1
|
Prömer JJ, Wolske S, Castets P, van Woerden GM, Barresi C, O'Connor KC, Herbst R. MuSK is a substrate for CaMK2β but this interaction is dispensable for MuSK activation in vivo. Sci Rep 2025; 15:14865. [PMID: 40295530 PMCID: PMC12037915 DOI: 10.1038/s41598-025-95053-3] [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: 11/01/2024] [Accepted: 03/18/2025] [Indexed: 04/30/2025] Open
Abstract
The neuromuscular junction (NMJ) is the unique interface between lower motor neurons and skeletal muscle fibers and is indispensable for muscle function. Tight control of its localized formation at the center of every muscle fiber, and maintenance throughout lifetime are sustained by muscle-specific kinase (MuSK). MuSK acts as central regulator of acetylcholine receptor clustering at the postsynapse. Localized and temporally controlled signaling of MuSK is primarily achieved by tyrosine autophosphorylation and inhibition thereof. Previous investigations suggested serine phosphorylation of the activation domain as an additional modulator of MuSK activation. Here we identified calcium/calmodulin dependent protein kinase II (CaMK2) and in particular CaMK2β as novel catalyst of MuSK activation and confirmed its capability to phosphorylate MuSK in heterologous cells. However, whereas CaMK2β absence in muscle cells reduced AChR clustering, MuSK phosphorylation was unchanged. Accordingly, we ruled out MuSK phosphorylation as the cause of synapse fragmentation in a mouse model for myotonic dystrophy type 1, in which the muscle-specific splice-variant of CaMK2β is missing, or as the cause of ataxia or delayed muscle development in CaMK2β knockout animals. Histological characterization of muscles of CaMK2β knockout mice indicated specific roles of CaMK2β in fast glycolytic versus slow oxidative muscle. Taken together, our data shows that MuSK can be phosphorylated by CaMK2β, but loss of CaMK2β is likely compensated by other CaMK2 paralogs at the NMJ.
Collapse
Affiliation(s)
- Jakob J Prömer
- Institute for Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Kinderspitalgasse 15, Vienna, 1090, Austria
| | - Sara Wolske
- Institute for Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Kinderspitalgasse 15, Vienna, 1090, Austria
| | - Perrine Castets
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1 rue Michel Servet, Geneva, CH-1211, Switzerland
| | - Geeske M van Woerden
- Departments of Clinical Genetics, ENCORE center of expertise, Erasmus, Rotterdam, 3015 GD, MC, The Netherlands
- Neuroscience, ENCORE center of expertise, Erasmus, Rotterdam, 3015 GD, MC, The Netherlands
| | - Cinzia Barresi
- Institute for Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Kinderspitalgasse 15, Vienna, 1090, Austria
| | - Kevin C O'Connor
- Departments of Neurology, Yale School of Medicine, New Haven, Connecticut, 06511, USA
- Immunobiology, Yale School of Medicine, New Haven, Connecticut, 06511, USA
| | - Ruth Herbst
- Institute for Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Kinderspitalgasse 15, Vienna, 1090, Austria.
| |
Collapse
|
2
|
Zheng Q, Zhang H, Zhao H, Chen Y, Yang H, Li T, Cai Q, Chen Y, Wang Y, Zhang M, Zhang H. Ca 2+/calmodulin-dependent protein kinase II β decodes ER Ca 2+ transients to trigger autophagosome formation. Mol Cell 2025; 85:620-637.e6. [PMID: 39742665 DOI: 10.1016/j.molcel.2024.12.005] [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: 05/01/2024] [Revised: 09/16/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025]
Abstract
In multicellular organisms, very little is known about how Ca2+ transients on the ER outer surface elicited by autophagy stimuli are sustained and decoded to trigger autophagosome formation. Here, we show that Ca2+/calmodulin-dependent protein kinase II β (CaMKIIβ) integrates ER Ca2+ transients to trigger liquid-liquid phase separation (LLPS) of the autophagosome-initiating FIP200 complex. In response to ER Ca2+ transients, CaMKIIβ is recruited from actin filaments and forms condensates, which serve as sites for the emergence of or interaction with FIP200 puncta. CaMKIIβ phosphorylates FIP200 at Thr269, Thr1127, and Ser1484 to modulate LLPS and properties of the FIP200 complex, thereby controlling its function in autophagosome formation. CaMKIIβ also controls the amplitude, duration, and propagation of ER Ca2+ transients during autophagy induction. CaMKIIβ mutations identified in the neurodevelopmental disorder MRD54 affect the function of CaMKIIβ in autophagy. Our study reveals that CaMKIIβ is essential for sustaining and decoding ER Ca2+ transients to specify autophagosome formation in mammalian cells.
Collapse
Affiliation(s)
- Qiaoxia Zheng
- National Laboratory of Biomacromolecules, New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Huan Zhang
- National Laboratory of Biomacromolecules, New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Hongyu Zhao
- National Laboratory of Biomacromolecules, New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yong Chen
- National Laboratory of Biomacromolecules, New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hongzhining Yang
- Department of Medical Bioinformatics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Tingting Li
- Department of Medical Bioinformatics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Qixu Cai
- School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yingyu Chen
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Youjun Wang
- College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Mingjie Zhang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hong Zhang
- National Laboratory of Biomacromolecules, New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
3
|
Storey GP, Riquelme R, Barria A. Activity-Dependent Internalization of Glun2B-Containing NMDARs Is Required for Synaptic Incorporation of Glun2A and Synaptic Plasticity. J Neurosci 2025; 45:e0823242024. [PMID: 39562042 PMCID: PMC11756629 DOI: 10.1523/jneurosci.0823-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 11/06/2024] [Accepted: 11/09/2024] [Indexed: 11/21/2024] Open
Abstract
NMDA-type glutamate receptors are heterotetrameric complexes composed of two GluN1 and two GluN2 subunits. The precise composition of the GluN2 subunits determines the channel's biophysical properties and influences its interaction with postsynaptic scaffolding proteins and signaling molecules involved in synaptic physiology and plasticity. The precise regulation of NMDAR subunit composition at synapses is crucial for proper synaptogenesis, neuronal circuit development, and synaptic plasticity, a cellular model of memory formation. In the forebrain during early development, NMDARs contain solely the GluN2B subunit, which is necessary for proper synaptogenesis and synaptic plasticity. In rodents, GluN2A subunit expression begins in the second postnatal week, replacing GluN2B-containing NMDARs at synapses in an activity- or sensory experience-dependent process. This switch in NMDAR subunit composition at synapses alters channel properties and reduces synaptic plasticity. The molecular mechanism regulating the switch remains unclear. We have investigated the role of activity-dependent internalization of GluN2B-containing receptors in shaping synaptic NMDAR subunit composition. Using molecular, pharmacological, and electrophysiological approaches in cultured organotypic hippocampal slices from rats of both sexes, we show that the process of incorporating GluN2A-containing NMDAR receptors requires activity-dependent internalization of GluN2B-containing NMDARs. Interestingly, blockade of GluN2A synaptic incorporation was associated with impaired potentiation of AMPA-mediated synaptic transmission, suggesting a potential coupling between the trafficking of AMPARs into synapses and that of GluN2A-containing NMDARs. These insights contribute to our understanding of the molecular mechanisms underlying synaptic trafficking of glutamate receptors and synaptic plasticity. They may also have implications for therapeutic strategies targeting NMDAR function in neurological disorders.
Collapse
Affiliation(s)
- Granville P Storey
- Department of Neurobiology and Biophysics, University of Washington School of Medicine, Seattle, Washington 98195-7290
| | - Raul Riquelme
- Department of Neurobiology and Biophysics, University of Washington School of Medicine, Seattle, Washington 98195-7290
| | - Andres Barria
- Department of Neurobiology and Biophysics, University of Washington School of Medicine, Seattle, Washington 98195-7290
| |
Collapse
|
4
|
Strucinska K, Kneis P, Pennington T, Cizio K, Szybowska P, Morgan A, Weertman J, Lewis TL. Fis1 is required for the development of the dendritic mitochondrial network in pyramidal cortical neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631801. [PMID: 39829888 PMCID: PMC11741399 DOI: 10.1101/2025.01.07.631801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Mitochondrial ATP production and calcium buffering are critical for metabolic regulation and neurotransmission making the formation and maintenance of the mitochondrial network a critical component of neuronal health. Cortical pyramidal neurons contain compartment-specific mitochondrial morphologies that result from distinct axonal and dendritic mitochondrial fission and fusion profiles. We previously showed that axonal mitochondria are maintained at a small size as a result of high axonal mitochondrial fission factor (Mff) activity. However, loss of Mff activity had little effect on cortical dendritic mitochondria, raising the question of how fission/fusion balance is controlled in the dendrites. Thus, we sought to investigate the role of another fission factor, fission 1 (Fis1), on mitochondrial morphology, dynamics and function in cortical neurons. We knocked down Fis1 in cortical neurons both in primary culture and in vivo, and unexpectedly found that Fis1 depletion decreased mitochondrial length in the dendrites, without affecting mitochondrial size in the axon. Further, loss of Fis1 activity resulted in both increased mitochondrial motility and dynamics in the dendrites. These results argue Fis1 exhibits dendrite selectivity and plays a more complex role in neuronal mitochondrial dynamics than previously reported. Functionally, Fis1 loss resulted in reduced mitochondrial membrane potential, increased sensitivity to complex III blockade, and decreased mitochondrial calcium uptake during neuronal activity. The altered mitochondrial network culminated in elevated resting calcium levels that increased dendritic branching but reduced spine density. We conclude that Fis1 regulates morphological and functional mitochondrial characteristics that influence dendritic tree arborization and connectivity.
Collapse
Affiliation(s)
- Klaudia Strucinska
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - Parker Kneis
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
- Molecular Biology & Biochemistry Department, Oklahoma University Health Sciences Campus, Oklahoma City, OK 73104
| | - Travis Pennington
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
- Neuroscience Program, Oklahoma University Health Sciences Campus, Oklahoma City, OK 73104
| | - Katarzyna Cizio
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
- Neuroscience Program, Oklahoma University Health Sciences Campus, Oklahoma City, OK 73104
| | - Patrycja Szybowska
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - Abigail Morgan
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
- Neuroscience Program, Oklahoma University Health Sciences Campus, Oklahoma City, OK 73104
| | - Joshua Weertman
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - Tommy L Lewis
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
- Molecular Biology & Biochemistry Department, Oklahoma University Health Sciences Campus, Oklahoma City, OK 73104
- Neuroscience Program, Oklahoma University Health Sciences Campus, Oklahoma City, OK 73104
- Physiology Department, Oklahoma University Health Sciences Campus, Oklahoma City, OK 73104
| |
Collapse
|
5
|
Yong J, Song J. CaMKII activity and metabolic imbalance-related neurological diseases: Focus on vascular dysfunction, synaptic plasticity, amyloid beta accumulation, and lipid metabolism. Biomed Pharmacother 2024; 175:116688. [PMID: 38692060 DOI: 10.1016/j.biopha.2024.116688] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/03/2024] Open
Abstract
Metabolic syndrome (MetS) is characterized by insulin resistance, hyperglycemia, excessive fat accumulation and dyslipidemia, and is known to be accompanied by neuropathological symptoms such as memory loss, anxiety, and depression. As the number of MetS patients is rapidly increasing globally, studies on the mechanisms of metabolic imbalance-related neuropathology are emerging as an important issue. Ca2+/calmodulin-dependent kinase II (CaMKII) is the main Ca2+ sensor and contributes to diverse intracellular signaling in peripheral organs and the central nervous system (CNS). CaMKII exerts diverse functions in cells, related to mechanisms such as RNA splicing, reactive oxygen species (ROS) generation, cytoskeleton, and protein-protein interactions. In the CNS, CaMKII regulates vascular function, neuronal circuits, neurotransmission, synaptic plasticity, amyloid beta toxicity, lipid metabolism, and mitochondrial function. Here, we review recent evidence for the role of CaMKII in neuropathologic issues associated with metabolic disorders.
Collapse
Affiliation(s)
- Jeongsik Yong
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun, Jeollanam-do, Republic of Korea.
| |
Collapse
|
6
|
Chen Y, Liu S, Jacobi AA, Jeng G, Ulrich JD, Stein IS, Patriarchi T, Hell JW. Rapid sequential clustering of NMDARs, CaMKII, and AMPARs upon activation of NMDARs at developing synapses. Front Synaptic Neurosci 2024; 16:1291262. [PMID: 38660466 PMCID: PMC11039796 DOI: 10.3389/fnsyn.2024.1291262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
Rapid, synapse-specific neurotransmission requires the precise alignment of presynaptic neurotransmitter release and postsynaptic receptors. How postsynaptic glutamate receptor accumulation is induced during maturation is not well understood. We find that in cultures of dissociated hippocampal neurons at 11 days in vitro (DIV) numerous synaptic contacts already exhibit pronounced accumulations of the pre- and postsynaptic markers synaptotagmin, synaptophysin, synapsin, bassoon, VGluT1, PSD-95, and Shank. The presence of an initial set of AMPARs and NMDARs is indicated by miniature excitatory postsynaptic currents (mEPSCs). However, AMPAR and NMDAR immunostainings reveal rather smooth distributions throughout dendrites and synaptic enrichment is not obvious. We found that brief periods of Ca2+ influx through NMDARs induced a surprisingly rapid accumulation of NMDARs within 1 min, followed by accumulation of CaMKII and then AMPARs within 2-5 min. Postsynaptic clustering of NMDARs and AMPARs was paralleled by an increase in their mEPSC amplitudes. A peptide that blocked the interaction of NMDAR subunits with PSD-95 prevented the NMDAR clustering. NMDAR clustering persisted for 3 days indicating that brief periods of elevated glutamate fosters permanent accumulation of NMDARs at postsynaptic sites in maturing synapses. These data support the model that strong glutamatergic stimulation of immature glutamatergic synapses results in a fast and substantial increase in postsynaptic NMDAR content that required NMDAR binding to PSD-95 or its homologues and is followed by recruitment of CaMKII and subsequently AMPARs.
Collapse
Affiliation(s)
- Yucui Chen
- Department of Pharmacology, University of Iowa, Iowa City, IA, United States
| | - Shangming Liu
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Ariel A. Jacobi
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Grace Jeng
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Jason D. Ulrich
- Department of Pharmacology, University of Iowa, Iowa City, IA, United States
| | - Ivar S. Stein
- Department of Pharmacology, University of Iowa, Iowa City, IA, United States
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Tommaso Patriarchi
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Johannes W. Hell
- Department of Pharmacology, University of Iowa, Iowa City, IA, United States
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
7
|
Xia X, Shi C, Tsien C, Sun CB, Xie L, Luo Z, Bian M, Russano K, Thakur HS, Benowitz LI, Goldberg JL, Kapiloff MS. Ca 2+/Calmodulin-Dependent Protein Kinase II Enhances Retinal Ganglion Cell Survival But Suppresses Axon Regeneration after Optic Nerve Injury. eNeuro 2024; 11:ENEURO.0478-23.2024. [PMID: 38548335 PMCID: PMC10978821 DOI: 10.1523/eneuro.0478-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 04/01/2024] Open
Abstract
Neuroprotection after injury or in neurodegenerative disease remains a major goal for basic and translational neuroscience. Retinal ganglion cells (RGCs), the projection neurons of the eye, degenerate in optic neuropathies after axon injury, and there are no clinical therapies to prevent their loss or restore their connectivity to targets in the brain. Here we demonstrate a profound neuroprotective effect of the exogenous expression of various Ca2+/calmodulin-dependent protein kinase II (CaMKII) isoforms in mice. A dramatic increase in RGC survival following the optic nerve trauma was elicited by the expression of constitutively active variants of multiple CaMKII isoforms in RGCs using adeno-associated viral (AAV) vectors across a 100-fold range of AAV dosing in vivo. Despite this neuroprotection, however, short-distance RGC axon sprouting was suppressed by CaMKII, and long-distance axon regeneration elicited by several pro-axon growth treatments was likewise inhibited even as CaMKII further enhanced RGC survival. Notably, in a dose-escalation study, AAV-expressed CaMKII was more potent for axon growth suppression than the promotion of survival. That diffuse overexpression of constitutively active CaMKII strongly promotes RGC survival after axon injury may be clinically valuable for neuroprotection per se. However, the associated strong suppression of the optic nerve axon regeneration demonstrates the need for understanding the intracellular domain- and target-specific CaMKII activities to the development of CaMKII signaling pathway-directed strategies for the treatment of optic neuropathies.
Collapse
Affiliation(s)
- Xin Xia
- Department of Ophthalmology, Byers Eye Institute, Mary M. and Sash A. Spencer Center for Vision Research, Stanford University School of Medicine, Palo Alto, California 94034
| | - Caleb Shi
- Department of Neurosurgery, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115
- Department of Neurosurgery and Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115
| | - Christina Tsien
- Department of Ophthalmology, Byers Eye Institute, Mary M. and Sash A. Spencer Center for Vision Research, Stanford University School of Medicine, Palo Alto, California 94034
| | - Catalina B Sun
- Department of Ophthalmology, Byers Eye Institute, Mary M. and Sash A. Spencer Center for Vision Research, Stanford University School of Medicine, Palo Alto, California 94034
| | - Lili Xie
- Department of Neurosurgery, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115
- Department of Neurosurgery and Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115
| | - Ziming Luo
- Department of Ophthalmology, Byers Eye Institute, Mary M. and Sash A. Spencer Center for Vision Research, Stanford University School of Medicine, Palo Alto, California 94034
| | - Minjuan Bian
- Department of Ophthalmology, Byers Eye Institute, Mary M. and Sash A. Spencer Center for Vision Research, Stanford University School of Medicine, Palo Alto, California 94034
| | - Kristina Russano
- Department of Ophthalmology, Byers Eye Institute, Mary M. and Sash A. Spencer Center for Vision Research, Stanford University School of Medicine, Palo Alto, California 94034
| | - Hrishikesh Singh Thakur
- Department of Ophthalmology, Byers Eye Institute, Mary M. and Sash A. Spencer Center for Vision Research, Stanford University School of Medicine, Palo Alto, California 94034
| | - Larry I Benowitz
- Department of Neurosurgery, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115
- Department of Neurosurgery and Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115
| | - Jeffrey L Goldberg
- Department of Ophthalmology, Byers Eye Institute, Mary M. and Sash A. Spencer Center for Vision Research, Stanford University School of Medicine, Palo Alto, California 94034
| | - Michael S Kapiloff
- Department of Ophthalmology, Byers Eye Institute, Mary M. and Sash A. Spencer Center for Vision Research, Stanford University School of Medicine, Palo Alto, California 94034
| |
Collapse
|
8
|
Lisek M, Tomczak J, Boczek T, Zylinska L. Calcium-Associated Proteins in Neuroregeneration. Biomolecules 2024; 14:183. [PMID: 38397420 PMCID: PMC10887043 DOI: 10.3390/biom14020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/27/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
The dysregulation of intracellular calcium levels is a critical factor in neurodegeneration, leading to the aberrant activation of calcium-dependent processes and, ultimately, cell death. Ca2+ signals vary in magnitude, duration, and the type of neuron affected. A moderate Ca2+ concentration can initiate certain cellular repair pathways and promote neuroregeneration. While the peripheral nervous system exhibits an intrinsic regenerative capability, the central nervous system has limited self-repair potential. There is evidence that significant variations exist in evoked calcium responses and axonal regeneration among neurons, and individual differences in regenerative capacity are apparent even within the same type of neurons. Furthermore, some studies have shown that neuronal activity could serve as a potent regulator of this process. The spatio-temporal patterns of calcium dynamics are intricately controlled by a variety of proteins, including channels, ion pumps, enzymes, and various calcium-binding proteins, each of which can exert either positive or negative effects on neural repair, depending on the cellular context. In this concise review, we focus on several calcium-associated proteins such as CaM kinase II, GAP-43, oncomodulin, caldendrin, calneuron, and NCS-1 in order to elaborate on their roles in the intrinsic mechanisms governing neuronal regeneration following traumatic damage processes.
Collapse
Affiliation(s)
| | | | | | - Ludmila Zylinska
- Department of Molecular Neurochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (M.L.); (J.T.); (T.B.)
| |
Collapse
|
9
|
Hamad MIK, Emerald BS, Kumar KK, Ibrahim MF, Ali BR, Bataineh MF. Extracellular molecular signals shaping dendrite architecture during brain development. Front Cell Dev Biol 2023; 11:1254589. [PMID: 38155836 PMCID: PMC10754048 DOI: 10.3389/fcell.2023.1254589] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/28/2023] [Indexed: 12/30/2023] Open
Abstract
Proper growth and branching of dendrites are crucial for adequate central nervous system (CNS) functioning. The neuronal dendritic geometry determines the mode and quality of information processing. Any defects in dendrite development will disrupt neuronal circuit formation, affecting brain function. Besides cell-intrinsic programmes, extrinsic factors regulate various aspects of dendritic development. Among these extrinsic factors are extracellular molecular signals which can shape the dendrite architecture during early development. This review will focus on extrinsic factors regulating dendritic growth during early neuronal development, including neurotransmitters, neurotrophins, extracellular matrix proteins, contact-mediated ligands, and secreted and diffusible cues. How these extracellular molecular signals contribute to dendritic growth has been investigated in developing nervous systems using different species, different areas within the CNS, and different neuronal types. The response of the dendritic tree to these extracellular molecular signals can result in growth-promoting or growth-limiting effects, and it depends on the receptor subtype, receptor quantity, receptor efficiency, the animal model used, the developmental time windows, and finally, the targeted signal cascade. This article reviews our current understanding of the role of various extracellular signals in the establishment of the architecture of the dendrites.
Collapse
Affiliation(s)
- Mohammad I. K. Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Kukkala K. Kumar
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Marwa F. Ibrahim
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Bassam R. Ali
- Department of Genetics and Genomics, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mo’ath F. Bataineh
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
10
|
Nicoll RA, Schulman H. Synaptic memory and CaMKII. Physiol Rev 2023; 103:2877-2925. [PMID: 37290118 PMCID: PMC10642921 DOI: 10.1152/physrev.00034.2022] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/26/2023] [Accepted: 04/30/2023] [Indexed: 06/10/2023] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) and long-term potentiation (LTP) were discovered within a decade of each other and have been inextricably intertwined ever since. However, like many marriages, it has had its up and downs. Based on the unique biochemical properties of CaMKII, it was proposed as a memory molecule before any physiological linkage was made to LTP. However, as reviewed here, the convincing linkage of CaMKII to synaptic physiology and behavior took many decades. New technologies were critical in this journey, including in vitro brain slices, mouse genetics, single-cell molecular genetics, pharmacological reagents, protein structure, and two-photon microscopy, as were new investigators attracted by the exciting challenge. This review tracks this journey and assesses the state of this marriage 40 years on. The collective literature impels us to propose a relatively simple model for synaptic memory involving the following steps that drive the process: 1) Ca2+ entry through N-methyl-d-aspartate (NMDA) receptors activates CaMKII. 2) CaMKII undergoes autophosphorylation resulting in constitutive, Ca2+-independent activity and exposure of a binding site for the NMDA receptor subunit GluN2B. 3) Active CaMKII translocates to the postsynaptic density (PSD) and binds to the cytoplasmic C-tail of GluN2B. 4) The CaMKII-GluN2B complex initiates a structural rearrangement of the PSD that may involve liquid-liquid phase separation. 5) This rearrangement involves the PSD-95 scaffolding protein, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs), and their transmembrane AMPAR-regulatory protein (TARP) auxiliary subunits, resulting in an accumulation of AMPARs in the PSD that underlies synaptic potentiation. 6) The stability of the modified PSD is maintained by the stability of the CaMKII-GluN2B complex. 7) By a process of subunit exchange or interholoenzyme phosphorylation CaMKII maintains synaptic potentiation in the face of CaMKII protein turnover. There are many other important proteins that participate in enlargement of the synaptic spine or modulation of the steps that drive and maintain the potentiation. In this review we critically discuss the data underlying each of the steps. As will become clear, some of these steps are more firmly grounded than others, and we provide suggestions as to how the evidence supporting these steps can be strengthened or, based on the new data, be replaced. Although the journey has been a long one, the prospect of having a detailed cellular and molecular understanding of learning and memory is at hand.
Collapse
Affiliation(s)
- Roger A Nicoll
- Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, California, United States
| | - Howard Schulman
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California, United States
- Panorama Research Institute, Sunnyvale, California, United States
| |
Collapse
|
11
|
Tullis JE, Larsen ME, Rumian NL, Freund RK, Boxer EE, Brown CN, Coultrap SJ, Schulman H, Aoto J, Dell'Acqua ML, Bayer KU. LTP induction by structural rather than enzymatic functions of CaMKII. Nature 2023; 621:146-153. [PMID: 37648853 PMCID: PMC10482691 DOI: 10.1038/s41586-023-06465-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 07/20/2023] [Indexed: 09/01/2023]
Abstract
Learning and memory are thought to require hippocampal long-term potentiation (LTP), and one of the few central dogmas of molecular neuroscience that has stood undisputed for more than three decades is that LTP induction requires enzymatic activity of the Ca2+/calmodulin-dependent protein kinase II (CaMKII)1-3. However, as we delineate here, the experimental evidence is surprisingly far from conclusive. All previous interventions inhibiting enzymatic CaMKII activity and LTP4-8 also interfere with structural CaMKII roles, in particular binding to the NMDA-type glutamate receptor subunit GluN2B9-14. Thus, we here characterized and utilized complementary sets of new opto-/pharmaco-genetic tools to distinguish between enzymatic and structural CaMKII functions. Several independent lines of evidence demonstrated LTP induction by a structural function of CaMKII rather than by its enzymatic activity. The sole contribution of kinase activity was autoregulation of this structural role via T286 autophosphorylation, which explains why this distinction has been elusive for decades. Directly initiating the structural function in a manner that circumvented this T286 role was sufficient to elicit robust LTP, even when enzymatic CaMKII activity was blocked.
Collapse
Affiliation(s)
- Jonathan E Tullis
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew E Larsen
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Program in Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Nicole L Rumian
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Program in Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ronald K Freund
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Emma E Boxer
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Program in Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Carolyn Nicole Brown
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Steven J Coultrap
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Howard Schulman
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jason Aoto
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Program in Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Program in Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - K Ulrich Bayer
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Program in Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
12
|
Borghi R, Trivisano M, Specchio N, Tartaglia M, Compagnucci C. Understanding the pathogenetic mechanisms underlying altered neuronal function associated with CAMK2B mutations. Neurosci Biobehav Rev 2023; 152:105299. [PMID: 37391113 DOI: 10.1016/j.neubiorev.2023.105299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/26/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
'Dominant mutations in CAMK2B, encoding a subunit of the calcium/calmodulin-dependent protein kinase II (CAMK2), a serine/threonine kinase playing a key role in synaptic plasticity, learning and memory, underlie a recently characterized neurodevelopmental disorder (MRD54) characterized by delayed psychomotor development, mild to severe intellectual disability, hypotonia, and behavioral abnormalities. Targeted therapies to treat MRD54 are currently unavailable. In this review, we revise current knowledge on the molecular and cellular mechanisms underlying the altered neuronal function associated with defective CAMKIIβ function. We also summarize the identified genotype-phenotype correlations and discuss the disease models that have been generated to profile the altered neuronal phenotype and understand the pathophysiology of this disease.
Collapse
Affiliation(s)
- Rossella Borghi
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Marina Trivisano
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesu' Children's Hospital, IRCCS, Rome, Italy
| | - Nicola Specchio
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesu' Children's Hospital, IRCCS, Rome, Italy
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Claudia Compagnucci
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| |
Collapse
|
13
|
Tullis JE, Bayer KU. Distinct synaptic pools of DAPK1 differentially regulate activity-dependent synaptic CaMKII accumulation. iScience 2023; 26:106723. [PMID: 37216104 PMCID: PMC10192646 DOI: 10.1016/j.isci.2023.106723] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/18/2023] [Accepted: 04/19/2023] [Indexed: 05/24/2023] Open
Abstract
The death-associated protein kinase 1 (DAPK1) regulates the synaptic movement of the Ca2+/calmodulin (CaM)-dependent protein kinase II (CaMKII). Synaptic CaMKII accumulation is mediated via binding to the NMDA-receptor subunit GluN2B and is required for long-term potentiation (LTP). By contrast, long-term depression (LTD) instead requires specific suppression of this movement, which is mediated by competitive DAPK1 binding to GluN2B. We find here that DAPK1 localizes to synapses via two distinct mechanisms: basal localization requires F-actin, but retention of DAPK1 at synapses during LTD requires an additional binding mode, likely to GluN2B. While F-actin binding mediates DAPK1 enrichment at synapses, it is not sufficient to suppress synaptic CaMKII movement. However, it is a prerequisite that enables the additional LTD-specific binding mode of DAPK1, which in turn mediates suppression of the CaMKII movement. Thus, both modes of synaptic DAPK1 localization work together to regulate synaptic CaMKII localization and thereby synaptic plasticity.
Collapse
Affiliation(s)
- Jonathan E. Tullis
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - K. Ulrich Bayer
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Program in Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
14
|
Hachem LD, Hong J, Velumian A, Mothe AJ, Tator CH, Fehlings MG. Excitotoxic glutamate levels drive spinal cord ependymal stem cell proliferation and fate specification through CP-AMPAR signaling. Stem Cell Reports 2023; 18:672-687. [PMID: 36764296 PMCID: PMC10031285 DOI: 10.1016/j.stemcr.2023.01.005] [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: 05/06/2022] [Revised: 01/11/2023] [Accepted: 01/11/2023] [Indexed: 02/11/2023] Open
Abstract
The adult spinal cord contains a population of ependymal-derived neural stem/progenitor cells (epNSPCs) that are normally quiescent, but are activated to proliferate, differentiate, and migrate after spinal cord injury. The mechanisms that regulate their response to injury cues, however, remain unknown. Here, we demonstrate that excitotoxic levels of glutamate promote the proliferation and astrocytic fate specification of adult spinal cord epNSPCs. We show that glutamate-mediated calcium influx through calcium-permeable alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors (CP-AMPARs) in concert with Notch signaling increases the proliferation of epNSPCs via pCREB, and induces astrocytic differentiation through Hes1 upregulation. Furthermore, the in vivo targeting of this pathway via positive modulation of AMPARs after spinal cord injury enhances epNSPC proliferation, astrogliogenesis, neurotrophic factor production and increases neuronal survival. Our study uncovers an important mechanism by which CP-AMPARs regulate the growth and phenotype of epNSPCs, which can be targeted therapeutically to harness the regenerative potential of these cells after injury.
Collapse
Affiliation(s)
- Laureen D Hachem
- Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON M5T 2S8, Canada
| | - James Hong
- Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada
| | - Alexander Velumian
- Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON M5T 2S8, Canada
| | - Andrea J Mothe
- Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada
| | - Charles H Tator
- Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON M5T 2S8, Canada.
| | - Michael G Fehlings
- Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON M5T 2S8, Canada.
| |
Collapse
|
15
|
Cai Q, Chen X, Zhu S, Nicoll RA, Zhang M. Differential roles of CaMKII isoforms in phase separation with NMDA receptors and in synaptic plasticity. Cell Rep 2023; 42:112146. [PMID: 36827181 DOI: 10.1016/j.celrep.2023.112146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 12/17/2022] [Accepted: 02/06/2023] [Indexed: 02/24/2023] Open
Abstract
Calcium calmodulin-dependent kinase II (CaMKII) is critical for synaptic transmission and plasticity. Two major isoforms of CaMKII, CaMKIIα and CaMKIIβ, play distinct roles in synaptic transmission and long-term potentiation (LTP) with unknown mechanisms. Here, we show that the length of the unstructured linker between the kinase domain and the oligomerizing hub determines the ability of CaMKII to rescue the basal synaptic transmission and LTP defects caused by removal of both CaMKIIα and CaMKIIβ (double knockout [DKO]). Remarkably, although CaMKIIβ binds to GluN2B with a comparable affinity as CaMKIIα does, only CaMKIIα with the short linker forms robust dense clusters with GluN2B via phase separation. Lengthening the linker of CaMKIIα with unstructured "Gly-Gly-Ser" repeats impairs its phase separation with GluN2B, and the mutant enzyme cannot rescue the basal synaptic transmission and LTP defects of DKO mice. Our results suggest that the phase separation capacity of CaMKII with GluN2B is critical for its cellular functions in the brain.
Collapse
Affiliation(s)
- Qixu Cai
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Laboratory Medicine, School of Public Heath, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiumin Chen
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Shihan Zhu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Roger A Nicoll
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Mingjie Zhang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
16
|
Beghi S, Furmanik M, Jaminon A, Veltrop R, Rapp N, Wichapong K, Bidar E, Buschini A, Schurgers LJ. Calcium Signalling in Heart and Vessels: Role of Calmodulin and Downstream Calmodulin-Dependent Protein Kinases. Int J Mol Sci 2022; 23:ijms232416139. [PMID: 36555778 PMCID: PMC9783221 DOI: 10.3390/ijms232416139] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Cardiovascular disease is the major cause of death worldwide. The success of medication and other preventive measures introduced in the last century have not yet halted the epidemic of cardiovascular disease. Although the molecular mechanisms of the pathophysiology of the heart and vessels have been extensively studied, the burden of ischemic cardiovascular conditions has risen to become a top cause of morbidity and mortality. Calcium has important functions in the cardiovascular system. Calcium is involved in the mechanism of excitation-contraction coupling that regulates numerous events, ranging from the production of action potentials to the contraction of cardiomyocytes and vascular smooth muscle cells. Both in the heart and vessels, the rise of intracellular calcium is sensed by calmodulin, a protein that regulates and activates downstream kinases involved in regulating calcium signalling. Among them is the calcium calmodulin kinase family, which is involved in the regulation of cardiac functions. In this review, we present the current literature regarding the role of calcium/calmodulin pathways in the heart and vessels with the aim to summarize our mechanistic understanding of this process and to open novel avenues for research.
Collapse
Affiliation(s)
- Sofia Beghi
- Cardiovascular Research Institute Maastricht (CARIM), Department of Biochemistry, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area Delle Scienze 11A, 43124 Parma, Italy
- Correspondence: ; Tel.: +39-3408473527
| | - Malgorzata Furmanik
- Cardiovascular Research Institute Maastricht (CARIM), Department of Biochemistry, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Armand Jaminon
- Cardiovascular Research Institute Maastricht (CARIM), Department of Biochemistry, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Rogier Veltrop
- Cardiovascular Research Institute Maastricht (CARIM), Department of Biochemistry, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Nikolas Rapp
- Cardiovascular Research Institute Maastricht (CARIM), Department of Biochemistry, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Kanin Wichapong
- Cardiovascular Research Institute Maastricht (CARIM), Department of Biochemistry, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Elham Bidar
- Department of Cardiothoracic Surgery, Heart and Vascular Centre, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Annamaria Buschini
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area Delle Scienze 11A, 43124 Parma, Italy
| | - Leon J. Schurgers
- Cardiovascular Research Institute Maastricht (CARIM), Department of Biochemistry, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
17
|
Cervilla-Martínez JF, Rodríguez-Gotor JJ, Wypijewski KJ, Fontán-Lozano Á, Wang T, Santamaría E, Fuller W, Mejías R. Altered Cortical Palmitoylation Induces Widespread Molecular Disturbances in Parkinson's Disease. Int J Mol Sci 2022; 23:ijms232214018. [PMID: 36430497 PMCID: PMC9696982 DOI: 10.3390/ijms232214018] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/16/2022] Open
Abstract
The relationship between Parkinson's disease (PD), the second-most common neurodegenerative disease after Alzheimer's disease, and palmitoylation, a post-translational lipid modification, is not well understood. In this study, to better understand the role of protein palmitoylation in PD and the pathways altered in this disease, we analyzed the differential palmitoyl proteome (palmitome) in the cerebral cortex of PD patients compared to controls (n = 4 per group). Data-mining of the cortical palmitome from PD patients and controls allowed us to: (i) detect a set of 150 proteins with altered palmitoylation in PD subjects in comparison with controls; (ii) describe the biological pathways and targets predicted to be altered by these palmitoylation changes; and (iii) depict the overlap between the differential palmitome identified in our study with protein interactomes of the PD-linked proteins α-synuclein, LRRK2, DJ-1, PINK1, GBA and UCHL1. In summary, we partially characterized the altered palmitome in the cortex of PD patients, which is predicted to impact cytoskeleton, mitochondrial and fibrinogen functions, as well as cell survival. Our study suggests that protein palmitoylation could have a role in the pathophysiology of PD, and that comprehensive palmitoyl-proteomics offers a powerful approach for elucidating novel cellular pathways modulated in this neurodegenerative disease.
Collapse
Affiliation(s)
- Juan F. Cervilla-Martínez
- Department of Physiology, School of Biology, University of Seville, Avenida de la Reina Mercedes, 6, 41012 Sevilla, Spain
| | - Juan J. Rodríguez-Gotor
- Department of Physiology, School of Biology, University of Seville, Avenida de la Reina Mercedes, 6, 41012 Sevilla, Spain
- Instituto de Neurociencias CSIC-UMH, Avenida Santiago Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain
| | - Krzysztof J. Wypijewski
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- School of Life Sciences, University of Dundee, Dundee DD2 5DA, UK
| | - Ángela Fontán-Lozano
- Department of Physiology, School of Biology, University of Seville, Avenida de la Reina Mercedes, 6, 41012 Sevilla, Spain
- Instituto de Biomedicina de Sevilla, Campus Hospital Universitario Virgen del Rocío, Avda. Manuel Siurot, s/n, 41013 Sevilla, Spain
| | - Tao Wang
- McKusick—Nathans Institute of Genetic Medicine and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Enrique Santamaría
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IDISNA, Irunlarrea Street, 3, 31008 Pamplona, Spain
| | - William Fuller
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Rebeca Mejías
- Department of Physiology, School of Biology, University of Seville, Avenida de la Reina Mercedes, 6, 41012 Sevilla, Spain
- Instituto de Biomedicina de Sevilla, Campus Hospital Universitario Virgen del Rocío, Avda. Manuel Siurot, s/n, 41013 Sevilla, Spain
- Correspondence: ; Tel.: +34-954-559-549
| |
Collapse
|
18
|
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: 17] [Impact Index Per Article: 5.7] [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
|
19
|
Zernov N, Bezprozvanny I, Popugaeva E. CaMKIIβ knockdown decreases store-operated calcium entry in hippocampal dendritic spines. IBRO Neurosci Rep 2022; 12:90-97. [PMID: 35079728 PMCID: PMC8777283 DOI: 10.1016/j.ibneur.2022.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/06/2022] [Accepted: 01/08/2022] [Indexed: 01/13/2023] Open
Abstract
Calcium/calmodulin-dependent protein kinase II (CaMKII) and neuronal store-operated calcium entry (nSOCE) have been implicated in the development of Alzheimer's disease (AD). nSOCE is involved in regulation of dendritic spine shape, particularly in stability of mushroom spines that play role in formation of strong synapses. CaMKII is involved in regulation of induction of long-term potentiation, that is needed for shaping of memory. In the present study, we demonstrated that inhibition of kinase activity of CaMKII by KN-62 decreases nSOCE amplitude in soma of primary hippocampal neurons. We have shown that knockdown of CaMKIIβ leads to the downregulation of nSOCE in dendritic spines. In agreement with previously published data, we have also observed that CaMKIIβ knockdown causes mushroom spine loss in primary hippocampal culture. The effect of CaMKIIβ knockdown on the nSOCE may be associated with a decrease of dendritic spine head size.
Collapse
Affiliation(s)
- Nikita Zernov
- Peter the Great St.Petersburg Polytechnic University, Laboratory of Molecular Neurodegeneration, St.Petersburg, Russia
| | - Ilya Bezprozvanny
- Peter the Great St.Petersburg Polytechnic University, Laboratory of Molecular Neurodegeneration, St.Petersburg, Russia
- UT Southwestern Medical Center, Department of Physiology, Dallas, USA
| | - Elena Popugaeva
- Peter the Great St.Petersburg Polytechnic University, Laboratory of Molecular Neurodegeneration, St.Petersburg, Russia
- Corresponding author.
| |
Collapse
|
20
|
Mehrotra S, Pierce ML, Cao Z, Jabba SV, Gerwick WH, Murray TF. Antillatoxin-Stimulated Neurite Outgrowth Involves the Brain-Derived Neurotrophic Factor (BDNF) - Tropomyosin Related Kinase B (TrkB) Signaling Pathway. JOURNAL OF NATURAL PRODUCTS 2022; 85:562-571. [PMID: 35239341 PMCID: PMC9245549 DOI: 10.1021/acs.jnatprod.1c01001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Voltage-gated sodium channel (VGSC) activators promote neurite outgrowth by augmenting intracellular Na+ concentration ([Na+]i) and upregulating N-methyl-d-aspartate receptor (NMDAR) function. NMDAR activation stimulates calcium (Ca2+) influx and increases brain-derived neurotrophic factor (BDNF) release and activation of tropomyosin receptor kinase B (TrkB) signaling. The BDNF-TrkB pathway has been implicated in activity-dependent neuronal development. We have previously shown that antillatoxin (ATX), a novel lipopeptide isolated from the cyanobacterium Moorea producens, is a VGSC activator that produces an elevation of [Na+]i. Here we address the effect of ATX on the synthesis and release of BDNF and determine the signaling mechanisms by which ATX enhances neurite outgrowth in immature cerebrocortical neurons. ATX treatment produced a concentration-dependent release of BDNF. Acute treatment with ATX also resulted in increased synthesis of BDNF. ATX stimulation of neurite outgrowth was prevented by pretreatment with a TrkB inhibitor or transfection with a dominant-negative Trk-B. The ATX activation of TrkB and Akt was blocked by both a NMDAR antagonist (MK-801) and a VGSC blocker (tetrodotoxin). These results suggest that VGSC activators such as the structurally novel ATX may represent a new pharmacological strategy to promote neuronal plasticity through a NMDAR-BDNF-TrkB-dependent mechanism.
Collapse
Affiliation(s)
- Suneet Mehrotra
- Omeros, 201 Elliott Ave. West, Seattle, Washington 98119, United States
| | - Marsha L Pierce
- Department of Pharmacology, College of Graduate Studies, Midwestern University, Downers Grove, Illinois 60515, United States
| | - Zhengyu Cao
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Sairam V Jabba
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina 27710, United States
| | - William H Gerwick
- Center for Marine Biotech and Biomedicine, Scripps Institute of Oceanography, University of California at San Diego, San Diego, California 92093-0212, United States
| | - Thomas F Murray
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, 2500 California Plaza, Omaha, Nebraska 68178, United States
| |
Collapse
|
21
|
Saldivar-Cerón HI, Villamar-Cruz O, Wells CM, Oguz I, Spaggiari F, Chernoff J, Patiño-López G, Huerta-Yepez S, Montecillo-Aguado M, Rivera-Pazos CM, Loza-Mejía MA, Vivar-Sierra A, Briseño-Díaz P, Zentella-Dehesa A, Leon-Del-Rio A, López-Saavedra A, Padierna-Mota L, Ibarra-Sánchez MDJ, Esparza-López J, Hernández-Rivas R, Arias-Romero LE. p21-Activated Kinase 1 Promotes Breast Tumorigenesis via Phosphorylation and Activation of the Calcium/Calmodulin-Dependent Protein Kinase II. Front Cell Dev Biol 2022; 9:759259. [PMID: 35111748 PMCID: PMC8802317 DOI: 10.3389/fcell.2021.759259] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/07/2021] [Indexed: 12/22/2022] Open
Abstract
p21-Activated kinase-1 (Pak1) is frequently overexpressed and/or amplified in human breast cancer and is necessary for transformation of mammary epithelial cells. Here, we show that Pak1 interacts with and phosphorylates the Calcium/Calmodulin-dependent Protein Kinase II (CaMKII), and that pharmacological inhibition or depletion of Pak1 leads to diminished activity of CaMKII. We found a strong correlation between Pak1 and CaMKII expression in human breast cancer samples, and combined inhibition of Pak1 and CaMKII with small-molecule inhibitors was synergistic and induced apoptosis more potently in Her2 positive and triple negative breast cancer (TNBC) cells. Co-adminstration of Pak and CaMKII small-molecule inhibitors resulted in a dramatic reduction of proliferation and an increase in apoptosis in a 3D cell culture setting, as well as an impairment in migration and invasion of TNBC cells. Finally, mice bearing xenografts of TNBC cells showed a significant delay in tumor growth when treated with small-molecule inhibitors of Pak and CaMKII. These data delineate a signaling pathway from Pak1 to CaMKII that is required for efficient proliferation, migration and invasion of mammary epithelial cells, and suggest new therapeutic strategies in breast cancer.
Collapse
Affiliation(s)
- Héctor I Saldivar-Cerón
- UBIMED, Facultad de Estudios Superiores-Iztacala, UNAM, Tlalnepantla, Mexico.,Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Olga Villamar-Cruz
- UBIMED, Facultad de Estudios Superiores-Iztacala, UNAM, Tlalnepantla, Mexico
| | - Claire M Wells
- Division of Cancer Studies, New Hunts House, Guy's Campus, King's College London, London, United Kingdom
| | - Ibrahim Oguz
- Division of Cancer Studies, New Hunts House, Guy's Campus, King's College London, London, United Kingdom
| | - Federica Spaggiari
- Division of Cancer Studies, New Hunts House, Guy's Campus, King's College London, London, United Kingdom
| | - Jonathan Chernoff
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Genaro Patiño-López
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México, Mexico City, Mexico
| | - Sara Huerta-Yepez
- Unidad de Investigación en Enfermedades Hemato-Oncológicas, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Mayra Montecillo-Aguado
- Unidad de Investigación en Enfermedades Hemato-Oncológicas, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Clara M Rivera-Pazos
- Unidad de Investigación en Enfermedades Hemato-Oncológicas, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Marco A Loza-Mejía
- Facultad de Ciencias Químicas, Universidad La Salle-México, Mexico City, Mexico
| | - Alonso Vivar-Sierra
- Facultad de Ciencias Químicas, Universidad La Salle-México, Mexico City, Mexico
| | - Paola Briseño-Díaz
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Alejandro Zentella-Dehesa
- Programa de Investigación en Cáncer de Mama, Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico.,Unidad de Bioquímica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City, Mexico
| | - Alfonso Leon-Del-Rio
- Programa de Investigación en Cáncer de Mama, Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico
| | - Alejandro López-Saavedra
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico
| | - Laura Padierna-Mota
- UNe Aplicaciones Biológicas, Laboratorios de Especialidades Inmunologicas, Mexico City, Mexico
| | - María de Jesús Ibarra-Sánchez
- Unidad de Bioquímica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City, Mexico
| | - José Esparza-López
- Unidad de Bioquímica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City, Mexico
| | - Rosaura Hernández-Rivas
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Luis E Arias-Romero
- UBIMED, Facultad de Estudios Superiores-Iztacala, UNAM, Tlalnepantla, Mexico
| |
Collapse
|
22
|
Buonarati OR, Miller AP, Coultrap SJ, Bayer KU, Reichow SL. Conserved and divergent features of neuronal CaMKII holoenzyme structure, function, and high-order assembly. Cell Rep 2021; 37:110168. [PMID: 34965414 PMCID: PMC8985225 DOI: 10.1016/j.celrep.2021.110168] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 08/30/2021] [Accepted: 12/03/2021] [Indexed: 11/23/2022] Open
Abstract
Neuronal CaMKII holoenzymes (α and β isoforms) enable molecular signal computation underlying learning and memory but also mediate excitotoxic neuronal death. Here, we provide a comparative analysis of these signaling devices, using single-particle electron microscopy (EM) in combination with biochemical and live-cell imaging studies. In the basal state, both isoforms assemble mainly as 12-mers (but also 14-mers and even 16-mers for the β isoform). CaMKIIα and β isoforms adopt an ensemble of extended activatable states (with average radius of 12.6 versus 16.8 nm, respectively), characterized by multiple transient intra- and inter-holoenzyme interactions associated with distinct functional properties. The extended state of CaMKIIβ allows direct resolution of intra-holoenzyme kinase domain dimers. These dimers could enable cooperative activation by calmodulin, which is observed for both isoforms. High-order CaMKII clustering mediated by inter-holoenzyme kinase domain dimerization is reduced for the β isoform for both basal and excitotoxicity-induced clusters, both in vitro and in neurons. The CaMKII holoenzyme enables neuronal signal computation. In a comparative structure-function analysis of the neuronal α and β isoforms, Buonarati et al. find evidence for kinase domain dimers within the holoenzyme that enable a cooperative activation mechanism in both isoforms and inter-holoenzyme interactions that enable high-order aggregate formation under ischemic conditions.
Collapse
Affiliation(s)
- Olivia R Buonarati
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Adam P Miller
- Department of Chemistry, Portland State University, Portland, OR 97201, USA
| | - Steven J Coultrap
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - K Ulrich Bayer
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Steve L Reichow
- Department of Chemistry, Portland State University, Portland, OR 97201, USA.
| |
Collapse
|
23
|
Lu X, Zhou M, Liu N, Zhang C, Zhao Z, Cai D. Synaptic Protein Phosphorylation Networks Are Associated With Electroacupuncture-Induced Circadian Control in the Suprachiasmatic Nucleus. Front Genet 2021; 12:762557. [PMID: 34976011 PMCID: PMC8717940 DOI: 10.3389/fgene.2021.762557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 11/29/2021] [Indexed: 11/23/2022] Open
Abstract
Phosphorylation is one of the most important posttranslational modifications and regulates the physiological process. While recent studies highlight a major role of phosphorylation in the regulation of sleep–wake cycles to a lesser extent, the phosphoproteome in the suprachiasmatic nucleus (SCN) is not well-understood. Herein, we reported that the EA treatment elicits partial reparation of circadian rhythmicity when mice were exposure to constant darkness for long time. We investigated the effects of EA on circadian rhythms in constant darkness between EA stimulation and free-running control. Next, mass spectrometry–based phosphoproteome was utilized to explore the molecular characteristics of EA-induced phosphorylation modification in the SCN. A total of 6,192 distinct phosphosites on 2,488 proteins were quantified. Functional annotation analysis and protein–protein interaction networks demonstrated the most significant enriched phosphor-proteins and phosphosites involved in postsynapse and glutamatergic synapse. The current data indicated that most of the altered molecules are structural proteins. The target proteins, NMDAR and CAMK2, were selected for verification, consistent with the results of LC–MS/MS. These findings revealed a complete profile of phosphorylation modification in response to EA.
Collapse
Affiliation(s)
| | | | | | | | | | - Dingjun Cai
- *Correspondence: Zhengyu Zhao, ; Dingjun Cai,
| |
Collapse
|
24
|
Parcerisas A, Ortega-Gascó A, Pujadas L, Soriano E. The Hidden Side of NCAM Family: NCAM2, a Key Cytoskeleton Organization Molecule Regulating Multiple Neural Functions. Int J Mol Sci 2021; 22:10021. [PMID: 34576185 PMCID: PMC8471948 DOI: 10.3390/ijms221810021] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 02/07/2023] Open
Abstract
Although it has been over 20 years since Neural Cell Adhesion Molecule 2 (NCAM2) was identified as the second member of the NCAM family with a high expression in the nervous system, the knowledge of NCAM2 is still eclipsed by NCAM1. The first studies with NCAM2 focused on the olfactory bulb, where this protein has a key role in axonal projection and axonal/dendritic compartmentalization. In contrast to NCAM1, NCAM2's functions and partners in the brain during development and adulthood have remained largely unknown until not long ago. Recent studies have revealed the importance of NCAM2 in nervous system development. NCAM2 governs neuronal morphogenesis and axodendritic architecture, and controls important neuron-specific processes such as neuronal differentiation, synaptogenesis and memory formation. In the adult brain, NCAM2 is highly expressed in dendritic spines, and it regulates synaptic plasticity and learning processes. NCAM2's functions are related to its ability to adapt to the external inputs of the cell and to modify the cytoskeleton accordingly. Different studies show that NCAM2 interacts with proteins involved in cytoskeleton stability and proteins that regulate calcium influx, which could also modify the cytoskeleton. In this review, we examine the evidence that points to NCAM2 as a crucial cytoskeleton regulation protein during brain development and adulthood. This key function of NCAM2 may offer promising new therapeutic approaches for the treatment of neurodevelopmental diseases and neurodegenerative disorders.
Collapse
Affiliation(s)
- Antoni Parcerisas
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain; (A.O.-G.); (L.P.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Department of Basic Sciences, Universitat Internacional de Catalunya, 08195 Sant Cugat del Vallès, Spain
| | - Alba Ortega-Gascó
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain; (A.O.-G.); (L.P.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Lluís Pujadas
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain; (A.O.-G.); (L.P.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Eduardo Soriano
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain; (A.O.-G.); (L.P.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| |
Collapse
|
25
|
Hayashi Y. Molecular mechanism of hippocampal long-term potentiation - Towards multiscale understanding of learning and memory. Neurosci Res 2021; 175:3-15. [PMID: 34375719 DOI: 10.1016/j.neures.2021.08.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 11/16/2022]
Abstract
Long-term potentiation (LTP) of synaptic transmission is considered to be a cellular counterpart of learning and memory. Activation of postsynaptic NMDA type glutamate receptor (NMDA-R) induces trafficking of AMPA type glutamate receptors (AMPA-R) and other proteins to the synapse in sequential fashion. At the same time, the dendritic spine expands for long-term and modulation of actin underlies this (structural LTP or sLTP). How these changes persist despite constant diffusion and turnover of the component proteins have been the central focus of the current LTP research. Signaling triggered by Ca2+-influx via NMDA-R triggers kinase including Ca2+/calmodulin-dependent protein kinase II (CaMKII). CaMKII can sustain longer-term biochemical signaling by forming a reciprocally-activating kinase-effector complex with its substrate proteins including Tiam1, thereby regulating persistence of the downstream signaling. Furthermore, activated CaMKII can condense at the synapse through the mechanism of liquid-liquid phase separation (LLPS). This increases the binding capacity at the synapse, thereby contributing to the maintenance of enlarged protein complexes. It may also serve as the synapse tag, which captures newly synthesized proteins.
Collapse
Affiliation(s)
- Yasunori Hayashi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan.
| |
Collapse
|
26
|
Izadi M, Seemann E, Schlobinski D, Schwintzer L, Qualmann B, Kessels MM. Functional interdependence of the actin nucleator Cobl and Cobl-like in dendritic arbor development. eLife 2021; 10:67718. [PMID: 34264190 PMCID: PMC8282341 DOI: 10.7554/elife.67718] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
Local actin filament formation is indispensable for development of the dendritic arbor of neurons. We show that, surprisingly, the action of single actin filament-promoting factors was insufficient for powering dendritogenesis. Instead, this required the actin nucleator Cobl and its only evolutionary distant ancestor Cobl-like acting interdependently. This coordination between Cobl-like and Cobl was achieved by physical linkage by syndapins. Syndapin I formed nanodomains at convex plasma membrane areas at the base of protrusive structures and interacted with three motifs in Cobl-like, one of which was Ca2+/calmodulin-regulated. Consistently, syndapin I, Cobl-like’s newly identified N terminal calmodulin-binding site and the single Ca2+/calmodulin-responsive syndapin-binding motif all were critical for Cobl-like’s functions. In dendritic arbor development, local Ca2+/CaM-controlled actin dynamics thus relies on regulated and physically coordinated interactions of different F-actin formation-promoting factors and only together they have the power to bring about the sophisticated neuronal morphologies required for neuronal network formation in mammals.
Collapse
Affiliation(s)
- Maryam Izadi
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Eric Seemann
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Dirk Schlobinski
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Lukas Schwintzer
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Britta Qualmann
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Michael M Kessels
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
27
|
Sandal P, Jong CJ, Merrill RA, Song J, Strack S. Protein phosphatase 2A - structure, function and role in neurodevelopmental disorders. J Cell Sci 2021; 134:270819. [PMID: 34228795 DOI: 10.1242/jcs.248187] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Neurodevelopmental disorders (NDDs), including intellectual disability (ID), autism and schizophrenia, have high socioeconomic impact, yet poorly understood etiologies. A recent surge of large-scale genome or exome sequencing studies has identified a multitude of mostly de novo mutations in subunits of the protein phosphatase 2A (PP2A) holoenzyme that are strongly associated with NDDs. PP2A is responsible for at least 50% of total Ser/Thr dephosphorylation in most cell types and is predominantly found as trimeric holoenzymes composed of catalytic (C), scaffolding (A) and variable regulatory (B) subunits. PP2A can exist in nearly 100 different subunit combinations in mammalian cells, dictating distinct localizations, substrates and regulatory mechanisms. PP2A is well established as a regulator of cell division, growth, and differentiation, and the roles of PP2A in cancer and various neurodegenerative disorders, such as Alzheimer's disease, have been reviewed in detail. This Review summarizes and discusses recent reports on NDDs associated with mutations of PP2A subunits and PP2A-associated proteins. We also discuss the potential impact of these mutations on the structure and function of the PP2A holoenzymes and the etiology of NDDs.
Collapse
Affiliation(s)
- Priyanka Sandal
- Department of Neuroscience and Pharmacology, and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242, USA
| | - Chian Ju Jong
- Department of Neuroscience and Pharmacology, and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242, USA
| | - Ronald A Merrill
- Department of Neuroscience and Pharmacology, and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242, USA
| | - Jianing Song
- Department of Neuroscience and Pharmacology, and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242, USA
| | - Stefan Strack
- Department of Neuroscience and Pharmacology, and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242, USA
| |
Collapse
|
28
|
Sakai Y, Li H, Inaba H, Funayama Y, Ishimori E, Kawatake-Kuno A, Yamagata H, Seki T, Hobara T, Nakagawa S, Watanabe Y, Tomita S, Murai T, Uchida S. Gene-environment interactions mediate stress susceptibility and resilience through the CaMKIIβ/TARPγ-8/AMPAR pathway. iScience 2021; 24:102504. [PMID: 34113835 PMCID: PMC8170005 DOI: 10.1016/j.isci.2021.102504] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 01/07/2021] [Accepted: 04/29/2021] [Indexed: 01/09/2023] Open
Abstract
Although stressful events predispose individuals to psychiatric disorders, such as depression, not all people who undergo a stressful life experience become depressed, suggesting that gene-environment interactions (GxE) determine depression risk. The ventral hippocampus (vHPC) plays key roles in motivation, sociability, anhedonia, despair-like behaviors, anxiety, sleep, and feeding, pointing to the involvement of this brain region in depression. However, the molecular mechanisms underlying the cross talk between the vHPC and GxE in shaping behavioral susceptibility and resilience to chronic stress remain elusive. Here, we show that Ca2+/calmodulin-dependent protein kinase IIβ (CaMKIIβ) activity in the vHPC is differentially modulated in GxE mouse models of depression susceptibility and resilience, and that CaMKIIβ-mediated TARPγ-8 phosphorylation enhances the expression of AMPA receptor subunit GluA1 in the postsynaptic sites to enable stress resilience. We present previously missing molecular mechanisms underlying chronic stress-elicited behavioral changes, providing strategies for preventing and treating stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Yusuke Sakai
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Haiyan Li
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hiromichi Inaba
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
- Department of Psychiatry, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yuki Funayama
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
- Department of Psychiatry, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Erina Ishimori
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
- Department of Psychiatry, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Ayako Kawatake-Kuno
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hirotaka Yamagata
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
- Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, 4-1-8 Hon-cho, Kawaguchi, Saitama 332-0012, Japan
| | - Tomoe Seki
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
- Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, 4-1-8 Hon-cho, Kawaguchi, Saitama 332-0012, Japan
| | - Teruyuki Hobara
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
- Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, 4-1-8 Hon-cho, Kawaguchi, Saitama 332-0012, Japan
| | - Shin Nakagawa
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Yoshifumi Watanabe
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Susumu Tomita
- Department of Cellular and Molecular Physiology, Program in Cellular Neuroscience, Neurodegeneration, and Repair, Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Toshiya Murai
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
- Department of Psychiatry, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shusaku Uchida
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
- Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, 4-1-8 Hon-cho, Kawaguchi, Saitama 332-0012, Japan
| |
Collapse
|
29
|
The Calcium/Calmodulin-Dependent Kinases II and IV as Therapeutic Targets in Neurodegenerative and Neuropsychiatric Disorders. Int J Mol Sci 2021; 22:ijms22094307. [PMID: 33919163 PMCID: PMC8122486 DOI: 10.3390/ijms22094307] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/13/2021] [Accepted: 04/17/2021] [Indexed: 12/14/2022] Open
Abstract
CaMKII and CaMKIV are calcium/calmodulin-dependent kinases playing a rudimentary role in many regulatory processes in the organism. These kinases attract increasing interest due to their involvement primarily in memory and plasticity and various cellular functions. Although CaMKII and CaMKIV are mostly recognized as the important cogs in a memory machine, little is known about their effect on mood and role in neuropsychiatric diseases etiology. Here, we aimed to review the structure and functions of CaMKII and CaMKIV, as well as how these kinases modulate the animals’ behavior to promote antidepressant-like, anxiolytic-like, and procognitive effects. The review will help in the understanding of the roles of the above kinases in the selected neurodegenerative and neuropsychiatric disorders, and this knowledge can be used in future drug design.
Collapse
|
30
|
Kaltezioti V, Foskolou IP, Lavigne MD, Ninou E, Tsampoula M, Fousteri M, Margarity M, Politis PK. Prox1 inhibits neurite outgrowth during central nervous system development. Cell Mol Life Sci 2021; 78:3443-3465. [PMID: 33247761 PMCID: PMC11072475 DOI: 10.1007/s00018-020-03709-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 12/24/2022]
Abstract
During central nervous system (CNS) development, proper and timely induction of neurite elongation is critical for generating functional, mature neurons, and neuronal networks. Despite the wealth of information on the action of extracellular cues, little is known about the intrinsic gene regulatory factors that control this developmental decision. Here, we report the identification of Prox1, a homeobox transcription factor, as a key player in inhibiting neurite elongation. Although Prox1 promotes acquisition of early neuronal identity and is expressed in nascent post-mitotic neurons, it is heavily down-regulated in the majority of terminally differentiated neurons, indicating a regulatory role in delaying neurite outgrowth in newly formed neurons. Consistently, we show that Prox1 is sufficient to inhibit neurite extension in mouse and human neuroblastoma cell lines. More importantly, Prox1 overexpression suppresses neurite elongation in primary neuronal cultures as well as in the developing mouse brain, while Prox1 knock-down promotes neurite outgrowth. Mechanistically, RNA-Seq analysis reveals that Prox1 affects critical pathways for neuronal maturation and neurite extension. Interestingly, Prox1 strongly inhibits many components of Ca2+ signaling pathway, an important mediator of neurite extension and neuronal maturation. In accordance, Prox1 represses Ca2+ entry upon KCl-mediated depolarization and reduces CREB phosphorylation. These observations suggest that Prox1 acts as a potent suppressor of neurite outgrowth by inhibiting Ca2+ signaling pathway. This action may provide the appropriate time window for nascent neurons to find the correct position in the CNS prior to initiation of neurites and axon elongation.
Collapse
Affiliation(s)
- Valeria Kaltezioti
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Iosifina P Foskolou
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Matthieu D Lavigne
- Institute for Fundamental Biomedical Research, BSRC 'Alexander Fleming', 34 Fleming Street, Vari, 16672, Athens, Greece
| | - Elpinickie Ninou
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Matina Tsampoula
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Maria Fousteri
- Institute for Fundamental Biomedical Research, BSRC 'Alexander Fleming', 34 Fleming Street, Vari, 16672, Athens, Greece
| | - Marigoula Margarity
- Laboratory of Human and Animal Physiology, Department of Biology, School of Natural Sciences, University of Patras, 26500, Rio Achaias, Greece
| | - Panagiotis K Politis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece.
| |
Collapse
|
31
|
Proietti Onori M, van Woerden GM. Role of calcium/calmodulin-dependent kinase 2 in neurodevelopmental disorders. Brain Res Bull 2021; 171:209-220. [PMID: 33774142 DOI: 10.1016/j.brainresbull.2021.03.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 01/28/2023]
Abstract
Neurodevelopmental disorders are a complex and heterogeneous group of neurological disorders characterized by their early-onset and estimated to affect more than 3% of children worldwide. The rapid advancement of sequencing technologies in the past years allowed the identification of hundreds of variants in several different genes causing neurodevelopmental disorders. Between those, new variants in the Calcium/calmodulin dependent protein kinase II (CAMK2) genes were recently linked to intellectual disability. Despite many years of research on CAMK2, this proves for the first time that this well-known and highly conserved molecule plays an important role in the human brain. In this review, we give an overview of the identified CAMK2 variants, and we speculate on potential mechanisms through which dysfunctions in CAMK2 result in neurodevelopmental disorders. Additionally, we discuss how the identification of CAMK2 variants might result in new exciting discoveries regarding the function of CAMK2 in the human brain.
Collapse
Affiliation(s)
- Martina Proietti Onori
- Department of Neuroscience, Erasmus MC, Rotterdam, 3015 GD, the Netherlands; The ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, 3015 GD, the Netherlands
| | - Geeske M van Woerden
- Department of Neuroscience, Erasmus MC, Rotterdam, 3015 GD, the Netherlands; The ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, 3015 GD, the Netherlands.
| |
Collapse
|
32
|
Abstract
Neurons develop dendritic morphologies that bear cell type-specific features in dendritic field size and geometry, branch placement and density, and the types and distributions of synaptic contacts. Dendritic patterns influence the types and numbers of inputs a neuron receives, and the ways in which neural information is processed and transmitted in the circuitry. Even subtle alterations in dendritic structures can have profound consequences on neuronal function and are implicated in neurodevelopmental disorders. In this chapter, I review how growing dendrites acquire their exquisite patterns by drawing examples from diverse neuronal cell types in vertebrate and invertebrate model systems. Dendrite morphogenesis is shaped by intrinsic and extrinsic factors such as transcriptional regulators, guidance and adhesion molecules, neighboring cells and synaptic partners. I discuss molecular mechanisms that regulate dendrite morphogenesis with a focus on five aspects of dendrite patterning: (1) Dendritic cytoskeleton and cellular machineries that build the arbor; (2) Gene regulatory mechanisms; (3) Afferent cues that regulate dendritic arbor growth; (4) Space-filling strategies that optimize dendritic coverage; and (5) Molecular cues that specify dendrite wiring. Cell type-specific implementation of these patterning mechanisms produces the diversity of dendrite morphologies that wire the nervous system.
Collapse
|
33
|
Nourbakhsh K, Yadav S. Kinase Signaling in Dendritic Development and Disease. Front Cell Neurosci 2021; 15:624648. [PMID: 33642997 PMCID: PMC7902504 DOI: 10.3389/fncel.2021.624648] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/06/2021] [Indexed: 01/19/2023] Open
Abstract
Dendrites undergo extensive growth and remodeling during their lifetime. Specification of neurites into dendrites is followed by their arborization, maturation, and functional integration into synaptic networks. Each of these distinct developmental processes is spatially and temporally controlled in an exquisite fashion. Protein kinases through their highly specific substrate phosphorylation regulate dendritic growth and plasticity. Perturbation of kinase function results in aberrant dendritic growth and synaptic function. Not surprisingly, kinase dysfunction is strongly associated with neurodevelopmental and psychiatric disorders. Herein, we review, (a) key kinase pathways that regulate dendrite structure, function and plasticity, (b) how aberrant kinase signaling contributes to dendritic dysfunction in neurological disorders and (c) emergent technologies that can be applied to dissect the role of protein kinases in dendritic structure and function.
Collapse
Affiliation(s)
| | - Smita Yadav
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| |
Collapse
|
34
|
Di Paolo A, Eastman G, Mesquita-Ribeiro R, Farias J, Macklin A, Kislinger T, Colburn N, Munroe D, Sotelo Sosa JR, Dajas-Bailador F, Sotelo-Silveira JR. PDCD4 regulates axonal growth by translational repression of neurite growth-related genes and is modulated during nerve injury responses. RNA (NEW YORK, N.Y.) 2020; 26:1637-1653. [PMID: 32747606 PMCID: PMC7566564 DOI: 10.1261/rna.075424.120] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 07/20/2020] [Indexed: 05/07/2023]
Abstract
Programmed cell death 4 (PDCD4) protein is a tumor suppressor that inhibits translation through the mTOR-dependent initiation factor EIF4A, but its functional role and mRNA targets in neurons remain largely unknown. Our work identified that PDCD4 is highly expressed in axons and dendrites of CNS and PNS neurons. Using loss- and gain-of-function experiments in cortical and dorsal root ganglia primary neurons, we demonstrated the capacity of PDCD4 to negatively control axonal growth. To explore PDCD4 transcriptome and translatome targets, we used Ribo-seq and uncovered a list of potential targets with known functions as axon/neurite outgrowth regulators. In addition, we observed that PDCD4 can be locally synthesized in adult axons in vivo, and its levels decrease at the site of peripheral nerve injury and before nerve regeneration. Overall, our findings demonstrate that PDCD4 can act as a new regulator of axonal growth via the selective control of translation, providing a target mechanism for axon regeneration and neuronal plasticity processes in neurons.
Collapse
Affiliation(s)
- Andrés Di Paolo
- Departamento de Proteínas y Ácidos Nucleicos, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay
| | - Guillermo Eastman
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay
| | | | - Joaquina Farias
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay
| | - Andrew Macklin
- Princess Margaret Cancer Centre, University Health Network, Toronto M5G 1L7, Canada
| | - Thomas Kislinger
- Princess Margaret Cancer Centre, University Health Network, Toronto M5G 1L7, Canada
- University of Toronto, Department of Medical Biophysics, Toronto M5S 1A1, Canada
| | - Nancy Colburn
- Former Chief of Laboratory of Cancer Prevention at the National Cancer Institute-NIH at Frederick, Maryland 21702, USA
| | - David Munroe
- Former Laboratory of Molecular Technologies, LEIDOS at Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, USA
| | - José R Sotelo Sosa
- Departamento de Proteínas y Ácidos Nucleicos, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay
| | | | - José R Sotelo-Silveira
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay
- Departamento de Biología Celular y Molecular, Facultad de Ciencias UdelaR, Montevideo 11400, Uruguay
| |
Collapse
|
35
|
Nicole O, Pacary E. CaMKIIβ in Neuronal Development and Plasticity: An Emerging Candidate in Brain Diseases. Int J Mol Sci 2020; 21:ijms21197272. [PMID: 33019657 PMCID: PMC7582470 DOI: 10.3390/ijms21197272] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 09/25/2020] [Accepted: 09/29/2020] [Indexed: 01/17/2023] Open
Abstract
The calcium/calmodulin-dependent protein kinase II (CaMKII) is a ubiquitous and central player in Ca2+ signaling that is best known for its functions in the brain. In particular, the α isoform of CaMKII has been the subject of intense research and it has been established as a central regulator of neuronal plasticity. In contrast, little attention has been paid to CaMKIIβ, the other predominant brain isoform that interacts directly with the actin cytoskeleton, and the functions of CaMKIIβ in this organ remain largely unexplored. However, recently, the perturbation of CaMKIIβ expression has been associated with multiple neuropsychiatric and neurodevelopmental diseases, highlighting CAMK2B as a gene of interest. Herein, after highlighting the main structural and expression differences between the α and β isoforms, we will review the specific functions of CaMKIIβ, as described so far, in neuronal development and plasticity, as well as its potential implication in brain diseases.
Collapse
Affiliation(s)
- Olivier Nicole
- CNRS, UMR5293 Institut des Maladies Neurodégénératives, University of Bordeaux, F-33000 Bordeaux, France;
| | - Emilie Pacary
- INSERM, Neurocentre Magendie, U1215, University of Bordeaux, F-33000 Bordeaux, France
- Correspondence:
| |
Collapse
|
36
|
AUTS2 Regulation of Synapses for Proper Synaptic Inputs and Social Communication. iScience 2020; 23:101183. [PMID: 32498016 PMCID: PMC7267731 DOI: 10.1016/j.isci.2020.101183] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/30/2020] [Accepted: 05/15/2020] [Indexed: 01/11/2023] Open
Abstract
Impairments in synapse development are thought to cause numerous psychiatric disorders. Autism susceptibility candidate 2 (AUTS2) gene has been associated with various psychiatric disorders, such as autism and intellectual disabilities. Although roles for AUTS2 in neuronal migration and neuritogenesis have been reported, its involvement in synapse regulation remains unclear. In this study, we found that excitatory synapses were specifically increased in the Auts2-deficient primary cultured neurons as well as Auts2 mutant forebrains. Electrophysiological recordings and immunostaining showed increases in excitatory synaptic inputs as well as c-fos expression in Auts2 mutant brains, suggesting that an altered balance of excitatory and inhibitory inputs enhances brain excitability. Auts2 mutant mice exhibited autistic-like behaviors including impairments in social interaction and altered vocal communication. Together, these findings suggest that AUTS2 regulates excitatory synapse number to coordinate E/I balance in the brain, whose impairment may underlie the pathology of psychiatric disorders in individuals with AUTS2 mutations. AUTS2 regulates excitatory synapse number in forebrain pyramidal neurons Loss of Auts2 leads to increased spine formation in development and adulthood Loss of Auts2 alters the balance of excitatory and inhibitory synaptic inputs Auts2 mutant mice exhibit cognitive and sociobehavioral deficits
Collapse
|
37
|
Bennison SA, Blazejewski SM, Smith TH, Toyo-Oka K. Protein kinases: master regulators of neuritogenesis and therapeutic targets for axon regeneration. Cell Mol Life Sci 2020; 77:1511-1530. [PMID: 31659414 PMCID: PMC7166181 DOI: 10.1007/s00018-019-03336-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/16/2019] [Accepted: 10/08/2019] [Indexed: 12/25/2022]
Abstract
Proper neurite formation is essential for appropriate neuronal morphology to develop and defects at this early foundational stage have serious implications for overall neuronal function. Neuritogenesis is tightly regulated by various signaling mechanisms that control the timing and placement of neurite initiation, as well as the various processes necessary for neurite elongation to occur. Kinases are integral components of these regulatory pathways that control the activation and inactivation of their targets. This review provides a comprehensive summary of the kinases that are notably involved in regulating neurite formation, which is a complex process that involves cytoskeletal rearrangements, addition of plasma membrane to increase neuronal surface area, coupling of cytoskeleton/plasma membrane, metabolic regulation, and regulation of neuronal differentiation. Since kinases are key regulators of these functions during neuromorphogenesis, they have high potential for use as therapeutic targets for axon regeneration after injury or disease where neurite formation is disrupted.
Collapse
Affiliation(s)
- Sarah A Bennison
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Sara M Blazejewski
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Trevor H Smith
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Kazuhito Toyo-Oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA.
| |
Collapse
|
38
|
Parcerisas A, Pujadas L, Ortega-Gascó A, Perelló-Amorós B, Viais R, Hino K, Figueiro-Silva J, La Torre A, Trullás R, Simó S, Lüders J, Soriano E. NCAM2 Regulates Dendritic and Axonal Differentiation through the Cytoskeletal Proteins MAP2 and 14-3-3. Cereb Cortex 2020; 30:3781-3799. [PMID: 32043120 DOI: 10.1093/cercor/bhz342] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/27/2019] [Accepted: 01/08/2020] [Indexed: 01/13/2023] Open
Abstract
Neural cell adhesion molecule 2 (NCAM2) is involved in the development and plasticity of the olfactory system. Genetic data have implicated the NCAM2 gene in neurodevelopmental disorders including Down syndrome and autism, although its role in cortical development is unknown. Here, we show that while overexpression of NCAM2 in hippocampal neurons leads to minor alterations, its downregulation severely compromises dendritic architecture, leading to an aberrant phenotype including shorter dendritic trees, retraction of dendrites, and emergence of numerous somatic neurites. Further, our data reveal alterations in the axonal tree and deficits in neuronal polarization. In vivo studies confirm the phenotype and reveal an unexpected role for NCAM2 in cortical migration. Proteomic and cell biology experiments show that NCAM2 molecules exert their functions through a protein complex with the cytoskeletal-associated proteins MAP2 and 14-3-3γ and ζ. We provide evidence that NCAM2 depletion results in destabilization of the microtubular network and reduced MAP2 signal. Our results demonstrate a role for NCAM2 in dendritic formation and maintenance, and in neural polarization and migration, through interaction of NCAM2 with microtubule-associated proteins.
Collapse
Affiliation(s)
- Antoni Parcerisas
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.,Vall d'Hebron Institut de Recerca (VHIR), 08035, Barcelona, Spain
| | - Lluís Pujadas
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.,Vall d'Hebron Institut de Recerca (VHIR), 08035, Barcelona, Spain
| | - Alba Ortega-Gascó
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.,Vall d'Hebron Institut de Recerca (VHIR), 08035, Barcelona, Spain
| | - Bartomeu Perelló-Amorós
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.,Vall d'Hebron Institut de Recerca (VHIR), 08035, Barcelona, Spain
| | - Ricardo Viais
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), 08028, Barcelona, Spain
| | - Keiko Hino
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA 95616, USA
| | - Joana Figueiro-Silva
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.,Neurobiology Unit, Institut d'Investigacions Biomèdiques de Barcelona, CSIC, IDIBAPS, 08036, Barcelona, Spain
| | - Anna La Torre
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA 95616, USA
| | - Ramón Trullás
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.,Neurobiology Unit, Institut d'Investigacions Biomèdiques de Barcelona, CSIC, IDIBAPS, 08036, Barcelona, Spain
| | - Sergi Simó
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA 95616, USA
| | - Jens Lüders
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), 08028, Barcelona, Spain
| | - Eduardo Soriano
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.,Vall d'Hebron Institut de Recerca (VHIR), 08035, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA) Academia, 08010, Barcelona, Spain
| |
Collapse
|
39
|
Djemil S, Chen X, Zhang Z, Lee J, Rauf M, Pak DTS, Dzakpasu R. Activation of nicotinic acetylcholine receptors induces potentiation and synchronization within in vitro hippocampal networks. J Neurochem 2019; 153:468-484. [PMID: 31821553 DOI: 10.1111/jnc.14938] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 01/08/2023]
Abstract
Nicotinic acetylcholine receptors (nAChRs) are known to play a role in cognitive functions of the hippocampus, such as memory consolidation. Given that they conduct Ca2+ and are capable of regulating the release of glutamate and γ-aminobutyric acid (GABA) within the hippocampus, thereby shifting the excitatory-inhibitory ratio, we hypothesized that the activation of nAChRs will result in the potentiation of hippocampal networks and alter synchronization. We used nicotine as a tool to investigate the impact of activation of nAChRs on neuronal network dynamics in primary embryonic rat hippocampal cultures prepared from timed-pregnant Sprague-Dawley rats. We perturbed cultured hippocampal networks with increasing concentrations of bath-applied nicotine and performed network extracellular recordings of action potentials using a microelectrode array. We found that nicotine modulated network dynamics in a concentration-dependent manner; it enhanced firing of action potentials as well as facilitated bursting activity. In addition, we used pharmacological agents to determine the contributions of discrete nAChR subtypes to the observed network dynamics. We found that β4-containing nAChRs are necessary for the observed increases in spiking, bursting, and synchrony, while the activation of α7 nAChRs augments nicotine-mediated network potentiation but is not necessary for its manifestation. We also observed that antagonists of N-methyl-D-aspartate receptors (NMDARs) and group I metabotropic glutamate receptors (mGluRs) partially blocked the effects of nicotine. Furthermore, nicotine exposure promoted autophosphorylation of Ca2+ /calmodulin-dependent kinase II (CaMKII) and serine 831 phosphorylation of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) subunit GluA1. These results suggest that nicotinic receptors induce potentiation and synchronization of hippocampal networks and glutamatergic synaptic transmission. Findings from this work highlight the impact of cholinergic signaling in generating network-wide potentiation in the form of enhanced spiking and bursting dynamics that coincide with molecular correlates of memory such as increased phosphorylation of CaMKII and GluA1. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Sarra Djemil
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Xin Chen
- Department of Physics, Georgetown University, Washington, DC, USA
| | - Ziyue Zhang
- Department of Physics, Georgetown University, Washington, DC, USA
| | - Jisoo Lee
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Mikael Rauf
- Department of Human Science, Georgetown University Medical Center, Washington, DC, USA
| | - Daniel T S Pak
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA.,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Rhonda Dzakpasu
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA.,Department of Physics, Georgetown University, Washington, DC, USA.,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
40
|
Vargas JY, Loria F, Wu Y, Córdova G, Nonaka T, Bellow S, Syan S, Hasegawa M, van Woerden GM, Trollet C, Zurzolo C. The Wnt/Ca 2+ pathway is involved in interneuronal communication mediated by tunneling nanotubes. EMBO J 2019; 38:e101230. [PMID: 31625188 PMCID: PMC6885744 DOI: 10.15252/embj.2018101230] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 09/08/2019] [Accepted: 09/12/2019] [Indexed: 01/15/2023] Open
Abstract
Tunneling nanotubes (TNTs) are actin-based transient tubular connections that allow direct communication between distant cells. TNTs play an important role in several physiological (development, immunity, and tissue regeneration) and pathological (cancer, neurodegeneration, and pathogens transmission) processes. Here, we report that the Wnt/Ca2+ pathway, an intracellular cascade that is involved in actin cytoskeleton remodeling, has a role in TNT formation and TNT-mediated transfer of cargoes. Specifically, we found that Ca2+ /calmodulin-dependent protein kinase II (CaMKII), a transducer of the Wnt/Ca2+ pathway, regulates TNTs in a neuronal cell line and in primary neurons. We identified the β isoform of CaMKII as a key molecule in modulating TNT formation and transfer, showing that this depends on the actin-binding activity of the protein. Finally, we found that the transfer of vesicles and aggregated α-synuclein between primary neurons can be regulated by the activation of the Wnt/Ca2+ pathway. Our findings suggest that Wnt/Ca2+ pathway could be a novel promising target for therapies designed to impair TNT-mediated propagation of pathogens.
Collapse
Affiliation(s)
- Jessica Y Vargas
- Unité de Trafic Membranaire et PathogénèseDépartement de Biologie Cellulaire et de l'InfectionInstitut PasteurParisFrance
| | - Frida Loria
- Unité de Trafic Membranaire et PathogénèseDépartement de Biologie Cellulaire et de l'InfectionInstitut PasteurParisFrance
- Present address:
Centro de Biología Molecular Severo Ochoa (CSIC‐UAM)Departamento de Biología MolecularUniversidad Autónoma de MadridMadridSpain
| | - Yuan‐Ju Wu
- Unité de Trafic Membranaire et PathogénèseDépartement de Biologie Cellulaire et de l'InfectionInstitut PasteurParisFrance
| | - Gonzalo Córdova
- Institut National de la Santé et de la Recherche MédicaleAssociation Institut de MyologieCentre de Recherche en MyologieUMRS974Sorbonne UniversitéParisFrance
| | - Takashi Nonaka
- Department of Dementia and Higher Brain FunctionTokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | | | - Sylvie Syan
- Unité de Trafic Membranaire et PathogénèseDépartement de Biologie Cellulaire et de l'InfectionInstitut PasteurParisFrance
| | - Masato Hasegawa
- Department of Dementia and Higher Brain FunctionTokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Geeske M van Woerden
- Department of NeuroscienceErasmus Medical CenterRotterdamThe Netherlands
- ENCORE Expertise Center for Neurodevelopmental DisordersErasmus Medical CenterRotterdamThe Netherlands
| | - Capucine Trollet
- Institut National de la Santé et de la Recherche MédicaleAssociation Institut de MyologieCentre de Recherche en MyologieUMRS974Sorbonne UniversitéParisFrance
| | - Chiara Zurzolo
- Unité de Trafic Membranaire et PathogénèseDépartement de Biologie Cellulaire et de l'InfectionInstitut PasteurParisFrance
| |
Collapse
|
41
|
Herbison R, Evans S, Doherty JF, Algie M, Kleffmann T, Poulin R. A molecular war: convergent and ontogenetic evidence for adaptive host manipulation in related parasites infecting divergent hosts. Proc Biol Sci 2019; 286:20191827. [PMID: 31744433 DOI: 10.1098/rspb.2019.1827] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mermithids (phylum Nematoda) and hairworms (phylum Nematomorpha) somehow drive their arthropod hosts into water, which is essential for the worms' survival after egression. The mechanisms behind this behavioural change have been investigated in hairworms, but not in mermithids. Establishing a similar mechanistic basis for host behavioural change between these two distantly related parasitic groups would provide strong convergent evidence for adaptive manipulation and insight into how these parasites modify and/or create behaviour. Here, we search for this convergence, and also contrast changes in physiology between hosts infected with immature and mature mermithids to provide the first ontogenetic evidence for adaptive manipulation by disentangling host response and pathology from the parasite's apparent manipulative effects. We used SWATH-mass spectrometry on brains of Forficula auricularia (earwig) and Bellorchestia quoyana (sandhopper), infected with the mermithids Mermis nigrescens and Thaumamermis zealandica, respectively, at both immature and mature stages of infection, to quantify proteomic changes resulting from mermithid infection. Across both hosts (and hairworm-infected hosts, from earlier studies), the general function of dysregulated proteins was conserved. Proteins involved in energy generation/mobilization were dysregulated, corroborating reports of erratic/hyperactive behaviour in infected hosts. Dysregulated proteins involved in axon/dendrite and synapse modulation were also common to all hosts, suggesting neuronal manipulation is involved in inducing positive hydrotaxis. Furthermore, downregulation of CamKII and associated proteins suggest manipulation of memory also contributes to the behavioural shift.
Collapse
Affiliation(s)
- Ryan Herbison
- Department of Zoology, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Steven Evans
- Department of Zoology, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | | | - Michael Algie
- Department of Biochemistry, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Torsten Kleffmann
- Department of Biochemistry, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Robert Poulin
- Department of Zoology, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| |
Collapse
|
42
|
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: 233] [Impact Index Per Article: 38.8] [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
|
43
|
Abstract
The structure of neuronal circuits that subserve cognitive functions in the brain is shaped and refined throughout development and into adulthood. Evidence from human and animal studies suggests that the cellular and synaptic substrates of these circuits are atypical in neuropsychiatric disorders, indicating that altered structural plasticity may be an important part of the disease biology. Advances in genetics have redefined our understanding of neuropsychiatric disorders and have revealed a spectrum of risk factors that impact pathways known to influence structural plasticity. In this Review, we discuss the importance of recent genetic findings on the different mechanisms of structural plasticity and propose that these converge on shared pathways that can be targeted with novel therapeutics.
Collapse
|
44
|
Di G, Liu G, Xu Y, Kim H. Effects of combined traffic noise on the synaptic ultrastructure and expressions of p-CaMKII and NMDAR1 in the hippocampus of young SD rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:22030-22039. [PMID: 31140091 DOI: 10.1007/s11356-019-05457-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 05/14/2019] [Indexed: 06/09/2023]
Abstract
In order to explore the effects of combined traffic noise (CTN) on learning and memory function, young Sprague-Dawley (SD) rats were exposed to CTN from highway and high-speed railway for 52 days, whose day-night equivalent continuous A-weighted sound pressure level (Ldn) was 70 dB(A) (corresponding sound pressure level was 80 dB). The synaptic ultrastructure and the expressions of phosphorylated calcium/calmodulin-dependent protein kinase II (p-CaMKII) and N-methyl-D-aspartate receptor 1 (NMDAR1 or NR1) in the hippocampus were tested by transmission electron microscopy (TEM) and Western blot, respectively. Results showed that there was no significant difference in the synaptic ultrastructure and the expressions of p-CaMKII and NR1 in the hippocampus of young rats between the experimental group and control group. Compared with single high-speed railway noise (HSRN) with Ldn of 70 dB(A), CTN had less influences on learning and memory function, which was closely related to smaller intermittency of CTN and less anxiety caused by CTN. In comparison with white noise with a sound pressure level of 80 dB, CTN had less impacts on learning and memory function, which was mainly associated with CTN's smaller R-weighted sound pressure level based on rats' auditory sensitivity.
Collapse
Affiliation(s)
- Guoqing Di
- Department of Environmental Science, College of Environmental and Resource Sciences, Zhejiang University, No. 866 Yuhangtang Road, Hangzhou, 310058, People's Republic of China.
| | - Guangxiang Liu
- Department of Environmental Science, College of Environmental and Resource Sciences, Zhejiang University, No. 866 Yuhangtang Road, Hangzhou, 310058, People's Republic of China
| | - Yaqian Xu
- Department of Environmental Science, College of Environmental and Resource Sciences, Zhejiang University, No. 866 Yuhangtang Road, Hangzhou, 310058, People's Republic of China
| | - Hakbong Kim
- Department of Environmental Science, College of Environmental and Resource Sciences, Zhejiang University, No. 866 Yuhangtang Road, Hangzhou, 310058, People's Republic of China
| |
Collapse
|
45
|
Xi F, Xu RJ, Xu JH, Ma JJ, Wang WH, Wang F, Ma YX, Qi SB, Zhang HC, Zhang HN, Qin XZ, Chen JQ, Li B, Liu CM, Yang HL, Meng B, Saijilafu. Calcium/calmodulin-dependent protein kinase II regulates mammalian axon growth by affecting F-actin length in growth cone. J Cell Physiol 2019; 234:23053-23065. [PMID: 31134625 DOI: 10.1002/jcp.28867] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/09/2019] [Accepted: 05/09/2019] [Indexed: 12/21/2022]
Abstract
While axon regeneration is a key determinant of functional recovery of the nervous system after injury, it is often poor in the mature nervous system. Influx of extracellular calcium (Ca2+ ) is one of the first phenomena that occur following axonal injury, and calcium/calmodulin-dependent protein kinase II (CaMKII), a target substrate for calcium ions, regulates the status of cytoskeletal proteins such as F-actin. Herein, we found that peripheral axotomy activates CaMKII in dorsal root ganglion (DRG) sensory neurons, and inhibition of CaMKII impairs axon outgrowth in both the peripheral and central nervous systems (PNS and CNS, respectively). Most importantly, we also found that the activation of CaMKII promotes PNS and CNS axon growth, and regulatory effects of CaMKII on axon growth occur via affecting the length of the F-actin. Thus, we believe our findings provide clear evidence that CaMKII is a critical modulator of mammalian axon regeneration.
Collapse
Affiliation(s)
- Feng Xi
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, P. R. China
| | - Ren-Jie Xu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, P. R. China.,Department of Orthopaedics, Suzhou Municipal Hospital, The Affiliated Hospital of Nanjing Medical University, Suzhou, Jiangsu, P. R. China
| | - Jin-Hui Xu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, P. R. China
| | - Jin-Jin Ma
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, P. R. China
| | - Wei-Hua Wang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, P. R. China
| | - Feng Wang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, P. R. China
| | - Yan-Xia Ma
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, P. R. China
| | - Shi-Bin Qi
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, P. R. China
| | - Hong-Cheng Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, P. R. China
| | - Hao-Nan Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, P. R. China
| | - Xu-Zhen Qin
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, P. R. China
| | - Jian-Quan Chen
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, P. R. China
| | - Bin Li
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, P. R. China
| | - Chang-Mei Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Science, Beijing, P. R. China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Hui-Lin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, P. R. China
| | - Bin Meng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, P. R. China
| | - Saijilafu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, P. R. China
| |
Collapse
|
46
|
Kudryashova IV. The Molecular Basis of Destabilization of Synapses as a Factor of Structural Plasticity. NEUROCHEM J+ 2019. [DOI: 10.1134/s1819712419010136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
47
|
Brzozowski JS, Skelding KA. The Multi-Functional Calcium/Calmodulin Stimulated Protein Kinase (CaMK) Family: Emerging Targets for Anti-Cancer Therapeutic Intervention. Pharmaceuticals (Basel) 2019; 12:ph12010008. [PMID: 30621060 PMCID: PMC6469190 DOI: 10.3390/ph12010008] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/02/2019] [Accepted: 01/04/2019] [Indexed: 01/25/2023] Open
Abstract
The importance of Ca2+ signalling in key events of cancer cell function and tumour progression, such as proliferation, migration, invasion and survival, has recently begun to be appreciated. Many cellular Ca2+-stimulated signalling cascades utilise the intermediate, calmodulin (CaM). The Ca2+/CaM complex binds and activates a variety of enzymes, including members of the multifunctional Ca2+/calmodulin-stimulated protein kinase (CaMK) family. These enzymes control a broad range of cancer-related functions in a multitude of tumour types. Herein, we explore the cancer-related functions of these kinases and discuss their potential as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Joshua S Brzozowski
- Priority Research Centre for Cancer Research, Innovation and Translation, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute (HMRI) and University of Newcastle, Callaghan, NSW 2308, Australia.
| | - Kathryn A Skelding
- Priority Research Centre for Cancer Research, Innovation and Translation, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute (HMRI) and University of Newcastle, Callaghan, NSW 2308, Australia.
| |
Collapse
|
48
|
van de Looij Y, Larpin C, Cabungcal JH, Sanches EF, Toulotte A, Do KQ, Sizonenko SV. Nutritional Intervention for Developmental Brain Damage: Effects of Lactoferrin Supplementation in Hypocaloric Induced Intrauterine Growth Restriction Rat Pups. Front Endocrinol (Lausanne) 2019; 10:46. [PMID: 30800096 PMCID: PMC6375847 DOI: 10.3389/fendo.2019.00046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 01/21/2019] [Indexed: 12/13/2022] Open
Abstract
Introduction: Intrauterine Growth Restriction (IUGR) refers to an impaired development of the fetus and hence results in adverse neurodevelopmental and psychiatric consequences later in life. Lactoferrin (Lf) is a glycoprotein present in milk that has already shown neuroprotective effects through its anti-inflammatory and antioxidant properties on impaired developing brains. The aim of this study was to characterize a rat model of IUGR and assess the neuroprotective effect of a nutritional supplementation with bovine Lf during pregnancy and lactation on this model. Methods: A model of 50% gestational caloric restriction (CR) was used. Three groups were designed, and pregnant rats had either ad libitum access to food (control group, CTL) or 50% of the controls' intake (restricted group, IUGR). The diet was isocaloric and supplemented with bovine Lf for the caloric restricted dams (restricted-Lf, IUGR_Lf). At postnatal day 7 and 21, advanced ex-vivo diffusion MRI techniques at 9.4T were used to investigate brain cortical and white matter microstructure. Further, genes and proteins involved in structure (synaptophysin, MBP), microglia (Iba-1), metabolism (MCT2, βCaMKII) and apoptosis (Bcl-2) were analyzed in the cortex and striatum. In the cortex, the number of parvalbumin immunoreactive interneurons and their perineuronal nets were quantified. Behavioral tests were performed at P31. Results: Effects of the CR were significant in the cortex and striatum with reduction of synaptophysin (marker of synaptogenesis) at P7 and MBP (marker of myelin) at P21 in the cortex. Indeed, MCT2 (energy metabolism), Bcl-2 (anti-apoptotic protein) and βCaMKII (synapse activity) expressions were reduced in IUGR groups at P7. In the striatum NG2 (marker of oligodendrocyte precursor cells) and Bcl-2 at P7 as well as βCaMKII at P21 were decreased following IUGR and restored by Lf. Cortical microstructure was impaired following CR with partial effect of Lf. Lf prevented oxidative stress induced parvalbumin interneurons impairments whereas striatum and external capsule showed alterations in microstructure depicted by diffusion MRI, which were also partially reversed by Lf. Discussion and Conclusion: The model of 50% caloric restriction induced mild impairment partially reversed by nutritional intervention using Lf during pregnancy and lactation.
Collapse
Affiliation(s)
- Yohan van de Looij
- Division of Child Development and Growth, Department of Pediatrics, School of Medicine, University of Geneva, Geneva, Switzerland
- Laboratory for Functional and Metabolic Imaging, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Camille Larpin
- Division of Child Development and Growth, Department of Pediatrics, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Jan-Harry Cabungcal
- Department of Psychiatry, Centre for Psychiatric Neuroscience, Lausanne University Hospital, Lausanne, Switzerland
| | - Eduardo F. Sanches
- Division of Child Development and Growth, Department of Pediatrics, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Audrey Toulotte
- Division of Child Development and Growth, Department of Pediatrics, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Kim Q. Do
- Department of Psychiatry, Centre for Psychiatric Neuroscience, Lausanne University Hospital, Lausanne, Switzerland
| | - Stéphane V. Sizonenko
- Division of Child Development and Growth, Department of Pediatrics, School of Medicine, University of Geneva, Geneva, Switzerland
- *Correspondence: Stéphane V. Sizonenko
| |
Collapse
|
49
|
McClatchy DB, Yu NK, Martínez-Bartolomé S, Patel R, Pelletier AR, Lavalle-Adam M, Powell SB, Roberto M, Yates JR. Structural Analysis of Hippocampal Kinase Signal Transduction. ACS Chem Neurosci 2018; 9:3072-3085. [PMID: 30053369 PMCID: PMC6374210 DOI: 10.1021/acschemneuro.8b00284] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Kinases are a major clinical target for human diseases. Identifying the proteins that interact with kinases in vivo will provide information on unreported substrates and will potentially lead to more specific methods for therapeutic kinase regulation. Here, endogenous immunoprecipitations of evolutionally distinct kinases (i.e., Akt, ERK2, and CAMK2) from rodent hippocampi were analyzed by mass spectrometry to generate three highly confident kinase protein-protein interaction networks. Proteins of similar function were identified in the networks, suggesting a universal model for kinase signaling complexes. Protein interactions were observed between kinases with reported symbiotic relationships. The kinase networks were significantly enriched in genes associated with specific neurodevelopmental disorders providing novel structural connections between these disease-associated genes. To demonstrate a functional relationship between the kinases and the network, pharmacological manipulation of Akt in hippocampal slices was shown to regulate the activity of potassium/sodium hyperpolarization-activated cyclic nucleotide-gated channel(HCN1), which was identified in the Akt network. Overall, the kinase protein-protein interaction networks provide molecular insight of the spatial complexity of in vivo kinase signal transduction which is required to achieve the therapeutic potential of kinase manipulation in the brain.
Collapse
Affiliation(s)
- Daniel B McClatchy
- Department of Molecular Medicine , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Nam-Kyung Yu
- Department of Molecular Medicine , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Salvador Martínez-Bartolomé
- Department of Molecular Medicine , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Reesha Patel
- Department of Neuroscience , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Alexander R Pelletier
- Department of Biochemistry, Microbiology and Immunology and Ottawa Institute of Systems Biology , University of Ottawa , Ottawa , ON K1H 8M5 , Canada
| | - Mathieu Lavalle-Adam
- Department of Biochemistry, Microbiology and Immunology and Ottawa Institute of Systems Biology , University of Ottawa , Ottawa , ON K1H 8M5 , Canada
| | - Susan B Powell
- Department of Psychiatry , UCSD , La Jolla , California 92093 , United States
| | - Marisa Roberto
- Department of Neuroscience , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - John R Yates
- Department of Molecular Medicine , The Scripps Research Institute , La Jolla , California 92037 , United States
| |
Collapse
|
50
|
Direct effects of Ca2+/calmodulin on actin filament formation. Biochem Biophys Res Commun 2018; 506:355-360. [DOI: 10.1016/j.bbrc.2018.07.159] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 07/31/2018] [Indexed: 01/06/2023]
|