1
|
Castilla F, Lugo V, Miranda-Laferte E, Jordan N, Huesgen PF, Santiago-Schübel B, Alfonso-Prieto M, Hidalgo P. Mapping the interaction surface between Ca Vβ and actin and its role in calcium channel clearance. Nat Commun 2025; 16:4352. [PMID: 40348749 PMCID: PMC12065904 DOI: 10.1038/s41467-025-59548-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/25/2025] [Indexed: 05/14/2025] Open
Abstract
Defective ion channel turnover and clearance of damaged proteins are associated with aging and neurodegeneration. The L-type CaV1.2 voltage-gated calcium channel mediates depolarization-induced calcium signals in heart and brain. Here, we determined the interaction surface between actin and two calcium channel subunits, CaVβ2 and CaVβ4, using cross-linking mass spectrometry and protein-protein docking, and uncovered a role in replenishing conduction-defective CaV1.2 channels. Computational and in vitro mutagenesis identified hotspots in CaVβ that decreased the affinity for actin but not for CaV1.2. When coexpressed with CaV1.2, none of the tested actin-association-deficient CaVβ mutants altered the single-channel properties or the total number of channels at the cell surface. However, coexpression with the CaVβ2 hotspot mutant downregulated current amplitudes, and with a concomitant reduction in the number of functionally available channels, indicating that current inhibition resulted from a build-up of conduction silent channels. Our findings established CaVβ2-actin interaction as a key player for clearing the plasma membrane of corrupted CaV1.2 proteins to ensure the maintenance of a functional pool of channels and proper calcium signal transduction. The CaVβ-actin molecular model introduces a potentially druggable protein-protein interface to intervene CaV-mediated signaling processes.
Collapse
Affiliation(s)
- Francisco Castilla
- Institute of Biological Information Processing (IBI-1)-Molecular and Cellular Physiology, Forschungszentrum Jülich, Jülich, Germany
- Graduate Program, Faculty of Mathematics and Natural Sciences, Heinrich-Heine University, Düsseldorf, Germany
- Biologics Analytical Research and Development, AbbVie Deutschland GmbH & Co. KG, Ludwigshafen, Germany
| | - Victor Lugo
- Institute of Biological Information Processing (IBI-1)-Molecular and Cellular Physiology, Forschungszentrum Jülich, Jülich, Germany
- Graduate Program, Faculty of Mathematics and Natural Sciences, Heinrich-Heine University, Düsseldorf, Germany
| | - Erick Miranda-Laferte
- Institute of Biological Information Processing (IBI-1)-Molecular and Cellular Physiology, Forschungszentrum Jülich, Jülich, Germany
| | - Nadine Jordan
- Institute of Biological Information Processing (IBI-1)-Molecular and Cellular Physiology, Forschungszentrum Jülich, Jülich, Germany
| | - Pitter F Huesgen
- Central Institute of Engineering, Electronics and Analytics (ZEA-3), Forschungszentrum Jülich, Jülich, Germany
- Institute of Biology II, University of Freiburg, Freiburg, Germany
| | - Beatrix Santiago-Schübel
- Central Institute of Engineering, Electronics and Analytics (ZEA-3), Forschungszentrum Jülich, Jülich, Germany.
- Institute of Biological Information Processing (IBI-7)-Structural Biochemistry, Forschungszentrum Jülich, Jülich, Germany.
| | - Mercedes Alfonso-Prieto
- Institute of Neuroscience and Medicine (INM-9)-Computational Biomedicine, Forschungszentrum Jülich, Jülich, Germany.
- Cécile and Vogt Institute for Brain Research, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany.
| | - Patricia Hidalgo
- Institute of Biological Information Processing (IBI-1)-Molecular and Cellular Physiology, Forschungszentrum Jülich, Jülich, Germany.
- Institute of Biochemistry, Heinrich-Heine University, Düsseldorf, Germany.
| |
Collapse
|
2
|
Huang Y, Chen SR, Pan HL. α2δ-1-Linked NMDA and AMPA Receptors in Neuropathic Pain and Gabapentinoid Action. J Neurochem 2025; 169:e70064. [PMID: 40191897 PMCID: PMC11995887 DOI: 10.1111/jnc.70064] [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: 02/06/2025] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 04/16/2025]
Abstract
Chronic neuropathic pain is a debilitating condition that presents a significant therapeutic challenge. Unlike nociceptive pain, neuropathic pain is predominantly driven by glutamate NMDA receptors (NMDARs) and/or Ca2+-permeable AMPA receptors (CP-AMPARs) at synapses between primary afferent nerves and excitatory neurons in the spinal dorsal horn. The α2δ-1 protein, encoded by Cacna2d1 and historically recognized as a subunit of voltage-activated Ca2+ channels, is the primary target of gabapentinoids, such as gabapentin and pregabalin, which are widely prescribed for neuropathic pain and epilepsy. However, gabapentinoids have minimal effects on Ca2+ channel activity. Recent studies reveal that α2δ-1 plays a pivotal role in amplifying nociceptive input to the spinal cord in neuropathic pain. This action is mediated through its dynamic physical interactions with phosphorylated NMDARs and GluA1/GluA2 subunits via its intrinsically disordered C-terminal region. α2δ-1 not only promotes synaptic trafficking of NMDARs but also disrupts heteromeric assembly of GluA1/GluA2 subunits in the spinal dorsal horn. The central function of α2δ-1 is to elevate intracellular Ca2+ concentrations at both presynaptic and postsynaptic sites, augmenting nociceptive transmission. Consequently, α2δ-1 serves as a dual regulator coordinating synaptic expression of NMDARs and GluA1 homomeric CP-AMPARs, a function that underlies the therapeutic actions of gabapentinoids. By inhibiting α2δ-1, gabapentinoids reduce the hyperactivity of synaptic α2δ-1-bound NMDARs and CP-AMPARs, thereby dampening the excessive excitatory synaptic transmission characteristic of neuropathic pain. These newly identified roles of α2δ-1 in orchestrating glutamatergic synaptic plasticity suggest that gabapentinoids could be repurposed for treating other neurological disorders involving dysregulated synaptic NMDARs and CP-AMPARs.
Collapse
Affiliation(s)
- Yuying Huang
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shao-Rui Chen
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hui-Lin Pan
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
3
|
Limpitikul WB, Dick IE. Inactivation of CaV1 and CaV2 channels. J Gen Physiol 2025; 157:e202313531. [PMID: 39883005 PMCID: PMC11781272 DOI: 10.1085/jgp.202313531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 01/02/2025] [Accepted: 01/15/2025] [Indexed: 01/31/2025] Open
Abstract
Voltage-gated Ca2+ channels (VGCCs) are highly expressed throughout numerous biological systems and play critical roles in synaptic transmission, cardiac excitation, and muscle contraction. To perform these various functions, VGCCs are highly regulated. Inactivation comprises a critical mechanism controlling the entry of Ca2+ through these channels and constitutes an important means to regulate cellular excitability, shape action potentials, control intracellular Ca2+ levels, and contribute to long-term potentiation and depression. For CaV1 and CaV2 channel families, inactivation proceeds via two distinct processes. Voltage-dependent inactivation (VDI) reduces Ca2+ entry through the channel in response to sustained or repetitive depolarization, while Ca2+-dependent inactivation (CDI) occurs in response to elevations in intracellular Ca2+ levels. These processes are critical for physiological function and undergo exquisite fine-tuning through multiple mechanisms. Here, we review known determinants and modulatory features of these two critical forms of channel regulation and their role in normal physiology and pathophysiology.
Collapse
Affiliation(s)
| | - Ivy E. Dick
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
4
|
Calderon-Rivera A, Gomez K, Rodríguez-Palma EJ, Khanna R. SUMOylation and DeSUMOylation: Tug of War of Pain Signaling. Mol Neurobiol 2025; 62:3305-3321. [PMID: 39276308 DOI: 10.1007/s12035-024-04478-w] [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: 05/31/2024] [Accepted: 09/02/2024] [Indexed: 09/16/2024]
Abstract
SUMOylation is a post-translational modification that attaches a small ubiquitin-like modifier (SUMO) group to a target protein via SUMO ligases, while deSUMOylation refers to the removal of this SUMO group by sentrin-specific proteases (SENPs). Although the functions of these processes have been well described in the nucleus, the role of SUMOylation and deSUMOylation in regulating ion channels is emerging as a novel area of study. Despite this, their contributions to pain signaling remain less clear. Therefore, this review consolidates the current evidence on the link(s) between SUMOylation, deSUMOylation, and pain, with a specific focus on ion channels expressed in the sensory system. Additionally, we explore the role of SUMOylation in the expression and function of kinases, vesicle proteins, and transcription factors, which result in the modulation of certain ion channels contributing to pain. Altogether, this review aims to highlight the relationship between SUMOylation and deSUMOylation in the modulation of ion channels, ultimately exploring the potential therapeutic role of these processes in chronic pain.
Collapse
Affiliation(s)
- Aida Calderon-Rivera
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, 1200 Newell Drive, Gainesville, FL, 32610, USA
| | - Kimberly Gomez
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, 1200 Newell Drive, Gainesville, FL, 32610, USA
| | - Erick J Rodríguez-Palma
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, 1200 Newell Drive, Gainesville, FL, 32610, USA
| | - Rajesh Khanna
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, 1200 Newell Drive, Gainesville, FL, 32610, USA.
- Pain and Addiction Therapeutics (PATH) Collaboratory, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
5
|
Martín‐Belmonte A, Aguado C, Alfaro‐Ruiz R, Kulik A, de la Ossa L, Moreno‐Martínez AE, Alberquilla S, García‐Carracedo L, Fernández M, Fajardo‐Serrano A, Aso E, Shigemoto R, Martín ED, Fukazawa Y, Ciruela F, Luján R. Nanoarchitecture of Ca V2.1 channels and GABA B receptors in the mouse hippocampus: Impact of APP/PS1 pathology. Brain Pathol 2025; 35:e13279. [PMID: 38887180 PMCID: PMC11835447 DOI: 10.1111/bpa.13279] [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: 02/06/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
Voltage-gated CaV2.1 (P/Q-type) Ca2+ channels play a crucial role in regulating neurotransmitter release, thus contributing to synaptic plasticity and to processes such as learning and memory. Despite their recognized importance in neural function, there is limited information on their potential involvement in neurodegenerative conditions such as Alzheimer's disease (AD). Here, we aimed to explore the impact of AD pathology on the density and nanoscale compartmentalization of CaV2.1 channels in the hippocampus in association with GABAB receptors. Histoblotting experiments showed that the density of CaV2.1 channel was significantly reduced in the hippocampus of APP/PS1 mice in a laminar-dependent manner. CaV2.1 channel was enriched in the active zone of the axon terminals and was present at a very low density over the surface of dendritic tree of the CA1 pyramidal cells, as shown by quantitative SDS-digested freeze-fracture replica labelling (SDS-FRL). In APP/PS1 mice, the density of CaV2.1 channel in the active zone was significantly reduced in the strata radiatum and lacunosum-moleculare, while it remained unaltered in the stratum oriens. The decline in Cav2.1 channel density was found to be associated with a corresponding impairment in the GABAergic synaptic function, as evidenced by electrophysiological experiments carried out in the hippocampus of APP/PS1 mice. Remarkably, double SDS-FRL showed a co-clustering of CaV2.1 channel and GABAB1 receptor in nanodomains (~40-50 nm) in wild type mice, while in APP/PS1 mice this nanoarchitecture was absent. Together, these findings suggest that the AD pathology-induced reduction in CaV2.1 channel density and CaV2.1-GABAB1 de-clustering may play a role in the synaptic transmission alterations shown in the AD hippocampus. Therefore, uncovering these layer-dependent changes in P/Q calcium currents associated with AD pathology can benefit the development of future strategies for AD management.
Collapse
Affiliation(s)
- Alejandro Martín‐Belmonte
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB‐UCLM)Universidad Castilla‐La ManchaAlbaceteSpain
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
- Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de BellvitgeBarcelonaSpain
| | - Carolina Aguado
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB‐UCLM)Universidad Castilla‐La ManchaAlbaceteSpain
- Laboratorio de Estructura Sináptica, Instituto de Investigación Sanitaria de Castilla‐La Mancha (IDISCAM)AlbaceteSpain
| | - Rocío Alfaro‐Ruiz
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB‐UCLM)Universidad Castilla‐La ManchaAlbaceteSpain
- Laboratorio de Estructura Sináptica, Instituto de Investigación Sanitaria de Castilla‐La Mancha (IDISCAM)AlbaceteSpain
| | - Akos Kulik
- Institute for Physiology II, Medical FacultyUniversity of FreiburgFreiburgGermany
| | - Luis de la Ossa
- Departamento de Sistemas Informáticos, Escuela Superior de Ingeniería InformáticaUniversidad de Castilla‐La ManchaAlbaceteSpain
| | - Ana Esther Moreno‐Martínez
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB‐UCLM)Universidad Castilla‐La ManchaAlbaceteSpain
- Laboratorio de Estructura Sináptica, Instituto de Investigación Sanitaria de Castilla‐La Mancha (IDISCAM)AlbaceteSpain
| | - Samuel Alberquilla
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones CientíficasMadridSpain
| | - Lucía García‐Carracedo
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones CientíficasMadridSpain
| | - Miriam Fernández
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB‐UCLM)Universidad Castilla‐La ManchaAlbaceteSpain
- Laboratorio de Estructura Sináptica, Instituto de Investigación Sanitaria de Castilla‐La Mancha (IDISCAM)AlbaceteSpain
| | - Ana Fajardo‐Serrano
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB‐UCLM)Universidad Castilla‐La ManchaAlbaceteSpain
| | - Ester Aso
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
- Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de BellvitgeBarcelonaSpain
| | - Ryuichi Shigemoto
- Institute of Science and Technology Austria (ISTA)KlosterneuburgAustria
| | - Eduardo D. Martín
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones CientíficasMadridSpain
| | - Yugo Fukazawa
- Division of Brain Structure and Function, Faculty of Medical ScienceUniversity of FukuiFukuiJapan
- Life Science Innovation CenterUniversity of FukuiFukuiJapan
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
- Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de BellvitgeBarcelonaSpain
| | - Rafael Luján
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB‐UCLM)Universidad Castilla‐La ManchaAlbaceteSpain
- Laboratorio de Estructura Sináptica, Instituto de Investigación Sanitaria de Castilla‐La Mancha (IDISCAM)AlbaceteSpain
| |
Collapse
|
6
|
Yu Y, Liao P, Jiang R. Ion Channels in Odor Information Processing of Neural Circuits of the Vertebrate Olfactory Bulb. Int J Mol Sci 2024; 25:13259. [PMID: 39769024 PMCID: PMC11675640 DOI: 10.3390/ijms252413259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Olfactory disorders and their associated complications present a considerable challenge to an individual's quality of life and emotional wellbeing. The current range of treatments, including surgical procedures, pharmacological interventions, and behavioral training, frequently proves ineffective in restoring olfactory function. The olfactory bulb (OB) is essential for odor processing and plays a pivotal role in the development of these disorders. Despite the acknowledged significance of ion channels in sensory functions and related pathologies, their specific involvement in OB remains unexplored. This review presents an overview of the functions of various ion channel families in regulating neuronal excitability, synaptic transmission, and the complex processes of olfactory perception. The objective of this review was to elucidate the role of ion channels in olfactory function, providing new insights into the diagnosis and treatment of olfactory dysfunction.
Collapse
Affiliation(s)
- Yunqing Yu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ping Liao
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ruotian Jiang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
7
|
Lauerer RJ, Lerche H. Voltage-gated calcium channels in genetic epilepsies. J Neurochem 2024; 168:3853-3871. [PMID: 37822150 PMCID: PMC11591408 DOI: 10.1111/jnc.15983] [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: 07/08/2023] [Revised: 09/17/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023]
Abstract
Voltage-gated calcium channels (VGCC) are abundant in the central nervous system and serve a broad spectrum of functions, either directly in cellular excitability or indirectly to regulate Ca2+ homeostasis. Ca2+ ions act as one of the main connections in excitation-transcription coupling, muscle contraction and excitation-exocytosis coupling, including synaptic transmission. In recent years, many genes encoding VGCCs main α or additional auxiliary subunits have been associated with epilepsy. This review sums up the current state of knowledge on disease mechanisms and provides guidance on disease-specific therapies where applicable.
Collapse
Affiliation(s)
- Robert J. Lauerer
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain ResearchUniversity and University Hospital TuebingenTuebingenGermany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain ResearchUniversity and University Hospital TuebingenTuebingenGermany
| |
Collapse
|
8
|
Medeiros AT, Gratz SJ, Delgado A, Ritt JT, O'Connor-Giles KM. Ca 2+ channel and active zone protein abundance intersects with input-specific synapse organization to shape functional synaptic diversity. eLife 2024; 12:RP88412. [PMID: 39291956 PMCID: PMC11410372 DOI: 10.7554/elife.88412] [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] [Indexed: 09/19/2024] Open
Abstract
Synaptic heterogeneity is a hallmark of nervous systems that enables complex and adaptable communication in neural circuits. To understand circuit function, it is thus critical to determine the factors that contribute to the functional diversity of synapses. We investigated the contributions of voltage-gated calcium channel (VGCC) abundance, spatial organization, and subunit composition to synapse diversity among and between synapses formed by two closely related Drosophila glutamatergic motor neurons with distinct neurotransmitter release probabilities (Pr). Surprisingly, VGCC levels are highly predictive of heterogeneous Pr among individual synapses of either low- or high-Pr inputs, but not between inputs. We find that the same number of VGCCs are more densely organized at high-Pr synapses, consistent with tighter VGCC-synaptic vesicle coupling. We generated endogenously tagged lines to investigate VGCC subunits in vivo and found that the α2δ-3 subunit Straightjacket along with the CAST/ELKS active zone (AZ) protein Bruchpilot, both key regulators of VGCCs, are less abundant at high-Pr inputs, yet positively correlate with Pr among synapses formed by either input. Consistently, both Straightjacket and Bruchpilot levels are dynamically increased across AZs of both inputs when neurotransmitter release is potentiated to maintain stable communication following glutamate receptor inhibition. Together, these findings suggest a model in which VGCC and AZ protein abundance intersects with input-specific spatial and molecular organization to shape the functional diversity of synapses.
Collapse
Affiliation(s)
- Audrey T Medeiros
- Neuroscience Graduate Training Program, Brown University, Providence, United States
| | - Scott J Gratz
- Department of Neuroscience, Brown University, Providence, United States
| | - Ambar Delgado
- Department of Neuroscience, Brown University, Providence, United States
| | - Jason T Ritt
- Department of Neuroscience, Brown University, Providence, United States
- Carney Institute for Brain Science, Brown University, Providence, United States
| | - Kate M O'Connor-Giles
- Neuroscience Graduate Training Program, Brown University, Providence, United States
- Department of Neuroscience, Brown University, Providence, United States
- Carney Institute for Brain Science, Brown University, Providence, United States
| |
Collapse
|
9
|
Ritzau-Jost A, Gsell F, Sell J, Sachs S, Montanaro J, Kirmann T, Maaß S, Irani SR, Werner C, Geis C, Sauer M, Shigemoto R, Hallermann S. LGI1 Autoantibodies Enhance Synaptic Transmission by Presynaptic K v1 Loss and Increased Action Potential Broadening. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200284. [PMID: 39141878 PMCID: PMC11379440 DOI: 10.1212/nxi.0000000000200284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 07/01/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND AND OBJECTIVES Autoantibodies against the protein leucine-rich glioma inactivated 1 (LGI1) cause the most common subtype of autoimmune encephalitis with predominant involvement of the limbic system, associated with seizures and memory deficits. LGI1 and its receptor ADAM22 are part of a transsynaptic protein complex that includes several proteins involved in presynaptic neurotransmitter release and postsynaptic glutamate sensing. Autoantibodies against LGI1 increase excitatory synaptic strength, but studies that genetically disrupt the LGI1-ADAM22 complex report a reduction in postsynaptic glutamate receptor-mediated responses. Thus, the mechanisms underlying the increased synaptic strength induced by LGI1 autoantibodies remain elusive, and the contributions of presynaptic molecules to the LGI1-transsynaptic complex remain unclear. We therefore investigated the presynaptic mechanisms that mediate autoantibody-induced synaptic strengthening. METHODS We studied the effects of patient-derived purified polyclonal LGI1 autoantibodies on synaptic structure and function by combining direct patch-clamp recordings from presynaptic boutons and somata of hippocampal neurons with super-resolution light and electron microscopy of hippocampal cultures and brain slices. We also identified the protein domain mediating the presynaptic effect using domain-specific patient-derived monoclonal antibodies. RESULTS LGI1 autoantibodies dose-dependently increased short-term depression during high-frequency transmission, consistent with increased release probability. The increased neurotransmission was not related to presynaptic calcium channels because presynaptic Cav2.1 channel density, calcium current amplitude, and calcium channel gating were unaffected by LGI1 autoantibodies. By contrast, application of LGI1 autoantibodies homogeneously reduced Kv1.1 and Kv1.2 channel density on the surface of presynaptic boutons. Direct presynaptic patch-clamp recordings revealed that LGI1 autoantibodies cause a pronounced broadening of the presynaptic action potential. Domain-specific effects of LGI1 autoantibodies were analyzed at the neuronal soma. Somatic action potential broadening was induced by polyclonal LGI1 autoantibodies and patient-derived monoclonal autoantibodies targeting the epitempin domain, but not the leucin-rich repeat domain. DISCUSSION Our results indicate that LGI1 autoantibodies reduce the density of both Kv1.1 and Kv1.2 on presynaptic boutons, without actions on calcium channel density or function, thereby broadening the presynaptic action potential and increasing neurotransmitter release. This study provides a molecular explanation for the neuronal hyperactivity observed in patients with LGI1 autoantibodies.
Collapse
Affiliation(s)
- Andreas Ritzau-Jost
- From the Carl-Ludwig-Institute of Physiology (A.R.-J., F.G., T.K., S.M., S.H.), Faculty of Medicine, Leipzig University; Section Translational Neuroimmunology (J.S., C.G.), Department of Neurology, Jena University Hospital; Department of Biotechnology and Biophysics (S.S., C.W., M.S.), University of Würzburg, Biocenter, Germany; Institute of Science and Technology Austria (ISTA) (J.M., R.S.), Klosterneuburg, Austria; Oxford Autoimmune Neurology Group (S.R.I.), Nuffield Department of Clinical Neurosciences, University of Oxford, ; Department of Neurology (S.R.I.), John Radcliffe Hospital, Oxford University Hospitals, United Kingdom; and Departments of Neurology and Neurosciences (S.R.I.), Mayo Clinic Jacksonville, FL
| | - Felix Gsell
- From the Carl-Ludwig-Institute of Physiology (A.R.-J., F.G., T.K., S.M., S.H.), Faculty of Medicine, Leipzig University; Section Translational Neuroimmunology (J.S., C.G.), Department of Neurology, Jena University Hospital; Department of Biotechnology and Biophysics (S.S., C.W., M.S.), University of Würzburg, Biocenter, Germany; Institute of Science and Technology Austria (ISTA) (J.M., R.S.), Klosterneuburg, Austria; Oxford Autoimmune Neurology Group (S.R.I.), Nuffield Department of Clinical Neurosciences, University of Oxford, ; Department of Neurology (S.R.I.), John Radcliffe Hospital, Oxford University Hospitals, United Kingdom; and Departments of Neurology and Neurosciences (S.R.I.), Mayo Clinic Jacksonville, FL
| | - Josefine Sell
- From the Carl-Ludwig-Institute of Physiology (A.R.-J., F.G., T.K., S.M., S.H.), Faculty of Medicine, Leipzig University; Section Translational Neuroimmunology (J.S., C.G.), Department of Neurology, Jena University Hospital; Department of Biotechnology and Biophysics (S.S., C.W., M.S.), University of Würzburg, Biocenter, Germany; Institute of Science and Technology Austria (ISTA) (J.M., R.S.), Klosterneuburg, Austria; Oxford Autoimmune Neurology Group (S.R.I.), Nuffield Department of Clinical Neurosciences, University of Oxford, ; Department of Neurology (S.R.I.), John Radcliffe Hospital, Oxford University Hospitals, United Kingdom; and Departments of Neurology and Neurosciences (S.R.I.), Mayo Clinic Jacksonville, FL
| | - Stefan Sachs
- From the Carl-Ludwig-Institute of Physiology (A.R.-J., F.G., T.K., S.M., S.H.), Faculty of Medicine, Leipzig University; Section Translational Neuroimmunology (J.S., C.G.), Department of Neurology, Jena University Hospital; Department of Biotechnology and Biophysics (S.S., C.W., M.S.), University of Würzburg, Biocenter, Germany; Institute of Science and Technology Austria (ISTA) (J.M., R.S.), Klosterneuburg, Austria; Oxford Autoimmune Neurology Group (S.R.I.), Nuffield Department of Clinical Neurosciences, University of Oxford, ; Department of Neurology (S.R.I.), John Radcliffe Hospital, Oxford University Hospitals, United Kingdom; and Departments of Neurology and Neurosciences (S.R.I.), Mayo Clinic Jacksonville, FL
| | - Jacqueline Montanaro
- From the Carl-Ludwig-Institute of Physiology (A.R.-J., F.G., T.K., S.M., S.H.), Faculty of Medicine, Leipzig University; Section Translational Neuroimmunology (J.S., C.G.), Department of Neurology, Jena University Hospital; Department of Biotechnology and Biophysics (S.S., C.W., M.S.), University of Würzburg, Biocenter, Germany; Institute of Science and Technology Austria (ISTA) (J.M., R.S.), Klosterneuburg, Austria; Oxford Autoimmune Neurology Group (S.R.I.), Nuffield Department of Clinical Neurosciences, University of Oxford, ; Department of Neurology (S.R.I.), John Radcliffe Hospital, Oxford University Hospitals, United Kingdom; and Departments of Neurology and Neurosciences (S.R.I.), Mayo Clinic Jacksonville, FL
| | - Toni Kirmann
- From the Carl-Ludwig-Institute of Physiology (A.R.-J., F.G., T.K., S.M., S.H.), Faculty of Medicine, Leipzig University; Section Translational Neuroimmunology (J.S., C.G.), Department of Neurology, Jena University Hospital; Department of Biotechnology and Biophysics (S.S., C.W., M.S.), University of Würzburg, Biocenter, Germany; Institute of Science and Technology Austria (ISTA) (J.M., R.S.), Klosterneuburg, Austria; Oxford Autoimmune Neurology Group (S.R.I.), Nuffield Department of Clinical Neurosciences, University of Oxford, ; Department of Neurology (S.R.I.), John Radcliffe Hospital, Oxford University Hospitals, United Kingdom; and Departments of Neurology and Neurosciences (S.R.I.), Mayo Clinic Jacksonville, FL
| | - Sebastian Maaß
- From the Carl-Ludwig-Institute of Physiology (A.R.-J., F.G., T.K., S.M., S.H.), Faculty of Medicine, Leipzig University; Section Translational Neuroimmunology (J.S., C.G.), Department of Neurology, Jena University Hospital; Department of Biotechnology and Biophysics (S.S., C.W., M.S.), University of Würzburg, Biocenter, Germany; Institute of Science and Technology Austria (ISTA) (J.M., R.S.), Klosterneuburg, Austria; Oxford Autoimmune Neurology Group (S.R.I.), Nuffield Department of Clinical Neurosciences, University of Oxford, ; Department of Neurology (S.R.I.), John Radcliffe Hospital, Oxford University Hospitals, United Kingdom; and Departments of Neurology and Neurosciences (S.R.I.), Mayo Clinic Jacksonville, FL
| | - Sarosh R Irani
- From the Carl-Ludwig-Institute of Physiology (A.R.-J., F.G., T.K., S.M., S.H.), Faculty of Medicine, Leipzig University; Section Translational Neuroimmunology (J.S., C.G.), Department of Neurology, Jena University Hospital; Department of Biotechnology and Biophysics (S.S., C.W., M.S.), University of Würzburg, Biocenter, Germany; Institute of Science and Technology Austria (ISTA) (J.M., R.S.), Klosterneuburg, Austria; Oxford Autoimmune Neurology Group (S.R.I.), Nuffield Department of Clinical Neurosciences, University of Oxford, ; Department of Neurology (S.R.I.), John Radcliffe Hospital, Oxford University Hospitals, United Kingdom; and Departments of Neurology and Neurosciences (S.R.I.), Mayo Clinic Jacksonville, FL
| | - Christian Werner
- From the Carl-Ludwig-Institute of Physiology (A.R.-J., F.G., T.K., S.M., S.H.), Faculty of Medicine, Leipzig University; Section Translational Neuroimmunology (J.S., C.G.), Department of Neurology, Jena University Hospital; Department of Biotechnology and Biophysics (S.S., C.W., M.S.), University of Würzburg, Biocenter, Germany; Institute of Science and Technology Austria (ISTA) (J.M., R.S.), Klosterneuburg, Austria; Oxford Autoimmune Neurology Group (S.R.I.), Nuffield Department of Clinical Neurosciences, University of Oxford, ; Department of Neurology (S.R.I.), John Radcliffe Hospital, Oxford University Hospitals, United Kingdom; and Departments of Neurology and Neurosciences (S.R.I.), Mayo Clinic Jacksonville, FL
| | - Christian Geis
- From the Carl-Ludwig-Institute of Physiology (A.R.-J., F.G., T.K., S.M., S.H.), Faculty of Medicine, Leipzig University; Section Translational Neuroimmunology (J.S., C.G.), Department of Neurology, Jena University Hospital; Department of Biotechnology and Biophysics (S.S., C.W., M.S.), University of Würzburg, Biocenter, Germany; Institute of Science and Technology Austria (ISTA) (J.M., R.S.), Klosterneuburg, Austria; Oxford Autoimmune Neurology Group (S.R.I.), Nuffield Department of Clinical Neurosciences, University of Oxford, ; Department of Neurology (S.R.I.), John Radcliffe Hospital, Oxford University Hospitals, United Kingdom; and Departments of Neurology and Neurosciences (S.R.I.), Mayo Clinic Jacksonville, FL
| | - Markus Sauer
- From the Carl-Ludwig-Institute of Physiology (A.R.-J., F.G., T.K., S.M., S.H.), Faculty of Medicine, Leipzig University; Section Translational Neuroimmunology (J.S., C.G.), Department of Neurology, Jena University Hospital; Department of Biotechnology and Biophysics (S.S., C.W., M.S.), University of Würzburg, Biocenter, Germany; Institute of Science and Technology Austria (ISTA) (J.M., R.S.), Klosterneuburg, Austria; Oxford Autoimmune Neurology Group (S.R.I.), Nuffield Department of Clinical Neurosciences, University of Oxford, ; Department of Neurology (S.R.I.), John Radcliffe Hospital, Oxford University Hospitals, United Kingdom; and Departments of Neurology and Neurosciences (S.R.I.), Mayo Clinic Jacksonville, FL
| | - Ryuichi Shigemoto
- From the Carl-Ludwig-Institute of Physiology (A.R.-J., F.G., T.K., S.M., S.H.), Faculty of Medicine, Leipzig University; Section Translational Neuroimmunology (J.S., C.G.), Department of Neurology, Jena University Hospital; Department of Biotechnology and Biophysics (S.S., C.W., M.S.), University of Würzburg, Biocenter, Germany; Institute of Science and Technology Austria (ISTA) (J.M., R.S.), Klosterneuburg, Austria; Oxford Autoimmune Neurology Group (S.R.I.), Nuffield Department of Clinical Neurosciences, University of Oxford, ; Department of Neurology (S.R.I.), John Radcliffe Hospital, Oxford University Hospitals, United Kingdom; and Departments of Neurology and Neurosciences (S.R.I.), Mayo Clinic Jacksonville, FL
| | - Stefan Hallermann
- From the Carl-Ludwig-Institute of Physiology (A.R.-J., F.G., T.K., S.M., S.H.), Faculty of Medicine, Leipzig University; Section Translational Neuroimmunology (J.S., C.G.), Department of Neurology, Jena University Hospital; Department of Biotechnology and Biophysics (S.S., C.W., M.S.), University of Würzburg, Biocenter, Germany; Institute of Science and Technology Austria (ISTA) (J.M., R.S.), Klosterneuburg, Austria; Oxford Autoimmune Neurology Group (S.R.I.), Nuffield Department of Clinical Neurosciences, University of Oxford, ; Department of Neurology (S.R.I.), John Radcliffe Hospital, Oxford University Hospitals, United Kingdom; and Departments of Neurology and Neurosciences (S.R.I.), Mayo Clinic Jacksonville, FL
| |
Collapse
|
10
|
Xiong A, Richmond JE, Kim H. Presynaptic neurons self-tune by inversely coupling neurotransmitter release with the abundance of CaV2 voltage-gated Ca 2+ channels. Proc Natl Acad Sci U S A 2024; 121:e2404969121. [PMID: 39172783 PMCID: PMC11363341 DOI: 10.1073/pnas.2404969121] [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: 03/09/2024] [Accepted: 07/12/2024] [Indexed: 08/24/2024] Open
Abstract
The abundance of CaV2 voltage-gated calcium channels is linked to presynaptic homeostatic plasticity (PHP), a process that recalibrates synaptic strength to maintain the stability of neural circuits. However, the molecular and cellular mechanisms governing PHP and CaV2 channels are not completely understood. Here, we uncover a previously not described form of PHP in Caenorhabditis elegans, revealing an inverse regulatory relationship between the efficiency of neurotransmitter release and the abundance of UNC-2/CaV2 channels. Gain-of-function unc-2SL(S240L) mutants, which carry a mutation analogous to the one causing familial hemiplegic migraine type 1 in humans, showed markedly reduced channel abundance despite increased channel functionality. Reducing synaptic release in these unc-2SL(S240L) mutants restored channel levels to those observed in wild-type animals. Conversely, loss-of-function unc-2DA(D726A) mutants, which harbor the D726A mutation in the channel pore, exhibited a marked increase in channel abundance. Enhancing synaptic release in unc-2DA mutants reversed this increase in channel levels. Importantly, this homeostatic regulation of UNC-2 channel levels is accompanied by the structural remodeling of the active zone (AZ); specifically, unc-2DA mutants, which exhibit increased channel abundance, showed parallel increases in select AZ proteins. Finally, our forward genetic screen revealed that WWP-1, a HECT family E3 ubiquitin ligase, is a key homeostatic mediator that removes UNC-2 from synapses. These findings highlight a self-tuning PHP regulating UNC-2/CaV2 channel abundance along with AZ reorganization, ensuring synaptic strength and stability.
Collapse
Affiliation(s)
- Ame Xiong
- Discipline of Cell Biology & Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| | - Janet E. Richmond
- Department of Biological Sciences, University of Illinois, Chicago, IL60607
| | - Hongkyun Kim
- Discipline of Cell Biology & Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| |
Collapse
|
11
|
Wang Y, Yang H, Li N, Wang L, Guo C, Ma W, Liu S, Peng C, Chen J, Song H, Chen H, Ma X, Yi J, Lian J, Kong W, Dong J, Tu X, Shah M, Tian X, Huang Z. A Novel Ubiquitin Ligase Adaptor PTPRN Suppresses Seizure Susceptibility through Endocytosis of Na V1.2 Sodium Channels. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400560. [PMID: 38874331 PMCID: PMC11304301 DOI: 10.1002/advs.202400560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/06/2024] [Indexed: 06/15/2024]
Abstract
Intrinsic plasticity, a fundamental process enabling neurons to modify their intrinsic properties, plays a crucial role in shaping neuronal input-output function and is implicated in various neurological and psychiatric disorders. Despite its importance, the underlying molecular mechanisms of intrinsic plasticity remain poorly understood. In this study, a new ubiquitin ligase adaptor, protein tyrosine phosphatase receptor type N (PTPRN), is identified as a regulator of intrinsic neuronal excitability in the context of temporal lobe epilepsy. PTPRN recruits the NEDD4 Like E3 Ubiquitin Protein Ligase (NEDD4L) to NaV1.2 sodium channels, facilitating NEDD4L-mediated ubiquitination, and endocytosis of NaV1.2. Knockout of PTPRN in hippocampal granule cells leads to augmented NaV1.2-mediated sodium currents and higher intrinsic excitability, resulting in increased seizure susceptibility in transgenic mice. Conversely, adeno-associated virus-mediated delivery of PTPRN in the dentate gyrus region decreases intrinsic excitability and reduces seizure susceptibility. Moreover, the present findings indicate that PTPRN exerts a selective modulation effect on voltage-gated sodium channels. Collectively, PTPRN plays a significant role in regulating intrinsic excitability and seizure susceptibility, suggesting a potential strategy for precise modulation of NaV1.2 channels' function.
Collapse
Affiliation(s)
- Yifan Wang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Hui Yang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Na Li
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Lili Wang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Chang Guo
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Weining Ma
- Department of NeurologyShengjing Hospital Affiliated to China Medical UniversityShenyang110022China
| | - Shiqi Liu
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Chao Peng
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Jiexin Chen
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Huifang Song
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Hedan Chen
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Xinyue Ma
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Jingyun Yi
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Jingjing Lian
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Weikaixin Kong
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Jie Dong
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Xinyu Tu
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Mala Shah
- UCL School of PharmacyUniversity College LondonLondonWC1N 1AXUK
| | - Xin Tian
- Department of NeurologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing Key Laboratory of NeurologyChongqing400016China
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| |
Collapse
|
12
|
Martus D, Williams SK, Pichi K, Mannebach-Götz S, Kaiser N, Wardas B, Fecher-Trost C, Meyer MR, Schmitz F, Beck A, Fairless R, Diem R, Flockerzi V, Belkacemi A. Cavβ3 Contributes to the Maintenance of the Blood-Brain Barrier and Alleviates Symptoms of Experimental Autoimmune Encephalomyelitis. Arterioscler Thromb Vasc Biol 2024; 44:1833-1851. [PMID: 38957986 DOI: 10.1161/atvbaha.124.321141] [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: 04/23/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Tight control of cytoplasmic Ca2+ concentration in endothelial cells is essential for the regulation of endothelial barrier function. Here, we investigated the role of Cavβ3, a subunit of voltage-gated Ca2+ (Cav) channels, in modulating Ca2+ signaling in brain microvascular endothelial cells (BMECs) and how this contributes to the integrity of the blood-brain barrier. METHODS We investigated the function of Cavβ3 in BMECs by Ca2+ imaging and Western blot, examined the endothelial barrier function in vitro and the integrity of the blood-brain barrier in vivo, and evaluated disease course after induction of experimental autoimmune encephalomyelitis in mice using Cavβ3-/- (Cavβ3-deficient) mice as controls. RESULTS We identified Cavβ3 protein in BMECs, but electrophysiological recordings did not reveal significant Cav channel activity. In vivo, blood-brain barrier integrity was reduced in the absence of Cavβ3. After induction of experimental autoimmune encephalomyelitis, Cavβ3-/- mice showed earlier disease onset with exacerbated clinical disability and increased T-cell infiltration. In vitro, the transendothelial resistance of Cavβ3-/- BMEC monolayers was lower than that of wild-type BMEC monolayers, and the organization of the junctional protein ZO-1 (zona occludens-1) was impaired. Thrombin stimulates inositol 1,4,5-trisphosphate-dependent Ca2+ release, which facilitates cell contraction and enhances endothelial barrier permeability via Ca2+-dependent phosphorylation of MLC (myosin light chain). These effects were more pronounced in Cavβ3-/- than in wild-type BMECs, whereas the differences were abolished in the presence of the MLCK (MLC kinase) inhibitor ML-7. Expression of Cacnb3 cDNA in Cavβ3-/- BMECs restored the wild-type phenotype. Coimmunoprecipitation and mass spectrometry demonstrated the association of Cavβ3 with inositol 1,4,5-trisphosphate receptor proteins. CONCLUSIONS Independent of its function as a subunit of Cav channels, Cavβ3 interacts with the inositol 1,4,5-trisphosphate receptor and is involved in the tight control of cytoplasmic Ca2+ concentration and Ca2+-dependent MLC phosphorylation in BMECs, and this role of Cavβ3 in BMECs contributes to blood-brain barrier integrity and attenuates the severity of experimental autoimmune encephalomyelitis disease.
Collapse
MESH Headings
- Animals
- Female
- Male
- Mice
- Blood-Brain Barrier/metabolism
- Calcium/metabolism
- Calcium Channels/metabolism
- Calcium Channels/genetics
- Calcium Signaling
- Capillary Permeability
- Cells, Cultured
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Endothelial Cells/metabolism
- Inositol 1,4,5-Trisphosphate Receptors/metabolism
- Inositol 1,4,5-Trisphosphate Receptors/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Myosin Light Chains/metabolism
- Myosin-Light-Chain Kinase/metabolism
- Myosin-Light-Chain Kinase/genetics
- Phosphorylation
Collapse
Affiliation(s)
- Damian Martus
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Sarah K Williams
- Neurologische Klinik, Universitätsklinikum Heidelberg, Germany (S.K.W., K.P., R.F., R.D.)
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany (R.F., S.K.W.)
| | - Kira Pichi
- Neurologische Klinik, Universitätsklinikum Heidelberg, Germany (S.K.W., K.P., R.F., R.D.)
| | - Stefanie Mannebach-Götz
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Nicolas Kaiser
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Barbara Wardas
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Claudia Fecher-Trost
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Markus R Meyer
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Frank Schmitz
- Institut für Anatomie und Zellbiologie (F.S.), Universität des Saarlandes, Homburg, Germany
| | - Andreas Beck
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Richard Fairless
- Neurologische Klinik, Universitätsklinikum Heidelberg, Germany (S.K.W., K.P., R.F., R.D.)
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany (R.F., S.K.W.)
| | - Ricarda Diem
- Neurologische Klinik, Universitätsklinikum Heidelberg, Germany (S.K.W., K.P., R.F., R.D.)
| | - Veit Flockerzi
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Anouar Belkacemi
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
- Now with Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, Germany (A. Belkacemi)
| |
Collapse
|
13
|
Perez-Miller S, Gomez K, Khanna R. Peptide and Peptidomimetic Inhibitors Targeting the Interaction of Collapsin Response Mediator Protein 2 with the N-Type Calcium Channel for Pain Relief. ACS Pharmacol Transl Sci 2024; 7:1916-1936. [PMID: 39022365 PMCID: PMC11249630 DOI: 10.1021/acsptsci.4c00181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/13/2024] [Accepted: 05/23/2024] [Indexed: 07/20/2024]
Abstract
Ion channels serve pleiotropic functions. Often found in complexes, their activities and functions are sculpted by auxiliary proteins. We discovered that collapsin response mediator protein 2 (CRMP2) is a binding partner and regulator of the N-type voltage-gated calcium channel (CaV2.2), a genetically validated contributor to chronic pain. Herein, we trace the discovery of a new peptidomimetic modulator of this interaction, starting from the identification and development of CBD3, a CRMP2-derived CaV binding domain peptide. CBD3 uncouples CRMP2-CaV2.2 binding to decrease CaV2.2 surface localization and calcium currents. These changes occur at presynaptic sites of nociceptive neurons and indeed, CBD3 ameliorates chronic pain in preclinical models. In pursuit of a CBD3 peptidomimetic, we exploited a unique approach to identify a dipeptide with low conformational flexibility and high solvent accessibility that anchors binding to CaV2.2. From a pharmacophore screen, we obtained CBD3063, a small-molecule that recapitulated CBD3's activity, reversing nociceptive behaviors in rodents of both sexes without sensory, affective, or cognitive effects. By disrupting the CRMP2-CaV2.2 interaction, CBD3063 exerts these effects indirectly through modulating CaV2.2 trafficking, supporting CRMP2 as an auxiliary subunit of CaV2.2. The parent peptide CBD3 was also found by us and others to have neuroprotective properties at postsynaptic sites, through N-methyl-d-aspartate receptor and plasmalemmal Na+/Ca2+ exchanger 3, potentially acting as an auxiliary subunit for these pathways as well. Our new compound is poised to address several open questions regarding CRMP2's role in regulating the CaV2.2 pathways to treat pain with the potential added benefit of neuroprotection.
Collapse
Affiliation(s)
- Samantha Perez-Miller
- Department
of Pharmacology & Therapeutics, College of Medicine, University of Florida, 1200 Newell Drive, ARB R5-234, Gainesville, Florida 32610-0267, United States
| | - Kimberly Gomez
- Department
of Pharmacology & Therapeutics, College of Medicine, University of Florida, 1200 Newell Drive, ARB R5-234, Gainesville, Florida 32610-0267, United States
| | - Rajesh Khanna
- Department
of Pharmacology & Therapeutics, College of Medicine, University of Florida, 1200 Newell Drive, ARB R5-234, Gainesville, Florida 32610-0267, United States
- Pain
and Addiction Therapeutics (PATH) Collaboratory, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
14
|
McCarthy CI, Kavalali ET. Nano-organization of synaptic calcium signaling. Biochem Soc Trans 2024; 52:1459-1471. [PMID: 38752834 PMCID: PMC11346461 DOI: 10.1042/bst20231385] [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: 03/04/2024] [Revised: 04/17/2024] [Accepted: 05/03/2024] [Indexed: 06/27/2024]
Abstract
Recent studies suggest an exquisite structural nano-organization within single synapses, where sites of evoked fusion - marked by clustering of synaptic vesicles, active zone proteins and voltage-gated calcium channels - are directly juxtaposed to postsynaptic receptor clusters within nanocolumns. This direct nanometer scale alignment between presynaptic fusion apparatus and postsynaptic receptors is thought to ensure the fidelity of synaptic signaling and possibly allow multiple distinct signals to occur without interference from each other within a single active zone. The functional specificity of this organization is made possible by the inherent nano-organization of calcium signals, where all the different calcium sources such as voltage-gated calcium channels, intracellular stores and store-operated calcium entry have dedicated local targets within their nanodomain to ensure precision of action. Here, we discuss synaptic nano-organization from the perspective of calcium signals, where some of the principal findings from early work in the 1980s continue to inspire current studies that exploit new genetic tools and super-resolution imaging technologies.
Collapse
Affiliation(s)
- Clara I. McCarthy
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, U.S.A
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240-7933, U.S.A
| | - Ege T. Kavalali
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, U.S.A
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240-7933, U.S.A
| |
Collapse
|
15
|
Trovò L, Kouvaros S, Schwenk J, Fernandez-Fernandez D, Fritzius T, Rem PD, Früh S, Gassmann M, Fakler B, Bischofberger J, Bettler B. Synaptotagmin-11 facilitates assembly of a presynaptic signaling complex in post-Golgi cargo vesicles. EMBO Rep 2024; 25:2610-2634. [PMID: 38698221 PMCID: PMC11169412 DOI: 10.1038/s44319-024-00147-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 05/05/2024] Open
Abstract
GABAB receptors (GBRs), the G protein-coupled receptors for GABA, regulate synaptic transmission throughout the brain. A main synaptic function of GBRs is the gating of Cav2.2-type Ca2+ channels. However, the cellular compartment where stable GBR/Cav2.2 signaling complexes form remains unknown. In this study, we demonstrate that the vesicular protein synaptotagmin-11 (Syt11) binds to both the auxiliary GBR subunit KCTD16 and Cav2.2 channels. Through these dual interactions, Syt11 recruits GBRs and Cav2.2 channels to post-Golgi vesicles, thus facilitating assembly of GBR/Cav2.2 signaling complexes. In addition, Syt11 stabilizes GBRs and Cav2.2 channels at the neuronal plasma membrane by inhibiting constitutive internalization. Neurons of Syt11 knockout mice exhibit deficits in presynaptic GBRs and Cav2.2 channels, reduced neurotransmitter release, and decreased GBR-mediated presynaptic inhibition, highlighting the critical role of Syt11 in the assembly and stable expression of GBR/Cav2.2 complexes. These findings support that Syt11 acts as a vesicular scaffold protein, aiding in the assembly of signaling complexes from low-abundance components within transport vesicles. This mechanism enables insertion of pre-assembled functional signaling units into the synaptic membrane.
Collapse
Affiliation(s)
- Luca Trovò
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Jochen Schwenk
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | | | | | - Simon Früh
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Martin Gassmann
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Bernd Fakler
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS Center for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation, Freiburg, Germany
| | | | - Bernhard Bettler
- Department of Biomedicine, University of Basel, Basel, Switzerland.
| |
Collapse
|
16
|
Belghazi M, Iborra C, Toutendji O, Lasserre M, Debanne D, Goaillard JM, Marquèze-Pouey B. High-Resolution Proteomics Unravel a Native Functional Complex of Cav1.3, SK3, and Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels in Midbrain Dopaminergic Neurons. Cells 2024; 13:944. [PMID: 38891076 PMCID: PMC11172389 DOI: 10.3390/cells13110944] [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: 03/04/2024] [Revised: 05/21/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
Pacemaking activity in substantia nigra dopaminergic neurons is generated by the coordinated activity of a variety of distinct somatodendritic voltage- and calcium-gated ion channels. We investigated whether these functional interactions could arise from a common localization in macromolecular complexes where physical proximity would allow for efficient interaction and co-regulations. For that purpose, we immunopurified six ion channel proteins involved in substantia nigra neuron autonomous firing to identify their molecular interactions. The ion channels chosen as bait were Cav1.2, Cav1.3, HCN2, HCN4, Kv4.3, and SK3 channel proteins, and the methods chosen to determine interactions were co-immunoprecipitation analyzed through immunoblot and mass spectrometry as well as proximity ligation assay. A macromolecular complex composed of Cav1.3, HCN, and SK3 channels was unraveled. In addition, novel potential interactions between SK3 channels and sclerosis tuberous complex (Tsc) proteins, inhibitors of mTOR, and between HCN4 channels and the pro-degenerative protein Sarm1 were uncovered. In order to demonstrate the presence of these molecular interactions in situ, we used proximity ligation assay (PLA) imaging on midbrain slices containing the substantia nigra, and we could ascertain the presence of these protein complexes specifically in substantia nigra dopaminergic neurons. Based on the complementary functional role of the ion channels in the macromolecular complex identified, these results suggest that such tight interactions could partly underly the robustness of pacemaking in dopaminergic neurons.
Collapse
Affiliation(s)
- Maya Belghazi
- CRN2M Centre de Recherche Neurobiologie-Neurophysiologie, CNRS, UMR7286, Aix-Marseille Université, 13015 Marseille, France;
- Institut de Microbiologie de la Méditerranée (IMM), CNRS, Aix-Marseille Université, 13009 Marseille, France
| | - Cécile Iborra
- Ion Channel and Synaptic Neurobiology, INSERM, UMR1072, Aix-Marseille Université, 13015 Marseille, France; (C.I.); (O.T.); (M.L.); (D.D.); (J.-M.G.)
| | - Ophélie Toutendji
- Ion Channel and Synaptic Neurobiology, INSERM, UMR1072, Aix-Marseille Université, 13015 Marseille, France; (C.I.); (O.T.); (M.L.); (D.D.); (J.-M.G.)
| | - Manon Lasserre
- Ion Channel and Synaptic Neurobiology, INSERM, UMR1072, Aix-Marseille Université, 13015 Marseille, France; (C.I.); (O.T.); (M.L.); (D.D.); (J.-M.G.)
| | - Dominique Debanne
- Ion Channel and Synaptic Neurobiology, INSERM, UMR1072, Aix-Marseille Université, 13015 Marseille, France; (C.I.); (O.T.); (M.L.); (D.D.); (J.-M.G.)
| | - Jean-Marc Goaillard
- Ion Channel and Synaptic Neurobiology, INSERM, UMR1072, Aix-Marseille Université, 13015 Marseille, France; (C.I.); (O.T.); (M.L.); (D.D.); (J.-M.G.)
- Institut de Neurosciences de la Timone, CNRS, Aix-Marseille Université, 13005 Marseille, France
| | - Béatrice Marquèze-Pouey
- Ion Channel and Synaptic Neurobiology, INSERM, UMR1072, Aix-Marseille Université, 13015 Marseille, France; (C.I.); (O.T.); (M.L.); (D.D.); (J.-M.G.)
| |
Collapse
|
17
|
Medeiros AT, Gratz S, Delgado A, Ritt J, O’Connor-Giles KM. Ca 2+ channel and active zone protein abundance intersects with input-specific synapse organization to shape functional synaptic diversity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.02.535290. [PMID: 37034654 PMCID: PMC10081318 DOI: 10.1101/2023.04.02.535290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Synaptic heterogeneity is a hallmark of nervous systems that enables complex and adaptable communication in neural circuits. To understand circuit function, it is thus critical to determine the factors that contribute to the functional diversity of synapses. We investigated the contributions of voltage-gated calcium channel (VGCC) abundance, spatial organization, and subunit composition to synapse diversity among and between synapses formed by two closely related Drosophila glutamatergic motor neurons with distinct neurotransmitter release probabilities (Pr). Surprisingly, VGCC levels are highly predictive of heterogeneous Pr among individual synapses of either low- or high-Pr inputs, but not between inputs. We find that the same number of VGCCs are more densely organized at high-Pr synapses, consistent with tighter VGCC-synaptic vesicle coupling. We generated endogenously tagged lines to investigate VGCC subunits in vivo and found that the α2δ-3 subunit Straightjacket along with the CAST/ELKS active zone (AZ) protein Bruchpilot, both key regulators of VGCCs, are less abundant at high-Pr inputs, yet positively correlate with Pr among synapses formed by either input. Consistently, both Straightjacket and Bruchpilot levels are dynamically increased across AZs of both inputs when neurotransmitter release is potentiated to maintain stable communication following glutamate receptor inhibition. Together, these findings suggest a model in which VGCC and AZ protein abundance intersects with input-specific spatial and molecular organization to shape the functional diversity of synapses.
Collapse
Affiliation(s)
- A. T. Medeiros
- Neuroscience Graduate Training Program, Brown University, Providence, RI
| | - S.J. Gratz
- Department of Neuroscience, Brown University, Providence, RI
| | - A. Delgado
- Department of Neuroscience, Brown University, Providence, RI
| | - J.T. Ritt
- Department of Neuroscience, Brown University, Providence, RI
- Carney Institute for Brain Science, Brown University, Providence, RI
| | - Kate M. O’Connor-Giles
- Neuroscience Graduate Training Program, Brown University, Providence, RI
- Department of Neuroscience, Brown University, Providence, RI
- Carney Institute for Brain Science, Brown University, Providence, RI
| |
Collapse
|
18
|
Koppensteiner P, Bhandari P, Önal C, Borges-Merjane C, Le Monnier E, Roy U, Nakamura Y, Sadakata T, Sanbo M, Hirabayashi M, Rhee J, Brose N, Jonas P, Shigemoto R. GABA B receptors induce phasic release from medial habenula terminals through activity-dependent recruitment of release-ready vesicles. Proc Natl Acad Sci U S A 2024; 121:e2301449121. [PMID: 38346189 PMCID: PMC10895368 DOI: 10.1073/pnas.2301449121] [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] [Received: 01/26/2023] [Accepted: 01/03/2024] [Indexed: 02/15/2024] Open
Abstract
GABAB receptor (GBR) activation inhibits neurotransmitter release in axon terminals in the brain, except in medial habenula (MHb) terminals, which show robust potentiation. However, mechanisms underlying this enigmatic potentiation remain elusive. Here, we report that GBR activation on MHb terminals induces an activity-dependent transition from a facilitating, tonic to a depressing, phasic neurotransmitter release mode. This transition is accompanied by a 4.1-fold increase in readily releasable vesicle pool (RRP) size and a 3.5-fold increase of docked synaptic vesicles (SVs) at the presynaptic active zone (AZ). Strikingly, the depressing phasic release exhibits looser coupling distance than the tonic release. Furthermore, the tonic and phasic release are selectively affected by deletion of synaptoporin (SPO) and Ca2+-dependent activator protein for secretion 2 (CAPS2), respectively. SPO modulates augmentation, the short-term plasticity associated with tonic release, and CAPS2 retains the increased RRP for initial responses in phasic response trains. The cytosolic protein CAPS2 showed a SV-associated distribution similar to the vesicular transmembrane protein SPO, and they were colocalized in the same terminals. We developed the "Flash and Freeze-fracture" method, and revealed the release of SPO-associated vesicles in both tonic and phasic modes and activity-dependent recruitment of CAPS2 to the AZ during phasic release, which lasted several minutes. Overall, these results indicate that GBR activation translocates CAPS2 to the AZ along with the fusion of CAPS2-associated SVs, contributing to persistency of the RRP increase. Thus, we identified structural and molecular mechanisms underlying tonic and phasic neurotransmitter release and their transition by GBR activation in MHb terminals.
Collapse
Affiliation(s)
| | - Pradeep Bhandari
- Institute of Science and Technology Austria, Klosterneuburg3400, Austria
| | - Cihan Önal
- Institute of Science and Technology Austria, Klosterneuburg3400, Austria
| | | | - Elodie Le Monnier
- Institute of Science and Technology Austria, Klosterneuburg3400, Austria
| | - Utsa Roy
- Institute of Science and Technology Austria, Klosterneuburg3400, Austria
| | - Yukihiro Nakamura
- Department of Pharmacology, Jikei University School of Medicine, Nishishinbashi, Minato-ku, Tokyo105-8461, Japan
| | - Tetsushi Sadakata
- Advanced Scientific Research Leaders Development Unit, Gunma University Graduate School of Medicine, Maebashi, Gunma371-8511, Japan
| | - Makoto Sanbo
- Section of Mammalian Transgenesis, National Institute for Physiological Sciences, Okazaki444-8585, Japan
| | - Masumi Hirabayashi
- Section of Mammalian Transgenesis, National Institute for Physiological Sciences, Okazaki444-8585, Japan
| | - JeongSeop Rhee
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen37077, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen37077, Germany
| | - Peter Jonas
- Institute of Science and Technology Austria, Klosterneuburg3400, Austria
| | - Ryuichi Shigemoto
- Institute of Science and Technology Austria, Klosterneuburg3400, Austria
| |
Collapse
|
19
|
Flockerzi V, Fakler B. TR(i)P Goes On: Auxiliary TRP Channel Subunits? Circ Res 2024; 134:346-350. [PMID: 38359093 DOI: 10.1161/circresaha.123.323178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 11/28/2023] [Indexed: 02/17/2024]
Abstract
Transient receptor potential (TRP) cation channels are a diverse family of channels whose members play prominent roles as cellular sensors and effectors. The important role of TRP channels (and mechanosensitive piezo channels) in the complex interaction of our senses with the environment was underlined by the award of the Nobel Prize in Physiology or Medicine to 2 pioneers in this field, David Julius and Ardem Patapoutian. There are many competent and comprehensive reviews on many aspects of the TRP channels, and there is no intention to expand on them. Rather, after an introduction to the nomenclature, the molecular architecture of native TRP channel/protein complexes in vivo will be summarized using TRP channels of the canonical transient receptor potential subfamily as an example. This molecular architecture provides the basis for the signatures of native canonical transient receptor potential currents and their control by endogenous modulators and potential drugs.
Collapse
Affiliation(s)
- Veit Flockerzi
- Experimental and Clinical Pharmacology and Toxicology, Saarland University, Homburg, Germany (V.F.)
| | - Bernd Fakler
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany (B.F.)
| |
Collapse
|
20
|
Li J, Veeraraghavan P, Young SM. Ca V 2.1 α 1 subunit motifs that control presynaptic Ca V 2.1 subtype abundance are distinct from Ca V 2.1 preference. J Physiol 2024; 602:485-506. [PMID: 38155373 PMCID: PMC10872416 DOI: 10.1113/jp284957] [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: 04/28/2023] [Accepted: 11/30/2023] [Indexed: 12/30/2023] Open
Abstract
Presynaptic voltage-gated Ca2+ channel (CaV ) subtype abundance at mammalian synapses regulates synaptic transmission in health and disease. In the mammalian central nervous system (CNS), most presynaptic terminals are CaV 2.1 dominant with a developmental reduction in CaV 2.2 and CaV 2.3 levels, and CaV 2 subtype levels are altered in various diseases. However, the molecular mechanisms controlling presynaptic CaV 2 subtype levels are largely unsolved. Because the CaV 2 α1 subunit cytoplasmic regions contain varying levels of sequence conservation, these regions are proposed to control presynaptic CaV 2 subtype preference and abundance. To investigate the potential role of these regions, we expressed chimeric CaV 2.1 α1 subunits containing swapped motifs with the CaV 2.2 and CaV 2.3 α1 subunit on a CaV 2.1/CaV 2.2 null background at the calyx of Held presynaptic terminals. We found that expression of CaV 2.1 α1 subunit chimeras containing the CaV 2.3 loop II-III region or cytoplasmic C-terminus (CT) resulted in a large reduction of presynaptic Ca2+ currents compared to the CaV 2.1 α1 subunit. However, the Ca2+ current sensitivity to the CaV 2.1 blocker agatoxin-IVA was the same between the chimeras and the CaV 2.1 α1 subunit. Additionally, we found no reduction in presynaptic Ca2+ currents with CaV 2.1/2.2 cytoplasmic CT chimeras. We conclude that the motifs in the CaV 2.1 loop II-III and CT do not individually regulate CaV 2.1 preference, although these motifs control CaV 2.1 levels and the CaV 2.3 CT contains motifs that negatively regulate presynaptic CaV 2.3 levels. We propose that the motifs controlling presynaptic CaV 2.1 preference are distinct from those regulating CaV 2.1 levels and may act synergistically to impact pathways regulating CaV 2.1 preference and abundance. KEY POINTS: Presynaptic CaV 2 subtype abundance regulates neuronal circuit properties, although the mechanisms regulating presynaptic CaV 2 subtype abundance and preference remain enigmatic. The CaV α1 subunit determines subtype and contains multiple motifs implicated in regulating presynaptic subtype abundance and preference. The CaV 2.1 α1 subunit domain II-III loop and cytoplasmic C-terminus are positive regulators of presynaptic CaV 2.1 abundance but do not regulate preference. The CaV 2.3 α1 subunit cytoplasmic C-terminus negatively regulates presynaptic CaV 2 subtype abundance but not preference, whereas the CaV 2.2 α1 subunit cytoplasmic C-terminus is not a key regulator of presynaptic CaV 2 subtype abundance or preference. The CaV 2 α1 subunit motifs determining the presynaptic CaV 2 preference are distinct from abundance.
Collapse
Affiliation(s)
- Jianing Li
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
- Cell Developmental Biology Graduate Program, University of Iowa, Iowa City, IA 52242, USA
| | | | - Samuel M. Young
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
- Department of Otolaryngology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
21
|
Lory P. The good company of T-type calcium channels : Commentary on: "T-type calcium channelosome," a review by N. Weiss and G. W. Zamponi. Pflugers Arch 2024; 476:149-150. [PMID: 38191755 DOI: 10.1007/s00424-023-02903-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 12/21/2023] [Accepted: 12/28/2023] [Indexed: 01/10/2024]
Affiliation(s)
- Philippe Lory
- IGF, Univ Montpellier, CNRS, INSERM, Montpellier, France.
- LabEx 'Ion Channel Science and Therapeutics', Montpellier, France.
- Institut de Génomique Fonctionnelle - CNRS, 141 rue de la Cardonille, 34094, Montpellier, France.
| |
Collapse
|
22
|
Weiss N, Zamponi GW. The T-type calcium channelosome. Pflugers Arch 2024; 476:163-177. [PMID: 38036777 DOI: 10.1007/s00424-023-02891-z] [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: 10/27/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
T-type calcium channels perform crucial physiological roles across a wide spectrum of tissues, spanning both neuronal and non-neuronal system. For instance, they serve as pivotal regulators of neuronal excitability, contribute to cardiac pacemaking, and mediate the secretion of hormones. These functions significantly hinge upon the intricate interplay of T-type channels with interacting proteins that modulate their expression and function at the plasma membrane. In this review, we offer a panoramic exploration of the current knowledge surrounding these T-type channel interactors, and spotlight certain aspects of their potential for drug-based therapeutic intervention.
Collapse
Affiliation(s)
- Norbert Weiss
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
23
|
Varadi G. Mechanism of Analgesia by Gabapentinoid Drugs: Involvement of Modulation of Synaptogenesis and Trafficking of Glutamate-Gated Ion Channels. J Pharmacol Exp Ther 2024; 388:121-133. [PMID: 37918854 DOI: 10.1124/jpet.123.001669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023] Open
Abstract
Gabapentinoids have clinically been used for treating epilepsy, neuropathic pain, and several other neurologic disorders for >30 years; however, the definitive molecular mechanism responsible for their therapeutic actions remained uncertain. The conventional pharmacological observation regarding their efficacy in chronic pain modulation is the weakening of glutamate release at presynaptic terminals in the spinal cord. While the α2/δ-1 subunit of voltage-gated calcium channels (VGCCs) has been identified as the primary drug receptor for gabapentinoids, the lack of consistent effect of this drug class on VGCC function is indicative of a minor role in regulating this ion channel's activity. The current review targets the efficacy and mechanism of gabapentinoids in treating chronic pain. The discovery of interaction of α2/δ-1 with thrombospondins established this protein as a major synaptogenic neuronal receptor for thrombospondins. Other findings identified α2/δ-1 as a powerful regulator of N-methyl-D-aspartate receptor (NMDA) and alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) by potentiating the synaptic expression, a putative pathophysiological mechanism of neuropathic pain. Further, the interdependent interactions between thrombospondin and α2/δ-1 contribute to chronic pain states, while gabapentinoid ligands efficaciously reverse such pain conditions. Gabapentin normalizes and even blocks NMDAR and AMPAR synaptic targeting and activity elicited by nerve injury. SIGNIFICANCE STATEMENT: Gabapentinoid drugs are used to treat various neurological conditions including chronic pain. In chronic pain states, gene expression of cacnα2/δ-1 and thrombospondins are upregulated and promote aberrant excitatory synaptogenesis. The complex trait of protein associations that involve interdependent interactions between α2/δ-1 and thrombospondins, further, association of N-methyl-D-aspartate receptor and alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor with the C-tail of α2/δ-1, constitutes a macromolecular signaling complex that forms the crucial elements for the pharmacological mode of action of gabapentinoids.
Collapse
|
24
|
Casas M, Dickson EJ. Channels, Transporters, and Receptors at Membrane Contact Sites. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2024; 7:25152564241305593. [PMID: 39742107 PMCID: PMC11686659 DOI: 10.1177/25152564241305593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 11/21/2024] [Indexed: 01/03/2025]
Abstract
Membrane contact sites (MCSs) are specialized regions where two or more organelle membranes come into close apposition, typically separated by only 10-30 nm, while remaining distinct and unfused. These sites play crucial roles in cellular homeostasis, signaling, and metabolism. This review focuses on ion channels, transporters, and receptors localized to MCSs, with particular emphasis on those associated with the plasma membrane and endoplasmic reticulum (ER). We discuss the molecular composition and functional significance of these proteins in shaping both organelle and cellular functions, highlighting their importance in excitable cells and their influence on intracellular calcium signaling. Key MCSs examined include ER-plasma membrane, ER-mitochondria, and ER-lysosome contacts. This review addresses our current knowledge of the ion channels found within these contacts, the dynamic regulation of MCSs, their importance in various physiological processes, and their potential implications in pathological conditions.
Collapse
Affiliation(s)
- Maria Casas
- Department of Physiology and Membrane Biology, University of California, Davis, CA, USA
| | - Eamonn James Dickson
- Department of Physiology and Membrane Biology, University of California, Davis, CA, USA
| |
Collapse
|
25
|
Sampedro-Castañeda M, Baltussen LL, Lopes AT, Qiu Y, Sirvio L, Mihaylov SR, Claxton S, Richardson JC, Lignani G, Ultanir SK. Epilepsy-linked kinase CDKL5 phosphorylates voltage-gated calcium channel Cav2.3, altering inactivation kinetics and neuronal excitability. Nat Commun 2023; 14:7830. [PMID: 38081835 PMCID: PMC10713615 DOI: 10.1038/s41467-023-43475-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 11/09/2023] [Indexed: 12/18/2023] Open
Abstract
Developmental and epileptic encephalopathies (DEEs) are a group of rare childhood disorders characterized by severe epilepsy and cognitive deficits. Numerous DEE genes have been discovered thanks to advances in genomic diagnosis, yet putative molecular links between these disorders are unknown. CDKL5 deficiency disorder (CDD, DEE2), one of the most common genetic epilepsies, is caused by loss-of-function mutations in the brain-enriched kinase CDKL5. To elucidate CDKL5 function, we looked for CDKL5 substrates using a SILAC-based phosphoproteomic screen. We identified the voltage-gated Ca2+ channel Cav2.3 (encoded by CACNA1E) as a physiological target of CDKL5 in mice and humans. Recombinant channel electrophysiology and interdisciplinary characterization of Cav2.3 phosphomutant mice revealed that loss of Cav2.3 phosphorylation leads to channel gain-of-function via slower inactivation and enhanced cholinergic stimulation, resulting in increased neuronal excitability. Our results thus show that CDD is partly a channelopathy. The properties of unphosphorylated Cav2.3 closely resemble those described for CACNA1E gain-of-function mutations causing DEE69, a disorder sharing clinical features with CDD. We show that these two single-gene diseases are mechanistically related and could be ameliorated with Cav2.3 inhibitors.
Collapse
Affiliation(s)
| | - Lucas L Baltussen
- Kinases and Brain Development Lab, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Laboratory for the Research of Neurodegenerative Diseases (VIB-KU Leuven), Department of Neurosciences, ON5 Herestraat 49, 3000, Leuven, Belgium
| | - André T Lopes
- Kinases and Brain Development Lab, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Yichen Qiu
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square House, London, WC1N 3BG, UK
| | - Liina Sirvio
- Kinases and Brain Development Lab, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Simeon R Mihaylov
- Kinases and Brain Development Lab, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Suzanne Claxton
- Kinases and Brain Development Lab, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Jill C Richardson
- Neuroscience, MSD Research Laboratories, 120 Moorgate, London, EC2M 6UR, UK
| | - Gabriele Lignani
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square House, London, WC1N 3BG, UK
| | - Sila K Ultanir
- Kinases and Brain Development Lab, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
26
|
Emperador-Melero J, Andersen JW, Metzbower SR, Levy AD, Dharmasri PA, de Nola G, Blanpied TA, Kaeser PS. Molecular definition of distinct active zone protein machineries for Ca 2+ channel clustering and synaptic vesicle priming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.27.564439. [PMID: 37961089 PMCID: PMC10634917 DOI: 10.1101/2023.10.27.564439] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Action potentials trigger neurotransmitter release with minimal delay. Active zones mediate this temporal precision by co-organizing primed vesicles with CaV2 Ca2+ channels. The presumed model is that scaffolding proteins directly tether primed vesicles to CaV2s. We find that CaV2 clustering and vesicle priming are executed by separate machineries. At hippocampal synapses, CaV2 nanoclusters are positioned at variable distances from those of the priming protein Munc13. The active zone organizer RIM anchors both proteins, but distinct interaction motifs independently execute these functions. In heterologous cells, Liprin-α and RIM from co-assemblies that are separate from CaV2-organizing complexes upon co-transfection. At synapses, Liprin-α1-4 knockout impairs vesicle priming, but not CaV2 clustering. The cell adhesion protein PTPσ recruits Liprin-α, RIM and Munc13 into priming complexes without co-clustering of CaV2s. We conclude that active zones consist of distinct complexes to organize CaV2s and vesicle priming, and Liprin-α and PTPσ specifically support priming site assembly.
Collapse
Affiliation(s)
| | | | - Sarah R. Metzbower
- Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| | - Aaron D. Levy
- Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| | - Poorna A. Dharmasri
- Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| | | | - Thomas A. Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| | | |
Collapse
|
27
|
Li J, Veeraraghavan P, Young SM. CaV2.1 α1 subunit motifs that control presynaptic CaV2.1 subtype abundance are distinct from CaV2.1 preference. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.28.538778. [PMID: 37162941 PMCID: PMC10168310 DOI: 10.1101/2023.04.28.538778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Presynaptic voltage-gated Ca2+ channels (CaV) subtype abundance at mammalian synapses regulates synaptic transmission in health and disease. In the mammalian central nervous system, most presynaptic terminals are CaV2.1 dominant with a developmental reduction in CaV2.2 and CaV2.3 levels, and CaV2 subtype levels are altered in various diseases. However, the molecular mechanisms controlling presynaptic CaV2 subtype levels are largely unsolved. Since the CaV2 α1 subunit cytoplasmic regions contain varying levels of sequence conservation, these regions are proposed to control presynaptic CaV2 subtype preference and abundance. To investigate the potential role of these regions, we expressed chimeric CaV2.1 α1subunits containing swapped motifs with the CaV2.2 and CaV2.3 α1 subunit on a CaV2.1/CaV2.2 null background at the calyx of Held presynaptic terminal. We found that expression of CaV2.1 α1 subunit chimeras containing the CaV2.3 loop II-III region or cytoplasmic C-terminus (CT) resulted in a large reduction of presynaptic Ca2+ currents compared to the CaV2.1 α1 subunit. However, the Ca2+ current sensitivity to the CaV2.1 blocker Agatoxin-IVA, was the same between the chimeras and the CaV2.1 α1 subunit. Additionally, we found no reduction in presynaptic Ca2+ currents with CaV2.1/2.2 cytoplasmic CT chimeras. We conclude that the motifs in the CaV2.1 loop II-III and CT do not individually regulate CaV2.1 preference, but these motifs control CaV2.1 levels and the CaV2.3 CT contains motifs that negatively regulate presynaptic CaV2.3 levels. We propose that the motifs controlling presynaptic CaV2.1 preference are distinct from those regulating CaV2.1 levels and may act synergistically to impact pathways regulating CaV2.1 preference and abundance.
Collapse
|
28
|
Zhang J, Zeng W, Han Y, Lee WR, Liou J, Jiang Y. Lysosomal LAMP proteins regulate lysosomal pH by direct inhibition of the TMEM175 channel. Mol Cell 2023; 83:2524-2539.e7. [PMID: 37390818 PMCID: PMC10528928 DOI: 10.1016/j.molcel.2023.06.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 04/03/2023] [Accepted: 06/02/2023] [Indexed: 07/02/2023]
Abstract
Maintaining a highly acidic lysosomal pH is central to cellular physiology. Here, we use functional proteomics, single-particle cryo-EM, electrophysiology, and in vivo imaging to unravel a key biological function of human lysosome-associated membrane proteins (LAMP-1 and LAMP-2) in regulating lysosomal pH homeostasis. Despite being widely used as a lysosomal marker, the physiological functions of the LAMP proteins have long been overlooked. We show that LAMP-1 and LAMP-2 directly interact with and inhibit the activity of the lysosomal cation channel TMEM175, a key player in lysosomal pH homeostasis implicated in Parkinson's disease. This LAMP inhibition mitigates the proton conduction of TMEM175 and facilitates lysosomal acidification to a lower pH environment crucial for optimal hydrolase activity. Disrupting the LAMP-TMEM175 interaction alkalinizes the lysosomal pH and compromises the lysosomal hydrolytic function. In light of the ever-increasing importance of lysosomes to cellular physiology and diseases, our data have widespread implications for lysosomal biology.
Collapse
Affiliation(s)
- Jiyuan Zhang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Weizhong Zeng
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Howard Hughes Medical Institute at University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yan Han
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wan-Ru Lee
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jen Liou
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Youxing Jiang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Howard Hughes Medical Institute at University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
29
|
Reyes Fernandez PC, Wright CS, Farach-Carson MC, Thompson WR. Examining Mechanisms for Voltage-Sensitive Calcium Channel-Mediated Secretion Events in Bone Cells. Calcif Tissue Int 2023; 113:126-142. [PMID: 37261463 PMCID: PMC11008533 DOI: 10.1007/s00223-023-01097-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/16/2023] [Indexed: 06/02/2023]
Abstract
In addition to their well-described functions in cell excitability, voltage-sensitive calcium channels (VSCCs) serve a critical role in calcium (Ca2+)-mediated secretion of pleiotropic paracrine and endocrine factors, including those produced in bone. Influx of Ca2+ through VSCCs activates intracellular signaling pathways to modulate a variety of cellular processes that include cell proliferation, differentiation, and bone adaptation in response to mechanical stimuli. Less well understood is the role of VSCCs in the control of bone and calcium homeostasis mediated through secreted factors. In this review, we discuss the various functions of VSCCs in skeletal cells as regulators of Ca2+ dynamics and detail how these channels might control the release of bioactive factors from bone cells. Because VSCCs are druggable, a better understanding of the multiple functions of these channels in the skeleton offers the opportunity for developing new therapies to enhance and maintain bone and to improve systemic health.
Collapse
Affiliation(s)
- Perla C Reyes Fernandez
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Christian S Wright
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Mary C Farach-Carson
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
- Departments of BioSciences and Bioengineering, Rice University, Houston, TX, 77005, USA
| | - William R Thompson
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA.
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
30
|
Hua J, Garcia de Paco E, Linck N, Maurice T, Desrumaux C, Manoury B, Rassendren F, Ulmann L. Microglial P2X4 receptors promote ApoE degradation and contribute to memory deficits in Alzheimer's disease. Cell Mol Life Sci 2023; 80:138. [PMID: 37145189 PMCID: PMC10163120 DOI: 10.1007/s00018-023-04784-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/06/2023]
Abstract
Numerous evidences support that microglia contributes to the progression of Alzheimer's disease. P2X4 receptors are ATP-gated channels with high calcium permeability, which are de novo expressed in a subset of reactive microglia associated with various pathological contexts, contributing to microglial functions. P2X4 receptors are mainly localized in lysosomes and trafficking to the plasma membrane is tightly regulated. Here, we investigated the role of P2X4 in the context of Alzheimer's disease (AD). Using proteomics, we identified Apolipoprotein E (ApoE) as a specific P2X4 interacting protein. We found that P2X4 regulates lysosomal cathepsin B (CatB) activity promoting ApoE degradation; P2rX4 deletion results in higher amounts of intracellular and secreted ApoE in both bone-marrow-derived macrophage (BMDM) and microglia from APPswe/PSEN1dE9 brain. In both human AD brain and APP/PS1 mice, P2X4 and ApoE are almost exclusively expressed in plaque-associated microglia. In 12-month-old APP/PS1 mice, genetic deletion of P2rX4 reverses topographical and spatial memory impairment and reduces amount of soluble small aggregates of Aß1-42 peptide, while no obvious alteration of plaque-associated microglia characteristics is observed. Our results support that microglial P2X4 promotes lysosomal ApoE degradation, indirectly altering Aß peptide clearance, which in turn might promotes synaptic dysfunctions and cognitive deficits. Our findings uncover a specific interplay between purinergic signaling, microglial ApoE, soluble Aß (sAß) species and cognitive deficits associated with AD.
Collapse
Affiliation(s)
- Jennifer Hua
- IGF, Univ Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Montpellier, France
| | - Elvira Garcia de Paco
- IGF, Univ Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Montpellier, France
| | - Nathalie Linck
- IGF, Univ Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Montpellier, France
| | - Tangui Maurice
- MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France
| | | | - Bénédicte Manoury
- Institut Necker Enfants Malades, INSERM, CNRS, Université de Paris, Paris, France
| | - François Rassendren
- IGF, Univ Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Montpellier, France
| | - Lauriane Ulmann
- IGF, Univ Montpellier, CNRS, INSERM, Montpellier, France.
- LabEx Ion Channel Science and Therapeutics, Montpellier, France.
| |
Collapse
|
31
|
Leyva-Leyva M, Sandoval A, Morales-Lázaro SL, Corzo-López A, Felix R, González-Ramírez R. Identification of Dp140 and α1-syntrophin as novel molecular interactors of the neuronal Ca V2.1 channel. Pflugers Arch 2023; 475:595-606. [PMID: 36964781 DOI: 10.1007/s00424-023-02803-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/21/2023] [Accepted: 03/06/2023] [Indexed: 03/26/2023]
Abstract
The primary function of dystrophin is to form a link between the cytoskeleton and the extracellular matrix. In addition to this crucial structural function, dystrophin also plays an essential role in clustering and organizing several signaling proteins, including ion channels. Proteomic analysis of the whole rodent brain has stressed the role of some components of the dystrophin-associated glycoprotein complex (DGC) as potential interacting proteins of the voltage-gated Ca2+ channels of the CaV2 subfamily. The interaction of CaV2 with signaling and scaffolding proteins, such as the DGC components, may influence their function, stability, and location in neurons. This work aims to study the interaction between dystrophin and CaV2.1. Our immunoprecipitation data showed the presence of a complex formed by CaV2.1, CaVα2δ-1, CaVβ4e, Dp140, and α1-syntrophin in the brain. Furthermore, proximity ligation assays (PLA) showed that CaV2.1 and CaVα2δ-1 interact with dystrophin in the hippocampus and cerebellum. Notably, Dp140 and α1-syntrophin increase CaV2.1 protein stability, half-life, permanence in the plasma membrane, and current density through recombinant CaV2.1 channels. Therefore, we have identified the Dp140 and α1-syntrophin as novel interaction partners of CaV2.1 channels in the mammalian brain. Consistent with previous findings, our work provides evidence of the role of DGC in anchoring and clustering CaV channels in a macromolecular complex.
Collapse
Affiliation(s)
- Margarita Leyva-Leyva
- Department of Molecular Biology and Histocompatibility, "Dr. Manuel Gea González" General Hospital, Mexico City, Mexico
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Universidad Nacional Autónoma de México (UNAM), Ciudad de Mexico, México
| | - Alejandro Sandoval
- School of Medicine FES Iztacala, National Autonomous University of México (UNAM), Tlalnepantla, Mexico
| | - Sara Luz Morales-Lázaro
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de Mexico, México
| | - Alejandra Corzo-López
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Ricardo Felix
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico.
| | - Ricardo González-Ramírez
- Department of Molecular Biology and Histocompatibility, "Dr. Manuel Gea González" General Hospital, Mexico City, Mexico.
| |
Collapse
|
32
|
Mayo S, Gómez-Manjón I, Marco-Hernández AV, Fernández-Martínez FJ, Camacho A, Martínez F. N-Type Ca Channel in Epileptic Syndromes and Epilepsy: A Systematic Review of Its Genetic Variants. Int J Mol Sci 2023; 24:6100. [PMID: 37047073 PMCID: PMC10094502 DOI: 10.3390/ijms24076100] [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: 02/20/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
N-type voltage-gated calcium channel controls the release of neurotransmitters from neurons. The association of other voltage-gated calcium channels with epilepsy is well-known. The association of N-type voltage-gated calcium channels and pain has also been established. However, the relationship between this type of calcium channel and epilepsy has not been specifically reviewed. Therefore, the present review systematically summarizes existing publications regarding the genetic associations between N-type voltage-dependent calcium channel and epilepsy.
Collapse
Affiliation(s)
- Sonia Mayo
- Genetics and Inheritance Research Group, Instituto de Investigación Sanitaria Hospital 12 de Octubre, 28041 Madrid, Spain
- Department of Genetics, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Irene Gómez-Manjón
- Genetics and Inheritance Research Group, Instituto de Investigación Sanitaria Hospital 12 de Octubre, 28041 Madrid, Spain
- Department of Genetics, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Ana Victoria Marco-Hernández
- Neuropediatric Unit, Hospital Universitario Doctor Peset, 46017 Valencia, Spain
- Translational Research in Genetics, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Francisco Javier Fernández-Martínez
- Genetics and Inheritance Research Group, Instituto de Investigación Sanitaria Hospital 12 de Octubre, 28041 Madrid, Spain
- Department of Genetics, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Ana Camacho
- Division of Pediatric Neurology, Department of Neurology, Hospital Universitario 12 de Octubre, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Francisco Martínez
- Translational Research in Genetics, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Genomic Unit, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Genetics Unit, Hospital Universitario y Politecnico La Fe, 46026 Valencia, Spain
| |
Collapse
|
33
|
Cunningham KL, Littleton JT. Mechanisms controlling the trafficking, localization, and abundance of presynaptic Ca 2+ channels. Front Mol Neurosci 2023; 15:1116729. [PMID: 36710932 PMCID: PMC9880069 DOI: 10.3389/fnmol.2022.1116729] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 12/26/2022] [Indexed: 01/14/2023] Open
Abstract
Voltage-gated Ca2+ channels (VGCCs) mediate Ca2+ influx to trigger neurotransmitter release at specialized presynaptic sites termed active zones (AZs). The abundance of VGCCs at AZs regulates neurotransmitter release probability (Pr ), a key presynaptic determinant of synaptic strength. Given this functional significance, defining the processes that cooperate to establish AZ VGCC abundance is critical for understanding how these mechanisms set synaptic strength and how they might be regulated to control presynaptic plasticity. VGCC abundance at AZs involves multiple steps, including channel biosynthesis (transcription, translation, and trafficking through the endomembrane system), forward axonal trafficking and delivery to synaptic terminals, incorporation and retention at presynaptic sites, and protein recycling. Here we discuss mechanisms that control VGCC abundance at synapses, highlighting findings from invertebrate and vertebrate models.
Collapse
Affiliation(s)
- Karen L. Cunningham
- The Picower Institute for Learning and Memory, Department of Biology, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | | |
Collapse
|
34
|
Zong P, Yue L. Regulation of Presynaptic Calcium Channels. ADVANCES IN NEUROBIOLOGY 2023; 33:171-202. [PMID: 37615867 DOI: 10.1007/978-3-031-34229-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Voltage-gated calcium channels (VGCCs), especially Cav2.1 and Cav2.2, are the major mediators of Ca2+ influx at the presynaptic membrane in response to neuron excitation, thereby exerting a predominant control on synaptic transmission. To guarantee the timely and precise release of neurotransmitters at synapses, the activity of presynaptic VGCCs is tightly regulated by a variety of factors, including auxiliary subunits, membrane potential, G protein-coupled receptors (GPCRs), calmodulin (CaM), Ca2+-binding proteins (CaBP), protein kinases, various interacting proteins, alternative splicing events, and genetic variations.
Collapse
Affiliation(s)
- Pengyu Zong
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Lixia Yue
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine, Farmington, CT, USA.
| |
Collapse
|
35
|
Wu T, Chen SR, Pan HL, Luo Y. The α2δ-1-NMDA receptor complex and its potential as a therapeutic target for ischemic stroke. Front Neurol 2023; 14:1148697. [PMID: 37153659 PMCID: PMC10157046 DOI: 10.3389/fneur.2023.1148697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/30/2023] [Indexed: 05/10/2023] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) play a critical role in excitotoxicity caused by ischemic stroke, but NMDAR antagonists have failed to be translated into clinical practice for treating stroke patients. Recent studies suggest that targeting the specific protein-protein interactions that regulate NMDARs may be an effective strategy to reduce excitotoxicity associated with brain ischemia. α2δ-1 (encoded by the Cacna2d1 gene), previously known as a subunit of voltage-gated calcium channels, is a binding protein of gabapentinoids used clinically for treating chronic neuropathic pain and epilepsy. Recent studies indicate that α2δ-1 is an interacting protein of NMDARs and can promote synaptic trafficking and hyperactivity of NMDARs in neuropathic pain conditions. In this review, we highlight the newly identified roles of α2δ-1-mediated NMDAR activity in the gabapentinoid effects and NMDAR excitotoxicity during brain ischemia as well as targeting α2δ-1-bound NMDARs as a potential treatment for ischemic stroke.
Collapse
Affiliation(s)
- Tao Wu
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shao-Rui Chen
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hui-Lin Pan
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Hui-Lin Pan
| | - Yi Luo
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- *Correspondence: Yi Luo
| |
Collapse
|
36
|
Kollewe A, Schwarz Y, Oleinikov K, Raza A, Haupt A, Wartenberg P, Wyatt A, Boehm U, Ectors F, Bildl W, Zolles G, Schulte U, Bruns D, Flockerzi V, Fakler B. Subunit composition, molecular environment, and activation of native TRPC channels encoded by their interactomes. Neuron 2022; 110:4162-4175.e7. [PMID: 36257322 DOI: 10.1016/j.neuron.2022.09.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/15/2022] [Accepted: 09/23/2022] [Indexed: 12/24/2022]
Abstract
In the mammalian brain TRPC channels, a family of Ca2+-permeable cation channels, are involved in a variety of processes from neuronal growth and synapse formation to transmitter release, synaptic transmission and plasticity. The molecular appearance and operation of native TRPC channels, however, remained poorly understood. Here, we used high-resolution proteomics to show that TRPC channels in the rodent brain are macro-molecular complexes of more than 1 MDa in size that result from the co-assembly of the tetrameric channel core with an ensemble of interacting proteins (interactome). The core(s) of TRPC1-, C4-, and C5-containing channels are mostly heteromers with defined stoichiometries for each subtype, whereas TRPC3, C6, and C7 preferentially form homomers. In addition, TRPC1/C4/C5 channels may co-assemble with the metabotropic glutamate receptor mGluR1, thus guaranteeing both specificity and reliability of channel activation via the phospholipase-Ca2+ pathway. Our results unveil the subunit composition of native TRPC channels and resolve the molecular details underlying their activation.
Collapse
Affiliation(s)
- Astrid Kollewe
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, 79104 Freiburg, Germany
| | - Yvonne Schwarz
- Institute of Physiology, Center for Integrative Physiology and Molecular Medicine, Saarland University, 66421 Homburg, Germany
| | - Katharina Oleinikov
- Institute of Physiology, Center for Integrative Physiology and Molecular Medicine, Saarland University, 66421 Homburg, Germany
| | - Ahsan Raza
- Experimental and Clinical Pharmacology and Toxicology, PZMS, Saarland University, 66421 Homburg, Germany
| | - Alexander Haupt
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, 79104 Freiburg, Germany
| | - Philipp Wartenberg
- Experimental and Clinical Pharmacology and Toxicology, PZMS, Saarland University, 66421 Homburg, Germany
| | - Amanda Wyatt
- Experimental and Clinical Pharmacology and Toxicology, PZMS, Saarland University, 66421 Homburg, Germany
| | - Ulrich Boehm
- Experimental and Clinical Pharmacology and Toxicology, PZMS, Saarland University, 66421 Homburg, Germany
| | - Fabien Ectors
- Transgenic facility, FARAH Research Center, Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Wolfgang Bildl
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, 79104 Freiburg, Germany
| | - Gerd Zolles
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, 79104 Freiburg, Germany
| | - Uwe Schulte
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, 79104 Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, Schänzlestr. 18, 79104 Freiburg, Germany
| | - Dieter Bruns
- Institute of Physiology, Center for Integrative Physiology and Molecular Medicine, Saarland University, 66421 Homburg, Germany
| | - Veit Flockerzi
- Experimental and Clinical Pharmacology and Toxicology, PZMS, Saarland University, 66421 Homburg, Germany.
| | - Bernd Fakler
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, 79104 Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, Schänzlestr. 18, 79104 Freiburg, Germany; Center for Basics in NeuroModulation, Breisacherstr. 4, 79106 Freiburg, Germany.
| |
Collapse
|
37
|
Pilch KS, Ramgoolam KH, Dolphin AC. Involvement of Ca V 2.2 channels and α 2 δ-1 in homeostatic synaptic plasticity in cultured hippocampal neurons. J Physiol 2022; 600:5333-5351. [PMID: 36377048 PMCID: PMC10107484 DOI: 10.1113/jp283600] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
In the mammalian brain, presynaptic CaV 2 channels play a pivotal role in synaptic transmission by mediating fast neurotransmitter exocytosis via influx of Ca2+ into the active zone of presynaptic terminals. However, the distribution and modulation of CaV 2.2 channels at plastic hippocampal synapses remains to be elucidated. Here, we assess CaV 2.2 channels during homeostatic synaptic plasticity, a compensatory form of homeostatic control preventing excessive or insufficient neuronal activity during which extensive active zone remodelling has been described. We show that chronic silencing of neuronal activity in mature hippocampal cultures resulted in elevated presynaptic Ca2+ transients, mediated by increased levels of CaV 2.2 channels at the presynaptic site. This work focused further on the role of α2 δ-1 subunits, important regulators of synaptic transmission and CaV 2.2 channel abundance at the presynaptic membrane. We found that α2 δ-1 overexpression reduces the contribution of CaV 2.2 channels to total Ca2+ flux without altering the amplitude of the Ca2+ transients. Levels of endogenous α2 δ-1 decreased during homeostatic synaptic plasticity, whereas the overexpression of α2 δ-1 prevented homeostatic synaptic plasticity in hippocampal neurons. Together, this study reveals a key role for CaV 2.2 channels and novel roles for α2 δ-1 during synaptic plastic adaptation. KEY POINTS: The roles of CaV 2.2 channels and α2 δ-1 in homeostatic synaptic plasticity in hippocampal neurons in culture were examined. Chronic silencing of neuronal activity resulted in elevated presynaptic Ca2+ transients, mediated by increased levels of CaV 2.2 channels at presynaptic sites. The level of endogenous α2 δ-1 decreased during homeostatic synaptic plasticity, whereas overexpression of α2 δ-1 prevented homeostatic synaptic plasticity in hippocampal neurons. Together, this study reveals a key role for CaV 2.2 channels and novel roles for α2 δ-1 during synaptic plastic adaptation.
Collapse
Affiliation(s)
- Kjara S. Pilch
- Department of NeurosciencePhysiology and PharmacologyUniversity College LondonLondonUK
| | - Krishma H. Ramgoolam
- Department of NeurosciencePhysiology and PharmacologyUniversity College LondonLondonUK
| | - Annette C. Dolphin
- Department of NeurosciencePhysiology and PharmacologyUniversity College LondonLondonUK
| |
Collapse
|
38
|
A slit-diaphragm-associated protein network for dynamic control of renal filtration. Nat Commun 2022; 13:6446. [PMID: 36307401 PMCID: PMC9616960 DOI: 10.1038/s41467-022-33748-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 09/29/2022] [Indexed: 12/25/2022] Open
Abstract
The filtration of blood in the kidney which is crucial for mammalian life is determined by the slit-diaphragm, a cell-cell junction between the foot processes of renal podocytes. The slit-diaphragm is thought to operate as final barrier or as molecular sensor of renal filtration. Using high-resolution proteomic analysis of slit-diaphragms affinity-isolated from rodent kidney, we show that the native slit-diaphragm is built from the junction-forming components Nephrin, Neph1 and Podocin and a co-assembled high-molecular weight network of proteins. The network constituents cover distinct classes of proteins including signaling-receptors, kinases/phosphatases, transporters and scaffolds. Knockout or knock-down of either the core components or the selected network constituents tyrosine kinase MER (MERTK), atrial natriuretic peptide-receptor C (ANPRC), integral membrane protein 2B (ITM2B), membrane-associated guanylate-kinase, WW and PDZ-domain-containing protein1 (MAGI1) and amyloid protein A4 resulted in target-specific impairment or disruption of the filtration process. Our results identify the slit-diaphragm as a multi-component system that is endowed with context-dependent dynamics via a co-assembled protein network.
Collapse
|
39
|
Zhang G, Liu JB, Yuan HL, Chen SY, Singer JH, Ke JB. Multiple Calcium Channel Types with Unique Expression Patterns Mediate Retinal Signaling at Bipolar Cell Ribbon Synapses. J Neurosci 2022; 42:6487-6505. [PMID: 35896423 PMCID: PMC9410755 DOI: 10.1523/jneurosci.0183-22.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 06/24/2022] [Accepted: 07/14/2022] [Indexed: 11/21/2022] Open
Abstract
Retinal bipolar cells (BCs) compose the canonical vertical excitatory pathway that conveys photoreceptor output to inner retinal neurons. Although synaptic transmission from BC terminals is thought to rely almost exclusively on Ca2+ influx through voltage-gated Ca2+ (CaV) channels mediating L-type currents, the molecular identity of CaV channels in BCs is uncertain. Therefore, we combined molecular and functional analyses to determine the expression profiles of CaV α1, β, and α2δ subunits in mouse rod bipolar (RB) cells, BCs from which the dynamics of synaptic transmission are relatively well-characterized. We found significant heterogeneity in CaV subunit expression within the RB population from mice of either sex, and significantly, we discovered that transmission from RB synapses was mediated by Ca2+ influx through P/Q-type (CaV2.1) and N-type (CaV2.2) conductances as well as the previously-described L-type (CaV1) and T-type (CaV3) conductances. Furthermore, we found both CaV1.3 and CaV1.4 proteins located near presynaptic ribbon-type active zones in RB axon terminals, indicating that the L-type conductance is mediated by multiple CaV1 subtypes. Similarly, CaV3 α1, β, and α2δ subunits also appear to obey a "multisubtype" rule, i.e., we observed a combination of multiple subtypes, rather than a single subtype as previously thought, for each CaV subunit in individual cells.SIGNIFICANCE STATEMENT Bipolar cells (BCs) transmit photoreceptor output to inner retinal neurons. Although synaptic transmission from BC terminals is thought to rely almost exclusively on Ca2+ influx through L-type voltage-gated Ca2+ (CaV) channels, the molecular identity of CaV channels in BCs is uncertain. Here, we report unexpectedly high molecular diversity of CaV subunits in BCs. Transmission from rod bipolar (RB) cell synapses can be mediated by Ca2+ influx through P/Q-type (CaV2.1) and N-type (CaV2.2) conductances as well as the previously-described L-type (CaV1) and T-type (CaV3) conductances. Furthermore, CaV1, CaV3, β, and α2δ subunits appear to obey a "multisubtype" rule, i.e., a combination of multiple subtypes for each subunit in individual cells, rather than a single subtype as previously thought.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Si-Yun Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Joshua H Singer
- Department of Biology, University of Maryland, College Park, Maryland 20742
| | - Jiang-Bin Ke
- 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
|
40
|
Synaptotagmin-7 Enhances Facilitation of Ca v2.1 Calcium Channels. eNeuro 2022; 9:ENEURO.0081-22.2022. [PMID: 35477860 PMCID: PMC9113918 DOI: 10.1523/eneuro.0081-22.2022] [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: 02/18/2022] [Revised: 04/15/2022] [Accepted: 04/22/2022] [Indexed: 11/21/2022] Open
Abstract
Voltage-gated calcium channel Cav2.1 undergoes Ca2+-dependent facilitation and inactivation, which are important in short-term synaptic plasticity. In presynaptic terminals, Cav2.1 forms large protein complexes that include synaptotagmins. Synaptotagmin-7 (Syt-7) is essential to mediate short-term synaptic plasticity in many synapses. Here, based on evidence that Cav2.1 and Syt-7 are both required for short-term synaptic facilitation, we investigated the direct interaction of Syt-7 with Cav2.1 and probed its regulation of Cav2.1 function. We found that Syt-7 binds specifically to the α1A subunit of Cav2.1 through interaction with the synaptic-protein interaction (synprint) site. Surprisingly, this interaction enhances facilitation in paired-pulse protocols and accelerates the onset of facilitation. Syt-7α induces a depolarizing shift in the voltage dependence of activation of Cav2.1 and slows Ca2+-dependent inactivation, whereas Syt-7β and Syt-7γ have smaller effects. Our results identify an unexpected, isoform-specific interaction between Cav2.1 and Syt-7 through the synprint site, which enhances Cav2.1 facilitation and modulates its inactivation.
Collapse
|
41
|
Despang P, Salamon S, Breitenkamp A, Kuzmenkina E, Matthes J. Inhibitory effects on L- and N-type calcium channels by a novel Ca Vβ 1 variant identified in a patient with autism spectrum disorder. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:459-470. [PMID: 35122502 PMCID: PMC8873119 DOI: 10.1007/s00210-022-02213-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 01/25/2022] [Indexed: 12/01/2022]
Abstract
Voltage-gated calcium channel (VGCC) subunits have been genetically associated with autism spectrum disorders (ASD). The properties of the pore-forming VGCC subunit are modulated by auxiliary β-subunits, which exist in four isoforms (CaVβ1-4). Our previous findings suggested that activation of L-type VGCCs is a common feature of CaVβ2 subunit mutations found in ASD patients. In the current study, we functionally characterized a novel CaVβ1b variant (p.R296C) identified in an ASD patient. We used whole-cell and single-channel patch clamp to study the effect of CaVβ1b_R296C on the function of L- and N-type VGCCs. Furthermore, we used co-immunoprecipitation followed by Western blot to evaluate the interaction of the CaVβ1b-subunits with the RGK-protein Gem. Our data obtained at both, whole-cell and single-channel levels, show that compared to a wild-type CaVβ1b, the CaVβ1b_R296C variant inhibits L- and N-type VGCCs. Interaction with and modulation by the RGK-protein Gem seems to be intact. Our findings indicate functional effects of the CaVβ1b_R296C variant differing from that attributed to CaVβ2 variants found in ASD patients. Further studies have to detail the effects on different VGCC subtypes and on VGCC expression.
Collapse
Affiliation(s)
- Patrick Despang
- Center of Pharmacology, Institute II, University of Cologne, Gleueler Strasse 24, 50931, Köln, Cologne, Germany
| | - Sarah Salamon
- Center of Pharmacology, Institute II, University of Cologne, Gleueler Strasse 24, 50931, Köln, Cologne, Germany
| | - Alexandra Breitenkamp
- Center of Pharmacology, Institute II, University of Cologne, Gleueler Strasse 24, 50931, Köln, Cologne, Germany
| | - Elza Kuzmenkina
- Center of Pharmacology, Institute II, University of Cologne, Gleueler Strasse 24, 50931, Köln, Cologne, Germany
| | - Jan Matthes
- Center of Pharmacology, Institute II, University of Cologne, Gleueler Strasse 24, 50931, Köln, Cologne, Germany.
| |
Collapse
|
42
|
Ji Y, Chen R, Wang Q, Wei Q, Tao R, Li B. Leveraging Gene-Level Prediction as Informative Covariate in Hypothesis Weighting Improves Power for Rare Variant Association Studies. Genes (Basel) 2022; 13:381. [PMID: 35205424 PMCID: PMC8872452 DOI: 10.3390/genes13020381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/31/2022] [Accepted: 02/09/2022] [Indexed: 02/05/2023] Open
Abstract
Gene-based rare variant association studies (RVASs) have low power due to the infrequency of rare variants and the large multiple testing burden. To correct for multiple testing, traditional false discovery rate (FDR) procedures which depend solely on P-values are often used. Recently, Independent Hypothesis Weighting (IHW) was developed to improve the detection power while maintaining FDR control by leveraging prior information for each hypothesis. Here, we present a framework to increase power of gene-based RVASs by incorporating prior information using IHW. We first build supervised machine learning models to assign each gene a prediction score that measures its disease risk, using the input of multiple biological features, fed with high-confidence risk genes and local background genes selected near GWAS significant loci as the training set. Then we use the prediction scores as covariates to prioritize RVAS results via IHW. We demonstrate the effectiveness of this framework through applications to RVASs in schizophrenia and autism spectrum disorder. We found sizeable improvements in the number of significant associations compared to traditional FDR approaches, and independent evidence supporting the relevance of the genes identified by our framework but not traditional FDR, demonstrating the potential of our framework to improve power of gene-based RVASs.
Collapse
Affiliation(s)
- Ying Ji
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA; (Y.J.); (R.C.); (Q.W.); (Q.W.)
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN 37203, USA
| | - Rui Chen
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA; (Y.J.); (R.C.); (Q.W.); (Q.W.)
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN 37203, USA
| | - Quan Wang
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA; (Y.J.); (R.C.); (Q.W.); (Q.W.)
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN 37203, USA
| | - Qiang Wei
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA; (Y.J.); (R.C.); (Q.W.); (Q.W.)
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN 37203, USA
| | - Ran Tao
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA; (Y.J.); (R.C.); (Q.W.); (Q.W.)
- Department of Biostatistics, Vanderbilt University, Nashville, TN 37203, USA
| | - Bingshan Li
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA; (Y.J.); (R.C.); (Q.W.); (Q.W.)
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN 37203, USA
| |
Collapse
|
43
|
Ion Channel Partnerships: Odd and Not-So-Odd Couples Controlling Neuronal Ion Channel Function. Int J Mol Sci 2022; 23:ijms23041953. [PMID: 35216068 PMCID: PMC8878034 DOI: 10.3390/ijms23041953] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/05/2022] [Accepted: 02/08/2022] [Indexed: 12/04/2022] Open
Abstract
The concerted function of the large number of ion channels expressed in excitable cells, including brain neurons, shapes diverse signaling events by controlling the electrical properties of membranes. It has long been recognized that specific groups of ion channels are functionally coupled in mediating ionic fluxes that impact membrane potential, and that these changes in membrane potential impact ion channel gating. Recent studies have identified distinct sets of ion channels that can also physically and functionally associate to regulate the function of either ion channel partner beyond that afforded by changes in membrane potential alone. Here, we review canonical examples of such ion channel partnerships, in which a Ca2+ channel is partnered with a Ca2+-activated K+ channel to provide a dedicated route for efficient coupling of Ca2+ influx to K+ channel activation. We also highlight examples of non-canonical ion channel partnerships between Ca2+ channels and voltage-gated K+ channels that are not intrinsically Ca2+ sensitive, but whose partnership nonetheless yields enhanced regulation of one or the other ion channel partner. We also discuss how these ion channel partnerships can be shaped by the subcellular compartments in which they are found and provide perspectives on how recent advances in techniques to identify proteins in close proximity to one another in native cells may lead to an expanded knowledge of other ion channel partnerships.
Collapse
|
44
|
Cruz-Garcia Y, Barkovits K, Kohlhaas M, Pickel S, Gulentz M, Heindl C, Pfeiffer K, Eder-Negrin P, Maack C, Marcus K, Kuhn M, Miranda-Laferte E. Nanoenviroments of the β-Subunit of L-Type Voltage-Gated Calcium Channels in Adult Cardiomyocytes. Front Cell Dev Biol 2022; 9:724778. [PMID: 35047492 PMCID: PMC8762238 DOI: 10.3389/fcell.2021.724778] [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: 06/14/2021] [Accepted: 12/06/2021] [Indexed: 11/23/2022] Open
Abstract
In cardiomyocytes, Ca2+ influx through L-type voltage-gated calcium channels (LTCCs) following membrane depolarization regulates crucial Ca2+-dependent processes including duration and amplitude of the action potentials and excitation-contraction coupling. LTCCs are heteromultimeric proteins composed of the Cavα1, Cavβ, Cavα2δ and Cavγ subunits. Here, using ascorbate peroxidase (APEX2)-mediated proximity labeling and quantitative proteomics, we identified 61 proteins in the nanoenvironments of Cavβ2 in cardiomyocytes. These proteins are involved in diverse cellular functions such as cellular trafficking, cardiac contraction, sarcomere organization and excitation-contraction coupling. Moreover, pull-down assays and co-immunoprecipitation analyses revealed that Cavβ2 interacts with the ryanodine receptor 2 (RyR2) in adult cardiomyocytes, probably coupling LTCCs and the RyR2 into a supramolecular complex at the dyads. This interaction is mediated by the Src-homology 3 domain of Cavβ2 and is necessary for an effective pacing frequency-dependent increase of the Ca2+-induced Ca2+ release mechanism in cardiomyocytes.
Collapse
Affiliation(s)
| | - Katalin Barkovits
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, Bochum, Germany.,Medical Proteome Analysis, Center for Proteindiagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Michael Kohlhaas
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Simone Pickel
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Michelle Gulentz
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Cornelia Heindl
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Kathy Pfeiffer
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, Bochum, Germany.,Medical Proteome Analysis, Center for Proteindiagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Petra Eder-Negrin
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Katrin Marcus
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, Bochum, Germany.,Medical Proteome Analysis, Center for Proteindiagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Michaela Kuhn
- Institute of Physiology, University of Würzburg, Würzburg, Germany.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Erick Miranda-Laferte
- Institute of Physiology, University of Würzburg, Würzburg, Germany.,Institut für Biologische Informationsprozesse, Molekular- und Zellphysiologie (IBI-1), Forschungszentrum Jülich, Jülich, Germany
| |
Collapse
|
45
|
Voltage-dependent Ca V3.2 and Ca V2.2 channels in nociceptive pathways. Pflugers Arch 2022; 474:421-434. [PMID: 35043234 DOI: 10.1007/s00424-022-02666-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 10/19/2022]
Abstract
Noxious stimuli like cold, heat, pH change, tissue damage, and inflammation depolarize a membrane of peripheral endings of specialized nociceptive neurons which eventually results in the generation of an action potential. The electrical signal is carried along a long axon of nociceptive neurons from peripheral organs to soma located in dorsal root ganglions and further to the dorsal horn of the spinal cord where it is transmitted through a chemical synapse and is carried through the spinal thalamic tract into the brain. Two subtypes of voltage-activated calcium play a major role in signal transmission: a low voltage-activated CaV3.2 channel and a high voltage-activated CaV2.2 channel. The CaV3.2 channel contributes mainly to the signal conductance along nociceptive neurons while the principal role of the CaV2.2 channel is in the synaptic transmission at the dorsal horn. Both channels contribute to the signal initiation at peripheral nerve endings. This review summarizes current knowledge about the expression and distribution of these channels in a nociceptive pathway, the regulation of their expression and gating during pain pathology, and their suitability as targets for pharmacological therapy.
Collapse
|
46
|
Zhang H, Wu ZS, Liu JQ, Huang H. Serum calcium channel subunit α2δ-1 concentrations and outcomes in patients with acute spontaneous intracerebral hemorrhage. Clin Chim Acta 2022; 527:17-22. [PMID: 35007528 DOI: 10.1016/j.cca.2022.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/24/2021] [Accepted: 01/03/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Voltage-gated calcium channel subunit α2δ-1 plays an important role in acute brain injury. We attempted to investigate whether serum α2δ-1 subunit concentrations are correlated with severity and prognosis following intracerebral hemorrhage (ICH). METHODS Serum α2δ-1 subunit concentrations were quantified in 103 ICH patients and 103 healthy controls. National Institutes of Health Stroke Scale (NIHSS) score and hematoma volume were estimated for assessing illness severity. Modified Rankin scale score of 3-6 at 90 days after stroke onset was defined as a worse outcome. RESULTS Serum α2δ-1 subunit concentrations were markedly higher in patients than in controls (median, 875.1 vs. 209.3 pg/ml). Serum α2δ-1 subunit concentrations of patients were tightly correlated with NIHSS score (r = 0.589) and hematoma volume (r = 0.594). Serum α2δ-1 subunit concentrations ≥ 875.1 pg/ml independently discriminated development of 90-day poor outcome with odds ratio of 5.228 (95% CI, 2.201-12.418) and area under the receiver operating characteristic curve of 0.794 (95% CI, 0.703-0.867). Serum α2δ-1 subunit concentrations > 973.4 pg/ml predicted 90-day poor outcome with 64.0% sensitivity and 90.6% specificity. The prognostic predictive ability of serum α2δ-1 concentrations was equivalent to those of NIHSS score and hematoma volume (both P > 0.05), and serum α2δ-1 concentrations also significantly improved the prognostic predictive capabilities of NIHSS score and hematoma volume (both P < 0.05). CONCLUSIONS Serum α2δ-1 subunit concentrations are intimately correlated with illness severity and are independently associated with poor 90-day outcome, substantializing serum α2δ-1 subunit as a potential prognostic biomarker for ICH.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Emergency Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 261 Huansha Road, Hangzhou 310006, China
| | - Ze-Sheng Wu
- Department of Emergency Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 261 Huansha Road, Hangzhou 310006, China
| | - Jing-Quan Liu
- Department of Urology, Hangzhou Ninth People's Hospital, 98 Yilong Road, Hangzhou 311225, China
| | - Huan Huang
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 261 Huansha Road, Hangzhou 310006, China.
| |
Collapse
|
47
|
Heck J, Palmeira Do Amaral AC, Weißbach S, El Khallouqi A, Bikbaev A, Heine M. More than a pore: How voltage-gated calcium channels act on different levels of neuronal communication regulation. Channels (Austin) 2021; 15:322-338. [PMID: 34107849 PMCID: PMC8205089 DOI: 10.1080/19336950.2021.1900024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
Voltage-gated calcium channels (VGCCs) represent key regulators of the calcium influx through the plasma membrane of excitable cells, like neurons. Activated by the depolarization of the membrane, the opening of VGCCs induces very transient and local changes in the intracellular calcium concentration, known as calcium nanodomains, that in turn trigger calcium-dependent signaling cascades and the release of chemical neurotransmitters. Based on their central importance as concierges of excitation-secretion coupling and therefore neuronal communication, VGCCs have been studied in multiple aspects of neuronal function and malfunction. However, studies on molecular interaction partners and recent progress in omics technologies have extended the actual concept of these molecules. With this review, we want to illustrate some new perspectives of VGCCs reaching beyond their function as calcium-permeable pores in the plasma membrane. Therefore, we will discuss the relevance of VGCCs as voltage sensors in functional complexes with ryanodine receptors, channel-independent actions of auxiliary VGCC subunits, and provide an insight into how VGCCs even directly participate in gene regulation. Furthermore, we will illustrate how structural changes in the intracellular C-terminus of VGCCs generated by alternative splicing events might not only affect the biophysical channel characteristics but rather determine their molecular environment and downstream signaling pathways.
Collapse
Affiliation(s)
- Jennifer Heck
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| | - Ana Carolina Palmeira Do Amaral
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| | - Stephan Weißbach
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
- Computational Genomics and Bioinformatics, Johannes Gutenberg-University Mainz, University Medical Center Mainz, Institute for Human Genetics, Mainz, Germany
| | - Abderazzaq El Khallouqi
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| | - Arthur Bikbaev
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| | - Martin Heine
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| |
Collapse
|
48
|
Kollewe A, Chubanov V, Tseung FT, Correia L, Schmidt E, Rössig A, Zierler S, Haupt A, Müller CS, Bildl W, Schulte U, Nicke A, Fakler B, Gudermann T. The molecular appearance of native TRPM7 channel complexes identified by high-resolution proteomics. eLife 2021; 10:68544. [PMID: 34766907 PMCID: PMC8616561 DOI: 10.7554/elife.68544] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 11/08/2021] [Indexed: 12/22/2022] Open
Abstract
The transient receptor potential melastatin-subfamily member 7 (TRPM7) is a ubiquitously expressed membrane protein consisting of ion channel and protein kinase domains. TRPM7 plays a fundamental role in the cellular uptake of divalent cations such as Zn2+, Mg2+, and Ca2+, and thus shapes cellular excitability, plasticity, and metabolic activity. The molecular appearance and operation of TRPM7 channels in native tissues have remained unresolved. Here, we investigated the subunit composition of endogenous TRPM7 channels in rodent brain by multi-epitope affinity purification and high-resolution quantitative mass spectrometry (MS) analysis. We found that native TRPM7 channels are high-molecular-weight multi-protein complexes that contain the putative metal transporter proteins CNNM1-4 and a small G-protein ADP-ribosylation factor-like protein 15 (ARL15). Heterologous reconstitution experiments confirmed the formation of TRPM7/CNNM/ARL15 ternary complexes and indicated that complex formation effectively and specifically impacts TRPM7 activity. These results open up new avenues towards a mechanistic understanding of the cellular regulation and function of TRPM7 channels.
Collapse
Affiliation(s)
- Astrid Kollewe
- Institute of Physiology II, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Vladimir Chubanov
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Fong Tsuen Tseung
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Leonor Correia
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Eva Schmidt
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Anna Rössig
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Susanna Zierler
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany.,Institute of Pharmacology, Johannes Kepler University Linz, Linz, Austria
| | - Alexander Haupt
- Institute of Physiology II, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Catrin Swantje Müller
- Institute of Physiology II, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wolfgang Bildl
- Institute of Physiology II, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Uwe Schulte
- Institute of Physiology II, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, Freiburg, Germany
| | - Annette Nicke
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Bernd Fakler
- Institute of Physiology II, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, Freiburg, Germany
| | - Thomas Gudermann
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany.,German Center for Lung Research, Munich, Germany
| |
Collapse
|
49
|
Corradi GR, Mazzitelli LR, Petrovich GD, de Tezanos Pinto F, Rochi L, Adamo HP. Plasma Membrane Ca 2+ Pump PMCA4z Is More Active Than Splicing Variant PMCA4x. Front Cell Neurosci 2021; 15:668371. [PMID: 34512262 PMCID: PMC8428515 DOI: 10.3389/fncel.2021.668371] [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: 02/16/2021] [Accepted: 07/22/2021] [Indexed: 11/13/2022] Open
Abstract
The plasma membrane Ca2+ pumps (PMCA) are P-ATPases that control Ca2+ signaling and homeostasis by transporting Ca2+ out of the eukaryotic cell. Humans have four genes that code for PMCA isoforms (PMCA1-4). A large diversity of PMCA isoforms is generated by alternative mRNA splicing at sites A and C. The different PMCA isoforms are expressed in a cell-type and developmental-specific manner and exhibit differential sensitivity to a great number of regulatory mechanisms. PMCA4 has two A splice variants, the forms "x" and "z". While PMCA4x is ubiquitously expressed and relatively well-studied, PMCA4z is less characterized and its expression is restricted to some tissues such as the brain and heart muscle. PMCA4z lacks a stretch of 12 amino acids in the so-called A-M3 linker, a conformation-sensitive region of the molecule connecting the actuator domain (A) with the third transmembrane segment (M3). We expressed in yeast PMCA4 variants "x" and "z", maintaining constant the most frequent splice variant "b" at the C-terminal end, and obtained purified preparations of both proteins. In the basal autoinhibited state, PMCA4zb showed a higher ATPase activity and a higher apparent Ca2+ affinity than PMCA4xb. Both isoforms were stimulated by calmodulin but PMCA4zb was more strongly activated by acidic lipids than PMCA4xb. The results indicate that a PMCA4 intrinsically more active and more responsive to acidic lipids is produced by the variant "z" of the splicing site A.
Collapse
Affiliation(s)
- Gerardo R Corradi
- Departamento de Química Biológica, Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Consejo Nacional de Investigaciones Científicas y Técnicas-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Luciana R Mazzitelli
- Departamento de Química Biológica, Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Consejo Nacional de Investigaciones Científicas y Técnicas-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Guido D Petrovich
- Departamento de Química Biológica, Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Consejo Nacional de Investigaciones Científicas y Técnicas-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Felicitas de Tezanos Pinto
- Departamento de Química Biológica, Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Consejo Nacional de Investigaciones Científicas y Técnicas-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Lucia Rochi
- Departamento de Química Biológica, Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Consejo Nacional de Investigaciones Científicas y Técnicas-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Hugo P Adamo
- Departamento de Química Biológica, Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Consejo Nacional de Investigaciones Científicas y Técnicas-Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
50
|
Abstract
Neurons are highly specialized cells equipped with a sophisticated molecular machinery for the reception, integration, conduction and distribution of information. The evolutionary origin of neurons remains unsolved. How did novel and pre-existing proteins assemble into the complex machinery of the synapse and of the apparatus conducting current along the neuron? In this review, the step-wise assembly of functional modules in neuron evolution serves as a paradigm for the emergence and modification of molecular machinery in the evolution of cell types in multicellular organisms. The pre-synaptic machinery emerged through modification of calcium-regulated large vesicle release, while the postsynaptic machinery has different origins: the glutamatergic postsynapse originated through the fusion of a sensory signaling module and a module for filopodial outgrowth, while the GABAergic postsynapse incorporated an ancient actin regulatory module. The synaptic junction, in turn, is built around two adhesion modules controlled by phosphorylation, which resemble septate and adherens junctions. Finally, neuronal action potentials emerged via a series of duplications and modifications of voltage-gated ion channels. Based on these origins, key molecular innovations are identified that led to the birth of the first neuron in animal evolution.
Collapse
|