Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Neuroscience ; 284: 311-324, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25301747

ABSTRACT

Converging evidence suggests that the Parkinson's disease-linked leucine-rich repeat kinase 2 (LRRK2) modulates cellular function by regulating actin dynamics. In the present study we investigate the role of LRRK2 in functional synaptic terminals of adult LRRK2-knockout and LRRK2(R1441G)-transgenic mice as well as in primary fibroblasts of LRRK2(G2019S) mutation carriers. We show that lack of LRRK2 decreases and overexpression of mutant LRRK2 age-dependently increases the effect of the actin depolymerizing agent Latrunculin A (LatA) on the synaptic cytoskeleton. Similarly, endogenous mutant LRRK2 increases sensitivity to LatA in primary fibroblasts. Under basal conditions however, these fibroblasts show an increase in F-actin bundles and a decrease in filopodial length which can be rescued by LatA treatment. Our data suggest that LRRK2 alters actin dynamics and F-actin structure both in brain neurons and skin fibroblasts. We hypothesize that increased F-actin bundling represents a compensatory mechanism to protect F-actin from the depolymerizing effect of mutant LRRK2 under basal conditions. Our data further indicate that LRRK2-dependent changes in the cytoskeleton might have functional consequences on postsynaptic NMDA receptor localization.


Subject(s)
Actins/metabolism , Presynaptic Terminals/metabolism , Protein Serine-Threonine Kinases/metabolism , Synaptosomes/metabolism , Aging/metabolism , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cells, Cultured , Central Nervous System Agents/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphatidylserines/metabolism , Presynaptic Terminals/ultrastructure , Protein Serine-Threonine Kinases/genetics , Pseudopodia/metabolism , Pseudopodia/ultrastructure , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptosomes/ultrastructure , Thiazolidines/pharmacology
2.
Neuroscience ; 208: 41-8, 2012 Apr 19.
Article in English | MEDLINE | ID: mdl-22342962

ABSTRACT

Mutations in leucine-rich repeat kinase 2 (LRRK2) have been causally linked to neuronal cell death in Parkinson's disease. LRRK2 expression has also been detected in B lymphocytes and macrophages, suggesting a role in immune responses. In the present study, we demonstrate that LRRK2 is expressed in primary microglial cells isolated from brains of adult mice. Moreover, lipopolysaccharide (LPS)-activated microglial cells from mice overexpressing the Parkinson's disease-linked LRRK2(R1441G) mutation exhibit increased expression and secretion of proinflammatory cytokines compared with wild-type control microglia. Expression of the LPS receptor Toll-like receptor 4 (TLR4) and downstream signaling proteins did not differ between LRRK2(R1441G) transgenic microglia and wild-type controls. Consistently, conditioned medium from LPS-stimulated LRRK2(R1441G) transgenic microglia induced significant cell death when added to neuronal cultures. These findings indicate that enhanced neuroinflammation may contribute to neurodegeneration in Parkinson's disease patients carrying LRRK2 mutations.


Subject(s)
Cytokines/biosynthesis , Cytokines/genetics , Inflammation/genetics , Inflammation/metabolism , Microglia/metabolism , Mutation/physiology , Neurotoxicity Syndromes/metabolism , Parkinson Disease/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Blotting, Western , Cells, Cultured , Chemokines/metabolism , Culture Media, Conditioned , Electrophoresis, Polyacrylamide Gel , Gene Expression Profiling , Humans , Immunohistochemistry , Interferon-gamma/pharmacology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Lipopolysaccharides/pharmacology , Macrophage Activation/physiology , Mice , Mice, Transgenic , Microglia/physiology , Neurons/physiology , Neurotoxicity Syndromes/genetics , Signal Transduction/drug effects , Toll-Like Receptor 4/biosynthesis , Toll-Like Receptor 4/genetics
3.
Neuroscience ; 163(2): 533-9, 2009 Oct 06.
Article in English | MEDLINE | ID: mdl-19559761

ABSTRACT

Autosomal dominant mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of late-onset Parkinson's disease. However, the regulators/effectors contributing to the (patho-)physiological functions of LRRK2 remain poorly defined. Here we show that human LRRK2 co-purifies/co-immunoprecipitates with elongation factor 1-alpha (EF1A). Co-incubation of recombinant LRRK2 and EF1A significantly reduces the kinase activity of LRRK2, whereas its GTPase activity remains unchanged. In addition to its canonical role in mRNA translation, EF1A maintains stability of the microtubule cytoskeleton. In the present study, EF1A promotes microtubule assembly in an in vitro tubulin polymerization assay which is impaired by co-incubation with LRRK2 at sub-stoichiometric concentrations. These findings suggest that the interaction between LRRK2 and EF1A may reciprocally modulate their physiological function.


Subject(s)
Microtubules/metabolism , Peptide Elongation Factor 1/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Brain/metabolism , Cell Line , GTP Phosphohydrolases/metabolism , Humans , Hydrolysis , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Insecta , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mass Spectrometry , Parkinson Disease/metabolism , Peptide Elongation Factor 1/isolation & purification , Protein Serine-Threonine Kinases/isolation & purification , Recombinant Proteins/metabolism , Tubulin/metabolism
4.
Neuroscience ; 154(4): 1450-7, 2008 Jul 17.
Article in English | MEDLINE | ID: mdl-18541383

ABSTRACT

Aggregation of alpha-synuclein may contribute to neuropathology in Parkinson's disease patients and in transgenic animal models. Natively unfolded alpha-synuclein binds to various proteins and conformational changes due to alpha-synuclein misfolding may alter physiological interactions. In the present study, we used protein arrays spotted with 5000 recombinant human proteins for a large scale interaction analysis of monomeric versus oligomeric alpha-synuclein. Monomeric alpha-synuclein bound to arrayed cAMP regulated phosphoprotein 19 and binding appears to be disrupted by alpha-synuclein oligomerization. Incubation with recombinant alpha-synuclein oligomers lead to the identification of several GTPase activating proteins and Cdc42 effector proteins as binding partners. Protein database searches revealed a Cdc42/Rac interactive binding domain in some interactors. To demonstrate in vivo relevance, we analyzed brainstem protein extracts from alpha-synuclein(A30P) transgenic mice. Pull-down assays using beads conjugated with a Cdc42/Rac interactive binding domain lead to an enrichment of endogenous alpha-synuclein oligomers. Cdc42 effector proteins were also co-immunoprecipitated with alpha-synuclein from brainstem lysates and were colocalized with alpha-synuclein aggregates in brain sections by double immunostaining. By two-dimensional gel electrophoretic analysis of synaptosomal fractions from transgenic mouse brains we detected additional isoforms of septin 6, a downstream target of Cdc42 effector proteins. Small GTPases have recently been identified in a genetic modifier screen to suppress alpha-synuclein toxicity in yeast. Our data indicate that components of small GTPase signal transduction pathways may be directly targeted by alpha-synuclein oligomers which potentially leads to signaling deficits and neurodegeneration.


Subject(s)
Brain/metabolism , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , cdc42 GTP-Binding Protein/metabolism , Animals , Blotting, Western , Brain/pathology , Electrophoresis, Gel, Two-Dimensional , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Transgenic , Microscopy, Atomic Force , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Array Analysis , Protein Conformation , Protein Folding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , cdc42 GTP-Binding Protein/chemistry
5.
Mult Scler ; 13(7): 840-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17881397

ABSTRACT

Cerebrospinal fluid (CSF) proteins may provide important information about the pathomechanisms present in multiple sclerosis (MS). Although diagnostic criteria for early MS are available, there is still a need for biomarkers, predicting disease subtype and progression to improve individually tailored treatment. Using the two-dimensional difference gel electrophoresis (2-D-DIGE) technology for comparative analysis, we compared CSF samples from patients with MS of the relapse-remitting type (RRMS, n = 12) and from patients with clinically isolated syndrome (CIS, n = 12) suggestive of a first demyelinating attack with neurologically normal controls. Protein spots that showed more than two-fold difference between patients and controls were selected for further analysis with MALDI-TOF mass spectrometry. Immunoblot analysis was performed to confirm the validity of individual candidate proteins. In RRMS, we identified 1 up-regulated and 10 down-regulated proteins. In CIS, 2 up-regulated and 11 down-regulated proteins were identified. One of these proteins (Apolipoprotein A1) was confirmed by immunoblot. Though the pathophysiological role of these proteins still remains to be elucidated in detail and further validation is needed, these findings may have a relevant impact on the identification of disease-specific markers.


Subject(s)
Biomarkers/cerebrospinal fluid , Cerebrospinal Fluid Proteins/cerebrospinal fluid , Multiple Sclerosis, Relapsing-Remitting/cerebrospinal fluid , Proteomics , Adolescent , Adult , Cerebrospinal Fluid Proteins/analysis , Down-Regulation , Electrophoresis, Gel, Two-Dimensional , Humans , Immunoblotting , Middle Aged , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Up-Regulation
6.
Cell Mol Neurobiol ; 27(4): 505-15, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17380380

ABSTRACT

1. Alpha-synuclein is known to play an important role in the pathogenesis of Parkinson's disease (PD). The pathogenicity of alpha-synuclein is related to its ability to form intraneuronal inclusions. The inclusions, which are found in brains of patients with PD and diffuse Lewy body disease consist partially of C-terminally truncated alpha-synuclein. This alpha-synuclein species has an increased ability to form aggregates compared to full length alpha-synuclein.2. We have used an adeno-associated virus (AAV) vector system to overexpress either C-terminally truncated or full length alpha-synuclein containing the A53T mutation, which have both been identified in brains of familial PD patients and transgenic mouse models. Dissociated mesencephalic neurons, cerebellar granule neurons, and organotypic midbrain slice cultures were infected with AAV containing the transgene under the control of the cytomegalovirus promoter.3. We demonstrate that viral overexpression of alpha-synuclein(A53T) leads to the formation of distorted neurites, intraneuritic swellings, and granular perikaryal deposits in cultured neurons. Our results indicate that these cell culture models may represent an early phase of PD reflecting pathologic neuritic alterations before significant neuronal cell loss occurs.


Subject(s)
Plaque, Amyloid/genetics , Transduction, Genetic , alpha-Synuclein/genetics , Animals , Animals, Newborn , Cells, Cultured , Dependovirus/genetics , Gene Expression Regulation , Green Fluorescent Proteins/genetics , Models, Biological , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neurons/metabolism , Organ Culture Techniques , Rats , Rats, Wistar , Recombinant Fusion Proteins/genetics
7.
J Neuroimmunol ; 185(1-2): 190-4, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17367871

ABSTRACT

We used two-dimensional difference in-gel electrophoresis (2-D-DIGE) for proteome analysis of cerebrospinal fluid (CSF) in Guillain-Barré syndrome (GBS). Spots showing >2-fold difference between GBS and controls were analysed using MALDI-TOF mass spectrometry. Proteins that were up-regulated in GBS included haptoglobin, serine/threonine kinase 10, alpha-1-antitrypsin, SNC73, alpha II spectrin, IgG kappa chain and cathepsin D preprotein, while transferrin, caldesmon, GALT, human heat shock protein 70, amyloidosis patient HL-heart-peptide 127aa and transthyretin were down-regulated. Some of these proteins are reported in CSF of GBS for the first time. Accordingly, the 2-D-DIGE technology may be useful to identify disease-specific proteins in patients with GBS.


Subject(s)
Cerebrospinal Fluid/chemistry , Guillain-Barre Syndrome/cerebrospinal fluid , Proteome/analysis , Adult , Aged , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Female , Humans , Image Processing, Computer-Assisted , Male , Middle Aged , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
8.
J Pharmacol Exp Ther ; 316(1): 189-99, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16188953

ABSTRACT

Pramipexole has been shown to possess neuroprotective properties in vitro that are partly independent of its dopaminergic agonism. The site of neuroprotective action is still unknown. Using [(3)H]pramipexole, we show that the drug enters and accumulates in cells and mitochondria. Detoxification of reactive oxygen species (ROS) by pramipexole is shown in vitro and in vivo by evaluating mitochondrial ROS release and aconitase-2 activity, respectively. Pramipexole and its (+)-enantiomer SND919CL2X [low-affinity dopamine agonist; (+)2-amino-4,5,6,7-tetrahydro-6-l-propylamino-benzathiazole dihydrochloride] possess equipotent efficacy toward hydrogen peroxide and nitric oxide generated in vitro and inhibit cell death in glutathione-depleted neuroblastoma cells. IC(50) values ranged from 15 to 1000 microM, consistent with the reactivity of the respective radical and the compartmentalization of ROS generation and ROS detoxification. Finally, both compounds were tested in superoxide dismutase 1-G93A mice, a model of familial amyotrophic lateral sclerosis. SND919CL2X (100 mg/kg) prolongs survival time and preserves motor function in contrast to pramipexole (3 mg/kg), which shows an increase in running wheel activity before disease onset, presumably caused by the dopaminergic agonism. We conclude that both enantiomers, in addition to their dopaminergic activity, are able to confer neuroprotective effects by their ability to accumulate in brain, cells, and mitochondria where they detoxify ROS. However, a clinical use of pramipexole as a mitochondria-targeted antioxidant is unlikely, because the high doses needed for antioxidative action in vitro are not accessible in vivo due to dopaminergic side effects. In contrast, SND919CL2X may represent the prototype of a mitochondria-targeted neuroprotectant because it has the same antioxidative properties without causing adverse effects.


Subject(s)
Antioxidants , Dopamine Agonists/pharmacology , Neuroprotective Agents , Thiazoles/pharmacology , Aconitate Hydratase/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Benzothiazoles , Brain Chemistry/drug effects , Cell Line, Tumor , Cerebellum/cytology , Cerebellum/drug effects , Cerebellum/metabolism , Culture Media , Dopamine Agonists/pharmacokinetics , Hydrogen Peroxide/pharmacology , Hyperkinesis/chemically induced , Hyperkinesis/psychology , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Motor Activity/drug effects , Oxidants/pharmacology , Pramipexole , Stereoisomerism , Superoxide Dismutase/genetics , Superoxide Dismutase/physiology , Superoxide Dismutase-1 , Thiazoles/pharmacokinetics
9.
Pharmazie ; 60(8): 637-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16124414

ABSTRACT

A new carbazole alkaloid, named clausine Z, has been isolated from the stems and leaves of Clausena excavata Burm. (Rutaceae). Its structure was established by spectroscopic methods. The compound exhibits inhibitory activity against cyclin-dependent kinase 5 (CDK5) and shows protective effects on cerebellar granule neurons in vitro.


Subject(s)
Carbazoles/chemistry , Carbazoles/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Rutaceae/chemistry , Animals , Cell Survival/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid , Cyclin-Dependent Kinase 5 , Indicators and Reagents , Magnetic Resonance Spectroscopy , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/drug effects , Neurons/metabolism , Plant Leaves/chemistry , Plant Stems/chemistry , Rats , Spectrometry, Mass, Electrospray Ionization , Spectrophotometry, Ultraviolet
10.
Eur J Pain ; 5(2): 155-67, 2001.
Article in English | MEDLINE | ID: mdl-11465981

ABSTRACT

The response of skin to ultraviolet (UV) irradiation is an inflammation with pronounced vasodilation and hyperalgesia. Volunteers underwent UV irradiation of patches of forearm skin 3 cm in diameter. The intensity of the UV irradiation (290-320 nm) ranged between 133 mJ/cm2 and 400 mJ/cm2. Changes in skin blood flow were measured with laser Doppler technique at 3, 6, 9, 12, 24, 30, 36, 48, 60, 72, 96 and 216 h post UV irradiation. The alteration in pain threshold was measured for heat with a Peltier thermode and for pressure with a metal stylus. The effect of repeated topical pre-treatment 4 days prior to UV irradiation with 1.5 g of 0.04% capsaicin containing ointment was also investigated.UV irradiation resulted in a dose-dependent increase of skin blood flow for more than 96 h post-irradiation. There were two peaks of blood flow at 12 h and 36 h. Topical application of capsaicin prior to UV irradiation attenuated blood flow from 30 h to 45 h post UV irradiation. Enhanced blood flow was also present 5 mm outside the irradiated area (flare reaction). The control level of heat pain threshold was 44.5+/-0.7;C in normal skin. Heat pain thresholds were lowered by up to 7.761.2;C in UV-irradiated areas in a dose-dependent fashion. The control level of pressure pain threshold was 15.260.3N. Pressure pain thresholds were lowered by up to 6 N in irradiated areas. Maximal hyperalgesia coincided with the second peak of skin blood flow between 30 h and 60 h post UV irradiation. The effects of topical application of capsaicin suggests an involvement of neuropeptide mechanisms in the late phase of the human sunburn reaction.


Subject(s)
Hyperalgesia/physiopathology , Skin/blood supply , Sunburn/physiopathology , Adult , Capsaicin/pharmacology , Dose-Response Relationship, Radiation , Erythema/physiopathology , Female , Humans , Laser-Doppler Flowmetry , Male , Middle Aged , Pain Threshold/drug effects , Pain Threshold/physiology , Pain Threshold/radiation effects , Regional Blood Flow/drug effects , Regional Blood Flow/physiology , Regional Blood Flow/radiation effects , Skin/radiation effects , Ultraviolet Rays
11.
Brain Res Mol Brain Res ; 88(1-2): 54-61, 2001 Mar 31.
Article in English | MEDLINE | ID: mdl-11295231

ABSTRACT

The transcriptional factor p53 is a regulatory protein which contributes to the preservation of tissue integrity by promoting either DNA repair or apoptosis. To establish the pathophysiological role of this protein in ischemia, we produced 1 h transient middle cerebral artery (MCA) occlusion in normal and in p53-deficient mice and investigated the resulting tissue damage by multiparametric imaging. Possible genetic influences on the angioarchitecture of the MCA territory and blood flow were examined by intravascular latex infusion and laser-Doppler flowmetry. Wild-type (p53(+/+)), heterozygous (p53(+/-)) and homozygous (p53(-/-)) mice deficient for the p53 gene did not differ in respect to angioarchitecture or the effect of vascular occlusion on blood flow and general physiological parameters. Twenty-four hours after 1 h MCA occlusion, mice revealed a gene dose-dependent decline in the size of metabolic disturbances (ATP depletion and inhibition of protein synthesis) and histological injury (Cresyl Violet staining). DNA fragmentations detected by terminal deoxynucleotidyl transferase-mediated UTP nick end labeling (TUNEL) did not differ in the three groups and were only present in ATP-depleted tissue. Our findings suggest that after transient focal brain ischemia p53 prevents rather than aggravates brain injury, and that this effect is brought about by mechanisms that are unrelated to the pro-apoptotic properties of this gene.


Subject(s)
Apoptosis/physiology , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/physiopathology , Tumor Suppressor Protein p53/genetics , Adenosine Triphosphate/metabolism , Animals , Cerebrovascular Circulation/physiology , Energy Metabolism/physiology , Heterozygote , Homozygote , In Situ Nick-End Labeling , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Laser-Doppler Flowmetry , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Stroke/pathology , Stroke/physiopathology
12.
Neuroscience ; 102(4): 779-87, 2001.
Article in English | MEDLINE | ID: mdl-11182242

ABSTRACT

The temporospatial expression pattern of the nuclear DNA repair enzyme redox factor-1 (ref-1), the p53-activated gene (pag) 608 and the effector caspase-3 was examined by in situ hybridization histochemistry in gerbils subjected to two 10-min episodes of unilateral common carotid artery occlusion, separated by 5h. Gene responses were correlated with the metabolic state, as revealed by regional adenosine 5'-triphosphate bioluminescent imaging, and with the degree of histological damage, as assessed by haematoxylin-eosin staining and terminal deoxynucleotidyl transferase-mediated-dUTP nick end labeling (TUNEL), in order to evaluate the role of these genes in the maturation of injury. Focal infarcts developed in the dorsolateral cerebral cortex at the bregma level and the nucleus caudate-putamen within four days after repeated unilateral ischemia, as indicated by a secondary adenosine 5'-triphosphate loss after initial adenosine 5'-triphosphate recovery and by histomorphological signs of pannecrosis. The more caudal cortex at hippocampal levels and the hippocampus (CA1>CA3 area), however, exhibited selective neuronal injury without adenosine 5'-triphosphate depletion. TUNEL+ cells appeared starting 5h after repeated unilateral ischemia. TUNEL+ cells reached maximum levels in the caudate-putamen at 12-24h, but much later in the cortex and hippocampus at two days after ischemia. Remarkably few TUNEL+ cells were noticed in the thalamus, where adenosine 5'-triphosphate state did not recover after reperfusion. Following repeated unilateral ischemia, a transient elevation of ref-1 mRNA was detected after 5h in the cerebral cortex and hippocampal CA1 area. Ref-1 mRNA levels decreased within 12-24h, before the onset of tissue damage. Subsequently, pag608 and caspase-3 mRNA levels increased, closely in parallel with the appearance of DNA fragmented cells, but slightly prior to the deterioration of adenosine 5'-triphosphate state. In the caudate-putamen, pag608 and caspase-3 mRNAs reached maximum levels already 12-24h after repeated common carotid artery occlusion, when DNA fragmentation was most prominent, and declined thereafter. In the cortex and hippocampal CA1-3 areas, where DNA damage appeared more slowly, pag608 and caspase-3 mRNAs were induced starting 24h after ischemia, and remained elevated even after two to four days. The levels of pag608 and caspase-3 mRNAs were similar at rostral and caudal levels of the cortex, as well as in the hippocampal CA1 and CA3 area, although the degree of injury differed considerably between these structures. Notably, pag608 and caspase-3 mRNAs were not elevated in the thalamus after repeated unilateral ischemia. The present report shows a close temporal association between the induction of ref-1, pag608 and caspase-3 mRNAs, the manifestation of cell injury and the secondary adenosine 5'-triphosphate depletion in infarcting brain areas, suggesting (i) that de novo responses of these genes may be involved in the maturation of cell injury and (ii) that apoptotic programs and the secondary deterioration of cerebral energy state may interfere with each other after ischemia.


Subject(s)
Brain Ischemia/pathology , Brain Ischemia/physiopathology , Carbon-Oxygen Lyases/genetics , Caspases/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase , DNA-Binding Proteins/genetics , Energy Metabolism/physiology , Nuclear Proteins/genetics , Adenosine Triphosphate/metabolism , Animals , Arterial Occlusive Diseases/metabolism , Arterial Occlusive Diseases/pathology , Arterial Occlusive Diseases/physiopathology , Brain/blood supply , Brain/enzymology , Brain/physiopathology , Brain Ischemia/metabolism , Carotid Artery, Common , Caspase 3 , Cerebral Infarction/metabolism , Cerebral Infarction/pathology , Cerebral Infarction/physiopathology , Coloring Agents , DNA Fragmentation , DNA Repair , Eosine Yellowish-(YS) , Fluorescent Dyes , Gene Expression/physiology , Gerbillinae , Hematoxylin , In Situ Hybridization , In Situ Nick-End Labeling , Luminescent Measurements , RNA, Messenger/analysis
14.
Brain Res Mol Brain Res ; 73(1-2): 138-43, 1999 Nov 10.
Article in English | MEDLINE | ID: mdl-10581407

ABSTRACT

Persistent activation of c-Jun N-terminal kinases (JNKs) and phosphorylation of c-Jun has been shown in various cell death paradigms. Inhibition of the JNK signal transduction pathway prevented neuronal cell death both in vitro and in vivo. In the present study, nuclear phospho-c-Jun immunoreactivity became apparent selectively in vulnerable hippocampal CA1 neurons at 24 h after transient global cerebral ischemia. A high constitutive expression of phospho-JNK1 was detected by immunoblot analysis of hippocampal extracts. Expression of JNK interacting protein-1 (JIP-1), which facilitates JNK signaling, remained unchanged in post-ischemic hippocampal neurons. By contrast, p53-activated gene 608 (PAG608), which promotes cell death in vitro, was strongly induced in post-ischemic CA1 neurons. Our data suggest that transcription factors p53 and phospho-c-Jun may contribute to programmed CA1 cell death following ischemia.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Death/genetics , DNA-Binding Proteins/genetics , Hippocampus/metabolism , Ischemic Attack, Transient/physiopathology , Neurons/metabolism , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-jun/genetics , Animals , Brain/metabolism , Carrier Proteins/metabolism , DNA-Binding Proteins/analysis , Gene Expression Regulation , Hippocampus/chemistry , Hippocampus/cytology , Immunohistochemistry , In Situ Hybridization , Male , Nuclear Proteins/analysis , Proto-Oncogene Proteins c-jun/analysis , RNA, Messenger/genetics , RNA-Binding Proteins , Rats , Rats, Wistar
15.
Metab Brain Dis ; 14(2): 117-24, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10488913

ABSTRACT

Neuronal death after brain ischemia is mainly due to necrosis but there is also evidence for involvement of apoptosis. To test the importance of apoptosis, we investigated the effect of targeted disruption of the apoptosis-suppressive gene bcl-2 on the severity of ischemic brain injury. Transient focal ischemia for 1 hour was induced by occlusion of the middle cerebral artery in homozygous (n=7) and heterozygous (n=6) bcl-2 knockout mice as well as in their wildtype littermates (n=5). Bcl-2 ablation did not influence cerebral blood flow but it significantly increased infarct size and neurological deficit score at 1 day after reperfusion in a gene-dose dependent manner. The exacerbation of tissue damage in the absence of Bcl-2 underscores the importance of apoptotic pathways for the manifestation of ischemic injury after transient vascular occlusion.


Subject(s)
Brain Ischemia/genetics , Brain Ischemia/pathology , Brain/pathology , Genes, bcl-2 , Mice, Knockout/genetics , Mice, Knockout/physiology , Animals , Apoptosis/physiology , Brain Ischemia/complications , Brain Ischemia/physiopathology , Cerebral Cortex/blood supply , Cerebral Infarction/pathology , Cerebrovascular Circulation , Female , Male , Mice , Nervous System Diseases/etiology
16.
Neuroscience ; 93(4): 1219-22, 1999.
Article in English | MEDLINE | ID: mdl-10501444

ABSTRACT

An essential role for caspases in programmed neuronal cell death has been demonstrated in various in vitro studies, and synthetic caspase inhibitors have recently been shown to prevent neuronal cell loss in animal models of focal cerebral ischemia and traumatic brain injury, respectively. The therapeutic utility of caspase inhibitors, however, will depend on preservation of both structural and functional integrity of neurons under stressful conditions. The present study demonstrates that expression and proteolytic activity of caspase-3 is up-regulated in the rat hippocampus after transient forebrain ischemia. Continuous i.c.v. infusion of the caspase inhibitor N-benzyloxycarbonyl-Asp(OMe)-Glu(OMe)-Val-Asp(OMe)-fluoromethyl ketone significantly attenuated caspase-3-like enzymatic activity, and blocked delayed cell loss of hippocampal CA1 neurons after ischemia. Administration of N-benzyloxycarbonyl-Asp(OMe)-Glu(OMe)-Val-Asp(OMe)-fluoromethyl ketone, however, did not prevent impairment of induction of long-term potentiation in post-ischemic CA1 cells, suggesting that caspase inhibition alone does not preserve neuronal functional plasticity.


Subject(s)
Apoptosis/drug effects , Caspase Inhibitors , Hippocampus/cytology , Ischemic Attack, Transient/physiopathology , Long-Term Potentiation/physiology , Animals , Caspase 3 , Caspases/analysis , Cysteine Proteinase Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic , Hippocampus/blood supply , Hippocampus/enzymology , In Situ Nick-End Labeling , Ischemic Attack, Transient/drug therapy , Male , Neurons/cytology , Neurons/drug effects , Neurons/enzymology , Oligopeptides/pharmacology , Proteins/genetics , RNA, Messenger/analysis , Rats , Rats, Wistar , X-Linked Inhibitor of Apoptosis Protein
17.
Cell Death Differ ; 6(7): 673-82, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10453078

ABSTRACT

Following transection of the optic nerve (ON), retinal ganglion cells (RGCs) upregulate Bax protein expression and undergo apoptosis. The present study aimed at reducing Bax expression in order to test whether Bax plays a causative role in the induction of secondary RGC apoptosis. Following injection into the vitreous, fluoresceinated oligonucleotides transfected RGCs in vivo at the injection site in the temporal superior retina. Following ON lesion, and repeated injections of a partially phosphorothioated Bax antisense oligonucleotide, but not following injection of control oligonucleotides, expression of Bax protein was locally inhibited, and the number of surviving RGCs was increased in Bax antisense treated rats 8 days after axotomy. Our results indicate that Bax induction is a prerequisite for the execution of RGC apoptosis following ON axotomy. While the Bax antisense strategy offers an exciting perspective to inhibit secondary neuronal degeneration in vivo, both limited transfection efficacy, and the temporal restriction of this effect currently limit the use of this approach with respect to clinical applications for the treatment of neurodegeneration.


Subject(s)
Apoptosis/drug effects , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Base Sequence , Cell Survival , Female , Gene Expression , Nerve Degeneration/prevention & control , Oligonucleotides, Antisense/genetics , Optic Nerve Injuries , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/drug effects , Time Factors , Transfection , bcl-2-Associated X Protein
18.
Br J Ophthalmol ; 83(2): 225-30, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10396203

ABSTRACT

BACKGROUND/AIMS: Ultraviolet (UV) irradiation of mammalian cells in culture evokes the transcriptional activation of different proto-oncogenes, among them members of the fos/jun family which are known to play an important role in cell proliferation and differentiation. To investigate in vivo UV induced proto-oncogene expression of irradiated ocular cells, the expression of JunB, JunD, and Egr-1 was analysed in the cornea, lens, and retina. Furthermore, UV radiation is known to induce pleiotrophic events in irradiated cells which include growth arrest, inflammation, and even cell death. In order to determine the type of cell death--for example, apoptosis versus necrosis, sections of UV irradiated rat eyes were further examined for distinct ultrastructural morphology of cell death and DNA fragmentation. METHODS: Eyes of anaesthetised rats were exposed to 1.5 J/cm2 of ultraviolet radiation (280-380 nm). Animals were perfused 6 and 16 hours after irradiation and tissue sections of enucleated bulbi were processed for light and electron microscopy. RESULTS: Under control conditions, Jun B was constitutively expressed in numerous superficial cells but also in scattered basal cells of the corneal epithelium. After UV exposure JunB expression was massively upregulated in many cells of the basal cell layers of the corneal epithelium, although during the entire experiment, both the corneal stroma and endothelium were JunB negative. In contrast, Egr-1 was expressed exclusively in lens epithelium showing only a faint expression pattern under control conditions. However, Egr-1 expression increased after UV exposure, so that many Egr-1 positive cells of the lens epithelium could be found several hours after UV illumination. JunD was expressed in single cells of both the ganglion cell layer and the inner nuclear layer of the retina, a pattern of expression which did not change after UV exposure. Regarding the type of cell death, features of apoptosis were only occasionally present in scattered superficial cells of the corneal epithelium of control eyes. After UV exposure, however, morphological signs of apoptosis and TUNEL positive cells were visible both in the stroma and epithelium of the rat cornea. In contrast, UV irradiated lens epithelial cells exhibited features typical of necrosis. The corneal endothelium and the retina did not show any indications of morphological changes indicative of cell death after UV irradiation. CONCLUSION: Each proto-oncogene encoded protein was found to be expressed in a tissue specific manner and UV irradiation differentially modulates the expression pattern of these transcriptional regulatory proteins. This temporospatial expression pattern of these proteins is accompanied by two morphologically distinct types of cell death in the cornea and lens after UV irradiation.


Subject(s)
Eye/radiation effects , Gene Expression Regulation/radiation effects , Proto-Oncogenes/radiation effects , Ultraviolet Rays , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Epithelium, Corneal/metabolism , Epithelium, Corneal/radiation effects , Immunohistochemistry , Lens, Crystalline/metabolism , Lens, Crystalline/radiation effects , Male , Proto-Oncogenes/physiology , Rats , Rats, Sprague-Dawley
19.
Brain Res Mol Brain Res ; 71(1): 61-8, 1999 Jul 23.
Article in English | MEDLINE | ID: mdl-10407187

ABSTRACT

The occurrence of radiation-induced apoptosis in normal brain was investigated using an animal model of radiosurgery. Adult male Fischer rats aged 3 to 4 months were subjected to single dose convergent beam irradiation (10 Gy). Apoptotic cell death was determined by in situ labeling of DNA nick ends (TUNEL) and light microscopic evaluation of cell morphology. Five hours after irradiation, a highly significant increase of apoptotic cells in the subgranular zone of the dentate gyrus was paralleled by a corresponding significant decrease of cells immunoreactive for the proliferation marker Ki-67. Morphology, location and distribution of cells affected by radiation-induced apoptosis in the dentate gyrus subgranular zone, together with NeuN-immunohistochemistry, support the contention that these cells belong to the immature progenitor population responsible for neurogenesis in the adult rat hippocampus.


Subject(s)
Apoptosis/radiation effects , Dentate Gyrus/radiation effects , Stem Cells/radiation effects , Animals , Dentate Gyrus/cytology , Dose-Response Relationship, Radiation , Immunohistochemistry , In Situ Nick-End Labeling , Ki-67 Antigen/analysis , Male , Particle Accelerators , Photons , Radiosurgery , Rats , Rats, Inbred F344 , Stem Cells/cytology , X-Rays
20.
J Neurosci Res ; 56(1): 21-7, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10213471

ABSTRACT

The expression of brain-derived neurotrophic factor (BDNF) and its receptor tyrosine kinase B are both increased after global ischemia. Therefore, a protective action of BDNF against the delayed degeneration of vulnerable neurons has been suggested. We have investigated the neuroprotective action of BDNF in global ischemia induced by a four-vessel occlusion in the rat. Following reperfusion, 0.06 microg/hr BDNF was continuously administered intracerebroventricularly with an osmotic minipump. Rats were sacrificed up to 7 days after ischemia and neuronal degeneration was identified by terminal transferase and biotin-dUTP nick end labeling (TUNEL) staining. Additionally, the glial reaction was investigated immunohistochemically and by measuring the activation of immunological nitric oxide synthase protein expression. Postischemic intracerebroventricular infusion of BDNF prevented neuronal death in the vulnerable CA1 region of the hippocampus. Additionally, astroglial activation and macrophage infiltration, which were observed in association with neuronal death, were inhibited by BDNF. This was paralleled by an inhibition of inducible nitric oxide synthase (iNOS) expression in the hippocampus. Thus, the observed neuroprotective effects of continuous BDNF administration after reperfusion suggest a therapeutic potential for BDNF in cerebral ischemia.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/physiopathology , Neuroglia/physiology , Neurons/physiology , Animals , Apoptosis/drug effects , DNA Fragmentation , In Situ Nick-End Labeling , Male , Nerve Degeneration/prevention & control , Neuroglia/drug effects , Neuroglia/pathology , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase Type II , Rats , Rats, Wistar , Reperfusion
SELECTION OF CITATIONS
SEARCH DETAIL
...