Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Neuron ; 111(11): 1760-1775.e8, 2023 06 07.
Article in English | MEDLINE | ID: mdl-36996810

ABSTRACT

The proteome of glutamatergic synapses is diverse across the mammalian brain and involved in neurodevelopmental disorders (NDDs). Among those is fragile X syndrome (FXS), an NDD caused by the absence of the functional RNA-binding protein FMRP. Here, we demonstrate how the brain region-specific composition of postsynaptic density (PSD) contributes to FXS. In the striatum, the FXS mouse model shows an altered association of the PSD with the actin cytoskeleton, reflecting immature dendritic spine morphology and reduced synaptic actin dynamics. Enhancing actin turnover with constitutively active RAC1 ameliorates these deficits. At the behavioral level, the FXS model displays striatal-driven inflexibility, a typical feature of FXS individuals, which is rescued by exogenous RAC1. Striatal ablation of Fmr1 is sufficient to recapitulate behavioral impairments observed in the FXS model. These results indicate that dysregulation of synaptic actin dynamics in the striatum, a region largely unexplored in FXS, contributes to the manifestation of FXS behavioral phenotypes.


Subject(s)
Fragile X Syndrome , Animals , Mice , Fragile X Mental Retardation Protein/genetics , Actins/metabolism , Brain/metabolism , Disease Models, Animal , Mice, Knockout , Dendritic Spines/metabolism , Mammals/metabolism
2.
Nat Commun ; 14(1): 763, 2023 02 20.
Article in English | MEDLINE | ID: mdl-36808152

ABSTRACT

Sleep behavior is conserved throughout evolution, and sleep disturbances are a frequent comorbidity of neuropsychiatric disorders. However, the molecular basis underlying sleep dysfunctions in neurological diseases remains elusive. Using a model for neurodevelopmental disorders (NDDs), the Drosophila Cytoplasmic FMR1 interacting protein haploinsufficiency (Cyfip85.1/+), we identify a mechanism modulating sleep homeostasis. We show that increased activity of the sterol regulatory element-binding protein (SREBP) in Cyfip85.1/+ flies induces an increase in the transcription of wakefulness-associated genes, such as the malic enzyme (Men), causing a disturbance in the daily NADP+/NADPH ratio oscillations and reducing sleep pressure at the night-time onset. Reduction in SREBP or Men activity in Cyfip85.1/+ flies enhances the NADP+/NADPH ratio and rescues the sleep deficits, indicating that SREBP and Men are causative for the sleep deficits in Cyfip heterozygous flies. This work suggests modulation of the SREBP metabolic axis as a new avenue worth exploring for its therapeutic potential in sleep disorders.


Subject(s)
Drosophila Proteins , Sterol Regulatory Element Binding Proteins , Animals , Sterol Regulatory Element Binding Proteins/metabolism , NADP/metabolism , Drosophila/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Sleep , Sterol Regulatory Element Binding Protein 2/metabolism , Drosophila Proteins/metabolism , Fragile X Mental Retardation Protein
3.
EMBO Rep ; 22(4): e51404, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33779029

ABSTRACT

Status epilepticus (SE) is a condition in which seizures are not self-terminating and thereby pose a serious threat to the patient's life. The molecular mechanisms underlying SE are likely heterogeneous and not well understood. Here, we reveal a role for the RNA-binding protein Fragile X-Related Protein 2 (FXR2P) in SE. Fxr2 KO mice display reduced sensitivity specifically to kainic acid-induced SE. Immunoprecipitation of FXR2P coupled to next-generation sequencing of associated mRNAs shows that FXR2P targets are enriched in genes that encode glutamatergic post-synaptic components. Of note, the FXR2P target transcriptome has a significant overlap with epilepsy and SE risk genes. In addition, Fxr2 KO mice fail to show sustained ERK1/2 phosphorylation induced by KA and present reduced burst activity in the hippocampus. Taken together, our findings show that the absence of FXR2P decreases the expression of glutamatergic proteins, and this decrease might prevent self-sustained seizures.


Subject(s)
Kainic Acid , Status Epilepticus , Animals , Hippocampus/metabolism , Kainic Acid/toxicity , Mice , Mice, Inbred C57BL , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Seizures/chemically induced , Seizures/genetics , Status Epilepticus/chemically induced , Status Epilepticus/genetics
4.
Nat Commun ; 10(1): 3454, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31371726

ABSTRACT

Copy-number variants of the CYFIP1 gene in humans have been linked to autism spectrum disorders (ASD) and schizophrenia (SCZ), two neuropsychiatric disorders characterized by defects in brain connectivity. Here, we show that CYFIP1 plays an important role in brain functional connectivity and callosal functions. We find that Cyfip1-heterozygous mice have reduced functional connectivity and defects in white matter architecture, similar to phenotypes found in patients with ASD, SCZ and other neuropsychiatric disorders. Cyfip1-deficient mice also present decreased myelination in the callosal axons, altered presynaptic function, and impaired bilateral connectivity. Finally, Cyfip1 deficiency leads to abnormalities in motor coordination, sensorimotor gating and sensory perception, which are also known neuropsychiatric disorder-related symptoms. These results show that Cyfip1 haploinsufficiency compromises brain connectivity and function, which might explain its genetic association to neuropsychiatric disorders.


Subject(s)
Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Brain/metabolism , Genetic Predisposition to Disease/genetics , Nerve Tissue Proteins/metabolism , Schizophrenia/metabolism , Adaptor Proteins, Signal Transducing , Animals , Autism Spectrum Disorder/diagnostic imaging , Axons , Behavior, Animal , Brain/diagnostic imaging , DNA Copy Number Variations , Disease Models, Animal , Genetic Association Studies , Haploinsufficiency , Heterozygote , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nervous System/metabolism , Nervous System Physiological Phenomena/genetics , Phenotype , Psychomotor Performance , Schizophrenia/diagnostic imaging , Schizophrenia/genetics , Sensory Gating , White Matter
5.
Nat Commun ; 8(1): 293, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28819097

ABSTRACT

The brain cytoplasmic (BC1) RNA is a non-coding RNA (ncRNA) involved in neuronal translational control. Absence of BC1 is associated with altered glutamatergic transmission and maladaptive behavior. Here, we show that pyramidal neurons in the barrel cortex of BC1 knock out (KO) mice display larger excitatory postsynaptic currents and increased spontaneous activity in vivo. Furthermore, BC1 KO mice have enlarged spine heads and postsynaptic densities and increased synaptic levels of glutamate receptors and PSD-95. Of note, BC1 KO mice show aberrant structural plasticity in response to whisker deprivation, impaired texture novel object recognition and altered social behavior. Thus, our study highlights a role for BC1 RNA in experience-dependent plasticity and learning in the mammalian adult neocortex, and provides insight into the function of brain ncRNAs regulating synaptic transmission, plasticity and behavior, with potential relevance in the context of intellectual disabilities and psychiatric disorders.Brain cytoplasmic (BC1) RNA is a non-coding RNA that has been implicated in translational regulation, seizure, and anxiety. Here, the authors show that in the cortex, BC1 RNA is required for sensory deprivation-induced structural plasticity of dendritic spines, as well as for correct sensory learning and social behaviors.


Subject(s)
Learning/physiology , Neocortex/physiology , Neuronal Plasticity/physiology , Pyramidal Cells/physiology , RNA, Small Cytoplasmic/genetics , Animals , Base Sequence , Cells, Cultured , Dendritic Spines/metabolism , Dendritic Spines/physiology , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , In Situ Hybridization, Fluorescence , Male , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Neocortex/cytology , Neocortex/metabolism , Neuronal Plasticity/genetics , Pyramidal Cells/metabolism , Pyramidal Cells/ultrastructure , Sensory Deprivation/physiology , Sequence Homology, Nucleic Acid , Social Behavior , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Vibrissae/metabolism , Vibrissae/physiology
6.
Sci Rep ; 6: 36489, 2016 11 07.
Article in English | MEDLINE | ID: mdl-27819338

ABSTRACT

Intracerebral injection of the excitotoxic, endogenous tryptophan metabolite, quinolinic acid (QA), constitutes a chemical model of neurodegenerative brain disease. Complementary techniques were combined to examine the consequences of QA injection into medial prefrontal cortex (mPFC) of C57BL6 mice. In accordance with the NMDAR-mediated synapto- and neurotoxic action of QA, we found an initial increase in excitability and an augmentation of hippocampal long-term potentiation, converting within two weeks into a reduction and impairment, respectively, of these processes. QA-induced mPFC excitotoxicity impaired behavioral flexibility in a reversal variant of the hidden-platform Morris water maze (MWM), whereas regular, extended MWM training was unaffected. QA-induced mPFC damage specifically affected the spatial-cognitive strategies that mice use to locate the platform during reversal learning. These behavioral and cognitive defects coincided with changes in cortical functional connectivity (FC) and hippocampal neuroplasticity. FC between various cortical regions was assessed by resting-state fMRI (rsfMRI) methodology, and mice that had received QA injection into mPFC showed increased FC between various cortical regions. mPFC and hippocampus (HC) are anatomically as well as functionally linked as part of a cortical network that controls higher-order cognitive functions. Together, these observations demonstrate the central functional importance of rodent mPFC as well as the validity of QA-induced mPFC damage as a preclinical rodent model of the early stages of neurodegeneration.


Subject(s)
Hippocampus/drug effects , Neuronal Plasticity/drug effects , Prefrontal Cortex/drug effects , Quinolinic Acid/pharmacology , Reversal Learning/drug effects , Animals , Female , Hippocampus/metabolism , Long-Term Potentiation/drug effects , Magnetic Resonance Imaging/methods , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Prefrontal Cortex/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
7.
Front Behav Neurosci ; 10: 83, 2016.
Article in English | MEDLINE | ID: mdl-27199692

ABSTRACT

Prenatal irradiation is known to perturb brain development. Epidemiological studies revealed that radiation exposure during weeks 8-15 of pregnancy was associated with an increased occurrence of mental disability and microcephaly. Such neurological deficits were reproduced in animal models, in which rodent behavioral testing is an often used tool to evaluate radiation-induced defective brain functionality. However, up to now, animal studies suggested a threshold dose of around 0.30 Gray (Gy) below which no behavioral alterations can be observed, while human studies hinted at late defects after exposure to doses as low as 0.10 Gy. Here, we acutely irradiated pregnant mice at embryonic day 11 with doses ranging from 0.10 to 1.00 Gy. A thorough investigation of the dose-response relationship of altered brain function and architecture following in utero irradiation was achieved using a behavioral test battery and volumetric 3D T2-weighted magnetic resonance imaging (MRI). We found dose-dependent changes in cage activity, social behavior, anxiety-related exploration, and spatio-cognitive performance. Although behavioral alterations in low-dose exposed animals were mild, we did unveil that both emotionality and higher cognitive abilities were affected in mice exposed to ≥0.10 Gy. Microcephaly was apparent from 0.33 Gy onwards and accompanied by deviations in regional brain volumes as compared to controls. Of note, total brain volume and the relative volume of the ventricles, frontal and posterior cerebral cortex, cerebellum, and striatum were most strongly correlated to altered behavioral parameters. Taken together, we present conclusive evidence for persistent low-dose effects after prenatal irradiation in mice and provide a better understanding of the correlation between their brain size and performance in behavioral tests.

8.
Sci Transl Med ; 7(309): 309ra164, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26468326

ABSTRACT

The orphan G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptor (GPCR) GPR3 regulates activity of the γ-secretase complex in the absence of an effect on Notch proteolysis, providing a potential therapeutic target for Alzheimer's disease (AD). However, given the vast resources required to develop and evaluate any new therapy for AD and the multiple failures involved in translational research, demonstration of the pathophysiological relevance of research findings in multiple disease-relevant models is necessary before initiating costly drug development programs. We evaluated the physiological consequences of loss of Gpr3 in four AD transgenic mouse models, including two that contain the humanized murine Aß sequence and express similar amyloid precursor protein (APP) levels as wild-type mice, thereby reducing potential artificial phenotypes. Our findings reveal that genetic deletion of Gpr3 reduced amyloid pathology in all of the AD mouse models and alleviated cognitive deficits in APP/PS1 mice. Additional three-dimensional visualization and analysis of the amyloid plaque burden provided accurate information on the amyloid load, distribution, and volume in the structurally intact adult mouse brain. Analysis of 10 different regions in healthy human postmortem brain tissue indicated that GPR3 expression was stable during aging. However, two cohorts of human AD postmortem brain tissue samples showed a correlation between elevated GPR3 and AD progression. Collectively, these studies provide evidence that GPR3 mediates the amyloidogenic proteolysis of APP in four AD transgenic mouse models as well as the physiological processing of APP in wild-type mice, suggesting that GPR3 may be a potential therapeutic target for AD drug development.


Subject(s)
Alzheimer Disease/physiopathology , Alzheimer Disease/therapy , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/physiology , Animals , Brain/physiology , Gene Deletion , Gene Expression , Humans , Mice , Mice, Transgenic , Models, Animal , Plaque, Amyloid/pathology
9.
Neurobiol Aging ; 36(1): 228-40, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25443293

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder hallmarked by the accumulation of extracellular amyloid-ß (Aß) peptide and intraneuronal hyperphosphorylated tau, as well as chronic neuroinflammation. Tauroursodeoxycholic acid (TUDCA) is an endogenous anti-apoptotic bile acid with potent neuroprotective properties in several experimental models of AD. We have previously reported the therapeutic efficacy of TUDCA treatment before amyloid plaque deposition in APP/PS1 double-transgenic mice. In the present study, we evaluated the protective effects of TUDCA when administrated after the onset of amyloid pathology. APP/PS1 transgenic mice with 7 months of age were injected intraperitoneally with TUDCA (500 mg/kg) every 3 days for 3 months. TUDCA treatment significantly attenuated Aß deposition in the brain, with a concomitant decrease in Aß1₋40 and Aß1₋42 levels. The amyloidogenic processing of amyloid precursor protein was also reduced, indicating that TUDCA interferes with Aß production. In addition, TUDCA abrogated GSK3ß hyperactivity, which is highly implicated in tau hyperphosphorylation and glial activation. This effect was likely dependent on the specific activation of the upstream kinase, Akt. Finally, TUDCA treatment decreased glial activation and reduced proinflammatory cytokine messenger RNA expression, while partially rescuing synaptic loss. Overall, our results suggest that TUDCA is a promising therapeutic strategy not only for prevention but also for treatment of AD after disease onset.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Protein Aggregation, Pathological/drug therapy , Protein Aggregation, Pathological/genetics , Taurochenodeoxycholic Acid/administration & dosage , Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Amyloid beta-Protein Precursor/genetics , Animals , Cholagogues and Choleretics , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Inflammation Mediators/metabolism , Mice, Transgenic , Molecular Targeted Therapy , Neuroprotective Agents , Presenilin-1/genetics , Protein Aggregation, Pathological/pathology , Protein Aggregation, Pathological/prevention & control , RNA, Messenger/metabolism , Taurochenodeoxycholic Acid/pharmacology , tau Proteins/metabolism
10.
Neuropharmacology ; 85: 178-89, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24863046

ABSTRACT

5-HT4 receptors (5-HT4R) are suggested to affect learning and memory processes. Earlier studies have shown that animals treated with 5-HT4R agonists, often with limited selectivity, show improved learning and memory with retention memory often being assessed immediately after or within 24 h after the last training session. In this study, we characterized the effect of pre-training treatment with the selective 5-HT4R agonist SSP-002392 on memory acquisition and the associated long-term memory retrieval in animal models of impaired cognition. Pre-training treatment with SSP-002392 (0.3 mg/kg, 1.5 mg/kg and 7.5 mg/kg p.o.) dose-dependently inhibited the cognitive deficits induced by scopolamine (0.5 mg/kg s.c.) in two different behavioral tasks: passive avoidance and Morris water maze. In the Morris water maze, spatial learning was significantly improved after treatment with SSP-002392 translating in an accelerated and more efficient localization of the hidden platform compared to scopolamine-treated controls. Moreover, retention memory was assessed 24 h (passive avoidance) and 72 h (Morris water maze) after the last training session of cognitive-impaired animals and this was significantly improved in animals treated with SSP-002392 prior to the training sessions. Furthermore, the effects of SSP-002392 were comparable to galanthamine hydrobromide. We conclude that SSP-002392 has potential as a memory-enhancing compound.


Subject(s)
Avoidance Learning/drug effects , Benzofurans/pharmacology , Maze Learning/drug effects , Memory Disorders/drug therapy , Nootropic Agents/pharmacology , Piperidines/pharmacology , Receptors, Serotonin, 5-HT4/metabolism , Animals , Anxiety/drug therapy , Anxiety/physiopathology , Avoidance Learning/physiology , Disease Models, Animal , Dose-Response Relationship, Drug , Fear/drug effects , Fear/physiology , Galantamine/pharmacology , Male , Maze Learning/physiology , Memory Disorders/physiopathology , Mice, Inbred C57BL , Random Allocation , Scopolamine , Serotonin 5-HT4 Receptor Agonists/pharmacology
11.
Neuropharmacology ; 75: 458-66, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24035915

ABSTRACT

In the Alzheimer's disease (AD) brain, accumulation of Aß1-42 peptides is suggested to initiate a cascade of pathological events. To date, no treatments are available that can reverse or delay AD-related symptoms in patients. In the current study, we introduce a new Aß toxicity inhibitor, SEN1500, which in addition to its block effect on Aß1-42 toxicity in synaptophysin assays, can be administered orally and cross the blood-brain barrier without adverse effects in mice. In a different set of animals, APPPS1-21 mice were fed with three different doses of SEN1500 (1 mg/kg, 5 mg/kg and 20 mg/kg) for a period of 5 months. Cognition was assessed in a variety of behavioral tests (Morris water maze, social recognition, conditioned taste aversion and passive avoidance). Results suggest a positive effect on cognition with 20 mg/kg SEN1500 compared to control APPPS1-21 mice. However, no changes in soluble or insoluble Aß1-40 and Aß1-42 were detected in the brains of SEN1500-fed mice. SEN1500 also attenuated the effect of Aß1-42 on synaptophysin levels in mouse cortical neurons, which indicated that the compound blocked the synaptic toxicity of Aß1-42. In vitro and in vivo effects presented here suggest that SEN1500 could be an interesting AD therapeutic.


Subject(s)
Alzheimer Disease/complications , Amyloid beta-Peptides/antagonists & inhibitors , Learning Disabilities/drug therapy , Learning Disabilities/etiology , Memory Disorders/etiology , Nitriles/administration & dosage , Peptide Fragments/antagonists & inhibitors , Administration, Oral , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Animals , Avoidance Learning/drug effects , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Maze Learning/drug effects , Mice , Mice, Transgenic , Mutation/genetics , Nitriles/chemistry , Presenilin-1/genetics , Pyrimidines/administration & dosage , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Synaptophysin/metabolism , Taste/drug effects
12.
J Alzheimers Dis ; 37(4): 777-88, 2013.
Article in English | MEDLINE | ID: mdl-23948912

ABSTRACT

Age-related cognitive decline and neurodegenerative diseases are a growing challenge for society. Accumulation of tau pathology has been proposed to partially contribute to these impairments. This study provides a behavioral characterization during aging of transgenic mice bearing tau mutations. THY-Tau22 mice were evaluated at ages wherein tau neuropathology in this transgenic mouse model is low (3-4 months), moderate (6-7 months), or extensive (>9 months). Spatial memory was found to be impaired only after 9 months of age in THY-Tau22 mice, whereas non-spatial memory was affected as early as 6 months, appearing to offer an opportunity for assessing potential therapeutic agents in attenuating or preventing tauopathies through modulation of tau kinetics.


Subject(s)
Aging/genetics , Aging/metabolism , Cognition Disorders/genetics , Cognition Disorders/metabolism , tau Proteins/genetics , tau Proteins/metabolism , Aging/pathology , Animals , Cognition Disorders/pathology , Disease Progression , Male , Maze Learning/physiology , Mice , Mice, Transgenic , Phosphorylation/genetics
13.
J Alzheimers Dis ; 37(1): 109-25, 2013.
Article in English | MEDLINE | ID: mdl-23788007

ABSTRACT

Alzheimer's disease (AD) is a consequence of degenerative brain pathology with amyloid plaque deposition and neurofibrillary tangle formation. These distinct aspects of AD neuropathology have been suggested to induce a cascade of pathological events ultimately leading to neurodegeneration as well as cognitive and behavioral decline. Amyloid and tau neuropathology is known to develop along distinct stages and affect parts of the brain differentially. In this study, we examined two mouse AD lines (AßPPPS1-21 and Tau22 mice), which mimic different partial aspects of AD pathology, at comparable stages of their pathology. Since prefrontal cortex (PFC) is one of the first regions to be affected in clinical AD, we compared long-term potentiation (LTP) of synaptic responses in medial PFC of AßPPPS1-21 and Tau22 mice. Frontal LTP was impaired in AßPPPS1-21 mice, but not in Tau22 mice. Consequently, we observed different behavioral defects between AßPPPS1-21 and Tau22 animals. Apart from spatial learning deficits, AßPPPS1-21 transgenic mice were impaired in fear learning, aversion learning, and extinction learning, whereas THY-Tau22 were impaired in appetitive responding. Discriminant function analysis identified critical behavioral variables that differentiated AßPPPS1-21 and THY-Tau22 mice from wild type littermates, and further confirmed that amyloid- versus tau-pathology differentially affects brain function.


Subject(s)
Alzheimer Disease/pathology , Cognition Disorders/pathology , Disease Models, Animal , Prefrontal Cortex/pathology , Synapses/pathology , tau Proteins , Alzheimer Disease/genetics , Alzheimer Disease/psychology , Amyloid beta-Protein Precursor/genetics , Animals , Cognition Disorders/genetics , Cognition Disorders/psychology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity/physiology , Plaque, Amyloid/genetics , Plaque, Amyloid/pathology , Presenilin-1/genetics , Synapses/genetics , tau Proteins/genetics
14.
Science ; 340(6135): 924-e, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23704554

ABSTRACT

Cramer et al. (Reports, 23 March 2012, p. 1503; published online 9 February 2012) tested bexarotene as a potential ß-amyloid-lowering drug for Alzheimer's disease (AD). We were not able to reproduce the described effects in several animal models. Drug formulation appears very critical. Our data call for extreme caution when considering this compound for use in AD patients.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apolipoproteins E/metabolism , Brain/metabolism , Tetrahydronaphthalenes/pharmacology , Tetrahydronaphthalenes/therapeutic use , Animals , Male
15.
Neurobiol Aging ; 34(7): 1779-89, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23474291

ABSTRACT

Lowering the production and accumulation of Aß has been explored as treatment for Alzheimer's disease (AD), because Aß is postulated to play an important role in the pathogenesis of AD. 5-HT4 receptors are an interesting drug target in this regard, as their activation might stimulate α-secretase processing, which increases sAPPα and reduces Aß, at least according to the central dogma in APP processing. Here we describe a novel high-affinity 5-HT4 receptor agonist SSP-002392 that, in cultured human neuroblastoma cells, potently increases the levels of cAMP and sAPPα at 100-fold lower concentrations than the effective concentrations of prucalopride, a known selective 5-HT4 receptor agonist. Chronic administration of this compound in a hAPP/PS1 mouse model of Alzheimer's disease decreased soluble and insoluble Aß in hippocampus, but the potential mechanisms underlying these observations seem to be complex. We found no evidence for direct α-secretase stimulation in the brain in vivo, but observed decreased APP and BACE-1 expression and elevated astroglia and microglia responses. Taken together these results provide support for a potential disease-modifying aspect when stimulating central 5-HT4 receptors; however, the complexity of the phenomena warrants further research.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Peptides/metabolism , Presenilin-1/genetics , Receptors, Serotonin, 5-HT4/metabolism , Serotonin Receptor Agonists/pharmacology , Serotonin Receptor Agonists/therapeutic use , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/antagonists & inhibitors , Amyloid beta-Protein Precursor/biosynthesis , Amyloid beta-Protein Precursor/genetics , Animals , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic
16.
BMC Genomics ; 14: 181, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23496794

ABSTRACT

BACKGROUND: Simultaneous isolation of nucleic acids and proteins from a single biological sample facilitates meaningful data interpretation and reduces time, cost and sampling errors. This is particularly relevant for rare human and animal specimens, often scarce, and/or irreplaceable. TRIzol(®) and TRIzol(®)LS are suitable for simultaneous isolation of RNA, DNA and proteins from the same biological sample. These reagents are widely used for RNA and/or DNA isolation, while reports on their use for protein extraction are limited, attributable to technical difficulties in protein solubilisation. RESULTS: TRIzol(®)LS was used for RNA isolation from 284 human colon cancer samples, including normal colon mucosa, tubulovillous adenomas, and colon carcinomas with proficient and deficient mismatch repair system. TRIzol(®) was used for RNA isolation from human colon cancer cells, from brains of transgenic Alzheimer's disease mice model, and from cultured mouse cortical neurons. Following RNA extraction, the TRIzol(®)-chloroform fractions from human colon cancer samples and from mouse hippocampus and frontal cortex were stored for 2 years and 3 months, respectively, at -80°C until used for protein isolation.Simple modifications to the TRIzol(®) manufacturer's protocol, including Urea:SDS solubilization and sonication, allowed improved protein recovery yield compared to the TRIzol(®) manufacturer's protocol. Following SDS-PAGE and Ponceau and Coomassie staining, recovered proteins displayed wide molecular weight range and staining pattern comparable to those obtainable with commonly used protein extraction protocols. We also show that nuclear and cytosolic proteins can be easily extracted and detected by immunoblotting, and that posttranslational modifications, such as protein phosphorylation, are detectable in proteins recovered from TRIzol(®)-chloroform fractions stored for up to 2 years at -80°C. CONCLUSIONS: We provide a novel approach to improve protein recovery from samples processed for nucleic acid extraction with TRIzol(®) and TRIzol(®)LS compared to the manufacturer`s protocol, allowing downstream immunoblotting and evaluation of steady-state relative protein expression levels. The method was validated in large sets of samples from multiple sources, including human colon cancer and brains of transgenic Alzheimer's disease mice model, stored in TRIzol(®)-chloroform for up to two years. Collectively, we provide a faster and cheaper alternative to the TRIzol(®) manufacturer`s protein extraction protocol, illustrating the high relevance, and wide applicability, of the present protein isolation method for the immunoblot evaluation of steady-state relative protein expression levels in samples from multiple sources, and following prolonged storage.


Subject(s)
Proteins/metabolism , RNA/metabolism , Adenoma, Villous/metabolism , Adenoma, Villous/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Brain/metabolism , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Cells, Cultured , Chloroform/chemistry , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Models, Animal , HCT116 Cells , Hippocampus/metabolism , Humans , Mice , Neurons/cytology , Neurons/metabolism , Phosphorylation , Protein Stability , Proteins/chemistry , Proteins/isolation & purification , RNA/isolation & purification , RNA Stability , Reagent Kits, Diagnostic , Urea/chemistry
17.
Neurobiol Dis ; 50: 21-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22974733

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease hallmarked by extracellular Aß(1-42) containing plaques, and intracellular neurofibrillary tangles (NFT) containing hyperphosphorylated tau protein. Progressively, memory deficits and cognitive disabilities start to occur as these hallmarks affect hippocampus and frontal cortex, regions highly involved in memory. Connective tissue growth factor (CTGF) expression, which is high in the vicinity of Aß plaques and NFTs, was found to influence γ-secretase activity, the molecular crux in Aß(1-42) production. Tauroursodeoxycholic acid (TUDCA) is an endogenous bile acid that downregulates CTGF expression in hepatocytes and has been shown to possess therapeutic efficacy in neurodegenerative models. To investigate the possible in vivo therapeutic effects of TUDCA, we provided 0.4% TUDCA-supplemented food to APP/PS1 mice, a well-established AD mouse model. Six months of TUDCA supplementation prevented the spatial, recognition and contextual memory defects observed in APP/PS1 mice at 8 months of age. Furthermore, TUDCA-supplemented APP/PS1 mice displayed reduced hippocampal and prefrontal amyloid deposition. These effects of TUDCA supplementation suggest a novel mechanistic route for Alzheimer therapeutics.


Subject(s)
Alzheimer Disease/complications , Amyloidogenic Proteins/drug effects , Cholagogues and Choleretics/pharmacology , Cognition Disorders/prevention & control , Taurochenodeoxycholic Acid/pharmacology , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloidogenic Proteins/metabolism , Animals , Behavior, Animal/drug effects , Cognition Disorders/pathology , Dietary Supplements , Disease Models, Animal , Male , Mice , Mice, Transgenic , Presenilin-1/genetics
18.
Neurobiol Aging ; 34(2): 551-61, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22621777

ABSTRACT

Synapses are considered the earliest site of Alzheimer's disease (AD) pathology, where synapse density is reduced, and synaptic loss is highly correlated with cognitive impairment. Tauroursodeoxycholic acid (TUDCA) has been shown to be neuroprotective in several models of AD, including neuronal exposure to amyloid ß (Aß) and amyloid precursor protein (APP)/presenilin 1 (PS1) double-transgenic mice. Here, we show that TUDCA modulates synaptic deficits induced by Aß in vitro. Specifically, TUDCA reduced the downregulation of the postsynaptic marker postsynaptic density-95 (PSD-95) and the decrease in spontaneous miniature excitatory postsynaptic currents (mEPSCs) frequency, while increasing the number of dendritic spines. This contributed to the induction of more robust and synaptically efficient neurons, reflected in inhibition of neuronal death. In vivo, TUDCA treatment of APP/PS1 mice abrogated the decrease in PSD-95 reactivity in the hippocampus. Taken together, these results expand the neuroprotective role of TUDCA to a synaptic level, further supporting the use of this molecule as a potential therapeutic strategy for the prevention and treatment of AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/pharmacology , Neuroprotective Agents/pharmacology , Synapses/drug effects , Taurochenodeoxycholic Acid/pharmacology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cell Death/drug effects , Cell Death/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Disease Models, Animal , Down-Regulation , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism , Rats , Rats, Wistar , Synapses/metabolism
19.
Mol Neurobiol ; 45(3): 440-54, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22438081

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by accumulation of amyloid-ß (Aß) peptide in the hippocampus and frontal cortex of the brain, leading to progressive cognitive decline. The endogenous bile acid tauroursodeoxycholic acid (TUDCA) is a strong neuroprotective agent in several experimental models of disease, including neuronal exposure to Aß. Nevertheless, the therapeutic role of TUDCA in AD pathology has not yet been ascertained. Here we report that feeding APP/PS1 double-transgenic mice with diet containing 0.4 % TUDCA for 6 months reduced accumulation of Aß deposits in the brain, markedly ameliorating memory deficits. This was accompanied by reduced glial activation and neuronal integrity loss in TUDCA-fed APP/PS1 mice compared to untreated APP/PS1 mice. Furthermore, TUDCA regulated lipid-metabolism mediators involved in Aß production and accumulation in the brains of transgenic mice. Overall amyloidogenic APP processing was reduced with TUDCA treatment, in association with, but not limited to, modulation of γ-secretase activity. Consequently, a significant decrease in Aß(1-40) and Aß(1-42) levels was observed in both hippocampus and frontal cortex of TUDCA-treated APP/PS1 mice, suggesting that chronic feeding of TUDCA interferes with Aß production, possibly through the regulation of lipid-metabolism mediators associated with APP processing. These results highlight TUDCA as a potential therapeutic strategy for the prevention and treatment of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Bile Acids and Salts/pharmacology , Presenilin-1/metabolism , Protein Processing, Post-Translational/drug effects , Taurochenodeoxycholic Acid/pharmacology , Alzheimer Disease/complications , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Bile Acids and Salts/therapeutic use , Brain/drug effects , Brain/metabolism , Brain/pathology , Cognition Disorders/complications , Cognition Disorders/drug therapy , Cognition Disorders/pathology , DNA-Binding Proteins , Humans , Lipid Metabolism/drug effects , Mice , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nuclear Proteins/metabolism , Synucleins/metabolism , Taurochenodeoxycholic Acid/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...