Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Med Chem ; 66(18): 13205-13246, 2023 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-37712656

RESUMEN

Huntington's disease (HD) is caused by an expanded CAG trinucleotide repeat in exon 1 of the huntingtin (HTT) gene. We report the design of a series of HTT pre-mRNA splicing modulators that lower huntingtin (HTT) protein, including the toxic mutant huntingtin (mHTT), by promoting insertion of a pseudoexon containing a premature termination codon at the exon 49-50 junction. The resulting transcript undergoes nonsense-mediated decay, leading to a reduction of HTT mRNA transcripts and protein levels. The starting benzamide core was modified to pyrazine amide and further optimized to give a potent, CNS-penetrant, and orally bioavailable HTT-splicing modulator 27. This compound reduced canonical splicing of the HTT RNA exon 49-50 and demonstrated significant HTT-lowering in both human HD stem cells and mouse BACHD models. Compound 27 is a structurally diverse HTT-splicing modulator that may help understand the mechanism of adverse effects such as peripheral neuropathy associated with branaplam.

2.
Front Neurosci ; 17: 1183023, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37325043

RESUMEN

Introduction: Cognitive impairment associated with old age or various brain disorders may be very disabling for affected individuals, placing their carers and public health services under considerable stress. The standard-of-care drugs produce only transient improvement of cognitive impairment in older people, so the search for novel, safe and effective therapeutics that would help to reverse or delay cognitive impairment is warranted. Repurposing pharmacological therapies with well-established safety record for additional indications is a promising recent trend in drug development. Vertigoheel (VH-04), a multicomponent drug made of Ambra grisea, Anamirta cocculus L., Conium maculatum, and Petroleum rectificatum, has been successfully used for several decades in the treatment of vertigo. Here, we investigated effects of VH-04 on cognitive performance in standard behavioral tests assessing different types of memory and explored cellular and molecular underpinnings of VH-04's biological activity. Methods: In the majority of behavioral experiments, namely in the spontaneous and rewarded alternation tests, passive avoidance test, contextual/cued fear conditioning, and social transmission of food preference, we examined the ability of single and repeated intraperitoneal administrations of VH-04 to improve cognitive parameters of mice and rats disrupted by the application of the muscarinic antagonist scopolamine. In addition, we also assessed how VH-04 affected novel object recognition and influenced performance of aged animals in Morris water maze. Furthermore, we also studied the effects of VH-04 on primary hippocampal neurons in vitro and mRNA expression of synaptophysin in the hippocampus. Results: Administration of VH-04 positively influenced visual recognition memory in the novel object recognition test and alleviated the impairments in spatial working memory and olfactory memory caused by the muscarinic antagonist scopolamine in the spontaneous alternation and social transmission of food preference tests. In addition, VH-04 improved retention of the spatial orientation memory of old rats in the Morris water maze. In contrast, VH-04 did not have significant effects on scopolamine-induced impairments in tests of fear-aggravated memory or rewarded alternation. Experiments in vitro showed that VH-04 stimulated neurite growth and possibly reversed the age-dependent decrease in hippocampal synaptophysin mRNA expression, which implies that VH-04 may preserve synaptic integrity in the aging brain. Discussion: Our findings allow a cautious conclusion that in addition to its ability to alleviate manifestations of vertigo, VH-04 may be also used as a cognitive enhancer.

3.
Cells ; 11(23)2022 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-36497038

RESUMEN

Body weight (BW) loss and reduced body mass index (BMI) are the most common peripheral alterations in Huntington disease (HD) and have been found in HD mutation carriers and HD animal models before the manifestation of neurological symptoms. This suggests that, at least in the early disease stage, these changes could be due to abnormal tissue growth rather than tissue atrophy. Moreover, BW and BMI are reported to be more affected in males than females in HD animal models and patients. Here, we confirmed sex-dependent growth alterations in the BACHD rat model for HD and investigated the associated contributing factors. Our results showed growth abnormalities along with decreased plasma testosterone and insulin-like growth factor 1 (IGF-1) levels only in males. Moreover, we demonstrated correlations between growth parameters, IGF-1, and testosterone. Our analyses further revealed an aberrant transcription of testosterone biosynthesis-related genes in the testes of BACHD rats with undisturbed luteinizing hormone (LH)/cAMP/PKA signaling, which plays a key role in regulating the transcription process of some of these genes. In line with the findings in BACHD rats, analyses in the R6/2 mouse model of HD showed similar results. Our findings support the view that mutant huntingtin may induce abnormal growth in males via the dysregulation of gene transcription in the testis, which in turn can affect testosterone biosynthesis.


Asunto(s)
Proteína Huntingtina , Enfermedad de Huntington , Testosterona , Animales , Femenino , Masculino , Ratones , Ratas , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Testosterona/biosíntesis , Proteína Huntingtina/genética
4.
PLoS One ; 16(10): e0258486, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34648564

RESUMEN

Huntington's disease (HD) results from an expansion mutation in the polyglutamine tract in huntingtin. Although huntingtin is ubiquitously expressed in the body, the striatum suffers the most severe pathology. Rhes is a Ras-related small GTP-binding protein highly expressed in the striatum that has been reported to modulate mTOR and sumoylation of mutant huntingtin to alter HD mouse model pathogenesis. Reports have varied on whether Rhes reduction is desirable for HD. Here we characterize multiple behavioral and molecular endpoints in the Q175 HD mouse model with genetic Rhes knockout (KO). Genetic RhesKO in the Q175 female mouse resulted in both subtle attenuation of Q175 phenotypic features, and detrimental effects on other kinematic features. The Q175 females exhibited measurable pathogenic deficits, as measured by MRI, MRS and DARPP32, however, RhesKO had no effect on these readouts. Additionally, RhesKO in Q175 mixed gender mice deficits did not affect mTOR signaling, autophagy or mutant huntingtin levels. We conclude that global RhesKO does not substantially ameliorate or exacerbate HD mouse phenotypes in Q175 mice.


Asunto(s)
Proteínas de Unión al GTP/genética , Enfermedad de Huntington/patología , Animales , Fenómenos Biomecánicos , Peso Corporal , Encéfalo/fisiología , Modelos Animales de Enfermedad , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Femenino , Proteínas de Unión al GTP/deficiencia , Proteínas de Unión al GTP/metabolismo , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
5.
PLoS One ; 15(12): e0243052, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33370315

RESUMEN

Huntington's disease (HD) is an inherited neurodegenerative disorder characterized by severe disruption of cognitive and motor functions, including changes in posture and gait. A number of HD mouse models have been engineered that display behavioral and neuropathological features of the disease, but gait alterations in these models are poorly characterized. Sensitive high-throughput tests of fine motor function and gait in mice might be informative in evaluating disease-modifying interventions. Here, we describe a hypothesis-free workflow that determines progressively changing locomotor patterns across 79 parameters in the R6/2 and Q175 mouse models of HD. R6/2 mice (120 CAG repeats) showed motor disturbances as early as at 4 weeks of age. Similar disturbances were observed in homozygous and heterozygous Q175 KI mice at 3 and 6 months of age, respectively. Interestingly, only the R6/2 mice developed forelimb ataxia. The principal components of the behavioral phenotypes produced two phenotypic scores of progressive postural instability based on kinematic parameters and trajectory waveform data, which were shared by both HD models. This approach adds to the available HD mouse model research toolbox and has a potential to facilitate the development of therapeutics for HD and other debilitating movement disorders with high unmet medical need.


Asunto(s)
Análisis de la Marcha/métodos , Proteína Huntingtina/genética , Enfermedad de Huntington/fisiopatología , Animales , Fenómenos Biomecánicos , Modelos Animales de Enfermedad , Femenino , Enfermedad de Huntington/genética , Masculino , Ratones , Ratones Transgénicos , Actividad Motora , Mutación , Postura
6.
Nat Med ; 25(7): 1131-1142, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31263285

RESUMEN

Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder caused by a CAG trinucleotide expansion in the huntingtin gene (HTT), which codes for the pathologic mutant HTT (mHTT) protein. Since normal HTT is thought to be important for brain function, we engineered zinc finger protein transcription factors (ZFP-TFs) to target the pathogenic CAG repeat and selectively lower mHTT as a therapeutic strategy. Using patient-derived fibroblasts and neurons, we demonstrate that ZFP-TFs selectively repress >99% of HD-causing alleles over a wide dose range while preserving expression of >86% of normal alleles. Other CAG-containing genes are minimally affected, and virally delivered ZFP-TFs are active and well tolerated in HD neurons beyond 100 days in culture and for at least nine months in the mouse brain. Using three HD mouse models, we demonstrate improvements in a range of molecular, histopathological, electrophysiological and functional endpoints. Our findings support the continued development of an allele-selective ZFP-TF for the treatment of HD.


Asunto(s)
Alelos , Proteína Huntingtina/genética , Enfermedad de Huntington/terapia , Mutación , Transcripción Genética , Dedos de Zinc , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Enfermedad de Huntington/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Neuroprotección , Repeticiones de Trinucleótidos
7.
Front Behav Neurosci ; 12: 226, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30333735

RESUMEN

Cognitive disturbances often predate characteristic motor dysfunction in individuals with Huntington's disease (HD) and place an increasing burden on the HD patients and caregivers with the progression of the disorder. Therefore, application of maximally translational cognitive tests to animal models of HD is imperative for the development of treatments that could alleviate cognitive decline in human patients. Here, we examined the performance of the Q175 mouse knock-in model of HD in the touch screen version of the paired associates learning (PAL) task. We found that 10-11-month-old heterozygous Q175 mice had severely attenuated learning curve in the PAL task, which was conceptually similar to previously documented impaired performance of individuals with HD in the PAL task of the Cambridge Neuropsychological Test Automated Battery (CANTAB). Besides high rate of errors in PAL task, Q175 mice exhibited considerably lower responding rate than age-matched wild-type (WT) animals. Our examination of effortful operant responding during fixed ratio (FR) and progressive ratio (PR) reinforcement schedules in a separate cohort of similar age confirmed slower and unselective performance of mutant animals, as observed during PAL task, but suggested that motivation to work for nutritional reward in the touch screen setting was similar in Q175 and WT mice. We also demonstrated that pronounced sensorimotor disturbances in Q175 mice can be detected at early touch screen testing stages, (e.g., during "Punish Incorrect" phase of operant pretraining), so we propose that shorter test routines may be utilised for more expedient studies of treatments aimed at the rescue of HD-related phenotype.

8.
PLoS One ; 12(2): e0171127, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28182673

RESUMEN

The aim of these studies was to demonstrate the therapeutic capacity of an antisense oligonucleotide with the sequence (CUG)7 targeting the expanded CAG repeat in huntingtin (HTT) mRNA in vivo in the R6/2 N-terminal fragment and Q175 knock-in Huntington's disease (HD) mouse models. In a first study, R6/2 mice received six weekly intracerebroventricular infusions with a low and high dose of (CUG)7 and were sacrificed 2 weeks later. A 15-60% reduction of both soluble and aggregated mutant HTT protein was observed in striatum, hippocampus and cortex of (CUG)7-treated mice. This correction at the molecular level resulted in an improvement of performance in multiple motor tasks, increased whole brain and cortical volume, reduced levels of the gliosis marker myo-inositol, increased levels of the neuronal integrity marker N-aceyl aspartate and increased mRNA levels of the striatal marker Darpp-32. These neuroanatomical and neurochemical changes, together with the improved motor performance, suggest that treatment with (CUG)7 ameliorates basal ganglia dysfunction. The HTT-lowering was confirmed by an independent study in Q175 mice using a similar (CUG)7 AON dosing regimen, further demonstrating a lasting reduction of mutant HTT protein in striatum, hippocampus and cortex for up to 18 weeks post last infusion along with an increase in motor activity. Based on these encouraging results, (CUG)7 may thus offer an interesting alternative HTT-lowering strategy for HD.


Asunto(s)
Terapia Genética , Proteína Huntingtina/genética , Enfermedad de Huntington/terapia , ARN sin Sentido/genética , Expansión de Repetición de Trinucleótido , Animales , Encéfalo/metabolismo , Encéfalo/patología , Femenino , Gliosis , Enfermedad de Huntington/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora
9.
Neuron ; 92(6): 1220-1237, 2016 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-27916455

RESUMEN

Huntington's disease (HD) symptoms are driven to a large extent by dysfunction of the basal ganglia circuitry. HD patients exhibit reduced striatal phoshodiesterase 10 (PDE10) levels. Using HD mouse models that exhibit reduced PDE10, we demonstrate the benefit of pharmacologic PDE10 inhibition to acutely correct basal ganglia circuitry deficits. PDE10 inhibition restored corticostriatal input and boosted cortically driven indirect pathway activity. Cyclic nucleotide signaling is impaired in HD models, and PDE10 loss may represent a homeostatic adaptation to maintain signaling. Elevation of both cAMP and cGMP by PDE10 inhibition was required for rescue. Phosphoproteomic profiling of striatum in response to PDE10 inhibition highlighted plausible neural substrates responsible for the improvement. Early chronic PDE10 inhibition in Q175 mice showed improvements beyond those seen with acute administration after symptom onset, including partial reversal of striatal deregulated transcripts and the prevention of the emergence of HD neurophysiological deficits. VIDEO ABSTRACT.


Asunto(s)
Corteza Cerebral/efectos de los fármacos , Enfermedad de Huntington/fisiopatología , Neostriado/efectos de los fármacos , Inhibidores de Fosfodiesterasa/farmacología , Pirazoles/farmacología , Quinolinas/farmacología , Animales , Ganglios Basales/diagnóstico por imagen , Ganglios Basales/efectos de los fármacos , Ganglios Basales/metabolismo , Ganglios Basales/fisiopatología , Corteza Cerebral/diagnóstico por imagen , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiopatología , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Enfermedad de Huntington/metabolismo , Ratones , Neostriado/diagnóstico por imagen , Neostriado/metabolismo , Neostriado/fisiopatología , Hidrolasas Diéster Fosfóricas , Tomografía de Emisión de Positrones , Núcleo Subtalámico/diagnóstico por imagen , Núcleo Subtalámico/efectos de los fármacos , Núcleo Subtalámico/metabolismo , Núcleo Subtalámico/fisiopatología , Tritio
10.
Exp Neurol ; 282: 99-118, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27163548

RESUMEN

Dysregulation of the kynurenine (Kyn) pathway has been associated with the progression of Huntington's disease (HD). In particular, elevated levels of the kynurenine metabolites 3-hydroxy kynurenine (3-OH-Kyn) and quinolinic acid (Quin), have been reported in the brains of HD patients as well as in rodent models of HD. The production of these metabolites is controlled by the activity of kynurenine mono-oxygenase (KMO), an enzyme which catalyzes the synthesis of 3-OH-Kyn from Kyn. In order to determine the role of KMO in the phenotype of mouse models of HD, we have developed a potent and selective KMO inhibitor termed CHDI-340246. We show that this compound, when administered orally to transgenic mouse models of HD, potently and dose-dependently modulates the Kyn pathway in peripheral tissues and in the central nervous system. The administration of CHDI-340246 leads to an inhibition of the formation of 3-OH-Kyn and Quin, and to an elevation of Kyn and Kynurenic acid (KynA) levels in brain tissues. We show that administration of CHDI-340246 or of Kyn and of KynA can restore several electrophysiological alterations in mouse models of HD, both acutely and after chronic administration. However, using a comprehensive panel of behavioral tests, we demonstrate that the chronic dosing of a selective KMO inhibitor does not significantly modify behavioral phenotypes or natural progression in mouse models of HD.


Asunto(s)
Fenómenos Electrofisiológicos/efectos de los fármacos , Inhibidores Enzimáticos/uso terapéutico , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/fisiopatología , Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Pirimidinas/uso terapéutico , Análisis de Varianza , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Fenómenos Electrofisiológicos/genética , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Hipocampo/efectos de los fármacos , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Técnicas In Vitro , Ácido Quinurénico/metabolismo , Quinurenina 3-Monooxigenasa/metabolismo , Masculino , Ratones , Ratones Transgénicos , Microdiálisis , Pirimidinas/química , Pirimidinas/metabolismo , Pirimidinas/farmacología , Ácido Quinolínico/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transfección , Repeticiones de Trinucleótidos/genética , Receptor Nicotínico de Acetilcolina alfa 7/genética , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
11.
J Neuropathol Exp Neurol ; 73(8): 752-69, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25003233

RESUMEN

Understanding the pathophysiologic mechanisms underlying Alzheimer disease relies on knowledge of disease onset and the sequence of development of brain pathologies. We present a comprehensive analysis of early and progressive changes in a mouse model that demonstrates a full spectrum of characteristic Alzheimer disease-like pathologies. This model demonstrates an altered immune redox state reminiscent of the human disease and capitalizes on data indicating critical differences between human and mouse immune responses, particularly in nitric oxide levels produced by immune activation of the NOS2 gene. Using the APPSwDI(+)/(+)mNos2(-/-) (CVN-AD) mouse strain, we show a sequence of pathologic events leading to neurodegeneration,which include pathologically hyperphosphorylated tau in the perforant pathway at 6 weeks of age progressing to insoluble tau, early appearance of ß-amyloid peptides in perivascular deposits around blood vessels in brain regions known to be vulnerable to Alzheimer disease, and progression to damage and overt loss in select vulnerable neuronal populations in these regions. The role of species differences between hNOS2 and mNos2 was supported by generating mice in which the human NOS2 gene replaced mNos2. When crossed with CVN-AD mice, pathologic characteristics of this new strain (APPSwDI(+)/(-)/HuNOS2(tg+)/(+)/mNos2(-/-)) mimicked the pathologic phenotypes found in the CVN-AD strain.


Asunto(s)
Enfermedad de Alzheimer/genética , Modelos Animales de Enfermedad , Mutación/genética , Óxido Nítrico Sintasa de Tipo II/deficiencia , Óxido Nítrico Sintasa de Tipo II/genética , Factores de Edad , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Condicionamiento Clásico/fisiología , Regulación de la Expresión Génica/genética , Humanos , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosforilación/genética
12.
PLoS Curr ; 62014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24558637

RESUMEN

Huntington's disease is a neurodegenerative disorder caused by mutations in the CAG tract of huntingtin. Several studies in HD cellular and rodent systems have identified disturbances in cyclic nucleotide signaling, which might be relevant to pathogenesis and therapeutic intervention. To investigate whether selective phosphodiesterase (PDE) inhibitors can improve some aspects of disease pathogenesis in HD models, we have systematically evaluated the effects of a variety of cAMP and cGMP selective PDE inhibitors in various HD models. Here we present the lack of effect in a variety of endpoints of the PDE subtype selective inhibitor SCH-51866, a PDE1/5 inhibitor, in the R6/2 mouse model of HD, after chronic oral dosing.

13.
PLoS One ; 7(12): e50717, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23284644

RESUMEN

Huntington's disease (HD) is an autosomal neurodegenerative disorder, characterized by severe behavioral, cognitive, and motor deficits. Since the discovery of the huntingtin gene (HTT) mutation that causes the disease, several mouse lines have been developed using different gene constructs of Htt. Recently, a new model, the zQ175 knock-in (KI) mouse, was developed (see description by Menalled et al, [1]) in an attempt to have the Htt gene in a context and causing a phenotype that more closely mimics HD in humans. Here we confirm the behavioral phenotypes reported by Menalled et al [1], and extend the characterization to include brain volumetry, striatal metabolite concentration, and early neurophysiological changes. The overall reproducibility of the behavioral phenotype across the two independent laboratories demonstrates the utility of this new model. Further, important features reminiscent of human HD pathology are observed in zQ175 mice: compared to wild-type neurons, electrophysiological recordings from acute brain slices reveal that medium spiny neurons from zQ175 mice display a progressive hyperexcitability; glutamatergic transmission in the striatum is severely attenuated; decreased striatal and cortical volumes from 3 and 4 months of age in homo- and heterozygous mice, respectively, with whole brain volumes only decreased in homozygotes. MR spectroscopy reveals decreased concentrations of N-acetylaspartate and increased concentrations of glutamine, taurine and creatine + phosphocreatine in the striatum of 12-month old homozygotes, the latter also measured in 12-month-old heterozygotes. Motor, behavioral, and cognitive deficits in homozygotes occur concurrently with the structural and metabolic changes observed. In sum, the zQ175 KI model has robust behavioral, electrophysiological, and histopathological features that may be valuable in both furthering our understanding of HD-like pathophyisology and the evaluation of potential therapeutic strategies to slow the progression of disease.


Asunto(s)
Conducta Animal , Encéfalo/patología , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Enfermedad de Huntington/patología , Enfermedad de Huntington/fisiopatología , Neurofisiología , Animales , Peso Corporal , Encéfalo/metabolismo , Encéfalo/fisiopatología , Recuento de Células , Progresión de la Enfermedad , Determinación de Punto Final , Femenino , Ácido Glutámico/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética , Masculino , Ratones , Neostriado/patología , Proteínas del Tejido Nervioso/genética , Neuronas/patología , Tamaño de los Órganos , Secuencias Repetitivas de Ácidos Nucleicos , Natación , Transmisión Sináptica
14.
J Alzheimers Dis ; 27(3): 499-510, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21821875

RESUMEN

The purpose of this study was to evaluate the efficacy of the radical scavenger IAC (bis(1-hydroxy-2,2,6,6-tetramethyl-4-piperidinyl) decantionate) in alleviating behavioral deficits and reducing amyloid-ß (Aß) accumulation in an Alzheimer's disease (AD) transgenic Tg2576 mouse model. Daily treatment with IAC (3-30 mg/kg, i.p.) was started at the age of 6 months and continued until the mice were 13 months old. At the age of 9 months and again at 12 months, the mice were tested in open field and water maze tests. At the age of 13 months, the mice were sacrificed and the brains processed for immunohistochemistry. Mortality was significantly reduced in all IAC-treated groups. In addition, IAC treatment improved the water maze hidden platform training performance but had no effect on motor activity in the open field or water maze swim speed in transgenic mice. Lastly, IAC treatment (10 mg/kg) significantly reduced the cortical Aß plaque burden. In vitro, IAC is able to increase the number of neurites and neurite branches in cultured cortical primary neurons. In conclusion, IAC slowed down the development of the AD-like phenotype in Tg2576 mice and accelerated neurite growth in cultured neurons.


Asunto(s)
Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Cognición/fisiología , Aprendizaje por Laberinto/fisiología , Piperidinas/uso terapéutico , Placa Amiloide/tratamiento farmacológico , Placa Amiloide/genética , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Cognición/efectos de los fármacos , Cricetinae , Modelos Animales de Enfermedad , Depuradores de Radicales Libres/farmacología , Depuradores de Radicales Libres/uso terapéutico , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Piperidinas/farmacología , Placa Amiloide/patología , Ratas , Ratas Wistar
15.
J Cereb Blood Flow Metab ; 30(7): 1318-28, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20145657

RESUMEN

In the traumatic brain injury (TBI) the initial impact causes both primary injury, and launches secondary injury cascades. One consequence, and a factor that may contribute to these secondary changes and functional outcome, is altered hemodynamics. The relative cerebral blood volume (CBV) changes in rat brain after severe controlled cortical impact injury were characterized to assess their interrelations with motor function impairment. Magnetic resonance imaging (MRI) was performed 1, 2, 4 h, and 1, 2, 3, 4, 7, and 14 days after TBI to quantify CBV and water diffusion. Neuroscore test was conducted before, and 2, 7, and 14 days after the TBI. We found distinct temporal profile of CBV in the perilesional area, hippocampus, and in the primary lesion. In all regions, the first response was drop of CBV. Perifocal CBV was reduced for over 4 days thereafter gradually recovering. After the initial drop, the hippocampal CBV was increased for 2 weeks. Neuroscore demonstrated severely impaired motor functions 2 days after injury (33% decrease), which then slowly recovered in 2 weeks. This recovery parallelled the recovery of perifocal CBV. CBV MRI can detect cerebrovascular pathophysiology after TBI in the vulnerable perilesional area, which seems to potentially associate with time course of sensory-motor deficit.


Asunto(s)
Conducta Animal/fisiología , Volumen Sanguíneo/fisiología , Lesiones Encefálicas , Encéfalo , Circulación Cerebrovascular/fisiología , Flujo Sanguíneo Regional , Animales , Encéfalo/irrigación sanguínea , Encéfalo/patología , Encéfalo/fisiología , Lesiones Encefálicas/patología , Lesiones Encefálicas/fisiopatología , Capilares/metabolismo , Hemodinámica , Humanos , Imagen por Resonancia Magnética , Masculino , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/fisiología
16.
Exp Gerontol ; 39(9): 1277-83, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15489050

RESUMEN

Spatial memory deficits occur earlier in female than male rodents as the animals age, and the cessation of estrous cycle has been suggested to play a role in this phenomenon. We examined the effects of long-term ovariectomy (OVX) and estrogen replacement therapy (ERT) with subcutaneous 17beta-estradiol minipellets on maze learning in aged (24-month-old) female C57BL/6J mice using a win-stay task (1/8 arms baited) in the radial arm maze (RAM) and a position discrimination task in the T-maze. ERT was started 40 days before the behavioral tests both in gonadally intact (sham-operated) and OVX mice. The effect of early OVX on RAM performance was investigated using three different age groups (7, 11 and 24 months) with different OVX durations (4, 8 and 19 months, respectively). ERT reduced the number of reference memory errors in RAM in aged sham-operated and OVX mice, but unlike in young mice (Heikkinen et al., 2002) it had no effect on working memory errors. Furthermore, OVX impaired the performance of aged mice in the T-maze. Comparison across the three age groups and three OVX durations indicated that the memory impairment induced by an early age OVX attenuates as the mice get close to their estropausal age.


Asunto(s)
Envejecimiento/psicología , Terapia de Reemplazo de Estrógeno , Estrógenos/fisiología , Aprendizaje por Laberinto/efectos de los fármacos , Envejecimiento/patología , Animales , Peso Corporal/efectos de los fármacos , Estradiol/farmacología , Femenino , Memoria a Corto Plazo/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos/efectos de los fármacos , Ovariectomía , Útero/patología
17.
Behav Brain Res ; 134(1-2): 433-45, 2002 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-12191831

RESUMEN

Transgenic mice carrying mutated human amyloid precursor protein (APPswe) and presenilin (PS1, A246E) genes develop first amyloid plaques around 9 months of age, but up to 18 months of age, amyloid depositions in these mice were largely restricted to the hippocampus, subiculum, and neocortex. To assess the behavioral consequences of amyloid accumulation in the hippocampal formation, we compared the effects of APP+PS1 (AP) genotype and fimbria-fornix (FFX) transection, either alone or combined, on various spatial learning and memory tasks. Both FFX-lesioned and AP mice were impaired in spatial navigation in the water maze, a typical hippocampal dependent task. Conversely, neither group of mice was impaired in a win-stay version of the radial arm maze (RAM) or position discrimination in the T-maze, tasks that do not depend on the hippocampus. FFX-lesioned mice were impaired in the win-shift version of the RAM, and in spontaneous and rewarded alternation in the T-maze, while AP mice performed equal to non-transgenic controls in all these working memory tasks, except long-term retention of the RAM task. AP mice thus appear to have a selective deficit in hippocampal dependent long-term memory, as do Alzheimer patients at early stage of the disease.


Asunto(s)
Amígdala del Cerebelo/fisiología , Neuropatías Amiloides/psicología , Precursor de Proteína beta-Amiloide/genética , Fórnix/fisiología , Proteínas de la Membrana/genética , Memoria/fisiología , Percepción Espacial/fisiología , Amígdala del Cerebelo/metabolismo , Neuropatías Amiloides/genética , Neuropatías Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Ansiedad/genética , Ansiedad/psicología , Química Encefálica/fisiología , Colina O-Acetiltransferasa/metabolismo , Aprendizaje por Laberinto/fisiología , Memoria a Corto Plazo/fisiología , Ratones , Ratones Transgénicos , Presenilina-1
18.
Neurobiol Dis ; 9(3): 339-47, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11950278

RESUMEN

We investigated the role of hippocampal amyloid pathology in spatial learning impairment of a new mouse line carrying mutated human amyloid precursor protein (APP) and presenilin-1 (PS1) transgenes. The APP + PS1 mice were tested in spatial navigation in the water maze and in position discrimination in the T-maze at ages of 3-4 and 11-12 months, before and after the appearance of first amyloid plaques. The APP + PS1 mice were impaired in water maze acquisition and retention only at the age of 11-12 months, but performed equally to controls in the T-maze task at both ages. In the impaired older age group, the levels of total Abeta1-42 in the hippocampus of APP + PS1 mice correlated negatively with the retention score. Here we show for the first time that the age-dependent impairment in memory retention in the traditional water maze of APP + PS1 mice correlates with the amount of total Abeta in hippocampus even at a stage when the amyloid deposits cover less than 1% of the hippocampal volume.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Hipocampo/metabolismo , Proteínas de la Membrana/genética , Trastornos de la Memoria/genética , Trastornos de la Memoria/metabolismo , Fragmentos de Péptidos/metabolismo , Factores de Edad , Péptidos beta-Amiloides/genética , Animales , Hipocampo/patología , Hipocampo/fisiopatología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fragmentos de Péptidos/genética , Placa Amiloide/genética , Placa Amiloide/metabolismo , Placa Amiloide/patología , Presenilina-1
19.
Horm Behav ; 41(1): 22-32, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11863380

RESUMEN

This study examined the effects of long-term estrogen treatment (sc 17 beta-estradiol minipellets) on learning in C57BL/6J female and male mice using a position discrimination task in the T-maze and a win-stay task (1/8 arms baited) in the radial arm maze (RAM). In addition, hippocampal monoamines and ChAT activity were measured at the end of the study and correlated to task performance. Female sham-operated (gonadally intact) and ovariectomized (OVX) mice were treated with estrogen either for 7 or 40 days before the behavioral tests and intact male mice for 7 days before the behavioral tests. In sham-operated mice the 40-day estrogen treatment improved RAM performance and in OVX mice both the 7- and 40-day estrogen treatments improved the performance in both maze tasks. The estrogen treatment also improved RAM performance in males. The hippocampal ChAT, NA, 5-HIAA, and DOPAC levels were decreased in OVX mice. Furthermore, the effects of estrogen treatment on the levels of hippocampal 5-HT and its metabolite 5-HIAA were different in sham-operated than in OVX mice. We could find no correlation between cognitive measures and neurochemical variables. This study gives new information about the effects of estrogen on learning and hippocampal neurotransmitters in mice.


Asunto(s)
Monoaminas Biogénicas/metabolismo , Estrógenos/farmacología , Hipocampo/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Norepinefrina/metabolismo , Ácido 3,4-Dihidroxifenilacético/metabolismo , Animales , Colina O-Acetiltransferasa/metabolismo , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Femenino , Ácido Hidroxiindolacético/metabolismo , Masculino , Aprendizaje por Laberinto/fisiología , Memoria/efectos de los fármacos , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos , Ovariectomía , Serotonina/metabolismo , Útero/anatomía & histología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...