1
|
Oestreicher D, Chepurwar S, Kusch K, Rankovic V, Jung S, Strenzke N, Pangrsic T. CaBP1 and 2 enable sustained Ca V1.3 calcium currents and synaptic transmission in inner hair cells. eLife 2024; 13:RP93646. [PMID: 39718549 DOI: 10.7554/elife.93646] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024] Open
Abstract
To encode continuous sound stimuli, the inner hair cell (IHC) ribbon synapses utilize calcium-binding proteins (CaBPs), which reduce the inactivation of their CaV1.3 calcium channels. Mutations in the CABP2 gene underlie non-syndromic autosomal recessive hearing loss DFNB93. Besides CaBP2, the structurally related CaBP1 is highly abundant in the IHCs. Here, we investigated how the two CaBPs cooperatively regulate IHC synaptic function. In Cabp1/2 double-knockout mice, we find strongly enhanced CaV1.3 inactivation, slowed recovery from inactivation and impaired sustained exocytosis. Already mild IHC activation further reduces the availability of channels to trigger synaptic transmission and may effectively silence synapses. Spontaneous and sound-evoked responses of spiral ganglion neurons in vivo are strikingly reduced and strongly depend on stimulation rates. Transgenic expression of CaBP2 leads to substantial recovery of IHC synaptic function and hearing sensitivity. We conclude that CaBP1 and 2 act together to suppress voltage- and calcium-dependent inactivation of IHC CaV1.3 channels in order to support sufficient rate of exocytosis and enable fast, temporally precise and indefatigable sound encoding.
Collapse
Affiliation(s)
- David Oestreicher
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Shashank Chepurwar
- Auditory Systems Physiology Group, Institute for Auditory Neuroscience, InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| | - Kathrin Kusch
- Functional Auditory Genomics, Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Vladan Rankovic
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Restorative Cochlear Genomics Group, Auditory Neuroscience and Optogenetics Laboratory, German Primate Cente, Göttingen, Germany
| | - Sangyong Jung
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Nicola Strenzke
- Auditory Systems Physiology Group, Institute for Auditory Neuroscience, InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| | - Tina Pangrsic
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
2
|
Liu JB, Yuan HL, Zhang G, Ke JB. Comprehensive Characterization of a Subfamily of Ca 2+-Binding Proteins in Mouse and Human Retinal Neurons at Single-Cell Resolution. eNeuro 2024; 11:ENEURO.0145-24.2024. [PMID: 39260891 PMCID: PMC11419601 DOI: 10.1523/eneuro.0145-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/20/2024] [Accepted: 09/03/2024] [Indexed: 09/13/2024] Open
Abstract
Ca2+-binding proteins (CaBPs; CaBP1-5) are a subfamily of neuronal Ca2+ sensors with high homology to calmodulin. Notably, CaBP4, which is exclusively expressed in rod and cone photoreceptors, is crucial for maintaining normal retinal functions. However, the functional roles of CaBP1, CaBP2, and CaBP5 in the retina remain elusive, primarily due to limited understanding of their expression patterns within inner retinal neurons. In this study, we conducted a comprehensive transcript analysis using single-cell RNA sequencing datasets to investigate the gene expression profiles of CaBPs in mouse and human retinal neurons. Our findings revealed notable similarities in the overall expression patterns of CaBPs across both species. Specifically, nearly all amacrine cell, ganglion cell, and horizontal cell types exclusively expressed CaBP1. In contrast, the majority of bipolar cell types, including rod bipolar (RB) cells, expressed distinct combinations of CaBP1, CaBP2, and CaBP5, rather than a single CaBP as previously hypothesized. Remarkably, mouse rods and human cones exclusively expressed CaBP4, whereas mouse cones and human rods coexpressed both CaBP4 and CaBP5. Our single-cell reverse transcription polymerase chain reaction analysis confirmed the coexpression CaBP1 and CaBP5 in individual RBs from mice of either sex. Additionally, all three splice variants of CaBP1, primarily L-CaBP1, were detected in mouse RBs. Taken together, our study offers a comprehensive overview of the distribution of CaBPs in mouse and human retinal neurons, providing valuable insights into their roles in visual functions.
Collapse
Affiliation(s)
- Jun-Bin Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - He-Lan Yuan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Gong Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Jiang-Bin Ke
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou 325000, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| |
Collapse
|
3
|
Lopez JA, Romero LO, Kaung WL, Maddox JW, Vásquez V, Lee A. Caldendrin Is a Repressor of PIEZO2 Channels and Touch Sensation in Mice. J Neurosci 2024; 44:e1402232023. [PMID: 38262725 PMCID: PMC10919251 DOI: 10.1523/jneurosci.1402-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/25/2024] Open
Abstract
The sense of touch is crucial for cognitive, emotional, and social development and relies on mechanically activated (MA) ion channels that transduce force into an electrical signal. Despite advances in the molecular characterization of these channels, the physiological factors that control their activity are poorly understood. Here, we used behavioral assays, electrophysiological recordings, and various mouse strains (males and females analyzed separately) to investigate the role of the calmodulin-like Ca2+ sensor, caldendrin, as a key regulator of MA channels and their roles in touch sensation. In mice lacking caldendrin (Cabp1 KO), heightened responses to tactile stimuli correlate with enlarged MA currents with lower mechanical thresholds in dorsal root ganglion neurons (DRGNs). The expression pattern of caldendrin in the DRG parallels that of the major MA channel required for touch sensation, PIEZO2. In transfected cells, caldendrin interacts with and inhibits the activity of PIEZO2 in a manner that requires an alternatively spliced sequence in the N-terminal domain of caldendrin. Moreover, targeted genetic deletion of caldendrin in Piezo2-expressing DRGNs phenocopies the tactile hypersensitivity of complete Cabp1 KO mice. We conclude that caldendrin is an endogenous repressor of PIEZO2 channels and their contributions to touch sensation in DRGNs.
Collapse
Affiliation(s)
- Josue A Lopez
- Department of Neuroscience and Center for Learning and Memory, University of Texas-Austin, Austin 78712, Texas
| | - Luis O Romero
- Department of Physiology, The University of Tennessee Health Science Center, Memphis 38163, Tennessee
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, Memphis 38163, Tennessee
| | - Wai-Lin Kaung
- Department of Neuroscience and Center for Learning and Memory, University of Texas-Austin, Austin 78712, Texas
| | - J Wesley Maddox
- Department of Neuroscience and Center for Learning and Memory, University of Texas-Austin, Austin 78712, Texas
| | - Valeria Vásquez
- Department of Physiology, The University of Tennessee Health Science Center, Memphis 38163, Tennessee
| | - Amy Lee
- Department of Neuroscience and Center for Learning and Memory, University of Texas-Austin, Austin 78712, Texas
| |
Collapse
|
4
|
Lisek M, Tomczak J, Boczek T, Zylinska L. Calcium-Associated Proteins in Neuroregeneration. Biomolecules 2024; 14:183. [PMID: 38397420 PMCID: PMC10887043 DOI: 10.3390/biom14020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/27/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
The dysregulation of intracellular calcium levels is a critical factor in neurodegeneration, leading to the aberrant activation of calcium-dependent processes and, ultimately, cell death. Ca2+ signals vary in magnitude, duration, and the type of neuron affected. A moderate Ca2+ concentration can initiate certain cellular repair pathways and promote neuroregeneration. While the peripheral nervous system exhibits an intrinsic regenerative capability, the central nervous system has limited self-repair potential. There is evidence that significant variations exist in evoked calcium responses and axonal regeneration among neurons, and individual differences in regenerative capacity are apparent even within the same type of neurons. Furthermore, some studies have shown that neuronal activity could serve as a potent regulator of this process. The spatio-temporal patterns of calcium dynamics are intricately controlled by a variety of proteins, including channels, ion pumps, enzymes, and various calcium-binding proteins, each of which can exert either positive or negative effects on neural repair, depending on the cellular context. In this concise review, we focus on several calcium-associated proteins such as CaM kinase II, GAP-43, oncomodulin, caldendrin, calneuron, and NCS-1 in order to elaborate on their roles in the intrinsic mechanisms governing neuronal regeneration following traumatic damage processes.
Collapse
Affiliation(s)
| | | | | | - Ludmila Zylinska
- Department of Molecular Neurochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (M.L.); (J.T.); (T.B.)
| |
Collapse
|
5
|
Vecchi JT, Mullan S, Lopez JA, Rhomberg M, Yamamoto A, Hallam A, Lee A, Sonka M, Hansen MR. Sensitivity of CNN image analysis to multifaceted measurements of neurite growth. BMC Bioinformatics 2023; 24:320. [PMID: 37620759 PMCID: PMC10464248 DOI: 10.1186/s12859-023-05444-4] [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: 12/14/2022] [Accepted: 08/11/2023] [Indexed: 08/26/2023] Open
Abstract
Quantitative analysis of neurite growth and morphology is essential for understanding the determinants of neural development and regeneration, however, it is complicated by the labor-intensive process of measuring diverse parameters of neurite outgrowth. Consequently, automated approaches have been developed to study neurite morphology in a high-throughput and comprehensive manner. These approaches include computer-automated algorithms known as 'convolutional neural networks' (CNNs)-powerful models capable of learning complex tasks without the biases of hand-crafted models. Nevertheless, their complexity often relegates them to functioning as 'black boxes.' Therefore, research in the field of explainable AI is imperative to comprehend the relationship between CNN image analysis output and predefined morphological parameters of neurite growth in order to assess the applicability of these machine learning approaches. In this study, drawing inspiration from the field of automated feature selection, we investigate the correlation between quantified metrics of neurite morphology and the image analysis results from NeuriteNet-a CNN developed to analyze neurite growth. NeuriteNet accurately distinguishes images of neurite growth based on different treatment groups within two separate experimental systems. These systems differentiate between neurons cultured on different substrate conditions and neurons subjected to drug treatment inhibiting neurite outgrowth. By examining the model's function and patterns of activation underlying its classification decisions, we discover that NeuriteNet focuses on aspects of neuron morphology that represent quantifiable metrics distinguishing these groups. Additionally, it incorporates factors that are not encompassed by neuron morphology tracing analyses. NeuriteNet presents a novel tool ideally suited for screening morphological differences in heterogeneous neuron groups while also providing impetus for targeted follow-up studies.
Collapse
Affiliation(s)
- Joseph T Vecchi
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Sean Mullan
- Iowa Institute for Biomedical Imaging, Electrical and Computer Engineering, University of Iowa, Iowa City, IA, USA
| | - Josue A Lopez
- Department of Neuroscience, University of Texas-Austin, Austin, TX, USA
| | - Madeline Rhomberg
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | | | - Annabelle Hallam
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Amy Lee
- Department of Neuroscience, University of Texas-Austin, Austin, TX, USA
| | - Milan Sonka
- Iowa Institute for Biomedical Imaging, Electrical and Computer Engineering, University of Iowa, Iowa City, IA, USA
| | - Marlan R Hansen
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
6
|
Lopez JA, Yamamoto A, Vecchi JT, Hagen J, Lee K, Sonka M, Hansen MR, Lee A. Caldendrin represses neurite regeneration and growth in dorsal root ganglion neurons. Sci Rep 2023; 13:2608. [PMID: 36788334 PMCID: PMC9929226 DOI: 10.1038/s41598-023-29622-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
Caldendrin is a Ca2+ binding protein that interacts with multiple effectors, such as the Cav1 L-type Ca2+ channel, which play a prominent role in regulating the outgrowth of dendrites and axons (i.e., neurites) during development and in response to injury. Here, we investigated the role of caldendrin in Cav1-dependent pathways that impinge upon neurite growth in dorsal root ganglion neurons (DRGNs). By immunofluorescence, caldendrin was localized in medium- and large- diameter DRGNs. Compared to DRGNs cultured from WT mice, DRGNs of caldendrin knockout (KO) mice exhibited enhanced neurite regeneration and outgrowth. Strong depolarization, which normally represses neurite growth through activation of Cav1 channels, had no effect on neurite growth in DRGN cultures from female caldendrin KO mice. Remarkably, DRGNs from caldendrin KO males were no different from those of WT males in terms of depolarization-dependent neurite growth repression. We conclude that caldendrin opposes neurite regeneration and growth, and this involves coupling of Cav1 channels to growth-inhibitory pathways in DRGNs of females but not males.
Collapse
Affiliation(s)
- Josue A Lopez
- Department of Neuroscience, University of Texas-Austin, 100 E. 24th St., Austin, TX, 78712, USA
| | - Annamarie Yamamoto
- Department of Neuroscience, University of Texas-Austin, 100 E. 24th St., Austin, TX, 78712, USA
| | - Joseph T Vecchi
- Department of Molecular Physiology and Biophysics and Otolaryngology Head-Neck Surgery, Iowa Neuroscience Institute, Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Jussara Hagen
- Department of Molecular Physiology and Biophysics and Otolaryngology Head-Neck Surgery, Iowa Neuroscience Institute, Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Kyungmoo Lee
- Electrical and Computer Engineering, Iowa Institute for Biomedical Imaging, University of Iowa, 51 Newton Rd. Iowa City, Iowa, 52242, USA
| | - Milan Sonka
- Electrical and Computer Engineering, Iowa Institute for Biomedical Imaging, University of Iowa, 51 Newton Rd. Iowa City, Iowa, 52242, USA
| | - Marlan R Hansen
- Department of Molecular Physiology and Biophysics and Otolaryngology Head-Neck Surgery, Iowa Neuroscience Institute, Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Amy Lee
- Department of Neuroscience, University of Texas-Austin, 100 E. 24th St., Austin, TX, 78712, USA.
| |
Collapse
|
7
|
Konietzny A, Grendel J, Kadek A, Bucher M, Han Y, Hertrich N, Dekkers DHW, Demmers JAA, Grünewald K, Uetrecht C, Mikhaylova M. Caldendrin and myosin V regulate synaptic spine apparatus localization via ER stabilization in dendritic spines. EMBO J 2022; 41:e106523. [PMID: 34935159 PMCID: PMC8844991 DOI: 10.15252/embj.2020106523] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/08/2021] [Accepted: 11/19/2021] [Indexed: 11/21/2022] Open
Abstract
Excitatory synapses of principal hippocampal neurons are frequently located on dendritic spines. The dynamic strengthening or weakening of individual inputs results in structural and molecular diversity of dendritic spines. Active spines with large calcium ion (Ca2+ ) transients are frequently invaded by a single protrusion from the endoplasmic reticulum (ER), which is dynamically transported into spines via the actin-based motor myosin V. An increase in synaptic strength correlates with stable anchoring of the ER, followed by the formation of an organelle referred to as the spine apparatus. Here, we show that myosin V binds the Ca2+ sensor caldendrin, a brain-specific homolog of the well-known myosin V interactor calmodulin. While calmodulin is an essential activator of myosin V motor function, we found that caldendrin acts as an inhibitor of processive myosin V movement. In mouse and rat hippocampal neurons, caldendrin regulates spine apparatus localization to a subset of dendritic spines through a myosin V-dependent pathway. We propose that caldendrin transforms myosin into a stationary F-actin tether that enables the localization of ER tubules and formation of the spine apparatus in dendritic spines.
Collapse
Affiliation(s)
- Anja Konietzny
- RG OptobiologyInstitute of BiologyHumboldt Universität zu BerlinBerlinGermany
- Guest Group Neuronal Protein TransportCenter for Molecular NeurobiologyZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Jasper Grendel
- RG OptobiologyInstitute of BiologyHumboldt Universität zu BerlinBerlinGermany
- Guest Group Neuronal Protein TransportCenter for Molecular NeurobiologyZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Alan Kadek
- Leibniz Institute for Experimental Virology (HPI)HamburgGermany
- European XFEL GmbHSchenefeldGermany
| | - Michael Bucher
- RG OptobiologyInstitute of BiologyHumboldt Universität zu BerlinBerlinGermany
- Guest Group Neuronal Protein TransportCenter for Molecular NeurobiologyZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Yuhao Han
- RG OptobiologyInstitute of BiologyHumboldt Universität zu BerlinBerlinGermany
- Guest Group Neuronal Protein TransportCenter for Molecular NeurobiologyZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- Centre for Structural Systems BiologyHamburgGermany
| | - Nathalie Hertrich
- RG OptobiologyInstitute of BiologyHumboldt Universität zu BerlinBerlinGermany
- Guest Group Neuronal Protein TransportCenter for Molecular NeurobiologyZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | | | | | - Kay Grünewald
- Leibniz Institute for Experimental Virology (HPI)HamburgGermany
- Centre for Structural Systems BiologyHamburgGermany
- Department of ChemistryUniversity of HamburgHamburgGermany
| | - Charlotte Uetrecht
- Leibniz Institute for Experimental Virology (HPI)HamburgGermany
- European XFEL GmbHSchenefeldGermany
- Centre for Structural Systems BiologyHamburgGermany
| | - Marina Mikhaylova
- RG OptobiologyInstitute of BiologyHumboldt Universität zu BerlinBerlinGermany
- Guest Group Neuronal Protein TransportCenter for Molecular NeurobiologyZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| |
Collapse
|
8
|
NeuriteNet: A convolutional neural network for assessing morphological parameters of neurite growth. J Neurosci Methods 2021; 363:109349. [PMID: 34480956 DOI: 10.1016/j.jneumeth.2021.109349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/19/2021] [Accepted: 08/30/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND During development or regeneration, neurons extend processes (i.e., neurites) via mechanisms that can be readily analyzed in culture. However, defining the impact of a drug or genetic manipulation on such mechanisms can be challenging due to the complex arborization and heterogeneous patterns of neurite growth in vitro. New Method: NeuriteNet is a Convolutional Neural Network (CNN) sorting model that uses a novel adaptation of the XRAI saliency map overlay, which is a region-based attribution method. NeuriteNet compares neuronal populations based on differences in neurite growth patterns, sorts them into respective groups, and overlays a saliency map indicating which areas differentiated the image for the sorting procedure. RESULTS In this study, we demonstrate that NeuriteNet effectively sorts images corresponding to dissociated neurons into control and treatment groups according to known morphological differences. Furthermore, the saliency map overlay highlights the distinguishing features of the neuron when sorting the images into treatment groups. NeuriteNet also identifies novel morphological differences in neurons cultured from control and genetically modified mouse strains. Comparison with Existing Methods: Unlike other neurite analysis platforms, NeuriteNet does not require manual manipulations, such as segmentation of neurites prior to analysis, and is more accurate than experienced researchers for categorizing neurons according to their pattern of neurite growth. CONCLUSIONS NeuriteNet can be used to effectively screen for morphological differences in a heterogeneous group of neurons and to provide feedback on the key features distinguishing those groups via the saliency map overlay.
Collapse
|
9
|
Complicity of α-synuclein oligomer and calcium dyshomeostasis in selective neuronal vulnerability in Lewy body disease. Arch Pharm Res 2021; 44:564-573. [PMID: 34114191 PMCID: PMC8254713 DOI: 10.1007/s12272-021-01334-6] [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: 02/22/2021] [Accepted: 05/27/2021] [Indexed: 11/18/2022]
Abstract
α-Synuclein oligomers and Ca2+ dyshomeostasis have been thoroughly investigated with respect to the pathogenesis of Lewy body disease (LBD). In LBD, α-synuclein oligomers exhibit a neuron-specific cytoplasmic distribution. Highly active neurons and neurons with a high Ca2+ burden are prone to damage in LBD. The neuronal vulnerability may be determined by transneuronal axonal transmission of the pathological processes; however, this hypothesis seems inconsistent with pathological findings that neurons anatomically connected to LBD-vulnerable neurons, such as neurons in the ventral tegmentum, are spared in LBD. This review focuses on and discusses the crucial roles played by α-synuclein oligomers and Ca2+ dyshomeostasis in early intraneural pathophysiology in LBD-vulnerable neurons. A challenging view is proposed on the synergy between retrograde transport of α-synuclein and vesicular Ca release, whereby neuronal vulnerability is propagated backward along repeatedly activated signaling pathway.
Collapse
|
10
|
Weyrer C, Turecek J, Niday Z, Liu PW, Nanou E, Catterall WA, Bean BP, Regehr WG. The Role of Ca V2.1 Channel Facilitation in Synaptic Facilitation. Cell Rep 2020; 26:2289-2297.e3. [PMID: 30811980 PMCID: PMC6597251 DOI: 10.1016/j.celrep.2019.01.114] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/04/2019] [Accepted: 01/30/2019] [Indexed: 02/05/2023] Open
Abstract
Activation of CaV2.1 voltage-gated calcium channels is facilitated by preceding calcium entry. Such self-modulatory facilitation is thought to contribute to synaptic facilitation. Using knockin mice with mutated CaV2.1 channels that do not facilitate (Ca IM-AA mice), we surprisingly found that, under conditions of physiological calcium and near-physiological temperatures, synaptic facilitation at hippocampal CA3 to CA1 synapses was not attenuated in Ca IM-AA mice and facilitation was paradoxically more prominent at two cerebellar synapses. Enhanced facilitation at these synapses is consistent with a decrease in initial calcium entry, suggested by an action-potential-evoked CaV2.1 current reduction in Purkinje cells from Ca IM-AA mice. In wild-type mice, CaV2.1 facilitation during high-frequency action potential trains was very small. Thus, for the synapses studied, facilitation of calcium entry through CaV2.1 channels makes surprisingly little contribution to synaptic facilitation under physiological conditions. Instead, CaV2.1 facilitation offsets CaV2.1 inactivation to produce remarkably stable calcium influx during high-frequency activation. Weyrer et al. use Ca IM-AA mice in which CaV2.1 calcium channel facilitation is eliminated to study synaptic facilitation at hippocampal and cerebellar synapses. Under conditions of physiological temperature, external calcium, and presynaptic waveforms, facilitation of CaV2.1 channels is small and does not contribute to synaptic facilitation at these synapses.
Collapse
Affiliation(s)
- Christopher Weyrer
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Josef Turecek
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Zachary Niday
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Pin W Liu
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Evanthia Nanou
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280, USA
| | - William A Catterall
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280, USA
| | - Bruce P Bean
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Wade G Regehr
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
11
|
Yamamoto K, Izumi Y, Arifuku M, Kume T, Sawada H. α-Synuclein oligomers mediate the aberrant form of spike-induced calcium release from IP 3 receptor. Sci Rep 2019; 9:15977. [PMID: 31685859 PMCID: PMC6828767 DOI: 10.1038/s41598-019-52135-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 10/12/2019] [Indexed: 01/10/2023] Open
Abstract
Emerging evidence implicates α-synuclein oligomers as potential culprits in the pathogenesis of Lewy body disease (LBD). Soluble oligomeric α-synuclein accumulation in cytoplasm is believed to modify neuronal activities and intraneural Ca2+ dynamics, which augment the metabolic burden in central neurons vulnerable to LBD, although this hypothesis remains to be fully tested. We evaluated how intracellular α-synuclein oligomers affect the neuronal excitabilities and Ca2+ dynamics of pyramidal neurons in neocortical slices from mice. Intracellular application of α-synuclein containing stable higher-order oligomers (αSNo) significantly reduced spike frequency during current injection, elongated the duration of spike afterhyperpolarization (AHP), and enlarged AHP current charge in comparison with that of α-synuclein without higher-order oligomers. This αSNo-mediated alteration was triggered by spike-induced Ca2+ release from inositol trisphosphate receptors (IP3R) functionally coupled with L-type Ca2+ channels and SK-type K+ channels. Further electrophysiological and immunochemical observations revealed that α-synuclein oligomers greater than 100 kDa were directly associated with calcium-binding protein 1, which is responsible for regulating IP3R gating. They also block Ca2+-dependent inactivation of IP3R, and trigger Ca2+-induced Ca2+ release from IP3R during multiple spikes. This aberrant machinery may result in intraneural Ca2+ dyshomeostasis and may be the molecular basis for the vulnerability of neurons in LBD brains.
Collapse
Affiliation(s)
- Kenji Yamamoto
- Department of Neurology and Clinical Research Center, National Hospital Organization Utano National Hospital, Kyoto, Japan.
| | - Yasuhiko Izumi
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Japan.,Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Monami Arifuku
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Toshiaki Kume
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan.,Department of Applied Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Hideyuki Sawada
- Department of Neurology and Clinical Research Center, National Hospital Organization Utano National Hospital, Kyoto, Japan
| |
Collapse
|
12
|
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
|
13
|
Fairless R, Williams SK, Diem R. Calcium-Binding Proteins as Determinants of Central Nervous System Neuronal Vulnerability to Disease. Int J Mol Sci 2019; 20:ijms20092146. [PMID: 31052285 PMCID: PMC6539299 DOI: 10.3390/ijms20092146] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 04/26/2019] [Accepted: 04/27/2019] [Indexed: 12/14/2022] Open
Abstract
Neuronal subpopulations display differential vulnerabilities to disease, but the factors that determine their susceptibility are poorly understood. Toxic increases in intracellular calcium are a key factor in several neurodegenerative processes, with calcium-binding proteins providing an important first line of defense through their ability to buffer incoming calcium, allowing the neuron to quickly achieve homeostasis. Since neurons expressing different calcium-binding proteins have been reported to be differentially susceptible to degeneration, it can be hypothesized that rather than just serving as markers of different neuronal subpopulations, they might actually be a key determinant of survival. In this review, we will summarize some of the evidence that expression of the EF-hand calcium-binding proteins, calbindin, calretinin and parvalbumin, may influence the susceptibility of distinct neuronal subpopulations to disease processes.
Collapse
Affiliation(s)
- Richard Fairless
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany.
- Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DFKZ), 69120 Heidelberg, Germany.
| | - Sarah K Williams
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany.
- Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DFKZ), 69120 Heidelberg, Germany.
| | - Ricarda Diem
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany.
- Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DFKZ), 69120 Heidelberg, Germany.
| |
Collapse
|
14
|
Miao J, Wang F, Wang R, Zeng J, Zheng C, Zhuang G. Pleiotrophin regulates functional heterogeneity of microglia cells in EAE animal models of multiple sclerosis by activating CCr-7/CD206 molecules and functional cytokines. Am J Transl Res 2019; 11:2013-2027. [PMID: 31105814 PMCID: PMC6511770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 01/20/2019] [Indexed: 06/09/2023]
Abstract
Multiple sclerosis (MS) is a neurodegenerative and immune-mediated disorder that characterizes by demyelination and neuro-inflammation. This study aimed to investigate the effects of pleiotrophin (PTN) on treatment of early injuries of white matter of MS patients. Experimental autoimmune encephalomyelitis (EAE) animal models were established by injecting 200 μg myelinoligodendrocyte glyeoprotein 33-35 (MOG35-55) and were divided into PTN+MOG group and PBS+MOG group. Meanwhile, normal mice group was assigned as control group (NC group). Immunofluorescence double label was used to examined co-expression of molecules. LV5-PTN and LV3-siPTN were established and transfected into microglia cells. All brain imaging data was acquired with MRI scanner. Quantitative real-time RT-PCR (qRT-PCR) and western blot were used to evaluate mRNA and protein expression, respectively. Lesion sites mainly appeared in NAWM of bilateral occipital lobes in EAE models. PTN treatment significantly enhanced CCr7 and reduced CD206 expression compared to PBS+MOG group (P<0.05). PTN participated in mitogen-activated protein kinase (MAPK) signaling pathway in EAE models. PTN treatment significantly regulated levels of functional cytokines in both M1 and M2 type microglia cells compared to PBS+MOG group (P<0.05). LV5-PTN and LV3-siPTN transfection modulated levels of PTN and MAPK molecule in microglia cells undergoing treatment of M1 or M2 inducer. PTN strengthened M1/M2 transformation by regulating functional cytokines. In conclusion, PTN regulated functional heterogeneity of microglia cells in EAE animal models of MS by activating CCr-7/CD206 molecules and functional cytokines. PTN could be considered as a promising candidate molecule for treating early injuries of white matter of patients with MS.
Collapse
Affiliation(s)
- Jiayin Miao
- Department of Neurology, Zhongshan Hospital, Xiamen University201 Hubinnan Road, Xiamen 361004, China
| | - Feng Wang
- College of Computer Engineering, Jimei UniversityXiamen 361021, China
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen UniversityXiamen 361005, China
| | - Ru Wang
- Department of Neurology, Zhongshan Hospital, Xiamen University201 Hubinnan Road, Xiamen 361004, China
| | - Jianqi Zeng
- Department of Neurology, Zhongshan Hospital, Xiamen University201 Hubinnan Road, Xiamen 361004, China
| | - Cina Zheng
- Magnetic Resonance Center, Zhongshan Hospital, Xiamen University201 Hubinnan Road, Xiamen 361004, China
| | - Guohong Zhuang
- Organ Transplantation Institute, Anti-Cancer Research Center, Medical College, Xiamen UniversityXiamen 361000, China
| |
Collapse
|
15
|
Mundhenk J, Fusi C, Kreutz MR. Caldendrin and Calneurons-EF-Hand CaM-Like Calcium Sensors With Unique Features and Specialized Neuronal Functions. Front Mol Neurosci 2019; 12:16. [PMID: 30787867 PMCID: PMC6372560 DOI: 10.3389/fnmol.2019.00016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/17/2019] [Indexed: 01/02/2023] Open
Abstract
The calmodulin (CaM)-like Ca2+-sensor proteins caldendrin, calneuron-1 and -2 are members of the neuronal calcium-binding protein (nCaBP)-family, a family that evolved relatively late during vertebrate evolution. All three proteins are abundant in brain but show a strikingly different subcellular localization. Whereas caldendrin is enriched in the postsynaptic density (PSD), calneuron-1 and -2 accumulate at the trans-Golgi-network (TGN). Caldendrin exhibit a unique bipartite structure with a basic and proline-rich N-terminus while calneurons are the only EF-Hand CaM-like transmembrane proteins. These uncommon structural features come along with highly specialized functions of calneurons in Golgi-to-plasma-membrane trafficking and for caldendrin in actin-remodeling in dendritic spine synapses. In this review article, we will provide a synthesis of available data on the structure and biophysical properties of all three proteins. We will then discuss their cellular function with special emphasis on synaptic neurotransmission. Finally, we will summarize the evidence for a role of these proteins in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jennifer Mundhenk
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Camilla Fusi
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Leibniz Group "Dendritic Organelles and Synaptic Function", Center for Molecular Neurobiology, ZMNH, Hamburg, Germany
| |
Collapse
|
16
|
Pangrsic T, Singer JH, Koschak A. Voltage-Gated Calcium Channels: Key Players in Sensory Coding in the Retina and the Inner Ear. Physiol Rev 2019; 98:2063-2096. [PMID: 30067155 DOI: 10.1152/physrev.00030.2017] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Calcium influx through voltage-gated Ca (CaV) channels is the first step in synaptic transmission. This review concerns CaV channels at ribbon synapses in primary sense organs and their specialization for efficient coding of stimuli in the physical environment. Specifically, we describe molecular, biochemical, and biophysical properties of the CaV channels in sensory receptor cells of the retina, cochlea, and vestibular apparatus, and we consider how such properties might change over the course of development and contribute to synaptic plasticity. We pay particular attention to factors affecting the spatial arrangement of CaV channels at presynaptic, ribbon-type active zones, because the spatial relationship between CaV channels and release sites has been shown to affect synapse function critically in a number of systems. Finally, we review identified synaptopathies affecting sensory systems and arising from dysfunction of L-type, CaV1.3, and CaV1.4 channels or their protein modulatory elements.
Collapse
Affiliation(s)
- Tina Pangrsic
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen and Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine , Göttingen, Germany ; Department of Biology, University of Maryland , College Park, Maryland ; and Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck , Innsbruck , Austria
| | - Joshua H Singer
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen and Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine , Göttingen, Germany ; Department of Biology, University of Maryland , College Park, Maryland ; and Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck , Innsbruck , Austria
| | - Alexandra Koschak
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen and Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine , Göttingen, Germany ; Department of Biology, University of Maryland , College Park, Maryland ; and Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck , Innsbruck , Austria
| |
Collapse
|
17
|
Yang T, Britt JK, Cintrón-Pérez CJ, Vázquez-Rosa E, Tobin KV, Stalker G, Hardie J, Taugher RJ, Wemmie J, Pieper AA, Lee A. Ca 2+-Binding Protein 1 Regulates Hippocampal-dependent Memory and Synaptic Plasticity. Neuroscience 2018; 380:90-102. [PMID: 29660444 DOI: 10.1016/j.neuroscience.2018.04.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/16/2018] [Accepted: 04/05/2018] [Indexed: 11/25/2022]
Abstract
Ca2+-binding protein 1 (CaBP1) is a Ca2+-sensing protein similar to calmodulin that potently regulates voltage-gated Ca2+ channels. Unlike calmodulin, however, CaBP1 is mainly expressed in neuronal cell-types and enriched in the hippocampus, where its function is unknown. Here, we investigated the role of CaBP1 in hippocampal-dependent behaviors using mice lacking expression of CaBP1 (C-KO). By western blot, the largest CaBP1 splice variant, caldendrin, was detected in hippocampal lysates from wild-type (WT) but not C-KO mice. Compared to WT mice, C-KO mice exhibited mild deficits in spatial learning and memory in both the Barnes maze and in Morris water maze reversal learning. In contextual but not cued fear-conditioning assays, C-KO mice showed greater freezing responses than WT mice. In addition, the number of adult-born neurons in the hippocampus of C-KO mice was ∼40% of that in WT mice, as measured by bromodeoxyuridine labeling. Moreover, hippocampal long-term potentiation was significantly reduced in C-KO mice. We conclude that CaBP1 is required for cellular mechanisms underlying optimal encoding of hippocampal-dependent spatial and fear-related memories.
Collapse
Affiliation(s)
- Tian Yang
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - Jeremiah K Britt
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Psychiatry, University of Iowa, Iowa City, IA 52242, USA
| | - Coral J Cintrón-Pérez
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Psychiatry, University of Iowa, Iowa City, IA 52242, USA
| | - Edwin Vázquez-Rosa
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Psychiatry, University of Iowa, Iowa City, IA 52242, USA
| | - Kevin V Tobin
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - Grant Stalker
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - Jason Hardie
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - Rebecca J Taugher
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Psychiatry, University of Iowa, Iowa City, IA 52242, USA
| | - John Wemmie
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Psychiatry, University of Iowa, Iowa City, IA 52242, USA
| | - Andrew A Pieper
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, University of Iowa, Iowa City, IA 52242, USA; Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Psychiatry, University of Iowa, Iowa City, IA 52242, USA; Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Free Radical, University of Iowa, Iowa City, IA 52242, USA; Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Radiation Biology Program, University of Iowa, Iowa City, IA 52242, USA; Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Radiation Oncology Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Veterans Affairs, University of Iowa, Iowa City, IA 52242, USA; Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Pappajohn Biomedical Institute and Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA
| | - Amy Lee
- Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
18
|
Control of Excitation/Inhibition Balance in a Hippocampal Circuit by Calcium Sensor Protein Regulation of Presynaptic Calcium Channels. J Neurosci 2018; 38:4430-4440. [PMID: 29654190 DOI: 10.1523/jneurosci.0022-18.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/15/2018] [Accepted: 04/01/2018] [Indexed: 12/19/2022] Open
Abstract
Activity-dependent regulation controls the balance of synaptic excitation to inhibition in neural circuits, and disruption of this regulation impairs learning and memory and causes many neurological disorders. The molecular mechanisms underlying short-term synaptic plasticity are incompletely understood, and their role in inhibitory synapses remains uncertain. Here we show that regulation of voltage-gated calcium (Ca2+) channel type 2.1 (CaV2.1) by neuronal Ca2+ sensor (CaS) proteins controls synaptic plasticity and excitation/inhibition balance in a hippocampal circuit. Prevention of CaS protein regulation by introducing the IM-AA mutation in CaV2.1 channels in male and female mice impairs short-term synaptic facilitation at excitatory synapses of CA3 pyramidal neurons onto parvalbumin (PV)-expressing basket cells. In sharp contrast, the IM-AA mutation abolishes rapid synaptic depression in the inhibitory synapses of PV basket cells onto CA1 pyramidal neurons. These results show that CaS protein regulation of facilitation and inactivation of CaV2.1 channels controls the direction of short-term plasticity at these two synapses. Deletion of the CaS protein CaBP1/caldendrin also blocks rapid depression at PV-CA1 synapses, implicating its upregulation of inactivation of CaV2.1 channels in control of short-term synaptic plasticity at this inhibitory synapse. Studies of local-circuit function revealed reduced inhibition of CA1 pyramidal neurons by the disynaptic pathway from CA3 pyramidal cells via PV basket cells and greatly increased excitation/inhibition ratio of the direct excitatory input versus indirect inhibitory input from CA3 pyramidal neurons to CA1 pyramidal neurons. This striking defect in local-circuit function may contribute to the dramatic impairment of spatial learning and memory in IM-AA mice.SIGNIFICANCE STATEMENT Many forms of short-term synaptic plasticity in neuronal circuits rely on regulation of presynaptic voltage-gated Ca2+ (CaV) channels. Regulation of CaV2.1 channels by neuronal calcium sensor (CaS) proteins controls short-term synaptic plasticity. Here we demonstrate a direct link between regulation of CaV2.1 channels and short-term synaptic plasticity in native hippocampal excitatory and inhibitory synapses. We also identify CaBP1/caldendrin as the calcium sensor interacting with CaV2.1 channels to mediate rapid synaptic depression in the inhibitory hippocampal synapses of parvalbumin-expressing basket cells to CA1 pyramidal cells. Disruption of this regulation causes altered short-term plasticity and impaired balance of hippocampal excitatory to inhibitory circuits.
Collapse
|
19
|
Yang T, Hu N, Pangršič T, Green S, Hansen M, Lee A. Functions of CaBP1 and CaBP2 in the peripheral auditory system. Hear Res 2018; 364:48-58. [PMID: 29661613 DOI: 10.1016/j.heares.2018.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/13/2018] [Accepted: 04/02/2018] [Indexed: 12/29/2022]
Abstract
CaBPs are a family of Ca2+ binding proteins related to calmodulin. Two CaBP family members, CaBP1 and CaBP2, are highly expressed in the cochlea. Here, we investigated the significance of CaBP1 and CaBP2 for hearing in mice lacking expression of these proteins (CaBP1 KO and CaBP2 KO) using auditory brain responses (ABRs) and distortion product otoacoustic emissions (DPOAEs). In CaBP1 KO mice, ABR wave I was larger in amplitude, and shorter in latency and faster in decay, suggestive of enhanced synchrony of auditory nerve fibers. This interpretation was supported by the greater excitability of CaBP1 KO than WT neurons in whole-cell patch clamp recordings of spiral ganglion neurons in culture, and normal presynaptic function of CaBP1 KO IHCs. DPOAEs and ABR thresholds were normal in 4-week old CaBP1 KO mice, but elevated ABR thresholds became evident at 32 kHz at 9 weeks, and at 8 and 16 kHz by 6 months of age. In contrast, CaBP2 KO mice exhibited significant ABR threshold elevations at 4 weeks of age that became more severe in the mid-frequency range by 9 weeks. Though normal at 4 weeks, DPOAEs in CaBP2 KO mice were significantly reduced in the mid-frequency range by 9 weeks. Our results reveal requirements for CaBP1 and CaBP2 in the peripheral auditory system and highlight the diverse modes by which CaBPs influence sensory processing.
Collapse
Affiliation(s)
- Tian Yang
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA; Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Ning Hu
- Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Tina Pangršič
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Steven Green
- Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Marlan Hansen
- Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Neurosurgery, University of Iowa, Iowa City, IA 52242, USA
| | - Amy Lee
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA; Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Neurology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
20
|
Yang T, Choi JE, Soh D, Tobin K, Joiner ML, Hansen M, Lee A. CaBP1 regulates Ca v1 L-type Ca 2+ channels and their coupling to neurite growth and gene transcription in mouse spiral ganglion neurons. Mol Cell Neurosci 2018; 88:342-352. [PMID: 29548764 DOI: 10.1016/j.mcn.2018.03.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 03/07/2018] [Accepted: 03/12/2018] [Indexed: 12/19/2022] Open
Abstract
CaBP1 is a Ca2+ binding protein that is widely expressed in neurons in the brain, retina, and cochlea. In heterologous expression systems, CaBP1 interacts with and regulates voltage-gated Cav Ca2+ channels but whether this is the case in neurons is unknown. Here, we investigated the cellular functions of CaBP1 in cochlear spiral ganglion neurons (SGNs), which express high levels of CaBP1. Consistent with the role of CaBP1 as a suppressor of Ca2+-dependent inactivation (CDI) of Cav1 (L-type) channels, Cav1 currents underwent greater CDI in SGNs from mice lacking CaBP1 (C-KO) than in wild-type (WT) SGNs. The coupling of Cav1 channels to downstream signaling pathways was also disrupted in C-KO SGNs. Activity-dependent repression of neurite growth was significantly blunted and unresponsive to Cav1 antagonists in C-KO SGNs in contrast to WT SGNs. Moreover, Cav1-mediated Ca2+ signals and phosphorylation of cAMP-response element binding protein were reduced in C-KO SGNs compared to WT SGNs. Our findings establish a role for CaBP1 as an essential regulator of Cav1 channels in SGNs and their coupling to downstream pathways controlling activity-dependent transcription and neurite growth.
Collapse
Affiliation(s)
- Tian Yang
- Departments of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Ji-Eun Choi
- Departments of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Daniel Soh
- Departments of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Kevin Tobin
- Departments of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Mei-Ling Joiner
- Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Marlan Hansen
- Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Neurosurgery, University of Iowa, Iowa City, IA 52242, USA
| | - Amy Lee
- Departments of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA; Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Neurology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
21
|
Kiran U, Regur P, Kreutz MR, Sharma Y, Chakraborty A. Intermotif Communication Induces Hierarchical Ca2+ Filling of Caldendrin. Biochemistry 2017; 56:2467-2476. [DOI: 10.1021/acs.biochem.7b00132] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Uday Kiran
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500007, India
| | - Phanindranath Regur
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500007, India
| | - Michael R. Kreutz
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Brenneckestrasse 6, 39118 Magdeburg, Germany
- Leibniz
Group ‘Dendritic Organelles and Synaptic Function’,
Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Yogendra Sharma
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Asima Chakraborty
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500007, India
| |
Collapse
|
22
|
Lack of CaBP1/Caldendrin or CaBP2 Leads to Altered Ganglion Cell Responses. eNeuro 2016; 3:eN-NWR-0099-16. [PMID: 27822497 PMCID: PMC5083949 DOI: 10.1523/eneuro.0099-16.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 09/24/2016] [Accepted: 10/08/2016] [Indexed: 12/01/2022] Open
Abstract
Calcium-binding proteins (CaBPs) form a subfamily of calmodulin-like proteins that were cloned from the retina. CaBP4 and CaBP5 have been shown to be important for normal visual function. Although CaBP1/caldendrin and CaBP2 have been shown to modulate various targets in vitro, it is not known whether they contribute to the transmission of light responses through the retina. Therefore, we generated mice that lack CaBP2 or CaBP1/caldendrin (Cabp2–/– and Cabp1–/–) to test whether these CaBPs are essential for normal retinal function. By immunohistochemistry, the overall morphology of Cabp1–/– and Cabp2–/– retinas and the number of synaptic ribbons appear normal; transmission electron microscopy shows normal tethered ribbon synapses and synaptic vesicles as in wild-type retinas. However, whole-cell patch clamp recordings showed that light responses of retinal ganglion cells of Cabp2–/– and Cabp1–/– mice differ in amplitude and kinetics from those of wild-type mice. We conclude that CaBP1/caldendrin and CaBP2 are not required for normal gross retinal and synapse morphology but are necessary for the proper transmission of light responses through the retina; like other CaBPs, CaBP1/caldendrin and CaBP2 likely act by modulating presynaptic Ca2+-dependent signaling mechanisms.
Collapse
|
23
|
Yang T, Scholl ES, Pan N, Fritzsch B, Haeseleer F, Lee A. Expression and Localization of CaBP Ca2+ Binding Proteins in the Mouse Cochlea. PLoS One 2016; 11:e0147495. [PMID: 26809054 PMCID: PMC4725724 DOI: 10.1371/journal.pone.0147495] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/05/2016] [Indexed: 11/19/2022] Open
Abstract
CaBPs are a family of EF-hand Ca2+ binding proteins that are structurally similar to calmodulin. CaBPs can interact with, and yet differentially modulate, effectors that are regulated by calmodulin, such as Cav1 voltage-gated Ca2+ channels. Immunolabeling studies suggest that multiple CaBP family members (CaBP1, 2, 4, and 5) are expressed in the cochlea. To gain insights into the respective auditory functions of these CaBPs, we characterized the expression and cellular localization of CaBPs in the mouse cochlea. By quantitative reverse transcription PCR, we show that CaBP1 and CaBP2 are the major CaBPs expressed in mouse cochlea both before and after hearing onset. Of the three alternatively spliced variants of CaBP1 (caldendrin, CaBP1-L, and CaBP1-S) and CaBP2 (CaBP2-alt, CaBP2-L, CaBP2-S), caldendrin and CaBP2-alt are the most abundant. By in situ hybridization, probes recognizing caldendrin strongly label the spiral ganglion, while probes designed to recognize all three isoforms of CaBP1 weakly label both the inner and outer hair cells as well as the spiral ganglion. Within the spiral ganglion, caldendrin/CaBP1 labeling is associated with cells resembling satellite glial cells. CaBP2-alt is strongly expressed in inner hair cells both before and after hearing onset. Probes designed to recognize all three variants of CaBP2 strongly label inner hair cells before hearing onset and outer hair cells after the onset of hearing. Thus, CaBP1 and CaBP2 may have overlapping roles in regulating Ca2+ signaling in the hair cells, and CaBP1 may have an additional function in the spiral ganglion. Our findings provide a framework for understanding the role of CaBP family members in the auditory periphery.
Collapse
Affiliation(s)
- Tian Yang
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa, United States of America
- Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, Iowa, United States of America
- Department of Neurology, University of Iowa, Iowa City, Iowa, United States of America
| | - Elizabeth S. Scholl
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa, United States of America
- Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, Iowa, United States of America
- Department of Neurology, University of Iowa, Iowa City, Iowa, United States of America
| | - Ning Pan
- Department of Biology, University of Iowa, Iowa City, Iowa, United States of America
| | - Bernd Fritzsch
- Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, Iowa, United States of America
- Department of Biology, University of Iowa, Iowa City, Iowa, United States of America
| | - Françoise Haeseleer
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington, United States of America
| | - Amy Lee
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa, United States of America
- Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, Iowa, United States of America
- Department of Neurology, University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
24
|
Abstract
Ca2+-dependent inactivation (CDI) is a negative feedback regulation of voltage-gated Cav1 and Cav2 channels that is mediated by the Ca2+ sensing protein, calmodulin (CaM), binding to the pore-forming Cav α1 subunit. David Yue and his colleagues made seminal contributions to our understanding of this process, as well as factors that regulate CDI. Important in this regard are members of a family of Ca2+ binding proteins (CaBPs) that are related to calmodulin. CaBPs are expressed mainly in neural tissues and can antagonize CaM-dependent CDI for Cav1 L-type channels. This review will focus on the roles of CaBPs as Cav1-interacting proteins, and the significance of these interactions for vision, hearing, and neuronal Ca2+ signaling events.
Collapse
Affiliation(s)
- Jason Hardie
- a Departments of Molecular Physiology and Biophysics ; Otolaryngology-Head and Neck Surgery and Neurology; University of Iowa ; Iowa City , IA USA
| | - Amy Lee
- a Departments of Molecular Physiology and Biophysics ; Otolaryngology-Head and Neck Surgery and Neurology; University of Iowa ; Iowa City , IA USA
| |
Collapse
|
25
|
Girard F, Venail J, Schwaller B, Celio M. The EF-hand Ca2+-binding protein super-family: A genome-wide analysis of gene expression patterns in the adult mouse brain. Neuroscience 2015; 294:116-55. [DOI: 10.1016/j.neuroscience.2015.02.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 02/10/2015] [Accepted: 02/10/2015] [Indexed: 01/13/2023]
|
26
|
Abstract
Voltage- and ligand-gated ion channels form the molecular basis of cellular excitability. With >400 members and accounting for ∼1.5% of the human genome, ion channels are some of the most well studied of all proteins in heterologous expression systems. Yet, ion channels often exhibit unexpected properties in vivo because of their interaction with a variety of signaling/scaffolding proteins. Such interactions can influence the function and localization of ion channels, as well as their coupling to intracellular second messengers and pathways, thus increasing the signaling potential of these ion channels in neurons. Moreover, functions have been ascribed to ion channels that are largely independent of their ion-conducting roles. Molecular and functional dissection of the ion channel proteome/interactome has yielded new insights into the composition of ion channel complexes and how their dysregulation leads to human disease.
Collapse
|
27
|
Korbolina EE, Ershov NI, Bryzgalov LO, Kolosova NG. Application of quantitative trait locus mapping and transcriptomics to studies of the senescence-accelerated phenotype in rats. BMC Genomics 2014; 15 Suppl 12:S3. [PMID: 25563673 PMCID: PMC4303943 DOI: 10.1186/1471-2164-15-s12-s3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Etiology of complex disorders, such as cataract and neurodegenerative diseases including age-related macular degeneration (AMD), remains poorly understood due to the paucity of animal models, fully replicating the human disease. Previously, two quantitative trait loci (QTLs) associated with early cataract, AMD-like retinopathy, and some behavioral aberrations in senescence-accelerated OXYS rats were uncovered on chromosome 1 in a cross between OXYS and WAG rats. To confirm the findings, we generated interval-specific congenic strains, WAG/OXYS-1.1 and WAG/OXYS-1.2, carrying OXYS-derived loci of chromosome 1 in the WAG strain. Both congenic strains displayed early cataract and retinopathy but differed clinically from OXYS rats. Here we applied a high-throughput RNA sequencing (RNA-Seq) strategy to facilitate nomination of the candidate genes and functional pathways that may be responsible for these differences and can contribute to the development of the senescence-accelerated phenotype of OXYS rats. Results First, the size and map position of QTL-derived congenic segments were determined by comparative analysis of coding single-nucleotide polymorphisms (SNPs), which were identified for OXYS, WAG, and congenic retinal RNAs after sequencing. The transferred locus was not what we expected in WAG/OXYS-1.1 rats. In rat retina, 15442 genes were expressed. Coherent sets of differentially expressed genes were identified when we compared RNA-Seq retinal profiles of 20-day-old WAG/OXYS-1.1, WAG/OXYS-1.2, and OXYS rats. The genes most different in the average expression level between the congenic strains included those generally associated with the Wnt, integrin, and TGF-β signaling pathways, widely involved in neurodegenerative processes. Several candidate genes (including Arhgap33, Cebpg, Gtf3c1, Snurf, Tnfaip3, Yme1l1, Cbs, Car9 and Fn1) were found to be either polymorphic in the congenic loci or differentially expressed between the strains. These genes may contribute to the development of cataract and retinopathy. Conclusions This study is the first RNA-Seq analysis of the rat retinal transcriptome generated with 40 mln sequencing read depth. The integration of QTL and transcriptomic analyses in our study forms the basis of future research into the relationship between the candidate genes within the congenic regions and specific changes in the retinal transcriptome as possible causal mechanisms that underlie age-associated disorders.
Collapse
|
28
|
Burgoyne RD, Haynes LP. Sense and specificity in neuronal calcium signalling. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:1921-32. [PMID: 25447549 PMCID: PMC4728190 DOI: 10.1016/j.bbamcr.2014.10.029] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 10/25/2014] [Accepted: 10/29/2014] [Indexed: 11/02/2022]
Abstract
Changes in the intracellular free calcium concentration ([Ca²⁺]i) in neurons regulate many and varied aspects of neuronal function over time scales from microseconds to days. The mystery is how a single signalling ion can lead to such diverse and specific changes in cell function. This is partly due to aspects of the Ca²⁺ signal itself, including its magnitude, duration, localisation and persistent or oscillatory nature. The transduction of the Ca²⁺ signal requires Ca²⁺binding to various Ca²⁺ sensor proteins. The different properties of these sensors are important for differential signal processing and determine the physiological specificity of Ca(2+) signalling pathways. A major factor underlying the specific roles of particular Ca²⁺ sensor proteins is the nature of their interaction with target proteins and how this mediates unique patterns of regulation. We review here recent progress from structural analyses and from functional analyses in model organisms that have begun to reveal the rules that underlie Ca²⁺ sensor protein specificity for target interaction. We discuss three case studies exemplifying different aspects of Ca²⁺ sensor/target interaction. This article is part of a special issue titled the 13th European Symposium on Calcium.
Collapse
Affiliation(s)
- Robert D Burgoyne
- Department of Cellular and Molecular Physiology, The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool, L69 3BX, United Kingdom.
| | - Lee P Haynes
- Department of Cellular and Molecular Physiology, The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool, L69 3BX, United Kingdom
| |
Collapse
|
29
|
Reddy PP, Raghuram V, Hradsky J, Spilker C, Chakraborty A, Sharma Y, Mikhaylova M, Kreutz MR. Molecular dynamics of the neuronal EF-hand Ca2+-sensor Caldendrin. PLoS One 2014; 9:e103186. [PMID: 25058677 PMCID: PMC4110014 DOI: 10.1371/journal.pone.0103186] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 06/29/2014] [Indexed: 11/18/2022] Open
Abstract
Caldendrin, L- and S-CaBP1 are CaM-like Ca2+-sensors with different N-termini that arise from alternative splicing of the Caldendrin/CaBP1 gene and that appear to play an important role in neuronal Ca2+-signaling. In this paper we show that Caldendrin is abundantly present in brain while the shorter splice isoforms L- and S-CaBP1 are not detectable at the protein level. Caldendrin binds both Ca2+ and Mg2+ with a global Kd in the low µM range. Interestingly, the Mg2+-binding affinity is clearly higher than in S-CaBP1, suggesting that the extended N-terminus might influence Mg2+-binding of the first EF-hand. Further evidence for intra- and intermolecular interactions of Caldendrin came from gel-filtration, surface plasmon resonance, dynamic light scattering and FRET assays. Surprisingly, Caldendrin exhibits very little change in surface hydrophobicity and secondary as well as tertiary structure upon Ca2+-binding to Mg2+-saturated protein. Complex inter- and intramolecular interactions that are regulated by Ca2+-binding, high Mg2+- and low Ca2+-binding affinity, a rigid first EF-hand domain and little conformational change upon titration with Ca2+ of Mg2+-liganted protein suggest different modes of binding to target interactions as compared to classical neuronal Ca2+-sensors.
Collapse
Affiliation(s)
| | - Vijeta Raghuram
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
- Centre for Cellular and Molecular Biology, CSIR, Hyderabad, India
| | - Johannes Hradsky
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Christina Spilker
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | | | - Yogendra Sharma
- Centre for Cellular and Molecular Biology, CSIR, Hyderabad, India
| | - Marina Mikhaylova
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
- Cell Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Michael R. Kreutz
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
- * E-mail:
| |
Collapse
|