1
|
Thomas RE, Mudlaff F, Schweers K, Farmer WT, Suvrathan A. Heterogeneity in Slow Synaptic Transmission Diversifies Purkinje Cell Timing. J Neurosci 2024; 44:e0455242024. [PMID: 39147589 PMCID: PMC11391503 DOI: 10.1523/jneurosci.0455-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/19/2024] [Accepted: 07/31/2024] [Indexed: 08/17/2024] Open
Abstract
The cerebellum plays an important role in diverse brain functions, ranging from motor learning to cognition. Recent studies have suggested that molecular and cellular heterogeneity within cerebellar lobules contributes to functional differences across the cerebellum. However, the specific relationship between molecular and cellular heterogeneity and diverse functional outputs of different regions of the cerebellum remains unclear. Here, we describe a previously unappreciated form of synaptic heterogeneity at parallel fiber synapses to Purkinje cells in the mouse cerebellum (both sexes). In contrast to uniform fast synaptic transmission, we found that the properties of slow synaptic transmission varied by up to threefold across different lobules of the mouse cerebellum, resulting in surprising heterogeneity. Depending on the location of a Purkinje cell, the time of peak of slow synaptic currents varied by hundreds of milliseconds. The duration and decay time of these currents also spanned hundreds of milliseconds, based on lobule. We found that, as a consequence of the heterogeneous synaptic dynamics, the same brief input stimulus was transformed into prolonged firing patterns over a range of timescales that depended on Purkinje cell location.
Collapse
Affiliation(s)
- Riya Elizabeth Thomas
- Centre for Research in Neuroscience, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montréal, Québec H3G 1A4, Canada
- Departments of Neurology and Neurosurgery, McGill University, Montréal, Québec H3G 1A4, Canada
- Pediatrics, McGill University, Montréal, Québec H3G 1A4, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Franziska Mudlaff
- Centre for Research in Neuroscience, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montréal, Québec H3G 1A4, Canada
- Departments of Neurology and Neurosurgery, McGill University, Montréal, Québec H3G 1A4, Canada
- Pediatrics, McGill University, Montréal, Québec H3G 1A4, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Kyra Schweers
- Centre for Research in Neuroscience, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montréal, Québec H3G 1A4, Canada
- Departments of Neurology and Neurosurgery, McGill University, Montréal, Québec H3G 1A4, Canada
- Pediatrics, McGill University, Montréal, Québec H3G 1A4, Canada
| | - William Todd Farmer
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Aparna Suvrathan
- Centre for Research in Neuroscience, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montréal, Québec H3G 1A4, Canada
- Departments of Neurology and Neurosurgery, McGill University, Montréal, Québec H3G 1A4, Canada
- Pediatrics, McGill University, Montréal, Québec H3G 1A4, Canada
| |
Collapse
|
2
|
Li Y, Zhou L, Deng H, Zhang Y, Li G, Yu H, Wu K, Wang F. A switch in the pathway of TRPC3-mediated calcium influx into brain pericytes contributes to capillary spasms after subarachnoid hemorrhage. Neurotherapeutics 2024; 21:e00380. [PMID: 38839450 DOI: 10.1016/j.neurot.2024.e00380] [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: 01/24/2024] [Revised: 05/07/2024] [Accepted: 05/25/2024] [Indexed: 06/07/2024] Open
Abstract
Calcium influx and subsequent elevation of the intracellular calcium concentration ([Ca2+]i) induce contractions of brain pericytes and capillary spasms following subarachnoid hemorrhage. This calcium influx is exerted through cation channels. However, the specific calcium influx pathways in brain pericytes after subarachnoid hemorrhage remain unknown. Transient receptor potential canonical 3 (TRPC3) is the most abundant cation channel potentially involved in calcium influx into brain pericytes and is involved in calcium influx into other cell types either via store-operated calcium entry (SOCE) or receptor-operated calcium entry (ROCE). Therefore, we hypothesized that TRPC3 is associated with [Ca2+]i elevation in brain pericytes, potentially mediating brain pericyte contraction and capillary spasms after subarachnoid hemorrhage. In this study, we isolated rat brain pericytes and demonstrated increased TRPC3 expression and its currents in brain pericytes after subarachnoid hemorrhage. Calcium imaging of brain pericytes revealed that changes in TRPC3 expression mediated a switch from SOCE-dominant to ROCE-dominant calcium influx after subarachnoid hemorrhage, resulting in significantly higher [Ca2+]i levels after SAH. TRPC3 activity in brain pericytes also contributed to capillary spasms and reduction in cerebral blood flow in an in vivo rat model of subarachnoid hemorrhage. Therefore, we suggest that the switch in TRPC3-mediated calcium influx pathways plays a crucial role in the [Ca2+]i elevation in brain pericytes after subarachnoid hemorrhage, ultimately leading to capillary spasms and a reduction in cerebral blood flow.
Collapse
Affiliation(s)
- Yuncong Li
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Lei Zhou
- The Key Laboratory of Stem Cell and Regenerative Medicine of Yunnan Province, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming 650500, China
| | - Hongji Deng
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Yongjin Zhang
- Department of Laboratory for Basic Research, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Guibo Li
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Hanfu Yu
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Kun Wu
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Fei Wang
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| |
Collapse
|
3
|
Sekerková G, Kilic S, Cheng YH, Fredrick N, Osmani A, Kim H, Opal P, Martina M. Phenotypical, genotypical and pathological characterization of the moonwalker mouse, a model of ataxia. Neurobiol Dis 2024; 195:106492. [PMID: 38575093 PMCID: PMC11089908 DOI: 10.1016/j.nbd.2024.106492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/13/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024] Open
Abstract
We performed a comprehensive study of the morphological, functional, and genetic features of moonwalker (MWK) mice, a mouse model of spinocerebellar ataxia caused by a gain of function of the TRPC3 channel. These mice show numerous behavioral symptoms including tremor, altered gait, circling behavior, impaired motor coordination, impaired motor learning and decreased limb strength. Cerebellar pathology is characterized by early and almost complete loss of unipolar brush cells as well as slowly progressive, moderate loss of Purkinje cell (PCs). Structural damage also includes loss of synaptic contacts from parallel fibers, swollen ER structures, and degenerating axons. Interestingly, no obvious correlation was observed between PC loss and severity of the symptoms, as the phenotype stabilizes around 2 months of age, while the cerebellar pathology is progressive. This is probably due to the fact that PC function is severely impaired much earlier than the appearance of PC loss. Indeed, PC firing is already impaired in 3 weeks old mice. An interesting feature of the MWK pathology that still remains to be explained consists in a strong lobule selectivity of the PC loss, which is puzzling considering that TRPC is expressed in every PC. Intriguingly, genetic analysis of MWK cerebella shows, among other alterations, changes in the expression of both apoptosis inducing and resistance factors possibly suggesting that damaged PCs initiate specific cellular pathways that protect them from overt cell loss.
Collapse
Affiliation(s)
- Gabriella Sekerková
- Department of Neuroscience, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA.
| | - Sumeyra Kilic
- Department of Neuroscience, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA
| | - Yen-Hsin Cheng
- Department of Neuroscience, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA
| | - Natalie Fredrick
- Department of Neurology, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA
| | - Anne Osmani
- Department of Neuroscience, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA
| | - Haram Kim
- Department of Neuroscience, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA
| | - Puneet Opal
- Department of Neurology, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA
| | - Marco Martina
- Department of Neuroscience, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA.
| |
Collapse
|
4
|
Tellios V, Maksoud MJE, Nagra R, Jassal G, Lu WY. Neuronal Nitric Oxide Synthase Critically Regulates the Endocannabinoid Pathway in the Murine Cerebellum During Development. CEREBELLUM (LONDON, ENGLAND) 2023; 22:1200-1215. [PMID: 36402869 DOI: 10.1007/s12311-022-01493-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/27/2022] [Indexed: 11/21/2022]
Abstract
The cerebellum is a major site of endocannabinoid (eCB) production and signaling. The predominant eCB within the cerebellum, 2-arachidonoylglycerol (2-AG), is produced by a metabotropic glutamate receptor type 1 (mGluR1)-initiated signaling cascade within Purkinje neurons (PNs). 2-AG retrogradely stimulates cannabinoid 1 receptors (CB1Rs) located on presynaptic terminals. The activated CB1R decreases neurotransmitter release and leads to the production of nitric oxide (NO), a gaseous molecule. Recently, our group discovered that during development in mice lacking neuronal nitric oxide synthase (nNOS-/-), PNs display an excitotoxic phenotype associated with overactivated mGluR1. Considering the importance of mGluR1 in 2-AG synthesis, the present study explored the role of nNOS-derived NO in regulating the eCB pathway within the cerebella of wildtype (WT) and nNOS-/- mice at postnatal day 7 (PD7), 2 weeks (2 W), and 7 weeks (7 W). Our analysis showed that diacylglycerol lipase α, the enzyme that catalyzes 2-AG production, was elevated at early postnatal ages, and followed by elevated levels of 2-AG in nNOS-/- cerebella compared to WT. CB1R expression in nNOS-/- cerebella was upregulated at PD7 but decreased at 2 W and 7 W when compared to age-matched WT mice cerebella. Importantly, treating organotypic nNOS-/- cerebellar slice cultures with an NO-donor-attenuated CB1R levels after 7 days in vitro. In addition, expression of the eCB hydrolases fatty acid amide hydrolase and monoacylglycerol lipase were significantly downregulated in nNOS-/- cerebella compared to WT cerebella at 7 W. Together, these results reveal a novel role for nNOS/NO in regulating eCB signaling in the cerebellum.
Collapse
Affiliation(s)
- Vasiliki Tellios
- Graduate Program of Neuroscience, The University of Western Ontario, London, N6A 5B7, Canada
- Robarts Research Institute, London, N6A 5B7, Canada
| | - Matthew J E Maksoud
- Graduate Program of Neuroscience, The University of Western Ontario, London, N6A 5B7, Canada
- Robarts Research Institute, London, N6A 5B7, Canada
| | | | - Gurneet Jassal
- Department of Physiology and Pharmacology, The University of Western Ontario, London, N6A 5B7, Canada
| | - Wei-Yang Lu
- Graduate Program of Neuroscience, The University of Western Ontario, London, N6A 5B7, Canada.
- Robarts Research Institute, London, N6A 5B7, Canada.
- Department of Physiology and Pharmacology, The University of Western Ontario, London, N6A 5B7, Canada.
| |
Collapse
|
5
|
Mansouri M, Kremser L, Nguyen TP, Kasugai Y, Caberlotto L, Gassmann M, Sarg B, Lindner H, Bettler B, Carboni L, Ferraguti F. Protein Networks Associated with Native Metabotropic Glutamate 1 Receptors (mGlu 1) in the Mouse Cerebellum. Cells 2023; 12:1325. [PMID: 37174725 PMCID: PMC10177021 DOI: 10.3390/cells12091325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/28/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023] Open
Abstract
The metabotropic glutamate receptor 1 (mGlu1) plays a pivotal role in synaptic transmission and neuronal plasticity. Despite the fact that several interacting proteins involved in the mGlu1 subcellular trafficking and intracellular transduction mechanisms have been identified, the protein network associated with this receptor in specific brain areas remains largely unknown. To identify novel mGlu1-associated protein complexes in the mouse cerebellum, we used an unbiased tissue-specific proteomic approach, namely co-immunoprecipitation followed by liquid chromatography/tandem mass spectrometry analysis. Many well-known protein complexes as well as novel interactors were identified, including G-proteins, Homer, δ2 glutamate receptor, 14-3-3 proteins, and Na/K-ATPases. A novel putative interactor, KCTD12, was further investigated. Reverse co-immunoprecipitation with anti-KCTD12 antibodies revealed mGlu1 in wild-type but not in KCTD12-knock-out homogenates. Freeze-fracture replica immunogold labeling co-localization experiments showed that KCTD12 and mGlu1 are present in the same nanodomain in Purkinje cell spines, although at a distance that suggests that this interaction is mediated through interposed proteins. Consistently, mGlu1 could not be co-immunoprecipitated with KCTD12 from a recombinant mammalian cell line co-expressing the two proteins. The possibility that this interaction was mediated via GABAB receptors was excluded by showing that mGlu1 and KCTD12 still co-immunoprecipitated from GABAB receptor knock-out tissue. In conclusion, this study identifies tissue-specific mGlu1-associated protein clusters including KCTD12 at Purkinje cell synapses.
Collapse
Affiliation(s)
- Mahnaz Mansouri
- Department of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.M.); (Y.K.)
| | - Leopold Kremser
- Institute of Medical Biochemistry, Protein Core Facility, Medical University of Innsbruck, 6020 Innsbruck, Austria; (L.K.); (B.S.); (H.L.)
| | | | - Yu Kasugai
- Department of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.M.); (Y.K.)
| | - Laura Caberlotto
- Centre for Computational and Systems Biology (COSBI), The Microsoft Research University of Trento, 38068 Rovereto, Italy;
| | - Martin Gassmann
- Department of Biomedicine, Pharmazentrum, University of Basel, 4056 Basel, Switzerland; (M.G.); (B.B.)
| | - Bettina Sarg
- Institute of Medical Biochemistry, Protein Core Facility, Medical University of Innsbruck, 6020 Innsbruck, Austria; (L.K.); (B.S.); (H.L.)
| | - Herbert Lindner
- Institute of Medical Biochemistry, Protein Core Facility, Medical University of Innsbruck, 6020 Innsbruck, Austria; (L.K.); (B.S.); (H.L.)
| | - Bernhard Bettler
- Department of Biomedicine, Pharmazentrum, University of Basel, 4056 Basel, Switzerland; (M.G.); (B.B.)
| | - Lucia Carboni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| | - Francesco Ferraguti
- Department of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.M.); (Y.K.)
| |
Collapse
|
6
|
Hahn S, Um KB, Kim SW, Kim HJ, Park MK. Proximal dendritic localization of NALCN channels underlies tonic and burst firing in nigral dopaminergic neurons. J Physiol 2023; 601:171-193. [PMID: 36398712 DOI: 10.1113/jp283716] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
In multipolar nigral dopamine (DA) neurons, the highly excitable proximal dendritic compartments (PDCs) and two Na+ -permeable leak channels, TRPC3 and NALCN, play a key role in pacemaking. However, the causal link between them is unknown. Here we report that the proximal dendritic localization of NALCN underlies pacemaking and burst firing in DA neurons. Our morphological analysis of nigral DA neurons reveals that TRPC3 is ubiquitously expressed in the whole somatodendritic compartment, but NALCN is localized within the PDCs. Blocking either TRPC3 or NALCN channels abolished pacemaking. However, only blocking NALCN, not TRPC3, degraded burst discharges. Furthermore, local glutamate uncaging readily induced burst discharges within the PDCs, compared with other parts of the neuron, and NALCN channel inhibition dissipated burst generation, indicating the importance of NALCN to the high excitability of PDCs. Therefore, we conclude that PDCs serve as a common base for tonic and burst firing in nigral DA neurons. KEY POINTS: Midbrain dopamine (DA) neurons are slow pacemakers that can generate tonic and burst firings, and the highly excitable proximal dendritic compartments (PDCs) and two Na+ -permeable leak channels, TRPC3 and NALCN, play a key role in pacemaking. We find that slow tonic firing depends on the basal activity of both the NALCN and TRPC3 channels, but that burst firing does not require TRPC3 channels but relies only on NALCN channels. We find that TRPC3 is ubiquitously expressed in the entire somatodendritic compartment, but that NALCN exists only within the PDCs in nigral DA neurons. We show that NALCN channel localization confers high excitability on PDCs and is essential for burst generation in nigral DA neurons. These results suggest that PDCs serve as a common base for tonic and burst firing in nigral DA neurons.
Collapse
Affiliation(s)
- Suyun Hahn
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Ki Bum Um
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - So Woon Kim
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Hyun Jin Kim
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea.,Samsung Medical Center, Samsung Biomedical Research Institute, Seoul, Korea
| | - Myoung Kyu Park
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea.,Samsung Medical Center, Samsung Biomedical Research Institute, Seoul, Korea
| |
Collapse
|
7
|
Yadav P, Podia M, Kumari SP, Mani I. Glutamate receptor endocytosis and signaling in neurological conditions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 196:167-207. [PMID: 36813358 DOI: 10.1016/bs.pmbts.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The non-essential amino acid glutamate acts as a major excitatory neurotransmitter and plays a significant role in the central nervous system (CNS). It binds with two different types of receptors, ionotropic glutamate receptors (iGluRs) and metabotropic glutamate receptors (mGluRs), responsible for the postsynaptic excitation of neurons. They are important for memory, neural development and communication, and learning. Endocytosis and subcellular trafficking of the receptor are essential for the regulation of receptor expression on the cell membrane and excitation of the cells. The endocytosis and trafficking of the receptor are dependent on its type, ligand, agonist, and antagonist present. This chapter discusses the types of glutamate receptors, their subtypes, and the regulation of their internalization and trafficking. The roles of glutamate receptors in neurological diseases are also briefly discussed.
Collapse
Affiliation(s)
- Prerna Yadav
- Department of Microbiology, University of Delhi, New Delhi, India
| | - Mansi Podia
- Department of Microbiology, University of Delhi, New Delhi, India
| | - Shashi Prabha Kumari
- Department of Microbiology, Ram Lal Anand College, University of Delhi, New Delhi, India
| | - Indra Mani
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India.
| |
Collapse
|
8
|
Fabian CB, Seney ML, Joffe ME. Sex differences and hormonal regulation of metabotropic glutamate receptor synaptic plasticity. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 168:311-347. [PMID: 36868632 PMCID: PMC10392610 DOI: 10.1016/bs.irn.2022.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Striking sex differences exist in presentation and incidence of several psychiatric disorders. For example, major depressive disorder is more prevalent in women than men, and women who develop alcohol use disorder progress through drinking milestones more rapidly than men. With regards to psychiatric treatment responses, women respond more favorably to selective serotonin reuptake inhibitors than men, whereas men have better outcomes when prescribed tricyclic antidepressants. Despite such well-documented biases in incidence, presentation, and treatment response, sex as a biological variable has long been neglected in preclinical and clinical research. An emerging family of druggable targets for psychiatric diseases, metabotropic glutamate (mGlu) receptors are G-protein coupled receptors broadly distributed throughout the central nervous system. mGlu receptors confer diverse neuromodulatory actions of glutamate at the levels of synaptic plasticity, neuronal excitability, and gene transcription. In this chapter, we summarize the current preclinical and clinical evidence for sex differences in mGlu receptor function. We first highlight basal sex differences in mGlu receptor expression and function and proceed to describe how gonadal hormones, notably estradiol, regulate mGlu receptor signaling. We then describe sex-specific mechanisms by which mGlu receptors differentially modulate synaptic plasticity and behavior in basal states and models relevant for disease. Finally, we discuss human research findings and highlight areas in need of further research. Taken together, this review emphasizes how mGlu receptor function and expression can differ across sex. Gaining a more complete understanding of how sex differences in mGlu receptor function contribute to psychiatric diseases will be critical in the development of novel therapeutics that are effective in all individuals.
Collapse
Affiliation(s)
- Carly B Fabian
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States; Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, United States
| | - Marianne L Seney
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States; Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, United States
| | - Max E Joffe
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States; Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
9
|
Hirai H, Fukai Y, Konno A, Hosoi N. Electrophysiological and Imaging Analysis of GFP-Tagged Protein Kinase C γ Translocation in Cerebellar Purkinje Cells. CEREBELLUM (LONDON, ENGLAND) 2022; 21:776-783. [PMID: 35218526 DOI: 10.1007/s12311-022-01384-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/16/2022] [Indexed: 06/14/2023]
Abstract
The cerebellum contains the highest density of protein kinase C (PKC) in the central nervous system. PKCγ, the major isotype accounting for over half of the PKCs in the cerebellum, is expressed exclusively in Purkinje cells (PCs). Inactivated PKCγ, which is localized in the cytoplasm of PC dendrites and soma, begins to translocate to the cell membrane upon activation. However, the physiological conditions that induce PKCγ translocation in PC remain largely unknown. Here, we virally expressed PKCγ-GFP in PCs and examined the conditions that induced its translocation to PC dendrites by whole-cell patch clamp analysis combined with confocal GFP fluorescence imaging. A single or repetitive (150 pulses at 5 Hz for 30 s) electrical stimulation to a climbing fiber (CF), which produced a complex spike(s) in PC, failed to induce translocation of PKCγ-GFP to the dendritic shaft of PCs. Direct current injection (+ 2 nA for 3 s) to PC also did not induce the translocation, although PCs generated simple spikes continuously at high rates. In contrast, high-frequency parallel fiber (PF) stimulation (50 pulses at 50 Hz for 1 s), which triggered action potentials followed by sustained depolarization (known as mGluR1-mediated slow depolarization), caused translocation of cytoplasmic PKCγ-GFP to the membrane. Low-frequency PF stimulation (150 pulses at 5 Hz for 30 s) induced continuous simple spike firing but did not induce translocation. These results suggest that CF-triggered depolarization, which causes Ca2+ influx through voltage-gated Ca2+ channels throughout PC dendrites and somas, is insufficient to induce the translocation of PKCγ, instead requiring high-frequency PF stimulation that activates mGluR1.
Collapse
Affiliation(s)
- Hirokazu Hirai
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan.
- Viral Vector Core, Gunma University Initiative for Advanced Research (GIAR), Maebashi, Gunma, 371-8511, Japan.
| | - Yuuki Fukai
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan
| | - Ayumu Konno
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan
- Viral Vector Core, Gunma University Initiative for Advanced Research (GIAR), Maebashi, Gunma, 371-8511, Japan
| | - Nobutake Hosoi
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan
| |
Collapse
|
10
|
Johnson SL, Tsou WL, Prifti MV, Harris AL, Todi SV. A survey of protein interactions and posttranslational modifications that influence the polyglutamine diseases. Front Mol Neurosci 2022; 15:974167. [PMID: 36187346 PMCID: PMC9515312 DOI: 10.3389/fnmol.2022.974167] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/27/2022] [Indexed: 01/20/2023] Open
Abstract
The presence and aggregation of misfolded proteins has deleterious effects in the nervous system. Among the various diseases caused by misfolded proteins is the family of the polyglutamine (polyQ) disorders. This family comprises nine members, all stemming from the same mutation—the abnormal elongation of a polyQ repeat in nine different proteins—which causes protein misfolding and aggregation, cellular dysfunction and disease. While it is the same type of mutation that causes them, each disease is distinct: it is influenced by regions and domains that surround the polyQ repeat; by proteins with which they interact; and by posttranslational modifications they receive. Here, we overview the role of non-polyQ regions that control the pathogenicity of the expanded polyQ repeat. We begin by introducing each polyQ disease, the genes affected, and the symptoms experienced by patients. Subsequently, we provide a survey of protein-protein interactions and posttranslational modifications that regulate polyQ toxicity. We conclude by discussing shared processes and pathways that bring some of the polyQ diseases together and may serve as common therapeutic entry points for this family of incurable disorders.
Collapse
Affiliation(s)
- Sean L. Johnson
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
| | - Wei-Ling Tsou
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
| | - Matthew V. Prifti
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
| | - Autumn L. Harris
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
- Maximizing Access to Research Careers (MARC) Program, Wayne State University, Detroit, MI, United States
| | - Sokol V. Todi
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
- Maximizing Access to Research Careers (MARC) Program, Wayne State University, Detroit, MI, United States
- Department of Neurology, Wayne State University, Detroit, MI, United States
- *Correspondence: Sokol V. Todi,
| |
Collapse
|
11
|
Wu QW, Kapfhammer JP. The Emerging Key Role of the mGluR1-PKCγ Signaling Pathway in the Pathogenesis of Spinocerebellar Ataxias: A Neurodevelopmental Viewpoint. Int J Mol Sci 2022; 23:ijms23169169. [PMID: 36012439 PMCID: PMC9409119 DOI: 10.3390/ijms23169169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 12/19/2022] Open
Abstract
Spinocerebellar ataxias (SCAs) are a heterogeneous group of autosomal dominantly inherited progressive disorders with degeneration and dysfunction of the cerebellum. Although different subtypes of SCAs are classified according to the disease-associated causative genes, the clinical syndrome of the ataxia is shared, pointing towards a possible convergent pathogenic pathway among SCAs. In this review, we summarize the role of SCA-associated gene function during cerebellar Purkinje cell development and discuss the relationship between SCA pathogenesis and neurodevelopment. We will summarize recent studies on molecules involved in SCA pathogenesis and will focus on the mGluR1-PKCγ signaling pathway evaluating the possibility that this might be a common pathway which contributes to these diseases.
Collapse
|
12
|
Indirect Negative Effect of Mutant Ataxin-1 on Short- and Long-Term Synaptic Plasticity in Mouse Models of Spinocerebellar Ataxia Type 1. Cells 2022; 11:cells11142247. [PMID: 35883691 PMCID: PMC9317252 DOI: 10.3390/cells11142247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 02/04/2023] Open
Abstract
Spinocerebellar ataxia type 1 (SCA1) is an intractable progressive neurodegenerative disease that leads to a range of movement and motor defects and is eventually lethal. Purkinje cells (PC) are typically the first to show signs of degeneration. SCA1 is caused by an expansion of the polyglutamine tract in the ATXN1 gene and the subsequent buildup of mutant Ataxin-1 protein. In addition to its toxicity, mutant Ataxin-1 protein interferes with gene expression and signal transduction in cells. Recently, it is evident that ATXN1 is not only expressed in neurons but also in glia, however, it is unclear the extent to which either contributes to the overall pathology of SCA1. There are various ways to model SCA1 in mice. Here, functional deficits at cerebellar synapses were investigated in two mouse models of SCA1 in which mutant ATXN1 is either nonspecifically expressed in all cell types of the cerebellum (SCA1 knock-in (KI)), or specifically in Bergmann glia with lentiviral vectors expressing mutant ATXN1 under the control of the astrocyte-specific GFAP promoter. We report impairment of motor performance in both SCA1 models. In both cases, prominent signs of astrocytosis were found using immunohistochemistry. Electrophysiological experiments revealed alteration of presynaptic plasticity at synapses between parallel fibers and PCs, and climbing fibers and PCs in SCA1 KI mice, which is not observed in animals expressing mutant ATXN1 solely in Bergmann glia. In contrast, short- and long-term synaptic plasticity was affected in both SCA1 KI mice and glia-targeted SCA1 mice. Thus, non-neuronal mechanisms may underlie some aspects of SCA1 pathology in the cerebellum. By combining the outcomes of our current work with our previous data from the B05 SCA1 model, we further our understanding of the mechanisms of SCA1.
Collapse
|
13
|
Ojima K, Kakegawa W, Yamasaki T, Miura Y, Itoh M, Michibata Y, Kubota R, Doura T, Miura E, Nonaka H, Mizuno S, Takahashi S, Yuzaki M, Hamachi I, Kiyonaka S. Coordination chemogenetics for activation of GPCR-type glutamate receptors in brain tissue. Nat Commun 2022; 13:3167. [PMID: 35710788 PMCID: PMC9203742 DOI: 10.1038/s41467-022-30828-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 05/19/2022] [Indexed: 11/20/2022] Open
Abstract
Direct activation of cell-surface receptors is highly desirable for elucidating their physiological roles. A potential approach for cell-type-specific activation of a receptor subtype is chemogenetics, in which both point mutagenesis of the receptors and designed ligands are used. However, ligand-binding properties are affected in most cases. Here, we developed a chemogenetic method for direct activation of metabotropic glutamate receptor 1 (mGlu1), which plays essential roles in cerebellar functions in the brain. Our screening identified a mGlu1 mutant, mGlu1(N264H), that was activated directly by palladium complexes. A palladium complex showing low cytotoxicity successfully activated mGlu1 in mGlu1(N264H) knock-in mice, revealing that activation of endogenous mGlu1 is sufficient to evoke the critical cellular mechanism of synaptic plasticity, a basis of motor learning in the cerebellum. Moreover, cell-type-specific activation of mGlu1 was demonstrated successfully using adeno-associated viruses in mice, which shows the potential utility of this chemogenetics for clarifying the physiological roles of mGlu1 in a cell-type-specific manner. Cell-type-specific activation of receptors is desirable for elucidating their roles in tissues or animals. Here, the authors developed a chemogenetic method for direct activation of mGlu1, a GPCR-type glutamate receptor subtype, and demonstrate its use in mouse brain tissue.
Collapse
Affiliation(s)
- Kento Ojima
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, 464-8603, Japan.,Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan
| | - Wataru Kakegawa
- Department of Neurophysiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Tokiwa Yamasaki
- Department of Neurophysiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Yuta Miura
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, 464-8603, Japan
| | - Masayuki Itoh
- Department of Neurophysiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Yukiko Michibata
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan
| | - Ryou Kubota
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan
| | - Tomohiro Doura
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, 464-8603, Japan
| | - Eriko Miura
- Department of Neurophysiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Hiroshi Nonaka
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center in Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba, 305-8575, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center in Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba, 305-8575, Japan
| | - Michisuke Yuzaki
- Department of Neurophysiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.
| | - Itaru Hamachi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan.
| | - Shigeki Kiyonaka
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, 464-8603, Japan. .,Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Nagoya, 464-8603, Japan.
| |
Collapse
|
14
|
Maksymetz J, Byun NE, Luessen DJ, Li B, Barry RL, Gore JC, Niswender CM, Lindsley CW, Joffe ME, Conn PJ. mGlu 1 potentiation enhances prelimbic somatostatin interneuron activity to rescue schizophrenia-like physiological and cognitive deficits. Cell Rep 2021; 37:109950. [PMID: 34731619 PMCID: PMC8628371 DOI: 10.1016/j.celrep.2021.109950] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 08/09/2021] [Accepted: 10/14/2021] [Indexed: 01/03/2023] Open
Abstract
Evidence for prefrontal cortical (PFC) GABAergic dysfunction is one of the most consistent findings in schizophrenia and may contribute to cognitive deficits. Recent studies suggest that the mGlu1 subtype of metabotropic glutamate receptor regulates cortical inhibition; however, understanding the mechanisms through which mGlu1 positive allosteric modulators (PAMs) regulate PFC microcircuit function and cognition is essential for advancing these potential therapeutics toward the clinic. We report a series of electrophysiology, optogenetic, pharmacological magnetic resonance imaging, and animal behavior studies demonstrating that activation of mGlu1 receptors increases inhibitory transmission in the prelimbic PFC by selective excitation of somatostatin-expressing interneurons (SST-INs). An mGlu1 PAM reverses cortical hyperactivity and concomitant cognitive deficits induced by N-methyl-d-aspartate (NMDA) receptor antagonists. Using in vivo optogenetics, we show that prelimbic SST-INs are necessary for mGlu1 PAM efficacy. Collectively, these findings suggest that mGlu1 PAMs could reverse cortical GABAergic deficits and exhibit efficacy in treating cognitive dysfunction in schizophrenia.
Collapse
Affiliation(s)
- James Maksymetz
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Nellie E Byun
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Deborah J Luessen
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Brianna Li
- Vanderbilt University, Nashville, TN 37232, USA
| | - Robert L Barry
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Radiology & Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - John C Gore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Radiology & Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Max E Joffe
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
15
|
Armelloni S, Mattinzoli D, Ikehata M, Alfieri C, Belingheri M, Moroni G, Cresseri D, Passerini P, Cerutti R, Messa P. Urinary mRNA Expression of Glomerular Podocyte Markers in Glomerular Disease and Renal Transplant. Diagnostics (Basel) 2021; 11:1499. [PMID: 34441433 PMCID: PMC8392587 DOI: 10.3390/diagnostics11081499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 11/20/2022] Open
Abstract
The research of novel markers in urinary samples, for the description of renal damage, is of high interest, and several works demonstrated the value of urinary mRNA quantification for the search of events related to renal disease or affecting the outcome of transplant kidneys. In the present pilot study, a comparison of the urine mRNA expression of specific podocyte markers among patients who had undergone clinical indication to renal transplanted (RTx, n = 20) and native (N, n = 18) renal biopsy was performed. The aim of this work was to identify genes involved in podocytes signaling and cytoskeletal regulation (NPHS1, NPHS2, SYNPO, WT1, TRPC6, GRM1, and NEUROD) in respect to glomerular pathology. We considered some genes relevant for podocytes signaling and for the function of the glomerular filter applying an alternative normalization approach. Our results demonstrate the WT1 urinary mRNA increases in both groups and it is helpful for podocyte normalization. Furthermore, an increase in the expression of TRPC6 after all kinds of normalizations was observed. According to our data, WT1 normalization might be considered an alternative approach to correct the expression of urinary mRNA. In addition, our study underlines the importance of slit diaphragm proteins involved in calcium disequilibrium, such as TRPC6.
Collapse
Affiliation(s)
- Silvia Armelloni
- Renal Research Laboratory, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.A.); (D.M.); (M.I.)
| | - Deborah Mattinzoli
- Renal Research Laboratory, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.A.); (D.M.); (M.I.)
| | - Masami Ikehata
- Renal Research Laboratory, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.A.); (D.M.); (M.I.)
| | - Carlo Alfieri
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy;
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.B.); (G.M.); (D.C.); (P.P.); (R.C.)
| | - Mirco Belingheri
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.B.); (G.M.); (D.C.); (P.P.); (R.C.)
| | - Gabrilella Moroni
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.B.); (G.M.); (D.C.); (P.P.); (R.C.)
| | - Donata Cresseri
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.B.); (G.M.); (D.C.); (P.P.); (R.C.)
| | - Patrizia Passerini
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.B.); (G.M.); (D.C.); (P.P.); (R.C.)
| | - Roberta Cerutti
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.B.); (G.M.); (D.C.); (P.P.); (R.C.)
| | - Piergiorgio Messa
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy;
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.B.); (G.M.); (D.C.); (P.P.); (R.C.)
| |
Collapse
|
16
|
Tellios V, Maksoud MJE, Xiang YY, Lu WY. Nitric Oxide Critically Regulates Purkinje Neuron Dendritic Development Through a Metabotropic Glutamate Receptor Type 1-Mediated Mechanism. THE CEREBELLUM 2021; 19:510-526. [PMID: 32270464 DOI: 10.1007/s12311-020-01125-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Nitric oxide (NO), specifically derived from neuronal nitric oxide synthase (nNOS), is a well-established regulator of synaptic transmission in Purkinje neurons (PNs), governing fundamental processes such as motor learning and coordination. Previous phenotypic analyses showed similar cerebellar structures between neuronal nitric oxide null (nNOS-/-) and wild-type (WT) adult male mice, despite prominent ataxic behavior within nNOS-/- mice. However, a study has yet to characterize PN molecular structure and their excitatory inputs during development in nNOS-/- mice. This study is the first to explore morphological abnormalities within the cerebellum of nNOS-/- mice, using immunohistochemistry and immunoblotting. This study sought to examine PN dendritic morphology and the expression of metabotropic glutamate receptor type 1 (mGluR1), vesicular glutamate transporter type 1 and 2 (vGluT1 and vGluT2), stromal interaction molecule 1 (STIM1), and calpain-1 within PNs of WT and nNOS-/- mice at postnatal day 7 (PD7), 2 weeks (2W), and 7 weeks (7W) of age. Results showed a decrease in PN dendritic branching at PD7 in nNOS-/- cerebella, while aberrant dendritic spine formation was noted in adult ages. Total protein expression of mGluR1 was decreased in nNOS-/- cerebella across development, while vGluT2, STIM1, and calpain-1 were significantly increased. Ex vivo treatment of WT slices with NOS inhibitor L-NAME increased calpain-1 expression, whereas treating nNOS-/- cerebellar slices with NO donor NOC-18 decreased calpain-1. Moreover, mGluR1 agonist DHPG increased calpain-1 in WT, but not in nNOS-/- slices. Together, these results indicate a novel role for nNOS/NO signaling in PN development, particularly by regulating an mGluR1-initiated calcium signaling mechanism.
Collapse
Affiliation(s)
- Vasiliki Tellios
- Graduate Program of Neuroscience, The University of Western Ontario, London, N6A 5B7, Canada.,Robarts Research Institute, London, N6A 5B7, Canada
| | - Matthew J E Maksoud
- Graduate Program of Neuroscience, The University of Western Ontario, London, N6A 5B7, Canada.,Robarts Research Institute, London, N6A 5B7, Canada
| | | | - Wei-Yang Lu
- Graduate Program of Neuroscience, The University of Western Ontario, London, N6A 5B7, Canada. .,Robarts Research Institute, London, N6A 5B7, Canada. .,Department of Physiology and Pharmacology, The University of Western Ontario, London, N6A 5B7, Canada.
| |
Collapse
|
17
|
Jitprapaikulsan J, Paul P, Thakolwiboon S, Mittal SO, Pittock SJ, Dubey D. Paraneoplastic neurological syndrome: an evolving story. Neurooncol Pract 2021; 8:362-374. [PMID: 34277016 DOI: 10.1093/nop/npab002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Paraneoplastic neurological syndrome (PNS) comprises a group of neurological disorders that result from a misguided immune response to the nervous system triggered by a distant tumor. These disorders frequently manifest before the diagnosis of the underlying neoplasm. Since the first reported case in 1888 by Oppenheim, the knowledge in this area has evolved rapidly. Several classic PNS have been described, such as limbic encephalitis, paraneoplastic cerebellar degeneration, encephalomyelitis, opsoclonus-myoclonus, sensory neuronopathy, Lambert-Eaton Myasthenic syndrome, and chronic gastrointestinal dysmotility. It is now recognized that PNS can have varied nonclassical manifestations that extend beyond the traditional syndromic descriptions. Multiple onconeural antibodies with high specificity for certain tumor types and neurological phenotypes have been discovered over the past 3 decades. Increasing use of immune checkpoint inhibitors (ICIs) has led to increased recognition of neurologic ICI-related adverse events. Some of these resemble PNS. In this article, we review the clinical, oncologic, and immunopathogenic associations of PNS.
Collapse
Affiliation(s)
- Jiraporn Jitprapaikulsan
- Department of Neurology, Mayo Clinic, Rochester, Minnesota.,Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pritikanta Paul
- Department of Neurology, Mayo Clinic, Rochester, Minnesota.,Department of Neurology and Rehabilitation, University of Illinois at Chicago, Chicago, Illinois
| | - Smathorn Thakolwiboon
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Shivam Om Mittal
- Department of Neurology, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
| | - Sean J Pittock
- Department of Neurology, Mayo Clinic, Rochester, Minnesota.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota.,Center for MS and Autoimmune Neurology, Mayo Clinic, Rochester, Minnesota
| | - Divyanshu Dubey
- Department of Neurology, Mayo Clinic, Rochester, Minnesota.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota.,Center for MS and Autoimmune Neurology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
18
|
Choi DW. Excitotoxicity: Still Hammering the Ischemic Brain in 2020. Front Neurosci 2020; 14:579953. [PMID: 33192266 PMCID: PMC7649323 DOI: 10.3389/fnins.2020.579953] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
Interest in excitotoxicity expanded following its implication in the pathogenesis of ischemic brain injury in the 1980s, but waned subsequent to the failure of N-methyl-D-aspartate (NMDA) antagonists in high profile clinical stroke trials. Nonetheless there has been steady progress in elucidating underlying mechanisms. This review will outline the historical path to current understandings of excitotoxicity in the ischemic brain, and suggest that this knowledge should be leveraged now to develop neuroprotective treatments for stroke.
Collapse
Affiliation(s)
- Dennis W Choi
- Department of Neurology, SUNY Stony Brook, Stony Brook, NY, United States
| |
Collapse
|
19
|
Tsumagari R, Maruo K, Kakizawa S, Ueda S, Yamanoue M, Saito H, Suzuki N, Shirai Y. Precise Regulation of the Basal PKCγ Activity by DGKγ Is Crucial for Motor Coordination. Int J Mol Sci 2020; 21:ijms21217866. [PMID: 33114041 PMCID: PMC7660329 DOI: 10.3390/ijms21217866] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/19/2020] [Accepted: 10/21/2020] [Indexed: 01/26/2023] Open
Abstract
Diacylglycerol kinase γ (DGKγ) is a lipid kinase to convert diacylglycerol (DG) to phosphatidic acid (PA) and indirectly regulates protein kinase C γ (PKCγ) activity. We previously reported that the basal PKCγ upregulation impairs cerebellar long-term depression (LTD) in the conventional DGKγ knockout (KO) mice. However, the precise mechanism in impaired cerebellar LTD by upregulated PKCγ has not been clearly understood. Therefore, we first produced Purkinje cell-specific DGKγ KO (tm1d) mice to investigate the specific function of DGKγ in Purkinje cells and confirmed that tm1d mice showed cerebellar motor dysfunction in the rotarod and beam tests, and the basal PKCγ upregulation but not PKCα in the cerebellum of tm1d mice. Then, the LTD-induced chemical stimulation, K-glu (50 mM KCl + 100 µM, did not induce phosphorylation of PKCα and dissociation of GluR2 and glutamate receptor interacting protein (GRIP) in the acute cerebellar slices of tm1d mice. Furthermore, treatment with the PKCγ inhibitor, scutellarin, rescued cerebellar LTD, with the phosphorylation of PKCα and the dissociation of GluR2 and GRIP. In addition, nonselective transient receptor potential cation channel type 3 (TRPC3) was negatively regulated by upregulated PKCγ. These results demonstrated that DGKγ contributes to cerebellar LTD by regulation of the basal PKCγ activity.
Collapse
Affiliation(s)
- Ryosuke Tsumagari
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences, Kobe University, Kobe 657-8501, Japan; (R.T.); (K.M.); (S.U.); (M.Y.)
| | - Kenta Maruo
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences, Kobe University, Kobe 657-8501, Japan; (R.T.); (K.M.); (S.U.); (M.Y.)
| | - Sho Kakizawa
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan;
| | - Shuji Ueda
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences, Kobe University, Kobe 657-8501, Japan; (R.T.); (K.M.); (S.U.); (M.Y.)
| | - Minoru Yamanoue
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences, Kobe University, Kobe 657-8501, Japan; (R.T.); (K.M.); (S.U.); (M.Y.)
| | - Hiromitsu Saito
- Department of Animal Functional Genomics of Advanced Science Research Promotion Center, Mie University Organization for the Promotion of Regional Innovation, Tsu 514-8507, Japan; (H.S.); (N.S.)
| | - Noboru Suzuki
- Department of Animal Functional Genomics of Advanced Science Research Promotion Center, Mie University Organization for the Promotion of Regional Innovation, Tsu 514-8507, Japan; (H.S.); (N.S.)
| | - Yasuhito Shirai
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences, Kobe University, Kobe 657-8501, Japan; (R.T.); (K.M.); (S.U.); (M.Y.)
- Correspondence: ; Tel.: +81-078-803-5887
| |
Collapse
|
20
|
Kumar A. Calcium Signaling During Brain Aging and Its Influence on the Hippocampal Synaptic Plasticity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:985-1012. [PMID: 31646542 DOI: 10.1007/978-3-030-12457-1_39] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Calcium (Ca2+) ions are highly versatile intracellular signaling molecules and are universal second messenger for regulating a variety of cellular and physiological functions including synaptic plasticity. Ca2+ homeostasis in the central nervous system endures subtle dysregulation with advancing age. Research has provided abundant evidence that brain aging is associated with altered neuronal Ca2+ regulation and synaptic plasticity mechanisms. Much of the work has focused on the hippocampus, a brain region critically involved in learning and memory, which is particularly susceptible to dysfunction during aging. The current chapter takes a specific perspective, assessing various Ca2+ sources and the influence of aging on Ca2+ sources and synaptic plasticity in the hippocampus. Integrating the knowledge of the complexity of age-related alterations in neuronal Ca2+ signaling and synaptic plasticity mechanisms will positively shape the development of highly effective therapeutics to treat brain disorders including cognitive impairment associated with aging and neurodegenerative disease.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
21
|
Environmental enrichment restores the reduced expression of cerebellar synaptophysin and the motor coordination impairment in rats prenatally treated with betamethasone. Physiol Behav 2019; 209:112590. [PMID: 31252027 DOI: 10.1016/j.physbeh.2019.112590] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 06/08/2019] [Accepted: 06/24/2019] [Indexed: 11/23/2022]
Abstract
Preterm babies treated with synthetic glucocorticoids in utero exhibit behavioural alterations and disturbances in brain maturation during postnatal life. Accordingly, it has been shown in preclinical studies that SGC exposure at a clinical dose alters the presynaptic and postsynaptic structures and results in synaptic impairments. However, the precise mechanism by which SGC exposure impairs synaptic protein expression and its implications are not fully elucidated. Therefore, the purpose of this study was to investigate the effect of prenatal exposure to a clinical dose of betamethasone on the pre- and postsynaptic proteins expression in the developing rat cerebellum and prefrontal cortex, whose synchronized synaptic activity is crucial for motor control and learning. Consequently, the first objective of the present study was to determine whether prenatal betamethasone -equivalent to the clinically used dose- alters cerebellar vermal and cortical expression of synaptophysin, synaptotagmin I, post-synaptic density protein 95 and gephyrin - four important pre- and post-synaptic proteins, respectively- at a relevant adolescent stage. In addition, our second objective was to assess whether prenatal betamethasone administration induced coordination impairment using a rotarod test. On the other hand, it has been shown that the environmental enrichment is capable of improving synaptic transmission and recovering various behavioural impairments. Nevertheless, there is not enough information about the effect of this non-pharmacological preclinical approach on the regulation of this cerebellar and cortical synaptic proteins. Therefore, the third objective of this study was to examine whether environmental enrichment exposure could recover the possible molecular and behavioural impairments in the offspring at the same developmental stage. The principal data showed that adolescent rats prenatally treated with betamethasone exhibited underexpression of synaptophysin in the vermal cerebellum, but not change in levels of synaptotagmin I, post-synaptic density protein 95 and gephyrin. Analysis of the same pre- and post-synaptic proteins no showed differences in the frontal cortex of the same rats. These results were accompanied by an increase in the number of falls in the rotarod test, when the speed of rotation was fixed and when it was in acceleration, which means motor coordination impairments. Importantly, we found that environmental enrichment restores the betamethasone-induced reduction in the cerebellar synaptophysin together with a recover in the motor coordination impairments in prenatally betamethasone-exposed adolescent rats.
Collapse
|
22
|
Wu B, Blot FG, Wong AB, Osório C, Adolfs Y, Pasterkamp RJ, Hartmann J, Becker EB, Boele HJ, De Zeeuw CI, Schonewille M. TRPC3 is a major contributor to functional heterogeneity of cerebellar Purkinje cells. eLife 2019; 8:45590. [PMID: 31486767 PMCID: PMC6733575 DOI: 10.7554/elife.45590] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 08/18/2019] [Indexed: 12/12/2022] Open
Abstract
Despite the canonical homogeneous character of its organization, the cerebellum plays differential computational roles in distinct sensorimotor behaviors. Previously, we showed that Purkinje cell (PC) activity differs between zebrin-negative (Z–) and zebrin-positive (Z+) modules (Zhou et al., 2014). Here, using gain-of-function and loss-of-function mouse models, we show that transient receptor potential cation channel C3 (TRPC3) controls the simple spike activity of Z–, but not Z+ PCs. In addition, TRPC3 regulates complex spike rate and their interaction with simple spikes, exclusively in Z– PCs. At the behavioral level, TRPC3 loss-of-function mice show impaired eyeblink conditioning, which is related to Z– modules, whereas compensatory eye movement adaptation, linked to Z+ modules, is intact. Together, our results indicate that TRPC3 is a major contributor to the cellular heterogeneity that introduces distinct physiological properties in PCs, conjuring functional heterogeneity in cerebellar sensorimotor integration.
Collapse
Affiliation(s)
- Bin Wu
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - François Gc Blot
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Aaron Benson Wong
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Catarina Osório
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Youri Adolfs
- Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Jana Hartmann
- Institute of Neuroscience, Technical University Munich, Munich, Germany
| | - Esther Be Becker
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Henk-Jan Boele
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Chris I De Zeeuw
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands.,Netherlands Institute for Neuroscience, Royal Dutch Academy for Arts and Sciences, Amsterdam, Netherlands
| | | |
Collapse
|
23
|
Phillips RS, John TT, Koizumi H, Molkov YI, Smith JC. Biophysical mechanisms in the mammalian respiratory oscillator re-examined with a new data-driven computational model. eLife 2019; 8:41555. [PMID: 30907727 PMCID: PMC6433470 DOI: 10.7554/elife.41555] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 02/07/2019] [Indexed: 12/11/2022] Open
Abstract
An autorhythmic population of excitatory neurons in the brainstem pre-Bötzinger complex is a critical component of the mammalian respiratory oscillator. Two intrinsic neuronal biophysical mechanisms—a persistent sodium current (INaP) and a calcium-activated non-selective cationic current (ICAN)—were proposed to individually or in combination generate cellular- and circuit-level oscillations, but their roles are debated without resolution. We re-examined these roles in a model of a synaptically connected population of excitatory neurons with ICAN and INaP. This model robustly reproduces experimental data showing that rhythm generation can be independent of ICAN activation, which determines population activity amplitude. This occurs when ICAN is primarily activated by neuronal calcium fluxes driven by synaptic mechanisms. Rhythm depends critically on INaP in a subpopulation forming the rhythmogenic kernel. The model explains how the rhythm and amplitude of respiratory oscillations involve distinct biophysical mechanisms.
Collapse
Affiliation(s)
- Ryan S Phillips
- Cellular and Systems Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, United States.,Department of Physics, University of New Hampshire, Durham, United States
| | - Tibin T John
- Cellular and Systems Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, United States
| | - Hidehiko Koizumi
- Cellular and Systems Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, United States
| | - Yaroslav I Molkov
- Department of Mathematics and Statistics, Georgia State University, Atlanta, United States.,Neuroscience Institute, Georgia State University, Atlanta, United States
| | - Jeffrey C Smith
- Cellular and Systems Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, United States
| |
Collapse
|
24
|
Jayakumar S, Richhariya S, Deb BK, Hasan G. A Multicomponent Neuronal Response Encodes the Larval Decision to Pupariate upon Amino Acid Starvation. J Neurosci 2018; 38:10202-10219. [PMID: 30301757 PMCID: PMC6246885 DOI: 10.1523/jneurosci.1163-18.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 09/21/2018] [Accepted: 09/24/2018] [Indexed: 12/12/2022] Open
Abstract
Organisms need to coordinate growth with development, particularly in the context of nutrient availability. Thus, multiple ways have evolved to survive extrinsic nutrient deprivation during development. In Drosophila, growth occurs during larval development. Larvae are thus critically dependent on nutritional inputs; but after critical weight, they pupariate even when starved. How nutrient availability is coupled to the internal metabolic state for the decision to pupariate needs better understanding. We had earlier identified glutamatergic interneurons in the ventral ganglion that regulate pupariation on a protein-deficient diet. Here we report that Drosophila third instar larvae (either sex) sense arginine to evaluate their nutrient environment using an amino acid transporter Slimfast. The glutamatergic interneurons integrate external protein availability with internal metabolic state through neuropeptide signals. IP3-mediated calcium release and store-operated calcium entry are essential in these glutamatergic neurons for such integration and alter neuronal function by reducing the expression of multiple ion channels.SIGNIFICANCE STATEMENT Coordinating growth with development, in the context of nutrient availability is a challenge for all organisms in nature. After attainment of "critical weight," insect larvae can pupariate, even in the absence of nutrition. Mechanism(s) that stimulate appropriate cellular responses and allow normal development on a nutritionally deficient diet remain to be understood. Here, we demonstrate that nutritional deprivation, in postcritical weight larvae, is sensed by special sensory neurons through an amino acid transporter that detects loss of environmental arginine. This information is integrated by glutamatergic interneurons with the internal metabolic state through neuropeptide signals. These glutamatergic interneurons require calcium-signaling-regulated expression of a host of neuronal channels to generate complex calcium signals essential for pupariation on a protein-deficient diet.
Collapse
Affiliation(s)
| | | | - Bipan Kumar Deb
- National Centre for Biological Sciences, TIFR, Bangalore 560065
| | - Gaiti Hasan
- National Centre for Biological Sciences, TIFR, Bangalore 560065
| |
Collapse
|
25
|
Chen Y, Yan Q, Zhou P, Li S, Zhu F. HERV-W env regulates calcium influx via activating TRPC3 channel together with depressing DISC1 in human neuroblastoma cells. J Neurovirol 2018; 25:101-113. [PMID: 30397826 DOI: 10.1007/s13365-018-0692-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/06/2018] [Accepted: 10/24/2018] [Indexed: 12/11/2022]
Abstract
The activation and involvement of human endogenous retroviruses W family envelope gene (HERV-W env, also called ERVWE1) have been reported in several neuropsychiatric disorders, including schizophrenia, as well as in multiple sclerosis (MS). Dysregulation of intracellular calcium content is also involved in the pathogenesis of these diseases. Our previous studies showed that HERV-W env overexpression results in activation of small conductance Ca2+-activated K+ channel protein 3 (SK3), a potential risk factor for schizophrenia. In the present study, we aimed to elucidate the relationship between HERV-W env and calcium signaling in schizophrenia. Our results showed that HERV-W env could induce Ca2+ influx in two human neuroblastoma cell lines and upregulate the expression and activation of TRPC3 in cells. The abnormal increase in intracellular Ca2+ concentration was inhibited by addition of the TRPC3 channel blocker pyr3, demonstrating that the Ca2+ influx induced by HERV-W env was TRPC3-dependent. Further experiments showed that HERV-W env overexpression downregulated DISC1, while knockdown of DISC1 promoted activation of TRPC3 without affecting TRPC3 expression. In conclusion, HERV-W env induced Ca2+ influx in human neuroblastoma cells by activating the TRPC3 channel through directly regulating its expression or downregulating DISC1, which could also increase TRPC3 activation without affecting TRPC3 expression. These findings provide new insights into how HERV-W env affects neuronal activity and contributes to the pathogenesis of schizophrenia.
Collapse
Affiliation(s)
- Yatang Chen
- Department of Medical Microbiology, School of Medicine, Wuhan University, 185 Donghu Road, Wuhan, 430071, People's Republic of China
| | - Qiujin Yan
- Department of Medical Microbiology, School of Medicine, Wuhan University, 185 Donghu Road, Wuhan, 430071, People's Republic of China
| | - Ping Zhou
- Department of Medical Microbiology, School of Medicine, Wuhan University, 185 Donghu Road, Wuhan, 430071, People's Republic of China
| | - Shan Li
- Department of Integrated Medicine, Dongfeng Hospital, Hubei University of Medicine, Hubei, 442000, People's Republic of China
| | - Fan Zhu
- Department of Medical Microbiology, School of Medicine, Wuhan University, 185 Donghu Road, Wuhan, 430071, People's Republic of China. .,Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, 430071, People's Republic of China.
| |
Collapse
|
26
|
Deurloo MHS, Turlova E, Chen WL, Lin YW, Tam E, Tassew NG, Wu M, Huang YC, Crawley JN, Monnier PP, Groffen AJA, Sun HS, Osborne LR, Feng ZP. Transcription Factor 2I Regulates Neuronal Development via TRPC3 in 7q11.23 Disorder Models. Mol Neurobiol 2018; 56:3313-3325. [PMID: 30120731 PMCID: PMC6477017 DOI: 10.1007/s12035-018-1290-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 07/31/2018] [Indexed: 12/24/2022]
Abstract
Williams syndrome (WS) and 7q11.23 duplication syndrome (Dup7q11.23) are neurodevelopmental disorders caused by the deletion and duplication, respectively, of ~ 25 protein-coding genes on chromosome 7q11.23. The general transcription factor 2I (GTF2I, protein TFII-I) is one of these proteins and has been implicated in the neurodevelopmental phenotypes of WS and Dup7q11.23. Here, we investigated the effect of copy number alterations in Gtf2i on neuronal maturation and intracellular calcium entry mechanisms known to be associated with this process. Mice with a single copy of Gtf2i (Gtf2i+/Del) had increased axonal outgrowth and increased TRPC3-mediated calcium entry upon carbachol stimulation. In contrast, mice with 3 copies of Gtf2i (Gtf2i+/Dup) had decreases in axon outgrowth and in TRPC3-mediated calcium entry. The underlying mechanism was that TFII-I did not affect TRPC3 protein expression, while it regulated TRPC3 membrane translocation. Together, our results provide novel functional insight into the cellular mechanisms that underlie neuronal maturation in the context of the 7q11.23 disorders.
Collapse
Affiliation(s)
- Marielle H S Deurloo
- Department of Physiology, University of Toronto, 1 King's College Circle, 3306 Medical Sciences Building, Toronto, ON, M5S 1A8, Canada.,Department of Functional Genomics, CNCR, Neuroscience Campus Amsterdam, VU University and VU Medical Center, 1081 HV, Amsterdam, Netherlands
| | - Ekaterina Turlova
- Department of Physiology, University of Toronto, 1 King's College Circle, 3306 Medical Sciences Building, Toronto, ON, M5S 1A8, Canada.,Department of Surgery, University of Toronto, 1 King's College Circle, 1184 Medical Sciences Building, Toronto, ON, M5S 1A8, Canada
| | - Wen-Liang Chen
- Department of Physiology, University of Toronto, 1 King's College Circle, 3306 Medical Sciences Building, Toronto, ON, M5S 1A8, Canada.,Department of Surgery, University of Toronto, 1 King's College Circle, 1184 Medical Sciences Building, Toronto, ON, M5S 1A8, Canada
| | - You Wei Lin
- Department of Physiology, University of Toronto, 1 King's College Circle, 3306 Medical Sciences Building, Toronto, ON, M5S 1A8, Canada
| | - Elaine Tam
- Department of Medicine, University of Toronto, 661 University Avenue, MaRS Centre, 1515 West Tower, Toronto, ON, M5G 1M1, Canada
| | - Nardos G Tassew
- Vision Division, Krembil Research Institute, Krembil Discovery Tower, KDT-8-418, 60 Leonard Street, Toronto, ON, M5T 2S8, Canada
| | - Michael Wu
- Department of Physiology, University of Toronto, 1 King's College Circle, 3306 Medical Sciences Building, Toronto, ON, M5S 1A8, Canada
| | - Ya-Chi Huang
- Department of Physiology, University of Toronto, 1 King's College Circle, 3306 Medical Sciences Building, Toronto, ON, M5S 1A8, Canada
| | - Jacqueline N Crawley
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Philippe P Monnier
- Vision Division, Krembil Research Institute, Krembil Discovery Tower, KDT-8-418, 60 Leonard Street, Toronto, ON, M5T 2S8, Canada
| | - Alexander J A Groffen
- Department of Functional Genomics, CNCR, Neuroscience Campus Amsterdam, VU University and VU Medical Center, 1081 HV, Amsterdam, Netherlands
| | - Hong-Shuo Sun
- Department of Physiology, University of Toronto, 1 King's College Circle, 3306 Medical Sciences Building, Toronto, ON, M5S 1A8, Canada. .,Department of Surgery, University of Toronto, 1 King's College Circle, 1184 Medical Sciences Building, Toronto, ON, M5S 1A8, Canada.
| | - Lucy R Osborne
- Department of Medicine, University of Toronto, 661 University Avenue, MaRS Centre, 1515 West Tower, Toronto, ON, M5G 1M1, Canada. .,Department of Molecular Genetics, University of Toronto, 661 University Avenue, MaRS Centre, 1515 West Tower, Toronto, ON, M5G 1M1, Canada.
| | - Zhong-Ping Feng
- Department of Physiology, University of Toronto, 1 King's College Circle, 3306 Medical Sciences Building, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
27
|
Wang Y, Han J, Chen X, Zeng X, Wang Y, Dong J, Chen J. Maternal iodine supplementation improves motor coordination in offspring by modulating the mGluR1 signaling pathway in mild iodine deficiency-induced hypothyroxinemia rats. J Nutr Biochem 2018; 58:80-89. [DOI: 10.1016/j.jnutbio.2018.04.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/17/2018] [Accepted: 04/22/2018] [Indexed: 02/02/2023]
|
28
|
Tian J, Zhu MX. GABA B Receptors Augment TRPC3-Mediated Slow Excitatory Postsynaptic Current to Regulate Cerebellar Purkinje Neuron Response to Type-1 Metabotropic Glutamate Receptor Activation. Cells 2018; 7:cells7080090. [PMID: 30060610 PMCID: PMC6116156 DOI: 10.3390/cells7080090] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 07/18/2018] [Accepted: 07/27/2018] [Indexed: 02/02/2023] Open
Abstract
During strong parallel fiber stimulation, glutamate released at parallel fiber-Purkinje cell synapses activates type-1 metabotropic glutamate receptor (mGluR1) to trigger a slow excitatory postsynaptic current (sEPSC) in cerebellar Purkinje neurons. The sEPSC is mediated by transient receptor potential canonical 3 (TRPC3) channels. Often co-localized with mGluR1 in Purkinje neuron dendrites are type B γ-aminobutyric acid receptors (GABABRs) that respond to inhibitory synaptic inputs from interneurons located in the molecular layer of cerebellar cortex. It has been shown that activation of postsynaptic GABABRs potentiates mGluR1 activation-evoked sEPSC in Purkinje cells, but the underlying molecular mechanism remains elusive. Here we report that the augmentation of mGluR1-sEPSC by GABABR activation in Purkinje neurons is completely absent in TRPC3 knockout mice, but totally intact in TRPC1-, TRPC4-, and TRPC1,4,5,6-knockout mice, suggesting that TRPC3 is the only TRPC isoform that mediates the potentiation. Moreover, our results indicate that the potentiation reflects a postsynaptic mechanism that requires both GABABRs and mGluR1 because it is unaffected by blocking neurotransmission with tetrodotoxin but blocked by inhibiting either GABABRs or mGluR1. Furthermore, we show that the co-stimulation of GABABRs has an effect on shaping the response of Purkinje cell firing to mGluR1-sEPSC, revealing a new function of inhibitory input on excitatory neurotransmission. We conclude that postsynaptic GABABRs regulate Purkinje cell responses to strong glutamatergic stimulation through modulation of mGluR1-TRPC3 coupling. Since mGluR1-TRPC3 coupling is essential in cerebellar long-term depression, synapse elimination, and motor coordination, our findings may have implications in essential cerebellar functions, such as motor coordination and learning.
Collapse
Affiliation(s)
- Jinbin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
29
|
Focal Ischaemic Infarcts Expand Faster in Cerebellar Cortex than Cerebral Cortex in a Mouse Photothrombotic Stroke Model. Transl Stroke Res 2018; 9:643-653. [PMID: 29455391 DOI: 10.1007/s12975-018-0615-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/02/2018] [Accepted: 02/04/2018] [Indexed: 12/31/2022]
Abstract
It is generally accepted that the cerebellum is particularly vulnerable to ischaemic injury, and this may contribute to the high mortality arising from posterior circulation strokes. However, this has not been systematically examined in an animal model. This study compared the development and resolution of matched photothrombotic microvascular infarcts in the cerebellar and cerebral cortices in adult 129/SvEv mice of both sexes. The photothrombotic lesions were made using tail vein injection of Rose Bengal with a 532 nm laser projected onto a 2 mm diameter aperture over the target region of the brain (with skull thinning). Infarct size was then imaged histologically following 2 h to 30-day survival using serial reconstruction of haematoxylin and eosin stained cryosections. This was complemented with immunohistochemistry for neuron and glial markers. At 2 h post-injury, the cerebellar infarct volume averaged ~ 2.7 times that of the cerebral cortex infarcts. Infarct volume reached maximum in the cerebellum in a quarter of the time (24 h) taken in the cerebral cortex (4 days). Remodelling resolved the infarcts within a month, leaving significantly larger residual injury volume in the cerebellum. The death of neurons in the core lesion at 2 h was confirmed by NeuN and Calbindin immunofluorescence, alongside activation of astrocytes and microglia. The latter persisted in the region within and surrounding the residual infarct at 30 days. This comparison of acute focal ischaemic injuries in cerebellar and cerebral cortices provides direct confirmation of exacerbation of neuropathology and faster kinetics in the cerebellum.
Collapse
|
30
|
Huang M, Verbeek DS. Why do so many genetic insults lead to Purkinje Cell degeneration and spinocerebellar ataxia? Neurosci Lett 2018; 688:49-57. [PMID: 29421540 DOI: 10.1016/j.neulet.2018.02.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/02/2018] [Indexed: 12/29/2022]
Abstract
The genetically heterozygous spinocerebellar ataxias are all characterized by cerebellar atrophy and pervasive Purkinje Cell degeneration. Up to date, more than 35 functionally diverse spinocerebellar ataxia genes have been identified. The main question that remains yet unsolved is why do some many genetic insults lead to Purkinje Cell degeneration and spinocerebellar ataxia? To address this question it is important to identify intrinsic pathways important for Purkinje Cell function and survival. In this review, we discuss the current consensus on shared mechanisms underlying the pervasive Purkinje Cell loss in spinocerebellar ataxia. Additionally, using recently published cell type specific expression data, we identified several Purkinje Cell-specific genes and discuss how the corresponding pathways might underlie the vulnerability of Purkinje Cells in response to the diverse genetic insults causing spinocerebellar ataxia.
Collapse
Affiliation(s)
- Miaozhen Huang
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Dineke S Verbeek
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands.
| |
Collapse
|
31
|
Scoles DR, Pulst SM. Spinocerebellar Ataxia Type 2. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1049:175-195. [PMID: 29427103 DOI: 10.1007/978-3-319-71779-1_8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Spinocerebellar ataxia type 2 (SCA2) is autosomal dominantly inherited and caused by CAG repeat expansion in the ATXN2 gene. Because the CAG repeat expansion is localized to an encoded region of ATXN2, the result is an expanded polyglutamine (polyQ) tract in the ATXN2 protein. SCA2 is characterized by progressive ataxia, and slow saccades. No treatment for SCA2 exists. ATXN2 mutation causes gains of new or toxic functions for the ATXN2 protein, resulting in abnormally slow Purkinje cell (PC) firing frequency and ultimately PC loss. This chapter describes the characteristics of SCA2 patients briefly, and reviews ATXN2 molecular features and progress toward the identification of a treatment for SCA2.
Collapse
Affiliation(s)
- Daniel R Scoles
- Department of Neurology, University of Utah, 175 North Medical Drive East, 5th Floor, Salt Lake City, UT, 84132, USA
| | - Stefan M Pulst
- Department of Neurology, University of Utah, 175 North Medical Drive East, 5th Floor, Salt Lake City, UT, 84132, USA.
| |
Collapse
|
32
|
Watson LM, Bamber E, Schnekenberg RP, Williams J, Bettencourt C, Lickiss J, Jayawant S, Fawcett K, Clokie S, Wallis Y, Clouston P, Sims D, Houlden H, Becker EB, Németh AH. Dominant Mutations in GRM1 Cause Spinocerebellar Ataxia Type 44. Am J Hum Genet 2017; 101:451-458. [PMID: 28886343 PMCID: PMC5591020 DOI: 10.1016/j.ajhg.2017.08.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 07/25/2017] [Indexed: 12/15/2022] Open
Abstract
The metabotropic glutamate receptor 1 (mGluR1) is abundantly expressed in the mammalian central nervous system, where it regulates intracellular calcium homeostasis in response to excitatory signaling. Here, we describe heterozygous dominant mutations in GRM1, which encodes mGluR1, that are associated with distinct disease phenotypes: gain-of-function missense mutations, linked in two different families to adult-onset cerebellar ataxia, and a de novo truncation mutation resulting in a dominant-negative effect that is associated with juvenile-onset ataxia and intellectual disability. Crucially, the gain-of-function mutations could be pharmacologically modulated in vitro using an existing FDA-approved drug, Nitazoxanide, suggesting a possible avenue for treatment, which is currently unavailable for ataxias.
Collapse
|
33
|
Meera P, Pulst S, Otis T. A positive feedback loop linking enhanced mGluR function and basal calcium in spinocerebellar ataxia type 2. eLife 2017; 6. [PMID: 28518055 PMCID: PMC5444899 DOI: 10.7554/elife.26377] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 05/16/2017] [Indexed: 12/11/2022] Open
Abstract
Metabotropic glutamate receptor 1 (mGluR1) function in Purkinje neurons (PNs) is essential for cerebellar development and for motor learning and altered mGluR1 signaling causes ataxia. Downstream of mGluR1, dysregulation of calcium homeostasis has been hypothesized as a key pathological event in genetic forms of ataxia but the underlying mechanisms remain unclear. We find in a spinocerebellar ataxia type 2 (SCA2) mouse model that calcium homeostasis in PNs is disturbed across a broad range of physiological conditions. At parallel fiber synapses, mGluR1-mediated excitatory postsynaptic currents (EPSCs) and associated calcium transients are increased and prolonged in SCA2 PNs. In SCA2 PNs, enhanced mGluR1 function is prevented by buffering [Ca2+] at normal resting levels while in wildtype PNs mGluR1 EPSCs are enhanced by elevated [Ca2+]. These findings demonstrate a deleterious positive feedback loop involving elevated intracellular calcium and enhanced mGluR1 function, a mechanism likely to contribute to PN dysfunction and loss in SCA2. DOI:http://dx.doi.org/10.7554/eLife.26377.001
Collapse
Affiliation(s)
- Pratap Meera
- Department of Neurobiology, Geffen School of Medicine, University of California, Los Angeles, United States
| | - Stefan Pulst
- Department of Neurology, University of Utah, Salt Lake, United States
| | - Thomas Otis
- Department of Neurobiology, Geffen School of Medicine, University of California, Los Angeles, United States.,Neuroscience, Ophthalmology, and Rare Diseases, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| |
Collapse
|
34
|
Shuvaev AN, Hosoi N, Sato Y, Yanagihara D, Hirai H. Progressive impairment of cerebellar mGluR signalling and its therapeutic potential for cerebellar ataxia in spinocerebellar ataxia type 1 model mice. J Physiol 2016; 595:141-164. [PMID: 27440721 DOI: 10.1113/jp272950] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 07/11/2016] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS Spinocerebellar ataxia type 1 (SCA1) is a progressive neurodegenerative disease caused by a gene defect, leading to movement disorder such as cerebellar ataxia. It remains largely unknown which functional defect contributes to the cerebellar ataxic phenotype in SCA1. In this study, we report progressive dysfunction of metabotropic glutamate receptor (mGluR) signalling, which leads to smaller slow synaptic responses, reduced dendritic Ca2+ signals and impaired synaptic plasticity at cerebellar synapses, in the early disease stage of SCA1 model mice. We also show that enhancement of mGluR signalling by a clinically available drug, baclofen, leads to improvement of motor performance in SCA1 mice. SCA1 is an incurable disease with no effective treatment, and our results may provide mechanistic grounds for targeting mGluRs and a novel drug therapy with baclofen to treat SCA1 patients in the future. ABSTRACT Spinocerebellar ataxia type 1 (SCA1) is a progressive neurodegenerative disease that presents with cerebellar ataxia and motor learning defects. Previous studies have indicated that the pathology of SCA1, as well as other ataxic diseases, is related to signalling pathways mediated by the metabotropic glutamate receptor type 1 (mGluR1), which is indispensable for proper motor coordination and learning. However, the functional contribution of mGluR signalling to SCA1 pathology is unclear. In the present study, we show that SCA1 model mice develop a functional impairment of mGluR signalling which mediates slow synaptic responses, dendritic Ca2+ signals, and short- and long-term synaptic plasticity at parallel fibre (PF)-Purkinje cell (PC) synapses in a progressive manner from the early disease stage (5 postnatal weeks) prior to PC death. Notably, impairment of mGluR-mediated dendritic Ca2+ signals linearly correlated with a reduction of PC capacitance (cell surface area) in disease progression. Enhancement of mGluR signalling by baclofen, a clinically available GABAB receptor agonist, led to an improvement of motor performance in SCA1 mice and the improvement lasted ∼1 week after a single application of baclofen. Moreover, the restoration of motor performance in baclofen-treated SCA1 mice matched the functional recovery of mGluR-mediated slow synaptic currents and mGluR-dependent short- and long-term synaptic plasticity. These results suggest that impairment of synaptic mGluR cascades is one of the important contributing factors to cerebellar ataxia in early and middle stages of SCA1 pathology, and that modulation of mGluR signalling by baclofen or other clinical interventions may be therapeutic targets to treat SCA1.
Collapse
Affiliation(s)
- Anton N Shuvaev
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan.,Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V. F. Voino-Yasenetsky, Krasnoyarsk, 660022, Russia
| | - Nobutake Hosoi
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan
| | - Yamato Sato
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Tokyo, 153-8902, Japan
| | - Dai Yanagihara
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Tokyo, 153-8902, Japan
| | - Hirokazu Hirai
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan.,Research Program for Neural Signalling, Division of Endocrinology, Metabolism and Signal Research, Gunma University Initiative for Advanced Research, Maebashi, Gunma, 371-8511, Japan
| |
Collapse
|
35
|
Abstract
Psychiatric and neurological disorders are mostly associated with the changes in neural calcium ion signaling pathways required for activity-triggered cellular events. One calcium channel family is the TRP cation channel family, which contains seven subfamilies. Results of recent papers have discovered that calcium ion influx through TRP channels is important. We discuss the latest advances in calcium ion influx through TRP channels in the etiology of psychiatric disorders. Activation of TRPC4, TRPC5, and TRPV1 cation channels in the etiology of psychiatric disorders such as anxiety, fear-associated responses, and depression modulate calcium ion influx. Evidence substantiates that anandamide and its analog (methanandamide) induce an anxiolytic-like effect via CB1 receptors and TRPV1 channels. Intracellular calcium influx induced by oxidative stress has an significant role in the etiology of bipolar disorders (BDs), and studies recently reported the important role of TRP channels such as TRPC3, TRPM2, and TRPV1 in converting oxidant or nitrogen radical signaling to cytosolic calcium ion homeostasis in BDs. The TRPV1 channel also plays a function in morphine tolerance and hyperalgesia. Among psychotropic drugs, amitriptyline and capsazepine seem to have protective effects on psychiatric disorders via the TRP channels. Some drugs such as cocaine and methamphetamine also seem to have an important role in alcohol addiction and substance abuse via activation of the TRPV1 channel. Thus, we explore the relationships between the etiology of psychiatric disorders and TRP channel-regulated mechanisms. Investigation of the TRP channels in psychiatric disorders holds the promise of the development of new drug treatments.
Collapse
Affiliation(s)
- Mustafa Nazıroğlu
- Neuroscience Research Center, Süleyman Demirel University, Dekanlık Binası, TR-32260, Isparta, Turkey.
| | | |
Collapse
|
36
|
Meera P, Pulst SM, Otis TS. Cellular and circuit mechanisms underlying spinocerebellar ataxias. J Physiol 2016; 594:4653-60. [PMID: 27198167 DOI: 10.1113/jp271897] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 03/13/2016] [Indexed: 12/12/2022] Open
Abstract
Degenerative ataxias are a common form of neurodegenerative disease that affect about 20 individuals per 100,000. The autosomal dominant spinocerebellar ataxias (SCAs) are caused by a variety of protein coding mutations (single nucleotide changes, deletions and expansions) in single genes. Affected genes encode plasma membrane and intracellular ion channels, membrane receptors, protein kinases, protein phosphatases and proteins of unknown function. Although SCA-linked genes are quite diverse they share two key features: first, they are highly, although not exclusively, expressed in cerebellar Purkinje neurons (PNs), and second, when mutated they lead ultimately to the degeneration of PNs. In this review we summarize ataxia-related changes in PN neurophysiology that have been observed in various mouse knockout lines and in transgenic models of human SCA. We also highlight emerging evidence that altered metabotropic glutamate receptor signalling and disrupted calcium homeostasis in PNs form a common, early pathophysiological mechanism in SCAs. Together these findings indicate that aberrant calcium signalling and profound changes in PN neurophysiology precede PN cell loss and are likely to lead to cerebellar circuit dysfunction that explains behavioural signs of ataxia characteristic of the disease.
Collapse
Affiliation(s)
- Pratap Meera
- Department of Neurobiology, Geffen School of Medicine, University of California, 650 Charles Young Drive, Los Angeles, CA, 90095, USA
| | - Stefan M Pulst
- Department of Neurology, University of Utah, 175 N Medical Drive E, Salt Lake City, UT, 84132, USA
| | - Thomas S Otis
- Department of Neurobiology, Geffen School of Medicine, University of California, 650 Charles Young Drive, Los Angeles, CA, 90095, USA.,Roche Pharmaceutical Research and Early Development (pRED), Neuroscience, Ophthalmology and Rare Diseases (NORD), Grenzacherstrasse 124, CH-4070, Basel, Switzerland
| |
Collapse
|
37
|
De-Miguel FF, Nicholls JG. Release of chemical transmitters from cell bodies and dendrites of nerve cells. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0181. [PMID: 26009760 DOI: 10.1098/rstb.2014.0181] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Papers in this issue concern extrasynaptic transmission, namely release of signalling molecules by exocytosis or diffusion from neuronal cell bodies, dendrites, axons and glia. Problems discussed concern the molecules, their secretion and importance for normal function and disease. Molecules secreted extrasynaptically include transmitters, peptides, hormones and nitric oxide. For extrasynaptic secretion, trains of action potentials are required, and the time course of release is slower than at synapses. Questions arise concerning the mechanism of extrasynaptic secretion: how does it differ from the release observed at synaptic terminals and gland cells? What kinds of vesicles take part? Is release accomplished through calcium entry, SNAP and SNARE proteins? A clear difference is in the role of molecules released synaptically and extrasynaptically. After extrasynaptic release, molecules reach distant as well as nearby cells, and thereby produce long-lasting changes over large volumes of brain. Such changes can affect circuits for motor performance and mood states. An example with clinical relevance is dyskinesia of patients treated with l-DOPA for Parkinson's disease. Extrasynaptically released transmitters also evoke responses in glial cells, which in turn release molecules that cause local vasodilatation and enhanced circulation in regions of the brain that are active.
Collapse
Affiliation(s)
- Francisco F De-Miguel
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Distrito Federal, Mexico
| | - John G Nicholls
- Scuola Internazionale Superiore di Studi Avanzati, SISSA, Trieste, Italy
| |
Collapse
|
38
|
Hartmann J, Konnerth A. TRPC3‐dependent synaptic transmission in central mammalian neurons. J Mol Med (Berl) 2015; 93:983-9. [DOI: 10.1007/s00109-015-1298-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 05/05/2015] [Accepted: 05/13/2015] [Indexed: 01/05/2023]
|
39
|
Becker EBE. The Moonwalker mouse: new insights into TRPC3 function, cerebellar development, and ataxia. THE CEREBELLUM 2015; 13:628-36. [PMID: 24797279 PMCID: PMC4155175 DOI: 10.1007/s12311-014-0564-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The Moonwalker (Mwk) mouse is a recent model of dominantly inherited cerebellar ataxia. The motor phenotype of the Mwk mouse is due to a gain-of-function mutation in the gene encoding the cation-permeable transient receptor potential channel (TRPC3). This mutation converts a threonine into an alanine in the highly conserved cytoplasmic S4–S5 linker of the channel, affecting channel gating. TRPC3 is highly expressed in cerebellar Purkinje cells and type II unipolar brush cells that both degenerate in the Mwk mouse. Studies of the Mwk mouse have provided new insights into the role of TRPC3 in cerebellar development and disease, which could not have been predicted from the Trpc3 knockout phenotype. Here, the genetic, behavioral, histological, and functional characterization of the Mwk mouse is reviewed. Moreover, the relationship of the Mwk mutant to other cerebellar mouse models and its relevance as a model for cerebellar ataxia are discussed.
Collapse
Affiliation(s)
- Esther B E Becker
- MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3PT, UK,
| |
Collapse
|
40
|
Zhang E, Liao P. Brain transient receptor potential channels and stroke. J Neurosci Res 2014; 93:1165-83. [PMID: 25502473 DOI: 10.1002/jnr.23529] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 09/10/2014] [Accepted: 11/04/2014] [Indexed: 02/06/2023]
Abstract
Transient receptor potential (TRP) channels have been increasingly implicated in the pathological mechanisms of CNS disorders. TRP expression has been detected in neurons, astrocytes, oligodendrocytes, microglia, and ependymal cells as well as in the cerebral vascular endothelium and smooth muscle. In stroke, TRPC3/4/6, TRPM2/4/7, and TRPV1/3/4 channels have been found to participate in ischemia-induced cell death, whereas other TRP channels, in particular those expressed in nonneuronal cells, have been less well studied. This review summarizes the current knowledge on the expression and functions of the TRP channels in various cell types in the brain and our current understanding of TRP channels in stroke pathophysiology. In an aging society, the occurrence of stroke is expected to increase steadily, and there is an urgent requirement to improve the current stroke management strategy. Therefore, elucidating the roles of TRP channels in stroke could shed light on the development of novel therapeutic strategies and ultimately improve stroke outcome.
Collapse
Affiliation(s)
- Eric Zhang
- Calcium Signalling Laboratory, National Neuroscience Institute, Singapore
| | - Ping Liao
- Calcium Signalling Laboratory, National Neuroscience Institute, Singapore.,Duke-NUS Graduate Medical School Singapore, Singapore
| |
Collapse
|
41
|
Nilius B, Szallasi A. Transient Receptor Potential Channels as Drug Targets: From the Science of Basic Research to the Art of Medicine. Pharmacol Rev 2014; 66:676-814. [DOI: 10.1124/pr.113.008268] [Citation(s) in RCA: 348] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
42
|
Clark KB. Basis for a neuronal version of Grover's quantum algorithm. Front Mol Neurosci 2014; 7:29. [PMID: 24860419 PMCID: PMC4029008 DOI: 10.3389/fnmol.2014.00029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/31/2014] [Indexed: 11/25/2022] Open
Abstract
Grover's quantum (search) algorithm exploits principles of quantum information theory and computation to surpass the strong Church–Turing limit governing classical computers. The algorithm initializes a search field into superposed N (eigen)states to later execute nonclassical “subroutines” involving unitary phase shifts of measured states and to produce root-rate or quadratic gain in the algorithmic time (O(N1/2)) needed to find some “target” solution m. Akin to this fast technological search algorithm, single eukaryotic cells, such as differentiated neurons, perform natural quadratic speed-up in the search for appropriate store-operated Ca2+ response regulation of, among other processes, protein and lipid biosynthesis, cell energetics, stress responses, cell fate and death, synaptic plasticity, and immunoprotection. Such speed-up in cellular decision making results from spatiotemporal dynamics of networked intracellular Ca2+-induced Ca2+ release and the search (or signaling) velocity of Ca2+ wave propagation. As chemical processes, such as the duration of Ca2+ mobilization, become rate-limiting over interstore distances, Ca2+ waves quadratically decrease interstore-travel time from slow saltatory to fast continuous gradients proportional to the square-root of the classical Ca2+ diffusion coefficient, D1/2, matching the computing efficiency of Grover's quantum algorithm. In this Hypothesis and Theory article, I elaborate on these traits using a fire-diffuse-fire model of store-operated cytosolic Ca2+ signaling valid for glutamatergic neurons. Salient model features corresponding to Grover's quantum algorithm are parameterized to meet requirements for the Oracle Hadamard transform and Grover's iteration. A neuronal version of Grover's quantum algorithm figures to benefit signal coincidence detection and integration, bidirectional synaptic plasticity, and other vital cell functions by rapidly selecting, ordering, and/or counting optional response regulation choices.
Collapse
Affiliation(s)
- Kevin B Clark
- Research and Development Service, Veterans Affairs Greater Los Angeles Healthcare System Los Angeles, CA, USA ; Complex Biological Systems Alliance North Andover, MA, USA
| |
Collapse
|
43
|
Ady V, Perroy J, Tricoire L, Piochon C, Dadak S, Chen X, Dusart I, Fagni L, Lambolez B, Levenes C. Type 1 metabotropic glutamate receptors (mGlu1) trigger the gating of GluD2 delta glutamate receptors. EMBO Rep 2013; 15:103-9. [PMID: 24357660 DOI: 10.1002/embr.201337371] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The orphan GluD2 receptor belongs to the ionotropic glutamate receptor family but does not bind glutamate. Ligand-gated GluD2 currents have never been evidenced, and whether GluD2 operates as an ion channel has been a long-standing question. Here, we show that GluD2 gating is triggered by type 1 metabotropic glutamate receptors, both in a heterologous expression system and in Purkinje cells. Thus, GluD2 is not only an adhesion molecule at synapses but also works as a channel. This gating mechanism reveals new properties of glutamate receptors that emerge from their interaction and opens unexpected perspectives regarding synaptic transmission and plasticity.
Collapse
Affiliation(s)
- Visou Ady
- Centre National de la Recherche Scientifique (CNRS) UMR 8119 Neurophysics and Physiology Laboratory, Université Paris Descartes, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
This minireview focuses on recent studies implicating class V myosins in organelle and macromolecule transport within neurons. These studies reveal that class V myosins play important roles in a wide range of fundamental processes occurring within neurons, including the transport into dendritic spines of organelles that support synaptic plasticity, the establishment of neuronal shape, the specification of polarized cargo transport, and the subcellular localization of mRNA.
Collapse
Affiliation(s)
- John A Hammer
- From the Cell Biology and Physiology Center, NHLBI, National Institutes of Health, Bethesda, Maryland 20892 and
| | | |
Collapse
|
45
|
NMDA receptor-dependent synaptic activation of TRPC channels in olfactory bulb granule cells. J Neurosci 2012; 32:5737-46. [PMID: 22539836 DOI: 10.1523/jneurosci.3753-11.2012] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Canonical transient receptor potential (TRPC) channels are widely expressed throughout the nervous system including the olfactory bulb where their function is largely unknown. Here, we describe their contribution to central synaptic processing at the reciprocal mitral and tufted cell-granule cell microcircuit, the most abundant synapse of the mammalian olfactory bulb. Suprathreshold activation of the synapse causes sodium action potentials in mouse granule cells and a subsequent long-lasting depolarization (LLD) linked to a global dendritic postsynaptic calcium signal recorded with two-photon laser-scanning microscopy. These signals are not observed after action potentials evoked by current injection in the same cells. The LLD persists in the presence of group I metabotropic glutamate receptor antagonists but is entirely absent from granule cells deficient for the NMDA receptor subunit NR1. Moreover, both depolarization and Ca²⁺ rise are sensitive to the blockade of NMDA receptors. The LLD and the accompanying Ca²⁺ rise are also absent in granule cells from mice deficient for both TRPC channel subtypes 1 and 4, whereas the deletion of either TRPC1 or TRPC4 results in only a partial reduction of the LLD. Recordings from mitral cells in the absence of both subunits reveal a reduction of asynchronous neurotransmitter release from the granule cells during recurrent inhibition. We conclude that TRPC1 and TRPC4 can be activated downstream of NMDA receptor activation and contribute to slow synaptic transmission in the olfactory bulb, including the calcium dynamics required for asynchronous release from the granule cell spine.
Collapse
|
46
|
Abstract
The Transient receptor potential (TRP) family of cation channels is a large protein family, which is mainly structurally uniform. Proteins consist typically of six transmembrane domains and mostly four subunits are necessary to form a functional channel. Apart from this, TRP channels display a wide variety of activation mechanisms (ligand binding, G-protein coupled receptor dependent, physical stimuli such as temperature, pressure, etc.) and ion selectivity profiles (from highly Ca(2+) selective to non-selective for cations). They have been described now in almost every tissue of the body, including peripheral and central neurons. Especially in the sensory nervous system the role of several TRP channels is already described on a detailed level. This review summarizes data that is currently available on their role in the central nervous system. TRP channels are involved in neurogenesis and brain development, synaptic transmission and they play a key role in the development of several neurological diseases.
Collapse
|