Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Neurobiol Aging ; 108: 189-195, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34340865

RESUMEN

Late-onset Alzheimer's disease (AD) has a significant genetic component, but the molecular mechanisms through which genetic risk factors contribute to AD pathogenesis are unclear. We screened for genetic sharing between AD and the blood levels of 615 metabolites to elucidate how the polygenic architecture of AD affects metabolomic profiles. We retrieved summary statistics from genome-wide association studies of AD and the metabolite blood levels and assessed for shared genetic etiology, using a polygenic risk score-based approach. For the blood levels of 31 metabolites, all of which were lipids, we identified and replicated genetic sharing with AD. We also found a positive genetic concordance - implying that genetic risk factors for AD are associated with higher blood levels - for 16 of the 31 replicated metabolites. In the brain, lipids and their intermediate metabolites have essential structural and functional roles, such as forming and dynamically regulating synaptic membranes. Our results imply that genetic risk factors for AD affect lipid levels, which may be leveraged to develop novel treatment strategies for AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Estudio de Asociación del Genoma Completo , Lípidos/sangre , Herencia Multifactorial/genética , Anciano , Encéfalo/metabolismo , Encéfalo/patología , Femenino , Humanos , Masculino , Metabolómica , Riesgo , Membranas Sinápticas/metabolismo , Membranas Sinápticas/patología
2.
Eur J Histochem ; 65(s1)2021 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-34346666

RESUMEN

The vertical ladder-based protocols contribute to the NMJ junction's adaptations, and when combined with and without load, can be potentiated. The present study aimed to investigate postsynaptic regions of the biceps brachii muscle in adult male Wistar rats submitted to different vertical ladder-based protocols (Sedentary - S; Climbing - C; Climbing with Load - LC and Combined Climbing - CC). The protocols (C, LC, CC) were performed in 24 sessions, 3 x/week, for 8 weeks. The myofibrillar ATPase analysis showed an increase in cross-sectional area (CSA) of the muscle fibers Type I in all trained Groups; Type II in C and LC and reduction in CC; Type IIx higher in all trained Groups. In the postsynaptic cleft, the stained area presents smaller in Groups C, LC, and CC; the total area showed smaller than LC and higher in C and CC. The stained and total perimeter, and dispersion showed a reduction in C, LC, and CC, higher maximum diameter in Groups C and CC, and decreased in LC. Regarding the postsynaptic cleft distribution, the stained area presented a decrease in all trained Groups. The integrated density presented higher principally in CC. The NMJ count showed an increase in all trained Groups. We concluded that the vertical ladder-based protocols combined contributed to the postsynaptic region adaptations.


Asunto(s)
Hipertrofia/fisiopatología , Actividad Motora , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/fisiopatología , Condicionamiento Físico Animal , Sinapsis/patología , Membranas Sinápticas/patología , Animales , Masculino , Modelos Teóricos , Ratas , Ratas Wistar
3.
Mol Med Rep ; 23(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33495808

RESUMEN

The release of neurotransmitters following the fusion of synaptic vesicles and the presynaptic membrane is an important process in the transmission of neuronal information. Syntaxin-binding protein 1 (Munc18-1) is a synaptic fusion protein binding protein, which mainly regulates synaptic vesicle fusion and neurotransmitter release by interacting with soluble N-ethylmaleimide sensitive factor attachment protein receptor. In addition to affecting neurotransmitter transmission, Munc18-1 is also involved in regulating neurosynaptic plasticity, neurodevelopment and neuroendocrine cell release functions (including thyroxine and insulin release). A number of previous studies have demonstrated that Munc18-1 has diverse and vital biological functions, and that its abnormal expression serves an important role in the pathogenesis of a variety of neurological diseases, including epileptic encephalopathy, schizophrenia, autism, Parkinson's disease, Alzheimer's disease, multiple sclerosis, Duchenne's muscular dystrophy and neuronal ceroid lipofuscinosis. The present review summarizes the function of Munc18-1 and its possible relationship to the pathogenesis of various neurological diseases.


Asunto(s)
Proteínas Munc18/metabolismo , Enfermedades del Sistema Nervioso/metabolismo , Membranas Sinápticas/metabolismo , Transmisión Sináptica , Animales , Humanos , Fusión de Membrana , Proteínas Munc18/genética , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/patología , Membranas Sinápticas/genética , Membranas Sinápticas/patología
4.
Proc Natl Acad Sci U S A ; 115(40): 10166-10171, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30224498

RESUMEN

The type 2 K+/Cl- cotransporter (KCC2) allows neurons to maintain low intracellular levels of Cl-, a prerequisite for efficient synaptic inhibition. Reductions in KCC2 activity are evident in epilepsy; however, whether these deficits directly contribute to the underlying pathophysiology remains controversial. To address this issue, we created knock-in mice in which threonines 906 and 1007 within KCC2 have been mutated to alanines (KCC2-T906A/T1007A), which prevents its phospho-dependent inactivation. The respective mice appeared normal and did not show any overt phenotypes, and basal neuronal excitability was unaffected. KCC2-T906A/T1007A mice exhibited increased basal neuronal Cl- extrusion, without altering total or plasma membrane accumulation of KCC2. Critically, activity-induced deficits in synaptic inhibition were reduced in the mutant mice. Consistent with this, enhanced KCC2 was sufficient to limit chemoconvulsant-induced epileptiform activity. Furthermore, this increase in KCC2 function mitigated induction of aberrant high-frequency activity during seizures, highlighting depolarizing GABA as a key contributor to the pathological neuronal synchronization seen in epilepsy. Thus, our results demonstrate that potentiating KCC2 represents a therapeutic strategy to alleviate seizures.


Asunto(s)
Epilepsia/metabolismo , Neuronas/metabolismo , Convulsiones/metabolismo , Simportadores/metabolismo , Membranas Sinápticas/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Sustitución de Aminoácidos , Animales , Epilepsia/genética , Epilepsia/patología , Técnicas de Sustitución del Gen , Ratones , Mutación Missense , Neuronas/patología , Convulsiones/genética , Convulsiones/patología , Simportadores/genética , Membranas Sinápticas/genética , Membranas Sinápticas/patología , Ácido gamma-Aminobutírico/genética , Cotransportadores de K Cl
5.
J Biol Chem ; 293(5): 1568-1569, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29414768

RESUMEN

Long-term depression (LTD) is a reduction in the efficacy of neuronal synapses, but the molecular basis of LTD signaling and how these signals lead to phenotypic outcomes, such as the shrinkage of synaptic regions, is not clear. In a new report, Woolfrey et al use chemically-induced LTD and a multitude of in vitro biochemical assays to provide evidence that synaptic removal of the scaffolding protein AKAP79/150 promotes LTD-induced spine shrinkage. The further identification of CaMKII, a kinase primarily associated with long-term potentiation (LTP), as a requirement for AKAP79/150 removal, uncovers unexpected interplay between different post-translational modifications and points to a new model of LTD.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Depresión Sináptica a Largo Plazo , Fosfolípidos/metabolismo , Membranas Sinápticas/metabolismo , Animales , Lipoilación , Fosforilación , Dominios Proteicos , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Columna Vertebral/metabolismo , Columna Vertebral/patología , Membranas Sinápticas/patología
6.
FASEB J ; 32(2): 888-893, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29025866

RESUMEN

Interactions of the presynaptic proteins, neuronal pentraxin 2 (NPTX2) and neurexin 2α (NRXN2α), with their respective postsynaptic functional partners, GluA4-containing glutamate (AMPA4) receptor and neuroligin 1 (NLGN1), enhance excitatory synaptic activity in some areas of the hippocampus and cerebral cortex. As early damage of such excitatory circuits in the brain tissues of participants with Alzheimer's disease (AD) correlates with cognitive losses, plasma neuron-derived exosome (NDE) levels of these 2 pairs of specialized synaptic proteins were quantified to assess their biomarker characteristics. The NDE contents of all 4 proteins were decreased significantly in AD dementia ( n = 46), and diminished levels of AMPA4 and NLGN1 correlated with the extent of cognitive loss. In a preclinical period, 6-11 yr before the onset of dementia, the NDE levels of all but NPTX2 were significantly lower than those of matched controls, and levels of all proteins declined significantly with the development of dementia. Reductions in NDE levels of these specialized excitatory synaptic proteins may therefore be indicative of the extent of cognitive loss and may reflect progression of the severity of AD.-Goetzl, E. J., Abner, E. L., Jicha, G. A., Kapogiannis, D., Schwartz, J. B. Declining levels of functionally specialized synaptic proteins in plasma neuronal exosomes with progression of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/sangre , Moléculas de Adhesión Celular Neuronal/sangre , Exosomas/metabolismo , Proteínas del Tejido Nervioso/sangre , Neuronas/metabolismo , Receptores AMPA/sangre , Membranas Sinápticas/metabolismo , Anciano , Enfermedad de Alzheimer/patología , Proteína C-Reactiva , Exosomas/patología , Femenino , Humanos , Masculino , Neuronas/patología , Membranas Sinápticas/patología
7.
J Cell Biol ; 216(11): 3831-3846, 2017 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-28972101

RESUMEN

Synapse assembly likely requires postsynaptic target recognition by incoming presynaptic afferents. Using newly generated conditional knock-in and knockout mice, we show in this study that latrophilin-2 (Lphn2), a cell-adhesion G protein-coupled receptor and presumptive α-latrotoxin receptor, controls the numbers of a specific subset of synapses in CA1-region hippocampal neurons, suggesting that Lphn2 acts as a synaptic target-recognition molecule. In cultured hippocampal neurons, Lphn2 maintained synapse numbers via a postsynaptic instead of a presynaptic mechanism, which was surprising given its presumptive role as an α-latrotoxin receptor. In CA1-region neurons in vivo, Lphn2 was specifically targeted to dendritic spines in the stratum lacunosum-moleculare, which form synapses with presynaptic entorhinal cortex afferents. In this study, postsynaptic deletion of Lphn2 selectively decreased spine numbers and impaired synaptic inputs from entorhinal but not Schaffer-collateral afferents. Behaviorally, loss of Lphn2 from the CA1 region increased spatial memory retention but decreased learning of sequential spatial memory tasks. Thus, Lphn2 appears to control synapse numbers in the entorhinal cortex/CA1 region circuit by acting as a domain-specific postsynaptic target-recognition molecule.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Corteza Entorrinal/metabolismo , Neuronas/metabolismo , Terminales Presinápticos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/metabolismo , Membranas Sinápticas/metabolismo , Animales , Conducta Animal , Región CA1 Hipocampal/patología , Región CA1 Hipocampal/fisiopatología , Células Cultivadas , Espinas Dendríticas/metabolismo , Espinas Dendríticas/patología , Corteza Entorrinal/patología , Corteza Entorrinal/fisiopatología , Miedo , Genotipo , Aprendizaje por Laberinto , Memoria , Ratones Mutantes , Actividad Motora , Neuronas/patología , Fenotipo , Terminales Presinápticos/patología , Receptores Acoplados a Proteínas G/genética , Receptores de Péptidos/genética , Prueba de Desempeño de Rotación con Aceleración Constante , Olfato , Membranas Sinápticas/patología , Potenciales Sinápticos , Factores de Tiempo , Transfección
8.
J Biochem Mol Toxicol ; 31(10)2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28621878

RESUMEN

Arthropod venoms are sources of molecules that may be useful tools to investigate molecular mechanisms of putative new medicines and laboratory drugs. Here we show the effects of the compound agelaiatoxin-8 (AVTx8), isolated from Agelaia vicina venom, on γ-aminobutyric acid (GABA) neurotransmission in rat brain synaptosomes. Analysis reveals that AvTx8 is composed by 14 amino acid residues with a molecular weight (MW) of 1567 Da. AvTx8 increased GABA release and inhibited GABA uptake in synaptosomes from rat cerebral cortex. AvTx8 inhibited GABA uptake and increased GABA release in the presence of Ca+ , Na+ , and K+ channel blockers, suggesting that it acts directly on GABA transporters. In addition, AvTx8 significantly decreases GABA binding in synaptic membranes from rat brain cortex, suggesting that it also modulates the activity of GABA receptors. Moreover, AvTx8 decreased GAT-1- and GAT-3-mediated GABA uptake in transfected COS-7 cells. Accordingly, we suggest that AvTx8 modulates GABA neurotransmission and might provide a novel entry point for identifying a new class of GABA-modulating neuroprotective drugs.


Asunto(s)
Membranas Sinápticas/metabolismo , Transmisión Sináptica/efectos de los fármacos , Sinaptosomas/metabolismo , Venenos de Avispas , Avispas/química , Ácido gamma-Aminobutírico/metabolismo , Animales , Células COS , Chlorocebus aethiops , Canales Iónicos/antagonistas & inhibidores , Canales Iónicos/metabolismo , Ratas , Ratas Wistar , Membranas Sinápticas/patología , Sinaptosomas/patología , Venenos de Avispas/química , Venenos de Avispas/aislamiento & purificación , Venenos de Avispas/toxicidad
9.
Proc Natl Acad Sci U S A ; 112(4): 1214-9, 2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25552561

RESUMEN

GABAA-receptor-based interneuron circuitry is essential for higher order function of the human nervous system and is implicated in schizophrenia, depression, anxiety disorders, and autism. Here we demonstrate that giant ankyrin-G (480-kDa ankyrin-G) promotes stability of somatodendritic GABAergic synapses in vitro and in vivo. Moreover, giant ankyrin-G forms developmentally regulated and cell-type-specific micron-scale domains within extrasynaptic somatodendritic plasma membranes of pyramidal neurons. We further find that giant ankyrin-G promotes GABAergic synapse stability through opposing endocytosis of GABAA receptors, and requires a newly described interaction with GABARAP, a GABAA receptor-associated protein. We thus present a new mechanism for stabilization of GABAergic interneuron synapses and micron-scale organization of extrasynaptic membrane that provides a rationale for studies linking ankyrin-G genetic variation with psychiatric disease and abnormal neurodevelopment.


Asunto(s)
Ancirinas/metabolismo , Endocitosis , Neuronas GABAérgicas/metabolismo , Células Piramidales/metabolismo , Receptores de GABA-A/metabolismo , Membranas Sinápticas/metabolismo , Animales , Ancirinas/genética , Proteínas Reguladoras de la Apoptosis , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Neuronas GABAérgicas/patología , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Trastornos Mentales/genética , Trastornos Mentales/metabolismo , Trastornos Mentales/patología , Ratones , Proteínas Asociadas a Microtúbulos , Células Piramidales/patología , Receptores de GABA-A/genética , Membranas Sinápticas/genética , Membranas Sinápticas/patología
10.
J Neurosci Res ; 92(5): 641-50, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24446209

RESUMEN

The APOE genotype is the major risk factor for Alzheimer's disease (AD); however, it remains unclarified how the ε4 allele accelerates whereas the ε2 allele suppresses AD development, compared with the more common ε3 allele. On the basis of the previous finding that the assembly of the amyloid-ß protein (Aß) into fibrils in the brain, an early and invariable pathological feature of AD, depends on the lipid environment, we determined the levels of synaptic membrane lipids in aged individuals of different APOE genotypes. In the comparison between amyloid-free ε2/ε3 and ε3/ε3 brains, the presence of the ε2 allele significantly decreased the level of cholesterol. Alternatively, in the comparison among ε3/ε3 brains, the presence of AD pathology substantially decreased the levels of cholesterol. This study suggests that the ε2 allele suppresses the initiation of AD development by lowering the cholesterol levels in synaptic membranes.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Apolipoproteínas E/genética , Encéfalo/patología , Lípidos , Sinaptosomas/metabolismo , Anciano , Anciano de 80 o más Años , Péptidos beta-Amiloides/metabolismo , Colesterol/metabolismo , Femenino , Gangliósidos/metabolismo , Genotipo , Humanos , Lípidos/genética , Masculino , Membranas Sinápticas/genética , Membranas Sinápticas/patología , Membranas Sinápticas/ultraestructura
11.
Autoimmun Rev ; 12(9): 912-7, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23537506

RESUMEN

The presence of autoantibodies against muscle-specific kinase (MuSK) at the neuromuscular junction (NMJ) results in myasthenia gravis (MG). MuSK antibody-associated MG (MuSK MG) patients often have severe symptoms, including bulbar dysfunction, respiratory insufficiency and atrophy of the facial and tongue muscles. MuSK antibodies in MG patients predominantly belong to the IgG4 subclass, and the unique properties of IgG4 antibodies are directly associated with the pathogenic mechanisms of MuSK MG. Histopathological studies in animal models of MuSK MG have revealed that anti-MuSK antibodies cause contraction of motor terminals, significant loss of acetylcholine receptor (AChR) expression, and a reduction in synaptic folds at the postsynaptic membrane in the absence of complement involvement. Failure of neuromuscular transmission at pre- and postsynaptic membranes of the NMJs has been observed in both patients and animal models of MuSK MG. A murine model of MuSK-MG revealed the mechanisms underlying cholinergic hypersensitivity after administration of acetylcholinesterase inhibitors, which has also been observed in MuSK-MG patients. Further studies of this model have provided evidence suggesting that 3,4-diaminopyridine may be effective as a symptomatic therapy for MuSK MG.


Asunto(s)
Autoanticuerpos/inmunología , Inmunoglobulina G/inmunología , Miastenia Gravis/inmunología , Miastenia Gravis/patología , Animales , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Miastenia Gravis/terapia , Unión Neuromuscular/patología , Unión Neuromuscular/fisiopatología , Proteínas Tirosina Quinasas Receptoras/inmunología , Receptores Colinérgicos/inmunología , Receptores Colinérgicos/metabolismo , Membranas Sinápticas/metabolismo , Membranas Sinápticas/patología
13.
J Biol Chem ; 287(13): 10265-10275, 2012 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-22287544

RESUMEN

Emerging evidence suggests that metabotropic glutamate receptors (mGluRs) are potential novel targets for brain disorders associated with the dysfunction of prefrontal cortex (PFC), a region critical for cognitive and emotional processes. Because N-methyl-D-aspartic acid receptor (NMDAR) dysregulation has been strongly associated with the pathophysiology of mental illnesses, we examined the possibility that mGluRs might be involved in modulating PFC functions by targeting postsynaptic NMDARs. We found that application of prototypical group III mGluR agonists significantly reduced NMDAR-mediated synaptic and ionic currents in PFC pyramidal neurons, which was mediated by mGluR7 localized at postsynaptic neurons and involved the ß-arrestin/ERK signaling pathway. The mGluR7 modulation of NMDAR currents was prevented by agents perturbing actin dynamics and by the inhibitor of cofilin, a major actin-depolymerizing factor. Consistently, biochemical and immunocytochemical results demonstrated that mGluR7 activation increased cofilin activity and F-actin depolymerization via an ERK-dependent mechanism. Furthermore, mGluR7 reduced the association of NMDARs with the scaffolding protein PSD-95 and the surface level of NMDARs in an actin-dependent manner. These data suggest that mGluR7, by affecting the cofilin/actin signaling, regulates NMDAR trafficking and function. Because ablation of mGluR7 leads to a variety of behavioral symptoms related to PFC dysfunction, such as impaired working memory and reduced anxiety and depression, our results provide a potential mechanism for understanding the role of mGluR7 in mental health and disorders.


Asunto(s)
Células Piramidales/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Membranas Sinápticas/metabolismo , Transmisión Sináptica , Factores Despolimerizantes de la Actina/genética , Factores Despolimerizantes de la Actina/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Ansiedad/genética , Ansiedad/metabolismo , Ansiedad/patología , Células Cultivadas , Depresión/genética , Depresión/metabolismo , Depresión/patología , Homólogo 4 de la Proteína Discs Large , Guanilato-Quinasas/genética , Guanilato-Quinasas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Trastornos de la Memoria/genética , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Ratones , Transporte de Proteínas/genética , Células Piramidales/patología , Ratas , Receptores de Glutamato Metabotrópico/genética , Receptores de N-Metil-D-Aspartato/genética , Membranas Sinápticas/genética , Membranas Sinápticas/patología
14.
Mol Neurobiol ; 45(1): 126-43, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22205299

RESUMEN

Alterations occur within distal neuronal compartments, including axons and synapses, during the course of neurodegenerative diseases such as Parkinson's disease (PD). These changes could hold important implications for the functioning of neural networks, especially since research studies have shown a loss of dendritic spines locating to medium spiny projection neurons and impaired axonal transport in PD-affected brains. However, despite ever-increasing awareness of the vulnerability of synapses and axons, inadequate understanding of the independent mechanisms regulating non-somatic neurodegeneration prevails. This has resulted in limited therapeutic strategies capable of targeting these distinct cellular compartments. Deregulated protein synthesis, folding and degrading proteins, and protein quality-control systems have repeatedly been linked with morphological and functional alterations of synapses in the PD-affected brains. Here, we review current understanding concerning the proteins involved in structural and functional changes that affect synaptic contact-points in PD. The collection of studies discussed emphasizes the need for developing therapeutics aimed at deregulated protein synthesis and degradation pathways operating at axonal and dendritic synapses for preserving "normal" circuitry and function, for as long as possible.


Asunto(s)
Degeneración Nerviosa/fisiopatología , Proteínas del Tejido Nervioso/metabolismo , Enfermedad de Parkinson/fisiopatología , Sinapsis/metabolismo , Animales , Humanos , Degeneración Nerviosa/patología , Proteínas del Tejido Nervioso/fisiología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Sinapsis/patología , Membranas Sinápticas/metabolismo , Membranas Sinápticas/patología , Transmisión Sináptica/fisiología , alfa-Sinucleína/metabolismo , alfa-Sinucleína/fisiología
15.
Curr Opin Neurobiol ; 20(5): 543-9, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20655194

RESUMEN

Chemical neurotransmission between neurons is a major point for modulation of neuronal activity. The neuronal synapse is the site of continuous cycles of rapid vesicle fusion (exocytosis) followed by their retrieval (endocytosis). Ongoing research efforts are largely focused on synaptic proteins involved in membrane fusion-and-fission, but it is now becoming clear that the dynamic lipid environment, where these proteins operate, also plays a key role in the modulation of chemical transmission. Growing evidence suggests that lipid metabolites regulate both vesicle fusion and retrieval highlighting the fact that membrane lipids have functions beyond the structural role. Furthermore, direct involvement of lipid metabolism in the pathogenesis of neurodegenerative disorders, such as Alzheimer's and Parkinson's diseases, necessitates rigorous investigation of the effects of lipids on synaptic mechanisms. Recent findings and possible lines of further investigation will be discussed in this review.


Asunto(s)
Lípidos de la Membrana/fisiología , Sinapsis/fisiología , Membranas Sinápticas/fisiología , Transmisión Sináptica/fisiología , Animales , Fusión Celular/métodos , Fusión Celular/tendencias , Exocitosis/genética , Exocitosis/fisiología , Humanos , Metabolismo de los Lípidos/genética , Metabolismo de los Lípidos/fisiología , Lípidos de la Membrana/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Sinapsis/patología , Membranas Sinápticas/patología
16.
J Neurochem ; 110(1): 157-69, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19453946

RESUMEN

Down syndrome (DS) is the most prevalent form of intellectual disability caused by the triplication of approximately 230 genes on chromosome 21. Recent data in Ts65Dn mice, the foremost mouse model of DS, strongly suggest that cognitive impairment in individuals with DS is a consequence of reduced synaptic plasticity because of chronic over-inhibition. It remains unclear however whether changes in plasticity are tied to global molecular changes at synapses, or are due to regional changes in the functional properties of synaptic circuits. One interesting framework for evaluating the activity state of the DS brain comes from in vitro studies showing that chronic pharmacological silencing of neuronal excitability orchestrates stereotyped changes in the protein composition of synaptic junctions. In the present study, we use proteomic strategies to evaluate whether synapses from the Ts65Dn cerebrum carry signatures characteristic of inactive cortical neurons. Our data reveal that synaptic junctions do not exhibit overt alterations in protein composition. Only modest changes in the levels of synaptic proteins and in their phosphorylation are observed. This suggests that subtle changes in the functional properties of specific synaptic circuits rather than large-scale homeostatic shifts in the expression of synaptic molecules contribute to cognitive impairment in people with DS.


Asunto(s)
Corteza Cerebral/metabolismo , Trastornos del Conocimiento/metabolismo , Síndrome de Down/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica/genética , Animales , Biomarcadores/análisis , Biomarcadores/metabolismo , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Síndrome de Down/genética , Síndrome de Down/fisiopatología , Femenino , Homeostasis/genética , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Mutantes Neurológicos , Proteínas del Tejido Nervioso/genética , Plasticidad Neuronal/genética , Fosforilación , Proteómica/métodos , Sinapsis/química , Sinapsis/genética , Membranas Sinápticas/genética , Membranas Sinápticas/metabolismo , Membranas Sinápticas/patología
17.
J Neurochem ; 110(2): 613-21, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19457102

RESUMEN

Parkin is the most common causative gene of juvenile and early-onset familial Parkinson's diseases and is thought to function as an E3 ubiquitin ligase in the ubiquitin-proteasome system. However, it remains unclear how loss of Parkin protein causes dopaminergic dysfunction and nigral neurodegeneration. To investigate the pathogenic mechanism underlying these mutations, we used parkin-/- mice to study its physiological function in the nigrostriatal circuit. Amperometric recordings showed decreases in evoked dopamine release in acute striatal slices of parkin-/- mice and reductions in the total catecholamine release and quantal size in dissociated chromaffin cells derived from parkin-/- mice. Intracellular recordings of striatal medium spiny neurons revealed impairments of long-term depression and long-term potentiation in parkin-/- mice, whereas long-term potentiation was normal in the Schaeffer collateral pathway of the hippocampus. Levels of dopamine receptors and dopamine transporters were normal in the parkin-/- striatum. These results indicate that Parkin is involved in the regulation of evoked dopamine release and striatal synaptic plasticity in the nigrostriatal pathway, and suggest that impairment in evoked dopamine release may represent a common pathophysiological change in recessive parkinsonism.


Asunto(s)
Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Plasticidad Neuronal/fisiología , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Cuerpo Estriado/fisiopatología , Estimulación Eléctrica , Femenino , Hipocampo/metabolismo , Hipocampo/fisiopatología , Depresión Sináptica a Largo Plazo/genética , Depresión Sináptica a Largo Plazo/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasticidad Neuronal/genética , Membranas Sinápticas/genética , Membranas Sinápticas/metabolismo , Membranas Sinápticas/patología , Ubiquitina-Proteína Ligasas/genética
18.
Neuromuscul Disord ; 19(5): 366-78, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19346129

RESUMEN

Mice deficient in the glycosyltransferase Large are characterized by severe muscle and central nervous system abnormalities. In this study, we show that the formation and maintenance of neuromuscular junctions in Large(myd) mice are greatly compromised. Neuromuscular junctions are not confined to the muscle endplate zone but are widely spread and are frequently accompanied by exuberant nerve sprouting. Nerve terminals are highly fragmented and binding of alpha-bungarotoxin to postsynaptic acetylcholine receptors (AChRs) is greatly reduced. In vitro, Large(myd) myotubes are responsive to agrin but produce aberrant AChR clusters, which are larger in area and less densely packed with AChRs. In addition, AChR expression on the cell surface is diminished suggesting that AChR assembly or transport is defective. These results together with the finding that O-linked glycosylation at neuromuscular junctions of Large(myd) mice is compromised indicate that the action of Large is necessary for proper neuromuscular junction development.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , N-Acetilglucosaminiltransferasas/genética , Enfermedades de la Unión Neuromuscular/genética , Enfermedades de la Unión Neuromuscular/metabolismo , Unión Neuromuscular/genética , Unión Neuromuscular/metabolismo , Agrina/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Mutantes , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/inervación , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Mutación/genética , Unión Neuromuscular/fisiopatología , Enfermedades de la Unión Neuromuscular/fisiopatología , Terminales Presinápticos/metabolismo , Terminales Presinápticos/patología , Receptores Nicotínicos/metabolismo , Membranas Sinápticas/metabolismo , Membranas Sinápticas/patología
19.
J Neurosci ; 29(7): 2193-204, 2009 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-19228972

RESUMEN

Huntington disease (HD) is a neurodegenerative disorder caused by an expanded CAG tract in the HD gene. Polyglutamine expansion of huntingtin (htt) results in early, progressive loss of medium spiny striatal neurons, as well as cortical neurons that project to the striatum. Excitotoxicity has been postulated to play a key role in the selective vulnerability of striatal neurons in HD. Early excitotoxic neuropathological changes observed in human HD brain include increased quinolinate (QUIN) concurrent with proliferative changes such as increased spine density and dendritic length. In later stages of the disease, degenerative-type changes are apparent, such as loss of dendritic arborization, a reduction in spine density and reduced levels of 3-hydroxykynurenine and QUIN. It is currently unknown whether sensitivity to excitotoxic stress varies between initiation and progression of disease. Here, we have assessed the excitotoxic phenotype in the YAC128 mouse model of HD by examining the response to excitotoxic stress at different stages of disease. Our results demonstrate that YAC128 mice display enhanced sensitivity to NMDA ex vivo and QUIN in vivo before obvious phenotypic changes. In contrast, 10-month-old symptomatic YAC128 mice are resistant to QUIN-induced neurotoxicity. These findings are paralleled by a significant increase in NMDAR-mediated membrane currents in presymptomatic YAC128 dissociated medium spiny neurons progressing to reduced NMDAR-mediated membrane currents with disease progression. These data highlight the dynamic nature of the mutant htt-mediated excitotoxic phenotype and suggests that therapeutic approaches to HD may need to be altered, depending on the stage and development of the disease.


Asunto(s)
Encéfalo/metabolismo , Predisposición Genética a la Enfermedad/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Estrés Fisiológico/genética , Animales , Encéfalo/fisiopatología , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatología , Células Cultivadas , Espinas Dendríticas/metabolismo , Espinas Dendríticas/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Enfermedad de Huntington/fisiopatología , Ratones , Ratones Transgénicos , N-Metilaspartato/metabolismo , N-Metilaspartato/toxicidad , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Neurotoxinas/metabolismo , Neurotoxinas/toxicidad , Técnicas de Cultivo de Órganos , Fenotipo , Ácido Quinolínico/metabolismo , Ácido Quinolínico/toxicidad , Membranas Sinápticas/metabolismo , Membranas Sinápticas/patología , Potenciales Sinápticos/genética , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
20.
J Biol Chem ; 284(13): 8495-506, 2009 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-19164281

RESUMEN

The amyloid precursor protein (APP) is cleaved to produce the Alzheimer disease-associated peptide Abeta, but the normal functions of uncleaved APP in the brain are unknown. We found that APP was present in the postsynaptic density of central excitatory synapses and coimmunoprecipitated with N-methyl-d-aspartate receptors (NMDARs). The presence of APP in the postsynaptic density was supported by the observation that NMDARs regulated trafficking and processing of APP; overexpression of the NR1 subunit increased surface levels of APP, whereas activation of NMDARs decreased surface APP and promoted production of Abeta. We transfected APP or APP RNA interference into primary neurons and used electrophysiological techniques to explore the effects of APP on postsynaptic function. Reduction of APP decreased (and overexpression of APP increased) NMDAR whole cell current density and peak amplitude of spontaneous miniature excitatory postsynaptic currents. The increase in NMDAR current by APP was due to specific recruitment of additional NR2B-containing receptors. Consistent with these findings, immunohistochemical experiments demonstrated that APP increased the surface levels and decreased internalization of NR2B subunits. These results demonstrate a novel physiological role of postsynaptic APP in enhancing NMDAR function.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Proteína Amiloide A Sérica/metabolismo , Membranas Sinápticas/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Encéfalo/patología , Células COS , Chlorocebus aethiops , Expresión Génica , Ratones , Ratones Noqueados , Transporte de Proteínas/genética , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética , Proteína Amiloide A Sérica/genética , Membranas Sinápticas/genética , Membranas Sinápticas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA