Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Prog Mol Biol Transl Sci ; 127: 183-210, 2014.
Article in English | MEDLINE | ID: mdl-25149218

ABSTRACT

The etiology of common, nonfamiliar late-onset Alzheimer's disease (LOAD) is only partly understood and seems to be extremely complex including many genetic and environmental factors. The most important environmental risk factor to develop LOAD is aging itself. Aging and LOAD are considered to be strongly linked to mitochondrial dysfunction and enhanced oxidative stress. In this review, we focus on the interaction between mitochondrial dysfunction in aging especially on defects of the respiratory chain of the oxidative phosphorylation system resulting in enhanced oxidative stress and the interplay between aging-associated mitochondrial defects and LOAD-associated mitochondrial failure. The deleterious effects of the two hallmarks of LOAD, amyloid beta, and hyperphosphorylated tau, on mitochondrial function, movement, and morphology are described as well as the toxic effects of the most relevant genetic risk factor of LOAD, the apolipoprotein E4 allele. Finally, the review provides an overview about drugs and nutritional ingredients which improve mitochondrial function or/and act as antioxidants and discusses their potential role in the treatment of LOAD.


Subject(s)
Alzheimer Disease/pathology , Mitochondria/pathology , Aging/pathology , Animals , Antioxidants/metabolism , Brain/pathology , Humans , Oxidative Phosphorylation
2.
J Alzheimers Dis ; 42(2): 395-411, 2014.
Article in English | MEDLINE | ID: mdl-24898661

ABSTRACT

Recent data suggest that the combined effect of oxidative stress due to aging and slightly elevated amyloid-ß (Aß) levels initiate Alzheimer's disease (AD) long before the clinical onset. Investigations of this early phase are hampered by the lack of cellular or animal models reflecting this scenario. We used SH-SY5Y cells stably transfected with an additional copy of the human AßPP gene and artificial aging by complex I inhibition. These cells show slightly elevated Aß levels, moderately decreased ATP levels, impaired mitochondrial membrane potential, and decreased mitochondrial respiration. Assessing mitochondrial dynamics with three different methods reveals a distinct shift toward mitochondrial fission and fragmentation in SH-SY5Y AßPPwt cells. We also performed electron cryo-tomography of isolated mitochondria to reveal that there were no major differences between SH-SY5Y control and SH-SY5Y AßPPwt mitochondria with respect to swelling or loss of cristae. Dystrophic neurites are an early pathological feature of AD. Interestingly, SH-SY5Y AßPPwt cells exhibit significantly longer neurites, likely due to substantially elevated levels of sAßPPα. Complex I inhibition also shows substantial effects on mitochondrial dynamics, impairs neuritogenesis, and elevates Aß levels in both cell types. In SH-SY5Y AßPPwt cells, these defects were more pronounced due to a relatively elevated Aß and a reduced sAßPPα production. Our findings suggest that the progression from low Aß levels to the beginning of AD takes place in the presence of oxidative stress during normal aging. This mechanism not only results from additive effects of both mechanisms on mitochondrial function but might also be additionally aggravated by altered amyloidogenic processing.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Models, Biological , Adenosine Triphosphate/metabolism , Cell Line, Tumor , Electron Transport Complex IV/metabolism , Humans , Membrane Potential, Mitochondrial/physiology , Microscopy, Electron , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Neuroblastoma/pathology , Transfection
3.
J Med Chem ; 57(11): 4861-75, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24831693

ABSTRACT

Dopamine D2 receptor-promoted activation of Gα(o) over Gα(i) may increase synaptic plasticity and thereby might improve negative symptoms of schizophrenia. Heterocyclic dopamine surrogates comprising a pyrazolo[1,5-a]pyridine moiety were synthesized and investigated for their binding properties when low- to subnanomolar K(i) values were determined for D(2L), D(2S), and D3 receptors. Measurement of [(35)S]GTPγS incorporation at D(2S) coexpressed with G-protein subunits indicated significant bias for promotion of Gα(o1) over Gα(i2) coupling for several test compounds. Functionally selective D(2S) activation was most striking for the carbaldoxime 8b (Gα(o1), pEC50 = 8.87, E(max) = 65%; Gα(i2), pEC50 = 6.63, E(max) = 27%). In contrast, the investigated 1,4-disubstituted aromatic piperazines (1,4-DAPs) behaved as antagonists for ß-arrestin-2 recruitment, implying significant ligand bias for G-protein activation over ß-arrestin-2 recruitment at D(2S) receptors. Ligand efficacy and selectivity between D(2S) and D3 activation were strongly influenced by regiochemistry and the nature of functional groups attached to the pyrazolo[1,5-a]pyridine moiety.


Subject(s)
Antipsychotic Agents/chemical synthesis , Heterocyclic Compounds, 2-Ring/chemical synthesis , Oximes/chemical synthesis , Piperazines/chemical synthesis , Pyrazoles/chemical synthesis , Receptors, Dopamine D2/agonists , Receptors, Dopamine D3/agonists , Animals , Antipsychotic Agents/chemistry , Antipsychotic Agents/pharmacology , Arrestins/metabolism , CHO Cells , Cricetulus , Drug Partial Agonism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , HEK293 Cells , Heterocyclic Compounds, 2-Ring/chemistry , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , Mice , Molecular Docking Simulation , Oximes/chemistry , Oximes/pharmacology , Piperazines/chemistry , Piperazines/pharmacology , Pyrazoles/chemistry , Pyrazoles/pharmacology , Radioligand Assay , Stereoisomerism , Structure-Activity Relationship , Swine , beta-Arrestin 2 , beta-Arrestins
4.
Mol Neurobiol ; 50(1): 107-18, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24633813

ABSTRACT

Huntington disease (HD) is a fatal neurodegenerative disorder caused by a CAG repeat expansion in exon 1 of the huntingtin gene (HTT). One prominent target of the mutant huntingtin protein (mhtt) is the mitochondrion, affecting its morphology, distribution, and function. Thus, mitochondria have been suggested as potential therapeutic targets for the treatment of HD. Olesoxime, a cholesterol-like compound, promotes motor neuron survival and neurite outgrowth in vitro, and its effects are presumed to occur via a direct interaction with mitochondrial membranes (MMs). We examined the properties of MMs isolated from cell and animal models of HD as well as the effects of olesoxime on MM fluidity and cholesterol levels. MMs isolated from brains of aged Hdh Q111/Q111 knock-in mice showed a significant decrease in 1,6-diphenyl-hexatriene (DPH) anisotropy, which is inversely correlated with membrane fluidity. Similar increases in MM fluidity were observed in striatal STHdh Q111/Q111 cells as well as in MMs isolated from brains of BACHD transgenic rats. Treatment of STHdh cells with olesoxime decreased the fluidity of isolated MMs. Decreased membrane fluidity was also measured in olesoxime-treated MMs isolated from brains of HD knock-in mice. In both models, treatment with olesoxime restored HD-specific changes in MMs. Accordingly, olesoxime significantly counteracted the mhtt-induced increase in MM fluidity of MMs isolated from brains of BACHD rats after 12 months of treatment in vivo, possibly by enhancing MM cholesterol levels. Thus, olesoxime may represent a novel pharmacological tool to treat mitochondrial dysfunction in HD.


Subject(s)
Brain/metabolism , Cholestenones/pharmacology , Huntington Disease/metabolism , Membrane Fluidity/drug effects , Mitochondrial Membranes/drug effects , Animals , Brain/drug effects , Cell Line , Cholestenones/therapeutic use , Disease Models, Animal , Huntington Disease/drug therapy , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Rats
5.
J Pharmacol Exp Ther ; 348(3): 393-400, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24259679

ABSTRACT

The acylated phloroglucinol, hyperforin, the main active ingredient of St. John's Wort, exerts antidepressant properties via indirect inhibition of serotonin reuptake by selectively activating the canonical transient receptor potential channel 6 (TRPC6). Hyperforin treatment can lead to drug-drug interactions due to potent activation of the nuclear receptor PXR (NR1I2), a key transcriptional regulator of genes involved in drug metabolism and transport. It was previously shown that synthetic acylated phloroglucinol derivatives activate TRPC6 with similar potency as hyperforin. However, their interaction potential with PXR remained unknown. Here we investigated five synthetic TRPC6-activating phloroglucinol derivatives and four TRPC6-nonactivating compounds compared with hyperforin and rifampicin for their potential to activate PXR in silico and in vitro. Computational PXR pharmacophore modeling did not indicate potent agonist or antagonist interactions for the TRPC6-activating derivatives, whereas one of them was suggested by docking studies to show both agonist and antagonist interactions. Hyperforin and rifampicin treatment of HepG2 cells cotransfected with human PXR expression vector and a CYP3A4 promoter-reporter construct resulted in potent PXR-dependent induction, whereas all TRPC6-activating compounds failed to show any PXR activation or to antagonize rifampicin-mediated CYP3A4 promoter induction. Hyperforin and rifampicin treatment of primary human hepatocytes resulted in highly correlated induction of PXR target genes, whereas treatment with the phloroglucinol derivatives elicited moderate gene expression changes that were only weakly correlated with those of rifampicin and hyperforin treatment. These results show that TRPC6-activating phloroglucinols do not activate PXR and should therefore be promising new candidates for further drug development.


Subject(s)
Phloroglucinol/analogs & derivatives , Phloroglucinol/pharmacology , Receptors, Steroid/agonists , Terpenes/pharmacology , Cytochrome P-450 CYP3A/genetics , Genes, Reporter , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Luciferases/genetics , Luciferases/metabolism , Molecular Docking Simulation , Phloroglucinol/chemistry , Pregnane X Receptor , Promoter Regions, Genetic , Receptors, Steroid/antagonists & inhibitors , Receptors, Steroid/chemistry , Terpenes/chemistry , Transcription, Genetic , Transcriptome
6.
Biochem Soc Trans ; 41(5): 1331-4, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24059528

ABSTRACT

The metabolic enhancer piracetam is used in many countries to treat cognitive impairment in aging, brain injuries, as well as dementia such as AD (Alzheimer's disease). As a specific feature of piracetam, beneficial effects are usually associated with mitochondrial dysfunction. In previous studies we were able to show that piracetam enhanced ATP production, mitochondrial membrane potential as well as neurite outgrowth in cell and animal models for aging and AD. To investigate further the effects of piracetam on mitochondrial function, especially mitochondrial fission and fusion events, we decided to assess mitochondrial morphology. Human neuroblastoma cells were treated with the drug under normal conditions and under conditions imitating aging and the occurrence of ROS (reactive oxygen species) as well as in stably transfected cells with the human wild-type APP (amyloid precursor protein) gene. This AD model is characterized by expressing only 2-fold more human Aß (amyloid ß-peptide) compared with control cells and therefore representing very early stages of AD when Aß levels gradually increase over decades. Interestingly, these cells exhibit an impaired mitochondrial function and morphology under baseline conditions. Piracetam is able to restore this impairment and shifts mitochondrial morphology back to elongated forms, whereas there is no effect in control cells. After addition of a complex I inhibitor, mitochondrial morphology is distinctly shifted to punctate forms in both cell lines. Under these conditions piracetam is able to ameliorate morphology in cells suffering from the mild Aß load, as well as mitochondrial dynamics in control cells.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Mitochondria/drug effects , Piracetam/therapeutic use , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Cell Line , Gene Expression Regulation , Humans , Membrane Potential, Mitochondrial/drug effects , Metabolism , Mitochondria/pathology , Reactive Oxygen Species/metabolism
7.
Chirality ; 25(11): 814-22, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24038285

ABSTRACT

In this study we addressed the role of chirality in the biological activity of RC-33, recently studied by us in its racemic form. An asymmetric synthesis procedure was the first experiment, leading to the desired enantioenriched RC-33 but with an enantiomeric excess (ee) not good enough for supporting the in vitro investigation. An enantioselective high-performance liquid chromatography (HPLC) procedure was then successfully carried out, yielding both RC-33 enantiomers in amounts and optical purity suitable for the pharmacological study. The absolute configuration of pure enantiomers was easily assigned exploiting the asymmetric synthesis previously devised. As emerged in the preliminary in vitro biological investigation, (S)- and (R)-RC-33 possess a comparable affinity towards the σ1 receptor and a very a similar behavior in the calcium influx assay, resulting in an equally effective σ1 receptor agonist. Overall, the results obtained so far suggest that the interaction with the biological target is nonstereoselective and leads us to hypothesize that there is a lack of stereoselectivity in the biological activity of RC-33.


Subject(s)
Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Animals , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/isolation & purification , Brain/drug effects , Brain/metabolism , Guinea Pigs , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/isolation & purification , PC12 Cells , Piperidines/chemical synthesis , Piperidines/isolation & purification , Rats , Receptors, sigma/agonists , Stereoisomerism
8.
J Neurochem ; 127(3): 303-13, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23875811

ABSTRACT

The non-selective cationic transient receptor canonical 6 (TRPC6) channels are involved in synaptic plasticity changes ranging from dendritic growth, spine morphology changes and increase in excitatory synapses. We previously showed that the TRPC6 activator hyperforin, the active antidepressant component of St. John's wort, induces neuritic outgrowth and spine morphology changes in PC12 cells and hippocampal CA1 neurons. However, the signaling cascade that transmits the hyperforin-induced transient rise in intracellular calcium into neuritic outgrowth is not yet fully understood. Several signaling pathways are involved in calcium transient-mediated changes in synaptic plasticity, ranging from calmodulin-mediated Ras-induced signaling cascades comprising the mitogen-activated protein kinase, PI3K signal transduction pathways as well as Ca(2+) /calmodulin-dependent protein kinase II (CAMKII) and CAMKIV. We show that several mechanisms are involved in TRPC6-mediated synaptic plasticity changes in PC12 cells and primary hippocampal neurons. Influx of calcium via TRPC6 channels activates different pathways including Ras/mitogen-activated protein kinase/extracellular signal-regulated kinases, phosphatidylinositide 3-kinase/protein kinase B, and CAMKIV in both cell types, leading to cAMP-response element binding protein phosphorylation. These findings are interesting not only in terms of the downstream targets of TRPC6 channels but also because of their potential to facilitate further understanding of St. John's wort extract-mediated antidepressant activity. Alterations in synaptic plasticity are considered to play an important role in the pathogenesis of depression. Beside several other proteins, TRPC6 channels regulate synaptic plasticity. This study demonstrates that different pathways including Ras/MEK/ERK, PI3K/Akt, and CAMKIV are involved in the improvement of synaptic plasticity by the TRPC6 activator hyperforin, the antidepressant active constituent of St. John's wort extract.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 4/physiology , Genes, ras/physiology , Hippocampus/physiology , Neurites/physiology , Neurons/physiology , Phosphatidylinositol 3-Kinases/physiology , TRPC Cation Channels/drug effects , TRPC Cation Channels/physiology , Animals , Anti-Bacterial Agents/pharmacology , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Activation/drug effects , Enzyme-Linked Immunosorbent Assay , Extracellular Signal-Regulated MAP Kinases/physiology , Hippocampus/drug effects , MAP Kinase Signaling System/physiology , Neurites/drug effects , Oncogene Protein v-akt/physiology , PC12 Cells , Phloroglucinol/analogs & derivatives , Phloroglucinol/pharmacology , Phosphorylation , Primary Cell Culture , Rats , Terpenes/pharmacology
9.
PLoS One ; 8(4): e59998, 2013.
Article in English | MEDLINE | ID: mdl-23637742

ABSTRACT

Recent clinical data support the clinical use of oral lavender oil in patients suffering from subsyndromal anxiety. We identified the molecular mechanism of action that will alter the perception of lavender oil as a nonspecific ingredient of aromatherapy to a potent anxiolytic inhibiting voltage dependent calcium channels (VOCCs) as highly selective drug target. In contrast to previous publications where exorbitant high concentrations were used, the effects of lavender oil in behavioral, biochemical, and electrophysiological experiments were investigated in physiological concentrations in the nanomolar range, which correlate to a single dosage of 80 mg/d in humans that was used in clinical trials. We show for the first time that lavender oil bears some similarities with the established anxiolytic pregabalin. Lavender oil inhibits VOCCs in synaptosomes, primary hippocampal neurons and stably overexpressing cell lines in the same range such as pregabalin. Interestingly, Silexan does not primarily bind to P/Q type calcium channels such as pregabalin and does not interact with the binding site of pregabalin, the α2δ subunit of VOCCs. Lavender oil reduces non-selectively the calcium influx through several different types of VOCCs such as the N-type, P/Q-type and T-type VOCCs. In the hippocampus, one brain region important for anxiety disorders, we show that inhibition by lavender oil is mainly mediated via N-type and P/Q-type VOCCs. Taken together, we provide a pharmacological and molecular rationale for the clinical use of the oral application of lavender oil in patients suffering from anxiety.


Subject(s)
Anti-Anxiety Agents/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Oils, Volatile/pharmacology , Plant Oils/pharmacology , Animals , Female , Hippocampus/cytology , Lavandula , Maze Learning/drug effects , Mice , Neurons/drug effects , Neurons/metabolism , Pregabalin , Synaptosomes/drug effects , Synaptosomes/metabolism , gamma-Aminobutyric Acid/analogs & derivatives , gamma-Aminobutyric Acid/pharmacology
10.
Eur J Pharm Sci ; 49(4): 603-13, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23643737

ABSTRACT

Given the fundamentally multifactorial character of Alzheimer's disease (AD), addressing more than one target for disease modification or therapy is expected to be highly advantageous. Here, following the cholinergic hypothesis, we aimed to inhibit both acetyl- and butyrylcholinesterase (AChE and BuChE) in order to increase the concentration of acetylcholine in the synaptic cleft. In addition, the formation of the amyloid ß fibrils should be inhibited and already preformed fibrils should be destroyed. Based on a recently identified AChE inhibitor with a 1,4-substituted 4-(1H)-pyridylene-hydrazone skeleton, a substance library has been generated and tested for inhibition of AChE, BuChE, and fibril formation. Blood-brain barrier mobility was ensured by a transwell assay. Whereas the p-nitrosubstituted compound 18C shows an anti-AChE activity in the nanomolar range of concentration (IC50=90 nM), the bisnaphthyl substituted compound 20L was found to be the best overall inhibitor of AChE/BuChE and enhances the fibril destruction.


Subject(s)
Acetylcholinesterase/metabolism , Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Hydrazones/pharmacology , Animals , Blood-Brain Barrier/metabolism , Endothelial Cells , HEK293 Cells , Humans , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
11.
Int J Pharm Pract ; 21(3): 169-77, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23419034

ABSTRACT

OBJECTIVE: To design and test the feasibility of two questionnaires in German community pharmacies exploring self-reported adherence to antihypertensives. METHODS: Two self-report questionnaires were designed for patients treated with antihypertensives. The 29-item-questionnaire (long form, LF) was completed by pharmacists interviewing patients who were on the premises filling a prescription. The short form (SF; 19 items) was sent by pharmacies to patients via mail. The acceptance of the instruments by patients and pharmacists as well as the feasibility to measure medication-taking behaviour was investigated. Adherence was investigated by using a modified 5-(LF) or 6-item (SF) Morisky score. RESULTS: Of 44 community pharmacies contacted, 18 agreed to participate. Patients' response rates were 428/915 (46.8%) for the SF and 249/760 (32.8%) for the LF. One hundred and seventy-nine patients (41.8%) and 70 patients (28.1%) reported adherence problems according to the SF and LF respectively. CONCLUSIONS: To our knowledge, this is the first attempt to develop a self-report instrument for the detection of non-adherence in patients taking antihypertensives in this setting in Germany. Patients were willing to provide detailed information about their medication-taking behaviour. Underestimation of non-adherence may be more pronounced when applying the questionnaire in the pharmacy.


Subject(s)
Antihypertensive Agents/administration & dosage , Hypertension/drug therapy , Medication Adherence , Surveys and Questionnaires , Aged , Antihypertensive Agents/therapeutic use , Community Pharmacy Services , Feasibility Studies , Female , Germany , Humans , Male , Middle Aged , Pharmacists , Self Report
12.
Int J Biochem Cell Biol ; 45(1): 76-80, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22743330

ABSTRACT

Mitochondria are membrane bound organelles that provide cellular energy in form of ATP. In addition to ATP synthesis mitochondria are key regulators of calcium homeostasis, free radical production, steroid synthesis and apoptosis, each of these factors could also be associated with essential mechanisms involved in neurodegenerative diseases. Recent studies revealed that changes in mitochondria membrane fluidity might have a direct impact on membrane-based processes such as fission-associated morphogenic changes, opening of the mitochondrial permeability transition pore or oxidative phosphorylation at the complexes of the electron transport chain. We investigated synaptosomal plasma and mitochondrial membranes isolated from brains of mouse models for ageing, Alzheimer's disease, Huntington's disease and Amyotrophic lateral sclerosis. Membrane properties are disease specifically altered, identifying mitochondrial membranes as targets for possible therapeutic strategies in neurodegenerative diseases. This article is part of a Directed Issue entitled: Bioenergetic dysfunction, adaptation and therapy.


Subject(s)
Aging/physiology , Brain/metabolism , Brain/pathology , Mitochondrial Membranes/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Animals , Apoptosis/physiology , Disease Models, Animal , Humans , Mice , Neurodegenerative Diseases/drug therapy
13.
Hippocampus ; 23(1): 40-52, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22815087

ABSTRACT

The standardized extract of the St. John's wort plant (Hypericum perforatum) is commonly used to treat mild to moderate depression. Its active constituent is hyperforin, a phloroglucinol derivative that reduces the reuptake of serotonin and norepinephrine by increasing intracellular Na(+) concentration through the activation of nonselective cationic TRPC6 channels. TRPC6 channels are also Ca(2+) -permeable, resulting in intracellular Ca(2+) elevations. Indeed, hyperforin activates TRPC6-mediated currents and Ca(2+) transients in rat PC12 cells, which induce their differentiation, mimicking the neurotrophic effect of nerve growth factor. Here, we show that hyperforin modulates dendritic spine morphology in CA1 and CA3 pyramidal neurons of hippocampal slice cultures through the activation of TRPC6 channels. Hyperforin also evoked intracellular Ca(2+) transients and depolarizing inward currents sensitive to the TRPC channel blocker La(3+) , thus resembling the actions of the neurotrophin brain-derived neurotrophic factor (BDNF) in hippocampal pyramidal neurons. These results suggest that the antidepressant actions of St. John's wort are mediated by a mechanism similar to that engaged by BDNF.


Subject(s)
Dendritic Spines/drug effects , Hypericum/chemistry , Phloroglucinol/analogs & derivatives , Plant Extracts/pharmacology , Pyramidal Cells/drug effects , TRPC Cation Channels/agonists , Terpenes/pharmacology , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Dendritic Spines/metabolism , Depression/drug therapy , Depression/metabolism , Gene Expression/physiology , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Lanthanum/pharmacology , Organ Culture Techniques , Phloroglucinol/pharmacology , Pyramidal Cells/metabolism , Pyramidal Cells/ultrastructure , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/genetics , Transfection
14.
Mol Neurobiol ; 46(1): 205-16, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22847631

ABSTRACT

Tauopathies like the "frontotemporal dementia with Parkinsonism linked to chromosome 17" (FTDP-17) are characterized by an aberrant accumulation of intracellular neurofibrillary tangles composed of hyperphosphorylated tau. For FTDP-17, a pathogenic tau mutation P301L was identified. Impaired mitochondrial function including disturbed dynamics such as fission and fusion are most likely major pathomechanisms of most neurodegenerative diseases. However, very little is known if tau itself affects mitochondrial function and dynamics. We addressed this question using SY5Y cells stably overexpressing wild-type (wt) and P301L mutant tau. P301L overexpression resulted in a substantial complex I deficit accompanied by decreased ATP levels and increased susceptibility to oxidative stress. This was paralleled by pronounced changes in mitochondrial morphology, decreased fusion and fission rates accompanied by reduced expression of several fission and fusion factors like OPA-1 or DRP-1. In contrast, overexpression of wt tau exhibits protective effects on mitochondrial function and dynamics including enhanced complex I activity. Our findings clearly link tau bidirectional to mitochondrial function and dynamics, identifying a novel aspect of the physiological role of tau and the pathomechanism of tauopathies.


Subject(s)
Mitochondria/pathology , Tauopathies/pathology , Cell Line, Tumor , Electron Transport , Humans , Mitochondria/ultrastructure , Mitochondrial Dynamics , Mutant Proteins/metabolism , Mutation/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Tauopathies/metabolism , tau Proteins/metabolism
15.
Mol Neurobiol ; 46(1): 186-93, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22833458

ABSTRACT

The non-Mendelian sporadic Alzheimer's disease (AD) is the most frequent form of dementia diagnosed worldwide. The most important risk factor to develop sporadic AD is aging itself. Next to hyperphosphorylated Tau, intracellular amyloid beta (Aß) oligomers are known to initiate a cascade of pathological events ranging from mitochondrial dysfunction, synaptic dysfunction, oxidative stress, and loss of calcium regulation, to inflammation. All these events are considered to play an important role in the progressive loss of neurons. The molecular mechanisms determining the balance between Aß production and clearance during the progression of the disease are not well understood. Furthermore, there is cumulating evidence that Aß formation impairs mitochondrial function and that mitochondrial dysfunction is an early event in the pathogenesis of AD. On the other hand, mitochondrial dysfunction, in particular increased formation of mitochondrially derived reactive oxygen species, promote Aß formation. Here, we review these latest findings linking mitochondrial dysfunction and Aß formation. We propose that mitochondrial dysfunction, which is well-known to increase with age, is an initial trigger for Aß production. As Aß itself further accelerates mitochondrial dysfunction and oxidative stress, its formation is self-stimulated. Taken together, a vicious cycle is initiated that originates from mitochondrial dysfunction, implying that AD can be viewed as an age-associated mitochondrial disorder. The proposed mechanism sheds new light on the pathophysiological changes taking place during the progression of AD as well as in the aging process.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Mitochondria/pathology , Aging/metabolism , Aging/pathology , Animals , Cognition , Humans , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Diseases/physiopathology , Reactive Oxygen Species/metabolism
16.
Mol Neurobiol ; 46(1): 194-204, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22821186

ABSTRACT

Alzheimer's disease (AD) is the most common progressive neurodegenerative disease. Today, AD affects millions of people worldwide and the number of AD cases will increase with increased life expectancy. The AD brain is marked by severe neurodegeneration like the loss of synapses and neurons, atrophy and depletion of neurotransmitter systems in the hippocampus and cerebral cortex. Recent findings suggest that these pathological changes are causally induced by mitochondrial dysfunction and increased oxidative stress. These changes are not only observed in the brain of AD patients but also in the periphery. In this review, we discuss the potential role of elevated apoptosis, increased oxidative stress and especially mitochondrial dysfunction as peripheral markers for the detection of AD in blood cells especially in lymphocytes. We discuss recent not otherwise published findings on the level of complex activities of the respiratory chain comprising mitochondrial respiration and the mitochondrial membrane potential (MMP). We obtained decreased basal MMP levels in lymphocytes from AD patients as well as enhanced sensitivity to different complex inhibitors of the respiratory chain. These changes are in line with mitochondrial defects obtained in AD cell and animal models, and in post-mortem AD tissue. Importantly, these mitochondrial alterations where not only found in AD patients but also in patients with mild cognitive impairment (MCI). These new findings point to a relevance of mitochondrial function as an early peripheral marker for the detection of AD and MCI.


Subject(s)
Alzheimer Disease/blood , Alzheimer Disease/physiopathology , Lymphocytes/pathology , Mitochondria/pathology , Aging/pathology , Alzheimer Disease/pathology , Animals , Biomarkers/blood , Humans , Mitochondria/metabolism , Oxidative Stress
17.
Int Psychogeriatr ; 24 Suppl 1: S21-4, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22784424

ABSTRACT

Neuroplasticity, the ability of synapses to undergo structural adaptations in response to functional demand or dysfunctions is increasingly impaired in aging and Alzheimer's disease. EGb761® has been shown in several preclinical reports to increase nearly all aspects of impaired neuroplasticity (long-term potentiation, spine density, neuritogenesis, neurogenesis). While all three fractions of constituents (ginkgolides, flavonoids, bilobalide) seem to be active, the flavonoids and specifically the aglycone isorhamnetin seem to be most relevant.


Subject(s)
Neuronal Plasticity/drug effects , Plant Extracts/pharmacology , Animals , Ginkgo biloba , Humans , Long-Term Potentiation/drug effects , Mitochondria/drug effects , Neurites/drug effects , Neurites/physiology , Neurogenesis/drug effects , Spinal Cord/drug effects , Spinal Cord/pathology
18.
Anal Chem ; 84(12): 5276-84, 2012 Jun 19.
Article in English | MEDLINE | ID: mdl-22594938

ABSTRACT

In the present work, the recently developed laser-induced liquid bead ion desorption mass spectrometry (LILBID MS) is applied as a novel technique to study Aß oligomerization, thought to be crucial in Alzheimer's disease (AD). The characterization of the earliest nucleation events of this peptide necessitates the application of several techniques to bridge the gap between small oligomers and large fibrils. We precisely monitored in time the transformation of monomeric Aß (1-42) into oligomeric Aß(n) (n < 20) and its dependence on concentration and agitation. The distribution shows signs of the hexamer being crucial in the assembly process. The intensity of the monomer decreases in time with a time constant of about 9 h. After a lag time of around 10 h, a phase transition occurred in which the total ion current of the oligomers goes to nearly zero. In this late stage of aggregation, protofibrils are formed and mass spectrometry is no longer sensitive. Here fluorescence correlation spectroscopy (FCS) and transmission electron microscopy (TEM) are complementary tools for detection and size characterization of these large species. We also utilized the oligomers of Aß (1-42) as a model of the corresponding in vivo process to screen the efficacy and specificity of small molecule inhibitors of oligomerization. The LILBID results are in excellent agreement with condensed phase behavior determined in other studies. Our data identified LILBID MS as a powerful technique that will advance the understanding of peptide oligomerization in neurodegenerative diseases and represents a powerful tool for the identification of small oligomerization inhibitors.


Subject(s)
Amyloid beta-Peptides/chemistry , Lasers , Mass Spectrometry/methods , Peptide Fragments/chemistry , Protein Multimerization , Amino Acid Sequence , Benzothiazoles , Humans , Hydrogen-Ion Concentration , Kinetics , Molecular Sequence Data , Protein Multimerization/drug effects , Protein Structure, Secondary , Small Molecule Libraries/pharmacology , Spectrometry, Fluorescence , Thiazoles/chemistry
19.
Mol Neurobiol ; 46(1): 136-50, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22552779

ABSTRACT

Increasing evidences suggest that mitochondrial dysfunction plays an important role in the pathogenesis of neurodegenerative diseases including Alzheimer's disease (AD). Alterations of mitochondrial efficiency and function are mainly related to alterations in mitochondrial content, amount of respiratory enzymes, or changes in enzyme activities leading to oxidative stress, mitochondrial permeability transition pore opening, and enhanced apoptosis. More recently, structural changes of the network are related to bioenergetic function, and its consequences are a matter of intensive research. Several mitochondria-targeting compounds with potential efficacy in AD including dimebon, methylene blue, piracetam, simvastatin, Ginkgo biloba, curcumin, and omega-3 polyunsaturated fatty acids have been identified. The majority of preclinical data indicate beneficial effects, whereas most controlled clinical trials did not meet the expectations. Since mitochondrial dysfunction represents an early event in disease progression, one reason for the disappointing clinical results could be that pharmacological interventions might came too late. Thus, more studies are needed that focus on therapeutic strategies starting before severe disease progress.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Mitochondria/pathology , Alzheimer Disease/drug therapy , Animals , Apoptosis/drug effects , Biological Products/pharmacology , Biological Products/therapeutic use , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Oxidative Stress/drug effects
20.
J Alzheimers Dis ; 31(1): 21-32, 2012.
Article in English | MEDLINE | ID: mdl-22475801

ABSTRACT

Due to their role in producing energy, as major sources of free radicals, and as critical regulators of apoptosis, mitochondria play a dominant role in the central nervous system (CNS). Mitochondrial dysfunction represents one major pathomechanism of Alzheimer's disease (AD), including impaired function of mitochondrial respiratory chain complexes and deficits of mitochondrial dynamics, such as impaired balance between fission and fusion mechanisms and reduced mitochondrial trafficking. Major consequences are enhanced depletion of mitochondria in axons and dendrites, synaptic dysfunction, and finally neuronal loss. Interfering with impaired mitochondrial dynamics has been proposed as novel strategy for antidementia drugs. Dimebon has been shown to improve cognition in animal models and seems to be beneficial in AD patients. Regardless of the final proof of Dimebon's clinical efficacy, it might specifically interfere with mechanisms relevant for the cognitive decline, especially by improving impaired mitochondrial function and/or dynamics in AD. Herein, we tested the effects of Dimebon on mitochondrial function and dynamics in a cellular model, overexpressing neurotoxic Aß peptides, one of the hallmarks of AD. Dimebon exerted pronounced effects on mitochondrial morphology, respiratory chain complex activities, and enlarged mitochondrial mass. In summary, form and function of mitochondria are altered in the Aß overexpressing cell model and precisely those changes are restored by nanomolar Dimebon treatment. Our findings support the idea that Dimebon improves mitochondrial function and that these "disease specific" effects might be relevant for interpretation and planning of future clinical trials.


Subject(s)
Amyloid beta-Peptides/toxicity , Mitochondria/drug effects , Oxygen Consumption/drug effects , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Drug Interactions , HEK293 Cells/ultrastructure , Humans , Mutation/genetics , Organic Chemicals , Oxygen Consumption/genetics , Spectrophotometry , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...