51
|
Reliability of neuromuscular transmission and how it is maintained. HANDBOOK OF CLINICAL NEUROLOGY 2008; 91:27-101. [PMID: 18631840 DOI: 10.1016/s0072-9752(07)01502-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
52
|
Serulle Y, Sugimori M, Llinás RR. Imaging synaptosomal calcium concentration microdomains and vesicle fusion by using total internal reflection fluorescent microscopy. Proc Natl Acad Sci U S A 2007; 104:1697-702. [PMID: 17242349 PMCID: PMC1785242 DOI: 10.1073/pnas.0610741104] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Transmitter release at chemical synapses is triggered by high calcium concentration microprofiles at the presynaptic cytosol. Such microprofiles, generated by the opening of voltage-dependent calcium channels at the presynaptic plasma membrane, have been defined as calcium concentration microdomains. Using total internal reflection fluorescent microscopy in conjunction with calcium and vesicular release indicator dyes, we have directly visualized the close apposition of calcium concentration microdomains and synaptic release sites at single synaptic terminals from the CNS from rat cerebellar mossy fiber and squid optic lobe. These findings demonstrate the close apposition of calcium entry and release sites and the dynamics of such site locations over time. Kinetic analysis shows that vesicles can be released via two distinct mechanisms: full-fusion and kiss-and-run. Calcium triggers vesicular motion toward the membrane, and the speed of such movement is calcium concentration-dependent. Moreover, the immediately available vesicular pool represents molecularly trapped vesicles that can be located at a larger distance from the plasma membrane than the field illuminated by total internal reflection fluorescent microscopy.
Collapse
Affiliation(s)
- Yafell Serulle
- *Program in Neuroscience and Physiology
- Department of Biochemistry, and
- Marine Biological Laboratory, Woods Hole, MA 02543
| | - Mutsuyuki Sugimori
- *Program in Neuroscience and Physiology
- Department of Physiology and Neuroscience, New York University School of Medicine, New York, NY 10016; and
- Marine Biological Laboratory, Woods Hole, MA 02543
| | - Rodolfo R. Llinás
- *Program in Neuroscience and Physiology
- Department of Physiology and Neuroscience, New York University School of Medicine, New York, NY 10016; and
- Marine Biological Laboratory, Woods Hole, MA 02543
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
53
|
Gracheva EO, Burdina AO, Holgado AM, Berthelot-Grosjean M, Ackley BD, Hadwiger G, Nonet ML, Weimer RM, Richmond JE. Tomosyn inhibits synaptic vesicle priming in Caenorhabditis elegans. PLoS Biol 2006; 4:e261. [PMID: 16895441 PMCID: PMC1514790 DOI: 10.1371/journal.pbio.0040261] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2006] [Accepted: 06/06/2006] [Indexed: 11/19/2022] Open
Abstract
Caenorhabditis elegans TOM-1 is orthologous to vertebrate tomosyn, a cytosolic syntaxin-binding protein implicated in the modulation of both constitutive and regulated exocytosis. To investigate how TOM-1 regulates exocytosis of synaptic vesicles in vivo, we analyzed
C. elegans tom-1 mutants. Our electrophysiological analysis indicates that evoked postsynaptic responses at
tom-1 mutant synapses are prolonged leading to a two-fold increase in total charge transfer. The enhanced response in
tom-1 mutants is not associated with any detectable changes in postsynaptic response kinetics, neuronal outgrowth, or synaptogenesis. However, at the ultrastructural level, we observe a concomitant increase in the number of plasma membrane-contacting vesicles in
tom-1 mutant synapses, a phenotype reversed by neuronal expression of TOM-1. Priming defective
unc-13 mutants show a dramatic reduction in plasma membrane-contacting vesicles, suggesting these vesicles largely represent the primed vesicle pool at the
C. elegans neuromuscular junction. Consistent with this conclusion, hyperosmotic responses in
tom-1 mutants are enhanced, indicating the primed vesicle pool is enhanced. Furthermore, the synaptic defects of
unc-13 mutants are partially suppressed in
tom-1 unc-13 double mutants. These data indicate that in the intact nervous system, TOM-1 negatively regulates synaptic vesicle priming.
This paper examines the in vivo role of the syntaxin binding protein tomosyn in synaptic transmission at the
C. elegans neuromuscular junction. Tomosyn inhibits vesicle priming by regulating the size of the readily releasable vesicle pool.
Collapse
Affiliation(s)
- Elena O Gracheva
- 1Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Anna O Burdina
- 1Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Andrea M Holgado
- 2Biology Department, Loyola University Chicago, Chicago, Illinois, United States of America
| | - Martine Berthelot-Grosjean
- 1Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Brian D Ackley
- 1Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Gayla Hadwiger
- 3Department of Anatomy and Neurobiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Michael L Nonet
- 3Department of Anatomy and Neurobiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Robby M Weimer
- 4Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
| | - Janet E Richmond
- 1Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
54
|
Pardo NE, Hajela RK, Atchison WD. Acetylcholine release at neuromuscular junctions of adult tottering mice is controlled by N-(cav2.2) and R-type (cav2.3) but not L-type (cav1.2) Ca2+ channels. J Pharmacol Exp Ther 2006; 319:1009-20. [PMID: 16982704 DOI: 10.1124/jpet.106.108670] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The mutation in the alpha(1A) subunit gene of the P/Q-type (Ca(v)2.1) Ca(2+) channel present in tottering (tg) mice causes ataxia and motor seizures that resemble absence epilepsy in humans. P/Q-type Ca(2+)channels are primarily involved in acetylcholine (ACh) release at mammalian neuromuscular junctions. Unmasking of L-type (Ca(v)1.1-1.2) Ca(2+) channels occurs in cerebellar Purkinje cells of tg mice. However, whether L-type Ca(2+) channels are also up-regulated at neuromuscular junctions of tg mice is unknown. We characterized thoroughly the pharmacological sensitivity of the Ca(2+) channels, which control ACh release at adult tg neuromuscular junctions. Block of N- and R-type (Ca(v)2.2-2.3), but not L-type Ca(2+) channels, significantly reduced quantal content of end-plate potentials in tg preparations. Neither resting nor KCl-evoked miniature end-plate potential frequency differed significantly between tg and wild type (WT). Immunolabeling of Ca(2+) channel subunits alpha(1A), alpha(1B), alpha(1C), and alpha(1E) revealed an apparent increase of alpha(1B), and alpha(1E) staining, at tg but not WT neuromuscular junctions. This presumably compensates for the deficit of P/Q-type Ca(2+)channels, which localized presynaptically at WT neuromuscular junctions. No alpha(1C) subunits juxtaposed with pre- or postsynaptic markers at either WT or tg neuromuscular junctions. Thus, in adult tg mice, immunocytochemical and electrophysiological data indicate that N- and R-type channels both assume control of ACh release at motor nerve terminals. Recruitment of alternate subtypes of Ca(2+) channels to control transmitter release seems to represent a commonly occurring method of neuronal plasticity. However, it is unclear which conditions underlie recruitment of Ca(v)2 as opposed to Ca(v)1-type Ca(2+) channels.
Collapse
Affiliation(s)
- Nicole E Pardo
- Department of Pharmacology and Toxicology, Michigan State University, B-331 Life Sciences Bldg., East Lansing, MI 48824-1317, USA
| | | | | |
Collapse
|
55
|
Abstract
Brain cells are highly energy dependent for maintaining ion homeostasis during high metabolic activity. During active periods, full mitochondrial function is essential to generate ATP from electrons that originate with the oxidation of NADH. Decreasing brain metabolism is a significant cause of cognitive abnormalities of Alzheimer disease (AD), but it remains uncertain whether this is the cause of further pathology or whether synaptic loss results in a lower energy demand. Synapses are the first to show pathological symptoms in AD before the onset of clinical symptoms. Because synaptic function has high energy demands, interruption in mitochondrial energy supply could be the major factor in synaptic failure in AD. A newly discovered age-related decline in neuronal NADH and redox ratio may jeopardize this function. Mitochondrial dehydrogenases and several mutations affecting energy transfer are frequently altered in aging and AD. Thus, with the accumulation of genetic defects in mitochondria at the level of energy transfer, the issue of neuronal susceptibility to damage as a function of age and age-related disease becomes important. In an aging rat neuron model, mitochondria are both chronically depolarized and produce more reactive oxygen species with age. These concepts suggest that multiple treatment targets may be needed to reverse this multifactorial disease. This review summarizes new insights based on the interaction of mitoenergetic failure, glutamate excitotoxicity, and amyloid toxicity in the exacerbation of AD.
Collapse
Affiliation(s)
- Mordhwaj S Parihar
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62794-9626, USA
| | | |
Collapse
|
56
|
Cho S, Meriney SD. The effects of presynaptic calcium channel modulation by roscovitine on transmitter release at the adult frog neuromuscular junction. Eur J Neurosci 2006; 23:3200-8. [PMID: 16820010 DOI: 10.1111/j.1460-9568.2006.04849.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Calcium (Ca2+) influx through presynaptic calcium channels triggers transmitter release, and any alterations in the gating of these calcium channels results in changes in the magnitude of transmitter released. We used (R)-roscovitine, a cyclin-dependent kinase inhibitor that also appears to act directly on calcium channels, as a tool to modulate presynaptic calcium influx and study effects on transmitter release. We show that this compound increased the quantal content of acetylcholine released from the Rana frog motor nerve terminal (by 149%) without changing paired-pulse facilitation (under low calcium conditions). In contrast, exposure to 3,4-diaminopyridine (DAP; which similarly affects transmitter release by partially blocking potassium channels, altering the shape of the presynaptic action potential, and indirectly increasing calcium entry) increased paired-pulse facilitation (by 23%). In addition, we show that (R)-roscovitine predominately slowed deactivation kinetics of calcium current (by 427%) recorded from Xenopus frog motoneurons, and as a result, increased the integral of calcium channel current evoked by a physiological action potential waveform (by 44%). Because we did not observe any significant effects of structurally related cyclin-dependent kinase inhibitors [(S)-roscovitine or olomoucine] on evoked transmitter release or calcium current kinetics, it appears that these effects of (R)-roscovitine are independent of cyclin-dependent kinases (cdks). In summary, we hypothesize that (R)-roscovitine effects on transmitter release at the adult frog neuromuscular junction (NMJ) are mediated by its effects on calcium channel gating, and these effects increase our understanding of calcium triggered secretion at this synapse.
Collapse
Affiliation(s)
- Soyoun Cho
- Department of Neuroscience, Center for Neuroscience, 446 Crawford Hall, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | | |
Collapse
|
57
|
Sun L, Li Q, Khanna R, Chan AW, Wong F, Stanley EF. Transmitter release face Ca2+channel clusters persist at isolated presynaptic terminals. Eur J Neurosci 2006; 23:1391-6. [PMID: 16553800 DOI: 10.1111/j.1460-9568.2006.04653.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Ca(2+) influx through N-type Ca(2+) channels (CaV2.2) is known to be critical for transmitter release at many synapses. These channels are known to be localized to transmitter release sites, but their anchoring mechanism remains unknown. Recent studies have demonstrated that presynaptic organization is subject to interactions with the postsynaptic cell or the intervening extracellular matrix. We used a previously described high-affinity antibody against the N-type Ca(2+) channels, Ab571, to localize Ca(2+) channel clusters at the release face of an isolated giant calyx-type synapse to test whether the maintenance of these clusters requires an intact extracellular matrix or contact with the postsynaptic cell. Because the number of Ca(2+) channel clusters was unchanged after extracellular matrix dispersal or nerve terminal isolation, we conclude that presynaptic transmitter release face Ca(2+) clusters can be maintained independently of extracellular influences. Our results suggest that a presynaptic molecular scaffold is responsible for the maintenance of release site Ca(2+) channel clusters.
Collapse
Affiliation(s)
- Li Sun
- Cellular and Molecular Biology Division, Toronto Western Research Institute, UHN Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
58
|
Oliveira L, Correia-de-Sá P. Protein kinase A and Ca(v)1 (L-Type) channels are common targets to facilitatory adenosine A2A and muscarinic M1 receptors on rat motoneurons. Neurosignals 2006; 14:262-72. [PMID: 16301841 DOI: 10.1159/000088642] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2005] [Accepted: 07/21/2005] [Indexed: 11/19/2022] Open
Abstract
At the rat motor endplate, pre-synaptic facilitatory adenosine A2A and muscarinic M1 receptors are mutually exclusive. We investigated whether these receptors share a common intracellular signalling pathway. Suppression of McN-A-343-induced M1 facilitation of [3H]ACh release was partially recovered when CGS21680C (an A2A agonist) was combined with the cyclic AMP antagonist Rp-cAMPS. Forskolin, rolipram and 8-bromo-cyclic AMP mimicked CGS21680C blockade of M1 facilitation. Both Rp-cAMPs and nifedipine reduced augmentation of [3H]ACh release by McN-A-343 and CGS21680C. Activation of M1 and A2A receptors enhanced Ca2+ recruitment through nifedipine-sensitive channels. Nifedipine inhibition revealed by McN-A-343 was prevented by chelerythrine (a PKC inhibitor) and Rp-cAMPS, suggesting that Ca(v)1 (L-type) channels phosphorylation by PKA and PKC is required. Rp-cAMPS inhibited [3H]ACh release in the presence of phorbol 12-myristate 13-acetate, but PKC inhibition by chelerythrine had no effect on release in the presence of 8-bromo-cyclic AMP. This suggests that the involvement of PKA may be secondary to M1-induced PKC activation. In conclusion, competition of M1 and A2A receptors to facilitate ACh release from motoneurons may occur by signal convergence to a common pathway involving PKA activation and Ca2+ influx through Ca(v)1 (L-type) channels.
Collapse
Affiliation(s)
- Laura Oliveira
- Laboratório de Farmacologia, Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | | |
Collapse
|
59
|
Khanna R, Sun L, Li Q, Guo L, Stanley EF. Long splice variant N type calcium channels are clustered at presynaptic transmitter release sites without modular adaptor proteins. Neuroscience 2006; 138:1115-25. [PMID: 16473471 DOI: 10.1016/j.neuroscience.2005.12.050] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2005] [Revised: 12/09/2005] [Accepted: 10/30/2005] [Indexed: 11/25/2022]
Abstract
The presynaptic N type Ca channel (CaV2.2) is associated with the transmitter release site apparatus and plays a critical role in the gating of transmitter release. It has been suggested that a distinct CaV2.2 long C terminal splice variant is targeted to the nerve terminal and is anchored at the release face by calcium/calmodulin-dependent serine protein kinase (CASK) and Munc-18-interacting protein (MINT), two modular adaptor proteins. We used the isolated chick ciliary ganglion calyx terminal together with two new antibodies (L4569, L4570) selective for CaV2.2 long C terminal splice variant to test these hypotheses. CaV2.2 long C terminal splice variant was present at the presynaptic transmitter release sites, as identified by Rab3a-interacting molecule (RIM) co-staining and quantitative immunocytochemistry. CASK was also present at the terminal both in conjunction with, and independent of its binding partner, MINT. Immunoprecipitation of CaV2.2 long C terminal splice variant from brain lysate coprecipitated CASK, confirming that these two proteins can form a complex. However, CASK was not colocalized either with CaV2.2 long C terminal splice variant or the transmitter release site marker RIM at the calyx terminal release face. Neither was MINT colocalized with CaV2.2 long C terminal splice variant. Our results show that native CaV2.2 long C terminal splice variant is targeted to the transmitter release sites at an intact presynaptic terminal. However, the lack of enrichment of CASK at the release site combined with the failure of this protein or its partner MINT to colocalize with CaV2.2 argues against the idea that these modular adaptor proteins anchor CaV2.2 at presynaptic nerve terminals.
Collapse
Affiliation(s)
- R Khanna
- Cellular and Molecular Biology Division, Toronto Western Research Institute, University Health Network, 399 Bathurst Street, MP14-320, Toronto, Ontario, Canada M5T 2S8
| | | | | | | | | |
Collapse
|
60
|
Khanna R, Li Q, Sun L, Collins TJ, Stanley EF. N type Ca2+ channels and RIM scaffold protein covary at the presynaptic transmitter release face but are components of independent protein complexes. Neuroscience 2006; 140:1201-8. [PMID: 16757118 DOI: 10.1016/j.neuroscience.2006.04.053] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2006] [Revised: 03/21/2006] [Accepted: 04/12/2006] [Indexed: 11/28/2022]
Abstract
Fast neurotransmitter release at presynaptic terminals occurs at specialized transmitter release sites where docked secretory vesicles are triggered to fuse with the membrane by the influx of Ca2+ ions that enter through local N type (CaV2.2) calcium channels. Thus, neurosecretion involves two key processes: the docking of vesicles at the transmitter release site, a process that involves the scaffold protein RIM (Rab3A interacting molecule) and its binding partner Munc-13, and the subsequent gating of vesicle fusion by activation of the Ca2+ channels. It is not known, however, whether the vesicle fusion complex with its attached Ca2+ channels and the vesicle docking complex are parts of a single multifunctional entity. The Ca2+ channel itself and RIM were used as markers for these two elements to address this question. We carried out immunostaining at the giant calyx-type synapse of the chick ciliary ganglion to localize the proteins at a native, undisturbed presynaptic nerve terminal. Quantitative immunostaining (intensity correlation analysis/intensity correlation quotient method) was used to test the relationship between these two proteins at the nerve terminal transmitter release face. The staining intensities for CaV2.2 and RIM covary strongly, consistent with the expectation that they are both components of the transmitter release sites. We then used immunoprecipitation to test if these proteins are also parts of a common molecular complex. However, precipitation of CaV2.2 failed to capture either RIM or Munc-13, a RIM binding partner. These findings indicate that although the vesicle fusion and the vesicle docking mechanisms coexist at the transmitter release face they are not parts of a common stable complex.
Collapse
Affiliation(s)
- R Khanna
- Cellular and Molecular Biology Division, MP14-320 Toronto Western Research Institute, University Health Network, 399 Bathurst Street, Toronto, Ontario, M5T 2S8 Canada
| | | | | | | | | |
Collapse
|
61
|
Photowala H, Freed R, Alford S. Location and function of vesicle clusters, active zones and Ca2+ channels in the lamprey presynaptic terminal. J Physiol 2005; 569:119-35. [PMID: 16141275 PMCID: PMC1464202 DOI: 10.1113/jphysiol.2005.091314] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/24/2005] [Accepted: 08/30/2005] [Indexed: 11/08/2022] Open
Abstract
Synaptic transmission requires spatial and temporal coordination of a specific sequence of events. The trigger for synaptic vesicle exocytosis is Ca(2)(+) entry into presynaptic terminals, leading to neurotransmitter release at highly specialized sites known as active zones. Ca(2)(+) channel proximity to exocytotic proteins and vesicle clusters at active zones have been inferred from biochemical, histological and ultrastructural data, but direct evidence about functional relationships between these elements in central synapses is absent. We have utilized the lamprey giant reticulospinal synapse to characterize functional colocalization of known synaptic markers in the presynaptic terminal, as well as their reliability during repeated activation. Recycling vesicle clusters, surrounding actin filaments, and physiologically relevant Ca(2)(+) influx all show identical morphological distribution. Ca(2)(+) influx is mediated by clusters of Ca(2)(+) channels that colocalize with the vesicle clusters, defined by imaged sites of vesicle recycling and actin localization. Synaptic transmission is inhibited by block of actin depolymerization, but Ca(2)(+) signalling is unaffected. Functional Ca(2)(+) channels are localized to presynaptic clusters, and Ca(2)(+) transients at these sites account for neurotransmitter release based on their spatial and temporal profiles. Ca(2)(+) transients evoked by single axonal action potentials are mediated solely by voltage-operated Ca(2)(+) channel activation, and slower Ca(2)(+) rises observed throughout the axon result from Ca(2)(+) diffusion from the synaptic regions. We conclude that at lamprey giant reticulospinal synapses, Ca(2)(+) channels and release sites colocalize, creating a close spatial relationship between active zones and Ca(2)(+) entry sites, which is necessary for site-specific, Ca(2)(+)-dependent secretion.
Collapse
Affiliation(s)
- Huzefa Photowala
- Department of Biological Sciences, University of Illinois, Chicago, IL 60607, USA
| | | | | |
Collapse
|
62
|
Mátyás F, Yanovsky Y, Mackie K, Kelsch W, Misgeld U, Freund TF. Subcellular localization of type 1 cannabinoid receptors in the rat basal ganglia. Neuroscience 2005; 137:337-61. [PMID: 16289348 DOI: 10.1016/j.neuroscience.2005.09.005] [Citation(s) in RCA: 147] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2005] [Revised: 09/08/2005] [Accepted: 09/09/2005] [Indexed: 11/29/2022]
Abstract
Endocannabinoids, acting via type 1 cannabinoid receptors (CB1), are known to be involved in short-term synaptic plasticity via retrograde signaling. Strong depolarization of the postsynaptic neurons is followed by the endocannabinoid-mediated activation of presynaptic CB1 receptors, which suppresses GABA and/or glutamate release. This phenomenon is termed depolarization-induced suppression of inhibition (DSI) or excitation (DSE), respectively. Although both phenomena have been reported to be present in the basal ganglia, the anatomical substrate for these actions has not been clearly identified. Here we investigate the high-resolution subcellular localization of CB1 receptors in the nucleus accumbens, striatum, globus pallidus and substantia nigra, as well as in the internal capsule, where the striato-nigral and pallido-nigral pathways are located. In all examined nuclei of the basal ganglia, we found that CB1 receptors were located on the membrane of axon terminals and preterminal axons. Electron microscopic examination revealed that the majority of these axon terminals were GABAergic, giving rise to mostly symmetrical synapses. Interestingly, preterminal axons showed far more intense staining for CB1, especially in the globus pallidus and substantia nigra, whereas their terminals were only faintly stained. Non-varicose, thin unmyelinated fibers in the internal capsule also showed strong CB1-labeling, and were embedded in bundles of myelinated CB1-negative axons. The majority of CB1 receptors labeled by immunogold particles were located in the axonal plasma membrane (92.3%), apparently capable of signaling cannabinoid actions. CB1 receptors in this location cannot directly modulate transmitter release, because the release sites are several hundred micrometers away. Interestingly, both the CB1 agonist, WIN55,212-2, as well as its antagonist, AM251, were able to block action potential generation, but via a CB1 independent mechanism, since the effects remained intact in CB1 knockout animals. Thus, our electrophysiological data suggest that these receptors are unable to influence action potential propagation, thus they may not be functional at these sites, but are likely being transported to the terminal fields. The present data are consistent with a role of endocannabinoids in the control of GABA, but not glutamate, release in the basal ganglia via presynaptic CB1 receptors, but also call the attention to possible non-CB1-mediated effects of widely used cannabinoid ligands on action potential generation.
Collapse
Affiliation(s)
- F Mátyás
- Institute of Experimental Medicine, Hungarian Academy of Sciences, PO Box 67, Budapest H-1450, Hungary
| | | | | | | | | | | |
Collapse
|
63
|
Wiersma-Meems R, Van Minnen J, Syed NI. Synapse formation and plasticity: the roles of local protein synthesis. Neuroscientist 2005; 11:228-37. [PMID: 15911872 DOI: 10.1177/1073858404274110] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
From simple reflexes in lower animals to complex motor patterns and learning and memory in higher animals, all nervous system functions hinge upon fundamental, albeit specialized, neuronal units termed synapses. The term synapse denotes the structural and functional building block upon which pivots the enormous information-processing capabilities of our brain. It is the neuronal communications through synapses that ultimately determine who we are and how we react and adapt to our ever-changing environment. Synapses are not only the epic center of our intellect, but they also control myriad traits of our personality, ranging from sinfulness to sainthood (see, e.g., Hamer 2004). Simply put-we are what our synapses deem us to be (LeDoux 2003)! Notwithstanding the reasoning that some aspects of the synaptic arrangement may be genetically hardwired, an overwhelming body of knowledge does nevertheless provide ample plausible evidence that synapses are highly plastic entities undergoing rapid adaptive changes throughout life. It is this adaptability that endows our brain with its "uncanny" powers.
Collapse
Affiliation(s)
- Ryanne Wiersma-Meems
- Department of Cell Biology and Anatomy, The Hotchkiss Brain Institute of Calgary, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | | |
Collapse
|
64
|
Yokoyama CT, Myers SJ, Fu J, Mockus SM, Scheuer T, Catterall WA. Mechanism of SNARE protein binding and regulation of Cav2 channels by phosphorylation of the synaptic protein interaction site. Mol Cell Neurosci 2005; 28:1-17. [PMID: 15607937 DOI: 10.1016/j.mcn.2004.08.019] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2003] [Revised: 06/02/2004] [Accepted: 08/02/2004] [Indexed: 11/30/2022] Open
Abstract
Ca(v)2.1 and Ca(v)2.2 channels conduct P/Q-type and N-type Ca(2+) currents that initiate neurotransmission and bind SNARE proteins through a synaptic protein interaction (synprint) site. PKC and CaMKII phosphorylate the synprint site and inhibit SNARE protein binding in vitro. Here we identify two separate microdomains that each bind syntaxin 1A and SNAP-25 in vitro and are regulated by PKC phosphorylation at serines 774 and 898 and CaMKII phosphorylation at serines 784 and 896. Activation of PKC resulted in its recruitment to and phosphorylation of Ca(V)2.2 channels, but PKC phosphorylation did not dissociate Ca(V)2.2 channel/syntaxin 1A complexes. Chimeric Ca(V)2.1a channels containing the synprint site of Ca(v)2.2 gain modulation by syntaxin 1A, which is blocked by PKC phosphorylation at the sites identified above. Our results support a bipartite model for the synprint site in which each SNARE-binding microdomain is controlled by a separate PKC and CaMKII phosphorylation site that regulates channel modulation by SNARE proteins.
Collapse
Affiliation(s)
- Charles T Yokoyama
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | |
Collapse
|
65
|
Leenders AM, Sheng ZH. Modulation of neurotransmitter release by the second messenger-activated protein kinases: implications for presynaptic plasticity. Pharmacol Ther 2005; 105:69-84. [PMID: 15626456 PMCID: PMC1804289 DOI: 10.1016/j.pharmthera.2004.10.012] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Activity-dependent modulation of synaptic function and structure is emerging as one of the key mechanisms underlying synaptic plasticity. Whereas over the past decade considerable progress has been made in identifying postsynaptic mechanisms for synaptic plasticity, the presynaptic mechanisms involved have remained largely elusive. Recent evidence implicates that second messenger regulation of the protein interactions in synaptic vesicle release machinery is one mechanism by which cellular events modulate synaptic transmission. Thus, identifying protein kinases and their targets in nerve terminals, particularly those functionally regulated by synaptic activity or intracellular [Ca2+], is critical to the elucidation of the molecular mechanisms underlying modulation of neurotransmitter release and presynaptic plasticity. The phosphorylation and dephosphorylation states of synaptic proteins that mediate vesicle exocytosis could regulate the biochemical pathways leading from synaptic vesicle docking to fusion. However, functional evaluation of the activity-dependent phosphorylation events for modulating presynaptic functions still represents a considerable challenge. Here, we present a brief overview of the data on the newly identified candidate targets of the second messenger-activated protein kinases in the presynaptic release machinery and discuss the potential impact of these phosphorylation events in synaptic strength and presynaptic plasticity.
Collapse
Affiliation(s)
| | - Zu-Hang Sheng
- * Corresponding author. Tel.: 301 435 4596; fax: 301 480 5763. E-mail address: (Z.-H. Sheng)
| |
Collapse
|
66
|
Nishimune H, Sanes JR, Carlson SS. A synaptic laminin-calcium channel interaction organizes active zones in motor nerve terminals. Nature 2005; 432:580-7. [PMID: 15577901 DOI: 10.1038/nature03112] [Citation(s) in RCA: 199] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2004] [Accepted: 10/13/2004] [Indexed: 11/10/2022]
Abstract
Synapse formation requires the differentiation of a functional nerve terminal opposite a specialized postsynaptic membrane. Here, we show that laminin beta2, a component of the synaptic cleft at the neuromuscular junction, binds directly to calcium channels that are required for neurotransmitter release from motor nerve terminals. This interaction leads to clustering of channels, which in turn recruit other presynaptic components. Perturbation of this interaction in vivo results in disassembly of neurotransmitter release sites, resembling defects previously observed in an autoimmune neuromuscular disorder, Lambert-Eaton myasthenic syndrome. These results identify an extracellular ligand of the voltage-gated calcium channel as well as a new laminin receptor. They also suggest a model for the development of nerve terminals, and provide clues to the pathogenesis of a synaptic disease.
Collapse
Affiliation(s)
- Hiroshi Nishimune
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | | | | |
Collapse
|
67
|
Cao YQ, Piedras-Rentería ES, Smith GB, Chen G, Harata NC, Tsien RW. Presynaptic Ca2+ channels compete for channel type-preferring slots in altered neurotransmission arising from Ca2+ channelopathy. Neuron 2004; 43:387-400. [PMID: 15294146 DOI: 10.1016/j.neuron.2004.07.014] [Citation(s) in RCA: 136] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2003] [Revised: 03/29/2004] [Accepted: 07/07/2004] [Indexed: 12/17/2022]
Abstract
Several human channelopathies result from mutations in alpha1A, the pore-forming subunit of P/Q-type Ca2+ channels, conduits of presynaptic Ca2+ entry for evoked neurotransmission. We found that wild-type human alpha1A subunits supported transmission between cultured mouse hippocampal neurons equally well as endogenous mouse alpha1A, whereas introduction of impermeant human alpha1A hampered the effect of endogenous subunits. Thus, presynaptic P/Q-type channels may compete for channel type-preferring "slots" that limit their synaptic effectiveness. The existence of slots generates predictions for how neurotransmission might be affected by changes in Ca2+ channel properties, which we tested by studying alpha1A mutations that are associated with familial hemiplegic migraine type 1 (FHM1). Mutant human P/Q-type channels were impaired in contributing to neurotransmission in precise accord with their deficiency in supporting whole-cell Ca2+ channel activity. Expression of mutant channels in wild-type neurons reduced the synaptic contribution of P/Q-type channels, suggesting that competition for type-preferring slots might support the dominant inheritance of FHM1.
Collapse
Affiliation(s)
- Yu-Qing Cao
- Department of Molecular and Cellular Physiology, Beckman Center, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | |
Collapse
|
68
|
Oliveira L, Timóteo MA, Correia-de-Sá P. Tetanic depression is overcome by tonic adenosine A(2A) receptor facilitation of L-type Ca(2+) influx into rat motor nerve terminals. J Physiol 2004; 560:157-68. [PMID: 15297571 PMCID: PMC1665215 DOI: 10.1113/jphysiol.2004.067595] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2004] [Accepted: 08/04/2004] [Indexed: 12/16/2022] Open
Abstract
Motor nerve terminals possess multiple voltage-sensitive calcium channels operating acetylcholine (ACh) release. In this study, we investigated whether facilitation of neuromuscular transmission by adenosine generated during neuronal firing was operated by Ca(2+) influx via 'prevalent' P-type or via the recruitment of 'silent' L-type channels. The release of [(3)H]ACh from rat phrenic nerve endings decreased upon increasing the stimulation frequency of the trains (750 pulses) from 5 Hz (83 +/- 4 x 10(3) disintegrations per minute per gram (d.p.m. g(-1)); n = 11) to 50 Hz (30 +/- 3 x 10(3) d.p.m. g(-1); n = 5). The P-type Ca(2+) channel blocker, omega-agatoxin IVA (100 nm) reduced (by 40 +/- 10%; n = 6) the release of [(3)H]ACh evoked by 50-Hz trains, while nifedipine (1 microM, an L-type blocker) was inactive. Tetanic depression was overcome (88 +/- 6 x 10(3) d.p.m. g(-1); n = 12) by stimulating the phrenic nerve with 50-Hz bursts (five bursts of 150 pulses, 20 s interburst interval). In these conditions, omega-agatoxin IVA (100 nM) failed to affect transmitter release, but nifedipine (1 microM) decreased [(3)H]ACh release by 21 +/- 7% (n = 4). Inactivation of endogenous adenosine with adenosine deaminase (ADA, 0.5 U ml(-1)) reduced (by 54 +/- 8%, n = 5) the release of [(3)H]ACh evoked with 50-Hz bursts. This effect was opposite to the excitatory actions of adenosine (0.5 mm), S-(p-nitrobenzyl)-6-thioinosine (5 microM, an adenosine uptake blocker) and CGS 21680C (3 nM, a selective A(2A) receptor agonist); as the A(1) receptor agonist R-N(6)-phenylisopropyl adenosine (R-PIA, 300 nM) failed to affect the release of [(3)H]ACh, the results indicate that adenosine generated during 50-Hz bursts exerts an A(2A)-receptor-mediated tonus. The effects of ADA (0.5 U ml(-1)) and CGS 21680C (3 nm) were prevented by nifedipine (1 microM). Blocking tonic A(2A) receptor activation, with ADA (0.5 U ml(-1)) or 3,7-dimethyl-1-propargyl xanthine (10 microM, an A(2A) antagonist), recovered omega-agatoxin IVA (100 nM) inhibition and caused the loss of function of nifedipine (1 microM). Data indicate that, in addition to the predominant P-type Ca(2+) current triggering ACh release during brief tetanic trains, motoneurones possess L-type channels that may be recruited to facilitate transmitter release during high-frequency bursts. The fine-tuning control of Ca(2+) influx through P- or L-type channels is likely to be mediated by endogenous adenosine. Therefore, tonic activation of presynaptic A(2A) receptors operating Ca(2+) influx via L-type channels may contribute to overcome tetanic depression during neuronal firing.
Collapse
Affiliation(s)
- Laura Oliveira
- Laboratório de Farmacologia, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, L. Prof Abel Salazar 2, 4099-003 Porto, Portugal
| | | | | |
Collapse
|
69
|
Zhai RG, Bellen HJ. The Architecture of the Active Zone in the Presynaptic Nerve Terminal. Physiology (Bethesda) 2004; 19:262-70. [PMID: 15381754 DOI: 10.1152/physiol.00014.2004] [Citation(s) in RCA: 194] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Active zones are highly specialized sites for release of neurotransmitter from presynaptic nerve terminals. The architecture of the active zone is exquisitely designed to facilitate the regulated tethering, docking, and fusing of the synaptic vesicles with the plasma membrane. Here we present our view of the structural and molecular organization of active zones across species and propose that all active zones are organized according to a common principle in which the structural differences correlate with the kinetics of transmitter release.
Collapse
Affiliation(s)
- R Grace Zhai
- Howard Hughes Medical Institute and Department of Molecular and Human Genetics, Division of Neuroscience, Program In Developmental Biology, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | |
Collapse
|
70
|
Bennett MR, Farnell L, Gibson WG. The facilitated probability of quantal secretion within an array of calcium channels of an active zone at the amphibian neuromuscular junction. Biophys J 2004; 86:2674-90. [PMID: 15111388 PMCID: PMC1304140 DOI: 10.1016/s0006-3495(04)74323-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2003] [Accepted: 12/18/2003] [Indexed: 10/21/2022] Open
Abstract
A Monte Carlo analysis has been made of the phenomenon of facilitation, whereby a conditioning impulse leaves nerve terminals in a state of heightened release of quanta by a subsequent test impulse, this state persisting for periods of hundreds of milliseconds. It is shown that a quantitative account of facilitation at the amphibian neuromuscular junction can be given if the exocytosis is triggered by the combined action of a low-affinity calcium-binding molecule at the site of exocytosis and a high-affinity calcium-binding molecule some distance away. The kinetic properties and spatial distribution of these molecules at the amphibian neuromuscular junction are arrived at by considering the appropriate values that the relevant parameters must take to successfully account for the experimentally observed amplitude and time course of decline of F1 and F2 facilitation after a conditioning impulse, as well as the growth of facilitation during short trains of impulses. This model of facilitation correctly predicts the effects on facilitation of exogenous buffers such as BAPTA during short trains of impulses. In addition, it accounts for the relative invariance of the kinetics of quantal release due to test-conditioning sequences of impulses as well as due to change in the extent of calcium influx during an impulse.
Collapse
Affiliation(s)
- M R Bennett
- Department of Physiology, The Institute for Biomedical Research, and The School of Mathematics and Statistics, University of Sydney, New South Wales, 2006, Australia.
| | | | | |
Collapse
|
71
|
Pinard A, Lévesque S, Vallée J, Robitaille R. Glutamatergic modulation of synaptic plasticity at a PNS vertebrate cholinergic synapse. Eur J Neurosci 2004; 18:3241-50. [PMID: 14686898 DOI: 10.1111/j.1460-9568.2003.03028.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The presence and the functionality of a glutamatergic regulation was studied at the frog neuromuscular junction (NMJ), a singly innervated cholinergic synapse. Bath application of glutamate reduced transmitter release without affecting nerve-evoked presynaptic Ca2+ entry and handling. (1S,3R)-aminocyclopentanedicarboxylic acid (ACPD), a metabotropic glutamate receptor (mGluR) agonist, mimicked the effects of glutamate while (S)-alpha-methyl-4-carboxyphenylglycine (MCPG), a mGluR antagonist, blocked glutamate effects. MCPG had no effect on transmitter release evoked at low frequency (0.2 Hz) but significantly reduced synaptic depression (10 Hz, 80 s). This suggests that a frequency-dependent endogenous glutamatergic modulation is present at the frog NMJ and is mediated through mGluRs. Immunohistochemical labelling revealed the presence of mGluRs at the end plate area, primarily on muscle fibers. Functional glutamate uptake machinery was also found at the NMJ as blockade of glutamate transport by the inhibitor dl-threo-beta-benzyloxyaspartate (DL-TBOA) increased high frequency-induced depression, suggesting that the transporters system is used to eliminate glutamate from the extracellular space. Moreover, immunohistochemical labelling revealed that glutamate-aspartate transporters (GLASTs) are predominantly present on perisynaptic Schwann cells (PSCs). However, local application of glutamate on PSCs unreliability evoked small Ca2+ responses. Hence, these data suggest that functional glutamatergic interactions at a purely cholinergic synapse, shape synaptic efficacy and short-term plasticity in a frequency-dependent fashion.
Collapse
Affiliation(s)
- Audrée Pinard
- Centre de Recherche en Sciences Neurologiques and Département de physiologie, Université de Montréal, Montréal, Canada,H3C 3J7
| | | | | | | |
Collapse
|
72
|
Kawasaki F, Zou B, Xu X, Ordway RW. Active zone localization of presynaptic calcium channels encoded by the cacophony locus of Drosophila. J Neurosci 2004; 24:282-5. [PMID: 14715960 PMCID: PMC6729574 DOI: 10.1523/jneurosci.3553-03.2004] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Presynaptic calcium channels play a central role in chemical synaptic transmission by providing the calcium trigger for evoked neurotransmitter release. These voltage-gated calcium channels are composed of a primary structural subunit, alpha1, as well as auxiliary beta and alpha2delta subunits. Our previous genetic, molecular, and functional analysis has shown that the cacophony (cac) gene encodes a primary presynaptic calcium channel alpha1 subunit in Drosophila. Here we report that transgenic expression of a cac-encoded alpha1 subunit fused with enhanced green fluorescent protein efficiently rescues cac lethal mutations and allows in vivo analysis of calcium channel localization at active zones. The results reported here further characterize the primary role of cac-encoded calcium channels in neurotransmitter release. In addition, these studies provide a unique genetic tool for live imaging of functional presynaptic calcium channels in vivo and define a molecular marker for immunolocalization of other presynaptic proteins relative to active zones. These findings are expected to facilitate additional analysis of synaptic development and function in this important model system.
Collapse
Affiliation(s)
- Fumiko Kawasaki
- Department of Biology and Genetics Graduate Program, Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | | | | | | |
Collapse
|
73
|
Carter TJ, Mynlieff M. ?-Aminobutyric acid type B receptors facilitate L-type and attenuate N-type Ca2+ currents in isolated hippocampal neurons. J Neurosci Res 2004; 76:323-33. [PMID: 15079861 DOI: 10.1002/jnr.20085] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Activation of presynaptic gamma-aminobutyric acid type B (GABA(B)) receptors inhibits neurotransmitter release at many synapses (both excitatory and inhibitory), and activation of postsynaptic GABA(B) receptors leads to a general inhibition of the postsynaptic cell in mature neurons. Although the action of GABA(B) receptors at the soma of excitatory hippocampal pyramidal cells has been resolved to be regulation of a potassium or calcium conductance, it is not clear that all neurons in the hippocampus demonstrate similar effects of GABA(B) receptor activation. In the current study, GABA(B) receptor-mediated effects on calcium currents in acute cultures composed of heterogeneous cells from the superior region of neonatal hippocampi were studied. In 54.5% of cells, the GABA(B) receptor agonist baclofen (10 microM) attenuated the whole-cell calcium current by 21.0% +/- 1.1%. In 29.9% of cells, baclofen facilitated the calcium current by 43.5% +/- 8.1%. The component of current attenuated by baclofen was blocked by the N-type calcium channel antagonist omega-conotoxin GVIA (3 microM). The component of current facilitated by baclofen was blocked by the L-type channel antagonist nimodipine (20 microM). For cells that showed calcium current facilitation, baclofen shifted the half-maximal activation by approximately -14 mV. The data indicate that activation of GABA(B) receptors in neurons of the superior hippocampus attenuates current through N-type channels and facilitates current through L-type channels. The two opposing effects of GABA(B) receptor activation may reflect the heterogeneity of the cultured cells or may be a developmentally regulated phenomenon.
Collapse
Affiliation(s)
- Thomas J Carter
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin 53201-1881, USA
| | | |
Collapse
|
74
|
Abstract
The neuromuscular junction (NMJ) is a cholinergic synapse that controls muscle contraction. Glial cells, called perisynaptic Schwann cells, surround nerve terminals at the NMJ. Transmitter release induced by repetitive nerve stimulation, elicit a frequency-dependent activation of G-protein-coupled receptors on perisynaptic Schwann cells and the release of calcium from internal stores. In return, perisynaptic Schwann cells modulate synaptic activity during and following high-frequency stimulation through short-term plasticity. In the present review, we discuss evidence of glial involvement in the short-term plasticity at the NMJ and the potential impact of such modulation on synaptic efficacy.
Collapse
Affiliation(s)
- Aurore Colomar
- Centre de Recherche en Sciences Neurologiques, Département de Physiologie, Université de Montréal, Pavillon Paul-G Desmarais, 2960 Chemin de la Tour, PO-Box 6128, Poste Centre-Ville H3C 3J7 Montreal, QC, Canada
| | - Richard Robitaille
- Centre de Recherche en Sciences Neurologiques, Département de Physiologie, Université de Montréal, Pavillon Paul-G Desmarais, 2960 Chemin de la Tour, PO-Box 6128, Poste Centre-Ville H3C 3J7 Montreal, QC, Canada
| |
Collapse
|
75
|
Troncone LRP, Georgiou J, Hua SY, Elrick D, Lebrun I, Magnoli F, Charlton MP. Promiscuous and reversible blocker of presynaptic calcium channels in frog and crayfish neuromuscular junctions from Phoneutria nigriventer spider venom. J Neurophysiol 2003; 90:3529-37. [PMID: 12890791 DOI: 10.1152/jn.00155.2003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Peptide channel blockers found in venoms of many predators are useful pharmacological tools and potential therapeutic agents. The venom of the Brazilian spider Phoneutria nigriventer contains a fraction, omega-phonetoxin-IIA (omega-Ptx-IIA, 8360 MW), which blocks Ca2+ channels. At frog neuromuscular junctions (NMJ) bathed in normal Ca2+ (1.8 mM) saline, omega-Ptx IIA did not affect spontaneous transmitter release but reversibly reduced evoked transmitter release by 75 and 95% at 12 and 24 nM, respectively. In contrast, toxin effects were irreversible in low-Ca2+ (0.5 mM) saline. Ca2+ imaging in normal-Ca2+ saline showed that omega-Ptx-IIA partially blocked stimulus-dependent presynaptic Ca2+ signals, and the blockade was almost completely reversible. Increases in spontaneous release frequency induced by high extracellular K+ were blocked by omega-Ptx-IIA. Therefore omega-Ptx-IIA blocks N-type Ca2+ channels, which admit Ca2+ that triggers transmitter release at the frog NMJ. Additional evidence predicts that omega-Ptx-IIA binds to N-type Ca2+ channels at a different site from that of omega-Conotoxin-GVIA. omega-Ptx-IIA also gave a low-affinity partial blockade of transmitter release and presynaptic Ca2+ signals at crayfish NMJs where P-type channels are blocked by omega-agatoxin-IVA. The Ca2+-dependent reversibility and promiscuity of this toxin may make it highly useful experimentally and therapeutically.
Collapse
|
76
|
Stanley EF, Reese TS, Wang GZ. Molecular scaffold reorganization at the transmitter release site with vesicle exocytosis or botulinum toxin C1. Eur J Neurosci 2003; 18:2403-7. [PMID: 14622203 DOI: 10.1046/j.1460-9568.2003.02948.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Neurotransmitter release sites at the freeze-fractured frog neuromuscular junction are composed of inner and outer paired rows of large membrane particles, the putative calcium channels, anchored by the ribs of an underlying protein scaffold. We analysed the locations of the release site particles as a reflection of the scaffold structure, comparing particle distributions in secreting terminals with those where secretion was blocked with botulinum toxin A, which cleaves a small segment off SNAP-25, or botulinum toxin C1, which cleaves the cytoplasmic domain of syntaxin. In the idle terminal the inner and outer paired rows were located approximately 25 and approximately 44 nm, respectively, from the release site midline. However, adjacent to vesicular fusion sites both particle rows were displaced towards the midline by approximately 25%. The intervals between the particles along each row were examined by a nearest-neighbour approach. In control terminals the peak interval along the inner row was approximately 17 nm, consistent with previous reports and the spacing of the scaffold ribs. While the average distance between particles in the outer row was also approximately 17 nm, a detailed analysis revealed short 'linear clusters' with a approximately 14 nm interval. These clusters were enriched at vesicle fusion sites, suggesting an association with the docking sites, and were eliminated by botulinum C1, but not A. Our findings suggest, first, that the release site scaffold ribs undergo a predictable, and possibly active, shortening during exocytosis and, second, that at the vesicle docking site syntaxin plays a role in the cross-linking of the rib tips to form the vesicle docking sites.
Collapse
Affiliation(s)
- Elise F Stanley
- Toronto Western Research Institute, University Health Network, MP14-320, 399 Bathurst Street, Toronto ON M5T 2S8, Canada.
| | | | | |
Collapse
|
77
|
Brelje TC, Wessendorf MW, Sorenson RL. Multicolor laser scanning confocal immunofluorescence microscopy: practical application and limitations. Methods Cell Biol 2003; 70:165-244. [PMID: 12512325 DOI: 10.1016/s0091-679x(02)70006-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- T Clark Brelje
- Department of Cell Biology and Neuroanatomy, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | | | | |
Collapse
|
78
|
Auld DS, Robitaille R. Perisynaptic Schwann cells at the neuromuscular junction: nerve- and activity-dependent contributions to synaptic efficacy, plasticity, and reinnervation. Neuroscientist 2003; 9:144-57. [PMID: 12708618 DOI: 10.1177/1073858403252229] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Glial cells are increasingly recognized for their important contributions to CNS and PNS synaptic function. Perisynaptic Schwann cells, which are glial cells at the neuromuscular junction, have proven to be an exceptionally useful model for studying these roles. Recent studies have shown that they detect and reciprocally modulate synaptic efficacy in an activity-dependent manner in the short term. In addition, perisynaptic Schwann cells guide reinnervating nerve sprouts after deinnervation, and many important parameters of this are dependent on synapse activity. Thus, it is hypothesized that perisynaptic Schwann cells are key integrators in a continuum of synaptic efficacy, stability, and plasticity at the neuromuscular junction, which is important for maintaining and restoring synaptic efficacy.
Collapse
Affiliation(s)
- Daniel S Auld
- Centre de Recherche en Sciences Neurologiques, Université de Montréal, Québec, Canada
| | | |
Collapse
|
79
|
Bouchard R, Pattarini R, Geiger JD. Presence and functional significance of presynaptic ryanodine receptors. Prog Neurobiol 2003; 69:391-418. [PMID: 12880633 DOI: 10.1016/s0301-0082(03)00053-4] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Ca(2+)-induced Ca(2+) release (CICR) mediated by sarcoplasmic reticulum resident ryanodine receptors (RyRs) has been well described in cardiac, skeletal and smooth muscle. In brain, RyRs are localised primarily to endoplasmic reticulum (ER) and have been demonstrated in postsynaptic entities, astrocytes and oligodendrocytes where they regulate intracellular Ca(2+) concentration ([Ca(2+)](i)), membrane potential and the activity of a variety of second messenger systems. Recently, the contribution of presynaptic RyRs and CICR to functions of central and peripheral presynaptic terminals, including neurotransmitter release, has received increased attention. However, there is no general agreement that RyRs are localised to presynaptic terminals, nor is it clear that RyRs regulate a large enough pool of intracellular Ca(2+) to be physiologically significant. Here, we review direct and indirect evidence that on balance favours the notion that ER and RyRs are found in presynaptic terminals and are physiologically significant. In so doing, it became obvious that some of the controversy originates from issues related to (i) the ability to demonstrate conclusively the physical presence of ER and RyRs, (ii) whether the biophysical properties of RyRs are such that they can contribute physiologically to regulation of presynaptic [Ca(2+)](i), (iii) how ER Ca(2+) load and feedback gain of CICR contributes to the ability to detect functionally relevant RyRs, (iv) the distance that Ca(2+) diffuses from plasma membranes to RyRs to trigger CICR and from RyRs to the Active Zone to enhance vesicle release, and (v) the experimental conditions used. The recognition that ER Ca(2+) stores are able to modulate local Ca(2+) levels and neurotransmitter release in presynaptic terminals will aid in the understanding of the cellular mechanisms controlling neuronal function.
Collapse
Affiliation(s)
- Ron Bouchard
- Division of Neuroscience Research, St. Boniface Research Centre, Winnipeg, Canada R2H 2A6
| | | | | |
Collapse
|
80
|
Mercier AJ, Friedrich R, Boldt M. Physiological functions of FMRFamide-like peptides (FLPs) in crustaceans. Microsc Res Tech 2003; 60:313-24. [PMID: 12539161 DOI: 10.1002/jemt.10270] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Neuropeptides play important roles in chemical signalling in the central and peripheral nervous systems. One of the largest families of neuropeptides is that of the FMRFamide-like peptides (FLPs). This paper reviews what is known about the physiological functions of FLPs in crustaceans, focussing on the cardiovascular, digestive and neuromuscular systems.
Collapse
Affiliation(s)
- A Joffre Mercier
- Department of Biological Sciences, Brock University, St Catharines, ON L2S 3A1, Canada.
| | | | | |
Collapse
|
81
|
Meunier FA, Feng ZP, Molgó J, Zamponi GW, Schiavo G. Glycerotoxin from Glycera convoluta stimulates neurosecretion by up-regulating N-type Ca2+ channel activity. EMBO J 2002; 21:6733-43. [PMID: 12485994 PMCID: PMC139097 DOI: 10.1093/emboj/cdf677] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
We report here the purification of glycerotoxin from the venom of Glycera convoluta, a novel 320 kDa protein capable of reversibly stimulating spontaneous and evoked neurotransmitter release at the frog neuromuscular junction. However, glycerotoxin is ineffective at the murine neuromuscular junction, which displays a different subtype of voltage- dependent Ca(2+) channels. By sequential and selective inhibition of various types of Ca(2+) channels, we found that glycerotoxin was acting via Ca(v)2.2 (N-type). In neuroendocrine cells, it elicits a robust, albeit transient, influx of Ca(2+) sensitive to the Ca(v)2.2 blockers omega-conotoxin GVIA and MVIIA. Moreover, glycerotoxin triggers a Ca(2+) transient in human embryonic kidney (HEK) cells over-expressing Ca(v)2.2 but not Ca(v)2.1 (P/Q-type). Whole-cell patch-clamp analysis of Ca(v)2.2 expressing HEK cells revealed an up-regulation of Ca(2+) currents due to a leftward shift of the activation peak upon glycerotoxin addition. A direct interaction between Ca(v)2.2 and this neurotoxin was revealed by co-immunoprecipitation experiments. Therefore, glycerotoxin is a unique addition to the arsenal of tools available to unravel the mechanism controlling Ca(2+)-regulated exocytosis via the specific activation of Ca(v)2.2.
Collapse
Affiliation(s)
- Frédéric A. Meunier
- Molecular Neuropathobiology Laboratory, Cancer Research UK, London Research Institute, Lincoln’s Inn Fields Laboratories, 44 Lincoln’s Inn Fields, London WC2A 3PX, UK, Department of Physiology and Biophysics, University of Calgary, 3330 Hospital Drive NW, Calgary T2N 4N1, Canada and Laboratoire de Neurobiologie Cellulaire et Moleculaire, UPR 9040, CNRS, 1 avenue de la Terrasse, 91198 Gif-sur-Yvette, France Present address: MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB2 2QH, UK Corresponding authors e-mail: or
| | - Zhong-Ping Feng
- Molecular Neuropathobiology Laboratory, Cancer Research UK, London Research Institute, Lincoln’s Inn Fields Laboratories, 44 Lincoln’s Inn Fields, London WC2A 3PX, UK, Department of Physiology and Biophysics, University of Calgary, 3330 Hospital Drive NW, Calgary T2N 4N1, Canada and Laboratoire de Neurobiologie Cellulaire et Moleculaire, UPR 9040, CNRS, 1 avenue de la Terrasse, 91198 Gif-sur-Yvette, France Present address: MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB2 2QH, UK Corresponding authors e-mail: or
| | - Jordi Molgó
- Molecular Neuropathobiology Laboratory, Cancer Research UK, London Research Institute, Lincoln’s Inn Fields Laboratories, 44 Lincoln’s Inn Fields, London WC2A 3PX, UK, Department of Physiology and Biophysics, University of Calgary, 3330 Hospital Drive NW, Calgary T2N 4N1, Canada and Laboratoire de Neurobiologie Cellulaire et Moleculaire, UPR 9040, CNRS, 1 avenue de la Terrasse, 91198 Gif-sur-Yvette, France Present address: MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB2 2QH, UK Corresponding authors e-mail: or
| | - Gerald W. Zamponi
- Molecular Neuropathobiology Laboratory, Cancer Research UK, London Research Institute, Lincoln’s Inn Fields Laboratories, 44 Lincoln’s Inn Fields, London WC2A 3PX, UK, Department of Physiology and Biophysics, University of Calgary, 3330 Hospital Drive NW, Calgary T2N 4N1, Canada and Laboratoire de Neurobiologie Cellulaire et Moleculaire, UPR 9040, CNRS, 1 avenue de la Terrasse, 91198 Gif-sur-Yvette, France Present address: MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB2 2QH, UK Corresponding authors e-mail: or
| | - Giampietro Schiavo
- Molecular Neuropathobiology Laboratory, Cancer Research UK, London Research Institute, Lincoln’s Inn Fields Laboratories, 44 Lincoln’s Inn Fields, London WC2A 3PX, UK, Department of Physiology and Biophysics, University of Calgary, 3330 Hospital Drive NW, Calgary T2N 4N1, Canada and Laboratoire de Neurobiologie Cellulaire et Moleculaire, UPR 9040, CNRS, 1 avenue de la Terrasse, 91198 Gif-sur-Yvette, France Present address: MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB2 2QH, UK Corresponding authors e-mail: or
| |
Collapse
|
82
|
Abstract
Most CNS synapses investigated thus far contain a large number of vesicles docked at the active zone, possibly forming individual release sites. At the present time, it is unclear whether these vesicles can be discharged independently of one another. To investigate this problem, we recorded miniature excitatory currents by whole-cell and single-synapse recordings from CA3-CA1 hippocampal neurons and analyzed their stochastic properties. In addition, spontaneous release was investigated by ultrastructural analysis of quickly frozen synapses, revealing vesicle intermediates in docking and spontaneous fusion states. In these experiments, no signs of inhibitory interactions between quanta could be detected up to 1 msec from the previous discharge. This suggests that exocytosis at one site does not per se inhibit vesicular fusion at neighboring sites. At longer intervals, the output of quanta diverged from a random memoryless Poisson process because of the presence of a bursting component. The latter, which could not be accounted for by random coincidences, was independent of Ca2+ elevations in the cytosol, whether from Ca2+ flux through the plasma membrane or release from internal stores. Results of these experiments, together with the observation of spontaneous pairs of omega profiles at the active zone, suggest that multimodal release is produced by an enduring activation of an integrated cluster of release sites.
Collapse
|
83
|
Flink MT, Atchison WD. Passive transfer of Lambert-Eaton syndrome to mice induces dihydropyridine sensitivity of neuromuscular transmission. J Physiol 2002; 543:567-76. [PMID: 12205190 PMCID: PMC2290502 DOI: 10.1113/jphysiol.2002.021048] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Lambert-Eaton myasthenic syndrome (LEMS) is a paraneoplastic disorder in which autoantibodies apparently target the voltage-gated Ca2+ channels that regulate acetylcholine (ACh) release at motor nerve terminals. P/Q-type Ca2+ channels are primarily involved in ACh release at mammalian neuromuscular junctions. Passive transfer of LEMS to mice by repeated administration of plasma from LEMS patients reduces the amplitude of the perineurial P/Q-type current, and unmasks a dihydropyridine (DHP)-sensitive L-type Ca2+ current at the motor nerve terminal. The present study sought to determine if this DHP-sensitive component contributes to ACh release. Mice were treated for 30 days with plasma from healthy human controls or patients with LEMS. For some studies, diaphragms from naive mice were incubated with LEMS or control human plasma for 2 or 24 h. End-plate potentials (EPPs) and miniature end-plate potentials (MEPPs) were recorded from neuromuscular junctions in the hemidiaphragm. Treatment of mice with LEMS plasma evoked the characteristic electrophysiological signs of LEMS: reduced quantal content and facilitation of EPP amplitudes at high-frequency stimulation. Quantal content was also reduced in muscles incubated acutely with LEMS plasma. Nimodipine, a DHP-type blocker of L-type Ca2+ channels, did not significantly affect the quantal content of muscles treated for 2 or 24 h with either control or LEMS plasma, or following chronic treatment with control plasma. However, following 30 days treatment with LEMS plasma, nimodipine significantly reduced the remaining quantal content to 57.7 +/- 3.3 % of pre-nimodipine control levels. Thus, DHP-sensitive Ca2+ channels become involved in synaptic transmission at the mouse neuromuscular junction after chronic, but not acute treatment with LEMS plasma. However, reductions in quantal release of ACh occur even after very short periods of exposure to LEMS plasma. As such, development of the L-type Ca2+ channel contribution to ACh release during passive transfer of LEMS appears to occur only after quantal release is significantly impaired for an extended duration, suggesting that an adaptive response of the ACh release apparatus occurs in LEMS.
Collapse
Affiliation(s)
- Michael T Flink
- Department of Pharmacology and Toxicology, Michigan State University, B-331 Life Sciences Building, East Lansing, MI 48824-1317, USA
| | | |
Collapse
|
84
|
Atwood HL, Karunanithi S. Diversification of synaptic strength: presynaptic elements. Nat Rev Neurosci 2002; 3:497-516. [PMID: 12094207 DOI: 10.1038/nrn876] [Citation(s) in RCA: 244] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Synapses are not static; their performance is modified adaptively in response to activity. Presynaptic mechanisms that affect the probability of transmitter release or the amount of transmitter that is released are important in synaptic diversification. Here, we address the diversity of presynaptic performance and its underlying mechanisms: how much of the variation can be accounted for by variation in synaptic morphology and how much by molecular differences? Significant progress has been made in defining presynaptic structural contributions to synaptic strength; by contrast, we know little about how presynaptic proteins produce normally observed functional differentiation, despite abundant information on presynaptic proteins and on the effects of their individual manipulation. Closing the gap between molecular and physiological synaptic diversification still represents a considerable challenge.
Collapse
Affiliation(s)
- Harold L Atwood
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada M5S 1A8.
| | | |
Collapse
|
85
|
Honda A, Yamada M, Saisu H, Takahashi H, Mori KJ, Abe T. Direct, Ca2+-dependent interaction between tubulin and synaptotagmin I: a possible mechanism for attaching synaptic vesicles to microtubules. J Biol Chem 2002; 277:20234-42. [PMID: 11925429 DOI: 10.1074/jbc.m112080200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The synaptic vesicle protein synaptotagmin I probably plays important roles in the synaptic vesicle cycle. However, the mechanisms of its action remain unclear. In this study, we have searched for cytoplasmic proteins that interact with synaptotagmin I. We found that the cytoskeletal protein tubulin directly and stoichiometrically bound to recombinant synaptotagmin I. The binding depended on mm Ca(2+), and 1 mol of tubulin dimer bound 2 mol of synaptotagmin I with half-maximal binding at 6.6 microm tubulin. The Ca(2+) dependence mainly resulted from Ca(2+) binding to the Ca(2+) ligands of synaptotagmin I. The C-terminal region of beta-tubulin and both C2 domains of synaptotagmin I were involved in the binding. The YVK motif in the C2 domains of synaptotagmin I was essential for tubulin binding. Tubulin and synaptotagmin I were co-precipitated from the synaptosome extract with monoclonal antibodies to tubulin and SNAP-25 (synaptosome-associated protein of 25 kDa), indicating the presence of tubulin/synaptotagmin I complex and tubulin binding to synaptotagmin I in SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) complexes. Synaptotagmin I promoted tubulin polymerization and bundled microtubules in the presence of Ca(2+). These results suggest that direct interaction between synaptotagmin I and tubulin provides a mechanism for attaching synaptic vesicles to microtubules in high Ca(2+) concentrations.
Collapse
Affiliation(s)
- Atsuko Honda
- Department of Cellular Neurobiology, Brain Research Institute, Faculty of Science, Niigata University, Niigata 951-8585, Japan
| | | | | | | | | | | |
Collapse
|
86
|
Yan Z, Chi P, Bibb JA, Ryan TA, Greengard P. Roscovitine: a novel regulator of P/Q-type calcium channels and transmitter release in central neurons. J Physiol 2002; 540:761-70. [PMID: 11986366 PMCID: PMC2290289 DOI: 10.1113/jphysiol.2001.013376] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Roscovitine is widely used for inhibition of cdk5, a cyclin-dependent kinase expressed predominantly in the brain. A novel function of roscovitine, i.e. an effect on Ca(2+) channels and transmitter release in central neurons, was studied by whole-cell voltage-clamp recordings and time-lapse fluorescence imaging techniques. Extracellular application of roscovitine markedly enhanced the tail calcium current following repolarization from depolarized voltages. This effect was rapid, reversible and dose dependent. Roscovitine dramatically slowed the deactivation kinetics of calcium channels. The deactivation time constant was increased 3- to 6-fold, suggesting that roscovitine could prolong the channel open state and increase the calcium influx. The potentiation of tail calcium currents caused by roscovitine and by the L-channel activator Bay K 8644 was not occluded but additive. Roscovitine-induced potentiation of tail calcium currents was significantly blocked by the P/Q-channel blocker CgTx-MVIIC, indicating that the major target of roscovitine is the P/Q-type calcium channel. In mutant mice with targeted deletion of p35, a neuronal specific activator of cdk5, roscovitine regulated calcium currents in a manner similar to that observed in wild-type mice. Moreover, intracellular perfusion of roscovitine failed to modulate calcium currents. These results suggest that roscovitine acts on extracellular site(s) of calcium channels via a cdk5-independent mechanism. Roscovitine potentiated glutamate release at presynaptic terminals of cultured hippocampal neurons detected with the vesicle trafficking dye FM1-43, consistent with the positive effect of roscovitine on the P/Q-type calcium channel, the major mediator of action potential-evoked transmitter release in the mammalian CNS.
Collapse
Affiliation(s)
- Zhen Yan
- Laboratory of Molecular & Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA.
| | | | | | | | | |
Collapse
|
87
|
Feng ZP, Grigoriev N, Munno D, Lukowiak K, MacVicar BA, Goldberg JI, Syed NI. Development of Ca2+ hotspots between Lymnaea neurons during synaptogenesis. J Physiol 2002; 539:53-65. [PMID: 11850501 PMCID: PMC2290139 DOI: 10.1113/jphysiol.2001.013125] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Calcium (Ca2+) channel clustering at specific presynaptic sites is a hallmark of mature synapses. However, the spatial distribution patterns of Ca2+ channels at newly formed synapses have not yet been demonstrated. Similarly, it is unclear whether Ca2+ 'hotspots' often observed at the presynaptic sites are indeed target cell contact specific and represent a specialized mechanism by which Ca2+ channels are targeted to select synaptic sites. Utilizing both soma-soma paired (synapsed) and single neurons from the mollusk Lymnaea, we have tested the hypothesis that differential gradients of voltage-dependent Ca2+ signals develop in presynaptic neuron at its contact point with the postsynaptic neuron; and that these Ca2+ hotspots are target cell contact specific. Fura-2 imaging, or two-photon laser scanning microscopy of Calcium Green, was coupled with electrophysiological techniques to demonstrate that voltage-induced Ca2+ gradients (hotspots) develop in the presynaptic cell at its contact point with the postsynaptic neuron, but not in unpaired single cells. The incidence of Ca2+ hotspots coincided with the appearance of synaptic transmission between the paired cells, and these gradients were target cell contact specific. In contrast, the voltage-induced Ca2+ signal in unpaired neurons was uniformly distributed throughout the somata; a similar pattern of Ca2+ gradient was observed in the presynaptic neuron when it was soma-soma paired with a non-synaptic partner cell. Moreover, voltage clamp recording techniques, in conjunction with a fast, optical differential perfusion system, were used to demonstrate that the total whole-cell Ca2+ (or Ba2+) current density in single and paired cells was not significantly different. However, the amplitude of Ba2+ current was significantly higher in the presynaptic cell at its contact side with the postsynaptic neurons, compared with non-contacted regions. In summary, this study demonstrates that voltage-induced Ca2+ hotspots develop in the presynaptic cell, concomitant with the appearance of synaptic transmission between the soma-soma paired cells. The appearance of Ca2+ gradients in presynaptic neurons is target cell contact specific and is probably due to a spatial redistribution of existing channels during synaptogenesis.
Collapse
Affiliation(s)
- Zhong-Ping Feng
- Respiratory and Neuroscience Research Groups, Faculty of Medicine, University of Calgary, 3330-Hospital Drive, NW, Calgary, Alberta, Canada T2N 4N1
| | | | | | | | | | | | | |
Collapse
|
88
|
Affiliation(s)
- C K Govind
- Life Sciences Division, University of Toronto at Scarborough, Scarborough, Ontario, Canada.
| | | |
Collapse
|
89
|
Affiliation(s)
- N Morel
- Laboratoire de Neurobiologie Cellulaire et Moléculaire, CNRS, Gif sur Yvette, France.
| | | | | |
Collapse
|
90
|
Tojima T, Ito E. A cyclic AMP-regulated negative feedforward system for neuritogenesis revealed in a neuroblastomaxglioma hybrid cell line. Neuroscience 2001; 104:583-91. [PMID: 11377857 DOI: 10.1016/s0306-4522(01)00061-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We examined the role of second messengers during the neuritogenesis that accompanies neuronal differentiation in a neuroblastomaxglioma hybrid cell line (NG108-15). NG108-15 cells extended neurites after treatment with dibutyryl cyclic AMP. This dibutyryl cyclic AMP treatment evoked the synthesis of voltage-dependent Ca(2+) channel proteins in the cells. The number of neurites was decreased by Ca(2+) influx under condition of high K(+). Interestingly, the increase of neurites stimulated by dibutyryl cyclic AMP and the decrease of neurites caused by high K(+) were both reversible. This is the first study to demonstrate that cyclic AMP regulates a negative feedforward system for neuritogenesis, which links with Ca(2+) signaling. Such a dual role of cyclic AMP may play an important part in precise neurite targeting.
Collapse
Affiliation(s)
- T Tojima
- Division of Biological Sciences, Graduate School of Science, Hokkaido University, 060-0810, Sapporo, Japan
| | | |
Collapse
|
91
|
DiGregorio DA, Negrete O, Jeromin A, Peng HB, Vergara JL. Contact-dependent aggregation of functional Ca2+ channels, synaptic vesicles and postsynaptic receptors in active zones of a neuromuscular junction. Eur J Neurosci 2001; 14:533-46. [PMID: 11553303 DOI: 10.1046/j.0953-816x.2001.01670.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
To examine whether Ca2+ channels aggregate in a contact-dependent manner, we characterized the distribution of synaptic vesicles and postsynaptic receptors, and compared it to the location of Ca2+ entry sites, in a Xenopus laevis nerve-muscle coculture preparation using a localized Ca2+ detection method. The majority (75%) of Ca2+ entry sites at spontaneously formed nerve-muscle contacts were associated with enhanced immunofluorescence to the synaptic vesicle protein, SV2. In contrast, only 11% of recorded sites without Ca2+ transients exhibited significant SV2 immunofluorescence. When comparing the spatial distribution of synaptic markers with that of Ca2+ entry sites, we found that the majority of Ca2+ entry sites (61%) were associated with both enhanced SV2 immunofluorescence and R-BTX fluorescence, thereby identifying putative neurotransmitter release sites where Ca2+ channels, synaptic vesicles and postsynaptic receptors are colocalized. Using polystyrene beads coated with a heparin binding protein known to mediate in vitro postsynaptic receptor clustering, we show that the location of Ca2+ domains was associated with enhanced SV2 immunofluorescence at neurite-to-bead contacts. We conclude that the localization of functional Ca2+ channels to putative active zones follows a contact-dependent signalling mechanism similar to that known to mediate vesicle aggregation and AChR clustering.
Collapse
Affiliation(s)
- D A DiGregorio
- Department of Physiology, UCLA School of Medicine, Los Angeles, CA 90095, USA
| | | | | | | | | |
Collapse
|
92
|
Patton BL, Cunningham JM, Thyboll J, Kortesmaa J, Westerblad H, Edström L, Tryggvason K, Sanes JR. Properly formed but improperly localized synaptic specializations in the absence of laminin alpha4. Nat Neurosci 2001; 4:597-604. [PMID: 11369940 DOI: 10.1038/88414] [Citation(s) in RCA: 162] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Precise apposition of pre- to postsynaptic specializations is required for optimal function of chemical synapses, but little is known about how it is achieved. At the skeletal neuromuscular junction, active zones (transmitter release sites) in the nerve terminal lie directly opposite junctional folds in the postsynaptic membrane. Few active zones or junctional folds form in mice lacking the laminin beta2 chain, which is normally concentrated in the synaptic cleft. beta2 and the broadly expressed gamma1 chain form heterotrimers with alpha chains, three of which, alpha2, alpha4 and alpha5, are present in the synaptic cleft. Thus, alpha2beta2gamma1, alpha4beta2gamma1 and alpha5beta2gamma1 heterotrimers are all lost in beta2 mutants. In mice lacking laminin alpha4, active zones and junctional folds form in normal numbers, but are not precisely apposed to each other. Thus, formation and localization of synaptic specializations are regulated separately, and alpha4beta2gamma1 (called laminin-9) is critical in the latter process.
Collapse
MESH Headings
- Animals
- Crosses, Genetic
- Exons
- Heterozygote
- Homozygote
- Laminin/analysis
- Lamins
- Macromolecular Substances
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle Contraction/physiology
- Muscle Fibers, Skeletal/pathology
- Muscle Fibers, Skeletal/physiology
- Muscle Fibers, Skeletal/ultrastructure
- Muscle, Skeletal/cytology
- Muscle, Skeletal/innervation
- Muscle, Skeletal/pathology
- Necrosis
- Nuclear Proteins/deficiency
- Nuclear Proteins/genetics
- Nuclear Proteins/physiology
- Protein Subunits
- Recombination, Genetic
- Stem Cells
- Synapses/pathology
- Synapses/physiology
- Synapses/ultrastructure
Collapse
Affiliation(s)
- B L Patton
- Department of Anatomy and Neurobiology, Washington University Medical Center, 660 South Euclid Avenue, St. Louis, Missouri 63110, USA.
| | | | | | | | | | | | | | | |
Collapse
|
93
|
Abstract
Reliable transmission of activity from nerve to muscle is necessary for the normal function of the body. The term 'safety factor' refers to the ability of neuromuscular transmission to remain effective under various physiological conditions and stresses. This is a result of the amount of transmitter released per nerve impulse being greater than that required to trigger an action potential in the muscle fibre. The safety factor is a measure of this excess of released transmitter. In this review we discuss the practical difficulties involved in estimating the safety factor in vitro. We then consider the factors that influence the safety factor in vivo. While presynaptic transmitter release may be modulated on a moment to moment basis, the postsynaptic features that determine the effect of released transmitter are not so readily altered to meet changing demands. Different strategies are used by different species to ensure reliable neuromuscular transmission. Some, like frogs, rely on releasing a large amount of transmitter while others, like man, rely on elaborate postsynaptic specialisations to enhance the response to transmitter. In normal adult mammals, the safety factor is generally 3-5. Both pre- and postsynaptic components change during development and may show plasticity in response to injury or disease. Thus, both acquired autoimmune and inherited congenital diseases of the neuromuscular junction (NMJ) can significantly reduce, or even transiently increase, safety factor.
Collapse
Affiliation(s)
- S J Wood
- Department of Physiology, School of Medical Sciences, University of Bristol, University Walk, BS8 1TD, Bristol, UK.
| | | |
Collapse
|
94
|
Differential frequency-dependent regulation of transmitter release by endogenous nitric oxide at the amphibian neuromuscular synapse. J Neurosci 2001. [PMID: 11160378 DOI: 10.1523/jneurosci.21-04-01087.2001] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Nitric oxide (NO) is a potent neuromodulator in the CNS and PNS. At the frog neuromuscular junction (nmj), exogenous application of NO reduces neurotransmitter release, and NO synthases (NOSs), the enzymes producing NO, are present at this synapse. This work aimed at studying the molecular mechanisms by which NO modulates synaptic efficacy at the nmj using electrophysiological recordings and Ca(2+)-imaging techniques. Bath application of the NO donors S-nitroso-N-acetylpenicillamine (SNAP) and sodium nitroprusside decreased end plate potential (EPP) amplitude as well as the frequency of miniature EPPs but not their amplitude. Ca(2+) responses elicited in presynaptic terminals by single action potentials were unaffected by NO, but responses evoked by a short train of stimuli were increased. Tonic endogenous production of NO was observed as suggested by the increase in EPP amplitude by bath application of the NO scavenger hemoglobin and the neuronal NOS inhibitor 3-bromo-7-nitroindazole sodium salt. A soluble guanylate cyclase inhibitor, 6-anilino-5,8-quinolinedione (LY-83583), increased EPP amplitude and occluded the effects of the NO donor, suggesting that NO acts via a cGMP-dependent mechanism. High-frequency-induced depression was reduced in the presence of the NO scavenger but not by LY-83583. However, adenosine-induced depression was significantly reduced after bath perfusion of SNAP and in the presence of LY-83583. Our results indicate that NO regulates transmitter release and adenosine-induced depression via a cGMP-dependent mechanism that occurs after Ca(2+) entry and that high-frequency-induced synaptic depression is regulated by NO in a cGMP-independent manner.
Collapse
|
95
|
Differential regulation of transmitter release by presynaptic and glial Ca2+ internal stores at the neuromuscular synapse. J Neurosci 2001. [PMID: 11245676 DOI: 10.1523/jneurosci.21-06-01911.2001] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The differential regulation of synaptic transmission by internal Ca(2+) stores of presynaptic terminals and perisynaptic Schwann cells (PSCs) was studied at the frog neuromuscular junction. Thapsigargin (tg), an inhibitor of Ca(2+)-ATPase pumps of internal stores, caused a transient Ca(2+) elevation in PSCs, whereas it had no effect on Ca(2+) stores of presynaptic terminals at rest. Tg prolonged presynaptic Ca(2+) responses evoked by single action potentials with no detectable increase in the resting Ca(2+) level in nerve terminals. However, Ca(2+) accumulation was observed during high frequency stimulation. Tg induced a rapid rise in endplate potential (EPP) amplitude, accompanied by a delayed and transient increase. The effects appeared presynaptic, as suggested by the lack of effects of tg on the amplitude and time course of miniature EPPs (MEPPs). However, MEPP frequency was increased when preparations were stimulated tonically (0.2 Hz). The delayed and transient increase in EPP amplitude was occluded by injections of the Ca(2+) chelator BAPTA into PSCs before tg application, whereas a rise in intracellular Ca(2+) in PSCs induced by inositol 1,4,5-triphosphate (IP(3)) injections potentiated transmitter release. Furthermore, increased Ca(2+) buffering capacity after BAPTA injection in PSCs resulted in a more pronounced synaptic depression induced by high frequency stimulation of the motor nerve (10 Hz/80 sec). It is concluded that presynaptic Ca(2+) stores act as a Ca(2+) clearance mechanism to limit the duration of transmitter release, whereas Ca(2+) release from glial stores initiates Ca(2+)-dependent potentiation of synaptic transmission.
Collapse
|
96
|
Herlitze S, Zhong H, Scheuer T, Catterall WA. Allosteric modulation of Ca2+ channels by G proteins, voltage-dependent facilitation, protein kinase C, and Ca(v)beta subunits. Proc Natl Acad Sci U S A 2001; 98:4699-704. [PMID: 11296298 PMCID: PMC31897 DOI: 10.1073/pnas.051628998] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
N-type and P/Q-type Ca(2+) channels are inhibited by neurotransmitters acting through G protein-coupled receptors in a membrane-delimited pathway involving Gbetagamma subunits. Inhibition is caused by a shift from an easily activated "willing" (W) state to a more-difficult-to-activate "reluctant" (R) state. This inhibition can be reversed by strong depolarization, resulting in prepulse facilitation, or by protein kinase C (PKC) phosphorylation. Comparison of regulation of N-type Ca(2+) channels containing Cav2.2a alpha(1) subunits and P/Q-type Ca(2+) channels containing Ca(v)2.1 alpha(1) subunits revealed substantial differences. In the absence of G protein modulation, Ca(v)2.1 channels containing Ca(v)beta subunits were tonically in the W state, whereas Ca(v)2.1 channels without beta subunits and Ca(v)2.2a channels with beta subunits were tonically in the R state. Both Ca(v)2.1 and Ca(v)2.2a channels could be shifted back toward the W state by strong depolarization or PKC phosphorylation. Our results show that the R state and its modulation by prepulse facilitation, PKC phosphorylation, and Ca(v)beta subunits are intrinsic properties of the Ca(2+) channel itself in the absence of G protein modulation. A common allosteric model of G protein modulation of Ca(2+)-channel activity incorporating an intrinsic equilibrium between the W and R states of the alpha(1) subunits and modulation of that equilibrium by G proteins, Ca(v)beta subunits, membrane depolarization, and phosphorylation by PKC accommodates our findings. Such regulation will modulate transmission at synapses that use N-type and P/Q-type Ca(2+) channels to initiate neurotransmitter release.
Collapse
Affiliation(s)
- S Herlitze
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280, USA
| | | | | | | |
Collapse
|
97
|
Abstract
Studies on the amphibian neuromuscular junction have indicated that N-type calcium channels are the sole mediators of stimulus-evoked neurotransmitter release. We show, via both presynaptic and postsynaptic voltage-clamp measurements, that dihydropyridine (DHP)-sensitive calcium channels also contribute to stimulus-evoked release at developing Xenopus neuromuscular junctions. Whereas inhibition of postsynaptic responses by omega-conotoxin (omega-Ctx) GVIA has been taken previously as evidence that only N-type channels mediate transmitter release, we find that both N-type and DHP-sensitive calcium currents are sensitive to this toxin. The unusual sensitivity of DHP-sensitive calcium channels to omega-Ctx GVIA in presynaptic terminals raises the possibility that this channel type may have escaped detection in previous physiological studies on adult frog neuromuscular junctions. Alternatively, the additional channel isoforms may be present only during early development, when they may serve to strengthen collectively presynaptic release during critical periods of synaptogenesis.
Collapse
|
98
|
Nayak SV, Dougherty JJ, McIntosh JM, Nichols RA. Ca(2+) changes induced by different presynaptic nicotinic receptors in separate populations of individual striatal nerve terminals. J Neurochem 2001; 76:1860-70. [PMID: 11259504 DOI: 10.1046/j.1471-4159.2001.00197.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Presynaptic nicotinic acetylcholine receptors likely play a modulatory role in the nerve terminal. Using laser-scanning confocal microscopy, we have characterized physiological responses obtained on activation of presynaptic nicotinic receptors by measuring calcium changes in individual nerve terminals (synaptosomes) isolated from the rat corpus striatum. Nicotine (500 nM) induced Ca(2+) changes in a subset (10-25%) of synaptosomes. The Ca(2+) responses were dependent on extracellular Ca(2+) and desensitized very slowly (several minutes) on prolonged exposure to agonist. The nicotine-induced Ca(2+) responses were dose-dependent and were completely blocked by dihydro-beta-erythroidine (5 microM), differentially affected by mecamylamine (10 microM) and alpha-conotoxin MII (100 nM), and not affected by alpha-bungarotoxin (500 nM). Immunocytochemical studies using well-characterized monoclonal antibodies revealed the presence of the alpha4 and alpha3/alpha5 nicotinic subunits. The nicotine-induced responses were unaffected by prior depolarization or by a mixture of Ca(2+) channel toxins including omega-conotoxin MVIIC (500 nM), omega-conotoxin GVIA (500 nM) and agatoxin TK (200 nM). Our results indicate that nicotinic receptors present on striatal nerve terminals induce Ca(2+) entry largely without involving voltage-gated Ca(2+) channels, most likely by direct permeation via the receptor channel itself. In addition, at least two subpopulations of presynaptic nicotinic receptors reside on separate terminals in the striatum, suggesting distinct modulatory roles.
Collapse
Affiliation(s)
- S V Nayak
- Department of Pharmacology and Physiology, Medical College of Pennsylvania/Hahnemann University, Philadelphia 19102, USA
| | | | | | | |
Collapse
|
99
|
Tojima T, Yamane Y, Takagi H, Takeshita T, Sugiyama T, Haga H, Kawabata K, Ushiki T, Abe K, Yoshioka T, Ito E. Three-dimensional characterization of interior structures of exocytotic apertures of nerve cells using atomic force microscopy. Neuroscience 2001; 101:471-81. [PMID: 11074169 DOI: 10.1016/s0306-4522(00)00320-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We examined the interior structure of exocytotic apertures in synaptic vesicles of neuroblastoma x glioma hybrid cells using atomic force microscopy. The atomic force microscopy detected apertures of 50-100nm in diameter at various depths within the varicosities of these cells. We were also able to image a regular radial pattern on the wall and lump-like structures at the bottom of these apertures. In contrast, scanning electron microscopy could only detect the apertures but not the fine details of their interior. The cells examined here exhibited the same electrophysiological properties and expression of synaptophysin and syntaxin 1 as presynaptic terminals, as studied by various electrophysiological and imaging techniques. Our results indicate that atomic force microscopy allows three-dimensional viewing of the fine structures located inside exocytotic apertures in nerve cells.
Collapse
Affiliation(s)
- T Tojima
- Division of Biological Sciences, Graduate School of Science, Hokkaido University, 060-0810, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Harlow ML, Ress D, Stoschek A, Marshall RM, McMahan UJ. The architecture of active zone material at the frog's neuromuscular junction. Nature 2001; 409:479-84. [PMID: 11206537 DOI: 10.1038/35054000] [Citation(s) in RCA: 313] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2000] [Accepted: 11/24/2000] [Indexed: 11/08/2022]
Abstract
Active zone material at the nervous system's synapses is situated next to synaptic vesicles that are docked at the presynaptic plasma membrane, and calcium channels that are anchored in the membrane. Here we use electron microscope tomography to show the arrangement and associations of structural components of this compact organelle at a model synapse, the frog's neuromuscular junction. Our findings indicate that the active zone material helps to dock the vesicles and anchor the channels, and that its architecture provides both a particular spatial relationship and a structural linkage between them. The structural linkage may include proteins that mediate the calcium-triggered exocytosis of neurotransmitter by the synaptic vesicles during synaptic transmission.
Collapse
Affiliation(s)
- M L Harlow
- Department of Neurobiology, Stanford University School of Medicine, California 94305, USA
| | | | | | | | | |
Collapse
|