Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
eNeuro ; 7(1)2020.
Article in English | MEDLINE | ID: mdl-31882533

ABSTRACT

While BDNF is receiving considerable attention for its role in synaptic plasticity and in nervous system dysfunction, identifying brain circuits involving BDNF-expressing neurons has been challenging. BDNF levels are very low in most brain areas, except for the large mossy fiber terminals in the hippocampus where BDNF accumulates at readily detectable levels. This report describes the generation of a mouse line allowing the detection of single brain cells synthesizing BDNF. A bicistronic construct encoding BDNF tagged with a P2A sequence preceding GFP allows the translation of BDNF and GFP as separate proteins. Following its validation with transfected cells, this construct was used to replace the endogenous Bdnf gene. Viable and fertile homozygote animals were generated, with the GFP signal marking neuronal cell bodies translating the Bdnf mRNA. Importantly, the distribution of immunoreactive BDNF remained unchanged, as exemplified by its accumulation in mossy fiber terminals in the transgenic animals. GFP-labeled neurons could be readily visualized in distinct layers in the cerebral cortex where BDNF has been difficult to detect with currently available reagents. In the hippocampal formation, quantification of the GFP signal revealed that <10% of the neurons do not translate the Bdnf mRNA at detectable levels, with the highest proportion of strongly labeled neurons found in CA3.


Subject(s)
Brain-Derived Neurotrophic Factor , Hippocampus , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/metabolism , Mice , Neuronal Plasticity , Neurons/metabolism
2.
Psychopharmacology (Berl) ; 236(12): 3451-3463, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31267156

ABSTRACT

RATIONALE: The N-methyl-D-aspartate receptor (NMDAR) antagonist ketamine is known to have not only a rapid antidepressant effect but also dissociative side effects. Traxoprodil and lanicemine, also NMDA antagonists, are candidate antidepressant drugs with fewer side effects. OBJECTIVES: In order to understand their mechanism of action, we investigated the acute effects of traxoprodil and lanicemine on brain connectivity using resting-state functional magnetic resonance imaging (rs-fMRI). METHODS: Functional connectivity (FC) alterations were examined using interregional correlation networks. Graph theoretical methods were used for whole brain network analysis. As interest in NMDAR antagonists as potential antidepressants was triggered by the antidepressant effect of ketamine, results were compared to previous findings from our ketamine studies. RESULTS: Similar to ketamine but to a smaller extent, traxoprodil increased hippocampal-prefrontal (Hc-PFC) coupling. Unlike ketamine, traxoprodil decreased connectivity within the PFC. Lanicemine had no effect on these properties. The improvement of Hc-PFC coupling corresponds well to clinical result, showing ketamine to have a greater antidepressant effect than traxoprodil, while lanicemine has a weak and transient effect. Connectivity changes overlapping between the drugs as well as alterations of local network properties occurred mostly in reward-related regions. CONCLUSION: The antidepressant effect of NMDA antagonists appears to be associated with enhanced Hc-PFC coupling. The effects on local network properties and regional connectivity suggest that improvement of reward processing might also be important for understanding the mechanisms underlying the antidepressant effects of these drugs.


Subject(s)
Hippocampus/drug effects , Phenethylamines/pharmacology , Piperidines/pharmacology , Prefrontal Cortex/drug effects , Pyridines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Reward , Animals , Antidepressive Agents/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/diagnostic imaging , Hippocampus/physiology , Magnetic Resonance Imaging/methods , Male , Prefrontal Cortex/diagnostic imaging , Prefrontal Cortex/physiology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/physiology
3.
J Alzheimers Dis ; 44(2): 649-60, 2015.
Article in English | MEDLINE | ID: mdl-25322927

ABSTRACT

BACKGROUND: Specific mechanisms behind the role of oxidative/nitrosative stress and mitochondrial dysfunction in Alzheimer's disease (AD) pathogenesis remain elusive. Mitochondrial aconitase (ACO2) is a Krebs cycle enzyme sensitive to free radical-mediated damage. OBJECTIVE: We assessed activity and expression of ACO2 extracted from blood lymphocytes of subjects with AD, mild cognitive impairment (MCI), older adults with normal cognition (OCN, age ≥65 years), and younger adults with normal cognition (YCN, age <65 years). Plasma levels and activities of antioxidants were also measured. METHODS: Blood samples were collected from 28 subjects with AD, 22 with MCI, 21 OCN, and 19 YCN. ACO2 activity was evaluated in a subsample before and after in vitro exposure to free radicals. RESULTS: ACO2 activity was significantly lower in AD and MCI cases than controls: ACO2 median activity was 0.64 ± 0.21 U/mg protein for AD, 0.93 ± 0.28 U/mg protein for MCI, 1.17 ± 0.78 U/mg protein for OCN subjects, and 1.23 ± 0.43 U/mg protein for YCN individuals. In subjects with AD and MCI, ACO2 expression was lower than OCN subjects, and ACO2 activity correlated with vitamin E plasma levels (rho: 0.64, p < 0.001) and Mini-Mental State Examination total score (rho: 0.82, p < 0.001). Furthermore, free radicals exposure reduced ACO2 activity more in individuals with AD than in OCN subjects. CONCLUSION: Our results suggest that ACO2 activity is reduced in peripheral lymphocytes of subjects with AD and MCI and correlates with antioxidant protection. Further studies are warranted to verify the role of ACO2 in AD pathogenesis and its importance as a marker of AD progression.


Subject(s)
Aconitate Hydratase/blood , Alzheimer Disease/blood , Cognitive Dysfunction/blood , Lymphocytes/metabolism , Aged , Biomarkers/blood , Blotting, Western , Disease Progression , Female , Free Radicals/metabolism , Humans , Male , Mental Status Schedule , Mitochondria/metabolism , Polymerase Chain Reaction , RNA, Messenger/metabolism , Vitamin E/blood
4.
Brain Res Bull ; 96: 28-38, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23639920

ABSTRACT

As nicotinic acetylcholine receptor (nAChR) agonists directly address cholinergic neurotransmission with potential impact on glutamatergic function, they are considered as potential new symptomatic treatment options for Alzheimer's disease compared to the indirectly operating acetylcholinesterase inhibitors such as the current gold standard donepezil. In order to evaluate the therapeutic value of nAChR activation to ameliorate cognitive dysfunction, a direct comparison between α4ß2, α7 nAChR agonists, and donepezil was performed on the level of an ex vivo experimental model of impaired memory formation. First, we demonstrated that amyloid beta (Aß)42 oligomers, which are believed to be the synaptotoxic Aß-species causally involved in the pathophysiology of Alzheimer's disease, have a detrimental effect on long-term potentiation (LTP) in the CA1 region of rat hippocampal slices, a widely used cellular model of learning and memory. Second, we investigated the potential of donepezil, the α4ß2 nAChR agonist TC-1827 and the α7 nAChR partial agonist SSR180711 to reverse Aß42 oligomer induced LTP impairment. Donepezil showed only a slight reversal of Aß42 oligomer induced impairment of early LTP, and had no effect on Aß42 oligomer induced impairment of late LTP. The same was demonstrated for the α4ß2 nAChR agonist TC-1827. In contrast, the α7 nAChR partial agonist SSR180711 completely rescued early as well as late LTP impaired by Aß42 oligomers. As activating α7 nAChRs was found to be most efficacious in restoring Aß42 oligomer induced LTP deficits, targeting α7 nAChRs might represent a powerful alternative approach for symptomatic treatment of AD.


Subject(s)
Amyloid beta-Peptides/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cholinesterase Inhibitors/pharmacology , Indans/pharmacology , Long-Term Potentiation/drug effects , Nicotinic Agonists/pharmacology , Piperidines/pharmacology , Acetylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Donepezil , Hippocampus/drug effects , Long-Term Potentiation/physiology , Male , Rats , Rats, Wistar , Receptors, Nicotinic/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism
5.
Biochemistry ; 52(8): 1466-76, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23374097

ABSTRACT

In Alzheimer's disease, substantial evidence indicates the causative role of soluble amyloid ß (Aß) aggregates. Although a variety of Aß assemblies have been described, the debate about their individual relevance is still ongoing. One critical issue hampering this debate is the use of different methods for the characterization of endogenous and synthetic peptide and their intrinsic limitations for distinguishing Aß aggregates. Here, we used different protocols for the establishment of prefibrillar Aß assemblies with varying morphologies and sizes and compared them in a head-to-head fashion. Aggregation was characterized via the monomeric peptide over time until spheroidal, protofibrillar, or fibrillar Aß aggregates were predominant. It could be shown that a change in the ionic environment induced a structural rearrangement, which consequently confounds the delineation of a measured neurotoxicity toward a distinct Aß assembly. Here, neuronal binding and hippocampal neurotransmission were found to be suitable to account for the synaptotoxicity to different Aß assemblies, based on the stability of the applied Aß aggregates in these settings. In contrast to monomeric or fibrillar Aß, different prefibrillar Aß aggregates targeted neurons and impaired hippocampal neurotransmission with nanomolar potency, albeit by different modalities. Spheroidal Aß aggregates inhibited NMDAR-dependent long-term potentiation, as opposed to protofibrillar Aß aggregates, which inhibited AMPAR-dominated basal neurotransmission. In addition, a provoked structural conversion of spheroidal to protofibrillar Aß assemblies resulted in a time-dependent suppression of basal neurotransmission, indicative of a mechanistic switch in synaptic impairment. Thus, we emphasize the importance of addressing the metastability of prefacto characterized Aß aggregates in assigning a biological effect.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Hippocampus/physiopathology , Neurons/pathology , Synaptic Transmission , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/chemistry , Animals , Cells, Cultured , Hippocampus/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Rats , Rats, Wistar
6.
J Neurol Sci ; 295(1-2): 46-52, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20570288

ABSTRACT

Point mutations in the mitochondrial genome are associated with a variety of metabolic disorders. The myopathy, encephalopathy, lactic acidosis, stroke-like episodes syndrome (MELAS), is most frequently associated with an A to G transition at position 3243 of the mitochondrial tRNA(Leu(UUR)) gene, and is characterized by biochemical and structural alterations of mitochondria. In the present study, we analyzed proteomic changes in an immortalized B-cell line harbouring the MELAS A3243G mutation by two-dimensional difference gel electrophoresis and immunoblot analysis. Although the cell line contained only 10% mutated mitochondrial genomes, we detected significant alterations in numerous proteins associated with the actin cytoskeleton and in nuclear-encoded subunits of mitochondrial respiratory chain complexes. Notably, mitochondrial Lon protease and Hsp60 were deregulated in MELAS cells, indicating an effect on the mitochondrial protein quality control system. By immunofluorescence microscopy, we detected mitochondrial Lon protease accumulation and changes in actin-binding proteins preferentially in MELAS cells containing numerous mitochondria with mutated genomes. Enzymatic assays revealed that Lon protease activity is increased in MELAS cell lysates. Although Lon protease has been shown to degrade misfolded proteins and to stabilize respiratory chain complexes within mitochondria, our MELAS cell line exhibited a higher sensitivity to mitochondrial stress. These findings provide novel insights into the cellular response to dysfunctional mitochondria containing mutated genomes.


Subject(s)
Actins/metabolism , Cytoskeleton/pathology , DNA, Mitochondrial/genetics , Lymphocytes/metabolism , MELAS Syndrome/pathology , Mitochondrial Proteins/metabolism , Mutation/genetics , Actins/genetics , Cytoskeleton/genetics , Electrophoresis, Gel, Two-Dimensional/methods , Humans , MELAS Syndrome/genetics , Mass Spectrometry/methods , Membrane Potential, Mitochondrial/genetics , Mitochondrial Proteins/genetics , Proteome/metabolism
7.
Proteomics ; 7(4): 605-616, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17309106

ABSTRACT

Synaptic dysfunction is an early event in Alzheimer's disease patients and has also been detected in transgenic mouse models. In the present study, we analyzed proteomic changes in synaptosomal fractions from Tg2576 mice that overexpress mutant human amyloid precursor protein (K670N, M671L) and from their nontransgenic littermates. Cortical and hippocampal tissue was microdissected at the onset of cognitive impairment, but before deposition of amyloid plaques. Crude synaptosomal fractions were prepared by differential centrifugation, proteins were separated by 2-D DIGE and identified by MS/MS. Significant alterations were detected in mitochondrial heat shock protein 70 pointing to a mitochondrial stress response. Subsequently, synaptosomal versus nonsynaptic mitochondria were purified from Tg2576 mice brains by density gradient centrifugation. Mitochondrial proteins were separated by IEF or Blue-native gel electrophoresis in the first dimension and SDS-PAGE in the second dimension. Numerous changes in the protein subunit composition of the respiratory chain complexes I and III were identified. Levels of corresponding mRNAs remain unchanged as shown by Affymetrix oligonucleotide array analysis. Functional examination revealed impaired state 3 respiration and uncoupled respiration in brain mitochondria from young Tg2576 mice. By immunoblotting, amyloid-beta oligomers were detected in synaptosomal fractions from Tg2576 mice and reduced glucose metabolism was observed in Tg2576 mice brains by [14C]-2-deoxyglucose infusion. Taken together, we demonstrate alterations in the mitochondrial proteome and function that occur in Tg2576 mice brains before amyloid plaque deposition suggesting that mitochondria are early targets of amyloid-beta aggregates.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Amyloid/metabolism , Brain/metabolism , Mitochondria/metabolism , Proteome/metabolism , Synaptosomes/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Brain/pathology , Carbon Radioisotopes/chemistry , Glucose/metabolism , Humans , Mice , Mice, Transgenic , Tandem Mass Spectrometry
8.
Proc Natl Acad Sci U S A ; 103(20): 7607-12, 2006 May 16.
Article in English | MEDLINE | ID: mdl-16682634

ABSTRACT

NADH:ubiquinone oxidoreductase (complex I) is a major source of reactive oxygen species in mitochondria and a significant contributor to cellular oxidative stress. Here, we describe the kinetic and molecular mechanism of superoxide production by complex I isolated from bovine heart mitochondria and confirm that it produces predominantly superoxide, not hydrogen peroxide. Redox titrations and electron paramagnetic resonance spectroscopy exclude the iron-sulfur clusters and flavin radical as the source of superoxide, and, in the absence of a proton motive force, superoxide formation is not enhanced during turnover. Therefore, superoxide is formed by the transfer of one electron from fully reduced flavin to O2. The resulting flavin radical is unstable, so the remaining electron is probably redistributed to the iron-sulfur centers. The rate of superoxide production is determined by a bimolecular reaction between O2 and reduced flavin in an empty active site. The proportion of the flavin that is thus competent for reaction is set by a preequilibrium, determined by the dissociation constants of NADH and NAD+, and the reduction potentials of the flavin and NAD+. Consequently, the ratio and concentrations of NADH and NAD+ determine the rate of superoxide formation. This result clearly links our mechanism for the isolated enzyme to studies on intact mitochondria, in which superoxide production is enhanced when the NAD+ pool is reduced. Therefore, our mechanism forms a foundation for formulating causative connections between complex I defects and pathological effects.


Subject(s)
Electron Transport Complex I/metabolism , Mitochondria, Heart/metabolism , Superoxides/metabolism , Animals , Cattle , Electrons , Flavins/chemistry , Flavins/metabolism , Humans , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Mitochondria, Heart/chemistry , NAD/chemistry , NAD/metabolism , Oxidants/chemistry , Oxidants/metabolism , Oxidation-Reduction , Oxygen/chemistry , Oxygen/metabolism
9.
Mol Cell Neurosci ; 24(2): 489-502, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14572469

ABSTRACT

Previous studies suggested that pro-apoptotic stimuli may trigger a fatal reactivation of cell cycle elements in postmitotic neurons. Supporting this hypothesis, small molecule inhibitors of cyclin-dependent kinases (CDKs), which are known primarily as cell cycle regulators, are neuroprotective. However, available CDK inhibitors cannot discriminate between the different members of the CDK family and inhibit also CDK5, which is not involved in cell cycle control. Testing a new class of CDK inhibitors, we find that inhibitory activity against CDK5, but not cell cycle-relevant CDKs, confers neuroprotection. Moreover, we demonstrate that cleavage of the CDK5 activator protein p35 to p25 is associated with CDK5 overactivation after focal cerebral ischemia, but not in other models used in this study. We find that blocking CDK5 activity, but not caspase inhibition, protects mitochondrial integrity of lesioned neurons. Thus, in our models, CDK5, rather than cell cycle-relevant CDKs, activates neuronal cell death pathways upstream of mitochondrial dysfunction, and inhibition of CDK5 may promote functional long-term rescue of injured neurons. Moreover, we present the first CDK5-selective small molecule inhibitor, lacking unwanted cytostatic effects due to cross-inhibition of mitotic CDKs.


Subject(s)
Apoptosis/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Mitochondria/drug effects , Neurons/drug effects , Animals , Apoptosis/physiology , Brain Ischemia/drug therapy , Brain Ischemia/enzymology , Buthionine Sulfoximine/pharmacology , Buthionine Sulfoximine/therapeutic use , Cell Death/drug effects , Cell Death/physiology , Cyclin-Dependent Kinase 5 , Cyclin-Dependent Kinases/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/therapeutic use , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mitochondria/enzymology , Necrosis , Neurons/enzymology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...