOI: 10.1126/science.aaf2669] [Citation(s) in RCA: 222] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 02/26/2016] [Indexed: 12/13/2022]
Abstract
Heterozygous SHANK3 mutations are associated with idiopathic autism and Phelan-McDermid syndrome. SHANK3 is a ubiquitously expressed scaffolding protein that is enriched in postsynaptic excitatory synapses. Here, we used engineered conditional mutations in human neurons and found that heterozygous and homozygous SHANK3 mutations severely and specifically impaired hyperpolarization-activated cation (Ih) channels. SHANK3 mutations caused alterations in neuronal morphology and synaptic connectivity; chronic pharmacological blockage of Ih channels reproduced these phenotypes, suggesting that they may be secondary to Ih-channel impairment. Moreover, mouse Shank3-deficient neurons also exhibited severe decreases in Ih currents. SHANK3 protein interacted with hyperpolarization-activated cyclic nucleotide-gated channel proteins (HCN proteins) that form Ih channels, indicating that SHANK3 functions to organize HCN channels. Our data suggest that SHANK3 mutations predispose to autism, at least partially, by inducing an Ih channelopathy that may be amenable to pharmacological intervention.
Collapse
Affiliation(s)
- Fei Yi
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Tamas Danko
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA. Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA. Department of Pathology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Salome Calado Botelho
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Christopher Patzke
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - ChangHui Pak
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA. Department of Pathology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA. Howard Hughes Medical Institute, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA.
| |
Collapse
|
36
|
Körber C, Kuner T. Molecular Machines Regulating the Release Probability of Synaptic Vesicles at the Active Zone. Front Synaptic Neurosci 2016; 8:5. [PMID: 26973506 PMCID: PMC4773589 DOI: 10.3389/fnsyn.2016.00005] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/17/2016] [Indexed: 11/13/2022] Open
Abstract
The fusion of synaptic vesicles (SVs) with the plasma membrane of the active zone (AZ) upon arrival of an action potential (AP) at the presynaptic compartment is a tightly regulated probabilistic process crucial for information transfer. The probability of a SV to release its transmitter content in response to an AP, termed release probability (Pr), is highly diverse both at the level of entire synapses and individual SVs at a given synapse. Differences in Pr exist between different types of synapses, between synapses of the same type, synapses originating from the same axon and even between different SV subpopulations within the same presynaptic terminal. The Pr of SVs at the AZ is set by a complex interplay of different presynaptic properties including the availability of release-ready SVs, the location of the SVs relative to the voltage-gated calcium channels (VGCCs) at the AZ, the magnitude of calcium influx upon arrival of the AP, the buffering of calcium ions as well as the identity and sensitivity of the calcium sensor. These properties are not only interconnected, but can also be regulated dynamically to match the requirements of activity patterns mediated by the synapse. Here, we review recent advances in identifying molecules and molecular machines taking part in the determination of vesicular Pr at the AZ.
Collapse
Affiliation(s)
- Christoph Körber
- Department of Functional Neuroanatomy, Institute of Anatomy and Cell Biology, Heidelberg University Heidelberg, Germany
| | - Thomas Kuner
- Department of Functional Neuroanatomy, Institute of Anatomy and Cell Biology, Heidelberg University Heidelberg, Germany
| |
Collapse
|
37
|
Takarada T, Kou M, Hida M, Fukumori R, Nakamura S, Kutsukake T, Kuramoto N, Hinoi E, Yoneda Y. Protective upregulation of activating transcription factor-3 against glutamate neurotoxicity in neuronal cells under ischemia. J Neurosci Res 2016; 94:378-88. [PMID: 26900013 DOI: 10.1002/jnr.23723] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 01/18/2016] [Accepted: 01/28/2016] [Indexed: 01/05/2023]
Abstract
This study evaluates the pathological role of the stress sensor activating transcription factor-3 (ATF3) in ischemic neurotoxicity. Upregulation of the transcript and protein for ATF3 was seen 2-10 hr after reperfusion in the ipsilateral cerebral hemisphere of mice with transient middle cerebral artery occlusion for 2 hr. Immunohistochemical analysis confirmed the expression of ATF3 by cells immunoreactive for a neuronal marker in neocortex, hippocampus, and striatum within 2 hr after reperfusion. In murine neocortical neurons previously cultured under ischemic conditions for 2 hr, transient upregulation of both Atf3 and ATF3 expression was similarly found during subsequent culture for 2-24 hr under normoxia. Lentiviral overexpression of ATF3 ameliorated the neurotoxicity of glutamate (Glu) in cultured murine neurons along with a slight but statistically significant inhibition of both Fluo-3 and rhodamine-2 fluorescence increases by N-methyl-D-aspartate. Similarly, transient upregulation was seen in Atf3 and ATF3 expression during the culture for 48 hr in neuronal Neuro2A cells previously cultured under ischemic conditions for 2 hr. Luciferase reporter analysis with ATF3 promoter together with immunoblotting revealed the possible involvement of several transcription factors responsive to extracellular and intracellular stressors in the transactivation of the Atf3 gene in Neuro2A cells. ATF3 could be upregulated to play a role in mechanisms underlying mitigation of the neurotoxicity mediated by the endogenous neurotoxin Glu at an early stage after ischemic signal inputs.
Collapse
Affiliation(s)
- Takeshi Takarada
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Medical, Pharmaceutical, and Health Sciences, Kanazawa, Ishikawa, Japan
| | - Miki Kou
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Medical, Pharmaceutical, and Health Sciences, Kanazawa, Ishikawa, Japan
| | - Miho Hida
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Medical, Pharmaceutical, and Health Sciences, Kanazawa, Ishikawa, Japan
| | - Ryo Fukumori
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Medical, Pharmaceutical, and Health Sciences, Kanazawa, Ishikawa, Japan
| | - Saki Nakamura
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Medical, Pharmaceutical, and Health Sciences, Kanazawa, Ishikawa, Japan
| | - Takaya Kutsukake
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Medical, Pharmaceutical, and Health Sciences, Kanazawa, Ishikawa, Japan
| | - Nobuyuki Kuramoto
- Department of Toxicology, Setsunan University, Hirakata, Osaka, Japan
| | - Eiichi Hinoi
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Medical, Pharmaceutical, and Health Sciences, Kanazawa, Ishikawa, Japan
| | - Yukio Yoneda
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Medical, Pharmaceutical, and Health Sciences, Kanazawa, Ishikawa, Japan
| |
Collapse
|
38
|
Nagre N, Wang S, Kellett T, Kanagasabai R, Deng J, Nishi M, Shilo K, Oeckler RA, Yalowich JC, Takeshima H, Christman J, Hubmayr RD, Zhao X. TRIM72 modulates caveolar endocytosis in repair of lung cells. Am J Physiol Lung Cell Mol Physiol 2015; 310:L452-64. [PMID: 26637632 DOI: 10.1152/ajplung.00089.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 12/01/2015] [Indexed: 01/11/2023] Open
Abstract
Alveolar epithelial and endothelial cell injury is a major feature of the acute respiratory distress syndrome, in particular when in conjunction with ventilation therapies. Previously we showed [Kim SC, Kellett T, Wang S, Nishi M, Nagre N, Zhou B, Flodby P, Shilo K, Ghadiali SN, Takeshima H, Hubmayr RD, Zhao X. Am J Physiol Lung Cell Mol Physiol 307: L449-L459, 2014.] that tripartite motif protein 72 (TRIM72) is essential for amending alveolar epithelial cell injury. Here, we posit that TRIM72 improves cellular integrity through its interaction with caveolin 1 (Cav1). Our data show that, in primary type I alveolar epithelial cells, lack of TRIM72 led to significant reduction of Cav1 at the plasma membrane, accompanied by marked attenuation of caveolar endocytosis. Meanwhile, lentivirus-mediated overexpression of TRIM72 selectively increases caveolar endocytosis in rat lung epithelial cells, suggesting a functional association between these two. Further coimmunoprecipitation assays show that deletion of either functional domain of TRIM72, i.e., RING, B-box, coiled-coil, or PRY-SPRY, abolishes the physical interaction between TRIM72 and Cav1, suggesting that all theoretical domains of TRIM72 are required to forge a strong interaction between these two molecules. Moreover, in vivo studies showed that injurious ventilation-induced lung cell death was significantly increased in knockout (KO) TRIM72(KO) and Cav1(KO) lungs compared with wild-type controls and was particularly pronounced in double KO mutants. Apoptosis was accompanied by accentuation of gross lung injury manifestations in the TRIM72(KO) and Cav1(KO) mice. Our data show that TRIM72 directly and indirectly modulates caveolar endocytosis, an essential process involved in repair of lung epithelial cells through removal of plasma membrane wounds. Given TRIM72's role in endomembrane trafficking and cell repair, we consider this molecule an attractive therapeutic target for patients with injured lungs.
Collapse
Affiliation(s)
- Nagaraja Nagre
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia; Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Shaohua Wang
- Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Thomas Kellett
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Ragu Kanagasabai
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Jing Deng
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Miyuki Nishi
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan; and
| | - Konstantin Shilo
- Division of Pulmonary Pathology, Department of Pathology, College of Medicine, The Ohio State University, Columbus, Ohio
| | | | - Jack C Yalowich
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Hiroshi Takeshima
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan; and
| | - John Christman
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Rolf D Hubmayr
- Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Xiaoli Zhao
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia; Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio; Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, College of Medicine, The Ohio State University, Columbus, Ohio;
| |
Collapse
|
39
|
Kulkarni C, Lo M, Fraseur JG, Tirrell DA, Kinzer-Ursem TL. Bioorthogonal Chemoenzymatic Functionalization of Calmodulin for Bioconjugation Applications. Bioconjug Chem 2015; 26:2153-60. [PMID: 26431265 DOI: 10.1021/acs.bioconjchem.5b00449] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Calmodulin (CaM) is a widely studied Ca(2+)-binding protein that is highly conserved across species and involved in many biological processes, including vesicle release, cell proliferation, and apoptosis. To facilitate biophysical studies of CaM, researchers have tagged and mutated CaM at various sites, enabling its conjugation to fluorophores, microarrays, and other reactive partners. However, previous attempts to add a reactive label to CaM for downstream studies have generally employed nonselective labeling methods or resulted in diminished CaM function. Here we report the first engineered CaM protein that undergoes site-specific and bioorthogonal labeling while retaining wild-type activity levels. By employing a chemoenzymatic labeling approach, we achieved selective and quantitative labeling of the engineered CaM protein with an N-terminal 12-azidododecanoic acid tag; notably, addition of the tag did not interfere with the ability of CaM to bind Ca(2+) or a partner protein. The specificity of our chemoenzymatic labeling approach also allowed for selective conjugation of CaM to reactive partners in bacterial cell lysates, without intermediate purification of the engineered protein. Additionally, we prepared CaM-affinity resins that were highly effective in purifying a representative CaM-binding protein, demonstrating that the engineered CaM remains active even after surface capture. Beyond studies of CaM and CaM-binding proteins, the protein engineering and surface capture methods described here should be translatable to other proteins and other bioconjugation applications.
Collapse
Affiliation(s)
- Chethana Kulkarni
- Division of Chemistry and Chemical Engineering, California Institute of Technology , 1200 East California Blvd., Pasadena, California 91125, United States
| | - Megan Lo
- Division of Chemistry and Chemical Engineering, California Institute of Technology , 1200 East California Blvd., Pasadena, California 91125, United States
| | - Julia G Fraseur
- Weldon School of Biomedical Engineering, Purdue University , 206 South Martin Jischke Drive, West Lafayette, Indiana 47907, United States
| | - David A Tirrell
- Division of Chemistry and Chemical Engineering, California Institute of Technology , 1200 East California Blvd., Pasadena, California 91125, United States
| | - Tamara L Kinzer-Ursem
- Division of Chemistry and Chemical Engineering, California Institute of Technology , 1200 East California Blvd., Pasadena, California 91125, United States.,Weldon School of Biomedical Engineering, Purdue University , 206 South Martin Jischke Drive, West Lafayette, Indiana 47907, United States
| |
Collapse
|
40
|
Nakazato R, Takarada T, Ikeno S, Nakamura S, Kutsukake T, Hinoi E, Yoneda Y. Upregulation of Runt-Related Transcription Factor-2 Through CCAAT Enhancer Binding Protein-β Signaling Pathway in Microglial BV-2 Cells Exposed to ATP. J Cell Physiol 2015; 230:2510-21. [PMID: 25802132 DOI: 10.1002/jcp.24988] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 03/10/2015] [Indexed: 01/01/2023]
Abstract
We have shown constitutive expression of the master regulator of osteoblastogenesis, runt-related transcription factor-2 (Runx2), by microglia cells outside bone. Here, we attempted to evaluate the pathological significance of Runx2 in microglial BV-2 cells exposed to ATP at a high concentration. Marked upregulation of Runx2 transcript and protein expression was seen in cells exposed to 1 mM ATP for a period longer than 30 min without inducing cytotoxicity. The Runx2 upregulation by ATP was prevented by extracellular and intracellular Ca(2+) chelators, while thapsigargin upregulated Runx2 expression alone without affecting the upregulation by ATP. A calmodulin antagonist prevented the upregulation by ATP, with calcineurin inhibitors being ineffective. Although ATP markedly increased nuclear levels of nuclear factor of activated T cell-2 (NFAT2), Runx2 promoter activity was not simulated by the introduction of either NFAT1 or NFAT2, but facilitated by that of CCAAT enhancer binding protein-α (C/EBPα), C/EBPβ and nuclear factor (erythroid-derived 2)-like-2 (Nrf2). Exposure to ATP up-regulated C/EBPβ and Nrf2, but not C/EBPα, expression, in addition to increasing nuclear levels of respective corresponding proteins. Runx2 upregulation by ATP was deteriorated by knockdown of C/EBPβ but not by that of Nrf2, however, while exposure to ATP up-regulated matrix metalloproteinase-13 (Mmp13) expression in a Runx2-dependent manner. Overexpression of Runx2 up-regulated Mmp13 expression with promoted incorporation of fluorescent beads into BV-2 cells without ATP. These results suggest that extracellular ATP up-regulates Runx2 expression through activation of the C/EBPβ signaling in a calmodulin-dependent manner to play a pivotal role in phagocytosis in microglial BV-2 cells.
Collapse
Affiliation(s)
- Ryota Nakazato
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Medical, Pharmaceutical and Health Sciences, Kanazawa, Japan
| | - Takeshi Takarada
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Medical, Pharmaceutical and Health Sciences, Kanazawa, Japan
| | - Shinsuke Ikeno
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Medical, Pharmaceutical and Health Sciences, Kanazawa, Japan
| | - Saki Nakamura
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Medical, Pharmaceutical and Health Sciences, Kanazawa, Japan
| | - Takaya Kutsukake
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Medical, Pharmaceutical and Health Sciences, Kanazawa, Japan
| | - Eiichi Hinoi
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Medical, Pharmaceutical and Health Sciences, Kanazawa, Japan
| | - Yukio Yoneda
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Medical, Pharmaceutical and Health Sciences, Kanazawa, Japan
| |
Collapse
|
41
|
Abstract
Postsynaptic AMPA-type glutamate receptors (AMPARs) are among the major determinants of synaptic strength and can be trafficked into and out of synapses. Neuronal activity regulates AMPAR trafficking during synaptic plasticity to induce long-term changes in synaptic strength, including long-term potentiation (LTP) and long-term depression (LTD). Rab family GTPases regulate most membrane trafficking in eukaryotic cells; particularly, Rab11 and its effectors are implicated in mediating postsynaptic AMPAR insertion during LTP. To explore the synaptic function of Rab11Fip5, a neuronal Rab11 effector and a candidate autism-spectrum disorder gene, we performed shRNA-mediated knock-down and genetic knock-out (KO) studies. Surprisingly, we observed robust shRNA-induced synaptic phenotypes that were rescued by a Rab11Fip5 cDNA but that were nevertheless not observed in conditional KO neurons. Both in cultured neurons and acute slices, KO of Rab11Fip5 had no significant effect on basic parameters of synaptic transmission, indicating that Rab11Fip5 is not required for fundamental synaptic operations, such as neurotransmitter release or postsynaptic AMPAR insertion. KO of Rab11Fip5 did, however, abolish hippocampal LTD as measured both in acute slices or using a chemical LTD protocol in cultured neurons but did not affect hippocampal LTP. The Rab11Fip5 KO mice performed normally in several behavioral tasks, including fear conditioning, but showed enhanced contextual fear extinction. These are the first findings to suggest a requirement for Rab11Fip5, and presumably Rab11, during LTD.
Collapse
|
42
|
Körber C, Horstmann H, Venkataramani V, Herrmannsdörfer F, Kremer T, Kaiser M, Schwenger DB, Ahmed S, Dean C, Dresbach T, Kuner T. Modulation of Presynaptic Release Probability by the Vertebrate-Specific Protein Mover. Neuron 2015. [PMID: 26212709 DOI: 10.1016/j.neuron.2015.07.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Mover, a member of the exquisitely small group of vertebrate-specific presynaptic proteins, has been discovered as an interaction partner of the scaffolding protein Bassoon, yet its function has not been elucidated. We used adeno-associated virus (AAV)-mediated shRNA expression to knock down Mover in the calyx of Held in vivo. Although spontaneous synaptic transmission remained unaffected, we found a strong increase of the evoked EPSC amplitude. The size of the readily releasable pool was unaltered, but short-term depression was accelerated and enhanced, consistent with an increase in release probability after Mover knockdown. This increase in release probability was not caused by alterations in Ca(2+) influx but rather by a higher Ca(2+) sensitivity of the release machinery, as demonstrated by presynaptic Ca(2+) uncaging. We therefore conclude that Mover expression in certain subsets of synapses negatively regulates synaptic release probability, constituting a novel mechanism to tune synaptic transmission.
Collapse
Affiliation(s)
- Christoph Körber
- Institute of Anatomy and Cell Biology, Heidelberg University, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany.
| | - Heinz Horstmann
- Institute of Anatomy and Cell Biology, Heidelberg University, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | - Varun Venkataramani
- Institute of Anatomy and Cell Biology, Heidelberg University, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | - Frank Herrmannsdörfer
- Institute of Anatomy and Cell Biology, Heidelberg University, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | - Thomas Kremer
- Institute of Anatomy and Cell Biology, Heidelberg University, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | - Michaela Kaiser
- Institute of Anatomy and Cell Biology, Heidelberg University, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | - Darius B Schwenger
- Institute of Anatomy and Cell Biology, Heidelberg University, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | - Saheeb Ahmed
- European Neuroscience Institute, Grisebachstrasse 5, 37077 Göttingen, Germany
| | - Camin Dean
- European Neuroscience Institute, Grisebachstrasse 5, 37077 Göttingen, Germany
| | - Thomas Dresbach
- Institute of Anatomy and Cell Biology, Heidelberg University, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany; Department of Anatomy and Embryology, Centre of Anatomy, University of Göttingen, Kreuzbergring 36, 37075 Göttingen, Germany
| | - Thomas Kuner
- Institute of Anatomy and Cell Biology, Heidelberg University, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany.
| |
Collapse
|
43
|
Timofeeva Y, Volynski KE. Calmodulin as a major calcium buffer shaping vesicular release and short-term synaptic plasticity: facilitation through buffer dislocation. Front Cell Neurosci 2015; 9:239. [PMID: 26190970 PMCID: PMC4486835 DOI: 10.3389/fncel.2015.00239] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 06/12/2015] [Indexed: 12/30/2022] Open
Abstract
Action potential-dependent release of synaptic vesicles and short-term synaptic plasticity are dynamically regulated by the endogenous Ca2+ buffers that shape [Ca2+] profiles within a presynaptic bouton. Calmodulin is one of the most abundant presynaptic proteins and it binds Ca2+ faster than any other characterized endogenous neuronal Ca2+ buffer. Direct effects of calmodulin on fast presynaptic Ca2+ dynamics and vesicular release however have not been studied in detail. Using experimentally constrained three-dimensional diffusion modeling of Ca2+ influx–exocytosis coupling at small excitatory synapses we show that, at physiologically relevant concentrations, Ca2+ buffering by calmodulin plays a dominant role in inhibiting vesicular release and in modulating short-term synaptic plasticity. We also propose a novel and potentially powerful mechanism for short-term facilitation based on Ca2+-dependent dynamic dislocation of calmodulin molecules from the plasma membrane within the active zone.
Collapse
Affiliation(s)
- Yulia Timofeeva
- Department of Computer Science, University of Warwick Coventry, UK ; Centre for Complexity Science, University of Warwick Coventry, UK
| | - Kirill E Volynski
- University College London Institute of Neurology, University College London London, UK
| |
Collapse
|
44
|
Dynamic binding mode of a Synaptotagmin-1-SNARE complex in solution. Nat Struct Mol Biol 2015; 22:555-64. [PMID: 26030874 PMCID: PMC4496268 DOI: 10.1038/nsmb.3035] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 04/27/2015] [Indexed: 12/21/2022]
Abstract
Rapid neurotransmitter release depends on the Ca2+-sensor Synaptotagmin-1 and the SNARE complex formed by synaptobrevin, syntaxin-1 and SNAP-25. How Synaptotagmin-1 triggers release remains unclear, in part because elucidating high-resolution structures of Synaptotagmin-1-SNARE complexes has been challenging. An NMR approach based on lanthanide-induced pseudocontact shifts now reveals a dynamic binding mode where basic residues in the concave side of the Synaptotagmin-1 C2B domain β-sandwich interact with a polyacidic region of the SNARE complex formed by syntaxin-1 and SNAP-25. The physiological relevance of this dynamic structural model is supported by mutations in basic residues of Synaptotagmin-1 that markedly impair SNARE-complex binding in vitro and Synaptotagmin-1 function in neurons. Mutations with milder effects on binding have correspondingly milder effects on Synaptotagmin-1 function. Our results support a model whereby their dynamic interaction facilitates cooperation between synaptotagmin-1 and the SNAREs in inducing membrane fusion.
Collapse
|
45
|
Pandalaneni S, Karuppiah V, Saleem M, Haynes LP, Burgoyne RD, Mayans O, Derrick JP, Lian LY. Neuronal Calcium Sensor-1 Binds the D2 Dopamine Receptor and G-protein-coupled Receptor Kinase 1 (GRK1) Peptides Using Different Modes of Interactions. J Biol Chem 2015; 290:18744-56. [PMID: 25979333 PMCID: PMC4513130 DOI: 10.1074/jbc.m114.627059] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Indexed: 11/25/2022] Open
Abstract
Neuronal calcium sensor-1 (NCS-1) is the primordial member of the neuronal calcium sensor family of EF-hand Ca2+-binding proteins. It interacts with both the G-protein-coupled receptor (GPCR) dopamine D2 receptor (D2R), regulating its internalization and surface expression, and the cognate kinases GRK1 and GRK2. Determination of the crystal structures of Ca2+/NCS-1 alone and in complex with peptides derived from D2R and GRK1 reveals that the differential recognition is facilitated by the conformational flexibility of the C-lobe-binding site. We find that two copies of the D2R peptide bind within the hydrophobic crevice on Ca2+/NCS-1, but only one copy of the GRK1 peptide binds. The different binding modes are made possible by the C-lobe-binding site of NCS-1, which adopts alternative conformations in each complex. C-terminal residues Ser-178–Val-190 act in concert with the flexible EF3/EF4 loop region to effectively form different peptide-binding sites. In the Ca2+/NCS-1·D2R peptide complex, the C-terminal region adopts a 310 helix-turn-310 helix, whereas in the GRK1 peptide complex it forms an α-helix. Removal of Ser-178–Val-190 generated a C-terminal truncation mutant that formed a dimer, indicating that the NCS-1 C-terminal region prevents NCS-1 oligomerization. We propose that the flexible nature of the C-terminal region is essential to allow it to modulate its protein-binding sites and adapt its conformation to accommodate both ligands. This appears to be driven by the variability of the conformation of the C-lobe-binding site, which has ramifications for the target specificity and diversity of NCS-1.
Collapse
Affiliation(s)
- Sravan Pandalaneni
- From the NMR Centre for Structural Biology, Institute of Integrative Biology, and
| | - Vijaykumar Karuppiah
- From the NMR Centre for Structural Biology, Institute of Integrative Biology, and the Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, and
| | - Muhammad Saleem
- the Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, and
| | - Lee P Haynes
- the Physiological Laboratory, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L37 4BY, United Kingdom
| | - Robert D Burgoyne
- the Physiological Laboratory, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L37 4BY, United Kingdom
| | - Olga Mayans
- Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB
| | - Jeremy P Derrick
- the Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, and
| | - Lu-Yun Lian
- From the NMR Centre for Structural Biology, Institute of Integrative Biology, and
| |
Collapse
|
46
|
Yang W, Thein S, Lim CY, Ericksen RE, Sugii S, Xu F, Robinson RC, Kim JB, Han W. Arp2/3 complex regulates adipogenesis by controlling cortical actin remodelling. Biochem J 2014; 464:179-192. [PMID: 25220164 DOI: 10.1042/bj20140805] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Extensive actin cytoskeleton remodelling occurs during adipocyte development. We have previously shown that disruption of stress fibres by the actin-severing protein cofilin is a requisite step in adipogenesis. However, it remains unclear whether actin nucleation and assembly into the cortical structure are essential for adipocyte development. In the present study we investigated the role of cortical actin assembly and of actin nucleation by the actin-related protein 2/3 (Arp2/3) complex in adipogenesis. Cortical actin structure formation started with accumulation of filamentous actin (F-actin) patches near the plasma membrane during adipogenesis. Depletion of Arp2/3 by knockdown of its subunits Arp3 or ARPC3 strongly impaired adipocyte differentiation, although adipogenesis-initiating factors were unaffected. Moreover, the assembly of F-actin-rich structures at the plasma membrane was suppressed and the cortical actin structure poorly developed after adipogenic induction in Arp2/3-deficient cells. Finally, we provide evidence that the cortical actin cytoskeleton is essential for efficient glucose transporter 4 (GLUT4) vesicle exocytosis and insulin signal transduction. These results show that the Arp2/3 complex is an essential regulator of adipocyte development through control of the formation of cortical actin structures, which may facilitate nutrient uptake and signalling events.
Collapse
Affiliation(s)
- Wulin Yang
- *Laboratory of Metabolic Medicine, Singapore Bioimaging Consortium, #02-02 Helios, 11 Biopolis Way, 138667 Singapore
| | - Shermaine Thein
- *Laboratory of Metabolic Medicine, Singapore Bioimaging Consortium, #02-02 Helios, 11 Biopolis Way, 138667 Singapore
| | | | - Russell E Ericksen
- *Laboratory of Metabolic Medicine, Singapore Bioimaging Consortium, #02-02 Helios, 11 Biopolis Way, 138667 Singapore
| | - Shigeki Sugii
- *Laboratory of Metabolic Medicine, Singapore Bioimaging Consortium, #02-02 Helios, 11 Biopolis Way, 138667 Singapore
| | - Feng Xu
- ‡Singapore Institute for Clinical Sciences, 30 Medical Drive, 117609 Singapore
| | | | - Jae Bum Kim
- ║National Creative Research Initiatives Center for Adipose Tissue Remodeling, Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul 151-742, Republic of Korea
| | | |
Collapse
|
47
|
|
48
|
Van Hook MJ, Parmelee CM, Chen M, Cork KM, Curto C, Thoreson WB. Calmodulin enhances ribbon replenishment and shapes filtering of synaptic transmission by cone photoreceptors. ACTA ACUST UNITED AC 2014; 144:357-78. [PMID: 25311636 PMCID: PMC4210432 DOI: 10.1085/jgp.201411229] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
At the first synapse in the vertebrate visual pathway, light-evoked changes in photoreceptor membrane potential alter the rate of glutamate release onto second-order retinal neurons. This process depends on the synaptic ribbon, a specialized structure found at various sensory synapses, to provide a supply of primed vesicles for release. Calcium (Ca(2+)) accelerates the replenishment of vesicles at cone ribbon synapses, but the mechanisms underlying this acceleration and its functional implications for vision are unknown. We studied vesicle replenishment using paired whole-cell recordings of cones and postsynaptic neurons in tiger salamander retinas and found that it involves two kinetic mechanisms, the faster of which was diminished by calmodulin (CaM) inhibitors. We developed an analytical model that can be applied to both conventional and ribbon synapses and showed that vesicle resupply is limited by a simple time constant, τ = 1/(Dρδs), where D is the vesicle diffusion coefficient, δ is the vesicle diameter, ρ is the vesicle density, and s is the probability of vesicle attachment. The combination of electrophysiological measurements, modeling, and total internal reflection fluorescence microscopy of single synaptic vesicles suggested that CaM speeds replenishment by enhancing vesicle attachment to the ribbon. Using electroretinogram and whole-cell recordings of light responses, we found that enhanced replenishment improves the ability of cone synapses to signal darkness after brief flashes of light and enhances the amplitude of responses to higher-frequency stimuli. By accelerating the resupply of vesicles to the ribbon, CaM extends the temporal range of synaptic transmission, allowing cones to transmit higher-frequency visual information to downstream neurons. Thus, the ability of the visual system to encode time-varying stimuli is shaped by the dynamics of vesicle replenishment at photoreceptor synaptic ribbons.
Collapse
Affiliation(s)
- Matthew J Van Hook
- Department of Ophthalmology and Visual Sciences and Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198
| | - Caitlyn M Parmelee
- Department of Mathematics, University of Nebraska-Lincoln, Lincoln, NE 68588
| | - Minghui Chen
- Department of Ophthalmology and Visual Sciences and Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 Department of Ophthalmology and Visual Sciences and Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198
| | - Karlene M Cork
- Department of Ophthalmology and Visual Sciences and Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 Department of Ophthalmology and Visual Sciences and Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198
| | - Carina Curto
- Department of Mathematics, University of Nebraska-Lincoln, Lincoln, NE 68588 Department of Mathematics, The Pennsylvania State University, University Park, State College, PA 16802
| | - Wallace B Thoreson
- Department of Ophthalmology and Visual Sciences and Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 Department of Ophthalmology and Visual Sciences and Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
49
|
Roy B, Ferdous J, Ali DW. NMDA receptors on zebrafish Mauthner cells require CaMKII-α for normal development. Dev Neurobiol 2014; 75:145-62. [PMID: 25047640 DOI: 10.1002/dneu.22214] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 07/15/2014] [Accepted: 07/20/2014] [Indexed: 12/31/2022]
Abstract
Calcium/calmodulin dependent protein kinase 2 (CaMKII) is a multifunctional protein that is highly enriched in the synapse. It plays important roles in neuronal functions such as synaptic plasticity, synaptogenesis, and neural development. Gene duplication in zebrafish has resulted in the occurrence of seven CaMKII genes (camk2a, camk2b1, camk2b2, camk2g1, camk2g2, camk2d1, and camk2d2) that are developmentally expressed. In this study, we used single cell, real-time quantitative PCR to investigate the expression of CaMKII genes in individual Mauthner cells (M-cells) of 2 days post fertilization (dpf) zebrafish embryos. We found that out of seven different CaMKII genes, only the mRNA for CaMKII-α was expressed in the M-cell at detectable levels, while all other isoforms were undetectable. Morpholino knockdown of CaMKII-α had no significant effect on AMPA synaptic currents (mEPSCs) but decreased the amplitude of NMDA mEPSCs. NMDA events exhibited a biexponential decay with τfast ≈ 30 ms and τslow ≈ 300 ms. Knockdown of CaMKII-α specifically reduced the amplitude of the slow component of the NMDA-mediated currents (mEPSCs), without affecting the fast component, the frequency, or the kinetics of the mEPSCs. Immunolabelling of the M-cell showed increased dendritic arborizations in the morphants compared with controls, and knockdown of CaMKII-α altered locomotor behaviors of touch responses. These results suggest that CaMKII-α is present in embryonic M-cells and that it plays a role in the normal development of excitatory synapses. Our findings pave the way for determining the function of specific CaMKII isoforms during the early stages of M-cell development.
Collapse
Affiliation(s)
- Birbickram Roy
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | | | | |
Collapse
|
50
|
Wang D, Epstein D, Khalaf O, Srinivasan S, Williamson WR, Fayyazuddin A, Quiocho FA, Hiesinger PR. Ca2+-Calmodulin regulates SNARE assembly and spontaneous neurotransmitter release via v-ATPase subunit V0a1. ACTA ACUST UNITED AC 2014; 205:21-31. [PMID: 24733584 PMCID: PMC3987144 DOI: 10.1083/jcb.201312109] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Ca2+–Calmodulin binding to neuronal v-ATPase V0 subunit a1 (V100) regulates SNARE complex assembly for a putative subset of synaptic vesicles that sustain spontaneous release in Drosophila. Most chemical neurotransmission occurs through Ca2+-dependent evoked or spontaneous vesicle exocytosis. In both cases, Ca2+ sensing is thought to occur shortly before exocytosis. In this paper, we provide evidence that the Ca2+ dependence of spontaneous vesicle release may partly result from an earlier requirement of Ca2+ for the assembly of soluble N-ethylmaleimide–sensitive fusion attachment protein receptor (SNARE) complexes. We show that the neuronal vacuolar-type H+-adenosine triphosphatase V0 subunit a1 (V100) can regulate the formation of SNARE complexes in a Ca2+–Calmodulin (CaM)-dependent manner. Ca2+–CaM regulation of V100 is not required for vesicle acidification. Specific disruption of the Ca2+-dependent regulation of V100 by CaM led to a >90% loss of spontaneous release but only had a mild effect on evoked release at Drosophila melanogaster embryo neuromuscular junctions. Our data suggest that Ca2+–CaM regulation of V100 may control SNARE complex assembly for a subset of synaptic vesicles that sustain spontaneous release.
Collapse
Affiliation(s)
- Dong Wang
- Department of Physiology and 2 Green Center Division for Systems Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | | | | | | | | | | | | | | |
Collapse
|