1
|
Qian X, Sheng X, Ding J, Yiming Z, Zheng J, Zhong J, Zhang T, Li X, He S, Li W, Zhang M. Tropisetron, an Antiemetic Drug, Exerts an Anti-Epileptic Effect Through the Activation of α7nAChRs in a Rat Model of Temporal Lobe Epilepsy. CNS Neurosci Ther 2024; 30:e70086. [PMID: 39445711 PMCID: PMC11500210 DOI: 10.1111/cns.70086] [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: 03/27/2024] [Revised: 08/28/2024] [Accepted: 09/28/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Temporal lobe epilepsy (TLE), a prevalent chronic neurological disorder, affects millions of individuals and is often resistant to anti-epileptic drugs. Increasing evidence has shown that acetylcholine (ACh) and cholinergic neurotransmission play a role in the pathophysiology of epilepsy. Tropisetron, an antiemetic drug used for chemotherapy in clinic, has displayed potential in the treatment of Alzheimer's disease, depression, and schizophrenia in animal models. However, as a partial agonist of α7 nicotinic acetylcholine receptors (α7nAChRs), whether tropisetron possesses the therapeutic potential for TLE has not yet been determined. METHODS In this study, tropisetron was intraperitoneally injected into pilocarpine-induced epileptic rats for 3 weeks. Alpha-bungarotoxin (α-bgt), a specific α7nAChR antagonist, was applied to investigate the mechanism of tropisetron. Rats were assessed for spontaneous recurrent seizures (SRS) and cognitive function using video surveillance and Morris's water maze testing. Hippocampal impairment and synaptic structure were evaluated by Nissl staining, immunohistochemistry, and Golgi staining. Additionally, the levels of glutamate, γ-aminobutyric acid (GABA), ACh, α7nAChRs, neuroinflammatory cytokines, glucocorticoids and their receptors, as well as synapse-associated protein (F-actin, cofilin-1) were quantified. RESULTS The results showed that tropisetron significantly reduced SRS, improved cognitive function, alleviated hippocampal sclerosis, and concurrently suppressed synaptic remodeling and the m6A modification of cofilin-1 in TLE rats. Furthermore, tropisetron lowered glutamate levels without affecting GABA levels, reduced neuroinflammation, and increased ACh levels and α7nAChR expression in the hippocampi of TLE rats. The effects of tropisetron treatment were counteracted by α-bgt. CONCLUSION In summary, these findings indicate that tropisetron exhibits an anti-epileptic effect and provides neuroprotection in TLE rats through the activation of α7nAChRs. The potential mechanism may involve the reduction of glutamate levels, enhancement of cholinergic transmission, and suppression of synaptic remodeling. Consequently, the present study not only highlights the potential of tropisetron as an anti-epileptic drug but also identifies α7nAChRs as a promising therapeutic target for the treatment of TLE.
Collapse
Affiliation(s)
- Xu Qian
- Department of Clinical Pharmacy, School of PharmaceuticalGuangzhou Medical University and Key Laboratory of Molecular Target &Clinical PharmacologyGuangzhouChina
| | - Xinwen Sheng
- Department of Clinical Pharmacy, School of PharmaceuticalGuangzhou Medical University and Key Laboratory of Molecular Target &Clinical PharmacologyGuangzhouChina
- Department of PharmacyThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Jiqiang Ding
- Department of Neurosurgery, The Six Affiliated Hospital (Dongguan Eastern Central Hospital)Jinan UniversityDongguanChina
| | - Zulipiya Yiming
- Department of Clinical Pharmacy, School of PharmaceuticalGuangzhou Medical University and Key Laboratory of Molecular Target &Clinical PharmacologyGuangzhouChina
| | - Jingjun Zheng
- Department of Clinical Pharmacy, School of PharmaceuticalGuangzhou Medical University and Key Laboratory of Molecular Target &Clinical PharmacologyGuangzhouChina
| | - Jiagui Zhong
- Department of Neurosurgery, The Six Affiliated Hospital (Dongguan Eastern Central Hospital)Jinan UniversityDongguanChina
| | - Tengyue Zhang
- Department of Clinical Pharmacy, School of PharmaceuticalGuangzhou Medical University and Key Laboratory of Molecular Target &Clinical PharmacologyGuangzhouChina
| | - Xuemei Li
- Department of Clinical Pharmacy, School of PharmaceuticalGuangzhou Medical University and Key Laboratory of Molecular Target &Clinical PharmacologyGuangzhouChina
| | - Shuqiao He
- Department of Clinical Pharmacy, School of PharmaceuticalGuangzhou Medical University and Key Laboratory of Molecular Target &Clinical PharmacologyGuangzhouChina
| | - Wei Li
- Department of Neurosurgery, The Six Affiliated Hospital (Dongguan Eastern Central Hospital)Jinan UniversityDongguanChina
| | - Mei Zhang
- Department of Clinical Pharmacy, School of PharmaceuticalGuangzhou Medical University and Key Laboratory of Molecular Target &Clinical PharmacologyGuangzhouChina
| |
Collapse
|
2
|
Chien CT, Puhl H, Vogel SS, Molloy JE, Chiu W, Khan S. Hub stability in the calcium calmodulin-dependent protein kinase II. Commun Biol 2024; 7:766. [PMID: 38918547 PMCID: PMC11199487 DOI: 10.1038/s42003-024-06423-y] [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: 06/20/2023] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
The calcium calmodulin protein kinase II (CaMKII) is a multi-subunit ring assembly with a central hub formed by the association domains. There is evidence for hub polymorphism between and within CaMKII isoforms, but the link between polymorphism and subunit exchange has not been resolved. Here, we present near-atomic resolution cryogenic electron microscopy (cryo-EM) structures revealing that hubs from the α and β isoforms, either standalone or within an β holoenzyme, coexist as 12 and 14 subunit assemblies. Single-molecule fluorescence microscopy of Venus-tagged holoenzymes detects intermediate assemblies and progressive dimer loss due to intrinsic holoenzyme lability, and holoenzyme disassembly into dimers upon mutagenesis of a conserved inter-domain contact. Molecular dynamics (MD) simulations show the flexibility of 4-subunit precursors, extracted in-silico from the β hub polymorphs, encompassing the curvature of both polymorphs. The MD explains how an open hub structure also obtained from the β holoenzyme sample could be created by dimer loss and analysis of its cryo-EM dataset reveals how the gap could open further. An assembly model, considering dimer concentration dependence and strain differences between polymorphs, proposes a mechanism for intrinsic hub lability to fine-tune the stoichiometry of αβ heterooligomers for their dynamic localization within synapses in neurons.
Collapse
Affiliation(s)
- Chih-Ta Chien
- Department of Bioengineering, and Department of Microbiology and Immunology, James H. Clark Center, Stanford University, Stanford, CA, 94305, USA
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institute of Health, Bethesda, MD, 20892, USA
| | - Henry Puhl
- Laboratory of Biophotonics and Quantum Biology, National Institutes on Alcohol, Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 208952, USA
| | - Steven S Vogel
- Laboratory of Biophotonics and Quantum Biology, National Institutes on Alcohol, Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 208952, USA
| | - Justin E Molloy
- The Francis Crick Institute, London, UK
- CMCB, Warwick Medical School, Coventry, CV4 7AL, UK
| | - Wah Chiu
- Department of Bioengineering, and Department of Microbiology and Immunology, James H. Clark Center, Stanford University, Stanford, CA, 94305, USA.
- CryoEM and Bioimaging Division, Stanford Synchrotron Radiation Light source, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, CA, 94025, USA.
| | - Shahid Khan
- Molecular Biology Consortium @ Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA.
| |
Collapse
|
3
|
Khan S, Molloy JE, Puhl H, Schulman H, Vogel SS. Real-time single-molecule imaging of CaMKII-calmodulin interactions. Biophys J 2024; 123:824-838. [PMID: 38414237 DOI: 10.1016/j.bpj.2024.02.021] [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: 08/03/2023] [Revised: 12/20/2023] [Accepted: 02/22/2024] [Indexed: 02/29/2024] Open
Abstract
The binding of calcium/calmodulin (CAM) to calcium/calmodulin-dependent protein kinase II (CaMKII) initiates an ATP-driven cascade that triggers CaMKII autophosphorylation. The autophosphorylation in turn increases the CaMKII affinity for CAM. Here, we studied the ATP dependence of CAM association with the actin-binding CaMKIIβ isoform using single-molecule total internal reflection fluorescence microscopy. Rhodamine-CAM associations/dissociations to surface-immobilized Venus-CaMKIIβ were resolved with 0.5 s resolution from video records, batch-processed with a custom algorithm. CAM occupancy was determined simultaneously with spot-photobleaching measurement of CaMKII holoenzyme stoichiometry. We show the ATP-dependent increase of the CAM association requires dimer formation for both the α and β isoforms. The study of mutant β holoenzymes revealed that the ATP-dependent increase in CAM affinity results in two distinct states. The phosphorylation-defective (T287.306-307A) holoenzyme resides only in the low-affinity state. CAM association is further reduced in the T287A holoenzyme relative to T287.306-307A. In the absence of ATP, the affinity of CAM for the T287.306-307A mutant and the wild-type monomer are comparable. The affinity of the ATP-binding impaired (K43R) mutant is even weaker. In ATP, the K43R holoenzyme resides in the low-affinity state. The phosphomimetic mutant (T287D) resides only in a 1000-fold higher-affinity state, with mean CAM occupancy of more than half of the 14-mer holoenzyme stoichiometry in picomolar CAM. ATP promotes T287D holoenzyme disassembly but does not elevate CAM occupancy. Single Poisson distributions characterized the ATP-dependent CAM occupancy of mutant holoenzymes. In contrast, the CAM occupancy of the wild-type population had a two-state distribution with both low- and high-affinity states represented. The low-affinity state was the dominant state, a result different from published in vitro assays. Differences in assay conditions can alter the balance between activating and inhibitory autophosphorylation. Bound ATP could be sufficient for CaMKII structural function, while antagonistic autophosphorylations may tune CaMKII kinase-regulated action-potential frequency decoding in vivo.
Collapse
Affiliation(s)
- Shahid Khan
- Molecular Biology Consortium at Lawrence Berkeley National Laboratory, Berkeley, California.
| | | | - Henry Puhl
- Laboratory of Biophotonics and Quantum Biology, National Institutes on Alcohol, Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland
| | - Howard Schulman
- Panorama Research Institute, Sunnyvale, California; Stanford University School of Medicine, Stanford, California
| | - Steven S Vogel
- Laboratory of Biophotonics and Quantum Biology, National Institutes on Alcohol, Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland
| |
Collapse
|
4
|
Lisek M, Tomczak J, Boczek T, Zylinska L. Calcium-Associated Proteins in Neuroregeneration. Biomolecules 2024; 14:183. [PMID: 38397420 PMCID: PMC10887043 DOI: 10.3390/biom14020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/27/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
The dysregulation of intracellular calcium levels is a critical factor in neurodegeneration, leading to the aberrant activation of calcium-dependent processes and, ultimately, cell death. Ca2+ signals vary in magnitude, duration, and the type of neuron affected. A moderate Ca2+ concentration can initiate certain cellular repair pathways and promote neuroregeneration. While the peripheral nervous system exhibits an intrinsic regenerative capability, the central nervous system has limited self-repair potential. There is evidence that significant variations exist in evoked calcium responses and axonal regeneration among neurons, and individual differences in regenerative capacity are apparent even within the same type of neurons. Furthermore, some studies have shown that neuronal activity could serve as a potent regulator of this process. The spatio-temporal patterns of calcium dynamics are intricately controlled by a variety of proteins, including channels, ion pumps, enzymes, and various calcium-binding proteins, each of which can exert either positive or negative effects on neural repair, depending on the cellular context. In this concise review, we focus on several calcium-associated proteins such as CaM kinase II, GAP-43, oncomodulin, caldendrin, calneuron, and NCS-1 in order to elaborate on their roles in the intrinsic mechanisms governing neuronal regeneration following traumatic damage processes.
Collapse
Affiliation(s)
| | | | | | - Ludmila Zylinska
- Department of Molecular Neurochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (M.L.); (J.T.); (T.B.)
| |
Collapse
|
5
|
Tullis JE, Bayer KU. Distinct synaptic pools of DAPK1 differentially regulate activity-dependent synaptic CaMKII accumulation. iScience 2023; 26:106723. [PMID: 37216104 PMCID: PMC10192646 DOI: 10.1016/j.isci.2023.106723] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/18/2023] [Accepted: 04/19/2023] [Indexed: 05/24/2023] Open
Abstract
The death-associated protein kinase 1 (DAPK1) regulates the synaptic movement of the Ca2+/calmodulin (CaM)-dependent protein kinase II (CaMKII). Synaptic CaMKII accumulation is mediated via binding to the NMDA-receptor subunit GluN2B and is required for long-term potentiation (LTP). By contrast, long-term depression (LTD) instead requires specific suppression of this movement, which is mediated by competitive DAPK1 binding to GluN2B. We find here that DAPK1 localizes to synapses via two distinct mechanisms: basal localization requires F-actin, but retention of DAPK1 at synapses during LTD requires an additional binding mode, likely to GluN2B. While F-actin binding mediates DAPK1 enrichment at synapses, it is not sufficient to suppress synaptic CaMKII movement. However, it is a prerequisite that enables the additional LTD-specific binding mode of DAPK1, which in turn mediates suppression of the CaMKII movement. Thus, both modes of synaptic DAPK1 localization work together to regulate synaptic CaMKII localization and thereby synaptic plasticity.
Collapse
Affiliation(s)
- Jonathan E. Tullis
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - K. Ulrich Bayer
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Program in Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
6
|
Khan R, Kulasiri D, Samarasinghe S. A multifarious exploration of synaptic tagging and capture hypothesis in synaptic plasticity: Development of an integrated mathematical model and computational experiments. J Theor Biol 2023; 556:111326. [PMID: 36279957 DOI: 10.1016/j.jtbi.2022.111326] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/25/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022]
Abstract
The synaptic tagging and capture (STC) hypothesis not only explain the integration and association of synaptic activities, but also the formation of learning and memory. The synaptic pathways involved in the synaptic tagging and capture phenomenon are called STC pathways. The STC hypothesis provides a potential explanation of the neuronal and synaptic processes underlying the synaptic consolidation of memories. Several mechanisms and molecules have been proposed to explain the process of memory allocation and synaptic tags, respectively. However, a clear link between the STC hypothesis and memory allocation is still missing because the encoding of memories in neural circuits is mainly associated with strongly recurrently connected groups of neurons. To explore the mechanisms of potential synaptic tagging candidates and their involvement in the process of memory allocation, we develop a mathematical model for a single dendritic spine based on five essential criteria of a synaptic tag. By developing a mathematical model, we attempt to understand the roles of the potentially critical molecular networks underlying the STC and the essential attributes of a synaptic tag. We include essential memory molecules in the STC model that have been identified in earlier studies as crucial for STC pathways. CaMKII activation is critical for the setting of the initial tag; however, coordinated activities with other kinases and the biochemical pathways are necessary for the tag to be stable. PKA modulates NMDAR-mediated Ca2+ signalling. Similarly, PKA and ERK crosstalk is essential for Ca2+ - mediated protein synthesis during l-LTP. Our theoretical model explains the quantitative contribution of Tags and protein synthesis during l-LTP in synaptic strength.
Collapse
Affiliation(s)
- Raheel Khan
- Centre for Advanced Computational Solutions (C-fACS), Department of Molecular Biosciences, Lincoln University, Christchurch, New Zealand
| | - D Kulasiri
- Centre for Advanced Computational Solutions (C-fACS), Department of Molecular Biosciences, Lincoln University, Christchurch, New Zealand.
| | - S Samarasinghe
- Centre for Advanced Computational Solutions (C-fACS), Department of Molecular Biosciences, Lincoln University, Christchurch, New Zealand
| |
Collapse
|
7
|
Khan S. Conformational spread drives the evolution of the calcium-calmodulin protein kinase II. Sci Rep 2022; 12:8499. [PMID: 35589775 PMCID: PMC9120016 DOI: 10.1038/s41598-022-12090-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 05/03/2022] [Indexed: 11/17/2022] Open
Abstract
The calcium calmodulin (Ca2+/CaM) dependent protein kinase II (CaMKII) decodes Ca2+ frequency oscillations. The CaMKIIα isoform is predominantly expressed in the brain and has a central role in learning. I matched residue and organismal evolution with collective motions deduced from the atomic structure of the human CaMKIIα holoenzyme to learn how its ring architecture abets function. Protein dynamic simulations showed its peripheral kinase domains (KDs) are conformationally coupled via lateral spread along the central hub. The underlying β-sheet motions in the hub or association domain (AD) were deconvolved into dynamic couplings based on mutual information. They mapped onto a coevolved residue network to partition the AD into two distinct sectors. A second, energetically stressed sector was added to ancient bacterial enzyme dimers for assembly of the ringed hub. The continued evolution of the holoenzyme after AD–KD fusion targeted the sector’s ring contacts coupled to the KD. Among isoforms, the α isoform emerged last and, it alone, mutated rapidly after the poikilotherm–homeotherm jump to match the evolution of memory. The correlation between dynamics and evolution of the CaMKII AD argues single residue substitutions fine-tune hub conformational spread. The fine-tuning could increase CaMKIIα Ca2+ frequency response range for complex learning functions.
Collapse
Affiliation(s)
- Shahid Khan
- Molecular Biology Consortium, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA. .,SBA School of Science and Engineering, LUMS, Lahore, Pakistan. .,Laboratory of Cell Biology, NINDS, NIH, Bethesda, MD, 20892, USA.
| |
Collapse
|
8
|
Buonarati OR, Miller AP, Coultrap SJ, Bayer KU, Reichow SL. Conserved and divergent features of neuronal CaMKII holoenzyme structure, function, and high-order assembly. Cell Rep 2021; 37:110168. [PMID: 34965414 PMCID: PMC8985225 DOI: 10.1016/j.celrep.2021.110168] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 08/30/2021] [Accepted: 12/03/2021] [Indexed: 11/23/2022] Open
Abstract
Neuronal CaMKII holoenzymes (α and β isoforms) enable molecular signal computation underlying learning and memory but also mediate excitotoxic neuronal death. Here, we provide a comparative analysis of these signaling devices, using single-particle electron microscopy (EM) in combination with biochemical and live-cell imaging studies. In the basal state, both isoforms assemble mainly as 12-mers (but also 14-mers and even 16-mers for the β isoform). CaMKIIα and β isoforms adopt an ensemble of extended activatable states (with average radius of 12.6 versus 16.8 nm, respectively), characterized by multiple transient intra- and inter-holoenzyme interactions associated with distinct functional properties. The extended state of CaMKIIβ allows direct resolution of intra-holoenzyme kinase domain dimers. These dimers could enable cooperative activation by calmodulin, which is observed for both isoforms. High-order CaMKII clustering mediated by inter-holoenzyme kinase domain dimerization is reduced for the β isoform for both basal and excitotoxicity-induced clusters, both in vitro and in neurons. The CaMKII holoenzyme enables neuronal signal computation. In a comparative structure-function analysis of the neuronal α and β isoforms, Buonarati et al. find evidence for kinase domain dimers within the holoenzyme that enable a cooperative activation mechanism in both isoforms and inter-holoenzyme interactions that enable high-order aggregate formation under ischemic conditions.
Collapse
Affiliation(s)
- Olivia R Buonarati
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Adam P Miller
- Department of Chemistry, Portland State University, Portland, OR 97201, USA
| | - Steven J Coultrap
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - K Ulrich Bayer
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Steve L Reichow
- Department of Chemistry, Portland State University, Portland, OR 97201, USA.
| |
Collapse
|
9
|
La Montanara P, Hervera A, Baltussen LL, Hutson TH, Palmisano I, De Virgiliis F, Kong G, Chadwick J, Gao Y, Bartus K, Majid QA, Gorgoraptis N, Wong K, Downs J, Pizzorusso T, Ultanir SK, Leonard H, Yu H, Millar DS, Istvan N, Mazarakis ND, Di Giovanni S. Cyclin-dependent-like kinase 5 is required for pain signaling in human sensory neurons and mouse models. Sci Transl Med 2021; 12:12/551/eaax4846. [PMID: 32641489 DOI: 10.1126/scitranslmed.aax4846] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 12/10/2019] [Accepted: 04/05/2020] [Indexed: 12/19/2022]
Abstract
Cyclin-dependent-like kinase 5 (CDKL5) gene mutations lead to an X-linked disorder that is characterized by infantile epileptic encephalopathy, developmental delay, and hypotonia. However, we found that a substantial percentage of these patients also report a previously unrecognized anamnestic deficiency in pain perception. Consistent with a role in nociception, we found that CDKL5 is expressed selectively in nociceptive dorsal root ganglia (DRG) neurons in mice and in induced pluripotent stem cell (iPS)-derived human nociceptors. CDKL5-deficient mice display defective epidermal innervation, and conditional deletion of CDKL5 in DRG sensory neurons impairs nociception, phenocopying CDKL5 deficiency disorder in patients. Mechanistically, CDKL5 interacts with calcium/calmodulin-dependent protein kinase II α (CaMKIIα) to control outgrowth and transient receptor potential cation channel subfamily V member 1 (TRPV1)-dependent signaling, which are disrupted in both CDKL5 mutant murine DRG and human iPS-derived nociceptors. Together, these findings unveil a previously unrecognized role for CDKL5 in nociception, proposing an original regulatory mechanism for pain perception with implications for future therapeutics in CDKL5 deficiency disorder.
Collapse
Affiliation(s)
- Paolo La Montanara
- Department of Brain Sciences, Division of Neuroscience, Imperial College London, London W12 0NN, UK.
| | - Arnau Hervera
- Department of Brain Sciences, Division of Neuroscience, Imperial College London, London W12 0NN, UK.,Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain.,Department of Cell Biology, Physiology and Immunology, Faculty of Biology & Institute of Neuroscience, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Lucas L Baltussen
- Kinases and Brain Development Laboratory, Francis Crick Institute, London NW1 1AT, UK
| | - Thomas H Hutson
- Department of Brain Sciences, Division of Neuroscience, Imperial College London, London W12 0NN, UK
| | - Ilaria Palmisano
- Department of Brain Sciences, Division of Neuroscience, Imperial College London, London W12 0NN, UK
| | - Francesco De Virgiliis
- Department of Brain Sciences, Division of Neuroscience, Imperial College London, London W12 0NN, UK
| | - Guiping Kong
- Department of Brain Sciences, Division of Neuroscience, Imperial College London, London W12 0NN, UK
| | - Jessica Chadwick
- Department of Brain Sciences, Division of Neuroscience, Imperial College London, London W12 0NN, UK
| | - Yunan Gao
- Gene Therapy, Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Department of Medicine, Imperial College London, London W12 0NN, UK
| | - Katalin Bartus
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London Bridge, London SE1 1UL, UK
| | - Qasim A Majid
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Nikos Gorgoraptis
- Department of Brain Sciences, Division of Neuroscience, Imperial College London, London W12 0NN, UK
| | - Kingsley Wong
- Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia
| | - Jenny Downs
- Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia
| | - Tommaso Pizzorusso
- Institute of Neuroscience, National Research Council (CNR), I-56124 Pisa, Italy.,Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, I-50135 Florence, Italy
| | - Sila K Ultanir
- Gene Therapy, Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Department of Medicine, Imperial College London, London W12 0NN, UK
| | - Helen Leonard
- Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - David S Millar
- Institute of Cancer and Genetics, Cardiff University, Cardiff F14 4ED, UK
| | - Nagy Istvan
- Nociception, Section of Anesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Nicholas D Mazarakis
- Gene Therapy, Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Department of Medicine, Imperial College London, London W12 0NN, UK
| | - Simone Di Giovanni
- Department of Brain Sciences, Division of Neuroscience, Imperial College London, London W12 0NN, UK.
| |
Collapse
|
10
|
Ordyan M, Bartol T, Kennedy M, Rangamani P, Sejnowski T. Interactions between calmodulin and neurogranin govern the dynamics of CaMKII as a leaky integrator. PLoS Comput Biol 2020; 16:e1008015. [PMID: 32678848 PMCID: PMC7390456 DOI: 10.1371/journal.pcbi.1008015] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 07/29/2020] [Accepted: 06/04/2020] [Indexed: 01/10/2023] Open
Abstract
Calmodulin-dependent kinase II (CaMKII) has long been known to play an important role in learning and memory as well as long term potentiation (LTP). More recently it has been suggested that it might be involved in the time averaging of synaptic signals, which can then lead to the high precision of information stored at a single synapse. However, the role of the scaffolding molecule, neurogranin (Ng), in governing the dynamics of CaMKII is not yet fully understood. In this work, we adopt a rule-based modeling approach through the Monte Carlo method to study the effect of Ca2+ signals on the dynamics of CaMKII phosphorylation in the postsynaptic density (PSD). Calcium surges are observed in synaptic spines during an EPSP and back-propagating action potential due to the opening of NMDA receptors and voltage dependent calcium channels. Using agent-based models, we computationally investigate the dynamics of phosphorylation of CaMKII monomers and dodecameric holoenzymes. The scaffolding molecule, Ng, when present in significant concentration, limits the availability of free calmodulin (CaM), the protein which activates CaMKII in the presence of calcium. We show that Ng plays an important modulatory role in CaMKII phosphorylation following a surge of high calcium concentration. We find a non-intuitive dependence of this effect on CaM concentration that results from the different affinities of CaM for CaMKII depending on the number of calcium ions bound to the former. It has been shown previously that in the absence of phosphatase, CaMKII monomers integrate over Ca2+ signals of certain frequencies through autophosphorylation (Pepke et al, Plos Comp. Bio., 2010). We also study the effect of multiple calcium spikes on CaMKII holoenzyme autophosphorylation, and show that in the presence of phosphatase, CaMKII behaves as a leaky integrator of calcium signals, a result that has been recently observed in vivo. Our models predict that the parameters of this leaky integrator are finely tuned through the interactions of Ng, CaM, CaMKII, and PP1, providing a mechanism to precisely control the sensitivity of synapses to calcium signals. Author Summary not valid for PLOS ONE submissions.
Collapse
Affiliation(s)
- Mariam Ordyan
- Institute for Neural Computation, University of California San Diego, La Jolla, California, United States of America
- Computational Neurobiology Laboratory, Salk Institute for Biological Sciences, La Jolla, California, United States of America
| | - Tom Bartol
- Computational Neurobiology Laboratory, Salk Institute for Biological Sciences, La Jolla, California, United States of America
| | - Mary Kennedy
- The Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, United States of America
- * E-mail: (PR), (TS)
| | - Terrence Sejnowski
- Institute for Neural Computation, University of California San Diego, La Jolla, California, United States of America
- Computational Neurobiology Laboratory, Salk Institute for Biological Sciences, La Jolla, California, United States of America
- * E-mail: (PR), (TS)
| |
Collapse
|
11
|
Fitzgerald ZT, Rose JK. Locally-Induced CaMKII Translocation Requires Nucleotide Binding. Front Synaptic Neurosci 2020; 12:4. [PMID: 32116640 PMCID: PMC7019030 DOI: 10.3389/fnsyn.2020.00004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 01/17/2020] [Indexed: 11/16/2022] Open
Abstract
Calcium-calmodulin-dependent protein kinase (CaMKII) is a molecule involved in several cell processes including plasticity related to learning and memory. Activation of NMDA-type glutamate receptors results in translocation of CaMKII to synapses. However, there are at least two distinct mechanisms by which glutamate-dependent CaMKII translocation occurs: one well-studied process resulting from whole-cell glutamate stimulation and one resulting from brief, local glutamate application. Unlike the relatively fast CaMKII translocation seen following whole-cell glutamate delivery (seconds), local application results in CaMKII translocation that occurs gradually within 6–10 min. This locally-induced translocation of CaMKII requires L-type Ca2+ channel co-activation but does not rely on GluN2B receptor subunit expression, unlike translocation following whole-cell application of glutamate. The current study examined if nucleotide binding is necessary for locally-induced CaMKII translocation, similar to CaMKII translocation resulting from whole-cell glutamate application. Three different mechanisms of inhibition were employed: staurosporine (ATP inhibitor), CaMKII(281–302) peptide inhibitor and expression of the K42M mutation. Locally-induced CaMKII translocation was moderately suppressed in the presence of either the broad-spectrum kinase inhibitor staurosporine (100 nm) or the CaMKII(281–302) peptide inhibitor. However, expression of the catalytically dead K42M mutation that prevents ATP-binding to CaMKII, significantly inhibited locally-induced translocation. Thus, CaMKII translocation following brief, local glutamate application requires nucleotide binding, providing support for future research into the molecular mechanisms of this distinct form of CaMKII translocation.
Collapse
Affiliation(s)
- Zachary T Fitzgerald
- Behavioral Neuroscience Program, Department of Psychology, Western Washington University, Bellingham, WA, United States
| | - Jacqueline K Rose
- Behavioral Neuroscience Program, Department of Psychology, Western Washington University, Bellingham, WA, United States
| |
Collapse
|
12
|
Dendritic Spines in Alzheimer's Disease: How the Actin Cytoskeleton Contributes to Synaptic Failure. Int J Mol Sci 2020; 21:ijms21030908. [PMID: 32019166 PMCID: PMC7036943 DOI: 10.3390/ijms21030908] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/24/2020] [Accepted: 01/26/2020] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by Aβ-driven synaptic dysfunction in the early phases of pathogenesis. In the synaptic context, the actin cytoskeleton is a crucial element to maintain the dendritic spine architecture and to orchestrate the spine’s morphology remodeling driven by synaptic activity. Indeed, spine shape and synaptic strength are strictly correlated and precisely governed during plasticity phenomena in order to convert short-term alterations of synaptic strength into long-lasting changes that are embedded in stable structural modification. These functional and structural modifications are considered the biological basis of learning and memory processes. In this review we discussed the existing evidence regarding the role of the spine actin cytoskeleton in AD synaptic failure. We revised the physiological function of the actin cytoskeleton in the spine shaping and the contribution of actin dynamics in the endocytosis mechanism. The internalization process is implicated in different aspects of AD since it controls both glutamate receptor membrane levels and amyloid generation. The detailed understanding of the mechanisms controlling the actin cytoskeleton in a unique biological context as the dendritic spine could pave the way to the development of innovative synapse-tailored therapeutic interventions and to the identification of novel biomarkers to monitor synaptic loss in AD.
Collapse
|
13
|
Regulation of Multifunctional Calcium/Calmodulin Stimulated Protein Kinases by Molecular Targeting. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:649-679. [PMID: 31646529 DOI: 10.1007/978-3-030-12457-1_26] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Multifunctional calcium/calmodulin-stimulated protein kinases control a broad range of cellular functions in a multitude of cell types. This family of kinases contain several structural similarities and all are regulated by phosphorylation, which either activates, inhibits or modulates their kinase activity. As these protein kinases are widely or ubiquitously expressed, and yet regulate a broad range of different cellular functions, additional levels of regulation exist that control these cell-specific functions. Of particular importance for this specificity of function for multifunctional kinases is the expression of specific binding proteins that mediate molecular targeting. These molecular targeting mechanisms allow pools of kinase in different cells, or parts of a cell, to respond differently to activation and produce different functional outcomes.
Collapse
|
14
|
Pharris MC, Patel NM, VanDyk TG, Bartol TM, Sejnowski TJ, Kennedy MB, Stefan MI, Kinzer-Ursem TL. A multi-state model of the CaMKII dodecamer suggests a role for calmodulin in maintenance of autophosphorylation. PLoS Comput Biol 2019; 15:e1006941. [PMID: 31869343 PMCID: PMC6957207 DOI: 10.1371/journal.pcbi.1006941] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 01/13/2020] [Accepted: 11/25/2019] [Indexed: 02/06/2023] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) accounts for up to 2 percent of all brain protein and is essential to memory function. CaMKII activity is known to regulate dynamic shifts in the size and signaling strength of neuronal connections, a process known as synaptic plasticity. Increasingly, computational models are used to explore synaptic plasticity and the mechanisms regulating CaMKII activity. Conventional modeling approaches may exclude biophysical detail due to the impractical number of state combinations that arise when explicitly monitoring the conformational changes, ligand binding, and phosphorylation events that occur on each of the CaMKII holoenzyme's subunits. To manage the combinatorial explosion without necessitating bias or loss in biological accuracy, we use a specialized syntax in the software MCell to create a rule-based model of a twelve-subunit CaMKII holoenzyme. Here we validate the rule-based model against previous experimental measures of CaMKII activity and investigate molecular mechanisms of CaMKII regulation. Specifically, we explore how Ca2+/CaM-binding may both stabilize CaMKII subunit activation and regulate maintenance of CaMKII autophosphorylation. Noting that Ca2+/CaM and protein phosphatases bind CaMKII at nearby or overlapping sites, we compare model scenarios in which Ca2+/CaM and protein phosphatase do or do not structurally exclude each other's binding to CaMKII. Our results suggest a functional mechanism for the so-called "CaM trapping" phenomenon, wherein Ca2+/CaM may structurally exclude phosphatase binding and thereby prolong CaMKII autophosphorylation. We conclude that structural protection of autophosphorylated CaMKII by Ca2+/CaM may be an important mechanism for regulation of synaptic plasticity.
Collapse
Affiliation(s)
- Matthew C. Pharris
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
| | - Neal M. Patel
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
| | - Tyler G. VanDyk
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
| | - Thomas M. Bartol
- Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Terrence J. Sejnowski
- Salk Institute for Biological Studies, La Jolla, California, United States of America
- Institute for Neural Computation, University of California San Diego, La Jolla, California, United States of America
- Division of Biological Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Mary B. Kennedy
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Melanie I. Stefan
- Salk Institute for Biological Studies, La Jolla, California, United States of America
- EMBL-European Bioinformatics Institute, Hinxton, United Kingdom
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
- ZJU-UoE Institute, Zhejiang University, Haining, China
- * E-mail: (MIS); (TLKU)
| | - Tamara L. Kinzer-Ursem
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
- * E-mail: (MIS); (TLKU)
| |
Collapse
|
15
|
Vargas JY, Loria F, Wu Y, Córdova G, Nonaka T, Bellow S, Syan S, Hasegawa M, van Woerden GM, Trollet C, Zurzolo C. The Wnt/Ca 2+ pathway is involved in interneuronal communication mediated by tunneling nanotubes. EMBO J 2019; 38:e101230. [PMID: 31625188 PMCID: PMC6885744 DOI: 10.15252/embj.2018101230] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 09/08/2019] [Accepted: 09/12/2019] [Indexed: 01/15/2023] Open
Abstract
Tunneling nanotubes (TNTs) are actin-based transient tubular connections that allow direct communication between distant cells. TNTs play an important role in several physiological (development, immunity, and tissue regeneration) and pathological (cancer, neurodegeneration, and pathogens transmission) processes. Here, we report that the Wnt/Ca2+ pathway, an intracellular cascade that is involved in actin cytoskeleton remodeling, has a role in TNT formation and TNT-mediated transfer of cargoes. Specifically, we found that Ca2+ /calmodulin-dependent protein kinase II (CaMKII), a transducer of the Wnt/Ca2+ pathway, regulates TNTs in a neuronal cell line and in primary neurons. We identified the β isoform of CaMKII as a key molecule in modulating TNT formation and transfer, showing that this depends on the actin-binding activity of the protein. Finally, we found that the transfer of vesicles and aggregated α-synuclein between primary neurons can be regulated by the activation of the Wnt/Ca2+ pathway. Our findings suggest that Wnt/Ca2+ pathway could be a novel promising target for therapies designed to impair TNT-mediated propagation of pathogens.
Collapse
Affiliation(s)
- Jessica Y Vargas
- Unité de Trafic Membranaire et PathogénèseDépartement de Biologie Cellulaire et de l'InfectionInstitut PasteurParisFrance
| | - Frida Loria
- Unité de Trafic Membranaire et PathogénèseDépartement de Biologie Cellulaire et de l'InfectionInstitut PasteurParisFrance
- Present address:
Centro de Biología Molecular Severo Ochoa (CSIC‐UAM)Departamento de Biología MolecularUniversidad Autónoma de MadridMadridSpain
| | - Yuan‐Ju Wu
- Unité de Trafic Membranaire et PathogénèseDépartement de Biologie Cellulaire et de l'InfectionInstitut PasteurParisFrance
| | - Gonzalo Córdova
- Institut National de la Santé et de la Recherche MédicaleAssociation Institut de MyologieCentre de Recherche en MyologieUMRS974Sorbonne UniversitéParisFrance
| | - Takashi Nonaka
- Department of Dementia and Higher Brain FunctionTokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | | | - Sylvie Syan
- Unité de Trafic Membranaire et PathogénèseDépartement de Biologie Cellulaire et de l'InfectionInstitut PasteurParisFrance
| | - Masato Hasegawa
- Department of Dementia and Higher Brain FunctionTokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Geeske M van Woerden
- Department of NeuroscienceErasmus Medical CenterRotterdamThe Netherlands
- ENCORE Expertise Center for Neurodevelopmental DisordersErasmus Medical CenterRotterdamThe Netherlands
| | - Capucine Trollet
- Institut National de la Santé et de la Recherche MédicaleAssociation Institut de MyologieCentre de Recherche en MyologieUMRS974Sorbonne UniversitéParisFrance
| | - Chiara Zurzolo
- Unité de Trafic Membranaire et PathogénèseDépartement de Biologie Cellulaire et de l'InfectionInstitut PasteurParisFrance
| |
Collapse
|
16
|
Shrestha A, Sultana R, Lee CC, Ogundele OM. SK Channel Modulates Synaptic Plasticity by Tuning CaMKIIα/β Dynamics. Front Synaptic Neurosci 2019; 11:18. [PMID: 31736736 PMCID: PMC6834780 DOI: 10.3389/fnsyn.2019.00018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/20/2019] [Indexed: 11/13/2022] Open
Abstract
N-Methyl-D-Aspartate Receptor 1 (NMDAR)-linked Ca++ current represents a significant percentage of post-synaptic transient that modulates synaptic strength and is pertinent to dendritic spine plasticity. In the hippocampus, Ca++ transient produced by glutamatergic ionotropic neurotransmission facilitates Ca++-Calmodulin-dependent kinase 2 (CaMKII) Thr286 phosphorylation and promote long-term potentiation (LTP) expression. At CA1 post-synaptic densities, Ca++ transients equally activate small conductance (SK2) channel which regulates excitability by suppressing Ca++ movement. Here, we demonstrate that upstream attenuation of GluN1 function in the hippocampus led to a decrease in Thr286 CaMKIIα phosphorylation, and increased SK2 expression. Consistent with the loss of GluN1 function, potentiation of SK channel in wild type hippocampus reduced CaMKIIα expression and abrogate synaptic localization of T286 pCaMKIIα. Our results demonstrate that positive modulation of SK channel at hippocampal synapses likely refine GluN1-linked plasticity by tuning dendritic localization of CaMKIIα.
Collapse
Affiliation(s)
| | | | | | - Olalekan M. Ogundele
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| |
Collapse
|
17
|
Alexander CJ, Wagner W, Copeland NG, Jenkins NA, Hammer JA. Creation of a myosin Va-TAP-tagged mouse and identification of potential myosin Va-interacting proteins in the cerebellum. Cytoskeleton (Hoboken) 2019; 75:395-409. [PMID: 29979496 DOI: 10.1002/cm.21474] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/19/2018] [Accepted: 06/27/2018] [Indexed: 12/29/2022]
Abstract
The actin-based motor myosin Va transports numerous cargos, including the smooth endoplasmic reticulum (SER) in cerebellar Purkinje neurons (PNs) and melanosomes in melanocytes. Identifying proteins that interact with this myosin is key to understanding its cellular functions. Toward that end, we used recombineering to insert via homologous recombination a tandem affinity purification (TAP) tag composed of the immunoglobulin G-binding domain of protein A, a tobacco etch virus cleavage site, and a FLAG tag into the mouse MYO5A locus immediately after the initiation codon. Importantly, we provide evidence that the TAP-tagged version of myosin Va (TAP-MyoVa) functions normally in terms of SER transport in PNs and melanosome positioning in melanocytes. Given this and other evidence that TAP-MyoVa is fully functional, we purified it together with associated proteins directly from juvenile mouse cerebella and subjected the samples to mass spectroscopic analyses. As expected, known myosin Va-binding partners like dynein light chain were identified. Importantly, numerous novel interacting proteins were also tentatively identified, including guanine nucleotide-binding protein G(o) subunit alpha (Gnao1), a biomarker for schizophrenia. Consistently, an antibody to Gnao1 immunoprecipitates myosin Va, and Gnao1's localization to PN dendritic spines depends on myosin Va. The mouse model created here should facilitate the identification of novel myosin Va-binding partners, which in turn should advance our understanding of the roles played by this important myosin in vivo.
Collapse
Affiliation(s)
- Christopher J Alexander
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Wolfgang Wagner
- Center for Molecular Neurobiology (ZMNH), Department of Molecular Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Neal G Copeland
- The University of Texas MD Anderson, Department of Genetics, Cancer Center, Houston, Texas
| | - Nancy A Jenkins
- The University of Texas MD Anderson, Department of Genetics, Cancer Center, Houston, Texas
| | - John A Hammer
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
18
|
Conformational coupling by trans-phosphorylation in calcium calmodulin dependent kinase II. PLoS Comput Biol 2019; 15:e1006796. [PMID: 31150387 PMCID: PMC6576796 DOI: 10.1371/journal.pcbi.1006796] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 06/17/2019] [Accepted: 03/28/2019] [Indexed: 12/16/2022] Open
Abstract
The calcium calmodulin-dependent protein kinase II (CaMKII) is a dodecameric holoenzyme important for encoding memory. Its activation, triggered by binding of calcium-calmodulin, persists autonomously after calmodulin dissociation. One (receiver) kinase captures and subsequently phosphorylates the regulatory domain peptide of a donor kinase forming a chained dimer as the first stage of autonomous activation. Protein dynamics simulations examined the conformational changes triggered by dimer formation and phosphorylation, aimed to provide a molecular rationale for human mutations that result in learning disabilities. Ensembles generated from X-ray crystal structures were characterized by network centrality and community analysis. Mutual information related collective motions to local fragment dynamics encoded with a structural alphabet. Implicit solvent tCONCOORD conformational ensembles revealed the dynamic architecture of inactive kinase domains was co-opted in the activated dimer but the network hub shifted from the nucleotide binding cleft to the captured peptide. Explicit solvent molecular dynamics (MD) showed nucleotide and substrate binding determinants formed coupled nodes in long-range signal relays between regulatory peptides in the dimer. Strain in the extended captured peptide was balanced by reduced flexibility of the receiver kinase C-lobe core. The relays were organized around a hydrophobic patch between the captured peptide and a key binding helix. The human mutations aligned along the relays. Thus, these mutations could disrupt the allosteric network alternatively, or in addition, to altered binding affinities. Non-binding protein sectors distant from the binding sites mediated the allosteric signalling; providing possible targets for inhibitor design. Phosphorylation of the peptide modulated the dielectric of its binding pocket to strengthen the patch, non-binding sectors, domain interface and temporal correlations between parallel relays. These results provide the molecular details underlying the reported positive kinase cooperativity to enrich the discussion on how autonomous activation by phosphorylation leads to long-term behavioural effects. Protein kinases play central roles in intracellular signalling. Auto-phosphorylation by bound nucleotide typically precedes phosphate transfer to multiple substrates. Protein conformational changes are central to kinase function, altering binding affinities to change cellular location and shunt from one signal pathway to another. In the brain, the multi-subunit kinase, CaMKII is activated by calcium-calmodulin upon calcium jumps produced by synaptic stimulation. Auto-transphosphorylation of a regulatory peptide enables the kinase to remain activated and mediate long-term behavioural effects after return to basal calcium levels. A database of mutated residues responsible for these effects is difficult to reconcile solely with impaired nucleotide or substrate binding. Therefore, we have computationally generated interaction networks to map the conformational plasticity of the kinase domains where most mutations localize. The network generated from the atomic structure of a phosphorylated dimer resolves protein sectors based on their collective motions. The sectors link nucleotide and substrate binding sites in self-reinforcing relays between regulatory peptides. The self-reinforcement is strengthened by phosphorylation consistent with the reported positive cooperativity of kinase activity with calcium-calmodulin concentration. The network gives a better match with the mutations and, in addition, reveals target sites for drug development.
Collapse
|
19
|
Khan S, Downing KH, Molloy JE. Architectural Dynamics of CaMKII-Actin Networks. Biophys J 2018; 116:104-119. [PMID: 30527447 PMCID: PMC6341221 DOI: 10.1016/j.bpj.2018.11.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 11/02/2018] [Accepted: 11/06/2018] [Indexed: 11/24/2022] Open
Abstract
Calcium-calmodulin-dependent kinase II (CaMKII) has an important role in dendritic spine remodeling upon synaptic stimulation. Using fluorescence video microscopy and image analysis, we investigated the architectural dynamics of rhodamine-phalloidin stabilized filamentous actin (F-actin) networks cross-linked by CaMKII. We used automated image analysis to identify F-actin bundles and crossover junctions and developed a dimensionless metric to characterize network architecture. Similar networks were formed by three different CaMKII species with a 10-fold length difference in the linker region between the kinase domain and holoenzyme hub, implying linker length is not a primary determinant of F-actin cross-linking. Electron micrographs showed that at physiological molar ratios, single CaMKII holoenzymes cross-linked multiple F-actin filaments at random, whereas at higher CaMKII/F-actin ratios, filaments bundled. Light microscopy established that the random network architecture resisted macromolecular crowding with polyethylene glycol and blocked ATP-powered compaction by myosin-II miniature filaments. Importantly, the networks disassembled after the addition of calcium-calmodulin and were then spaced within 3 min into compacted foci by myosin motors or more slowly (30 min) aggregated by crowding. Single-molecule total internal reflection fluorescence microscopy showed CaMKII dissociation from surface-immobilized globular actin exhibited a monoexponential dwell-time distribution, whereas CaMKII bound to F-actin networks had a long-lived fraction, trapped at crossover junctions. Release of CaMKII from F-actin, triggered by calcium-calmodulin, was too rapid to measure with flow-cell exchange (<20 s). The residual bound fraction was reduced substantially upon addition of an N-methyl-D-aspartate receptor peptide analog but not ATP. These results provide mechanistic insights to CaMKII-actin interactions at the collective network and single-molecule level. Our findings argue that CaMKII-actin networks in dendritic spines maintain spine size against physical stress. Upon synaptic stimulation, CaMKII is disengaged by calcium-calmodulin, triggering network disassembly, expansion, and subsequent compaction by myosin motors with kinetics compatible with the times recorded for the poststimulus changes in spine volume.
Collapse
Affiliation(s)
- Shahid Khan
- Molecular Biology Consortium, Lawrence Berkeley National Laboratory, Berkeley, California; The Francis Crick Institute, London, United Kingdom.
| | - Kenneth H Downing
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California
| | | |
Collapse
|
20
|
Borovac J, Bosch M, Okamoto K. Regulation of actin dynamics during structural plasticity of dendritic spines: Signaling messengers and actin-binding proteins. Mol Cell Neurosci 2018; 91:122-130. [PMID: 30004015 DOI: 10.1016/j.mcn.2018.07.001] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 06/25/2018] [Accepted: 07/06/2018] [Indexed: 12/17/2022] Open
Abstract
Activity-dependent plasticity of synaptic structure and function plays an essential role in neuronal development and in cognitive functions including learning and memory. The formation, maintenance and modulation of dendritic spines are mainly controlled by the dynamics of actin filaments (F-actin) through interaction with various actin-binding proteins (ABPs) and postsynaptic signaling messengers. Induction of long-term potentiation (LTP) triggers a cascade of events involving Ca2+ signaling, intracellular pathways such as cAMP and cGMP, and regulation of ABPs such as CaMKII, Cofilin, Aip1, Arp2/3, α-actinin, Profilin and Drebrin. We review here how these ABPs modulate the rate of assembly, disassembly, stabilization and bundling of F-actin during LTP induction. We highlight the crucial role that CaMKII exerts in both functional and structural plasticity by directly coupling Ca2+ signaling with F-actin dynamics through the β subunit. Moreover, we show how cAMP and cGMP second messengers regulate postsynaptic structural potentiation. Brain disorders such as Alzheimer's disease, schizophrenia or autism, are associated with alterations in the regulation of F-actin dynamics by these ABPs and signaling messengers. Thus, a better understanding of the molecular mechanisms controlling actin cytoskeleton can provide cues for the treatment of these disorders.
Collapse
Affiliation(s)
- Jelena Borovac
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Miquel Bosch
- Institute for Bioengineering of Catalonia, Barcelona 08028, Spain.
| | - Kenichi Okamoto
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON M5G 1X5, Canada.
| |
Collapse
|
21
|
Joensuu M, Lanoue V, Hotulainen P. Dendritic spine actin cytoskeleton in autism spectrum disorder. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:362-381. [PMID: 28870634 DOI: 10.1016/j.pnpbp.2017.08.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/21/2017] [Accepted: 08/30/2017] [Indexed: 01/01/2023]
Abstract
Dendritic spines are small actin-rich protrusions from neuronal dendrites that form the postsynaptic part of most excitatory synapses. Changes in the shape and size of dendritic spines correlate with the functional changes in excitatory synapses and are heavily dependent on the remodeling of the underlying actin cytoskeleton. Recent evidence implicates synapses at dendritic spines as important substrates of pathogenesis in neuropsychiatric disorders, including autism spectrum disorder (ASD). Although synaptic perturbations are not the only alterations relevant for these diseases, understanding the molecular underpinnings of the spine and synapse pathology may provide insight into their etiologies and could reveal new drug targets. In this review, we will discuss recent findings of defective actin regulation in dendritic spines associated with ASD.
Collapse
Affiliation(s)
- Merja Joensuu
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland; Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Vanessa Lanoue
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Pirta Hotulainen
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland.
| |
Collapse
|
22
|
Analysis of the CaMKIIα and β splice-variant distribution among brain regions reveals isoform-specific differences in holoenzyme formation. Sci Rep 2018; 8:5448. [PMID: 29615706 PMCID: PMC5882894 DOI: 10.1038/s41598-018-23779-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/20/2018] [Indexed: 12/21/2022] Open
Abstract
Four CaMKII isoforms are encoded by distinct genes, and alternative splicing within the variable linker-region generates additional diversity. The α and β isoforms are largely brain-specific, where they mediate synaptic functions underlying learning, memory and cognition. Here, we determined the α and β splice-variant distribution among different mouse brain regions. Surprisingly, the nuclear variant αB was detected in all regions, and even dominated in hypothalamus and brain stem. For CaMKIIβ, the full-length variant dominated in most regions (with higher amounts of minor variants again seen in hypothalamus and brain stem). The mammalian but not fish CaMKIIβ gene lacks exon v3N that encodes the nuclear localization signal in αB, but contains three exons not found in the CaMKIIα gene (exons v1, v4, v5). While skipping of exons v1 and/or v5 generated the minor splice-variants β’, βe and βe’, essentially all transcripts contained exon v4. However, we instead detected another minor splice-variant (now termed βH), which lacks part of the hub domain that mediates formation of CaMKII holoenzymes. Surprisingly, in an optogenetic cellular assay of protein interactions, CaMKIIβH was impaired for binding to the β hub domain, but still bound CaMKIIα. This provides the first indication for isoform-specific differences in holoenzyme formation.
Collapse
|
23
|
Sáez PJ, Vargas P, Shoji KF, Harcha PA, Lennon-Duménil AM, Sáez JC. ATP promotes the fast migration of dendritic cells through the activity of pannexin 1 channels and P2X 7 receptors. Sci Signal 2017; 10:10/506/eaah7107. [PMID: 29162744 DOI: 10.1126/scisignal.aah7107] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Upon its release from injured cells, such as infected, transformed, inflamed, or necrotic cells, extracellular adenosine-5'-triphosphate (ATP) acts as a danger signal that recruits phagocytes, such as neutrophils, macrophages, and dendritic cells (DCs), to the site of injury. The sensing of extracellular ATP occurs through purinergic (P2) receptors. We investigated the cellular mechanisms linking purinergic signaling to DC motility. We found that ATP stimulated fast DC motility through an autocrine signaling loop, which was initiated by the activation of P2X7 receptors and further amplified by pannexin 1 (Panx1) channels. Upon stimulation of the P2X7 receptor by ATP, Panx1 contributed to fast DC motility by increasing the permeability of the plasma membrane, which resulted in supplementary ATP release. In the absence of Panx1, DCs failed to increase their speed of migration in response to ATP, despite exhibiting a normal P2X7 receptor-mediated Ca2+ response. In addition to DC migration, Panx1 channel- and P2X7 receptor-dependent signaling was further required to stimulate the reorganization of the actin cytoskeleton. In vivo, functional Panx1 channels were required for the homing of DCs to lymph nodes, although they were dispensable for DC maturation. These data suggest that P2X7 receptors and Panx1 channels are crucial players in the regulation of DC migration to endogenous danger signals.
Collapse
Affiliation(s)
- Pablo J Sáez
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 6513677, Chile. .,INSERM U932 Immunité et Cancer, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 12 Rue Lhomond, Paris 75005, France
| | - Pablo Vargas
- INSERM U932 Immunité et Cancer, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 12 Rue Lhomond, Paris 75005, France.,CNRS UMR144, Institut Curie, PSL Research University, 12 Rue Lhomond, Paris 75005, France
| | - Kenji F Shoji
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 6513677, Chile
| | - Paloma A Harcha
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 6513677, Chile.,Instituto Milenio, Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso 2360103, Chile
| | - Ana-María Lennon-Duménil
- INSERM U932 Immunité et Cancer, Institut Curie, Paris Sciences et Lettres (PSL) Research University, 12 Rue Lhomond, Paris 75005, France.
| | - Juan C Sáez
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 6513677, Chile. .,Instituto Milenio, Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso 2360103, Chile
| |
Collapse
|