Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 112
Filter
1.
Br J Nutr ; 103(1): 25-31, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19747415

ABSTRACT

Oxidative damage is believed to be associated with ageing, cancer and several degenerative diseases. Previous reports have shown that safflower-seed extract and its major antioxidant constituents, serotonin hydroxycinnamic amides, possess a powerful free radical-scavenging and antioxidative activity, paying particular attention to atherosclerotic reactive oxygen species (ROS)-related dysfunctions. In the present report, we examined a still unknown cell-based mechanism of serotonin derivatives against ROS-related neuronal damage, phenomena that represent a crucial event in neurodegenerative diseases. Serotonin derivatives N-(p-coumaroyl)serotonin and N-feruloylserotonin exerted a protective effect on high glucose-induced cell death, inhibited the activation of caspase-3 which represents the last and crucial step within the cascade of events leading to apoptosis, and inhibited the overproduction of the mitochondrial superoxide, which represents the most dangerous radical produced by hyperglycaemia, by acting as scavengers of the superoxide radical. In addition, serotonin derivative concentration inside the cells and inside the mitochondria was increased in a time-dependent manner. Since recent studies support the assertion that mitochondrial dysfunctions related to oxidative damage are the major contributors to neurodegenerative diseases, these preliminary cell-based results identify a mitochondria-targeted antioxidant property of serotonin derivatives that could represent a novel therapeutic approach against the neuronal disorders and complications related to ROS.


Subject(s)
Glucose/toxicity , PC12 Cells/metabolism , Serotonin/pharmacology , Animals , Caspases/metabolism , Cell Differentiation , Cell Survival/drug effects , Glucose/pharmacology , Kinetics , Mitochondria/drug effects , Mitochondria/metabolism , PC12 Cells/drug effects , PC12 Cells/pathology , Rats , Reactive Oxygen Species/metabolism , Serotonin/analogs & derivatives , Superoxides/metabolism
2.
Neuroscience ; 158(4): 1436-45, 2009 Feb 18.
Article in English | MEDLINE | ID: mdl-19095046

ABSTRACT

Intermittent hypoxia (IH) is a major pathological factor in the development of neural deficits associated with sleep-disordered breathing. Here we demonstrate that IH lasting 2 or 30 days, but not sustained hypoxia (SH) of the same duration, was accompanied by several posttranslational modifications of the large subunit of RNA polymerase II, Rpb1, including hydroxylation of proline 1465, phosphorylation of serine 5 residues within the C-terminal domain, and nondegradative ubiquitylation. These modifications were found to occur in two regions of the brain, hippocampal region CA1 and the prefrontal cortex, but not in neocortex, brainstem and CA3 region of hippocampus. We also found that mice exposed to 14 or 30 days of IH, but not SH, demonstrated cognitive deficits in behavioral assays. Furthermore, by using the pheochromocytoma-derived PC12 cell line, we showed that, under in vitro IH conditions, induction of Rpb1 hydroxylation, phosphorylation, and ubiquitylation required that the von Hippel-Lindau protein be present. We hypothesize that the observed modifications of Rpb1 participate in regulating the expression of genes involved in mediating cognitive deficits evoked by chronic IH.


Subject(s)
Gene Expression Regulation/physiology , Hippocampus/enzymology , Hypoxia/pathology , Prefrontal Cortex/enzymology , RNA Polymerase II/metabolism , Animals , Carrier Proteins/metabolism , Cullin Proteins/metabolism , Hypoxia/physiopathology , Learning Disabilities/etiology , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , PC12 Cells/enzymology , PC12 Cells/pathology , RNA Polymerase II/genetics , Rats , Retinol-Binding Proteins, Cellular/metabolism , Serine/metabolism , Time Factors , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
3.
Z Naturforsch C J Biosci ; 64(5-6): 395-8, 2009.
Article in English | MEDLINE | ID: mdl-19678545

ABSTRACT

The neurotoxicity induced by beta-amyloid (Abeta), which is one of the major causes of Alzheimer's disease (AD), leads to synaptic loss and subsequent neuronal death. Therefore, modulation of Abeta-induced neurotoxicity, as well as regeneration of damaged synapses could be important therapeutic approaches to control AD. In this study, we found that isorosmanol, an abietane-type diterpene, protected PC12 cells against Abeta-induced toxicity. Furthermore, isorosmanol promoted the generation of neurites. The neurotrophic effect of isorosmanol was enhanced by co-treatment with nerve growth factor (NGF). In addition, the neurite outgrowth induced by isorosmanol was accompanied by F-actin redistribution and increased expression of neurofilaments. Taken together, these results suggest that isorosmanol possesses both neuroprotective and neurotrophic effects, that might be beneficial for controlling AD.


Subject(s)
Diterpenes/pharmacology , Neurites/physiology , PC12 Cells/pathology , Abietanes , Actins/drug effects , Actins/metabolism , Animals , Cell Survival/drug effects , Nerve Growth Factors/pharmacology , Neurites/drug effects , PC12 Cells/drug effects , Rats
4.
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
5.
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
6.
Mil Med ; 183(suppl_1): 269-275, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29635567

ABSTRACT

Threshold shock-impulse levels required to induce cellular injury and cumulative effects upon single and/or multiple exposures are not well characterized. Currently, there are few in vitro experimental models with blast pressure waves generated by using real explosives in the laboratory for investigating the effects of primary blast-induced traumatic brain injury. An in vitro indoor experimental platform is developed using real military explosive charges to accurately represent battlefield blast exposure and to probe the effects of primary explosive blast on dissociated neurons and tissue slices. Preliminary results indicate that physical insults altered membrane permeability, impacted cellular viability, created axonal beadings, and led to synaptic protein loss in hippocampal slice cultures. Injuries from blast under the conditions that were examined did not appear to cause immediate or sustained damage to the cells. Three consecutive primary blasts failed to disrupt the overall cellular integrity in the hippocampal slice cultures and produced a unique type of pathology comprised with distinct reduction in synaptic proteins before cellular deterioration set in. These observed changes might add to the challenges in regard to enhancing our understanding of the complex biochemical and molecular mechanisms caused by primary blast-induced injury.


Subject(s)
Explosions , Hippocampus/pathology , Neurons/pathology , Sound/adverse effects , Animals , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Hippocampus/physiopathology , Neurons/cytology , PC12 Cells/pathology , Rats , Rats, Sprague-Dawley/abnormalities , Rats, Sprague-Dawley/injuries , Triazines/adverse effects
7.
Cell Death Differ ; 13(12): 2118-28, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16729028

ABSTRACT

p53 is necessary for the elimination of neural cells inappropriately differentiated or in response to stimuli. However, the role of p53 in neuronal differentiation is not certain. Here, we showed that nerve growth factor (NGF)-mediated differentiation in PC12 cells is enhanced by overexpression of wild-type p53 but inhibited by mutant p53 or knockdown of endogenous wild-type p53, the latter of which can be rescued by expression of exogenous wild-type p53. Interestingly, p53 knockdown or overexpression of mutant p53 attenuates NGF-mediated activation of TrkA, the high-affinity receptor for NGF and a tyrosine kinase, and activation of the mitogen-activated protein kinase pathway. In addition, p53 knockdown reduces the constitutive levels of TrkA, which renders PC12 cells inert to NGF. And finally, we showed that both constitutive and stimuli-induced expressions of TrkA are regulated by p53 and that induction of TrkA by activated endogenous p53 enhances NGF-mediated differentiation. Taken together, our data demonstrate that p53 plays a critical role in NGF-mediated neuronal differentiation in PC12 cells at least in part via regulation of TrkA levels.


Subject(s)
Cell Differentiation/physiology , Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Differentiation/genetics , Gene Expression Regulation , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Nerve Growth Factor/genetics , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Neurons/metabolism , Neurons/pathology , PC12 Cells/metabolism , PC12 Cells/pathology , Rats , Receptor, trkA/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Tumor Suppressor Protein p53/genetics
8.
Brain Res ; 1185: 359-65, 2007 Dec 14.
Article in English | MEDLINE | ID: mdl-17996855

ABSTRACT

Manganese has been known to induce neurological disorders similar to Parkinson's disease. The dysfunction of ubiquitin-proteasome system, a pathway involved in detoxification and targeting of damaged proteins, is connected with Parkinson's disease pathogenesis. Oxidative stress may be involved in Parkinson's disease, and may also be associated with manganese-induced neurotoxicity. In the present study, we determined the effects of manganese chloride on proteasome activity in PC12 cells. Furthermore, we investigated the relationship between oxidative stress and the change of proteasome activity. The proteasome activity of PC12 cells was measured by an ELISA method. Selective oxidative stress parameters, including malondialdehyde and protein carbonyl, were measured in PC12 cells treated with manganese chloride. Cell survival and apoptosis were measured by methyl thiazolyl tetrazolium and terminal transferase-mediated dUTP nick end-labeling. In our research, manganese chloride exposure inhibited the activity of proteasome and induced oxidative stress. Both can be reversed by antioxidant agent N-acetylcysteine. N-acetylcysteine also inhibited the cytotoxicity induced by manganese chloride. In conclusion, our results imply that proteasome inhibition may be associated with manganese-induced cytotoxicity in dopaminergic neurons, which may be connected with oxidative damage.


Subject(s)
Chlorides/adverse effects , Manganese Compounds/adverse effects , Oxidative Stress/drug effects , Proteasome Endopeptidase Complex/metabolism , Trace Elements/adverse effects , Acetylcysteine/pharmacology , Analysis of Variance , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Count/methods , Dose-Response Relationship, Drug , Drug Interactions , Enzyme-Linked Immunosorbent Assay/methods , In Situ Nick-End Labeling , PC12 Cells/drug effects , PC12 Cells/pathology , Protein Carbonylation/drug effects , Rats , Tetrazolium Salts , Thiazoles
9.
Mol Cell Biol ; 20(16): 6170-83, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10913198

ABSTRACT

HES-1 is a Hairy-related basic helix-loop-helix protein with three evolutionarily conserved regions known to define its function as a transcription repressor. The basic region, helix-loop-helix domain, and WRPW motif have been characterized for their molecular function in DNA binding, dimer formation, and corepressor recruitment, respectively. In contrast, the function conferred by a fourth conserved region, the helix 3-helix 4 (H-3/4) domain, is not known. To better understand H-3/4 domain function, we expressed HES-1 variants under tetracycline-inducible control in PC12 cells. As expected, the induced expression of moderate levels of wild-type HES-1 in PC12 cells strongly inhibited nerve growth factor-induced differentiation. This repression was dependent on the H-3/4 domain. Unexpectedly, expression of HES-1 also arrested cell growth, an effect that could be reversed upon down regulation of HES-1. Concomitant with growth arrest, there was a strong reduction in bromodeoxyuridine incorporation and PCNA protein levels, although not in cyclin D1 expression. Expression of a HES-1 protein carrying the H-3/4 domain, but not the WRPW domain, still partially inhibited both proliferation and differentiation. Transcription assays in PC12 cells directly demonstrated that the H-3/4 domain can mediate DNA-binding-dependent transcription repression, even in the absence of corepressor recruitment by the WRPW motif. HES-1 expression strongly repressed transcription of the p21(cip1) promoter, a cyclin-cyclin-dependent kinase inhibitor up regulated during NGF-induced differentiation, and the H-3/4 domain is necessary for this repression. Thus, the H-3/4 domain of HES-1 contributes to transcription repression independently of WRPW function, inhibits neurite formation, and facilitates two distinct and previously uncharacterized roles for HES-1: the inhibition of cell proliferation and the direct transcriptional repression of the NGF-induced gene, p21.


Subject(s)
Homeodomain Proteins/genetics , PC12 Cells/pathology , PC12 Cells/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation/genetics , Cell Division/genetics , Helix-Loop-Helix Motifs , Rats , Repressor Proteins , Transcription Factor HES-1 , Transcription, Genetic
10.
Food Chem Toxicol ; 102: 93-101, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28137608

ABSTRACT

Silymarin (SM) is a well-known antioxidant, anti-inflammatory and anti-cancer compound extracted from the milk thistle. Here, we investigated the protective effect of SM against acrylamide (AA)-induced neurotoxicity, mainly caused by oxidative stress, via activation of the nuclear transcription factor E2-related factor 2 (Nrf2) signalling pathway in PC12 cells. The MTT reduction assay was used to measure cell viability in various drug-treated groups and demonstrated that SM could increase cell viability in AA-treated PC12 cells. We then measured the reactive oxygen species (ROS) levels by the peroxide-sensitive fluorescent probe DCFH-DA and intracellular glutathione (GSH) and malondialdehyde (MDA) levels by absorption spectrophotometry. Our data revealed that SM could reduce ROS and MDA levels and increase GSH levels in AA-induced PC12 cells. To identify a potential mechanism for SM-induced protection, we measured the mRNA and protein expression levels of Nrf2 and its downstream target antioxidants glutathione peroxidase (Gpx), glutamate cysteine ligase catalytic subunit (GCLC) and glutamate cysteine ligase modifier subunit (GCLM) by quantitative real-time PCR and Western blot, respectively. The results suggested that SM could activate Nrf2 signalling and increase the expression of Nrf2, Gpx, GCLC and GCLM in AA-treated PC12 cells. In conclusion, SM can effectively alleviate AA-induced neurotoxicity in PC12 cells.


Subject(s)
Acrylamide/toxicity , NF-E2-Related Factor 2/metabolism , Neurotoxicity Syndromes/prevention & control , PC12 Cells/drug effects , Silymarin/pharmacology , Animals , Gene Expression Regulation/drug effects , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Malondialdehyde/metabolism , NF-E2-Related Factor 2/genetics , Neurotoxicity Syndromes/metabolism , PC12 Cells/metabolism , PC12 Cells/pathology , Rats , Reactive Oxygen Species/metabolism
11.
Nan Fang Yi Ke Da Xue Xue Bao ; 37(1): 93-96, 2017 01 20.
Article in Zh | MEDLINE | ID: mdl-28109106

ABSTRACT

OBJECTIVE: To study the effect of methamphetamine (METH) exposure on S-nitrosylation of protein disulphide isomerase and the neurotoxicity of METH in PC12 cells. METHODS: PC12 cells were exposed to different concentrations of METH, and the cell viability was assessed using the cell-counting kit-8. PC12 cells exposed to METH in the presence of the NOS inhibitor N-nitro-L-arginine (L-NNA) were examined for cell viability and S-nitrosylation of protein disulphide isomerase using the biotin-switch method, and the changes in cell morphology were examined with HE staining. RESULTS: METH exposure obviously decreased the cell viability and increased S-nitrosylation of protein disulphide isomerase, and the effect of METH was obviously inhibited by L-NNA treatment. CONCLUSION: METH can cause obvious neurotoxicity and promote S-nitrosylation of protein disulphide isomerase in PC12 cells.


Subject(s)
Cell Survival/drug effects , Methamphetamine/toxicity , PC12 Cells/drug effects , Protein Disulfide-Isomerases/metabolism , Animals , Cell Count , Cell Shape/drug effects , Enzyme Inhibitors/pharmacology , Methamphetamine/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitroarginine/pharmacology , PC12 Cells/metabolism , PC12 Cells/pathology , Rats
12.
Brain Res ; 1085(1): 57-67, 2006 Apr 26.
Article in English | MEDLINE | ID: mdl-16574083

ABSTRACT

Recently, we have reported that minocycline, a semi-synthetic tetracycline with neuroprotective effects, inhibits the in vitro ischemic-like injury and 5-lipoxygenase (5-LOX) activation in PC12 cells. In the present study, we further determined whether minocycline protects PC12 cells from excitotoxicity via inhibiting 5-LOX activation. We used N-methyl-d-aspartate (NMDA, 200 microM) to induce early (exposure for 6 h) and delayed (exposure for 6 h followed by 24 h recovery) injuries. We found that NMDA receptor antagonist ketamine, 5-LOX inhibitor caffeic acid and minocycline concentration dependently attenuated NMDA-induced early and delayed cell injuries (viability reduction and cell death). However, only ketamine (1 microM) inhibited NMDA-evoked elevation of intracellular calcium. In addition, immunohistochemical analysis showed that NMDA induced 5-LOX translocation to the nuclear membrane after 1- to 6-h exposure which was confirmed by Western blotting, indicating that 5-LOX was activated. Ketamine, caffeic acid and minocycline (each at 1 microM) inhibited 5-LOX translocation after early injury. After delayed injury, PC12 cells were shrunk, and 5-LOX was translocated to the nuclei and nuclear membrane; ketamine, caffeic acid and minocycline inhibited both cell shrinking and 5-LOX translocation. As a control, 12-LOX inhibitor baicalein showed a weak effect on cell viability and death, but no effect on 5-LOX translocation. Therefore, we conclude that the protective effect of minocycline on NMDA-induced injury is partly mediated by inhibiting 5-LOX activation.


Subject(s)
Arachidonate 5-Lipoxygenase/physiology , Excitatory Amino Acid Agonists/toxicity , Minocycline/pharmacology , N-Methylaspartate/toxicity , Neuroprotective Agents/pharmacology , PC12 Cells/drug effects , Analysis of Variance , Animals , Blotting, Western/methods , Calcium/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Flavanones/pharmacology , Immunohistochemistry/methods , Ketamine/pharmacology , PC12 Cells/pathology , Rats , Tetrazolium Salts , Thiazoles , Time Factors
13.
Life Sci ; 79(24): 2293-302, 2006 Nov 10.
Article in English | MEDLINE | ID: mdl-16935310

ABSTRACT

Oxidative stress induced cell injury is reported to contribute to the pathogenesis of cerebral ischemia. Reactive oxygen species such as hydrogen peroxide (H2O2) and superoxide radical along with nitric oxide and peroxynitrite generated during ischemia-reperfusion injury, causes the overactivation of poly (ADP-ribose) polymerase (PARP) leading to neuronal cell death. In the present study we have evaluated the effects of PARP inhibitor, 8-hydroxy-2 methyl-quinazolin-4-[3H]one (NU1025) in H2O2 and 3-morphilinosyndonimine (SIN-1) induced cytotoxicity in PC12 cells as well as in middle cerebral artery occlusion (MCAO) induced focal cerebral ischemia in rats. Exposure of PC12 cells to H2O2 (0.4 mM) and SIN-1 (0.8 mM) resulted in a significant decrease in cell viability after 6 h. Pretreatment with NU1025 (0.2 mM) restored cell viability to approximately 73 and 82% in H2O2 and SIN-1 injured cells, respectively. In MCAO studies, NU1025 was administered at different time points (1 h before reperfusion, immediately before reperfusion, 3 h after reperfusion and 6 h after reperfusion). NU1025 at 1 and 3 mg/kg reduced total infarct volume to 25% and 45%, respectively, when administered 1 h before reperfusion. NU1025 also produced significant improvement in neurological deficits. Neuroprotection with NU1025 was associated with reduction in PAR accumulation, reversal of brain NAD depletion and reduction in DNA fragmentation. Results of this study demonstrate the neuroprotective activity of NU1025 and suggest its potential in cerebral ischemia.


Subject(s)
Apoptosis/drug effects , Brain Ischemia/drug therapy , Enzyme Inhibitors/therapeutic use , NAD/metabolism , Neuroprotective Agents/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors , Quinazolines/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Survival/drug effects , DNA Fragmentation , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Combinations , Enzyme Inhibitors/pharmacology , Hydrogen Peroxide/pharmacology , Male , Middle Cerebral Artery/injuries , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , Neuroprotective Agents/pharmacology , PC12 Cells/drug effects , PC12 Cells/pathology , Quinazolines/pharmacology , Rats , Rats, Sprague-Dawley , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
14.
Eur J Med Chem ; 41(2): 241-52, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16412536

ABSTRACT

Several 1-alkyl-1,2,3,4-tetrahydroisoquinoline (TIQ) derivatives, which may play a role in Parkinson's disease, have been synthesized via Pummerer-type cyclization of the sulfonium ion formed in situ from N-formyl sulfoxide. Using an in vitro trypan blue exclusion assay, high concentrations of TIQ derivatives possessing bulky alkyl group substituents such as 1-cyclobutyl-, 1-cyclohexyl-, 1-phenyl- or 1-benzyl- at the C-1 position were found to significantly affect the viability of PC12 cells. Moreover, TIQ derivatives that moderately or strongly induced apoptosis (e.g., 1-phenyl-TIQ and 1-cyclohexyl-TIQ, respectively) paralleled the results obtained using the trypan blue exclusion assay. These results suggest that the size and electron-donating properties of functional groups may affect the cytotoxicity of TIQ derivatives.


Subject(s)
Apoptosis/drug effects , Brain/pathology , Neurotoxins/metabolism , Parkinson Disease/pathology , Tetrahydroisoquinolines/chemical synthesis , Tetrahydroisoquinolines/pharmacology , Animals , Cells, Cultured , Membrane Transport Proteins/metabolism , Microscopy, Fluorescence, Multiphoton , PC12 Cells/pathology , Parkinson Disease/drug therapy , Rats
15.
Basic Clin Pharmacol Toxicol ; 99(5): 353-7, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17076686

ABSTRACT

Lignophenol, prepared using a phase-separation system, is a derivative of lignin, which is one of the components in the plant cell wall, and possesses high phenolic function, high stability and antioxidant properties. However, little is known about the beneficial effect of lignophenol. In this study, we investigated the protective effect of lignophenol from the beech tree (Fagus crenata Blume) on copper- and zinc-mediated apoptosis in PC12 cells by using DNA fragmentation and TUNEL assays. In DNA fragmentation assays, the DNA ladder patterns in the PC12 cells treated with 200 microM Cu and 200 microM Zn were enhanced, whereas the DNA ladder pattern was hardly observed in these cells treated with 20 mM lignophenol. In the TUNEL assay, TUNEL signals increased significantly in the untreated PC12 cells exposed to 200 microM Cu compared with the control. In contrast, the degree of apoptosis in the 20 mM lignophenol-treated cells was significantly lower than in the untreated cells, indicating that lignophenol inhibited Cu-induced apoptotic cell death in PC 12 cells. In the 200 microM Zn-exposed group, the degree of apoptosis in the 20 mM lignophenol-treated cells was also low compared with the untreated cells. In conclusion, these results suggest that lignophenol plays a role in protecting against Cu- and Zn-mediated PC12 apoptotic cell death.


Subject(s)
Apoptosis/drug effects , Copper/pharmacology , Fagus/chemistry , Lignin/pharmacology , PC12 Cells/drug effects , Zinc/pharmacology , Animals , Cell Survival/drug effects , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Drug Combinations , Electrophoresis, Agar Gel , In Situ Nick-End Labeling , Lignin/analogs & derivatives , Lignin/chemistry , PC12 Cells/pathology , Plant Extracts/pharmacology , Rats
16.
Cancer Res ; 56(9): 2161-6, 1996 May 01.
Article in English | MEDLINE | ID: mdl-8616866

ABSTRACT

The molecular mechanism of cell death due to hypoxia has not been elucidated. Our recent observations that overexpression of the anti-apoptotic proto-oncogene bcl-2 and a bcl-2-related gene, bcl-x, prevents hypoxic cell death suggest that hypoxia induces apoptosis. Using electron microscopy and confocal and nonconfocal fluorescence microscopy, we show here that hypoxia does, in fact, induce both necrosis and apoptosis, and that the proportion of these two modes is highly dependent on the cell type. Overexpression of Bcl-2 or Bcl-Xl blocks hypoxia-induced apoptosis in a dose-dependent manner.


Subject(s)
Apoptosis , Necrosis , Proto-Oncogene Proteins/biosynthesis , Animals , Cell Hypoxia , Gene Transfer Techniques , Microscopy, Confocal , Microscopy, Fluorescence , PC12 Cells/metabolism , PC12 Cells/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2 , Rats , bcl-X Protein
17.
Zhong Yao Cai ; 29(10): 1051-5, 2006 Oct.
Article in Zh | MEDLINE | ID: mdl-17326406

ABSTRACT

OBJECTIVE: To study the protective effects of flavonoids from Cuscuta chinensis (CF) on oxidative stress in cultured PC12 cells and investigate the mechanism of the effects. METHODS: The cell viability was analyzed by MTT method and the radical scavenging activity of CF was examined by DPPH (1, 1-diphenyl-2-picrylhydrazyl). The morphological changes were observed by Hoechst 33258 staining assay, and the apoptosis rate of PC12 cells was detected by propidium iodide stain flow cytometry (FCM). RESULTS: Application with 0.3-0.5 mM H2O2 induced a dose and time dependent viability loss in PC12 cells; Treatment with 0.5 mM H2O2 for 24 h was shown to cause nearly 50% viabliity loss and apoptosis in PC12 cells; Pretreatment with different concentrations of CF for 0.5 h increased the survival rate of PC12 cells, inhibited apoptosis induced by H2O2; CF had the activity of scavenging free radicals generated by DPPH in a dose-dependent manner. CONCLUSION: CF can protect PC12 cells against oxidative stress. The mechanism of it may be the ability of scavenging ROS and increasing the activity of antioxidant enzyme.


Subject(s)
Apoptosis/drug effects , Cuscuta/chemistry , Flavonoids/pharmacology , Free Radical Scavengers/pharmacology , PC12 Cells/drug effects , Animals , Cell Survival/drug effects , Dose-Response Relationship, Drug , Flavonoids/administration & dosage , Flow Cytometry , Free Radicals/metabolism , Hydrogen Peroxide , Oxidative Stress/drug effects , PC12 Cells/pathology , Plants, Medicinal/chemistry , Rats
18.
J Neurosci ; 19(24): 10747-56, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10594058

ABSTRACT

Protein 4.1N is a neuronal selective isoform of the erythrocyte membrane cytoskeleton protein 4.1R. In the present study, we demonstrate an interaction between 4.1N and nuclear mitotic apparatus protein (NuMA), a nuclear protein required for mitosis. The binding involves the C-terminal domain of 4.1N. In PC12 cells treatment with nerve growth factor (NGF) elicits translocation of 4. 1N to the nucleus and promotes its association with NuMA. Specific targeting of 4.1N to the nucleus arrests PC12 cells at the G1 phase and produces an aberrant nuclear morphology. Inhibition of 4.1N nuclear translocation prevents the NGF-mediated arrest of cell division, which can be reversed by overexpression of 4.1N. Thus, nuclear 4.1N appears to mediate the antiproliferative actions of NGF by antagonizing the role of NuMA in mitosis.


Subject(s)
Cytoskeletal Proteins/metabolism , Membrane Proteins , Nerve Growth Factor/pharmacology , Neuropeptides/metabolism , Nuclear Proteins/metabolism , PC12 Cells/metabolism , Animals , Biological Transport/drug effects , Biological Transport/physiology , Cell Cycle Proteins , Cell Division/drug effects , Cell Division/physiology , Cell Nucleus/metabolism , G1 Phase/physiology , Nerve Growth Factor/antagonists & inhibitors , PC12 Cells/pathology , Rats
19.
Endocrinology ; 146(12): 5332-40, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16150914

ABSTRACT

We here report a new physiological system that governs catecholamine synthesis involving bone morphogenetic proteins (BMPs) and activin in the rat pheochromocytoma cell line, PC12. BMP type I receptors, including activin receptor-like kinase-2 (ALK-2) (also referred to as ActRIA) and ALK-3 (BMPRIA), both type II receptors, ActRII and BMPRII, as well as the ligands BMP-2, -4, and -7 and inhibin/activin subunits were expressed in PC12 cells. PC12 cells predominantly secrete dopamine, whereas noradrenaline and adrenaline production is negligible. BMP-2, -4, -6, and -7 and activin A each suppressed dopamine and cAMP synthesis in a dose-dependent fashion. The BMP ligands also decreased 3,4-dihydroxyphenylalanine decarboxylase mRNA expression, whereas activin suppressed tyrosine hydroxylase expression. BMPs induced both Smad1/5/8 phosphorylation and Tlx2-Luc activation, whereas activin stimulated 3TP-Luc activity and p38 MAPK phosphorylation. ERK signaling was not affected by BMPs or activin. Dexamethasone enhanced catecholamine synthesis, accompanying increases in tyrosine hydroxylase and 3,4-dihydroxyphenylalanine decarboxylase transcription without cAMP accumulation. In the presence of dexamethasone, BMPs and activin failed to reduce dopamine as well as cAMP production. In addition, dexamethasone modulated mitotic suppression of PC12 induced by BMPs in a ligand-dependent manner. Furthermore, intracellular BMP signaling was markedly suppressed by dexamethasone treatment and the expression of ALK-2, ALK-3, and BMPRII was significantly inhibited by dexamethasone. Collectively, the endogenous BMP/activin system plays a key role in the regulation of catecholamine production. Controlling activity of the BMP system may be critical for glucocorticoid-induced catecholamine synthesis by adrenomedullar cells.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Catecholamines/biosynthesis , Glucocorticoids/metabolism , PC12 Cells/metabolism , Activins/metabolism , Activins/pharmacology , Animals , Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/pharmacology , Cyclic AMP/antagonists & inhibitors , Dexamethasone/pharmacology , Dopamine Antagonists/pharmacology , Glucocorticoids/pharmacology , Inhibin-beta Subunits/pharmacology , Ligands , Mitosis/drug effects , PC12 Cells/pathology , Rats , Signal Transduction , Smad Proteins/metabolism
20.
J Cereb Blood Flow Metab ; 25(2): 154-162, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15647748

ABSTRACT

Although ischemic tolerance has been described in a variety of primary cell culture systems, no similar in vitro models have been reported with any cell line. A model of ischemic preconditioning in the rat pheochromocytoma PC12 cell line is described here. When compared to nonpreconditioned cells, preexposure of PC12 cells to 6 hours of oxygen and glucose deprivation (OGD) significantly increased cell viability after 15 hours of OGD 24 hours later. Flow cytometry analysis of cells labeled with specific markers for apoptosis, Annexin V, and Hoechst 33342, and of DNA content, revealed that apoptosis is involved in OGD-induced PC12 cell death and that preconditioning of the cells mainly counteracts the effect of apoptosis. Immunocytochemistry of caspase-3, a central executioner in the apoptotic process, further confirmed the activation of apoptotic pathways in OGD-induced PC12 cell death. This model may be useful to investigate the cellular mechanisms involved in neuronal transient tolerance following ischemia.


Subject(s)
Ischemic Preconditioning/methods , Models, Biological , PC12 Cells/pathology , Animals , Annexin A5/metabolism , Apoptosis/physiology , Benzimidazoles/metabolism , Caspase 3 , Caspases/metabolism , Flow Cytometry , Immunohistochemistry , Rats
SELECTION OF CITATIONS
SEARCH DETAIL