1
|
Virga DM, Hamilton S, Osei B, Morgan A, Kneis P, Zamponi E, Park NJ, Hewitt VL, Zhang D, Gonzalez KC, Russell FM, Grahame Hardie D, Prudent J, Bloss E, Losonczy A, Polleux F, Lewis TL. Activity-dependent compartmentalization of dendritic mitochondria morphology through local regulation of fusion-fission balance in neurons in vivo. Nat Commun 2024; 15:2142. [PMID: 38459070 PMCID: PMC10923867 DOI: 10.1038/s41467-024-46463-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 02/27/2024] [Indexed: 03/10/2024] Open
Abstract
Neuronal mitochondria play important roles beyond ATP generation, including Ca2+ uptake, and therefore have instructive roles in synaptic function and neuronal response properties. Mitochondrial morphology differs significantly between the axon and dendrites of a given neuronal subtype, but in CA1 pyramidal neurons (PNs) of the hippocampus, mitochondria within the dendritic arbor also display a remarkable degree of subcellular, layer-specific compartmentalization. In the dendrites of these neurons, mitochondria morphology ranges from highly fused and elongated in the apical tuft, to more fragmented in the apical oblique and basal dendritic compartments, and thus occupy a smaller fraction of dendritic volume than in the apical tuft. However, the molecular mechanisms underlying this striking degree of subcellular compartmentalization of mitochondria morphology are unknown, precluding the assessment of its impact on neuronal function. Here, we demonstrate that this compartment-specific morphology of dendritic mitochondria requires activity-dependent, Ca2+ and Camkk2-dependent activation of AMPK and its ability to phosphorylate two direct effectors: the pro-fission Drp1 receptor Mff and the recently identified anti-fusion, Opa1-inhibiting protein, Mtfr1l. Our study uncovers a signaling pathway underlying the subcellular compartmentalization of mitochondrial morphology in dendrites of neurons in vivo through spatially precise and activity-dependent regulation of mitochondria fission/fusion balance.
Collapse
Affiliation(s)
- Daniel M Virga
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Stevie Hamilton
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Bertha Osei
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Abigail Morgan
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Neuroscience, Biochemistry & Molecular Biology, Oklahoma University Health Science Campus, Oklahoma City, OK, USA
| | - Parker Kneis
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Neuroscience, Biochemistry & Molecular Biology, Oklahoma University Health Science Campus, Oklahoma City, OK, USA
| | - Emiliano Zamponi
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Natalie J Park
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Victoria L Hewitt
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - David Zhang
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Kevin C Gonzalez
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Fiona M Russell
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, Scotland, UK
| | - D Grahame Hardie
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, Scotland, UK
| | - Julien Prudent
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Hills Road, CB2 0XY, Cambridge, UK
| | - Erik Bloss
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Attila Losonczy
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Franck Polleux
- Department of Neuroscience, Columbia University, New York, NY, USA.
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA.
| | - Tommy L Lewis
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
- Neuroscience, Biochemistry & Molecular Biology, Oklahoma University Health Science Campus, Oklahoma City, OK, USA.
| |
Collapse
|
2
|
Ohtsuka S, Miyai Y, Mima H, Magari M, Chiba Y, Suizu F, Sakagami H, Ueno M, Tokumitsu H. Transcriptional, biochemical, and immunohistochemical analyses of CaMKKβ/2 splice variants that co-localize with CaMKIV in spermatids. Cell Calcium 2024; 117:102820. [PMID: 37979343 DOI: 10.1016/j.ceca.2023.102820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/18/2023] [Accepted: 10/29/2023] [Indexed: 11/20/2023]
Abstract
Ca2+/calmodulin-dependent protein kinase kinase (CaMKK) phosphorylates and activates downstream protein kinases, including CaMKI, CaMKIV, PKB/Akt, and AMPK; thus, regulates various Ca2+-dependent physiological and pathophysiological pathways. Further, CaMKKβ/2 in mammalian species comprises multiple alternatively spliced variants; however, their functional differences or redundancy remain unclear. In this study, we aimed to characterize mouse CaMKKβ/2 splice variants (CaMKKβ-3 and β-3x). RT-PCR analyses revealed that mouse CaMKKβ-1, consisting of 17 exons, was predominantly expressed in the brain; whereas, mouse CaMKKβ-3 and β-3x, lacking exon 16 and exons 14/16, respectively, were primarily expressed in peripheral tissues. At the protein level, the CaMKKβ-3 or β-3x variants showed high expression levels in mouse cerebrum and testes. This was consistent with the localization of CaMKKβ-3/-3x in spermatids in seminiferous tubules, but not the localization of CaMKKβ-1. We also observed the co-localization of CaMKKβ-3/-3x with a target kinase, CaMKIV, in elongating spermatids. Biochemical characterization further revealed that CaMKKβ-3 exhibited Ca2+/CaM-induced kinase activity similar to CaMKKβ-1. Conversely, we noted that CaMKKβ-3x impaired Ca2+/CaM-binding ability, but exhibited significantly weak autonomous activity (approximately 500-fold lower than CaMKKβ-1 or β-3) due to the absence of C-terminal of the catalytic domain and a putative residue (Ile478) responsible for the kinase autoinhibition. Nevertheless, CaMKKβ-3x showed the ability to phosphorylate downstream kinases, including CaMKIα, CaMKIV, and AMPKα in transfected cells comparable to CaMKKβ-1 and β-3. Collectively, CaMKKβ-3/-3x were identified as functionally active and could be bona fide CaMKIV-kinases in testes involved in the activation of the CaMKIV cascade in spermatids, resulting in the regulation of spermiogenesis.
Collapse
Affiliation(s)
- Satomi Ohtsuka
- Applied Cell Biology, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
| | - Yumi Miyai
- Inflammation Pathology, Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa, 761-0793, Japan
| | - Hiroyuki Mima
- Applied Cell Biology, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
| | - Masaki Magari
- Applied Cell Biology, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
| | - Yoichi Chiba
- Inflammation Pathology, Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa, 761-0793, Japan
| | - Futoshi Suizu
- Oncology Pathology, Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa, 761-0793, Japan
| | - Hiroyuki Sakagami
- Department of Anatomy, Kitasato University School of Medicine, Kanagawa, 252-0374, Japan
| | - Masaki Ueno
- Inflammation Pathology, Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa, 761-0793, Japan
| | - Hiroshi Tokumitsu
- Applied Cell Biology, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan.
| |
Collapse
|
3
|
Ageta-Ishihara N, Takemoto-Kimura S, Kondo Y, Okamura M, Bito H. Lipidation states orchestrate CLICK-III/CaMKIγ's stepwise association with Golgi and rafts-enriched membranes and specify its functional coupling to STEF-Rac1-dependent neurite extension. Front Cell Neurosci 2023; 17:1204302. [PMID: 37601281 PMCID: PMC10435254 DOI: 10.3389/fncel.2023.1204302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/13/2023] [Indexed: 08/22/2023] Open
Abstract
CLICK-III/CaMKIγ is a lipid-anchored neuronal isoform of multifunctional Ca2+/calmodulin-dependent protein kinases, which mediates BDNF-dependent dendritogenesis in cultured cortical neurons. We found that two distinct lipidation states of CaMKIγ, namely, prenylation and palmitoylation, controlled its association with detergent-resistant microdomains in the dendrites and were essential for its dendritogenic activity. However, the impact of each lipid modification on membrane targeting/trafficking and how it specifies functional coupling leading to polarized changes in neuronal morphology are not clear. Here, we show that prenylation induces membrane anchoring of CaMKIγ, permitting access to the Golgi apparatus, and a subsequent palmitoylation facilitates association with cholesterol-enriched lipid microdomains or lipid rafts, in particular at the Golgi. To specifically test the role of palmitoylated CaMKγ in neurite extension, we identified and took advantage of a cell system, PC12, which, unlike neurons, conveniently lacked CaMKIγ and was deficient in the activity-dependent release of a neuritogenic growth factor while possessing the ability to activate polarized rafts signaling for morphogenesis. This system allowed us to rigorously demonstrate that an activity-dependent, lipid rafts-restricted Rac activation leading to neuritogenesis could be functionally rescued by dually lipidated CaMKIγ expression, revealing that not only prenylation but also palmitoylation is essential for CaMKIγ to activate a compartmentalized STEF-Rac1 pathway. These results shed light on the significance of recruiting prenylated and palmitoylated CaMKIγ into the coalescing signalosomes at lipid rafts together with Rac1 and its specific GEF and STEF and forming a compartmentalized Ca2+ signaling pathway that underlies activity-dependent neuritogenesis and morphogenesis during axodendritic polarization critical for brain development and circuitogenesis.
Collapse
Affiliation(s)
- Natsumi Ageta-Ishihara
- Department of Biomolecular Science, Faculty of Science, Toho University, Funabashi, Japan
- PRESTO, Japan Science and Technology Agency, Kawaguchi, Japan
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Bunkyo, Japan
| | - Sayaka Takemoto-Kimura
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Bunkyo, Japan
- Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Yayoi Kondo
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Bunkyo, Japan
| | - Michiko Okamura
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Bunkyo, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Bunkyo, Japan
| |
Collapse
|
4
|
Zhao Y, Liu J, Liu S, Yang P, Liang Y, Ma J, Mao S, Sun C, Yang Y. Fibroblast exosomal TFAP2C induced by chitosan oligosaccharides promotes peripheral axon regeneration via the miR-132-5p/CAMKK1 axis. Bioact Mater 2023; 26:249-263. [PMID: 36936807 PMCID: PMC10020534 DOI: 10.1016/j.bioactmat.2023.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 03/03/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Chitosan and its degradation product, oligosaccharides, have been shown to facilitate peripheral nerve regeneration. However, the underlying mechanisms are not well understood. In this study, we analyzed the protein expression profiles in sciatic nerves after injury using proteomics. A group of proteins related to exosome packaging and transport is up-regulated by chitosan oligosaccharides (COS), implying that exosomes are involved in COS-induced peripheral nerve regeneration. In fact, exosomes derived from fibroblasts (f-EXOs) treated with COS significantly promoted axon extension and regeneration. Exosomal protein identification and functional studies, revealed that TFAP2C is a key factor in neurite outgrowth induced by COS-f-EXOs. Furthermore, we showed that TFAP2C targets the pri-miRNA-132 gene and represses miR-132-5p expression in dorsal root ganglion neurons. Camkk1 is a downstream substrate of miR-132-5p that positively affects axon extension. In rats, miR-132-5p antagomir stimulates CAMKK1 expression and improves axon regeneration and functional recovery in sciatic nerves after injury. Our data reveal the mechanism for COS in axon regeneration, that is COS induce fibroblasts to produce TFAP2C-enriched EXOs, which are then transferred into axons to promote axon regeneration via miR-132-5p/CAMKK1. Moreover, these results show a new facet of fibroblasts in axon regeneration in peripheral nerves.
Collapse
Affiliation(s)
- Yahong Zhao
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Jina Liu
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Sha Liu
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Panpan Yang
- School of Medicine, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Yunyun Liang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Jinyu Ma
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Susu Mao
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Cheng Sun
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| |
Collapse
|
5
|
Gaff J, Octaviana F, Jackaman C, Kamerman P, Papadimitriou J, Lee S, Mountford J, Price P. Expression in skin biopsies supports genetic evidence linking CAMKK2, P2X7R and P2X4R with HIV-associated sensory neuropathy. J Neurovirol 2023; 29:241-251. [PMID: 37166584 PMCID: PMC10404215 DOI: 10.1007/s13365-023-01134-2] [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: 12/19/2022] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 05/12/2023]
Abstract
HIV-associated sensory neuropathy (HIV-SN) affects 14-38% of HIV+ individuals stable on therapy with no neurotoxic drugs. Polymorphisms in CAMKK2, P2X7R and P2X4R associated with altered risk of HIV-SN in Indonesian and South African patients. The role of CaMKK2 in neuronal repair makes this an attractive candidate, but a direct role for any protein is predicated on expression in affected tissues. Here, we describe expression of CaMKK2, P2X7R and P2X4R proteins in skin biopsies from the lower legs of HIV+ Indonesians with and without HIV-SN, and healthy controls (HC). HIV-SN was diagnosed using the Brief Peripheral Neuropathy Screen. Biopsies were stained to detect protein gene product 9.5 on nerve fibres and CaMKK2, P2X7R or P2X4R, and were examined using 3-colour sequential scanning confocal microscopy. Intraepidermal nerve fibre densities (IENFD) were lower in HIV+ donors than HC and correlated directly with nadir CD4 T-cell counts (r = 0.69, p = 0.004). However, IENFD counts were similar in HIV-SN+ and HIV-SN- donors (p = 0.19) and so did not define neuropathy. CaMKK2+ cells were located close to dermal and epidermal nerve fibres and were rare in HC and HIV-SN- donors, consistent with a role for the protein in nerve damage and/or repair. P2X7R was expressed by cells in blood vessels of HIV-SN- donors, but rarely in HC or HIV-SN+ donors. P2X4R expression by cells in the epidermal basal layer appeared greatest in HIV-SN+ donors. Overall, the differential expression of CaMKK2, P2X7R and P2X4R supports the genetic evidence of a role for these proteins in HIV-SN.
Collapse
Affiliation(s)
- Jessica Gaff
- Curtin Medical School, Curtin University, Bentley, 6102, Australia
| | - Fitri Octaviana
- Neurology Department, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Neurology Department, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Connie Jackaman
- Curtin Medical School, Curtin University, Bentley, 6102, Australia
- Curtin Health Innovation Research Institute, Bentley, Australia
| | - Peter Kamerman
- School of Physiology, University of Witwatersrand, Johannesburg, South Africa
| | | | - Silvia Lee
- Curtin Medical School, Curtin University, Bentley, 6102, Australia
- Department of Microbiology, Pathwest Laboratory Medicine, Perth, Australia
| | | | - Patricia Price
- Curtin Medical School, Curtin University, Bentley, 6102, Australia.
- Neurology Department, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.
- Curtin Health Innovation Research Institute, Bentley, Australia.
- School of Physiology, University of Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
6
|
Virga DM, Hamilton S, Osei B, Morgan A, Zamponi E, Park NJ, Hewitt VL, Zhang D, Gonzalez KC, Bloss E, Polleux F, Lewis TL. Activity-dependent subcellular compartmentalization of dendritic mitochondria structure in CA1 pyramidal neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.25.534233. [PMID: 36993655 PMCID: PMC10055421 DOI: 10.1101/2023.03.25.534233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Neuronal mitochondria play important roles beyond ATP generation, including Ca2+ uptake, and therefore have instructive roles in synaptic function and neuronal response properties. Mitochondrial morphology differs significantly in the axon and dendrites of a given neuronal subtype, but in CA1 pyramidal neurons (PNs) of the hippocampus, mitochondria within the dendritic arbor also display a remarkable degree of subcellular, layer-specific compartmentalization. In the dendrites of these neurons, mitochondria morphology ranges from highly fused and elongated in the apical tuft, to more fragmented in the apical oblique and basal dendritic compartments, and thus occupy a smaller fraction of dendritic volume than in the apical tuft. However, the molecular mechanisms underlying this striking degree of subcellular compartmentalization of mitochondria morphology are unknown, precluding the assessment of its impact on neuronal function. Here, we demonstrate that this compartment-specific morphology of dendritic mitochondria requires activity-dependent, Camkk2-dependent activation of AMPK and its ability to phosphorylate two direct effectors: the pro-fission Drp1 receptor Mff and the recently identified anti-fusion, Opa1-inhibiting protein, Mtfr1l. Our study uncovers a new activity-dependent molecular mechanism underlying the extreme subcellular compartmentalization of mitochondrial morphology in dendrites of neurons in vivo through spatially precise regulation of mitochondria fission/fusion balance.
Collapse
Affiliation(s)
- Daniel M. Virga
- Department of Neuroscience, Columbia Medical School, New York, NY- USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY- USA
| | - Stevie Hamilton
- Department of Neuroscience, Columbia Medical School, New York, NY- USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY- USA
| | - Bertha Osei
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Abigail Morgan
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Neuroscience, Oklahoma University Health Science Campus, Oklahoma City, OK, USA
| | - Emiliano Zamponi
- Department of Neuroscience, Columbia Medical School, New York, NY- USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY- USA
| | - Natalie J. Park
- Department of Neuroscience, Columbia Medical School, New York, NY- USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY- USA
| | - Victoria L. Hewitt
- Department of Neuroscience, Columbia Medical School, New York, NY- USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY- USA
| | - David Zhang
- Department of Neuroscience, Columbia Medical School, New York, NY- USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY- USA
| | - Kevin C. Gonzalez
- Department of Neuroscience, Columbia Medical School, New York, NY- USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY- USA
| | - Erik Bloss
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Franck Polleux
- Department of Neuroscience, Columbia Medical School, New York, NY- USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY- USA
| | - Tommy L. Lewis
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Neuroscience, Oklahoma University Health Science Campus, Oklahoma City, OK, USA
| |
Collapse
|
7
|
Molecular Mechanisms Underlying Ca2+/Calmodulin-Dependent Protein Kinase Kinase Signal Transduction. Int J Mol Sci 2022; 23:ijms231911025. [PMID: 36232320 PMCID: PMC9570080 DOI: 10.3390/ijms231911025] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 12/03/2022] Open
Abstract
Ca2+/calmodulin-dependent protein kinase kinase (CaMKK) is the activating kinase for multiple downstream kinases, including CaM-kinase I (CaMKI), CaM-kinase IV (CaMKIV), protein kinase B (PKB/Akt), and 5′AMP-kinase (AMPK), through the phosphorylation of their activation-loop Thr residues in response to increasing the intracellular Ca2+ concentration, as CaMKK itself is a Ca2+/CaM-dependent enzyme. The CaMKK-mediated kinase cascade plays important roles in a number of Ca2+-dependent pathways, such as neuronal morphogenesis and plasticity, transcriptional activation, autophagy, and metabolic regulation, as well as in pathophysiological pathways, including cancer progression, metabolic syndrome, and mental disorders. This review focuses on the molecular mechanism underlying CaMKK-mediated signal transduction in normal and pathophysiological conditions. We summarize the current knowledge of the structural, functional, and physiological properties of the regulatory kinase, CaMKK, and the development and application of its pharmacological inhibitors.
Collapse
|
8
|
Suzuki Y, Kurata T, Koide T, Okada I, Nakajima N, Imaizumi Y, Yamamura H. Local Ca<sup>2+</sup> Signals within Caveolae Cause Nuclear Translocation of CaMK1α in Mouse Vascular Smooth Muscle Cells. Biol Pharm Bull 2022; 45:1354-1363. [DOI: 10.1248/bpb.b22-00319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yoshiaki Suzuki
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Tomo Kurata
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Tsukasa Koide
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Itsuki Okada
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Nanami Nakajima
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Yuji Imaizumi
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Hisao Yamamura
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| |
Collapse
|
9
|
Han JH, Kim YK, Kim H, Lee J, Oh MJ, Kim SB, Kim M, Kim KH, Yoon HJ, Lee MS, Minna JD, White MA, Kim HS. Snail acetylation by autophagy-derived acetyl-coenzyme A promotes invasion and metastasis of KRAS-LKB1 co-mutated lung cancer cells. CANCER COMMUNICATIONS (LONDON, ENGLAND) 2022; 42:716-749. [PMID: 35838183 PMCID: PMC9395322 DOI: 10.1002/cac2.12332] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/21/2022] [Accepted: 06/26/2022] [Indexed: 11/29/2022]
Abstract
Background Autophagy is elevated in metastatic tumors and is often associated with active epithelial‐to‐mesenchymal transition (EMT). However, the extent to which EMT is dependent on autophagy is largely unknown. This study aimed to identify the mechanisms by which autophagy facilitates EMT. Methods We employed a liquid chromatography‐based metabolomic approach with kirsten rat sarcoma viral oncogene (KRAS) and liver kinase B1 (LKB1) gene co‐mutated (KL) cells that represent an autophagy/EMT‐coactivated invasive lung cancer subtype for the identification of metabolites linked to autophagy‐driven EMT activation. Molecular mechanisms of autophagy‐driven EMT activation were further investigated by quantitative real‐time polymerase chain reaction (qRT‐PCR), Western blotting analysis, immunoprecipitation, immunofluorescence staining, and metabolite assays. The effects of chemical and genetic perturbations on autophagic flux were assessed by two orthogonal approaches: microtubule‐associated protein 1A/1B‐light chain 3 (LC3) turnover analysis by Western blotting and monomeric red fluorescent protein‐green fluorescent protein (mRFP‐GFP)‐LC3 tandem fluorescent protein quenching assay. Transcription factor EB (TFEB) activity was measured by coordinated lysosomal expression and regulation (CLEAR) motif‐driven luciferase reporter assay. Experimental metastasis (tail vein injection) mouse models were used to evaluate the impact of calcium/calmodulin‐dependent protein kinase kinase 2 (CAMKK2) or ATP citrate lyase (ACLY) inhibitors on lung metastasis using IVIS luciferase imaging system. Results We found that autophagy in KL cancer cells increased acetyl‐coenzyme A (acetyl‐CoA), which facilitated the acetylation and stabilization of the EMT‐inducing transcription factor Snail. The autophagy/acetyl‐CoA/acetyl‐Snail axis was further validated in tumor tissues and in autophagy‐activated pancreatic cancer cells. TFEB acetylation in KL cancer cells sustained pro‐metastatic autophagy in a mammalian target of rapamycin complex 1 (mTORC1)‐independent manner. Pharmacological inhibition of this axis via CAMKK2 inhibitors or ACLY inhibitors consistently reduced the metastatic capacity of KL cancer cells in vivo. Conclusions This study demonstrates that autophagy‐derived acetyl‐CoA promotes Snail acetylation and thereby facilitates invasion and metastasis of KRAS‐LKB1 co‐mutated lung cancer cells and that inhibition of the autophagy/acetyl‐CoA/acetyl‐Snail axis using CAMKK2 or ACLY inhibitors could be a potential therapeutic strategy to suppress metastasis of KL lung cancer.
Collapse
Affiliation(s)
- Jang Hee Han
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.,Department of Medical Science, Yonsei University Graduate School, Seoul, 03722, Korea.,Department of Urology, Seoul National University Hospital, Seoul, 03722, Korea
| | - Yong Keon Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.,Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Hakhyun Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.,Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Jooyoung Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.,Checkmate Therapeutics Inc., Seoul, 07207, Korea
| | - Myung Joon Oh
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.,Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Sang Bum Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Minjee Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.,Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Kook Hwan Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Hyun Ju Yoon
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.,Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Myung-Shik Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - John D Minna
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
| | - Michael A White
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
| | - Hyun Seok Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.,Checkmate Therapeutics Inc., Seoul, 07207, Korea.,Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
| |
Collapse
|
10
|
Shang W, Dai Z, Zhang J, Shen F, Sui N, Liang J. Embryonic opioid exposure impairs inhibitory transmission of striatum in day‐old chicks. Dev Psychobiol 2022; 64:e22273. [DOI: 10.1002/dev.22273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 03/05/2022] [Accepted: 03/13/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Wen Shang
- CAS Key Laboratory of Mental Health Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| | - Zhonghua Dai
- CAS Key Laboratory of Mental Health Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| | - Jianjun Zhang
- CAS Key Laboratory of Mental Health Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| | - Fang Shen
- CAS Key Laboratory of Mental Health Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| | - Nan Sui
- CAS Key Laboratory of Mental Health Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| | - Jing Liang
- CAS Key Laboratory of Mental Health Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
- Sino‐Danish Center for Education and Research University of Chinese Academy of Sciences Beijing China
| |
Collapse
|
11
|
Guo S, Moore RM, Charlesworth MC, Johnson KL, Spinner RJ, Windebank AJ, Wang H. The proteome of distal nerves: implication in delayed repair and poor functional recovery. Neural Regen Res 2022; 17:1998-2006. [PMID: 35142689 PMCID: PMC8848594 DOI: 10.4103/1673-5374.335159] [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] [Indexed: 11/04/2022] Open
Abstract
Chronic denervation is one of the key factors that affect nerve regeneration. Chronic axotomy deteriorates the distal nerve stump, causes protein changes, and renders the microenvironment less permissive for regeneration. Some of these factors/proteins have been individually studied. To better delineate the comprehensive protein expression profiles and identify proteins that contribute to or are associated with this detrimental effect, we carried out a proteomic analysis of the distal nerve using an established delayed rat sciatic nerve repair model. Four rats that received immediate repair after sciatic nerve transection served as control, whereas four rats in the experimental group (chronic denervation) had their sciatic nerve repaired after a 12-week delay. All the rats were sacrificed after 16 weeks to harvest the distal nerves for extracting proteins. Twenty-five micrograms of protein from each sample were fractionated in SDS-PAGE gels. NanoLC-MS/MS analysis was applied to the gels. Protein expression levels of nerves on the surgery side were compared to those on the contralateral side. Any protein with a P value of less than 0.05 and a fold change of 4 or higher was deemed differentially expressed. All the differentially expressed proteins in both groups were further stratified according to the biological processes. A PubMed search was also conducted to identify the differentially expressed proteins that have been reported to be either beneficial or detrimental to nerve regeneration. Ingenuity Pathway Analysis (IPA) software was used for pathway analysis. The results showed that 709 differentially expressed proteins were identified in the delayed repair group, with a bigger proportion of immune and inflammatory process-related proteins and a smaller proportion of proteins related to axon regeneration and lipid metabolism in comparison to the control group where 478 differentially expressed proteins were identified. The experimental group also had more beneficial proteins that were downregulated and more detrimental proteins that were upregulated. IPA revealed that protective pathways such as LXR/RXR, acute phase response, RAC, ERK/MAPK, CNTF, IL-6, and FGF signaling were inhibited in the delayed repair group, whereas three detrimental pathways, including the complement system, PTEN, and apoptosis signaling, were activated. An available database of the adult rodent sciatic nerve was used to assign protein changes to specific cell types. The poor regeneration seen in the delayed repair group could be associated with the down-regulation of beneficial proteins and up-regulation of detrimental proteins. The proteins and pathways identified in this study may offer clues for future studies to identify therapeutic targets.
Collapse
Affiliation(s)
- Song Guo
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Raymond M Moore
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | | | | | - Robert J Spinner
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Huan Wang
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
12
|
Nakagawa-Tamagawa N, Kirino E, Sugao K, Nagata H, Tagawa Y. Involvement of Calcium-Dependent Pathway and β Subunit-Interaction in Neuronal Migration and Callosal Projection Deficits Caused by the Cav1.2 I1166T Mutation in Developing Mouse Neocortex. Front Neurosci 2021; 15:747951. [PMID: 34955712 PMCID: PMC8692569 DOI: 10.3389/fnins.2021.747951] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction: Gain-of-function mutations in the L-type Ca2+ channel Cav1.2 cause Timothy syndrome (TS), a multisystem disorder associated with neurologic symptoms, including autism spectrum disorder (ASD), seizures, and intellectual disability. Cav1.2 plays key roles in neural development, and its mutation can affect brain development and connectivity through Ca2+-dependent and -independent mechanisms. Recently, a gain-of-function mutation, I1166T, in Cav1.2 was identified in patients with TS-like disorder. Its channel properties have been analyzed in vitro but in vivo effects of this mutation on brain development remain unexplored. Methods:In utero electroporation was performed on ICR mice at embryonic day 15 to express GFP, wild-type, and mutant Cav1.2 channels into cortical layer 2/3 excitatory neurons in the primary somatosensory area. The brain was fixed at postnatal days 14–16, sliced, and scanned using confocal microscopy. Neuronal migration of electroporated neurons was examined in the cortex of the electroporated hemisphere, and callosal projection was examined in the white matter and contralateral hemisphere. Results: Expression of the I1166T mutant in layer 2/3 neurons caused migration deficits in approximately 20% of electroporated neurons and almost completely diminished axonal arborization in the contralateral hemisphere. Axonal projection in the white matter was not affected. We introduced second mutations onto Cav1.2 I1166T; L745P mutation blocks Ca2+ influx through Cav1.2 channels and inhibits the Ca2+-dependent pathway, and the W440A mutation blocks the interaction of the Cav1.2 α1 subunit to the β subunit. Both second mutations recovered migration and projection. Conclusion: This study demonstrated that the Cav1.2 I1166T mutation could affect two critical steps during cerebrocortical development, migration and axonal projection, in the mouse brain. This is mediated through Ca2+-dependent pathway downstream of Cav1.2 and β subunit-interaction.
Collapse
Affiliation(s)
- Nao Nakagawa-Tamagawa
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan.,Laboratory for Molecular Analysis of Higher Brain Function, RIKEN Center for Brain Science, Saitama, Japan
| | - Emi Kirino
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Kohtaroh Sugao
- Platform Technology Research Unit, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Hidetaka Nagata
- Platform Technology Research Unit, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Yoshiaki Tagawa
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
13
|
Alçada-Morais S, Gonçalves N, Moreno-Juan V, Andres B, Ferreira S, Marques JM, Magalhães J, Rocha JMM, Xu X, Partidário M, Cunha RA, López-Bendito G, Rodrigues RJ. Adenosine A2A Receptors Contribute to the Radial Migration of Cortical Projection Neurons through the Regulation of Neuronal Polarization and Axon Formation. Cereb Cortex 2021; 31:5652-5663. [PMID: 34184030 DOI: 10.1093/cercor/bhab188] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 01/02/2023] Open
Abstract
Cortical interneurons born in the subpallium reach the cortex through tangential migration, whereas pyramidal cells reach their final position by radial migration. Purinergic signaling via P2Y1 receptors controls the migration of intermediate precursor cells from the ventricular zone to the subventricular zone. It was also reported that the blockade of A2A receptors (A2AR) controls the tangential migration of somatostatin+ interneurons. Here we found that A2AR control radial migration of cortical projection neurons. In A2AR-knockout (KO) mouse embryos or naïve mouse embryos exposed to an A2AR antagonist, we observed an accumulation of early-born migrating neurons in the lower intermediate zone at late embryogenesis. In utero knockdown of A2AR also caused an accumulation of neurons at the lower intermediate zone before birth. This entails the presently identified ability of A2AR to promote multipolar-bipolar transition and axon formation, critical for the transition of migrating neurons from the intermediate zone to the cortical plate. This effect seems to require extracellular ATP-derived adenosine since a similar accumulation of neurons at the lower intermediate zone was observed in mice lacking ecto-5'-nucleotidase (CD73-KO). These findings frame adenosine as a fine-tune regulator of the wiring of cortical inhibitory and excitatory networks.
Collapse
Affiliation(s)
- Sofia Alçada-Morais
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra 3030-789, Portugal
| | - Nélio Gonçalves
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal
| | | | - Belén Andres
- Instituto de Neurociencias, CSIC-UMH, San Juan de Alicante 03550, Spain
| | - Sofia Ferreira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra 3030-789, Portugal
| | - Joana M Marques
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal
| | - Joana Magalhães
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal
| | - João M M Rocha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal
| | - Xinli Xu
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra 3030-789, Portugal
| | - Matilde Partidário
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra 3004-504, Portugal
| | | | - Ricardo J Rodrigues
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra 3030-789, Portugal
| |
Collapse
|
14
|
Brief Report: Demographic and Genetic Associations With Markers of Small and Large Fiber Sensory Neuropathy in HIV Patients Treated Without Stavudine. J Acquir Immune Defic Syndr 2021; 85:612-616. [PMID: 32925363 DOI: 10.1097/qai.0000000000002503] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Neurotoxic antiretroviral therapy (ART) such as stavudine has been now replaced with safer therapies, reducing the prevalence of neuropathy from 34% to 15% in HIV+ Indonesians. However, it is unclear whether the residual cases display damage to small or large nerve fibers and whether both are influenced by known risk factors, including alleles of CAMKK2 associated with neuropathy in HIV patients. The encoded protein influences the growth and repair of nerve fibers. HIV-positive adults on ART for >12 months without exposure to stavudine were screened for neuropathy using the AIDS Clinical Trials Group Brief Peripheral Neuropathy Screen (BPNS). Large fiber neuropathy was assessed by nerve conduction (NC) and small fiber neuropathy using stimulated skin wrinkling (SSW) applied to the fingers. CAMKK2 alleles were assessed by TaqMan OpenArray technology. Neuropathy diagnoses were more common with SSW than BPNS (49/173 vs 26/185, χ; P = 0.0009), with poor alignment between these outcomes (P = 0.60). NC and BPNS diagnosed neuropathy at similar frequencies (29/151 vs 26/185; P = 0.12) and were aligned (P < 0.0001). In bivariate analyses, all diagnoses were associated with patients' age and persistent HIV replication, with minor effects from CD4 T-cell counts and time on ART. CAMKK2 alleles associated with neuropathy diagnosed with BPNS and SSW but not NC. Multivariable analyses confirmed the importance of age and HIV replication, with distinct CAMKK2 polymorphisms affecting BPNS and SSW. Paradoxically, height was protective against skin wrinkling. Overall the data link CAMKK2 genotypes with small rather than large fiber damage. SSW may reflect pathology distinct from that identified using BPNS.
Collapse
|
15
|
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
|
16
|
Peerboom C, Wierenga CJ. The postnatal GABA shift: A developmental perspective. Neurosci Biobehav Rev 2021; 124:179-192. [PMID: 33549742 DOI: 10.1016/j.neubiorev.2021.01.024] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/13/2021] [Accepted: 01/28/2021] [Indexed: 12/13/2022]
Abstract
GABA is the major inhibitory neurotransmitter that counterbalances excitation in the mature brain. The inhibitory action of GABA relies on the inflow of chloride ions (Cl-), which hyperpolarizes the neuron. In early development, GABA signaling induces outward Cl- currents and is depolarizing. The postnatal shift from depolarizing to hyperpolarizing GABA is a pivotal event in brain development and its timing affects brain function throughout life. Altered timing of the postnatal GABA shift is associated with several neurodevelopmental disorders. Here, we argue that the postnatal shift from depolarizing to hyperpolarizing GABA represents the final shift in a sequence of GABA shifts, regulating proliferation, migration, differentiation, and finally plasticity of developing neurons. Each developmental GABA shift ensures that the instructive role of GABA matches the circumstances of the developing network. Sensory input may be a crucial factor in determining proper timing of the postnatal GABA shift. A developmental perspective is necessary to interpret the full consequences of a mismatch between connectivity, activity and GABA signaling during brain development.
Collapse
Affiliation(s)
- Carlijn Peerboom
- Cell Biology, Neurobiology and Biophysics, Biology Department, Faculty of Science, Utrecht University, 3584 CH, Utrecht, the Netherlands
| | - Corette J Wierenga
- Cell Biology, Neurobiology and Biophysics, Biology Department, Faculty of Science, Utrecht University, 3584 CH, Utrecht, the Netherlands.
| |
Collapse
|
17
|
Wang R, Lin Q. Prolonged ketamine exposure induces enhanced excitatory GABAergic synaptic activity in the anterior cingulate cortex of neonatal rats. Neurosci Lett 2021; 745:135647. [PMID: 33444673 DOI: 10.1016/j.neulet.2021.135647] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/06/2021] [Indexed: 11/28/2022]
Abstract
Experimental studies have indicated that prolonged ketamine exposure in neonates at anesthetic doses causes neuronal apoptosis, which contributes to long-term impairments of learning and memory later in life. The neuronal excitotoxicity mediated by compensatory upregulation of N-methyl-d-aspartate receptors (NMDARs) is proposed to be the underlying mechanism. However, this view does not convincingly explain why excitotoxicity-related apoptotic injury develops selectively in immature neurons. We proposed that the GABAA receptors (GABAARs)-mediated excitatory synaptic signaling due to high expression of the Na+-K+-2Cl- co-transporter (NKCC1), occurring during the early neuronal development period, plays a distinct role in the susceptibility of immature neurons to ketamine-induced injury. Using whole-cell patch-clamp recordings from the forebrain slices containing the anterior cingulate cortex, we found that in vivo repeated ketamine administration significantly induced neuronal hyperexcitability in neonatal, but not adolescent, rats. Such hyperexcitability was accompanied by the increase both in GABAAR- and NMDAR-mediated synaptic transmissions. An interference with the NKCC1 by bumetanide treatment completely reversed these enhanced effects of ketamine exposure and blocked GABAAR-mediated postsynaptic current activity. Thus, these findings were significant as they showed, for the first time, that GABAAR-mediated excitatory action may contribute distinctly to neuronal excitotoxic effects of ketamine on immature neurons in the developing brain.
Collapse
Affiliation(s)
- Ruirui Wang
- Department of Psychology, The University of Texas at Arlington, TX, USA
| | - Qing Lin
- Department of Psychology, The University of Texas at Arlington, TX, USA.
| |
Collapse
|
18
|
Takata T, Araki S, Tsuchiya Y, Watanabe Y. Persulfide Signaling in Stress-Initiated Calmodulin Kinase Response. Antioxid Redox Signal 2020; 33:1308-1319. [PMID: 32460522 DOI: 10.1089/ars.2020.8138] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Significance: Calcium ion (Ca2+)/calmodulin (CaM)-dependent protein kinases (CaMKs) are activated by phosphorylation of a crucial threonine residue either by itself (CaMKII) or by upstream kinases, CaMK kinases (CaMKKs) (CaMKI and CaMKIV). CaMKs, present in most mammalian tissues, can phosphorylate many downstream targets, thereby regulating numerous cellular functions. Recent Advances: Aside from canonical post-translational modifications, cysteine-based redox switches in CaMKs affect their enzyme activities. In addition to reactive oxygen species (ROS) and reactive nitrogen species (RNS), reactive sulfur species (RSS) are also recognized as key signaling molecules, regulating protein function through polysulfidation, formation of polysulfides [-S-(S)n-H] on their reactive cysteine residues. To comprehend the biological significance of RSS signaling-related CaMK regulation, here we introduce a novel concept defining CaMKs as RSS targets in stress responses. The stress responses include an irreversible electrophile attack for CaMKI, inflammation for CaMKII, and endoplasmic reticulum stress for CaMKIV. Critical Issues: Development of various human diseases is associated with increased ROS, RNS, and RSS generation. Therefore, depending on specific pathophysiology, RSS could have very particular effects on CaMK functions. Future Directions: How multiple sources and mutual reactions of ROS, RNS, and RSS are coordinated is obscure. Elucidating the mechanisms through applications of enzymology, chemical biology, and mass spectrometry enables to uncover the complexities of redox regulation of CaMK cascades.
Collapse
Affiliation(s)
- Tsuyoshi Takata
- Department of Pharmacology, Showa Pharmaceutical University, Tokyo, Japan.,Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shoma Araki
- Department of Pharmacology, Showa Pharmaceutical University, Tokyo, Japan
| | - Yukihiro Tsuchiya
- Department of Pharmacology, Showa Pharmaceutical University, Tokyo, Japan
| | - Yasuo Watanabe
- Department of Pharmacology, Showa Pharmaceutical University, Tokyo, Japan
| |
Collapse
|
19
|
Bortolasci CC, Spolding B, Kidnapillai S, Connor T, Truong TT, Liu ZS, Panizzutti B, Richardson MF, Gray L, Berk M, Dean OM, Walder K. Transcriptional Effects of Psychoactive Drugs on Genes Involved in Neurogenesis. Int J Mol Sci 2020; 21:ijms21218333. [PMID: 33172123 PMCID: PMC7672551 DOI: 10.3390/ijms21218333] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022] Open
Abstract
Although neurogenesis is affected in several psychiatric diseases, the effects and mechanisms of action of psychoactive drugs on neurogenesis remain unknown and/or controversial. This study aims to evaluate the effects of psychoactive drugs on the expression of genes involved in neurogenesis. Neuronal-like cells (NT2-N) were treated with amisulpride (10 µM), aripiprazole (0.1 µM), clozapine (10 µM), lamotrigine (50 µM), lithium (2.5 mM), quetiapine (50 µM), risperidone (0.1 µM), or valproate (0.5 mM) for 24 h. Genome wide mRNA expression was quantified and analysed using gene set enrichment analysis, with the neurogenesis gene set retrieved from the Gene Ontology database and the Mammalian Adult Neurogenesis Gene Ontology (MANGO) database. Transcription factors that are more likely to regulate these genes were investigated to better understand the biological processes driving neurogenesis. Targeted metabolomics were performed using gas chromatography-mass spectrometry. Six of the eight drugs decreased the expression of genes involved in neurogenesis in both databases. This suggests that acute treatment with these psychoactive drugs negatively regulates the expression of genes involved in neurogenesis in vitro. SOX2 and three of its target genes (CCND1, BMP4, and DKK1) were also decreased after treatment with quetiapine. This can, at least in part, explain the mechanisms by which these drugs decrease neurogenesis at a transcriptional level in vitro. These results were supported by the finding of increased metabolite markers of mature neurons following treatment with most of the drugs tested, suggesting increased proportions of mature relative to immature neurons consistent with reduced neurogenesis.
Collapse
Affiliation(s)
- Chiara C. Bortolasci
- The Institute for Mental and Physical Health and Clinical Translation, Barwon Health, Deakin University, Geelong 3220, Australia; (B.S.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (B.P.); (L.G.); (M.B.); (O.M.D.); (K.W.)
- School of Medicine, Centre for Molecular and Medical Research, Deakin University, Geelong 3220, Australia;
- Correspondence:
| | - Briana Spolding
- The Institute for Mental and Physical Health and Clinical Translation, Barwon Health, Deakin University, Geelong 3220, Australia; (B.S.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (B.P.); (L.G.); (M.B.); (O.M.D.); (K.W.)
- School of Medicine, Centre for Molecular and Medical Research, Deakin University, Geelong 3220, Australia;
| | - Srisaiyini Kidnapillai
- School of Medicine, Centre for Molecular and Medical Research, Deakin University, Geelong 3220, Australia;
| | - Timothy Connor
- The Institute for Mental and Physical Health and Clinical Translation, Barwon Health, Deakin University, Geelong 3220, Australia; (B.S.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (B.P.); (L.G.); (M.B.); (O.M.D.); (K.W.)
- School of Medicine, Centre for Molecular and Medical Research, Deakin University, Geelong 3220, Australia;
| | - Trang T.T. Truong
- The Institute for Mental and Physical Health and Clinical Translation, Barwon Health, Deakin University, Geelong 3220, Australia; (B.S.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (B.P.); (L.G.); (M.B.); (O.M.D.); (K.W.)
- School of Medicine, Centre for Molecular and Medical Research, Deakin University, Geelong 3220, Australia;
| | - Zoe S.J. Liu
- The Institute for Mental and Physical Health and Clinical Translation, Barwon Health, Deakin University, Geelong 3220, Australia; (B.S.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (B.P.); (L.G.); (M.B.); (O.M.D.); (K.W.)
- School of Medicine, Centre for Molecular and Medical Research, Deakin University, Geelong 3220, Australia;
| | - Bruna Panizzutti
- The Institute for Mental and Physical Health and Clinical Translation, Barwon Health, Deakin University, Geelong 3220, Australia; (B.S.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (B.P.); (L.G.); (M.B.); (O.M.D.); (K.W.)
- School of Medicine, Centre for Molecular and Medical Research, Deakin University, Geelong 3220, Australia;
| | - Mark F. Richardson
- School of Life and Environmental Sciences, Genomics Centre, Deakin University, Geelong 3220, Australia;
| | - Laura Gray
- The Institute for Mental and Physical Health and Clinical Translation, Barwon Health, Deakin University, Geelong 3220, Australia; (B.S.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (B.P.); (L.G.); (M.B.); (O.M.D.); (K.W.)
- School of Medicine, Centre for Molecular and Medical Research, Deakin University, Geelong 3220, Australia;
| | - Michael Berk
- The Institute for Mental and Physical Health and Clinical Translation, Barwon Health, Deakin University, Geelong 3220, Australia; (B.S.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (B.P.); (L.G.); (M.B.); (O.M.D.); (K.W.)
- Department of Psychiatry, Royal Melbourne Hospital, University of Melbourne, Parkville 3052, Australia
- Centre of Youth Mental Health, University of Melbourne, Parkville 3052, Australia
- Orygen Youth Health Research Centre, Parkville 3052, Australia
| | - Olivia M. Dean
- The Institute for Mental and Physical Health and Clinical Translation, Barwon Health, Deakin University, Geelong 3220, Australia; (B.S.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (B.P.); (L.G.); (M.B.); (O.M.D.); (K.W.)
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Australia
| | - Ken Walder
- The Institute for Mental and Physical Health and Clinical Translation, Barwon Health, Deakin University, Geelong 3220, Australia; (B.S.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (B.P.); (L.G.); (M.B.); (O.M.D.); (K.W.)
- School of Medicine, Centre for Molecular and Medical Research, Deakin University, Geelong 3220, Australia;
| |
Collapse
|
20
|
Horigane SI, Hamada S, Kamijo S, Yamada H, Yamasaki M, Watanabe M, Bito H, Ohtsuka T, Takemoto-Kimura S. Development of an L-type Ca 2+ channel-dependent Ca 2+ transient during the radial migration of cortical excitatory neurons. Neurosci Res 2020; 169:17-26. [PMID: 32598973 DOI: 10.1016/j.neures.2020.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 01/16/2023]
Abstract
Increasing evidence has shown that voltage-gated L-type Ca2+ channels (LTCCs) are crucial for neurodevelopmental events, including neuronal differentiation/migration and neurite morphogenesis/extension. However, the time course of their functional maturation during the development of excitatory neurons remains unknown. Using a combination of fluorescence in situ hybridization and in utero electroporation-based labeling, we found that the transcripts of Cacna1c and Cacna1d, which encode the LTCC pore-forming subunits, were upregulated in the intermediate zone (IZ) during radial migration. Ca2+ imaging using GCaMP6s in acute brain slices showed spontaneous Ca2+ transients in migrating neurons throughout the IZ. Neurons in the IZ upper layer, especially in the multipolar-to-bipolar transition layer (TL), exhibited more frequent Ca2+ transients than adjacent layers and responded to FPL64176, a potent activator of LTCC. Consistently, nimodipine, an LTCC blocker, inhibited spontaneous Ca2+ transients in neurons in the TL. Collectively, we showed a hitherto unknown increased prevalence of LTCC-dependent Ca2+ transients in the TL of the IZ upper layer during the radial migration of excitatory neurons, which could be essential for the regulation of Ca2+-dependent neurodevelopmental processes.
Collapse
Affiliation(s)
- Shin-Ichiro Horigane
- Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601, Japan; Molecular/cellular Neuroscience, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan; Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Shun Hamada
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Satoshi Kamijo
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hirokazu Yamada
- Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601, Japan; Molecular/cellular Neuroscience, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Miwako Yamasaki
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Toshihisa Ohtsuka
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Sayaka Takemoto-Kimura
- Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601, Japan; Molecular/cellular Neuroscience, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan; PRESTO, Japan Science and Technology Agency, Chiyoda-ku, Tokyo, 102-0076, Japan.
| |
Collapse
|
21
|
Gaff J, Pillay P, Cherry C, Laws SM, Price P, Kamerman P. The role of CAMKK2 polymorphisms in HIV-associated sensory neuropathy in South Africans. J Neurol Sci 2020; 416:116987. [PMID: 32585444 DOI: 10.1016/j.jns.2020.116987] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 12/30/2022]
Abstract
Human immunodeficiency virus-associated sensory neuropathy (HIV-SN) is a common neurological complication of HIV infection. It affected 57% of South African patients whose antiretroviral therapy (ART) included stavudine and was influenced by genotypes of the P2X-block (P2X7R, P2X4R and CAMKK2). We investigate associations between HIV-SN and P2X-block genotypes in patients who never received stavudine. An adjacent gene, ANAPC5, was included. 75 HIV+ individuals were assessed using the Brief Peripheral Neuropathy Screen before treatment and after 6-8 months on stavudine-free regimens. DNA was genotyped for 48 polymorphisms across the four genes using an OpenArray™ platform. Haplotypes were derived using fastPHASE. Associations with HIV-SN were assessed using bivariate and multivariate analyses. Nine individuals (12%) were diagnosed with HIV-SN prior to ART and a further 20 individuals (27%) developed HIV-SN within 6-8 months. Five polymorphisms, rs503720*G (OR = 133) in P2X7R, rs10849861*A (OR = 5.99), rs1653586*T (OR = 67.8) and rs11065504*C (OR = 0.02) in CAMKK2, and rs2089886*A (OR = 6.68) in ANAPC5, associated with HIV-SN after adjusting for body weight, nadir CD4 T-cell counts and prior tuberculosis (model p < 0.0001, n = 69, Pseudo R2 = 0.54). Three CAMKK2 haplotypes were associated with HIV-SN (OR = 2.82, 3.42 and 6.85) after adjusting for body weight, nadir CD4 T-cell counts and prior tuberculosis (model p < 0.0005, n = 71, Pseudo R2 = 0.26). The results support a role for CAMKK2 in HIV-SN, independent of mechanisms invoked by stavudine. SIGNIFICANCE STATEMENT: HIV-associated sensory neuropathy (HIV-SN) remains a clinically relevant complication of HIV infection and its treatment, affecting 38% of patients treated without neurotoxic stavudine. HIV-SN can impact an individual's ability to work and quality of life, with few effective therapeutic options, so an understanding of the underlying mechanisms would have clinical value. We confirm that CAMKK2 polymorphisms and haplotypes influence susceptibility to HIV-SN in South Africans treated without stavudine. This provides further evidence for a role for the protein encoded by CAMKK2 in the pathogenesis of HIV-SN, independent of mechanisms initiated by stavudine.
Collapse
Affiliation(s)
- Jessica Gaff
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia
| | - Prinisha Pillay
- Brain Function Research Group, School of Physiology, University of Witwatersrand, Johannesburg, South Africa
| | - Catherine Cherry
- Brain Function Research Group, School of Physiology, University of Witwatersrand, Johannesburg, South Africa; Department of Infectious Diseases, Alfred Health and Monash University, Melbourne, Australia; Burnet Institute, Melbourne, Australia
| | - Simon M Laws
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia; Collaborative Genomics Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Patricia Price
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia; Brain Function Research Group, School of Physiology, University of Witwatersrand, Johannesburg, South Africa.
| | - Peter Kamerman
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia; Brain Function Research Group, School of Physiology, University of Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
22
|
Poston RG, Murphy L, Rejepova A, Ghaninejad-Esfahani M, Segales J, Mulligan K, Saha RN. Certain ortho-hydroxylated brominated ethers are promiscuous kinase inhibitors that impair neuronal signaling and neurodevelopmental processes. J Biol Chem 2020; 295:6120-6137. [PMID: 32229587 PMCID: PMC7196656 DOI: 10.1074/jbc.ra119.011138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 03/23/2020] [Indexed: 12/20/2022] Open
Abstract
The developing nervous system is remarkably sensitive to environmental signals, including disruptive toxins, such as polybrominated diphenyl ethers (PBDEs). PBDEs are an environmentally pervasive class of brominated flame retardants whose neurodevelopmental toxicity mechanisms remain largely unclear. Using dissociated cortical neurons from embryonic Rattus norvegicus, we found here that chronic exposure to 6-OH-BDE-47, one of the most prevalent hydroxylated PBDE metabolites, suppresses both spontaneous and evoked neuronal electrical activity. On the basis of our previous work on mitogen-activated protein kinase (MAPK)/extracellular signal-related kinase (ERK) (MEK) biology and our observation that 6-OH-BDE-47 is structurally similar to kinase inhibitors, we hypothesized that certain hydroxylated PBDEs mediate neurotoxicity, at least in part, by impairing the MEK-ERK axis of MAPK signal transduction. We tested this hypothesis on three experimental platforms: 1) in silico, where modeling ligand-protein docking suggested that 6-OH-BDE-47 is a promiscuous ATP-competitive kinase inhibitor; 2) in vitro in dissociated neurons, where 6-OH-BDE-47 and another specific hydroxylated BDE metabolite similarly impaired phosphorylation of MEK/ERK1/2 and activity-induced transcription of a neuronal immediate early gene; and 3) in vivo in Drosophila melanogaster, where developmental exposures to 6-OH-BDE-47 and a MAPK inhibitor resulted in offspring displaying similarly increased frequency of mushroom-body β-lobe midline crossing, a metric of axonal guidance. Taken together, our results support that certain ortho-hydroxylated PBDE metabolites are promiscuous kinase inhibitors and can cause disruptions of critical neurodevelopmental processes, including neuronal electrical activity, pre-synaptic functions, MEK-ERK signaling, and axonal guidance.
Collapse
Affiliation(s)
- Robert G Poston
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343
| | - Lillian Murphy
- Department of Biological Sciences, Center for Interdisciplinary Molecular Biology: Education, Research and Advancement (CIMERA), California State University, Sacramento, California 95819
| | - Ayna Rejepova
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343
| | - Mina Ghaninejad-Esfahani
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343
| | - Joshua Segales
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343
| | - Kimberly Mulligan
- Department of Biological Sciences, Center for Interdisciplinary Molecular Biology: Education, Research and Advancement (CIMERA), California State University, Sacramento, California 95819
| | - Ramendra N Saha
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343.
| |
Collapse
|
23
|
Ohtsuka S, Ozeki Y, Fujiwara M, Miyagawa T, Kanayama N, Magari M, Hatano N, Suizu F, Ishikawa T, Tokumitsu H. Development and Characterization of Novel Molecular Probes for Ca 2+/Calmodulin-Dependent Protein Kinase Kinase, Derived from STO-609. Biochemistry 2020; 59:1701-1710. [PMID: 32298102 DOI: 10.1021/acs.biochem.0c00149] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ca2+/calmodulin-dependent protein kinase kinase (CaMKK) activates particular multifunctional kinases, including CaMKI, CaMKIV, and 5'AMP-activated protein kinase (AMPK), resulting in the regulation of various Ca2+-dependent cellular processes, including neuronal, metabolic, and pathophysiological pathways. We developed and characterized a novel pan-CaMKK inhibitor, TIM-063 (2-hydroxy-3-nitro-7H-benzo[de]benzo[4,5]imidazo[2,1-a]isoquinolin-7-one) derived from STO-609 (7H-benzimidazo[2,1-a]benz[de]isoquinoline-7-one-3-carboxylic acid), and an inactive analogue (TIM-062) as molecular probes for the analysis of CaMKK-mediated cellular responses. Unlike STO-609, TIM-063 had an inhibitory activity against CaMKK isoforms (CaMKKα and CaMKKβ) with a similar potency (Ki = 0.35 μM for CaMKKα, and Ki = 0.2 μM for CaMKKβ) in vitro. Two TIM-063 analogues lacking a nitro group (TIM-062) or a hydroxy group (TIM-064) completely impaired CaMKK inhibitory activities, indicating that both substituents are necessary for the CaMKK inhibitory activity of TIM-063. Enzymatic analysis revealed that TIM-063 is an ATP-competitive inhibitor that directly targets the catalytic domain of CaMKK, similar to STO-609. TIM-063 suppressed the ionomycin-induced phosphorylation of exogenously expressed CaMKI, CaMKIV, and endogenous AMPKα in HeLa cells with an IC50 of ∼0.3 μM, and it suppressed CaMKK isoform-mediated CaMKIV phosphorylation in transfected COS-7 cells. Thus, TIM-063, but not the inactive analogue (TIM-062), displayed cell permeability and the ability to inhibit CaMKK activity in cells. Taken together, these results indicate that TIM-063 could be a useful tool for the precise analysis of CaMKK-mediated signaling pathways and may be a promising lead compound for the development of therapeutic agents for the treatment of CaMKK-related diseases.
Collapse
Affiliation(s)
- Satomi Ohtsuka
- Applied Cell Biology, Graduate School of Interdisciplinary Science & Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
| | | | | | | | - Naoki Kanayama
- Applied Cell Biology, Graduate School of Interdisciplinary Science & Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
| | - Masaki Magari
- Applied Cell Biology, Graduate School of Interdisciplinary Science & Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
| | - Naoya Hatano
- Applied Cell Biology, Graduate School of Interdisciplinary Science & Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
| | - Futoshi Suizu
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
| | | | - Hiroshi Tokumitsu
- Applied Cell Biology, Graduate School of Interdisciplinary Science & Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
| |
Collapse
|
24
|
In vivo epigenetic editing of Sema6a promoter reverses transcallosal dysconnectivity caused by C11orf46/Arl14ep risk gene. Nat Commun 2019; 10:4112. [PMID: 31511512 PMCID: PMC6739341 DOI: 10.1038/s41467-019-12013-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 08/12/2019] [Indexed: 12/15/2022] Open
Abstract
Many neuropsychiatric risk genes contribute to epigenetic regulation but little is known about specific chromatin-associated mechanisms governing the formation of neuronal connectivity. Here we show that transcallosal connectivity is critically dependent on C11orf46, a nuclear protein encoded in the chromosome 11p13 WAGR risk locus. C11orf46 haploinsufficiency was associated with hypoplasia of the corpus callosum. C11orf46 knockdown disrupted transcallosal projections and was rescued by wild type C11orf46 but not the C11orf46R236H mutant associated with intellectual disability. Multiple genes encoding key regulators of axonal development, including Sema6a, were hyperexpressed in C11orf46-knockdown neurons. RNA-guided epigenetic editing of Sema6a gene promoters via a dCas9-SunTag system with C11orf46 binding normalized SEMA6A expression and rescued transcallosal dysconnectivity via repressive chromatin remodeling by the SETDB1 repressor complex. Our study demonstrates that interhemispheric communication is sensitive to locus-specific remodeling of neuronal chromatin, revealing the therapeutic potential for shaping the brain's connectome via gene-targeted designer activators and repressor proteins.
Collapse
|
25
|
Gaff J, Octaviana F, Ariyanto I, Cherry C, Laws SM, Price P. Polymorphisms in CAMKK2 associate with susceptibility to sensory neuropathy in HIV patients treated without stavudine. J Neurovirol 2019; 25:814-824. [PMID: 31309408 DOI: 10.1007/s13365-019-00771-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 05/14/2019] [Accepted: 06/03/2019] [Indexed: 01/20/2023]
Abstract
HIV-associated sensory neuropathy (HIV-SN) is a debilitating neurological complication of HIV infection potentiated by the antiretroviral drug stavudine. While stavudine is no longer used, HIV-SN now affects around 15% of HIV+ Indonesians. Here, we investigate whether polymorphisms within the P2X-block (P2X4R, P2X7R, CAMKK2) and/or ANAPC5 mark susceptibility to HIV-SN in this setting. As polymorphisms in these genes associated with HIV-SN in African HIV patients receiving stavudine, the comparison can identify mechanisms independent of stavudine. HIV patients who had never used stavudine (n = 202) attending clinics in Jakarta were screened for neuropathy using the AIDS Clinical Trials Group Brief Peripheral Neuropathy Screen. Open-array technology was used to type 48 polymorphisms spanning the four genes. Haplotypes were derived for each gene using fastPHASE. Haplogroups were constructed with median-joining methods. Multivariable models optimally predicting HIV-SN were based on factors achieving p < 0.2 in bivariate analyses. Minor alleles of three co-inherited polymorphisms in CAMKK2 (rs7975295*C, rs1560568*A, rs1132780*T) associated with a reduced prevalence of HIV-SN individually and after adjusting for lower CD4 T cell count and viremia (p = 0.0002, pseudo R2 = 0.11). The optimal model for haplotypes linked HIV-SN with viremia and lower current CD4 T cell count, plus CAMKK2 haplotypes 6 and 11 and P2X7R haplotypes 2 and 12 (p = 0.0002; pseudo R2 = 0.11). CAMKK2 haplogroup A (includes 16 haplotypes and all instances of rs7975295*C, rs1560568*A, rs1132780*T) associated with reduced rates of HIV-SN (p = 0.02, OR = 0.43 CI = 0.21-0.88). These findings support a protective role for these three alleles, suggesting a role in the pathogenesis of HIV-SN that is independent of stavudine.
Collapse
Affiliation(s)
- Jessica Gaff
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia
| | - Fitri Octaviana
- Neurology Department, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Neurology Department, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Ibnu Ariyanto
- Virology and Cancer Pathobiology Research Center, Universitas Indonesia, Jakarta, Indonesia
| | - Catherine Cherry
- Department of Infectious Diseases, Alfred Health and Monash University, Melbourne, Australia.,Burnet Institute, Melbourne, Australia
| | - Simon M Laws
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia.,Collaborative Genomics Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Patricia Price
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia. .,Virology and Cancer Pathobiology Research Center, Universitas Indonesia, Jakarta, Indonesia. .,School of Physiology, University of Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
26
|
Horigane SI, Ozawa Y, Yamada H, Takemoto-Kimura S. Calcium signalling: a key regulator of neuronal migration. J Biochem 2019; 165:401-409. [PMID: 30753600 DOI: 10.1093/jb/mvz012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 02/10/2019] [Indexed: 12/12/2022] Open
Abstract
Neuronal migration is a crucial event in neuronal development for the construction of brain architecture and neuronal networks. Newborn neurons proliferate in the germinal zone and start migration toward their final destination. Migrating neurons adopt different routes, cell shapes and migratory modes depending on extracellular factors and outer physical substrates. Intracellular Ca2+ is an essential second messenger that regulates diverse cellular functions by activating Ca2+-dependent signalling molecules that underlie Ca2+-responsive cellular functions. Neuronal migration during brain architecture construction is no exception. Spontaneous Ca2+ transients are observed in several types of migrating neurons, and a series of Ca2+-dependent signalling molecules governing neuronal migration has been identified. In this review, we first summarize the molecular mechanisms that trigger intracellular Ca2+ elevation in migrating neurons. In the latter half of this review, we provide an overview of the literature on Ca2+-dependent signalling molecules underlying neuronal migration.
Collapse
Affiliation(s)
- Shin-Ichiro Horigane
- Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Molecular/Cellular Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukihiro Ozawa
- Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Molecular/Cellular Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Pathology and Laboratory Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Hirokazu Yamada
- Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Molecular/Cellular Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sayaka Takemoto-Kimura
- Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Molecular/Cellular Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Saitama, Japan
| |
Collapse
|
27
|
Takata T, Tsukuda A, Tsuchiya Y, Akaike T, Watanabe Y. The active-site cysteine residue of Ca 2+/calmodulin-dependent protein kinase I is protected from irreversible modification via generation of polysulfidation. Nitric Oxide 2019; 86:68-75. [PMID: 30844494 DOI: 10.1016/j.niox.2019.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/14/2019] [Accepted: 02/21/2019] [Indexed: 11/30/2022]
Abstract
Ca2+/calmodulin (CaM)-dependent protein kinase (CaMK) I is activated by the phosphorylation of a crucial activation loop Thr177 by upstream kinases, CaMK kinase (CaMKK), and regulates axonal or dendritic extension and branching. Reactive sulfur species (RSS) modulate protein functions via polysulfidation of the reactive Cys residues. Here, we report that the activity of CaMKI was reversibly inhibited via its polysulfidation at Cys179 by RSS. In vitro incubation of CaMKI with the exogenous RSS donor Na2S3 resulted in a dose-dependent inhibition of the phosphorylation at Thr177 by CaMKK and inactivation of the enzymatic activity. Dithiothreitol (DTT), a small molecule reducing reagent, rescued these inhibitions. Conversely, mutated CaMKI (C179V) was resistant to the Na2S3-induced inactivation. In transfected cells expressing CaMKI, ionomycin-induced CaMKI activity was decreased upon treatment with Na2S4, whereas cells expressing mutant CaMKI (C179V) proved resistant to this treatment. A biotin-polyethylene glycol-conjugated maleimide capture assay revealed that CaMKI was a target for polysulfidation in cells. Furthermore, the polysulfidation of CaMKI protected Cys179 from its irreversible modification, known as protein succination. Thus, we propose that CaMKI was reversibly inhibited via polysulfidation of Cys179 by RSS, thereby protecting it from irreversible modification.
Collapse
Affiliation(s)
- Tsuyoshi Takata
- Department of Pharmacology, High Technology Research Center, Showa Pharmaceutical University, Machida, Tokyo, 194-8543, Japan
| | - Ayaka Tsukuda
- Department of Pharmacology, High Technology Research Center, Showa Pharmaceutical University, Machida, Tokyo, 194-8543, Japan
| | - Yukihiro Tsuchiya
- Department of Pharmacology, High Technology Research Center, Showa Pharmaceutical University, Machida, Tokyo, 194-8543, Japan
| | - Takaaki Akaike
- Department of Environmental Health Sciences and Molecular Toxicology, Graduate School of Medicine, Tohoku University, Miyagi, 980-8575, Japan
| | - Yasuo Watanabe
- Department of Pharmacology, High Technology Research Center, Showa Pharmaceutical University, Machida, Tokyo, 194-8543, Japan.
| |
Collapse
|
28
|
Gonzalez-Escamilla G, Muthuraman M, Chirumamilla VC, Vogt J, Groppa S. Brain Networks Reorganization During Maturation and Healthy Aging-Emphases for Resilience. Front Psychiatry 2018; 9:601. [PMID: 30519196 PMCID: PMC6258799 DOI: 10.3389/fpsyt.2018.00601] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 10/29/2018] [Indexed: 12/31/2022] Open
Abstract
Maturation and aging are important life periods that are linked to drastic brain reorganization processes which are essential for mental health. However, the development of generalized theories for delimiting physiological and pathological brain remodeling through life periods linked to healthy states and resilience on one side or mental dysfunction on the other remains a challenge. Furthermore, important processes of preservation and compensation of brain function occur continuously in the cerebral brain networks and drive physiological responses to life events. Here, we review research on brain reorganization processes across the lifespan, demonstrating brain circuits remodeling at the structural and functional level that support mental health and are parallelized by physiological trajectories during maturation and healthy aging. We show evidence that aberrations leading to mental disorders result from the specific alterations of cerebral networks and their pathological dynamics leading to distinct excitability patterns. We discuss how these series of large-scale responses of brain circuits can be viewed as protective or malfunctioning mechanisms for the maintenance of mental health and resilience.
Collapse
Affiliation(s)
| | - Muthuraman Muthuraman
- Department of Neurology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Venkata C. Chirumamilla
- Department of Neurology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Johannes Vogt
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sergiu Groppa
- Department of Neurology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
29
|
Courchet V, Roberts AJ, Meyer-Dilhet G, Del Carmine P, Lewis TL, Polleux F, Courchet J. Haploinsufficiency of autism spectrum disorder candidate gene NUAK1 impairs cortical development and behavior in mice. Nat Commun 2018; 9:4289. [PMID: 30327473 PMCID: PMC6191442 DOI: 10.1038/s41467-018-06584-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 09/05/2018] [Indexed: 01/14/2023] Open
Abstract
Recently, numerous rare de novo mutations have been identified in patients diagnosed with autism spectrum disorders (ASD). However, despite the predicted loss-of-function nature of some of these de novo mutations, the affected individuals are heterozygous carriers, which would suggest that most of these candidate genes are haploinsufficient and/or lead to expression of dominant-negative forms of the protein. Here, we tested this hypothesis with the candidate ASD gene Nuak1 that we previously identified for its role in the development of cortical connectivity. We report that Nuak1 is haploinsufficient in mice with regard to its function in cortical development. Furthermore Nuak1+/− mice show a combination of abnormal behavioral traits ranging from defective spatial memory consolidation, defects in social novelty (but not social preference) and abnormal sensorimotor gating. Overall, our results demonstrate that Nuak1 haploinsufficiency leads to defects in the development of cortical connectivity and a complex array of behavorial deficits. Nuak1 is an autism spectrum disorder candidate gene. Here the authors report behavioral and cortical development in mice heterozygous for Nuak1, suggesting loss of function mutations in one copy of Nuak1 may contribute to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Virginie Courchet
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, F-69008, Lyon, France
| | - Amanda J Roberts
- Department of Neurosciences, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Géraldine Meyer-Dilhet
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, F-69008, Lyon, France
| | - Peggy Del Carmine
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, F-69008, Lyon, France
| | - Tommy L Lewis
- Department of Neuroscience, Zuckerman Mind Brain Behavior Institute and Kavli Institute for Brain Science, Columbia University, New York, NY, 10032, USA
| | - Franck Polleux
- Department of Neuroscience, Zuckerman Mind Brain Behavior Institute and Kavli Institute for Brain Science, Columbia University, New York, NY, 10032, USA.
| | - Julien Courchet
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, F-69008, Lyon, France.
| |
Collapse
|
30
|
A Critical Neurodevelopmental Role for L-Type Voltage-Gated Calcium Channels in Neurite Extension and Radial Migration. J Neurosci 2018; 38:5551-5566. [PMID: 29773754 DOI: 10.1523/jneurosci.2357-17.2018] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 12/28/2017] [Accepted: 04/21/2018] [Indexed: 11/21/2022] Open
Abstract
Despite many association studies linking gene polymorphisms and mutations of L-type voltage-gated Ca2+ channels (VGCCs) in neurodevelopmental disorders such as autism and schizophrenia, the roles of specific L-type VGCC during brain development remain unclear. Calcium signaling has been shown to be essential for neurodevelopmental processes such as sculpting of neurites, functional wiring, and fine tuning of growing networks. To investigate this relationship, we performed submembraneous calcium imaging using a membrane-tethered genetically encoded calcium indicator (GECI) Lck-G-CaMP7. We successfully recorded spontaneous regenerative calcium transients (SRCaTs) in developing mouse excitatory cortical neurons prepared from both sexes before synapse formation. SRCaTs originated locally in immature neurites independently of somatic calcium rises and were significantly more elevated in the axons than in dendrites. SRCaTs were not blocked by tetrodoxin, a Na+ channel blocker, but were strongly inhibited by hyperpolarization, suggesting a voltage-dependent source. Pharmacological and genetic manipulations revealed the critical importance of the Cav1.2 (CACNA1C) pore-forming subunit of L-type VGCCs, which were indeed expressed in immature mouse brains. Consistently, knocking out Cav1.2 resulted in significant alterations of neurite outgrowth. Furthermore, expression of a gain-of-function Cav1.2 mutant found in Timothy syndrome, an autosomal dominant multisystem disorder exhibiting syndromic autism, resulted in impaired radial migration of layer 2/3 excitatory neurons, whereas postnatal abrogation of Cav1.2 enhancement could rescue cortical malformation. Together, these lines of evidence suggest a critical role for spontaneous opening of L-type VGCCs in neural development and corticogenesis and indicate that L-type VGCCs might constitute a perinatal therapeutic target for neuropsychiatric calciochannelopathies.SIGNIFICANCE STATEMENT Despite many association studies linking gene polymorphisms and mutations of L-type voltage-gated Ca2+ channels (VGCCs) in neurodevelopmental disorders such as autism and schizophrenia, the roles of specific L-type VGCCs during brain development remain unclear. We here combined the latest Ca2+ indicator technology, quantitative pharmacology, and in utero electroporation and found a hitherto unsuspected role for L-type VGCCs in determining the Ca2+ signaling landscape of mouse immature neurons. We found that malfunctional L-type VGCCs in immature neurons before birth might cause errors in neuritic growth and cortical migration. Interestingly, the retarded corticogenesis phenotype was rescued by postnatal correction of L-type VGCC signal aberration. These findings suggest that L-type VGCCs might constitute a perinatal therapeutic target for neurodevelopment-associated psychiatric disorders.
Collapse
|
31
|
Vogt J, Kirischuk S, Unichenko P, Schlüter L, Pelosi A, Endle H, Yang JW, Schmarowski N, Cheng J, Thalman C, Strauss U, Prokudin A, Bharati BS, Aoki J, Chun J, Lutz B, Luhmann HJ, Nitsch R. Synaptic Phospholipid Signaling Modulates Axon Outgrowth via Glutamate-dependent Ca2+-mediated Molecular Pathways. Cereb Cortex 2018; 27:131-145. [PMID: 27909001 PMCID: PMC5939201 DOI: 10.1093/cercor/bhw370] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Indexed: 12/28/2022] Open
Abstract
Altered synaptic bioactive lipid signaling has been recently shown to augment neuronal excitation in the hippocampus of adult animals by activation of presynaptic LPA2-receptors leading to increased presynaptic glutamate release. Here, we show that this results in higher postsynaptic Ca2+ levels and in premature onset of spontaneous neuronal activity in the developing entorhinal cortex. Interestingly, increased synchronized neuronal activity led to reduced axon growth velocity of entorhinal neurons which project via the perforant path to the hippocampus. This was due to Ca2+-dependent molecular signaling to the axon affecting stabilization of the actin cytoskeleton. The spontaneous activity affected the entire entorhinal cortical network and thus led to reduced overall axon fiber numbers in the mature perforant path that is known to be important for specific memory functions. Our data show that precise regulation of early cortical activity by bioactive lipids is of critical importance for proper circuit formation.
Collapse
Affiliation(s)
- Johannes Vogt
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Sergei Kirischuk
- Institute of Physiology, University Medical Center, Johannes Gutenberg-University, 55128 Mainz, Germany
| | - Petr Unichenko
- Institute of Physiology, University Medical Center, Johannes Gutenberg-University, 55128 Mainz, Germany
| | - Leslie Schlüter
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Assunta Pelosi
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Heiko Endle
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Jenq-Wei Yang
- Institute of Physiology, University Medical Center, Johannes Gutenberg-University, 55128 Mainz, Germany
| | - Nikolai Schmarowski
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Jin Cheng
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Carine Thalman
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Ulf Strauss
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin, 10119 Berlin, Germany
| | - Alexey Prokudin
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - B Suman Bharati
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Junken Aoki
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Jerold Chun
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center, Johannes Gutenberg-University, 55128 Mainz, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center, Johannes Gutenberg-University, 55128 Mainz, Germany
| | - Robert Nitsch
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| |
Collapse
|
32
|
GABA A receptor subunit deregulation in the hippocampus of human foetuses with Down syndrome. Brain Struct Funct 2017; 223:1501-1518. [PMID: 29168008 PMCID: PMC5869939 DOI: 10.1007/s00429-017-1563-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 11/05/2017] [Indexed: 11/01/2022]
Abstract
The function, regulation and cellular distribution of GABAA receptor subunits have been extensively documented in the adult rodent brain and are linked to numerous neurological disorders. However, there is a surprising lack of knowledge on the cellular (sub-) distribution of GABAA receptor subunits and of their expressional regulation in developing healthy and diseased foetal human brains. To propose a role for GABAA receptor subunits in neurodevelopmental disorders, we studied the developing hippocampus of normal and Down syndrome foetuses. Among the α1-3 and γ2 subunits probed, we find significantly altered expression profiles of the α1, α3 and γ2 subunits in developing Down syndrome hippocampi, with the α3 subunit being most affected. α3 subunits were selectively down-regulated in all hippocampal subfields and developmental periods tested in Down syndrome foetuses, presenting a developmental mismatch by their adult-like distribution in early foetal development. We hypothesized that increased levels of the amyloid precursor protein (APP), and particularly its neurotoxic β-amyloid (1-42) fragment, could disrupt α3 gene expression, likely by facilitating premature neuronal differentiation. Indeed, we find increased APP content in the hippocampi of the Down foetuses. In a corresponding cellular model, soluble β-amyloid (1-42) administered to cultured SH-SY5Y neuroblastoma cells, augmented by retinoic acid-induced differentiation towards a neuronal phenotype, displayed a reduction in α3 subunit levels. In sum, this study charts a comprehensive regional and subcellular map of key GABAA receptor subunits in identified neuronal populations in the hippocampus of healthy and Down syndrome foetuses and associates increased β-amyloid load with discordant down-regulation of α3 subunits.
Collapse
|
33
|
SAD-B Phosphorylation of CAST Controls Active Zone Vesicle Recycling for Synaptic Depression. Cell Rep 2017; 16:2901-2913. [PMID: 27626661 DOI: 10.1016/j.celrep.2016.08.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 06/28/2016] [Accepted: 08/05/2016] [Indexed: 12/25/2022] Open
Abstract
Short-term synaptic depression (STD) is a common form of activity-dependent plasticity observed widely in the nervous system. Few molecular pathways that control STD have been described, but the active zone (AZ) release apparatus provides a possible link between neuronal activity and plasticity. Here, we show that an AZ cytomatrix protein CAST and an AZ-associated protein kinase SAD-B coordinately regulate STD by controlling reloading of the AZ with release-ready synaptic vesicles. SAD-B phosphorylates the N-terminal serine (S45) of CAST, and S45 phosphorylation increases with higher firing rate. A phosphomimetic CAST (S45D) mimics CAST deletion, which enhances STD by delaying reloading of the readily releasable pool (RRP), resulting in a pool size decrease. A phosphonegative CAST (S45A) inhibits STD and accelerates RRP reloading. Our results suggest that the CAST/SAD-B reaction serves as a brake on synaptic transmission by temporal calibration of activity and synaptic depression via RRP size regulation.
Collapse
|
34
|
Takemoto-Kimura S, Suzuki K, Horigane SI, Kamijo S, Inoue M, Sakamoto M, Fujii H, Bito H. Calmodulin kinases: essential regulators in health and disease. J Neurochem 2017; 141:808-818. [PMID: 28295333 DOI: 10.1111/jnc.14020] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 02/24/2017] [Accepted: 03/08/2017] [Indexed: 01/22/2023]
Abstract
Neuronal activity induces intracellular Ca2+ increase, which triggers activation of a series of Ca2+ -dependent signaling cascades. Among these, the multifunctional Ca2+ /calmodulin-dependent protein kinases (CaMKs, or calmodulin kinases) play key roles in neuronal transmission, synaptic plasticity, circuit development and cognition. The most investigated CaMKs for these roles in neuronal functions are CaMKI, CaMKII, CaMKIV and we will shed light on these neuronal CaMKs' functions in this review. Catalytically active members of CaMKs currently are CaMKI, CaMKII, CaMKIV and CaMKK. Although they all necessitate the binding of Ca2+ and calmodulin complex (Ca2+ /CaM) for releasing autoinhibition, each member of CaMK has distinct activation mechanisms-autophosphorylation mediated autonomy of multimeric CaMKII and CaMKK-dependent phosphoswitch-induced activation of CaMKI or CaMKIV. Furthermore, each CaMK shows distinct subcellular localization that underlies specific compartmentalized function in each activated neuron. In this review, we first summarize these molecular characteristics of each CaMK as to regulation and subcellular localization, and then describe each biological function. In the last section, we also focus on the emerging role of CaMKs in pathophysiological conditions by introducing the recent studies, especially focusing on drug addiction and depression, and discuss how dysfunctional CaMKs may contribute to the pathology of the neuropsychological disorders. This article is part of the mini review series "60th Anniversary of the Japanese Society for Neurochemistry".
Collapse
Affiliation(s)
- Sayaka Takemoto-Kimura
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Chikusa-ku, Nagoya, Japan.,PRESTO-Japan Science and Technology Agency, Chiyoda-ku, Tokyo, Japan
| | - Kanzo Suzuki
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shin-Ichiro Horigane
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Chikusa-ku, Nagoya, Japan
| | - Satoshi Kamijo
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masatoshi Inoue
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masayuki Sakamoto
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hajime Fujii
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
35
|
Seno T, Ikeno T, Mennya K, Kurishita M, Sakae N, Sato M, Takada H, Konishi Y. Kinesin-1 sorting in axons controls the differential retraction of arbor terminals. J Cell Sci 2016; 129:3499-510. [PMID: 27505885 DOI: 10.1242/jcs.183806] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 08/02/2016] [Indexed: 01/13/2023] Open
Abstract
The ability of neurons to generate multiple arbor terminals from a single axon is crucial for establishing proper neuronal wiring. Although growth and retraction of arbor terminals are differentially regulated within the axon, the mechanisms by which neurons locally control their structure remain largely unknown. In the present study, we found that the kinesin-1 (Kif5 proteins) head domain (K5H) preferentially marks a subset of arbor terminals. Time-lapse imaging clarified that these arbor terminals were more stable than others, because of a low retraction rate. Local inhibition of kinesin-1 in the arbor terminal by chromophore-assisted light inactivation (CALI) enhanced the retraction rate. The microtubule turnover was locally regulated depending on the length from the branching point to the terminal end, but did not directly correlate with the presence of K5H. By contrast, F-actin signal values in arbor terminals correlated spatiotemporally with K5H, and inhibition of actin turnover prevented retraction. Results from the present study reveal a new system mediated by kinesin-1 sorting in axons that differentially controls stability of arbor terminals.
Collapse
Affiliation(s)
- Takeshi Seno
- Department of Human and Artificial Intelligence Systems, University of Fukui, Fukui 910-8507, Japan
| | - Tatsuki Ikeno
- Department of Human and Artificial Intelligence Systems, University of Fukui, Fukui 910-8507, Japan
| | - Kousuke Mennya
- Department of Human and Artificial Intelligence Systems, University of Fukui, Fukui 910-8507, Japan
| | - Masayuki Kurishita
- Department of Human and Artificial Intelligence Systems, University of Fukui, Fukui 910-8507, Japan
| | - Narumi Sakae
- Department of Human and Artificial Intelligence Systems, University of Fukui, Fukui 910-8507, Japan
| | - Makoto Sato
- Life Science Innovation Center, University of Fukui, Fukui 910-8507, Japan Research Center for Child Mental Development, University of Fukui, Fukui 910-1193, Japan Department of Anatomy and Neuroscience, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan United Graduate School of Child Development, Osaka University, Kanazawa University-Hamamatsu University School of Medicine, Chiba University and University of Fukui, Osaka University, Osaka 565-0871, Japan
| | - Hiroki Takada
- Department of Human and Artificial Intelligence Systems, University of Fukui, Fukui 910-8507, Japan
| | - Yoshiyuki Konishi
- Department of Human and Artificial Intelligence Systems, University of Fukui, Fukui 910-8507, Japan Life Science Innovation Center, University of Fukui, Fukui 910-8507, Japan Department of Materials Science and Biotechnology, Faculty of Engineering, University of Fukui, Fukui 910-8507, Japan
| |
Collapse
|
36
|
Gonçalves SA, Outeiro TF. Traffic jams and the complex role of α-Synuclein aggregation in Parkinson disease. Small GTPases 2016; 8:78-84. [PMID: 27314512 DOI: 10.1080/21541248.2016.1199191] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
A common pathological event among various neurodegenerative disorders (NDs) is the misfolding and aggregation of different proteins in the brain. This is thought to potentiate aberrant protein-protein interactions that culminate in the disruption of several biological processes and, ultimately, in neuronal cell loss. Although protein aggregates are a common hallmark in several disorders, the molecular pathways leading to their generation remain unclear. The misfolding and aggregation of α-Synuclein (aSyn) is the pathological hallmark of Parkinson disease (PD), the second most common age related ND. It has been postulated that oligomeric species of aSyn, rather than more mature aggregated forms of the protein, are the causative agents of cytotoxicity. In recent years, we have been investigating the molecular mechanisms underlying the initial steps of aSyn accumulation in living cells. Using an unbiased genome-wide lentiviral RNAi screen we identified trafficking and kinase genes as modulators of aSyn oligomerization, aggregation, and toxicity. Among those, Rab8b, Rab11a, Rab13 and Slp5 were found to promote the clearance of aSyn inclusions and reduce aSyn toxicity. Moreover, we found that endocytic recycling and secretion of aSyn was enhanced upon expression of Rab11a or Rab13 in cells accumulating aSyn inclusions. Altogether, our findings suggest specific trafficking steps may prove beneficial as targets for therapeutic intervention in synucleinopathies, and should be further investigated in other models.
Collapse
Affiliation(s)
- Susana A Gonçalves
- a CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas , Universidade NOVA de Lisboa , Lisboa , Portugal
| | - Tiago Fleming Outeiro
- a CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas , Universidade NOVA de Lisboa , Lisboa , Portugal.,b Department of Neurodegeneration and Restorative Research , University Medical Center Göttingen , Göttingen , Germany.,c Max Planck Institute for Experimental Medicine , Göttingen , Germany
| |
Collapse
|
37
|
Gonçalves SA, Macedo D, Raquel H, Simões PD, Giorgini F, Ramalho JS, Barral DC, Ferreira Moita L, Outeiro TF. shRNA-Based Screen Identifies Endocytic Recycling Pathway Components That Act as Genetic Modifiers of Alpha-Synuclein Aggregation, Secretion and Toxicity. PLoS Genet 2016; 12:e1005995. [PMID: 27123591 PMCID: PMC4849646 DOI: 10.1371/journal.pgen.1005995] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 03/28/2016] [Indexed: 12/15/2022] Open
Abstract
Alpha-Synuclein (aSyn) misfolding and aggregation is common in several neurodegenerative diseases, including Parkinson's disease and dementia with Lewy bodies, which are known as synucleinopathies. Accumulating evidence suggests that secretion and cell-to-cell trafficking of pathological forms of aSyn may explain the typical patterns of disease progression. However, the molecular mechanisms controlling aSyn aggregation and spreading of pathology are still elusive. In order to obtain unbiased information about the molecular regulators of aSyn oligomerization, we performed a microscopy-based large-scale RNAi screen in living cells. Interestingly, we identified nine Rab GTPase and kinase genes that modulated aSyn aggregation, toxicity and levels. From those, Rab8b, Rab11a, Rab13 and Slp5 were able to promote the clearance of aSyn inclusions and rescue aSyn induced toxicity. Furthermore, we found that endocytic recycling and secretion of aSyn was enhanced upon Rab11a and Rab13 expression in cells accumulating aSyn inclusions. Overall, our study resulted in the identification of new molecular players involved in the aggregation, toxicity, and secretion of aSyn, opening novel avenues for our understanding of the molecular basis of synucleinopathies.
Collapse
Affiliation(s)
- Susana A. Gonçalves
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Lisboa, Portugal
- CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Diana Macedo
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Lisboa, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de lisboa, Oeiras, Portugal
| | | | - Pedro D. Simões
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Lisboa, Portugal
| | - Flaviano Giorgini
- Department of Genetics, University of Leicester, Leicester, United Kingdom
| | - José S. Ramalho
- CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Duarte C. Barral
- CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
| | | | - Tiago Fleming Outeiro
- CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
- Department of Neurodegeneration and Restorative Research, University Medical Center Göttingen, Göttingen, Germany
- Max Planck Institute for Experimental Medicine, Göttingen, Germany
| |
Collapse
|
38
|
Goullee H, Wadley AL, Cherry CL, Allcock RJN, Black M, Kamerman PR, Price P. Polymorphisms in CAMKK2 may predict sensory neuropathy in African HIV patients. J Neurovirol 2016; 22:508-17. [PMID: 26785644 DOI: 10.1007/s13365-015-0421-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Revised: 12/18/2015] [Accepted: 12/21/2015] [Indexed: 01/06/2023]
Abstract
HIV-associated sensory neuropathy (HIV-SN) is the most common neurological condition associated with HIV. HIV-SN has characteristics of an inflammatory pathology caused by the virus itself and/or by antiretroviral treatment (ART). Here, we assess the impact of single-nucleotide polymorphisms (SNPs) in a cluster of three genes that affect inflammation and neuronal repair: P2X7R, P2X4R and CAMKK2. HIV-SN status was assessed using the Brief Peripheral Neuropathy Screening tool, with SN defined by bilateral symptoms and signs. Forty-five SNPs in P2X7R, P2X4R and CAMKK2 were genotyped using TaqMan fluorescent probes, in DNA samples from 153 HIV(+) black Southern African patients exposed to stavudine. Haplotypes were derived using the fastPHASE algorithm, and SNP genotypes and haplotypes associated with HIV-SN were identified. Optimal logistic regression models included demographics (age and height), with SNPs (model p < 0.0001; R (2) = 0.19) or haplotypes (model p < 0.0001; R (2) = 0.18, n = 137 excluding patients carrying CAMKK2 haplotypes perfectly associated with SN). Overall, CAMKK2 exhibited the strongest associations with HIV-SN, with two SNPs and six haplotypes predicting SN status in black Southern Africans. This gene warrants further study.
Collapse
Affiliation(s)
- Hayley Goullee
- School of Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia
| | - Antonia L Wadley
- Brain Function Research Group, School of Physiology, University of the Witwatersrand, Johannesburg, South Africa
| | - Catherine L Cherry
- Brain Function Research Group, School of Physiology, University of the Witwatersrand, Johannesburg, South Africa.,Department of Infectious Diseases, Alfred Hospital and Monash University, and Centre for Biomedical Research, Burnet Institute, Melbourne, Australia
| | - Richard J N Allcock
- School of Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia
| | - Michael Black
- Centre for Comparative Genomics, Murdoch University, Perth, Australia
| | - Peter R Kamerman
- Brain Function Research Group, School of Physiology, University of the Witwatersrand, Johannesburg, South Africa
| | - Patricia Price
- Brain Function Research Group, School of Physiology, University of the Witwatersrand, Johannesburg, South Africa. .,School of Biomedical Science, Curtin University of Technology, Bentley, Western Australia, 6845, Australia.
| |
Collapse
|
39
|
Horigane SI, Ageta-Ishihara N, Kamijo S, Fujii H, Okamura M, Kinoshita M, Takemoto-Kimura S, Bito H. Facilitation of axon outgrowth via a Wnt5a-CaMKK-CaMKIα pathway during neuronal polarization. Mol Brain 2016; 9:8. [PMID: 26772170 PMCID: PMC4715351 DOI: 10.1186/s13041-016-0189-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 01/10/2016] [Indexed: 11/10/2022] Open
Abstract
Background Wnt5a, originally identified as a guidance cue for commissural axons, activates a non-canonical pathway critical for cortical axonal morphogenesis. The molecular signaling cascade underlying this event remains obscure. Results Through Ca2+ imaging in acute embryonic cortical slices, we tested if radially migrating cortical excitatory neurons that already bore primitive axons were sensitive to Wnt5a. While Wnt5a only evoked brief Ca2+ transients in immature neurons present in the intermediate zone (IZ), Wnt5a-induced Ca2+ oscillations were sustained in neurons that migrated out to the cortical plate (CP). We wondered whether this early Wnt5a-Ca2+ signaling during neuronal polarization has a morphogenetic consequence. During transition from round to polarized shape, Wnt5a administration to immature cultured cortical neurons specifically promoted axonal, but not dendritic, outgrowth. Pharmacological and genetic inhibition of the CaMKK-CaMKIα pathway abolished Wnt5a-induced axonal elongation, and rescue of CaMKIα in CaMKIα-knockdown neurons restored Wnt5a-mediated axon outgrowth. Conclusions This study suggests that Wnt5a activates Ca2+ signaling during a neuronal morphogenetic time window when axon outgrowth is critically facilitated. Furthermore, the CaMKK-CaMKIα cascade is required for the axonal growth effect of Wnt5a during neuronal polarization. Electronic supplementary material The online version of this article (doi:10.1186/s13041-016-0189-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shin-ichiro Horigane
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan.
| | - Natsumi Ageta-Ishihara
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Department of Molecular Biology, Division of Biological Sciences, Nagoya University Graduate School of Science, Furo-cho, Chikusa, Nagoya, 464-8602, Japan.
| | - Satoshi Kamijo
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, 100-0004, Japan.
| | - Hajime Fujii
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, 100-0004, Japan.
| | - Michiko Okamura
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Makoto Kinoshita
- Department of Molecular Biology, Division of Biological Sciences, Nagoya University Graduate School of Science, Furo-cho, Chikusa, Nagoya, 464-8602, Japan.
| | - Sayaka Takemoto-Kimura
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan. .,PRESTO, Japan Science and Technology Agency, Chiyoda-ku, Tokyo, 102-0076, Japan.
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, 100-0004, Japan.
| |
Collapse
|
40
|
Namba T, Funahashi Y, Nakamuta S, Xu C, Takano T, Kaibuchi K. Extracellular and Intracellular Signaling for Neuronal Polarity. Physiol Rev 2015; 95:995-1024. [PMID: 26133936 DOI: 10.1152/physrev.00025.2014] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neurons are one of the highly polarized cells in the body. One of the fundamental issues in neuroscience is how neurons establish their polarity; therefore, this issue fascinates many scientists. Cultured neurons are useful tools for analyzing the mechanisms of neuronal polarization, and indeed, most of the molecules important in their polarization were identified using culture systems. However, we now know that the process of neuronal polarization in vivo differs in some respects from that in cultured neurons. One of the major differences is their surrounding microenvironment; neurons in vivo can be influenced by extrinsic factors from the microenvironment. Therefore, a major question remains: How are neurons polarized in vivo? Here, we begin by reviewing the process of neuronal polarization in culture conditions and in vivo. We also survey the molecular mechanisms underlying neuronal polarization. Finally, we introduce the theoretical basis of neuronal polarization and the possible involvement of neuronal polarity in disease and traumatic brain injury.
Collapse
Affiliation(s)
- Takashi Namba
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Funahashi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinichi Nakamuta
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Chundi Xu
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsuya Takano
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
41
|
Fujiwara Y, Hiraoka Y, Fujimoto T, Kanayama N, Magari M, Tokumitsu H. Analysis of Distinct Roles of CaMKK Isoforms Using STO-609-Resistant Mutants in Living Cells. Biochemistry 2015; 54:3969-77. [DOI: 10.1021/acs.biochem.5b00149] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Yuya Fujiwara
- Division
of Medical Bioengineering, Graduate School of Natural Science and
Technology, Okayama University, Okayama 700-8530, Japan
| | - Yuri Hiraoka
- Division
of Medical Bioengineering, Graduate School of Natural Science and
Technology, Okayama University, Okayama 700-8530, Japan
| | | | - Naoki Kanayama
- Division
of Medical Bioengineering, Graduate School of Natural Science and
Technology, Okayama University, Okayama 700-8530, Japan
| | - Masaki Magari
- Division
of Medical Bioengineering, Graduate School of Natural Science and
Technology, Okayama University, Okayama 700-8530, Japan
| | - Hiroshi Tokumitsu
- Division
of Medical Bioengineering, Graduate School of Natural Science and
Technology, Okayama University, Okayama 700-8530, Japan
| |
Collapse
|
42
|
Uhlén P, Fritz N, Smedler E, Malmersjö S, Kanatani S. Calcium signaling in neocortical development. Dev Neurobiol 2015; 75:360-8. [DOI: 10.1002/dneu.22273] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 01/16/2015] [Accepted: 01/17/2015] [Indexed: 11/10/2022]
Affiliation(s)
- Per Uhlén
- Department of Medical Biochemistry and Biophysics; Karolinska Institutet; SE-171 77 Stockholm Sweden
| | - Nicolas Fritz
- The Science for Life Laboratory; The Royal Institute of Technology; SE-171 77 Stockholm Sweden
| | - Erik Smedler
- Department of Medical Biochemistry and Biophysics; Karolinska Institutet; SE-171 77 Stockholm Sweden
| | - Seth Malmersjö
- Department of Chemical and Systems Biology; School of Medicine, Stanford University; Stanford California 94305
| | - Shigeaki Kanatani
- Department of Medical Biochemistry and Biophysics; Karolinska Institutet; SE-171 77 Stockholm Sweden
| |
Collapse
|
43
|
Inoue M, Takeuchi A, Horigane SI, Ohkura M, Gengyo-Ando K, Fujii H, Kamijo S, Takemoto-Kimura S, Kano M, Nakai J, Kitamura K, Bito H. Rational design of a high-affinity, fast, red calcium indicator R-CaMP2. Nat Methods 2014; 12:64-70. [PMID: 25419959 DOI: 10.1038/nmeth.3185] [Citation(s) in RCA: 195] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 10/25/2014] [Indexed: 12/11/2022]
Abstract
Fluorescent Ca(2+) reporters are widely used as readouts of neuronal activities. Here we designed R-CaMP2, a high-affinity red genetically encoded calcium indicator (GECI) with a Hill coefficient near 1. Use of the calmodulin-binding sequence of CaMKK-α and CaMKK-β in lieu of an M13 sequence resulted in threefold faster rise and decay times of Ca(2+) transients than R-CaMP1.07. These features allowed resolving single action potentials (APs) and recording fast AP trains up to 20-40 Hz in cortical slices. Somatic and synaptic activities of a cortical neuronal ensemble in vivo were imaged with similar efficacy as with previously reported sensitive green GECIs. Combining green and red GECIs, we successfully achieved dual-color monitoring of neuronal activities of distinct cell types, both in the mouse cortex and in freely moving Caenorhabditis elegans. Dual imaging using R-CaMP2 and green GECIs provides a powerful means to interrogate orthogonal and hierarchical neuronal ensembles in vivo.
Collapse
Affiliation(s)
- Masatoshi Inoue
- 1] Department of Neurochemistry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan. [2] Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo, Japan
| | - Atsuya Takeuchi
- Department of Neurophysiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Shin-ichiro Horigane
- Department of Neurochemistry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | | | | - Hajime Fujii
- Department of Neurochemistry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Satoshi Kamijo
- 1] Department of Neurochemistry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan. [2] Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo, Japan
| | - Sayaka Takemoto-Kimura
- 1] Department of Neurochemistry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan. [2] Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Saitama, Japan
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Junichi Nakai
- Brain Science Institute, Saitama University, Saitama, Japan
| | - Kazuo Kitamura
- 1] Department of Neurophysiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan. [2] Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Saitama, Japan
| | - Haruhiko Bito
- 1] Department of Neurochemistry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan. [2] Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo, Japan
| |
Collapse
|
44
|
Nonaka M, Kim R, Fukushima H, Sasaki K, Suzuki K, Okamura M, Ishii Y, Kawashima T, Kamijo S, Takemoto-Kimura S, Okuno H, Kida S, Bito H. Region-Specific Activation of CRTC1-CREB Signaling Mediates Long-Term Fear Memory. Neuron 2014; 84:92-106. [DOI: 10.1016/j.neuron.2014.08.049] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2014] [Indexed: 11/29/2022]
|
45
|
Hyun TK, Lee S, Kumar D, Rim Y, Kumar R, Lee SY, Lee CH, Kim JY. RNA-seq analysis of Rubus idaeus cv. Nova: transcriptome sequencing and de novo assembly for subsequent functional genomics approaches. PLANT CELL REPORTS 2014; 33:1617-28. [PMID: 25023872 DOI: 10.1007/s00299-014-1641-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 05/19/2014] [Accepted: 05/28/2014] [Indexed: 05/09/2023]
Abstract
Using Illumina sequencing technology, we have generated the large-scale transcriptome sequencing data containing abundant information on genes involved in the metabolic pathways in R. idaeus cv. Nova fruits. Rubus idaeus (Red raspberry) is one of the important economical crops that possess numerous nutrients, micronutrients and phytochemicals with essential health benefits to human. The molecular mechanism underlying the ripening process and phytochemical biosynthesis in red raspberry is attributed to the changes in gene expression, but very limited transcriptomic and genomic information in public databases is available. To address this issue, we generated more than 51 million sequencing reads from R. idaeus cv. Nova fruit using Illumina RNA-Seq technology. After de novo assembly, we obtained 42,604 unigenes with an average length of 812 bp. At the protein level, Nova fruit transcriptome showed 77 and 68 % sequence similarities with Rubus coreanus and Fragaria versa, respectively, indicating the evolutionary relationship between them. In addition, 69 % of assembled unigenes were annotated using public databases including NCBI non-redundant, Cluster of Orthologous Groups and Gene ontology database, suggesting that our transcriptome dataset provides a valuable resource for investigating metabolic processes in red raspberry. To analyze the relationship between several novel transcripts and the amounts of metabolites such as γ-aminobutyric acid and anthocyanins, real-time PCR and target metabolite analysis were performed on two different ripening stages of Nova. This is the first attempt using Illumina sequencing platform for RNA sequencing and de novo assembly of Nova fruit without reference genome. Our data provide the most comprehensive transcriptome resource available for Rubus fruits, and will be useful for understanding the ripening process and for breeding R. idaeus cultivars with improved fruit quality.
Collapse
Affiliation(s)
- Tae Kyung Hyun
- Division of Applied Life Science (BK21plus/WCU Program), Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Neuronal polarization in vivo: Growing in a complex environment. Curr Opin Neurobiol 2014; 27:215-23. [DOI: 10.1016/j.conb.2014.04.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 04/10/2014] [Accepted: 04/10/2014] [Indexed: 01/06/2023]
|
47
|
Ageta-Ishihara N, Miyata T, Ohshima C, Watanabe M, Sato Y, Hamamura Y, Higashiyama T, Mazitschek R, Bito H, Kinoshita M. Septins promote dendrite and axon development by negatively regulating microtubule stability via HDAC6-mediated deacetylation. Nat Commun 2014; 4:2532. [PMID: 24113571 PMCID: PMC3826633 DOI: 10.1038/ncomms3532] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 09/02/2013] [Indexed: 11/11/2022] Open
Abstract
Neurite growth requires two guanine nucleotide-binding protein polymers of tubulins and septins. However, whether and how those cytoskeletal systems are coordinated was unknown. Here we show that the acute knockdown or knockout of the pivotal septin subunit SEPT7 from cerebrocortical neurons impairs their interhemispheric and cerebrospinal axon projections and dendritogenesis in perinatal mice, when the microtubules are severely hyperacetylated. The resulting hyperstabilization and growth retardation of microtubules are demonstrated in vitro. The phenotypic similarity between SEPT7 depletion and the pharmacological inhibition of α-tubulin deacetylase HDAC6 reveals that HDAC6 requires SEPT7 not for its enzymatic activity, but to associate with acetylated α-tubulin. These and other findings indicate that septins provide a physical scaffold for HDAC6 to achieve efficient microtubule deacetylation, thereby negatively regulating microtubule stability to an optimal level for neuritogenesis. Our findings shed light on the mechanisms underlying the HDAC6-mediated coupling of the two ubiquitous cytoskeletal systems during neural development. Septins are a family of heteropolymerizing GTP/GDP-binding proteins and are implicated in neuritogenesis in nematodes. Ageta-Ishihara et al. show that septins also facilitate this process in the developing mouse brain as scaffolds that coordinate HDAC6-mediated deacetylation of microtubules.
Collapse
Affiliation(s)
- Natsumi Ageta-Ishihara
- Division of Biological Sciences, Nagoya University Graduate School of Science, Nagoya 464-8602, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Namba T, Kibe Y, Funahashi Y, Nakamuta S, Takano T, Ueno T, Shimada A, Kozawa S, Okamoto M, Shimoda Y, Oda K, Wada Y, Masuda T, Sakakibara A, Igarashi M, Miyata T, Faivre-Sarrailh C, Takeuchi K, Kaibuchi K. Pioneering axons regulate neuronal polarization in the developing cerebral cortex. Neuron 2014; 81:814-29. [PMID: 24559674 DOI: 10.1016/j.neuron.2013.12.015] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2013] [Indexed: 01/06/2023]
Abstract
The polarization of neurons, which mainly includes the differentiation of axons and dendrites, is regulated by cell-autonomous and non-cell-autonomous factors. In the developing central nervous system, neuronal development occurs in a heterogeneous environment that also comprises extracellular matrices, radial glial cells, and neurons. Although many cell-autonomous factors that affect neuronal polarization have been identified, the microenvironmental cues involved in neuronal polarization remain largely unknown. Here, we show that neuronal polarization occurs in a microenvironment in the lower intermediate zone, where the cell adhesion molecule transient axonal glycoprotein-1 (TAG-1) is expressed in cortical efferent axons. The immature neurites of multipolar cells closely contact TAG-1-positive axons and generate axons. Inhibition of TAG-1-mediated cell-to-cell interaction or its downstream kinase Lyn impairs neuronal polarization. These results show that the TAG-1-mediated cell-to-cell interaction between the unpolarized multipolar cells and the pioneering axons regulates the polarization of multipolar cells partly through Lyn kinase and Rac1.
Collapse
Affiliation(s)
- Takashi Namba
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Yuji Kibe
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Yasuhiro Funahashi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Shinichi Nakamuta
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Tetsuya Takano
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Takuji Ueno
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Akiko Shimada
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Sachi Kozawa
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Mayumi Okamoto
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Yasushi Shimoda
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomiokamachi, Nagaoka, Niigata 940-2188, Japan
| | - Kanako Oda
- Experimental Animal Resource, Brain Research Institute, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahi-machi, Chuo-ku, Niigata, Niigata 951-8510, Japan
| | - Yoshino Wada
- Division of Molecular and Cellular Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahi-machi, Chuo-ku, Niigata, Niigata 951-8510, Japan
| | - Tomoyuki Masuda
- Department of Neurobiology, University of Tsukuba School of Medicine, Ibaraki 305-8577, Japan
| | - Akira Sakakibara
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Michihiro Igarashi
- Division of Molecular and Cellular Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahi-machi, Chuo-ku, Niigata, Niigata 951-8510, Japan
| | - Takaki Miyata
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Catherine Faivre-Sarrailh
- Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, UMR 7286 CNRS, Marseille, France
| | - Kosei Takeuchi
- Division of Molecular and Cellular Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahi-machi, Chuo-ku, Niigata, Niigata 951-8510, Japan; Department of Biology, School of Medicine, Aichi Medical University, Yazako, Nagakute, Aichi 480-1195, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan.
| |
Collapse
|
49
|
Thong JYJ, Qiu A, Sum MY, Kuswanto CN, Tuan TA, Donohoe G, Sitoh YY, Sim K. Effects of the neurogranin variant rs12807809 on thalamocortical morphology in schizophrenia. PLoS One 2013; 8:e85603. [PMID: 24386483 PMCID: PMC3875583 DOI: 10.1371/journal.pone.0085603] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 11/28/2013] [Indexed: 12/31/2022] Open
Abstract
Although the genome wide supported psychosis susceptibility neurogranin (NRGN) gene is expressed in human brains, it is unclear how it impacts brain morphology in schizophrenia. We investigated the influence of NRGN rs12807809 on cortical thickness, subcortical volumes and shapes in patients with schizophrenia. One hundred and fifty six subjects (91 patients with schizophrenia and 65 healthy controls) underwent structural MRI scans and their blood samples were genotyped. A brain mapping algorithm, large deformation diffeomorphic metric mapping, was used to perform group analysis of subcortical shapes and cortical thickness. Patients with risk TT genotype were associated with widespread cortical thinning involving frontal, parietal and temporal cortices compared with controls with TT genotype. No volumetric difference in subcortical structures (hippocampus, thalamus, amygdala, basal ganglia) was observed between risk TT genotype in patients and controls. However, patients with risk TT genotype were associated with thalamic shape abnormalities involving regions related to pulvinar and medial dorsal nuclei. Our results revealed the influence of the NRGN gene on thalamocortical morphology in schizophrenia involving widespread cortical thinning and thalamic shape abnormalities. These findings help to clarify underlying NRGN mediated pathophysiological mechanisms involving cortical-subcortical brain networks in schizophrenia.
Collapse
Affiliation(s)
- Jamie Yu Jin Thong
- Department of Bioengineering, National University of Singapore, Singapore
| | - Anqi Qiu
- Department of Bioengineering, National University of Singapore, Singapore
- Clinical Imaging Research Center, National University of Singapore, Singapore
- Singapore Institute for Clinical Sciences, the Agency for Science, Technology and Research, Singapore
- * E-mail:
| | - Min Yi Sum
- Research Division, Institute of Mental Health, Singapore
| | | | - Ta Ahn Tuan
- Department of Bioengineering, National University of Singapore, Singapore
| | - Gary Donohoe
- Department of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Republic of Ireland
| | - Yih Yian Sitoh
- Department of Neuroradiology, National Neuroscience Institute, Singapore
| | - Kang Sim
- Research Division, Institute of Mental Health, Singapore
- Department of General Psychiatry, Institute of Mental Health, Singapore
| |
Collapse
|
50
|
Riascos D, Nicholas A, Samaeekia R, Yukhananov R, Mesulam MM, Bigio EH, Weintraub S, Guo L, Geula C. Alterations of Ca²⁺-responsive proteins within cholinergic neurons in aging and Alzheimer's disease. Neurobiol Aging 2013; 35:1325-33. [PMID: 24461366 DOI: 10.1016/j.neurobiolaging.2013.12.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 12/07/2013] [Accepted: 12/19/2013] [Indexed: 01/05/2023]
Abstract
The molecular basis of selective neuronal vulnerability in Alzheimer's disease (AD) remains poorly understood. Using basal forebrain cholinergic neurons (BFCNs) as a model and immunohistochemistry, we have demonstrated significant age-related loss of the calcium-binding protein calbindin-D(28K) (CB) from BFCN, which was associated with tangle formation and degeneration in AD. Here, we determined alterations in RNA and protein for CB and the Ca(2+)-responsive proteins Ca(2+)/calmodulin-dependent protein kinase I (CaMKI), growth-associated protein-43 (GAP43), and calpain in the BF. We observed progressive downregulation of CB and CaMKI RNA in laser-captured BFCN in the normal-aged-AD continuum. We also detected progressive loss of CB, CaMKIδ, and GAP43 proteins in BF homogenates in aging and AD. Activated μ-calpain, a calcium-sensitive protease that degrades CaMKI and GAP43, was significantly increased in the normal aged BF and was 10 times higher in AD BF. Overactivation of μ-calpain was confirmed using proteolytic fragments of its substrate spectrin. Substantial age- and AD-related alterations in Ca(2+)-sensing proteins most likely contribute to selective vulnerability of BFCN to degeneration in AD.
Collapse
Affiliation(s)
- David Riascos
- Cognitive Neurology and Alzheimer's Disease Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Alexander Nicholas
- Department of Medicine, Harvard Medical School and Division of Gerontology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Ravand Samaeekia
- Cognitive Neurology and Alzheimer's Disease Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - M-Marsel Mesulam
- Cognitive Neurology and Alzheimer's Disease Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Eileen H Bigio
- Cognitive Neurology and Alzheimer's Disease Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Sandra Weintraub
- Cognitive Neurology and Alzheimer's Disease Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ling Guo
- Cognitive Neurology and Alzheimer's Disease Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Changiz Geula
- Cognitive Neurology and Alzheimer's Disease Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|