Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Neurosurg ; 120(3): 628-38, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24266544

ABSTRACT

OBJECT: New experimental models and diagnostic methods are needed to better understand the pathophysiology of focal neocortical epilepsies in a search for improved epilepsy treatment options. The authors hypothesized that a focal disruption of adenosine homeostasis in the neocortex might be sufficient to trigger electrographic seizures. They further hypothesized that a focal disruption of adenosine homeostasis might affect microcirculation and thus offer a diagnostic opportunity for the detection of a seizure focus located in the neocortex. METHODS: Focal disruption of adenosine homeostasis was achieved by injecting an adeno-associated virus (AAV) engineered to overexpress adenosine kinase (ADK), the major metabolic clearance enzyme for the brain's endogenous anticonvulsant adenosine, into the neocortex of mice. Eight weeks following virus injection, the affected brain area was imaged via optical microangiography (OMAG) to detect changes in microcirculation. After completion of imaging, cortical electroencephalography (EEG) recordings were obtained from the imaged brain area. RESULTS: Viral expression of the Adk cDNA in astrocytes generated a focal area (~ 2 mm in diameter) of ADK overexpression within the neocortex. OMAG scanning revealed a reduction in vessel density within the affected brain area of approximately 23% and 29% compared with control animals and the contralateral hemisphere, respectively. EEG recordings revealed electrographic seizures within the focal area of ADK overexpression at a rate of 1.3 ± 0.2 seizures per hour (mean ± SEM). CONCLUSIONS: The findings of this study suggest that focal adenosine deficiency is sufficient to generate a neocortical focus of hyperexcitability, which is also characterized by reduced vessel density. The authors conclude that their model constitutes a useful tool to study neocortical epilepsies and that OMAG constitutes a noninvasive diagnostic tool for the imaging of seizure foci with disrupted adenosine homeostasis.


Subject(s)
Adenosine Kinase/genetics , Adenosine/deficiency , Astrocytes/enzymology , Epilepsies, Partial/metabolism , Neocortex/metabolism , Adenosine/metabolism , Adenosine Kinase/metabolism , Animals , Cerebrovascular Circulation/genetics , Dependovirus/genetics , Disease Models, Animal , Electroencephalography , Epilepsies, Partial/diagnosis , Epilepsies, Partial/genetics , Genetic Vectors , Homeostasis/genetics , Male , Mice , Mice, Inbred C57BL , Microcirculation/genetics , Neocortex/blood supply , Neocortex/cytology
2.
PLoS One ; 8(11): e80902, 2013.
Article in English | MEDLINE | ID: mdl-24312250

ABSTRACT

Adenosine A2A receptors (A2AR) are located postsynaptically in striatopallidal GABAergic neurons, antagonizing dopamine D2 receptor functions, and are also located presynaptically at corticostriatal terminals, facilitating glutamate release. To address the hypothesis that these two A2AR populations differently control the action of psychostimulants, we characterized A2AR modulation of cocaine-induced effects at the level of DARPP-32 phosphorylation at Thr-34 and Thr-75, c-Fos expression, and psychomotor activity using two lines of cell-type selective A2AR knockout (KO) mice with selective A2AR deletion in GABAergic neurons (striatum-A2AR-KO mice), or with A2AR deletion in both striatal GABAergic neurons and projecting cortical glutamatergic neurons (forebrain-A2AR-KO mice). We demonstrated that striatum-A2AR KO mice lacked A2ARs exclusively in striatal GABAergic terminals whereas forebrain-A2AR KO mice lacked A2ARs in both striatal GABAergic and glutamatergic terminals leading to a blunted A2AR-mediated facilitation of synaptosomal glutamate release. The inactivation of A2ARs in GABAergic neurons reduced striatal DARPP-32 phosphorylation at Thr-34 and increased its phosphorylation at Thr-75. Conversely, the additional deletion of corticostriatal glutamatergic A2ARs produced opposite effects on DARPP-32 phosphorylation at Thr-34 and Thr-75. This distinct modulation of DARPP-32 phosphorylation was associated with opposite responses to cocaine-induced striatal c-Fos expression and psychomotor activity in striatum-A2AR KO (enhanced) and forebrain-A2AR KO mice (reduced). Thus, A2ARs in glutamatergic corticostriatal terminals and in GABAergic striatal neurons modulate the action of psychostimulants and DARPP-32 phosphorylation in opposite ways. We conclude that A2ARs in glutamatergic terminals prominently control the action of psychostimulants and define a novel mechanism by which A2ARs fine-tune striatal activity by integrating GABAergic, dopaminergic and glutamatergic signaling.


Subject(s)
Corpus Striatum/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , GABAergic Neurons/metabolism , Glutamic Acid/metabolism , Receptor, Adenosine A2A/metabolism , Animals , Cocaine/pharmacology , Corpus Striatum/drug effects , Gene Expression , Mice , Mice, Knockout , Phosphorylation , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Psychomotor Performance , Receptor, Adenosine A2A/genetics , Synaptosomes/metabolism
3.
J Vasc Interv Radiol ; 24(10): 1537-42.e2, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24070510

ABSTRACT

Molecular analysis of apoptosis and autophagy pathways was performed from a single hepatocellular carcinoma treated with yttrium-90 and doxorubicin-loaded beads before resection and compared with normal liver tissue from the margins. Both bead formulations activated apoptosis-associated mechanisms and increased autophagy pathway protein levels. Increased DNA fragmentation and autophagy markers were seen in tumor treated with drug-eluting beads compared with yttrium-90-treated tumor. These results suggest that both microembolic therapies activate cell death signaling, although differences in apoptosis and autophagy pathways were seen in this patient. Knowledge of mechanisms of action for each treatment may enhance future therapeutic strategies.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/therapy , Doxorubicin/administration & dosage , Liver Neoplasms/metabolism , Liver Neoplasms/therapy , Yttrium Radioisotopes/therapeutic use , Autophagy/drug effects , Autophagy/radiation effects , Capsules , Chemoradiotherapy/methods , Delayed-Action Preparations/administration & dosage , Humans , Male , Middle Aged , Radiopharmaceuticals/therapeutic use , Treatment Outcome
4.
J Clin Invest ; 122(7): 2567-77, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22706302

ABSTRACT

An emerging theory of schizophrenia postulates that hypofunction of adenosine signaling may contribute to its pathophysiology. This study was designed to test the "adenosine hypothesis" of schizophrenia and to evaluate focal adenosine-based strategies for therapy. We found that augmentation of adenosine by pharmacologic inhibition of adenosine kinase (ADK), the key enzyme of adenosine clearance, exerted antipsychotic-like activity in mice. Further, overexpression of ADK in transgenic mice was associated with attentional impairments linked to schizophrenia. We observed that the striatal adenosine A2A receptor links adenosine tone and psychomotor response to amphetamine, an indicator of dopaminergic signaling. Finally, intrastriatal implants of engineered adenosine-releasing cells restored the locomotor response to amphetamine in mice overexpressing ADK, whereas the same grafts placed proximal to the hippocampus of transgenic mice reversed their working memory deficit. This functional double dissociation between striatal and hippocampal adenosine demonstrated in Adk transgenic mice highlights the independent contributions of these two interconnected brain regions in the pathophysiology of schizophrenia and thus provides the rationale for developing local adenosine augmentation therapies for the treatment of schizophrenia.


Subject(s)
Adenosine Kinase/antagonists & inhibitors , Adenosine/metabolism , Cognition Disorders/therapy , Endophenotypes , Psychotic Disorders/therapy , Schizophrenia/drug therapy , Schizophrenic Psychology , Adenosine/deficiency , Adenosine Kinase/metabolism , Amphetamines/pharmacology , Animals , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Basal Ganglia/metabolism , Basal Ganglia/pathology , Cell Transplantation , Cells, Cultured , Cognition Disorders/genetics , Cricetinae , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morpholines/pharmacology , Morpholines/therapeutic use , Psychotic Disorders/genetics , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Receptor, Adenosine A2A/metabolism , Schizophrenia/genetics , Schizophrenia/therapy
5.
Exp Neurol ; 235(2): 497-507, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22309833

ABSTRACT

MicroRNAs are small non-coding RNAs that regulate post-transcriptional gene expression. In the short time since the discovery of microRNAs, the literature has burgeoned with studies focused on the biosynthesis of microRNAs, target prediction and binding, and mechanisms of translational repression by microRNAs. Given the prominent role of microRNAs in all areas of cell biology, it is not surprising that microRNAs are also linked to human diseases, including those of the nervous system. One of the least-studied areas of microRNA research is how their expression is regulated outside of development and cancer. Thus, we examined a role for regulation of microRNAs by neurotransmitter receptor activation in mouse brain. We focused on the group I metabotropic glutamate receptors by using intracerebroventricular injection of the selective agonist, (S)-3,5-dihydroxyphenylglycine (DHPG) in mouse brain. We then examined the expression of microRNAs in the cerebral cortex by Ambion and Invitrogen microarrays, and the expression of mature microRNA sequences by SABiosciences qPCR arrays, at 4, 8 and 24 h after DHPG injection. These studies revealed that the largest number of significantly regulated microRNAs was detected 8h after DHPG injection in the microarrays and qPCR arrays. We then used RNA blots to quantify microRNA expression, and in situ hybridization to examine cellular distribution of the microRNAs regulated by DHPG. Bioinformatic analysis of the microRNAs regulated 8 h after DHPG in all three arrays revealed KEGG pathways that are known to correlate with group I mGluR effects, as well as recently described and novel pathways. These studies are the first to show that DHGP regulates the expression of microRNAs in mouse cerebral cortex, and support the hypothesis that group I mGluRs may regulate microRNA expression in mouse brain.


Subject(s)
Brain/metabolism , Gene Expression Regulation , Glycine/analogs & derivatives , MicroRNAs/biosynthesis , Resorcinols/administration & dosage , Animals , Brain/drug effects , Excitatory Amino Acid Agonists/administration & dosage , Glycine/administration & dosage , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/biosynthesis
6.
Glia ; 60(1): 83-95, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21964979

ABSTRACT

Astrogliosis and associated dysfunction of adenosine homeostasis are pathological hallmarks of the epileptic brain and thought to contribute to seizure generation in epilepsy. The authors hypothesized that astrogliosis-an early component of the epileptogenic cascade-might be linked to focal seizure onset. To isolate the contribution of astrogliosis to ictogenesis from other pathological events involved in epilepsy, the authors used a minimalistic model of epileptogenesis in mice, based on a focal onset status epilepticus triggered by intra-amygdaloid injection of kainic acid. The authors demonstrate acute neuronal cell loss restricted to the injected amygdala and ipsilateral CA3, followed 3 weeks later by focal astrogliosis and overexpression of the adenosine-metabolizing enzyme adenosine kinase (ADK). Using synchronous electroencephalographic recordings from multiple depth electrodes, the authors identify the KA-injected amygdala and ipsilateral CA3 as two independent foci for the initiation of non-synchronized electrographic subclinical seizures. Importantly, seizures remained focal and restricted to areas of ADK overexpression. However, after systemic application of a non-convulsive dose of an adenosine A(1) -receptor antagonist, seizures in amygdala and CA3 immediately synchronized and spread throughout the cortex, leading to convulsive seizures. This focal seizure phenotype remained stable over at least several weeks. We conclude that astrogliosis via disruption of adenosine homeostasis per se and in the absence of any other overt pathology, is associated with the emergence of spontaneous recurrent subclinical seizures, which remain stable over space and time. A secondary event, here mimicked by brain-wide disruption of adenosine signaling, is likely required to turn pre-existing subclinical seizures into a clinical seizure phenotype.


Subject(s)
Adenosine/metabolism , Gliosis/complications , Neuroglia/metabolism , Seizures/etiology , Seizures/pathology , Adenosine Kinase/metabolism , Amygdala/drug effects , Amygdala/pathology , Animals , Disease Models, Animal , Electroencephalography , Gene Expression Regulation/drug effects , Gliosis/chemically induced , Hippocampus/drug effects , Hippocampus/pathology , Kainic Acid/toxicity , Male , Mice , Mice, Inbred C57BL , Time Factors
7.
J Cereb Blood Flow Metab ; 31(7): 1648-59, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21427729

ABSTRACT

Adenosine kinase (ADK) is the major negative metabolic regulator of the endogenous neuroprotectant and homeostatic bioenergetic network regulator adenosine. We used three independent experimental approaches to determine the role of ADK as a molecular target for predicting the brain's susceptibility to ischemic stroke. First, when subjected to a middle cerebral artery occlusion model of focal cerebral ischemia, transgenic fb-Adk-def mice, which have increased ADK expression in striatum (164%) and reduced ADK expression in cortical forebrain (65%), demonstrate increased striatal infarct volume (126%) but almost complete protection of cortex (27%) compared with wild-type (WT) controls, indicating that cerebral injury levels directly correlate to levels of ADK in the CNS. Second, we demonstrate abrogation of lipopolysaccharide (LPS)-induced ischemic preconditioning in transgenic mice with brain-wide ADK overexpression (Adk-tg), indicating that ADK activity negatively regulates LPS-induced tolerance to stroke. Third, using adeno-associated virus-based vectors that carry Adk-sense or -antisense constructs to overexpress or knockdown ADK in vivo, we demonstrate increased (126%) or decreased (51%) infarct volume, respectively, 4 weeks after injection into the striatum of WT mice. Together, our data define ADK as a possible therapeutic target for modulating the degree of stroke-induced brain injury.


Subject(s)
Adenosine Kinase/metabolism , Brain Ischemia/enzymology , Brain Ischemia/pathology , Brain/enzymology , Brain/pathology , Adenosine/metabolism , Adenosine Kinase/genetics , Animals , Brain/blood supply , Brain Ischemia/therapy , Cerebral Cortex/blood supply , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Down-Regulation , Gene Deletion , Gene Expression , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/therapy , Ischemic Preconditioning , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Up-Regulation
8.
Int J Physiol Pathophysiol Pharmacol ; 2(2): 148-160, 2010 Jun 30.
Article in English | MEDLINE | ID: mdl-20740046

ABSTRACT

In retina, an ischemic injury-resistant condition (ischemic tolerance) can be induced by a sub-lethal ischemic treatment (preconditioning) prior to an otherwise injurious ischemic insult. In this work, we compared retinal proteomic changes under three different ischemic conditions, as a means to identify the effector mechanisms that underlie retinal ischemic tolerance. Transient retinal ischemia was induced by elevating the intraocular pressure (IOP) in three groups of adult rats as follows: Group 1, ischemic-preconditioned, 110 mmHg for 8 minutes followed by 48 hours reperfusion; Group 2, ischemic-injured, 110 mmHg for 60 minutes followed by 24 hours reperfusion; Group 3, ischemic-tolerant, preconditioning treatment followed by another 60 minutes of 110 mmHg and 24 hours reperfusion. Protein quantities in each of the afore-mentioned retinal ischemic conditions, as determined by quantitative mass spectrometry, were compared with that of the contralateral control eyes (sham-treated). As a result, a total of 328 proteins were identified and quantified; among them, 30-60% of proteins showed a change in abundance under one or more retinal ischemic conditions. In particular, in ischemic-tolerant retinas, histone proteins H2B, H3 and H4 demonstrated an increase in abundance, whereas histone H2A showed a decrease in abundance. Further immunohistochemical analyses confirmed the results of proteomic analyses, and detected an up regulation of tri-methylated histone H3, mono-ubiquitinated histone H2A and Polycomb group protein RING2. Together, these results suggest a role of epigenetic regulation in the induction of retinal ischemic tolerance that involves histone and polycomb proteins.

9.
Sci Signal ; 3(111): ra15, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20197544

ABSTRACT

Exposing the brain to sublethal ischemia affects the response to a subsequent, otherwise injurious ischemia, resulting in transcriptional suppression and neuroprotection, a response called ischemic tolerance. Here, we show that the proteomic signature of the ischemic-tolerant brain is characterized by increased abundance of transcriptional repressors, particularly polycomb group (PcG) proteins. Knocking down PcG proteins precluded the induction of ischemic tolerance, whereas in an in vitro model, overexpressing the PcG proteins SCMH1 or BMI1 induced tolerance to ischemia without preconditioning. We found that PcG proteins are associated with the promoter regions of genes encoding two potassium channel proteins that show decreased abundance in ischemic-tolerant brains. Furthermore, PcG proteins decreased potassium currents in cultured neuronal cells, and knocking down potassium channels elicited tolerance without preconditioning. These findings reveal a previously unknown mechanism of neuroprotection that involves gene repressors of the PcG family.


Subject(s)
Brain Ischemia/physiopathology , Repressor Proteins/genetics , Repressor Proteins/physiology , Animals , Base Sequence , Brain Ischemia/genetics , Epigenesis, Genetic , In Vitro Techniques , Kv1.5 Potassium Channel/genetics , Kv1.5 Potassium Channel/metabolism , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/metabolism , Nuclear Proteins/genetics , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Promoter Regions, Genetic , Proteomics , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , Repressor Proteins/antagonists & inhibitors , Shaker Superfamily of Potassium Channels/genetics , Shaker Superfamily of Potassium Channels/metabolism , Signal Transduction
10.
Biomaterials ; 29(26): 3609-16, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18514814

ABSTRACT

Adenosine augmentation therapies (AAT) make rational use of the brain's own adenosine-based seizure control system and hold promise for the therapy of refractory epilepsy. In an effort to develop an AAT compatible with future clinical application, we developed a novel silk protein-based release system for adenosine. Adenosine releasing brain implants with target release doses of 0, 40, 200, and 1000ng adenosine per day were prepared by embedding adenosine containing microspheres into nanofilm-coated silk fibroin scaffolds. In vitro, the respective polymers released 0, 33.4, 170.5, and 819.0ng adenosine per day over 14 days. The therapeutic potential of the implants was validated in a dose-response study in the rat model of kindling epileptogenesis. Four days prior to the onset of kindling, adenosine releasing polymers were implanted into the infrahippocampal cleft and progressive acquisition of kindled seizures was monitored over a total of 48 stimulations. We document a dose-dependent retardation of seizure acquisition. In recipients of polymers releasing 819ng adenosine per day, kindling epileptogenesis was delayed by one week corresponding to 18 kindling stimulations. Histological analysis of brain samples confirmed the correct location of implants and electrodes. We conclude that silk-based delivery of around 1000ng adenosine per day is a safe and efficient strategy to suppress seizures.


Subject(s)
Adenosine/therapeutic use , Drug Carriers/chemistry , Drug Delivery Systems , Epilepsy/drug therapy , Polymers/chemistry , Silk/chemistry , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/therapeutic use , Epilepsy/physiopathology , Fibroins/chemistry , Hippocampus/surgery , Humans , Kindling, Neurologic , Male , Materials Testing , Microspheres , Prostheses and Implants , Rats , Rats, Sprague-Dawley
11.
Neuron Glia Biol ; 4(2): 91-9, 2008 May.
Article in English | MEDLINE | ID: mdl-19674507

ABSTRACT

The astrocytic enzyme adenosine kinase (ADK) is a key negative regulator of the brain's endogenous anticonvulsant adenosine. Astrogliosis with concomitant upregulation of ADK is part of the epileptogenic cascade and contributes to seizure generation. To molecularly dissect the respective roles of astrogliosis and ADK-expression for seizure generation, we used a transgenic approach to uncouple ADK-expression from astrogliosis: in Adk-tg mice the endogenous Adk-gene was deleted and replaced by a ubiquitously expressed Adk-transgene with novel ectopic expression in pyramidal neurons, resulting in spontaneous seizures. Here, we followed a unique approach to selectively injure the CA3 of these Adk-tg mice. Using this strategy, we had the opportunity to study astrogliosis and epileptogenesis in the absence of the endogenous astrocytic Adk-gene. After triggering epileptogenesis we demonstrate astrogliosis without upregulation of ADK, but lack of seizures, whereas matching wild-type animals developed astrogliosis with upregulation of ADK and spontaneous recurrent seizures. By uncoupling ADK-expression from astrogliosis, we demonstrate that global expression levels of ADK rather than astrogliosis per se contribute to seizure generation.


Subject(s)
Adenosine Kinase/metabolism , Astrocytes , Epilepsy/etiology , Gliosis/complications , Adenosine Kinase/deficiency , Adenosine Kinase/genetics , Animals , Astrocytes/enzymology , Brain/enzymology , Cell Death , Chronic Disease , Epilepsy/enzymology , Gliosis/enzymology , Kainic Acid , Male , Mice , Mice, Knockout , Mice, Transgenic , Pyramidal Cells/enzymology , Recurrence , Seizures/chemically induced , Seizures/etiology , Seizures/physiopathology , Seizures/prevention & control , Severity of Illness Index , Status Epilepticus/complications , Status Epilepticus/physiopathology , Time Factors , Tissue Distribution , Transgenes , Up-Regulation
12.
J Cereb Blood Flow Metab ; 25(2): 234-46, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15647742

ABSTRACT

Bcl-2 plays a pivotal role in the control of cell death and is upregulated by ischemic tolerance. Because Bcl-2 expression is regulated by the transcription factor cyclic AMP response element-binding protein (CREB), we investigated the role of CREB activation in two models of ischemic preconditioning: focal ischemic tolerance after middle cerebral artery occlusion (MCAO) and in vitro ischemic tolerance modeled by oxygen-glucose deprivation (OGD). After preconditioning ischemia (30 minutes MCAO or 30 minutes OGD), phosphorylation of CREB was increased, and there was an increased interaction between the bcl-2 cyclic AMP-responsive element (CRE) promoter and nuclear proteins after preconditioning ischemia in vivo and in vitro. Chromatin immunoprecipitation revealed an increased interaction between CREB-binding protein and the bcl-2 CRE rather than CREB, after preconditioning ischemia. Ischemic tolerance was blocked by a CRE decoy oligonucleotide, which also blocked Bcl-2 expression. The protein kinase A inhibitor H89, the calcium/calmodulin kinase inhibitor KN62, and the MEK inhibitor U0126 blocked ischemic tolerance, but not the phosphatidylinositol 3-kinase inhibitor LY294002. H89, KN62, and U0126 reduced CREB activation and Bcl-2 expression. Taken together, these data suggest that after ischemic preconditioning CREB activation regulates the expression of the prosurvival protein Bcl-2.


Subject(s)
Brain Ischemia/physiopathology , Cyclic AMP Response Element-Binding Protein/metabolism , Ischemic Preconditioning , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Blotting, Western , Brain Ischemia/pathology , Cells, Cultured , Electrophoretic Mobility Shift Assay , Immunohistochemistry , In Situ Nick-End Labeling , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Male , Neurons/metabolism , Neurons/pathology , Protein Kinases/drug effects , Protein Kinases/metabolism , Rats
13.
J Clin Invest ; 113(7): 1059-68, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15057313

ABSTRACT

Programmed cell death pathways have been implicated in the mechanism by which neurons die following brief and prolonged seizures, but the significance of proapoptotic Bcl-2 family proteins in the process remains poorly defined. Expression of the death agonist Bcl-2-interacting mediator of cell death (Bim) is under the control of the forkhead in rhabdomyosarcoma (FKHR) transcription factors. This prompted us to examine the response of this pathway to experimental seizures and in hippocampi from patients with intractable temporal lobe epilepsy. A short period of status epilepticus in rats that damaged the hippocampus activated FKHR/FKHRL-1 and induced a significant increase in expression of Bim. Blocking of FKHR/FKHRL-1 dephosphorylation after seizures improved hippocampal neuronal survival in vivo, and Bim antisense oligonucleotides were neuroprotective against seizures in vitro. Inhibition of Akt increased the FKHR/Bim response and DNA fragmentation within the normally resistant cortex. Analysis of hippocampi from patients with intractable epilepsy revealed that Bim levels were significantly lower than in controls and FKHR was inhibited; we were able to reproduce these results experimentally in rats by evoking multiple brief, noninjurious electroshock seizures. We conclude that Bim expression may be a critical determinant of whether seizures damage the brain, and that its control may be neuroprotective in status epilepticus and epilepsy.


Subject(s)
Carrier Proteins/metabolism , Epilepsy/metabolism , Hippocampus/metabolism , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Temporal Lobe/metabolism , Transcription Factors/metabolism , Animals , Apoptosis Regulatory Proteins , Bcl-2-Like Protein 11 , Cell Death/physiology , Forkhead Transcription Factors , Humans , Male , Neurons/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2 , Rats , Rats, Sprague-Dawley , Up-Regulation
14.
Ann Neurol ; 55(4): 485-94, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15048887

ABSTRACT

Experimental and human data suggest programmed (active) cell death may contribute to the progressive hippocampal atrophy seen in patients with refractory temporal lobe epilepsy. Death-associated protein (DAP) kinase is a novel calcium/calmodulin-activated kinase that functions in apoptosis mediated by death receptors. Because seizure-induced neuronal death involves both death receptor activation and calcium, we examined DAP kinase expression, localization, and interactions in hippocampal resections from patients with intractable temporal lobe epilepsy (n = 10) and autopsy controls (n = 6). Expression and phosphorylation of DAP kinase was significantly increased in epilepsy brain compared with control. DAP kinase and DAP kinase-interacting protein 1 (DIP-1) localized to mitochondria in control brain, whereas levels of both were increased in the cytoplasm and microsomal (endoplasmic reticulum) fraction in epilepsy samples. Coimmunoprecipitation analysis showed increased DAP kinase binding to calmodulin, DIP-1, and the Fas-associated protein with death domain (FADD) in epilepsy samples. Finally, immunohistochemistry determined DAP kinase was coexpressed with DIP-1 in neurons. This study provides the first description of DAP kinase and DIP-1 in human brain and suggests DAP kinase is a novel molecular regulator of neuronal death in epilepsy.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/biosynthesis , Epilepsy, Temporal Lobe/enzymology , Hippocampus/enzymology , Adolescent , Adult , Apoptosis Regulatory Proteins , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Death-Associated Protein Kinases , Epilepsy, Temporal Lobe/genetics , Epilepsy, Temporal Lobe/pathology , Female , Gene Expression Regulation, Enzymologic/physiology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Middle Aged , Phosphorylation
15.
J Neurosci Res ; 76(1): 121-8, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15048936

ABSTRACT

Although mice are amenable to gene knockout, they have not been exploited in the setting of seizure-induced neurodegeneration due to the resistance to injury of key mouse strains. We refined and developed models of seizure-induced neuronal death in the C57BL/6 and BALB/c strains by focally evoking seizures using intra-amygdala kainic acid. Seizures in adult male BALB/c mice, or C57BL/6 mice as reference, caused ipsilateral death of CA1 and CA3 neurons within the hippocampus. Termination of seizures by lorazepam was more effective than diazepam in both strains, largely restricting neuronal loss to the CA3 sector. Electroencephalography (EEG) recordings defined injurious and non-injurious seizure patterns, which could not be separated adequately by behavioral observation alone. Degenerating neurons in the hippocampus were positive for DNA fragmentation and approximately a third of these exhibited morphologic features of programmed cell death. Western blot analysis revealed the cleavage of caspase-8 after seizures in both strains. These data refine our C57BL/6 model and establish a companion model of focally evoked limbic seizures in the BALB/c mouse that provides further evidence for activation of programmed cell death after seizures.


Subject(s)
Apoptosis/physiology , Hippocampus/pathology , Seizures/pathology , Animals , Anticonvulsants/pharmacology , Blotting, Western , Caspase 8 , Caspases/metabolism , DNA Fragmentation/drug effects , Diazepam/pharmacology , Electroencephalography/drug effects , Genes, bcl-2/genetics , Immunohistochemistry , In Situ Nick-End Labeling , Lorazepam/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Neurological , Neurons/pathology
16.
Space Med Med Eng (Beijing) ; 17(3): 180-3, 2004 Jun.
Article in Chinese | MEDLINE | ID: mdl-15920843

ABSTRACT

OBJECTIVE: To study the ultrastructural changes in the cerebral cortex and cerebellar cortex of rats under simulated weightlessness and the possible mechanism. METHOD: The tail-suspended rats model (-30 degrees head down tilt) was adopted to simulate weightlessness in the experiment. The rats were suspended for 7 d, 14 d, 21 d, and 28 d, and then were perfused through the hearts. The specimens were drawn from the rats' cerebral cortex and cerebellar cortex for electron microscopy. RESULT: The results showed that under simulated weightlessness, the main changes in the neuron can be described as follows: swelling of mitochondria, endoplasmic reticulum and Golgi complex, even formation of big empty vesicles; reduction of number of synaptic vesicles in IV layer; increase corrugation of capillary lumen and thickening of basement membrane. Degranulation of rough endoplasmic reticulum in Purkinje's cells of the cerebellar cortex occurred obviously. On the 14th and the 21st day of suspension, the changes were most significant and tended to return to normal on the 28th day. CONCLUSION: The experimental results demonstrated that simulated weightlessness led to ultrastructural changes in the cerebral cortex and cerebella cortex of rats. The ultrastructure changed with the course of simulated weightlessness and tended to return to normal. It showed an adaption to the simulated weightlessness.


Subject(s)
Cerebral Cortex/ultrastructure , Hindlimb Suspension , Neurons/ultrastructure , Weightlessness Simulation , Adaptation, Physiological , Animals , Endoplasmic Reticulum/pathology , Golgi Apparatus/pathology , Microscopy, Electron , Mitochondria/pathology , Purkinje Cells/pathology , Rats , Rats, Wistar , Time Factors
17.
J Neurochem ; 86(5): 1260-70, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12911633

ABSTRACT

Death-associated protein (DAP) kinase is calcium-regulated and known to function downstream of death receptors, prompting us to examine its role in the mechanism of seizure-induced neuronal death. Brief seizures were focally evoked in rats, eliciting neuronal death within the CA3 subfield of the hippocampus, and to a lesser extent, cortex. Western blotting confirmed expression of DAP kinase within hippocampus and cortex at the predicted weight of approximately 160 kDa. Immunohistochemistry revealed seizures triggered a significant increase in numbers of DAP kinase-expressing cells within CA3 and cortex, without affecting cell counts within seizure-resistant CA2 or the dentate gyrus. Numbers of DAP kinase-expressing cells were increased in relation to specific patterns of injury-causing seizure activity, electrographically defined. Seizures caused an early increase in DAP kinase binding to actin, and association with calmodulin. Co-immunoprecipitation studies also revealed seizures triggered binding of DAP kinase to the tumor necrosis factor receptor 1 and the Fas-associated death domain protein, commensurate with caspase-8 proteolysis. In contrast, within surviving fields of the hippocampus, DAP kinase interacted with the molecular chaperone 14-3-3. These data suggest DAP kinase is involved in the molecular pathways activated during seizure-induced neuronal death.


Subject(s)
Adaptor Proteins, Signal Transducing , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Seizures/physiopathology , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism , 14-3-3 Proteins , Amygdala/drug effects , Amygdala/pathology , Animals , Antigens, CD/metabolism , Apoptosis Regulatory Proteins , Carrier Proteins/metabolism , Cell Count , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Death-Associated Protein Kinases , Disease Models, Animal , Electroencephalography , Enzyme Activation , Fas-Associated Death Domain Protein , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/pathology , Kainic Acid , Male , Rats , Rats, Sprague-Dawley , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Seizures/chemically induced , Seizures/classification , Tyrosine 3-Monooxygenase/metabolism
18.
Eur J Neurosci ; 17(10): 2065-76, 2003 May.
Article in English | MEDLINE | ID: mdl-12786973

ABSTRACT

The consequences of activation of tumour necrosis factor receptor 1 (TNFR1) during neuronal injury remain controversial. The apoptosis signal-regulating kinase 1 (ASK1), a mitogen-activated protein kinase kinase kinase, can mediate cell death downstream of TNFR1. Presently, we examined the formation of the TNFR1 signalling cascade and response of ASK1 during seizure-induced neuronal death. Brief (40 min) seizures were induced in rats by intra-amygdala microinjection of kainic acid, which elicited unilateral hippocampal CA3 neuronal death. Seizures caused a rapid decline in the expression of the silencer of death domains protein within injured CA3. Co-immunoprecipitation analysis revealed a commensurate assembly of a TNFR1 scaffold complex containing TNFR-associated death domain protein, receptor interacting protein and TNFR-activating factor 2. In addition, recruitment of TNFR-activating factor 2 was likely promoted by Bcl10-mediated sequestering of cellular inhibitor of apoptosis protein 2. Apoptosis signal-regulating kinase 1 was sequestered in a complex that contained the molecular chaperone 14-3-3beta and protein phosphatase 5. Seizures triggered its dissociation, and the phosphorylation of the ASK1 substrates, mitogen-activated protein kinase kinase 3/6 and 4. Subsequently, protein phosphatase 5 translocated into the nuclei of degenerating CA3 neurons, while ASK1 colocalized with the adaptor proteins Daxx and TNFR-activating factor 2 at the outer membrane of injured CA3 neurons. Neutralizing antibodies to TNFalpha reduced the numbers of DNA damaged cells within the injured hippocampus. These data suggest ASK1 may be involved in the mechanism of seizure-induced neuronal death downstream of a TNFR1 death-signalling complex.


Subject(s)
Antigens, CD/metabolism , Apoptosis/physiology , Epilepsy/metabolism , MAP Kinase Kinase Kinases/metabolism , Pyramidal Cells/enzymology , Receptors, Tumor Necrosis Factor/metabolism , 14-3-3 Proteins , Animals , Antibodies/pharmacology , Carrier Proteins/metabolism , Cell Nucleus/enzymology , Epilepsy/chemically induced , Epilepsy/pathology , Fas Ligand Protein , Immunohistochemistry , MAP Kinase Kinase Kinase 5 , Male , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Proteins/metabolism , Pyramidal Cells/pathology , Rats , Rats, Sprague-Dawley , Receptors, Tumor Necrosis Factor, Type I , Signal Transduction , TNF Receptor-Associated Factor 2 , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Tyrosine 3-Monooxygenase/metabolism , Up-Regulation
19.
J Neurosci ; 22(19): 8458-65, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12351720

ABSTRACT

Bcl-2 family gene products are critical to the integration of cell death stimuli that target the mitochondrion. Proapoptotic BAD (Bcl-2-associated death protein) has been shown to dissociate from its sequestered site with the molecular chaperone protein 14-3-3 and displace proapoptotic BAX (Bcl-2-associated X protein) from antiapoptotic BCL-Xl. BAX subsequently translocates to the mitochondrion and induces cytochrome c release and caspase activation. Herein we report the response of the key members of this proposed pathway after seizures. Seizures evoked by microinjection of kainic acid into the amygdala of the rat induced unilateral CA3 pyramidal neuron death with features of apoptosis. In control hippocampus and cortex, BAD was found constitutively bound to 14-3-3, whereas BCL-Xl bound BAX. Within damaged hippocampus, seizures induced the dissociation of BAD from 14-3-3 and the subsequent dimerization of BAD with BCL-Xl as determined by immunoprecipitation and immunohistochemical colocalization. 14-3-3 was found to translocate to the nucleus of degenerating neurons, whereas BAX accumulated at mitochondrial membranes. In contrast, the primarily uninjured cortex exhibited increased phosphorylation of Akt (protein kinase B), which may phosphorylate and inhibit BAD, and no altered binding of BAD to BCL-Xl. Finally, administration of an inhibitor of phosphatidylinositol 3-kinase (LY294002), thought to be an upstream activator of Akt, exacerbated cortical apoptosis after seizures. These data suggest that seizures elicit divergent cell death and survival responses within neuronal populations and that the BAD cell death pathway may perform an instigator or reinforcement role in seizure-induced neuronal death.


Subject(s)
Carrier Proteins/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Seizures/metabolism , 14-3-3 Proteins , Active Transport, Cell Nucleus , Amygdala/drug effects , Animals , Apoptosis/drug effects , Cell Death , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , In Situ Nick-End Labeling , Kainic Acid , Male , Microinjections , Mitochondria/metabolism , Mitochondria/ultrastructure , Neurons/drug effects , Neurons/pathology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Rats , Rats, Sprague-Dawley , Seizures/chemically induced , Signal Transduction/drug effects , Tyrosine 3-Monooxygenase/metabolism , bcl-2-Associated X Protein , bcl-Associated Death Protein , bcl-X Protein
20.
J Neurosci Res ; 69(5): 614-21, 2002 Sep 01.
Article in English | MEDLINE | ID: mdl-12210827

ABSTRACT

Research into the molecular mechanisms of epileptic brain injury is hampered by the resistance of key mouse strains to seizure-induced neuronal death evoked by systemically administered excitotoxins such as kainic acid. Because C57BL/6 mice are extensively employed as the genetic background for transgenic/knockout modeling in cell death research but are seizure resistant, we sought to develop a seizure model in this strain characterized by injury to the hippocampal CA subfields. Adult male C57BL/6 mice underwent focally evoked seizures induced by intraamygdala microinjection of kainic acid. Kainic acid (KA) effectively elicited ipsilateral CA3 pyramidal neuronal death within a narrow dose range of 0.1-0.3 microg, with mortality < 10%. With employment of the most consistent (0.3 microg) dose, seizures were terminated 15, 30, 60, or 90 min after KA by diazepam. Damage was largely restricted to the ipsilateral CA3 subfield of the hippocampus, but injury was also consistent within CA1, suggesting that this mouse model better reflects the hippocampal neuropathology of human temporal lobe epilepsy than does the rat, in which CA1 is typically spared. Confirming this CA1 injury as seizure specific and not a consequence of ischemia, we used laser-Doppler flowmetry to determine that cerebral perfusion did not significantly change (97% to 118%) over control. Degenerating cells were > 95% neuronal as determined by neuron-specific nuclear protein (NeuN) counterstaining of terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeled (TUNEL) brain sections. Furthermore, TUNEL-positive cells often exhibited the morphological features of apoptosis, and small numbers were positive for cleaved caspase-3. These data establish a mouse model of focally evoked seizures in the C57BL/6 strain associated with a restricted pattern of apoptotic neurodegeneration within the hippocampal subfields that may be applied to research into the molecular basis of neuronal death after seizures.


Subject(s)
Epilepsy/physiopathology , Neurons/physiology , Analysis of Variance , Animals , Anticonvulsants/pharmacology , Cell Death/physiology , Diazepam/pharmacology , Disease Models, Animal , Electroencephalography , Epilepsy/chemically induced , Hippocampus/drug effects , Hippocampus/pathology , In Situ Nick-End Labeling , Kainic Acid/pharmacology , Laser-Doppler Flowmetry , Limbic System/physiopathology , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...