1
|
Pays E. The Janus-faced functions of Apolipoproteins L in membrane dynamics. Cell Mol Life Sci 2024; 81:134. [PMID: 38478101 PMCID: PMC10937811 DOI: 10.1007/s00018-024-05180-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/06/2024] [Accepted: 02/18/2024] [Indexed: 03/17/2024]
Abstract
The functions of human Apolipoproteins L (APOLs) are poorly understood, but involve diverse activities like lysis of bloodstream trypanosomes and intracellular bacteria, modulation of viral infection and induction of apoptosis, autophagy, and chronic kidney disease. Based on recent work, I propose that the basic function of APOLs is the control of membrane dynamics, at least in the Golgi and mitochondrion. Together with neuronal calcium sensor-1 (NCS1) and calneuron-1 (CALN1), APOL3 controls the activity of phosphatidylinositol-4-kinase-IIIB (PI4KB), involved in both Golgi and mitochondrion membrane fission. Whereas secreted APOL1 induces African trypanosome lysis through membrane permeabilization of the parasite mitochondrion, intracellular APOL1 conditions non-muscular myosin-2A (NM2A)-mediated transfer of PI4KB and APOL3 from the Golgi to the mitochondrion under conditions interfering with PI4KB-APOL3 interaction, such as APOL1 C-terminal variant expression or virus-induced inflammatory signalling. APOL3 controls mitophagy through complementary interactions with the membrane fission factor PI4KB and the membrane fusion factor vesicle-associated membrane protein-8 (VAMP8). In mice, the basic APOL1 and APOL3 activities could be exerted by mAPOL9 and mAPOL8, respectively. Perspectives regarding the mechanism and treatment of APOL1-related kidney disease are discussed, as well as speculations on additional APOLs functions, such as APOL6 involvement in adipocyte membrane dynamics through interaction with myosin-10 (MYH10).
Collapse
Affiliation(s)
- Etienne Pays
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041, Gosselies, Belgium.
| |
Collapse
|
2
|
Lisek M, Tomczak J, Boczek T, Zylinska L. Calcium-Associated Proteins in Neuroregeneration. Biomolecules 2024; 14:183. [PMID: 38397420 PMCID: PMC10887043 DOI: 10.3390/biom14020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/27/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
The dysregulation of intracellular calcium levels is a critical factor in neurodegeneration, leading to the aberrant activation of calcium-dependent processes and, ultimately, cell death. Ca2+ signals vary in magnitude, duration, and the type of neuron affected. A moderate Ca2+ concentration can initiate certain cellular repair pathways and promote neuroregeneration. While the peripheral nervous system exhibits an intrinsic regenerative capability, the central nervous system has limited self-repair potential. There is evidence that significant variations exist in evoked calcium responses and axonal regeneration among neurons, and individual differences in regenerative capacity are apparent even within the same type of neurons. Furthermore, some studies have shown that neuronal activity could serve as a potent regulator of this process. The spatio-temporal patterns of calcium dynamics are intricately controlled by a variety of proteins, including channels, ion pumps, enzymes, and various calcium-binding proteins, each of which can exert either positive or negative effects on neural repair, depending on the cellular context. In this concise review, we focus on several calcium-associated proteins such as CaM kinase II, GAP-43, oncomodulin, caldendrin, calneuron, and NCS-1 in order to elaborate on their roles in the intrinsic mechanisms governing neuronal regeneration following traumatic damage processes.
Collapse
Affiliation(s)
| | | | | | - Ludmila Zylinska
- Department of Molecular Neurochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (M.L.); (J.T.); (T.B.)
| |
Collapse
|
3
|
Li G, Wu Y, Zhang Y, Wang H, Li M, He D, Guan W, Yao H. Research progress on phosphatidylinositol 4-kinase inhibitors. Biochem Pharmacol 2024; 220:115993. [PMID: 38151075 DOI: 10.1016/j.bcp.2023.115993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 12/29/2023]
Abstract
Phosphatidylinositol 4-kinases (PI4Ks) could phosphorylate phosphatidylinositol (PI) to produce phosphatidylinositol 4-phosphate (PI4P) and maintain its metabolic balance and location. PI4P, the most abundant monophosphate inositol in eukaryotic cells, is a precursor of higher phosphoinositols and an essential substrate for the PLC/PKC and PI3K/Akt signaling pathways. PI4Ks regulate vesicle transport, signal transduction, cytokinesis, and cell unity, and are involved in various physiological and pathological processes, including infection and growth of parasites such as Plasmodium and Cryptosporidium, replication and survival of RNA viruses, and the development of tumors and nervous system diseases. The development of novel drugs targeting PI4Ks and PI4P has been the focus of the research and clinical application of drugs, especially in recent years. In particular, PI4K inhibitors have made great progress in the treatment of malaria and cryptosporidiosis. We describe the biological characteristics of PI4Ks; summarize the physiological functions and effector proteins of PI4P; and analyze the structural basis of selective PI4K inhibitors for the treatment of human diseases in this review. Herein, this review mainly summarizes the developments in the structure and enzyme activity of PI4K inhibitors.
Collapse
Affiliation(s)
- Gang Li
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, 510260, China
| | - Yanting Wu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, 510260, China; Department of Chemistry, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong, 999077, China
| | - Yali Zhang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, 510260, China
| | - Huamin Wang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, 510260, China
| | - Mengjie Li
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, 510260, China
| | - Dengqin He
- School of Biotechnology and Health Science, Wuyi University, 22 Dongchengcun, Jiangmen, Guangdong, 529020, China
| | - Wen Guan
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, 510260, China
| | - Hongliang Yao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, 510260, China.
| |
Collapse
|
4
|
Lecordier L, Heo P, Graversen JH, Hennig D, Skytthe MK, Cornet d'Elzius A, Pincet F, Pérez-Morga D, Pays E. Apolipoproteins L1 and L3 control mitochondrial membrane dynamics. Cell Rep 2023; 42:113528. [PMID: 38041817 PMCID: PMC10765320 DOI: 10.1016/j.celrep.2023.113528] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/08/2023] [Accepted: 11/17/2023] [Indexed: 12/04/2023] Open
Abstract
Apolipoproteins L1 and L3 (APOLs) are associated at the Golgi with the membrane fission factors phosphatidylinositol 4-kinase-IIIB (PI4KB) and non-muscular myosin 2A. Either APOL1 C-terminal truncation (APOL1Δ) or APOL3 deletion (APOL3-KO [knockout]) reduces PI4KB activity and triggers actomyosin reorganization. We report that APOL3, but not APOL1, controls PI4KB activity through interaction with PI4KB and neuronal calcium sensor-1 or calneuron-1. Both APOLs are present in Golgi-derived autophagy-related protein 9A vesicles, which are involved in PI4KB trafficking. Like APOL3-KO, APOL1Δ induces PI4KB dissociation from APOL3, linked to reduction of mitophagy flux and production of mitochondrial reactive oxygen species. APOL1 and APOL3, respectively, can interact with the mitophagy receptor prohibitin-2 and the mitophagosome membrane fusion factor vesicle-associated membrane protein-8 (VAMP8). While APOL1 conditions PI4KB and APOL3 involvement in mitochondrion fission and mitophagy, APOL3-VAMP8 interaction promotes fusion between mitophagosomal and endolysosomal membranes. We propose that APOL3 controls mitochondrial membrane dynamics through interactions with the fission factor PI4KB and the fusion factor VAMP8.
Collapse
Affiliation(s)
- Laurence Lecordier
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Paul Heo
- Laboratoire de Physique de l'Ecole Normale Supérieure, Ecole Normale Supérieure (ENS), Université Paris Sciences et Lettres (PSL), CNRS, Sorbonne Université, Université Paris-Cité, 75005 Paris, France; Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Jonas H Graversen
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Dorle Hennig
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Maria Kløjgaard Skytthe
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense C, Denmark
| | | | - Frédéric Pincet
- Laboratoire de Physique de l'Ecole Normale Supérieure, Ecole Normale Supérieure (ENS), Université Paris Sciences et Lettres (PSL), CNRS, Sorbonne Université, Université Paris-Cité, 75005 Paris, France
| | - David Pérez-Morga
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium; Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Etienne Pays
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium.
| |
Collapse
|
5
|
Muñoz-Reyes D, McClelland LJ, Arroyo-Urea S, Sánchez-Yepes S, Sabín J, Pérez-Suárez S, Menendez M, Mansilla A, García-Nafría J, Sprang S, Sanchez-Barrena MJ. The neuronal calcium sensor NCS-1 regulates the phosphorylation state and activity of the Gα chaperone and GEF Ric-8A. eLife 2023; 12:e86151. [PMID: 38018500 PMCID: PMC10732572 DOI: 10.7554/elife.86151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 11/24/2023] [Indexed: 11/30/2023] Open
Abstract
The neuronal calcium sensor 1 (NCS-1), an EF-hand Ca2+ binding protein, and Ric-8A coregulate synapse number and probability of neurotransmitter release. Recently, the structures of Ric-8A bound to Gα have revealed how Ric-8A phosphorylation promotes Gα recognition and activity as a chaperone and guanine nucleotide exchange factor. However, the molecular mechanism by which NCS-1 regulates Ric-8A activity and its interaction with Gα subunits is not well understood. Given the interest in the NCS-1/Ric-8A complex as a therapeutic target in nervous system disorders, it is necessary to shed light on this molecular mechanism of action at atomic level. We have reconstituted NCS-1/Ric-8A complexes to conduct a multimodal approach and determine the sequence of Ca2+ signals and phosphorylation events that promote the interaction of Ric-8A with Gα. Our data show that the binding of NCS-1 and Gα to Ric-8A are mutually exclusive. Importantly, NCS-1 induces a structural rearrangement in Ric-8A that traps the protein in a conformational state that is inaccessible to casein kinase II-mediated phosphorylation, demonstrating one aspect of its negative regulation of Ric-8A-mediated G-protein signaling. Functional experiments indicate a loss of Ric-8A guanine nucleotide exchange factor (GEF) activity toward Gα when complexed with NCS-1, and restoration of nucleotide exchange activity upon increasing Ca2+ concentration. Finally, the high-resolution crystallographic data reported here define the NCS-1/Ric-8A interface and will allow the development of therapeutic synapse function regulators with improved activity and selectivity.
Collapse
Affiliation(s)
- Daniel Muñoz-Reyes
- Department of Crystallography and Structural Biology, Institute of Physical-Chemistry 'Blas Cabrera', CSICMadridSpain
| | - Levi J McClelland
- Center for Biomolecular Structure and Dynamics, and Division of Biological Sciences, University of MontanaMissoulaUnited States
| | - Sandra Arroyo-Urea
- Institute for Biocomputation and Physics of Complex Systems (BIFI) and Laboratorio de Microscopías Avanzadas (LMA), University of ZaragozaZaragozaSpain
| | - Sonia Sánchez-Yepes
- Department of Neurobiology, Instituto Ramón y Cajal de Investigación Sanitaria, Hospital Universitario Ramón y CajalMadridSpain
| | - Juan Sabín
- AFFINImeter Scientific & Development team, Software 4 Science DevelopmentsSantiago de CompostelaSpain
- Departamento de Física Aplicada, Universidad de Santiago de CompostelaSantiago de CompostelaSpain
| | - Sara Pérez-Suárez
- Department of Crystallography and Structural Biology, Institute of Physical-Chemistry 'Blas Cabrera', CSICMadridSpain
| | - Margarita Menendez
- Department of Biological Physical-Chemisty, Institute of Physical-Chemistry 'Blas Cabrera', CSICMadridSpain
- Ciber of Respiratory Diseases, ISCIIIMadridSpain
| | - Alicia Mansilla
- Department of Neurobiology, Instituto Ramón y Cajal de Investigación Sanitaria, Hospital Universitario Ramón y CajalMadridSpain
- Department of Systems Biology, Universidad de AlcalaMadridSpain
| | - Javier García-Nafría
- Institute for Biocomputation and Physics of Complex Systems (BIFI) and Laboratorio de Microscopías Avanzadas (LMA), University of ZaragozaZaragozaSpain
| | - Stephen Sprang
- Center for Biomolecular Structure and Dynamics, and Division of Biological Sciences, University of MontanaMissoulaUnited States
| | - Maria Jose Sanchez-Barrena
- Department of Crystallography and Structural Biology, Institute of Physical-Chemistry 'Blas Cabrera', CSICMadridSpain
| |
Collapse
|
6
|
Chen M, Xu L, Wu Y, Soba P, Hu C. The organization and function of the Golgi apparatus in dendrite development and neurological disorders. Genes Dis 2023; 10:2425-2442. [PMID: 37554209 PMCID: PMC10404969 DOI: 10.1016/j.gendis.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/13/2022] [Accepted: 11/05/2022] [Indexed: 12/24/2022] Open
Abstract
Dendrites are specialized neuronal compartments that sense, integrate and transfer information in the neural network. Their development is tightly controlled and abnormal dendrite morphogenesis is strongly linked to neurological disorders. While dendritic morphology ranges from relatively simple to extremely complex for a specified neuron, either requires a functional secretory pathway to continually replenish proteins and lipids to meet dendritic growth demands. The Golgi apparatus occupies the center of the secretory pathway and is regulating posttranslational modifications, sorting, transport, and signal transduction, as well as acting as a non-centrosomal microtubule organization center. The neuronal Golgi apparatus shares common features with Golgi in other eukaryotic cell types but also forms distinct structures known as Golgi outposts that specifically localize in dendrites. However, the organization and function of Golgi in dendrite development and its impact on neurological disorders is just emerging and so far lacks a systematic summary. We describe the organization of the Golgi apparatus in neurons, review the current understanding of Golgi function in dendritic morphogenesis, and discuss the current challenges and future directions.
Collapse
Affiliation(s)
- Meilan Chen
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
- Department of Ophthalmology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510320, China
| | - Lu Xu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
| | - Yi Wu
- Department of Ophthalmology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510320, China
| | - Peter Soba
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Bonn 53115, Germany
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Chun Hu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
| |
Collapse
|
7
|
Andres-Alonso M, Borgmeyer M, Mirzapourdelavar H, Lormann J, Klein K, Schweizer M, Hoffmeister-Ullerich S, Oelschlegel AM, Dityatev A, Kreutz MR. Golgi satellites are essential for polysialylation of NCAM and expression of LTP at distal synapses. Cell Rep 2023; 42:112692. [PMID: 37355986 DOI: 10.1016/j.celrep.2023.112692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/28/2023] [Accepted: 06/08/2023] [Indexed: 06/27/2023] Open
Abstract
The complex cytoarchitecture of neurons poses significant challenges for the maturation of synaptic membrane proteins. It is currently unclear whether locally secreted synaptic proteins bypass the Golgi or whether they traffic through Golgi satellites (GSs). Here, we create a transgenic GS reporter mouse line and show that GSs are widely distributed along dendrites and are capable of mature glycosylation, in particular sialylation. We find that polysialylation of locally secreted NCAM takes place at GSs. Accordingly, in mice lacking a component of trans-Golgi network-to-plasma membrane trafficking, we find fewer GSs and significantly reduced PSA-NCAM levels in distal dendrites of CA1 neurons that receive input from the temporoammonic pathway. Induction of long-term potentiation at those, but not more proximal, synapses is severely impaired. We conclude that GSs serve the need for local mature glycosylation of synaptic membrane proteins in distal dendrites and thereby contribute to rapid changes in synaptic strength.
Collapse
Affiliation(s)
- Maria Andres-Alonso
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany.
| | - Maximilian Borgmeyer
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | | | - Jakob Lormann
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Kim Klein
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Michaela Schweizer
- Core Facility Morphology und Electron Microscopy, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Sabine Hoffmeister-Ullerich
- Core Facility Bioanalytik, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Anja M Oelschlegel
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto von Guericke University, 39120 Magdeburg, Germany; Medical Faculty, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Michael R Kreutz
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto von Guericke University, 39120 Magdeburg, Germany.
| |
Collapse
|
8
|
Huang Z, Wang H, Wang D, Zhao X, Liu W, Zhong X, He D, Mu B, Lu M. Identification of core genes in prefrontal cortex and hippocampus of Alzheimer's disease based on mRNA‐miRNA network. J Cell Mol Med 2022; 26:5779-5793. [DOI: 10.1111/jcmm.17593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/19/2021] [Accepted: 04/24/2022] [Indexed: 11/21/2022] Open
Affiliation(s)
- Zhi‐Hang Huang
- Chongqing Key Laboratory of Sichuan‐Chongqing Co‐construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Hai Wang
- Chongqing Key Laboratory of Sichuan‐Chongqing Co‐construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology Chengdu University of Traditional Chinese Medicine Chengdu China
- School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Dong‐Mei Wang
- School of Basic Medical Sciences Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Xiu‐Yun Zhao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience Soochow University Suzhou China
| | - Wen‐Wen Liu
- Chongqing Key Laboratory of Sichuan‐Chongqing Co‐construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Xin Zhong
- Chongqing Key Laboratory of Sichuan‐Chongqing Co‐construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Dong‐Mei He
- School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Ben‐Rong Mu
- Chongqing Key Laboratory of Sichuan‐Chongqing Co‐construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Mei‐Hong Lu
- Chongqing Key Laboratory of Sichuan‐Chongqing Co‐construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology Chengdu University of Traditional Chinese Medicine Chengdu China
| |
Collapse
|
9
|
Grochowska KM, Andres‐Alonso M, Karpova A, Kreutz MR. The needs of a synapse—How local organelles serve synaptic proteostasis. EMBO J 2022; 41:e110057. [PMID: 35285533 PMCID: PMC8982616 DOI: 10.15252/embj.2021110057] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/24/2021] [Accepted: 02/10/2022] [Indexed: 12/12/2022] Open
Abstract
Synaptic function crucially relies on the constant supply and removal of neuronal membranes. The morphological complexity of neurons poses a significant challenge for neuronal protein transport since the machineries for protein synthesis and degradation are mainly localized in the cell soma. In response to this unique challenge, local micro‐secretory systems have evolved that are adapted to the requirements of neuronal membrane protein proteostasis. However, our knowledge of how neuronal proteins are synthesized, trafficked to membranes, and eventually replaced and degraded remains scarce. Here, we review recent insights into membrane trafficking at synaptic sites and into the contribution of local organelles and micro‐secretory pathways to synaptic function. We describe the role of endoplasmic reticulum specializations in neurons, Golgi‐related organelles, and protein complexes like retromer in the synthesis and trafficking of synaptic transmembrane proteins. We discuss the contribution of autophagy and of proteasome‐mediated and endo‐lysosomal degradation to presynaptic proteostasis and synaptic function, as well as nondegradative roles of autophagosomes and lysosomes in signaling and synapse remodeling. We conclude that the complexity of neuronal cyto‐architecture necessitates long‐distance protein transport that combines degradation with signaling functions.
Collapse
Affiliation(s)
- Katarzyna M Grochowska
- Leibniz Group “Dendritic Organelles and Synaptic Function” Center for Molecular Neurobiology ZMNH University Medical Center Hamburg‐Eppendorf Hamburg Germany
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
| | - Maria Andres‐Alonso
- Leibniz Group “Dendritic Organelles and Synaptic Function” Center for Molecular Neurobiology ZMNH University Medical Center Hamburg‐Eppendorf Hamburg Germany
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
| | - Anna Karpova
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
- Center for Behavioral Brain Sciences Otto von Guericke University Magdeburg Germany
| | - Michael R Kreutz
- Leibniz Group “Dendritic Organelles and Synaptic Function” Center for Molecular Neurobiology ZMNH University Medical Center Hamburg‐Eppendorf Hamburg Germany
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
- Center for Behavioral Brain Sciences Otto von Guericke University Magdeburg Germany
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg Germany
| |
Collapse
|
10
|
Fabbretti E, Antognolli G, Tongiorgi E. Amyloid-β Impairs Dendritic Trafficking of Golgi-Like Organelles in the Early Phase Preceding Neurite Atrophy: Rescue by Mirtazapine. Front Mol Neurosci 2021; 14:661728. [PMID: 34149353 PMCID: PMC8209480 DOI: 10.3389/fnmol.2021.661728] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/30/2021] [Indexed: 12/20/2022] Open
Abstract
Neurite atrophy with loss of neuronal polarity is a pathological hallmark of Alzheimer's disease (AD) and other neurological disorders. While there is substantial agreement that disruption of intracellular vesicle trafficking is associated with axonal pathology in AD, comparatively less is known regarding its role in dendritic atrophy. This is a significant gap of knowledge because, unlike axons, dendrites are endowed with the complete endomembrane system comprising endoplasmic reticulum (ER), ER-Golgi intermediate compartment (ERGIC), Golgi apparatus, post-Golgi vesicles, and a recycling-degradative route. In this study, using live-imaging of pGOLT-expressing vesicles, indicative of Golgi outposts and satellites, we investigate how amyloid-β (Aβ) oligomers affect the trafficking of Golgi-like organelles in the different dendritic compartments of cultured rat hippocampal neurons. We found that short-term (4 h) treatment with Aβ led to a decrease in anterograde trafficking of Golgi vesicles in dendrites of both resting and stimulated (with 50 mM KCl) neurons. We also characterized the ability of mirtazapine, a noradrenergic and specific serotonergic tetracyclic antidepressant (NaSSA), to rescue Golgi dynamics in dendrites. Mirtazapine treatment (10 μM) increased the number and both anterograde and retrograde motility, reducing the percentage of static Golgi vesicles. Finally, mirtazapine reverted the neurite atrophy induced by 24 h treatment with Aβ oligomers, suggesting that this drug is able to counteract the effects of Aβ by improving the dendritic trafficking of Golgi-related vesicles.
Collapse
Affiliation(s)
- Elsa Fabbretti
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | | | - Enrico Tongiorgi
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
11
|
Pays E. The function of apolipoproteins L (APOLs): relevance for kidney disease, neurotransmission disorders, cancer and viral infection. FEBS J 2021; 288:360-381. [PMID: 32530132 PMCID: PMC7891394 DOI: 10.1111/febs.15444] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/24/2020] [Accepted: 06/03/2020] [Indexed: 12/17/2022]
Abstract
The discovery that apolipoprotein L1 (APOL1) is the trypanolytic factor of human serum raised interest about the function of APOLs, especially following the unexpected finding that in addition to their protective action against sleeping sickness, APOL1 C-terminal variants also cause kidney disease. Based on the analysis of the structure and trypanolytic activity of APOL1, it was proposed that APOLs could function as ion channels of intracellular membranes and be involved in mechanisms triggering programmed cell death. In this review, the recent finding that APOL1 and APOL3 inversely control the synthesis of phosphatidylinositol-4-phosphate (PI(4)P) by the Golgi PI(4)-kinase IIIB (PI4KB) is commented. APOL3 promotes Ca2+ -dependent activation of PI4KB, but due to their increased interaction with APOL3, APOL1 C-terminal variants can inactivate APOL3, leading to reduction of Golgi PI(4)P synthesis. The impact of APOLs on several pathological processes that depend on Golgi PI(4)P levels is discussed. I propose that through their effect on PI4KB activity, APOLs control not only actomyosin activities related to vesicular trafficking, but also the generation and elongation of autophagosomes induced by inflammation.
Collapse
Affiliation(s)
- Etienne Pays
- Laboratory of Molecular ParasitologyIBMMUniversité Libre de BruxellesGosseliesBelgium
| |
Collapse
|
12
|
Uzureau S, Lecordier L, Uzureau P, Hennig D, Graversen JH, Homblé F, Mfutu PE, Oliveira Arcolino F, Ramos AR, La Rovere RM, Luyten T, Vermeersch M, Tebabi P, Dieu M, Cuypers B, Deborggraeve S, Rabant M, Legendre C, Moestrup SK, Levtchenko E, Bultynck G, Erneux C, Pérez-Morga D, Pays E. APOL1 C-Terminal Variants May Trigger Kidney Disease through Interference with APOL3 Control of Actomyosin. Cell Rep 2020; 30:3821-3836.e13. [PMID: 32187552 PMCID: PMC7090385 DOI: 10.1016/j.celrep.2020.02.064] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/17/2020] [Accepted: 02/14/2020] [Indexed: 11/18/2022] Open
Abstract
The C-terminal variants G1 and G2 of apolipoprotein L1 (APOL1) confer human resistance to the sleeping sickness parasite Trypanosoma rhodesiense, but they also increase the risk of kidney disease. APOL1 and APOL3 are death-promoting proteins that are partially associated with the endoplasmic reticulum and Golgi membranes. We report that in podocytes, either APOL1 C-terminal helix truncation (APOL1Δ) or APOL3 deletion (APOL3KO) induces similar actomyosin reorganization linked to the inhibition of phosphatidylinositol-4-phosphate [PI(4)P] synthesis by the Golgi PI(4)-kinase IIIB (PI4KB). Both APOL1 and APOL3 can form K+ channels, but only APOL3 exhibits Ca2+-dependent binding of high affinity to neuronal calcium sensor-1 (NCS-1), promoting NCS-1-PI4KB interaction and stimulating PI4KB activity. Alteration of the APOL1 C-terminal helix triggers APOL1 unfolding and increased binding to APOL3, affecting APOL3-NCS-1 interaction. Since the podocytes of G1 and G2 patients exhibit an APOL1Δ or APOL3KO-like phenotype, APOL1 C-terminal variants may induce kidney disease by preventing APOL3 from activating PI4KB, with consecutive actomyosin reorganization of podocytes.
Collapse
Affiliation(s)
- Sophie Uzureau
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Laurence Lecordier
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Pierrick Uzureau
- Laboratory of Experimental Medicine (ULB222), CHU Charleroi, Université Libre de Bruxelles, Montigny le Tilleul, Belgium
| | - Dorle Hennig
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Jonas H Graversen
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Fabrice Homblé
- Laboratory of Structure and Function of Biological Membranes, Université Libre de Bruxelles, 1050 Brussels, Belgium
| | - Pepe Ekulu Mfutu
- Pediatric Nephrology, University Hospital Leuven, 3000 Leuven, Belgium
| | | | - Ana Raquel Ramos
- Institute of Interdisciplinary Research in Human and Molecular Biology, Campus Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Rita M La Rovere
- Laboratory of Molecular and Cellular Signalling, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Tomas Luyten
- Laboratory of Molecular and Cellular Signalling, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Marjorie Vermeersch
- Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Patricia Tebabi
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Marc Dieu
- URBC-Narilis, University of Namur, 5000 Namur, Belgium
| | - Bart Cuypers
- Biomedical Sciences Department, Institute of Tropical Medicine, 2000 Antwerpen, Belgium; Adrem Data Lab, Department of Mathematics and Computer Science, University of Antwerp, 2000 Antwerpen, Belgium
| | - Stijn Deborggraeve
- Biomedical Sciences Department, Institute of Tropical Medicine, 2000 Antwerpen, Belgium
| | - Marion Rabant
- Adult Nephrology-Transplantation Department, Paris Hospitals and Paris Descartes University, 75006 Paris, France
| | - Christophe Legendre
- Pathology Department, Paris Hospitals and Paris Descartes University, 75006 Paris, France
| | - Søren K Moestrup
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense C, Denmark; Department of Biomedicine, University of Aarhus, 8000 Aarhus, Denmark
| | - Elena Levtchenko
- Pediatric Nephrology, University Hospital Leuven, 3000 Leuven, Belgium
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signalling, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Christophe Erneux
- Institute of Interdisciplinary Research in Human and Molecular Biology, Campus Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - David Pérez-Morga
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium; Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Etienne Pays
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium.
| |
Collapse
|
13
|
Cornejo VH, González C, Campos M, Vargas-Saturno L, Juricic MDLÁ, Miserey-Lenkei S, Pertusa M, Madrid R, Couve A. Non-conventional Axonal Organelles Control TRPM8 Ion Channel Trafficking and Peripheral Cold Sensing. Cell Rep 2020; 30:4505-4517.e5. [DOI: 10.1016/j.celrep.2020.03.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 10/07/2019] [Accepted: 03/05/2020] [Indexed: 12/31/2022] Open
|
14
|
The Great Escape: how phosphatidylinositol 4-kinases and PI4P promote vesicle exit from the Golgi (and drive cancer). Biochem J 2019; 476:2321-2346. [DOI: 10.1042/bcj20180622] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/06/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022]
Abstract
Abstract
Phosphatidylinositol 4-phosphate (PI4P) is a membrane glycerophospholipid and a major regulator of the characteristic appearance of the Golgi complex as well as its vesicular trafficking, signalling and metabolic functions. Phosphatidylinositol 4-kinases, and in particular the PI4KIIIβ isoform, act in concert with PI4P to recruit macromolecular complexes to initiate the biogenesis of trafficking vesicles for several Golgi exit routes. Dysregulation of Golgi PI4P metabolism and the PI4P protein interactome features in many cancers and is often associated with tumour progression and a poor prognosis. Increased expression of PI4P-binding proteins, such as GOLPH3 or PITPNC1, induces a malignant secretory phenotype and the release of proteins that can remodel the extracellular matrix, promote angiogenesis and enhance cell motility. Aberrant Golgi PI4P metabolism can also result in the impaired post-translational modification of proteins required for focal adhesion formation and cell–matrix interactions, thereby potentiating the development of aggressive metastatic and invasive tumours. Altered expression of the Golgi-targeted PI 4-kinases, PI4KIIIβ, PI4KIIα and PI4KIIβ, or the PI4P phosphate Sac1, can also modulate oncogenic signalling through effects on TGN-endosomal trafficking. A Golgi trafficking role for a PIP 5-kinase has been recently described, which indicates that PI4P is not the only functionally important phosphoinositide at this subcellular location. This review charts new developments in our understanding of phosphatidylinositol 4-kinase function at the Golgi and how PI4P-dependent trafficking can be deregulated in malignant disease.
Collapse
|
15
|
Caracci MO, Fuentealba LM, Marzolo MP. Golgi Complex Dynamics and Its Implication in Prevalent Neurological Disorders. Front Cell Dev Biol 2019; 7:75. [PMID: 31134199 PMCID: PMC6514153 DOI: 10.3389/fcell.2019.00075] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 04/18/2019] [Indexed: 12/13/2022] Open
Abstract
Coupling of protein synthesis with protein delivery to distinct subcellular domains is essential for maintaining cellular homeostasis, and defects thereof have consistently been shown to be associated with several diseases. This function is particularly challenging for neurons given their polarized nature and differential protein requirements in synaptic boutons, dendrites, axons, and soma. Long-range trafficking is greatly enhanced in neurons by discrete mini-organelles resembling the Golgi complex (GC) referred to as Golgi outposts (GOPs) which play an essential role in the development of dendritic arborization. In this context, the morphology of the GC is highly plastic, and the polarized distribution of this organelle is necessary for neuronal migration and polarized growth. Furthermore, synaptic components are readily trafficked and modified at GOP suggesting a function for this organelle in synaptic plasticity. However, little is known about GOPs properties and biogenesis and the role of GOP dysregulation in pathology. In this review, we discuss current literature supporting a role for GC dynamics in prevalent neurological disorders such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and epilepsy, and examine the association of these disorders with the wide-ranging effects of GC function on common cellular pathways regulating neuronal excitability, polarity, migration, and organellar stress. First, we discuss the role of Golgins and Golgi-associated proteins in the regulation of GC morphology and dynamics. Then, we consider abnormal GC arrangements observed in neurological disorders and associations with common neuronal defects therein. Finally, we consider the cell signaling pathways involved in the modulation of GC dynamics and argue for a master regulatory role for Reelin signaling, a well-known regulator of neuronal polarity and migration. Determining the cellular pathways involved in shaping the Golgi network will have a direct and profound impact on our current understanding of neurodevelopment and neuropathology and aid the development of novel therapeutic strategies for improved patient care and prognosis.
Collapse
Affiliation(s)
- Mario O Caracci
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luz M Fuentealba
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María-Paz Marzolo
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
16
|
Burgoyne RD, Helassa N, McCue HV, Haynes LP. Calcium Sensors in Neuronal Function and Dysfunction. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a035154. [PMID: 30833454 DOI: 10.1101/cshperspect.a035154] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Calcium signaling in neurons as in other cell types can lead to varied changes in cellular function. Neuronal Ca2+ signaling processes have also become adapted to modulate the function of specific pathways over a wide variety of time domains and these can have effects on, for example, axon outgrowth, neuronal survival, and changes in synaptic strength. Ca2+ also plays a key role in synapses as the trigger for fast neurotransmitter release. Given its physiological importance, abnormalities in neuronal Ca2+ signaling potentially underlie many different neurological and neurodegenerative diseases. The mechanisms by which changes in intracellular Ca2+ concentration in neurons can bring about diverse responses is underpinned by the roles of ubiquitous or specialized neuronal Ca2+ sensors. It has been established that synaptotagmins have key functions in neurotransmitter release, and, in addition to calmodulin, other families of EF-hand-containing neuronal Ca2+ sensors, including the neuronal calcium sensor (NCS) and the calcium-binding protein (CaBP) families, play important physiological roles in neuronal Ca2+ signaling. It has become increasingly apparent that these various Ca2+ sensors may also be crucial for aspects of neuronal dysfunction and disease either indirectly or directly as a direct consequence of genetic variation or mutations. An understanding of the molecular basis for the regulation of the targets of the Ca2+ sensors and the physiological roles of each protein in identified neurons may contribute to future approaches to the development of treatments for a variety of human neuronal disorders.
Collapse
Affiliation(s)
- Robert D Burgoyne
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Nordine Helassa
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Hannah V McCue
- Centre for Genomic Research, University of Liverpool, Liverpool, United Kingdom
| | - Lee P Haynes
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
17
|
Nakamura TY, Nakao S, Wakabayashi S. Emerging Roles of Neuronal Ca 2+ Sensor-1 in Cardiac and Neuronal Tissues: A Mini Review. Front Mol Neurosci 2019; 12:56. [PMID: 30886571 PMCID: PMC6409499 DOI: 10.3389/fnmol.2019.00056] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 02/15/2019] [Indexed: 11/13/2022] Open
Abstract
The EF-hand calcium (Ca2+)-binding protein, neuronal Ca2+ sensor-1 (NCS-1/frequenin), is predominantly expressed in neuronal tissues and plays a crucial role in neuronal functions, including synaptic transmission and plasticity. NCS-1 has diverse functional roles, as elucidated in the past 15 years, which include the regulation of phosphatidylinositol 4-kinase IIIβ (PI-4K-β) and several ion channels such as voltage-gated K+ and Ca2+ channels, the D2 dopamine receptors, and inositol 1,4,5-trisphosphate receptors (InsP3Rs). Functional analyses demonstrated that NCS-1 enhances exocytosis and neuronal survival after injury, as well as promotes learning and memory in mice. NCS-1 is also expressed in the heart including the Purkinje fibers (PFs) of the conduction system. NCS-1 interacts with KV4 K+ channels together with dipeptidyl peptidase-like protein-6 (DPP-6), and this macromolecule then composes the transient outward current in PFs and contributes to the repolarization of PF action potential, thus being responsible for idiopathic arrhythmia. Moreover, NCS-1 expression was reported to be significantly high at the immature stage and at hypertrophy in adults. That report demonstrated that NCS-1 positively regulates cardiac contraction in immature hearts by increasing intracellular Ca2+ signals through interaction with InsP3Rs. With the related signals, NCS-1 activates nuclear Ca2+ signals, which would be a mechanism underlying hormone-induced cardiac hypertrophy. Furthermore, NCS-1 contributes to stress tolerance in cardiomyocytes by activating mitochondrial detoxification pathways, with a key role in Ca2+-dependent pathways. In this review, we will discuss recent findings supporting the functional significance of NCS-1 in the brain and heart and will address possible underlying molecular mechanisms.
Collapse
Affiliation(s)
- Tomoe Y Nakamura
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Shu Nakao
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan.,Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Japan
| | - Shigeo Wakabayashi
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan.,Department of Pharmacology, Osaka Medical College, Takatsuki, Japan
| |
Collapse
|
18
|
Mundhenk J, Fusi C, Kreutz MR. Caldendrin and Calneurons-EF-Hand CaM-Like Calcium Sensors With Unique Features and Specialized Neuronal Functions. Front Mol Neurosci 2019; 12:16. [PMID: 30787867 PMCID: PMC6372560 DOI: 10.3389/fnmol.2019.00016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/17/2019] [Indexed: 01/02/2023] Open
Abstract
The calmodulin (CaM)-like Ca2+-sensor proteins caldendrin, calneuron-1 and -2 are members of the neuronal calcium-binding protein (nCaBP)-family, a family that evolved relatively late during vertebrate evolution. All three proteins are abundant in brain but show a strikingly different subcellular localization. Whereas caldendrin is enriched in the postsynaptic density (PSD), calneuron-1 and -2 accumulate at the trans-Golgi-network (TGN). Caldendrin exhibit a unique bipartite structure with a basic and proline-rich N-terminus while calneurons are the only EF-Hand CaM-like transmembrane proteins. These uncommon structural features come along with highly specialized functions of calneurons in Golgi-to-plasma-membrane trafficking and for caldendrin in actin-remodeling in dendritic spine synapses. In this review article, we will provide a synthesis of available data on the structure and biophysical properties of all three proteins. We will then discuss their cellular function with special emphasis on synaptic neurotransmission. Finally, we will summarize the evidence for a role of these proteins in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jennifer Mundhenk
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Camilla Fusi
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Leibniz Group "Dendritic Organelles and Synaptic Function", Center for Molecular Neurobiology, ZMNH, Hamburg, Germany
| |
Collapse
|
19
|
Franco R, Aguinaga D, Reyes I, Canela EI, Lillo J, Tarutani A, Hasegawa M, Del Ser-Badia A, Del Rio JA, Kreutz MR, Saura CA, Navarro G. N-Methyl-D-Aspartate Receptor Link to the MAP Kinase Pathway in Cortical and Hippocampal Neurons and Microglia Is Dependent on Calcium Sensors and Is Blocked by α-Synuclein, Tau, and Phospho-Tau in Non-transgenic and Transgenic APP Sw,Ind Mice. Front Mol Neurosci 2018; 11:273. [PMID: 30233307 PMCID: PMC6127644 DOI: 10.3389/fnmol.2018.00273] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 07/18/2018] [Indexed: 11/14/2022] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) respond to glutamate to allow the influx of calcium ions and the signaling to the mitogen-activated protein kinase (MAPK) cascade. Both MAPK- and Ca2+-mediated events are important for both neurotransmission and neural cell function and fate. Using a heterologous expression system, we demonstrate that NMDAR may interact with the EF-hand calcium-binding proteins calmodulin, calneuron-1, and NCS1 but not with caldendrin. NMDARs were present in primary cultures of both neurons and microglia from cortex and hippocampus. Calmodulin in microglia, and calmodulin and NCS1 in neurons, are necessary for NMDA-induced MAP kinase pathway activation. Remarkably, signaling to the MAP kinase pathway was blunted in primary cultures of cortical and hippocampal neurons and microglia from wild-type animals by proteins involved in neurodegenerative diseases: α-synuclein, Tau, and p-Tau. A similar blockade by pathogenic proteins was found using samples from the APPSw,Ind transgenic Alzheimer’s disease model. Interestingly, a very marked increase in NMDAR–NCS1 complexes was identified in neurons and a marked increase of both NMDAR–NCS1 and NMDAR–CaM complexes was identified in microglia from the transgenic mice. The results show that α-synuclein, Tau, and p-Tau disrupt the signaling of NMDAR to the MAPK pathway and that calcium sensors are important for NMDAR function both in neurons and microglia. Finally, it should be noted that the expression of receptor–calcium sensor complexes, specially those involving NCS1, is altered in neural cells from APPSw,Ind mouse embryos/pups.
Collapse
Affiliation(s)
- Rafael Franco
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - David Aguinaga
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Irene Reyes
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Enric I Canela
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Jaume Lillo
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain
| | - Airi Tarutani
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masato Hasegawa
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Anna Del Ser-Badia
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Department de Bioquímica i Biologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - José A Del Rio
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Leibniz Group Dendritic Organelles and Synaptic Function, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carlos A Saura
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Department de Bioquímica i Biologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Gemma Navarro
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
20
|
Angelats E, Requesens M, Aguinaga D, Kreutz MR, Franco R, Navarro G. Neuronal Calcium and cAMP Cross-Talk Mediated by Cannabinoid CB 1 Receptor and EF-Hand Calcium Sensor Interactions. Front Cell Dev Biol 2018; 6:67. [PMID: 30073165 PMCID: PMC6060245 DOI: 10.3389/fcell.2018.00067] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 06/12/2018] [Indexed: 11/19/2022] Open
Abstract
Endocannabinoids are important players in neural development and function. They act via receptors, whose activation inhibits cAMP production. The aim of the paper was to look for calcium- and cAMP-signaling cross-talk mediated by cannabinoid CB1 receptors (CB1R) and to assess the relevance of EF-hand CaM-like calcium sensors in this regard. Using a heterologous expression system, we demonstrated that CB1R interacts with calneuron-1 and NCS1 but not with caldendrin. Furthermore, interaction motives were identified in both calcium binding proteins and the receptor, and we showed that the first two sensors competed for binding to the receptor in a Ca2+-dependent manner. Assays in neuronal primary cultures showed that, CB1R-NCS1 complexes predominate at basal Ca2+ levels, whereas in the presence of ionomycin, a calcium ionophore, CB1R-calneuron-1 complexes were more abundant. Signaling assays following forskolin-induced intracellular cAMP levels showed in mouse striatal neurons that binding of CB1R to NCS1 is required for CB1R-mediated signaling, while the binding of CB1R to calneuron-1 completely blocked Gi-mediated signaling in response to a selective receptor agonist, arachidonyl-2-chloroethylamide. Calcium levels and interaction with calcium sensors may even lead to apparent Gs coupling after CB1R agonist challenge.
Collapse
Affiliation(s)
- Edgar Angelats
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Requesens
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - David Aguinaga
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Leibniz Group "Dendritic Organelles and Synaptic Function", Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rafael Franco
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Gemma Navarro
- Centro de Investigación en Red, Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain.,Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
| |
Collapse
|
21
|
Kobuke K, Oki K, Gomez-Sanchez CE, Gomez-Sanchez EP, Ohno H, Itcho K, Yoshii Y, Yoneda M, Hattori N. Calneuron 1 Increased Ca 2+ in the Endoplasmic Reticulum and Aldosterone Production in Aldosterone-Producing Adenoma. Hypertension 2017; 71:125-133. [PMID: 29109191 DOI: 10.1161/hypertensionaha.117.10205] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 09/01/2017] [Accepted: 10/10/2017] [Indexed: 11/16/2022]
Abstract
Aldosterone production is initiated by angiotensin II stimulation and activation of intracellular Ca2+ signaling. In aldosterone-producing adenoma (APA) cells, the activation of intracellular Ca2+ signaling is independent of the renin-angiotensin-aldosterone systems. The purpose of our study was to clarify molecular mechanisms of aldosterone production related to Ca2+ signaling. Transcriptome analysis revealed that the CALN1 gene encoding calneuron 1 had the strongest correlation with CYP11B2 (aldosterone synthase) among genes encoding Ca2+-binding proteins in APA. CALN1 modulation and synthetic or fluorescent compounds were used for functional studies in human adrenocortical carcinoma (HAC15) cells. CALN1 expression was 4.4-fold higher in APAs than nonfunctioning adrenocortical adenomas. CALN1 expression colocalized with CYP11B2 expression as investigated using immunohistochemistry in APA and zona glomerulosa of male rats fed by a low-salt diet. CALN1 expression was detected in the endoplasmic reticulum (ER) by using GFP-fused CALN1, CellLight ER-RFP, and the corresponding antibodies. CALN1-overexpressing HAC15 cells showed increased Ca2+ in the ER and cytosol fluorescence-based studies. Aldosterone production was potentiated in HAC15 cells by CALN1 expression, and dose-responsive inhibition with TMB-8 showed that CALN1-mediated Ca2+ storage in ER involved sarcoendoplasmic reticulum calcium transport ATPase. The silencing of CALN1 decreased Ca2+ in ER, and abrogated angiotensin II- or KCNJ5 T158A-mediated aldosterone production in HAC15 cells. Increased CALN1 expression in APA was associated with elevated Ca2+ storage in ER and aldosterone overproduction. Suppression of CALN1 expression prevented angiotensin II- or KCNJ5 T158A-mediated aldosterone production in HAC15 cells, suggesting that CALN1 is a potential therapeutic target for excess aldosterone production.
Collapse
Affiliation(s)
- Kazuhiro Kobuke
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.K., K.O., H.O., K.I., Y.Y., M.Y., N.H.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S., E.P.G.-S.); and University of Mississippi Medical Center, Jackson (C.E.G.-S., E.P.G.-S.)
| | - Kenji Oki
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.K., K.O., H.O., K.I., Y.Y., M.Y., N.H.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S., E.P.G.-S.); and University of Mississippi Medical Center, Jackson (C.E.G.-S., E.P.G.-S.).
| | - Celso E Gomez-Sanchez
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.K., K.O., H.O., K.I., Y.Y., M.Y., N.H.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S., E.P.G.-S.); and University of Mississippi Medical Center, Jackson (C.E.G.-S., E.P.G.-S.)
| | - Elise P Gomez-Sanchez
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.K., K.O., H.O., K.I., Y.Y., M.Y., N.H.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S., E.P.G.-S.); and University of Mississippi Medical Center, Jackson (C.E.G.-S., E.P.G.-S.)
| | - Haruya Ohno
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.K., K.O., H.O., K.I., Y.Y., M.Y., N.H.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S., E.P.G.-S.); and University of Mississippi Medical Center, Jackson (C.E.G.-S., E.P.G.-S.)
| | - Kiyotaka Itcho
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.K., K.O., H.O., K.I., Y.Y., M.Y., N.H.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S., E.P.G.-S.); and University of Mississippi Medical Center, Jackson (C.E.G.-S., E.P.G.-S.)
| | - Yoko Yoshii
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.K., K.O., H.O., K.I., Y.Y., M.Y., N.H.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S., E.P.G.-S.); and University of Mississippi Medical Center, Jackson (C.E.G.-S., E.P.G.-S.)
| | - Masayasu Yoneda
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.K., K.O., H.O., K.I., Y.Y., M.Y., N.H.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S., E.P.G.-S.); and University of Mississippi Medical Center, Jackson (C.E.G.-S., E.P.G.-S.)
| | - Noboru Hattori
- From the Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan (K.K., K.O., H.O., K.I., Y.Y., M.Y., N.H.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS (C.E.G.-S., E.P.G.-S.); and University of Mississippi Medical Center, Jackson (C.E.G.-S., E.P.G.-S.)
| |
Collapse
|
22
|
Abstract
BACKGROUND Prepulse inhibition (PPI) of the startle reflex has been suggested as a candidate endophenotype for schizophrenia research, as it shows high heritability and has been found deficient in schizophrenia spectrum disorders. The objectives of the study were to 1) identify common genetic variants associated with baseline startle and PPI; 2) estimate the single nucleotide polymorphism heritability; and 3) examine the relationship of polygenic score for schizophrenia with baseline startle and PPI. METHODS A cohort of healthy young male subjects (n = 1493) originating from the Learning on Genetics of Schizophrenia Spectrum project was assessed for baseline startle and PPI. The most recent genome-wide association study in schizophrenia from the Psychiatric Genomics Consortium 2 was used to calculate polygenic scores. RESULTS Eleven loci showed suggestive association (p < 10(-6)) with baseline startle and PPI in the discovery cohort. Additional genotyping in a replication cohort identified genome-wide significant association at two loci (rs61810702 and rs4718984). These loci were co-localized with expression quantitative trait loci associated with gene expression of nerve growth factor (NGF) and calneuron 1 (CALN1) genes. Estimation of the genetic and environmental contributions to baseline startle and PPI showed a substantial single nucleotide polymorphism heritability for 120-ms PPI stimuli. Increased polygenic risk score for schizophrenia was associated with reduced PPI. CONCLUSIONS Common genetic variation has an important role in the etiology of schizophrenia and PPI impairments. Overall, these data support the idea that PPI is a valid endophenotype that can be used to explore the genetic architecture of schizophrenia.
Collapse
|
23
|
Bera S, Raghuram V, Mikhaylova M, Kreutz MR. A plasmid-based expression system to study protein-protein interactions at the Golgi in vivo. Anal Biochem 2016; 502:50-52. [PMID: 26973219 DOI: 10.1016/j.ab.2016.02.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 02/20/2016] [Accepted: 02/24/2016] [Indexed: 01/23/2023]
Abstract
There is still an unmet need for simple methods to verify, visualize, and confirm protein-protein interactions in vivo. Here we describe a plasmid-based system to study such interactions. The system is based on the transmembrane domain (TMD) of the EF-hand Ca(2+) sensor protein calneuron-2. We show that fusion of 28 amino acids that include the TMD of calneuron-2 to proteins of interest results in prominent localization on the cytoplasmic side of the Golgi. The recruitment of binding partners to the protein of interest fused to this sequence can then be easily visualized by fluorescent tags.
Collapse
Affiliation(s)
- Sujoy Bera
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Vijeta Raghuram
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany; CSIR-Centre for Cellular and Molecular Biology (CCMB), Hyderabad, 500007, India
| | - Marina Mikhaylova
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany; Emmy Noether Group "Neuronal Protein Transport", Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Michael R Kreutz
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany; Leibniz Group "Dendritic Organelles and Synaptic Function", Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| |
Collapse
|
24
|
A Dendritic Golgi Satellite between ERGIC and Retromer. Cell Rep 2016; 14:189-99. [DOI: 10.1016/j.celrep.2015.12.024] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 11/08/2015] [Accepted: 11/25/2015] [Indexed: 11/20/2022] Open
|
25
|
Yang Z, Kirton HM, MacDougall DA, Boyle JP, Deuchars J, Frater B, Ponnambalam S, Hardy ME, White E, Calaghan SC, Peers C, Steele DS. The Golgi apparatus is a functionally distinct Ca2+ store regulated by the PKA and Epac branches of the β1-adrenergic signaling pathway. Sci Signal 2015; 8:ra101. [PMID: 26462734 PMCID: PMC4869832 DOI: 10.1126/scisignal.aaa7677] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ca(2+) release from the Golgi apparatus regulates key functions of the organelle, including vesicle trafficking. We found that the Golgi apparatus was the source of prolonged Ca(2+) release events that originated near the nuclei of primary cardiomyocytes. Golgi Ca(2+) release was unaffected by depletion of sarcoplasmic reticulum Ca(2+), and disruption of the Golgi apparatus abolished Golgi Ca(2+) release without affecting sarcoplasmic reticulum function, suggesting functional and spatial independence of Golgi and sarcoplasmic reticulum Ca(2+) stores. β1-Adrenoceptor stimulation triggers the production of the second messenger cAMP, which activates the Epac family of Rap guanine nucleotide exchange factors and the kinase PKA (protein kinase A). Phosphodiesterases (PDEs), including those in the PDE3 and PDE4 families, degrade cAMP. Activation of β1-adrenoceptors stimulated Golgi Ca(2+) release, an effect that required activation of Epac, PKA, and the kinase CaMKII. Inhibition of PDE3s or PDE4s potentiated β1-adrenergic-induced Golgi Ca(2+) release, which is consistent with compartmentalization of cAMP signaling near the Golgi apparatus. Interventions that stimulated Golgi Ca(2+) release appeared to increase the trafficking of vascular endothelial growth factor receptor-1 (VEGFR-1) from the Golgi apparatus to the surface membrane of cardiomyocytes. In cardiomyocytes from rats with heart failure, decreases in the abundance of PDE3s and PDE4s were associated with increased Golgi Ca(2+) release events. These data suggest that the Golgi apparatus is a focal point for β1-adrenergic-stimulated Ca(2+) signaling and that the Golgi Ca(2+) store functions independently from the sarcoplasmic reticulum and the global Ca(2+) transients that trigger contraction in cardiomyocytes.
Collapse
Affiliation(s)
- Zhaokang Yang
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK.
| | - Hannah M Kirton
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
| | | | - John P Boyle
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK
| | - James Deuchars
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Brenda Frater
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
| | | | - Matthew E Hardy
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Edward White
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Sarah C Calaghan
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Chris Peers
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK
| | - Derek S Steele
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
26
|
Prydz K. Determinants of Glycosaminoglycan (GAG) Structure. Biomolecules 2015; 5:2003-22. [PMID: 26308067 PMCID: PMC4598785 DOI: 10.3390/biom5032003] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 08/17/2015] [Accepted: 08/18/2015] [Indexed: 01/05/2023] Open
Abstract
Proteoglycans (PGs) are glycosylated proteins of biological importance at cell surfaces, in the extracellular matrix, and in the circulation. PGs are produced and modified by glycosaminoglycan (GAG) chains in the secretory pathway of animal cells. The most common GAG attachment site is a serine residue followed by a glycine (-ser-gly-), from which a linker tetrasaccharide extends and may continue as a heparan sulfate, a heparin, a chondroitin sulfate, or a dermatan sulfate GAG chain. Which type of GAG chain becomes attached to the linker tetrasaccharide is influenced by the structure of the protein core, modifications occurring to the linker tetrasaccharide itself, and the biochemical environment of the Golgi apparatus, where GAG polymerization and modification by sulfation and epimerization take place. The same cell type may produce different GAG chains that vary, depending on the extent of epimerization and sulfation. However, it is not known to what extent these differences are caused by compartmental segregation of protein cores en route through the secretory pathway or by differential recruitment of modifying enzymes during synthesis of different PGs. The topic of this review is how different aspects of protein structure, cellular biochemistry, and compartmentalization may influence GAG synthesis.
Collapse
Affiliation(s)
- Kristian Prydz
- Department of Biosciences, University of Oslo, Box 1066, Blindern OSLO 0316, Norway.
| |
Collapse
|
27
|
Yan J, Leal K, Magupalli VG, Nanou E, Martinez GQ, Scheuer T, Catterall WA. Modulation of CaV2.1 channels by neuronal calcium sensor-1 induces short-term synaptic facilitation. Mol Cell Neurosci 2015; 63:124-31. [PMID: 25447945 DOI: 10.1016/j.mcn.2014.11.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 10/29/2014] [Accepted: 11/03/2014] [Indexed: 12/01/2022] Open
Abstract
Facilitation and inactivation of P/Q-type Ca2+ currents mediated by Ca2+/calmodulin binding to Ca(V)2.1 channels contribute to facilitation and rapid depression of synaptic transmission, respectively. Other calcium sensor proteins displace calmodulin from its binding site and differentially modulate P/Q-type Ca2 + currents, resulting in diverse patterns of short-term synaptic plasticity. Neuronal calcium sensor-1 (NCS-1, frequenin) has been shown to enhance synaptic facilitation, but the underlying mechanism is unclear. We report here that NCS-1 directly interacts with IQ-like motif and calmodulin-binding domain in the C-terminal domain of Ca(V)2.1 channel. NCS-1 reduces Ca2 +-dependent inactivation of P/Q-type Ca2+ current through interaction with the IQ-like motif and calmodulin-binding domain without affecting peak current or activation kinetics. Expression of NCS-1 in presynaptic superior cervical ganglion neurons has no effect on synaptic transmission, eliminating effects of this calcium sensor protein on endogenous N-type Ca2+ currents and the endogenous neurotransmitter release machinery. However, in superior cervical ganglion neurons expressing wild-type Ca(V)2.1 channels, co-expression of NCS-1 induces facilitation of synaptic transmission in response to paired pulses and trains of depolarizing stimuli, and this effect is lost in Ca(V)2.1 channels with mutations in the IQ-like motif and calmodulin-binding domain. These results reveal that NCS-1 directly modulates Ca(V)2.1 channels to induce short-term synaptic facilitation and further demonstrate that CaS proteins are crucial in fine-tuning short-term synaptic plasticity.
Collapse
|
28
|
Xia S, Zhou X, Wang T, Zhang Q, Li Q, Liu Y, Xing Q, Wang L, He L, Zhao X. Experimental validation of candidate schizophrenia gene CALN1 as a target for microRNA-137. Neurosci Lett 2015; 602:110-4. [PMID: 26163462 DOI: 10.1016/j.neulet.2015.07.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 06/18/2015] [Accepted: 07/02/2015] [Indexed: 10/23/2022]
Abstract
MIR137, which encodes microRNA-137 (miR-137), and several of its target genes exhibit genome-wide significant associations with schizophrenia. In a previous study, we analyzed the SNPs in a group of predicted MIR137 target genes and detected genome-wide significant association of schizophrenia with rs2944829 in the CALN1 gene. However, no experimental evidence for CALN1 and MIR137 interaction has yet been reported. In this study, we first computationally analyzed the putative miR-137 target site on CALN1 and predicted that miR-137 binds CALN1 at nucleotide (nt) position 236-242 in the 3'UTR. Then we assayed gene expression by transfecting miR-137 mimics into HEK293 and SH-SY5Y cell lines. Quantitative real-time RT-PCR results showed that the expression level of CALN1 significantly decreased in cells co-transfected with miR-137 mimics compared to cells transfected with the blank control (P=.0046 in HEK293 cell lines, P=.038 in SH-SY5Y cells lines). Finally, we co-transfected different combinations of miRNA mimics and either wild type CALN1 3'UTR or mutant 3'UTR reporters into HEK293 and SH-SY5Y cell lines and assessed the specificity of miRNA binding using a luciferase reporter assay. The transfection of miR-137 mimics corresponded with a considerable reduction of luciferase activity on vectors carrying the target fragment (P=1.17×10(-5), 68% reduction in HEK293 cell line, and P=5.09×10(-6), 32% reduction in SH-SY5Y cell line). This inhibition was impaired by site-directed mutagenesis of the miR-137 target fragment. Our results provide strong evidence that CALN1 is a target of miR-137.
Collapse
Affiliation(s)
- Shihui Xia
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, PR China
| | - Xinyao Zhou
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, PR China
| | - Teng Wang
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, PR China
| | - Qiang Zhang
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, PR China
| | - Qiaoli Li
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, PR China
| | - Yun Liu
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, PR China
| | - Qinghe Xing
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, PR China
| | - Lei Wang
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, PR China
| | - Lin He
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, PR China
| | - Xinzhi Zhao
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, PR China.
| |
Collapse
|
29
|
Hradsky J, Bernstein HG, Marunde M, Mikhaylova M, Kreutz MR. Alternative splicing, expression and cellular localization of Calneuron-1 in the rat and human brain. J Histochem Cytochem 2015; 63:793-804. [PMID: 26116628 DOI: 10.1369/0022155415595841] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 06/20/2015] [Indexed: 01/29/2023] Open
Abstract
Calneuron-1 and -2 are members of the neuronal calcium-binding protein family (nCaBP). They are transmembrane Calmodulin-like EF-hand Ca(2+)-sensors, and a function in the control of Golgi-to-plasma membrane vesicle trafficking has been assigned to both proteins. In this paper, we describe the distribution of Calneuron-1 in rat and human brains. We show that Calneuron-1 is ubiquitously expressed in all brain regions examined. The protein is most abundant in Purkinje cells of the cerebellum and principal neurons of the cortex and limbic brain whereas no expression in glial cells is apparent. In addition, we identify two novel splice isoforms of Calneuron-1 with extended N-termini. These isoforms are particular abundant in the cerebellum. Taken together, these data set grounds for a better understanding of the cellular function of Calneurons.
Collapse
Affiliation(s)
- Johannes Hradsky
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, Magdeburg, Germany (JH, MMarunde, MMikhaylova, MRK)
| | - Hans-Gert Bernstein
- Department of Psychiatry, Faculty of Medicine, Otto-von-Guericke University, Magdeburg, Germany (HGB)
| | - Monika Marunde
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, Magdeburg, Germany (JH, MMarunde, MMikhaylova, MRK)
| | - Marina Mikhaylova
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, Magdeburg, Germany (JH, MMarunde, MMikhaylova, MRK),Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (MMikhaylova)
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, Magdeburg, Germany (JH, MMarunde, MMikhaylova, MRK)
| |
Collapse
|
30
|
Rajamanoharan D, McCue HV, Burgoyne RD, Haynes LP. Modulation of phosphatidylinositol 4-phosphate levels by CaBP7 controls cytokinesis in mammalian cells. Mol Biol Cell 2015; 26:1428-39. [PMID: 25717182 PMCID: PMC4395124 DOI: 10.1091/mbc.e14-07-1243] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 02/13/2015] [Indexed: 12/29/2022] Open
Abstract
For more than 25 years, lysosomes have been known to cluster at the intercellular bridge during cytokinesis, but why has remained a mystery. This study provides evidence that phosphoinositide metabolism is important for this clustering and that lysosome activity is required for cytokinesis. Calcium and phosphoinositide signaling regulate cell division in model systems, but their significance in mammalian cells is unclear. Calcium-binding protein-7 (CaBP7) is a phosphatidylinositol 4-kinaseIIIβ (PI4KIIIβ) inhibitor required during cytokinesis in mammalian cells, hinting at a link between these pathways. Here we characterize a novel association of CaBP7 with lysosomes that cluster at the intercellular bridge during cytokinesis in HeLa cells. We show that CaBP7 regulates lysosome clustering and that PI4KIIIβ is essential for normal cytokinesis. CaBP7 depletion induces lysosome mislocalization, extension of intercellular bridge lifetime, and cytokinesis failure. These data connect phosphoinositide and calcium pathways to lysosome localization and normal cytokinesis in mammalian cells.
Collapse
Affiliation(s)
- Dayani Rajamanoharan
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | - Hannah V McCue
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | - Robert D Burgoyne
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | - Lee P Haynes
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, United Kingdom
| |
Collapse
|
31
|
Rajanikanth V, Sharma AK, Rajyalakshmi M, Chandra K, Chary KVR, Sharma Y. Liaison between Myristoylation and Cryptic EF-Hand Motif Confers Ca2+ Sensitivity to Neuronal Calcium Sensor-1. Biochemistry 2015; 54:1111-22. [DOI: 10.1021/bi501134g] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | - Anand Kumar Sharma
- CSIR-Centre for
Cellular and Molecular Biology (CCMB), Hyderabad 500007, India
| | - Meduri Rajyalakshmi
- CSIR-Centre for
Cellular and Molecular Biology (CCMB), Hyderabad 500007, India
| | - Kousik Chandra
- Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai 400005, India
| | - Kandala V. R. Chary
- Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai 400005, India
- Center
for Interdisciplinary Sciences, Tata Institute of Fundamental Research, Hyderabad 500075, India
| | - Yogendra Sharma
- CSIR-Centre for
Cellular and Molecular Biology (CCMB), Hyderabad 500007, India
| |
Collapse
|
32
|
Navarro G, Aguinaga D, Moreno E, Hradsky J, Reddy PP, Cortés A, Mallol J, Casadó V, Mikhaylova M, Kreutz MR, Lluís C, Canela EI, McCormick PJ, Ferré S. Intracellular calcium levels determine differential modulation of allosteric interactions within G protein-coupled receptor heteromers. CHEMISTRY & BIOLOGY 2014; 21:1546-56. [PMID: 25457181 PMCID: PMC9875831 DOI: 10.1016/j.chembiol.2014.10.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 10/03/2014] [Accepted: 10/06/2014] [Indexed: 01/27/2023]
Abstract
The pharmacological significance of the adenosine A2A receptor (A2AR)-dopamine D2 receptor (D2R) heteromer is well established and it is being considered as an important target for the treatment of Parkinson’s disease and other neuropsychiatric disorders. However, the physiological factors that control its distinctive biochemical properties are still unknown. We demonstrate that different intracellular Ca2+ levels exert a differential modulation of A2AR-D2R heteromer-mediated adenylyl-cyclase and MAPK signaling in striatal cells. This depends on the ability of low and high Ca2+ levels to promote a selective interaction of the heteromer with the neuronal Ca2+-binding proteins NCS-1 and calneuron-1, respectively. These Ca2+-binding proteins differentially modulate allosteric interactions within the A2AR-D2R heteromer, which constitutes a unique cellular device that integrates extracellular (adenosine and dopamine) and intracellular (Ca+2) signals to produce a specific functional response.
Collapse
Affiliation(s)
- Gemma Navarro
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas and Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona 08028, Spain
- Corresponding authors: Dr. Gemma Navarro, Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Diagonal 645, 08028 Barcelona, Spain; ; Dr. Sergi Ferré, Integrative Neurobiology Section, NIDA, IRP, Triad Technology Building, 333 Cassell Dive, Baltimore, MD 21224;
| | - David Aguinaga
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas and Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona 08028, Spain
| | - Estefania Moreno
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas and Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona 08028, Spain
| | - Johannes Hradsky
- Research Group Neuroplasticity, Leibniz-Institute for Neurobiology, Magdeburg 39118, Germany
| | - Pasham P. Reddy
- Research Group Neuroplasticity, Leibniz-Institute for Neurobiology, Magdeburg 39118, Germany
| | - Antoni Cortés
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas and Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona 08028, Spain
| | - Josefa Mallol
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas and Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona 08028, Spain
| | - Vicent Casadó
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas and Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona 08028, Spain
| | - Marina Mikhaylova
- Research Group Neuroplasticity, Leibniz-Institute for Neurobiology, Magdeburg 39118, Germany
- Cell Biology, Utrecht University, Utrecht 3584CH, The Netherlands
| | - Michael R. Kreutz
- Research Group Neuroplasticity, Leibniz-Institute for Neurobiology, Magdeburg 39118, Germany
| | - Carme Lluís
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas and Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona 08028, Spain
| | - Enric I. Canela
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas and Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona 08028, Spain
| | - Peter J. McCormick
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas and Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona 08028, Spain
- School of Pharmacy, University of East Anglia, Norwich NR47TJ, United Kingdom
| | - Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland 21224, USA
- Corresponding authors: Dr. Gemma Navarro, Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Diagonal 645, 08028 Barcelona, Spain; ; Dr. Sergi Ferré, Integrative Neurobiology Section, NIDA, IRP, Triad Technology Building, 333 Cassell Dive, Baltimore, MD 21224;
| |
Collapse
|
33
|
Burgoyne RD, Haynes LP. Sense and specificity in neuronal calcium signalling. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:1921-32. [PMID: 25447549 PMCID: PMC4728190 DOI: 10.1016/j.bbamcr.2014.10.029] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 10/25/2014] [Accepted: 10/29/2014] [Indexed: 11/02/2022]
Abstract
Changes in the intracellular free calcium concentration ([Ca²⁺]i) in neurons regulate many and varied aspects of neuronal function over time scales from microseconds to days. The mystery is how a single signalling ion can lead to such diverse and specific changes in cell function. This is partly due to aspects of the Ca²⁺ signal itself, including its magnitude, duration, localisation and persistent or oscillatory nature. The transduction of the Ca²⁺ signal requires Ca²⁺binding to various Ca²⁺ sensor proteins. The different properties of these sensors are important for differential signal processing and determine the physiological specificity of Ca(2+) signalling pathways. A major factor underlying the specific roles of particular Ca²⁺ sensor proteins is the nature of their interaction with target proteins and how this mediates unique patterns of regulation. We review here recent progress from structural analyses and from functional analyses in model organisms that have begun to reveal the rules that underlie Ca²⁺ sensor protein specificity for target interaction. We discuss three case studies exemplifying different aspects of Ca²⁺ sensor/target interaction. This article is part of a special issue titled the 13th European Symposium on Calcium.
Collapse
Affiliation(s)
- Robert D Burgoyne
- Department of Cellular and Molecular Physiology, The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool, L69 3BX, United Kingdom.
| | - Lee P Haynes
- Department of Cellular and Molecular Physiology, The Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool, L69 3BX, United Kingdom
| |
Collapse
|
34
|
Reddy PP, Raghuram V, Hradsky J, Spilker C, Chakraborty A, Sharma Y, Mikhaylova M, Kreutz MR. Molecular dynamics of the neuronal EF-hand Ca2+-sensor Caldendrin. PLoS One 2014; 9:e103186. [PMID: 25058677 PMCID: PMC4110014 DOI: 10.1371/journal.pone.0103186] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 06/29/2014] [Indexed: 11/18/2022] Open
Abstract
Caldendrin, L- and S-CaBP1 are CaM-like Ca2+-sensors with different N-termini that arise from alternative splicing of the Caldendrin/CaBP1 gene and that appear to play an important role in neuronal Ca2+-signaling. In this paper we show that Caldendrin is abundantly present in brain while the shorter splice isoforms L- and S-CaBP1 are not detectable at the protein level. Caldendrin binds both Ca2+ and Mg2+ with a global Kd in the low µM range. Interestingly, the Mg2+-binding affinity is clearly higher than in S-CaBP1, suggesting that the extended N-terminus might influence Mg2+-binding of the first EF-hand. Further evidence for intra- and intermolecular interactions of Caldendrin came from gel-filtration, surface plasmon resonance, dynamic light scattering and FRET assays. Surprisingly, Caldendrin exhibits very little change in surface hydrophobicity and secondary as well as tertiary structure upon Ca2+-binding to Mg2+-saturated protein. Complex inter- and intramolecular interactions that are regulated by Ca2+-binding, high Mg2+- and low Ca2+-binding affinity, a rigid first EF-hand domain and little conformational change upon titration with Ca2+ of Mg2+-liganted protein suggest different modes of binding to target interactions as compared to classical neuronal Ca2+-sensors.
Collapse
Affiliation(s)
| | - Vijeta Raghuram
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
- Centre for Cellular and Molecular Biology, CSIR, Hyderabad, India
| | - Johannes Hradsky
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Christina Spilker
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | | | - Yogendra Sharma
- Centre for Cellular and Molecular Biology, CSIR, Hyderabad, India
| | - Marina Mikhaylova
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
- Cell Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Michael R. Kreutz
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
- * E-mail:
| |
Collapse
|
35
|
Mikhaylova M, Schumacher A, Borutzki C, Neumann JR, Macharadze T, El-Mousleh T, Wahle P, Zenclussen AC, Kreutz MR. Analysis of Y-P30/Dermcidin expression and properties of the Y-P30 peptide. BMC Res Notes 2014; 7:400. [PMID: 24969620 PMCID: PMC4082292 DOI: 10.1186/1756-0500-7-400] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 06/20/2014] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND The survival promoting peptide Y-P30 has a variety of neuritogenic and neuroprotective effects in vitro and in vivo. In previous work we reported the expression of Y-P30/dermcidin in maternal peripheral blood mononuclear cells (PBMCs) and the transport of the protein to the fetal brain. In this study we analyzed hormonal regulation of Y-P30 in human immune cells and expression of Y-P30 in the placenta. We further studied the stability and secretion of the Y-P30 peptide. RESULTS We found indications that Y-P30 might be produced in human placenta. The Y-P30 mRNA was rarely found in isolated human PBMCs and alpha-feto-protein, human chorionic gonadotropin as well as estradiol combined with progesterone could not induce Y-P30 expression. Y-P30 was found to be extraordinarily stable; therefore, contamination with the peptide and the Y-P30/Dermcidin precursor mRNA is a serious concern in experiments looking at the expression of Y-P30/Dermcidin. In cultured cell lines and primary neurons we found that Y-P30 could be released, but neuronal uptake of Y-P30 was not observed. CONCLUSIONS Our data suggest that a source of Y-P30 apart from eccrine glands might be the placenta. The peptide can be secreted together with the signaling peptide and it might reach the fetal brain where it can exert its neuritogenic functions by binding to neuronal membranes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ana C Zenclussen
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, Magdeburg 39118, Germany.
| | | |
Collapse
|
36
|
Tan J, Brill JA. Cinderella story: PI4P goes from precursor to key signaling molecule. Crit Rev Biochem Mol Biol 2013; 49:33-58. [PMID: 24219382 DOI: 10.3109/10409238.2013.853024] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Phosphatidylinositol lipids are signaling molecules involved in nearly all aspects of cellular regulation. Production of phosphatidylinositol 4-phosphate (PI4P) has long been recognized as one of the first steps in generating poly-phosphatidylinositol phosphates involved in actin organization, cell migration, and signal transduction. In addition, progress over the last decade has brought to light independent roles for PI4P in membrane trafficking and lipid homeostasis. Here, we describe recent advances that reveal the breadth of processes regulated by PI4P, the spectrum of PI4P effectors, and the mechanisms of spatiotemporal control that coordinate crosstalk between PI4P and cellular signaling pathways.
Collapse
Affiliation(s)
- Julie Tan
- Department of Molecular Genetics, University of Toronto , Toronto, Ontario , Canada and
| | | |
Collapse
|
37
|
Extracellular pH and neuronal depolarization serve as dynamic switches to rapidly mobilize trkA to the membrane of adult sensory neurons. J Neurosci 2013; 33:8202-15. [PMID: 23658159 DOI: 10.1523/jneurosci.4408-12.2013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Activation of the nerve growth factor (NGF) receptor trkA and tissue acidosis are critically linked to inflammation-associated nociceptor sensitization. This study explored how increased acidity is linked to sensory neuron sensitization to NGF. Adult Wistar rat primary sensory neurons grown at physiological pH 7.4, then either kept at pH 7.4 or challenged for 30 min in pH 6.5 medium, provided a model of acidosis. Nonpermeabilizing trkA immunofluorescence revealed a significant increase in trkA mobilization to the plasma membrane from intracellular stores in response to proton challenge. This was confirmed using a surface protein biotinylation assay and Brefeldin A disruption of the rough endoplasmic reticulum-Golgi-trans-Golgi network. Mobilization of trkA to the membrane at pH 6.5 was abolished in neurons treated with the acid-sensitive ion channel blocker, amiloride. While elevated levels of NGF-independent trkA phosphorylation occurred at pH 6.5 alone, the level of activation was significantly increased in response to NGF challenge. Exposure of sensory neurons to pH 6.5 medium also resulted in strong calcium (Ca(2+)) transients that were reversible upon reintroduction to physiological pH. The pH 6.5-induced mobilization of trkA to the membrane was Ca(2+) dependent, as BAPTA-AM Ca(2+) chelation abrogated the response. Interestingly, KCl-induced depolarization was sufficient to induce mobilization of trkA to the cell surface at pH 7.4, but did not augment the response to pH 6.5. In conclusion, increased mobilization of trkA to neuronal membranes in response to either acidosis or neuronal depolarization provides two novel mechanisms by which sensory neurons can rapidly sensitize to NGF and has important implications for inflammatory pain states.
Collapse
|
38
|
Clayton EL, Minogue S, Waugh MG. Mammalian phosphatidylinositol 4-kinases as modulators of membrane trafficking and lipid signaling networks. Prog Lipid Res 2013; 52:294-304. [PMID: 23608234 PMCID: PMC3989048 DOI: 10.1016/j.plipres.2013.04.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 04/08/2013] [Indexed: 12/19/2022]
Abstract
The four mammalian phosphatidylinositol 4-kinases modulate inter-organelle lipid trafficking, phosphoinositide signalling and intracellular vesicle trafficking. In addition to catalytic domains required for the synthesis of PI4P, the phosphatidylinositol 4-kinases also contain isoform-specific structural motifs that mediate interactions with proteins such as AP-3 and the E3 ubiquitin ligase Itch, and such structural differences determine isoform-specific roles in membrane trafficking. Moreover, different permutations of phosphatidylinositol 4-kinase isozymes may be required for a single cellular function such as occurs during distinct stages of GPCR signalling and in Golgi to lysosome trafficking. Phosphatidylinositol 4-kinases have recently been implicated in human disease. Emerging paradigms include increased phosphatidylinositol 4-kinase expression in some cancers, impaired functioning associated with neurological pathologies, the subversion of PI4P trafficking functions in bacterial infection and the activation of lipid kinase activity in viral disease. We discuss how the diverse and sometimes overlapping functions of the phosphatidylinositol 4-kinases present challenges for the design of isoform-specific inhibitors in a therapeutic context.
Collapse
Affiliation(s)
- Emma L Clayton
- UCL Institute for Liver & Digestive Health, UCL Royal Free Campus, Rowland Hill Street, London NW3 2PF, United Kingdom
| | | | | |
Collapse
|
39
|
Lin TY, Li BR, Tsai ST, Chen CW, Chen CH, Chen YT, Pan CY. Improved silicon nanowire field-effect transistors for fast protein-protein interaction screening. LAB ON A CHIP 2013; 13:676-684. [PMID: 23235921 DOI: 10.1039/c2lc40772h] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Understanding how proteins interact with each other is the basis for studying the biological mechanisms behind various physiological activities. Silicon nanowire field-effect transistors (SiNW-FETs) are sensitive sensors used to detect biomolecular interactions in real-time. However, the majority of the applications that use SiNW-FETs are for known interactions between different molecules. To explore the capability of SiNW-FETs as fast screening devices to identify unknown interacting molecules, we applied mass spectrometry (MS) to analyze molecules reversibly bound to the SiNW-FETs. Calmodulin (CaM) is a Ca(2+)-sensing protein that is ubiquitously expressed in cells and its interaction with target molecules is Ca(2+)-dependent. By modifying the SiNW-FET surface with glutathione, glutathione S-transferase (GST)-tagged CaM binds reversibly to the SiNW-FET. We first verified the Ca(2+)-dependent interaction between GST-CaM and purified troponin I, which is involved in muscle contraction, through the conductance changes of the SiNW-FET. Furthermore, the cell lysate containing overexpressed Ca(2+)/CaM-dependent protein kinase IIα induced a conductance change in the GST-CaM-modified SiNW-FET. The bound proteins were eluted and subsequently identified by MS as CaM and kinase. In another example, candidate proteins from neuronal cell lysates interacting with calneuron I (CalnI), a CaM-like protein, were captured with a GST-CalnI-modified SiNW-FET. The proteins that interacted with CalnI were eluted and verified by MS. The Ca(2+)-dependent interaction between GST-CalnI and one of the candidates, heat shock protein 70, was re-confirmed via the SiNW-FET measurement. Our results demonstrate the effectiveness of combining MS with SiNW-FETs to quickly screen interacting molecules from cell lysates.
Collapse
Affiliation(s)
- Ti-Yu Lin
- Institute of Zoology, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
40
|
Karpova A, Mikhaylova M, Bera S, Bär J, Reddy P, Behnisch T, Rankovic V, Spilker C, Bethge P, Sahin J, Kaushik R, Zuschratter W, Kähne T, Naumann M, Gundelfinger E, Kreutz M. Encoding and Transducing the Synaptic or Extrasynaptic Origin of NMDA Receptor Signals to the Nucleus. Cell 2013; 152:1119-33. [DOI: 10.1016/j.cell.2013.02.002] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 12/11/2012] [Accepted: 02/01/2013] [Indexed: 10/27/2022]
|
41
|
Hradsky J, Mikhaylova M, Karpova A, Kreutz MR, Zuschratter W. Super-resolution microscopy of the neuronal calcium-binding proteins Calneuron-1 and Caldendrin. Methods Mol Biol 2013; 963:147-169. [PMID: 23296610 DOI: 10.1007/978-1-62703-230-8_10] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Calcium (Ca(2+)) signaling in neurons is mediated by plethora of calcium binding proteins with many of them belonging to the Calmodulin family of calcium sensors. Many studies have shown that the subcellular localization of neuronal EF-hand Ca(2+)-sensors is crucial for their cellular function. To overcome the resolution limit of classical fluorescence and confocal microscopy various imaging techniques have been developed recently that improve the resolution by an order of magnitude in all dimensions. This new microscope techniques make co-localization studies of Ca(2+)-binding proteins more reliable and help to get insights into the macromolecular organization of intracellular structures and signaling pathways beyond the diffraction limit of visible light.
Collapse
Affiliation(s)
- Johannes Hradsky
- Research Group, Neuroplasticity, Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany
| | | | | | | | | |
Collapse
|
42
|
Cancino J, Luini A. Signaling Circuits on the Golgi Complex. Traffic 2012; 14:121-34. [DOI: 10.1111/tra.12022] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 10/12/2012] [Accepted: 10/12/2012] [Indexed: 01/21/2023]
|
43
|
McCue HV, Patel P, Herbert AP, Lian LY, Burgoyne RD, Haynes LP. Solution NMR structure of the Ca2+-bound N-terminal domain of CaBP7: a regulator of golgi trafficking. J Biol Chem 2012; 287:38231-43. [PMID: 22989873 PMCID: PMC3488092 DOI: 10.1074/jbc.m112.402289] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 09/12/2012] [Indexed: 12/12/2022] Open
Abstract
Calcium-binding protein 7 (CaBP7) is a member of the calmodulin (CaM) superfamily that harbors two high affinity EF-hand motifs and a C-terminal transmembrane domain. CaBP7 has been previously shown to interact with and modulate phosphatidylinositol 4-kinase III-β (PI4KIIIβ) activity in in vitro assays and affects vesicle transport in neurons when overexpressed. Here we show that the N-terminal domain (NTD) of CaBP7 is sufficient to mediate the interaction of CaBP7 with PI4KIIIβ. CaBP7 NTD encompasses the two high affinity Ca(2+) binding sites, and structural characterization through multiangle light scattering, circular dichroism, and NMR reveals unique properties for this domain. CaBP7 NTD binds specifically to Ca(2+) but not Mg(2+) and undergoes significant conformational changes in both secondary and tertiary structure upon Ca(2+) binding. The Ca(2+)-bound form of CaBP7 NTD is monomeric and exhibits an open conformation similar to that of CaM. Ca(2+)-bound CaBP7 NTD has a solvent-exposed hydrophobic surface that is more expansive than observed in CaM or CaBP1. Within this hydrophobic pocket, there is a significant reduction in the number of methionine residues that are conserved in CaM and CaBP1 and shown to be important for target recognition. In CaBP7 NTD, these residues are replaced with isoleucine and leucine residues with branched side chains that are intrinsically more rigid than the flexible methionine side chain. We propose that these differences in surface hydrophobicity, charge, and methionine content may be important in determining highly specific interactions of CaBP7 with target proteins, such as PI4KIIIβ.
Collapse
Affiliation(s)
- Hannah V. McCue
- From the Physiological Laboratory, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, and
| | - Pryank Patel
- From the Physiological Laboratory, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, and
| | - Andrew P. Herbert
- From the Physiological Laboratory, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, and
| | - Lu-Yun Lian
- the NMR Centre for Structural Biology, Institute of Integrative Biology, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | - Robert D. Burgoyne
- From the Physiological Laboratory, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, and
| | - Lee P. Haynes
- From the Physiological Laboratory, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, and
| |
Collapse
|
44
|
Phosphatidylinositol 4-Kinases and PI4P Metabolism in the Nervous System: Roles in Psychiatric and Neurological Diseases. Mol Neurobiol 2012; 47:361-72. [DOI: 10.1007/s12035-012-8358-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 09/27/2012] [Indexed: 01/18/2023]
|
45
|
Raghuram V, Sharma Y, Kreutz MR. Ca(2+) sensor proteins in dendritic spines: a race for Ca(2+). Front Mol Neurosci 2012; 5:61. [PMID: 22586368 PMCID: PMC3347464 DOI: 10.3389/fnmol.2012.00061] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2012] [Accepted: 04/18/2012] [Indexed: 12/21/2022] Open
Abstract
Dendritic spines are believed to be micro-compartments of Ca2+ regulation. In a recent study, it was suggested that the ubiquitous and evolutionarily conserved Ca2+ sensor, calmodulin (CaM), is the first to intercept Ca2+ entering the spine and might be responsible for the fast decay of Ca2+ transients in spines. Neuronal calcium sensor (NCS) and neuronal calcium-binding protein (nCaBP) families consist of Ca2+ sensors with largely unknown synaptic functions despite an increasing number of interaction partners. Particularly how these sensors operate in spines in the presence of CaM has not been discussed in detail before. The limited Ca2+ resources and the existence of common targets create a highly competitive environment where Ca2+ sensors compete with each other for Ca2+ and target binding. In this review, we take a simple numerical approach to put forth possible scenarios and their impact on signaling via Ca2+ sensors of the NCS and nCaBP families. We also discuss the ways in which spine geometry and properties of ion channels, their kinetics and distribution, alter the spatio-temporal aspects of Ca2+ transients in dendritic spines, whose interplay with Ca2+ sensors in turn influences the race for Ca2+.
Collapse
Affiliation(s)
- Vijeta Raghuram
- Centre for Cellular and Molecular Biology, CSIR Hyderabad, India
| | | | | |
Collapse
|
46
|
Navarro G, Hradsky J, Lluís C, Casadó V, McCormick PJ, Kreutz MR, Mikhaylova M. NCS-1 associates with adenosine A(2A) receptors and modulates receptor function. Front Mol Neurosci 2012; 5:53. [PMID: 22529776 PMCID: PMC3328853 DOI: 10.3389/fnmol.2012.00053] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 04/02/2012] [Indexed: 11/13/2022] Open
Abstract
Modulation of G protein-coupled receptor (GPCR) signaling by local changes in intracellular calcium concentration is an established function of Calmodulin (CaM) which is known to interact with many GPCRs. Less is known about the functional role of the closely related neuronal EF-hand Ca2+-sensor proteins that frequently associate with CaM targets with different functional outcome. In the present study we aimed to investigate if a target of CaM—the A2A adenosine receptor is able to associate with two other neuronal calcium binding proteins (nCaBPs), namely NCS-1 and caldendrin. Using bioluminescence resonance energy transfer (BRET) and co-immunoprecipitation experiments we show the existence of A2A—NCS-1 complexes in living cells whereas caldendrin did not associate with A2A receptors under the conditions tested. Interestingly, NCS-1 binding modulated downstream A2A receptor intracellular signaling in a Ca2+-dependent manner. Taken together this study provides further evidence that neuronal Ca2+-sensor proteins play an important role in modulation of GPCR signaling.
Collapse
Affiliation(s)
- Gemma Navarro
- Faculty of Biology, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas and Department of Biochemistry and Molecular Biology, University of Barcelona Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
47
|
Haynes LP, McCue HV, Burgoyne RD. Evolution and functional diversity of the Calcium Binding Proteins (CaBPs). Front Mol Neurosci 2012; 5:9. [PMID: 22375103 PMCID: PMC3284769 DOI: 10.3389/fnmol.2012.00009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 01/25/2012] [Indexed: 02/01/2023] Open
Abstract
The mammalian central nervous system (CNS) exhibits a remarkable ability to process, store, and transfer information. Key to these activities is the use of highly regulated and unique patterns of calcium signals encoded by calcium channels and decoded by families of specific calcium-sensing proteins. The largest family of eukaryotic calcium sensors is those related to the small EF-hand containing protein calmodulin (CaM). In order to maximize the usefulness of calcium as a signaling species and to permit the evolution and fine tuning of the mammalian CNS, families of related proteins have arisen that exhibit characteristic calcium binding properties and tissue-, cellular-, and sub-cellular distribution profiles. The Calcium Binding Proteins (CaBPs) represent one such family of vertebrate specific CaM like proteins that have emerged in recent years as important regulators of essential neuronal target proteins. Bioinformatic analyses indicate that the CaBPs consist of two subfamilies and that the ancestral members of these are CaBP1 and CaBP8. The CaBPs have distinct intracellular localizations based on different targeting mechanisms including a novel type-II transmembrane domain in CaBPs 7 and 8 (otherwise known as calneuron II and calneuron I, respectively). Recent work has led to the identification of new target interactions and possible functions for the CaBPs suggesting that they have multiple physiological roles with relevance for the normal functioning of the CNS.
Collapse
Affiliation(s)
- Lee P Haynes
- The Physiological Laboratory, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool Liverpool, UK
| | | | | |
Collapse
|
48
|
Cellular Mechanisms for the Biogenesis and Transport of Synaptic and Dense-Core Vesicles. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 299:27-115. [DOI: 10.1016/b978-0-12-394310-1.00002-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
49
|
Hradsky J, Raghuram V, Reddy PP, Navarro G, Hupe M, Casado V, McCormick PJ, Sharma Y, Kreutz MR, Mikhaylova M. Post-translational membrane insertion of tail-anchored transmembrane EF-hand Ca2+ sensor calneurons requires the TRC40/Asna1 protein chaperone. J Biol Chem 2011; 286:36762-76. [PMID: 21878631 DOI: 10.1074/jbc.m111.280339] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Calneuron-1 and -2 are neuronal EF-hand-type calcium sensor proteins that are prominently targeted to trans-Golgi network membranes and impose a calcium threshold at the Golgi for phosphatidylinositol 4-OH kinase IIIβ activation and the regulated local synthesis of phospholipids that are crucial for TGN-to-plasma membrane trafficking. In this study, we show that calneurons are nonclassical type II tail-anchored proteins that are post-translationally inserted into the endoplasmic reticulum membrane via an association of a 23-amino acid-long transmembrane domain (TMD) with the TRC40/Asna1 chaperone complex. Following trafficking to the Golgi, calneurons are probably retained in the TGN because of the length of the TMD and phosphatidylinositol 4-phosphate lipid binding. Both calneurons rapidly self-associate in vitro and in vivo via their TMD and EF-hand containing the N terminus. Although dimerization and potentially multimerization precludes TRC40/Asna1 binding and thereby membrane insertion, we found no evidence for a cytosolic pool of calneurons and could demonstrate that self-association of calneurons is restricted to membrane-inserted protein. The dimerization properties and the fact that they, unlike every other EF-hand calmodulin-like Ca(2+) sensor, are always associated with membranes of the secretory pathway, including vesicles and plasma membrane, suggests a high degree of spatial segregation for physiological target interactions.
Collapse
Affiliation(s)
- Johannes Hradsky
- Research Group Neuroplasticity, Leibniz-Institute for Neurobiology, 39118 Magdeburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Mikhaylova M, Hradsky J, Kreutz MR. Between promiscuity and specificity: novel roles of EF-hand calcium sensors in neuronal Ca2+ signalling. J Neurochem 2011; 118:695-713. [PMID: 21722133 DOI: 10.1111/j.1471-4159.2011.07372.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In recent years, substantial progress has been made towards an understanding of the physiological function of EF-hand calcium sensor proteins of the Calmodulin (CaM) superfamily in neurons. This deeper appreciation is based on the identification of novel target interactions, structural studies and the discovery of novel signalling mechanisms in protein trafficking and synaptic plasticity, in which CaM-like sensor proteins appear to play a role. However, not all interactions are of plausible physiological relevance and in many cases it is not yet clear how the CaM signaling network relates to the proposed function of other EF-hand sensors. In this review, we will summarize these findings and address some of the open questions on the functional role of EF-hand calcium binding proteins in neurons.
Collapse
Affiliation(s)
- Marina Mikhaylova
- PG Neuroplasticity, Leibniz-Institute for Neurobiology, Magdeburg, Germany
| | | | | |
Collapse
|