Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Mol Ther ; 31(4): 1088-1105, 2023 04 05.
Article in English | MEDLINE | ID: mdl-36641623

ABSTRACT

Angelman syndrome (AS) is a neurogenetic disorder caused by the loss of ubiquitin ligase E3A (UBE3A) gene expression in the brain. The UBE3A gene is paternally imprinted in brain neurons. Clinical features of AS are primarily due to the loss of maternally expressed UBE3A in the brain. A healthy copy of paternal UBE3A is present in the brain but is silenced by a long non-coding antisense transcript (UBE3A-ATS). Here, we demonstrate that an artificial transcription factor (ATF-S1K) can silence Ube3a-ATS in an adult mouse model of Angelman syndrome (AS) and restore endogenous physiological expression of paternal Ube3a. A single injection of adeno-associated virus (AAV) expressing ATF-S1K (AAV-S1K) into the tail vein enabled whole-brain transduction and restored UBE3A protein in neurons to ∼25% of wild-type protein. The ATF-S1K treatment was highly specific to the target site with no detectable inflammatory response 5 weeks after AAV-S1K administration. AAV-S1K treatment of AS mice showed behavioral rescue in exploratory locomotion, a task involving gross and fine motor abilities, similar to low ambulation and velocity in AS patients. The specificity and tolerability of a single injection of AAV-S1K therapy for AS demonstrate the use of ATFs as a promising translational approach for AS.


Subject(s)
Angelman Syndrome , Animals , Mice , Angelman Syndrome/genetics , Angelman Syndrome/therapy , Angelman Syndrome/metabolism , Brain/metabolism , Gene Expression Regulation , Transcription Factors/genetics , Phenotype , Ubiquitin-Protein Ligases/genetics
2.
Trends Genet ; 35(7): 527-541, 2019 07.
Article in English | MEDLINE | ID: mdl-31128888

ABSTRACT

Epigenome editing refers to the generation of precise chromatin alterations and their effects on gene expression and cell biology. Until recently, much of the efforts in epigenome editing were limited to tissue culture models of disease. However, the convergence of techniques from different fields including mammalian genetics, virology, and CRISPR engineering is advancing epigenome editing into a new era. Researchers are increasingly embracing the use of multicellular model organisms to test the role of specific chromatin alterations in development and disease. The challenge of successful live-animal epigenomic editing will depend on a well-informed foundation of the current methodologies for cell-specific delivery and editing accuracy. Here we review the opportunities for basic research and therapeutic applications.


Subject(s)
Epigenome , Epigenomics/methods , Gene Editing/methods , Animals , Cell Transplantation/methods , Clustered Regularly Interspaced Short Palindromic Repeats , Dependovirus/genetics , Mice, Transgenic , Zinc Fingers/genetics
3.
Hum Mol Genet ; 28(22): 3842-3852, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31625566

ABSTRACT

Ubiquitin E3 ligase 3A (UBE3A) encodes an E3 ubiquitin ligase whose loss from the maternal allele causes the neurodevelopmental disorder Angelman syndrome (AS). Previous studies of UBE3A function have not examined full Ube3a deletion in mouse, the complexity of imprinted gene networks in brain nor the molecular basis of systems-level cognitive dysfunctions in AS. We therefore utilized a systems biology approach to elucidate how UBE3A loss impacts the early postnatal brain in a novel CRISPR/Cas9-engineered rat Angelman model of a complete Ube3a deletion. Strand-specific transcriptome analysis of offspring from maternally or paternally inherited Ube3a deletions revealed the expected parental expression patterns of Ube3a sense and antisense transcripts by postnatal day 2 (P2) in hypothalamus and day 9 (P9) in cortex, compared to wild-type littermates. The dependency of genome-wide effects on parent-of-origin, Ube3a genotype and time (P2 and P9) was investigated through transcriptome (RNA sequencing of cortex and hypothalamus) and methylome (whole-genome bisulfite sequencing of hypothalamus). Weighted gene co-expression and co-methylation network analyses identified co-regulated networks in maternally inherited Ube3a deletion offspring enriched in postnatal developmental processes including Wnt signaling, synaptic regulation, neuronal and glial functions, epigenetic regulation, ubiquitin, circadian entrainment and splicing. Furthermore, we showed that loss of the paternal Ube3a antisense transcript resulted in both unique and overlapping dysregulated gene pathways with maternal loss, predominantly at the level of differential methylation. Together, these results provide a holistic examination of the molecular impacts of UBE3A loss in brain, supporting the existence of interactive epigenetic networks between maternal and paternal transcripts at the Ube3a locus.


Subject(s)
Genomic Imprinting , Ubiquitin-Protein Ligases/genetics , Angelman Syndrome/genetics , Angelman Syndrome/metabolism , Animals , Brain/metabolism , Cerebral Cortex/metabolism , Epigenesis, Genetic , Female , Gene Expression , Gene Expression Profiling/methods , Gene Regulatory Networks/genetics , Hypothalamus/metabolism , Neuroglia/metabolism , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Synapses/genetics , Synapses/metabolism , Systems Biology , Transcriptome , Ubiquitin-Protein Ligases/metabolism , Wnt Signaling Pathway
4.
Pharmacol Res ; 113(Pt B): 781-787, 2016 11.
Article in English | MEDLINE | ID: mdl-27345857

ABSTRACT

The prevalence of Alzheimer's disease (AD) is higher in females than in males, and causes more severe cognitive, memory and behavioral impairments. Previously, in male transgenic (Tg) APPSweDI mice, we reported that the novel lipophilic 1,4-dihydropyridine (DHP) derivative AP-12 crossed the blood-brain barrier, blocked neuronal and vascular calcium channels, changed brain protein expression and improved behavior. In this study, we used female Tg APPSweDI mice to assess the effects of AP-12 on behavior, and brain protein expression, with a particular focus on those of the GABAergic system. The results showed that in female Tg mice, similar to male Tg mice, AP-12 improved spatial learning/memory performance in the water maze test and demonstrated anxiolytic effect in the elevated zero maze (after single administration of AP-12) and elevated plus maze (after chronic injections of AP-12). In addition, we demonstrated upregulated expression of glutamate decarboxylase 67 (GAD67) and vesicular GABA transporter (VGAT) in the cingulate cortex and hippocampus, pointing to the role of the GABAergic system as one of the neural networks dysregulated in AD. In both female and male mice, AP-12 did not change the expression of hippocampal Homer-1, a protein which is involved in synaptic plasticity. However, in cingulate cortex, the staining density of Homer-1 was significantly increased in female mice. Further, female mice (similar to male mice) did not show changes in brain AChE expression and in the amyloid beta load in the hippocampus and cingulate cortex. In conclusion, the memory enhancing, anxiolytic and protein expression effects of AP-12 did not show sex specificity in APPSweDI mice. Considering the ability of AP-12 to block brain calcium channels and improve memory by enhancing the GABAergic and synaptic plasticity processes, AP-12 is a promising compound which merits further pre-clinical studies to investigate its usefulness in the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Calcium Channel Blockers/pharmacology , Calcium/metabolism , Gyrus Cinguli/drug effects , Hippocampus/drug effects , Memory/drug effects , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Anti-Anxiety Agents/pharmacology , Blood-Brain Barrier/metabolism , Dihydropyridines/pharmacology , Disease Models, Animal , Female , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Glutamate Decarboxylase/metabolism , Gyrus Cinguli/metabolism , Hippocampus/metabolism , Male , Maze Learning/drug effects , Mice , Mice, Transgenic , Neuronal Plasticity/drug effects , Up-Regulation/drug effects , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
5.
J Neurosci Res ; 92(3): 338-46, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24273007

ABSTRACT

Mildronate, a carnitine congener drug, previously has been shown to provide neuroprotection in an azidothymidine-induced mouse model of neurotoxicity and in a Parkinson's disease rat model. The aim of this study was to investigate the effects of mildronate treatment on cognition and pathology in Alzheimer's disease (AD) model mice (APP(SweDI)). Mildronate was administered i.p. daily at 50 or 100 mg/kg for 28 days. At the end of treatment, the animals were behaviorally and cognitively tested, and brains were assessed for AD-related pathology, inflammation, synaptic markers, and acetylcholinesterase (AChE). The data show that mildronate treatment significantly improved animal performance in water maze and social recognition tests, lowered amyloid-ß deposition in the hippocampus, increased expression of the microglia marker Iba-1, and decreased AChE staining, although it did not alter expression of proteins involved in synaptic plasticity (GAP-43, synaptophysin, and GAD67). Taken together, these findings indicate mildronate's ability to improve cognition and reduce amyloid-ß pathology in a mouse model of AD and its possible therapeutic utility as a disease-modifying drug in AD patients.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Cognition Disorders/drug therapy , Methylhydrazines/therapeutic use , Acetylcholinesterase/metabolism , Adjuvants, Immunologic/pharmacology , Alzheimer Disease/complications , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Cognition Disorders/etiology , Disease Models, Animal , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Female , Glial Fibrillary Acidic Protein/metabolism , Glutamate Decarboxylase/metabolism , Humans , Locomotion/drug effects , Locomotion/genetics , Methylhydrazines/pharmacology , Mice , Mice, Transgenic , Social Behavior
6.
bioRxiv ; 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38586056

ABSTRACT

Human cell line models, including the neuronal precursor line LUHMES, are important for investigating developmental transcriptional dynamics within imprinted regions, particularly the 15q11-q13 Angelman (AS) and Prader-Willi (PWS) syndrome locus. AS results from loss of maternal UBE3A in neurons, where the paternal allele is silenced by a convergent antisense transcript UBE3A-ATS, a lncRNA that normally terminates at PWAR1 in non-neurons. qRTPCR analysis confirmed the exclusive and progressive increase in UBE3A-ATS in differentiating LUHMES neurons, validating their use for studying UBE3A silencing. Genome-wide transcriptome analyses revealed changes to 11,834 genes during neuronal differentiation, including the upregulation of most genes within the 15q11-q13 locus. To identify dynamic changes in chromatin loops linked to transcriptional activity, we performed a HiChIP validated by 4C, which identified two neuron-specific CTCF loops between MAGEL2-SNRPN and PWAR1-UBE3A. To determine if allele-specific differentially methylated regions (DMR) may be associated with CTCF loop anchors, whole genome long-read nanopore sequencing was performed. We identified a paternally hypomethylated DMR near the SNRPN upstream loop anchor exclusive to neurons and a paternally hypermethylated DMR near the PWAR1 CTCF anchor exclusive to undifferentiated cells, consistent with increases in neuronal transcription. Additionally, DMRs near CTCF loop anchors were observed in both cell types, indicative of allele-specific differences in chromatin loops regulating imprinted transcription. These results provide an integrated view of the 15q11-q13 epigenetic landscape during LUHMES neuronal differentiation, underscoring the complex interplay of transcription, chromatin looping, and DNA methylation. They also provide insights for future therapeutic approaches for AS and PWS.

7.
Medicina (Kaunas) ; 49(7): 301-9, 2013.
Article in English | MEDLINE | ID: mdl-24375241

ABSTRACT

This review for the first time summarizes the data obtained in the neuropharmacological studies of mildronate, a drug previously known as a cardioprotective agent. In different animal models of neurotoxicity and neurodegenerative diseases, we demonstrated its neuroprotecting activity. By the use of immunohistochemical methods and Western blot analysis, as well as some selected behavioral tests, the new mechanisms of mildronate have been demonstrated: a regulatory effect on mitochondrial processes and on the expression of nerve cell proteins, which are involved in cell survival, functioning, and inflammation processes. Particular attention is paid to the capability of mildronate to stimulate learning and memory and to the expression of neuronal proteins involved in synaptic plasticity and adult neurogenesis. These properties can be useful in neurological practice to protect and treat neurological disorders, particularly those associated with neurodegeneration and a decline in cognitive functions.


Subject(s)
Adjuvants, Immunologic/pharmacology , Learning/drug effects , Methylhydrazines/pharmacology , Mitochondria/drug effects , Nerve Tissue Proteins/biosynthesis , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Disease Models, Animal , Humans , Mice , Mitochondria/metabolism , Nerve Regeneration/drug effects , Neuritis/metabolism , Neuritis/pathology , Neurons/metabolism , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Rats
8.
mSystems ; 8(1): e0060822, 2023 02 23.
Article in English | MEDLINE | ID: mdl-36598241

ABSTRACT

A large subset of patients with Angelman syndrome (AS) suffer from concurrent gastrointestinal (GI) issues, including constipation, poor feeding, and reflux. AS is caused by the loss of ubiquitin ligase E3A (UBE3A) gene expression in the brain. Clinical features of AS, which include developmental delays, intellectual disability, microcephaly, and seizures, are primarily due to the deficient expression or function of the maternally inherited UBE3A allele. The association between neurodevelopmental delay and GI disorders is part of the increasing evidence suggesting a link between the brain and the gut microbiome via the microbiota-gut-brain axis. To investigate the associations between colonization of the gut microbiota in AS, we characterized the fecal microbiome in three animal models of AS involving maternal deletions of Ube3A, including mouse, rat, and pig, using 16S rRNA amplicon sequencing. Overall, we identified changes in bacterial abundance across all three animal models of AS. Specific bacterial groups were significantly increased across all animal models, including Lachnospiraceae Incertae sedis, Desulfovibrios sp., and Odoribacter, which have been correlated with neuropsychiatric disorders. Taken together, these findings suggest that specific changes to the local environment in the gut are driven by a Ube3a maternal deletion, unaffected by varying housing conditions, and are prominent and detectable across multiple small and large animal model species. These findings begin to uncover the underlying mechanistic causes of GI disorders in AS patients and provide future therapeutic options for AS patients. IMPORTANCE Angelman syndrome (AS)-associated gastrointestinal (GI) symptoms significantly impact quality of life in patients. In AS models in mouse, rat, and pig, AS animals showed impaired colonization of the gut microbiota compared to wild-type (healthy) control animals. Common changes in AS microbiomes across all three animal models may play a causal effect for GI symptoms and may help to identify ways to treat these comorbidities in patients in the future.


Subject(s)
Angelman Syndrome , Gastrointestinal Diseases , Gastrointestinal Microbiome , Mice , Rats , Animals , Swine , Angelman Syndrome/genetics , Gastrointestinal Microbiome/genetics , RNA, Ribosomal, 16S/genetics , Quality of Life , Disease Models, Animal , Ubiquitin-Protein Ligases/genetics
9.
Medicina (Kaunas) ; 47(10): 552-9, 2011.
Article in English | MEDLINE | ID: mdl-22186119

ABSTRACT

BACKGROUND: Mildronate (3-[2,2,2-trimethylhydrazinium] propionate dihydrate) traditionally is a well-known cardioprotective drug. However, our recent studies convincingly demonstrated its neuroprotective properties. The aim of the present study was to evaluate the influence of mildronate on the expression of proteins that are involved in the differentiation and survival of the nigrostriatal dopaminergic neurons in the rat model of Parkinson's disease (PD). The following biomarkers were used: heat shock protein 70 (Hsp70, a molecular chaperone), glial cell line-derived nerve growth factor (GDNF, a growth factor promoting neuronal differentiation, regeneration, and survival), and neural cell adhesion molecule (NCAM). MATERIAL AND METHODS: PD was modeled by 6-hydroxydopamine (6-OHDA) unilateral intrastriatal injection in rats. Mildronate was administered at doses of 10, 20, and 50 mg/kg for 2 weeks intraperitoneally before 6-OHDA injection. Rat brains were dissected on day 28 after discontinuation of mildronate injections. The expression of biomarkers was assessed immunohistochemically and by western blot assay. RESULTS: 6-OHDA decreased the expression of Hsp70 and GDNF in the lesioned striatum and substantia nigra, whereas in mildronate-pretreated (20 and 50 mg/kg) rats, the expression of Hsp70 and GDNF was close to the control group values. NCAM expression also was decreased by 6-OHDA in the striatum and it was totally protected by mildronate at a dose of 50 mg/kg. In contrast, in the substantia nigra, 6-OHDA increased the expression of NCAM, while mildronate pretreatment (20 and 50 mg/kg) reversed the 6-OHDA-induced overexpression of NCAM close to the control values. CONCLUSION: The obtained data showed that mildronate was capable to regulate the expression of proteins that play a role in the homeostasis of neuro-glial processes.


Subject(s)
Cardiovascular Agents/administration & dosage , Methylhydrazines/administration & dosage , Neuroprotective Agents/administration & dosage , Parkinson Disease, Secondary/drug therapy , Protein Biosynthesis/drug effects , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Glial Cell Line-Derived Neurotrophic Factor/antagonists & inhibitors , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/biosynthesis , Male , Neural Cell Adhesion Molecules/antagonists & inhibitors , Neural Cell Adhesion Molecules/biosynthesis , Oxidopamine/antagonists & inhibitors , Oxidopamine/pharmacology , Parkinson Disease, Secondary/metabolism , Rats , Rats, Wistar , Substantia Nigra/drug effects , Substantia Nigra/metabolism
10.
Front Mol Neurosci ; 14: 789913, 2021.
Article in English | MEDLINE | ID: mdl-35153670

ABSTRACT

Zinc finger (ZF), transcription activator-like effectors (TALE), and CRISPR/Cas9 therapies to regulate gene expression are becoming viable strategies to treat genetic disorders, although effective in vivo delivery systems for these proteins remain a major translational hurdle. We describe the use of a mesenchymal stem/stromal cell (MSC)-based delivery system for the secretion of a ZF protein (ZF-MSC) in transgenic mouse models and young rhesus monkeys. Secreted ZF protein from mouse ZF-MSC was detectable within the hippocampus 1 week following intracranial or cisterna magna (CM) injection. Secreted ZF activated the imprinted paternal Ube3a in a transgenic reporter mouse and ameliorated motor deficits in a Ube3a deletion Angelman Syndrome (AS) mouse. Intrathecally administered autologous rhesus MSCs were well-tolerated for 3 weeks following administration and secreted ZF protein was detectable within the cerebrospinal fluid (CSF), midbrain, and spinal cord. This approach is less invasive when compared to direct intracranial injection which requires a surgical procedure.

11.
Int J Mol Sci ; 11(11): 4465-87, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-21151450

ABSTRACT

Previously, we have found that mildronate [3-(2,2,2-trimethylhydrazinium) propionate dihydrate], a small molecule with charged nitrogen and oxygen atoms, protects mitochondrial metabolism that is altered by inhibitors of complex I and has neuroprotective effects in an azidothymidine-neurotoxicity mouse model. In the present study, we investigated the effects of mildronate in a rat model of Parkinson's disease (PD) that was generated via a unilateral intrastriatal injection of the neurotoxin 6-hydroxydopamine (6-OHDA). We assessed the expression of cell biomarkers that are involved in signaling cascades and provide neural and glial integration: the neuronal marker TH (tyrosine hydroxylase); ubiquitin (a regulatory peptide involved in the ubiquitin-proteasome degradation system); Notch-3 (a marker of progenitor cells); IBA-1 (a marker of microglial cells); glial fibrillary acidic protein, GFAP (a marker of astrocytes); and inducible nitric oxide synthase, iNOS (a marker of inflammation). The data show that in the 6-OHDA-lesioned striatum, mildronate completely prevented the loss of TH, stimulated Notch-3 expression and decreased the expression of ubiquitin, GFAP and iNOS. These results provide evidence for the ability of mildronate to control the expression of an array of cellular proteins and, thus, impart multi-faceted homeostatic mechanisms in neurons and glial cells in a rat model of PD. We suggest that the use of mildronate provides a protective effect during the early stages of PD that can delay or halt the progression of this neurodegenerative disease.


Subject(s)
Methylhydrazines/pharmacology , Neuroglia/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease, Secondary/metabolism , Animals , Biomarkers/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Corpus Striatum/cytology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Male , Methylhydrazines/therapeutic use , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Neuroglia/metabolism , Neurons/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Oxidopamine/toxicity , Rats , Rats, Wistar , Receptor, Notch3 , Receptors, Notch/genetics , Receptors, Notch/metabolism , Substantia Nigra/cytology , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
12.
Eur J Pharmacol ; 818: 381-399, 2018 Jan 05.
Article in English | MEDLINE | ID: mdl-29133125

ABSTRACT

Recent studies devoted to neuroprotection have focused on the role of the gamma-aminobutyric acid (GABA) system in regulating neuroinflammatory processes which play a key role in the neurodegenerative processes observed in Alzheimer's disease (AD) by inducing glial cell overactivation and impairing neurotransmission. Data on the efficacy of classical GABA-A and GABA-B receptor agonists (muscimol and baclofen, respectively) in animal models of AD are not available. Moreover, no published studies have examined the ability of optimal doses of these compounds to prevent neuroinflammation, the alterations in neurotransmission and cognitive deficits. In the present study, we used a non-transgenic rat model of AD obtained by intracerebroventricular streptozocin (STZ) injection and assessed the effects of muscimol and baclofen at very low doses (0.01-0.05mg/kg) on spatial memory and the expression of cortical and hippocampal proteins related to neuroinflammation, namely proteins involved in astroglial functions (glial fibrillary acidic protein, GFAP), GABA synthesis (GABA synthesizing enzyme, glutamic acid decarboxylase 67, GAD67) and acetylcholine degradation (acetylcholine esterase). The presented study demonstrated that in a rat model of STZ-induced AD both muscimol and baclofen at the tested doses exerted memory-enhancing and anti-inflammatory effects, as well as normalization of acetylcholine esterase and GABA expression. We suggested that the function of very low doses of GABA receptor agonists differs from typical GABA-related inhibition and may be mediated by the allosteric sites of GABA receptors or other non-specific cell regulatory pathways.


Subject(s)
Alzheimer Disease/physiopathology , Baclofen/pharmacology , Brain/drug effects , Cognition/drug effects , Gene Expression Regulation/drug effects , Muscimol/pharmacology , Streptozocin/adverse effects , Alzheimer Disease/chemically induced , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Brain/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Glial Fibrillary Acidic Protein/metabolism , Male , Memory/drug effects , Rats , Rats, Wistar , Spatial Learning/drug effects
13.
Exp Gerontol ; 86: 39-49, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27167583

ABSTRACT

Clinical and epidemiological evidence suggests that lifestyle factors, including nutrition, may influence the chances of developing of Alzheimer's disease (AD), and also likely affect the aging process. Whereas it is clear that high-fat diets are increasing both body weight and the risk of developing Alzheimer's disease, to date, there have been very few studies comparing diets high with different sources of calories (i.e., high fat versus high protein versus high carbohydrates) to determine whether dietary composition has importance beyond the known effect of high caloric intake to increase body weight, AD pathology and cognitive deficits. In the current study we examined the effects that different diets high in carbohydrate, protein or fat content, but similar in caloric value, have on the development of cognitive impairment and brain pathology in wild-type and Tg AD model mice. The results demonstrate that long term feeding with balanced diets similar in caloric content but with significant changes in the source of calories, all negatively influence cognition compared to the control diet, and that this effect is more pronounced in Tg animals with AD pathology.


Subject(s)
Alzheimer Disease/etiology , Cognition Disorders/etiology , Diet/adverse effects , Dietary Carbohydrates/pharmacology , Dietary Fats/pharmacology , Dietary Proteins/pharmacology , Adiposity/physiology , Alzheimer Disease/pathology , Analysis of Variance , Animals , Behavior, Animal/physiology , Body Composition/physiology , Body Weight/physiology , Brain/pathology , Cognition Disorders/pathology , Diet, High-Fat/adverse effects , Disease Models, Animal , Energy Intake/physiology , Male , Mice, Transgenic
14.
PLoS One ; 10(6): e0127686, 2015.
Article in English | MEDLINE | ID: mdl-26042808

ABSTRACT

Ca2+ blockers, particularly those capable of crossing the blood-brain barrier (BBB), have been suggested as a possible treatment or disease modifying agents for neurodegenerative disorders, e.g., Alzheimer's disease. The present study investigated the effects of a novel 4-(N-dodecyl) pyridinium group-containing 1,4-dihydropyridine derivative (AP-12) on cognition and synaptic protein expression in the brain. Treatment of AP-12 was investigated in wild type C57BL/6J mice and transgenic Alzheimer's disease model mice (Tg APPSweDI) using behavioral tests and immunohistochemistry, as well as mass spectrometry to assess the blood-brain barrier (BBB) penetration. The data demonstrated the ability of AP-12 to cross the BBB, improve spatial learning and memory in both mice strains, induce anxiolytic action in transgenic mice, and increase expression of hippocampal and cortical proteins (GAD67, Homer-1) related to synaptic plasticity. The compound AP-12 can be seen as a prototype molecule for use in the design of novel drugs useful to halt progression of clinical symptoms (more specifically, anxiety and decline in memory) of neurodegenerative diseases, particularly Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/genetics , Brain/metabolism , Carrier Proteins/metabolism , Dihydropyridines/pharmacology , Glutamate Decarboxylase/metabolism , Memory/drug effects , Spatial Learning/drug effects , Animals , Anti-Anxiety Agents/pharmacology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain/pathology , Gyrus Cinguli/drug effects , Gyrus Cinguli/metabolism , Gyrus Cinguli/pathology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Homer Scaffolding Proteins , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Time Factors
15.
Eur J Pharmacol ; 745: 76-83, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25446926

ABSTRACT

The present study investigates the efficacy of mildronate, a carnitine congener, to protect stress and haloperidol-induced impairment of memory in rats and the expression of brain protein biomarkers involved in synaptic plasticity, such as brain-derived neurotrophic factor (BDNF), acetylcholine esterase and glutamate decarboxylase 67 (GAD67). Two amnesia models were used: 2h immobilization stress and 3-week haloperidol treatment. Stress caused memory impairment in the passive avoidance test and induced a significant 2-fold BDNF elevation in hippocampal and striatal tissues that was completely inhibited by mildronate. Mildronate decreased the level of GAD67 (but not acetylcholine esterase) expression by stress. Haloperidol decrease by a third hippocampal BDNF and acetylcholine esterase (but not GAD67) expression, which was normalized by mildronate; it also reversed the haloperidol-induced memory impairment in Barnes test. The results suggest the usefulness of mildronate as protector against neuronal disturbances caused by stress or haloperidol.


Subject(s)
Brain/drug effects , Memory/drug effects , Methylhydrazines/pharmacology , Acetylcholinesterase/metabolism , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Biomarkers/metabolism , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Carnitine/analogs & derivatives , Carnitine/pharmacology , GPI-Linked Proteins/metabolism , Glutamate Decarboxylase/metabolism , Haloperidol/toxicity , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/physiology , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/pharmacology , Rats , Rats, Wistar , Stress, Physiological
16.
Pharmacol Biochem Behav ; 106: 68-76, 2013 May.
Article in English | MEDLINE | ID: mdl-23537732

ABSTRACT

Previously we demonstrated that mildronate [3-(2,2,2-trimethylhydrazinium) propionate dihydrate], a representative of the aza-butyrobetaine class of compounds, protects mitochondrial metabolism under conditions such as ischemia. Mildronate also acted as a neuroprotective agent in an azidothymidine-induced mouse model of neurotoxicity, as well as in a rat model of Parkinson's disease. These observations suggest that mildronate may stimulate processes involved in cell survival and change expression of proteins involved in neurogenic processes. The present study investigated the influence of mildronate on learning and memory in the passive avoidance response (PAR) test and the active conditioned avoidance response (CAR) test in rats. The CAR test employed also bromodeoxyuridine (BrdU)-treated animals. Hippocampal cell BrdU incorporation was then immunohistochemically assessed in BrdU-treated, CAR-trained rats to identify proliferating cells. In addition, the expression of hippocampal proteins which could serve as memory enhancement biomarkers was evaluated and compared to non-trained animals' data. These biomarkers included glutamic acid decarboxylase 65/67 (GAD65/67), acetylcholine esterase (AChE), growth-associated protein-43 (GAP-43) and the transcription factor c-jun/activator protein-1 (AP-1). The results showed that mildronate enhanced learning/memory formation that coincided with the proliferation of neural progenitor cells, changing/regulating of the expression of biomarker proteins which are involved in the activation of glutamatergic and cholinergic pathways, transcription factors and adhesion molecule. The data from our study suggest that mildronate may be useful as a possible cognitive enhancer for the treatment of patients with neurodegenerative diseases with dementia.


Subject(s)
Hippocampus/drug effects , Learning/drug effects , Memory/drug effects , Methylhydrazines/pharmacology , Nerve Tissue Proteins/metabolism , Acetylcholinesterase/metabolism , Animals , Blotting, Western , Bromodeoxyuridine/metabolism , Glutamate Decarboxylase/metabolism , Hippocampus/metabolism , Male , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL