1
|
Alkadhi KA. Giant miniature endplate potentials at vertebrate neuromuscular junctions: A review. Eur J Neurosci 2024; 60:7183-7194. [PMID: 39600045 DOI: 10.1111/ejn.16624] [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: 07/10/2024] [Revised: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
An unusually large amplitude spontaneous miniature endplate potentials (gMEPPs) occur naturally at low frequency at the vertebrate neuromuscular junction. Unlike the normal miniature endplate potentials (nMEPPs), these gMEPPs have long duration and long time to peak. More strikingly, gMEPPs seem to be independent of extracellular and intracellular Ca+2. and have a greater temperature sensitivity than nMEPPs. They are potentiated by tetrodotoxin but inhibited by acetylcholine (ACh) receptor blockers indicating ACh is the neurotransmitter responsible for gMEPPs. The frequency of gMEPPs is greatly increased in muscles weakened by various drugs, toxins or disease conditions suggesting that gMEPPs may be a part of possible neurotrophic mechanism to preserve effective neuromuscular transmission when the normal function is compromised.
Collapse
Affiliation(s)
- Karim A Alkadhi
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| |
Collapse
|
2
|
Grønbæk-Thygesen M, Hartmann-Petersen R. Cellular and molecular mechanisms of aspartoacylase and its role in Canavan disease. Cell Biosci 2024; 14:45. [PMID: 38582917 PMCID: PMC10998430 DOI: 10.1186/s13578-024-01224-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/24/2024] [Indexed: 04/08/2024] Open
Abstract
Canavan disease is an autosomal recessive and lethal neurological disorder, characterized by the spongy degeneration of the white matter in the brain. The disease is caused by a deficiency of the cytosolic aspartoacylase (ASPA) enzyme, which catalyzes the hydrolysis of N-acetyl-aspartate (NAA), an abundant brain metabolite, into aspartate and acetate. On the physiological level, the mechanism of pathogenicity remains somewhat obscure, with multiple, not mutually exclusive, suggested hypotheses. At the molecular level, recent studies have shown that most disease linked ASPA gene variants lead to a structural destabilization and subsequent proteasomal degradation of the ASPA protein variants, and accordingly Canavan disease should in general be considered a protein misfolding disorder. Here, we comprehensively summarize the molecular and cell biology of ASPA, with a particular focus on disease-linked gene variants and the pathophysiology of Canavan disease. We highlight the importance of high-throughput technologies and computational prediction tools for making genotype-phenotype predictions as we await the results of ongoing trials with gene therapy for Canavan disease.
Collapse
Affiliation(s)
- Martin Grønbæk-Thygesen
- The Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, 2200N, Copenhagen, Denmark.
| | - Rasmus Hartmann-Petersen
- The Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, 2200N, Copenhagen, Denmark.
| |
Collapse
|
3
|
Becker I, Wang-Eckhardt L, Eckhardt M. NAAG synthetase deficiency has only low influence on pathogenesis in a Canavan disease mouse model. J Inherit Metab Dis 2024; 47:230-243. [PMID: 38011891 DOI: 10.1002/jimd.12693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023]
Abstract
Canavan disease (CD) is a leukodystrophy caused by mutations in the N-acetylaspartate (NAA) hydrolase aspartoacylase (ASPA). Inability to degrade NAA and its accumulation in the brain results in spongiform myelin degeneration. NAA is mainly synthesized by neurons, where it is also a precursor of the neuropeptide N-acetylaspartylglutamate (NAAG). Hydrolysis of this peptide by glutamate carboxypeptidases is an additional source of extracellular NAA besides the instant neuronal release of NAA. This study examines to what extent NAA released from NAAG contributes to NAA accumulation and pathogenesis in the brain of Aspanur7/nur7 mutant mice, an established model of CD. Towards this aim, Aspanur7/nur7 mice with additional deficiencies in NAAG synthetase genes Rimklb and/or Rimkla were generated. Loss of myelin in Aspanur7/nur7 mice was not significantly affected by Rimkla and Rimklb deficiency and there was also no obvious change in the extent of brain vacuolation. Astrogliosis was slightly reduced in the forebrain of Rimkla and Rimklb double deficient Aspanur7/nur7 mice. However, only minor improvements at the behavioral level were found. The brain NAA accumulation in CD mice was, however, not significantly reduced in the absence of NAAG synthesis. In summary, there was only a weak tendency towards reduced pathogenic symptoms in Aspanur7/nur7 mice deficient in NAAG synthesis. Therefore, we conclude that NAAG metabolism has little influence on NAA accumulation in Aspanur7/nur7 mice and development of pathological symptoms in CD.
Collapse
Affiliation(s)
- Ivonne Becker
- Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Lihua Wang-Eckhardt
- Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Matthias Eckhardt
- Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
4
|
Petrov AM. Oxysterols in Central and Peripheral Synaptic Communication. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:91-123. [PMID: 38036877 DOI: 10.1007/978-3-031-43883-7_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Cholesterol is a key molecule for synaptic transmission, and both central and peripheral synapses are cholesterol rich. During intense neuronal activity, a substantial portion of synaptic cholesterol can be oxidized by either enzymatic or non-enzymatic pathways to form oxysterols, which in turn modulate the activities of neurotransmitter receptors (e.g., NMDA and adrenergic receptors), signaling molecules (nitric oxide synthases, protein kinase C, liver X receptors), and synaptic vesicle cycling involved in neurotransmitters release. 24-Hydroxycholesterol, produced by neurons in the brain, could directly affect neighboring synapses and change neurotransmission. 27-Hydroxycholesterol, which can cross the blood-brain barrier, can alter both synaptogenesis and synaptic plasticity. Increased generation of 25-hydroxycholesterol by activated microglia and macrophages could link inflammatory processes to learning and neuronal regulation. Amyloids and oxidative stress can lead to an increase in the levels of ring-oxidized sterols and some of these oxysterols (4-cholesten-3-one, 5α-cholestan-3-one, 7β-hydroxycholesterol, 7-ketocholesterol) have a high potency to disturb or modulate neurotransmission at both the presynaptic and postsynaptic levels. Overall, oxysterols could be used as "molecular prototypes" for therapeutic approaches. Analogs of 24-hydroxycholesterol (SGE-301, SGE-550, SAGE718) can be used for correction of NMDA receptor hypofunction-related states, whereas inhibitors of cholesterol 24-hydroxylase, cholestane-3β,5α,6β-triol, and cholest-4-en-3-one oxime (olesoxime) can be utilized as potential anti-epileptic drugs and (or) protectors from excitotoxicity.
Collapse
Affiliation(s)
- Alexey M Petrov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", Kazan, RT, Russia.
- Kazan State Medial University, Kazan, RT, Russia.
- Kazan Federal University, Kazan, RT, Russia.
| |
Collapse
|
5
|
Personius KE, Siebert D, Koch DW, Udin SB. Blockage of neuromuscular glutamate receptors impairs reinnervation following nerve crush in adult mice. Front Cell Neurosci 2022; 16:1000218. [PMID: 36212695 PMCID: PMC9535682 DOI: 10.3389/fncel.2022.1000218] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
Motor axons in peripheral nerves are capable of regeneration following injury. However, complete recovery of motor function is rare, particularly when reinnervation is delayed. We have previously found that glutamate receptors play a crucial role in the successful innervation of muscle during mouse development. In particular, blocking N-methyl-D-aspartate (NMDA) receptor activity delays the normal elimination of excess innervation of each neuromuscular junction. Here, we use behavioral, immunohistochemical, electrophysiological, and calcium imaging methods to test whether glutamate receptors play a similar role in the transition from polyneuronal to mono-innervation and in recovery of function following peripheral nerve injury in mature muscle.
Collapse
Affiliation(s)
- Kirkwood E. Personius
- Program in Neuroscience, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
- Department of Rehabilitation Science, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
- *Correspondence: Kirkwood E. Personius,
| | - Danielle Siebert
- Program in Neuroscience, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Dennis W. Koch
- Department of Kinesiology, Canisius College, Buffalo, NY, United States
| | - Susan B. Udin
- Program in Neuroscience, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, Buffalo, NY, United States
| |
Collapse
|
6
|
Morland C, Nordengen K. N-Acetyl-Aspartyl-Glutamate in Brain Health and Disease. Int J Mol Sci 2022; 23:ijms23031268. [PMID: 35163193 PMCID: PMC8836185 DOI: 10.3390/ijms23031268] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 02/04/2023] Open
Abstract
N-acetyl-aspartyl-glutamate (NAAG) is the most abundant dipeptide in the brain, where it acts as a neuromodulator of glutamatergic synapses by activating presynaptic metabotropic glutamate receptor 3 (mGluR3). Recent data suggest that NAAG is selectively localized to postsynaptic dendrites in glutamatergic synapses and that it works as a retrograde neurotransmitter. NAAG is released in response to glutamate and provides the postsynaptic neuron with a feedback mechanisms to inhibit excessive glutamate signaling. A key regulator of synaptically available NAAG is rapid degradation by the extracellular enzyme glutamate carboxypeptidase II (GCPII). Increasing endogenous NAAG—for instance by inhibiting GCPII—is a promising treatment option for many brain disorders where glutamatergic excitotoxicity plays a role. The main effect of NAAG occurs through increased mGluR3 activation and thereby reduced glutamate release. In the present review, we summarize the transmitter role of NAAG and discuss the involvement of NAAG in normal brain physiology. We further present the suggested roles of NAAG in various neurological and psychiatric diseases and discuss the therapeutic potential of strategies aiming to enhance NAAG levels.
Collapse
Affiliation(s)
- Cecilie Morland
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, The Faculty of Mathematics and Natural Sciences, University of Oslo, 1068 Oslo, Norway
- Correspondence: (C.M.); (K.N.); Tel.: +47-22844937; (C.M.); +47-23073580 (K.N.)
| | - Kaja Nordengen
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318 Oslo, Norway
- Correspondence: (C.M.); (K.N.); Tel.: +47-22844937; (C.M.); +47-23073580 (K.N.)
| |
Collapse
|
7
|
Yang S, Datta D, Elizabeth Woo, Duque A, Morozov YM, Arellano J, Slusher BS, Wang M, Arnsten AFT. Inhibition of glutamate-carboxypeptidase-II in dorsolateral prefrontal cortex: potential therapeutic target for neuroinflammatory cognitive disorders. Mol Psychiatry 2022; 27:4252-4263. [PMID: 35732693 PMCID: PMC9718677 DOI: 10.1038/s41380-022-01656-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 05/27/2022] [Indexed: 02/07/2023]
Abstract
Glutamate carboxypeptidase-II (GCPII) expression in brain is increased by inflammation, e.g. by COVID19 infection, where it reduces NAAG stimulation of metabotropic glutamate receptor type 3 (mGluR3). GCPII-mGluR3 signaling is increasingly linked to higher cognition, as genetic alterations that weaken mGluR3 or increase GCPII signaling are associated with impaired cognition in humans. Recent evidence from macaque dorsolateral prefrontal cortex (dlPFC) shows that mGluR3 are expressed on dendritic spines, where they regulate cAMP-PKA opening of potassium (K+) channels to enhance neuronal firing during working memory. However, little is known about GCPII expression and function in the primate dlPFC, despite its relevance to inflammatory disorders. The present study used multiple label immunofluorescence and immunoelectron microscopy to localize GCPII in aging macaque dlPFC, and examined the effects of GCPII inhibition on dlPFC neuronal physiology and working memory function. GCPII was observed in astrocytes as expected, but also on neurons, including extensive expression in dendritic spines. Recordings in dlPFC from aged monkeys performing a working memory task found that iontophoresis of the GCPII inhibitors 2-MPPA or 2-PMPA markedly increased working memory-related neuronal firing and spatial tuning, enhancing neural representations. These beneficial effects were reversed by an mGluR2/3 antagonist, or by a cAMP-PKA activator, consistent with mGluR3 inhibition of cAMP-PKA-K+ channel signaling. Systemic administration of the brain penetrant inhibitor, 2-MPPA, significantly improved working memory performance without apparent side effects, with largest effects in the oldest monkeys. Taken together, these data endorse GCPII inhibition as a potential strategy for treating cognitive disorders associated with aging and/or neuroinflammation.
Collapse
Affiliation(s)
- Shengtao Yang
- grid.47100.320000000419368710Department Neuroscience, Yale University School of Medicine, New Haven, CT USA
| | - Dibyadeep Datta
- grid.47100.320000000419368710Department Neuroscience, Yale University School of Medicine, New Haven, CT USA ,grid.47100.320000000419368710Department Psychiatry, Yale University School of Medicine, New Haven, CT USA
| | - Elizabeth Woo
- grid.47100.320000000419368710Department Neuroscience, Yale University School of Medicine, New Haven, CT USA
| | - Alvaro Duque
- grid.47100.320000000419368710Department Neuroscience, Yale University School of Medicine, New Haven, CT USA
| | - Yury M. Morozov
- grid.47100.320000000419368710Department Neuroscience, Yale University School of Medicine, New Haven, CT USA
| | - Jon Arellano
- grid.47100.320000000419368710Department Neuroscience, Yale University School of Medicine, New Haven, CT USA
| | - Barbara S. Slusher
- grid.21107.350000 0001 2171 9311Department Neurology and Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD USA
| | - Min Wang
- grid.47100.320000000419368710Department Neuroscience, Yale University School of Medicine, New Haven, CT USA
| | - Amy F. T. Arnsten
- grid.47100.320000000419368710Department Neuroscience, Yale University School of Medicine, New Haven, CT USA
| |
Collapse
|
8
|
Becker I, Wang-Eckhardt L, Lodder-Gadaczek J, Wang Y, Grünewald A, Eckhardt M. Mice deficient in the NAAG synthetase II gene Rimkla are impaired in a novel object recognition task. J Neurochem 2021; 157:2008-2023. [PMID: 33638175 DOI: 10.1111/jnc.15333] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/11/2021] [Accepted: 02/21/2021] [Indexed: 12/27/2022]
Abstract
N-acetylaspartylglutamate (NAAG) is an abundant neuropeptide in the mammalian nervous system, synthesized by two related NAAG synthetases I and II (NAAGS-I and -II) encoded by the genes Rimklb and Rimkla, respectively. NAAG plays a role in cognition and memory, according to studies using inhibitors of the NAAG hydrolase glutamate carboxypeptidase II that increase NAAG concentration. To examine consequences of reduced NAAG concentration, Rimkla-deficient (Rimkla-/- ) mice were generated. These mice exhibit normal NAAG level at birth, likely because of the intact Rimklb gene, but have significantly reduced NAAG levels in all brain regions in adulthood. In wild type mice NAAGS-II was most abundant in brainstem and spinal cord, as demonstrated using a new NAAGS-II antiserum. In the hippocampus, NAAGS-II was only detectable in neurons expressing parvalbumin, a marker of GABAergic interneurons. Apart from reduced open field activity, general behavior of adult (6 months old) Rimkla-/- mice examined in different tests (dark-light transition, optokinetic behavior, rotarod, and alternating T-maze) was not significantly altered. However, Rimkla-/- mice were impaired in a short-term novel object recognition test. This was also the case for mice lacking NAA synthase Nat8l, which are devoid of NAAG. Together with results from previous studies showing that inhibition of the NAAG degrading enzyme glutamate carboxypeptidase II is associated with a significant improvement in object recognition, these results suggest a direct involvement of NAAG synthesized by NAAGS-II in the memory consolidation underlying the novel object recognition task.
Collapse
Affiliation(s)
- Ivonne Becker
- Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Lihua Wang-Eckhardt
- Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Julia Lodder-Gadaczek
- Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Yong Wang
- Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Agathe Grünewald
- Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Matthias Eckhardt
- Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
9
|
Rainey AN, Fukui SM, Mark K, King HM, Blitz DM. Intrinsic sources of tachykinin-related peptide in the thoracic ganglion mass of the crab, Cancer borealis. Gen Comp Endocrinol 2021; 302:113688. [PMID: 33275935 DOI: 10.1016/j.ygcen.2020.113688] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 11/30/2022]
Abstract
Neuropeptides comprise the largest class of neural and neuroendocrine signaling molecules. Vertebrate tachykinins (TKs) and the structurally-related invertebrate tachykinin-related peptides (TRPs) together form the largest neuropeptide superfamily, with a number of conserved neural and neuroendocrine functions across species. Arthropods, including crustaceans, have provided many insights into neuropeptide signaling and function. Crustacean tachykinin-related peptide occurs in endocrine organs and cells and in two of the major crustacean CNS components, the supraoesophageal ganglion ("brain") and the stomatogastric nervous system. However, little is known about TRP sources in the remaining major CNS component, the thoracic ganglion mass (TGM). To gain further insight into the function of this peptide, we aimed to identify intrinsic TRP sources in the TGM of the Jonah crab, Cancer borealis. We first adapted a clearing protocol to improve TRP immunoreactivity specifically in the TGM, which is a dense, fused mass of multiple ganglia in short-bodied crustaceans such as Cancer species of crabs. We verified that the clearing protocol avoided distortion of cell body morphology yet increased visibility of TRP immunoreactivity. Using confocal microscopy, we found TRP-immunoreactive (TRP-IR) axon tracts running the length of the TGM, TRP-IR neuropil in all ganglia, and approximately 110 TRP-IR somata distributed throughout the TGM, within and between ganglia. These somata likely represent both neural and neuroendocrine sources of TRP. Thus, there are many potential intrinsic sources of TRP in the TGM that are positioned to regulate behaviors such as food intake, locomotion, respiration, and reproduction.
Collapse
Affiliation(s)
- Amanda N Rainey
- Department of Biology and Center for Neuroscience, Miami University, Oxford, OH 45056, United States
| | - Stephanie M Fukui
- Department of Biology and Center for Neuroscience, Miami University, Oxford, OH 45056, United States
| | - Katie Mark
- Department of Biology and Center for Neuroscience, Miami University, Oxford, OH 45056, United States
| | - Hailey M King
- Department of Biology and Center for Neuroscience, Miami University, Oxford, OH 45056, United States
| | - Dawn M Blitz
- Department of Biology and Center for Neuroscience, Miami University, Oxford, OH 45056, United States.
| |
Collapse
|
10
|
Kozik EM, Marzluff EM, Lindgren CA. Evidence of NAAG-family tripeptide NAAG 2 in the Drosophila nervous system. J Neurochem 2020; 156:38-47. [PMID: 32885844 DOI: 10.1111/jnc.15173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 01/02/2023]
Abstract
N-acetylaspartylglutamate (NAAG) is a common neurotransmitter in the mammalian nervous system; however, it has never been reported in the nervous system of the fruit fly, Drosophila melanogaster. Using antiserum against NAAG, we localized NAAG-like immunoreactivity to neurons in the ventral nerve cord and to type Is glutamatergic nerve terminals in larval neuromuscular junctions. Using liquid chromatography tandem mass spectrometry (LC-MS), we failed to find NAAG but found the related peptide N-acetylaspartylglutamylglutamate (NAAG2 ) in Drosophila CNS and body wall tissue. This is the first report of any NAAG-family peptide in the nervous system of Drosophila and is also the first report of NAAG2 being present in a much higher concentration than NAAG in the nervous system of any species. Thus, the larval fruit fly presents an interesting model for the study of the functional role of NAAG2 of which very little is known-especially in the absence of an abundance of NAAG.
Collapse
Affiliation(s)
- Emily M Kozik
- Biology Department, Grinnell College, Grinnell, IA, USA
| | | | | |
Collapse
|
11
|
Hammond-Weinberger DR, Wang Y, Glavis-Bloom A, Spitzer NC. Mechanism for neurotransmitter-receptor matching. Proc Natl Acad Sci U S A 2020; 117:4368-4374. [PMID: 32041885 PMCID: PMC7049162 DOI: 10.1073/pnas.1916600117] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Synaptic communication requires the expression of functional postsynaptic receptors that match the presynaptically released neurotransmitter. The ability of neurons to switch the transmitter they release is increasingly well documented, and these switches require changes in the postsynaptic receptor population. Although the activity-dependent molecular mechanism of neurotransmitter switching is increasingly well understood, the basis of specification of postsynaptic neurotransmitter receptors matching the newly expressed transmitter is unknown. Using a functional assay, we show that sustained application of glutamate to embryonic vertebrate skeletal muscle cells cultured before innervation is necessary and sufficient to up-regulate ionotropic glutamate receptors from a pool of different receptors expressed at low levels. Up-regulation of these ionotropic receptors is independent of signaling by metabotropic glutamate receptors. Both imaging of glutamate-induced calcium elevations and Western blots reveal ionotropic glutamate receptor expression prior to immunocytochemical detection. Sustained application of glutamate to skeletal myotomes in vivo is necessary and sufficient for up-regulation of membrane expression of the GluN1 NMDA receptor subunit. Pharmacological antagonists and morpholinos implicate p38 and Jun kinases and MEF2C in the signal cascade leading to ionotropic glutamate receptor expression. The results suggest a mechanism by which neuronal release of transmitter up-regulates postsynaptic expression of appropriate transmitter receptors following neurotransmitter switching and may contribute to the proper expression of receptors at the time of initial innervation.
Collapse
Affiliation(s)
- Dena R Hammond-Weinberger
- Neurobiology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093-0357;
| | - Yunxin Wang
- Neurobiology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093-0357
| | - Alex Glavis-Bloom
- Neurobiology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093-0357
| | - Nicholas C Spitzer
- Neurobiology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093-0357;
- Center for Neural Circuits and Behavior, Kavli Institute for Brain and Mind, University of California San Diego, La Jolla, CA 92161
| |
Collapse
|
12
|
Glutamate at the Vertebrate Neuromuscular Junction: From Modulation to Neurotransmission. Cells 2019; 8:cells8090996. [PMID: 31466388 PMCID: PMC6770210 DOI: 10.3390/cells8090996] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/21/2019] [Accepted: 08/27/2019] [Indexed: 12/23/2022] Open
Abstract
Although acetylcholine is the major neurotransmitter operating at the skeletal neuromuscular junction of many invertebrates and of vertebrates, glutamate participates in modulating cholinergic transmission and plastic changes in the last. Presynaptic terminals of neuromuscular junctions contain and release glutamate that contribute to the regulation of synaptic neurotransmission through its interaction with pre- and post-synaptic receptors activating downstream signaling pathways that tune synaptic efficacy and plasticity. During vertebrate development, the chemical nature of the neurotransmitter at the vertebrate neuromuscular junction can be experimentally shifted from acetylcholine to other mediators (including glutamate) through the modulation of calcium dynamics in motoneurons and, when the neurotransmitter changes, the muscle fiber expresses and assembles new receptors to match the nature of the new mediator. Finally, in adult rodents, by diverting descending spinal glutamatergic axons to a denervated muscle, a functional reinnervation can be achieved with the formation of new neuromuscular junctions that use glutamate as neurotransmitter and express ionotropic glutamate receptors and other markers of central glutamatergic synapses. Here, we summarize the past and recent experimental evidences in support of a role of glutamate as a mediator at the synapse between the motor nerve ending and the skeletal muscle fiber, focusing on the molecules and signaling pathways that are present and activated by glutamate at the vertebrate neuromuscular junction.
Collapse
|
13
|
Wang JS, Bojovic D, Chen Y, Lindgren CA. Homocysteine sensitizes the mouse neuromuscular junction to oxidative stress by nitric oxide. Neuroreport 2018; 29:1030-1035. [PMID: 29939872 PMCID: PMC6044473 DOI: 10.1097/wnr.0000000000001073] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Homocysteine (HCY), a redox-active metabolite of the methionine cycle, is of particular clinical interest because of its association with various neurodegenerative diseases including amyotrophic lateral sclerosis. It has been previously established that HCY exacerbates damage to motor neurons from reactive oxygen species (ROS) such as hydrogen peroxide. To assess the role of HCY at the mammalian neuromuscular junction, neurotransmission was monitored by electrophysiology at the mouse epitrochleoanconeus muscle. Preparations were preincubated in HCY before inducing ROS and recordings were taken before and after ROS treatment. In this study, HCY was observed to sensitize the neuromuscular junction to ROS-induced depression of spontaneous transmission frequency, an effect we found to be mediated by a N-methyl-D-aspartate receptor (NMDAR) and nitric oxide (NO). The NMDAR antagonist D, L-2-amino-5-phosphonopentanoic acid prevented the HCY-induced sensitization to oxidative stress. Disrupting NO activity with either the nitric oxide synthase I antagonist Nω-nitro-L-arginine methyl ester hydrochloride or the NO scavenger 2-(4-carboxyphenyl)-4,4,5,5-tetramethyl-imidazoline-1-oxyl-3-oxide potassium salt also prevented sensitization. Moreover, replacing HCY with the exogenous NO donor Diethylamine NONOate diethylammonium was sufficient to reconstitute the effects of HCY-induced sensitization to ROS. Interestingly, a novel secondary effect was observed where HCY itself depresses quantal content, an effect found to be mediated by NMDARs independently of nitric oxide and ROS. Collectively, these data present a novel model of two distinct pathways through which HCY alters neurotransmission at the neuromuscular junction. Characterizing HCY's mechanism of action is of particular clinical relevance as many treatments for amyotrophic lateral sclerosis are centered on mitigating HCY-induced pathologies.
Collapse
Affiliation(s)
- John S Wang
- Department of Biology, Grinnell College, Grinnell, Iowa, USA
| | | | | | | |
Collapse
|
14
|
Lanuza MA, Tomàs J, Garcia N, Cilleros-Mañé V, Just-Borràs L, Tomàs M. Axonal competition and synapse elimination during neuromuscular junction development. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2018.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
15
|
Tomàs J, Garcia N, Lanuza MA, Santafé MM, Tomàs M, Nadal L, Hurtado E, Simó-Ollé A, Cilleros-Mañé V, Just-Borràs L. Adenosine Receptors in Developing and Adult Mouse Neuromuscular Junctions and Functional Links With Other Metabotropic Receptor Pathways. Front Pharmacol 2018; 9:397. [PMID: 29740322 PMCID: PMC5928480 DOI: 10.3389/fphar.2018.00397] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 04/05/2018] [Indexed: 12/30/2022] Open
Abstract
In the last few years, we have studied the presence and involvement in synaptogenesis and mature transmitter release of the adenosine autoreceptors (AR) in the mammalian neuromuscular junction (NMJ). Here, we review and bring together the previously published data to emphasize the relevance of these receptors for developmental axonal competition, synaptic loss and mature NMJ functional modulation. However, in addition to AR, activity-dependent mediators originating from any of the three cells that make the synapse (nerve, muscle, and glial cells) cross the extracellular cleft to generate signals in target metabotropic receptors. Thus, the integrated interpretation of the complementary function of all these receptors is needed. We previously studied, in the NMJ, the links of AR with mAChR and the neurotrophin receptor TrkB in the control of synapse elimination and transmitter release. We conclude that AR cooperate with these receptors through synergistic and antagonistic effects in the developmental synapse elimination process. In the adult NMJ, this cooperation is manifested so as that the functional integrity of a given receptor group depends on the other receptors operating normally (i.e., the functional integrity of mAChR depends on AR operating normally). These observations underlie the relevance of AR in the NMJ function.
Collapse
Affiliation(s)
- Josep Tomàs
- *Correspondence: Josep Tomàs, Neus Garcia, Maria A. Lanuza,
| | - Neus Garcia
- *Correspondence: Josep Tomàs, Neus Garcia, Maria A. Lanuza,
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Neuromuscular NMDA Receptors Modulate Developmental Synapse Elimination. J Neurosci 2017; 36:8783-9. [PMID: 27559162 DOI: 10.1523/jneurosci.1181-16.2016] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/27/2016] [Indexed: 02/03/2023] Open
Abstract
UNLABELLED At birth, each mammalian skeletal muscle fiber is innervated by multiple motor neurons, but in a few weeks, all but one of those axons retracts (Redfern, 1970) and differential activity between inputs controls this phenomenon (Personius and Balice-Gordon, 2001; Sanes and Lichtman, 2001; Personius et al., 2007; Favero et al., 2012). Acetylcholine, the primary neuromuscular transmitter, has long been presumed to mediate this activity-dependent process (O'Brien et al., 1978), but glutamatergic transmission also occurs at the neuromuscular junction (Berger et al., 1995; Grozdanovic and Gossrau, 1998; Mays et al., 2009). To test the role of neuromuscular NMDA receptors, we assessed their contribution to muscle calcium fluxes in mice and tested whether they influence removal of excess innervation at the end plate. Developmental synapse pruning was slowed by reduction of NMDA receptor activation or expression and by reduction of glutamate production. Conversely, pruning is accelerated by application of exogenous NMDA. We also found that NMDA induced increased muscle calcium only during the first 2 postnatal weeks. Therefore, neuromuscular NMDA receptors play previously unsuspected roles in neuromuscular activity and synaptic pruning during development. SIGNIFICANCE STATEMENT In normal adult muscle, each muscle fiber is innervated by a single axon, but at birth, fibers are multiply innervated. Elimination of excess connections requires neural activity; because the neuromuscular junction (NMJ) is a cholinergic synapse, acetylcholine has been assumed to be the critical mediator of activity. However, glutamate receptors are also expressed at the NMJ. We found that axon removal in mice is slowed by pharmacological and molecular manipulations that decrease signaling through neuromuscular NMDA receptors, whereas application of exogenous NMDA at the NMJ accelerates synapse elimination and increases muscle calcium levels during the first 2 postnatal weeks. Therefore, neuromuscular NMDA receptors play previously unsuspected roles in neuromuscular activity and elimination of excess synaptic input during development.
Collapse
|
17
|
Tomàs J, Garcia N, Lanuza MA, Santafé MM, Tomàs M, Nadal L, Hurtado E, Simó A, Cilleros V. Presynaptic Membrane Receptors Modulate ACh Release, Axonal Competition and Synapse Elimination during Neuromuscular Junction Development. Front Mol Neurosci 2017; 10:132. [PMID: 28559796 PMCID: PMC5432534 DOI: 10.3389/fnmol.2017.00132] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/20/2017] [Indexed: 12/17/2022] Open
Abstract
During the histogenesis of the nervous system a lush production of neurons, which establish an excessive number of synapses, is followed by a drop in both neurons and synaptic contacts as maturation proceeds. Hebbian competition between axons with different activities leads to the loss of roughly half of the neurons initially produced so connectivity is refined and specificity gained. The skeletal muscle fibers in the newborn neuromuscular junction (NMJ) are polyinnervated but by the end of the competition, 2 weeks later, the NMJ are innervated by only one axon. This peripheral synapse has long been used as a convenient model for synapse development. In the last few years, we have studied transmitter release and the local involvement of the presynaptic muscarinic acetylcholine autoreceptors (mAChR), adenosine autoreceptors (AR) and trophic factor receptors (TFR, for neurotrophins and trophic cytokines) during the development of NMJ and in the adult. This review article brings together previously published data and proposes a molecular background for developmental axonal competition and loss. At the end of the first week postnatal, these receptors modulate transmitter release in the various nerve terminals on polyinnervated NMJ and contribute to axonal competition and synapse elimination.
Collapse
Affiliation(s)
- Josep Tomàs
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i VirgiliReus, Spain
| | - Neus Garcia
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i VirgiliReus, Spain
| | - Maria A Lanuza
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i VirgiliReus, Spain
| | - Manel M Santafé
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i VirgiliReus, Spain
| | - Marta Tomàs
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i VirgiliReus, Spain
| | - Laura Nadal
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i VirgiliReus, Spain
| | - Erica Hurtado
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i VirgiliReus, Spain
| | - Anna Simó
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i VirgiliReus, Spain
| | - Víctor Cilleros
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i VirgiliReus, Spain
| |
Collapse
|
18
|
Kasimov M, Fatkhrakhmanova M, Mukhutdinova K, Petrov A. 24S-Hydroxycholesterol enhances synaptic vesicle cycling in the mouse neuromuscular junction: Implication of glutamate NMDA receptors and nitric oxide. Neuropharmacology 2017; 117:61-73. [DOI: 10.1016/j.neuropharm.2017.01.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/24/2017] [Accepted: 01/27/2017] [Indexed: 12/29/2022]
|
19
|
Tsentsevitsky A, Nurullin L, Nikolsky E, Malomouzh A. Metabotropic and ionotropic glutamate receptors mediate the modulation of acetylcholine release at the frog neuromuscular junction. J Neurosci Res 2016; 95:1391-1401. [DOI: 10.1002/jnr.23977] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 09/19/2016] [Accepted: 10/05/2016] [Indexed: 12/28/2022]
Affiliation(s)
- Andrei Tsentsevitsky
- Laboratory of Biophysics of Synaptic Processes; Kazan Institute of Biochemistry and Biophysics, Russian Academy of Sciences; Kazan Russia
- Open Laboratory of Neuropharmacology; Kazan Federal University; Kazan Russia
| | - Leniz Nurullin
- Laboratory of Biophysics of Synaptic Processes; Kazan Institute of Biochemistry and Biophysics, Russian Academy of Sciences; Kazan Russia
- Open Laboratory of Neuropharmacology; Kazan Federal University; Kazan Russia
- Department of Biology; Kazan State Medical University; Kazan Russia
| | - Evgeny Nikolsky
- Laboratory of Biophysics of Synaptic Processes; Kazan Institute of Biochemistry and Biophysics, Russian Academy of Sciences; Kazan Russia
- Open Laboratory of Neuropharmacology; Kazan Federal University; Kazan Russia
- Department of Medical and Biological Physics; Kazan State Medical University; Kazan Russia
| | - Artem Malomouzh
- Laboratory of Biophysics of Synaptic Processes; Kazan Institute of Biochemistry and Biophysics, Russian Academy of Sciences; Kazan Russia
- Open Laboratory of Neuropharmacology; Kazan Federal University; Kazan Russia
| |
Collapse
|
20
|
Perez-Siles G, Grant A, Ellis M, Ly C, Kidambi A, Khalil M, Llanos RM, Fontaine SL, Strickland AV, Züchner S, Bermeo S, Neist E, Brennan-Speranza TC, Takata RI, Speck-Martins CE, Mercer JFB, Nicholson GA, Kennerson ML. Characterizing the molecular phenotype of an Atp7a(T985I) conditional knock in mouse model for X-linked distal hereditary motor neuropathy (dHMNX). Metallomics 2016; 8:981-92. [PMID: 27293072 DOI: 10.1039/c6mt00082g] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
ATP7A is a P-type ATPase essential for cellular copper (Cu) transport and homeostasis. Loss-of-function ATP7A mutations causing systemic Cu deficiency are associated with severe Menkes disease or its milder allelic variant, occipital horn syndrome. We previously identified two rare ATP7A missense mutations (P1386S and T994I) leading to a non-fatal form of motor neuron disorder, X-linked distal hereditary motor neuropathy (dHMNX), without overt signs of systemic Cu deficiency. Recent investigations using a tissue specific Atp7a knock out model have demonstrated that Cu plays an essential role in motor neuron maintenance and function, however the underlying pathogenic mechanisms of ATP7A mutations causing axonal degeneration remain unknown. We have generated an Atp7a conditional knock in mouse model of dHMNX expressing Atp7a(T985I), the orthologue of the human ATP7A(T994I) identified in dHMNX patients. Although a degenerative motor phenotype is not observed, the knock in Atp7a(T985I/Y) mice show altered Cu levels within the peripheral and central nervous systems, an increased diameter of the muscle fibres and altered myogenin and myostatin gene expression. Atp7a(T985I/Y) mice have reduced Atp7a protein levels and recapitulate the defective trafficking and altered post-translational regulatory mechanisms observed in the human ATP7A(T994I) patient fibroblasts. Our model provides a unique opportunity to characterise the molecular phenotype of dHMNX and the time course of cellular events leading to the process of axonal degeneration in this disease.
Collapse
Affiliation(s)
- Gonzalo Perez-Siles
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Concord, NSW, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Still NAAG’ing After All These Years. NEUROPSYCHOPHARMACOLOGY: A TRIBUTE TO JOSEPH T. COYLE 2016; 76:215-55. [DOI: 10.1016/bs.apha.2016.01.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
22
|
Bukharaeva E, Shakirzyanova A, Khuzakhmetova V, Sitdikova G, Giniatullin R. Homocysteine aggravates ROS-induced depression of transmitter release from motor nerve terminals: potential mechanism of peripheral impairment in motor neuron diseases associated with hyperhomocysteinemia. Front Cell Neurosci 2015; 9:391. [PMID: 26500495 PMCID: PMC4594498 DOI: 10.3389/fncel.2015.00391] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/18/2015] [Indexed: 12/13/2022] Open
Abstract
Homocysteine (HCY) is a pro-inflammatory sulphur-containing redox active endogenous amino acid, which concentration increases in neurodegenerative disorders including amyotrophic lateral sclerosis (ALS). A widely held view suggests that HCY could contribute to neurodegeneration via promotion of oxidative stress. However, the action of HCY on motor nerve terminals has not been investigated so far. We previously reported that oxidative stress inhibited synaptic transmission at the neuromuscular junction, targeting primarily the motor nerve terminals. In the current study, we investigated the effect of HCY on oxidative stress-induced impairment of transmitter release at the mouse diaphragm muscle. The mild oxidant H2O2 decreased the intensity of spontaneous quantum release from nerve terminals (measured as the frequency of miniature endplate potentials, MEPPs) without changes in the amplitude of MEPPs, indicating a presynaptic effect. Pre-treatment with HCY for 2 h only slightly affected both amplitude and frequency of MEPPs but increased the inhibitory potency of H2O2 almost two fold. As HCY can activate certain subtypes of glutamate N-methyl D-aspartate (NMDA) receptors we tested the role of NMDA receptors in the sensitizing action of HCY. Remarkably, the selective blocker of NMDA receptors, AP-5 completely removed the sensitizing effect of HCY on the H2O2-induced presynaptic depressant effect. Thus, at the mammalian neuromuscular junction HCY largely increases the inhibitory effect of oxidative stress on transmitter release, via NMDA receptors activation. This combined effect of HCY and local oxidative stress can specifically contribute to the damage of presynaptic terminals in neurodegenerative motoneuron diseases, including ALS.
Collapse
Affiliation(s)
- Ellya Bukharaeva
- Department of Physiology, Kazan Federal UniversityKazan, Russia
- A. I. Virtanen Institute for Molecular Sciences, Department of Neurobiology, University of Eastern FinlandKuopio, Finland
- Kazan Institute of Biochemistry and BiophysicsKazan, Russia
| | - Anastasia Shakirzyanova
- Department of Physiology, Kazan Federal UniversityKazan, Russia
- A. I. Virtanen Institute for Molecular Sciences, Department of Neurobiology, University of Eastern FinlandKuopio, Finland
| | - Venera Khuzakhmetova
- Department of Physiology, Kazan Federal UniversityKazan, Russia
- Kazan Institute of Biochemistry and BiophysicsKazan, Russia
| | - Guzel Sitdikova
- A. I. Virtanen Institute for Molecular Sciences, Department of Neurobiology, University of Eastern FinlandKuopio, Finland
| | - Rashid Giniatullin
- Department of Physiology, Kazan Federal UniversityKazan, Russia
- A. I. Virtanen Institute for Molecular Sciences, Department of Neurobiology, University of Eastern FinlandKuopio, Finland
| |
Collapse
|
23
|
Khacho P, Wang B, Ahlskog N, Hristova E, Bergeron R. Differential effects of N-acetyl-aspartyl-glutamate on synaptic and extrasynaptic NMDA receptors are subunit- and pH-dependent in the CA1 region of the mouse hippocampus. Neurobiol Dis 2015; 82:580-592. [PMID: 26303888 DOI: 10.1016/j.nbd.2015.08.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 08/13/2015] [Accepted: 08/17/2015] [Indexed: 11/30/2022] Open
Abstract
Ischemic strokes cause excessive release of glutamate, leading to overactivation of N-methyl-d-aspartate receptors (NMDARs) and excitotoxicity-induced neuronal death. For this reason, inhibition of NMDARs has been a central focus in identifying mechanisms to avert this extensive neuronal damage. N-acetyl-aspartyl-glutamate (NAAG), the most abundant neuropeptide in the brain, is neuroprotective in ischemic conditions in vivo. Despite this evidence, the exact mechanism underlying its neuroprotection, and more specifically its effect on NMDARs, is currently unknown due to conflicting results in the literature. Here, we uncover a pH-dependent subunit-specific action of NAAG on NMDARs. Using whole-cell electrophysiological recordings on acute hippocampal slices from adult mice and on HEK293 cells, we found that NAAG increases synaptic GluN2A-containing NMDAR EPSCs, while effectively decreasing extrasynaptic GluN2B-containing NMDAR EPSCs in physiological pH. Intriguingly, the results of our study further show that in low pH, which is a physiological occurrence during ischemia, NAAG depresses GluN2A-containing NMDAR EPSCs and amplifies its inhibitory effect on GluN2B-containing NMDAR EPSCs, as well as upregulates the surface expression of the GluN2A subunit. Altogether, our data demonstrate that NAAG has differential effects on NMDAR function based on subunit composition and pH. These findings suggest that the role of NAAG as a neuroprotective agent during an ischemic stroke is likely mediated by its ability to reduce NMDAR excitation. The inhibitory effect of NAAG on NMDARs and its enhanced function in acidic conditions make NAAG a prime therapeutic agent for the treatment of ischemic events.
Collapse
Affiliation(s)
- Pamela Khacho
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Boyang Wang
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Nina Ahlskog
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Elitza Hristova
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Richard Bergeron
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Department of Psychiatry, University of Ottawa, Ottawa, ON K1Z 7K4, Canada; Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON K1Y 4E9, Canada.
| |
Collapse
|
24
|
Dynnik VV, Kononov AV, Sergeev AI, Teplov IY, Tankanag AV, Zinchenko VP. To Break or to Brake Neuronal Network Accelerated by Ammonium Ions? PLoS One 2015. [PMID: 26217943 PMCID: PMC4517767 DOI: 10.1371/journal.pone.0134145] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Purpose The aim of present study was to investigate the effects of ammonium ions on in vitro neuronal network activity and to search alternative methods of acute ammonia neurotoxicity prevention. Methods Rat hippocampal neuronal and astrocytes co-cultures in vitro, fluorescent microscopy and perforated patch clamp were used to monitor the changes in intracellular Ca2+- and membrane potential produced by ammonium ions and various modulators in the cells implicated in neural networks. Results Low concentrations of NH4Cl (0.1–4 mM) produce short temporal effects on network activity. Application of 5–8 mM NH4Cl: invariably transforms diverse network firing regimen to identical burst patterns, characterized by substantial neuronal membrane depolarization at plateau phase of potential and high-amplitude Ca2+-oscillations; raises frequency and average for period of oscillations Ca2+-level in all cells implicated in network; results in the appearance of group of «run out» cells with high intracellular Ca2+ and steadily diminished amplitudes of oscillations; increases astrocyte Ca2+-signalling, characterized by the appearance of groups of cells with increased intracellular Ca2+-level and/or chaotic Ca2+-oscillations. Accelerated network activity may be suppressed by the blockade of NMDA or AMPA/kainate-receptors or by overactivation of AMPA/kainite-receptors. Ammonia still activate neuronal firing in the presence of GABA(A) receptors antagonist bicuculline, indicating that «disinhibition phenomenon» is not implicated in the mechanisms of networks acceleration. Network activity may also be slowed down by glycine, agonists of metabotropic inhibitory receptors, betaine, L-carnitine, L-arginine, etc. Conclusions Obtained results demonstrate that ammonium ions accelerate neuronal networks firing, implicating ionotropic glutamate receptors, having preserved the activities of group of inhibitory ionotropic and metabotropic receptors. This may mean, that ammonia neurotoxicity might be prevented by the activation of various inhibitory receptors (i.e. by the reinforcement of negative feedback control), instead of application of various enzyme inhibitors and receptor antagonists (breaking of neural, metabolic and signaling systems).
Collapse
Affiliation(s)
- Vladimir V. Dynnik
- Laboratory of intracellular signaling, Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
- Laboratory of bioenergetics, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
- * E-mail:
| | - Alexey V. Kononov
- Laboratory of intracellular signaling, Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Alexander I. Sergeev
- Laboratory of intracellular signaling, Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Iliya Y. Teplov
- Laboratory of intracellular signaling, Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Arina V. Tankanag
- Laboratory of intracellular signaling, Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Valery P. Zinchenko
- Laboratory of intracellular signaling, Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
| |
Collapse
|
25
|
Blizzard CA, Southam KA, Dawkins E, Lewis KE, King AE, Clark JA, Dickson TC. Identifying the primary site of pathogenesis in amyotrophic lateral sclerosis - vulnerability of lower motor neurons to proximal excitotoxicity. Dis Model Mech 2015; 8:215-24. [PMID: 25740331 PMCID: PMC4348560 DOI: 10.1242/dmm.018606] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 01/20/2015] [Indexed: 12/13/2022] Open
Abstract
There is a desperate need for targeted therapeutic interventions that slow the progression of amyotrophic lateral sclerosis (ALS). ALS is a disorder with heterogeneous onset, which then leads to common final pathways involving multiple neuronal compartments that span both the central and peripheral nervous system. It is believed that excitotoxic mechanisms might play an important role in motor neuron death in ALS. However, little is known about the mechanisms by which excitotoxicity might lead to the neuromuscular junction degeneration that characterizes ALS, or about the site at which this excitotoxic cascade is initiated. Using a novel compartmentalised model of site-specific excitotoxin exposure in lower motor neurons in vitro, we found that spinal motor neurons are vulnerable to somatodendritic, but not axonal, excitotoxin exposure. Thus, we developed a model of somatodendritic excitotoxicity in vivo using osmotic mini pumps in Thy-1-YFP mice. We demonstrated that in vivo cell body excitotoxin exposure leads to significant motor neuron death and neuromuscular junction (NMJ) retraction. Using confocal real-time live imaging of the gastrocnemius muscle, we found that NMJ remodelling preceded excitotoxin-induced NMJ degeneration. These findings suggest that excitotoxicity in the spinal cord of individuals with ALS might result in a die-forward mechanism of motor neuron death from the cell body outward, leading to initial distal plasticity, followed by subsequent pathology and degeneration.
Collapse
Affiliation(s)
- Catherine A Blizzard
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, TAS 7000, Australia
| | - Katherine A Southam
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, TAS 7000, Australia
| | - Edgar Dawkins
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, TAS 7000, Australia
| | - Katherine E Lewis
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, TAS 7000, Australia
| | - Anna E King
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, TAS 7000, Australia
| | - Jayden A Clark
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, TAS 7000, Australia
| | - Tracey C Dickson
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, TAS 7000, Australia
| |
Collapse
|
26
|
Lainiola M, Procaccini C, Linden AM. mGluR3 knockout mice show a working memory defect and an enhanced response to MK-801 in the T- and Y-maze cognitive tests. Behav Brain Res 2014; 266:94-103. [DOI: 10.1016/j.bbr.2014.03.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Revised: 02/28/2014] [Accepted: 03/04/2014] [Indexed: 12/29/2022]
|
27
|
Moffett JR, Arun P, Ariyannur PS, Namboodiri AMA. N-Acetylaspartate reductions in brain injury: impact on post-injury neuroenergetics, lipid synthesis, and protein acetylation. FRONTIERS IN NEUROENERGETICS 2013; 5:11. [PMID: 24421768 PMCID: PMC3872778 DOI: 10.3389/fnene.2013.00011] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 12/09/2013] [Indexed: 12/22/2022]
Abstract
N-Acetylaspartate (NAA) is employed as a non-invasive marker for neuronal health using proton magnetic resonance spectroscopy (MRS). This utility is afforded by the fact that NAA is one of the most concentrated brain metabolites and that it produces the largest peak in MRS scans of the healthy human brain. NAA levels in the brain are reduced proportionately to the degree of tissue damage after traumatic brain injury (TBI) and the reductions parallel the reductions in ATP levels. Because NAA is the most concentrated acetylated metabolite in the brain, we have hypothesized that NAA acts in part as an extensive reservoir of acetate for acetyl coenzyme A synthesis. Therefore, the loss of NAA after TBI impairs acetyl coenzyme A dependent functions including energy derivation, lipid synthesis, and protein acetylation reactions in distinct ways in different cell populations. The enzymes involved in synthesizing and metabolizing NAA are predominantly expressed in neurons and oligodendrocytes, respectively, and therefore some proportion of NAA must be transferred between cell types before the acetate can be liberated, converted to acetyl coenzyme A and utilized. Studies have indicated that glucose metabolism in neurons is reduced, but that acetate metabolism in astrocytes is increased following TBI, possibly reflecting an increased role for non-glucose energy sources in response to injury. NAA can provide additional acetate for intercellular metabolite trafficking to maintain acetyl CoA levels after injury. Here we explore changes in NAA, acetate, and acetyl coenzyme A metabolism in response to brain injury.
Collapse
Affiliation(s)
- John R. Moffett
- Neuroscience Program, Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health SciencesBethesda, MD, USA
| | | | | | | |
Collapse
|
28
|
Lindgren CA, Newman ZL, Morford JJ, Ryan SB, Battani KA, Su Z. Cyclooxygenase-2, prostaglandin E2 glycerol ester and nitric oxide are involved in muscarine-induced presynaptic enhancement at the vertebrate neuromuscular junction. J Physiol 2013; 591:4749-64. [PMID: 23818695 DOI: 10.1113/jphysiol.2013.256727] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Previous work has demonstrated that activation of muscarinic acetylcholine receptors at the lizard neuromuscular junction (NMJ) induces a biphasic modulation of evoked neurotransmitter release: an initial depression followed by a delayed enhancement. The depression is mediated by the release of the endocannabinoid 2-arachidonylglycerol (2-AG) from the muscle and its binding to cannabinoid type 1 receptors on the motor nerve terminal. The work presented here suggests that the delayed enhancement of neurotransmitter release is mediated by cyclooxygenase-2 (COX-2) as it converts 2-AG to the glycerol ester of prostaglandin E2 (PGE2-G). Using immunofluorescence, COX-2 was detected in the perisynaptic Schwann cells (PSCs) surrounding the NMJ. Pretreatment with either of the selective COX-2 inhibitors, nimesulide or DuP 697, prevents the delayed increase in endplate potential (EPP) amplitude normally produced by muscarine. In keeping with its putative role as a mediator of the delayed muscarinic effect, PGE2-G enhances evoked neurotransmitter release. Specifically, PGE2-G increases the amplitude of EPPs without altering that of spontaneous miniature EPPs. As shown previously for the muscarinic effect, the enhancement of evoked neurotransmitter release by PGE2-G depends on nitric oxide (NO) as the response is abolished by application of either N(G)-nitro-l-arginine methyl ester (l-NAME), an inhibitor of NO synthesis, or carboxy-PTIO, a chelator of NO. Intriguingly, the enhancement is not prevented by AH6809, a prostaglandin receptor antagonist, but is blocked by capsazepine, a TRPV1 and TRPM8 receptor antagonist. Taken together, these results suggest that the conversion of 2-AG to PGE2-G by COX-2 underlies the muscarine-induced enhancement of neurotransmitter release at the vertebrate NMJ.
Collapse
Affiliation(s)
- Clark A Lindgren
- C. A. Lindgren: Grinnell College, Department of Biology, 1116 8th Ave., Grinnell College, Grinnell, IA 50112, USA.
| | | | | | | | | | | |
Collapse
|