Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
Add more filters

Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
J Cell Biol ; 139(4): 907-16, 1997 Nov 17.
Article in English | MEDLINE | ID: mdl-9362509

ABSTRACT

Previous studies have indicated that neuro-endocrine cells store monoamines and acetylcholine (ACh) in different secretory vesicles, suggesting that the transport proteins responsible for packaging these neurotransmitters sort to distinct vesicular compartments. Molecular cloning has recently demonstrated that the vesicular transporters for monoamines and ACh show strong sequence similarity, and studies of the vesicular monoamine transporters (VMATs) indicate preferential localization to large dense core vesicles (LDCVs) rather than synaptic-like microvesicles (SLMVs) in rat pheochromocytoma PC12 cells. We now report the localization of the closely related vesicular ACh transporter (VAChT). In PC12 cells, VAChT differs from the VMATs by immunofluorescence and fractionates almost exclusively to SLMVs and endosomes by equilibrium sedimentation. Immunoisolation further demonstrates colocalization with synaptophysin on SLMVs as well as other compartments. However, small amounts of VAChT also occur on LDCVs. Thus, VAChT differs in localization from the VMATs, which sort predominantly to LDCVs. In addition, we demonstrate ACh transport activity in stable PC12 transformants overexpressing VAChT. Since previous work has suggested that VAChT expression confers little if any transport activity in non-neural cells, we also determined its localization in transfected CHO fibroblasts. In CHO cells, VAChT localizes to the same endosomal compartment as the VMATs by immunofluorescence, density gradient fractionation, and immunoisolation with an antibody to the transferrin receptor. We have also detected ACh transport activity in the transfected CHO cells, indicating that localization to SLMVs is not required for function. In summary, VAChT differs in localization from the VMATs in PC12 cells but not CHO cells.


Subject(s)
Acetylcholine/metabolism , CHO Cells/ultrastructure , Carrier Proteins/metabolism , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Neuropeptides , PC12 Cells/ultrastructure , Synaptic Vesicles/metabolism , Vesicular Transport Proteins , Animals , Biological Transport , Cell Compartmentation , Cell Membrane/metabolism , Cricetinae , Endosomes/metabolism , Fluorescent Antibody Technique, Indirect , Intracellular Membranes/metabolism , Peptides/immunology , Rats , Receptors, Transferrin/metabolism , Synaptophysin/metabolism , Vesicular Acetylcholine Transport Proteins , Vesicular Biogenic Amine Transport Proteins
10.
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
11.
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
12.
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
13.
Mol Cell Biol ; 20(19): 7378-87, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10982854

ABSTRACT

The Rho family of small GTPases has been implicated in cytoskeletal reorganization and subsequent morphological changes in various cell types. Among them, Rac and Cdc42 have been shown to be involved in neurite outgrowth in neuronal cells. In this study, we examined the role of RhoG, another member of Rho family GTPases, in nerve growth factor (NGF)-induced neurite outgrowth in PC12 cells. Expression of wild-type RhoG in PC12 cells induced neurite outgrowth in the absence of NGF, and the morphology of wild-type RhoG-expressing cells was similar to that of NGF-differentiated cells. Constitutively active RhoG-transfected cells extended short neurites but developed large lamellipodial or filopodial structures at the tips of neurites. RhoG-induced neurite outgrowth was inhibited by coexpression with dominant-negative Rac1 or Cdc42. In addition, expression of constitutively active RhoG elevated endogenous Rac1 and Cdc42 activities. We also found that the NGF-induced neurite outgrowth was enhanced by expression of wild-type RhoG whereas expression of dominant-negative RhoG suppressed the neurite outgrowth. Furthermore, constitutively active Ras-induced neurite outgrowth was also suppressed by dominant-negative RhoG. Taken together, these results suggest that RhoG is a key regulator in NGF-induced neurite outgrowth, acting downstream of Ras and upstream of Rac1 and Cdc42 in PC12 cells.


Subject(s)
GTP Phosphohydrolases/physiology , Neoplasm Proteins/physiology , Nerve Tissue Proteins/physiology , Neurites/metabolism , PC12 Cells/metabolism , Animals , Cell Differentiation/drug effects , GTP Phosphohydrolases/biosynthesis , GTP Phosphohydrolases/genetics , Genes, Dominant , Mutagenesis, Site-Directed , Nerve Growth Factors/pharmacology , PC12 Cells/drug effects , PC12 Cells/ultrastructure , Rats , Recombinant Fusion Proteins/physiology , Signal Transduction , Transfection , cdc42 GTP-Binding Protein/physiology , rac1 GTP-Binding Protein/physiology
14.
Sci STKE ; 2005(313): pe55, 2005 Dec 06.
Article in English | MEDLINE | ID: mdl-16333019

ABSTRACT

Long membrane tethers between cells, known as membrane nantotubes or tunneling nanotubules, create supracellular structures that allow multiple cell bodies to act in a synchronized manner. Calcium fluxes, vesicles, and cell-surface components can all traffic between cells connected by nanotubes. Thus, complex and specific messages can be transmitted between multiple cells, and the strength of signal will suffer relatively little with the distance traveled, as compared to the use of soluble factors to transmit messages. Connecting multiple antigen-presenting cells, for example, can help amplify and coordinate immune responses that are distal to an antigenic site. Conversely, because the ability of a pathogen to spread between cells is a key determinant of its capacity to multiply, pathogens may exploit nanotubes for their own transmission.


Subject(s)
Cell Communication/physiology , Cell Surface Extensions/physiology , Nanotubes , Animals , Biological Transport , Calcium/metabolism , Cell Surface Extensions/ultrastructure , Eukaryotic Cells/physiology , Eukaryotic Cells/ultrastructure , Humans , Inositol 1,4,5-Trisphosphate/metabolism , PC12 Cells/ultrastructure , Prokaryotic Cells/physiology , Prokaryotic Cells/ultrastructure , Rats , Virus Physiological Phenomena
15.
J Neurosci Methods ; 156(1-2): 55-63, 2006 Sep 30.
Article in English | MEDLINE | ID: mdl-16554096

ABSTRACT

Cellular migration is central to a wide range of biological and pathological processes in vivo. In vitro cell migration assays can be used to obtain invaluable information relating to the mechanism of cell movement, but current available methods can be limiting. Here we describe a novel motility assay that allows the simultaneous investigation of both quantitative and qualitative aspects of a population of motile cells as they move across a variety of substrates. By plating cells in a confluent monolayer on a coverslip, the monolayer can then be inverted to migrate over a larger substrate-coated coverslip, which can subsequently be reliably quantified, and subjected to immunocytochemistry and confocal imaging. This assay can be used to assess multiple aspects of motility, including distance, quantity, morphology, polarization and component colocalization. To demonstrate the utility of this assay, it was applied to the study of a stimulator of PC12 cell migration, nerve growth factor (NGF), and how this migration is influenced by the extracellular substrate, laminin. Furthermore, since mutations to the NGF receptor, TrkA, have been noted to alter the behaviour of PC12 cells in response to NGF, a PC12 subline that expresses a mutated TrkA receptor was utilized to illustrate that a Y785F mutation in the cytoplasmic tail of TrkA results in increased migration in response to the stimulus compared to the control PC12s.


Subject(s)
Cell Movement/physiology , PC12 Cells/physiology , Animals , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Movement/genetics , Immunohistochemistry , Laminin/pharmacology , Microscopy, Confocal , Mutation/physiology , Nerve Growth Factors/pharmacology , PC12 Cells/ultrastructure , Rats , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/genetics
16.
Brain Res Bull ; 69(6): 687-94, 2006 May 31.
Article in English | MEDLINE | ID: mdl-16716839

ABSTRACT

Defects in mitochondrial function have been shown to participate in the induction of neuronal cell injury. The effect of econazole against the cytotoxicity of 1-methyl-4-phenylpyridinium (MPP(+)) in differentiated PC12 cells was assessed in relation to the mitochondrial membrane permeability changes. Treatment of PC12 cells with MPP(+) resulted in the nuclear damage, decrease in the mitochondrial transmembrane potential, cytosolic accumulation of cytochrome c, activation of caspase-3, increase in the formation of reactive oxygen species (ROS) and depletion of GSH. Econazole (0.25-2.5 microM) inhibited the cytotoxicity of MPP(+) or rotenone. The addition of econazole (0.5 microM) significantly attenuated the MPP(+)-induced mitochondrial damage, elevation of intracellular Ca(2+) level and cell death. However, because of the cytotoxicity, econazole at 5 microM did not attenuate the toxicity of MPP(+). The results show that econazole at the low concentrations may reduce the MPP(+)-induced viability loss in PC12 cells by suppressing the mitochondrial permeability transition, leading to activation of caspase-3 and the elevation of intracellular Ca(2+) levels, which are associated with the increased formation of ROS and depletion of GSH.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Econazole/pharmacology , Herbicides/toxicity , Mitochondrial Membranes/drug effects , Animals , Apoptosis/drug effects , Calcium/metabolism , Caspase 3 , Caspases/metabolism , Cell Survival/drug effects , Cytochromes c/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Flow Cytometry/methods , Membrane Potentials/drug effects , PC12 Cells/ultrastructure , Permeability/drug effects , Rats , Reactive Oxygen Species/metabolism
18.
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
19.
Oncogene ; 20(50): 7307-17, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11704860

ABSTRACT

Guanine nucleotide exchange factors from the Dbl family are proto-oncogenic proteins that activate small GTPases of the Rho family. Here we report the characterization of GEF720, a novel Dbl-like protein related to p115Rho-GEF. GEF720 activated RhoA both in our recently developed Yeast Exchange Assay and in biochemical in vitro exchange assays. GEF720 induced RhoA dependent assembly of actin stress fibers in REF52 fibroblastic cells. In NIH3T3 cells this Dbl-like protein elicited formation of transformation foci with a morphology similar to RhoA-V14 induced foci. In the PC12 neuron-like cell line, expression of GEF720, whose mRNA is brain specific, inhibited NGF-induced neurite outgrowth. Finally, GEF720 gene is located on human chromosome 1 on band 1p36, between Tumor Protein 73 and Tumor Necrosis Factor Receptor 12, two genes rearranged in many neuroblastoma cell lines. Together, these results show that this new Dbl related protein, GEF720, is an exchange factor that can directly activate RhoA in vivo and is potentially involved in the control of neuronal cell differentiation. GEF720 is also a new candidate gene involved in the progression of neuroblastoma and developmental abnormalities associated with rearrangements in the 1p36 chromosomal region.


Subject(s)
Brain Chemistry , Chromosomes, Human, Pair 1/genetics , Guanine Nucleotide Exchange Factors/genetics , Nerve Tissue Proteins/genetics , rhoA GTP-Binding Protein/metabolism , 3T3 Cells , Actins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Brain/enzymology , Cell Differentiation , Cell Line, Transformed , Cell Transformation, Neoplastic/genetics , Chromosome Mapping , Disease Progression , Enzyme Activation , Exons/genetics , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Genes , Guanine Nucleotide Exchange Factors/metabolism , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Humans , Mice , Molecular Sequence Data , Multigene Family , Neurites/ultrastructure , Neuroblastoma/genetics , Neuroblastoma/pathology , PC12 Cells/ultrastructure , Protein Binding , Protein Structure, Tertiary , Rats , Recombinant Fusion Proteins/physiology , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Stress Fibers/metabolism , Transfection , Tumor Cells, Cultured
20.
Endocrinology ; 146(11): 4727-36, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16109784

ABSTRACT

GLUT8 is a high-affinity glucose transporter present mostly in testes and a subset of brain neurons. At the cellular level, it is found in a poorly defined intracellular compartment in which it is retained by an N-terminal dileucine motif. Here we assessed GLUT8 colocalization with markers for different cellular compartments and searched for signals, which could trigger its cell surface expression. We showed that when expressed in PC12 cells, GLUT8 was located in a perinuclear compartment in which it showed partial colocalization with markers for the endoplasmic reticulum but not with markers for the trans-Golgi network, early endosomes, lysosomes, and synaptic-like vesicles. To evaluate its presence at the plasma membrane, we generated a recombinant adenovirus for the expression of GLUT8 containing an extracellular myc epitope. Cell surface expression was evaluated by immunofluorescence microscopy of transduced PC12 cells or primary hippocampal neurons exposed to different stimuli. Those included substances inducing depolarization, activation of protein kinase A and C, activation or inhibition of tyrosine kinase-linked signaling pathways, glucose deprivation, AMP-activated protein kinase stimulation, and osmotic shock. None of these stimuli-induced GLUT8 cell surface translocation. Furthermore, when GLUT8myc was cotransduced with a dominant-negative form of dynamin or GLUT8myc-expressing PC-12 cells or neurons were incubated with an anti-myc antibody, no evidence for constitutive recycling of the transporter through the cell surface could be obtained. Thus, in cells normally expressing it, GLUT8 was associated with a specific intracellular compartment in which it may play an as-yet-uncharacterized role.


Subject(s)
Cell Membrane/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Hippocampus/metabolism , Neurons/metabolism , PC12 Cells/metabolism , Subcellular Fractions/metabolism , AMP-Activated Protein Kinases , Animals , Biological Transport , Cell Membrane/physiology , Electrophysiology , Enzyme Activation , Glucose/deficiency , Glucose Transport Proteins, Facilitative/genetics , Hippocampus/cytology , Humans , Intracellular Membranes/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Multienzyme Complexes/metabolism , Neurons/ultrastructure , Osmotic Pressure , PC12 Cells/physiology , PC12 Cells/ultrastructure , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/genetics , Rats , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL