Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
J Neural Transm (Vienna) ; 130(8): 1003-1012, 2023 08.
Article in English | MEDLINE | ID: mdl-37014414

ABSTRACT

Therapeutic approaches providing effective medication for Alzheimer's disease (AD) patients after disease onset are urgently needed. Previous studies in AD mouse models and in humans suggested that physical exercise or changed lifestyle can delay AD-related synaptic and memory dysfunctions when treatment started in juvenile animals or in elderly humans before onset of disease symptoms. However, a pharmacological treatment that can reverse memory deficits in AD patients was thus far not identified. Importantly, AD disease-related dysfunctions have increasingly been associated with neuro-inflammatory mechanisms and searching for anti-inflammatory medication to treat AD seems promising. Like for other diseases, repurposing of FDA-approved drugs for treatment of AD is an ideally suited strategy to reduce the time to bring such medication into clinical practice. Of note, the sphingosine-1-phosphate analogue fingolimod (FTY720) was FDA-approved in 2010 for treatment of multiple sclerosis patients. It binds to the five different isoforms of Sphingosine-1-phosphate receptors (S1PRs) that are widely distributed across human organs. Interestingly, recent studies in five different mouse models of AD suggest that FTY720 treatment, even when starting after onset of AD symptoms, can reverse synaptic deficits and memory dysfunction in these AD mouse models. Furthermore, a very recent multi-omics study identified mutations in the sphingosine/ceramide pathway as a risk factor for sporadic AD, suggesting S1PRs as promising drug target in AD patients. Therefore, progressing with FDA-approved S1PR modulators into human clinical trials might pave the way for these potential disease modifying anti-AD drugs.


Subject(s)
Alzheimer Disease , Multiple Sclerosis , Mice , Animals , Humans , Aged , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Alzheimer Disease/drug therapy , Drug Repositioning , Sclerosis , Multiple Sclerosis/drug therapy , Inflammation/drug therapy , Inflammation/metabolism
2.
Polymers (Basel) ; 14(7)2022 Mar 22.
Article in English | MEDLINE | ID: mdl-35406157

ABSTRACT

The present study covers the synthesis, purification and evaluation of a novel aminomethacrylate-based copolymer in terms of its suitability for improving the solubility and in vitro release of poorly water-soluble drug compounds. The new copolymer was synthesized by solvent polymerization with radical initiation and by use of a chain transfer agent. Based on its composition, it can be considered as a modified type of dimethylaminoethyl methacrylate-butyl methacrylate-methyl methacrylate "EUDRAGIT® E PO" (ModE). ModE was specifically developed to provide a copolymer with processing and application properties that exceed those of commercially available (co-)polymers in solubility enhancement technologies where possible. By varying the concentration of the chain transfer agent in the radical polymerization process, the molecular weight of ModE was varied in a range of 173-305 kDa. To evaluate the solubility-enhancing properties of ModE, a series of drug-loaded extrudates were prepared by hot melt extrusion using the novel-as well as several commercially available-(co-)polymers. These extrudates were then subjected to comparative tests for amorphousness, solubility-enhancing properties, storage stability, and drug release. Celecoxib, efavirenz, and fenofibrate were used as model drugs in all experiments. Of all the (co-)polymers included in the study, ModE with a molecular weight of 173 kDa showed the best performance in terms of desired properties and was shown to be particularly suitable for preparing amorphous solid dispersions (ASDs) of the three model drugs, which in a first set of dissolution experiments showed better release behavior under pH conditions of the fasting stomach than higher molecular weight ModE types, as well as a variety of commercially available (co-)polymers. Therefore, the results demonstrate the successful synthesis of a new copolymer, which in future studies will be investigated in more detail for universal application in the field of solubility enhancement.

3.
Eval Program Plann ; 91: 102049, 2022 04.
Article in English | MEDLINE | ID: mdl-35217287

ABSTRACT

Strategies for sustaining a program beyond initial implementation remain one of the most poorly understood aspects of high-quality program implementation. This paper describes the Quality Improvement and Fidelity Assessment Process (QIFAP), a program purveyor-agency partnership that uses a unique, multi-step method for supporting sustained implementation of the Therapeutic Crisis Intervention (TCI) system to manage crises in child serving organizations. It outlines the steps of the process and highlights how specific activities are linked to current knowledge and principles from implementation science. The QIFAP occurs over a period of about three months, during which time program developers and agency representatives conduct staff surveys, a two-day site visit, and fidelity assessments in order to gather information, discuss findings, and plan steps for improving the TCI system in the organization. The process is guided by principles that emphasize the importance of organization leadership, building relationships, co-learning, using an individualized approach, data informed decision making, acknowledging risk, and congruence. We describe ways in which the strategies and approaches within the QIFAP are rooted in implementation science literature. Thus, the model represents an illustration of how research-based knowledge can work in practice to support long-term, high-quality program implementation.


Subject(s)
Crisis Intervention , Implementation Science , Child , Health Services , Humans , Program Evaluation , Quality Improvement
4.
Cereb Cortex ; 32(7): 1350-1364, 2022 03 30.
Article in English | MEDLINE | ID: mdl-34470044

ABSTRACT

Neurotrophins are secreted proteins that control survival, differentiation, and synaptic plasticity. While mature neurotrophins regulate these functions via tyrosine kinase signaling (Trk), uncleaved pro-neurotrophins bind preferentially to the p75 neurotrophin receptor (p75NTR) and often exert opposite effects to those of mature neurotrophins. In the amygdala, brain-derived neurotrophic factor (BDNF) enables long-term potentiation as well as fear and fear extinction learning. In the present study, we focused on the impact of mature BDNF and proBDNF signaling on long-term depression (LTD) in the lateral amygdala (LA). Hence, we conducted extracellular field potential recordings in an in vitro slice preparation and recorded LTD in cortical and thalamic afferents to the LA. LTD was unchanged by acute block of BDNF/TrkB signaling. In contrast, LTD was inhibited by blocking p75NTR signaling, by disinhibition of the proteolytic cleavage of proBDNF into mature BDNF, and by preincubation with a function-blocking anti-proBDNF antibody. Since LTD-like processes in the amygdala are supposed to be related to fear extinction learning, we locally inhibited p75NTR signaling in the amygdala during or after fear extinction training, resulting in impaired fear extinction memory. Overall, these results suggest that in the amygdala proBDNF/p75NTR signaling plays a pivotal role in LTD and fear extinction learning.


Subject(s)
Extinction, Psychological , Fear , Amygdala/metabolism , Animals , Extinction, Psychological/physiology , Fear/physiology , Learning/physiology , Mice , Neuronal Plasticity
5.
Transl Psychiatry ; 11(1): 233, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33888685

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is implicated in a number of processes that are crucial for healthy functioning of the brain. Schizophrenia is associated with low BDNF levels in the brain and blood, however, not much is known about BDNF's role in the different symptoms of schizophrenia. Here, we used BDNF-haploinsufficient (BDNF+/-) mice to investigate the role of BDNF in different mouse behavioral endophenotypes of schizophrenia. Furthermore, we assessed if an enriched environment can prevent the observed changes. In this study, male mature adult wild-type and BDNF+/- mice were tested in mouse paradigms for cognitive flexibility (attentional set shifting), sensorimotor gating (prepulse inhibition), and associative emotional learning (safety and fear conditioning). Before these tests, half of the mice had a 2-month exposure to an enriched environment, including running wheels. After the tests, BDNF brain levels were quantified. BDNF+/- mice had general deficits in the attentional set-shifting task, increased startle magnitudes, and prepulse inhibition deficits. Contextual fear learning was not affected but safety learning was absent. Enriched environment housing completely prevented the observed behavioral deficits in BDNF+/- mice. Notably, the behavioral performance of the mice was negatively correlated with BDNF protein levels. These novel findings strongly suggest that decreased BDNF levels are associated with several behavioral endophenotypes of schizophrenia. Furthermore, an enriched environment increases BDNF protein to wild-type levels and is thereby able to rescue these behavioral endophenotypes.


Subject(s)
Brain-Derived Neurotrophic Factor , Schizophrenia , Animals , Brain-Derived Neurotrophic Factor/genetics , Endophenotypes , Haploinsufficiency , Male , Mice , Mice, Inbred C57BL , Reflex, Startle , Schizophrenia/genetics
6.
Drug Deliv Transl Res ; 11(5): 2239-2251, 2021 10.
Article in English | MEDLINE | ID: mdl-33469893

ABSTRACT

The use of alcoholic beverages can cause uncontrolled release of drugs from sustained-release solid oral dosage forms and pose severe risks to patient health. The aim of this work was to design a new polymeric excipient with ethanol resistance inherent to the polymer. Polymers were systematically designed, manufactured via emulsion polymerization, and fully characterized. Glass transition temperatures between 10 and 18 °C and minimum film forming temperatures between 10 and 25 °C were chosen because these parameters are ideal for aqueous film-coating processing. Three model drug formulations were made with the new polymer excipients and tested in the presence and absence of ethanol. The concept of an alcohol resistance factor based on Weibull regression analysis was introduced. In vitro results confirmed the hypothesized structure-function relationship between comonomer composition and ethanol resistance. That is, nonionic hydrophilic functional groups interacted more strongly with the ethanolic solvent, as compared with cationic hydrophilic comonomer that interacted more strongly with the surrounding water molecules. The alcohol resistance factor varied between - 44 ± 2% (slower drug release in presence of ethanol) and + 34 ± 0% (faster drug release in presence of ethanol) depending on the comonomer ratio. The main advantages of these new excipients compared with ethanol-resistant excipient blends include ease of use, plasticizers are not necessary, and shorter coating times.


Subject(s)
Excipients , Polymers , Delayed-Action Preparations , Drug Compounding , Ethanol , Humans , Solubility
7.
Int J Mol Sci ; 21(23)2020 Nov 25.
Article in English | MEDLINE | ID: mdl-33255764

ABSTRACT

Therapeutic approaches providing effective medication for Alzheimer's disease (AD) patients after disease onset are urgently needed. Previous studies in AD mouse models suggested that physical exercise or changed lifestyle can delay AD-related synaptic and memory dysfunctions when treatment started in juvenile animals long before onset of disease symptoms, while a pharmacological treatment that can reverse synaptic and memory deficits in AD mice was thus far not identified. Repurposing food and drug administration (FDA)-approved drugs for treatment of AD is a promising way to reduce the time to bring such medication into clinical practice. The sphingosine-1 phosphate analog fingolimod (FTY720) was approved recently for treatment of multiple sclerosis patients. Here, we addressed whether fingolimod rescues AD-related synaptic deficits and memory dysfunction in an amyloid precursor protein/presenilin-1 (APP/PS1) AD mouse model when medication starts after onset of symptoms (at five months). Male mice received intraperitoneal injections of fingolimod for one to two months starting at five to six months. This treatment rescued spine density as well as long-term potentiation in hippocampal cornu ammonis-1 (CA1) pyramidal neurons, that were both impaired in untreated APP/PS1 animals at six to seven months of age. Immunohistochemical analysis with markers of microgliosis (ionized calcium-binding adapter molecule 1; Iba1) and astrogliosis (glial fibrillary acid protein; GFAP) revealed that our fingolimod treatment regime strongly down regulated neuroinflammation in the hippocampus and neocortex of this AD model. These effects were accompanied by a moderate reduction of Aß accumulation in hippocampus and neocortex. Our results suggest that fingolimod, when applied after onset of disease symptoms in an APP/PS1 mouse model, rescues synaptic pathology that is believed to underlie memory deficits in AD mice, and that this beneficial effect is mediated via anti-neuroinflammatory actions of the drug on microglia and astrocytes.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor/genetics , Inflammation/drug therapy , Memory Disorders/drug therapy , Presenilin-1/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , Anti-Inflammatory Agents/pharmacology , Astrocytes/metabolism , Astrocytes/pathology , Disease Models, Animal , Fingolimod Hydrochloride/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Memory Disorders/genetics , Memory Disorders/metabolism , Memory Disorders/pathology , Mice , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Synapses/genetics , Synapses/pathology
8.
Cell Tissue Res ; 382(1): 161-172, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32845430

ABSTRACT

The amygdala is a central hub for fear learning assessed by Pavlovian fear conditioning. Indeed, the prevailing hypothesis that learning and memory are mediated by changes in synaptic strength was shown most convincingly at thalamic and cortical afferents to the lateral amygdala. The neurotrophin brain-derived neurotrophic factor (BDNF) is known to regulate synaptic plasticity and memory formation in many areas of the mammalian brain including the amygdala, where BDNF signalling via tropomyosin-related kinase B (TrkB) receptors is prominently involved in fear learning. This review updates the current understanding of BDNF/TrkB signalling in the amygdala related to fear learning and extinction. In addition, actions of proBDNF/p75NTR and NGF/TrkA as well as NT-3/TrkC signalling in the amygdala are introduced.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Fear/physiology , Learning/physiology , Neurotrophin 3/physiology , Amygdala , Animals , Humans , Signal Transduction
9.
J Neurosci Methods ; 341: 108797, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32479974

ABSTRACT

BACKGROUND: Spine loss is a hallmark of Alzheimer´s and other neurodegenerative diseases, and testing candidate therapeutic drugs needs quantitative analysis of dendritic spine densities. Golgi-Cox impregnation of neurons is a classical method to visualize dendritic spines in diseased brains. Importantly, at early disease stages spine loss occurs locally in the vicinity of amyloid plaques, and concomitant fluorescence labeling of amyloid plaques is required to detect local spine damage. NEW METHOD: Because Golgi-Cox impregnation is done on unsectioned brains, whereas fluorescence staining is performed on sectioned material, the combination is technically challenging. We have now developed a novel combination of Golgi-Cox impregnation with methoxy-X04 fluorescence labeling of plaques that is performed on unsectioned brains. RESULTS: We used this new combination method to quantify dendritic spine densities in mouse hippocampal CA1 pyramidal neurons. Comparison of neurons from wildtype and APP/PS1 mice revealed local spine loss in the vicinity of amyloid plaques in both male and female APP/PS1 mice. COMPARISON WITH EXISTING METHOD: Golgi-Cox impregnation of neurons combined with methoxy-X04 staining of amyloid plaques is a highly reliable, easy-to-use method for permanent visualization of spines as compared to the technically more sophisticated and less stable fluorescence imaging of spines. CONCLUSION: Our novel combination method will be highly useful for testing potential therapeutic drugs in Alzheimer mouse models.


Subject(s)
Alzheimer Disease , Plaque, Amyloid , Alzheimer Disease/diagnostic imaging , Animals , Dendritic Spines , Female , Male , Mice , Mice, Transgenic , Staining and Labeling
10.
Neurosci Biobehav Rev ; 115: 25-33, 2020 08.
Article in English | MEDLINE | ID: mdl-32439371

ABSTRACT

A better understanding of context in decision-making-that is, the internal and external conditions that modulate decisions-is required to help bridge the gap between natural behaviors that evolved by natural selection and more arbitrary laboratory models of anxiety and fear. Because anxiety and fear are mechanisms evolved to manage threats from predators and other exigencies, the large behavioral, ecological and evolutionary literature on predation risk is useful for re-framing experimental research on human anxiety-related disorders. We review the trade-offs that are commonly made during antipredator decision-making in wild animals along with the context under which the behavior is performed and measured, and highlight their relevance for focused laboratory models of fear and anxiety. We then develop an integrative mechanistic model of decision-making under risk which, when applied to laboratory and field settings, should improve studies of the biological basis of normal and pathological anxiety and may therefore improve translational outcomes.


Subject(s)
Fear , Predatory Behavior , Animals , Anxiety , Anxiety Disorders , Humans
11.
J Biol Chem ; 295(14): 4383-4397, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32094224

ABSTRACT

Mitochondrial oxidative phosphorylation (OXPHOS) and cellular workload are tightly balanced by the key cellular regulator, calcium (Ca2+). Current models assume that cytosolic Ca2+ regulates workload and that mitochondrial Ca2+ uptake precedes activation of matrix dehydrogenases, thereby matching OXPHOS substrate supply to ATP demand. Surprisingly, knockout (KO) of the mitochondrial Ca2+ uniporter (MCU) in mice results in only minimal phenotypic changes and does not alter OXPHOS. This implies that adaptive activation of mitochondrial dehydrogenases by intramitochondrial Ca2+ cannot be the exclusive mechanism for OXPHOS control. We hypothesized that cytosolic Ca2+, but not mitochondrial matrix Ca2+, may adapt OXPHOS to workload by adjusting the rate of pyruvate supply from the cytosol to the mitochondria. Here, we studied the role of malate-aspartate shuttle (MAS)-dependent substrate supply in OXPHOS responses to changing Ca2+ concentrations in isolated brain and heart mitochondria, synaptosomes, fibroblasts, and thymocytes from WT and MCU KO mice and the isolated working rat heart. Our results indicate that extramitochondrial Ca2+ controls up to 85% of maximal pyruvate-driven OXPHOS rates, mediated by the activity of the complete MAS, and that intramitochondrial Ca2+ accounts for the remaining 15%. Of note, the complete MAS, as applied here, included besides its classical NADH oxidation reaction the generation of cytosolic pyruvate. Part of this largely neglected mechanism has previously been described as the "mitochondrial gas pedal." Its implementation into OXPHOS control models integrates seemingly contradictory results and warrants a critical reappraisal of metabolic control mechanisms in health and disease.


Subject(s)
Calcium/metabolism , Cytosol/metabolism , Mitochondria/metabolism , Pyruvic Acid/metabolism , Animals , Aspartic Acid/metabolism , Brain/metabolism , Calcium Channels/deficiency , Calcium Channels/genetics , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Heart/physiology , Malates/chemistry , Malates/metabolism , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Oxidative Phosphorylation , Rats , Substrate Specificity , Synaptosomes/metabolism
12.
Neurosci Lett ; 713: 134521, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31563673

ABSTRACT

Disturbed iron (Fe) ion homeostasis and mitochondrial dysfunction have been implicated in neurodegeneration. Both processes are related, because central Fe ion consuming biogenetic pathways take place in mitochondria and affect their oxidative energy metabolism. Iron is imported into mitochondria by the two homologous Fe ion importers mitoferrin-1 and mitoferrin-2. To elucidate more specifically the role of mitochondrial Fe ions for brain energy metabolism and for proper neuronal function, we generated mice with a neuron-specific knockout of mitoferrin-1 (Slc25a37-/- or mfrn-1-/-) and compared them with corresponding control littermates (mfrn-1flox/flox). Mice lacking neuronal mfrn-1 exhibited no obvious anatomical or behavioral abnormalities as neonates, young or adult animals. However, they exhibited a moderate decrease in brain mitochondrial O2-consumption with complex-I substrates of the electron transport chain (p < 0.05), indicating a moderate suppression of electron transport. While these mice did not exhibit altered basal fear levels, inquisitiveness or motor skills in specific neurobiological test batteries, they clearly exhibited decreased spatial learning skills and missing establishment of stable spatial memory in Morris water maze, as compared to floxed controls (p < 0.05). We thus conclude that mitochondrial Fe ion supply is an important player in neuronal energy metabolism and proper brain function and that the carrier mitoferrin-1 cannot be completely replaced by mitoferrin-2 or other as yet unknown Fe ion carriers.


Subject(s)
Brain/metabolism , Energy Metabolism/physiology , Membrane Transport Proteins/physiology , Memory/physiology , Spatial Learning/physiology , Animals , Behavior, Animal/physiology , Female , Male , Maze Learning/physiology , Membrane Transport Proteins/genetics , Mice , Mice, Knockout/genetics , Mitochondria/metabolism , Oxygen Consumption/physiology
13.
Int J Mol Sci ; 20(17)2019 Sep 03.
Article in English | MEDLINE | ID: mdl-31484392

ABSTRACT

Brain-derived neurotrophic factor (BDNF) has previously been shown to play an important role in glutamatergic synaptic plasticity in the amygdala, correlating with cued fear learning. While glutamatergic neurotransmission is facilitated by BDNF signaling in the amygdala, its mechanism of action at inhibitory synapses in this nucleus is far less understood. We therefore analyzed the impact of chronic BDNF depletion on GABAA-mediated synaptic transmission in BDNF heterozygous knockout mice (BDNF+/-). Analysis of miniature and evoked inhibitory postsynaptic currents (IPSCs) in the lateral amygdala (LA) revealed neither pre- nor postsynaptic differences in BDNF+/- mice compared to wild-type littermates. In addition, long-term potentiation (LTP) of IPSCs was similar in both genotypes. In contrast, facilitation of spontaneous IPSCs (sIPSCs) by norepinephrine (NE) was significantly reduced in BDNF+/- mice. These results argue against a generally impaired efficacy and plasticity at GABAergic synapses due to a chronic BDNF deficit. Importantly, the increase in GABAergic tone mediated by NE is reduced in BDNF+/- mice. As release of NE is elevated during aversive behavioral states in the amygdala, effects of a chronic BDNF deficit on GABAergic inhibition may become evident in response to states of high arousal, leading to amygdala hyper-excitability and impaired amygdala function.


Subject(s)
Amygdala/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Long-Term Potentiation/physiology , Animals , Brain-Derived Neurotrophic Factor/genetics , Female , Long-Term Potentiation/genetics , Mice , Mice, Knockout , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Patch-Clamp Techniques , Synaptic Transmission/genetics , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism
14.
Mol Neurobiol ; 56(10): 6833-6855, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30929164

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is a secreted messenger molecule that is crucial for neuronal function and induction of synaptic plasticity. Although altered availability of BDNF underlies many neurological deficits and neurodegenerative disorders, secretion dynamics of endogenous BDNF are unexplored. We generated a BDNF-GFP knock-in (KiBE) mouse, in which GFP-labeled BDNF is expressed under the control of the unaltered endogenous mouse BDNF gene regulatory elements. This KiBE mouse model enables for the first time live cell imaging analysis of endogenous BDNF dynamics. We show that BDNF-GFP release and biological activity in vivo are unaffected by the GFP tag, since homozygous KiBE mice, which lack wild-type BDNF, are healthy and have a normal life expectancy. STED superresolution microscopy shows that 70% of BDNF-GFP vesicles in KiBE mouse neurites are localized in dendrites, being typically 200 nm away from synaptic release sites. Live cell imaging in hippocampal slices also reveals prominent targeting of endogenous BDNF-GFP vesicles to dendrites. Fusion pore opening and cargo release of dendritic BDNF vesicles start within 30 s after a strong depolarizing stimulus and continue for > 100 s thereafter, revealing an astonishingly delayed and prolonged release of endogenous BDNF.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Dendrites/metabolism , Exocytosis , Gene Knock-In Techniques , Green Fluorescent Proteins/metabolism , Synaptic Vesicles/metabolism , Animals , Axons/metabolism , Cells, Cultured , Chromosomes, Mammalian/genetics , Gene Targeting , Genome , Hippocampus/metabolism , Mice
15.
Sci Adv ; 4(10): eaat6994, 2018 10.
Article in English | MEDLINE | ID: mdl-30417089

ABSTRACT

Cognitive impairments can be devastating for quality of life, and thus, preventing or counteracting them is of great value. To this end, the present study exploits the potential of the plant Rhodiola rosea and identifies the constituent ferulic acid eicosyl ester [icosyl-(2E)-3-(4-hydroxy-3-methoxyphenyl)-prop-2-enoate (FAE-20)] as a memory enhancer. We show that food supplementation with dried root material from R. rosea dose-dependently improves odor-taste reward associative memory scores in larval Drosophila and prevents the age-related decline of this appetitive memory in adult flies. Task-relevant sensorimotor faculties remain unaltered. From a parallel approach, a list of candidate compounds has been derived, including R. rosea-derived FAE-20. Here, we show that both R. rosea-derived FAE-20 and synthetic FAE-20 are effective as memory enhancers in larval Drosophila. Synthetic FAE-20 also partially compensates for age-related memory decline in adult flies, as well as genetically induced early-onset loss of memory function in young flies. Furthermore, it increases excitability in mouse hippocampal CA1 neurons, leads to more stable context-shock aversive associative memory in young adult (3-month-old) mice, and increases memory scores in old (>2-year-old) mice. Given these effects, and given the utility of R. rosea-the plant from which we discovered FAE-20-as a memory enhancer, these results may hold potential for clinical applications.


Subject(s)
Coumaric Acids/pharmacology , Esters/pharmacology , Memory/drug effects , Rhodiola/chemistry , Age Factors , Animals , Bees , Behavior, Animal/drug effects , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/drug effects , Dietary Supplements , Drosophila melanogaster , Fear/drug effects , Larva/drug effects , Male , Mice, Inbred C57BL , Plant Extracts/pharmacology , Species Specificity
17.
Front Behav Neurosci ; 9: 58, 2015.
Article in English | MEDLINE | ID: mdl-25852506

ABSTRACT

There is increasing evidence that brain-derived neurotrophic factor (BDNF) plays a crucial role in Alzheimer's disease (AD) pathology. A number of studies demonstrated that AD patients exhibit reduced BDNF levels in the brain and the blood serum, and in addition, several animal-based studies indicated a potential protective effect of BDNF against Aß-induced neurotoxicity. In order to further investigate the role of BDNF in the etiology of AD, we created a novel mouse model by crossing a well-established AD mouse model (APP/PS1) with a mouse exhibiting a chronic BDNF deficiency (BDNF(+/-)). This new triple transgenic mouse model enabled us to further analyze the role of BDNF in AD in vivo. We reasoned that in case BDNF has a protective effect against AD pathology, an AD-like phenotype in our new mouse model should occur earlier and/or in more severity than in the APP/PS1-mice. Indeed, the behavioral analysis revealed that the APP/PS1-BDNF(+/-)-mice show an earlier onset of learning impairments in a two-way active avoidance task in comparison to APP/PS1- and BDNF(+/-)-mice. However in the Morris water maze (MWM) test, we could not observe an overall aggrevated impairment in spatial learning and also short-term memory in an object recognition task remained intact in all tested mouse lines. In addition to the behavioral experiments, we analyzed the amyloid plaque pathology in the APP/PS1 and APP/PS1-BDNF(+/-)-mice and observed a comparable plaque density in the two genotypes. Moreover, our results revealed a higher plaque density in prefrontal cortical compared to hippocampal brain regions. Our data reveal that higher cognitive tasks requiring the recruitment of cortical networks appear to be more severely affected in our new mouse model than learning tasks requiring mainly sub-cortical networks. Furthermore, our observations of an accelerated impairment in active avoidance learning in APP/PS1-BDNF(+/-)-mice further supports the hypothesis that BDNF deficiency amplifies AD-related cognitive dysfunctions.

18.
Neurobiol Learn Mem ; 120: 52-60, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25724412

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is a crucial mediator of neural plasticity and, consequently, of memory formation. In hippocampus-dependent learning tasks BDNF also seems to play an essential role. However, there are conflicting results concerning the spatial learning ability of aging BDNF(+/-) mice in the Morris water maze paradigm. To evaluate the effect of chronic BDNF deficiency in the hippocampus on spatial learning throughout life, we conducted a comprehensive study to test differently aged BDNF(+/-) mice and their wild type littermates in the Morris water maze and to subsequently quantify their hippocampal BDNF protein levels as well as expression levels of TrkB receptors. We observed an age-dependent learning deficit in BDNF(+/-) animals, starting at seven months of age, despite stable hippocampal BDNF protein expression and continual decline of TrkB receptor expression throughout aging. Furthermore, we detected a positive correlation between hippocampal BDNF protein levels and learning performance during the probe trial in animals that showed a good learning performance during the long-term memory test.


Subject(s)
Brain-Derived Neurotrophic Factor/deficiency , Maze Learning/physiology , Age Factors , Animals , Blotting, Western , Brain-Derived Neurotrophic Factor/analysis , Brain-Derived Neurotrophic Factor/physiology , Hippocampus/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
19.
Mol Pharm ; 11(4): 1273-81, 2014 Apr 07.
Article in English | MEDLINE | ID: mdl-24592902

ABSTRACT

Amphiphilic triblock copolymers represent a versatile delivery platform capable of co-delivery of nucleic acids, drugs, and/or dyes. Multifunctional cationic triblock copolymers based on poly(ethylene glycol), poly-ε-caprolactone, and polyethylene imine, designed for the delivery of siRNA, were evaluated in vitro and in vivo. Moreover, a nucleic acid-unpacking-sensitive imaging technique based on quantum dot-mediated fluorescence resonance energy transfer (QD-FRET) was established. Cell uptake in vitro was measured by flow cytometry, whereas transfection efficiencies of nanocarriers with different hydrophilic block lengths were determined in vitro and in vivo by quantitative real-time PCR. Furthermore, after the proof of concept was demonstrated by fluorescence spectroscopy/microscopy, a prototype FRET pair was established by co-loading QDs and fluorescently labeled siRNA. The hydrophobic copolymer mediated a 5-fold higher cellular uptake and good knockdown efficiency (61 ± 5% in vitro, 55 ± 18% in vivo) compared to its hydrophilic counterpart (13 ± 6% in vitro, 30 ± 17% in vivo), which exhibited poor performance. FRET was demonstrated by UV-induced emission of the acceptor dye. Upon complex dissociation, which was simulated by the addition of heparin, a dose-dependent decrease in FRET efficiency was observed. We believe that in vitro/in vivo correlation of the structure and function of polymeric nanocarriers as well as sensitive imaging functionality for mechanistic investigations are prerequisites for a more rational design of amphiphilic gene carriers.


Subject(s)
Fluorescence Resonance Energy Transfer , Gene Transfer Techniques , Polyesters/administration & dosage , Polyethyleneimine/analogs & derivatives , Quantum Dots/administration & dosage , RNA, Small Interfering/administration & dosage , Animals , Mice , Mice, Inbred BALB C , Polyethyleneimine/administration & dosage
20.
Neuropharmacology ; 71: 247-54, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23587649

ABSTRACT

Brain-derived neurotrophic factor (BDNF) signaling via TrkB crucially regulates synaptic plasticity in the brain. Although BDNF is abundant at hippocampal mossy fiber (MF) synapses, which critically contribute to hippocampus dependent memory, its role in MF synaptic plasticity (long-term potentiation, LTP) remained largely unclear. Using field potential recordings in CA3 of adult heterozygous BDNF knockout (ko, BDNF+/-) mice we observed impaired (∼50%) NMDAR-independent MF-LTP. In contrast to MF synapses, LTP at neighboring associative/commissural (A/C) fiber synapses remained unaffected. To exclude that impaired MF-LTP in BDNF+/- mice was due to developmental changes in response to chronically reduced BDNF levels, and to prove the importance of acute availability of BDNF in MF-LTP, we also tested effects of acute interference with BDNF/TrkB signaling. Inhibition of TrkB tyrosine kinase signaling with k252a, or with the selective BDNF scavenger TrkB-Fc, both inhibited MF-LTP to the same extent as observed in BDNF+/- mice. Basal synaptic transmission, short-term plasticity, and synaptic fatigue during LTP induction were not significantly altered by treatment with k252a or TrkB-Fc, or by chronic BDNF reduction in BDNF+/- mice. Since the acute interference with BDNF-signaling did not completely block MF-LTP, our results provide evidence that an additional mechanism besides BDNF induced TrkB signaling contributes to this type of LTP. Our results prove for the first time a mechanistic action of acute BDNF/TrkB signaling in presynaptic expression of MF-LTP in adult hippocampus.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , CA3 Region, Hippocampal/metabolism , Long-Term Potentiation , Mossy Fibers, Hippocampal/metabolism , Receptor, trkB/metabolism , Signal Transduction , Synapses/metabolism , Animals , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Brain-Derived Neurotrophic Factor/genetics , CA3 Region, Hippocampal/drug effects , Heterozygote , Humans , Long-Term Potentiation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mossy Fibers, Hippocampal/drug effects , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Inhibition/drug effects , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Protein Kinase Inhibitors/pharmacology , Receptor, trkB/antagonists & inhibitors , Signal Transduction/drug effects , Synapses/drug effects , Synaptic Transmission/drug effects , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...