Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 171
Filter
1.
Int J Mol Sci ; 21(21)2020 Nov 07.
Article in English | MEDLINE | ID: mdl-33171774

ABSTRACT

Neuritogenesis is the process underling nervous system regeneration; however, optimal extracellular signals that can promote neuronal regenerative activities require further investigation. Previously, we developed a novel method for inducing neuronal differentiation in rat PC12 cells using temperature-controlled repeated thermal stimulation (TRTS) with a heating plate. Based on neurogenic sensitivity to TRTS, PC12 cells were classified as either hyper- or hyposensitive. In this study, we aimed to investigate the mechanism of hyposensitivity by establishing two PC12-derived subclones according to TRTS sensitivity during differentiation: PC12-P1F1, a hypersensitive subclone, and PC12-P1D10, a hyposensitive subclone. To characterize these subclones, cell size and neuritogenesis were evaluated in subclones treated with nerve growth factor (NGF), bone morphogenetic protein (BMP), or various TRTS. No significant differences in cell size were observed among the parental cells and subclones. BMP4- or TRTS-induced neuritogenesis was increased in PC12-P1F1 cells compared to that in the parental cells, while no neuritogenesis was observed in PC12-P1D10 cells. In contrast, NGF-induced neuritogenesis was observed in all three cell lines. Furthermore, a BMP inhibitor, LDN-193189, considerably inhibited TRTS-induced neuritogenesis. These results suggest that the BMP pathway might be required for TRTS-induced neuritogenesis, demonstrating the useful aspects of these novel subclones for TRTS research.


Subject(s)
Nerve Regeneration/physiology , PC12 Cells/metabolism , Thermosensing/physiology , Animals , Cell Differentiation/physiology , Neurites/metabolism , Neurogenesis/physiology , Neurons/metabolism , PC12 Cells/physiology , Rats , Temperature
2.
Acta Pharmacol Sin ; 33(1): 34-40, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22120967

ABSTRACT

AIM: To investigate the effects of M3, a derivative of huperzine A, on the apoptosis induced by sodium nitroprusside (SNP) in PC12 cells. METHODS: Cell viability was detected using MTT method. Apoptosis was examined with annexin V/prodium iodide (PI) stain. The levels of reactive oxygen species (ROS) were measured using fluorophotometric quantitation. The amount of malonaldehyde (MDA) was determined with MDA detection kits. The expression of caspase-3 and Hsp70 were analyzed using Western blotting. RESULTS: Exposure of PC12 cells to SNP (200 µmol/L) for 24 h decreased the cell viability to 69.0% of that in the control group. Pretreatment with M3 (10 µmol/L) or huperzine A (10 µmol/L) significantly protected the cells against SNP-induced injury and apoptosis; the ratio of apoptotic bodies in PC12 cells was decreased from 27.3% to 15.0%. Pretreatment with M3 (10 µmol/L) significantly decreased ROS and MDA levels, and increased the expression of Hsp70 in the cells. Quercetin (10 µmol/L) blocked the protective effect of M3, while did not influence on that of huperzine A. CONCLUSION: M3 protects PC12 cells against SNP-induced apoptosis, possible due to ROS scavenging and Hsp70 induction.


Subject(s)
Alkaloids/chemistry , Alkaloids/pharmacology , Apoptosis/drug effects , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Nitroprusside/pharmacology , PC12 Cells/drug effects , Sesquiterpenes/chemistry , Sesquiterpenes/pharmacology , Animals , Antioxidants/pharmacology , Cell Survival/drug effects , HSP70 Heat-Shock Proteins/metabolism , Humans , Malondialdehyde/metabolism , Molecular Structure , Nitric Oxide Donors/pharmacology , PC12 Cells/physiology , Quercetin/pharmacology , Rats , Reactive Oxygen Species/metabolism
3.
Proc Natl Acad Sci U S A ; 106(41): 17308-13, 2009 Oct 13.
Article in English | MEDLINE | ID: mdl-19805029

ABSTRACT

Ca(2+)-dependent activator protein for secretion (CAPS) is an essential factor for regulated vesicle exocytosis that functions in priming reactions before Ca(2+)-triggered fusion of vesicles with the plasma membrane. However, the precise events that CAPS regulates to promote vesicle fusion are unclear. In the current work, we reconstituted CAPS function in a SNARE-dependent liposome fusion assay using VAMP2-containing donor and syntaxin-1/SNAP-25-containing acceptor liposomes. The CAPS stimulation of fusion required PI(4,5)P(2) in acceptor liposomes and was independent of Ca(2+), but Ca(2+) dependence was restored by inclusion of synaptotagmin. CAPS stimulated trans-SNARE complex formation concomitant with the stimulation of full membrane fusion at physiological SNARE densities. CAPS bound syntaxin-1, and CAPS truncations that competitively inhibited syntaxin-1 binding also inhibited CAPS-dependent fusion. The results revealed an unexpected activity of a priming protein to accelerate fusion by efficiently promoting trans-SNARE complex formation. CAPS may function in priming by organizing SNARE complexes on the plasma membrane.


Subject(s)
Qa-SNARE Proteins/metabolism , SNARE Proteins/metabolism , Synaptosomal-Associated Protein 25/metabolism , Animals , Cell Membrane/metabolism , Cell Membrane/physiology , Exocytosis/physiology , Homeostasis , Lecithins/metabolism , Liposomes/metabolism , Membrane Fusion/physiology , PC12 Cells/physiology , Phosphatidylserines/metabolism , Rats , Synaptotagmins/metabolism , Syntaxin 1/metabolism , Vesicle-Associated Membrane Protein 2/metabolism
4.
J Neurochem ; 119(4): 697-707, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21668890

ABSTRACT

Dp71 has an important role in the central nervous system. To better understand the function of Dp71 domains in neuronal differentiation, PC12 cells were stably transfected with a dystrophin mutant, Dp71Δ(78-79) , which lacks exons 78 and 79. Based on the percentage of cells bearing neurites and neurite length analyses, we found that cells stably expressing Dp71Δ(78-79) (PC12-C11) differentiate more efficiently than non-transfected cells. While wild-type cells reach their maximum differentiation 9-12 days after initiating the differentiation process, the PC12-C11 cells reach differentiation in 4-6 days. Protein expression analysis showed a down-regulation of Dp71a and an up-regulation of Dp71ab and/or Up71, ß-dystroglycan and neuron-specific enolase in undifferentiated and in neural growth factor differentiated PC12-C11 cells. No change was observed in the expression of Grb2 and Up400. The subcellular localization of Dp71Δ(78-79) was in the cell periphery, and there was no change in localization during the differentiation process, which was also observed throughout the neurite extensions.


Subject(s)
Cell Differentiation/genetics , Dystrophin/genetics , Gene Expression Regulation/genetics , Mutation/genetics , Animals , Cell Differentiation/drug effects , Dystroglycans/genetics , Dystroglycans/metabolism , Exons/genetics , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , Gene Expression Regulation/drug effects , Nerve Growth Factor/pharmacology , Neurites , PC12 Cells/physiology , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Time Factors , Transfection/methods
5.
Neurochem Res ; 36(2): 223-31, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21057871

ABSTRACT

Recently, it was reported that in a 4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model, neuronal cell death is associated with the cdk5-mediated hyperphosphorylation of myocyte enhancer factor 2 (MEF2), a transcription factor that is critically required for neuronal survival. In the present study, we investigated the possible involvement of cdk5-mediated MEF2D down-regulation on 6-hydroxydopamine (6-OHDA)-induced PC12 cell death. 6-OHDA was found to significantly increase nitric oxide (NO) production and to induce apoptosis in a time-dependent manner in PC12 cells. Furthermore, 6-OHDA was found to markedly reduce MEF2D levels under conditions that could induce PC12 cell apoptosis. In addition, PC12 cell death and MEF2D degradation by 6-OHDA were prevented by the cdk5 inhibitor roscovitine, but roscovitine could not restore the 6-OHDA-induced inactivation of Akt. These results suggest that the cell death and MEF2D degradation caused by 6-OHDA are dependent on cdk5 activity. On the other hand, roscovitine enhanced the 6-OHDA-induced activations of ERK1/2 and JNK, but reduced the 6-OHDA-induced activation of p38. These results suggest that PC12 cell death by 6-OHDA appears to be regulated by the down-regulation of MEF2D via some interaction between cdk5 and MAP kinase.


Subject(s)
Apoptosis/drug effects , Myogenic Regulatory Factors/metabolism , Oxidopamine/pharmacology , PC12 Cells/drug effects , PC12 Cells/physiology , Animals , Apoptosis/physiology , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , DNA Fragmentation , Down-Regulation , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , MEF2 Transcription Factors , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Myogenic Regulatory Factors/genetics , Nitric Oxide/metabolism , Protein Kinase Inhibitors/metabolism , Purines/metabolism , Rats , Roscovitine , Signal Transduction/physiology
6.
Mol Biol Rep ; 38(5): 3561-7, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21120620

ABSTRACT

In this study, the effect of aucubin on H(2)O(2)-induced apoptosis was studied by using a rat pheochromocytoma (PC12) cell line. We have analyzed the apoptosis of H(2)O(2)-induced PC12 cells, H(2)O(2)-induced apoptosis appeared to correlate with lower Bcl-2 expression, higher Bax expression and sequential activation of caspase-3 leading to cleavage of poly-ADP-ribose polymerase (PARP). Aucubin not only inhibited lower Bcl-2 expression, high Bax expression, but also modulated caspase-3 activation, PARP cleavage, and eventually protected against H(2)O(2)-induced apoptosis. These results indicated that aucubin can obstruct H(2)O(2)-induced apoptosis by regulating of the expression of Bcl-2 and Bax, as well as suppression of caspases cascade activation.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Hydrogen Peroxide/pharmacology , Iridoid Glucosides/pharmacology , PC12 Cells/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , Animals , Cell Survival/drug effects , Chromatin/ultrastructure , Iridoid Glucosides/chemistry , Molecular Structure , Oxidants/pharmacology , PC12 Cells/physiology , PC12 Cells/ultrastructure , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , bcl-2-Associated X Protein/metabolism
7.
Mol Cell Proteomics ; 8(10): 2350-67, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19525549

ABSTRACT

MS-based quantitative proteomics is widely used for large scale identification of proteins. However, an integrated approach that offers comprehensive proteome coverage, a tool for the quick categorization of the identified proteins, and a standardized biological study method is needed for helping the researcher focus on investigating the proteins with biologically important functions. In this study, we utilized isobaric tagging for relative and absolute quantification (iTRAQ)-based quantitative differential LC/MS/MS, functional annotation with a proprietary gene ontology tool (Molecular Annotation by Gene Ontology (MANGO)), and standard biochemical methods to identify proteins related to neuronal differentiation in nerve growth factor-treated rat pheochromocytoma (PC12) cells, which serve as a representative model system for studying neuronal biological processes. We performed MS analysis by using both nano-LC-MALDI-MS/MS and nano-LC-ESI-MS/MS for maximal proteome coverage. Of 1,482 non-redundant proteins semiquantitatively identified, 72 were differentially expressed with 39 up- and 33 down-regulated, including 64 novel nerve growth factor-responsive PC12 proteins. Gene ontology analysis of the differentially expressed proteins by MANGO indicated with statistical significance that the up-regulated proteins were mostly related to the biological processes of cell morphogenesis, apoptosis/survival, and cell differentiation. Some of the up-regulated proteins of unknown function, such as PAIRBP1, translationally controlled tumor protein, prothymosin alpha, and MAGED1, were further analyzed to validate their significant functions in neuronal differentiation by immunoblotting and immunocytochemistry using each antibody combined with a specific short interfering RNA technique. Knockdown of these proteins caused abnormal cell morphological changes, inhibition of neurite formation, and cell death during each course of the differentiation, confirming their important roles in neurite formation and survival of PC12 cells. These results show that our iTRAQ-MANGO-biological analysis framework, which integrates a number of standard proteomics strategies, is effective for targeting and elucidating the functions of proteins involved in the cellular biological process being studied.


Subject(s)
Cell Differentiation/physiology , Cell Survival/physiology , Chromatography, Liquid/methods , Neurons/physiology , Proteomics/methods , Software , Tandem Mass Spectrometry/methods , Animals , Apoptosis/physiology , Molecular Sequence Data , Nerve Growth Factor/metabolism , PC12 Cells/physiology , Proteome/analysis , Rats
8.
J Neurochem ; 112(3): 797-806, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19943848

ABSTRACT

The photopigment melanopsin is expressed in a subtype of mammalian ganglion cells in the retina that project to the circadian clock in the hypothalamic suprachiasmatic nucleus to mediate non-visual light information. Melanopsin renders these retinal ganglion cells intrinsically photosensitive and the cells respond to light by a membrane depolarization and induction of the immediate early response gene Fos. Previous studies showed that the light activated melanopsin-induced signaling, the phototransduction, leading to depolarization of the membrane resembles the invertebrate opsins, which involves a Galpha(q/11) coupled phospholipase C activation. However, the signaling proteins mediating melanopsin-induced Fos expression are unresolved. In this study, we examined the phototransduction leading to Fos expression in melanopsin-transfected PC12 cells. A pivotal role of the extracellular signal-regulated protein kinase 1/2 (ERK1/2) was found as pharmacological blockage of this kinase suppressed the light-induced Fos expression. Illumination increased the inositol phosphate turnover and induced phosphorylation of ERK1/2 and p38 but not the c-Jun N-terminal kinase. The Galpha(q/11) protein inhibitor YM254890 attenuated these intracellular light responses. Our data strongly indicate that Galpha(q/11)-mediated ERK1/2 activation is essential for expression of Fos upon illumination of melanopsin-expressing PC12 cells.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Light , Oncogene Proteins v-fos/metabolism , Rod Opsins/metabolism , Animals , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/genetics , Gene Expression Regulation/physiology , Inositol Phosphates/metabolism , Oncogene Proteins v-fos/genetics , PC12 Cells/drug effects , PC12 Cells/physiology , PC12 Cells/radiation effects , Phosphorylation/drug effects , Phosphorylation/radiation effects , Rats , Rod Opsins/genetics , Statistics, Nonparametric , Transfection/methods
9.
Neurosignals ; 18(1): 32-42, 2010.
Article in English | MEDLINE | ID: mdl-20389133

ABSTRACT

Rat pheochromocytoma (PC12) cells characteristically undergo differentiation when cultured with nerve growth factor (NGF). Here we show that NGF dramatically increased the adenylyl cyclase-activating property of forskolin in PC12 cells. This effect of NGF was well maintained even when NGF was removed after 4 days, even though the morphological features of neuronal differentiation were rapidly lost on removal of NGF. The enhanced cAMP production in response to forskolin could be due to a synergistic interaction between forskolin and endogenously released agonists acting on G(s)-coupled receptors. However, responses to forskolin were not attenuated by antagonists of adenosine A2 receptors or pituitary adenylate cyclase-activating polypeptide (PACAP) receptors, suggesting that adenosine and PACAP were not involved. Adenylyl cyclases 3, 6 and 9 were the predominant isoforms expressed in PC12 cells, but we found no evidence for NGF-induced changes in expression levels of any of the 9 adenylyl cyclase isoforms, nor in the expression of Gα(s). These findings highlight that NGF has a subtle influence on adenylyl cyclase activity in PC12 cells which may influence more than the neurite extension process classically associated with neuronal differentiation.


Subject(s)
Adenylyl Cyclases/metabolism , Cell Differentiation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Nerve Growth Factor/pharmacology , Analysis of Variance , Animals , Cell Count/methods , Colforsin/pharmacology , Cyclic AMP/metabolism , Drug Interactions , PC12 Cells/drug effects , PC12 Cells/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Protein Isoforms/metabolism , Rats , Time Factors , Tritium/metabolism
10.
J Biomed Sci ; 17 Suppl 1: S17, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20804591

ABSTRACT

BACKGROUND: Taurine is a free amino acid present in high concentrations in a variety of organs of mammalians. As an antioxidant, taurine has been found to protect cells against oxidative stress, but the underlying mechanism is still unclear. METHODS: In this report, we present evidence to support the conclusion that taurine exerts a protective function against endoplasmic reticulum (ER) stress induced by H2O2 in PC 12 cells. Oxidative stress was introduced by exposure of PC 12 cells to 250 uM H2O2 for 4 hours. RESULTS: It was found that the cell viability of PC 12 cells decreased with an increase of H2O2 concentration ranging from approximately 76% cell viability at 100 uM H2O2 down to 18% at 500 uM H2O2. At 250 uM H2O2, cell viability was restored to 80% by taurine at 25 mM. Furthermore, H2O2 treatment also caused a marked reduction in the expression of Bcl-2 while no significant change of Bax was observed. Treatment with taurine restored the reduced expression of Bcl-2 close to the control level without any obvious effect on Bax. Furthermore, taurine was also found to suppress up-regulation of GRP78, GADD153/CHOP and Bim induced by H2O2, suggesting that taurine may also exert a protective function against oxidative stress by reducing the ER stress. CONCLUSION: In summary, taurine was shown to protect PC12 cells against oxidative stress induced by H2O2. ER stress was induced by oxidative stress and can be suppressed by taurine.


Subject(s)
Antioxidants/pharmacology , Endoplasmic Reticulum/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Taurine/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Cell Survival/drug effects , Dose-Response Relationship, Drug , Heat-Shock Proteins/metabolism , Hydrogen Peroxide/pharmacology , Membrane Proteins/metabolism , Oxidants/pharmacology , PC12 Cells/drug effects , PC12 Cells/physiology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Transcription Factor CHOP/metabolism , bcl-2-Associated X Protein/metabolism
11.
Neuroscience ; 159(2): 550-8, 2009 Mar 17.
Article in English | MEDLINE | ID: mdl-19171180

ABSTRACT

Excitability and axon/dendrite specification are the most distinctive features in the establishment of neuronal polarization. Conditioned medium from rat sciatic nerve (CM) induced a neuronal-like morphology in PC12 cells. Here we show that CM neuritogenic activity is limited to the induction of dendrites in PC12 cells. However, treatment of these cells with CM in combination with a generic inhibitor for tyrosine kinase receptors (k252a) promoted the enhancement of neurite length, development of axons and induction of sodium currents. On the other hand, specific inhibition of TrkA and p75(NTR) receptors in CM-treated cells reduced the neurite length in comparison with cells treated only with CM, although the effect over the induction of sodium currents was continuously observed. These results suggested that CM had some components that, even though are able to start the morphological cell differentiation and produce short neurites (likely acting through TrkA and p75(NTR)), can restrain further neurite extension. Depletion of pro-NGF isoforms from CM produced a similar effect as the exerted by k252a, TrkA and p75(NTR) receptor inhibitors in CM-treated cells, inducing the elicitation of sodium currents. These results suggested that the effect of CM might be mediated through pro-NGF. The difference between the results obtained with the generic inhibitor for Trk receptors and the specific inhibitors for TrkA and p75(NTR) receptors in CM-treated cells, suggested that alternative pathways could be used to regulate neurite elongation, axon specification and sodium currents in PC12 cells. These findings represent important clues to improve the understanding of the initiation of neuronal polarity.


Subject(s)
Nerve Growth Factors/antagonists & inhibitors , Neurites/physiology , PC12 Cells/cytology , Protein Precursors/antagonists & inhibitors , Sciatic Nerve/chemistry , Sodium Channels/metabolism , Analysis of Variance , Animals , Carbazoles/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Culture Media, Conditioned/pharmacology , Enzyme Inhibitors/pharmacology , Immunoprecipitation/methods , Indole Alkaloids/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Microtubule-Associated Proteins/metabolism , Nerve Growth Factor/pharmacology , Nerve Growth Factors/metabolism , Neurites/drug effects , Neurofilament Proteins/metabolism , PC12 Cells/drug effects , PC12 Cells/physiology , Patch-Clamp Techniques/methods , Protein Precursors/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Nerve Growth Factor/antagonists & inhibitors , Receptor, trkA/antagonists & inhibitors , Time Factors , Tissue Culture Techniques
12.
J Mol Neurosci ; 37(3): 225-37, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18629654

ABSTRACT

There is a paucity of quantitative methods for evaluating the morphological differentiation of neuronal cells in a three-dimensional (3-D) system to assist in quality control of neural tissue engineering constructs for use in reparative medicine. Neuronal cells tend to aggregate in the 3-D scaffolds, hindering the application of two-dimensional (2-D) morphological methods to quantitate neuronal differentiation. To address this problem, we developed a stable transfectant green fluorescence protein (GFP)-PC12 neuronal cell model, in which the differentiation process in 3-D can be monitored with high sensitivity by fluorescence microscopy. Under 2-D conditions, the green cells showed collagen adherence, round morphology, proliferation properties, expression of the nerve growth factor (NGF) receptors TrkA and p75(NTR), stimulation of extracellular signal-regulated kinase phosphorylation by NGF and were able to differentiate in a dose-dependent manner upon NGF treatment, like wild-type (wt)-PC12 cells. When grown within 3-D collagen gels, upon NGF treatment, the GFP-PC12 cells differentiated, expressing long neurite outgrowths. We describe here a new validated method to measure NGF-induced differentiation in 3-D. Having properties similar to those of wt-PC12 and an ability to grow and differentiate in 3-D structures, these highly visualized GFP-expressing PC12 cells may serve as an ideal model for investigating various aspects of differentiation to serve in neural engineering.


Subject(s)
Cell Differentiation/physiology , Collagen/metabolism , Gels/metabolism , Green Fluorescent Proteins/metabolism , Neurons/physiology , PC12 Cells , Animals , Cell Proliferation , Green Fluorescent Proteins/genetics , Neurons/cytology , PC12 Cells/cytology , PC12 Cells/physiology , Rats , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism
13.
J Mol Neurosci ; 37(3): 254-62, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18636346

ABSTRACT

A method was developed to assess mitochondrial movement in the living cell that is dependent, in part, on microtubule and/or associating protein interactions. The leader sequence from cytochrome-c was used to drive DsRed2 fluorescent proteins to accumulate in the mitochondria, thus enabling to follow mitochondrial (cytochrome-c's) movement. For calculating the percentage of mitochondrial movement, an image-processing program was used (ImageJ). Paclitaxel, an antitumor agent, is a potent microtubule-stabilizing agent that increases the stability of tubulin polymers, inhibiting mitosis and mitochondrial activity in dividing cells. Here, we tested whether paclitaxel inhibits mitochondrial movement in pheochromocytoma cells (a neuronal model, when tested in a differentiated state). While a 2-day exposure to paclitaxel resulted in cellular toxicity (measured as inhibition of mitochondrial activity), 2-3 h exposure to paclitaxel were sufficient to inhibit mitochondrial movement as assessed in 10-20-s imaging sessions in living cells. Mitotracker deep-red staining validated the staining obtained with DsRed2-cytochrome-c and identified intact mitochondria. Results showed a significant paclitaxel dose-dependent inhibition of mitochondrial movement. This new method should enable further assessment of microtubule-interacting drugs and other cytoskeletal components for their potential influence of mitochondrial movement as a test for activity and side effects.


Subject(s)
Adrenal Gland Neoplasms/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Mitochondria , Paclitaxel/pharmacology , Pheochromocytoma/metabolism , Animals , Cell Line, Tumor , Fluorescent Dyes/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , PC12 Cells/cytology , PC12 Cells/drug effects , PC12 Cells/physiology , Rats
14.
Mol Cell Endocrinol ; 300(1-2): 126-31, 2009 Mar 05.
Article in English | MEDLINE | ID: mdl-19022340

ABSTRACT

The major source for dehydroepiandrosterone (DHEA) and its sulphate compound DHEA-S is the inner zone of the adrenal cortex, which is in direct contact to adrenomedullary chromaffin cells. Due to their close proximity, direct interactions of DHEA and DHEA-S with chromaffin cells during adrenal gland development and throughout the whole life span are hypothesized. A possible direct effect of DHEA-S and the cellular and molecular mechanisms of DHEA-S action on chromaffin cells remain unresolved. Therefore, in this study, we aimed at clarifying DHEA-S effects and mechanisms of action on rat chromaffin PC12 cells. DHEA-S (10(-6)mol/l) inhibited nerve growth factor (NGF, 20ng/ml)-induced cell proliferation by 66% (n=4, p<0.001). In NGF-stimulated cells, neuronal differentiation was inhibited by DHEA-S, as demonstrated by a 22% reduction (n=3; p<0.05) of neuronal differentiation marker expression, synaptosome-associated protein of 25kDa (SNAP-25), and a 59% (n=6; p<0.001) decrease in neurite outgrowth. Moreover, DHEA-S stimulated expression of endocrine marker chromogranin A (CgA) by 31% (n=4; p<0.05 vs. control) and catecholamine release from NGF-treated PC12 cells by 229% (n=3-5; p<0.001), indicating a DHEA-S-induced shift towards neuroendocrine differentiation. On a molecular level, DHEA-S diminished NGF-induced ERK1/2 phosphorylation. Taken together, DHEA-S inhibited NGF-induced proliferation and neuronal differentiation and shifted cells towards a more endocrine phenotype. Interference of DHEA-S with NGF-stimulated ERK1/2 activation might be involved in this effect. Our study provides support for the notion that adrenocortical-derived DHEA-S impacts adrenomedullary chromaffin cells during development and differentiation.


Subject(s)
Cell Differentiation/drug effects , Dehydroepiandrosterone Sulfate/pharmacology , PC12 Cells , Animals , Chromogranin A/metabolism , Dopamine/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , PC12 Cells/drug effects , PC12 Cells/physiology , Rats , Synaptosomal-Associated Protein 25/metabolism
15.
Exp Mol Med ; 41(4): 269-76, 2009 Apr 30.
Article in English | MEDLINE | ID: mdl-19299916

ABSTRACT

Differentiation of neuronal cells has been shown to accelerate stress-induced cell death, but the underlying mechanisms are not completely understood. Here, we find that early and sustained increase in cytosolic ([Ca2(+)]c) and mitochondrial Ca2(+) levels ([Ca2(+)]m) is essential for the increased sensitivity to staurosporine- induced cell death following neuronal differentiation in PC12 cells. Consistently, pretreatment of differentiated PC12 cells with the intracellular Ca2(+)-chelator EGTA-AM diminished staurosporine-induced PARP cleavage and cell death. Furthermore, Ca2(+) overload and enhanced vulnerability to staurosporine in differentiated cells were prevented by Bcl-XL overexpression. Our data reveal a new regulatory role for differentiation-dependent alteration of Ca2(+) signaling in cell death in response to staurosporine.


Subject(s)
Calcium/metabolism , Cell Differentiation/physiology , Neurons , PC12 Cells , Staurosporine/pharmacology , Animals , Caspase 3/metabolism , DNA Fragmentation , Mitochondria/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/physiology , PC12 Cells/cytology , PC12 Cells/drug effects , PC12 Cells/physiology , Rats , bcl-X Protein/metabolism
16.
J Cell Biol ; 135(4): 883-93, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8922374

ABSTRACT

Nearly all of the known activities required for mitochondrial DNA (mtDNA) replication and expression are nuclear-encoded gene products, necessitating communication between these two physically distinct intracellular compartments. A significant amount of both general and specific biochemical information about mtDNA replication in mammalian cells has been known for almost two decades. Early studies achieved selective incorporation of the thymidine analog 5-Bromo-2-deoxy-Uridine (BrdU) into mtDNA of thymidine kinase-deficient (TK[-]) cells. We have revisited this approach from a cellular perspective to determine whether there exist spatiotemporal constraints on mtDNA replication. Laser-scanning confocal microscopy was used to selectively detect mtDNA synthesis in situ in cultured mammalian cells using an immunocytochemical double-labeling approach to visualize the incorporation of BrdU into mtDNA of dye-labeled mitochondria. In situ detection of BrdU-incorporated mtDNA was feasible after a minimum of 1-2 h treatment with BrdU, consistent with previous biochemical studies that determined the time required for completion of a round of mtDNA replication. Interestingly, the pattern of BrdU incorporation into the mtDNA of cultured mammalian cells consistently radiated outward from a perinuclear position, suggesting that mtDNA replication first occurs in the vicinity of nuclear-provided materials. Newly replicated mtDNA then appears to rapidly distribute throughout the dynamic cellular mitochondrial network.


Subject(s)
DNA Replication/physiology , DNA, Mitochondrial/analysis , Animals , Antibodies, Antinuclear , Blood Platelets/cytology , Blood Platelets/physiology , Blood Platelets/ultrastructure , Bromodeoxyuridine , Cell Differentiation/physiology , Cell Division/physiology , DNA, Mitochondrial/biosynthesis , DNA, Mitochondrial/immunology , Fluorescent Antibody Technique , Genome , HeLa Cells/cytology , HeLa Cells/physiology , HeLa Cells/ultrastructure , Humans , Mammals , Mitochondria/genetics , Mitochondria/metabolism , Osteosarcoma , PC12 Cells/cytology , PC12 Cells/physiology , PC12 Cells/ultrastructure , Rats
17.
J Cell Biol ; 119(6): 1669-80, 1992 Dec.
Article in English | MEDLINE | ID: mdl-1469055

ABSTRACT

Previous studies have shown that in neuronal cells the developmental phenomenon of programmed cell death is an active process, requiring synthesis of both RNA and protein. This presumably reflects a requirement for novel gene products to effect cell death. It is shown here that the death of nerve growth factor-deprived neuronal PC12 cells occurs at the same rate as that of rat sympathetic neurons and, like rat sympathetic neurons, involves new transcription and translation. In nerve growth factor-deprived neuronal PC12 cells, a decline in metabolic activity, assessed by uptake of [3H]2-deoxyglucose, precedes the decline in cell number, assessed by counts of trypan blue-excluding cells. Both declines are prevented by actinomycin D and anisomycin. In contrast, the death of nonneuronal (chromaffin-like) PC12 cells is not inhibited by transcription or translation inhibitors and thus does not require new protein synthesis. DNA fragmentation by internucleosomal cleavage does not appear to be a consistent or significant aspect of cell death in sympathetic neurons, neuronal PC12 cells, or nonneuronal PC12 cells, notwithstanding that the putative nuclease inhibitor aurintricarboxylic acid protects sympathetic neurons, as well as neuronal and nonneuronal PC12 cells, from death induced by trophic factor removal. Both phenotypic classes of PC12 cells respond to aurintricarboxylic acid with similar dose-response characteristics. Our results indicate that programmed cell death in neuronal PC12 cells, but not in nonneuronal PC12 cells, resembles programmed cell death in sympathetic neurons in significant mechanistic aspects: time course, role of new protein synthesis, and lack of a significant degree of DNA fragmentation.


Subject(s)
Apoptosis/physiology , Nerve Growth Factors/pharmacology , Neurons/physiology , PC12 Cells/physiology , Sympathetic Nervous System/physiology , Animals , Anisomycin/pharmacology , Apoptosis/drug effects , Aurintricarboxylic Acid/pharmacology , Cell Differentiation , Cell Separation , DNA Damage , Dactinomycin/pharmacology , Deoxyglucose/metabolism , PC12 Cells/drug effects , Protein Biosynthesis , Rats , Rats, Inbred Strains , Rats, Wistar , Transcription, Genetic
18.
Neuron ; 22(1): 125-37, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10027295

ABSTRACT

Elementary Ca2+ release signals in nerve growth factor- (NGF-) differentiated PC12 cells and hippocampal neurons, functionally analogous to the "Ca2+ sparks" and "Ca2+ puffs" identified in other cell types, were characterized by confocal microscopy. They either occurred spontaneously or could be activated by caffeine and metabotropic agonists. The release events were dissimilar to the sparks and puffs described so far, as many arose from clusters of both ryanodine receptors (RyRs) and inositol 1,4,5-trisphosphate receptors (InsP3Rs). Increasing either the stimulus strength or loading of the intracellular stores enhanced the frequency of and coupling between elementary release sites and evoked global Ca2+ signals. In the PC12 cells, the elementary Ca2+ release preferentially occurred around the branch points. Spatio-temporal recruitment of such elementary release events may regulate neuronal activities.


Subject(s)
Calcium Signaling/physiology , Hippocampus/physiology , Nerve Growth Factors/pharmacology , Neurons/physiology , PC12 Cells/pathology , PC12 Cells/physiology , Animals , Calcium/metabolism , Calcium Channels/metabolism , Calcium Channels/physiology , Cell Differentiation/drug effects , Electrophysiology , Endoplasmic Reticulum/metabolism , Hippocampus/cytology , Inositol 1,4,5-Trisphosphate Receptors , Neurites/metabolism , Rats , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Ryanodine Receptor Calcium Release Channel/metabolism , Ryanodine Receptor Calcium Release Channel/physiology
19.
Neuron ; 11(5): 865-75, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8240810

ABSTRACT

The product of the Wnt-1 proto-oncogene is a secreted glycoprotein that is normally produced in regions of the embryonic neural tube. We show here that expression of mouse Wnt-1 cDNA in the rat PC12 pheochromocytoma cell line causes a dramatic conversion from a round to a flat cell morphology. In addition, PC12 cells expressing Wnt-1 (PC12/Wnt-1) fail to extend neurites after treatment with NGF, despite the presence and activation of high affinity NGF receptors encoded by the trk gene and the induction of early response genes. Furthermore, PC12/Wnt-1 cells fail to express several neuron- and chromaffin-specific genes, indicating that PC12/Wnt-1 cells have assumed a new phenotype. Although NGF and FGF utilize similar signal transduction pathways in PC12 cells, only FGF is capable of inducing a morphological response and synthesis of transin mRNA in PC12/Wnt-1 cells.


Subject(s)
Fibroblast Growth Factors/pharmacology , Gene Expression/drug effects , Immediate-Early Proteins , Nerve Growth Factors/pharmacology , PC12 Cells/pathology , PC12 Cells/physiology , Proto-Oncogene Proteins/pharmacology , Zebrafish Proteins , Animals , Calcium-Binding Proteins , Carrier Proteins , Chromaffin System/cytology , Chromaffin System/metabolism , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , Intracellular Signaling Peptides and Proteins , Matrix Metalloproteinase 3 , Membrane Proteins , Metalloendopeptidases/genetics , Microtubule Proteins , Nerve Tissue Proteins/genetics , PC12 Cells/drug effects , Phosphorylation , RNA, Messenger/metabolism , Receptors, Nerve Growth Factor/metabolism , Stathmin , Transcription Factors/genetics , Tyrosine/metabolism , Wnt Proteins , Wnt1 Protein
20.
Apoptosis ; 13(3): 364-76, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18293087

ABSTRACT

The fungal alkaloid militarinone A (MiliA) was recently found to stimulate neuronal outgrowth in PC-12 cells by persistant activation of pathways that are also involved in NGF-mediated differentiation, namely the PI3-K/PKB and the MEK/ERK pathways. Application of equal concentrations of MiliA to other cells such as the murine neuroblastoma cell line N2a resulted in immediate onset of apoptosis by nuclear translocation of apoptosis inducing factor (AIF), activation of caspases and c-Jun/AP-1 transcription factor without an intermediate differentiated phenotype, although minor transient phosphorylation of PKB and MAPK as well as activation of NF-kappaB were also observed. Translocation of AIF was preceded by p53 phosphorylation at Ser15 and blocked by pifithrin alpha, a known inhibitor of p53-transcriptional activity. We here show that both cell types activate the same pathways albeit in different time scales. This is mainly due to contrasting basal expression levels of p53, which in turn regulates expression of AIF. In PC-12 cells, continuous activation of these pathways after prolonged treatment with 40 muM MiliA first led to up-regulation of p53, phosphorylation of p53, release of AIF from mitochondria and its translocation into the nucleus. Additionally, also activation of the c-Jun/AP-1 transcription factor was observed, and PC-12 cells subsequently underwent apoptosis 48-72 h post-treatment. We report that similar pathways working on different levels are able to initially shape very divergent cellular responses.


Subject(s)
Alkaloids/pharmacology , Apoptosis/drug effects , Neurites/physiology , PC12 Cells/drug effects , PC12 Cells/physiology , Pyridones/pharmacology , Tumor Suppressor Protein p53/biosynthesis , Animals , Apoptosis Inducing Factor/metabolism , Cell Line, Tumor , Chromones/pharmacology , Down-Regulation , Mice , Mitogen-Activated Protein Kinases/biosynthesis , Morpholines/pharmacology , Neurites/drug effects , Proto-Oncogene Proteins c-akt/biosynthesis , Rats
SELECTION OF CITATIONS
SEARCH DETAIL