首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 571 毫秒
1.
GABAB receptors assemble from principle and auxiliary subunits. The principle subunits GABAB1 and GABAB2 form functional heteromeric GABAB(1,2) receptors that associate with homotetramers of auxiliary KCTD8, -12, -12b, or -16 (named after their K+ channel tetramerization domain) subunits. These auxiliary subunits constitute receptor subtypes with distinct functional properties. KCTD12 and -12b generate desensitizing receptor responses while KCTD8 and -16 generate largely non-desensitizing receptor responses. The structural elements of the KCTDs underlying these differences in desensitization are unknown. KCTDs are modular proteins comprising a T1 tetramerization domain, which binds to GABAB2, and a H1 homology domain. KCTD8 and -16 contain an additional C-terminal H2 homology domain that is not sequence-related to the H1 domains. No functions are known for the H1 and H2 domains. Here we addressed which domains and sequence motifs in KCTD proteins regulate desensitization of the receptor response. We found that the H1 domains in KCTD12 and -12b mediate desensitization through a particular sequence motif, T/NFLEQ, which is not present in the H1 domains of KCTD8 and -16. In addition, the H2 domains in KCTD8 and -16 inhibit desensitization when expressed C-terminal to the H1 domains but not when expressed as a separate protein in trans. Intriguingly, the inhibitory effect of the H2 domain is sequence-independent, suggesting that the H2 domain sterically hinders desensitization by the H1 domain. Evolutionary analysis supports that KCTD12 and -12b evolved desensitizing properties by liberating their H1 domains from antagonistic H2 domains and acquisition of the T/NFLEQ motif.  相似文献   

2.
Inhibitory GABAB receptors (GABABRs) can down-regulate most excitatory synapses in the CNS by reducing postsynaptic excitability. Functional GABABRs are heterodimers of GABAB1 and GABAB2 subunits and here we show that the trafficking and surface expression of GABABRs is differentially regulated by synaptic or pathophysiological activation of NMDA receptors (NMDARs). Activation of synaptic NMDARs using a chemLTP protocol increases GABABR recycling and surface expression. In contrast, excitotoxic global activation of synaptic and extrasynaptic NMDARs by bath application of NMDA causes the loss of surface GABABRs. Intriguingly, exposing neurons to extreme metabolic stress using oxygen/glucose deprivation (OGD) increases GABAB1 but decreases GABAB2 surface expression. The increase in surface GABAB1 involves enhanced recycling and is blocked by the NMDAR antagonist AP5. The decrease in surface GABAB2 is also blocked by AP5 and by inhibiting degradation pathways. These results indicate that NMDAR activity is critical in GABABR trafficking and function and that the individual subunits can be separately controlled to regulate neuronal responsiveness and survival.  相似文献   

3.
4.
Metabotropic GABAB receptors are crucial for controlling the excitability of neurons by mediating slow inhibition in the CNS. The strength of receptor signaling depends on the number of cell surface receptors, which is thought to be regulated by trafficking and degradation mechanisms. Although the mechanisms of GABAB receptor trafficking are studied to some extent, it is currently unclear whether receptor degradation actively controls the number of GABAB receptors available for signaling. Here we tested the hypothesis that proteasomal degradation contributes to the regulation of GABAB receptor expression levels. Blocking proteasomal activity in cultured cortical neurons considerably enhanced total and cell surface expression of GABAB receptors, indicating the constitutive degradation of the receptors by proteasomes. Proteasomal degradation required Lys48-linked polyubiquitination of lysines 767/771 in the C-terminal domain of the GABAB2 subunit. Inactivation of these ubiquitination sites increased receptor levels and GABAB receptor signaling in neurons. Proteasomal degradation was mediated by endoplasmic reticulum-associated degradation (ERAD) as shown by the accumulation of receptors in the endoplasmic reticulum upon inhibition of proteasomes, by the increase of receptor levels, as well as receptor signaling upon blocking ERAD function, and by the interaction of GABAB receptors with the essential ERAD components Hrd1 and p97. In conclusion, the data support a model in which the fraction of GABAB receptors available for plasma membrane trafficking is regulated by degradation via the ERAD machinery. Thus, modulation of ERAD activity by changes in physiological conditions may represent a mechanism to adjust receptor numbers and thereby signaling strength.  相似文献   

5.
Metabotropic GABAB receptors are abundantly expressed at glutamatergic synapses where they control excitability of the synapse. Here, we tested the hypothesis that glutamatergic neurotransmission may regulate GABAB receptors. We found that application of glutamate to cultured cortical neurons led to rapid down-regulation of GABAB receptors via lysosomal degradation. This effect was mimicked by selective activation of AMPA receptors and further accelerated by coactivation of group I metabotropic glutamate receptors. Inhibition of NMDA receptors, blockade of L-type Ca2+ channels, and removal of extracellular Ca2+ prevented glutamate-induced down-regulation of GABAB receptors, indicating that Ca2+ influx plays a critical role. We further established that glutamate-induced down-regulation depends on the internalization of GABAB receptors. Glutamate did not affect the rate of GABAB receptor endocytosis but led to reduced recycling of the receptors back to the plasma membrane. Blockade of lysosomal activity rescued receptor recycling, indicating that glutamate redirects GABAB receptors from the recycling to the degradation pathway. In conclusion, the data indicate that sustained activation of AMPA receptors down-regulates GABAB receptors by sorting endocytosed GABAB receptors preferentially to lysosomes for degradation on the expense of recycling. This mechanism may relieve glutamatergic synapses from GABAB receptor-mediated inhibition resulting in increased synaptic excitability.  相似文献   

6.
GABAB receptors function as heterodimeric G-protein-coupled receptors for the neurotransmitter γ-aminobutyric acid (GABA). Receptor subtypes, based on isoforms of the ligand-binding subunit GABAB1, are thought to involve a differential set of associated proteins. Here, we describe two mouse lines that allow a straightforward biochemical isolation of GABAB receptors. The transgenic mice express GABAB1 isoforms that contain sequences for a two-step affinity purification, in addition to their endogenous subunit repertoire. Comparative analyses of purified samples from the transgenic mice and wild-type control animals revealed two novel components of the GABAB1 complex. One of the identified proteins, potassium channel tetramerization domain-containing protein 12, associates with heterodimeric GABAB receptors via the GABAB2 subunit. In transfected hippocampal neurons, potassium channel tetramerization domain-containing protein 12 augmented axonal surface targeting of GABAB2. The mice equipped with tags on GABAB1 facilitate validation and identification of native binding partners of GABAB receptors, providing insight into the molecular mechanisms of synaptic modulation.  相似文献   

7.
Regulation of cell surface expression of neurotransmitter receptors is crucial for determining synaptic strength and plasticity, but the underlying mechanisms are not well understood. We previously showed that proteasomal degradation of GABAB receptors via the endoplasmic reticulum (ER)-associated protein degradation (ERAD) machinery determines the number of cell surface GABAB receptors and thereby GABAB receptor-mediated neuronal inhibition. Here, we show that proteasomal degradation of GABAB receptors requires the interaction of the GABAB2 C terminus with the proteasomal AAA-ATPase Rpt6. A mutant of Rpt6 lacking ATPase activity prevented degradation of GABAB receptors but not the removal of Lys48-linked ubiquitin from GABAB2. Blocking ERAD activity diminished the interaction of Rtp6 with GABAB receptors resulting in increased total as well as cell surface expression of GABAB receptors. Modulating neuronal activity affected proteasomal activity and correspondingly the interaction level of Rpt6 with GABAB2. This resulted in altered cell surface expression of the receptors. Thus, neuronal activity-dependent proteasomal degradation of GABAB receptors by the ERAD machinery is a potent mechanism regulating the number of GABAB receptors available for signaling and is expected to contribute to homeostatic neuronal plasticity.  相似文献   

8.
In this study, we investigated the distribution and developmental expression of the GABAB receptor subunits, GABAB1 and GABAB2, in the main and accessory olfactory bulbs of the rat. Antibodies raised against these subunits strongly labelled the glomerular layer, suggesting that olfactory and vomeronasal nerve fibers express functional GABAB receptors. Using postembedding immunogold cytochemistry, we found that GABAB receptors can be present at both extrasynaptic and presynaptic sites of olfactory nerve terminals, and in the latter case they are preferentially associated with the peripheral part of the synaptic specialization. Olfactory nerve fibers expressed GABAB1 and GABAB2 at early developmental stages, suggesting that GABAB receptors may play a role in olfactory development. Output and local neurons of the main and accessory olfactory bulbs were also labelled for GABAB1 and GABAB2, although the subcellular distribution patterns of the two subunits were not completely overlapping. These results indicate that presynaptically located GABAB receptors modulate neurotransmitter release from olfactory and vomeronasal nerve fibers and that, in addition to this presynaptic role, GABAB receptors may regulate neuronal excitability in infraglomerular circuits.  相似文献   

9.
Recent investigations have shown that members of the KCTD family play important roles in fundamental biological processes. Despite their roles, very limited information is available on their structures and molecular organization. By combining different experimental and theoretical techniques, we have here characterized the two folded domains of KCTD12, an integral component and modulator of the GABAB2 receptor. Secondary prediction methods and CD spectroscopy have shown that the N‐terminal domain KCTD12BTB assumes an α/β structure, whereas the C‐terminal domain KCTD12H1 is predominantly characterized by a β‐structure. Binding assays indicate that the two domains independently expressed show a good affinity for each other. This suggests that the overall protein is likely endowed with a rather compact structure with two interacting structured domains joint by a long disordered region. Notably, both KCTD12BTB and KCTD12H1 are tetrameric when individually expressed. This finding could modify the traditional view that ascribes only to POZ/BTB domain a specific oligomerization role. The first quantification of the affinity of KCTD12POZ/BTB for the C‐terminal region of GABAB2 shows that it falls in the low micromolar range. Interestingly, we also demonstrate that a GABAB2‐related peptide is able to bind KCTD12BTB with a very high affinity. This peptide may represent a useful tool for modulating KCTD12/GABAB2 interaction in vitro and may also constitute the starting point for the development of peptidomimetic compounds with a potential for therapeutic applications. Copyright © 2013 John Wiley & Sons, Ltd.  相似文献   

10.
GABAB receptors are the G‐protein‐coupled receptors for the neurotransmitter γ‐aminobutyric acid (GABA). Receptor subtypes are based on the subunit isoforms GABAB1a and GABAB1b, which combine with GABAB2 subunits to form heteromeric receptors. Here, we used a modified bacterial artificial chromosome (BAC) containing the GABAB1 gene to generate transgenic mice expressing GABAB1a and GABAB1b subunits fused to the enhanced green fluorescence protein (eGFP). We demonstrate that the GABAB1‐eGFP fusion proteins reproduce the cellular expression patterns of endogenous GABAB1 proteins in the brain and in peripheral tissue. Crossing the GABAB1‐eGFP BAC transgene into the GABAB1?/? background restores pre and postsynaptic GABAB functions, showing that the GABAB1‐eGFP fusion proteins substitute for the lack of endogenous GABAB1 proteins. Finally, we demonstrate that the GABAB1‐eGFP fusion proteins replicate the temporal expression patterns of native GABAB receptors in cultured neurons. These transgenic mice therefore provide a validated tool for direct visualization of native GABAB receptors. genesis 47:595–602, 2009. © 2009 Wiley‐Liss, Inc.  相似文献   

11.
γ-Aminobutyric acid type A (GABAA) receptors are pentameric ligand-gated ion channels that mediate fast inhibition in the central nervous system. Depending on their subunit composition, these receptors exhibit distinct pharmacological properties and differ in their ability to interact with proteins involved in receptor anchoring at synaptic or extra-synaptic sites. Whereas GABAA receptors containing α1, α2, or α3 subunits are mainly located synaptically where they interact with the submembranous scaffolding protein gephyrin, receptors containing α5 subunits are predominantly found extra-synaptically and seem to interact with radixin for anchorage. Neuroplastin is a cell adhesion molecule of the immunoglobulin superfamily that is involved in hippocampal synaptic plasticity. Our results reveal that neuroplastin and GABAA receptors can be co-purified from rat brain and exhibit a direct physical interaction as demonstrated by co-precipitation and Förster resonance energy transfer (FRET) analysis in a heterologous expression system. The brain-specific isoform neuroplastin-65 co-localizes with GABAA receptors as shown in brain sections as well as in neuronal cultures, and such complexes can either contain gephyrin or be devoid of gephyrin. Neuroplastin-65 specifically co-localizes with α1 or α2 but not with α3 subunits at GABAergic synapses. In addition, neuroplastin-65 also co-localizes with GABAA receptor α5 subunits at extra-synaptic sites. Down-regulation of neuroplastin-65 by shRNA causes a loss of GABAA receptor α2 subunits at GABAergic synapses. These results suggest that neuroplastin-65 can co-localize with a subset of GABAA receptor subtypes and might contribute to anchoring and/or confining GABAA receptors to particular synaptic or extra-synaptic sites, thus affecting receptor mobility and synaptic strength.  相似文献   

12.
Benke D  Zeilhofer HU 《The EMBO journal》2012,31(15):3234-3236
EMBO J 31: 3239–3251 (2012); published online June122012It is now well established that G protein-coupled receptors can exist not only as homodimers, but also as heterodimers or higher order oligomers. However, whether and how dimerization of the receptors is regulated is poorly understood. In this issue of The EMBO Journal, the team of Marc Landry provides evidence for an intriguing mechanism by which—under pathological conditions—GABAB receptor heterodimers at the cell surface are disrupted and thereby inactivated. An impressive set of experiments thus reveals a novel mechanism regulating the number of functional GABAB receptors in the plasma membrane and shows that the receptor heterodimer may not be as stable as we previously thought.It is evident that dimerization and oligomerization at least of class A and C G protein-coupled receptors (GPCRs) play important roles in permitting or enhancing their cell surface trafficking (Milligan, 2010). The assembly process is thought to serve as a quality control mechanism to ensure that only fully mature and functional receptors reach the plasma membrane. The prototype of an obligatory heterodimer among GPCRs is the GABAB receptor, which controls excitability of neurons by mediating slow inhibitory neurotransmission (Gassmann and Bettler, 2012). Functional GABAB receptors are built from two related proteins termed as GABAB1 and GABAB2. Although both subunits display a similar structural organization—with a large extracellular domain containing a Venus fly-trap structure, seven transmembrane domains and a large intracellular located C-terminal domain—they serve distinct, complementary functions. GABAB1 binds the orthosteric ligands whereas GABAB2 recruits the G protein and is required for cell surface trafficking of the receptor complex by masking an ER retention signal present in the C-terminal domain of GABAB1. This is a striking example that dimerization is essential for and control of expression of a functional GPCR. The availability of GABAB receptors at the cell surface is also determined by receptor trafficking, which includes endocytosis, recycling and degradation of the receptors (Benke, 2010). To maintain the required number of cell surface receptors for signalling under a given physiological status, all levels of receptor trafficking need to be precisely balanced. Changing the balance of the different trafficking mechanisms is one means to adjust receptor numbers to changing physiological conditions. An example of such regulation is the recently uncovered mechanism of the glutamate receptor-mediated downregulation of GABAB receptors where the balance of recycling and degradation of the receptors is shifted towards degradation (Benke et al, 2012). Laffray et al (2012) propose a novel and unexpected mechanism regulating the number of functional receptors at the cell surface. This mechanism is operative in vivo under pathological conditions and is based on the disruption of GABAB receptor heterodimers present in the plasma membrane by the GABAB1 interacting protein 14-3-3ζ.Seven members of 14-3-3 proteins (14-3-3β, γ, ɛ, ζ, η, σ and τ) are ubiquitously expressed in mammals. 14-3-3 proteins bind predominantly to phosphoserine and phosphothreonine containing sequences and interact with hundreds of different partners to regulate a variety of cellular processes ranging from protein trafficking, apoptosis, cell cycle, signal transduction, cell adhesion and metabolism. It is therefore not surprising that alterations in the expression levels of 14-3-3 proteins and/or changes in the interaction status with target proteins are increasingly observed in diseases such as cancer, neurodegenerative diseases and epilepsy (Zhao et al, 2011).Among 14-3-3 proteins, 14-3-3ζ interacts with the C-terminal domain of GABAB1 and has been shown in vitro to inhibit the heterodimerization of GABAB1 and GABAB2 C-terminal domains (Couve et al, 2001). However, the physiological and potential pathological function of this interaction was entirely unresolved. In a rat model of neuropathic pain (spinal nerve ligation), Laffray et al (2012) observed a significant upregulation of 14-3-3ζ selectively in the ipsilateral dorsal horn of the lumbar spinal cord, the area where nociceptive signal processing in response to the injury takes place. Using several complementary methodologies including coimmunoprecipitation, colocalization immunofluorescence analysis, electron microscopy and two-photon fluorescence lifetime imaging, the authors demonstrated in vitro and in vivo that upregulation of 14-3-3ζ results in an increased interaction with GABAB1 in the plasma membrane and in a concomitant loss of GABAB1/GABAB2 association. This finding suggests that 14-3-3ζ disrupts existing heterodimers in the plasma membrane (Figure 1). As a consequence, the increased GABAB1/14-3-3ζ interaction rendered cell surface GABAB receptors non-functional and impaired GABAB receptor signalling.Open in a separate windowFigure 1Novel mechanism regulating GABAB receptor signalling by disrupting the functional receptor heterodimer via interaction with 14-3-3ζ. Functional GABAB receptors are obligatory heterodimers built from GABAB1 and GABAB2 subunits. Under normal conditions, binding of GABA to the Venus fly trap-like structure in the N-terminal domain of GABAB1 activates Gi/o proteins recruited by GABAB2 and thereby modulates distinct effector systems (adenylyl cyclases, potassium channels and voltage-gated Ca2+ channels). After induction of neuropathic pain by spinal nerve ligation, 14-3-3ζ is selectively upregulated in the spinal dorsal horn where painful sensory signals are processed and transmitted to the brain. 14-3-3ζ binds to the C-terminal domain of GABAB1 and disrupts by a yet-to-be identified mechanism the receptor dimer. This results in non-fuctional receptors and prevents GABAB receptor signalling.The main unresolved and extremely interesting issue concerns the mechanism of heterodimer disruption by 14-3-3ζ. The interaction site of 14-3-3ζ partially overlaps with the coiled-coil domain in the C-terminal domain of GABAB1 (Couve et al, 2001). Coiled-coil domains are protein–protein interaction sites and are one of the domains thought to be involved in the heterodimerization of GABAB1 and GABAB2. The most obvious mechanism is a direct competition of 14-3-3ζ and GABAB2 for interaction with GABAB1. 14-3-3 proteins are inherently rigid proteins able to stabilize a given conformation after binding to its partner protein (Obsil and Obsilova, 2011). Thus, binding of 14-3-3ζ might arrest GABAB1 in a conformation that is non-permissive for GABAB2 heterodimerization. However, the apparent affinity of the interaction of 14-3-3ζ with GABAB1 is rather low and relatively high concentrations of 14-3-3ζ are required to prevent heterodimerization of GABAB1 and GABAB2 C-terminal domains in vitro (Couve et al, 2001). Given the relatively moderate increase of 14-3-3ζ in neuropathic spinal cord, a direct competition mechanism per se appears unlikely. On the other hand, 14-3-3 proteins predominantly bind to motifs containing phosphorserine and phosphothreonine. Therefore, phosphorylation of GABAB1 within the 14-3-3ζ binding site might thus foster the GABAB1/14-3-3ζ interaction. In this regard, it would be important to test whether serine or threonine residues within the 14-3-3ζ binding site are phosphorylated in chronic pain states and whether phosphorylation is required for 14-3-3ζ interaction.An alternative mechanism may be based on the scaffolding properties of 14-3-3 proteins. 14-3-3 proteins act as dimers and thus harbour at least two protein interaction sites (Obsil and Obsilova, 2011). Therefore, 14-3-3ζ may target a second protein or a protein complex to GABAB receptors that forces the receptor complex to dissociate and prevent reassociation. Proteomic analyses of isolated GABAB1/14-3-3ζ complexes are needed to address this issue.Another important aspect of the paper is that it sheds some light on the involvement of GABAB receptors in neuropathic pain. So far, there is no coherent picture on the contribution of GABAB receptors to chronic pain states. However, there is increasing evidence that diminished GABAB receptor activity due to downregulation of the receptors might play a role in at least some models of neuropathic pain (Zeilhofer et al, 2012). Although there might be distinct mechanisms downregulating functional GABAB receptors in chronic pain conditions, disruption of GABAB receptor heterodimers via upregulation of 14-3-3ζ appears to be a contributing factor. In their neuropathic pain model, Laffray et al (2012) observed a diminished analgesic activity of the intrathecally injected GABAB receptor agonist baclofen. Preventing the binding of 14-3-3ζ to GABAB receptors via knocking-down 14-3-3ζ with siRNA or by using a synthetic peptide disrupting the GABAB1/14-3-3 interaction restored expression of GABAB receptor heterodimers in the plasma membrane and consequently enhanced the analgesic effect of baclofen. Even more important, disruption of the 14-3-3ζ/GABAB1 interaction by injection of the interfering synthetic peptide alone in the absence of baclofen partially reversed pain in the neuropathic rats. This finding implies that diminished GABAB receptor signalling contributes to the expression of neuropathic pain. These results might be a starting point for a therapeutic strategy to reduce neuropathic pain based on reversing the GABAB1/14-3-3ζ interaction. There are already small molecule inhibitors of 14-3-3 protein–protein interactions under development (Zhao et al, 2011), which might be useful for testing the feasibility of such an approach.  相似文献   

13.
Understanding the mechanisms that control synaptic efficacy through the availability of neurotransmitter receptors depends on uncovering their specific intracellular trafficking routes. γ-Aminobutyric acid type B (GABAB) receptors (GABABRs) are obligatory heteromers present at dendritic excitatory and inhibitory postsynaptic sites. It is unknown whether synthesis and assembly of GABABRs occur in the somatic endoplasmic reticulum (ER) followed by vesicular transport to dendrites or whether somatic synthesis is followed by independent transport of the subunits for assembly and ER export throughout the somatodendritic compartment. To discriminate between these possibilities we studied the association of GABABR subunits in dendrites of hippocampal neurons combining live fluorescence microscopy, biochemistry, quantitative colocalization, and bimolecular fluorescent complementation. We demonstrate that GABABR subunits are segregated and differentially mobile in dendritic intracellular compartments and that a high proportion of non-associated intracellular subunits exist in the brain. Assembled heteromers are preferentially located at the plasma membrane, but blockade of ER exit results in their intracellular accumulation in the cell body and dendrites. We propose that GABABR subunits assemble in the ER and are exported from the ER throughout the neuron prior to insertion at the plasma membrane. Our results are consistent with a bulk flow of segregated subunits through the ER and rule out a post-Golgi vesicular transport of preassembled GABABRs.The efficacy of synaptic transmission depends on the intracellular trafficking of neurotransmitter receptors (1, 2). The trafficking of glutamatergic and GABAA6 receptors has been extensively studied, and their implications for synaptic plasticity have been well documented (3, 4). For example, differential trafficking of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors modifies synaptic strength and influences experience-dependent plasticity in vivo (5). The molecular mechanisms that govern the trafficking of metabotropic GABABRs and their consequences for synaptic inhibition remain less clear. In particular, limited information is available regarding the relationship between the trafficking of GABABRs and the topological complexity of the secretory pathway in neurons.GABABRs mediate the slow component of synaptic inhibition by acting on pre- and postsynaptic targets (68). They are implicated in epilepsy, anxiety, stress, sleep disorders, nociception, depression, and cognition (9). They also represent attractive targets for the treatment of withdrawal symptoms from drugs of addiction such as cocaine (10). They are obligatory heteromers composed of GABABR1 and GABABR2 subunits. GABABR1 contains an RXR-type sequence in the intracellular C-terminal domain that functions as an ER retention motif (11, 12). The ER retention sequence is masked upon assembly with GABABR2 resulting in the appearance of functional receptors at the plasma membrane. Only GABABR1 binds GABA with high affinity, whereas G protein signaling is exclusively mediated by the second and third intracellular loops of GABABR2 (1315). GABABRs are located in dendrites and axons, but their distribution does not coincide with the active zone or the postsynaptic density. Rather, they are adjacent to both compartments constituting perisynaptic receptors (16, 17).If GABABR subunits are synthesized in the soma, at least two possibilities exist for their anterograde transport, assembly, and insertion in dendrites. First, the subunits may be synthesized in the cell body, assembled in the somatic ER, and targeted preassembled in post-Golgi vesicles to their site of insertion in dendrites. Alternatively, they may be synthesized in the soma and transported through the ER membrane as non-heteromeric subunits. In the latter scenario, newly assembled receptors may exit the ER throughout the somatodendritic compartment prior to insertion at the plasma membrane and diffuse laterally for retention at functional sites. No evidence exists to discriminate between these possibilities. We reasoned that a prevalence of associated subunits in post-Golgi vesicles in dendrites would favor the first alternative, whereas the existence of non-associated subunits in intracellular compartments would support a somatodendritic assembly mechanism. Here we explore the presence of associated GABABR subunits using fluorescence recovery after photobleaching (FRAP), biochemistry, and quantitative colocalization. In addition, we report for the first time the use of BiFC (18) to study GABABR assembly in neurons. Our results demonstrate that GABABR subunits are differentially mobile in dendrites and that a high proportion of non-associated subunits prevail in an intracellular fraction of the adult brain. They also show that GABABR subunits are heteromeric at the plasma membrane but segregated in intracellular compartments of dendrites of hippocampal neurons. Importantly, treatment with brefeldin A (BFA) or interference of the coatomer protein complex II impair ER export and result in the accumulation of assembled subunits in intracellular compartments throughout the somatodendritic arbor. We conclude that GABABR subunits are synthesized in the soma and remain segregated in intracellular compartments prior to somatodendritic assembly. Our observations rule out a post-Golgi vesicular transport of preassembled GABABRs and suggest an alternative mechanism of receptor targeting.  相似文献   

14.
Emerging evidence suggests that functional γ-aminobutyric acid B receptors (GABABRs) are expressed by astrocytes within the mammalian brain. GABABRs are heterodimeric G-protein-coupled receptors that are composed of R1/R2 subunits. To date, they have been characterized in neurons as the principal mediators of sustained inhibitory signaling; however their roles in astrocytic physiology have been ill defined. Here we reveal that the cytoplasmic tail of the GABABR2 subunit binds directly to the astrocytic protein glutamine synthetase (GS) and that this interaction determines the subcellular localization of GS. We further demonstrate that the binding of GS to GABABR2 increases the steady state expression levels of GS in heterologous cells and in mouse primary astrocyte culture. Mechanistically this increased stability of GS in the presence of GABABR2 occurs via reduced proteasomal degradation. Collectively, our results suggest a novel role for GABABRs as regulators of GS stability. Given the critical role that GS plays in the glutamine-glutamate cycle, astrocytic GABABRs may play a critical role in supporting both inhibitory and excitatory neurotransmission.  相似文献   

15.
A GABAA receptor (GABAAR) α1 subunit mutation, A322D (AD), causes an autosomal dominant form of juvenile myoclonic epilepsy (ADJME). Previous studies demonstrated that the mutation caused α1(AD) subunit misfolding and rapid degradation, reducing its total and surface expression substantially. Here, we determined the effects of the residual α1(AD) subunit expression on wild type GABAAR expression to determine whether the AD mutation conferred a dominant negative effect. We found that although the α1(AD) subunit did not substitute for wild type α1 subunits on the cell surface, it reduced the surface expression of α1β2γ2 and α3β2γ2 receptors by associating with the wild type subunits within the endoplasmic reticulum and preventing them from trafficking to the cell surface. The α1(AD) subunit reduced surface expression of α3β2γ2 receptors by a greater amount than α1β2γ2 receptors, thus altering cell surface GABAAR composition. When transfected into cultured cortical neurons, the α1(AD) subunit altered the time course of miniature inhibitory postsynaptic current kinetics and reduced miniature inhibitory postsynaptic current amplitudes. These findings demonstrated that, in addition to causing a heterozygous loss of function of α1(AD) subunits, this epilepsy mutation also elicited a modest dominant negative effect that likely shapes the epilepsy phenotype.  相似文献   

16.
The GABAA receptors are the major inhibitory receptors in the brain and are localized at both synaptic and extrasynaptic membranes. Synaptic GABAA receptors mediate phasic inhibition, whereas extrasynaptic GABAA receptors mediate tonic inhibition. Both phasic and tonic inhibitions regulate neuronal activity, but whether they regulate each other is not very clear. Here, we investigated the functional interaction between synaptic and extrasynaptic GABAA receptors through various molecular manipulations. Overexpression of extrasynaptic α6β3δ-GABAA receptors in mouse hippocampal pyramidal neurons significantly increased tonic currents. Surprisingly, the increase of tonic inhibition was accompanied by a dramatic reduction of the phasic inhibition, suggesting a possible homeostatic regulation of the total inhibition. Overexpressing the α6 subunit alone induced an up-regulation of δ subunit expression and suppressed phasic inhibition similar to overexpressing the α6β3δ subunits. Interestingly, blocking all GABAA receptors after overexpressing α6β3δ receptors could not restore the synaptic GABAergic transmission, suggesting that receptor activation is not required for the homeostatic interplay. Furthermore, insertion of a gephyrin-binding-site (GBS) into the α6 and δ subunits recruited α6GBSβ3δGBS receptors to postsynaptic sites but failed to rescue synaptic GABAergic transmission. Thus, it is not the positional effect of extrasynaptic α6β3δ receptors that causes the down-regulation of phasic inhibition. Overexpressing α5β3γ2 subunits similarly reduced synaptic GABAergic transmission. We propose a working model that both synaptic and extrasynaptic GABAA receptors may compete for limited receptor slots on the plasma membrane to maintain a homeostatic range of the total inhibition.  相似文献   

17.
δ subunit-containing γ-aminobutyric acid, type A (GABAA)receptors are expressed extrasynaptically and mediate tonic inhibition. In cerebellar granule cells, they often form receptors together with α1 and/or α6 subunits. We were interested in determining the architecture of receptors containing both subunits. We predefined the subunit arrangement of several different GABAA receptor pentamers by concatenation. These receptors composed of α1, α6, β3, and δ subunits were expressed in Xenopus oocytes. Currents elicited in response to GABA were determined in the presence and absence of 3α,21-dihydroxy-5α-pregnan-20-one (THDOC) or ethanol, or currents were elicited by 4,5,6,7-tetrahydroisoxazolo[5,4-c]-pyridin-3-ol (THIP). Several subunit configurations formed active channels. We therefore conclude that δ can assume multiple positions in a receptor pentamer made up of α1, α6, β3, and δ subunits. The different receptors differ in their functional properties. Functional expression of one receptor type was only evident in the combined presence of the neurosteroid THDOC with the channel agonist GABA. Most, but not all, receptors active with GABA/THDOC responded to THIP. None of the receptors was modulated by ethanol concentrations up to 30 mm. Several observations point to a preferred position of δ subunits between two α subunits in α1α6β3δ receptors. This property is shared by α1β3δ and α6β3δ receptors, but there are differences in the additionally expressed isoforms.  相似文献   

18.
GABA-gated chloride channels (GABAARs) trafficking is involved in the regulation of fast inhibitory transmission. Here, we took advantage of a γ2(R43Q) subunit mutation linked to epilepsy in humans that considerably reduces the number of GABAARs on the cell surface to better understand the trafficking of GABAARs. Using recombinant expression in cultured rat hippocampal neurons and COS-7 cells, we showed that receptors containing γ2(R43Q) were addressed to the cell membrane but underwent clathrin-mediated dynamin-dependent endocytosis. The γ2(R43Q)-dependent endocytosis was reduced by GABAAR antagonists. These data, in addition to a new homology model, suggested that a conformational change in the extracellular domain of γ2(R43Q)-containing GABAARs increased their internalization. This led us to show that endogenous and recombinant wild-type GABAAR endocytosis in both cultured neurons and COS-7 cells can be amplified by their agonists. These findings revealed not only a direct relationship between endocytosis of GABAARs and a genetic neurological disorder but also that trafficking of these receptors can be modulated by their agonist.  相似文献   

19.
Synaptic GABAA receptors (GABAARs) mediate most of the inhibitory neurotransmission in the brain. The majority of these receptors are comprised of α1, β2, and γ2 subunits. The amygdala, a structure involved in processing emotional stimuli, expresses α2 and γ1 subunits at high levels. The effect of these subunits on GABAAR-mediated synaptic transmission is not known. Understanding the influence of these subunits on GABAAR-mediated synaptic currents may help in identifying the roles and locations of amygdala synapses that contain these subunits. Here, we describe the biophysical and synaptic properties of pure populations of α1β2γ2, α2β2γ2, α1β2γ1 and α2β2γ1 GABAARs. Their synaptic properties were examined in engineered synapses, whereas their kinetic properties were studied using rapid agonist application, and single channel recordings. All macropatch currents activated rapidly (<1 ms) and deactivated as a function of the α-subunit, with α2-containing GABAARs consistently deactivating ∼10-fold more slowly. Single channel analysis revealed that the slower current decay of α2-containing GABAARs was due to longer burst durations at low GABA concentrations, corresponding to a ∼4-fold higher affinity for GABA. Synaptic currents revealed a different pattern of activation and deactivation to that of macropatch data. The inclusion of α2 and γ1 subunits slowed both the activation and deactivation rates, suggesting that receptors containing these subunits cluster more diffusely at synapses. Switching the intracellular domains of the γ2 and γ1 subunits substantiated this inference. Because this region determines post-synaptic localization, we hypothesize that GABAARs containing γ1 and γ2 use different mechanisms for synaptic clustering.  相似文献   

20.
γ-Aminobutyric acid type B (GABA(B)) receptors are important for slow synaptic inhibition in the CNS. The efficacy of inhibition is directly related to the stability of cell surface receptors. For GABA(B) receptors, heterodimerization between R1 and R2 subunits is critical for cell surface expression and signaling, but how this determines the rate and extent of receptor internalization is unknown. Here, we insert a high affinity α-bungarotoxin binding site into the N terminus of the R2 subunit and reveal its dominant role in regulating the internalization of GABA(B) receptors in live cells. To simultaneously study R1a and R2 trafficking, a new α-bungarotoxin binding site-labeling technique was used, allowing α-bungarotoxin conjugated to different fluorophores to selectively label R1a and R2 subunits. This approach demonstrated that R1a and R2 are internalized as dimers. In heterologous expression systems and neurons, the rates and extents of internalization for R1aR2 heteromers and R2 homomers are similar, suggesting a regulatory role for R2 in determining cell surface receptor stability. The fast internalization rate of R1a, which has been engineered to exit the endoplasmic reticulum, was slowed to that of R2 by truncating the R1a C-terminal tail or by removing a dileucine motif in its coiled-coil domain. Slowing the rate of internalization by co-assembly with R2 represents a novel role for GPCR heterodimerization whereby R2 subunits, via their C terminus coiled-coil domain, mask a dileucine motif on R1a subunits to determine the surface stability of the GABA(B) receptor.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号