Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
Nat Prod Res ; 34(11): 1521-1527, 2020 Jun.
Article in English | MEDLINE | ID: mdl-30445866

ABSTRACT

A new ester (1) and a terpenoid (2) were isolated from the dried whole plant of Disporopsis aspersa (HUA) ENGL. ex DIELS for the first time and their structures were elucidated, as well as their biological activities are described. The two compounds all showed good antifungal activities, especially furanone (2) exhibited better antifungal activity against Pseudoperonospora cubensis and Phytophthora infestans with EC50 value of 22.82, 18.90 µg/mL, respectively. Compound 1 exhibited a significant promotion on the neurite outgrowth in NGF-induced PC-12 cells, and moderate inhibition on the NO production induced by lipopolysaccharide (LPS) in BV-2 microglial cells.


Subject(s)
Anti-Inflammatory Agents/isolation & purification , Antifungal Agents/isolation & purification , Asparagaceae/chemistry , Neuronal Outgrowth/drug effects , Plant Extracts/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Antifungal Agents/pharmacology , Esters/isolation & purification , Esters/pharmacology , Microglia/drug effects , Nitric Oxide/antagonists & inhibitors , PC12 Cells/drug effects , PC12 Cells/ultrastructure , Phytophthora infestans/drug effects , Plant Extracts/chemistry , Rats , Terpenes/isolation & purification , Terpenes/pharmacology
2.
J Neurochem ; 146(3): 235-250, 2018 08.
Article in English | MEDLINE | ID: mdl-29808474

ABSTRACT

Prohibitin (PHB) is a ubiquitously expressed and evolutionarily conserved mitochondrial protein with multiple functions. We have recently shown that PHB up-regulation offers robust protection against neuronal injury in models of cerebral ischemia in vitro and in vivo, but the mechanism by which PHB affords neuroprotection remains to be elucidated. Here, we manipulated PHB expression in PC12 neural cells to investigate its impact on mitochondrial function and the mechanisms whereby it protects cells exposed to oxidative stress. PHB over-expression promoted cell survival, whereas PHB down-regulation diminished cell viability. Functionally, manipulation of PHB levels did not affect basal mitochondrial respiration, but it increased spare respiratory capacity. Moreover, PHB over-expression preserved mitochondrial respiratory function of cells exposed to oxidative stress. Preserved respiratory capacity in differentiated PHB over-expressing cells exposed to oxidative stress was associated with an elongated mitochondrial morphology, whereas PHB down-regulation enhanced fragmentation. Mitochondrial complex I oxidative degradation was attenuated by PHB over-expression and increased in PHB knockdown cells. Changes in complex I degradation were associated with alterations of respiratory chain supercomplexes. Furthermore, we showed that PHB directly interacts with cardiolipin and that down-regulation of PHB results in loss of cardiolipin in mitochondria, which may contribute to destabilizing respiratory chain supercomplexes. Taken together, these data demonstrate that PHB modulates mitochondrial integrity and bioenergetics under oxidative stress, and suggest that the protective effect of PHB is mediated by stabilization of the mitochondrial respiratory machinery and its functional capacity, by the regulation of cardiolipin content. Open Data: Materials are available on https://cos.io/our-services/open-science-badges/ https://osf.io/93n6m/.


Subject(s)
Mitochondria/metabolism , Neurons/ultrastructure , Oxidative Stress/physiology , PC12 Cells/ultrastructure , Repressor Proteins/metabolism , Animals , Cardiolipins/metabolism , Cell Survival , Cells, Cultured , Dose-Response Relationship, Drug , Electron Transport Chain Complex Proteins/metabolism , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/genetics , Neurons/drug effects , Neurons/metabolism , Oligomycins/pharmacology , Oxidants/pharmacology , Oxidative Stress/drug effects , Oxygen Consumption/physiology , Prohibitins , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Repressor Proteins/genetics , Time Factors , Transfection
3.
Mol Cell Neurosci ; 80: 89-99, 2017 04.
Article in English | MEDLINE | ID: mdl-28254618

ABSTRACT

Parkin is an E3 ubiquitin ligase whose mutations cause autosomal recessive juvenile Parkinson's disease (PD). Unlike the human phenotype, parkin knockout (KO) mice show no apparent dopamine neuron degeneration, although they demonstrate reduced expression and activity of striatal mitochondrial proteins believed to be necessary for neuronal survival. Instead, parkin-KO mice show reduced striatal evoked dopamine release, abnormal synaptic plasticity, and non-motor symptoms, all of which appear to mimic the preclinical features of Parkinson's disease. Extensive studies have screened candidate synaptic proteins responsible for reduced evoked dopamine release, and synaptotagmin XI (Syt XI), an isoform of Syt family regulating membrane trafficking, has been identified as a substrate of parkin in humans. However, its expression level is unaltered in the striatum of parkin-KO mice. Thus, the target(s) of parkin and the molecular mechanisms underlying the impaired dopamine release in parkin-KO mice remain unknown. In this study, we focused on Syt IV because of its highly homology to Syt XI, and because they share an evolutionarily conserved lack of Ca2+-binding capacity; thus, Syt IV plays an inhibitory role in Ca2+-dependent neurotransmitter release in PC12 cells and neurons in various brain regions. We found that a proteasome inhibitor increased Syt IV protein, but not Syt XI protein, in neuron-like, differentiated PC12 cells, and that parkin interacted with and polyubiquitinated Syt IV, thereby accelerating its protein turnover. Parkin overexpression selectively degraded Syt IV protein, but not Syt I protein (indispensable for Ca2+-dependent exocytosis), thus enhancing depolarization-dependent exocytosis. Furthermore, in parkin-KO mice, the level of striatal Syt IV protein was increased. Our data indicate a crucial role for parkin in the proteasomal degradation of Syt IV, and provide a potential mechanism of parkin-regulated, evoked neurotransmitter release.


Subject(s)
Neurons/metabolism , Proteolysis , Synaptotagmins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/physiology , Animals , Autoantigens/pharmacology , COS Cells , Chlorocebus aethiops , Corpus Striatum/cytology , Exocytosis/genetics , Membrane Proteins/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factor/pharmacology , Oligopeptides/pharmacology , PC12 Cells/drug effects , PC12 Cells/ultrastructure , Proteasome Inhibitors/pharmacology , Protein Transport , Proteolysis/drug effects , Rats , Synaptotagmins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/drug effects , Vesicle-Associated Membrane Protein 2/metabolism
4.
ACS Chem Neurosci ; 7(9): 1255-63, 2016 09 21.
Article in English | MEDLINE | ID: mdl-27433833

ABSTRACT

The Aß complexes of some redox-active species, such as Cu, cause oxidative stress and induce severe toxicity by generating reactive oxygen species (ROS). Thus, Cu chelation therapy should be considered as a valuable strategy for the treatment of Alzheimer's disease (AD). However, more attention should be paid to the specific chelating ability of these chelating agents. Herein, a tripeptide GGH was used to selectively chelate the Cu(2+) in Aß-Cu complex in the presence of other metal ions (e.g., K(+), Ca(2+), Ni(2+), Mg(2+), and Zn(2+)) as shown by isothermal titration calorimetry results. GGH decreased the level of HO(•) radicals by preventing the formation of intermediate Cu(I) ion. Thus, the Cu species completely lost its catalytic activity at a superequimolar GGH/Cu(II) ratio (4:1) as observed by UV-visible spectroscopy, coumarin-3-carboxylic acid fluorescence, and BCA assay. Moreover, (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) (MTT) assay indicates that GGH increased PC-12 cell viability from 36% to 63%, and neurotoxicity partly triggered by ROS decreased. These results indicate potential development of peptide chelation therapy for AD treatment.


Subject(s)
Amyloid beta-Peptides/metabolism , Chelating Agents/pharmacology , Copper/metabolism , Oligopeptides/pharmacology , Oxidation-Reduction/drug effects , PC12 Cells/drug effects , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/ultrastructure , Animals , Cell Differentiation/drug effects , Chelating Agents/chemistry , Copper/chemistry , Copper/pharmacology , Dose-Response Relationship, Drug , Metals/metabolism , Metals/pharmacology , Microscopy, Electron, Transmission , Neurons/drug effects , Oxidative Stress/drug effects , PC12 Cells/ultrastructure , Protein Binding/drug effects , Rats , Reactive Oxygen Species/metabolism , Time Factors
5.
Neurosignals ; 23(1): 1-10, 2015.
Article in English | MEDLINE | ID: mdl-26656295

ABSTRACT

BACKGROUND: Chorein, a protein supporting activation of phosphoinositide 3 kinase (PI3K), participates in the regulation of actin polymerization and cell survival. A loss of function mutation of the chorein encoding gene VPS13A (vacuolar protein sorting-associated protein 13A) leads to chorea-acanthocytosis (ChAc), a neurodegenerative disorder with simultaneous erythrocyte akanthocytosis. In blood platelets chorein deficiency has been shown to compromise expression of vesicle-associated membrane protein 8 (VAMP8) and thus degranulation. The present study explored whether chorein is similarly involved in VAMP8 expression and dopamine release of pheochromocytoma (PC12) cells. METHODS: Chorein was down-regulated by silencing in PC12 cells. Transmission electron microscopy was employed to quantify the number of vesicles, RT-PCR to determine transcript levels, Western blotting to quantify protein expression and ELISA to determine dopamine release. RESULTS: Chorein silencing significantly reduced the number of vesicles, VAMP8 transcript levels and VAMP8 protein abundance. Increase of extracellular K+ from 5 mM to 40 mM resulted in marked stimulation of dopamine release, an effect significantly blunted by chorein silencing. CONCLUSIONS: Chorein deficiency down-regulates VAMP8 expression, vesicle numbers and dopamine release in pheochromocytoma cells.


Subject(s)
Dopamine/metabolism , Vesicular Transport Proteins/metabolism , Animals , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Microscopy, Electron, Transmission , PC12 Cells/drug effects , PC12 Cells/ultrastructure , Potassium Chloride/pharmacology , R-SNARE Proteins/genetics , R-SNARE Proteins/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Small Interfering/ultrastructure , Rats , Transfection , Vesicular Transport Proteins/genetics
6.
Hear Res ; 320: 11-7, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25576787

ABSTRACT

Modulation of a materials surface topography can be used to steer various aspects of adherent cell behaviour, such as cell directional organization. Especially nanometric sized topographies, featuring sizes similar to for instance the axons of the spiral ganglion cells, are interesting for such purpose. Here, we utilized nanosized grooves in the range of 75-500 nm, depth of 30-150 nm, and pitches between 150 nm and 1000 nm for cell culture of neuron-like PC12 cells. The organizational behaviour was evaluated after 7 days of culture by bright field and scanning electron microscopy. Nanotopographies were shown to induce aligned cell-body/axon orientation and an increased axonal outgrowth. Our findings suggest that a threshold for cell body alignment of neuronal cells exists on grooved topographies with a groove width of 130 nm, depth of 70 nm and pitch of 300 nm, while axon alignment can already be induced by grooves with 135 nm width, 52 nm depth and 200 nm pitch. However, no threshold has been found for axonal outgrowth, as all of the used patterns increased outgrowth of PC12-axons. In conclusion, surface nanopatterns have the potential to be utilized as an electrode modification for a stronger separation of cells, and can be used to direct cells towards the electrode contacts of cochlear implants.


Subject(s)
Axons/ultrastructure , Cell Culture Techniques/methods , Cell Differentiation/physiology , Nanostructures/ultrastructure , Neurons/cytology , PC12 Cells/cytology , Animals , Axons/physiology , Cell Adhesion/physiology , Cells, Cultured , Cochlear Implants , Electrodes , Microscopy, Atomic Force , Microscopy, Electron, Scanning , Models, Animal , Neurons/physiology , Neurons/ultrastructure , PC12 Cells/physiology , PC12 Cells/ultrastructure , Polystyrenes , Rats
7.
Am J Chin Med ; 41(6): 1343-59, 2013.
Article in English | MEDLINE | ID: mdl-24228605

ABSTRACT

Rotenone, an inhibitor of mitochondrial complex I, has been widely regarded as a neurotoxin because it induces a Parkinson's disease-like syndrome. The fruit and root bark of Lycium chinense Miller have been used as traditional medicines in Asia to treat neurodegenerative diseases. In this study, we examined the neuroprotective effects of Lycium chinense Miller extracts in rotenone-treated PC12 cells. Treatment with rotenone reduced PC12 cell viability and cellular ATP levels. Conversely, caspase 3/7 activity, the ratio of Bax:Bcl-2 expression levels, mitochondrial superoxide level, and intracellular calcium (Ca(2+)) concentration were elevated. Pretreatment with Lycium chinense Miller extracts significantly increased cell viability and ATP levels. Additionally, they attenuated caspase activation, mitochondrial membrane depolarization and mitochondrial superoxide production. Moreover, confocal microscopy showed that the mitochondrial staining pattern was restored from that of extracts treated cells and that the increase in intracellular Ca (2+) level was blunted by treatment with the extracts. Our results suggest that Lycium chinense Miller extracts may have the possible beneficial effects in Parkinson's disease by attenuating rotenone induced toxicity.


Subject(s)
Cell Survival/drug effects , Lycium , Neuroprotective Agents , PC12 Cells/drug effects , PC12 Cells/metabolism , Phytotherapy , Plant Extracts/pharmacology , Rotenone/antagonists & inhibitors , Rotenone/toxicity , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Caspases/metabolism , Cells, Cultured , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , PC12 Cells/ultrastructure , Parkinson Disease/drug therapy , Plant Extracts/therapeutic use , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Superoxides/metabolism , bcl-2-Associated X Protein/metabolism
9.
Arch Ital Biol ; 150(2-3): 194-217, 2012.
Article in English | MEDLINE | ID: mdl-23165879

ABSTRACT

Mutations in the PTEN-induced putative kinase1 (PINK1) represent the second most frequent cause of autosomal recessive Parkinson's disease. The PINK1 protein mainly localizes to mitochondria and interacts with a variety of proteins, including the pro-autophagy protein beclin1 and the ubiquitin-ligase parkin. Upon stress conditions, PINK1 is known to recruit parkin at the surface of dysfunctional mitochondria and to activate the mitophagy cascade. Aim of this study was to use a simple and highly reproducible catecholamine cell model and transmission electron microscopy to characterize whether PINK1 could affect mitochondrial homeostasis, the recruitment of specific proteins at mitochondria, mitophagy and apoptosis. Samples were analyzed both in baseline conditions and following treatment with methamphetamine (METH), a neurotoxic compound which strongly activates autophagy and produces mitochondrial damage. Our data provide robust sub-cellular evidence that the modulation of PINK1 levels dramatically affects the morphology and number of mitochondria and the amount of cell death. In particular, especially upon METH exposure, PINK1 is able to increase the total number of mitochondria, concurrently recruit beclin1, parkin and ubiquitin and enhance the clearance of damaged mitochondria. In the absence of functional PINK1 and upon autophagy stress, we observe a failure of the autophagy system at large, with marked accumulation of dysfunctional mitochondria and dramatic increase of apoptotic cell death. These findings highlight the strong neuroprotective role of PINK1 as a key protein in the surveillance and regulation of mitochondrial homeostasis.


Subject(s)
Autophagy/genetics , Mitochondria/genetics , Mutation/genetics , Protein Kinases/genetics , Animals , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Cell Death/genetics , Central Nervous System Stimulants/pharmacology , Humans , Membrane Proteins/metabolism , Methamphetamine/pharmacology , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/ultrastructure , PC12 Cells/drug effects , PC12 Cells/ultrastructure , RNA, Small Interfering/genetics , Rats , Subcellular Fractions/metabolism , Subcellular Fractions/ultrastructure , Transfection , Ubiquitin-Protein Ligases/metabolism
10.
Brain Res Bull ; 87(1): 30-6, 2012 Jan 04.
Article in English | MEDLINE | ID: mdl-21985983

ABSTRACT

GPR12, an orphan G protein-coupled receptor, constitutively activates the Gs signaling pathway and further increases intracellular cyclic AMP. GPR12 overexpression has been reported to promote neurite extension in neurons or transform neuro2a neuroblastoma cells into neuron-like cells. However, the possible effects and mechanisms of GPR12 in the differentiation of PC12 cells are still unknown. The present study shows that GPR12 overexpression induced PC12 cells differentiation into neuron-like cells with enlarged cell sizes and neuritogenesis possibly via activation of Erk1/2 signaling and significantly increased the expression of several neurite outgrowth-related genes, including Bcl-xL, Bcl-2 and synaptophysin. These findings indicate that GPR12 may play a role in neurite outgrowth during PC12 cell differentiation.


Subject(s)
Cell Differentiation/physiology , Neurites/physiology , PC12 Cells/physiology , PC12 Cells/ultrastructure , Receptors, G-Protein-Coupled/metabolism , Animals , Neurons/cytology , Neurons/physiology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Receptors, G-Protein-Coupled/genetics , Signal Transduction/physiology , bcl-X Protein/genetics , bcl-X Protein/metabolism
12.
J Alzheimers Dis ; 27(1): 61-73, 2011.
Article in English | MEDLINE | ID: mdl-21750376

ABSTRACT

Amyloid-ß (Aß) peptide-induced neurotoxicity is typically associated with cell death through mechanisms not entirely understood. Here, we investigated stress signaling events triggered by soluble Aß in differentiated rat neuronal-like PC12 cells. Morphologic evaluation of apoptosis confirmed that Aß induced nuclear fragmentation that was prevented by pre-treatment with the antiapoptotic bile acid tauroursodeoxycholic acid (TUDCA). In addition, Aß exposure triggered an early signaling response by the endoplasmic reticulum (ER) and caspase-12-mediated apoptosis, which, however, was independent of the ER-stress pathway. Furthermore, ER stress markers, including GRP94, ATF-6α, CHOP, and eIF2α, were strongly downregulated by Aß, independent of protein degradation, and partially restored by TUDCA. Calpain inhibition prevented caspase-12 activation and reduced levels of ATF-6α. Importantly, Aß-induced GRP94 downregulation was related to protein secretion and partially rescued through inhibition of the secretory pathway by geldanamycin and brefeldin. In conclusion, we showed that the ER is a proximal stress sensor for soluble Aß-induced toxicity, resulting in caspase-12 activation and cell death in PC12 neuronal cells. Moreover, ER chaperone GRP94 secretion was associated with Aß-induced apoptotic signaling. These data provide new information linking apoptotic properties of Aß peptide to distinct subcellular mechanisms of toxicity. Further characterization of this signaling pathway is likely to provide new perspectives for modulation of amyloid-induced apoptosis.


Subject(s)
Amyloid beta-Peptides/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , HSP70 Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Peptide Fragments/pharmacology , Activating Transcription Factor 6/metabolism , Animals , Apoptosis/drug effects , Brefeldin A/pharmacology , Calcium/metabolism , Caspase 12/metabolism , Cell Survival/drug effects , Cholagogues and Choleretics/pharmacology , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/drug effects , Lactones/pharmacology , PC12 Cells/drug effects , PC12 Cells/ultrastructure , Rats , Sesquiterpenes/pharmacology , Taurochenodeoxycholic Acid/pharmacology , Transcription Factor CHOP/metabolism
13.
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
14.
Synapse ; 64(10): 765-72, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20698031

ABSTRACT

PC12 cells have been used as a model of sympathetic neurons. Nerve growth factor (NGF), basic fibroblast growth factor (bFGF), and cAMP induce neurite outgrowth from PC12 cells. cAMP induced a greater number of neurites than did NGF. In particular, we attempted to elucidate whether PC12 cell neurites, induced by several factors including NGF, bFGF, and cAMP, form synapses, and whether each neurite has presynaptic and postsynaptic properties. Using scanning electron microscopy (SEM) and transmission electron microscopy (TEM), we observed that neurites are connected to each other. The connected regions presented dense core vesicles and a clathrin-coated membrane invagination. In addition, typical maker proteins for axon and dendrite were identified by an immuno-staining method. Tau-1, an axonal marker in neurons, was localized at a high concentration in the terminal tips of neurites from PC12 cells, which were connected to neurite processes containing MAP-2, a dendritic marker in neurons. Furthermore, neurites containing SV2 and synaptotagmin, markers of synaptic vesicles, were in contact with neurites harboring drebrin, a marker of the postsynaptic membrane, suggesting that neurites from PC12 cells induced by NGF, bFGF, and cAMP may form synapse-like structures. Tat-C3 toxin, a Rho inhibitor, augmented neurite outgrowth induced by NGF, bFGF, and cAMP. Tat-C3 toxin together with neurotrophins also exhibited synapse-like structures between neurites. However, it remains to be studied whether RhoA inhibition plays a role in the formation of synapse-like structures in PC12 cells.


Subject(s)
Neurites/ultrastructure , Synapses/ultrastructure , Animals , Antibodies, Monoclonal/metabolism , Cyclic AMP/pharmacology , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 2/pharmacology , Membrane Glycoproteins/metabolism , Microscopy, Electron/methods , Microtubule-Associated Proteins/metabolism , Nerve Growth Factor/pharmacology , Nerve Tissue Proteins/metabolism , Neurites/drug effects , Neurites/physiology , PC12 Cells/drug effects , PC12 Cells/ultrastructure , Rats , Synapses/drug effects , Synapses/metabolism , Synaptotagmins/metabolism , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
15.
Neurosci Res ; 66(1): 14-21, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19766678

ABSTRACT

The glucose analog 2-deoxy-d-glucose (2DG) depletes cells of glucose. Inhibition of glycosylation caused by glucose depletion induces endoplasmic reticulum (ER) stress with subsequent apoptosis. Glucose-regulated protein 78 (GRP78) is a molecular chaperone that acts within the ER. During ER stress, GRP78 expression is induced as part of the unfolded protein response (UPR). We found that nerve growth factor (NGF) prevented 2DG-triggered ER stress-mediated apoptosis, but not the induction of GRP78 expression, in PC12 cells. Surprisingly, GRP78 expression was further up-regulated when NGF was added to 2DG-treated PC12 cells. When a specific inhibitor of phosphatidylinositol 3-kinase (PI3-K), LY294002, was added to 2DG plus NGF-treated cells, both the effects of NGF on 2DG-induced apoptosis and GRP78 expression were significantly diminished. In addition, versipelostatin (VST), a specific inhibitor of GRP78 expression, and small interfering RNA (siRNA) against GRP78 mRNA also decreased both the effects of NGF on 2DG-induced apoptosis and GRP78 expression. RT-PCR and Western blot analyses revealed that enhanced production of nuclear p50 ATF6, but not spliced XBP1, mainly contributed to the NGF-induced enhancement of GRP78 expression in 2DG-treated cells. These results suggest that the NGF-activated PI3-K/Akt signaling pathway plays a protective role against ER stress-mediated apoptosis via enhanced expression of GRP78 in PC12 cells.


Subject(s)
Antimetabolites/pharmacology , Apoptosis/drug effects , Deoxyglucose/pharmacology , Endoplasmic Reticulum/drug effects , Heat-Shock Proteins/metabolism , Nerve Growth Factor/pharmacology , Analysis of Variance , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Endoplasmic Reticulum Chaperone BiP , Enzyme Inhibitors/pharmacology , Heat-Shock Proteins/genetics , In Situ Nick-End Labeling/methods , PC12 Cells/ultrastructure , RNA, Messenger/metabolism , Rats , Regulatory Factor X Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection/methods , Up-Regulation/drug effects , X-Box Binding Protein 1
16.
Neurotoxicology ; 30(4): 581-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19501118

ABSTRACT

Herein we demonstrate that PC12 cells, which overexpress human wild-type amyloid-beta precursor protein (AbetaPPwt) or AbetaPP bearing double Swedish mutation (AbetaPPsw), reveal phenotype characteristic for Alzheimer's disease (AD). The examination of cell ultrastructure showed the presence of peptide aggregates within the cells, activation of endosomal-lysosomal system and extensive exocytosis. Furthermore, the autophagy induction was also characteristic hallmark of amyloid-beta-induced cytotoxicity. Morphological changes were positively correlated with the extent of phosphorylated glycogen synthase kinase-3beta (phospho-Tyr(216)-GSK-3beta, GSK-3beta-P(Y216)). The activity of GSK-3beta is believed to cause tau protein hyper-phosphorylation, increased amyloid-beta production and local plaque-associated microglial-mediated inflammatory responses. All of them are symptomatic for AD. In our studies, the highly significant Y216 phosphorylation and over-expression of total GSK-3beta were observed in AbetaPPsw-transfected PC12 cells. In addition, the immuocytochemical analysis showed co-localization of GSK-3beta-P(Y216) and amyloid-beta deposits. Thus, our data support a functional role of GSK-3beta in AbetaPP processing, further implicating this kinase in the amyloid-beta-dependent pathogenesis.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Glycogen Synthase Kinase 3/metabolism , Mutation , Tyrosine/metabolism , Amyloid beta-Peptides/genetics , Animals , Autophagy/genetics , Electrokymography , Endosomes/metabolism , Endosomes/ultrastructure , Glycogen Synthase Kinase 3 beta , Humans , Lysosomes/metabolism , Lysosomes/ultrastructure , Microscopy, Immunoelectron/methods , PC12 Cells/metabolism , PC12 Cells/ultrastructure , Phosphorylation/genetics , Rats , Subcellular Fractions/metabolism , Transfection/methods , Tyrosine/genetics
18.
Neurotoxicology ; 29(6): 998-1007, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18706927

ABSTRACT

It has been postulated that dihydroxyphenylacetic acid (DOPAC), a major dopamine metabolite, and nitric oxide (NO) induce mitochondrial dysfunction in a synergistic manner. We examined the combined effects of NO and DOPAC on PC-12 cells in terms of cell viability, nuclear morphology, mitochondrial parameters and cell death mechanisms. The apoptotic cell death induced by the NO-donor, S-nitroso-N-acetylpenicillamine (SNAP), was differently modulated by DOPAC as a function of DOPAC/cell ratios. Whereas below 200nmol/10(6) cells, DOPAC inhibited a typical apoptotic pathway induced by exposure the cells to the NO donor, above 200nmol DOPAC/10(6) cells, the cell death was not only enhanced but encompassed a distinct mechanism. Loading the cells with dopamine mimicked the effects of DOPAC. Specifically, the combination of DOPAC and NO induced an early mitochondrial membrane potential dissipation and ATP depletion followed by loss of cellular membrane integrity. Mitochondrial dysfunction was accompanied by the release of cytochrome c in both cases, NO individually and in combination with DOPAC, but caspase-3 and caspase-9 activation were only observed in the absence of DOPAC. DOPAC alone was ineffective. Thus, our results suggest a role for DOPAC as a modulator of cell fate and point to a pathway of cell death involving DOPAC and NO, via mechanisms that include mitochondrial dysfunction but do not involve the activation of the typical apoptotic caspase cascade. The significance of these results is discussed in connection with the mechanisms of cell death underlying Parkinson's disease.


Subject(s)
3,4-Dihydroxyphenylacetic Acid/pharmacology , Free Radical Scavengers/toxicity , Mitochondria/drug effects , Nitric Oxide/toxicity , Analysis of Variance , Animals , Apoptosis Inducing Factor/metabolism , Caspases/metabolism , Cell Death/drug effects , Cell Size/drug effects , Cytochromes c/metabolism , Dose-Response Relationship, Drug , Drug Interactions , L-Lactate Dehydrogenase/metabolism , Membrane Potential, Mitochondrial/drug effects , PC12 Cells/drug effects , PC12 Cells/ultrastructure , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Rats
19.
Neurosci Lett ; 436(3): 340-4, 2008 May 16.
Article in English | MEDLINE | ID: mdl-18406529

ABSTRACT

Neurite outgrowth is crucial for neural circuit formation. Intracellular membrane trafficking is involved in the cell surface expansion that is necessary for neurite outgrowth. It is known that syntaxin 6 is predominantly located in the Golgi region in undifferentiated PC12 cells and that it regulates trans-Golgi network trafficking and the secretory pathway via its coiled-coil domains. However, whether it also regulates neurite outgrowth remains unknown. In this paper, we found that syntaxin 6 was located both in the Golgi apparatus and the distal tips of the neurites of nerve growth factor (NGF)-treated PC12 cells. We also showed that the overexpression of the first coiled-coil domain of syntaxin 6 inhibited NGF-dependent neurite outgrowth. However, the coiled-coil domain-disrupting mutant had little effect on neurite outgrowth. These results suggest that the first coiled-coil domain of syntaxin 6 plays a crucial role in NGF-dependent neurite outgrowth.


Subject(s)
Gene Expression Regulation/drug effects , Nerve Growth Factor/pharmacology , Neurites/drug effects , Neurites/physiology , Qa-SNARE Proteins/metabolism , Amino Acid Sequence , Analysis of Variance , Animals , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Mutation/physiology , PC12 Cells/ultrastructure , Protein Structure, Tertiary/physiology , Qa-SNARE Proteins/genetics , Rats , Transfection
20.
Brain Res ; 1151: 46-54, 2007 Jun 02.
Article in English | MEDLINE | ID: mdl-17408597

ABSTRACT

Membrane composition serves to identify intracellular compartments, signal cell death, as well as to alter a cell's electrical and physical properties. Here we use amperometry to show that supplementation with the phospholipids phosphatidylcholine (PC), phosphatidylethanolamine (PE), sphingomyelin (SM), and phosphatidylserine (PS) can alter several aspects of exocytosis. Changes in the amperometric peak shape derived from individual exocytosing vesicles reveal that PC slows expulsion of neurotransmitter while PE accelerates expulsion of neurotransmitter. Amperometry data reveal a reduced amount of catecholamine released per event from PC-treated cells while electron micrographs indicate the vesicles in these cells are 50% larger than controls, thus providing evidence of pharmacological changes in vesicle concentration. Addition of SM appears to affect the rate of fusion pore expansion, indicated by slower peak rise times, but does not affect decay times or quantal size. Addition of PS results in a 1.7-fold increase in the number of events elicited by high-K(+) depolarization. Electron micrographs of PS-treated cells suggest that increased vesicle recruitment underlies enhanced secretion. We did not observe any effect of phosphatidylinositol (PI) treatment. Together these data suggest that differences in membrane composition affect exocytosis and might be involved in mechanisms of cell function controlling the dynamics of communication via exocytosis.


Subject(s)
Exocytosis/drug effects , Phospholipids/pharmacology , Analysis of Variance , Animals , Electrochemistry/methods , Microscopy, Electron, Transmission/methods , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , PC12 Cells/drug effects , PC12 Cells/ultrastructure , Rats
SELECTION OF CITATIONS
SEARCH DETAIL