Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
Add more filters










Publication year range
1.
Neuroscience ; 295: 58-71, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-25800309

ABSTRACT

Weight-loss dieting often leads to loss of control, rebound weight gain, and is a risk factor for binge pathology. Based on findings that food restriction (FR) upregulates sucrose-induced trafficking of glutamatergic AMPA receptors to the nucleus accumbens (NAc) postsynaptic density (PSD), this study was an initial test of the hypothesis that episodic "breakthrough" intake of forbidden food during dieting interacts with upregulated mechanisms of synaptic plasticity to increase reward-driven feeding. Ad libitum (AL) fed and FR subjects consumed a limited amount of 10% sucrose, or had access to water, every other day for 10 occasions. Beginning three weeks after return of FR rats to AL feeding, when 24-h chow intake and rate of body weight gain had normalized, subjects with a history of sucrose intake during FR consumed more sucrose during a four week intermittent access protocol than the two AL groups and the group that had access to water during FR. In an experiment that substituted noncontingent administration of d-amphetamine for sucrose, FR subjects displayed an enhanced locomotor response during active FR but a blunted response, relative to AL subjects, during recovery from FR. This result suggests that the enduring increase in sucrose consumption is unlikely to be explained by residual enhancing effects of FR on dopamine signaling. In a biochemical experiment which paralleled the sucrose behavioral experiment, rats with a history of sucrose intake during FR displayed increased abundance of pSer845-GluA1, GluA2, and GluA3 in the NAc PSD relative to rats with a history of FR without sucrose access and rats that had been AL throughout, whether they had a history of episodic sucrose intake or not. A history of FR, with or without a history of sucrose intake, was associated with increased abundance of GluA1. A terminal 15-min bout of sucrose intake produced a further increase in pSer845-GluA1 and GluA2 in subjects with a history of sucrose intake during FR. Generally, neither a history of sucrose intake nor a terminal bout of sucrose intake affected AMPA receptor abundance in the NAc PSD of AL subjects. Together, these results are consistent with the hypothesis, but the functional contribution of increased synaptic incorporation of AMPA receptors remains to be established.


Subject(s)
Feeding Behavior/physiology , Food Deprivation/physiology , Nucleus Accumbens/cytology , Post-Synaptic Density/metabolism , Receptors, AMPA/metabolism , Sucrose/administration & dosage , Sweetening Agents/administration & dosage , Amphetamine/pharmacology , Analysis of Variance , Animals , Body Weight/drug effects , Body Weight/physiology , Central Nervous System Stimulants/pharmacology , Eating/drug effects , Eating/physiology , Feeding Behavior/drug effects , Hyperkinesis/chemically induced , Male , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Subcellular Fractions , Time Factors
2.
J Neurovirol ; 19(4): 314-27, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23868513

ABSTRACT

In the era of greatly improved pharmacological treatment of HIV infection through highly active antiretroviral therapy (HAART), HIV patients experience reduced viral loads, reduced opportunistic infections, increased CD4+ T cell count, and greater life expectancy. Although life expectancy is increased, patients often develop neurological disturbances that may persist for long periods, seriously jeopardizing quality of life and adherence to the medication protocols of HAART. For these reasons, HIV-associated neurological disorders have gained importance in both clinical and basic investigations of HIV infection. Depression is the most prevalent neuropsychiatric disorder among people living with HIV. In this review, we discuss how HIV can predispose infected individuals to depression by several interrelated mechanisms. These include inducing chronic elevation of cytokines through activation of microglia and astrocytes; decreasing monoaminergic function; inducing neurotoxicity, especially in dopaminergic neurons; and reducing brain-derived neurotrophic factor. These viral pathways interact with psychosocial factors to create the depressive state. HIV depression has a great impact on quality of life and implementation of antiretroviral therapy, and thus, recognition of these modes of action is significant for understanding HIV neuropathology and for selecting modalities for pharmacologic treatment.


Subject(s)
Depression/physiopathology , Depression/virology , HIV Infections/psychology , Humans , Nervous System Diseases/virology
3.
Neuroscience ; 165(4): 1074-86, 2010 Feb 17.
Article in English | MEDLINE | ID: mdl-19931598

ABSTRACT

Previous findings suggest that neuroadaptations downstream of D-1 dopamine (DA) receptor stimulation in nucleus accumbens (NAc) are involved in the enhancement of drug reward by chronic food restriction (FR). Given the high co-expression of D-1 and GluR1 AMPA receptors in NAc, and the regulation of GluR1 channel conductance and trafficking by D-1-linked intracellular signaling cascades, the present study examined effects of the D-1 agonist, SKF-82958, on NAc GluR1 phosphorylation, intracranial electrical self-stimulation reward (ICSS), and reversibility of reward effects by a polyamine GluR1 antagonist, 1-NA-spermine, in ad libitum fed (AL) and FR rats. Systemically administered SKF-82958, or brief ingestion of a 10% sucrose solution, increased NAc GluR1 phosphorylation on Ser845, but not Ser831, with a greater effect in FR than AL rats. Microinjection of SKF-82958 in NAc shell produced a reward-potentiating effect that was greater in FR than AL rats, and was reversed by co-injection of 1-NA-spermine. GluR1 abundance in whole cell and synaptosomal fractions of NAc did not differ between feeding groups, and microinjection of AMPA, while affecting ICSS, did not exert greater effects in FR than AL rats. These results suggest a role of NAc GluR1 in the reward-potentiating effect of D-1 DA receptor stimulation and its enhancement by FR. Moreover, GluR1 involvement appears to occur downstream of D-1 DA receptor stimulation rather than reflecting a basal increase in GluR1 expression or function. Based on evidence that phosphorylation of GluR1 on Ser845 primes synaptic strengthening, the present results may reflect a mechanism via which FR normally facilitates reward-related learning to re-align instrumental behavior with environmental contingencies under the pressure of negative energy balance.


Subject(s)
Food Deprivation/physiology , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology , Receptors, AMPA/metabolism , Receptors, Dopamine D1/metabolism , Reward , Animals , Benzazepines/pharmacology , Dietary Sucrose , Dopamine Agonists/pharmacology , Eating/physiology , Electric Stimulation , Excitatory Amino Acid Antagonists/pharmacology , Male , Neurons/drug effects , Neurons/physiology , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Receptors, AMPA/antagonists & inhibitors , Receptors, Dopamine D1/agonists , Self Administration , Spermine/pharmacology , Synaptosomes/drug effects , Synaptosomes/physiology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism
4.
Mol Cell Neurosci ; 35(3): 470-81, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17544687

ABSTRACT

RNA editing modifies the GluR2 AMPA receptor subunit pore loop at the Q/R site and limits receptor Ca(2+) permeability. Editing failure is implicated in neurodegenerative diseases, including amyotrophic lateral sclerosis. We show that channels with unedited GluR2 are highly toxic in cultured hippocampal neurons. Toxicity exceeds that of other Ca(2+)-permeable AMPA receptor types and is influenced by agonist binding site mutations, ability to desensitize, and extracellular Ca(2+) levels. Significantly, toxicity also depends on GluR2's constitutive surface trafficking, a function dependent on GluR2 C-terminal domain interaction with NSF, a specialized chaperone. We have exploited the interaction between unedited GluR2 and NSF to reduce GluR2 surface levels. We show that a peptide that blocks the GluR2-NSF interaction reduces unedited GluR2 toxicity by reducing receptor surface expression. Peptide block of trafficking provides a model for design of drugs to reduce unedited GluR2 excitotoxicity in neurodegenerative diseases that result from editing failure.


Subject(s)
Calcium/metabolism , Excitatory Amino Acid Agonists/pharmacology , Permeability/drug effects , Receptors, AMPA/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/toxicity , Animals , Cell Death/drug effects , Dose-Response Relationship, Drug , Embryo, Mammalian , Hippocampus/cytology , In Situ Nick-End Labeling/methods , Mutagenesis/physiology , Neurons/drug effects , Neurons/metabolism , Protein Transport/drug effects , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Receptors, AMPA/genetics , Sindbis Virus/physiology , Time Factors , Transcriptional Activation
5.
J Neurosci ; 21(15): 5417-28, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11466413

ABSTRACT

The PICK1 protein interacts in neurons with the AMPA-type glutamate receptor subunit 2 (GluR2) and with several other membrane receptors via its single PDZ domain. We show that PICK1 also binds in neurons and in heterologous cells to protein kinase Calpha (PKCalpha) and that the interaction is highly dependent on the activation of the kinase. The formation of PICK1-PKCalpha complexes is strongly induced by TPA, and PICK1-PKCalpha complexes are cotargeted with PICK1-GluR2 complexes to spines, where GluR2 is found to be phosphorylated by PKC on serine 880. PICK1 also reduces the plasma membrane levels of the GluR2 subunit, consistent with a targeting function of PICK1 and a PKC-facilitated release of GluR2 from the synaptic anchoring proteins ABP and GRIP. This work indicates that PICK1 functions as a targeting and transport protein that directs the activated form of PKCalpha to GluR2 in spines, leading to the activity-dependent release of GluR2 from synaptic anchor proteins and the PICK1-dependent transport of GluR2 from the synaptic membrane.


Subject(s)
Carrier Proteins/metabolism , Hippocampus/metabolism , Isoenzymes/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Protein Kinase C/metabolism , Receptors, AMPA/metabolism , 3T3 Cells , Animals , Carrier Proteins/genetics , Cell Cycle Proteins , Cells, Cultured , Cricetinae , Cytoskeletal Proteins , Dimerization , Enzyme Activation/drug effects , Enzyme Activators/pharmacology , Hippocampus/cytology , Humans , Isoenzymes/genetics , Macromolecular Substances , Mice , Models, Molecular , Mutagenesis, Site-Directed , Neurons/cytology , Nuclear Proteins/genetics , Protein Binding/drug effects , Protein Binding/genetics , Protein Kinase C/genetics , Protein Kinase C-alpha , Protein Transport/physiology , Rats , Receptor Aggregation , Receptors, AMPA/genetics , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Two-Hybrid System Techniques
6.
Mech Dev ; 101(1-2): 47-59, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11231058

ABSTRACT

A transgenic mouse model was used to examine the roles of the murine transcription factors Pax-3 and Mitf in melanocyte development. Transgenic mice expressing beta-galactosidase from the dopachrome tautomerase (Dct) promoter were generated and found to express the transgene in developing melanoblasts as early as embryonic day (E) 9.5. These mice express the transgene in a pattern characteristic of endogenous Dct expression. Transgenic mice were intercrossed with two murine coat color mutants, Splotch (Sp), containing a mutation in the murine Pax3 gene, and Mitf(mi), with a mutation in the basic-helix-loop-helix-leucine zipper gene Mitf. Transgenic heterozygous mutant animals were crossed to generate transgenic embryos for analysis. Examination of beta-galactosidase-expressing melanoblasts in mutant embryos reveals that Mitf is required in vivo for survival of melanoblasts up to the migration staging area in neural crest development. Examination of Mitf(mi)/+ embryos shows that there are diminished numbers of melanoblasts in the heterozygous state early in melanocyte development, consistent with a gene dosage-dependent effect upon cell survival. However, quantification and analysis of melanoblast growth during the migratory phase suggests that melanoblasts then increase in number more rapidly in the heterozygous embryo. In contrast to Mitf(mi)/Mitf(mi) embryos, Sp/Sp embryos exhibit melanoblasts that have migrated to characteristic locations along the melanoblast migratory pathway, but are greatly reduced in number compared to control littermates. Together, these results support a model for melanocyte development whereby Pax3 is required to expand a pool of committed melanoblasts or restricted progenitor cells early in development, whereas Mitf facilitates survival of the melanoblast in a gene dosage-dependent manner within and immediately after emigration from the dorsal neural tube, and may also directly or indirectly affect the rate at which melanoblast number increases during dorsolateral pathway migration.


Subject(s)
DNA-Binding Proteins/physiology , Melanocytes/metabolism , Melanocytes/physiology , Transcription Factors , Animals , Cell Movement , Cell Survival , Crosses, Genetic , Dose-Response Relationship, Drug , Female , Heterozygote , Intramolecular Oxidoreductases/genetics , Male , Mice , Mice, Transgenic , Microphthalmia-Associated Transcription Factor , Microscopy, Fluorescence , Models, Biological , Mutation , PAX3 Transcription Factor , Paired Box Transcription Factors , Promoter Regions, Genetic , Time Factors , Transgenes , beta-Galactosidase/metabolism
7.
J Neurosci ; 21(2): 495-503, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11160429

ABSTRACT

Excitatory synaptic currents in the mammalian brain are typically mediated by the neurotransmitter glutamate, acting at AMPA receptors. We used immunocytochemistry to investigate the distribution of AMPA receptor-binding protein (ABP) in the cerebral neocortex. ABP was most prominent in pyramidal neurons, although it was also present (at lower levels) in interneurons. ABP and its putative binding partners, the GluR2/3 subunits of the AMPA receptor, exhibited prominent cellular colocalization. Under appropriate processing conditions, colocalization could also be documented in puncta, many of which could be recognized as dendritic spines. However, a sizable minority of GluR2/3-positive puncta were immunonegative for ABP. Because glutamate receptor-interacting protein (GRIP) may also anchor GluR2, we studied the relative distribution of ABP and GRIP. There was extensive colocalization of these two antigens at the cellular level, although GRIP, unlike ABP, was strongest in nonpyramidal neurons. Different parts of a single dendrite could stain selectively for ABP or GRIP. To further characterize this heterogeneity, we investigated punctate staining of neuropil using synaptophysin and the membrane tracer DiA to identify probable synapses. Some puncta were comparably positive for both ABP and GRIP, but the majority were strongly positive for one antigen and only weakly positive or immunonegative for the other. This heterogeneity could be seen even within adjacent spines of a single dendrite. These data suggest that ABP may act as a scaffold for AMPA receptors either in concert with or independently from GRIP.


Subject(s)
Carrier Proteins/metabolism , Neocortex/metabolism , Nerve Tissue Proteins/metabolism , Animals , Contraindications , Dendrites/metabolism , Dendrites/ultrastructure , Immunohistochemistry , Intercellular Signaling Peptides and Proteins , Interneurons/cytology , Interneurons/metabolism , Intracellular Signaling Peptides and Proteins , Male , Neocortex/cytology , Neuropil/metabolism , Neuropil/ultrastructure , Organ Specificity , Pyramidal Cells/cytology , Pyramidal Cells/metabolism , Pyridinium Compounds , Rats , Rats, Sprague-Dawley , Synapses/metabolism , Synaptophysin/metabolism
8.
Neurosurgery ; 47(6): 1400-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11126911

ABSTRACT

OBJECTIVE: Few molecular targets are both consistently and selectively expressed in a majority of central nervous system (CNS) neoplasms. Receptor tyrosine kinases have been implicated in brain tumor oncogenesis. We previously isolated one such receptor, discoidin domain receptor-1 (DDR1), from high-grade pediatric brain tumors. Here, we analyze the cellular origin and distribution of DDR1 expression in human brain tumors and its expression in tumor cells relative to surrounding brain. METHODS: By use of a digoxigenin-labeled DDR1 riboprobe, we investigated the expression of DDR1 messenger ribonucleic acid in a prospective series of 30 resected human primary and metastatic brain neoplasms, nonneoplastic human brain, and mouse embryonic brain, as well as a mouse glioblastoma model, by in situ hybridization. RESULTS: All the high-grade primary brain and metastatic brain tumors showed unequivocal, intense DDR1 expression within the majority of tumor cells, whereas expression was not observed in hyperplastic tumor blood vessels, normal brain blood vessels, inflammatory cells, or in the normal brain tissue that surrounded the tumor. Receptor expression was limited to tumor cells located within solid tumor tissue. Overall, 27 of 29 resected CNS tumors exhibited tumor cell-specific DDR1 expression, whereas one specimen composed of isolated glioblastoma cells within invaded brain parenchyma showed no detectable staining for this receptor. DDR1 was also expressed preferentially in the ventricular zone (a region of highly proliferating precursor cells) of mice at embryonic Day 15.5. CONCLUSION: We found that DDR1 is consistently expressed in all high-grade brain neoplasms studied and is selective for tumor cells in the specimens analyzed. The expression of DDR1 by tumor cells of CNS neoplasms and by primitive cells of the embryonic ventricular zone suggests that DDR1 is a potentially useful marker of tumor cells within the CNS.


Subject(s)
Brain Neoplasms/metabolism , Genes, Tumor Suppressor , Nuclear Proteins , Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases , Receptors, Mitogen/metabolism , Adolescent , Adult , Aged , Animals , Brain/cytology , Brain/embryology , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Cerebral Ventricle Neoplasms/embryology , Cerebral Ventricle Neoplasms/metabolism , Cerebral Ventricle Neoplasms/pathology , Child , Child, Preschool , Discoidin Domain Receptors , Embryo, Mammalian/metabolism , Female , Glioma/embryology , Glioma/pathology , Humans , Male , Mice/embryology , Middle Aged , Neoplasm Transplantation , Protein-Tyrosine Kinases/genetics , Proteins , RNA, Messenger/metabolism , Receptors, Mitogen/genetics , Tumor Cells, Cultured
9.
Neuron ; 27(2): 313-25, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10985351

ABSTRACT

We studied the role of PDZ proteins GRIP, ABP, and PICK1 in GluR2 AMPA receptor trafficking. An epitope-tagged MycGluR2 subunit, when expressed in hippocampal cultured neurons, was specifically targeted to the synaptic surface. With the mutant MycGluR2delta1-10, which lacks the PDZ binding site, the overall dendritic intracellular transport and the synaptic surface targeting were not affected. However, over time, Myc-GluR2delta1-10 accumulated at synapses significantly less than MycGluR2. Notably, a single residue substitution, S880A, which blocks binding to ABP/GRIP but not to PICK1, reduced synaptic accumulation to the same extent as the PDZ site truncation. We conclude that the association of GluR2 with ABP and/or GRIP but not PICK1 is essential for maintaining the synaptic surface accumulation of the receptor, possibly by limiting its endocytotic rate.


Subject(s)
Carrier Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, AMPA/metabolism , Synaptic Membranes/metabolism , Adaptor Proteins, Signal Transducing , Animals , Binding Sites/genetics , Cells, Cultured , Cytoskeletal Proteins , Hippocampus/cytology , Hippocampus/metabolism , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Mutagenesis, Site-Directed , Neurons/cytology , Neurons/metabolism , Nuclear Proteins/metabolism , Protein Structure, Tertiary/genetics , Proto-Oncogene Proteins c-myc/genetics , Rats , Rats, Sprague-Dawley , Receptors, AMPA/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sindbis Virus/genetics
10.
Neuropharmacology ; 39(12): 2255-66, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10974309

ABSTRACT

The mechanisms by which the NMDA receptor (NMDAR) induces excitotoxicity were investigated using a novel assay. We quantitated the capacity of wild type and mutant receptors for cell killing in CHO cells and cultured cortical neurons by measuring the activity of a co-transfected firefly luciferase expression plasmid. NR1 subunit pore mutations that block Ca(2+) influx, and deletion of the NR1 cytoplasmic C-terminal domain, which functions in Ca(2+) regulation of receptor currents, decreased NMDAR mediated cell killing. We also transfected the NR1 pore mutants and C-terminal truncations in the presence of co-expressed exogenous wild type subunits. The pore and C-terminal truncation mutants acted in a dominant negative fashion and increased the survival of NMDAR-expressing CHO cells. Although physiological studies of similar NMDA receptor mutants have been carried out in heterologous cell lines, their functions in neurons remain relatively unknown. We show that expression of pore mutants and specific C terminal truncation mutants in cultured cortical neurons also exerts dominant negative function and protects these primary cells from endogenous receptor induced excitotoxic death. These results implicate positive actions of the selectivity filter and of the NR1 C-terminal domain in a Ca(2+)-dependent mechanism for NMDAR excitotoxicity. They also indicate that the mutant receptors which show diminished excitotoxicity and dominant negative action in heterologous cells can co-assemble with endogenous subunits in primary neurons and block NMDAR-dependent excitotoxic death.


Subject(s)
Excitatory Amino Acids/toxicity , Receptors, N-Methyl-D-Aspartate/physiology , Animals , CHO Cells , Cell Death/physiology , Cell Survival , Cerebral Cortex/cytology , Cricetinae , DNA/biosynthesis , DNA/genetics , Luciferases/genetics , Mutation/genetics , Neurons/drug effects , Plasmids/genetics , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Transfection
11.
J Invest Dermatol ; 115(1): 106-12, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10886516

ABSTRACT

The expression of the dopachrome tautomerase gene (Dct) and its protein product, tyrosinase-related protein-2, was studied in the cultured, phorbol-ester-dependent murine melanocyte cell line melan-a. Increased cell density was found to stimulate Dct expression both in cells stably transfected with a Dct promoter-lacZ construct and endogenously in nontransfected cells. Increased Dct expression under these conditions corresponds to increased tyrosinase-related protein-2 production. Tyrosinase-related protein-2 was found to exist in two distinct glycoforms with different endoglycosidase sensitivities. Density-dependent expression of tyrosinase-related protein-2 was independent of time of cell growth, cell proliferation, and soluble factors, implying that cell-cell contact is the important determinant governing increased Dct expression under these conditions. Tyrp1 gene expression and tyrosinase-related protein-1 production were also induced under similar conditions. The results show that cell-cell contact between melanocytes induces a coordinated response at both transcriptional and nontranscriptional levels that induces production of the tyrosinase-related proteins that have a significant role in melanization.


Subject(s)
Cell Count/drug effects , Intramolecular Oxidoreductases/genetics , Animals , Cell Communication , Cells, Cultured , Gene Expression Regulation , Glycosylation/drug effects , Intramolecular Oxidoreductases/biosynthesis , Phorbol Esters/pharmacology
12.
Proc Natl Acad Sci U S A ; 97(11): 6120-5, 2000 May 23.
Article in English | MEDLINE | ID: mdl-10823955

ABSTRACT

The GTPase dynamin I is required for synaptic vesicle (SV) endocytosis. Our observation that dynamin binds to the SV protein synaptophysin in a Ca(2+)-dependent fashion suggested the possibility that a dynamin/synaptophysin complex functions in SV recycling. In this paper we show that disruption of the dynamin/synaptophysin interaction by peptide injection into the squid giant synapse preterminal results in a decrease in transmitter release during high-frequency stimulation, indicating an inhibition of SV recycling. Electron microscopy of these synapses reveals a depletion of SVs, demonstrating a block of vesicle retrieval after fusion. In addition, we observed an increase in clathrin-coated vesicles, indicating that the peptide does not block clathrin-dependent endocytosis. We conclude that the dynamin/synaptophysin complex functions in a clathrin-independent mechanism of SV endocytosis that is required for efficient synaptic transmission.


Subject(s)
Endocytosis/physiology , Nerve Tissue Proteins/physiology , Synaptic Vesicles/physiology , Synaptophysin/physiology , Animals , Calcium/physiology , Clathrin/physiology , Decapodiformes , Dynamin I , Dynamins , GTP Phosphohydrolases/physiology , Macromolecular Substances , Membrane Fusion , Recombinant Fusion Proteins/physiology , Stellate Ganglion/cytology , Synaptic Transmission/physiology
13.
J Neurophysiol ; 82(6): 3367-77, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10601468

ABSTRACT

Peptide growth factors such as the neurotrophins and fibroblast growth factors have potent effects on synaptic transmission, development, and cell survival. We report that chronic (hours) treatment with basic fibroblast growth factor (FGF-2) potentiates Ca(2+)-dependent N-methyl-D-aspartate (NMDA) receptor inactivation in cultured hippocampal neurons. This effect is specific for the NMDA-subtype of ionotropic glutamate receptor and FGF-2. The potentiated inactivation requires ongoing protein synthesis during growth factor treatment and the activity of protein phosphatase 2B (PP2B or calcineurin) during agonist application. These results suggest a mechanism by which FGF-2 receptor signaling may regulate neuronal survival and synaptic plasticity.


Subject(s)
Calcium/physiology , Fibroblast Growth Factor 2/pharmacology , Hippocampus/drug effects , Neurons/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Actinin/physiology , Animals , Calcineurin/metabolism , Calcium Channels/physiology , Cells, Cultured , Electric Stimulation , Electrophysiology , Hippocampus/cytology , Immunohistochemistry , Ion Channel Gating/drug effects , Membrane Potentials/physiology , Nerve Tissue Proteins/metabolism , Patch-Clamp Techniques , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
14.
Ann N Y Acad Sci ; 868: 465-73, 1999 Apr 30.
Article in English | MEDLINE | ID: mdl-10414323

ABSTRACT

The synapse is a specialized cellular junction with an elaborate and highly evolved capacity for signal transduction. At excitatory synapses, the neurotransmitter glutamate is released from the presynaptic nerve terminal and stimulates several types of glutamate receptors in the postsynaptic membrane. These include the ionotropic receptors, which are glutamate-gated cation channels, and the metabotropic receptors, which are G protein-coupled seven-transmembrane receptors. The ionotropic glutamate receptors have received special attention because of growing evidence that changes in their synaptic abundance, posttranslational modification, or molecular interactions can provide long-term changes in synaptic strength. This review summarizes new information about the ionotropic glutamate receptors and relates receptor function to the organization of the postsynaptic membrane and the regulation of electrophysiologic and biochemical signaling at the synapse.


Subject(s)
Receptors, Kainic Acid/chemistry , Animals , Glutamic Acid/metabolism , Protein Binding , Protein-Tyrosine Kinases/metabolism , Receptors, AMPA/chemistry , Receptors, N-Methyl-D-Aspartate/chemistry , Synaptic Transmission/physiology , GluK2 Kainate Receptor
16.
Ann N Y Acad Sci ; 868: 561-4, 1999 Apr 30.
Article in English | MEDLINE | ID: mdl-10414337

ABSTRACT

We review the cloning of a novel AMPA receptor binding protein (ABP) that interacts with GluR2/3 and is homologous to GRIP. ABP is enriched in the PSD with GluR2 and is localized to the PSD by EM. ABP binds GluR2 via the C-terminal VXI motif through a Class I PDZ interaction. ABP and GRIP can also homo- and heteromultimerize. Thus, ABP and GRIP may be involved in AMPA receptor regulation and localization, by linking it to other cytoskeletal or signaling molecules. We suggest that the ABP/GRIP and PSD-95 families form distinct scaffolds that anchor, respectively, AMPA and NMDA receptors. We are currently investigating proteins that bind ABP and that may regulate the AMPA receptor.


Subject(s)
Carrier Proteins/genetics , Nerve Tissue Proteins/genetics , Receptors, AMPA/metabolism , Animals , Brain/metabolism , Carrier Proteins/chemistry , Cloning, Molecular , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Models, Molecular , Nerve Tissue Proteins/chemistry , Protein Binding , Rats
17.
J Biol Chem ; 274(2): 595-606, 1999 Jan 08.
Article in English | MEDLINE | ID: mdl-9872993

ABSTRACT

The basic region/helix-loop-helix/leucine zipper (B-HLH-LZ) oncoprotein c-Myc is abundant in proliferating cells and forms heterodimers with Max protein that bind to E-box sites in DNA and stimulate genes required for proliferation. A second B-HLH-LZ protein, Mxi1, is induced during terminal differentiation, and forms heterodimers with Max that also bind E-boxes but tether the mSin3 transcriptional repressor protein along with histone deacetylase thereby antagonizing Myc-dependent activation. We show that Mxi1 also antagonizes Myc by a second pathway, repression of transcription from the major c-myc promoter, P2. Repression was independent of Mxi1 binding to mSin3 but dependent on the Mxi1 LZ and COOH-terminal sequences, including putative casein kinase II phosphorylation sites. Repression targeted elements of the myc P2 promoter core (-35/+10), where it reversed transactivation by the constitutive transcription factor, USF. We show that Zn2+ induction of a stably transfected, metallothionein promoter-regulated mxi1 gene blocked the ability of serum to induce transcription of the endogenous c-myc gene and cell entry into S phase. Thus, induction of Mxi1 in terminally differentiating cells may block Myc function by repressing the c-myc gene P2 promoter, as well as by antagonizing Myc-dependent transactivation through E-boxes.


Subject(s)
DNA-Binding Proteins/metabolism , Genes, myc , Promoter Regions, Genetic , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins , Transcription Factors/metabolism , 3T3 Cells , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors , DNA Primers , DNA-Binding Proteins/genetics , Gene Expression Regulation , Histone Deacetylases , Mice , Sequence Deletion , Transcription Factors/genetics , Tumor Suppressor Proteins , Upstream Stimulatory Factors
18.
Cell Growth Differ ; 9(10): 827-36, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9790494

ABSTRACT

PC12 cells differentiate in response to nerve growth factor from a chromaffin cell to a sympathetic neuronal phenotype. Wnt-1 is a secreted signaling factor required for development of mammalian midbrain and cerebellum. PC12 cells transformed by Wnt-1 fail to express several differentiation-specific genes in response to nerve growth factor. We have previously shown that HES-1, a negative regulator of neuronal differentiation, is increased in Wnt-1/PC12 cells (P. S. Issack and E. B. Ziff. Altered expression of helix-loop-helix transcriptional regulators and cyclin D1 in Wnt-1-transformed PC12 cells. Cell Growth & Differ., 9: 837-845). Here, we show that the HES-1 promoter is more active in Wnt-1/PC12 cells relative to PC12 and that the binding sites for the transcription factor RBP-J kappa contribute to this induction. We also identify two additional promoter elements required for elevated HES-1 expression. One element binds Wnt-1-induced protein complexes in a sequence-specific manner. Identification of Wnt-1 responsive elements in potential target genes may provide clues to nuclear pathways regulated by Wnt-1.


Subject(s)
Gene Expression Regulation , Homeodomain Proteins/genetics , Neurons/cytology , Nuclear Proteins , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Zebrafish Proteins , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation , Cell Line, Transformed , DNA/metabolism , DNA-Binding Proteins/metabolism , Helix-Loop-Helix Motifs , Immunoglobulin J Recombination Signal Sequence-Binding Protein , Neurons/metabolism , PC12 Cells , Proto-Oncogene Proteins/physiology , Rats , Sequence Deletion , Transcription Factor HES-1 , Transcription Factors/metabolism , Transformation, Genetic , Wnt Proteins , Wnt1 Protein
19.
Cell Growth Differ ; 9(10): 837-45, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9790495

ABSTRACT

Nerve growth factor induces PC12 cells to differentiate from a chromaffin cell to a sympathetic neuronal phenotype. In contrast, PC12 cells, which stably express Wnt-1, a secreted signaling factor required for development of mammalian midbrain and cerebellum, fail to express differentiation-specific genes in response to nerve growth factor. Analysis of factors binding to E box-containing regulatory elements of the terminal differentiation gene encoding peripherin suggested a differentiation-specific control of expression of helix-loop-helix transcriptional regulators. Specifically, the MASH-1 (mammalian achaete-scute homologue) helix-loop-helix transcription factor, which plays a positive role in neuronal differentiation, is reduced in Wnt-1/PC12 cells, and HES-1, a negative regulator of MASH-1, is increased. These data suggest that the differentiation block may result from induction of HES-1. Wnt-1/PC12 cells also proliferate more rapidly and express increased levels of cyclin D1. Thus, Wnt-1 may block the differentiation and enhance the proliferation of PC12 cells by activating HES-1 and cyclin D1 and repressing MASH-1.


Subject(s)
Cyclin D1/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation , Homeodomain Proteins/genetics , Membrane Glycoproteins , Neurons/cytology , Proto-Oncogene Proteins/genetics , Transcription Factors/genetics , Zebrafish Proteins , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation , Cell Division , Cell Line, Transformed , Helix-Loop-Helix Motifs , Intermediate Filament Proteins/genetics , Nerve Tissue Proteins/genetics , Neurons/metabolism , PC12 Cells , Peripherins , Proto-Oncogene Proteins/physiology , Rats , Transcription Factor HES-1 , Transformation, Genetic , Wnt Proteins , Wnt1 Protein
20.
Neuron ; 21(3): 581-91, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9768844

ABSTRACT

We report the cloning of alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor-binding protein (ABP), a postsynaptic density (PSD) protein related to glutamate receptor-interacting protein (GRIP) with two sets of three PDZ domains, which binds the GluR2/3 AMPA receptor subunits. ABP exhibits widespread CNS expression and is found at the postsynaptic membrane. We show that the protein interactions of the ABP/GRIP family differ from the PSD-95 family, which binds N-methyl-D-aspartate (NMDA) receptors. ABP binds to the GluR2/3 C-terminal VKI-COOH motif via class II hydrophobic PDZ interactions, distinct from the class I PSD-95-NMDA receptor interaction. ABP and GRIP also form homo- and heteromultimers through PDZ-PDZ interactions but do not bind PSD-95. We suggest that the ABP/GRIP and PSD-95 families form distinct scaffolds that anchor, respectively, AMPA and NMDA receptors.


Subject(s)
Brain/metabolism , Carrier Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, AMPA/chemistry , Receptors, AMPA/metabolism , Spinal Cord/metabolism , Synapses/metabolism , Amino Acid Sequence , Animals , Binding Sites , Brain/cytology , Brain/growth & development , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cloning, Molecular , Gene Expression Regulation, Developmental , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Macromolecular Substances , Models, Molecular , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Peptide Fragments/chemistry , Protein Conformation , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Spinal Cord/cytology , Spinal Cord/growth & development , Synapses/ultrastructure , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...