1
|
mGluR5 is transiently confined in perisynaptic nanodomains to shape synaptic function. Nat Commun 2023; 14:244. [PMID: 36646691 PMCID: PMC9842668 DOI: 10.1038/s41467-022-35680-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 12/19/2022] [Indexed: 01/18/2023] Open
Abstract
The unique perisynaptic distribution of postsynaptic metabotropic glutamate receptors (mGluRs) at excitatory synapses is predicted to directly shape synaptic function, but mechanistic insight into how this distribution is regulated and impacts synaptic signaling is lacking. We used live-cell and super-resolution imaging approaches, and developed molecular tools to resolve and acutely manipulate the dynamic nanoscale distribution of mGluR5. Here we show that mGluR5 is dynamically organized in perisynaptic nanodomains that localize close to, but not in the synapse. The C-terminal domain of mGluR5 critically controlled perisynaptic confinement and prevented synaptic entry. We developed an inducible interaction system to overcome synaptic exclusion of mGluR5 and investigate the impact on synaptic function. We found that mGluR5 recruitment to the synapse acutely increased synaptic calcium responses. Altogether, we propose that transient confinement of mGluR5 in perisynaptic nanodomains allows flexible modulation of synaptic function.
Collapse
|
2
|
Membrane trafficking and positioning of mGluRs at presynaptic and postsynaptic sites of excitatory synapses. Neuropharmacology 2021; 200:108799. [PMID: 34592242 DOI: 10.1016/j.neuropharm.2021.108799] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/31/2021] [Accepted: 09/17/2021] [Indexed: 01/21/2023]
Abstract
The plethora of functions of glutamate in the brain are mediated by the complementary actions of ionotropic and metabotropic glutamate receptors (mGluRs). The ionotropic glutamate receptors carry most of the fast excitatory transmission, while mGluRs modulate transmission on longer timescales by triggering multiple intracellular signaling pathways. As such, mGluRs mediate critical aspects of synaptic transmission and plasticity. Interestingly, at synapses, mGluRs operate at both sides of the cleft, and thus bidirectionally exert the effects of glutamate. At postsynaptic sites, group I mGluRs act to modulate excitability and plasticity. At presynaptic sites, group II and III mGluRs act as auto-receptors, modulating release properties in an activity-dependent manner. Thus, synaptic mGluRs are essential signal integrators that functionally couple presynaptic and postsynaptic mechanisms of transmission and plasticity. Understanding how these receptors reach the membrane and are positioned relative to the presynaptic glutamate release site are therefore important aspects of synapse biology. In this review, we will discuss the currently known mechanisms underlying the trafficking and positioning of mGluRs at and around synapses, and how these mechanisms contribute to synaptic functioning. We will highlight outstanding questions and present an outlook on how recent technological developments will move this exciting research field forward.
Collapse
|
3
|
McCullock TW, Kammermeier PJ. The evidence for and consequences of metabotropic glutamate receptor heterodimerization. Neuropharmacology 2021; 199:108801. [PMID: 34547332 DOI: 10.1016/j.neuropharm.2021.108801] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 09/07/2021] [Accepted: 09/17/2021] [Indexed: 12/15/2022]
Abstract
Metabotropic glutamate receptors (mGluRs) are an essential component of the mammalian central nervous system. These receptors modulate neuronal excitability in response to extracellular glutamate through the activation of intracellular heterotrimeric G proteins. Like most other class C G protein-coupled receptors, mGluRs function as obligate dimer proteins, meaning they need to form dimer complexes before becoming functional receptors. All mGluRs possess the ability to homodimerize, but studies over the past ten years have demonstrated these receptors are also capable of forming heterodimers in specific patterns. These mGluR heterodimers appear to have their own unique biophysical behavior and pharmacology with both native and synthetic compounds with few rules having been identified that allow for prediction of the consequences of any particular mGluR pair forming heterodimers. Here, we review the relevant literature demonstrating the existence and consequences of mGluR heterodimerization. By collecting biophysical and pharmacological data of several mGluR heterodimers we demonstrate the lack of generalizable behavior of these complexes indicating that each individual dimeric pair needs to be investigated independently. Additionally, by combining sequence alignment and structural analysis, we propose that interactions between the β4-A Helix Loop and the D Helix in the extracellular domain of these receptors are the structural components that dictate heterodimerization compatibility. Finally, we discuss the potential implications of mGluR heterodimerization from the viewpoints of further developing our understanding of neuronal physiology and leveraging mGluRs as a therapeutic target for the treatment of pathophysiology.
Collapse
Affiliation(s)
- Tyler W McCullock
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave, Box 711, Rochester, NY, 14642, USA.
| | - Paul J Kammermeier
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave, Box 711, Rochester, NY, 14642, USA.
| |
Collapse
|
4
|
An engineered channelrhodopsin optimized for axon terminal activation and circuit mapping. Commun Biol 2021; 4:461. [PMID: 33846537 PMCID: PMC8042110 DOI: 10.1038/s42003-021-01977-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 03/11/2021] [Indexed: 11/09/2022] Open
Abstract
Optogenetic tools such as channelrhodopsin-2 (ChR2) enable the manipulation and mapping of neural circuits. However, ChR2 variants selectively transported down a neuron’s long-range axonal projections for precise presynaptic activation remain lacking. As a result, ChR2 activation is often contaminated by the spurious activation of en passant fibers that compromise the accurate interpretation of functional effects. Here, we explored the engineering of a ChR2 variant specifically localized to presynaptic axon terminals. The metabotropic glutamate receptor 2 (mGluR2) C-terminal domain fused with a proteolytic motif and axon-targeting signal (mGluR2-PA tag) localized ChR2-YFP at axon terminals without disturbing normal transmission. mGluR2-PA-tagged ChR2 evoked transmitter release in distal projection areas enabling lower levels of photostimulation. Circuit connectivity mapping in vivo with the Spike Collision Test revealed that mGluR2-PA-tagged ChR2 is useful for identifying axonal projection with significant reduction in the polysynaptic excess noise. These results suggest that the mGluR2-PA tag helps actuate trafficking to the axon terminal, thereby providing abundant possibilities for optogenetic experiments. Hamada et al. engineer and utilise a channelrhodopsin-2 variant that is localized to presynaptic axon terminals. They demonstrate its use for circuitry mapping in vivo and thus provide a useful tool for future optogenetic experiments
Collapse
|
5
|
Rai D, Akagi T, Shimohata A, Ishii T, Gangi M, Maruyama T, Wada-Kiyama Y, Ogiwara I, Kaneda M. Involvement of the C-terminal domain in cell surface localization and G-protein coupling of mGluR6. J Neurochem 2020; 158:837-848. [PMID: 33067823 DOI: 10.1111/jnc.15217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 05/25/2020] [Accepted: 10/11/2020] [Indexed: 01/05/2023]
Abstract
Metabotropic glutamate receptor 6, mGluR6, interacts with scaffold proteins and Gβγ subunits via its intracellular C-terminal domain (CTD). The mGluR6 pathway is critically involved in the retinal processing of visual signals. We herein investigated whether the CTD (residues 840-871) was necessary for mGluR6 cell surface localization and G-protein coupling using mGluR6-CTD mutants with immunocytochemistry, surface biotinylation assays, and electrophysiological approaches. We used 293T cells and primary hippocampal neurons as model systems. We examined C-terminally truncated mGluR6 and showed that the removal of up to residue 858 did not affect surface localization or glutamate-induced G-protein-mediated responses, whereas a 15-amino acid deletion (Δ857-871) impaired these functions. However, a 21-amino acid deletion (Δ851-871) restored surface localization and glutamate-dependent responses, which were again attenuated when the entire CTD was removed. The sequence alignment of group III mGluRs showed conserved amino acids resembling an ER retention motif in the CTD. These results suggest that the intracellular CTD is required for the cell surface transportation and receptor function of mGluR6, whereas it may contain regulatory elements for intracellular trafficking and signaling.
Collapse
Affiliation(s)
- Dilip Rai
- Department of Physiology, Nippon Medical School, Tokyo, Japan
| | - Takumi Akagi
- Department of Physiology, Nippon Medical School, Tokyo, Japan
| | | | - Toshiyuki Ishii
- Department of Physiology, Nippon Medical School, Tokyo, Japan
| | - Mie Gangi
- Department of Physiology, Nippon Medical School, Tokyo, Japan
| | - Takuma Maruyama
- Department of Physiology, Nippon Medical School, Tokyo, Japan
| | | | - Ikuo Ogiwara
- Department of Physiology, Nippon Medical School, Tokyo, Japan
| | | |
Collapse
|
6
|
Of Molecules and Mechanisms. J Neurosci 2019; 40:81-88. [PMID: 31630114 DOI: 10.1523/jneurosci.0743-19.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 08/25/2019] [Accepted: 08/31/2019] [Indexed: 11/21/2022] Open
Abstract
Without question, molecular biology drives modern neuroscience. The past 50 years has been nothing short of revolutionary as key findings have moved the field from correlation toward causation. Most obvious are the discoveries and strategies that have been used to build tools for visualizing circuits, measuring activity, and regulating behavior. Less flashy, but arguably as important are the myriad investigations uncovering the actions of single molecules, macromolecular structures, and integrated machines that serve as the basis for constructing cellular and signaling pathways identified in wide-scale gene or RNA studies and for feeding data into informational networks used in systems biology. This review follows the pathways that were opened in neuroscience by major discoveries and set the stage for the next 50 years.
Collapse
|
7
|
Ha S, Baver S, Huo L, Gata A, Hairston J, Huntoon N, Li W, Zhang T, Benecchi EJ, Ericsson M, Hentges ST, Bjørbæk C. Somato-dendritic localization and signaling by leptin receptors in hypothalamic POMC and AgRP neurons. PLoS One 2013; 8:e77622. [PMID: 24204898 PMCID: PMC3812230 DOI: 10.1371/journal.pone.0077622] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 09/13/2013] [Indexed: 11/18/2022] Open
Abstract
Leptin acts via neuronal leptin receptors to control energy balance. Hypothalamic pro-opiomelanocortin (POMC) and agouti-related peptide (AgRP)/Neuropeptide Y (NPY)/GABA neurons produce anorexigenic and orexigenic neuropeptides and neurotransmitters, and express the long signaling form of the leptin receptor (LepRb). Despite progress in the understanding of LepRb signaling and function, the sub-cellular localization of LepRb in target neurons has not been determined, primarily due to lack of sensitive anti-LepRb antibodies. Here we applied light microscopy (LM), confocal-laser scanning microscopy (CLSM), and electron microscopy (EM) to investigate LepRb localization and signaling in mice expressing a HA-tagged LepRb selectively in POMC or AgRP/NPY/GABA neurons. We report that LepRb receptors exhibit a somato-dendritic expression pattern. We further show that LepRb activates STAT3 phosphorylation in neuronal fibers within several hypothalamic and hindbrain nuclei of wild-type mice and rats, and specifically in dendrites of arcuate POMC and AgRP/NPY/GABA neurons of Leprb+/+ mice and in Leprbdb/db mice expressing HA-LepRb in a neuron specific manner. We did not find evidence of LepRb localization or STAT3-signaling in axon-fibers or nerve-terminals of POMC and AgRP/NPY/GABA neurons. Three-dimensional serial EM-reconstruction of dendritic segments from POMC and AgRP/NPY/GABA neurons indicates a high density of shaft synapses. In addition, we found that the leptin activates STAT3 signaling in proximity to synapses on POMC and AgRP/NPY/GABA dendritic shafts. Taken together, these data suggest that the signaling-form of the leptin receptor exhibits a somato-dendritic expression pattern in POMC and AgRP/NPY/GABA neurons. Dendritic LepRb signaling may therefore play an important role in leptin’s central effects on energy balance, possibly through modulation of synaptic activity via post-synaptic mechanisms.
Collapse
Affiliation(s)
- Sangdeuk Ha
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Scott Baver
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lihong Huo
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Adriana Gata
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Joyce Hairston
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Nicholas Huntoon
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Wenjing Li
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Thompson Zhang
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Elizabeth J. Benecchi
- Electron Microscopy Facility, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Maria Ericsson
- Electron Microscopy Facility, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Shane T. Hentges
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Christian Bjørbæk
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail :
| |
Collapse
|
8
|
Garzón M, Pickel VM. Somatodendritic targeting of M5 muscarinic receptor in the rat ventral tegmental area: implications for mesolimbic dopamine transmission. J Comp Neurol 2013; 521:2927-46. [PMID: 23504804 PMCID: PMC4038040 DOI: 10.1002/cne.23323] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 01/29/2013] [Accepted: 02/26/2013] [Indexed: 11/10/2022]
Abstract
Muscarinic modulation of mesolimbic dopaminergic neurons in the ventral tegmental area (VTA) plays an important role in reward, potentially mediated through the M5 muscarinic acetylcholine receptor (M5R). However, the key sites for M5R-mediated control of dopamine neurons within this region are still unknown. To address this question we examined the electron microscopic immunocytochemical localization of antipeptide antisera against M5R and the plasmalemmal dopamine transporter (DAT) in single sections through the rat VTA. M5R was located mainly to VTA somatodendritic profiles (71%; n = 627), at least one-third (33.2%; n = 208) of which also contained DAT. The M5R immunoreactivity was distributed along cytoplasmic tubulovesicular endomembrane systems in somata and large dendrites, but was more often located at plasmalemmal sites in small dendrites, the majority of which did not express DAT. The M5R-immunoreactive dendrites received a balanced input from unlabeled terminals forming either asymmetric or symmetric synapses. Compared with dendrites, M5R was less often seen in axon terminals, comprising only 10.8% (n = 102) of the total M5R-labeled profiles. These terminals were usually presynaptic to unlabeled dendrites, suggesting that M5R activation can indirectly modulate non-DAT-containing dendrites through presynaptic mechanisms. Our results provide the first ultrastructural evidence that in the VTA, M5R has a subcellular location conducive to major involvement in postsynaptic signaling in many dendrites, only some of which express DAT. These findings suggest that cognitive and rewarding effects ascribed to muscarinic activation in the VTA can primarily be credited to M5R activation at postsynaptic plasma membranes distinct from dopamine transport.
Collapse
Affiliation(s)
- Miguel Garzón
- Department of Anatomy, Histology, and Neuroscience, Medical School, Universidad Autónoma de Madrid (UAM), Madrid, 28029, Spain.
| | | |
Collapse
|
9
|
Ma M, Shofer FS, Neumar RW. Calpastatin overexpression protects axonal transport in an in vivo model of traumatic axonal injury. J Neurotrauma 2012; 29:2555-63. [PMID: 22776025 DOI: 10.1089/neu.2012.2473] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Traumatic brain injury (TBI) causes substantial morbidity and mortality worldwide. A key component of both mild and severe TBI is diffuse axonal injury. Except in cases of extreme mechanical strain, when axons are torn at the moment of trauma, axonal stretch injury is characterized by early cytoskeletal proteolysis, transport disruption, and secondary axotomy. Calpains, a family of Ca(2+)-dependent proteases, have been implicated in this pathologic cascade, but direct in vivo evidence is lacking. To test the hypothesis that calpains play a causal role in axonal stretch injury in vivo, we used our rat optic nerve stretch model following adeno-associated viral (AAV) vector-mediated overexpression of the endogenous calpain inhibitor calpastatin in optic nerve axons. AAV vectors were designed for optimal expression of human calpastatin (hCAST) in retinal ganglion cells (RGCs). Calpain inhibition by the expressed protein was then confirmed in primary cortical cultures. Finally, we performed bilateral intravitreal injections of AAV vectors expressing hCAST or the reporter protein ZsGreen 3 weeks prior to unilateral optic nerve stretch. Immediately after stretch injury, Fluoro-Gold was injected into the superior colliculi for assessment of retrograde axonal transport. Rats were euthanized 4 days after stretch injury. Both hCAST and ZsGreen were detected in axons throughout the optic nerve to the chiasm. Calpastatin overexpression partially preserved axonal transport after stretch injury (58.3±15.6% reduction in Fluoro-Gold labeling relative to uninjured contralateral controls in ZsGreen-expressing RGCs, versus 33.8±23.9% in hCAST-expressing RGCs; p=0.038). These results provide direct evidence that axonal calpains play a causal role in transport disruption after in vivo stretch injury.
Collapse
Affiliation(s)
- Marek Ma
- Department of Emergency Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | | | | |
Collapse
|
10
|
Differential trafficking of transport vesicles contributes to the localization of dendritic proteins. Cell Rep 2012; 2:89-100. [PMID: 22840400 DOI: 10.1016/j.celrep.2012.05.018] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 02/04/2012] [Accepted: 05/22/2012] [Indexed: 11/21/2022] Open
Abstract
In neurons, transmembrane proteins are targeted to dendrites in vesicles that traffic solely within the somatodendritic compartment. How these vesicles are retained within the somatodendritic domain is unknown. Here, we use a novel pulse-chase system, which allows synchronous release of exogenous transmembrane proteins from the endoplasmic reticulum to follow movements of post-Golgi transport vesicles. Surprisingly, we found that post-Golgi vesicles carrying dendritic proteins were equally likely to enter axons and dendrites. However, once such vesicles entered the axon, they very rarely moved beyond the axon initial segment but instead either halted or reversed direction in an actin and Myosin Va-dependent manner. In contrast, vesicles carrying either an axonal or a nonspecifically localized protein only rarely halted or reversed and instead generally proceeded to the distal axon. Thus, our results are consistent with the axon initial segment behaving as a vesicle filter that mediates the differential trafficking of transport vesicles.
Collapse
|
11
|
Seabold GK, Wang PY, Petralia RS, Chang K, Zhou A, McDermott MI, Wang YX, Milgram SL, Wenthold RJ. Dileucine and PDZ-binding motifs mediate synaptic adhesion-like molecule 1 (SALM1) trafficking in hippocampal neurons. J Biol Chem 2012; 287:4470-84. [PMID: 22174418 PMCID: PMC3281672 DOI: 10.1074/jbc.m111.279661] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 11/22/2011] [Indexed: 12/18/2022] Open
Abstract
Synaptic adhesion-like molecules (SALMs) are a family of cell adhesion molecules involved in neurite outgrowth and synapse formation. Of the five family members, only SALM1, -2, and -3 contain a cytoplasmic C-terminal PDZ-binding motif. We have found that SALM1 is unique among the SALMs because deletion of its PDZ-binding motif (SALM1ΔPDZ) blocks its surface expression in heterologous cells. When expressed in hippocampal neurons, SALM1ΔPDZ had decreased surface expression in dendrites and the cell soma but not in axons, suggesting that the PDZ-binding domain may influence cellular trafficking of SALMs to specific neuronal locations. Endoglycosidase H digestion assays indicated that SALM1ΔPDZ is retained in the endoplasmic reticulum (ER) in heterologous cells. However, when the entire C-terminal tail of SALM1 was deleted, SALM1 was detected on the cell surface. Using serial deletions, we identified a region of SALM1 that contains a putative dileucine ER retention motif, which is not present in the other SALMs. Mutation of this DXXXLL motif allowed SALM1 to leave the ER and enhanced its surface expression in heterologous cells and neurons. An increase in the number of protrusions at the dendrites and cell body was observed when this SALM1 mutant was expressed in hippocampal neurons. With electron microscopy, these protrusions appeared to be irregular, enlarged spines and filopodia. Thus, enrichment of SALM1 on the cell surface affects dendritic arborization, and intracellular motifs regulate its dendritic versus axonal localization.
Collapse
Affiliation(s)
- Gail K Seabold
- Laboratory of Neurochemistry, NIDCD/National Institutes of Health, 50 South Dr., Bldg. 50, Rm. 4144, Bethesda, MD20892-8027, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
van den Berg R, Hoogenraad CC. Molecular motors in cargo trafficking and synapse assembly. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 970:173-96. [PMID: 22351056 DOI: 10.1007/978-3-7091-0932-8_8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Every production process, be it cellular or industrial, depends on a constant supply of energy and resources. Synapses, specialized junctions in the central nervous system through which neurons signal to each other, are no exception to this rule. In order to form new synapses and alter the strength of synaptic transmission, neurons need a regulatory mechanism to deliver and remove synaptic proteins at synaptic sites. Neurons make use of active transport driven by molecular motor proteins to move synaptic cargo over either microtubules (kinesin, dynein) or actin filaments (myosin) to their specific site of action. These mechanisms are crucial for the initial establishment of synaptic specializations during synaptogenesis and for activity-dependent changes in synaptic strength during plasticity. In this chapter, we address the organization of the neuronal cytoskeleton, focus on synaptic cargo transport activities that operate in axons and dendrites, and discuss the spatial and temporal regulation of motor protein-based transport.
Collapse
Affiliation(s)
- Robert van den Berg
- Cell Biology, Utrecht University, Padualaan 8, 3584CH, Utrecht, The Netherlands
| | | |
Collapse
|
13
|
Shen Y, Lindemeyer AK, Spigelman I, Sieghart W, Olsen RW, Liang J. Plasticity of GABAA receptors after ethanol pre-exposure in cultured hippocampal neurons. Mol Pharmacol 2010; 79:432-42. [PMID: 21163967 DOI: 10.1124/mol.110.068650] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Alcohol use causes many physiological changes in brain with behavioral sequelae. We previously observed (J Neurosci 27:12367-12377, 2007) plastic changes in hippocampal slice recordings paralleling behavioral changes in rats treated with a single intoxicating dose of ethanol (EtOH). Here, we were able to reproduce in primary cultured hippocampal neurons many of the effects of in vivo EtOH exposure on GABA(A) receptors (GABA(A)Rs). Cells grown 11 to 15 days in vitro demonstrated GABA(A)R δ subunit expression and sensitivity to enhancement by short-term exposure to EtOH (60 mM) of GABA(A)R-mediated tonic current (I(tonic)) using whole-cell patch-clamp techniques. EtOH gave virtually no enhancement of mIPSCs. Cells pre-exposed to EtOH (60 mM) for 30 min showed, 1 h after EtOH withdrawal, a 50% decrease in basal I(tonic) magnitude and tolerance to short-term EtOH enhancement of I(tonic), followed by reduced basal mIPSC area at 4 h. At 24 h, we saw considerable recovery in mIPSC area and significant potentiation by short-term EtOH; in addition, GABA(A)R currents exhibited reduced enhancement by benzodiazepines. These changes paralleled significant decreases in cell-surface expression of normally extrasynaptic δ and α4 GABA(A)R subunits as early as 20 min after EtOH exposure and reduced α5-containing GABA(A)Rs at 1 h, followed by a larger reduction of normally synaptic α1 subunit at 4 h, and then by increases in α4γ2-containing cell-surface receptors by 24 h. Measuring internalization of biotinylated GABA(A)Rs, we showed for the first time that the EtOH-induced loss of I(tonic) and cell-surface δ/α4 20 min after withdrawal results from increased receptor endocytosis rather than decreased exocytosis.
Collapse
Affiliation(s)
- Yi Shen
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095-1735, USA
| | | | | | | | | | | |
Collapse
|
14
|
Kapitein LC, Hoogenraad CC. Which way to go? Cytoskeletal organization and polarized transport in neurons. Mol Cell Neurosci 2010; 46:9-20. [PMID: 20817096 DOI: 10.1016/j.mcn.2010.08.015] [Citation(s) in RCA: 163] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Accepted: 08/25/2010] [Indexed: 01/01/2023] Open
Abstract
To establish and maintain their polarized morphology, neurons employ active transport driven by cytoskeletal motor proteins to sort cargo between axons and dendrites. These motors can move in a specific direction over either microtubules (kinesins, dynein) or actin filaments (myosins). The basic traffic rules governing polarized transport on the neuronal cytoskeleton have long remained unclear, but recent work has revealed several fundamental sorting principles based on differences in the cytoskeletal organization in axons versus dendrites. We will highlight the basic characteristics of the neuronal cytoskeleton and review existing evidence for microtubule and actin based traffic rules in polarized neuronal transport. We will propose a model in which polarized sorting of cargo is established by recruiting or activating the proper subset of motor proteins, which are subsequently guided to specific directions by the polarized organization of the neuronal cytoskeleton.
Collapse
Affiliation(s)
- Lukas C Kapitein
- Department of Neuroscience, Erasmus Medical Center, Dr. Molewaterplein 50, 3015 GE, Rotterdam, The Netherlands
| | | |
Collapse
|
15
|
Kurisu J, Fukuda T, Yokoyama S, Hirano T, Kengaku M. Polarized targeting of DNER into dendritic plasma membrane in hippocampal neurons depends on endocytosis. J Neurochem 2010; 113:1598-610. [PMID: 20367751 DOI: 10.1111/j.1471-4159.2010.06714.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The targeting of membrane proteins into axons and dendrites is of critical importance for directional signal transmission within specific neural circuits. Many dendritic proteins have been shown to reach the somatodendritic membrane based on selective sorting and transport of carrier vesicles. Using rat hippocampal neurons in culture, we investigated the trafficking pathways of Delta/Notch-like EGF-related receptor (DNER), a transmembrane Notch ligand which is specifically expressed in CNS dendrites. Mutations in the cytoplasmic domain of DNER that abolished somatodendritic localization also increased its surface expression. Furthermore, inhibition of endocytosis resulted in disruption of the somatodendritic localization of DNER, indicating that the somatodendritic targeting of DNER is dependent on endocytosis. The DNER cytoplasmic domain binds to a clathrin adaptor protein complex-2 via a proximal tyrosine motif and a 40 amino acid stretch in the mid-domain, but not by the C-terminal tail. Molecular and pharmacological inhibition revealed that the surface expression of DNER is regulated by clathrin-dependent and -independent endocytosis. In contrast, the somatodendritic targeting of DNER is predominantly regulated by clathrin- and adaptor protein complex-2-independent endocytosis via the C-terminal tail of DNER. Our data suggest that clathrin-independent endocytosis is critical for the polarized targeting of somatodendritic proteins.
Collapse
Affiliation(s)
- Junko Kurisu
- Laboratory for Neural Cell Polarity, RIKEN Brain Science Institute, Wako, Saitama, Japan
| | | | | | | | | |
Collapse
|
16
|
Ishii M, Morigiwa K, Takao M, Nakanishi S, Fukuda Y, Mimura O, Tsukamoto Y. Ectopic synaptic ribbons in dendrites of mouse retinal ON- and OFF-bipolar cells. Cell Tissue Res 2009; 338:355-75. [PMID: 19859741 PMCID: PMC2779389 DOI: 10.1007/s00441-009-0880-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Accepted: 09/08/2009] [Indexed: 11/12/2022]
Abstract
The ectopic distribution of synaptic ribbons in dendrites of mouse retinal bipolar cells was examined by using genetic ablation of metabotropic glutamate receptor subtype 6 (mGluR6), electron microscopy, and immunocytochemistry. Ectopic ribbons were observed in dendrites of rod and ON-cone bipolar cells in the mGluR6-deficient mouse but not in those of wild-type mice. The number of rod spherules facing the ectopic ribbons in mGluR6-deficient rod bipolar dendrites increased gradually during early growth and reached a plateau level of about 20% at 12 weeks. These ectopic ribbons were immunopositive for RIBEYE, a ribbon-specific protein, but the associated vesicles were immunonegative for synaptophysin, a synaptic-vesicle-specific protein. The presence of ectopic ribbons was correlated with an increase in the roundness of the invaginating dendrites of the rod bipolar cells. We further confirmed ectopic ribbons in dendrites of OFF-cone bipolar cells in wild-type retinas. Of the four types of OFF-cone bipolar cells (T1-T4), only the T2-type, which had a greater number of synaptic ribbons at the axon terminal and a thicker axon cylinder than the other types, had ectopic ribbons. Light-adapted experiments revealed that, in wild-type mice under enhanced-light adaptation (considered similar to the mGluR6-deficient state), the roundness in the invaginating dendrites and axon terminals of rod bipolar cells increased, but no ectopic ribbons were detected. Based on these findings and known mechanisms for neurotransmitter release and protein trafficking, the possible mechanisms underlying the ectopic ribbons are discussed on the basis of intracellular transport for the replenishment of synaptic proteins.
Collapse
Affiliation(s)
- Masaaki Ishii
- Department of Biology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501 Japan
- Department of Ophthalmology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501 Japan
| | | | - Motoharu Takao
- Department of Human and Information Science, Tokai University, Hiratsuka, Kanagawa 259-1292 Japan
| | | | - Yutaka Fukuda
- Department of Physiology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871 Japan
| | - Osamu Mimura
- Department of Ophthalmology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501 Japan
| | - Yoshihiko Tsukamoto
- Department of Biology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501 Japan
| |
Collapse
|
17
|
Bovolin P, Bovetti S, Fasolo A, Katarova Z, Szabo G, Shipley MT, Margolis FL, Puche AC. Developmental regulation of metabotropic glutamate receptor 1 splice variants in olfactory bulb mitral cells. J Neurosci Res 2009; 87:369-79. [PMID: 18816797 DOI: 10.1002/jnr.21864] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Alternative splicing of the metabotropic glutamate receptor 1 (mGluR1) receptor gene generates two major receptor isoforms, mGluR1a and mGluR1b, differing in intracellular function and distribution. However, little is known on the expression profiles of these variants during development. We examined the mRNA expression profile of mGluR1a/b in microdissected layers and acutely isolated mitral cells in the developing mouse olfactory bulb. This analysis showed that the two mGluR1 variants are differentially regulated within each bulb layer. During the first postnatal week, the mGluR1a isoform replaces GluR1b in the microdissected mitral cell layer (MCL) and in isolated identified mitral cells, coinciding with a developmental epoch of mitral cell dendritic reorganization. Although mGluR1a mRNA is expressed at high levels in both the adult external plexiform layer (EPL) and MCL, Western blotting analysis reveals a marked reduction of the mGluR1a protein in the MCL, where mitral cell bodies are located, and strong labeling in the EPL, which contains mitral cell dendrites. This suggests that there is increased dendritic trafficking efficiency of the receptor in adult. The temporal and spatial shift in mGluR1b/a expression suggests distinct roles of the mGluR1 isoforms, with mGluR1b potentially involved in the early mitral cell maturation and mGluR1a in dendritic and synapse function.
Collapse
Affiliation(s)
- P Bovolin
- Department of Animal and Human Biology, University of Turin, Turin, Italy
| | | | | | | | | | | | | | | |
Collapse
|
18
|
A Selective Filter for Cytoplasmic Transport at the Axon Initial Segment. Cell 2009; 136:1148-60. [DOI: 10.1016/j.cell.2009.01.016] [Citation(s) in RCA: 259] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2008] [Revised: 11/15/2008] [Accepted: 01/05/2009] [Indexed: 11/22/2022]
|
19
|
Allen J, Chilton JK. The specific targeting of guidance receptors within neurons: who directs the directors? Dev Biol 2008; 327:4-11. [PMID: 19121301 DOI: 10.1016/j.ydbio.2008.12.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2008] [Revised: 11/27/2008] [Accepted: 12/09/2008] [Indexed: 02/04/2023]
Abstract
Guidance molecules present in both axonal and dendritic growth cones mediate neuronal responses to extracellular cues thereby ensuring correct neurite pathfinding and development of the nervous system. Little is known though about the mechanisms employed by neurons to deliver these receptors, specifically and efficiently, to the extending growth cone. A deeper understanding of this process is crucial if guidance receptors are to be manipulated to promote nervous system repair. Studies in other polarised cells, notably epithelial, have elucidated fundamental routes to the intracellular segregation of molecules mediated by endosomal pathways. Due to their extreme complexity and specialisation, neurons appear to have built upon these generic systems to evolve sophisticated trafficking networks. A striking feature is the axon initial segment which acts like a valve to tightly regulate the flux of molecules both entering and leaving the axon. Once in the growth cone, further controls operate to enhance the retention or rejection, as appropriate, of membrane receptors. We discuss the current state of knowledge regarding the intracellular trafficking of axon guidance receptors and how this relates to their developmental roles. We highlight the various facets still to be properly elucidated and by building on existing data regarding neuronal polarity and intracellular sorting mechanisms suggest ways to fill these gaps.
Collapse
Affiliation(s)
- James Allen
- Institute of Biomedical and Clinical Science, Peninsula Medical School, Research Way, Plymouth PL6 8BU, UK
| | | |
Collapse
|
20
|
Yang JSJ, Bai JM, Lee T. Dynein-dynactin complex is essential for dendritic restriction of TM1-containing Drosophila Dscam. PLoS One 2008; 3:e3504. [PMID: 18946501 PMCID: PMC2566808 DOI: 10.1371/journal.pone.0003504] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2008] [Accepted: 09/29/2008] [Indexed: 01/14/2023] Open
Abstract
Background Many membrane proteins, including Drosophila Dscam, are enriched in dendrites or axons within neurons. However, little is known about how the differential distribution is established and maintained. Methodology/Principal Findings Here we investigated the mechanisms underlying the dendritic targeting of Dscam[TM1]. Through forward genetic mosaic screens and by silencing specific genes via targeted RNAi, we found that several genes, encoding various components of the dynein-dynactin complex, are required for restricting Dscam[TM1] to the mushroom body dendrites. In contrast, compromising dynein/dynactin function did not affect dendritic targeting of two other dendritic markers, Nod and Rdl. Tracing newly synthesized Dscam[TM1] further revealed that compromising dynein/dynactin function did not affect the initial dendritic targeting of Dscam[TM1], but disrupted the maintenance of its restriction to dendrites. Conclusions/Significance The results of this study suggest multiple mechanisms of dendritic protein targeting. Notably, dynein-dynactin plays a role in excluding dendritic Dscam, but not Rdl, from axons by retrograde transport.
Collapse
Affiliation(s)
- Jacob Shun-Jen Yang
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Jia-Min Bai
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Tzumin Lee
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
21
|
The axon-dendrite targeting of Kv3 (Shaw) channels is determined by a targeting motif that associates with the T1 domain and ankyrin G. J Neurosci 2008; 27:14158-70. [PMID: 18094255 DOI: 10.1523/jneurosci.3675-07.2007] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Kv3 (Shaw) channels regulate rapid spiking, transmitter release and dendritic integration of many central neurons. Crucial to functional diversity are the complex targeting patterns of channel proteins. However, the targeting mechanisms are not known. Here we report that the axon-dendrite targeting of Kv3.1 is controlled by a conditional interaction of a C-terminal axonal targeting motif (ATM) with the N-terminal T1 domain and adaptor protein ankyrin G. In cultured hippocampal neurons, although the two splice variants of Kv3.1, Kv3.1a and Kv3.1b, are differentially targeted to the somatodendritic and axonal membrane, respectively, the lysine-rich ATM is surprisingly common for both splice variants. The ATM not only directly binds to the T1 domain in a Zn2+-dependent manner, but also associates with the ankyrin-repeat domain of ankyrin G. However, the full-length channel proteins of Kv3.1b display stronger association to ankyrin G than those of Kv3.1a, suggesting that the unique splice domain at Kv3.1b C terminus influences ATM binding to T1 and ankyrin G. Because ankyrin G mainly resides at the axon initial segment, we propose that it may function as a barrier for axon-dendrite targeting of Kv3.1 channels. In support of this idea, disrupting ankyrin G function either by over-expressing a dominant-negative mutant or by siRNA knockdown decreases polarized axon-dendrite targeting of both Kv3.1a and Kv3.1b. We conclude that the conditional ATM masked by the T1 domain in Kv3.1a is exposed by the splice domain in Kv3.1b, and is subsequently recognized by ankyrin G to target Kv3.1b into the axon.
Collapse
|
22
|
Armsen W, Himmel B, Betz H, Eulenburg V. The C-terminal PDZ-ligand motif of the neuronal glycine transporter GlyT2 is required for efficient synaptic localization. Mol Cell Neurosci 2007; 36:369-80. [PMID: 17851090 DOI: 10.1016/j.mcn.2007.07.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Revised: 07/13/2007] [Accepted: 07/24/2007] [Indexed: 11/28/2022] Open
Abstract
The neuronal glycine transporter 2 (GlyT2) belongs to the large SLC6 family of Na+/Cl--dependent neurotransmitter transporters. At its extreme C-terminus, GlyT2 carries a type III PDZ domain binding motif (PDZ-ligand motif), which interacts with the PDZ domain protein syntenin-1. Here, we investigated the physiological role of the GlyT2 PDZ-ligand motif by a loss-of-function approach. Inactivation of the PDZ-ligand motif did not impair the localization, glycosylation and transport function of recombinant GlyT2 expressed in HEK293T cells. However, in transfected hippocampal neurons, the synaptic localization of GlyT2 was significantly reduced upon PDZ-ligand motif inactivation. Co-localization of GlyT2 with marker proteins of excitatory and inhibitory synapses was decreased by down to 50% upon PDZ-ligand motif deletion as compared to the wild-type protein. These data indicate that the C-terminal PDZ-ligand motif of GlyT2 plays an important role in transporter trafficking to and/or stabilization at synaptic sites.
Collapse
Affiliation(s)
- Wencke Armsen
- Department of Neurochemistry, Max-Planck Institute for Brain Research, Deutschordenstrasse 46, 60528 Frankfurt am Main, Germany
| | | | | | | |
Collapse
|
23
|
Tsukamoto Y, Morigiwa K, Ishii M, Takao M, Iwatsuki K, Nakanishi S, Fukuda Y. A novel connection between rods and ON cone bipolar cells revealed by ectopic metabotropic glutamate receptor 7 (mGluR7) in mGluR6-deficient mouse retinas. J Neurosci 2007; 27:6261-7. [PMID: 17553999 PMCID: PMC6672139 DOI: 10.1523/jneurosci.5646-06.2007] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Since the discovery of direct chemical synapses between rod photoreceptor and OFF cone bipolar cells in mouse retinas, whether the ON cone bipolar cell also receive direct chemical input from rod has been a pending question. In finding that metabotropic glutamate receptor 7 (mGluR7) was uniquely expressed in dendrites of ON cone bipolar cells in the mGluR6-deficient mouse retina, we used this ectopic mGluR7 immunoreactivity as a specific marker for the ON cone bipolar to search for its rod connection. Here, we show that a certain type of ON cone bipolar cell forms ribbon-associated synapses not only with cones, but also rods. This finding was verified in the wild-type mouse retina by three-dimensional reconstruction of bipolar cells from serial electron micrographs. These ON cone bipolars were further identified as corresponding to type 7 of mouse bipolar cell described by Ghosh et al. (2004) and also to the green fluorescent protein (GFP)-labeled type 7 bipolars in the alpha-gustducin-GFP transgenic mouse. Our findings suggest that, in mice, rod signals bifurcate into a third ON and OFF pathway in addition to the two known routes to cone bipolar cells: (1) via rod chemical synapse --> rod bipolar --> AII amacrine --> ON and OFF cone bipolar cells; (2) via rod-cone gap junction --> cone chemical synapse --> ON and OFF cone bipolar cells; and (3) via rod chemical synapse --> ON and OFF cone bipolar cells. This third novel pathway is thought to transmit fast and moderately light-sensitive rod signals, functioning to smooth out the intensity changes at the scotopic-mesopic interface.
Collapse
Affiliation(s)
- Yoshihiko Tsukamoto
- Department of Biology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan.
| | | | | | | | | | | | | |
Collapse
|
24
|
Nomura S, Fukaya M, Tsujioka T, Wu D, Watanabe M. Phospholipase Cβ3 is distributed in both somatodendritic and axonal compartments and localized around perisynapse and smooth endoplasmic reticulum in mouse Purkinje cell subsets. Eur J Neurosci 2007; 25:659-72. [PMID: 17298601 DOI: 10.1111/j.1460-9568.2007.05334.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Phospholipase Cbeta3 (PLCbeta3) and PLCbeta4 are the two major isoforms in cerebellar Purkinje cells (PCs), displaying reciprocal expression across the cerebellum. Here, we examined subcellular distribution of PLCbeta3 in the mouse cerebellum by producing specific antibody. PLCbeta3 was detected as a particulate pattern of immunostaining in various PC elements. Like PLCbeta4, PLCbeta3 was richly distributed in somatodendritic compartments, where it was colocalized with molecules constituting the metabotropic glutamate receptor (mGluR1) signalling pathway, i.e. mGluR1alpha, G alpha q/G alpha 11 subunits of G q protein, inositol 1,4,5-trisphosphate receptor IP3R1, Homer1, protein kinase C PKCgamma, and diacylglycerol lipase DAGLalpha. Unlike PLCbeta4, PLCbeta3 was also distributed at low to moderate levels in PC axons, which were intense for IP3R1 and PKCgamma, low for G alpha q/G alpha 11, and negative for mGluR1alpha, Homer1, and DAGLalpha. By immunoelectron microscopy, PLCbeta3 was preferentially localized around the smooth endoplasmic reticulum in spines, dendrites, and axons of PCs, and also accumulated at the perisynapse of parallel fibre-PC synapses. Consistent with the ultrastructural localization, PLCbeta3 was biochemically enriched in the microsomal and postsynaptic density fractions. These results suggest that PLCbeta3 plays a major role in mediating mGluR1-dependent synaptic transmission, plasticity, and integration in PLCbeta3-dominant PCs, through eliciting Ca2+ release, protein phosphorylation, and endocannabinoid production at local somatodendritic compartments. Because PLCbeta3 can be activated by G betagamma subunits liberated from Gi/o and Gs proteins as well, axonal PLCbeta3 seems to modulate the conduction of action potentials through mediating local Ca2+ release and protein phosphorylation upon activation of a variety of G protein-coupled receptors other than mGluR1.
Collapse
Affiliation(s)
- Sachi Nomura
- Department of Anatomy, Hokkaido University School of Medicine, Sapporo 060-8638, Japan
| | | | | | | | | |
Collapse
|
25
|
Xu W, Wong TP, Chery N, Gaertner T, Wang YT, Baudry M. Calpain-Mediated mGluR1α Truncation: A Key Step in Excitotoxicity. Neuron 2007; 53:399-412. [PMID: 17270736 DOI: 10.1016/j.neuron.2006.12.020] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2006] [Revised: 10/31/2006] [Accepted: 12/05/2006] [Indexed: 10/23/2022]
Abstract
Excitotoxicity mediated by glutamate receptors plays crucial roles in ischemia and other neurodegenerative diseases. Whereas overactivation of ionotropic glutamate receptors is neurotoxic, the role of metabotropic glutamate receptors (mGluRs), and especially mGluR1, remains equivocal. Here we report that activation of NMDA receptors results in calpain-mediated truncation of the C-terminal domain of mGluR1alpha at Ser(936). The truncated mGluR1alpha maintains its ability to increase cytosolic calcium while it no longer activates the neuroprotective PI(3)K-Akt signaling pathways. Full-length and truncated forms of mGluR1alpha play distinct roles in excitotoxic neuronal degeneration in cultured neurons. A fusion peptide derived from the calpain cleavage site of mGluR1alpha efficiently blocks NMDA-induced truncation of mGluR1alpha in primary neuronal cultures and exhibits neuroprotection against excitotoxicity both in vitro and in vivo. These findings shed light on the relationship between NMDA and mGluR1alpha and indicate the existence of a positive feedback regulation in excitotoxicity involving calpain and mGluR1alpha.
Collapse
Affiliation(s)
- Wei Xu
- Neuroscience Program, University of Southern California, Los Angeles, CA 90089-2520, USA
| | | | | | | | | | | |
Collapse
|
26
|
Enz R. The trick of the tail: protein-protein interactions of metabotropic glutamate receptors. Bioessays 2007; 29:60-73. [PMID: 17187376 DOI: 10.1002/bies.20518] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
It was initially believed that G-protein-coupled receptors, such as metabotropic glutamate receptors, could simply be described as individual proteins that are associated with intracellular signal cascades via G-proteins. This view is no longer tenable. Today we know that metabotropic glutamate receptors (mGluRs) can dimerize and bind to a variety of proteins in addition to trimeric G-proteins. These newly identified protein interactions led to the discovery of new regulatory mechanisms that are independent of and sometimes synergistic with the classical G-protein-coupled second messenger pathways. Notably, several of these mechanisms connect mGluR-mediated signaling to other receptor classes, thereby creating a network of different receptor types and associated signal cascades. The intracellular C-termini of mGluRs play a key role in the regulation of these networks, and various new protein interactions of these domains were described recently. Because mGluRs are involved in a variety of physiological and pathophysiological processes, some of the proteins interacting with this receptor class have potential as valuable pharmaceutical targets. This review will give a comprehensive overview of proteins interacting with mGluR C-termini, highlight new evolving regulatory mechanisms for glutamatergic signal transduction and discuss possibilities for future drug development.
Collapse
Affiliation(s)
- Ralf Enz
- Emil-Fischer-Zentrum, Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, 91054 Erlangen, Germany.
| |
Collapse
|
27
|
Decimo I, Roncarati R, Grasso S, Clemens M, Chiamulera C, Fumagalli G. SK3 trafficking in hippocampal cells: the role of different molecular domains. Biosci Rep 2007; 26:399-412. [PMID: 17061167 DOI: 10.1007/s10540-006-9029-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
The regulative steps that control trafficking of ion channels are fundamental determinants of their qualitative and quantitative expression on the cell membrane. In this work the trafficking of the small conductance calcium-activated potassium channel, SK3 was studied in neurons in order to identify relevant molecular domains involved in this process. Hippocampal cell cultures were transfected with fusion proteins of green fluorescent protein (GFP) and different SK3 subunit truncations. The differential distribution of the mutants was analyzed by confocal microscopy and compared to the localization of the control fusion protein with full length SK3. The transport of chimeric proteins was quantified from fluorescence images by developing a morphometric analytical method. We found that the full length SK3 was distributed in cell body, axon and dendrites, whereas the deleted forms GFPDelta578-736 (deletion of the entire C-terminal domain), GFPDeltaCaMBD (deletion of the calmodulin-binding site) and GFPDeltaN (deletion of the N-terminal domain) were not transported into cell processes but accumulated in the cell body. The GFPDelta640-736 (deletion of the distal C-terminal domain) showed a distribution similar to control. The quantification and statistical analysis confirmed the differences in distribution across the three groups. In conclusion, the current work provides evidence for a fundamental role of the N-terminal domain and the calmodulin binding domain in SK3 trafficking in neurons.
Collapse
Affiliation(s)
- Ilaria Decimo
- Department of Medicine and Public Health, University of Verona, P.le L. Scuro 10, 37100 Verona, Italy.
| | | | | | | | | | | |
Collapse
|
28
|
McDonald NA, Henstridge CM, Connolly CN, Irving AJ. An essential role for constitutive endocytosis, but not activity, in the axonal targeting of the CB1 cannabinoid receptor. Mol Pharmacol 2006; 71:976-84. [PMID: 17182888 DOI: 10.1124/mol.106.029348] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In central neurons, the cell-surface distribution of cannabinoid receptor subtype-1 (CB(1)) is highly polarized toward axons and is associated with synaptic terminals, in which it is well-positioned to modulate neurotransmitter release. It has been suggested that high levels of constitutive activity mediate CB(1) receptor axonal targeting, leading to domain-specific endocytosis. We have investigated further the mechanisms that underlie CB(1) receptor axonal polarization in hippocampal neurons and found that constitutive activity is not an essential requirement for this process. We demonstrate that the cell-surface distribution of an N-terminally tagged, fluorescent CB(1) receptor fusion-protein is almost exclusively localized to the axon when expressed in cultured hippocampal neurons. Inhibition of endocytosis by cotransfection with a dominant-negative dynamin-1 (K44A) mutant traps both recombinant and endogenous CB(1) receptors at the somatodendritic cell surface. However, this effect could not be mimicked by inhibiting constitutive activity or receptor activation, either by expressing mutant receptors that lack these properties or by treatment with CB(1) receptor antagonists possessing inverse agonist activity. These data are consistent with a revised model in which domain-specific endocytosis regulates the functional polarization of CB(1) receptors, but this process is distinct from constitutive activity.
Collapse
Affiliation(s)
- Neil A McDonald
- Neurosciences Institute, Division of Pathology and Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, United Kingdom
| | | | | | | |
Collapse
|
29
|
Kayadjanian N, Lee HS, Piña-Crespo J, Heinemann SF. Localization of glutamate receptors to distal dendrites depends on subunit composition and the kinesin motor protein KIF17. Mol Cell Neurosci 2006; 34:219-30. [PMID: 17174564 PMCID: PMC2692377 DOI: 10.1016/j.mcn.2006.11.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2006] [Revised: 11/02/2006] [Accepted: 11/06/2006] [Indexed: 11/24/2022] Open
Abstract
Correct glutamate receptor localization in neurons is crucial for neurotransmission in the brain. Here we investigated the mechanisms underlying localization of kainate GluR5 receptors to dendrites in cultured hippocampal neurons. We find that the GluR5 distribution depends on association with GluR6 and KA2 subunits. The GluR5 subunit was expressed in distal dendrites only when GluR6 and KA2 subunits were present, whereas it was restricted to proximal dendrites in the absence of these subunits. The overlap between GluR5 distribution and the organization of microtubules in dendrites led us to examine whether KIF17, a microtubule motor protein expressed in distal dendrites, is involved in GluR5 localization to distal dendrites. We show here, for the first time that the microtubule motor protein KIF17 interacts with GluR6 and KA2 subunits and is required for GluR5 localization to distal dendrites, defining a novel mechanism that controls receptor localization in neurons.
Collapse
Affiliation(s)
- N Kayadjanian
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, PO Box 85800, La Jolla, CA 92186-5800, USA.
| | | | | | | |
Collapse
|
30
|
Garzón M, Pickel VM. Subcellular distribution of M2 muscarinic receptors in relation to dopaminergic neurons of the rat ventral tegmental area. J Comp Neurol 2006; 498:821-39. [PMID: 16927256 PMCID: PMC2577061 DOI: 10.1002/cne.21082] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Acetylcholine can affect cognitive functions and reward, in part, through activation of muscarinic receptors in the ventral tegmental area (VTA) to evoke changes in mesocorticolimbic dopaminergic transmission. Among the known muscarinic receptor subtypes present in the VTA, the M2 receptor (M2R) is most implicated in autoregulation and also may play a heteroreceptor role in regulation of the output of the dopaminergic neurons. We sought to determine the functionally relevant sites for M2R activation in relation to VTA dopaminergic neurons by examining the electron microscopic immunolabeling of M2R and the dopamine transporter (DAT) in the VTA of rat brain. The M2R was localized to endomembranes in DAT-containing somatodendritic profiles but showed a more prominent, size-dependent plasmalemmal location in nondopaminergic dendrites. M2R also was located on the plasma membrane of morphologically heterogenous axon terminals contacting unlabeled as well as M2R- or DAT-labeled dendrites. Some of these terminals formed asymmetric synapses resembling those of cholinergic terminals in the VTA. The majority, however, formed symmetric, inhibitory-type synapses or were apposed without recognized junctions. Our results provide the first ultrastructural evidence that the M2R is expressed, but largely not available for local activation, on the plasma membrane of VTA dopaminergic neurons. Instead, the M2R in this region has a distribution suggesting more indirect regulation of mesocorticolimbic transmission through autoregulation of acetylcholine release and changes in the physiological activity or release of other, largely inhibitory transmitters. These findings could have implications for understanding the muscarinic control of cognitive and goal-directed behaviors within the VTA.
Collapse
Affiliation(s)
- Miguel Garzón
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, New York 10021, USA.
| | | |
Collapse
|
31
|
Xu J, Zhu Y, Heinemann SF. Identification of sequence motifs that target neuronal nicotinic receptors to dendrites and axons. J Neurosci 2006; 26:9780-93. [PMID: 16988049 PMCID: PMC6674458 DOI: 10.1523/jneurosci.0840-06.2006] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neuronal nicotinic acetylcholine receptors (nAChRs) belong to a family of ligand-gated ion channels that play important roles in central and peripheral nervous systems. The subcellular distribution of neuronal nAChRs has important implications for function and is not well understood. Here, we analyzed the targeting of two major types of neuronal nAChRs by expressing epitope-tagged subunits in cultured hippocampal neurons. Surprisingly, the alpha7 nAChR (alpha7) and alpha4/beta2 nAChR (alpha4beta2) displayed distinct patterns of expression, with alpha7 targeted preferentially to the somatodendritic compartments, whereas alpha4beta2 was localized to both axonal and dendritic domains. When fused to CD4 or IL2RA (interleukin 2 receptor alpha subunit) proteins, which are normally distributed ubiquitously, the M3-M4 intracellular loop from the alpha7 subunit promoted dendritic expression, whereas the homologous M3-M4 loop from the alpha4 subunit led to surface axonal expression. Systemic screening and alanine substitution further identified a 25-residue leucine motif ([DE]XXXL[LI]) containing an axonal targeting sequence within the alpha4 loop and a 48-residue dileucine and tyrosine motif (YXXØ) containing a dendritic targeting sequence from the alpha7 loop. These results provide valuable information in understanding diverse roles of neuronal nAChRs in mediating and modulating synaptic transmission, synaptic plasticity, and nicotine addiction.
Collapse
Affiliation(s)
- Jian Xu
- Molecular Neurobiology Laboratory, The Salk Institute, La Jolla, California 92037
| | - Yongling Zhu
- Molecular Neurobiology Laboratory, The Salk Institute, La Jolla, California 92037
| | - Stephen F. Heinemann
- Molecular Neurobiology Laboratory, The Salk Institute, La Jolla, California 92037
| |
Collapse
|
32
|
Das SS, Banker GA. The role of protein interaction motifs in regulating the polarity and clustering of the metabotropic glutamate receptor mGluR1a. J Neurosci 2006; 26:8115-25. [PMID: 16885225 PMCID: PMC6673791 DOI: 10.1523/jneurosci.1015-06.2006] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
When expressed in cultured hippocampal neurons, the metabotropic glutamate receptor mGluR1a is polarized to dendrites and concentrated at postsynaptic sites. We used a mutational analysis to determine how previously identified protein interaction motifs in the C terminus of mGluR1a contribute to its localization. Our results show that the polyproline motif that mediates interaction with Homer family proteins is critical for the synaptic clustering of mGluR1a. A single point mutation in this motif, which prevents the binding of Homer with mGluR1a, reduced its colocalization with a postsynaptic marker to near-chance levels but did not affect its dendritic polarity. In contrast, deleting the PDZ (postsynaptic density-95/Discs large/zona occludens-1) binding domain, which interacts with Tamalin and Shank, had no effect on synaptic localization. Neither of these protein interaction motifs is important for trafficking to the plasma membrane or for polarization to dendrites. Although deleting the entire C terminus of mGluR1a only modestly reduced its dendritic polarity, this domain was sufficient to redirect an unpolarized reporter protein to dendrites. These observations suggest that mGluR1a contains redundant dendritic targeting signals. Together, our results indicate that the localization of mGluR1a involves two distinct steps, one that targets the protein to dendrites and a second that sequesters it at postsynaptic sites; different protein interactions motifs mediate each step.
Collapse
Affiliation(s)
- Sonal S Das
- Center for Research on Occupational and Environmental Toxicology, Oregon Health and Science University, Portland, Oregon 97239, USA
| | | |
Collapse
|
33
|
Abstract
Voltage-gated ion channels have to be at the right place in the right number to endow individual neurons with their specific character. Their biophysical properties together with their spatial distribution define the signalling characteristics of a neuron. Improper channel localization could cause communication defects in a neuronal network. This review covers recent studies of mechanisms for targeting voltage-gated ion channels to axons and dendrites, including trafficking, retention and endocytosis pathways for the preferential localization of particular ion channels. We also discuss how the spatial localization of these channels might contribute to the electrical excitability of neurons, and consider the need for future work in this emerging field.
Collapse
Affiliation(s)
- Helen C Lai
- Center for Basic Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | |
Collapse
|
34
|
Misonou H, Menegola M, Buchwalder L, Park EW, Meredith A, Rhodes KJ, Aldrich RW, Trimmer JS. Immunolocalization of the Ca2+-activated K+ channel Slo1 in axons and nerve terminals of mammalian brain and cultured neurons. J Comp Neurol 2006; 496:289-302. [PMID: 16566008 PMCID: PMC2605666 DOI: 10.1002/cne.20931] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Ca(2+)-activated voltage-dependent K(+) channels (Slo1, KCa1.1, Maxi-K, or BK channel) play a crucial role in controlling neuronal signaling by coupling channel activity to both membrane depolarization and intracellular Ca(2+) signaling. In mammalian brain, immunolabeling experiments have shown staining for Slo1 channels predominantly localized to axons and presynaptic terminals of neurons. We have developed anti-Slo1 mouse monoclonal antibodies that have been extensively characterized for specificity of staining against recombinant Slo1 in heterologous cells, and native Slo1 in mammalian brain, and definitively by the lack of detectable immunoreactivity against brain samples from Slo1 knockout mice. Here we provide precise immunolocalization of Slo1 in rat brain with one of these monoclonal antibodies and show that Slo1 is accumulated in axons and synaptic terminal zones associated with glutamatergic synapses in hippocampus and GABAergic synapses in cerebellum. By using cultured hippocampal pyramidal neurons as a model system, we show that heterologously expressed Slo1 is initially targeted to the axonal surface membrane, and with further development in culture, become localized in presynaptic terminals. These studies provide new insights into the polarized localization of Slo1 channels in mammalian central neurons and provide further evidence for a key role in regulating neurotransmitter release in glutamatergic and GABAergic terminals.
Collapse
Affiliation(s)
- Hiroaki Misonou
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616
- Department of Biochemistry and Cell Biology, State University of New York at Stony Brook, Stony Brook, NY 11794
| | - Milena Menegola
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616
| | - Lynn Buchwalder
- Department of Biochemistry and Cell Biology, State University of New York at Stony Brook, Stony Brook, NY 11794
| | - Eunice W. Park
- Department of Biochemistry and Cell Biology, State University of New York at Stony Brook, Stony Brook, NY 11794
| | - Andrea Meredith
- Department of Molecular and Cellular Physiology and Howard Hughes Medical Institute, Stanford School of Medicine, Stanford, CA 94305
| | | | - Richard W. Aldrich
- Department of Molecular and Cellular Physiology and Howard Hughes Medical Institute, Stanford School of Medicine, Stanford, CA 94305
| | - James S. Trimmer
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616
- Department of Biochemistry and Cell Biology, State University of New York at Stony Brook, Stony Brook, NY 11794
| |
Collapse
|
35
|
Abstract
Although kinesins are known to transport neuronal proteins, it is not known what role they play in the targeting of their cargos to specific subcellular compartments in neurons. Here we present evidence that the K+ channel Kv4.2, which is a major regulator of dendritic excitability, is transported to dendrites by the kinesin isoform Kif17. We show that a dominant negative construct against Kif17 dramatically inhibits localization to dendrites of both introduced and endogenous Kv4.2, but those against other kinesins found in dendrites do not. Kv4.2 colocalizes with Kif17 but not with other kinesin isoforms in dendrites of cortical neurons. Native Kv4.2 and Kif17 coimmunoprecipitate from brain lysate, and introduced, tagged versions of the two proteins coimmunoprecipitate from COS cell lysate, indicating that the two proteins interact, either directly or indirectly. The interaction between Kif17 and Kv4.2 appears to occur through the extreme C terminus of Kv4.2 and not through the dileucine motif. Thus, the dileucine motif does not determine the localization of Kv4.2 by causing the channel to interact with a specific motor protein. In support of this conclusion, we found that the dileucine motif mediates dendritic targeting of CD8 independent of Kif17. Together our data show that Kif17 is probably the motor that transports Kv4.2 to dendrites but suggest that this motor does not, by itself, specify dendritic localization of the channel.
Collapse
Affiliation(s)
- Po-Ju Chu
- Department of Biology and Program in Molecular and Computational Biology, University of Southern California, Los Angeles, California 90089-2910, USA
| | | | | |
Collapse
|
36
|
Abstract
Shaker K+ channels play an important role in modulating electrical excitability of axons. Recent work has demonstrated that the T1 tetramerization domain of Kv1.2 is both necessary and sufficient for targeting of the channel to the axonal surface [Gu, C., Jan, Y.N. & Jan, L.Y. (2003) Science,301, 646-649]. Here we use a related channel, Kv1.3, as a model to investigate cellular mechanisms that mediate axonal targeting. We show that the T1 domain of Kv1.3 is necessary and sufficient to mediate targeting of the channel to the axonal surface in pyramidal neurons in slices of cortex from neonatal rat. The T1 domain is also sufficient to cause preferential axonal localization of intracellular protein, which indicates that the domain probably does not work through compartment-specific endocytosis or compartment-specific vesicle docking. To determine whether the T1 domain mediates axonal trafficking of transport vesicles, we compared the trafficking of vesicles containing green fluorescent protein-labelled transferrin receptor with those containing the same protein fused with the T1 domain in living cortical neurons. Vesicles containing the wild-type transferrin receptor did not traffic to the axon, in accord with previously published results; however, those containing the transferrin receptor fused to T1 did traffic to the axon. These results are consistent with the T1 domain of Kv1.3 mediating axonal targeting by causing transport vesicles to traffic to axons and they represent the first evidence that such a mechanism might underlie axonal targeting.
Collapse
Affiliation(s)
- Jacqueline F Rivera
- University of Southern California, Department of Biological Sciences and Program in Molecular and Computational Biology, 1050 Childs Way MCB 204, Los Angeles, CA 90089-1340, USA
| | | | | |
Collapse
|
37
|
Sakagami Y, Yamamoto K, Sugiura S, Inokuchi K, Hayashi T, Kato N. Essential roles of Homer-1a in homeostatic regulation of pyramidal cell excitability: a possible link to clinical benefits of electroconvulsive shock. Eur J Neurosci 2005; 21:3229-39. [PMID: 16026461 DOI: 10.1111/j.1460-9568.2005.04165.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Homer-1a/Vesl1S, a member of the scaffold protein family Homer/Vesl, is expressed during seizure and serves to reduce seizure susceptibility. Cellular mechanisms for this feedback regulation were studied in neocortex pyramidal cells by injecting Homer-1a protein intracellularly. The injection reduced membrane excitability as demonstrated in two ways. First, the resting potential was hyperpolarized by 5-10 mV. Second, the mean frequency of spikes evoked by depolarizing current injection was decreased. This reduction of excitability was prevented by applying each of the followings: the calcium chelator BAPTA, the calcium store depletor cyclopiazonic acid (CPA), the insitol-1,4,5-trisphosphate receptor (IP(3)R) blocker heparin, the phospholipase C (PLC) inhibitor U-73122, the metabotropic glutamate receptor (mGluR) antagonist 2-methyl-6-(phenylethynyl)-pyridine (MPEP), and the large-conductance calcium activated potassium channel (BK channel) antagonist charybdotoxin. The small-conductance calcium activated potassium channel (SK channel) blocker dequalinium was ineffective. These findings suggest that activation of mGluR by Homer-1a produced IP(3), which caused inositol-induced calcium release and a consequent BK channel opening, thus hyperpolarizing the injected neurons. In slices from rats subjected to electroconvulsive shock (ECS), a comparable reduction of excitability was observed without Homer-1a injection. The ECS-induced reduction of excitability was abolished by MPEP, charybdotoxin, heparin or BAPTA. Intracellular injection of anti-Homer-1a antibody was suppressive as well, but anti-Homer-1b/c antibody was not. We propose that ECS-induced Homer-1a stimulated the same pathway as did the injected Homer-1a, thereby driving a feedback regulation of excitability.
Collapse
Affiliation(s)
- Yu Sakagami
- Department of Integrative Brain Science, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | | | | | | | | | | |
Collapse
|
38
|
Mitsui S, Saito M, Hayashi K, Mori K, Yoshihara Y. A novel phenylalanine-based targeting signal directs telencephalin to neuronal dendrites. J Neurosci 2005; 25:1122-31. [PMID: 15689548 PMCID: PMC6725959 DOI: 10.1523/jneurosci.3853-04.2005] [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] [Indexed: 11/21/2022] Open
Abstract
Neurons sort out a variety of functional molecules to appropriate subcellular destinations. Telencephalin (TLCN; intercellular adhesion molecule-5) is a cell adhesion molecule specifically localized to somatodendritic membranes in the telencephalic neurons. Here, we established a new in vivo strategy to analyze neuronal sorting mechanisms by ectopic expression of molecules of interest in the cerebellar Purkinje cells of transgenic mice. By using this system, we identified a novel dendritic targeting determinant in the cytoplasmic tail region of TLCN. A full-length TLCN ectopically expressed in the Purkinje cells was localized exclusively to dendrites but not to axons. In contrast, a deletion of cytoplasmic C-terminal 12 amino acids (residues 901-912) or a point mutation of Phe905 to Ala abrogated the dendrite-specific targeting with appearance of the truncated and point-mutated TLCN in both axons and dendrites. Furthermore, an addition of the C-terminal 17 amino acids (residues 896-912) of TLCN to an unrelated molecule (CD8) was sufficient for its specific targeting to dendrites in several types of neurons. Because the C-terminal region of TLCN does not contain any canonical dendritic targeting sequences such as the tyrosine-based motif or the dileucine motif, this study suggests a novel mechanism of protein trafficking to the dendritic compartment of neurons.
Collapse
Affiliation(s)
- Sachiko Mitsui
- Laboratory for Neurobiology of Synapse, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | | | | | | | | |
Collapse
|
39
|
Routtenberg A, Rekart JL. Post-translational protein modification as the substrate for long-lasting memory. Trends Neurosci 2005; 28:12-9. [PMID: 15626492 DOI: 10.1016/j.tins.2004.11.006] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Prevailing models of memory identify mRNA translation as necessary for long-lasting information storage. However, there are enough instances of memory storage in the virtual absence of protein synthesis to prompt consideration of alternative models. A comprehensive review of the protein synthesis literature leads us to conclude that the translational mechanism is exclusively a permissive, replenishment step. Therefore, we propose that post-translational modification (PTM) of proteins already at the synapse is the crucial instructive mechanism underlying long-lasting memory. A novel feature of this model is that non-random spontaneous (or endogenous) brain activity operates as a regulated positive-feedback rehearsal mechanism, updating network configurations by fine-tuning the PTM state of previously modified proteins. Synapses participating in memory storage are therefore supple, a feature required for networks to alter complexity and update continuously. In analogy with codons for amino acids, a long-lasting memory is represented by a 'degenerate code' - a set of pseudo-redundant networks that can ensure its longevity.
Collapse
Affiliation(s)
- Aryeh Routtenberg
- Department of Psychology, Northwestern University, Evanston, IL 60208, USA.
| | | |
Collapse
|
40
|
Hirokawa N, Takemura R. Molecular motors and mechanisms of directional transport in neurons. Nat Rev Neurosci 2005; 6:201-14. [PMID: 15711600 DOI: 10.1038/nrn1624] [Citation(s) in RCA: 598] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Intracellular transport is fundamental for neuronal morphogenesis, function and survival. Many proteins are selectively transported to either axons or dendrites. In addition, some specific mRNAs are transported to dendrites for local translation. Proteins of the kinesin superfamily participate in selective transport by using adaptor or scaffolding proteins to recognize and bind cargoes. The molecular components of RNA-transporting granules have been identified, and it is becoming clear how cargoes are directed to axons and dendrites by kinesin superfamily proteins. Here we discuss the molecular mechanisms of directional axonal and dendritic transport with specific emphasis on the role of motor proteins and their mechanisms of cargo recognition.
Collapse
Affiliation(s)
- Nobutaka Hirokawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan.
| | | |
Collapse
|
41
|
Abstract
Modulating protein-protein interactions involved in disease pathways is an attractive strategy for developing drugs, but remains a challenge to achieve. One approach is to target certain domains within proteins that mediate these interactions. One example of such a domain is the PDZ domain, which is involved in interactions between many different proteins in a variety of cellular contexts. Because PDZ domains have well-defined binding sites, they are promising targets for drug discovery. However, there is still much to learn about the function of these domains before drugs targeting PDZ interactions can become a reality.
Collapse
Affiliation(s)
- Kumlesh K Dev
- Neuroscience Research, Novartis Institutes for BioMedical Research, Novartis Pharma AG, WSJ-386.7.43, CH-4002 Basel, Switzerland.
| |
Collapse
|
42
|
Herlitze S, Xie M, Han J, Hümmer A, Melnik-Martinez KV, Moreno RL, Mark MD. Targeting mechanisms of high voltage-activated Ca2+ channels. J Bioenerg Biomembr 2004; 35:621-37. [PMID: 15000523 DOI: 10.1023/b:jobb.0000008027.19384.c0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Functional voltage-dependent Ca2+ channel complexes are assembled by three to four subunits: alpha1, beta, alpha2delta subunits (C. Leveque et al., 1994, J. Biol Chem. 269, 6306-6312; M. W. McEnery et al., 1991, Proc. Natl. Acad. Sci. U.S.A. 88, 11095-11099) and at least in muscle cells also y subunits (B. M. Curtis and W. A. Catterall, 1984, Biochemistry 23, 2113-2118). Ca2+ channels mediate the voltage-dependent Ca2+ influx in subcellular compartments, triggering such diverse processes as neurotransmitter release, dendritic action potentials, excitation-contraction, and excitation-transcription coupling. The targeting of biophysically defined Ca2+ channel complexes to the correct subcellular structures is, thus, critical to proper cell and physiological functioning. Despite their importance, surprisingly little is known about the targeting mechanisms by which Ca2+ channel complexes are transported to their site of function. Here we summarize what we know about the targeting of Ca2+ channel complexes through the cell to the plasma membrane and subcellular structures.
Collapse
Affiliation(s)
- Stefan Herlitze
- Department of Neurosciences, Case Western Reserve University, School of Medicine, Room E604, 10900 Euclid Avenue, Cleveland, Ohio 44106-4975, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
G protein-coupled receptors (GPCRs) modulate diverse physiological and behavioral signaling pathways by virtue of changes in receptor activation and inactivation states. Functional changes in receptor properties include dynamic interactions with regulatory molecules and trafficking to various cellular compartments at various stages of the life cycle of a GPCR. This review focuses on trafficking of GPCRs to the cell surface, stabilization there, and agonist-regulated turnover. GPCR interactions with a variety of newly revealed partners also are reviewed with the intention of provoking further analysis of the relevance of these interactions in GPCR trafficking, signaling, or both. The disease consequences of mislocalization of GPCRs also are described.
Collapse
Affiliation(s)
- Christopher M Tan
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | | | | | | | | |
Collapse
|
44
|
Schlag BD, Lou Z, Fennell M, Dunlop J. Ligand Dependency of 5-Hydroxytryptamine 2C Receptor Internalization. J Pharmacol Exp Ther 2004; 310:865-70. [PMID: 15113845 DOI: 10.1124/jpet.104.067306] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Agonist-induced internalization of G protein-coupled receptors (GPCRs) is a well characterized phenomenon believed to contribute to receptor desensitization. The 5-hydroxytryptamine (5-HT)2C subtype of serotonin receptor is a GPCR that we have shown to internalize upon agonist incubation. In this study, we have examined the effects of 5-HT2C receptor agonists serotonin, Ro 60-0175 [(S)-2-(6-chloro-5-fluoroindol-1-yl)-1-methylethylamine], and WAY-161503 [(4aR)-8,9-dichloro-2,3,4,4a-tetrahydro-1H-pyrazino[1,2-a]quinoxalin-5(6H)-one]; partial agonists mCPP [1-(m-chlorophenyl)piperazine] and DOI [(+)-1-(2,5-dimethoxy-4-iodophenyl)-2-amino-propane]; inverse agonists SB-206553 [N-3-pyridinyl-3,5-dihydro-5-methylbenzo(1,2-b:4,5-b')dipyrrole-1(2H)carboxamide] and mianserin; and neutral antagonists SB-242084 [6-chloro-5-methyl-1-[[2-[(2-methyl-3-pyridyl)oxy]-5-pyridyl]carbamoyl]-indoline] and 5-methoxygramine on the internalization of a C-terminal green fluorescent protein (GFP)-tagged 5-HT2C receptor (VSV isoform) expressed in transiently transfected human embryonic kidney cells. We detected internalization with an automated, cell-based fluorescence-imaging system (Arrayscan) and monitored function with intracellular Ca2+ measurements (flourometric imaging plate reader). The 5-HT2C-GFP construct exhibited appropriate pharmacology, and we observed that although all three agonists resulted in similar magnitudes of dose-dependent internalization, the partial agonists resulted in approximately 50% less internalization, and the inverse agonists and neutral antagonists failed to induce internalization. These results were confirmed by confocal microscopy. They demonstrate that the 5-HT2C receptor is internalized by incubation with agonists and partial agonists but not with inverse agonists or neutral antagonists.
Collapse
Affiliation(s)
- Brian D Schlag
- Neuroscience Discovery Research, Wyeth Research, CN 8000, Princeton, NJ 08543, USA
| | | | | | | |
Collapse
|
45
|
Abstract
Like axons, dendrites need guidance for proper orientation and positioning within the brain. Guidance determines synaptic connectivity as well as the strength of transmission. Recent in vivo studies have demonstrated that several cell-surface receptors, previously known as axon guidance molecules, are also responsible for the directed outgrowth of dendrites. Collectively, these studies reveal that the function of guidance molecules in individual neurons and individual processes is diverse and likely to be specifically regulated. Here, these studies are reviewed and emerging issues and implications are discussed.
Collapse
Affiliation(s)
- Susan Kim
- Department of Cell and Structural Biology, University of Illinois, Urbana, IL 61801, USA
| | | |
Collapse
|
46
|
Alonso-Nanclares L, White EL, Elston GN, DeFelipe J. Synaptology of the proximal segment of pyramidal cell basal dendrites. Eur J Neurosci 2004; 19:771-6. [PMID: 14984428 DOI: 10.1111/j.0953-816x.2003.03166.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Pyramidal neurons are covered with dendritic spines, the main postsynaptic targets of excitatory (asymmetrical) synapses. However, the proximal portion of both the apical and basal dendrites is devoid of spines, suggesting a lack of excitatory inputs to this region. In the present study we used electron microscopy to analyse the proximal region of the basal dendrites of supra- and infragranular pyramidal cells to determine if this is the case. The proximal region of 80 basal dendrites sampled from the rat hindlimb representation in the primary somatosensory cortex was studied by electron microscopy. A total of 317 synapses were found within this region of the dendrites, all of which were of the symmetrical type. These results suggest that glutamate receptors, although present in the cytoplasm, are not involved in synaptic junctions in the proximal portion of the dendrites. These data further support the idea that inhibitory terminals exclusively innervate the proximal region of basal dendrites.
Collapse
|
47
|
Tamamaki N, Yanagawa Y, Tomioka R, Miyazaki JI, Obata K, Kaneko T. Green fluorescent protein expression and colocalization with calretinin, parvalbumin, and somatostatin in the GAD67-GFP knock-in mouse. J Comp Neurol 2004; 467:60-79. [PMID: 14574680 DOI: 10.1002/cne.10905] [Citation(s) in RCA: 1035] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Gamma-aminobutyric acid (GABA)ergic neurons in the central nervous system regulate the activity of other neurons and play a crucial role in information processing. To assist an advance in the research of GABAergic neurons, here we produced two lines of glutamic acid decarboxylase-green fluorescence protein (GAD67-GFP) knock-in mouse. The distribution pattern of GFP-positive somata was the same as that of the GAD67 in situ hybridization signal in the central nervous system. We encountered neither any apparent ectopic GFP expression in GAD67-negative cells nor any apparent lack of GFP expression in GAD67-positive neurons in the two GAD67-GFP knock-in mouse lines. The timing of GFP expression also paralleled that of GAD67 expression. Hence, we constructed a map of GFP distribution in the knock-in mouse brain. Moreover, we used the knock-in mice to investigate the colocalization of GFP with NeuN, calretinin (CR), parvalbumin (PV), and somatostatin (SS) in the frontal motor cortex. The proportion of GFP-positive cells among NeuN-positive cells (neocortical neurons) was approximately 19.5%. All the CR-, PV-, and SS-positive cells appeared positive for GFP. The CR-, PV, and SS-positive cells emitted GFP fluorescence at various intensities characteristics to them. The proportions of CR-, PV-, and SS-positive cells among GFP-positive cells were 13.9%, 40.1%, and 23.4%, respectively. Thus, the three subtypes of GABAergic neurons accounted for 77.4% of the GFP-positive cells. They accounted for 6.5% in layer I. In accord with unidentified GFP-positive cells, many medium-sized spherical somata emitting intense GFP fluorescence were observed in layer I.
Collapse
Affiliation(s)
- Nobuaki Tamamaki
- Department of Morphological Brain Science, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan.
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
Targeting of proteins to specific subcellular locations within pre- and postsynaptic neurons is essential for synapse formation. The heparan sulfate proteoglycan agrin orchestrates postsynaptic differentiation of the neuromuscular junction and may be involved in synaptic development and signaling in the central nervous system (CNS). Agrin is expressed as transmembrane and secretory isoforms with distinct N-termini. We examined the distribution of recombinant agrin in cultured motor and hippocampal neurons by transfection with agrin-GFP constructs. Immunostaining revealed a vesicular transport compartment within all neurites. Plasma membrane insertion and secretion of recombinant agrin were targeted to axonal growth cones of motor neurons; transmembrane agrin-GFP was targeted predominantly to axons and axonal growth cones in hippocampal neurons. We used agrin deletion mutants to show that axonal targeting of agrin depends on multiple domains that function in an additive fashion, including the very N-terminal portions and the C-terminal half of the molecule.
Collapse
Affiliation(s)
- Birgit Neuhuber
- Laboratory of Cell Biology, NHLBI-NIH, Bethesda, MD 20892, USA
| | | |
Collapse
|
49
|
Wisco D, Anderson ED, Chang MC, Norden C, Boiko T, Fölsch H, Winckler B. Uncovering multiple axonal targeting pathways in hippocampal neurons. ACTA ACUST UNITED AC 2003; 162:1317-28. [PMID: 14517209 PMCID: PMC2173963 DOI: 10.1083/jcb.200307069] [Citation(s) in RCA: 136] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Neuronal polarity is, at least in part, mediated by the differential sorting of membrane proteins to distinct domains, such as axons and somata/dendrites. We investigated the pathways underlying the subcellular targeting of NgCAM, a cell adhesion molecule residing on the axonal plasma membrane. Following transport of NgCAM kinetically, surprisingly we observed a transient appearance of NgCAM on the somatodendritic plasma membrane. Down-regulation of endocytosis resulted in loss of axonal accumulation of NgCAM, indicating that the axonal localization of NgCAM was dependent on endocytosis. Our data suggest the existence of a dendrite-to-axon transcytotic pathway to achieve axonal accumulation. NgCAM mutants with a point mutation in a crucial cytoplasmic tail motif (YRSL) are unable to access the transcytotic route. Instead, they were found to travel to the axon on a direct route. Therefore, our results suggest that multiple distinct pathways operate in hippocampal neurons to achieve axonal accumulation of membrane proteins.
Collapse
Affiliation(s)
- Dolora Wisco
- Brookdale Department of Molecular, Cell, and Developmental Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Among the most morphologically complex cells, neurons are masters of membrane specialization. Nowhere is this more striking than in the division of cellular labor between the axon and the dendrites. In morphology, signaling properties, cytoskeletal organization, and physiological function, axons and dendrites (or more properly, the somatodendritic compartment) are radically different. Such polarization of neurons into domains specialized for either receiving (dendrites) or transmitting (axons) cellular signals provides the underpinning for all neural circuitry. The initial specification of axonal and dendritic identity occurs early in neuronal life, persists for decades, and is manifested by the presence of very different sets of cell surface proteins. Yet, how neuronal polarity is established, how distinct axonal and somatodendritic domains are maintained, and how integral membrane proteins are directed to dendrites or accumulate in axons remain enduring and formidable questions in neuronal cell biology.
Collapse
Affiliation(s)
- April C Horton
- Department of Neurobiology, Duke University Medical Center, Box 3209, Durham, NC 27710, USA
| | | |
Collapse
|