1
|
Narayanan D, Larsen ASG, Gauger SJ, Adafia R, Hammershøi RB, Hamborg L, Bruus‐Jensen J, Griem‐Krey N, Gee CL, Frølund B, Stratton MM, Kuriyan J, Kastrup JS, Langkilde AE, Wellendorph P, Solbak SMØ. Ligand-induced CaMKIIα hub Trp403 flip, hub domain stacking, and modulation of kinase activity. Protein Sci 2024; 33:e5152. [PMID: 39275999 PMCID: PMC11400628 DOI: 10.1002/pro.5152] [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: 04/10/2024] [Revised: 07/09/2024] [Accepted: 08/08/2024] [Indexed: 09/16/2024]
Abstract
γ-Hydroxybutyric acid (GHB) analogs are small molecules that bind competitively to a specific cavity in the oligomeric CaMKIIα hub domain. Binding affects conformation and stability of the hub domain, which may explain the neuroprotective action of some of these compounds. Here, we describe molecular details of interaction of the larger-type GHB analog 2-(6-(4-chlorophenyl)imidazo[1,2-b]pyridazine-2-yl)acetic acid (PIPA). Like smaller-type analogs, PIPA binding to the CaMKIIα hub domain promoted thermal stability. PIPA additionally modulated CaMKIIα activity under sub-maximal CaM concentrations and ultimately led to reduced substrate phosphorylation. A high-resolution X-ray crystal structure of a stabilized CaMKIIα (6x mutant) hub construct revealed details of the binding mode of PIPA, which involved outward placement of tryptophan 403 (Trp403), a central residue in a flexible loop close to the upper hub cavity. Small-angle X-ray scattering (SAXS) solution structures and mass photometry of the CaMKIIα wild-type hub domain in the presence of PIPA revealed a high degree of ordered self-association (stacks of CaMKIIα hub domains). This stacking neither occurred with the smaller compound 3-hydroxycyclopent-1-enecarboxylic acid (HOCPCA), nor when Trp403 was replaced with leucine (W403L). Additionally, CaMKIIα W403L hub was stabilized to a larger extent by PIPA compared to CaMKIIα hub wild type, indicating that loop flexibility is important for holoenzyme stability. Thus, we propose that ligand-induced outward placement of Trp403 by PIPA, which promotes an unforeseen mechanism of hub domain stacking, may be involved in the observed reduction in CaMKIIα kinase activity. Altogether, this sheds new light on allosteric regulation of CaMKIIα activity via the hub domain.
Collapse
Affiliation(s)
- Dilip Narayanan
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Anne Sofie G. Larsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Stine Juul Gauger
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Ruth Adafia
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
- Chemistry‐Biology Interface Training ProgramUniversity of MassachusettsAmherstMassachusettsUSA
| | - Rikke Bartschick Hammershøi
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Louise Hamborg
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Jesper Bruus‐Jensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Nane Griem‐Krey
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Christine L. Gee
- HHMIUniversity of CaliforniaBerkeleyCaliforniaUSA
- Department of Molecular and Cell BiologyUniversity of CaliforniaBerkeleyCaliforniaUSA
- California Institute for Quantitative BiosciencesUniversity of CaliforniaBerkeleyCaliforniaUSA
- Department of BiochemistryVanderbilt University School of MedicineNashvilleTennesseeUSA
| | - Bente Frølund
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Margaret M. Stratton
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
| | - John Kuriyan
- HHMIUniversity of CaliforniaBerkeleyCaliforniaUSA
- Department of Molecular and Cell BiologyUniversity of CaliforniaBerkeleyCaliforniaUSA
- California Institute for Quantitative BiosciencesUniversity of CaliforniaBerkeleyCaliforniaUSA
- Department of BiochemistryVanderbilt University School of MedicineNashvilleTennesseeUSA
- Department of ChemistryUniversity of CaliforniaBerkeleyCaliforniaUSA
- Physical Biosciences DivisionLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Jette Sandholm Kastrup
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Annette E. Langkilde
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Petrine Wellendorph
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Sara M. Ø. Solbak
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| |
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
|
Brown CN, Bayer KU. Studying CaMKII: Tools and standards. Cell Rep 2024; 43:113982. [PMID: 38517893 PMCID: PMC11088445 DOI: 10.1016/j.celrep.2024.113982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/19/2024] [Accepted: 03/06/2024] [Indexed: 03/24/2024] Open
Abstract
The Ca2+/calmodulin (CaM)-dependent protein kinase II (CaMKII) is a ubiquitous mediator of cellular Ca2+ signals with both enzymatic and structural functions. Here, we briefly introduce the complex regulation of CaMKII and then provide a comprehensive overview of the expanding toolbox to study CaMKII. Beyond a variety of distinct mutants, these tools now include optical methods for measurement and manipulation, with the latter including light-induced inhibition, stimulation, and sequestration. Perhaps most importantly, there are now three mechanistically distinct classes of specific CaMKII inhibitors, and their combined use enables the interrogation of CaMKII functions in a manner that is powerful and sophisticated yet also accessible. This review aims to provide guidelines for the interpretation of the results obtained with these tools, with careful consideration of their direct and indirect effects.
Collapse
Affiliation(s)
- Carolyn Nicole Brown
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Karl Ulrich Bayer
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
4
|
Chacar S, Abdi A, Almansoori K, Alshamsi J, Al Hageh C, Zalloua P, Khraibi AA, Holt SG, Nader M. Role of CaMKII in diabetes induced vascular injury and its interaction with anti-diabetes therapy. Rev Endocr Metab Disord 2024; 25:369-382. [PMID: 38064002 PMCID: PMC10943158 DOI: 10.1007/s11154-023-09855-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2023] [Indexed: 03/16/2024]
Abstract
Diabetes mellitus is a metabolic disorder denoted by chronic hyperglycemia that drives maladaptive structural changes and functional damage to the vasculature. Attenuation of this pathological remodeling of blood vessels remains an unmet target owing to paucity of information on the metabolic signatures of this process. Ca2+/calmodulin-dependent kinase II (CaMKII) is expressed in the vasculature and is implicated in the control of blood vessels homeostasis. Recently, CaMKII has attracted a special attention in view of its chronic upregulated activity in diabetic tissues, yet its role in the diabetic vasculature remains under investigation.This review highlights the physiological and pathological actions of CaMKII in the diabetic vasculature, with focus on the control of the dialogue between endothelial (EC) and vascular smooth muscle cells (VSMC). Activation of CaMKII enhances EC and VSMC proliferation and migration, and increases the production of extracellular matrix which leads to maladaptive remodeling of vessels. This is manifested by activation of genes/proteins implicated in the control of the cell cycle, cytoskeleton organization, proliferation, migration, and inflammation. Endothelial dysfunction is paralleled by impaired nitric oxide signaling, which is also influenced by CaMKII signaling (activation/oxidation). The efficiency of CaMKII inhibitors is currently being tested in animal models, with a focus on the genetic pathways involved in the regulation of CaMKII expression (microRNAs and single nucleotide polymorphisms). Interestingly, studies highlight an interaction between the anti-diabetic drugs and CaMKII expression/activity which requires further investigation. Together, the studies reviewed herein may guide pharmacological approaches to improve health-related outcomes in patients with diabetes.
Collapse
Affiliation(s)
- Stephanie Chacar
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.
- Center for Biotechnology, Khalifa University of Science and Technology, 127788, Abu Dhabi, United Arab Emirates.
| | - Abdulhamid Abdi
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Khalifa Almansoori
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Jawaher Alshamsi
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Cynthia Al Hageh
- Department of Molecular Biology and Genetics, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Pierre Zalloua
- Department of Molecular Biology and Genetics, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University of Science and Technology, 127788, Abu Dhabi, United Arab Emirates
| | - Ali A Khraibi
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University of Science and Technology, 127788, Abu Dhabi, United Arab Emirates
| | - Stephen G Holt
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- SEHA Kidney Care, SEHA, Abu Dhabi, UAE
| | - Moni Nader
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.
- Center for Biotechnology, Khalifa University of Science and Technology, 127788, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
5
|
Özden C, MacManus S, Adafia R, Samkutty A, Torres‐Ocampo AP, Garman SC, Stratton MM. Ca2+/CaM dependent protein kinase II (CaMKII)α and CaMKIIβ hub domains adopt distinct oligomeric states and stabilities. Protein Sci 2024; 33:e4960. [PMID: 38501502 PMCID: PMC10962473 DOI: 10.1002/pro.4960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/05/2024] [Accepted: 02/22/2024] [Indexed: 03/20/2024]
Abstract
Ca2+ /calmodulin-dependent protein kinase II (CaMKII) is a multidomain serine/threonine kinase that plays important roles in the brain, heart, muscle tissue, and eggs/sperm. The N-terminal kinase and regulatory domain is connected by a flexible linker to the C-terminal hub domain. The hub domain drives the oligomeric organization of CaMKII, assembling the kinase domains into high local concentration. Previous structural studies have shown multiple stoichiometries of the holoenzyme as well as the hub domain alone. Here, we report a comprehensive study of the hub domain stoichiometry and stability in solution. We solved two crystal structures of the CaMKIIβ hub domain that show 14-mer (3.1 Å) and 16-mer (3.4 Å) assemblies. Both crystal structures were determined from crystals grown in the same drop, which suggests that CaMKII oligomers with different stoichiometries likely coexist. To further interrogate hub stability, we employed mass photometry and temperature denaturation studies of CaMKIIβ and CaMKIIα hubs, which highlight major differences between these highly similar domains. We created a dimeric CaMKIIβ hub unit using rational mutagenesis, which is significantly less stable than the oligomer. Both hub domains populate an intermediate during unfolding. We found that multiple CaMKIIβ hub stoichiometries are present in solution and that larger oligomers are more stable. CaMKIIα had a narrower distribution of molecular weight and was distinctly more stable than CaMKIIβ.
Collapse
Affiliation(s)
- Can Özden
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
- Molecular and Cellular Biology Graduate ProgramUniversity of MassachusettsAmherstMassachusettsUSA
| | - Sara MacManus
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
| | - Ruth Adafia
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
- Molecular and Cellular Biology Graduate ProgramUniversity of MassachusettsAmherstMassachusettsUSA
| | - Alfred Samkutty
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
| | - Ana P. Torres‐Ocampo
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
- Molecular and Cellular Biology Graduate ProgramUniversity of MassachusettsAmherstMassachusettsUSA
| | - Scott C. Garman
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
| | - Margaret M. Stratton
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
| |
Collapse
|
6
|
Lučić I, Héluin L, Jiang PL, Castro Scalise AG, Wang C, Franz A, Heyd F, Wahl MC, Liu F, Plested AJR. CaMKII autophosphorylation can occur between holoenzymes without subunit exchange. eLife 2023; 12:e86090. [PMID: 37566455 PMCID: PMC10468207 DOI: 10.7554/elife.86090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 08/10/2023] [Indexed: 08/12/2023] Open
Abstract
The dodecameric protein kinase CaMKII is expressed throughout the body. The alpha isoform is responsible for synaptic plasticity and participates in memory through its phosphorylation of synaptic proteins. Its elaborate subunit organization and propensity for autophosphorylation allow it to preserve neuronal plasticity across space and time. The prevailing hypothesis for the spread of CaMKII activity, involving shuffling of subunits between activated and naive holoenzymes, is broadly termed subunit exchange. In contrast to the expectations of previous work, we found little evidence for subunit exchange upon activation, and no effect of restraining subunits to their parent holoenzymes. Rather, mass photometry, crosslinking mass spectrometry, single molecule TIRF microscopy and biochemical assays identify inter-holoenzyme phosphorylation (IHP) as the mechanism for spreading phosphorylation. The transient, activity-dependent formation of groups of holoenzymes is well suited to the speed of neuronal activity. Our results place fundamental limits on the activation mechanism of this kinase.
Collapse
Affiliation(s)
- Iva Lučić
- Institute of Biology, Cellular Biophysics, Humboldt Universität zu BerlinBerlinGermany
- Leibniz-Forschungsinstitut für Molekulare PharmakologieBerlinGermany
| | - Léonie Héluin
- Institute of Biology, Cellular Biophysics, Humboldt Universität zu BerlinBerlinGermany
- Leibniz-Forschungsinstitut für Molekulare PharmakologieBerlinGermany
| | - Pin-Lian Jiang
- Leibniz-Forschungsinstitut für Molekulare PharmakologieBerlinGermany
| | - Alejandro G Castro Scalise
- Institute of Biology, Cellular Biophysics, Humboldt Universität zu BerlinBerlinGermany
- Leibniz-Forschungsinstitut für Molekulare PharmakologieBerlinGermany
| | - Cong Wang
- Leibniz-Forschungsinstitut für Molekulare PharmakologieBerlinGermany
| | - Andreas Franz
- Institute of Chemistry and Biochemistry, Freie Universität BerlinBerlinGermany
| | - Florian Heyd
- Institute of Chemistry and Biochemistry, Freie Universität BerlinBerlinGermany
| | - Markus C Wahl
- Institute of Chemistry and Biochemistry, Freie Universität BerlinBerlinGermany
- Helmholtz-Zentrum Berlin für Materialien und Energie, Macromolecular CrystallographyBerlinGermany
| | - Fan Liu
- Leibniz-Forschungsinstitut für Molekulare PharmakologieBerlinGermany
- Charité-Universitätsmedizin BerlinBerlinGermany
| | - Andrew JR Plested
- Institute of Biology, Cellular Biophysics, Humboldt Universität zu BerlinBerlinGermany
- Leibniz-Forschungsinstitut für Molekulare PharmakologieBerlinGermany
- NeuroCure, Charité UniversitätsmedizinBerlinGermany
| |
Collapse
|
7
|
Ameen SS, Griem-Krey N, Dufour A, Hossain MI, Hoque A, Sturgeon S, Nandurkar H, Draxler DF, Medcalf RL, Kamaruddin MA, Lucet IS, Leeming MG, Liu D, Dhillon A, Lim JP, Basheer F, Zhu HJ, Bokhari L, Roulston CL, Paradkar PN, Kleifeld O, Clarkson AN, Wellendorph P, Ciccotosto GD, Williamson NA, Ang CS, Cheng HC. N-Terminomic Changes in Neurons During Excitotoxicity Reveal Proteolytic Events Associated With Synaptic Dysfunctions and Potential Targets for Neuroprotection. Mol Cell Proteomics 2023; 22:100543. [PMID: 37030595 PMCID: PMC10199228 DOI: 10.1016/j.mcpro.2023.100543] [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: 07/07/2022] [Revised: 02/23/2023] [Accepted: 04/04/2023] [Indexed: 04/10/2023] Open
Abstract
Excitotoxicity, a neuronal death process in neurological disorders such as stroke, is initiated by the overstimulation of ionotropic glutamate receptors. Although dysregulation of proteolytic signaling networks is critical for excitotoxicity, the identity of affected proteins and mechanisms by which they induce neuronal cell death remain unclear. To address this, we used quantitative N-terminomics to identify proteins modified by proteolysis in neurons undergoing excitotoxic cell death. We found that most proteolytically processed proteins in excitotoxic neurons are likely substrates of calpains, including key synaptic regulatory proteins such as CRMP2, doublecortin-like kinase I, Src tyrosine kinase and calmodulin-dependent protein kinase IIβ (CaMKIIβ). Critically, calpain-catalyzed proteolytic processing of these proteins generates stable truncated fragments with altered activities that potentially contribute to neuronal death by perturbing synaptic organization and function. Blocking calpain-mediated proteolysis of one of these proteins, Src, protected against neuronal loss in a rat model of neurotoxicity. Extrapolation of our N-terminomic results led to the discovery that CaMKIIα, an isoform of CaMKIIβ, undergoes differential processing in mouse brains under physiological conditions and during ischemic stroke. In summary, by identifying the neuronal proteins undergoing proteolysis during excitotoxicity, our findings offer new insights into excitotoxic neuronal death mechanisms and reveal potential neuroprotective targets for neurological disorders.
Collapse
Affiliation(s)
- S Sadia Ameen
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Nane Griem-Krey
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Antoine Dufour
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - M Iqbal Hossain
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia; Department of Pharmacology and Toxicology, University of Alabama, Birmingham, Alabama, USA
| | - Ashfaqul Hoque
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Sharelle Sturgeon
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Harshal Nandurkar
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Dominik F Draxler
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Robert L Medcalf
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Mohd Aizuddin Kamaruddin
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Isabelle S Lucet
- Chemical Biology Division, The Walter and Eliza Hall Institute for Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Michael G Leeming
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Dazhi Liu
- Department of Neurology, School of Medicine, University of California, Davis, California, USA
| | - Amardeep Dhillon
- Faculty of Health, Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Jet Phey Lim
- Faculty of Health, Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Faiza Basheer
- Faculty of Health, Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Hong-Jian Zhu
- Department of Surgery (Royal Melbourne Hospital), University of Melbourne, Parkville, Victoria, Australia
| | - Laita Bokhari
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Carli L Roulston
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Prasad N Paradkar
- CSIRO Health & Biosecurity, Australian Centre for Disease Preparedness, East Geelong, Victoria, Australia
| | - Oded Kleifeld
- Faculty of Biology, Technion-Israel Institute of Technology, Technion City, Haifa, Israel
| | - Andrew N Clarkson
- Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Petrine Wellendorph
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Giuseppe D Ciccotosto
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia.
| | - Nicholas A Williamson
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia.
| | - Ching-Seng Ang
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia.
| | - Heung-Chin Cheng
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
8
|
Calcium/Calmodulin-Stimulated Protein Kinase II (CaMKII): Different Functional Outcomes from Activation, Depending on the Cellular Microenvironment. Cells 2023; 12:cells12030401. [PMID: 36766743 PMCID: PMC9913510 DOI: 10.3390/cells12030401] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/20/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Calcium/calmodulin-stimulated protein kinase II (CaMKII) is a family of broad substrate specificity serine (Ser)/threonine (Thr) protein kinases widely expressed in many tissues that is capable of mediating diverse functional responses depending on its cellular and molecular microenvironment. This review briefly summarises current knowledge on the structure and regulation of CaMKII and focuses on how the molecular environment, and interaction with binding partner proteins, can produce different populations of CaMKII in different cells, or in different subcellular locations within the same cell, and how these different populations of CaMKII can produce diverse functional responses to activation following an increase in intracellular calcium concentration. This review also explores the possibility that identifying and characterising the molecular interactions responsible for the molecular targeting of CaMKII in different cells in vivo, and identifying the sites on CaMKII and/or the binding proteins through which these interactions occur, could lead to the development of highly selective inhibitors of specific CaMKII-mediated functional responses in specific cells that would not affect CaMKII-mediated responses in other cells. This may result in the development of new pharmacological agents with therapeutic potential for many clinical conditions.
Collapse
|
9
|
The CaMKIIα hub ligand Ph-HTBA promotes neuroprotection after focal ischemic stroke by a distinct molecular interaction. Biomed Pharmacother 2022; 156:113895. [DOI: 10.1016/j.biopha.2022.113895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 11/23/2022] Open
|
10
|
Griem-Krey N, Clarkson AN, Wellendorph P. CaMKIIα as a Promising Drug Target for Ischemic Grey Matter. Brain Sci 2022; 12:1639. [PMID: 36552099 PMCID: PMC9775128 DOI: 10.3390/brainsci12121639] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/26/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a major mediator of Ca2+-dependent signaling pathways in various cell types throughout the body. Its neuronal isoform CaMKIIα (alpha) centrally integrates physiological but also pathological glutamate signals directly downstream of glutamate receptors and has thus emerged as a target for ischemic stroke. Previous studies provided evidence for the involvement of CaMKII activity in ischemic cell death by showing that CaMKII inhibition affords substantial neuroprotection. However, broad inhibition of this central kinase is challenging because various essential physiological processes like synaptic plasticity rely on intact CaMKII regulation. Thus, specific strategies for targeting CaMKII after ischemia are warranted which would ideally only interfere with pathological activity of CaMKII. This review highlights recent advances in the understanding of how ischemia affects CaMKII and how pathospecific pharmacological targeting of CaMKII signaling could be achieved. Specifically, we discuss direct targeting of CaMKII kinase activity with peptide inhibitors versus indirect targeting of the association (hub) domain of CaMKIIα with analogues of γ-hydroxybutyrate (GHB) as a potential way to achieve more specific pharmacological modulation of CaMKII activity after ischemia.
Collapse
Affiliation(s)
- Nane Griem-Krey
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Andrew N. Clarkson
- Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin 9016, New Zealand
| | - Petrine Wellendorph
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark
| |
Collapse
|
11
|
Bredow M, Monaghan J. Cross-kingdom regulation of calcium- and/or calmodulin-dependent protein kinases by phospho-switches that relieve autoinhibition. CURRENT OPINION IN PLANT BIOLOGY 2022; 68:102251. [PMID: 35767936 DOI: 10.1016/j.pbi.2022.102251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/04/2022] [Accepted: 05/26/2022] [Indexed: 06/15/2023]
Abstract
Mechanisms to sense and respond to calcium have evolved in all organisms. Calmodulin is a universal calcium sensor across eukaryotes that directly binds calcium and associates with many downstream signal transducers including protein kinases. All eukaryotes encode calcium-dependent and/or calmodulin-dependent kinases, however there are distinct protein families across kingdoms. Here, we compare the activation mechanisms of calmodulin-dependent protein kinases (CaMKs), calcium- and calmodulin-dependent protein kinases (CCaMKs) and calcium-dependent protein kinases (CDPKs), noting striking similarities regarding phosphorylation in a regulatory segment known as the autoinhibitory junction. We thus propose that conserved regulation by phosphorylation underlies the activation of calcium-responsive proteins from different kingdoms.
Collapse
Affiliation(s)
- Melissa Bredow
- Department of Plant Pathology and Microbiology, Iowa State University, Ames IA, USA.
| | | |
Collapse
|
12
|
Mohanan AG, Gunasekaran S, Jacob RS, Omkumar RV. Role of Ca2+/Calmodulin-Dependent Protein Kinase Type II in Mediating Function and Dysfunction at Glutamatergic Synapses. Front Mol Neurosci 2022; 15:855752. [PMID: 35795689 PMCID: PMC9252440 DOI: 10.3389/fnmol.2022.855752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/21/2022] [Indexed: 01/25/2023] Open
Abstract
Glutamatergic synapses harbor abundant amounts of the multifunctional Ca2+/calmodulin-dependent protein kinase type II (CaMKII). Both in the postsynaptic density as well as in the cytosolic compartment of postsynaptic terminals, CaMKII plays major roles. In addition to its Ca2+-stimulated kinase activity, it can also bind to a variety of membrane proteins at the synapse and thus exert spatially restricted activity. The abundance of CaMKII in glutamatergic synapse is akin to scaffolding proteins although its prominent function still appears to be that of a kinase. The multimeric structure of CaMKII also confers several functional capabilities on the enzyme. The versatility of the enzyme has prompted hypotheses proposing several roles for the enzyme such as Ca2+ signal transduction, memory molecule function and scaffolding. The article will review the multiple roles played by CaMKII in glutamatergic synapses and how they are affected in disease conditions.
Collapse
Affiliation(s)
- Archana G. Mohanan
- Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Sowmya Gunasekaran
- Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- Research Scholar, Manipal Academy of Higher Education, Manipal, India
| | - Reena Sarah Jacob
- Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- Research Scholar, Manipal Academy of Higher Education, Manipal, India
| | - R. V. Omkumar
- Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- *Correspondence: R. V. Omkumar,
| |
Collapse
|
13
|
Jensen EL, Receveur-Brechot V, Hachemane M, Wils L, Barbier P, Parsiegla G, Gontero B, Launay H. Structural Contour Map of the Iota Carbonic Anhydrase from the Diatom Thalassiosira pseudonana Using a Multiprong Approach. Int J Mol Sci 2021; 22:ijms22168723. [PMID: 34445427 PMCID: PMC8395977 DOI: 10.3390/ijms22168723] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 02/07/2023] Open
Abstract
Carbonic anhydrases (CAs) are a family of ubiquitous enzymes that catalyze the interconversion of CO2 and HCO3−. The “iota” class (ι-CA) was first found in the marine diatom Thalassiosira pseudonana (tpι-CA) and is widespread among photosynthetic microalgae and prokaryotes. The ι-CA has a domain COG4875 (or COG4337) that can be repeated from one to several times and resembles a calcium–calmodulin protein kinase II association domain (CaMKII-AD). The crystal structure of this domain in the ι-CA from a cyanobacterium and a chlorarachniophyte has been recently determined. However, the three-dimensional organization of the four domain-containing tpι-CA is unknown. Using biophysical techniques and 3-D modeling, we show that the homotetrameric tpι-CA in solution has a flat “drone-like” shape with a core formed by the association of the first two domains of each monomer, and four protruding arms formed by domains 3 and 4. We also observe that the short linker between domains 3 and 4 in each monomer confers high flexibility, allowing for different conformations to be adopted. We propose the possible 3-D structure of a truncated tpι-CA containing fewer domain repeats using experimental data and discuss the implications of this atypical shape on the activity and metal coordination of the ι-CA.
Collapse
Affiliation(s)
- Erik L. Jensen
- Aix Marseille Univ, CNRS, BIP, UMR 7281, IMM, FR 3479, 31 Chemin J. Aiguier, CEDEX 20, 13 402 Marseille, France; (E.L.J.); (V.R.-B.); (M.H.); (L.W.); (G.P.)
| | - Véronique Receveur-Brechot
- Aix Marseille Univ, CNRS, BIP, UMR 7281, IMM, FR 3479, 31 Chemin J. Aiguier, CEDEX 20, 13 402 Marseille, France; (E.L.J.); (V.R.-B.); (M.H.); (L.W.); (G.P.)
| | - Mohand Hachemane
- Aix Marseille Univ, CNRS, BIP, UMR 7281, IMM, FR 3479, 31 Chemin J. Aiguier, CEDEX 20, 13 402 Marseille, France; (E.L.J.); (V.R.-B.); (M.H.); (L.W.); (G.P.)
| | - Laura Wils
- Aix Marseille Univ, CNRS, BIP, UMR 7281, IMM, FR 3479, 31 Chemin J. Aiguier, CEDEX 20, 13 402 Marseille, France; (E.L.J.); (V.R.-B.); (M.H.); (L.W.); (G.P.)
| | - Pascale Barbier
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13 402 Marseille, France;
| | - Goetz Parsiegla
- Aix Marseille Univ, CNRS, BIP, UMR 7281, IMM, FR 3479, 31 Chemin J. Aiguier, CEDEX 20, 13 402 Marseille, France; (E.L.J.); (V.R.-B.); (M.H.); (L.W.); (G.P.)
| | - Brigitte Gontero
- Aix Marseille Univ, CNRS, BIP, UMR 7281, IMM, FR 3479, 31 Chemin J. Aiguier, CEDEX 20, 13 402 Marseille, France; (E.L.J.); (V.R.-B.); (M.H.); (L.W.); (G.P.)
- Correspondence: (B.G.); (H.L.)
| | - Hélène Launay
- Aix Marseille Univ, CNRS, BIP, UMR 7281, IMM, FR 3479, 31 Chemin J. Aiguier, CEDEX 20, 13 402 Marseille, France; (E.L.J.); (V.R.-B.); (M.H.); (L.W.); (G.P.)
- Correspondence: (B.G.); (H.L.)
| |
Collapse
|
14
|
GHB analogs confer neuroprotection through specific interaction with the CaMKIIα hub domain. Proc Natl Acad Sci U S A 2021; 118:2108079118. [PMID: 34330837 PMCID: PMC8346900 DOI: 10.1073/pnas.2108079118] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
GHB is a natural brain metabolite of GABA, previously reported to be neuroprotective. However, the high-affinity binding site for GHB has remained elusive for almost 40 y. We here unveil CaMKIIα, a highly important neuronal kinase, as the long-sought-after GHB high-affinity target. Via a specific interaction within the central hub domain of CaMKIIα, GHB analogs act to stabilize the hub oligomer complex. This interaction potentially explains pronounced neuroprotective effects of GHB analogs in cultured neurons exposed to a chemical insult and in mice exposed to ischemia. The postischemic treatment effects of GHB analogs underline these compounds as selective and high-affinity potential drug candidates and CaMKIIα as a relevant pharmacological target for stroke therapy. Ca2+/calmodulin-dependent protein kinase II alpha subunit (CaMKIIα) is a key neuronal signaling protein and an emerging drug target. The central hub domain regulates the activity of CaMKIIα by organizing the holoenzyme complex into functional oligomers, yet pharmacological modulation of the hub domain has never been demonstrated. Here, using a combination of photoaffinity labeling and chemical proteomics, we show that compounds related to the natural substance γ-hydroxybutyrate (GHB) bind selectively to CaMKIIα. By means of a 2.2-Å x-ray crystal structure of ligand-bound CaMKIIα hub, we reveal the molecular details of the binding site deep within the hub. Furthermore, we show that binding of GHB and related analogs to this site promotes concentration-dependent increases in hub thermal stability believed to alter holoenzyme functionality. Selectively under states of pathological CaMKIIα activation, hub ligands provide a significant and sustained neuroprotection, which is both time and dose dependent. This is demonstrated in neurons exposed to excitotoxicity and in a mouse model of cerebral ischemia with the selective GHB analog, HOCPCA (3-hydroxycyclopent-1-enecarboxylic acid). Together, our results indicate a hitherto unknown mechanism for neuroprotection by a highly specific and unforeseen interaction between the CaMKIIα hub domain and small molecule brain-penetrant GHB analogs. This establishes GHB analogs as powerful tools for investigating CaMKII neuropharmacology in general and as potential therapeutic compounds for cerebral ischemia in particular.
Collapse
|
15
|
Zoidl GR, Spray DC. The Roles of Calmodulin and CaMKII in Cx36 Plasticity. Int J Mol Sci 2021; 22:4473. [PMID: 33922931 PMCID: PMC8123330 DOI: 10.3390/ijms22094473] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/17/2021] [Accepted: 04/20/2021] [Indexed: 01/07/2023] Open
Abstract
Anatomical and electrophysiological evidence that gap junctions and electrical coupling occur between neurons was initially confined to invertebrates and nonmammals and was thought to be a primitive form of synaptic transmission. More recent studies revealed that electrical communication is common in the mammalian central nervous system (CNS), often coexisting with chemical synaptic transmission. The subsequent progress indicated that electrical synapses formed by the gap junction protein connexin-36 (Cx36) and its paralogs in nonmammals constitute vital elements in mammalian and fish synaptic circuitry. They govern the collective activity of ensembles of coupled neurons, and Cx36 gap junctions endow them with enormous adaptive plasticity, like that seen at chemical synapses. Moreover, they orchestrate the synchronized neuronal network activity and rhythmic oscillations that underlie the fundamental integrative processes, such as memory and learning. Here, we review the available mechanistic evidence and models that argue for the essential roles of calcium, calmodulin, and the Ca2+/calmodulin-dependent protein kinase II in integrating calcium signals to modulate the strength of electrical synapses through interactions with the gap junction protein Cx36.
Collapse
Affiliation(s)
- Georg R. Zoidl
- Department of Biology & Center for Vision Research (CVR), York University, Toronto, ON M3J 1P3, Canada
| | - David C. Spray
- Dominick P. Purpura Department of Neuroscience & Department of Medicine (Cardiology), Albert Einstein College of Medicine, New York, NY 10461, USA;
| |
Collapse
|
16
|
Wang L, Ginnan RG, Wang YX, Zheng YM. Interactive Roles of CaMKII/Ryanodine Receptor Signaling and Inflammation in Lung Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:305-317. [PMID: 33788199 DOI: 10.1007/978-3-030-63046-1_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a multifunctional protein kinase and has been recently recognized to play a vital role in pathological events in the pulmonary system. CaMKII has diverse downstream targets that promote vascular disease, asthma, and cancer, so improved understanding of CaMKII signaling has the potential to lead to new therapies for lung diseases. Multiple studies have demonstrated that CaMKII is involved in redox modulation of ryanodine receptors (RyRs). CaMKII can be directly activated by reactive oxygen species (ROS) which then regulates RyR activity, which is essential for Ca2+-dependent processes in lung diseases. Furthermore, both CaMKII and RyRs participate in the inflammation process. However, their role in the pulmonary physiology in response to ROS is still an ambiguous one. Because CaMKII and RyRs are important in pulmonary biology, cell survival, cell cycle control, and inflammation, it is possible that the relationship between ROS and CaMKII/RyRs signal complex will be necessary for understanding and treating lung diseases. Here, we review roles of CaMKII/RyRs in lung diseases to understand with how CaMKII/RyRs may act as a transduction signal to connect prooxidant conditions into specific downstream pathological effects that are relevant to rare and common forms of pulmonary disease.
Collapse
Affiliation(s)
- Lan Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.,Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Roman G Ginnan
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| | - Yun-Min Zheng
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
17
|
Sloutsky R, Dziedzic N, Dunn MJ, Bates RM, Torres-Ocampo AP, Boopathy S, Page B, Weeks JG, Chao LH, Stratton MM. Heterogeneity in human hippocampal CaMKII transcripts reveals allosteric hub-dependent regulation. Sci Signal 2020; 13:eaaz0240. [PMID: 32694170 PMCID: PMC7654443 DOI: 10.1126/scisignal.aaz0240] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Calcium/calmodulin-dependent protein kinase II (CaMKII) plays a central role in Ca2+ signaling throughout the body. In the hippocampus, CaMKII is required for learning and memory. Vertebrate genomes encode four CaMKII homologs: CaMKIIα, CaMKIIβ, CaMKIIγ, and CaMKIIδ. All CaMKIIs consist of a kinase domain, a regulatory segment, a variable linker region, and a hub domain, which is responsible for oligomerization. The four proteins differ primarily in linker length and composition because of extensive alternative splicing. Here, we report the heterogeneity of CaMKII transcripts in three complex samples of human hippocampus using deep sequencing. We showed that hippocampal cells contain a diverse collection of over 70 CaMKII transcripts from all four CaMKII-encoding genes. We characterized the Ca2+/CaM sensitivity of hippocampal CaMKII variants spanning a broad range of linker lengths and compositions. The effect of the variable linker on Ca2+/CaM sensitivity depended on the kinase and hub domains. Moreover, we revealed a previously uncharacterized role for the hub domain as an allosteric regulator of kinase activity, which may provide a pharmacological target for modulating CaMKII activity. Using small-angle x-ray scattering and single-particle cryo-electron microscopy (cryo-EM), we present evidence for extensive interactions between the kinase and the hub domains, even in the presence of a 30-residue linker. Together, these data suggest that Ca2+/CaM sensitivity in CaMKII is homolog dependent and includes substantial contributions from the hub domain. Our sequencing approach, combined with biochemistry, provides insights into understanding the complex pool of endogenous CaMKII splice variants.
Collapse
Affiliation(s)
- Roman Sloutsky
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, USA
| | - Noelle Dziedzic
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
| | - Matthew J Dunn
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, USA
| | - Rachel M Bates
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, USA
| | - Ana P Torres-Ocampo
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
| | - Sivakumar Boopathy
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Brendan Page
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, USA
| | - John G Weeks
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, USA
| | - Luke H Chao
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA.
- Department of Genetics Harvard Medical School, Boston, MA 02115, USA
| | - Margaret M Stratton
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, USA.
| |
Collapse
|
18
|
Bhattacharyya M, Karandur D, Kuriyan J. Structural Insights into the Regulation of Ca 2+/Calmodulin-Dependent Protein Kinase II (CaMKII). Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035147. [PMID: 31653643 DOI: 10.1101/cshperspect.a035147] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a highly conserved serine/threonine kinase that is ubiquitously expressed throughout the human body. Specialized isoforms of CaMKII play key roles in neuronal and cardiac signaling. The distinctive holoenzyme architecture of CaMKII, with 12-14 kinase domains attached by flexible linkers to a central hub, poses formidable challenges for structural characterization. Nevertheless, progress in determining the structural mechanisms underlying CaMKII functions has come from studying the kinase domain and the hub separately, as well as from a recent electron microscopic investigation of the intact holoenzyme. In this review, we discuss our current understanding of the structure of CaMKII. We also discuss the intriguing finding that the CaMKII holoenzyme can undergo activation-triggered subunit exchange, a process that has implications for the potentiation and perpetuation of CaMKII activity.
Collapse
Affiliation(s)
- Moitrayee Bhattacharyya
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720.,California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, California 94720.,Howard Hughes Medical Institute, University of California, Berkeley, California 94720
| | - Deepti Karandur
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720.,California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, California 94720.,Howard Hughes Medical Institute, University of California, Berkeley, California 94720
| | - John Kuriyan
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720.,California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, California 94720.,Howard Hughes Medical Institute, University of California, Berkeley, California 94720.,Department of Chemistry, University of California, Berkeley, California 94720.,Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| |
Collapse
|
19
|
Torres‐Ocampo AP, Özden C, Hommer A, Gardella A, Lapinskas E, Samkutty A, Esposito E, Garman SC, Stratton MM. Characterization of CaMKIIα holoenzyme stability. Protein Sci 2020; 29:1524-1534. [PMID: 32282091 PMCID: PMC7255518 DOI: 10.1002/pro.3869] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 12/29/2022]
Abstract
Ca2+ /calmodulin-dependent protein kinase II (CaMKII) is a Ser/Thr kinase necessary for long-term memory formation and other Ca2+ -dependent signaling cascades such as fertilization. Here, we investigated the stability of CaMKIIα using a combination of differential scanning calorimetry (DSC), X-ray crystallography, and mass photometry (MP). The kinase domain has a low thermal stability (apparent Tm = 36°C), which is slightly stabilized by ATP/MgCl2 binding (apparent Tm = 40°C) and significantly stabilized by regulatory segment binding (apparent Tm = 60°C). We crystallized the kinase domain of CaMKII bound to p-coumaric acid in the active site. This structure reveals solvent-exposed hydrophobic residues in the substrate-binding pocket, which are normally buried in the autoinhibited structure when the regulatory segment is present. This likely accounts for the large stabilization that we observe in DSC measurements comparing the kinase alone with the kinase plus regulatory segment. The hub domain alone is extremely stable (apparent Tm ~ 90°C), and the holoenzyme structure has multiple unfolding transitions ranging from ~60°C to 100°C. Using MP, we compared a CaMKIIα holoenzyme with different variable linker regions and determined that the dissociation of both these holoenzymes occurs at a higher concentration (is less stable) compared with the hub domain alone. We conclude that within the context of the holoenzyme structure, the kinase domain is stabilized, whereas the hub domain is destabilized. These data support a model where domains within the holoenzyme interact.
Collapse
Affiliation(s)
- Ana P. Torres‐Ocampo
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
- Molecular and Cellular Biology Graduate ProgramUniversity of MassachusettsAmherstMassachusettsUSA
| | - Can Özden
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
- Molecular and Cellular Biology Graduate ProgramUniversity of MassachusettsAmherstMassachusettsUSA
| | - Alexandra Hommer
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
| | - Anne Gardella
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
| | - Emily Lapinskas
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
| | - Alfred Samkutty
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
| | | | - Scott C Garman
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
| | - Margaret M Stratton
- Department of Biochemistry and Molecular BiologyUniversity of MassachusettsAmherstMassachusettsUSA
- Molecular and Cellular Biology Graduate ProgramUniversity of MassachusettsAmherstMassachusettsUSA
| |
Collapse
|
20
|
Röhm S, Krämer A, Knapp S. Function, Structure and Topology of Protein Kinases. PROTEINKINASE INHIBITORS 2020. [DOI: 10.1007/7355_2020_97] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
21
|
Bayer KU, Schulman H. CaM Kinase: Still Inspiring at 40. Neuron 2019; 103:380-394. [PMID: 31394063 DOI: 10.1016/j.neuron.2019.05.033] [Citation(s) in RCA: 200] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 02/12/2019] [Accepted: 05/21/2019] [Indexed: 01/07/2023]
Abstract
The Ca2+/calmodulin (CaM)-dependent protein kinase II (CaMKII) was touted as a memory molecule, even before its involvement in long-term potentiation (LTP) was shown. The enzyme has not disappointed, with subsequent demonstrations of remarkable structural and regulatory properties. Its neuronal functions now extend to long-term depression (LTD), and last year saw the first direct evidence for memory storage by CaMKII. Although CaMKII may have taken the spotlight, it is a member of a large family of diverse and interesting CaM kinases. Our aim is to place CaMKII in context of the other CaM kinases and then review certain aspects of this kinase that are of current interest.
Collapse
Affiliation(s)
- K Ulrich Bayer
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | | |
Collapse
|
22
|
Ardestani G, West MC, Maresca TJ, Fissore RA, Stratton MM. FRET-based sensor for CaMKII activity (FRESCA): A useful tool for assessing CaMKII activity in response to Ca 2+ oscillations in live cells. J Biol Chem 2019; 294:11876-11891. [PMID: 31201271 DOI: 10.1074/jbc.ra119.009235] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/11/2019] [Indexed: 01/22/2023] Open
Abstract
Ca2+ oscillations and consequent Ca2+/calmodulin-dependent protein kinase II (CaMKII) activation are required for embryogenesis, as well as neuronal, immunological, and cardiac signaling. Fertilization directly results in Ca2+ oscillations, but the resultant pattern of CaMKII activity remains largely unclear. To address this gap, we first employed the one existing biosensor for CaMKII activation. This sensor, Camui, comprises CaMKIIα and therefore solely reports on the activation of this CaMKII variant. Additionally, to detect the activity of all endogenous CaMKII variants simultaneously, we constructed a substrate-based sensor for CaMKII activity, FRESCA (FRET-based sensor for CaMKII activity). To examine the differential responses of the Camui and FRESCA sensors, we used several approaches to stimulate Ca2+ release in mouse eggs, including addition of phospholipase Cζ cRNA, which mimics natural fertilization. We found that the Camui response is delayed or terminates earlier than the FRESCA response. FRESCA enables assessment of endogenous CaMKII activity in real-time by both fertilization and artificial reagents, such as Sr2+, which also leads to CaMKII activation. FRESCA's broad utility will be important for optimizing artificial CaMKII activation for clinical use to manage infertility. Moreover, FRESCA provides a new view on CaMKII activity, and its application in additional biological systems may reveal new signaling paradigms in eggs, as well as in neurons, cardiomyocytes, immune cells, and other CaMKII-expressing cells.
Collapse
Affiliation(s)
- Goli Ardestani
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts 01003.,Veterinary and Animal Sciences Graduate Program, University of Massachusetts, Amherst, Massachusetts 01003
| | - Megan C West
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, Massachusetts 01003
| | - Thomas J Maresca
- Department of Biology, University of Massachusetts, Amherst, Massachusetts 01003
| | - Rafael A Fissore
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts 01003
| | - Margaret M Stratton
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, Massachusetts 01003
| |
Collapse
|
23
|
Jensen EL, Clement R, Kosta A, Maberly SC, Gontero B. A new widespread subclass of carbonic anhydrase in marine phytoplankton. ISME JOURNAL 2019; 13:2094-2106. [PMID: 31024153 PMCID: PMC6776030 DOI: 10.1038/s41396-019-0426-8] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/28/2019] [Accepted: 03/30/2019] [Indexed: 11/09/2022]
Abstract
Most aquatic photoautotrophs depend on CO2-concentrating mechanisms (CCMs) to maintain productivity at ambient concentrations of CO2, and carbonic anhydrase (CA) plays a key role in these processes. Here we present different lines of evidence showing that the protein LCIP63, identified in the marine diatom Thalassiosira pseudonana, is a CA. However, sequence analysis showed that it has a low identity with any known CA and therefore belongs to a new subclass that we designate as iota-CA. Moreover, LCIP63 unusually prefers Mn2+ to Zn2+ as a cofactor, which is potentially of ecological relevance since Mn2+ is more abundant than Zn2+ in the ocean. LCIP63 is located in the chloroplast and only expressed at low concentrations of CO2. When overexpressed using biolistic transformation, the rate of photosynthesis at limiting concentrations of dissolved inorganic carbon increased, confirming its role in the CCM. LCIP63 homologs are present in the five other sequenced diatoms and in other algae, bacteria, and archaea. Thus LCIP63 is phylogenetically widespread but overlooked. Analysis of the Tara Oceans database confirmed this and showed that LCIP63 is widely distributed in marine environments and is therefore likely to play an important role in global biogeochemical carbon cycling.
Collapse
Affiliation(s)
- Erik L Jensen
- Aix Marseille Univ, CNRS, BIP, UMR 7281, IMM, FR3479, 31 Chemin J. Aiguier, 13402, Marseille Cedex 20, France
| | - Romain Clement
- Aix Marseille Univ, CNRS, BIP, UMR 7281, IMM, FR3479, 31 Chemin J. Aiguier, 13402, Marseille Cedex 20, France
| | - Artemis Kosta
- Microscopy Core Facility, Aix Marseille Univ, CNRS, IMM, FR3479, 31 Chemin J. Aiguier, 13402, Marseille Cedex 20, France
| | - Stephen C Maberly
- Lake Ecosystems Group, Centre for Ecology & Hydrology, Lancaster Environment Centre, Library Avenue, Bailrigg, Lancaster, LA1 4AP, UK
| | - Brigitte Gontero
- Aix Marseille Univ, CNRS, BIP, UMR 7281, IMM, FR3479, 31 Chemin J. Aiguier, 13402, Marseille Cedex 20, France.
| |
Collapse
|
24
|
McSpadden ED, Xia Z, Chi CC, Susa AC, Shah NH, Gee CL, Williams ER, Kuriyan J. Variation in assembly stoichiometry in non-metazoan homologs of the hub domain of Ca 2+ /calmodulin-dependent protein kinase II. Protein Sci 2019; 28:1071-1082. [PMID: 30942928 DOI: 10.1002/pro.3614] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/01/2019] [Indexed: 11/12/2022]
Abstract
The multi-subunit Ca2+ /calmodulin-dependent protein kinase II (CaMKII) holoenzyme plays a critical role in animal learning and memory. The kinase domain of CaMKII is connected by a flexible linker to a C-terminal hub domain that assembles into a 12- or 14-subunit scaffold that displays the kinase domains around it. Studies on CaMKII suggest that the stoichiometry and dynamic assembly/disassembly of hub oligomers may be important for CaMKII regulation. Although CaMKII is a metazoan protein, genes encoding predicted CaMKII-like hub domains, without associated kinase domains, are found in the genomes of some green plants and bacteria. We show that the hub domains encoded by three related green algae, Chlamydomonas reinhardtii, Volvox carteri f. nagarensis, and Gonium pectoral, assemble into 16-, 18-, and 20-subunit oligomers, as assayed by native protein mass spectrometry. These are the largest known CaMKII hub domain assemblies. A crystal structure of the hub domain from C. reinhardtii reveals an 18-subunit organization. We identified four intra-subunit hydrogen bonds in the core of the fold that are present in the Chlamydomonas hub domain, but not in metazoan hubs. When six point mutations designed to recapitulate these hydrogen bonds were introduced into the human CaMKII-α hub domain, the mutant protein formed assemblies with 14 and 16 subunits, instead of the normal 12- and 14-subunit assemblies. Our results show that the stoichiometric balance of CaMKII hub assemblies can be shifted readily by small changes in sequence.
Collapse
Affiliation(s)
- Ethan D McSpadden
- Department of Molecular and Cell Biology, University of California, Berkeley, California.,California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, California.,Howard Hughes Medical Institute, University of California, Berkeley, California
| | - Zijie Xia
- Department of Chemistry, University of California, Berkeley, California
| | - Chris C Chi
- Department of Molecular and Cell Biology, University of California, Berkeley, California.,California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, California.,Howard Hughes Medical Institute, University of California, Berkeley, California
| | - Anna C Susa
- Department of Chemistry, University of California, Berkeley, California
| | - Neel H Shah
- Department of Molecular and Cell Biology, University of California, Berkeley, California.,California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, California.,Howard Hughes Medical Institute, University of California, Berkeley, California
| | - Christine L Gee
- Department of Molecular and Cell Biology, University of California, Berkeley, California.,California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, California.,Howard Hughes Medical Institute, University of California, Berkeley, California
| | - Evan R Williams
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, California.,Department of Chemistry, University of California, Berkeley, California
| | - John Kuriyan
- Department of Molecular and Cell Biology, University of California, Berkeley, California.,California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, California.,Howard Hughes Medical Institute, University of California, Berkeley, California.,Department of Chemistry, University of California, Berkeley, California.,Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California
| |
Collapse
|
25
|
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
|
26
|
Deciphering CaMKII Multimerization Using Fluorescence Correlation Spectroscopy and Homo-FRET Analysis. Biophys J 2017; 112:1270-1281. [PMID: 28355553 DOI: 10.1016/j.bpj.2017.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 02/03/2017] [Accepted: 02/03/2017] [Indexed: 11/20/2022] Open
Abstract
While kinases are typically composed of one or two subunits, calcium-calmodulin (CaM)-dependent protein kinase II (CaMKII) is composed of 8-14 subunits arranged as pairs around a central core. It is not clear if the CaMKII holoenzyme functions as an assembly of independent subunits, as catalytic pairs, or as a single unit. One strategy to address this question is to genetically engineer monomeric and dimeric CaMKII and evaluate how their activity compares to the wild-type (WT) holoenzyme. Here a technique that combines fluorescence correlation spectroscopy and homo-FRET analysis was used to characterize assembly mutants of Venus-tagged CaMKIIα to identify a dimeric CaMKII. Spectroscopy was then used to compare how holoenzyme structure and function changes in response to activation with CaM in the dimeric mutant, WT-holoenzyme, and a monomeric CaMKII oligomerization-domain deletion mutant control. CaM triggered an increase in hydrodynamic volume in both WT and dimeric CaMKII without altering subunit stoichiometry or the net homo-FRET between Venus-tagged catalytic domains. Biochemical analysis revealed that the dimeric mutant also functioned like WT holoenzyme in terms of its kinase activity with an exogenous substrate, and for endogenous T286 autophosphorylation. We conclude that the fundamental functional units of CaMKII holoenzyme are paired catalytic-domains.
Collapse
|
27
|
The CaMKII holoenzyme structure in activation-competent conformations. Nat Commun 2017; 8:15742. [PMID: 28589927 PMCID: PMC5467236 DOI: 10.1038/ncomms15742] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 04/25/2017] [Indexed: 12/18/2022] Open
Abstract
The Ca2+/calmodulin-dependent protein kinase II (CaMKII) assembles into large 12-meric holoenzymes, which is thought to enable regulatory processes required for synaptic plasticity underlying learning, memory and cognition. Here we used single particle electron microscopy (EM) to determine a pseudoatomic model of the CaMKIIα holoenzyme in an extended and activation-competent conformation. The holoenzyme is organized by a rigid central hub complex, while positioning of the kinase domains is highly flexible, revealing dynamic holoenzymes ranging from 15–35 nm in diameter. While most kinase domains are ordered independently, ∼20% appear to form dimers and <3% are consistent with a compact conformation. An additional level of plasticity is revealed by a small fraction of bona-fide 14-mers (<4%) that may enable subunit exchange. Biochemical and cellular FRET studies confirm that the extended state of CaMKIIα resolved by EM is the predominant form of the holoenzyme, even under molecular crowding conditions. Ca2+/calmodulin-dependent protein kinase II (CaMKII) forms a 12 subunit holoenzyme central to synaptic plasticity. Here the authors report a 3D structure of the CaMKII holoenzyme in an activation-competent state obtained by single particle EM, and suggest a role for the intrinsically disordered linker domain in facilitating cooperative activation.
Collapse
|
28
|
Pellegrini E, Signor L, Singh S, Boeri Erba E, Cusack S. Structures of the inactive and active states of RIP2 kinase inform on the mechanism of activation. PLoS One 2017; 12:e0177161. [PMID: 28545134 PMCID: PMC5436651 DOI: 10.1371/journal.pone.0177161] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 04/24/2017] [Indexed: 12/14/2022] Open
Abstract
Innate immune receptors NOD1 and NOD2 are activated by bacterial peptidoglycans leading to recruitment of adaptor kinase RIP2, which, upon phosphorylation and ubiquitination, becomes a scaffold for downstream effectors. The kinase domain (RIP2K) is a pharmaceutical target for inflammatory diseases caused by aberrant NOD2-RIP2 signalling. Although structures of active RIP2K in complex with inhibitors have been reported, the mechanism of RIP2K activation remains to be elucidated. Here we analyse RIP2K activation by combining crystal structures of the active and inactive states with mass spectrometric characterization of their phosphorylation profiles. The active state has Helix αC inwardly displaced and the phosphorylated Activation Segment (AS) disordered, whilst in the inactive state Helix αC is outwardly displaced and packed against the helical, non-phosphorylated AS. Biophysical measurements show that the active state is a stable dimer whilst the inactive kinase is in a monomer-dimer equilibrium, consistent with the observed structural differences at the dimer interface. We conclude that RIP2 kinase auto-phosphorylation is intimately coupled to dimerization, similar to the case of BRAF. Our results will help drug design efforts targeting RIP2 as a potential treatment for NOD2-RIP2 related inflammatory diseases.
Collapse
Affiliation(s)
| | - Luca Signor
- University Grenoble Alpes, IBS, Grenoble, France
- CNRS, IBS, Grenoble, France
- CEA, IBS, Grenoble, France
| | - Saurabh Singh
- European Molecular Biology Laboratory, Grenoble, France
| | - Elisabetta Boeri Erba
- University Grenoble Alpes, IBS, Grenoble, France
- CNRS, IBS, Grenoble, France
- CEA, IBS, Grenoble, France
| | - Stephen Cusack
- European Molecular Biology Laboratory, Grenoble, France
- * E-mail:
| |
Collapse
|
29
|
Amara CS, Fabritius C, Houben A, Wolff LI, Hartmann C. CaMKII Signaling Stimulates Mef2c Activity In Vitro but Only Minimally Affects Murine Long Bone Development in vivo. Front Cell Dev Biol 2017; 5:20. [PMID: 28361052 PMCID: PMC5352711 DOI: 10.3389/fcell.2017.00020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 03/02/2017] [Indexed: 01/17/2023] Open
Abstract
The long bones of vertebrate limbs form by endochondral ossification, whereby mesenchymal cells differentiate into chondrogenic progenitors, which then differentiate into chondrocytes. Chondrocytes undergo further differentiation from proliferating to prehypertrophic, and finally to hypertrophic chondrocytes. Several signaling pathways and transcription factors regulate this process. Previously, we and others have shown in chicken that overexpression of an activated form of Calcium/calmodulin-dependent kinase II (CaMKII) results in ectopic chondrocyte maturation. Here, we show that this is not the case in the mouse. Although, in vitro Mef2c activity was upregulated by about 55-fold in response to expression of an activated form of CaMKII (DACaMKII), transgenic mice that expressed a dominant-active form of CaMKII under the control of the Col2a1 regulatory elements display only a very transient and mild phenotype. Here, only the onset of chondrocyte hypertrophy at E12.5 is accelerated. It is also this early step in chondrocyte differentiation that is temporarily delayed around E13.5 in transgenic mice expressing the peptide inhibitor CaM-KIIN from rat (rKIIN) under the control of the Col2a1 regulatory elements. Yet, ultimately DACaMKII, as well as rKIIN transgenic mice are born with completely normal skeletal elements with regard to their length and growth plate organization. Hence, our in vivo analysis suggests that CaMKII signaling plays a minor role in chondrocyte maturation in mice.
Collapse
Affiliation(s)
- Chandra S Amara
- Department Bone and Skeletal Research, Medical Faculty of the University of Münster (WWU), Institute of Experimental Musculoskeletal Medicine Münster, Germany
| | - Christine Fabritius
- Department Bone and Skeletal Research, Medical Faculty of the University of Münster (WWU), Institute of Experimental Musculoskeletal Medicine Münster, Germany
| | - Astrid Houben
- Department Bone and Skeletal Research, Medical Faculty of the University of Münster (WWU), Institute of Experimental Musculoskeletal Medicine Münster, Germany
| | - Lena I Wolff
- Department Bone and Skeletal Research, Medical Faculty of the University of Münster (WWU), Institute of Experimental Musculoskeletal Medicine Münster, Germany
| | - Christine Hartmann
- Department Bone and Skeletal Research, Medical Faculty of the University of Münster (WWU), Institute of Experimental Musculoskeletal Medicine Münster, Germany
| |
Collapse
|
30
|
Responses of the marine diatom Thalassiosira pseudonana to changes in CO 2 concentration: a proteomic approach. Sci Rep 2017; 7:42333. [PMID: 28181560 PMCID: PMC5299434 DOI: 10.1038/srep42333] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 01/10/2017] [Indexed: 12/18/2022] Open
Abstract
The concentration of CO2 in many aquatic systems is variable, often lower than the KM of the primary carboxylating enzyme Rubisco, and in order to photosynthesize efficiently, many algae operate a facultative CO2 concentrating mechanism (CCM). Here we measured the responses of a marine diatom, Thalassiosira pseudonana, to high and low concentrations of CO2 at the level of transcripts, proteins and enzyme activity. Low CO2 caused many metabolic pathways to be remodeled. Carbon acquisition enzymes, primarily carbonic anhydrase, stress, degradation and signaling proteins were more abundant while proteins associated with nitrogen metabolism, energy production and chaperones were less abundant. A protein with similarities to the Ca2+/ calmodulin dependent protein kinase II_association domain, having a chloroplast targeting sequence, was only present at low CO2. This protein might be a specific response to CO2 limitation since a previous study showed that other stresses caused its reduction. The protein sequence was found in other marine diatoms and may play an important role in their response to low CO2 concentration.
Collapse
|
31
|
Khan S, Conte I, Carter T, Bayer KU, Molloy JE. Multiple CaMKII Binding Modes to the Actin Cytoskeleton Revealed by Single-Molecule Imaging. Biophys J 2016; 111:395-408. [PMID: 27463141 PMCID: PMC4968397 DOI: 10.1016/j.bpj.2016.06.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 11/22/2022] Open
Abstract
Localization of the Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) to dendritic spine synapses is determined in part by the actin cytoskeleton. We determined binding of GFP-tagged CaMKII to tag-RFP-labeled actin cytoskeleton within live cells using total internal reflection fluorescence microscopy and single-molecule tracking. Stepwise photobleaching showed that CaMKII formed oligomeric complexes. Photoactivation experiments demonstrated that diffusion out of the evanescent field determined the track lifetimes. Latrunculin treatment triggered a coupled loss of actin stress fibers and the colocalized, long-lived CaMKII tracks. The CaMKIIα (α) isoform, which was previously thought to lack F-actin interactions, also showed binding, but this was threefold weaker than that observed for CaMKIIβ (β). The βE' splice variant bound more weakly than α, showing that binding by β depends critically on the interdomain linker. The mutations βT287D and αT286D, which mimic autophosphorylation states, also abolished F-actin binding. Autophosphorylation triggers autonomous CaMKII activity, but does not impair GluN2B binding, another important synaptic protein interaction of CaMKII. The CaMKII inhibitor tatCN21 or CaMKII mutations that inhibit GluN2B association by blocking binding of ATP (βK43R and αK42M) or Ca(2+)/calmodulin (βA303R) had no effect on the interaction with F-actin. These results provide the first rationale for the reduced synaptic spine localization of the αT286D mutant, indicating that transient F-actin binding contributes to the synaptic localization of the CaMKIIα isoform. The track lifetime distributions had a stretched exponential form consistent with a heterogeneously diffusing population. This heterogeneity suggests that CaMKII adopts different F-actin binding modes, which is most easily rationalized by multiple subunit contacts between the CaMKII dodecamer and the F-actin cytoskeleton that stabilize the initial weak (micromolar) monovalent interaction.
Collapse
Affiliation(s)
- Shahid Khan
- Molecular Biology Consortium, Lawrence Berkeley National Laboratory, Berkeley, California.
| | - Ianina Conte
- Cardiovascular and Cell Science Research Institute, St. George's University of London, London, UK
| | - Tom Carter
- Cell Biology and Genetics, St. George's University of London, London, UK
| | - K Ulrich Bayer
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado
| | - Justin E Molloy
- The Francis Crick Institute, Mill Hill Laboratory, London, UK
| |
Collapse
|
32
|
Differentiation of Human Adipose Derived Stem Cells into Smooth Muscle Cells Is Modulated by CaMKIIγ. Stem Cells Int 2016; 2016:1267480. [PMID: 27493668 PMCID: PMC4963582 DOI: 10.1155/2016/1267480] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 06/02/2016] [Accepted: 06/07/2016] [Indexed: 12/13/2022] Open
Abstract
The multifunctional Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) is known to participate in maintenance and switches of smooth muscle cell (SMC) phenotypes. However, which isoform of CaMKII is involved in differentiation of adult mesenchymal stem cells into contractile SMCs remains unclear. In the present study, we detected γ isoform of CaMKII in differentiation of human adipose derived stem cells (hASCs) into SMCs that resulted from treatment with TGF-β1 and BMP4 in combination for 7 days. The results showed that CaMKIIγ increased gradually during differentiation of hASCs as determined by real-time PCR and western blot analysis. The siRNA-mediated knockdown of CaMKIIγ decreased the protein levels and transcriptional levels of smooth muscle contractile markers (a-SMA, SM22a, calponin, and SM-MHC), while CaMKIIγ overexpression increases the transcriptional and protein levels of smooth muscle contractile markers. These results suggested that γ isoform of CaMKII plays a significant role in smooth muscle differentiation of hASCs.
Collapse
|
33
|
MicroRNA-30 inhibits neointimal hyperplasia by targeting Ca(2+)/calmodulin-dependent protein kinase IIδ (CaMKIIδ). Sci Rep 2016; 6:26166. [PMID: 27199283 PMCID: PMC4873751 DOI: 10.1038/srep26166] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 04/27/2016] [Indexed: 01/13/2023] Open
Abstract
The multifunctional Ca2+/calmodulin-dependent protein kinase II δ-isoform (CaMKIIδ) promotes vascular smooth muscle (VSM) proliferation, migration, and injury-induced vascular wall neointima formation. The objective of this study was to test if microRNA-30 (miR-30) family members are endogenous regulators of CaMKIIδ expression following vascular injury and whether ectopic expression of miR-30 can inhibit CaMKIIδ-dependent VSM cell function and neointimal VSM hyperplasia induced by vascular injury. The CaMKIIδ 3′UTR contains a consensus miR-30 binding sequence that is highly conserved across species. A significant decrease in miR-30 family members and increase in CaMKIIδ2 protein expression, with no change in CaMKIIδ mRNA expression, was observed in medial layers of VSM 7 days post-injury. In vitro, overexpression of miR-30c or miR-30e inhibited CaMKIIδ2 protein expression by ~50% in cultured rat aortic VSM cells, and inhibited VSM cell proliferation and migration. In vivo, lenti-viral delivery of miR-30c into injured rat carotid arteries prevented the injury-induced increase in CaMKIIδ2. Furthermore, neointima formation was dramatically inhibited by lenti-viral delivery of miR-30c in the injured medial smooth muscle. These studies define a novel mechanism for regulating CaMKIIδ expression in VSM and provide a new potential therapeutic strategy to reduce progression of vascular proliferative diseases, including atherosclerosis and restenosis.
Collapse
|
34
|
Bhattacharyya M, Stratton MM, Going CC, McSpadden ED, Huang Y, Susa AC, Elleman A, Cao YM, Pappireddi N, Burkhardt P, Gee CL, Barros T, Schulman H, Williams ER, Kuriyan J. Molecular mechanism of activation-triggered subunit exchange in Ca(2+)/calmodulin-dependent protein kinase II. eLife 2016; 5. [PMID: 26949248 PMCID: PMC4859805 DOI: 10.7554/elife.13405] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/03/2016] [Indexed: 12/04/2022] Open
Abstract
Activation triggers the exchange of subunits in Ca2+/calmodulin-dependent protein kinase II (CaMKII), an oligomeric enzyme that is critical for learning, memory, and cardiac function. The mechanism by which subunit exchange occurs remains elusive. We show that the human CaMKII holoenzyme exists in dodecameric and tetradecameric forms, and that the calmodulin (CaM)-binding element of CaMKII can bind to the hub of the holoenzyme and destabilize it to release dimers. The structures of CaMKII from two distantly diverged organisms suggest that the CaM-binding element of activated CaMKII acts as a wedge by docking at intersubunit interfaces in the hub. This converts the hub into a spiral form that can release or gain CaMKII dimers. Our data reveal a three-way competition for the CaM-binding element, whereby phosphorylation biases it towards the hub interface, away from the kinase domain and calmodulin, thus unlocking the ability of activated CaMKII holoenzymes to exchange dimers with unactivated ones. DOI:http://dx.doi.org/10.7554/eLife.13405.001 How does memory outlast the lifetime of the molecules that encode it? One enzyme that is found in neurons and has been suggested to help long-term memories to form is called CaMKII. Each CaMKII assembly is typically composed of 12 to 14 protein subunits associated in a ring and can exist in either an “unactivated” or “activated” state. In 2014, researchers showed that CaMKII assemblies can exchange subunits with each other. Importantly, an active CaMKII can mix with an unactivated CaMKII and share its activation state. CaMKII may use this mechanism to spread information to the next generation of proteins – thereby allowing activation to outlast the lifespan of the initially activated proteins. However the molecular mechanism that underlies this process was not clear. Now, Bhattacharyya et al. – including some of the researchers involved in the 2014 work – address two questions about this mechanism. How do subunits exchange between CaMKII assemblies? And how does the activation of CaMKII initiate subunit exchange? A closed-ring hub ties the subunits of CaMKII together, similar to the organization of the segments in an orange. To undergo subunit exchange, the hub must open up to release and accept subunits. Bhattacharyya et al. have now uncovered an intrinsic flexibility in the hub that is triggered by a short peptide segment in CaMKII. This segment, which is exposed in activated CaMKII but not in the unactivated form, can crack open the hub ring by binding between the hub subunits, like a finger separating the segments of an orange. This allows the hub to flex and expand, and once open, the hub’s flexibility allows room for subunits to be released or accepted. Although this subunit exchange mechanism could be a powerful means for spreading the activated state throughout signaling pathways, the biological relevance of this phenomenon has not been clarified. However, the mechanistic framework provided by Bhattacharyya et al. may allow new experiments to be performed that test the consequences of subunit exchange in live cells and organisms. It could also enable investigations into the importance of subunit exchange in long-term memory. DOI:http://dx.doi.org/10.7554/eLife.13405.002
Collapse
Affiliation(s)
- Moitrayee Bhattacharyya
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Margaret M Stratton
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Catherine C Going
- Department of Chemistry, University of California, Berkeley, Berkeley, United States
| | - Ethan D McSpadden
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Yongjian Huang
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States.,Biophysics Graduate Group, University of California, Berkeley, Berkeley, United States
| | - Anna C Susa
- Department of Chemistry, University of California, Berkeley, Berkeley, United States
| | - Anna Elleman
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Yumeng Melody Cao
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Nishant Pappireddi
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Pawel Burkhardt
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Christine L Gee
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Tiago Barros
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | | | - Evan R Williams
- Department of Chemistry, University of California, Berkeley, Berkeley, United States
| | - John Kuriyan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States.,Biophysics Graduate Group, University of California, Berkeley, Berkeley, United States.,Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, United States
| |
Collapse
|
35
|
Olinares PDB, Dunn AD, Padovan JC, Fernandez-Martinez J, Rout MP, Chait BT. A Robust Workflow for Native Mass Spectrometric Analysis of Affinity-Isolated Endogenous Protein Assemblies. Anal Chem 2016; 88:2799-807. [PMID: 26849307 PMCID: PMC4790104 DOI: 10.1021/acs.analchem.5b04477] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The central players in most cellular events are assemblies of macromolecules. Structural and functional characterization of these assemblies requires knowledge of their subunit stoichiometry and intersubunit connectivity. One of the most direct means for acquiring such information is so-called "native mass spectrometry (MS)", wherein the masses of the intact assemblies and parts thereof are accurately determined. It is of particular interest to apply native MS to the study of endogenous protein assemblies-i.e., those wherein the component proteins are expressed at endogenous levels in their natural functional states, rather than the overexpressed (sometimes partial) constructs commonly employed in classical structural studies, whose assembly can introduce stoichiometry artifacts and other unwanted effects. To date, the application of native MS to the elucidation of endogenous protein complexes has been limited by the difficulty in obtaining pristine cell-derived assemblies at sufficiently high concentrations for effective analysis. Here, to address this challenge, we present a robust workflow that couples rapid and efficient affinity isolation of endogenous protein complexes with a sensitive native MS readout. The resulting workflow has the potential to provide a wealth of data on the stoichiometry and intersubunit connectivity of endogenous protein assemblies-information that is key to successful integrative structural elucidation of biological systems.
Collapse
Affiliation(s)
- Paul Dominic B. Olinares
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, NY 10065 USA
| | - Amelia D. Dunn
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, NY 10065 USA
| | - Júlio C. Padovan
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, NY 10065 USA
| | | | - Michael P. Rout
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY 10065 USA
| | - Brian T. Chait
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, NY 10065 USA
| |
Collapse
|
36
|
Mollova MY, Katus HA, Backs J. Regulation of CaMKII signaling in cardiovascular disease. Front Pharmacol 2015; 6:178. [PMID: 26379551 PMCID: PMC4548452 DOI: 10.3389/fphar.2015.00178] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 08/10/2015] [Indexed: 01/08/2023] Open
Abstract
Heart failure (HF) is a major cause of death in the developed countries (Murray and Lopez, 1996; Koitabashi and Kass, 2012). Adverse cardiac remodeling that precedes heart muscle dysfunction is characterized by a myriad of molecular changes affecting the cardiomyocyte. Among these, alterations in protein kinase pathways play often an important mediator role since they link upstream pathologic stress signaling with downstream regulatory programs and thus affect both the structural and functional integrity of the heart muscle. In the context of cardiac disease, a profound understanding for the overriding mechanisms that regulate protein kinase activity (protein-protein interactions, post-translational modifications, or targeting via anchoring proteins) is crucial for the development of specific and effective pharmacological treatment strategies targeting the failing myocardium. In this review, we focus on several mechanisms of upstream regulation of Ca2+-calmodulin-dependent protein kinase II that play a relevant pathophysiological role in the development and progression of cardiovascular disease; precise targeting of these mechanisms might therefore represent novel and promising tools for prevention and treatment of HF.
Collapse
Affiliation(s)
- Mariya Y Mollova
- Research Unit Cardiac Epigenetics, Department of Cardiology, Angiology and Pneumology, University of Heidelberg , Heidelberg, Germany ; Department of Cardiology, Angiology and Pneumology, University of Heidelberg , Heidelberg, Germany ; Partner Site Heidelberg/Mannheim, German Center for Cardiovascular Research , Heidelberg, Germany
| | - Hugo A Katus
- Department of Cardiology, Angiology and Pneumology, University of Heidelberg , Heidelberg, Germany ; Partner Site Heidelberg/Mannheim, German Center for Cardiovascular Research , Heidelberg, Germany
| | - Johannes Backs
- Research Unit Cardiac Epigenetics, Department of Cardiology, Angiology and Pneumology, University of Heidelberg , Heidelberg, Germany ; Partner Site Heidelberg/Mannheim, German Center for Cardiovascular Research , Heidelberg, Germany
| |
Collapse
|
37
|
Hoover CM, Edwards SL, Yu SC, Kittelmann M, Richmond JE, Eimer S, Yorks RM, Miller KG. A novel CaM kinase II pathway controls the location of neuropeptide release from Caenorhabditis elegans motor neurons. Genetics 2014; 196:745-65. [PMID: 24653209 PMCID: PMC3948804 DOI: 10.1534/genetics.113.158568] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 11/17/2013] [Indexed: 12/29/2022] Open
Abstract
Neurons release neuropeptides via the regulated exocytosis of dense core vesicles (DCVs) to evoke or modulate behaviors. We found that Caenorhabditis elegans motor neurons send most of their DCVs to axons, leaving very few in the cell somas. How neurons maintain this skewed distribution and the extent to which it can be altered to control DCV numbers in axons or to drive release from somas for different behavioral impacts is unknown. Using a forward genetic screen, we identified loss-of-function mutations in UNC-43 (CaM kinase II) that reduce axonal DCV levels by ∼90% and cell soma/dendrite DCV levels by ∼80%, leaving small synaptic vesicles largely unaffected. Blocking regulated secretion in unc-43 mutants restored near wild-type axonal levels of DCVs. Time-lapse video microscopy showed no role for CaM kinase II in the transport of DCVs from cell somas to axons. In vivo secretion assays revealed that much of the missing neuropeptide in unc-43 mutants is secreted via a regulated secretory pathway requiring UNC-31 (CAPS) and UNC-18 (nSec1). DCV cargo levels in unc-43 mutants are similarly low in cell somas and the axon initial segment, indicating that the secretion occurs prior to axonal transport. Genetic pathway analysis suggests that abnormal neuropeptide function contributes to the sluggish basal locomotion rate of unc-43 mutants. These results reveal a novel pathway controlling the location of DCV exocytosis and describe a major new function for CaM kinase II.
Collapse
Affiliation(s)
- Christopher M. Hoover
- Genetic Models of Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| | - Stacey L. Edwards
- Genetic Models of Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| | - Szi-chieh Yu
- Department of Biological Sciences, University of Illinois, Chicago, Illinois 60607
| | - Maike Kittelmann
- European Neuroscience Institute, Center for Molecular Physiology of the Brain, Georg-August University, Goettingen, Germany 37073
| | - Janet E. Richmond
- Department of Biological Sciences, University of Illinois, Chicago, Illinois 60607
| | - Stefan Eimer
- European Neuroscience Institute, Center for Molecular Physiology of the Brain, Georg-August University, Goettingen, Germany 37073
- BIOSS Center for Biological Signaling Studies, Albert-Ludwigs-University, Freiburg, Germany 79085
| | - Rosalina M. Yorks
- Genetic Models of Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| | - Kenneth G. Miller
- Genetic Models of Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| |
Collapse
|
38
|
Stratton M, Lee IH, Bhattacharyya M, Christensen SM, Chao LH, Schulman H, Groves JT, Kuriyan J. Activation-triggered subunit exchange between CaMKII holoenzymes facilitates the spread of kinase activity. eLife 2014; 3:e01610. [PMID: 24473075 PMCID: PMC3901001 DOI: 10.7554/elife.01610] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The activation of the dodecameric Ca(2+)/calmodulin dependent kinase II (CaMKII) holoenzyme is critical for memory formation. We now report that CaMKII has a remarkable property, which is that activation of the holoenzyme triggers the exchange of subunits between holoenzymes, including unactivated ones, enabling the calcium-independent phosphorylation of new subunits. We show, using a single-molecule TIRF microscopy technique, that the exchange process is triggered by the activation of CaMKII, and that exchange is modulated by phosphorylation of two residues in the calmodulin-binding segment, Thr 305 and Thr 306. Based on these results, and on the analysis of molecular dynamics simulations, we suggest that the phosphorylated regulatory segment of CaMKII interacts with the central hub of the holoenzyme and weakens its integrity, thereby promoting exchange. Our results have implications for an earlier idea that subunit exchange in CaMKII may have relevance for information storage resulting from brief coincident stimuli during neuronal signaling. DOI: http://dx.doi.org/10.7554/eLife.01610.001.
Collapse
Affiliation(s)
- Margaret Stratton
- Department of Molecular and Cell Biology, Berkeley, Berkeley, United States
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Learning and memory require the formation of new neural networks in the brain. A key mechanism underlying this process is synaptic plasticity at excitatory synapses, which connect neurons into networks. Excitatory synaptic transmission happens when glutamate, the excitatory neurotransmitter, activates receptors on the postsynaptic neuron. Synaptic plasticity is a higher-level process in which the strength of excitatory synapses is altered in response to the pattern of activity at the synapse. It is initiated in the postsynaptic compartment, where the precise pattern of influx of calcium through activated glutamate receptors leads either to the addition of new receptors and enlargement of the synapse (long-term potentiation) or the removal of receptors and shrinkage of the synapse (long-term depression). Calcium/calmodulin-regulated enzymes and small GTPases collaborate to control this highly tuned mechanism.
Collapse
Affiliation(s)
- Mary B Kennedy
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| |
Collapse
|
40
|
Recombinant probes reveal dynamic localization of CaMKIIα within somata of cortical neurons. J Neurosci 2013; 33:14579-90. [PMID: 24005308 DOI: 10.1523/jneurosci.2108-13.2013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In response to NMDA receptor stimulation, CaMKIIα moves rapidly from a diffuse distribution within the shafts of neuronal dendrites to a clustered postsynaptic distribution. However, less is known about CaMKIIα localization and trafficking within neuronal somata. Here we use a novel recombinant probe capable of labeling endogenous CaMKIIα in living rat neurons to examine its localization and trafficking within the somata of cortical neurons. This probe, which was generated using an mRNA display selection, binds to endogenous CaMKIIα at high affinity and specificity following expression in rat cortical neurons in culture. In ∼45% of quiescent cortical neurons, labeled clusters of CaMKIIα 1-4 μm in diameter were present. Upon exposure to glutamate and glycine, CaMKIIα clusters disappeared in a Ca(2+)-dependent manner within seconds. Moreover, minutes after the removal of glutamate and glycine, the clusters returned to their original configuration. The clusters, which also appear in cortical neurons in sections taken from mouse brains, contain actin and disperse upon exposure to cytochalasin D, an actin depolymerizer. In conclusion, within the soma, CaMKII localizes and traffics in a manner that is distinct from its localization and trafficking within the dendrites.
Collapse
|
41
|
Nonspecific, reversible inhibition of voltage-gated calcium channels by CaMKII inhibitor CK59. Cell Mol Neurobiol 2013; 33:723-9. [PMID: 23657616 DOI: 10.1007/s10571-013-9941-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 04/24/2013] [Indexed: 10/26/2022]
Abstract
Investigation of kinase-related processes often uses pharmacological inhibition to reveal pathways in which kinases are involved. However, one concern about using such kinase inhibitors is their potential lack of specificity. Here, we report that the calcium-calmodulin-dependent kinase II (CaMKII) inhibitor CK59 inhibited multiple voltage-gated calcium channels, including the L-type channel during depolarization in a dose-dependent manner. The use of another CaMKII inhibitor, cell-permeable autocamtide-2 related inhibitory peptide II (Ant-AIP-II), failed to similarly decrease calcium current or entry in hippocampal cultures, as shown by ratiometric calcium imaging and whole-cell patch clamp electrophysiology. Notably, inhibition due to CK59 was reversible; washout of the drug brought calcium levels back to control values upon depolarization. Furthermore, the IC50 for CK59 was approximately 50 μM, which is only fivefold larger than the reported IC50 values for CaMKII inhibition. Similar nonspecific actions of other CaMKII inhibitors KN93 and KN62 have previously been reported. In the case of all three kinase inhibitors, the IC50 for calcium current inhibition falls near that of CaMKII inhibition. Our findings demonstrate that CK59 attenuates activity of voltage-gated calcium channels, and thus provide more evidence for caution when relying on pharmacological inhibition to examine kinase-dependent phenomena.
Collapse
|
42
|
Stratton MM, Chao LH, Schulman H, Kuriyan J. Structural studies on the regulation of Ca2+/calmodulin dependent protein kinase II. Curr Opin Struct Biol 2013; 23:292-301. [PMID: 23632248 DOI: 10.1016/j.sbi.2013.04.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 03/24/2013] [Accepted: 04/02/2013] [Indexed: 11/15/2022]
Abstract
Ca(2+)/calmodulin dependent protein kinase II (CaMKII) is a broadly distributed metazoan Ser/Thr protein kinase that is important in neuronal and cardiac signaling. CaMKII forms oligomeric assemblies, typically dodecameric, in which the calcium-responsive kinase domains are organized around a central hub. We review the results of crystallographic analyses of CaMKII, including the recently determined structure of a full-length and autoinhibited form of the holoenzyme. These structures, when combined with other data, allow informed speculation about how CaMKII escapes calcium-dependence when calcium spikes exceed threshold frequencies.
Collapse
Affiliation(s)
- Margaret M Stratton
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | | | | | | |
Collapse
|
43
|
Fera A, Dosemeci A, Sousa AA, Yang C, Leapman RD, Reese TS. Direct visualization of CaMKII at postsynaptic densities by electron microscopy tomography. J Comp Neurol 2013; 520:4218-25. [PMID: 22627922 DOI: 10.1002/cne.23151] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Ca(2+) /calmodulin-dependent protein kinase II (CaMKII) is a major component of postsynaptic densities (PSDs) involved in synaptic regulation. It has been previously shown that upon activity CaMKII from the spine reversibly aggregates at the cytoplasmic surfaces of PSDs, where it encounters various targets for phosphorylation. Targets for CaMKII are also present within the PSD, but there has been no reliable method to pinpoint whether, or where, CaMKII is located inside the PSD. Here we show that CaMKII can be mapped molecule-by-molecule within isolated PSDs using negative stain electron microscopy tomography. CaMKII molecules found in the core of the PSD may represent a pool distinct from the CaMKII residing at the cytoplasmic surface.
Collapse
Affiliation(s)
- Andrea Fera
- Laboratory of Neurobiology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Yasuda R. Studying signal transduction in single dendritic spines. Cold Spring Harb Perspect Biol 2012; 4:cshperspect.a005611. [PMID: 22843821 DOI: 10.1101/cshperspect.a005611] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Many forms of synaptic plasticity are triggered by biochemical signaling that occurs in small postsynaptic compartments called dendritic spines, each of which typically houses the postsynaptic terminal associated with a single glutamatergic synapse. Recent advances in optical techniques allow investigators to monitor biochemical signaling in single dendritic spines and thus reveal the signaling mechanisms that link synaptic activity and the induction of synaptic plasticity. This is mostly in the study of Ca2+-dependent forms of synaptic plasticity for which many of the steps between Ca2+ influx and changes to the synapse are now known. This article introduces the new techniques used to investigate signaling in single dendritic spines and the neurobiological insights that they have produced.
Collapse
Affiliation(s)
- Ryohei Yasuda
- Neurobiology Department, Howard Hughes Medical Institute, Duke University Medical Center, Durham, North Carolina 27710, USA.
| |
Collapse
|
45
|
Coultrap SJ, Bayer KU. CaMKII regulation in information processing and storage. Trends Neurosci 2012; 35:607-18. [PMID: 22717267 DOI: 10.1016/j.tins.2012.05.003] [Citation(s) in RCA: 246] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 05/07/2012] [Accepted: 05/11/2012] [Indexed: 11/29/2022]
Abstract
The Ca(2+)/Calmodulin(CaM)-dependent protein kinase II (CaMKII) is activated by Ca(2+)/CaM, but becomes partially autonomous (Ca(2+)-independent) upon autophosphorylation at T286. This hallmark feature of CaMKII regulation provides a form of molecular memory and is indeed important in long-term potentiation (LTP) of excitatory synapse strength and memory formation. However, emerging evidence supports a direct role in information processing, while storage of synaptic information may instead be mediated by regulated interaction of CaMKII with the NMDA receptor (NMDAR) complex. These and other CaMKII regulation mechanisms are discussed here in the context of the kinase structure and their impact on postsynaptic functions. Recent findings also implicate CaMKII in long-term depression (LTD), as well as functional roles at inhibitory synapses, lending renewed emphasis on better understanding the spatiotemporal control of CaMKII regulation.
Collapse
Affiliation(s)
- Steven J Coultrap
- Department of Pharmacology, University of Colorado Denver School of Medicine, Aurora, CO 80045, USA
| | | |
Collapse
|
46
|
Singer HA. Ca2+/calmodulin-dependent protein kinase II function in vascular remodelling. J Physiol 2011; 590:1349-56. [PMID: 22124148 DOI: 10.1113/jphysiol.2011.222232] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Vascular smooth muscle (VSM) undergoes a phenotypic switch in response to injury, a process that contributes to pathophysiological vascular wall remodelling. VSM phenotype switching is a consequence of changes in gene expression, including an array of ion channels and pumps affecting spatiotemporal features of intracellular Ca(2+) signals. Ca(2+) signalling promotes vascular wall remodelling by regulating cell proliferation, motility, and/or VSM gene transcription, although the mechanisms are not clear. In this review, the functions of multifunctional Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) in VSM phenotype switching and synthetic phenotype function are considered. CaMKII isozymes have complex structural and autoregulatory properties. Vascular injury in vivo results in rapid changes in CaMKII isoform expression with reduced expression of CaMKIIγ and upregulation of CaMKIIδ in medial wall VSM. SiRNA-mediated suppression of CaMKIIδ or gene deletion attenuates VSM proliferation and consequent neointimal formation. In vitro studies support functions for CaMKII in the regulation of cell proliferation, motility and gene expression via phosphorylation of CREB1 and HDACIIa/MEF2 complexes. These studies support the concept, and provide potential mechanisms, whereby Ca(2+) signalling through CaMKIIδ promotes VSM phenotype transitions and vascular remodelling.
Collapse
Affiliation(s)
- Harold A Singer
- Center for Cardiovascular Sciences, Albany Medical College (MC-8), 47 New Scotland Avenue, Albany, NY 12208, USA.
| |
Collapse
|
47
|
Abstract
Phenylketonuria is the most common, inherited aminoacidopathy associated with brain injury. To date, no study has focused on the neuropathology of the genetic mouse model of phenylketonuria, BTBR-Pah(enu2). We examined dendritic spines and synapses in the CA1 and prefrontal cortex among the wild-type, heterozygous, and BTBR-Pah(enu2) mice. A reduced density of dendritic spines, a shortened length of the presynaptic active zone, a widened synaptic cleft, and decreased thickness of postsynaptic density were revealed in BTBR-Pah(enu2) mice. Meanwhile, the phosphorylation at Thr286 of Ca(2+)/calmodulin-dependent protein kinase IIα was alerted in BTBR-Pah(enu2) mice. These findings revealed that phenylketonuria-related brain impairment is accompanied with abnormalities of dendritic spines and synapses. The dysfunction of Ca(2+)/calmodulin-dependent protein kinase IIα may result in an impaired synaptic function.
Collapse
|
48
|
A CaMKIIβ signaling pathway at the centrosome regulates dendrite patterning in the brain. Nat Neurosci 2011; 14:973-83. [PMID: 21725312 PMCID: PMC3391735 DOI: 10.1038/nn.2857] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 04/28/2011] [Indexed: 01/13/2023]
Abstract
The protein kinase calcium/calmodulin-dependent kinase II (CaMKII) predominantly consists of the α and β isoforms in the brain. Although CaMKIIα functions have been elucidated, the unique catalytic functions of CaMKIIβ have remained unknown. Using knockdown analyses in primary neurons and in the rat cerebellar cortex in vivo, we report that CaMKIIβ operates at the centrosome in a CaMKIIα-independent manner to drive dendrite retraction and pruning. We also find that the targeting protein PCM1 localizes CaMKIIβ at the centrosome. Finally, we uncover the E3 ubiquitin ligase Cdc20-APC as a novel centrosomal substrate of CaMKIIβ. CaMKIIβ phosphorylates Cdc20 at Ser51, which induces Cdc20 dispersion from the centrosome, thereby inhibiting centrosomal Cdc20-APC activity and triggering the transition from growth to retraction of dendrites. Our findings define a novel isoform-specific function for CaMKIIβ that regulates ubiquitin signaling at the centrosome and thereby orchestrates dendrite patterning, with important implications for neuronal connectivity in the brain.
Collapse
|
49
|
Abstract
Ischemic insults on neurons trigger excessive, pathological glutamate release that causes Ca²⁺ overload resulting in neuronal cell death (excitotoxicity). The Ca²⁺/calmodulin (CaM)-dependent protein kinase II (CaMKII) is a major mediator of physiological excitatory glutamate signals underlying neuronal plasticity and learning. Glutamate stimuli trigger autophosphorylation of CaMKII at T286, a process that makes the kinase "autonomous" (partially active independent from Ca²⁺ stimulation) and that is required for forms of synaptic plasticity. Recent studies suggested autonomous CaMKII activity also as potential drug target for post-insult neuroprotection, both after glutamate insults in neuronal cultures and after focal cerebral ischemia in vivo. However, CaMKII and other members of the CaM kinase family have been implicated in regulation of both neuronal death and survival. Here, we discuss past findings and possible mechanisms of CaM kinase functions in excitotoxicity and cerebral ischemia, with a focus on CaMKII and its regulation.
Collapse
|
50
|
Delarosa S, Guillemette J, Papillon J, Han YS, Kristof AS, Cybulsky AV. Activity of the Ste20-like kinase, SLK, is enhanced by homodimerization. Am J Physiol Renal Physiol 2011; 301:F554-64. [PMID: 21677149 DOI: 10.1152/ajprenal.00062.2011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The expression and activation of the Ste20-like kinase, SLK, is increased during renal development and recovery from ischemic acute renal failure. SLK promotes apoptosis, and during renal injury and repair, transcriptional induction or posttranscriptional control of SLK may, therefore, regulate cell survival. SLK contains protein interaction (coiled-coil) domains, suggesting that posttranslational homodimerization may also modulate SLK activity. We therefore expressed coiled-coil regions in the C-terminal domain of SLK as fusion proteins and demonstrated their homodimerization. By gel-filtration chromatography, endogenous and heterologously expressed SLK were detected in a macromolecular protein complex. To test the role of homodimerization in kinase activation, we constructed a fusion protein consisting of the SLK catalytic domain (amino acids 1-373) and a modified FK506 binding protein, Fv (Fv-SLK 1-373). Addition of AP20187 (an analog of FK506) enhanced the homodimerization of Fv-SLK 1-373. In an in vitro kinase assay, the dimeric Fv-SLK 1-373 displayed greater kinase activity than the monomeric form. In cells expressing Fv-SLK 1-373, homodimerization increased activation-specific phosphorylation of the proapoptotic kinases, c-Jun N-terminal kinase and p38 kinase. Compared with the monomer, dimeric Fv-SLK 1-373 enhanced the activation of a Bax promoter-luciferase reporter. Finally, expression of Fv-SLK 1-373 induced apoptosis, and the effect was increased by homodimerization. Thus the activity, downstream signaling, and functional effects of SLK are enhanced by dimerization of the kinase domain.
Collapse
Affiliation(s)
- Sierra Delarosa
- Department of Medicine, McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | |
Collapse
|