Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Hum Mol Genet ; 29(19): 3224-3248, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-32959884

RESUMEN

Genome-wide association studies have reported that, amongst other microglial genes, variants in TREM2 can profoundly increase the incidence of developing Alzheimer's disease (AD). We have investigated the role of TREM2 in primary microglial cultures from wild type mice by using siRNA to decrease Trem2 expression, and in parallel from knock-in mice heterozygous or homozygous for the Trem2 R47H AD risk variant. The prevailing phenotype of Trem2 R47H knock-in mice was decreased expression levels of Trem2 in microglia, which resulted in decreased density of microglia in the hippocampus. Overall, primary microglia with reduced Trem2 expression, either by siRNA or from the R47H knock-in mice, displayed a similar phenotype. Comparison of the effects of decreased Trem2 expression under conditions of lipopolysaccharide (LPS) pro-inflammatory or IL-4 anti-inflammatory stimulation revealed the importance of Trem2 in driving a number of the genes up-regulated in the anti-inflammatory phenotype. RNA-seq analysis showed that IL-4 induced the expression of a program of genes including Arg1 and Ap1b1 in microglia, which showed an attenuated response to IL-4 when Trem2 expression was decreased. Genes showing a similar expression profile to Arg1 were enriched for STAT6 transcription factor recognition elements in their promoter, and Trem2 knockdown decreased levels of STAT6. LPS-induced pro-inflammatory stimulation suppressed Trem2 expression, thus preventing TREM2's anti-inflammatory drive. Given that anti-inflammatory signaling is associated with tissue repair, understanding the signaling mechanisms downstream of Trem2 in coordinating the pro- and anti-inflammatory balance of microglia, particularly mediating effects of the IL-4-regulated anti-inflammatory pathway, has important implications for fighting neurodegenerative disease.


Asunto(s)
Regulación de la Expresión Génica , Mediadores de Inflamación/metabolismo , Inflamación/inmunología , Glicoproteínas de Membrana/fisiología , Microglía/inmunología , Mutación , Receptores Inmunológicos/fisiología , Transcriptoma , Animales , Animales Recién Nacidos , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Microglía/patología , RNA-Seq , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/metabolismo
2.
J Neurosci Res ; 97(12): 1728-1741, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31392765

RESUMEN

Dementia is now the leading cause of death in the United Kingdom, accounting for over 12% of all deaths and is the fifth most common cause of death worldwide. As treatments for heart disease and cancers improve and the population ages, the number of sufferers will only increase, with the chance of developing dementia doubling every 5 years after the age of 65. Finding an effective treatment is ever more critical to avert this pandemic health (and economic) crisis. To date, most dementia-related research has focused on the cortex and the hippocampus; however, with dementia becoming more fully recognized as aspects of diseases historically categorized as motor disorders (e.g., Parkinson's and Huntington's diseases), the role of the basal ganglia in dementia is coming to the fore. Conversely, it is highly likely that neuronal pathways in these structures traditionally considered as spared in Alzheimer's disease are also affected, particularly in later stages of the disease. In this review, we examine some of the limited evidence linking the basal ganglia to dementia.


Asunto(s)
Ganglios Basales/fisiopatología , Demencia/fisiopatología , Enfermedades Neurodegenerativas/fisiopatología , Animales , Ganglios Basales/patología , Demencia/genética , Demencia/patología , Dopamina/fisiología , Neuronas Dopaminérgicas/fisiología , Predisposición Genética a la Enfermedad , Humanos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Neuronas/fisiología
3.
J Neurosci Res ; 97(12): 1624-1635, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31353533

RESUMEN

Huntington's disease (HD), an inherited neurodegenerative disorder that principally affects striatum and cerebral cortex, is generally thought to have an adult onset. However, a small percentage of cases develop symptoms before 20 years of age. This juvenile variant suggests that brain development may be altered in HD. Indeed, recent evidence supports an important role of normal huntingtin during embryonic brain development and mutations in this protein cause cortical abnormalities. Functional studies also demonstrated that the cerebral cortex becomes hyperexcitable with disease progression. In this review, we examine clinical and experimental evidence that cortical development is altered in HD. We also provide preliminary evidence that cortical pyramidal neurons from R6/2 mice, a model of juvenile HD, are hyperexcitable and display dysmorphic processes as early as postnatal day 7. Further, some symptomatic mice present with anatomical abnormalities reminiscent of human focal cortical dysplasia, which could explain the occurrence of epileptic seizures in this genetic mouse model and in children with juvenile HD. Finally, we discuss recent treatments aimed at correcting abnormal brain development.


Asunto(s)
Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/fisiopatología , Excitabilidad Cortical , Enfermedad de Huntington/fisiopatología , Neuronas/fisiología , Animales , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/patología , Ratones Transgénicos , Neuronas/patología
4.
Cereb Cortex ; 27(6): 3437-3448, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28334103

RESUMEN

Neuronal pentraxin 1 (NPTX1) has been implicated in Alzheimer's disease, being present in and around dystrophic neurons in plaques, affecting glutamatergic transmission postsynaptically and mediating effects of amyloidß. Here, we confirm the presence of NPTX1 around plaques in postmortem Alzheimer's disease brain and report that acutely applied human NPTX1 increases paired-pulse ratio at mouse CA3-CA1 hippocampal synapses, indicating a decrease in glutamate release. In contrast, chronic exposure to NPTX1, NPTX2, or NPTX receptor decreases paired-pulse ratio, mimicking some of the earliest changes in mice expressing familial Alzheimer's disease genes. The peripheral pentraxin, serum amyloid P component (SAP), causes similar synaptic effects to NPTX1. The presence of SAP on amyloid plaques in Alzheimer's disease confirms that it can enter the brain. We show that SAP and neuronal pentraxins can interact and that SAP can enter the brain if the blood-brain barrier is compromised, suggesting that peripheral pentraxins could affect central synaptic transmission via this interaction, especially in the event of blood-brain barrier breakdown.


Asunto(s)
Barrera Hematoencefálica/fisiopatología , Proteína C-Reactiva/metabolismo , Ácido Glutámico/metabolismo , Hipocampo/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Animales , Animales Recién Nacidos , Barrera Hematoencefálica/patología , Proteína C-Reactiva/genética , Proteína C-Reactiva/farmacología , Potenciales Evocados/efectos de los fármacos , Potenciales Evocados/fisiología , Femenino , Antagonistas del GABA/farmacología , Células HEK293 , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/farmacología , Neuronas/efectos de los fármacos , Piridazinas/farmacología , Componente Amiloide P Sérico/farmacología , Sinapsis/efectos de los fármacos , Sinapsis/genética , Sinapsis/metabolismo
5.
Brain ; 138(Pt 7): 1992-2004, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25981962

RESUMEN

Detecting and treating Alzheimer's disease, before cognitive deficits occur, has become the health challenge of our time. The earliest known event in Alzheimer's disease is rising amyloid-ß. Previous studies have suggested that effects on synaptic transmission may precede plaque deposition. Here we report how relative levels of different soluble amyloid-ß peptides in hippocampus, preceding plaque deposition, relate to synaptic and genomic changes. Immunoprecipitation-mass spectrometry was used to measure the early rise of different amyloid-ß peptides in a mouse model of increasing amyloid-ß ('TASTPM', transgenic for familial Alzheimer's disease genes APP/PSEN1). In the third postnatal week, several amyloid-ß peptides were above the limit of detection, including amyloid-ß40, amyloid-ß38 and amyloid-ß42 with an intensity ratio of 6:3:2, respectively. By 2 months amyloid-ß levels had only increased by 50% and although the ratio of the different peptides remained constant, the first changes in synaptic currents, compared to wild-type mice could be detected with patch-clamp recordings. Between 2 and 4 months old, levels of amyloid-ß40 rose by ∼7-fold, but amyloid-ß42 rose by 25-fold, increasing the amyloid-ß42:amyloid-ß40 ratio to 1:1. Only at 4 months did plaque deposition become detectable and only in some mice; however, synaptic changes were evident in all hippocampal fields. These changes included increased glutamate release probability (P < 0.001, n = 7-9; consistent with the proposed physiological effect of amyloid-ß) and loss of spontaneous action potential-mediated activity in the cornu ammonis 1 (CA1) and dentate gyrus regions of the hippocampus (P < 0.001, n = 7). Hence synaptic changes occur when the amyloid-ß levels and amyloid-ß42:amyloid-ß40 ratio are still low compared to those necessary for plaque deposition. Genome-wide microarray analysis revealed changes in gene expression at 2-4 months including synaptic genes being strongly affected but often showing significant changes only by 4 months. We thus demonstrate that, in a mouse model of rising amyloid-ß, the initial deposition of plaques does not occur until several months after the first amyloid-ß becomes detectable but coincides with a rapid acceleration in the rise of amyloid-ß levels and the amyloid-ß42:amyloid-ß40 ratio. Prior to acceleration, however, there is already a pronounced synaptic dysfunction, reflected as changes in synaptic transmission and altered gene expression, indicating that restoring synaptic function early in the disease progression may represent the earliest possible target for intervention in the onset of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Placa Amiloide/patología , Transmisión Sináptica/fisiología , Animales , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Hipocampo/patología , Inmunoprecipitación , Espectrometría de Masas , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Técnicas de Placa-Clamp , Placa Amiloide/genética , Placa Amiloide/metabolismo , Transcriptoma
6.
Neural Plast ; 2016: 6170509, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26881123

RESUMEN

The laying down of memory requires strong stimulation resulting in specific changes in synaptic strength and corresponding changes in size of dendritic spines. Strong stimuli can also be pathological, causing a homeostatic response, depressing and shrinking the synapse to prevent damage from too much Ca(2+) influx. But do all types of dendritic spines serve both of these apparently opposite functions? Using confocal microscopy in organotypic slices from mice expressing green fluorescent protein in hippocampal neurones, the size of individual spines along sections of dendrite has been tracked in response to application of tetraethylammonium. This strong stimulus would be expected to cause both a protective homeostatic response and long-term potentiation. We report separation of these functions, with spines of different sizes reacting differently to the same strong stimulus. The immediate shrinkage of large spines suggests a homeostatic protective response during the period of potential danger. In CA1, long-lasting growth of small spines subsequently occurs consolidating long-term potentiation but only after the large spines return to their original size. In contrast, small spines do not change in dentate gyrus where potentiation does not occur. The separation in time of these changes allows clear functional differentiation of spines of different sizes.


Asunto(s)
Espinas Dendríticas/fisiología , Hipocampo/citología , Hipocampo/fisiología , Homeostasis , Potenciación a Largo Plazo , Células Piramidales/citología , Células Piramidales/fisiología , Animales , Células Cultivadas , Espinas Dendríticas/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , Hipocampo/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Ratones , Células Piramidales/efectos de los fármacos , Tetraetilamonio/farmacología
7.
Neurodegener Dis ; 15(2): 93-108, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25871323

RESUMEN

BACKGROUND: Huntington's disease (HD) is a late-onset fatal neurodegenerative disorder caused by a CAG trinucleotide repeat expansion in the gene coding for the protein huntingtin and is characterised by progressive motor, psychiatric and cognitive decline. We previously demonstrated that normal synaptic function in HD could be restored by application of dopamine receptor agonists, suggesting that changes in the release or bioavailability of dopamine may be a contributing factor to the disease process. OBJECTIVE: In the present study, we examined the properties of midbrain dopaminergic neurones and dopamine release in presymptomatic and symptomatic transgenic HD mice. METHODS AND RESULTS: Using intracellular sharp recordings and immunohistochemistry, we found that neuronal excitability was increased due to a loss of slow afterhyperpolarisation and that these changes were related to an apparent functional loss and abnormal distribution of SK3 channels (KCa2.3 encoded by the KCNN3 gene), a class of small-conductance calcium-activated potassium channels. Electrochemical detection of dopamine showed that this observation was associated with an enhanced dopamine release in presymptomatic transgenic mice and a drastic reduction in symptomatic animals. These changes occurred in the context of a progressive expansion in the CAG repeat number and nuclear localisation of mutant protein within the substantia nigra pars compacta. CONCLUSIONS: Dopaminergic neuronal dysfunction is a key early event in HD disease progression. The initial increase in dopamine release appears to be related to a loss of SK3 channel function, a protein containing a polyglutamine tract. Implications for polyglutamine-mediated sequestration of SK3 channels, dopamine-associated DNA damage and CAG expansion are discussed in the context of HD.


Asunto(s)
Encéfalo/patología , Neuronas Dopaminérgicas/fisiología , Enfermedad de Huntington/patología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Animales , Fenómenos Biofísicos/genética , Modelos Animales de Enfermedad , Dopamina/metabolismo , Estimulación Eléctrica , Femenino , Regulación de la Expresión Génica/genética , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Técnicas In Vitro , Masculino , Potenciales de la Membrana/genética , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Expansión de Repetición de Trinucleótido/genética , Tirosina 3-Monooxigenasa/metabolismo
8.
Hippocampus ; 24(12): 1413-6, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25208523

RESUMEN

Glycogen synthase kinase-3 (GSK3), particularly the isoform GSK3ß, has been implicated in a wide range of physiological systems and neurological disorders including Alzheimer's Disease. However, the functional importance of GSK3α has been largely untested. The multifunctionality of GSK3 limits its potential as a drug target because of inevitable side effects. Due to its greater expression in the CNS, GSK3ß rather than GSK3α has also been assumed to be of primary importance in synaptic plasticity. Here, we investigate bidirectional long-term synaptic plasticity in knockin mice with a point mutation in GSK3α or GSK3ß that prevents their inhibitory regulation. We report that only the mutation in GSK3α affects long-term potentiation (LTP) and depression (LTD). This stresses the importance of investigating isoform specificity for GSK3 in all systems and suggests that GSK3α should be investigated as a drug target in cognitive disorders including Alzheimer's Disease.


Asunto(s)
Región CA1 Hipocampal/enzimología , Región CA3 Hipocampal/enzimología , Glucógeno Sintasa Quinasa 3/metabolismo , Plasticidad Neuronal/fisiología , Sinapsis/enzimología , Envejecimiento/fisiología , Animales , Región CA1 Hipocampal/crecimiento & desarrollo , Región CA3 Hipocampal/crecimiento & desarrollo , Potenciales Postsinápticos Excitadores/fisiología , Técnicas de Sustitución del Gen , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Isoenzimas , Masculino , Ratones Transgénicos , Mutación , Técnicas de Cultivo de Tejidos
9.
Proc Natl Acad Sci U S A ; 107(29): 13141-6, 2010 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-20624961

RESUMEN

Neurofibromatosis type I (NF1) is one of the most common single-gene causes of learning disabilities. Here, we use behavioral working memory probes and electrophysiological studies in a mouse model of NF1 (Nf1 heterozygous null mutants; Nf1(+/-)) to demonstrate that (i) Neurofibromin regulates prefrontal and striatal inhibitory networks, specifically activity-dependent GABA release and (ii) is required for working memory performance, with inhibition-dependent working memory deficits seen in Nf1(+/-) mice. We find that increased inhibition in medial prefrontal cortex (mPFC) is sufficient to alter persistent activity in a biophysical model of an mPFC microcircuit, suggesting a possible mechanism for Nf1(+/-) working memory deficits. Accordingly, working memory assays applied during functional MRI (fMRI) studies in human subjects with NF1 reveal hypoactivation of corticostriatal networks, which is associated with impaired working memory performance. Collectively, these integrative mouse and human studies reveal molecular and cellular mechanisms contributing to working memory deficits in NF1.


Asunto(s)
Memoria a Corto Plazo/fisiología , Neostriado/metabolismo , Inhibición Neural/fisiología , Neurofibromina 1/metabolismo , Animales , Conducta Animal/fisiología , Simulación por Computador , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Humanos , Potenciales Postsinápticos Inhibidores/fisiología , Masculino , Ratones , Modelos Biológicos , Neostriado/fisiopatología , Neurofibromatosis 1/fisiopatología , Neurofibromina 1/deficiencia , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiopatología , Transducción de Señal , Adulto Joven , Ácido gamma-Aminobutírico/metabolismo , Proteínas ras/metabolismo
10.
J Neurophysiol ; 107(2): 677-91, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22072510

RESUMEN

The R6/2 mouse is the most frequently used model for experimental and preclinical drug trials in Huntington's disease (HD). When the R6/2 mouse was first developed, it carried exon 1 of the huntingtin gene with ~150 cytosine-adenine-guanine (CAG) repeats. The model presented with a rapid and aggressive phenotype that shared many features with the human condition and was particularly similar to juvenile HD. However, instability in the CAG repeat length due to different breeding practices has led to both decreases and increases in average CAG repeat lengths among colonies. Given the inverse relationship in human HD between CAG repeat length and age at onset and to a degree, the direct relationship with severity of disease, we have investigated the effect of altered CAG repeat length. Four lines, carrying ~110, ~160, ~210, and ~310 CAG repeats, were examined using a battery of tests designed to assess the basic R6/2 phenotype. These included electrophysiological properties of striatal medium-sized spiny neurons, motor activity, inclusion formation, and protein expression. The results showed an unpredicted, inverted "U-shaped" relationship between CAG repeat length and phenotype; increasing the CAG repeat length from 110 to 160 exacerbated the R6/2 phenotype, whereas further increases to 210 and 310 CAG repeats greatly ameliorated the phenotype. These findings demonstrate that the expected relationship between CAG repeat length and disease severity observed in humans is lost in the R6/2 mouse model and highlight the importance of CAG repeat-length determination in preclinical drug trials that use this model.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/fisiopatología , Fenotipo , Expansión de Repetición de Trinucleótido/genética , Análisis de Varianza , Animales , Peso Corporal/genética , Cuerpo Estriado/patología , Cuerpo Estriado/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Agonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/fisiología , Conducta Exploratoria/fisiología , Genotipo , Humanos , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/genética , Fuerza Muscular/genética , N-Metilaspartato/farmacología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/patología , Neuronas/fisiología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Técnicas de Placa-Clamp , Prueba de Desempeño de Rotación con Aceleración Constante , Convulsiones/etiología , Convulsiones/genética
11.
Cell Rep ; 41(8): 111686, 2022 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-36417868

RESUMEN

Using spatial cell-type-enriched transcriptomics, we compare plaque-induced gene (PIG) expression in microglia-touching plaques, neighboring plaques, and far from plaques in an aged Alzheimer's mouse model with late plaque development. In 18-month-old APPNL-F/NL-F knockin mice, with and without the Alzheimer's disease risk mutation Trem2R47H/R47H, we report that expression of 38/55 PIGs have plaque-induced microglial upregulation, with a subset only upregulating in microglia directly contacting plaques. For seven PIGs, including Trem2, this upregulation is prevented in APPNL-F/NL-FTrem2R47H/R47H mice. These TREM2-dependent genes are all involved in phagocytic and degradative processes that we show correspond to a decrease in phagocytic markers and an increase in the density of small plaques in Trem2-mutated mice. Furthermore, despite the R47H mutation preventing increased Trem2 gene expression, TREM2 protein levels and microglial density are still marginally increased on plaques. Hence, both microglial contact with plaques and functioning TREM2 are necessary for microglia to respond appropriately to amyloid pathology.


Asunto(s)
Enfermedad de Alzheimer , Amiloidosis , Animales , Ratones , Microglía/metabolismo , Enfermedad de Alzheimer/metabolismo , Placa Amiloide/metabolismo , Proteínas Amiloidogénicas/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo
12.
Mol Neurodegener ; 16(1): 47, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34266459

RESUMEN

BACKGROUND: Microglia are active modulators of Alzheimer's disease but their role in relation to amyloid plaques and synaptic changes due to rising amyloid beta is unclear. We add novel findings concerning these relationships and investigate which of our previously reported results from transgenic mice can be validated in knock-in mice, in which overexpression and other artefacts of transgenic technology are avoided. METHODS: AppNL-F and AppNL-G-F knock-in mice expressing humanised amyloid beta with mutations in App that cause familial Alzheimer's disease were compared to wild type mice throughout life. In vitro approaches were used to understand microglial alterations at the genetic and protein levels and synaptic function and plasticity in CA1 hippocampal neurones, each in relationship to both age and stage of amyloid beta pathology. The contribution of microglia to neuronal function was further investigated by ablating microglia with CSF1R inhibitor PLX5622. RESULTS: Both App knock-in lines showed increased glutamate release probability prior to detection of plaques. Consistent with results in transgenic mice, this persisted throughout life in AppNL-F mice but was not evident in AppNL-G-F with sparse plaques. Unlike transgenic mice, loss of spontaneous excitatory activity only occurred at the latest stages, while no change could be detected in spontaneous inhibitory synaptic transmission or magnitude of long-term potentiation. Also, in contrast to transgenic mice, the microglial response in both App knock-in lines was delayed until a moderate plaque load developed. Surviving PLX5266-depleted microglia tended to be CD68-positive. Partial microglial ablation led to aged but not young wild type animals mimicking the increased glutamate release probability in App knock-ins and exacerbated the App knock-in phenotype. Complete ablation was less effective in altering synaptic function, while neither treatment altered plaque load. CONCLUSIONS: Increased glutamate release probability is similar across knock-in and transgenic mouse models of Alzheimer's disease, likely reflecting acute physiological effects of soluble amyloid beta. Microglia respond later to increased amyloid beta levels by proliferating and upregulating Cd68 and Trem2. Partial depletion of microglia suggests that, in wild type mice, alteration of surviving phagocytic microglia, rather than microglial loss, drives age-dependent effects on glutamate release that become exacerbated in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen/métodos , Microglía/metabolismo , Placa Amiloide/patología , Transmisión Sináptica/fisiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Humanos , Ratones
13.
J Neurosci ; 29(33): 10371-86, 2009 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-19692612

RESUMEN

Previously, we identified progressive alterations in spontaneous EPSCs and IPSCs in the striatum of the R6/2 mouse model of Huntington's disease (HD). Medium-sized spiny neurons from these mice displayed a lower frequency of EPSCs, and a population of cells exhibited an increased frequency of IPSCs beginning at approximately 40 d, a time point when the overt behavioral phenotype begins. The cortex provides the major excitatory drive to the striatum and is affected during disease progression. We examined spontaneous EPSCs and IPSCs of somatosensory cortical pyramidal neurons in layers II/III in slices from three different mouse models of HD: the R6/2, the YAC128, and the CAG140 knock-in. Results revealed that spontaneous EPSCs occurred at a higher frequency, and evoked EPSCs were larger in behaviorally phenotypic mice whereas spontaneous IPSCs were initially increased in frequency in all models and subsequently decreased in R6/2 mice after they displayed the typical R6/2 overt behavioral phenotype. Changes in miniature IPSCs and evoked IPSC paired-pulse ratios suggested altered probability of GABA release. Also, in R6/2 mice, blockade of GABA(A) receptors induced complex discharges in slices and seizures in vivo at all ages. In conclusion, altered excitatory and inhibitory inputs to pyramidal neurons in the cortex in HD appear to be a prevailing deficit throughout the development of the disease. Furthermore, the differences between synaptic phenotypes in cortex and striatum are important for the development of future therapeutic approaches, which may need to be targeted early in the development of the phenotype.


Asunto(s)
Corteza Cerebral/fisiología , Modelos Animales de Enfermedad , Enfermedad de Huntington/genética , Enfermedad de Huntington/fisiopatología , Inhibición Neural/fisiología , Potenciales Sinápticos/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Células Piramidales/fisiología
14.
J Neurosci ; 29(8): 2414-27, 2009 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-19244517

RESUMEN

Huntington disease is a genetic neurodegenerative disorder that produces motor, neuropsychiatric, and cognitive deficits and is caused by an abnormal expansion of the CAG tract in the huntingtin (htt) gene. In humans, mutated htt induces a preferential loss of medium spiny neurons in the striatum and, to a lesser extent, a loss of cortical neurons as the disease progresses. The mechanisms causing these degenerative changes remain unclear, but they may involve synaptic dysregulation. We examined the activity of the corticostriatal pathway using a combination of electrophysiological and optical imaging approaches in brain slices and acutely dissociated neurons from the YAC128 mouse model of Huntington disease. The results demonstrated biphasic age-dependent changes in corticostriatal function. At 1 month, before the behavioral phenotype develops, synaptic currents and glutamate release were increased. At 7 and 12 months, after the development of the behavioral phenotype, evoked synaptic currents were reduced. Glutamate release was decreased by 7 months and was markedly reduced by 12 months. These age-dependent alterations in corticostriatal activity were paralleled by a decrease in dopamine D(2) receptor modulation of the presynaptic terminal. Together, these findings point to dynamic alterations at the corticostriatal pathway and emphasize that therapies directed toward preventing or alleviating symptoms need to be specifically designed depending on the stage of disease progression.


Asunto(s)
Envejecimiento/fisiología , Corteza Cerebral/fisiopatología , Cuerpo Estriado/fisiopatología , Enfermedad de Huntington/patología , Vías Nerviosas/fisiopatología , Factores de Edad , Análisis de Varianza , Animales , Biofisica , Cadmio/farmacología , Células Cultivadas , Cromosomas Artificiales de Levadura/genética , Modelos Animales de Enfermedad , Dopaminérgicos/farmacología , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/fisiopatología , Técnicas In Vitro , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Potenciales de la Membrana/fisiología , Ratones , Neuronas/efectos de los fármacos , Neuronas/patología , Neuronas/fisiología , Compuestos de Piridinio/metabolismo , Compuestos de Amonio Cuaternario/metabolismo , Estadísticas no Paramétricas , Factores de Tiempo , Expansión de Repetición de Trinucleótido/genética
15.
Eur J Neurosci ; 31(1): 14-28, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20092552

RESUMEN

Striatal medium-sized spiny neurons (MSSNs) receive glutamatergic inputs modulated presynaptically and postsynaptically by dopamine. Mice expressing the gene for enhanced green fluorescent protein as a reporter gene to identify MSSNs containing D1 or D2 receptor subtypes were used to examine dopamine modulation of spontaneous excitatory postsynaptic currents (sEPSCs) in slices and postsynaptic N-methyl-d-aspartate (NMDA) and alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) currents in acutely isolated cells. The results demonstrated dopamine receptor-specific modulation of sEPSCs. Dopamine and D1 agonists increased sEPSC frequency in D1 receptor-expressing MSSNs (D1 cells), whereas dopamine and D2 agonists decreased sEPSC frequency in D2 receptor-expressing MSSNs (D2 cells). These effects were fully (D1 cells) or partially (D2 cells) mediated through retrograde signaling via endocannabinoids. A cannabinoid 1 receptor (CB1R) agonist and a blocker of anandamide transporter prevented the D1 receptor-mediated increase in sEPSC frequency in D1 cells, whereas a CB1R antagonist partially blocked the decrease in sEPSC frequency in D2 cells. At the postsynaptic level, low concentrations of a D1 receptor agonist consistently increased NMDA and AMPA currents in acutely isolated D1 cells, whereas a D2 receptor agonist decreased these currents in acutely isolated D2 cells. These results show that both glutamate release and postsynaptic excitatory currents are regulated in opposite directions by activation of D1 or D2 receptors. The direction of this regulation is also specific to D1 and D2 cells. We suggest that activation of postsynaptic dopamine receptors controls endocannabinoid mobilization, acting on presynaptic CB1Rs, thus modulating glutamate release differently in glutamate terminals projecting to D1 and D2 cells.


Asunto(s)
Moduladores de Receptores de Cannabinoides/metabolismo , Cuerpo Estriado/fisiología , Dopamina/metabolismo , Endocannabinoides , Neuronas/fisiología , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Animales , Células Cultivadas , Cuerpo Estriado/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Ácido Glutámico/metabolismo , Proteínas Fluorescentes Verdes/genética , Técnicas In Vitro , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Transgénicos , N-Metilaspartato/metabolismo , Neuronas/efectos de los fármacos , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/metabolismo , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D2/agonistas , Transducción de Señal , Sinapsis/efectos de los fármacos , Sinapsis/fisiología , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/metabolismo
16.
Prog Neurobiol ; 81(5-6): 253-71, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17169479

RESUMEN

The corticostriatal pathway provides most of the excitatory glutamatergic input into the striatum and it plays an important role in the development of the phenotype of Huntington's disease (HD). This review summarizes results obtained from genetic HD mouse models concerning various alterations in this pathway. Evidence indicates that dysfunctions of striatal circuits and cortical neurons that make up the corticostriatal pathway occur during the development of the HD phenotype, well before there is significant neuronal cell loss. Morphological changes in the striatum are probably primed initially by alterations in the intrinsic functional properties of striatal medium-sized spiny neurons. Some of these alterations, including increased sensitivity of N-methyl-D-aspartate receptors in subpopulations of neurons, might be constitutively present but ultimately require abnormalities in the corticostriatal inputs for the phenotype to be expressed. Dysfunctions of the corticostriatal pathway are complex and there are multiple changes as demonstrated by significant age-related transient and more chronic interactions with the disease state. There also is growing evidence for changes in cortical microcircuits that interact to induce dysfunctions of the corticostriatal pathway. The conclusions of this review emphasize, first, the general role of neuronal circuits in the expression of the HD phenotype and, second, that both cortical and striatal circuits must be included in attempts to establish a framework for more rational therapeutic strategies in HD. Finally, as changes in cortical and striatal circuitry are complex and in some cases biphasic, therapeutic interventions should be regionally specific and take into account the temporal progression of the phenotype.


Asunto(s)
Corteza Cerebral/fisiopatología , Cuerpo Estriado/fisiopatología , Enfermedad de Huntington/patología , Vías Nerviosas/fisiopatología , Animales , Corteza Cerebral/patología , Cuerpo Estriado/patología , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/fisiopatología , Vías Nerviosas/patología
17.
Brain Commun ; 1(1): fcz022, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32274467

RESUMEN

Genome-wide association studies of late-onset Alzheimer's disease risk have previously identified genes primarily expressed in microglia that form a transcriptional network. Using transgenic mouse models of amyloid deposition, we previously showed that many of the mouse orthologues of these risk genes are co-expressed and associated with amyloid pathology. In this new study, we generate an improved RNA-seq-derived network that is expressed in amyloid-responsive mouse microglia and we statistically compare this with gene-level variation in previous human Alzheimer's disease genome-wide association studies to predict at least four new risk genes for the disease (OAS1, LAPTM5, ITGAM/CD11b and LILRB4). Of the mouse orthologues of these genes Oas1a is likely to respond directly to amyloid at the transcriptional level, similarly to established risk gene Trem2, because the increase in Oas1a and Trem2 transcripts in response to amyloid deposition in transgenic mice is significantly higher than both the increase of the average microglial transcript and the increase in microglial number. In contrast, the mouse orthologues of LAPTM5, ITGAM/CD11b and LILRB4 (Laptm5, Itgam/CD11b and Lilra5) show increased transcripts in the presence of amyloid plaques similar in magnitude to the increase of the average microglial transcript and the increase in microglia number, except that Laptm5 and Lilra5 transcripts increase significantly quicker than the average microglial transcript as the plaque load becomes dense. This work suggests that genetic variability in the microglial response to amyloid deposition is a major determinant for Alzheimer's disease risk, and identification of these genes may help to predict the risk of developing Alzheimer's disease. These findings also provide further insights into the mechanisms underlying Alzheimer's disease for potential drug discovery.

18.
EBioMedicine ; 39: 422-435, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30555043

RESUMEN

BACKGROUND: Progression of Alzheimer's disease is thought initially to depend on rising amyloidß and its synaptic interactions. Transgenic mice (TASTPM; APPSwe/PSEN1M146V) show altered synaptic transmission, compatible with increased physiological function of amyloidß, before plaques are detected. Recently, the importance of microglia has become apparent in the human disease. Similarly, TASTPM show a close association of plaque load with upregulated microglial genes. METHODS: CA1 synaptic transmission and plasticity were investigated using in vitro electrophysiology. Microglial relationship to plaques was examined with immunohistochemistry. Behaviour was assessed with a forced-alternation T-maze, open field, light/dark box and elevated plus maze. FINDINGS: The most striking finding is the increase in microglial numbers in TASTPM, which, like synaptic changes, begins before plaques are detected. Further increases and a reactive phenotype occur later, concurrent with development of larger plaques. Long-term potentiation is initially enhanced at pre-plaque stages but decrements with the initial appearance of plaques. Finally, despite altered plasticity, TASTPM have little cognitive deficit, even with a heavy plaque load, although they show altered non-cognitive behaviours. INTERPRETATION: The pre-plaque synaptic changes and microglial proliferation are presumably related to low, non-toxic amyloidß levels in the general neuropil and not directly associated with plaques. However, as plaques grow, microglia proliferate further, clustering around plaques and becoming phagocytic. Like in humans, even when plaque load is heavy, without development of neurofibrillary tangles and neurodegeneration, these alterations do not result in cognitive deficits. Behaviours are seen that could be consistent with pre-diagnosis changes in the human condition. FUNDING: GlaxoSmithKline; BBSRC; UCL; ARUK; MRC.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Cognición/fisiología , Hipocampo/fisiología , Microglía/fisiología , Presenilina-1/genética , Animales , Conducta Animal , Modelos Animales de Enfermedad , Hemicigoto , Hipocampo/metabolismo , Humanos , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos , Microglía/metabolismo , Transmisión Sináptica
19.
J Neurosci Res ; 86(10): 2147-58, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18381762

RESUMEN

Gap junctions provide a means for electrotonic coupling between neurons, allowing for the generation of synchronous activity, an important contributor to learning and memory. Connexin36 (Cx36) is largely neuron specific and provides a target for genetic manipulation to determine the physiological relevance of neuronal coupling. Within the striatum, Cx36 is more specifically localized to the interneuronal population, which provides the main inhibitory input to the principal projection medium-sized spiny neurons. In the present study, we examined the impact of genetic ablation of Cx36 on striatal spontaneous synaptic activity. Patch-clamp recordings were performed from medium-sized spiny neurons, the primary target of interneurons. In Cx36 knockout mice, the frequencies of both excitatory and inhibitory spontaneous postsynaptic currents were reduced. We also showed that activation of dopamine receptors differentially modulated the frequency of GABAergic currents in Cx36 knockout mice compared with their wild-type littermates, suggesting that dopamine plays a role in altering the coupling of interneurons. Taken together, the present findings demonstrate that electrical coupling of neuronal populations is important for the maintenance of normal chemical synaptic interactions within the striatum.


Asunto(s)
Comunicación Celular/fisiología , Conexinas/metabolismo , Cuerpo Estriado/metabolismo , Neuronas/metabolismo , Transmisión Sináptica/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Antimaláricos/farmacología , Comunicación Celular/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Dopamina/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/metabolismo , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/fisiología , Mefloquina/farmacología , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Sodio/farmacología , Transmisión Sináptica/efectos de los fármacos , Tetrodotoxina/farmacología , Proteína delta-6 de Union Comunicante
20.
Artículo en Inglés | MEDLINE | ID: mdl-30745408

RESUMEN

Mouse models of Alzheimer's disease have commonly used transgenic overexpression of genes involved in production of amyloid ß (APP and/or PSEN1/2) or Tau (MAPT) with mutations that result in familial forms of dementia. We discuss possible improvements that may create full models while avoiding the problems of overexpression and report synaptic results in APPKI models. We stress use of inappropriate controls without overexpression of the normal human protein and the mismatch between the learning deficits reported in mice with plaques but no tangles and the human condition. We focus on Tau overexpression, including new data that support previous reports of the grossly nonlinear relationship between Tau overexpression and neurofibrillary tangle load, with a twofold increase in Tau protein, resulting in a 100-fold increase in tangle density. These data also support the hypothesis that a high concentration of soluble Tau, in overexpression models, plays an important direct role in neurodegeneration, rather than only via aggregation. Finally, we hypothesize that there is an optimal concentration range over which Tau can bind to microtubules and a threshold beyond which much of the overexpressed protein is unable to bind. The excess thus causes toxicity in ways not necessarily related to the process in human dementias.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA