Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
3.
Nat Cancer ; 4(10): 1418-1436, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37697045

ABSTRACT

Glioblastoma (GBM) is an incurable brain cancer that lacks effective therapies. Here we show that EAG2 and Kvß2, which are predominantly expressed by GBM cells at the tumor-brain interface, physically interact to form a potassium channel complex due to a GBM-enriched Kvß2 isoform. In GBM cells, EAG2 localizes at neuron-contacting regions in a Kvß2-dependent manner. Genetic knockdown of the EAG2-Kvß2 complex decreases calcium transients of GBM cells, suppresses tumor growth and invasion and extends the survival of tumor-bearing mice. We engineered a designer peptide to disrupt EAG2-Kvß2 interaction, thereby mitigating tumor growth in patient-derived xenograft and syngeneic mouse models across GBM subtypes without overt toxicity. Neurons upregulate chemoresistant genes in GBM cells in an EAG2-Kvß2-dependent manner. The designer peptide targets neuron-associated GBM cells and possesses robust efficacy in treating temozolomide-resistant GBM. Our findings may lead to the next-generation therapeutic agent to benefit patients with GBM.


Subject(s)
Glioblastoma , Humans , Mice , Animals , Glioblastoma/drug therapy , Temozolomide/pharmacology , Temozolomide/therapeutic use , Ether-A-Go-Go Potassium Channels/therapeutic use , Disease Models, Animal , Peptides/therapeutic use , Neurons/pathology
4.
Int J Mol Sci ; 24(13)2023 Jul 04.
Article in English | MEDLINE | ID: mdl-37446272

ABSTRACT

ATP, as a paracrine signalling molecule, induces intracellular Ca2+ elevation via the activation of purinergic receptors on the surface of glia-like cochlear supporting cells. These cells, including the Deiters' cells (DCs), are also coupled by gap junctions that allow the propagation of intercellular Ca2+ waves via diffusion of Ca2+ mobilising second messenger IP3 between neighbouring cells. We have compared the ATP-evoked Ca2+ transients and the effect of two different gap junction (GJ) blockers (octanol and carbenoxolone, CBX) on the Ca2+ transients in DCs located in the apical and middle turns of the hemicochlea preparation of BALB/c mice (P14-19). Octanol had no effect on Ca2+ signalling, while CBX inhibited the ATP response, more prominently in the middle turn. Based on astrocyte models and using our experimental results, we successfully simulated the Ca2+ dynamics in DCs in different cochlear regions. The mathematical model reliably described the Ca2+ transients in the DCs and suggested that the tonotopical differences could originate from differences in purinoceptor and Ca2+ pump expressions and in IP3-Ca2+ release mechanisms. The cochlear turn-dependent effect of CBX might be the result of the differing connexin isoform composition of GJs along the tonotopic axis. The contribution of IP3-mediated Ca2+ signalling inhibition by CBX cannot be excluded.


Subject(s)
Calcium , Gap Junctions , Mice , Animals , Mice, Inbred BALB C , Calcium/metabolism , Gap Junctions/metabolism , Receptors, Purinergic/metabolism , Organ of Corti/metabolism , Hearing , Adenosine Triphosphate/metabolism
5.
Neuron ; 111(1): 30-48.e14, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36323321

ABSTRACT

Major obstacles in brain cancer treatment include the blood-tumor barrier (BTB), which limits the access of most therapeutic agents, and quiescent tumor cells, which resist conventional chemotherapy. Here, we show that Sox2+ tumor cells project cellular processes to ensheathe capillaries in mouse medulloblastoma (MB), a process that depends on the mechanosensitive ion channel Piezo2. MB develops a tissue stiffness gradient as a function of distance to capillaries. Sox2+ tumor cells perceive substrate stiffness to sustain local intracellular calcium, actomyosin tension, and adhesion to promote cellular process growth and cell surface sequestration of ß-catenin. Piezo2 knockout reverses WNT/ß-catenin signaling states between Sox2+ tumor cells and endothelial cells, compromises the BTB, reduces the quiescence of Sox2+ tumor cells, and markedly enhances the MB response to chemotherapy. Our study reveals that mechanosensitive tumor cells construct the BTB to mask tumor chemosensitivity. Targeting Piezo2 addresses the BTB and tumor quiescence properties that underlie treatment failures in brain cancer.


Subject(s)
Brain Neoplasms , beta Catenin , Mice , Animals , beta Catenin/metabolism , beta Catenin/therapeutic use , Endothelial Cells/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain/metabolism , Ion Channels/metabolism , Blood-Brain Barrier/metabolism
6.
Sci Data ; 8(1): 217, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34385453

ABSTRACT

The Open Databases Integration for Materials Design (OPTIMADE) consortium has designed a universal application programming interface (API) to make materials databases accessible and interoperable. We outline the first stable release of the specification, v1.0, which is already supported by many leading databases and several software packages. We illustrate the advantages of the OPTIMADE API through worked examples on each of the public materials databases that support the full API specification.

7.
J Neurosci ; 40(45): 8652-8668, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33060174

ABSTRACT

Inhibitory interneurons integrate into developing circuits in specific ratios and distributions. In the neocortex, inhibitory network formation occurs concurrently with the apoptotic elimination of a third of GABAergic interneurons. The cell surface molecules that select interneurons to survive or die are unknown. Here, we report that members of the clustered Protocadherins (cPCDHs) control GABAergic interneuron survival during developmentally-regulated cell death. Conditional deletion of the gene cluster encoding the γ-Protocadherins (Pcdhgs) from developing GABAergic neurons in mice of either sex causes a severe loss of inhibitory populations in multiple brain regions and results in neurologic deficits such as seizures. By focusing on the neocortex and the cerebellar cortex, we demonstrate that reductions of inhibitory interneurons result from elevated apoptosis during the critical postnatal period of programmed cell death (PCD). By contrast, cortical interneuron (cIN) populations are not affected by removal of Pcdhgs from pyramidal neurons or glial cells. Interneuron loss correlates with reduced AKT signaling in Pcdhg mutant interneurons, and is rescued by genetic blockade of the pro-apoptotic factor BAX. Together, these findings identify the PCDHGs as pro-survival transmembrane proteins that select inhibitory interneurons for survival and modulate the extent of PCD. We propose that the PCDHGs contribute to the formation of balanced inhibitory networks by controlling the size of GABAergic interneuron populations in the developing brain.SIGNIFICANCE STATEMENT A pivotal step for establishing appropriate excitatory-inhibitory ratios is adjustment of neuronal populations by cell death. In the mouse neocortex, a third of GABAergic interneurons are eliminated by BAX-dependent apoptosis during the first postnatal week. Interneuron cell death is modulated by neural activity and pro-survival pathways but the cell-surface molecules that select interneurons for survival or death are unknown. We demonstrate that members of the cadherin superfamily, the clustered γ-Protocadherins (PCDHGs), regulate the survival of inhibitory interneurons and the balance of cell death. Deletion of the Pcdhgs in mice causes inhibitory interneuron loss in the cortex and cerebellum, and leads to motor deficits and seizures. Our findings provide a molecular basis for controlling inhibitory interneuron population size during circuit formation.


Subject(s)
Cadherins/physiology , Cell Death/physiology , Interneurons/physiology , gamma-Aminobutyric Acid/physiology , Animals , Apoptosis/genetics , Cadherin Related Proteins , Cadherins/genetics , Cerebral Cortex/cytology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/growth & development , Electroencephalography , Female , Magnetic Resonance Imaging , Male , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Nerve Net/physiology , Nervous System Diseases/etiology , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/physiology , Seizures/etiology , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/physiology
8.
J Neuroinflammation ; 17(1): 51, 2020 Feb 07.
Article in English | MEDLINE | ID: mdl-32028989

ABSTRACT

BACKGROUND: Radiotherapy is widely used and effective for treating brain tumours, but inevitably impairs cognition as it arrests cellular processes important for learning and memory. This is particularly evident in the aged brain with limited regenerative capacity, where radiation produces irreparable neuronal damage and activation of neighbouring microglia. The latter is responsible for increased neuronal death and contributes to cognitive decline after treatment. To date, there are few effective means to prevent cognitive deficits after radiotherapy. METHODS: Here we implanted hematopoietic stem cells (HSCs) from young or old (2- or 18-month-old, respectively) donor mice expressing green fluorescent protein (GFP) into old recipients and assessed cognitive abilities 3 months post-reconstitution. RESULTS: Regardless of donor age, GFP+ cells homed to the brain of old recipients and expressed the macrophage/microglial marker, Iba1. However, only young cells attenuated deficits in novel object recognition and spatial memory and learning in old mice post-irradiation. Mechanistically, old recipients that received young HSCs, but not old, displayed significantly greater dendritic spine density and long-term potentiation (LTP) in CA1 neurons of the hippocampus. Lastly, we found that GFP+/Iba1+ cells from young and old donors were differentially polarized to an anti- and pro-inflammatory phenotype and produced neuroprotective factors and reactive nitrogen species in vivo, respectively. CONCLUSION: Our results suggest aged peripherally derived microglia-like cells may exacerbate cognitive impairments after radiotherapy, whereas young microglia-like cells are polarized to a reparative phenotype in the irradiated brain, particularly in neural circuits associated with rewards, learning, and memory. These findings present a proof-of-principle for effectively reinstating central cognitive function of irradiated brains with peripheral stem cells from young donor bone marrow.


Subject(s)
Cognitive Dysfunction/therapy , Hematopoietic Stem Cell Transplantation , Maze Learning/physiology , Radiotherapy/adverse effects , Recovery of Function/physiology , Animals , Behavior, Animal/physiology , Cognitive Dysfunction/etiology , Dendritic Spines/physiology , Hippocampus/physiology , Humans , Long-Term Potentiation/physiology , Memory/physiology , Mice , Neurons/physiology , Spinocerebellar Ataxias/genetics , Treatment Outcome
9.
Mol Psychiatry ; 25(9): 2017-2035, 2020 09.
Article in English | MEDLINE | ID: mdl-30224722

ABSTRACT

Principal neurons encode information by varying their firing rate and patterns precisely fine-tuned through GABAergic interneurons. Dysregulation of inhibition can lead to neuropsychiatric disorders, yet little is known about the molecular basis underlying inhibitory control. Here, we find that excessive GABA release from basket cells (BCs) attenuates the firing frequency of Purkinje neurons (PNs) in the cerebellum of Fragile X Mental Retardation 1 (Fmr1) knockout (KO) mice, a model of Fragile X Syndrome (FXS) with abrogated expression of the Fragile X Mental Retardation Protein (FMRP). This over-inhibition originates from increased excitability and Ca2+ transients in the presynaptic terminals, where Kv1.2 potassium channels are downregulated. By paired patch-clamp recordings, we further demonstrate that acutely introducing an N-terminal fragment of FMRP into BCs normalizes GABA release in the Fmr1-KO synapses. Conversely, direct injection of an inhibitory FMRP antibody into BCs, or membrane depolarization of BCs, enhances GABA release in the wild type synapses, leading to abnormal inhibitory transmission comparable to the Fmr1-KO neurons. We discover that the N-terminus of FMRP directly binds to a phosphorylated serine motif on the C-terminus of Kv1.2; and that loss of this interaction in BCs exaggerates GABA release, compromising the firing activity of PNs and thus the output from the cerebellar circuitry. An allosteric Kv1.2 agonist, docosahexaenoic acid, rectifies the dysregulated inhibition in vitro as well as acoustic startle reflex and social interaction in vivo of the Fmr1-KO mice. Our results unravel a novel molecular locus for targeted intervention of FXS and perhaps autism.


Subject(s)
Fragile X Mental Retardation Protein , Fragile X Syndrome , Animals , Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Interneurons/metabolism , Mice , Mice, Knockout , Synaptic Transmission , gamma-Aminobutyric Acid
10.
Cells ; 8(10)2019 10 17.
Article in English | MEDLINE | ID: mdl-31627326

ABSTRACT

Exploring the development of the hearing organ helps in the understanding of hearing and hearing impairments and it promotes the development of the regenerative approaches-based therapeutic efforts. The role of supporting cells in the development of the organ of Corti is much less elucidated than that of the cochlear sensory receptor cells. The use of our recently published method of single-cell electroporation loading of a fluorescent Ca2+ probe in the mouse hemicochlea preparation provided an appropriate means to investigate the Deiters' cells at the subcellular level in two different cochlear turns (apical, middle). Deiters' cell's soma and process elongated, and the process became slimmer by maturation without tonotopic preference. The tonotopically heterogeneous spontaneous Ca2+ activity less frequently occurred by maturation and implied subcellular difference. The exogenous ATP- and UTP-evoked Ca2+ responses were maturation-dependent and showed P2Y receptor dominance in the apical turn. By monitoring the basic structural dimensions of this supporting cell type as well as its spontaneous and evoked purinergic Ca2+ signaling in the hemicochlea preparation in different stages in the critical postnatal P5-25 developmental period for the first time, we showed that the soma and the phalangeal process of the Deiters' cells go through age- and tonotopy-dependent changes in the morphometric parameters and purinergic signaling.


Subject(s)
Cochlea/metabolism , Hair Cells, Auditory/metabolism , Adenosine Triphosphate/pharmacology , Animals , Calcium/metabolism , Cochlea/drug effects , Electroporation , In Vitro Techniques , Mice , Mice, Inbred BALB C , Models, Theoretical , Receptors, Purinergic/metabolism , Signal Transduction/drug effects , Uridine Triphosphate/pharmacology
11.
Nat Commun ; 10(1): 826, 2019 02 18.
Article in English | MEDLINE | ID: mdl-30778063

ABSTRACT

Synaptic heterogeneity is widely observed but its underpinnings remain elusive. We addressed this issue using mature calyx of Held synapses whose numbers of bouton-like swellings on stalks of the nerve terminals inversely correlate with release probability (Pr). We examined presynaptic Ca2+ currents and transients, topology of fluorescently tagged knock-in Ca2+ channels, and Ca2+ channel-synaptic vesicle (SV) coupling distance using Ca2+ chelator and inhibitor of septin cytomatrix in morphologically diverse synapses. We found that larger clusters of Ca2+ channels with tighter coupling distance to SVs elevate Pr in stalks, while smaller clusters with looser coupling distance lower Pr in swellings. Septin is a molecular determinant of the differences in coupling distance. Supported by numerical simulations, we propose that varying the ensemble of two morphological modules containing distinct Ca2+ channel-SV topographies diversifies Pr in the terminal, thereby establishing a morpho-functional continuum that expands the coding capacity within a single synapse population.

12.
Hear Res ; 371: 75-86, 2019 01.
Article in English | MEDLINE | ID: mdl-30504093

ABSTRACT

Ca2+ is an important intracellular messenger and regulator in both physiological and pathophysiological mechanisms in the hearing organ. Investigation of cellular Ca2+ homeostasis in the mature cochlea is hampered by the special anatomy and high vulnerability of the organ. A quick, straightforward and reliable Ca2+ imaging method with high spatial and temporal resolution in the mature organ of Corti is missing. Cell cultures or isolated cells do not preserve the special microenvironment and intercellular communication, while cochlear explants are excised from only a restricted portion of the organ of Corti and usually from neonatal pre-hearing murines. The hemicochlea, prepared from hearing mice allows tonotopic experimental approach on the radial perspective in the basal, middle and apical turns of the organ. We used the preparation recently for functional imaging in supporting cells of the organ of Corti after bulk loading of the Ca2+ indicator. However, bulk loading takes long time, is variable and non-selective, and causes the accumulation of the indicator in the extracellular space. In this study we show the improved labeling of supporting cells of the organ of Corti by targeted single-cell electroporation in mature mouse hemicochlea. Single-cell electroporation proved to be a reliable way of reducing the duration and variability of loading and allowed subcellular Ca2+ imaging by increasing the signal-to-noise ratio, while cell viability was retained during the experiments. We demonstrated the applicability of the method by measuring the effect of purinergic, TRPA1, TRPV1 and ACh receptor stimulation on intracellular Ca2+ concentration at the cellular and subcellular level. In agreement with previous results, ATP evoked reversible and repeatable Ca2+ transients in Deiters', Hensen's and Claudius' cells. TRPA1 and TRPV1 stimulation by AITC and capsaicin, respectively, failed to induce any Ca2+ response in the supporting cells, except in a single Hensen's cell in which AITC evoked transients with smaller amplitude. AITC also caused the displacement of the tissue. Carbachol, agonist of ACh receptors induced Ca2+ transients in about a third of Deiters' and fifth of Hensen's cells. Here we have presented a fast and cell-specific indicator loading method allowing subcellular functional Ca2+ imaging in supporting cells of the organ of Corti in the mature hemicochlea preparation, thus providing a straightforward tool for deciphering the poorly understood regulation of Ca2+ homeostasis in these cells.


Subject(s)
Calcium/metabolism , Cochlea/cytology , Cochlea/metabolism , Adenosine Triphosphate/metabolism , Aniline Compounds/administration & dosage , Animals , Calcium Chelating Agents/administration & dosage , Calcium Signaling/drug effects , Carbachol/administration & dosage , Cochlea/drug effects , Electroporation/methods , Fluoresceins/administration & dosage , Fluorescent Dyes/administration & dosage , Fura-2/administration & dosage , In Vitro Techniques , Labyrinth Supporting Cells/cytology , Labyrinth Supporting Cells/drug effects , Labyrinth Supporting Cells/metabolism , Mice , Mice, Inbred BALB C , Organ of Corti/cytology , Organ of Corti/drug effects , Organ of Corti/metabolism , Receptors, Cholinergic/metabolism , Single-Cell Analysis/methods , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/metabolism
14.
J Chem Phys ; 148(24): 241739, 2018 Jun 28.
Article in English | MEDLINE | ID: mdl-29960375

ABSTRACT

We assess Gaussian process (GP) regression as a technique to model interatomic forces in metal nanoclusters by analyzing the performance of 2-body, 3-body, and many-body kernel functions on a set of 19-atom Ni cluster structures. We find that 2-body GP kernels fail to provide faithful force estimates, despite succeeding in bulk Ni systems. However, both 3- and many-body kernels predict forces within an ∼0.1 eV/Šaverage error even for small training datasets and achieve high accuracy even on out-of-sample, high temperature structures. While training and testing on the same structure always provide satisfactory accuracy, cross-testing on dissimilar structures leads to higher prediction errors, posing an extrapolation problem. This can be cured using heterogeneous training on databases that contain more than one structure, which results in a good trade-off between versatility and overall accuracy. Starting from a 3-body kernel trained this way, we build an efficient non-parametric 3-body force field that allows accurate prediction of structural properties at finite temperatures, following a newly developed scheme [A. Glielmo et al., Phys. Rev. B 95, 214302 (2017)]. We use this to assess the thermal stability of Ni19 nanoclusters at a fractional cost of full ab initio calculations.

15.
Biomed Opt Express ; 6(3): 1021-9, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25798323

ABSTRACT

We created a computational model to investigate the characteristics of label-free molecular detection by stimulated emission, which is the fundamental process of stimulated emission microscopy proposed and experimentally demonstrated by Min et al. In our model the molecule is considered to be a two-state quantum system with finite number of vibrational states. The laser excitations are modelled as zero order Gaussian beams. The field-molecule interaction is considered to be an electric dipole interaction. Based on these assumptions we constructed a Liouville-von Neumann master equation for the reduced density operator. The numerical solution of the master equation determines the expectation value of additional photons produced by stimulated emission. Based on this model algorithms are proposed to evaluate relative excitations. Linear dependence in concentration and quadratic dependence in space resolution were obtained at weak excitations. Time delay dependent relative excitation can be evaluated by taking into account only a single vibrational mode. However, to calculate the spectrum of relative excitation two entangled vibrational modes are necessary. An algorithm is proposed that overcomes the problem of computational complexity and enables to evaluate the spectrum on a high-end computer. High correlation between calculated and measured data of time delay and frequency dependent relative excitation, confirm the validity of the proposed model.

16.
J Neurosci ; 34(42): 14032-45, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25319700

ABSTRACT

Mitral cells express low-voltage activated Cav3.3 channels on their distal apical tuft dendrites (McKay et al., 2006; Johnston and Delaney, 2010). They also discharge Na(+)-dependent dendritic action potentials and release glutamate from these dendrites. Around resting membrane potentials, between -65 and -50 mV, Cav3.x channels are a primary determinant of cytoplasmic [Ca(2+)]. In this study using C57 mice, we present evidence that subthreshold Cav3.x-mediated Ca(2+) influx modulates action potential evoked transmitter release and directly drives asynchronous release from distal tuft dendrites. Presynaptic hyperpolarization and selective block of Cav3.x channels with Z941 (Tringham et al., 2012) reduce mitral-to-mitral EPSP amplitude, increase the coefficient of variation of EPSPs, and increase paired-pulse ratios, consistent with a reduced probability of transmitter release. Both hyperpolarization and Cav3.x channel blockade reduce steady-state cytoplasmic [Ca(2+)] in the tuft dendrite without reducing action potential evoked Ca(2+) influx, suggesting that background [Ca(2+)] modulates evoked release. We demonstrate that Cav3.x-mediated Ca(2+) influx from even one mitral cell at membrane potentials between -65 and -50 mV is sufficient to produce feedback inhibition from periglomerular neurons. Deinactivation of Cav3.x channels by hyperpolarization increases T-type Ca(2+) influx upon repolarization and increases feedback inhibition to produce subthreshold modulation of the mitral-periglomerular reciprocal circuit.


Subject(s)
Calcium Channels, T-Type/physiology , Caveolin 3/physiology , Dendrites/physiology , Olfactory Bulb/physiology , Olfactory Pathways/physiology , Presynaptic Terminals/physiology , Action Potentials/physiology , Animals , Excitatory Postsynaptic Potentials/physiology , Female , Male , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Neural Inhibition/physiology , Organ Culture Techniques
17.
J Neurochem ; 111(3): 745-56, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19719824

ABSTRACT

Although the role of Na(+) in several aspects of Ca(2+) regulation has already been shown, the exact mechanism of intracellular Ca(2+) concentration ([Ca(2+)](i)) increase resulting from an enhancement in the persistent, non-inactivating Na(+) current (I(Na,P)), a decisive factor in certain forms of epilepsy, has yet to be resolved. Persistent Na(+) current, evoked by veratridine, induced bursts of action potentials and sustained membrane depolarization with monophasic intracellular Na(+) concentration ([Na(+)](i)) and biphasic [Ca(2+)](i) increase in CA1 pyramidal cells in acute hippocampal slices. The Ca(2+) response was tetrodotoxin- and extracellular Ca(2+)-dependent and ionotropic glutamate receptor-independent. The first phase of [Ca(2+)](i) rise was the net result of Ca(2+) influx through voltage-gated Ca(2+) channels and mitochondrial Ca(2+) sequestration. The robust second phase in addition involved reverse operation of the Na(+)-Ca(2+) exchanger and mitochondrial Ca(2+) release. We excluded contribution of the endoplasmic reticulum. These results demonstrate a complex interaction between persistent, non-inactivating Na(+) current and [Ca(2+)](i) regulation in CA1 pyramidal cells. The described cellular mechanisms are most likely part of the pathomechanism of certain forms of epilepsy that are associated with I(Na,P). Describing the magnitude, temporal pattern and sources of Ca(2+) increase induced by I(Na,P) may provide novel targets for antiepileptic drug therapy.


Subject(s)
Calcium/metabolism , Hippocampus/drug effects , Membrane Potentials/drug effects , Sodium/metabolism , Veratridine/pharmacology , Animals , Animals, Newborn , Anticonvulsants/pharmacology , Biophysics , Cadmium Chloride/pharmacology , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Clonazepam/analogs & derivatives , Clonazepam/pharmacology , Electric Stimulation/methods , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/cytology , In Vitro Techniques , Ionophores/pharmacology , Male , Membrane Potentials/physiology , Patch-Clamp Techniques , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Rats , Rats, Wistar , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Thapsigargin/pharmacology , Thiazepines/pharmacology
18.
Free Radic Biol Med ; 44(6): 1010-22, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18206124

ABSTRACT

The major role of reactive oxygen species (ROS) in the pathomechanism of ischemia have been widely recognized. Still, measurements of the precise time course and regional distribution of ischemia-induced ROS level changes in acute brain slices have been missing. By using acute hippocampal slices and the fluorescent dye CM-H2DCFDA, we showed that reoxygenation after in vitro ischemia (oxygen-glucose deprivation; OGD) increased ROS levels in the hippocampal CA1 layers vulnerable to ischemia but did not have significant effects in the resistant stratum granulosum in the dentate gyrus (DG). Production of ROS started during OGD, but, contrary to reoxygenation, it manifested as a ROS level increase exclusively in the presence of catalase and glutathione peroxidase inhibition. The mechanism of ROS production involves the activation of NMDA receptors and nitric oxide synthases. The inhibition of ROS response by either AP-5 or L-NAME together with the ROS sensitivity profile of the dye suggest that peroxynitrite, the reaction product of superoxide and nitric oxide, plays a role in the response. Direct visualization of layer-specific effects of ROS production and its scavenging, shown for the first time in acute hippocampal slices, suggests that distinct ROS homeostasis may underlie the different ischemic vulnerability of CA1 and DG.


Subject(s)
Brain Ischemia/physiopathology , Hippocampus/metabolism , Reactive Oxygen Species/metabolism , Animals , Glucose/deficiency , Hippocampus/physiopathology , Male , Organ Culture Techniques , Oxygen , Rats , Rats, Wistar , Reactive Oxygen Species/analysis
19.
J Neurochem ; 105(2): 360-8, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18036194

ABSTRACT

Diclofenac (DCF) is a widely used non-steroidal anti-inflammatory drug, which also act as a mitochondrial toxin. As it is known that selective mitochondrial complex I inhibition combined with mild oxidative stress causes striatal dopaminergic dysfunction, we tested whether DCF also compromise dopaminergic function in the striatum. [3H]Dopamine ([3H]DA) release was measured from rat striatal slices after in vitro (2 h, 10-25 micromol/L) or in vivo (3 mg/kg i.v. for 28 days) DCF treatment. In vitro treatment significantly decreased [3H]DA uptake and dopamine (DA) content of the slices. H2O2 (0.1 mmol/L)-evoked DA release was enhanced. Intracellular reactive oxygen species production was not significantly changed in the presence of DCF. After in vivo DCF treatment no apparent decrease in striatal DA content was observed and the uptake of [3H]DA into slices was increased. The intensity of tyrosine hydroxylase immunoreactivity in the striatum was highly variable, and both decrease and increase were observed in individual rats. The H2O2-evoked [3H]DA release was significantly decreased and the effluent contained a significant amount of [3H]octopamine, [3H]tyramine, and [3H]beta-phenylethylamine. The ATP content and adenylate energy charge were decreased. In conclusion, whereas in vitro DCF pre-treatment resembles the effect of the mitochondrial toxin rotenone, in vivo it rather counteracts than aggravates dopaminergic dysfunction.


Subject(s)
Corpus Striatum/drug effects , Cyclooxygenase Inhibitors/pharmacology , Diclofenac/pharmacology , Dopamine/metabolism , Adenosine Triphosphate/metabolism , Analysis of Variance , Animals , Dose-Response Relationship, Drug , Hydrogen Peroxide/pharmacology , In Vitro Techniques , Male , Octopamine/metabolism , Phenelzine/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Tritium/metabolism , Tyramine/metabolism , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...