Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Restor Neurol Neurosci ; 34(6): 965-976, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27834787

ABSTRACT

PURPOSE: Astrocytes perform a plethora of important functions in the central nervous system (CNS) and are involved in cocaine-evoked synaptic plasticity. Previously, we showed that while cocaine decreased cyclin A2 expression in primary human neural progenitor cells, it increased cyclin A2 expression in human astrocytes. Since cyclin A2 is an essential regulator of the cell cycle, the aim of the present study is to clarify the effect of cocaine on proliferation of human astrocytes and elucidate the underlying molecular mechanisms. METHODS: Primary human astrocytes were treated with either 1, 10, or 100 µM cocaine for 48 hr, and cell proliferation was measured using the CyQUANT cell proliferation assay. To elucidate the molecular mechanisms through which cocaine affects the proliferation of astrocytes, we analyzed gene expression profiles in cocaine-treated primary human astrocytes using a human focused cDNA array. Gene ontology/pathway enrichment analysis, STRING protein-protein interaction analysis, RT-qPCR, and western blotting were used to identify signal transduction pathways that are involved in cocaine-induced astrocyte dysfunction. RESULTS: Cocaine at 10 and 100 µM significantly increased human astrocyte proliferation. Gene expression profiling revealed the JNK MAP kinase pathway as a driver of cell proliferation affected by cocaine in human astrocytes. Further experiments showed that cocaine-induced JNK activation induced up-regulation of cyclin A2, leading to enhanced proliferation of human astrocytes. CONCLUSION: Cocaine-induced abnormal increases in the number of astrocytes may cause disruption in neuron-glia signaling and contribute to synaptic impairment in the CNS. Understanding the mechanisms of cocaine's effects on human astrocytes may help to reveal the involvement of glial cells in addictive behaviors.


Subject(s)
Anesthetics, Local/pharmacology , Astrocytes/drug effects , Cell Proliferation/drug effects , Cocaine/pharmacology , Cyclin A2/metabolism , MAP Kinase Kinase 4/metabolism , Up-Regulation/drug effects , Analysis of Variance , Dose-Response Relationship, Drug , Gene Expression Profiling , Glial Fibrillary Acidic Protein/metabolism , Humans , Oligonucleotide Array Sequence Analysis , RNA, Messenger , Signal Transduction/drug effects
2.
Cell Rep ; 10(4): 616-32, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25640183

ABSTRACT

Human pluripotent stem cell (hPSC) lines exhibit repeated patterns of genetic variation, which can alter in vitro properties as well as suitability for clinical use. We examined associations between copy-number variations (CNVs) on chromosome 17 and hPSC mesodiencephalic dopaminergic (mDA) differentiation. Among 24 hPSC lines, two karyotypically normal lines, BG03 and CT3, and BG01V2, with trisomy 17, exhibited amplification of the WNT3/WNT9B region and rapid mDA differentiation. In hPSC lines with amplified WNT3/WNT9B, basic fibroblast growth factor (bFGF) signaling through mitogen-activated protein kinase (MAPK)/ERK amplifies canonical WNT signaling by phosphorylating LRP6, resulting in enhanced undifferentiated proliferation. When bFGF is absent, noncanonical WNT signaling becomes dominant due to upregulation of SIAH2, enhancing JNK signaling and promoting loss of pluripotency. When bFGF is present during mDA differentiation, stabilization of canonical WNT signaling causes upregulation of LMX1A and mDA induction. Therefore, CNVs in 17q21.31, a "hot spot" for genetic variation, have multiple and complex effects on hPSC cellular phenotype.


Subject(s)
Neurons/cytology , Neurons/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Wnt Proteins/metabolism , Wnt3 Protein/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , Wnt Proteins/genetics , Wnt3 Protein/genetics
3.
PLoS Med ; 5(6): e117, 2008 Jun 10.
Article in English | MEDLINE | ID: mdl-18593214

ABSTRACT

BACKGROUND: Prenatal exposure of the developing brain to cocaine causes morphological and behavioral abnormalities. Recent studies indicate that cocaine-induced proliferation inhibition and/or apoptosis in neural progenitor cells may play a pivotal role in causing these abnormalities. To understand the molecular mechanism through which cocaine inhibits cell proliferation in neural progenitors, we sought to identify the molecules that are responsible for mediating the effect of cocaine on cell cycle regulation. METHODS AND FINDINGS: Microarray analysis followed by quantitative real-time reverse transcription PCR was used to screen cocaine-responsive and cell cycle-related genes in a neural progenitor cell line where cocaine exposure caused a robust anti-proliferative effect by interfering with the G1-to-S transition. Cyclin A2, among genes related to the G1-to-S cell cycle transition, was most strongly down-regulated by cocaine. Down-regulation of cyclin A was also found in cocaine-treated human primary neural and A2B5+ progenitor cells, as well as in rat fetal brains exposed to cocaine in utero. Reversing cyclin A down-regulation by gene transfer counteracted the proliferation inhibition caused by cocaine. Further, we found that cocaine-induced accumulation of reactive oxygen species, which involves N-oxidation of cocaine via cytochrome P450, promotes cyclin A down-regulation by causing an endoplasmic reticulum (ER) stress response, as indicated by increased phosphorylation of eIF2alpha and expression of ATF4. In the developing rat brain, the P450 inhibitor cimetidine counteracted cocaine-induced inhibition of neural progenitor cell proliferation as well as down-regulation of cyclin A. CONCLUSIONS: Our results demonstrate that down-regulation of cyclin A underlies cocaine-induced proliferation inhibition in neural progenitors. The down-regulation of cyclin A is initiated by N-oxidative metabolism of cocaine and consequent ER stress. Inhibition of cocaine N-oxidative metabolism by P450 inhibitors may provide a preventive strategy for counteracting the adverse effects of cocaine on fetal brain development.


Subject(s)
Cocaine/pharmacology , Neurons/drug effects , Stem Cells/drug effects , Animals , Cell Cycle , Cell Proliferation/drug effects , Cell Survival , Cells, Cultured , Cimetidine/pharmacology , Cyclin A/genetics , Cyclin A/metabolism , Cyclin A2 , Down-Regulation , Female , Humans , Phosphorylation , Pregnancy , Rats , Rats, Sprague-Dawley , Transfection
4.
J Neurosci Res ; 85(8): 1724-33, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17455326

ABSTRACT

The molecular mechanisms involved in N-methyl-D-aspartate (NMDA)-induced cell death and Delta9-tetrahydrocannabinol (THC)-induced neuroprotection were investigated in vitro with an AF5 neural progenitor cell line model. By microarray analysis, Ywhah, CK1, Hsp60, Pdcd 4, and Pdcd 7 were identified as being strongly regulated by both NMDA toxicity and THC neuroprotection. The 14-3-3 eta (14-3-3eta; gene symbol Ywhah) and 14-3-3 zeta (14-3-3zeta; gene symbol Ywhaz) transcripts were deceased by NMDA treatment and increased by THC treatment prior to NMDA, as measured by cDNA microarray analysis and quantitative real-time RT-PCR. Other 14-3-3 isoforms were unchanged. Whereas up-regulation of 14-3-3zeta expression was observed 30 min after treatment with THC plus NMDA, down-regulation by NMDA alone was not seen until 16 hr after treatment. By Western blotting, THC increased 14-3-3 protein only in cells that were also treated with NMDA. Overexpression of 14-3-3eta or 14-3-3zeta by transient plasmid transfection increased 14-3-3 protein levels and decreased NMDA-induced cell death. These data suggest that increases in 14-3-3 proteins mediate THC-induced neuroprotection under conditions of NMDA-induced cellular stress.


Subject(s)
14-3-3 Proteins/biosynthesis , Dronabinol/pharmacology , N-Methylaspartate/physiology , Neuroprotective Agents/pharmacology , 14-3-3 Proteins/genetics , Animals , Cell Line , Mesencephalon/cytology , N-Methylaspartate/antagonists & inhibitors , N-Methylaspartate/toxicity , Oligonucleotide Array Sequence Analysis , Plasmids , Rats , Transfection
5.
Neurosci Lett ; 389(2): 99-103, 2005 Dec 02.
Article in English | MEDLINE | ID: mdl-16098661

ABSTRACT

NMDA causes oxidative stress in neurons, and produces cell death involving elements of both necrosis and apoptosis. To examine the neuroprotective mechanism of Delta9-tetrahydrocannabinol (THC) in NMDA-induced death of AF5 cells, we measured reactive oxygen species (ROS) formation after exposure to NMDA. ROS generation was increased by NMDA, and NMDA-induced ROS generation was significantly decreased by THC. Western blotting revealed an increase in phosphorylated p38 MAPK after NMDA treatment, which was also blocked by pretreatment with THC. The time course of ROS generation and activation of MAPK signaling pathways were similar. SB203580, a p38 inhibitor, partially blocked glutamate excitotoxicity in AF5 cells. The present data suggest that THC protects against NMDA-induced apoptosis in AF5 cells by blocking ROS generation and inhibiting the activation of p38-MAPK.


Subject(s)
Apoptosis/drug effects , Dronabinol/pharmacology , N-Methylaspartate/antagonists & inhibitors , Neurotoxins/antagonists & inhibitors , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Apoptosis/physiology , Cell Line, Transformed , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , N-Methylaspartate/toxicity , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Neuroprotective Agents/pharmacology , Neurotoxins/toxicity , Oxidative Stress/drug effects , Oxidative Stress/physiology , Phosphorylation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Exp Neurol ; 191 Suppl 1: S45-59, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15629761

ABSTRACT

Immortalized central nervous system (CNS) cell lines are useful as in vitro models for innumerable purposes such as elucidating biochemical pathways, studies of effects of drugs, and ultimately, such cells may also be useful for neural transplantation. The SV40 large T (LT) oncoprotein, commonly used for immortalization, interacts with several cell cycle regulatory factors, including binding and inactivating p53 and retinoblastoma family cell-cycle regulators. In an attempt to define the minimal requirements of SV40 T antigen for immortalizing cells of CNS origin, we constructed T155c, encoding the N-terminal 155 amino acids of LT. The p53 binding region is known to reside in the C-terminal region of LT. An additional series of mutants was produced to further narrow the molecular targets for immortalization, and plasmid vectors were constructed for each. In a p53 temperature sensitive cell line model, T64-7B, expression of T155c and all constructs having mutations outside of the first 82 amino acids were capable of overriding cell-cycle block at the non-permissive growth temperature. Several cell lines were produced from fetal rat mesencephalic and cerebral cortical cultures using the T155c construct. The E107K construct contained a mutation in the Rb binding region, but was nonetheless capable of overcoming cell cycle block in T64-7B cell and immortalizing primary cultured cells. Cells immortalized with T155c were often highly dependent on the presence of bFGF for growth. Telomerase activity, telomere length, growth rates, and integrity of the p53 gene in cells immortalized with T155c did not change over 100 population doublings in culture, indicating that cells immortalized with T155c were generally stable during long periods of continuous culture.


Subject(s)
Antigens, Viral, Tumor/genetics , Cell Transformation, Viral/genetics , Cerebral Cortex/cytology , Mesencephalon/cytology , Peptide Fragments/genetics , Simian virus 40/genetics , Animals , Cell Cycle/genetics , Cell Division/drug effects , Cell Line, Transformed , Cells, Cultured , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Clone Cells , Fibroblast Growth Factor 2/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Mesencephalon/embryology , Mesencephalon/metabolism , Mutagenesis, Site-Directed , Peptide Fragments/biosynthesis , Rats , Rats, Sprague-Dawley , Telomerase/metabolism , Telomere/chemistry , Telomere/metabolism , Temperature , Transfection , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...