Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 291
Filter
Add more filters










Publication year range
1.
J Evol Biol ; 29(3): 560-71, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26663312

ABSTRACT

Social animals vary in their ability to compete with group members over shared resources and also vary in their cooperative efforts to produce these resources. Competition among groups can promote within-group cooperation, but many existing models of intergroup cooperation do not explicitly account for observations that group members invest differentially in cooperation and that there are often within-group competitive or power asymmetries. We present a game theoretic model of intergroup competition that investigates how such asymmetries affect within-group cooperation. In this model, group members adopt one of two roles, with relative competitive efficiency and the number of individuals varying between roles. Players in each role make simultaneous, coevolving decisions. The model predicts that although intergroup competition increases cooperative contributions to group resources by both roles, contributions are predominantly from individuals in the less competitively efficient role, whereas individuals in the more competitively efficient role generally gain the larger share of these resources. When asymmetry in relative competitive efficiency is greater, a group's per capita cooperation (averaged across both roles) is higher, due to increased cooperation from the competitively inferior individuals. For extreme asymmetry in relative competitive efficiency, per capita cooperation is highest in groups with a single competitively superior individual and many competitively inferior individuals, because the latter acquiesce and invest in cooperation rather than within-group competition. These predictions are consistent with observed features of many societies, such as monogynous Hymenoptera with many workers and caste dimorphism.


Subject(s)
Behavior, Animal , Models, Biological , Animals , Competitive Behavior , Cooperative Behavior , Hymenoptera
2.
Neuroscience ; 153(4): 1126-34, 2008 Jun 02.
Article in English | MEDLINE | ID: mdl-18440154

ABSTRACT

Posttraumatic stress disorder (PTSD) is one of the most common psychiatric disorders. Despite the extensive study of the neurobiological correlates of this disorder, the underlying mechanisms of PTSD are still poorly understood. Recently, a study demonstrated that dexamethasone (Dex), a synthetic glucocorticoid, can up-regulate p11, known as S100A10-protein which is down-regulated in patients with depression, (Yao et al., 1999; Huang et al., 2003) a common comorbid disorder in PTSD. These observations led to our hypothesis that traumatic stress may alter expression of p11 mediated through a glucocorticoid receptor. Here, we demonstrate that inescapable tail shock increased both prefrontal cortical p11 mRNA levels and plasma corticosterone levels in rats. We also found that Dex up-regulated p11 expression in SH-SY5Y cells through glucocorticoid response elements (GREs) within the p11 promoter. This response was attenuated by either RU486, a glucocorticoid receptor (GR) antagonist or mutating two of three glucocorticoid response elements (GRE2 and GRE3) in the p11 promoter. Finally, we showed that p11 mRNA levels were increased in postmortem prefrontal cortical tissue (area 46) of patients with PTSD. The data obtained from our work in a rat model of inescapable tail shock, a p11-transfected cell line and postmortem brain tissue from PTSD patients outline a possible mechanism by which p11 is regulated by glucocorticoids elevated by traumatic stress.


Subject(s)
Annexin A2/metabolism , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Nuclear Proteins/metabolism , Promoter Regions, Genetic/drug effects , Prosencephalon/metabolism , S100 Proteins/metabolism , Stress, Psychological/pathology , Up-Regulation/drug effects , Animals , Animals, Newborn , Annexin A2/genetics , Cells, Cultured , Chromatin Immunoprecipitation/methods , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Electroshock/adverse effects , Hormone Antagonists/pharmacology , Humans , Male , Mifepristone/pharmacology , Nuclear Proteins/genetics , Prosencephalon/cytology , Prosencephalon/drug effects , Rats , Rats, Sprague-Dawley , S100 Proteins/genetics , Stress, Psychological/etiology , Time Factors , Up-Regulation/physiology
3.
Neuroscience ; 131(2): 275-82, 2005.
Article in English | MEDLINE | ID: mdl-15708472

ABSTRACT

17Beta-estradiol (E2) is a major neuroregulator, exerting both genomic and non-genomic actions. E2 regulation of Slack (sequence like a calcium-activated potassium channel) potassium channels has not been identified in the CNS. We demonstrate E2-induced activation of Slack channels, which display a unitary conductance of about 60 pS, are inhibited by intracellular calcium, and are abundantly expressed in the nervous system. In lipid bilayers derived from rat cortical neuronal membranes, E2 increases Slack open probability and appears to decrease channel inactivation. Additionally, E2 binds to the Slack channel and activates outward currents in human embryonic kidney-293 cells that express Slack channels but not classical estrogen receptors (i.e. ERalpha or ERbeta). Neither E2-induced activation nor the binding intensity of E2 to the Slack channel is blocked by tamoxifen, an ER antagonist/agonist. Thus, E2 activates a potassium channel, Slack, through a non-traditional membrane binding site, adding to known non-genomic mechanisms by which E2 exerts pharmacological and toxicological effects in the CNS.


Subject(s)
Estradiol/metabolism , Nerve Tissue Proteins/metabolism , Potassium Channels/metabolism , Animals , Cell Line , Dose-Response Relationship, Drug , Humans , Nerve Tissue Proteins/genetics , Neurons/metabolism , Potassium Channels/genetics , Potassium Channels, Sodium-Activated , Protein Binding/physiology
4.
Exp Neurol ; 190(2): 276-88, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15530869

ABSTRACT

The mammalian central nervous system (CNS) has little capacity for self-repair after injury, and neurons are not capable of proliferating. Therefore, neural tissue engineering that combines neural stem and progenitor cells and biologically derived polymer scaffolds may revolutionize the medical approach to the treatment of damaged CNS tissues. Neural stem and progenitor cells isolated from embryonic rat cortical or subcortical neuroepithelium were dispersed within type I collagen, and the cell-collagen constructs were cultured in serum-free medium containing basic fibroblast growth factor. The collagen-entrapped stem and progenitors actively expanded and efficiently generated neurons, which developed neuronal polarity, neurotransmitters, ion channels/receptors, and excitability. Ca2+ imaging showed that differentiation from BrdU+/TuJ1- to BrdU-/TuJ1+ cells was accompanied by a shift in expression of functional receptors for neurotransmitters from cholinergic and purinergic to predominantly GABAergic and glutamatergic. Spontaneous postsynaptic currents were recorded by patch-clamping from precursor cell-derived neurons and these currents were partially blocked by 10-microM bicuculline, and completely blocked by additional 10 microM of the kainate receptor antagonist CNQX, indicating an appearance of both GABAergic and glutamatergic synaptic activities. Staining with endocytotic marker FM1-43 demonstrated active synaptic vesicle recycling occurring among collagen-entrapped neurons. These results show that neural stem and progenitor cells cultured in 3D collagen gels recapitulate CNS stem cell development; this is the first demonstration of CNS stem and progenitor cell-derived functional synapse and neuronal network formation in a 3D matrix. The proliferative capacity and neuronal differentiating potential of neural progenitors in 3D collagen gels suggest their potential use in attempts to promote neuronal regeneration in vivo.


Subject(s)
Cell Differentiation/physiology , Collagen Type I , Gels , Neurons/cytology , Stem Cells/cytology , Tissue Engineering/methods , Animals , Astrocytes/cytology , Brain/cytology , Brain/metabolism , Cell Proliferation , Cells, Cultured , Embryo, Mammalian , Immunohistochemistry , Microscopy, Confocal , Neurons/metabolism , Oligodendroglia/cytology , Patch-Clamp Techniques , Rats , Stem Cells/metabolism
5.
Neurology ; 63(8): 1446-51, 2004 Oct 26.
Article in English | MEDLINE | ID: mdl-15505163

ABSTRACT

OBJECTIVE: To determine if the CD4+CD28- T-cell subset is expanded in patients with recurrent stroke or death after acute ischemic stroke. This subset of the peripheral blood T-cell lymphocyte population has a strong pro-inflammatory and tissue-damaging potential. METHODS: Consecutive patients within the first 48 hours of ischemic stroke were prospectively studied. Peripheral blood CD4+CD28- cells were quantified by flow cytometry. The study endpoint was recurrent stroke or death from any cause during 1 year of follow-up. RESULTS: One hundred six patients (mean age 75.0 +/- 13.5 years; 50 women) were studied. The median CD4+CD28- cell count was 4.5% (range 0.2 to 72.2%). Twenty-seven endpoints (10 recurrent strokes and 17 deaths) occurred during follow-up. Stroke recurrence/death rates were significantly associated with increasing CD4+CD28- counts, rising from 14.2% in patients with CD4+CD28- levels of <1.0 to 48.1% for those with CD4+CD28- counts of >8.0% (p = 0.003, Cochran linear test of trend). Higher CD4+CD28- counts were also present in patients with a history of prior stroke (p = 0.03). After adjustment for age, admission NIH Stroke Scale score, prior stroke, and atrial fibrillation, CD4+CD28- counts of >8.0% were associated with a cumulative hazard ratio of 5.81 (95% CI: 1.58 to 21.32) for stroke recurrence or death. CONCLUSIONS: Rising counts of circulating CD4+CD28- cells are associated with an increasing risk of stroke recurrence and death, in addition to an observed association with prior stroke. Expansion of this T-cell subset presumably represents a biomarker and possibly a contributory pathogenic mechanism of recurrent stroke and death after ischemic stroke.


Subject(s)
Brain Ischemia/immunology , CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Lymphocytes/immunology , Stroke/immunology , T-Lymphocyte Subsets/immunology , Acute Disease , Aged , Aged, 80 and over , Atrial Fibrillation/complications , Biomarkers/blood , Brain Ischemia/blood , Brain Ischemia/mortality , CD4 Lymphocyte Count , Encephalitis/immunology , Encephalitis/physiopathology , Female , Humans , Lymphocyte Activation/immunology , Male , Middle Aged , Mortality , Predictive Value of Tests , Prospective Studies , Recurrence , Risk Factors , Stroke/blood , Stroke/mortality , Up-Regulation/immunology
6.
Neuroscience ; 118(1): 37-47, 2003.
Article in English | MEDLINE | ID: mdl-12676135

ABSTRACT

We have expanded neuroepithelial cells dissociated from the embryonic rat telencephalon in serum-free defined medium containing basic fibroblast growth factor (bFGF) in order to generate a model neuroepithelium to study the interaction of ethanol with both growth factor- and transmitter-stimulated proliferation. Ethanol blocked proliferation stimulated by bFGF and by carbachol, an agonist at muscarinic acetylcholine receptors, in a dose-dependent manner. In addition, ethanol attenuated autonomous expansion of neuroepithelial cells occurring following withdrawal of bFGF. The latter effect was associated with an increase in the number of apoptotic cells identified by terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling labeling. We studied the effects of ethanol on carbachol-stimulated signaling pathways critical to its proliferative effects. Ethanol significantly reduced carbachol-stimulated Ca(2+) signaling, as well as Erk1/Erk2, Akt and cyclic AMP-response element-binding phosphorylations in a dose-dependent manner. Comparison of the potency of ethanol in attenuating carbachol-stimulated proliferation and signal transduction showed that mitogen-activated protein kinase phosphorylation was less sensitive to ethanol than the other parameters. The results indicate that ethanol's suppression of proliferation induced by carbachol in this model neuroepithelium likely involves multiple signaling pathways. These effects in vitro may help to explain the devastating effects of prenatal ethanol exposure in vivo, which contribute to the fetal alcohol syndrome.


Subject(s)
Alcohol-Induced Disorders, Nervous System/metabolism , Brain/drug effects , Cell Division/drug effects , Ethanol/toxicity , Fetal Alcohol Spectrum Disorders/metabolism , Neurons/drug effects , Protein Serine-Threonine Kinases , Stem Cells/drug effects , Acetylcholine/antagonists & inhibitors , Acetylcholine/metabolism , Alcohol-Induced Disorders, Nervous System/physiopathology , Animals , Brain/embryology , Brain/physiopathology , Calcium Signaling/drug effects , Calcium Signaling/physiology , Carbachol/antagonists & inhibitors , Cell Division/physiology , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Fetal Alcohol Spectrum Disorders/physiopathology , Fetus , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/deficiency , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Muscarinic Antagonists/toxicity , Neurons/metabolism , Pregnancy , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/metabolism , Stem Cells/metabolism
7.
Neurosci Lett ; 332(2): 103-6, 2002 Oct 31.
Article in English | MEDLINE | ID: mdl-12384221

ABSTRACT

It has been shown that the inhalation of toluene in rats can cause neuronal apoptosis in the central nervous system. However, the cellular and molecular effects of toluene directly on astrocytes are relatively unknown. We used primary cultures of astrocytes isolated from the neonatal rat cortex as a model to study the toluene effects on cell outcome and associated signal transduction pathways using immunostaining and Western blotting. We observed that acute toluene exposure significantly induced caspase-dependent cell apoptosis and transiently stimulated the activation of p42/44 mitogen-activated protein kinase (MAPK) in the primary astrocytes. Interestingly, the inhibition of the p42/44 MAPK signaling cascade by PD98059 in conjunction with the toluene treatment evoked more cellular apoptosis than toluene alone, suggesting that the toluene-induced transient MAPK activation may play a role in promoting cell survival during the toluene exposure.


Subject(s)
Astrocytes/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Toluene/pharmacology , Animals , Astrocytes/drug effects , Cell Death/drug effects , Cell Division/drug effects , Cells, Cultured , Flavonoids/pharmacology , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Rats , Solvents , Stimulation, Chemical
8.
Biotechnol Bioeng ; 76(4): 376-90, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11745165

ABSTRACT

A series of recombinant Escherichia coli strains have been constructed and evaluated for their ability to synthesize p-hydroxybenzoic acid from glucose under fed-batch fermentor conditions. The maximum concentration of p-hydroxybenzoic acid synthesized was 12 g/L and corresponded to a yield of 13% (mol/mol). Synthesis of p-hydroxybenzoic acid began with direction of increased carbon flow into the common pathway of aromatic amino acid biosynthesis. This was accomplished in all constructs with overexpression of a feedback-insensitive isozyme of 3-deoxy-D-arabino-heptulosonic acid 7-phosphate synthase. Expression levels of enzymes in the common pathway of aromatic amino acid biosynthesis were also increased in all constructs to deliver increased carbon flow from the beginning to the end of the common pathway. A previously unreported inhibition of 3-dehydroquinate synthase by L-tyrosine was discovered to be a significant impediment to the flow of carbon through the common pathway. Chorismic acid, the last metabolite of the common pathway, was converted into p-hydroxybenzoic acid by ubiC-encoded chorismate lyase. Constructs differed in the strategy used for overexpression of chorismate lyase and also differed as to whether mutations were present in the host E. coli to inactivate other chorismate-utilizing enzymes. Use of overexpressed chorismate lyase to increase the rate of chorismic acid aromatization was mitigated by attendant decreases in the specific activity of DAHP synthase and feedback inhibition caused by p-hydroxybenzoic acid. The toxicity of p-hydroxybenzoic acid towards E. coli metabolism and growth was also evaluated.


Subject(s)
Biotechnology , Escherichia coli/metabolism , Glucose/metabolism , Parabens/metabolism , 3-Deoxy-7-Phosphoheptulonate Synthase/metabolism , Catalysis , Fermentation , Models, Chemical , Oxo-Acid-Lyases/metabolism , Parabens/chemistry , Phosphorus-Oxygen Lyases/metabolism , Plasmids/metabolism , Time Factors , Nicotiana/metabolism , Tyrosine/metabolism
9.
Brain Res ; 920(1-2): 183-93, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11716824

ABSTRACT

The chronic, lupus-like autoimmune disease in MRL-lpr mice is associated with leucocyte infiltration into the choroid plexus, brain cell death, and deficits in motivated behavior. The presence of lymphoid cells in the ventricular lumen and the increased number of TUNEL-positive cells in periventricular areas led to the hypothesis that immune cells enter into the cerebrospinal fluid (CSF) and induce primary neuronal damage in regions bordering the cerebral ventricles. Using an in vitro approach, we presently examine the possibility that CSF from autoimmune mice is neurotoxic and/or gliotoxic. The CSF and serum from diseased MRL-lpr mice, less symptomatic MRL +/+ controls, and healthy Swiss/Webster mice (non-autoimmune controls) were frozen until their effects on the viability of pyramidal neurons and astrocytes were assessed in a two-color fluorescence assay. Significant reduction in neuronal viability (in some cases as low as 67%) was observed in the co-cultures of hippocampal neurons and astrocytes incubated for 24 h with CSF from autoimmune MRL-lpr mice. The viability of astrocytes did not differ among the groups, and the CSF from autoimmune mice appeared more toxic than the serum. The behavior of MRL-lpr mice differed significantly from the control groups, as indicated by impaired exploration, reduced intake of palatable food, and excessive immobility in the forced swim test. The present results suggest that CSF from the behaviorally impaired lupus-prone mice is neurotoxic and are consistent with the hypothesis that neuroactive metabolites are produced intrathecally in neuropsychiatric lupus erythematosus.


Subject(s)
Autoimmunity/physiology , Behavior, Animal/physiology , Cerebrospinal Fluid/physiology , Neurotoxicity Syndromes/psychology , Animals , Astrocytes/physiology , Brain/cytology , Brain/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Coculture Techniques , Drinking/physiology , Eating/physiology , Hippocampus/pathology , Male , Mice , Mice, Inbred MRL lpr , Motor Activity/physiology , Neurons/physiology , Pyramidal Cells/physiology , Swimming/psychology
10.
J Am Chem Soc ; 123(42): 10173-82, 2001 Oct 24.
Article in English | MEDLINE | ID: mdl-11603966

ABSTRACT

The expense and limited availability of shikimic acid isolated from plants has impeded utilization of this hydroaromatic as a synthetic starting material. Although recombinant Escherichia coli catalysts have been constructed that synthesize shikimic acid from glucose, the yield, titer, and purity of shikimic acid are reduced by the sizable concentrations of quinic acid and 3-dehydroshikimic acid that are formed as byproducts. The 28.0 g/L of shikimic acid synthesized in 14% yield by E. coli SP1.1/pKD12.138 in 48 h as a 1.6:1.0:0.65 (mol/mol/mol) shikimate/quinate/dehydroshikimate mixture is typical of synthesized product mixtures. Quinic acid formation results from the reduction of 3-dehydroquinic acid catalyzed by aroE-encoded shikimate dehydrogenase. Is quinic acid derived from reduction of 3-dehydroquinic acid prior to synthesis of shikimic acid? Alternatively, does quinic acid result from a microbe-catalyzed equilibration involving transport of initially synthesized shikimic acid back into the cytoplasm and operation of the common pathway of aromatic amino acid biosynthesis in the reverse of its normal biosynthetic direction? E. coli SP1.1/pSC5.214A, a construct incapable of de novo synthesis of shikimic acid, catalyzed the conversion of shikimic acid added to its culture medium into a 1.1:1.0:0.70 molar ratio of shikimate/quinate/dehydroshikimate within 36 h. Further mechanistic insights were afforded by elaborating the relationship between transport of shikimic acid and formation of quinic acid. These experiments indicate that formation of quinic acid during biosynthesis of shikimic acid results from a microbe-catalyzed equilibration of initially synthesized shikimic acid. By apparently repressing shikimate transport, the aforementioned E. coli SP1.1/pKD12.138 synthesized 52 g/L of shikimic acid in 18% yield from glucose as a 14:1.0:3.0 shikimate/quinate/dehydroshikimate mixture.


Subject(s)
Escherichia coli Proteins , Membrane Transport Proteins , Shikimic Acid/analogs & derivatives , Shikimic Acid/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Silencing , Glucose/metabolism , Glucose/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Kinetics , Methylglucosides/metabolism , Methylglucosides/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Quinic Acid/metabolism , Nicotiana/genetics , Nicotiana/metabolism , Transketolase/biosynthesis , Transketolase/genetics , Transketolase/metabolism
11.
Neurosci Lett ; 312(3): 125-8, 2001 Oct 26.
Article in English | MEDLINE | ID: mdl-11602326

ABSTRACT

Substantial evidence suggests that the accumulation of beta-amyloid (Abeta)-derived peptides contributes to the aetiology of Alzheimer's disease (AD) by stimulating formation of free radicals. Thus, the antioxidant alpha-lipoate, which is able to cross the blood-brain barrier, would seem an ideal substance in the treatment of AD. We have investigated the potential effectiveness of alpha-lipoic acid (LA) against cytotoxicity induced by Abeta peptide (31-35) (30 microM) and hydrogen peroxide (H(2)O(2)) (100 microM) with the cellular 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) reduction and fluorescence dye propidium iodide assays in primary neurons of rat cerebral cortex. We found that treatment with LA protected cortical neurons against cytotoxicity induced by Abeta or H(2)O(2). In addition, LA-induced increase in the level of Akt in the neurons was observed by Western blot. The LA-induced neuroprotection and Akt increase were attenuated by pre-treatment with the phosphatidylinositol 3-kinase inhibitor, LY294002 (50 microM). Our data suggest that the neuroprotective effects of the antioxidant LA are partly mediated through activation of the PKB/Akt signaling pathway.


Subject(s)
Alzheimer Disease/drug therapy , Antioxidants/pharmacology , Cell Death/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/drug effects , Thioctic Acid/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Animals , Biological Assay , Cell Death/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cells, Cultured/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Coloring Agents , Dose-Response Relationship, Drug , Drug Interactions/physiology , Fetus , Hydrogen Peroxide/antagonists & inhibitors , Hydrogen Peroxide/metabolism , Neurons/metabolism , Neurons/pathology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , Tetrazolium Salts , Thiazoles
12.
Biosens Bioelectron ; 16(7-8): 481-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11544042

ABSTRACT

GABA(A) receptors are known targets for certain classes of environmental neurotoxins and pharmaceutical compounds. Since few neural cell lines express functional GABA(A) receptors, the capacity to rapidly screen for compounds that affect GABA(A) receptor function is presently limited. Previous work has demonstrated that rat neural precursor cells express functional GABA(A) receptors that can be monitored via Ca(2+) imaging. This study examined GABA(A) receptor subunit expression to determine whether GABA(A) receptor function and its interactions with neurotoxins is preserved after passaging. Neural precursor cells isolated from embryonic day 13 rat brain were expanded in serum-free medium containing basic fibroblast growth factor and passaged three times. Reverse transcription-polymerase chain reaction analysis demonstrated early expression of abundant mRNAs encoding various GABA(A) receptor subunits. Ca(2+) imaging showed that the highly proliferating precursor cells in passaged cultures maintained expression of functional GABA(A) receptors. In addition, we showed that trimethylolpropane phosphate, a neurotoxin generated during partial pyrolysis of a synthetic ester turbine engine lubricant, potently inhibited muscimol (GABA(A) receptor agonist) but not depolarization-induced cytosolic Ca(2+) increase. The findings of this study suggest that neural precursor cells may be well suited for the evaluation of certain environmental neurotoxins with convulsant activity. The potential use of neural precursor cells in high-throughput screens for compounds acting on GABA(A) receptors is discussed.


Subject(s)
Biosensing Techniques/methods , Receptors, GABA-A/genetics , Animals , Bridged Bicyclo Compounds, Heterocyclic/toxicity , Calcium/metabolism , Cells, Cultured , Gene Expression , Neurons/cytology , Neurotoxins/toxicity , Protein Subunits , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, GABA-A/chemistry , Receptors, GABA-A/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism
13.
Neuroscience ; 104(3): 913-22, 2001.
Article in English | MEDLINE | ID: mdl-11440820

ABSTRACT

GABA(A) receptor/Cl- channels and voltage-gated Ca2+ channels are believed to be important sites of ethanol action in the CNS. Acute exposure of ethanol potentiates GABA(A) receptor/Cl- channel activity and inhibits voltage-gated Ca2+ channels in a number of preparations, mostly post-mitotic neurons. The effects of ethanol on these channels in primary cultures of undifferentiated neural precursor cells remain unknown. To address this issue, we examined the effects of ethanol on GABA(A) agonist-activated elevation of cytosolic Ca2+ in an in vitro model of the cortical neuroepithelium derived from rat basic fibroblast growth factor-expanded neural precursor cells. We found a potent inhibition of GABA(A)-activated elevation of cytosolic Ca2+ by ethanol in actively proliferating cells. Since we had recently demonstrated that GABA(A) receptor activation depolarizes these cells and elevates their cytosolic Ca2+, we tested whether the effects of ethanol involved both GABA(A) receptors and voltage-gated Ca2+ channels. Both extracellular K+- and muscimol-induced cytosolic Ca2+ elevations were abolished by nitrendipine, indicating that both depolarizing stimuli triggered Ca2+ influx through L-type voltage-gated Ca2+ channels. Exposure of proliferating cells to different concentrations of ethanol revealed that the drug was more potent in blocking muscimol-induced compared to K+-evoked cytosolic Ca2+ elevations. These results raise the possibility that ethanol blocks GABAergic stimulation of cytosolic Ca2+ levels in proliferating precursors primarily by interacting with GABA(A) receptor/Cl- channels and secondarily with voltage-gated Ca2+ channels.


Subject(s)
Calcium/metabolism , Chloride Channels/drug effects , Cytosol/drug effects , Ethanol/pharmacology , Neurons/drug effects , Receptors, GABA-A/drug effects , Stem Cells/drug effects , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured/cytology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/embryology , Chloride Channels/metabolism , Cytosol/metabolism , Disease Models, Animal , Extracellular Space/drug effects , Extracellular Space/metabolism , Female , Fetal Alcohol Spectrum Disorders/metabolism , Fetal Alcohol Spectrum Disorders/physiopathology , Fetus , Immunohistochemistry , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/metabolism , Potassium Chloride/pharmacology , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Stem Cells/metabolism
14.
Neuroreport ; 12(9): 1919-23, 2001 Jul 03.
Article in English | MEDLINE | ID: mdl-11435923

ABSTRACT

The cellular mechanisms underlying the neuroprotective effects of estrogen are only beginning to be elucidated. Here we examined the role of protein kinase B (Akt) activation in 17beta-estradiol (E2) inhibition of beta-amyloid peptide (31-35) (Abeta31-35)-induced neurotoxicity in cultured rat hippocampal neurons. Abeta31-35 (25-30 betaM) significantly decreased the total number of microtubule associated protein-2 positive cells (MAP2+). This decrease was significantly reversed by pre-treatment with 100 nM E2. Further, 100 nM E2 alone significantly increased the total number of protein kinase B and microtubule associated protein-2 positive cells compared with controls. Such E2-induced increases were inhibited by LY294002 (20 microM), a specific PI3-K inhibitor, as well as by tamoxifen, an estrogen receptor antagonist/selective estrogen receptor modulator. These results indicate that the neuroprotective effects of E2 may be mediated at least in part via estrogen receptor-mediated protein kinase B activation.


Subject(s)
Amyloid beta-Peptides/pharmacology , Estradiol/pharmacology , Hippocampus/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Neurotoxins/pharmacology , Peptide Fragments/pharmacology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/drug effects , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Cell Count , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured/cytology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Chromones/pharmacology , Drug Interactions/physiology , Enzyme Inhibitors/pharmacology , Estradiol/metabolism , Estrogen Antagonists/pharmacology , Fetus , Hippocampus/cytology , Hippocampus/metabolism , Immunohistochemistry , Microtubule-Associated Proteins/metabolism , Morpholines/pharmacology , Neurons/cytology , Neurons/metabolism , Peptide Fragments/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism , Tamoxifen/pharmacology
15.
Cereb Cortex ; 11(8): 744-53, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11459764

ABSTRACT

During development, postmitotic neurons migrate from germinal regions into the cortical plate (cp), where lamination occurs. In rats, GABA is transiently expressed in the cp, near target destinations for migrating neurons. In vitro GABA stimulates neuronal motility, suggesting cp cells release GABA, which acts as a chemoattractant during corticogenesis. Pharmacological studies indicate GABA stimulates migration via GABA(B)-receptor (GABA(B)-R) activation. Using immunohistochemistry, RT-PCR and Western blotting, we examined embryonic cortical cell expression of GABA(B)-Rs in vivo. At E17, GABA(B)-R1(+) cells were identified in the ventricular zone (vz) and cp. RT-PCR and Western blotting demonstrated the presence of GABA(B)-R1a and GABA(B)-R1b mRNA and proteins. Using immuno- cytochemistry, GABA(B)-R expression was examined in vz and cp cell dissociates before and after migration to GABA in an in vitro chemotaxis assay. GABA-induced migration resulted in an increase of GABA(B)-R(+) cells in the migrated population. While <20% of each starting dissociate was GABA(B)-R(+), >70% of migrated cells were immunopositive. We used a microchemotaxis assay to analyze cp cell release of diffusible chemotropic factor(s). In vitro, cp dissociates induced vz cell migration in a cell density-dependent manner that was blocked by micromolar saclofen (a GABA(B)-R antagonist). HPLC demonstrated cp cells release micromolar levels of GABA and taurine in several hours. Micromolar levels of both molecules stimulated cell migration that was blocked by micromolar saclofen. Thus, migratory cortical cells express GABA(B)-Rs, cp cells release GABA and taurine, and both molecules stimulate cortical cell movement. Together these findings suggest GABA and/or taurine act as chemoattractants for neurons during rat cortical histogenesis via mechanisms involving GABA(B)-Rs.


Subject(s)
Cell Movement/physiology , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Receptors, GABA-B/physiology , Amino Acids/analysis , Amino Acids/isolation & purification , Animals , Antimetabolites , Blotting, Western , Bromodeoxyuridine , Cell Separation , Chemotactic Factors/pharmacology , Chromatography, High Pressure Liquid , Female , Immunohistochemistry , Pregnancy , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Taurine/metabolism , gamma-Aminobutyric Acid/metabolism
16.
J Neurosci ; 21(7): 2343-60, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11264309

ABSTRACT

GABA emerges as a trophic signal during rat neocortical development in which it modulates proliferation of neuronal progenitors in the ventricular/subventricular zone (VZ/SVZ) and mediates radial migration of neurons from the VZ/SVZ to the cortical plate/subplate (CP/SP) region. In this study we investigated the role of GABA in the earliest phases of neuronal differentiation in the CP/SP. GABAergic-signaling components emerging during neuronal lineage progression were comprehensively characterized using flow cytometry and immunophenotyping together with physiological indicator dyes. During migration from the VZ/SVZ to the CP/SP, differentiating cortical neurons became predominantly GABAergic, and their dominant GABA(A) receptor subunit expression pattern changed from alpha4beta1gamma1 to alpha3beta3gamma2gamma3 coincident with an increasing potency of GABA on GABA(A) receptor-mediated depolarization. GABA(A) autoreceptor/Cl(-) channel activity in cultured CP/SP neurons dominated their baseline potential and indirectly their cytosolic Ca(2+) (Ca(2+)c) levels via Ca(2+) entry through L-type Ca(2+) channels. Block of this autocrine circuit at the level of GABA synthesis, GABA(A) receptor activation, intracellular Cl(-) ion homeostasis, or L-type Ca(2+) channels attenuated neurite outgrowth in most GABAergic CP/SP neurons. In the absence of autocrine GABAergic signaling, neuritogenesis could be preserved by depolarizing cells and elevating Ca(2+)c. These results reveal a morphogenic role for GABA during embryonic neocortical neuron development that involves GABA(A) autoreceptors and L-type Ca(2+) channels.


Subject(s)
Chloride Channels/physiology , Embryo, Mammalian/physiology , Neocortex/physiology , Neurites/physiology , Neurons/physiology , Receptors, GABA-A/physiology , gamma-Aminobutyric Acid/physiology , Action Potentials/physiology , Animals , Electrophysiology , Microscopy, Electron , Neurons/ultrastructure , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
17.
J Neurosci ; 21(5): 1569-79, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11222647

ABSTRACT

Muscarinic acetylcholine receptor (mAChR), a member of the G-protein-coupled receptors (GPCRs) gene superfamily, has been shown to mediate the effects of acetylcholine on differentiation and proliferation in the CNS. However, the mechanism or mechanisms whereby mAChRs regulate cell proliferation remain poorly understood. Here we show that in vitro bFGF-expanded neural progenitor cells dissociated from rat cortical neuroepithelium express muscarinic acetylcholine receptor subtype mRNAs. We demonstrate that stimulation of these mAChRs with carbachol, a muscarinic agonist, activated extracellular-regulated kinases (Erk1/2) and phosphatidylinositol-3 kinase (PI-3K). This, in turn, stimulated DNA synthesis in neural progenitor cells. MEK inhibitor PD98059 and PI-3K inhibitors wortmannin and LY294002 inhibited a carbachol-induced increase in DNA synthesis. These findings indicate that the activation of both PI-3 kinase and MEK signaling pathways via muscarinic receptors is involved in stimulating DNA synthesis in the neural progenitor cells during early neurogenesis.


Subject(s)
DNA/biosynthesis , Mitogen-Activated Protein Kinases/metabolism , Nerve Tissue Proteins , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Muscarinic/metabolism , Stem Cells/metabolism , Animals , Cell Division/drug effects , Cell Division/physiology , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 2/pharmacology , Intermediate Filament Proteins/biosynthesis , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Muscarinic Agonists/pharmacology , Nestin , Neurons/cytology , Neurons/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Muscarinic/genetics , Stem Cells/cytology , Stem Cells/drug effects , Telencephalon/cytology
18.
J Neurosci Methods ; 102(2): 187-95, 2000 Oct 30.
Article in English | MEDLINE | ID: mdl-11040415

ABSTRACT

To investigate the ability to culture neural precursor cells in a three-dimensional (3D) collagen gel, neuroepithelial cells were isolated from embryonic day 13 rat cortex, dispersed within type I collagen and maintained for up to 30 days in vitro. Cultured in Neuorobasal medium supplemented with B27 containing basic fibroblast growth factor, the collagen-entrapped precursor cells actively expanded and formed clone-like clusters. Many cells in the center of the cluster were proliferating as revealed by 5-bromo-2'-deoxyuridine uptake. Some cells began to migrate away from the center at 5 days and were labeled by either neuronal marker neuron-specific beta-tubulin (TuJ1) or astrocytic marker glial fibrillary acidic protein. The differentiated neurons (TuJ1(+)) exhibited characteristic cytosolic Ca(2+) oscillations in response to excitatory neurotransmitter glutamate. These findings suggest the suitability of the 3D culture system for the proliferation and differentiation of neural precursor cells.


Subject(s)
Calcium/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Cytosol/metabolism , Stem Cells/cytology , Stem Cells/physiology , Animals , Astrocytes/cytology , Cell Differentiation , Cell Division/drug effects , Cell Survival , Cells, Cultured , Cerebral Cortex/drug effects , Collagen , Culture Media, Serum-Free , Fibroblast Growth Factor 2/pharmacology , Gels , Glutamic Acid/pharmacology , Neurons/cytology , Oscillometry , Rats , Rats, Sprague-Dawley , Stem Cells/drug effects
19.
J Neurophysiol ; 84(3): 1392-403, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10980012

ABSTRACT

Whole cell patch-clamp recordings using Cl(-)-filled pipettes revealed more negative levels of baseline current and associated current variance in embryonic rat hippocampal neurons co-cultured on a monolayer of astrocytes than those cultured on poly-D-lysine. These effects were mimicked by culturing neurons on poly-D-lysine in astrocyte-conditioned medium (ACM). The baseline current and variance decreased immediately in all cells after either local perfusion with saline or exposure to bicuculline, an antagonist of GABA at GABA(A) receptor/Cl(-) channels. Baseline current and variance in all cells reached a nadir at approximately 0 mV, the calculated equilibrium potential for Cl(-). Perfusion of ACM rapidly induced a sustained current in neurons, which also reversed polarity at approximately 0 mV. Bicuculline attenuated or eliminated the ACM-induced current at a concentration that completely blocked micromolar GABA-induced current. Quantitative analyses of spontaneously occurring fluctuations superimposed on the ACM-induced current revealed estimated unitary properties of the underlying channel activity similar to those calculated for GABA's activation of GABA(A) receptor/Cl(-) channels. Bicuculline-sensitive synaptic-like transients, which reversed at approximately 0 mV, were also detected in neurons cultured in ACM, and these were immediately eliminated along with the negative baseline current and superimposed current fluctuations by perfusion. Furthermore bicuculline-sensitive synaptic-like transients were rapidly and reversibly triggered when ACM was acutely applied. ACM induced an increase in cytoplasmic Ca(2+) in cultured embryonic hippocampal neurons that was completely blocked by bicuculline and strychnine. We conclude that astrocytes release diffusible substances, most likely GABA, that persistently activate GABA(A) receptor/Cl(-) channels in co-cultured neurons.


Subject(s)
Astrocytes/metabolism , Chloride Channels/metabolism , Egtazic Acid/analogs & derivatives , Hippocampus/metabolism , Neurons/metabolism , Receptors, GABA-A/metabolism , gamma-Aminobutyric Acid/metabolism , 3-Mercaptopropionic Acid/pharmacology , Animals , Animals, Newborn , Bicuculline/pharmacology , Calcium/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Chloride Channels/antagonists & inhibitors , Chlorides/metabolism , Coculture Techniques , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , GABA Antagonists/pharmacology , GABA-A Receptor Antagonists , Glycine Agents/pharmacology , Hippocampus/cytology , Hippocampus/embryology , Membrane Potentials/physiology , Neurons/cytology , Patch-Clamp Techniques , Rats , Strychnine/pharmacology
20.
Cereb Cortex ; 10(9): 899-909, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10982750

ABSTRACT

Recent studies indicate that GABA acts as a chemoattractant during rat cortical histogenesis. In vivo, GABA localizes in appropriate locations for a chemoattractant, along migratory routes and near target destinations for migrating cortical neurons. In vitro, GABA induces dissociated embryonic cortical neurons to migrate. Here, embryonic rat cortical slices were cultured in the presence or absence of GABA receptor (GABA-R) antagonists to assess GABA's effects on neuronal migration in situ. Gestational day 18 (E18) cortical slices were incubated overnight in bromodeoxyuridine (BrdU)-containing medium to label ventricular zone (vz) cells as they underwent terminal mitosis. The slices were then cultured in BrdU-free medium with or without GABA-R antagonists. In control slices, most BrdU(+) cells were observed in the cortical plate (cp) after 48 h. In contrast, cultures maintained in either saclofen (a GABA(B)-R antagonist) or picrotoxin (a GABA(A/C)-R antagonist) had few BrdU-labeled cp cells. However, the effects of the two antagonists were distinct. In the picrotoxin-treated slices, nearly half of all BrdU(+) cells remained in the vz and subventricular zone (svz), whereas saclofen treatment resulted in an accumulation of BrdU(+) cells in the intermediate zone (iz). Bicuculline, a GABA(A)-R antagonist, did not block, but rather enhanced migration of BrdU(+) cells into the cp. These results provide evidence that picrotoxin-sensitive receptors promote the migration of vz/svz cells into the iz, while saclofen-sensitive receptors signal cells to migrate into the cp. Thus, as cortical cells differentiate, changing receptor expression appears to modulate migratory responses to GABA.


Subject(s)
Bicuculline/pharmacology , Cell Movement/drug effects , Cerebral Cortex/cytology , GABA Antagonists/pharmacology , Nerve Tissue Proteins , Neurons/cytology , Receptors, GABA-A/physiology , Animals , Baclofen/analogs & derivatives , Baclofen/pharmacology , Bromodeoxyuridine/analysis , Cell Movement/physiology , Cellular Senescence/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/embryology , Chemotaxis/drug effects , Female , Glial Fibrillary Acidic Protein/analysis , Intermediate Filament Proteins/analysis , Mitosis , Nestin , Neuroglia/cytology , Neurons/chemistry , Neurons/physiology , Organ Culture Techniques , Picrotoxin/pharmacology , Pregnancy , Rats , gamma-Aminobutyric Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...