Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 274
Filter
1.
Int J Mol Sci ; 21(7)2020 Apr 04.
Article in English | MEDLINE | ID: mdl-32260390

ABSTRACT

The rhizome of Anemarrhena asphodeloides Bunge, used in Traditional Chinese Medicine as a brain function-improving herb, is a promising source of neuroprotective substances. The aim of this study was to evaluate the protective action of xanthones from A. asphodeloides rhizomes on the PC12 cell line exposed to the neurotoxic agent-3-nitropropionic acid (3-NP). The xanthone-enriched fraction of the ethanolic extract of A. asphodeloides (abbreviated from now on as XF, for the Xanthone Fraction), rich in polyphenolic xanthone glycosides, in concentrations from 5 to 100 µg/mL, and 3-NP in concentrations from 2.5 to 15 mM, were examined. After 8, 16, 24, 48, and 72 h of exposure of cells to various combinations of 3-NP and XF, the MTT viability assay was performed and morphological changes were estimated by confocal fluorescence microscopy. The obtained results showed a significant increase in the number of cells surviving after treatment with XF with exposure to neurotoxic 3-NP and decreased morphological changes in PC12 cells in a dose and time dependent manner. The most effective protective action was observed when PC12 cells were pre-incubated with the XF. This effect may contribute to the traditional indications of this herb for neurological and cognitive complaints. However, a significant cytotoxicity observed at higher XF concentrations (over 10 µg/mL) and longer incubation time (48 h) requires caution in future research and thorough investigation into potential adverse effects.


Subject(s)
Anemarrhena/chemistry , Neuroprotective Agents/pharmacology , Nitro Compounds/adverse effects , PC12 Cells/cytology , Propionates/adverse effects , Xanthones/pharmacology , Animals , Apoptosis/drug effects , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Molecular Structure , Neuroprotective Agents/chemistry , PC12 Cells/drug effects , Rats , Rhizome/chemistry , Time Factors , Xanthones/chemistry
2.
Molecules ; 23(5)2018 04 26.
Article in English | MEDLINE | ID: mdl-29701695

ABSTRACT

The leaves of Morus alba L. are an important herbal medicine in Asia. The systematic isolation of the metabolites of the leaves of Morus alba L. was achieved using a combination of liquid chromatography techniques. The structures were elucidated by spectroscopic data analysis and the absolute configuration was determined based on electronic circular dichroism (ECD) spectroscopic data and hydrolysis experiments. Their biological activity was evaluated using different biological assays, such as the assessment of their capacity to inhibit the aldose reductase enzyme; the determination of their cytotoxic activity and the evaluation of their neuroprotective effects against the deprivation of serum or against the presence of nicouline. Chemical investigation of the leaves of Morus alba L. resulted in four new structures 1⁻4 and a known molecule 5. Compounds 2 and 5 inhibited aldose reductase with IC50 values of 4.33 µM and 6.0 µM compared with the potent AR inhibitor epalrestat (IC50 1.88 × 10−3 µM). Pretreatment with compound 3 decreased PC12 cell apoptosis subsequent serum deprivation condition and pretreatment with compound 5 decreased nicouline-induced PC12 cell apoptosis as compared with control cells (p < 0.001).


Subject(s)
Enzyme Inhibitors/chemistry , Morus/chemistry , Neuroprotective Agents/chemistry , Plant Extracts/chemistry , Plant Leaves/chemistry , Aldehyde Reductase/antagonists & inhibitors , Animals , Apoptosis/drug effects , Chromatography, Liquid , Circular Dichroism , Enzyme Inhibitors/pharmacology , Molecular Structure , Neuroprotective Agents/pharmacology , PC12 Cells/cytology , PC12 Cells/drug effects , Plant Extracts/pharmacology , Rats
3.
Sci Rep ; 8(1): 574, 2018 01 12.
Article in English | MEDLINE | ID: mdl-29330409

ABSTRACT

Neurodegenerative diseases (NDD) are typically associated with neuron loss in nervous system areas. Interventions with related death mechanisms may ameliorate NDD progression. Oxidative stress plays an important role in NDD cell death routines. However, tert-butylhydroperoxide (t-BHP), a widely used oxidative stress stimulus, induces neural cell death through a mechanism that remains elusive. In our study, the ferroptosis marker events occurred after co-treatment with 100 µM t-BHP for 1 h, all of which were reversed in the presence of the ferroptosis inhibitor ferrostatin-1 (Fer-1) and the iron chelator deferoxamine, implying the occurrence of ferroptosis. Moreover, mitochondrial dysfunction accompanied by a decreased in membrane potential and ATP production, increased mitochondrial ROS generation. Furthermore, this mitochondrial dysfunction could be reversed by Fer-1. In addition, JNK1/2 and ERK1/2 were activated upstream of the ferroptosis and mitochondrial dysfunction. In summary, these data suggest that ferroptosis, coupled with mitochondrial dysfunction, was involved in t-BHP-induced PC12 death. JNK1/2 and ERK1/2 played important roles in t-BHP-induced cell death. Overall, this study might provide clues to the oxidative stress-based strategies for cell protection in NDD.


Subject(s)
Adenosine Triphosphate/metabolism , Mitochondria/drug effects , PC12 Cells/drug effects , Reactive Oxygen Species/metabolism , tert-Butylhydroperoxide/pharmacology , Animals , Cell Death , Cell Survival/drug effects , Cyclohexylamines/pharmacology , Deferoxamine/pharmacology , Gene Expression Regulation/drug effects , MAP Kinase Signaling System/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Oxidative Stress , PC12 Cells/cytology , PC12 Cells/metabolism , Phenylenediamines/pharmacology , Rats
4.
Neuroreport ; 28(9): 533-539, 2017 Jun 14.
Article in English | MEDLINE | ID: mdl-28489665

ABSTRACT

Neuronal regeneration and axonal regrowth mechanisms in the injured mammalian central nervous system are largely unknown. As part of a major pathway for inhibiting axonal regeneration, activated neuronal glycosylphosphatidylinositol-anchored Nogo receptor (NgR) interacts with LINGO-1 and p75NTR to form a complex at the cell surface. However, it was found in our previous report that upregulation of NgR stimulated by injury plays a key role in neuronal regeneration in the neonatal cortex freeze-lesion model, but its downstream signalling remains elusive. In the present study, the novel regulatory role of NgR in a serine-threonine kinase WNK1 was identified. NgR's transcriptional regulation of WNK1 was identified by RT-qPCR and semiquantitative western blot after the overexpression or knockdown of NgR, and the regulation is specific to WNK1, which is not the same for its family members, WNK2, WNK3 and WNK4. Furthermore, NgR inhibition by NEP fails to affect WNK1, which indicates that WNK1 functions outside of the Nogo-A/NgR pathway. By performing a proliferation, migration and axonal extension assay, we also identified that overexpressed NgR critically regulated these processes and impairment by overexpressing NgR was rescued with coexpression of WNK1, indicating the partial role of WNK1 in NgR-mediated morphological regulation. Our study identifies a separation of functions for the NgR-regulated WNK1 in mediating proliferation, migration and axonal extension in PC12 cells as well as a specific regulatory role between NgR and WNK1 that is important for recovery from central nervous system injury.


Subject(s)
Axons/physiology , Cell Movement/physiology , Cell Proliferation/physiology , Gene Expression Regulation/physiology , Nogo Receptors/metabolism , WNK Lysine-Deficient Protein Kinase 1/metabolism , Animals , Axons/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Nerve Growth Factor/pharmacology , Nogo Receptors/genetics , PC12 Cells/cytology , RNA, Messenger/metabolism , Rats , Sincalide/metabolism , Transfection , WNK Lysine-Deficient Protein Kinase 1/genetics
5.
J Vis Exp ; (120)2017 02 26.
Article in English | MEDLINE | ID: mdl-28287599

ABSTRACT

Substrate and cell patterning techniques are widely used in cell biology to study cell-to-cell and cell-to-substrate interactions. Conventional patterning techniques work well only with simple shapes, small areas and selected bio-materials. This article describes a method to distribute cell suspensions as well as substrate solutions into complex, long, closed (dead-end) polydimethylsiloxane (PDMS) microchannels using negative pressure. This method enables researchers to pattern multiple substrates including fibronectin, collagen, antibodies (Sal-1), poly-D-lysine (PDL), and laminin. Patterning of substrates allows one to indirectly pattern a variety of cells. We have tested C2C12 myoblasts, the PC12 neuronal cell line, embryonic rat cortical neurons, and amphibian retinal neurons. In addition, we demonstrate that this technique can directly pattern fibroblasts in microfluidic channels via brief application of a low vacuum on cell suspensions. The low vacuum does not significantly decrease cell viability as shown by cell viability assays. Modifications are discussed for application of the method to different cell and substrate types. This technique allows researchers to pattern cells and proteins in specific patterns without the need for exotic materials or equipment and can be done in any laboratory with a vacuum.


Subject(s)
PC12 Cells/cytology , PC12 Cells/metabolism , Proteins/metabolism , Animals , Cell Adhesion , Cell Count , Cells, Cultured , Fibroblasts/cytology , Models, Animal , Rats
6.
J Biol Chem ; 291(44): 22970-22976, 2016 10 28.
Article in English | MEDLINE | ID: mdl-27624933

ABSTRACT

When treated with nerve growth factor, PC12 cells will differentiate over the course of several days. Here, we have followed changes during differentiation in the cellular levels of phosphoinositide-specific phospholipase Cß (PLCß) and its activator, Gαq, which together mediate Ca2+ release. We also followed changes in the level of the novel PLCß binding partner TRAX (translin-associated factor X), which promotes RNA-induced gene silencing. We find that the level of PLCß increases 4-fold within 24 h, whereas Gαq increases only 1.4-fold, and this increase occurs ∼24 h later than PLCß. Alternately, the level of TRAX remains constant over the 72 h tested. When PLCß1 or TRAX is down-regulated, differentiation does not occur. The impact of PLCß on differentiation appears independent of Gαq as down-regulating Gαq at constant PLCß does not affect differentiation. Förster resonance energy transfer studies after PLCß association with its partners indicate that PLCß induced soon after nerve growth factor treatment associates with TRAX rather than Gαq Functional measurements of Ca2+ signals to assess the activity of PLCß-Gαq complexes and measurements of the reversal of siRNA(GAPDH) to assess the activity of PLCß-TRAX complexes additionally suggest that the newly synthesized PLCß associates with TRAX to impact RNA-induced silencing. Taken together, our studies show that PLCß, through its ability to bind TRAX and reverse RNA silencing of specific genes, plays a key role in switching PC12 cells to their differentiated state.


Subject(s)
Cell Differentiation , PC12 Cells/enzymology , Phospholipase C beta/metabolism , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , PC12 Cells/cytology , Phospholipase C beta/genetics , Protein Binding , Rats
7.
PLoS One ; 10(6): e0130599, 2015.
Article in English | MEDLINE | ID: mdl-26087007

ABSTRACT

Luteolin and apigenin are dietary flavones and exhibit a broad spectrum of biological activities including antioxidant, anti-inflammatory, anti-cancer and neuroprotective effects. The lipid peroxidation product 4-hydroxy-2-nonenal (4-HNE) has been implicated as a causative agent in the development of neurodegenerative disorders. This study investigates the cytoprotective effects of luteolin and apigenin against 4-HNE-mediated cytotoxicity in neuronal-like catecholaminergic PC12 cells. Both flavones restored cell viability and repressed caspase-3 and PARP-1 activation in 4-HNE-treated cells. Luteolin also mitigated 4-HNE-mediated LC3 conversion and reactive oxygen species (ROS) production. Luteolin and apigenin up-regulated 4-HNE-mediated unfolded protein response (UPR), leading to an increase in endoplasmic reticulum chaperone GRP78 and decrease in the expression of UPR-targeted pro-apoptotic genes. They also induced the expression of Nrf2-targeted HO-1 and xCT in the absence of 4-HNE, but counteracted their expression in the presence of 4-HNE. Moreover, we found that JNK and p38 MAPK inhibitors significantly antagonized the increase in cell viability induced by luteolin and apigenin. Consistently, enhanced phosphorylation of JNK and p38 MAPK was observed in luteolin- and apigenin-treated cells. In conclusion, this result shows that luteolin and apigenin activate MAPK and Nrf2 signaling, which elicit adaptive cellular stress response pathways, restore 4-HNE-induced ER homeostasis and inhibit cytotoxicity. Luteolin exerts a stronger cytoprotective effect than apigenin possibly due to its higher MAPK, Nrf2 and UPR activation, and ROS scavenging activity.


Subject(s)
Antioxidants/pharmacology , Apigenin/pharmacology , Cell Death/drug effects , Luteolin/pharmacology , Neuroprotective Agents/pharmacology , PC12 Cells/drug effects , Aldehydes/metabolism , Animals , Caspase 3/metabolism , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation/drug effects , Lipid Peroxidation , MAP Kinase Signaling System/drug effects , NF-E2-Related Factor 2/metabolism , PC12 Cells/cytology , PC12 Cells/metabolism , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Unfolded Protein Response/drug effects
8.
Biomaterials ; 55: 33-43, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25934450

ABSTRACT

At high concentrations, manganese (Mn) promotes cellular neurodevelopment but causes toxicity. Here, we report that Mn ion at high concentrations can be delivered to pheochromocytoma 12 (PC12) cells using gold nanoparticles (AuNPs) to enhance cellular neurodevelopment without toxicity. Mn(2+) release from AuNPs was designed to be pH-responsive so that low pH condition of the cell endosomes can trigger in situ release of Mn(2+) from AuNPs after cellular uptake of Mn-incorporated AuNPs (MnAuNPs). Due to the differences in reduction potentials of Mn and Au, only Mn ionized and released while Au remained intact when MnAuNPs were uptaken by cells. Compared to PC12 cells treated with a high concentration of free Mn(2+), PC12 cells treated with an equal concentration of MnAuNPs resulted in significantly enhanced cellular neurodevelopment with decreased apoptosis and necrosis. Treatment with a high concentration of free Mn(2+) led to an abrupt consumption of a large amount of ATP for the intracellular transport of Mn(2+) through the ion channel of the cell membrane and to mitochondrial damage caused by the high intracellular concentration of Mn(2+), both of which resulted in cell necrosis and apoptosis. In contrast, MnAuNP-treated cells consumed much smaller amount of ATP for the intracellular transport of MnAuNPs by endocytosis and showed pH-triggered in situ release of Mn(2+) from the MnAuNPs in the endosomes of the cells, both of which prevented the cell death caused by ATP depletion and mitochondrial damage. To our knowledge, this is the first report on the use of AuNPs as a vehicle for pH-responsive, intracellular delivery of metal ion, which may open a new window for drug delivery and clinical therapy.


Subject(s)
Cell Differentiation , Drug Delivery Systems , Gold/chemistry , Manganese/chemistry , Metal Nanoparticles/chemistry , Neurons/cytology , Adenosine Triphosphate/chemistry , Animals , Apoptosis , Cell Membrane/metabolism , Endocytosis , Hydrogen-Ion Concentration , Ions , Lactic Acid/chemistry , Mitochondria/metabolism , Mitochondria/pathology , Necrosis , Neurons/drug effects , PC12 Cells/cytology , PC12 Cells/drug effects , Rats
9.
Rev Sci Instrum ; 86(3): 033705, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25832236

ABSTRACT

Atomic Force Microscopy (AFM) has a great potential as a tool to characterize mechanical and morphological properties of living cells; these properties have been shown to correlate with cells' fate and patho-physiological state in view of the development of novel early-diagnostic strategies. Although several reports have described experimental and technical approaches for the characterization of cellular elasticity by means of AFM, a robust and commonly accepted methodology is still lacking. Here, we show that micrometric spherical probes (also known as colloidal probes) are well suited for performing a combined topographic and mechanical analysis of living cells, with spatial resolution suitable for a complete and accurate mapping of cell morphological and elastic properties, and superior reliability and accuracy in the mechanical measurements with respect to conventional and widely used sharp AFM tips. We address a number of issues concerning the nanomechanical analysis, including the applicability of contact mechanical models and the impact of a constrained contact geometry on the measured Young's modulus (the finite-thickness effect). We have tested our protocol by imaging living PC12 and MDA-MB-231 cells, in order to demonstrate the importance of the correction of the finite-thickness effect and the change in Young's modulus induced by the action of a cytoskeleton-targeting drug.


Subject(s)
Microscopy, Atomic Force/instrumentation , Microscopy, Atomic Force/methods , Single-Cell Analysis/instrumentation , Single-Cell Analysis/methods , Animals , Biomechanical Phenomena , Cell Adhesion , Cell Line, Tumor , Colloids , Elastic Modulus , Humans , PC12 Cells/cytology , PC12 Cells/physiology , Rats
10.
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
11.
CNS Neurol Disord Drug Targets ; 13(8): 1458-64, 2014.
Article in English | MEDLINE | ID: mdl-25345505

ABSTRACT

There is heightened interest in the field of stroke recovery as there is need for agents that would prevent the debilitating effects of the disorder, thereby tremendously reducing the societal and economic costs associated with it. In this study, the isolation of two flavonoids--quercetin-3-O-galactoside (1) and quercetin-3-O-arabinoside (2)--from Rumex aquaticus (western dock) and their neuroprotective effects were reported in the oxygen-glucose deprivation (OGD) model of in vitro ischemia using rat pheochromocytoma (PC12) cell line. Bioassay-guided fractionation of the ethyl-acetate extract of Rumex aquaticus L. afforded the isolation of compounds 1 and 2. The structures of compounds were established on the basis of spectroscopic analyses (UV, mass spectrometry (MS) and nuclear magnetic resonance spectroscopy (NMR). Both compounds were isolated for the first time from this species. In the course of the pharmacological experiments it was detected that these flavonoids at 10 µM concentration significantly improved cell survival in the oxygen-glucose deprivation model of ischemia. Moreover, they also increased neurite outgrowth in differentiated PC12 cells subjected to ischemic insult. Investigations on the cellular mechanism for the observed effect revealed that compound 1 (10 µM) enhances the expression of synaptophysin - a marker of synapses, and an indicator of synaptic plasticity. Rapid restoration of neurological function following injury is paramount to the prevention of debilitating consequences of ischemic stroke. This combination of neuroprotection and neuritogenic potential could be particularly useful in the recovery phase of stroke.


Subject(s)
Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Neurites/drug effects , Neuroprostanes/pharmacology , Rumex/chemistry , Synaptophysin/metabolism , Actins/metabolism , Animals , Cell Death/drug effects , Dose-Response Relationship, Drug , Glucose/deficiency , Hypoxia , PC12 Cells/cytology , PC12 Cells/drug effects , Plant Extracts/chemistry , Rats
12.
Braz J Med Biol Res ; 47(9): 773-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25075574

ABSTRACT

The present study focuses on the neuroprotective effect of glycyrrhizic acid (GA, a major compound separated from Glycyrrhiza Radix, which is a crude Chinese traditional drug) against glutamate-induced cytotoxicity in differentiated PC12 (DPC12) cells. The results showed that GA treatment improved cell viability and ameliorated abnormal glutamate-induced alterations in mitochondria in DPC12 cells. GA reversed glutamate-suppressed B-cell lymphoma 2 levels, inhibited glutamate-enhanced expressions of Bax and cleaved caspase 3, and reduced cytochrome C (Cyto C) release. Exposure to glutamate strongly inhibited phosphorylation of AKT (protein kinase B) and extracellular signal-regulated kinases (ERKs); however, GA pretreatment enhanced activation of ERKs but not AKT. The presence of PD98059 (a mitogen-activated protein/extracellular signal-regulated kinase kinase [MEK] inhibitor) but not LY294002 (a phosphoinositide 3-kinase [PI3K] inhibitor) diminished the potency of GA for improving viability of glutamate-exposed DPC12 cells. These results indicated that ERKs and mitochondria-related pathways are essential for the neuroprotective effect of GA against glutamate-induced toxicity in DPC12 cells. The present study provides experimental evidence supporting GA as a potential therapeutic agent for use in the treatment of neurodegenerative diseases.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Glutamic Acid/toxicity , Glycyrrhizic Acid/therapeutic use , Neuroprotective Agents/therapeutic use , PC12 Cells/drug effects , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Caspase 3/isolation & purification , Cell Differentiation/drug effects , Cell Survival/drug effects , Chromones/pharmacology , Cytochromes c/drug effects , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , MAP Kinase Signaling System/drug effects , Mitochondria/drug effects , Morpholines/pharmacology , PC12 Cells/classification , PC12 Cells/cytology , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-bcl-2/isolation & purification , Rats , bcl-2-Associated X Protein/isolation & purification
13.
J Alzheimers Dis ; 40(4): 929-39, 2014.
Article in English | MEDLINE | ID: mdl-24531159

ABSTRACT

Autophagy is a lysosomal degradative process essential for neuronal homeostasis, whereas autophagic failure has been linked to accumulating neurodegenerative disorders. However, the precise role of autophagy in axonal and dendritic degeneration in Alzheimer's disease (AD) remains unclear. In this study, we aim to investigate the precise effect of autophagy in amyloid-ß peptide (Aß)25-35-mediated neurite degeneration. Aß35-25, the non-neurotoxic reverse sequence analogue of Aß25-35, was used as a negative control. Our results showed that Aß25-35 dose-dependently suppressed PC12 proliferation and induced autophagy induction in neurites (axons and dendrites). A high proportion of autophagic structures in PC12 neurites were autolysosomes after 24 h of Aß25-35 treatment. Autophagy inhibition by 3-methyladenine (3MA), LY294002, and chloroquine (CQ) could not relieve the Aß25-35-induced neurite degeneration, while administration of autophagy stimulator rapamycin or AR-12 efficiently suppressed neurite degeneration. Autophagosomes colocalized with fragmented mitochondria after Aß25-35 treatment. Similar results were obtained using in vitro cultured superior cervical ganglion neurons. These findings demonstrate that autophagy stimulation may prevent neuritic degeneration following Aß25-35 treatment. Upregulation of autophagic activity may provide a valuable approach for the treatment of axonal and dendritic dystrophy in AD patients.


Subject(s)
Amyloid beta-Peptides/toxicity , Autophagy/physiology , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Neurites/pathology , Peptide Fragments/toxicity , Animals , Animals, Newborn , Autophagy/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/pathology , Nerve Degeneration/prevention & control , Neurites/drug effects , Neurites/ultrastructure , Neurons/pathology , Organ Culture Techniques , PC12 Cells/cytology , Rats , Superior Cervical Ganglion/cytology
14.
PLoS One ; 8(3): e57037, 2013.
Article in English | MEDLINE | ID: mdl-23469182

ABSTRACT

A wide range of growth factors encode information into specific temporal patterns of MAP kinase (MAPK) and CREB phosphorylation, which are further decoded by expression of immediate early gene products (IEGs) to exert biological functions. However, the IEG decoding system remain unknown. We built a data-driven based on time courses of MAPK and CREB phosphorylation and IEG expression in response to various growth factors to identify how signal is processed. We found that IEG expression uses common decoding systems regardless of growth factors and expression of each IEG differs in upstream dependency, switch-like response, and linear temporal filters. Pulsatile ERK phosphorylation was selectively decoded by expression of EGR1 rather than c-FOS. Conjunctive NGF and PACAP stimulation was selectively decoded by synergistic JUNB expression through switch-like response to c-FOS. Thus, specific temporal patterns and combinations of MAPKs and CREB phosphorylation can be decoded by selective IEG expression via distinct temporal filters and switch-like responses. The data-driven modeling is versatile for analysis of signal processing and does not require detailed prior knowledge of pathways.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Genes, Immediate-Early , Mitogen-Activated Protein Kinases/genetics , Models, Biological , PC12 Cells/metabolism , Animals , Anisomycin/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation/drug effects , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factor/pharmacology , PC12 Cells/cytology , PC12 Cells/drug effects , Phosphorylation/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rats , Signal Transduction/drug effects , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Biosens Bioelectron ; 44: 122-6, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23416312

ABSTRACT

We report on a sensitive, simple, label-free cell-based electrochemical sensor to monitor the toxic effect of acrylamide on the Pheochromocytoma cells. The surface of the electrode was modified with gold nanoparticles and electrochemically reduced graphene oxide. Cyclic voltammetry, impedance spectroscopy and differential pulse voltammetry were applied to characterize the modified electrode. Reduced graphene oxide was proved to increase electron-transfer rate between the cell and the surface of electrode, while gold nanoparticle retain cell bioactivity. The sensor exhibited good correlation to the logarithmic value of cell numbers ranging from 1.6×10(4) to 1.6×10(7) cells mL(-1), with R.S.D value of 1.68%. The value of differential pulse voltammetry (cell adsorption concentration of 1.6×10(7) cells mL(-1)) decreased with the concentration of acrylamide in range of 0.1-5 mM with the detection limit as 0.04 mM. Scanning electron microscope-based morphological and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide analysis confirmed the results of the electrochemical study. This sensor was proved to be a useful tool for probing the toxicity of cells, and assisted in the development of a labeling-free, simple, rapid and immediate detection method.


Subject(s)
Acrylamide/toxicity , Biosensing Techniques/methods , Cell Survival/drug effects , Electrochemical Techniques/methods , Nanocomposites/chemistry , PC12 Cells/drug effects , Animals , Cell Count , Cells, Immobilized/cytology , Cells, Immobilized/drug effects , Gold/chemistry , Graphite/chemistry , PC12 Cells/cytology , Rats
16.
Biofactors ; 38(3): 219-25, 2012.
Article in English | MEDLINE | ID: mdl-22488872

ABSTRACT

Huntington's disease (HD) is characterized by the dysfunction of mitochondrial energy metabolism, which is associated with the functional impairment of succinate dehydrogenase (mitochondrial complex II), and pyruvate dehydrogenase (PDH). Treatment with 3-nitropropionic acid (3-NP), a potent irreversible inhibitor of succinate dehydrogenase, replicates most of the pathophysiological features of HD. In the present study, we investigated the effect of (-)schisandrin B [(-)Sch B, a potent enantiomer of schisandrin B] on 3-NP-induced cell injury in rat differentiated neuronal PC12 cells. The 3-NP caused cell necrosis, as assessed by lactate dehydrogenase (LDH) leakage, and mitochondrion-dependent cell apoptosis, as assessed by caspase-3 and caspase-9 activation, in differentiated PC12 cells. The cytotoxicity induced by 3-NP was associated with a depletion of cellular reduced glutathione (GSH) as well as the activation of redox-sensitive c-Jun N-terminal kinase (JNK) pathway and the inhibition of PDH. (-)Sch B pretreatment (5 and 15 µM) significantly reduced the extent of necrotic and apoptotic cell death in 3-NP-challenged cells. The cytoprotection afforded by (-)Sch B pretreatment was associated with the attenuation of 3-NP-induced GSH depletion as well as JNK activation and PDH inhibition. (-)Sch B pretreatment enhanced cellular glutathione redox status and ameliorated the 3-NP-induced cellular energy crisis, presumably by suppressing the activated JNK-mediated PDH inhibition, thereby protecting against necrotic and apoptotic cell death in differentiated PC12 cells.


Subject(s)
Lignans/pharmacology , Nitro Compounds/toxicity , Polycyclic Compounds/pharmacology , Propionates/toxicity , Animals , Apoptosis , Cell Death , Cyclooctanes/pharmacology , Cytoprotection , Glutathione/metabolism , Glutathione Reductase/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Ketone Oxidoreductases/antagonists & inhibitors , Ketone Oxidoreductases/metabolism , PC12 Cells/cytology , PC12 Cells/metabolism , Rats
17.
Neurotoxicol Teratol ; 34(2): 232-41, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22222554

ABSTRACT

Developmental organophosphate exposure reduces the numbers of neural cells, contributing to neurobehavioral deficits. We administered chlorpyrifos or diazinon to newborn rats on postnatal days 1-4, in doses straddling the threshold for barely-detectable cholinesterase inhibition, and evaluated gene expression in the cell cycle and apoptosis pathways on postnatal day 5. Both organophosphates evoked transcriptional changes in 20-25% of the genes in each category; chlorpyrifos and diazinon targeted the same genes, with similar magnitudes of change, as evidenced by high concordance. Furthermore, the same effects were obtained with doses above or below the threshold for cholinesterase inhibition, indicating a mechanism unrelated to anticholinesterase actions. We then evaluated the effects of chlorpyrifos in undifferentiated and differentiating PC12 cells and found even greater targeting of cell cycle and apoptosis genes, affecting up to 40% of all genes in the pathways. Notably, the genes affected in undifferentiated cells were not concordant with those in differentiating cells, pointing to dissimilar outcomes dependent on developmental stage. The in vitro model successfully identified 60-70% of the genes affected by chlorpyrifos in vivo, indicating that the effects are exerted directly on developing neural cells. Our results show that organophosphates target the genes regulating the cell cycle and apoptosis in the developing brain and in neuronotypic cells in culture, with the pattern of vulnerability dependent on the specific stage of development. Equally important, these effects do not reflect actions on cholinesterase and operate at exposures below the threshold for any detectable inhibition of this enzyme.


Subject(s)
Apoptosis/drug effects , Brain/drug effects , Brain/growth & development , Cell Cycle/drug effects , Neurons/pathology , Neurotoxicity Syndromes/etiology , Organophosphates/toxicity , Animals , Animals, Newborn , Brain/metabolism , Brain/pathology , Cell Differentiation/drug effects , Cells, Cultured , Chlorpyrifos/toxicity , Cholinesterase Inhibitors/toxicity , Diazinon/toxicity , Gene Expression/drug effects , Neurons/cytology , Neurons/drug effects , Neurotoxicity Syndromes/metabolism , PC12 Cells/cytology , PC12 Cells/drug effects , Rats , Transcription, Genetic/drug effects
18.
Behav Brain Res ; 229(1): 82-90, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22185695

ABSTRACT

The present study examined the effects of Tremella fuciformis (TF) on the learning and memory function and the neural activity in rats with trimethyltin (TMT)-induced memory deficits. The rats were administered saline or TF (TF 25, 50, 100 mg/kg, p.o.) daily for 21 days. The cognitive improving efficacy of TF on the amnesic rats, which was induced by TMT, was investigated by assessing the Morris water maze test and by performing Choline acetyltransferase (ChAT) and cAMP responsive element binding protein (CREB) immunohistochemistry. In order to confirm the underlying mechanisms of the memory enhancing effects of TF, we assessed the neurite outgrowth of PC12 cells. We also administered 18F-fluorodeoxyglucose and performed a PET scan of the frontal lobe. The rats with TMT injection showed impaired learning and memory of the tasks and treatment with TF produced a significant improvement of the escape latency to find the platform in the Morris water maze compared to that of the control group. In the retention test, the TF50 group showed increased time spent around the platform compared to that of the control group. Consistent with the behavioral data, TF50 mg/kg significantly alleviated the loss of ChAT-ir neurons in the hippocampus compared to that of the control group. Treatment with TF significantly increased the CREB positive neurons in the hippocampal CA1 area as compared to that of the control group. In addition, TF treatment (50 mg/kg) increased the glucose uptake approximately sevenfold in the frontal lobe and it significantly promoted neurite outgrowth of the PC12 cells, as compared to that of the controls. These results suggest that TF may be useful for improving the cognitive function via regulation of the CREB signaling pathway and cholinergic system in the hippocampus.


Subject(s)
CREB-Binding Protein/metabolism , Cholinergic Agents/metabolism , Memory Disorders/drug therapy , Neurites/drug effects , Neuroprotective Agents/therapeutic use , Polysaccharides/therapeutic use , Analysis of Variance , Animals , Choline O-Acetyltransferase/metabolism , Dose-Response Relationship, Drug , Fluorodeoxyglucose F18 , Gene Expression Regulation/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Male , Maze Learning/drug effects , Memory Disorders/chemically induced , Memory Disorders/diagnostic imaging , Neuroprotective Agents/pharmacology , PC12 Cells/cytology , Polysaccharides/pharmacology , Positron-Emission Tomography , Rats , Rats, Sprague-Dawley , Time Factors , Trimethyltin Compounds/toxicity
19.
J Biomed Mater Res A ; 99(4): 598-606, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-21953886

ABSTRACT

Substrate factors such as surface energy distribution can affect cell functions, such as neuronal differentiation of PC12 cells. However, the surface effects that trigger such cell responses need to be clarified and analyzed. Here we show that the total surface tension is not a critical parameter. Self-assembled monolayers of alkylsiloxanes on glass were used as culture substrates. By changing the nanoscale structure and ordering of the monolayer, we designed surfaces with a range of dispersive (γ(d) ) and nondispersive (γ(nd) ) potentials, but with a similar value for total free-energy (50 ≤ γ(d) + γ(nd) ≤ 55 mN m⁻¹). When seeded on surfaces displaying γ(d) /γ(nd) ≤ 3.7, PC12 cells underwent low level of neuritogenesis. On surfaces exhibiting γ(d) /γ(nd) ≥ 5.4, neurite outgrowth was greatly enhanced and apparent by only 24 h of culture in absence of nerve growth-factor treatment. These data indicate how the spatial distribution of surface potentials may control neuritogenesis, thus providing a new criterion to address nerve regeneration issues on rigid biocompatible surfaces.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/physiology , Neurites/physiology , Animals , Cell Culture Techniques/instrumentation , Glass/chemistry , Materials Testing , Molecular Structure , Nanostructures , Neurites/ultrastructure , PC12 Cells/cytology , Rats , Siloxanes/chemistry , Surface Properties
20.
BMB Rep ; 44(5): 312-6, 2011 May.
Article in English | MEDLINE | ID: mdl-21615985

ABSTRACT

1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and its toxic metabolite 1-methyl-4-phenylpyridium ion (MPP(+)) have been shown to induce Parkinson's disease-like symptoms as well as neurotoxicity in humans and animal species. Recently, we reported that maintenance of redox balance and cellular defense against oxidative damage are primary functions of the novel antioxidant enzyme cytosolic NADP(+) -dependent isocitrate dehydrogenase (IDPc). In this study, we examined the role of IDPc in cellular defense against MPP(+) -induced oxidative injury using PC12 cells transfected with IDPc small interfering RNA (siRNA). Our results demonstrate that MPP(+) -mediated disruption of cellular redox status, oxidative damage to cells, and apoptotic cell death were significantly enhanced by knockdown of IDPc.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Cytosol/enzymology , Isocitrate Dehydrogenase/metabolism , NADP/metabolism , Oxidative Stress , PC12 Cells/drug effects , PC12 Cells/enzymology , Animals , Apoptosis/drug effects , Gene Knockdown Techniques , Isocitrate Dehydrogenase/genetics , Oxidation-Reduction , PC12 Cells/cytology , RNA, Small Interfering/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...