Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Dev Neurobiol ; 76(2): 166-89, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26014473

ABSTRACT

Relating changes in gene expression to discrete developmental events remains an elusive challenge in neuroscience, in part because most neural territories are comprised of multiple cell types that mature over extended periods of time. The medial nucleus of the trapezoid body (MNTB) is an attractive vertebrate model system that contains a nearly homogeneous population of neurons, which are innervated by large glutamatergic nerve terminals called calyces of Held (CH). Key steps in maturation of CHs and MNTB neurons, including CH growth and competition, occur very quickly for most cells between postnatal days (P)2 and P6. Therefore, we characterized genome-wide changes in this system, with dense temporal sampling during the first postnatal week. We identified 541 genes whose expression changed significantly between P0-6 and clustered them into eight groups based on temporal expression profiles. Candidate genes from each of the eight profile groups were validated in separate samples by qPCR. Our tissue sample permitted comparison of known glial and neuronal transcripts and revealed that monotonically increasing or decreasing expression profiles tended to be associated with glia and neurons, respectively. Gene ontology revealed enrichment of genes involved in axon pathfinding, cell differentiation, cell adhesion and extracellular matrix. The latter category included elements of perineuronal nets, a prominent feature of MNTB neurons that is morphologically distinct by P6, when CH growth and competition are resolved onto nearly all MNTB neurons. These results provide a genetic framework for investigation of general mechanisms responsible for nerve terminal growth and maturation.


Subject(s)
Auditory Pathways/physiology , Axons/metabolism , Gene Expression Regulation, Developmental/physiology , Neuroglia/metabolism , Neurons/metabolism , Synapses/physiology , Animals , Brain Stem/growth & development , Extracellular Matrix/metabolism , Mice
2.
J Neurosci ; 33(32): 12954-69, 2013 Aug 07.
Article in English | MEDLINE | ID: mdl-23926251

ABSTRACT

Hallmark features of neural circuit development include early exuberant innervation followed by competition and pruning to mature innervation topography. Several neural systems, including the neuromuscular junction and climbing fiber innervation of Purkinje cells, are models to study neural development in part because they establish a recognizable endpoint of monoinnervation of their targets and because the presynaptic terminals are large and easily monitored. We demonstrate here that calyx of Held (CH) innervation of its target, which forms a key element of auditory brainstem binaural circuitry, exhibits all of these characteristics. To investigate CH development, we made the first application of serial block-face scanning electron microscopy to neural development with fine temporal resolution and thereby accomplished the first time series for 3D ultrastructural analysis of neural circuit formation. This approach revealed a growth spurt of added apposed surface area (ASA)>200 µm2/d centered on a single age at postnatal day 3 in mice and an initial rapid phase of growth and competition that resolved to monoinnervation in two-thirds of cells within 3 d. This rapid growth occurred in parallel with an increase in action potential threshold, which may mediate selection of the strongest input as the winning competitor. ASAs of competing inputs were segregated on the cell body surface. These data suggest mechanisms to select "winning" inputs by regional reinforcement of postsynaptic membrane to mediate size and strength of competing synaptic inputs.


Subject(s)
Brain Stem/cytology , Models, Neurological , Neurons/physiology , Synapses/physiology , Age Factors , Animals , Animals, Newborn , Axons/ultrastructure , Brain Stem/growth & development , Calcium/metabolism , Computer Simulation , Embryo, Mammalian , Excitatory Postsynaptic Potentials/physiology , Female , Imaging, Three-Dimensional , In Vitro Techniques , Male , Mice , Microscopy, Electron, Scanning , Nerve Net/ultrastructure , Neurons/ultrastructure , Nonlinear Dynamics , Pregnancy , Presynaptic Terminals/ultrastructure , Statistics, Nonparametric , Synapses/ultrastructure , Synaptic Vesicles/ultrastructure
3.
Front Psychiatry ; 4: 39, 2013.
Article in English | MEDLINE | ID: mdl-23761764

ABSTRACT

Pain alters opioid reinforcement, presumably via neuroadaptations within ascending pain pathways interacting with the limbic system. Nerve injury increases expression of glutamate receptors and their associated Homer scaffolding proteins throughout the pain processing pathway. Homer proteins, and their associated glutamate receptors, regulate behavioral sensitivity to various addictive drugs. Thus, we investigated a potential role for Homers in the interactions between pain and drug reward in mice. Chronic constriction injury (CCI) of the sciatic nerve elevated Homer1b/c and/or Homer2a/b expression within all mesolimbic structures examined and for the most part, the Homer increases coincided with elevated mGluR5, GluN2A/B, and the activational state of various down-stream kinases. Behaviorally, CCI mice showed pain hypersensitivity and a conditioned place-aversion (CPA) at a low heroin dose that supported conditioned place-preference (CPP) in naïve controls. Null mutations of Homer1a, Homer1, and Homer2, as well as transgenic disruption of mGluR5-Homer interactions, either attenuated or completely blocked low-dose heroin CPP, and none of the CCI mutant strains exhibited heroin-induced CPA. However, heroin CPP did not depend upon full Homer1c expression within the nucleus accumbens (NAC), as CPP occurred in controls infused locally with small hairpin RNA-Homer1c, although intra-NAC and/or intrathecal cDNA-Homer1c, -Homer1a, and -Homer2b infusions (to best mimic CCI's effects) were sufficient to blunt heroin CPP in uninjured mice. However, arguing against a simple role for CCI-induced increases in either spinal or NAC Homer expression for heroin CPA, cDNA infusion of our various cDNA constructs either did not affect (intrathecal) or attenuated (NAC) heroin CPA. Together, these data implicate increases in glutamate receptor/Homer/kinase activity within limbic structures, perhaps outside the NAC, as possibly critical for switching the incentive motivational properties of heroin following nerve injury, which has relevance for opioid psychopharmacology in individuals suffering from neuropathic pain.

4.
Alcohol ; 46(4): 377-87, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22444953

ABSTRACT

Neuronal activity dependent pentraxin (Narp) interacts with α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) glutamate receptors to facilitate excitatory synapse formation by aggregating them at established synapses. Alcohol is well-characterized to influence central glutamatergic transmission, including AMPA receptor function. Herein, we examined the influence of injected and ingested alcohol upon Narp protein expression, as well as basal Narp expression in mouse lines selectively bred for high blood alcohol concentrations under limited access conditions. Alcohol up-regulated accumbens Narp levels, concomitant with increases in levels of the GluR1 AMPA receptor subunit. However, accumbens Narp or GluR1 levels did not vary as a function of selectively bred genotype. We next employed a Narp knock-out (KO) strategy to begin to understand the behavioral relevance of alcohol-induced changes in protein expression in several assays of alcohol reward. Compared to wild-type mice, Narp KO animals: fail to escalate daily intake of high alcohol concentrations under free-access conditions; shift their preference away from high alcohol concentrations with repeated alcohol experience; exhibit a conditioned place-aversion in response to the repeated pairing of 3 g/kg alcohol with a distinct environment and fail to exhibit alcohol-induced locomotor hyperactivity following repeated alcohol treatment. Narp deletion did not influence the daily intake of either food or water, nor did it alter any aspect of spontaneous or alcohol-induced motor activity, including the development of tolerance to its motor-impairing effects with repeated treatment. Taken together, these data indicate that Narp induction, and presumably subsequent aggregation of AMPA receptors, may be important for neuroplasticity within limbic subcircuits mediating or maintaining the rewarding properties of alcohol.


Subject(s)
Behavior, Animal/drug effects , C-Reactive Protein/drug effects , Ethanol/pharmacology , Nerve Tissue Proteins/drug effects , Nucleus Accumbens/drug effects , Receptors, AMPA/drug effects , Up-Regulation/genetics , Animals , Behavior, Animal/physiology , C-Reactive Protein/genetics , C-Reactive Protein/metabolism , Conditioning, Operant/drug effects , Ethanol/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Neurons/drug effects , Neurons/metabolism , Nucleus Accumbens/metabolism , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Reward , Up-Regulation/drug effects , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism
5.
Cell ; 147(3): 615-28, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22036569

ABSTRACT

Assemblies of ß-amyloid (Aß) peptides are pathological mediators of Alzheimer's Disease (AD) and are produced by the sequential cleavages of amyloid precursor protein (APP) by ß-secretase (BACE1) and γ-secretase. The generation of Aß is coupled to neuronal activity, but the molecular basis is unknown. Here, we report that the immediate early gene Arc is required for activity-dependent generation of Aß. Arc is a postsynaptic protein that recruits endophilin2/3 and dynamin to early/recycling endosomes that traffic AMPA receptors to reduce synaptic strength in both hebbian and non-hebbian forms of plasticity. The Arc-endosome also traffics APP and BACE1, and Arc physically associates with presenilin1 (PS1) to regulate γ-secretase trafficking and confer activity dependence. Genetic deletion of Arc reduces Aß load in a transgenic mouse model of AD. In concert with the finding that patients with AD can express anomalously high levels of Arc, we hypothesize that Arc participates in the pathogenesis of AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Cytoskeletal Proteins/metabolism , Endosomes/metabolism , Nerve Tissue Proteins/metabolism , Protein Transport , Animals , Cell Membrane/metabolism , Humans , Mice , Mice, Knockout
6.
Neuron ; 68(6): 1128-42, 2010 Dec 22.
Article in English | MEDLINE | ID: mdl-21172614

ABSTRACT

Homeostatic scaling is a non-Hebbian form of neural plasticity that maintains neuronal excitability and informational content of synaptic arrays in the face of changes of network activity. Here, we demonstrate that homeostatic scaling is dependent on group I metabotropic glutamate receptor activation that is mediated by the immediate early gene Homer1a. Homer1a is transiently upregulated during increases in network activity and evokes agonist-independent signaling of group I mGluRs that scales down the expression of synaptic AMPA receptors. Homer1a effects are dynamic and play a role in the induction of scaling. Similar to mGluR-LTD, Homer1a-dependent scaling involves a reduction of tyrosine phosphorylation of GluA2 (GluR2), but is distinct in that it exploits a unique signaling property of group I mGluR to confer cell-wide, agonist-independent activation of the receptor. These studies reveal an elegant interplay of mechanisms that underlie Hebbian and non-Hebbian plasticity.


Subject(s)
Carrier Proteins/physiology , Homeostasis/physiology , Neuronal Plasticity/physiology , Receptors, Metabotropic Glutamate/metabolism , Animals , Carrier Proteins/genetics , Cells, Cultured , Cerebral Cortex/metabolism , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , Homeostasis/genetics , Homer Scaffolding Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/genetics , Signal Transduction/genetics , Signal Transduction/physiology
7.
J Neurosci ; 29(27): 8655-68, 2009 Jul 08.
Article in English | MEDLINE | ID: mdl-19587272

ABSTRACT

The glutamate receptor-associated protein Homer2 regulates alcohol-induced neuroplasticity within the nucleus accumbens (NAC), but the precise intracellular signaling cascades involved are not known. This study examined the role for NAC metabotropic glutamate receptor (mGluR)-Homer2-phosphatidylinositol 3-kinase (PI3K) signaling in regulating excessive alcohol consumption within the context of the scheduled high alcohol consumption (SHAC) model of binge alcohol drinking. Repeated bouts of binge drinking ( approximately 1.5 g/kg per 30 min) elevated NAC Homer2a/b expression and increased PI3K activity in this region. Virus-mediated knockdown of NAC Homer2b expression attenuated alcohol intake, as did an intra-NAC infusion of the mGluR5 antagonist MPEP [2-methyl-6-(phenylethynyl)pyridine hydrochloride] (0.1-1 microg/side) and the PI3K antagonist wortmannin (50 ng/side), supporting necessary roles for mGluR5/Homer2/PI3K in binge alcohol drinking. Moreover, when compared with wild-type littermates, transgenic mice with an F1128R point mutation in mGluR5 that markedly reduces Homer binding exhibited a 50% reduction in binge alcohol drinking, which was related to reduced NAC basal PI3K activity. Consistent with the hypothesis that mGluR5-Homer-PI3K signaling may be a mechanism governing excessive alcohol intake, the "anti-binge" effects of MPEP and wortmannin were not additive, nor were they observed in the mGluR5(F1128R) transgenic mice. Finally, mice genetically selected for a high versus low SHAC phenotype differed in NAC mGluR, Homer2, and PI3K activity, consistent with the hypothesis that augmented NAC mGluR5-Homer2-PI3K signaling predisposes a high binge alcohol-drinking phenotype. Together, these data point to an important role for NAC mGluR5-Homer2-PI3K signaling in regulating binge-like alcohol consumption that has relevance for our understanding of the neurobiology of alcoholism and its pharmacotherapy.


Subject(s)
Alcoholism/metabolism , Carrier Proteins/physiology , Ethanol/toxicity , Nucleus Accumbens/metabolism , Phosphatidylinositol 3-Kinases/physiology , Receptors, Metabotropic Glutamate/physiology , Signal Transduction/drug effects , Up-Regulation/physiology , Alcoholism/enzymology , Alcoholism/genetics , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Homer Scaffolding Proteins , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nucleus Accumbens/drug effects , Nucleus Accumbens/enzymology , Phenotype , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphatidylinositol 3-Kinases/genetics , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/biosynthesis , Receptors, Metabotropic Glutamate/genetics , Signal Transduction/physiology , Up-Regulation/drug effects
8.
Science ; 319(5862): 476-81, 2008 Jan 25.
Article in English | MEDLINE | ID: mdl-18218901

ABSTRACT

T cell receptor (TCR) and costimulatory receptor (CD28) signals cooperate in activating T cells, although understanding of how these pathways are themselves regulated is incomplete. We found that Homer2 and Homer3, members of the Homer family of cytoplasmic scaffolding proteins, are negative regulators of T cell activation. This is achieved through binding of nuclear factor of activated T cells (NFAT) and by competing with calcineurin. Homer-NFAT binding was also antagonized by active serine-threonine kinase AKT, thereby enhancing TCR signaling via calcineurin-dependent dephosphorylation of NFAT. This corresponded with changes in cytokine expression and an increase in effector-memory T cell populations in Homer-deficient mice, which also developed autoimmune-like pathology. These results demonstrate a further means by which costimulatory signals are regulated to control self-reactivity.


Subject(s)
Carrier Proteins/metabolism , Lymphocyte Activation , NFATC Transcription Factors/metabolism , T-Lymphocytes/immunology , Animals , CD28 Antigens/immunology , CD3 Complex/immunology , Calcineurin/metabolism , Calcium/metabolism , Carrier Proteins/chemistry , Cell Line , Cells, Cultured , Crystallography, X-Ray , Homer Scaffolding Proteins , Humans , Jurkat Cells , Mice , Mice, Knockout , NFATC Transcription Factors/chemistry , Phosphorylation , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Fusion Proteins/metabolism , Signal Transduction , T-Lymphocytes/metabolism
9.
J Neurosci ; 27(23): 6249-60, 2007 Jun 06.
Article in English | MEDLINE | ID: mdl-17553998

ABSTRACT

Group I metabotropic glutamate receptors (mGluRs), mGluR1 and mGluR5, regulate activity in the globus pallidus (GP) and subthalamic nucleus (STN). To test whether the localization of group I mGluRs is altered in parkinsonism, we used immunoelectron microscopy to analyze the subcellular and subsynaptic distribution of mGluR1a and mGluR5 in GP and STN of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice. Homer1 and Homer2 knock-out mice were used to assess the role of Homer in MPTP-induced redistribution of group I mGluRs. We also examined the effects of MPTP on the expression levels of group I mGluRs and Homer proteins in GP and striatum. MPTP treatment significantly reduced the expression levels of H1a and mGluR1a in striatum but not in GP. Although light microscopy did not reveal noticeable effects of MPTP treatment on the distribution of group I mGluRs and Homer proteins in GP and STN, specific changes in the ultrastructural localization of mGluR1a were found in MPTP-treated normal and Homer knock-out mice. An increase in the expression of presynaptic axonal and terminal mGluR1a labeling and an increased level of mGluR1a immunoreactivity in the postsynaptic specialization of putative GABAergic synapses were among the most significant effects induced by dopamine depletion. However, neither of these changes was found for mGluR5, which, in contrast, displayed complex regulatory alterations in its subsynaptic distribution in response to Homer deletion and MPTP lesion. Thus, nigrostriatal dopaminergic lesion and Homer deletion lead to changes in the trafficking of group I mGluRs in vivo that are specific to receptor subtypes and brain areas.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Carrier Proteins/genetics , Corpus Striatum/metabolism , Globus Pallidus/metabolism , Receptors, Metabotropic Glutamate/biosynthesis , Subthalamic Nucleus/metabolism , Animals , Carrier Proteins/metabolism , Corpus Striatum/chemistry , Corpus Striatum/drug effects , Female , Gene Deletion , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Globus Pallidus/chemistry , Globus Pallidus/drug effects , Homer Scaffolding Proteins , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Metabotropic Glutamate/analysis , Receptors, Metabotropic Glutamate/genetics , Subthalamic Nucleus/chemistry , Subthalamic Nucleus/drug effects
10.
J Biol Chem ; 282(19): 14283-90, 2007 May 11.
Article in English | MEDLINE | ID: mdl-17355963

ABSTRACT

Excitation-contraction (E-C) coupling and Ca(2+)-induced Ca(2+) release in smooth and cardiac muscles is mediated by the L-type Ca(2+) channel isoform Ca(v)1.2 and the ryanodine receptor isoform RyR2. Although physical coupling between Ca(v)1.1 and RyR1 in skeletal muscle is well established, it is generally assumed that Ca(v)1.2 and RyR2 do not directly communicate either passively or dynamically during E-C coupling. In the present work, we re-examined this assumption by studying E-C coupling in the detrusor muscle of wild type and Homer1(-/-) mice and by demonstrating a Homer1-mediated dynamic interaction between Ca(v)1.2 and RyR2 using the split green fluorescent protein technique. Deletion of Homer1 in mice (but not of Homer2 or Homer3) resulted in impaired urinary bladder function, which was associated with higher sensitivity of the detrusor muscle to muscarinic stimulation and membrane depolarization. This was not due to an altered expression or function of RyR2 and Ca(v)1.2. Most notably, expression of Ca(v)1.2 and RyR2 tagged with the complementary C- and N-terminal halves of green fluorescent protein and in the presence and absence of Homer1 isoforms revealed that H1a and H1b/c reciprocally modulates a dynamic interaction between Ca(v)1.2 and RyR2 to regulate the intensity of Ca(2+)-induced Ca(2+) release and its dependence on membrane depolarization. These findings define the molecular basis of a "two-state" model of E-C coupling by Ca(v)1.2 and RyR2. In one state, Ca(v)1.2 couples to RyR2 by H1b/c, which results in reduced responsiveness to membrane depolarization and in the other state H1a uncouples Ca(v)1.2 and RyR2 to enhance responsiveness to membrane depolarization. These findings reveal an unexpected and novel mode of interaction and communication between Ca(v)1.2 and RyR2 with important implications for the regulation of smooth and possibly cardiac muscle E-C coupling.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Signaling , Calcium/metabolism , Carrier Proteins/physiology , Muscle Contraction , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Carrier Proteins/genetics , Cells, Cultured , Homer Scaffolding Proteins , Kidney/metabolism , Membrane Microdomains , Mice , Mice, Knockout , Urinary Bladder/metabolism , Urinary Incontinence
11.
J Biol Chem ; 281(43): 32540-9, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-16887806

ABSTRACT

Store-operated Ca(2+) channels (SOCs) mediate receptor-stimulated Ca(2+) influx. Accumulating evidence indicates that members of the transient receptor potential (TRP) channel family are components of SOCs in mammalian cells. Agonist stimulation activates SOCs and TRP channels directly and by inducing translocation of channels in intracellular vesicles to the plasma membrane (PM). The mechanism of TRP channel translocation in response to store depletion and agonist stimulation is not known. Here we use TRPC3 as a model to show that IP(3) and the scaffold Homer 1 (H1) regulate the rate of translocation and retrieval of TRPC3 from the PM. In resting cells, TRPC3 exists in TRPC3-H1b/c-IP(3)Rs complexes that are located in part at the PM and in part in intracellular vesicles. Binding of IP(3) to the IP(3)Rs dissociates the interaction between IP(3)Rs and H1 but not between H1 and TRPC3 to form IP(3)Rs-TRPC3-H1b/c. TIRFM and biotinylation assays show robust receptor- and store-dependent translocation of the TRPC3 to the PM and their retrieval upon termination of cell stimulation. The translocation requires depletion of stored Ca(2+) and is prevented by inhibition of the IP(3)Rs. In HEK293, dissociating the H1b/c-IP(3)R complex with H1a results in TRPC3 translocation to the PM, where it is spontaneously active. The TRPC3-H1b/c-IP(3)Rs complex is reconstituted by infusing H1c into these cells. Reconstitution is inhibited by IP(3). Deletion of H1 in mice markedly reduces the rates of translocation and retrieval of TRPC3. Conversely, infusion of H1c into H1(-/-) cells eliminates spontaneous channel activity and increases the rate of channel activation by agonist stimulation. The effects of H1c are inhibited by IP(3). These findings together with our earlier studies demonstrating gating of TRPC3 by IP(3)Rs were used to develop a model in which assembly of the TRPC3-H1b/c-IP(3)Rs complexes by H1b/c mediates both the translocation of TRPC3-containing vesicles to the PM and gating of TRPC3 by IP(3)Rs.


Subject(s)
Carrier Proteins/metabolism , Cell Membrane/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , TRPC Cation Channels/metabolism , TRPC Cation Channels/physiology , Animals , Cell Culture Techniques , Cell Line , Collagenases/pharmacology , Homer Scaffolding Proteins , Humans , Mice , Models, Biological , Pancreas/cytology , Pancreas/drug effects , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , TRPC Cation Channels/genetics , Trypsin/pharmacology
12.
J Neurosci ; 25(30): 7054-61, 2005 Jul 27.
Article in English | MEDLINE | ID: mdl-16049182

ABSTRACT

Homer proteins are integral to the assembly of proteins regulating glutamate signaling and synaptic plasticity. Constitutive Homer2 gene deletion [knock-out (KO)] and rescue with adeno-associated viral (AAV) transfection of Homer2b was used to demonstrate the importance of Homer proteins in neuroplasticity produced by repeated ethanol (EtOH) administration. Homer2 KO mice avoided drinking high concentrations of EtOH and did not develop place preference or locomotor sensitization after repeated EtOH administration. The deficient behavioral plasticity to EtOH after Homer2 deletion was paralleled by a lack of augmentation in the rise in extracellular dopamine and glutamate elicited by repeated EtOH injections. The genotypic differences in EtOH-induced change in behavior and neurochemistry were essentially reversed by AAV-mediated transfection of Homer2b into accumbens cells including, differences in EtOH preference, locomotor sensitization, and EtOH-induced elevations in extracellular glutamate and dopamine. These data demonstrate a necessary and active role for accumbens Homer2 expression in regulating EtOH-induced behavioral and cellular neuroplasticity.


Subject(s)
Carrier Proteins/physiology , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Alcohol Drinking/genetics , Alcohol Drinking/physiopathology , Animals , Avoidance Learning/drug effects , Carrier Proteins/genetics , Carrier Proteins/pharmacology , Drug Synergism , Excitatory Amino Acid Antagonists/pharmacology , Female , Gene Deletion , Homer Scaffolding Proteins , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology , Piperazines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Reflex/drug effects
13.
Neuron ; 43(3): 401-13, 2004 Aug 05.
Article in English | MEDLINE | ID: mdl-15294147

ABSTRACT

Drug addiction involves complex interactions between pharmacology and learning in genetically susceptible individuals. Members of the Homer gene family are regulated by acute and chronic cocaine administration. Here, we report that deletion of Homer1 or Homer2 in mice caused the same increase in sensitivity to cocaine-induced locomotion, conditioned reward, and augmented extracellular glutamate in nucleus accumbens as that elicited by withdrawal from repeated cocaine administration. Moreover, adeno-associated virus-mediated restoration of Homer2 in the accumbens of Homer2 KO mice reversed the cocaine-sensitized phenotype. Further analysis of Homer2 KO mice revealed extensive additional behavioral and neurochemical similarities to cocaine-sensitized animals, including accelerated acquisition of cocaine self-administration and altered regulation of glutamate by metabotropic glutamate receptors and cystine/glutamate exchange. These data show that Homer deletion mimics the behavioral and neurochemical phenotype produced by repeated cocaine administration and implicate Homer in regulating addiction to cocaine.


Subject(s)
Carrier Proteins/physiology , Cocaine-Related Disorders/metabolism , Cocaine/administration & dosage , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cocaine-Related Disorders/genetics , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Dose-Response Relationship, Drug , Glutamic Acid/metabolism , Homer Scaffolding Proteins , Locomotion/drug effects , Locomotion/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Self Administration
14.
Cell ; 114(6): 777-89, 2003 Sep 19.
Article in English | MEDLINE | ID: mdl-14505576

ABSTRACT

Receptor signaling at the plasma membrane often releases calcium from intracellular stores. For example, inositol triphosphate (IP3) produced by receptor-coupled phospholipase C activates an intracellular store calcium channel, the IP(3)R. Conversely, stores can induce extracellular calcium to enter the cell through plasma membrane channels, too. How this "reverse" coupling works was unclear, but store IP(3)Rs were proposed to bind and regulate plasma membrane TRP cation channels. Here, we demonstrate that the adaptor protein, termed Homer, facilitates a physical association between TRPC1 and the IP(3)R that is required for the TRP channel to respond to signals. The TRPC1-Homer-IP(3)R complex is dynamic and its disassembly parallels TRPC1 channel activation. Homer's action depends on its ability to crosslink and is blocked by the dominant-negative immediate early gene form, H1a. Since H1a is transcriptionally regulated by cellular activity, this mechanism can affect both short and long-term regulation of TRPC1 function.


Subject(s)
Calcium Channels/metabolism , Carrier Proteins/metabolism , Ion Channel Gating/physiology , Neuropeptides/metabolism , Receptors, Cell Surface/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Binding Sites/physiology , Brain/metabolism , Calcium/metabolism , Calcium Channels/genetics , Calcium Signaling/genetics , Cell Membrane/metabolism , Cells, Cultured , Homer Scaffolding Proteins , Inositol 1,4,5-Trisphosphate Receptors , Macromolecular Substances , Membrane Potentials/genetics , Mice , Mice, Mutant Strains , Mutation/genetics , Patch-Clamp Techniques , Protein Binding/genetics , Protein Structure, Tertiary/physiology , Rats , TRPC Cation Channels
15.
J Cell Biol ; 162(2): 293-303, 2003 Jul 21.
Article in English | MEDLINE | ID: mdl-12860966

ABSTRACT

Homers are scaffolding proteins that bind G protein-coupled receptors (GPCRs), inositol 1,4,5-triphosphate (IP3) receptors (IP3Rs), ryanodine receptors, and TRP channels. However, their role in Ca2+ signaling in vivo is not known. Characterization of Ca2+ signaling in pancreatic acinar cells from Homer2-/- and Homer3-/- mice showed that Homer 3 has no discernible role in Ca2+ signaling in these cells. In contrast, we found that Homer 2 tunes intensity of Ca2+ signaling by GPCRs to regulate the frequency of [Ca2+]i oscillations. Thus, deletion of Homer 2 increased stimulus intensity by increasing the potency for agonists acting on various GPCRs to activate PLCbeta and evoke Ca2+ release and oscillations. This was not due to aberrant localization of IP3Rs in cellular microdomains or IP3R channel activity. Rather, deletion of Homer 2 reduced the effectiveness of exogenous regulators of G proteins signaling proteins (RGS) to inhibit Ca2+ signaling in vivo. Moreover, Homer 2 preferentially bound to PLCbeta in pancreatic acini and brain extracts and stimulated GAP activity of RGS4 and of PLCbeta in an in vitro reconstitution system, with minimal effect on PLCbeta-mediated PIP2 hydrolysis. These findings describe a novel, unexpected function of Homer proteins, demonstrate that RGS proteins and PLCbeta GAP activities are regulated functions, and provide a molecular mechanism for tuning signal intensity generated by GPCRs and, thus, the characteristics of [Ca2+]i oscillations.


Subject(s)
Carrier Proteins/metabolism , GTP-Binding Proteins/metabolism , GTPase-Activating Proteins/metabolism , Isoenzymes/metabolism , Neuropeptides/metabolism , RGS Proteins/metabolism , Type C Phospholipases/metabolism , Animals , Bombesin/pharmacology , Calcium/metabolism , Calcium/pharmacokinetics , Calcium Signaling , Calcium-Transporting ATPases/antagonists & inhibitors , Calcium-Transporting ATPases/metabolism , Carbachol/pharmacology , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cholecystokinin/pharmacology , Cholinergic Agonists/pharmacology , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/agonists , Gene Deletion , Homer Scaffolding Proteins , Indoles/pharmacology , Mice , Mice, Knockout , Neuropeptides/chemistry , Neuropeptides/genetics , Pancreas/cytology , Pancreas/drug effects , Pancreas/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase C beta , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases
SELECTION OF CITATIONS
SEARCH DETAIL
...