Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 501
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pharmacol Res ; 158: 104863, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32407957

RESUMEN

Neural stem cell (NSC) neuronal differentiation requires a metabolic shift towards oxidative phosphorylation. We now show that a branched-chain amino acids-driven, persistent metabolic shift toward energy metabolism is required for full neuronal maturation. We increased energy metabolism of differentiating neurons derived both from murine NSCs and human induced pluripotent stem cells (iPSCs) by supplementing the cell culture medium with a mixture composed of branched-chain amino acids, essential amino acids, TCA cycle precursors and co-factors. We found that treated differentiating neuronal cells with enhanced energy metabolism increased: i) total dendritic length; ii) the mean number of branches and iii) the number and maturation of the dendritic spines. Furthermore, neuronal spines in treated neurons appeared more stable with stubby and mushroom phenotype and with increased expression of molecules involved in synapse formation. Treated neurons modified their mitochondrial dynamics increasing the mitochondrial fusion and, consistently with the increase of cellular ATP content, they activated cellular mTORC1 dependent p70S6 K1 anabolism. Global transcriptomic analysis further revealed that treated neurons induce Nrf2 mediated gene expression. This was correlated with a functional increase in the Reactive Oxygen Species (ROS) scavenging mechanisms. In conclusion, persistent branched-chain amino acids-driven metabolic shift toward energy metabolism enhanced neuronal differentiation and antioxidant defences. These findings offer new opportunities to pharmacologically modulate NSC neuronal differentiation and to develop effective strategies for treating neurodegenerative diseases.


Asunto(s)
Aminoácidos de Cadena Ramificada/farmacología , Diferenciación Celular/fisiología , Metabolismo Energético/efectos de los fármacos , Células-Madre Neurales/fisiología , Adenosina Trifosfato/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/ultraestructura , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neurogénesis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Sinapsis/genética , Sinapsis/fisiología , Sinapsis/ultraestructura , Transcriptoma
2.
J Comput Neurosci ; 48(1): 1-20, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31797200

RESUMEN

Information transfer may not be limited only to synapses. Therefore, the processes and dynamics of biological neuron-astrocyte coupling and intercellular interaction within this domain are worth investigating. Existing models of tripartite synapse consider an astrocyte as a point process. Here, we extended the tripartite synapse model by considering the astrocytic processes (synaptic and perinodal) as compartments. The scattered extrinsic signals in the extracellular space and the presence of calcium stores in different astrocytic sites create local transient [Ca2+]. We investigated the Ca2+ dynamics and found that the increase in astrocytic intracellular [Ca2+] enhances the probability of neurotransmitter release. However, the period in which the extrasynaptic glutamate lingers in the extracellular space may cause excitotoxicity. We propose further biological investigation on intercellular communication, considering that unconventional sources (nonsynaptic) of glutamate may improve information processing in neuron-astrocyte networks.


Asunto(s)
Astrocitos/fisiología , Comunicación Celular/fisiología , Modelos Neurológicos , Sinapsis/fisiología , Algoritmos , Animales , Astrocitos/ultraestructura , Calcio/metabolismo , Señalización del Calcio/fisiología , Simulación por Computador , Espacio Extracelular/fisiología , Ácido Glutámico/fisiología , Humanos , Vaina de Mielina , Terminales Presinápticos/fisiología , Nódulos de Ranvier , Sinapsis/ultraestructura , Transmisión Sináptica
3.
Cell Tissue Res ; 375(1): 103-122, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30519837

RESUMEN

We examine evolutionary aspects of two primordial neuropeptides, arginine vasopressin (AVP) and pituitary adenylate cyclase-activating polypeptide (PACAP); the distribution of AVP and PACAP and their receptors in mammals; AVP and PACAP release patterns relevant to their roles in neuroendocrine control in brain and periphery; and finally the intricate interlocking of homeostatic and allostatic regulation created by extrahypothalamic AVP and PACAP projections to brain circuit nodes important in controlling appetitive, avoidance and aggressive motor responses. A cardinal feature of peptide neurotransmission important in regulatory control of organismic responses and emphasized in this review, is that neuropeptides are released from large dense-core vesicles docked not only within axonal varicosities and dendrites but also at presynaptic nerve terminal sites, along with small clear synaptic vesicles, at active zones. Peptide transmitter nerve terminals, from hypothalamic and other projections, are distributed widely to multiple brain areas important in integrative control of behavior. They converge with heterologous inputs that release other transmitters, including other peptides, in the same areas. The concept of a quasi-hormonal effect of peptide neurotransmission through coordinated release at multiple synapses throughout the brain echoes earlier conceptualizations of "action-at-a-distance" by diffusion following peptide release at non-synaptic sites. Yet, it recognizes that peptide delivery occurs with neuroanatomical precision, from discrete peptide-containing brain nuclei, via highly distributed projections to multiple extrahypothalamic nodes, registering multiple homeostatic, hedonistic, aversive and reproductive drives that modulate real-time motor decisions. There is paradigmatic value in the discussion of these two particular ancient neuropeptides, for peptide-centric translational neuroendocrinology and peptide GPCR-based neurotherapeutics.


Asunto(s)
Arginina Vasopresina/metabolismo , Hipotálamo/metabolismo , Motivación , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Sinapsis/metabolismo , Animales , Modelos Neurológicos , Sinapsis/ultraestructura
4.
Int J Immunopathol Pharmacol ; 32: 2058738418780066, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29873261

RESUMEN

Reduced glucose utilization and deficient energy metabolism that occur in the early stages of Alzheimer's disease correlate with impaired cognition, and this information is evidence that Alzheimer's disease is a metabolic disease that is associated with brain insulin/insulin-like growth factor resistance. This research aimed to investigate the effects of Banxia Xiexin decoction (BXD) on cognitive deficits in APPswe/PS1dE9 double transgenic mice and verify the hypothesis that BXD treatment improves cognitive function via improving insulin signalling, glucose metabolism and synaptic plasticity in the hippocampus of APPswe/PS1dE9 double transgenic mice. We used 3-month-old APPswe/PS1dE9 double transgenic mice as the case groups and wild-type littermates of the double transgenic mice from the same colony as the control group. Forty-five APPswe/PS1dE9 double transgenic mice were randomly divided into the model group, donepezil group and BXD group. The mice in the control and model groups were administered 0.5% carboxymethyl cellulose orally. The Morris water maze and step-down test were conducted to evaluate the cognitive performance of APPswe/PS1dE9 double transgenic mice after BXD treatment. Ultrastructure of synapses was observed in the hippocampal CA1 area. Proteins involved in insulin signalling pathways and glucose transports in the hippocampus were assessed through immunohistochemical staining and western blot. After 3 months intervention, we found that BXD treatment improved cognitive performance and the synaptic quantity and ultrastructure, restored insulin signalling and increased the expression of glucose transporter 1 (GLUT1) and GLUT3 levels. These findings suggest that the beneficial effect of BXD on cognition may be due to the improvement of insulin signalling, glucose metabolism and synaptic plasticity.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Cognición/efectos de los fármacos , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Hipocampo/efectos de los fármacos , Insulina/metabolismo , Extractos Vegetales/uso terapéutico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Hipocampo/ultraestructura , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones Transgénicos , Presenilina-1/genética , Sinapsis/efectos de los fármacos , Sinapsis/ultraestructura
5.
Mol Pain ; 14: 1744806918765837, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29592784

RESUMEN

Objective To observe the analgesic effect of early hyperbaric oxygen (HBO) treatment in chronic constriction injury (CCI) rats, and to analyze the influence of HBO on the expression of neuronal nitric oxide synthase and inducible nitric oxide synthase and on the levels of inflammatory factors. Methods Rats were assigned into three groups randomly: sham, CCI, and HBO groups. The CCI rat model was established, and HBO treatment at 2.5 ATA (60 min) was given one day after surgery, lasting for five consecutive days. The pain behaviors of the rats were observed at predetermined time points, and the activation of astrocytes at dorsal horns as well as the changes of the synaptic ultrastructures were observed. The expressions of inducible nitric oxide synthase and neuronal nitric oxide synthase were detected by Western blot, and the levels of tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1ß) were detected by quantitative real-time PCR. Results Rats in the CCI group developed hyperalgesia when compared with the sham group. Mechanical withdrawal threshold decreased and thermal withdrawal latency shortened in CCI group. Also, astrocytes at the dorsal horn were activated, the synaptic structure was disordered, the expressions of inducible nitric oxide synthase and neuronal nitric oxide synthase were increased significantly, and the release of inflammatory factor (TNF-α and IL-1ß) was up-regulated. However, with early initiation of HBO treatment, rats in the HBO group showed significantly alleviated hyperalgesia, increased mechanical withdrawal threshold, and prolonged thermal withdrawal latency. HBO treatment inhibited astrocyte expression and maintained normal synaptic structure. The expressions of inducible nitric oxide synthase and neuronal nitric oxide synthase were decreased in the dorsal horn, and the release of inflammatory factor (TNF-α and IL-1ß) was reduced. Conclusions Early HBO treatment significantly improves hyperalgesia in rats with neuropathic pain. The decreased expressions of inducible nitric oxide synthase and neuronal nitric oxide synthase and reduced levels of inflammatory factors are important mechanisms by which early HBO helps to alleviate neuropathic pain.


Asunto(s)
Analgésicos/farmacología , Oxigenoterapia Hiperbárica , Mediadores de Inflamación/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Animales , Conducta Animal , Enfermedad Crónica , Constricción , Interleucina-1beta/metabolismo , Masculino , Neuralgia/terapia , Ratas Sprague-Dawley , Asta Dorsal de la Médula Espinal/metabolismo , Asta Dorsal de la Médula Espinal/patología , Sinapsis/metabolismo , Sinapsis/ultraestructura , Factor de Necrosis Tumoral alfa/metabolismo
6.
Nat Neurosci ; 21(3): 353-363, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29459763

RESUMEN

CA1 pyramidal neurons are a major output of the hippocampus and encode features of experience that constitute episodic memories. Feature-selective firing of these neurons results from the dendritic integration of inputs from multiple brain regions. While it is known that synchronous activation of spatially clustered inputs can contribute to firing through the generation of dendritic spikes, there is no established mechanism for spatiotemporal synaptic clustering. Here we show that single presynaptic axons form multiple, spatially clustered inputs onto the distal, but not proximal, dendrites of CA1 pyramidal neurons. These compound connections exhibit ultrastructural features indicative of strong synapses and occur much more commonly in entorhinal than in thalamic afferents. Computational simulations revealed that compound connections depolarize dendrites in a biophysically efficient manner, owing to their inherent spatiotemporal clustering. Our results suggest that distinct afferent projections use different connectivity motifs that differentially contribute to dendritic integration.


Asunto(s)
Axones/fisiología , Región CA1 Hipocampal/fisiología , Dendritas/fisiología , Células Piramidales/patología , Sinapsis/fisiología , Animales , Axones/ultraestructura , Región CA1 Hipocampal/ultraestructura , Simulación por Computador , Dendritas/ultraestructura , Potenciales Postsinápticos Excitadores , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas Aferentes/fisiología , Neuronas Aferentes/ultraestructura , Terminales Presinápticos/fisiología , Terminales Presinápticos/ultraestructura , Células Piramidales/ultraestructura , Sinapsis/ultraestructura , Tálamo/citología , Tálamo/fisiología
7.
Brain Struct Funct ; 223(5): 2143-2156, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29380121

RESUMEN

Kisspeptin (KP) synthesizing neurons of the hypothalamic infundibular region are critically involved in the central regulation of fertility; these cells regulate pulsatile gonadotropin-releasing hormone (GnRH) secretion and mediate sex steroid feedback signals to GnRH neurons. Fine structural analysis of the human KP system is complicated by the use of post mortem tissues. To gain better insight into the neuroanatomy of the somato-dendritic cellular compartment, we introduced the diolistic labeling of immunohistochemically identified KP neurons using a gene gun loaded with the lipophilic dye, DiI. Confocal microscopic studies of primary dendrites in 100-µm-thick tissue sections established that 79.3% of KP cells were bipolar, 14.1% were tripolar, and 6.6% were unipolar. Primary dendrites branched sparsely, contained numerous appendages (9.1 ± 1.1 spines/100 µm dendrite), and received rich innervation from GABAergic, glutamatergic, and KP-containing terminals. KP neuron synaptology was analyzed with immunoelectron microscopy on perfusion-fixed specimens. KP axons established frequent contacts and classical synapses on unlabeled, and on KP-immunoreactive somata, dendrites, and spines. Synapses were asymmetric and the presynaptic structures contained round and regular synaptic vesicles, in addition to dense-core granules. Although immunofluorescent studies failed to detect vesicular glutamate transporter isoforms in KP axons, ultrastructural characteristics of synaptic terminals suggested use of glutamatergic, in addition to peptidergic, neurotransmission. In summary, immunofluorescent and DiI labeling of KP neurons in thick hypothalamic sections and immunoelectron microscopic studies of KP-immunoreactive neurons in brains perfusion-fixed shortly post mortem allowed us to identify previously unexplored fine structural features of KP neurons in the mediobasal hypothalamus of humans.


Asunto(s)
Hipotálamo/citología , Kisspeptinas/metabolismo , Neuronas/citología , Neuronas/metabolismo , Anciano , Anciano de 80 o más Años , Autopsia , Axones/metabolismo , Axones/ultraestructura , Carbocianinas/metabolismo , Cuerpo Celular/ultraestructura , Dendritas/metabolismo , Dendritas/ultraestructura , Ácido Glutámico/metabolismo , Humanos , Imagenología Tridimensional , Kisspeptinas/ultraestructura , Lisina/análogos & derivados , Lisina/metabolismo , Masculino , Microscopía Confocal , Microscopía Inmunoelectrónica , Persona de Mediana Edad , Red Nerviosa/metabolismo , Red Nerviosa/ultraestructura , Sinapsis/metabolismo , Sinapsis/ultraestructura , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/ultraestructura , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/ultraestructura , Ácido gamma-Aminobutírico/metabolismo
8.
Elife ; 72018 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-29328020

RESUMEN

We studied the role of the synaptic ribbon for sound encoding at the synapses between inner hair cells (IHCs) and spiral ganglion neurons (SGNs) in mice lacking RIBEYE (RBEKO/KO). Electron and immunofluorescence microscopy revealed a lack of synaptic ribbons and an assembly of several small active zones (AZs) at each synaptic contact. Spontaneous and sound-evoked firing rates of SGNs and their compound action potential were reduced, indicating impaired transmission at ribbonless IHC-SGN synapses. The temporal precision of sound encoding was impaired and the recovery of SGN-firing from adaptation indicated slowed synaptic vesicle (SV) replenishment. Activation of Ca2+-channels was shifted to more depolarized potentials and exocytosis was reduced for weak depolarizations. Presynaptic Ca2+-signals showed a broader spread, compatible with the altered Ca2+-channel clustering observed by super-resolution immunofluorescence microscopy. We postulate that RIBEYE disruption is partially compensated by multi-AZ organization. The remaining synaptic deficit indicates ribbon function in SV-replenishment and Ca2+-channel regulation.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Células Ciliadas Auditivas Internas/fisiología , Audición , Fosfoproteínas/deficiencia , Ganglio Espiral de la Cóclea/citología , Sinapsis/fisiología , Estimulación Acústica , Oxidorreductasas de Alcohol , Animales , Proteínas Co-Represoras , Ratones , Ratones Noqueados , Microscopía Electrónica , Microscopía Fluorescente , Sinapsis/ultraestructura
9.
J Acoust Soc Am ; 142(5): EL434, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29195459

RESUMEN

Noise exposure and aging can damage cochlear synapses required for suprathreshold listening, even when cochlear structures needed for hearing at threshold remain unaffected. To control for effects of aging, behavioral amplitude modulation (AM) detection and subcortical envelope following responses (EFRs) to AM tones in 25 age-restricted (18-19 years) participants with normal thresholds, but different self-reported noise exposure histories were studied. Participants with more noise exposure had smaller EFRs and tended to have poorer AM detection than less-exposed individuals. Simulations of the EFR using a well-established cochlear model were consistent with more synaptopathy in participants reporting greater noise exposure.


Asunto(s)
Corteza Auditiva/fisiopatología , Percepción Auditiva , Cóclea/fisiopatología , Potenciales Evocados Auditivos , Pérdida Auditiva Provocada por Ruido/etiología , Pérdida Auditiva Sensorineural/etiología , Audición , Ruido/efectos adversos , Sinapsis/ultraestructura , Estimulación Acústica , Adolescente , Factores de Edad , Audiometría de Tonos Puros , Umbral Auditivo , Simulación por Computador , Electroencefalografía , Femenino , Pérdida Auditiva Provocada por Ruido/diagnóstico , Pérdida Auditiva Provocada por Ruido/fisiopatología , Pérdida Auditiva Provocada por Ruido/psicología , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/fisiopatología , Pérdida Auditiva Sensorineural/psicología , Humanos , Masculino , Modelos Neurológicos , Psicoacústica , Factores de Riesgo , Adulto Joven
10.
J Alzheimers Dis ; 59(2): 683-694, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28671132

RESUMEN

Alzheimer's disease (AD) is the most common cause of progressive cognitive impairment in the aged. The aggregation of the amyloid ß-protein (Aß) is a hallmark of AD and is linked to synapse loss and cognitive impairment. Capsaicin, a specific agonist of the transient receptor potential vanilloid 1 (TRPV1), has been proven to ameliorate stress-induced AD-like pathological and cognitive impairments, but it is unclear whether TRPV1 activation can affect cognitive and synaptic functions in Aß-induced mouse model of AD. In this study, we investigated the effects of TRPV1 activation on spatial memory and synaptic plasticity in mice treated with Aß. To induce AD-like pathological and cognitive impairments, adult C57Bl/6 mice were microinjected with Aß42 (100 µM, 2.5 µl/mouse, i.c.v.). Two weeks after Aß42 microinjection, spatial learning and memory as well as hippocampal long-term potentiation (LTP) were examined. The results showed that Aß42 microinjection significantly impaired spatial learning and memory in the Morris water maze and novel object recognition tests compared with controls. These behavioral changes were accompanied by synapse loss and impaired LTP in the CA1 area of hippocampus. More importantly, daily capsaicin (1 mg/kg, i.p.) treatment throughout the experiment dramatically improved spatial learning and memory and synaptic function, as reflected by enhanced hippocampal LTP and reduced synapse loss, whereas the TRPV1 antagonist capsazepine (1 mg/kg, i.p.) treatment had no effects on cognitive and synaptic function in Aß42-treated mice. These results indicate that TRPV1 activation by capsaicin rescues cognitive deficit in the Aß42-induced mouse model of AD both structurely and functionally.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Encéfalo/patología , Capsaicina/uso terapéutico , Disfunción Cognitiva , Fragmentos de Péptidos/toxicidad , Fármacos del Sistema Sensorial/uso terapéutico , Sinapsis/efectos de los fármacos , Análisis de Varianza , Animales , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/patología , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large/metabolismo , Estimulación Eléctrica , Conducta Exploratoria/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Reconocimiento en Psicología/efectos de los fármacos , Sinapsis/patología , Sinapsis/ultraestructura , Sinaptofisina/metabolismo , Factores de Tiempo
11.
Phytomedicine ; 25: 15-24, 2017 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-28190466

RESUMEN

BACKGROUND: X. sorbifolia is a widely cultivated ecologicalcrop in the north of China which is used to produce biodiesel fuel. It also possesses special medicinal value and has attracted keen interests of researchers to explore its bioactivity. PURPOSE: To extract the total triterpenoid saponins from the husk of X. sorbifolia (TSX) and investigate its effects on Alzheimer's disease (AD). STUDY DESIGN: TSX was prepared via modern extraction techniques. Its effects on two AD animal models, as well as the preliminary mechanism were investigated comprehensively. METHODS: The behavioral experiments including Y maze test, Morris water maze test and passive avoidance test were performed to observe the learning and memory abilities of the animals. ELISA assays, transmission electron microscope observation and Western blotting were employed in mechanism study. RESULTS: TSX, the main composition of X. sorbifolia, accounted for 88.77% in the plant material. It could significantly increase the spontaneous alternation in Y maze test (F (6, 65)=3.209, P<0.01), prolong the swimming time in the fourth quadrant in probe test of Morris water maze test (F (6, 71)=4.019, P<0.01), and increase the escape latency in passive avoidance test (F (6, 65)=3.684, P<0.01) in AD model animals. The preliminary mechanism research revealed that TSX could significantly increase the contents of hippocampal Ach and ChAT, and enhance activity of ChAT in hippocampus of quinolinic acid injected rats (F (5, 61)=3.915, P 0.01; F (5, 61)=3.623, P<0.01, F (5, 61)=4.344, P<0.01, respectively). It could also increase the activities of T-AOC and T-SOD, and decrease the content of MDA in hippocampus of Aß1-42 injected mice (F (5, 30)=5.193, P<0.01, F (5, 30)=2.865, P<0.05, F (5, 30)=4.735, P<0.01, respectively). Moreover, it significantly increased the expressions of SYP, PSD-95 and GAP-43 in hippocampus (F (4, 27)=3.495, P<0.05; F (4, 27)=2.965, P<0.05; F (4, 27)=4.365, P<0.01, respectively), and improved the synaptic ultra-structure damage in model rats. CONCLUSION: TSX could significantly improve the impairments of learning and memory. The preliminary mechanism might associate with its protection effects against oxidative stress damage, cholinergic system deficiency and synaptic damage. TSX are perfectly suitable for AD patients as medicine or functional food, which would be a new candidate to treat AD.


Asunto(s)
Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria , Estrés Oxidativo/efectos de los fármacos , Sapindaceae/química , Saponinas/farmacología , Sinapsis/patología , Triterpenos/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , China , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/etiología , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Ratones , Fitoterapia , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Ratas Sprague-Dawley , Superóxido Dismutasa/metabolismo , Sinapsis/ultraestructura , Triterpenos/uso terapéutico
12.
J Neurosci ; 37(9): 2435-2448, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28137974

RESUMEN

The traditional classification of primary motor cortex (M1) as an agranular area has been challenged recently when a functional layer 4 (L4) was reported in M1. L4 is the principal target for thalamic input in sensory areas, which raises the question of how thalamocortical synapses formed in M1 in the mouse compare with those in neighboring sensory cortex (S1). We identified thalamic boutons by their immunoreactivity for the vesicular glutamate transporter 2 (VGluT2) and performed unbiased disector counts from electron micrographs. We discovered that the thalamus contributed proportionately only half as many synapses to the local circuitry of L4 in M1 compared with S1. Furthermore, thalamic boutons in M1 targeted spiny dendrites exclusively, whereas ∼9% of synapses were formed with dendrites of smooth neurons in S1. VGluT2+ boutons in M1 were smaller and formed fewer synapses per bouton on average (1.3 vs 2.1) than those in S1, but VGluT2+ synapses in M1 were larger than in S1 (median postsynaptic density areas of 0.064 µm2 vs 0.042 µm2). In M1 and S1, thalamic synapses formed only a small fraction (12.1% and 17.2%, respectively) of all of the asymmetric synapses in L4. The functional role of the thalamic input to L4 in M1 has largely been neglected, but our data suggest that, as in S1, the thalamic input is amplified by the recurrent excitatory connections of the L4 circuits. The lack of direct thalamic input to inhibitory neurons in M1 may indicate temporal differences in the inhibitory gating in L4 of M1 versus S1.SIGNIFICANCE STATEMENT Classical interpretations of the function of primary motor cortex (M1) emphasize its lack of the granular layer 4 (L4) typical of sensory cortices. However, we show here that, like sensory cortex (S1), mouse M1 also has the canonical circuit motif of a core thalamic input to the middle cortical layer and that thalamocortical synapses form a small fraction (M1: 12%; S1: 17%) of all asymmetric synapses in L4 of both areas. Amplification of thalamic input by recurrent local circuits is thus likely to be a significant mechanism in both areas. Unlike M1, where thalamocortical boutons typically form a single synapse, thalamocortical boutons in S1 usually formed multiple synapses, which means they can be identified with high probability in the electron microscope without specific labeling.


Asunto(s)
Corteza Motora/ultraestructura , Corteza Somatosensorial/ultraestructura , Tálamo/ultraestructura , Animales , Imagenología Tridimensional , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Modelos Anatómicos , Corteza Motora/metabolismo , Vías Nerviosas/fisiología , Vías Nerviosas/ultraestructura , Fosfopiruvato Hidratasa/metabolismo , Terminales Presinápticos/metabolismo , Terminales Presinápticos/ultraestructura , Corteza Somatosensorial/metabolismo , Sinapsis/metabolismo , Sinapsis/ultraestructura , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestructura , Tálamo/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/ultraestructura
13.
Brain Res ; 1657: 361-367, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28063855

RESUMEN

High dietary intake of plant estrogens (phytoestrogens) can affect brain structure and function. The effects of phytoestrogen intake within the range of normal animal and human dietary consumption, however, remain uncertain. The aim of the present study was to determine the effects of the isoflavonoids present in a standard low phytoestrogen laboratory rat chow on spine synapse density in the stratum radiatum of area CA1 of the hippocampus. Weanling rats (22days old) were fed either standard chow (Teklad 2018), a nutritionally comparable diet without soy (Teklad 2016) or a custom diet containing Teklad 2016 supplemented with the principal soy isoflavonoids, daidzein and genistein, for 40days. Rats were ovariectomized at 54days of age. Eight days later, spine synapse density on the apical dendrites of hippocampal pyramidal neurons in the stratum radiatum of area CA1 was measured by electron microscopic stereological analysis. Animals maintained on Teklad 2016 exhibited an approximately 60% lower CA1 spine synapse density than animals consuming Teklad 2018. Replacing genistein and daidzein in Teklad 2016 returned synapse density to levels indistinguishable from those in animals on Teklad 2018. These results indicate that the isoflavonoids in a standard laboratory rat diet exert significant effects on spine synapse density in the CA1 region of the hippocampus. Since changes in spine synapse density in this region of the hippocampus have been linked to cognitive performance and mood state, these data suggest that even relatively low daily consumption of soy phytoestrogens may be sufficient to influence hippocampal function.


Asunto(s)
Región CA1 Hipocampal/ultraestructura , Espinas Dendríticas/ultraestructura , Dieta , Fitoestrógenos/administración & dosificación , Proteínas de Soja/administración & dosificación , Sinapsis/ultraestructura , Alimentación Animal , Animales , Femenino , Genisteína/administración & dosificación , Isoflavonas/administración & dosificación , Microscopía Electrónica , Ovariectomía , Células Piramidales/ultraestructura , Ratas Sprague-Dawley
14.
Mol Cell Neurosci ; 79: 64-80, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28087334

RESUMEN

Acute total sleep deprivation (SD) impairs memory consolidation, attention, working memory and perception. Structural, electrophysiological and molecular experimental approaches provided evidences for the involvement of sleep in synaptic functions. Despite the wide scientific interest on the effects of sleep on the synapse, there is a lack of systematic investigation of sleep-related changes in the synaptic proteome. We isolated parietal cortical and thalamic synaptosomes of rats after 8h of total SD by gentle handling and 16h after the end of deprivation to investigate the short- and longer-term effects of SD on the synaptic proteome, respectively. The SD efficiency was verified by electrophysiology. Protein abundance alterations of the synaptosomes were analyzed by fluorescent two-dimensional differential gel electrophoresis and by tandem mass spectrometry. As several altered proteins were found to be involved in synaptic strength regulation, our data can support the synaptic homeostasis hypothesis function of sleep and highlight the long-term influence of SD after the recovery sleep period, mostly on cortical synapses. Furthermore, the large-scale and brain area-specific protein network change in the synapses may support both ideas of sleep-related synaptogenesis and molecular maintenance and reorganization in normal rat brain.


Asunto(s)
Corteza Cerebral/metabolismo , Proteoma/metabolismo , Privación de Sueño/metabolismo , Sinapsis/metabolismo , Tálamo/metabolismo , Animales , Corteza Cerebral/ultraestructura , Masculino , Proteoma/genética , Ratas , Ratas Sprague-Dawley , Privación de Sueño/patología , Sinapsis/ultraestructura , Tálamo/ultraestructura
15.
Curr Neuropharmacol ; 15(1): 47-56, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-26997507

RESUMEN

BACKGROUND: Rapastinel (GLYX-13) is a NMDA receptor modulator with glycine-site partial agonist properties. It is a robust cognitive enhancer and shows rapid and long-lasting antidepressant properties in both animal models and in humans. METHODS: Rapastinel was derived from a monoclonal antibody, B6B21, is a tetrapeptide (threonine-proline-proline-threonine-amide) obtained from amino acid sequence information obtained from sequencing one of the hypervariable regions of the light chain of B6B21. The in-vivo and in-vitro pharmacology of rapastinel was examined. RESULTS: Rapastinel was found to be a robust cognitive enhancer in a variety of learning and memory paradigms and shows marked antidepressant-like properties in multiple models including the forced swim (Porsolt), learned helplessness and chronic unpredictable stress. Rapastinel's rapid-acting antidepressant properties appear to be mediated by its ability to activate NMDA receptors leading to enhancement in synaptic plasticity processes associated with learning and memory. This is further substantiated by the increase in mature dendritic spines found 24 hrs after rapastinel treatment in both the rat dentate gyrus and layer five of the medial prefrontal cortex. Moreover, ex vivo LTP studies showed that the effects of rapastinel persisted at least two weeks post-dosing. CONCLUSION: These data suggest that rapastinel has significant effects on metaplasticity processes that may help explain the long lasting antidepressant effects of rapastinel seen in the human clinical trial results.


Asunto(s)
Depresión/tratamiento farmacológico , Oligopéptidos/farmacología , Oligopéptidos/uso terapéutico , Factores de Edad , Animales , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Depresión/patología , Depresión/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Conducta Exploratoria/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Oligopéptidos/química , Ratas , Receptores de N-Metil-D-Aspartato/metabolismo , Natación , Sinapsis/efectos de los fármacos , Sinapsis/ultraestructura , Vocalización Animal/efectos de los fármacos
16.
Brain Struct Funct ; 222(2): 735-748, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27255751

RESUMEN

T-type calcium channels (Cav3) are key mediators of thalamic bursting activity, but also regulate single cells excitability, dendritic integration, synaptic strength and transmitter release. These functions are strongly influenced by the subcellular and subsynaptic localization of Cav3 channels along the somatodendritic domain of thalamic cells. In Parkinson's disease, T-type calcium channels dysfunction in the basal ganglia-receiving thalamic nuclei likely contributes to pathological thalamic bursting activity. In this study, we analyzed the cellular, subcellular, and subsynaptic localization of the Cav3.1 channel in the ventral anterior (VA) and centromedian/parafascicular (CM/Pf) thalamic nuclei, the main thalamic targets of basal ganglia output, in normal and parkinsonian monkeys. All thalamic nuclei displayed strong Cav3.1 neuropil immunoreactivity, although the intensity of immunolabeling in CM/Pf was significantly lower than in VA. Ultrastructurally, 70-80 % of the Cav3.1-immunoreactive structures were dendritic shafts. Using immunogold labeling, Cav3.1 was commonly found perisynaptic to asymmetric and symmetric axo-dendritic synapses, suggesting a role of Cav3.1 in regulating excitatory and inhibitory neurotransmission. Significant labeling was also found at non-synaptic sites along the plasma membrane of thalamic neurons. There was no difference in the overall pattern and intensity of immunostaining between normal and parkinsonian monkeys, suggesting that the increased rebound bursting in the parkinsonian state is not driven by changes in Cav3.1 expression. Thus, T-type calcium channels are located to subserve neuronal bursting, but also regulate glutamatergic and non-glutamatergic transmission along the whole somatodendritic domain of basal ganglia-receiving neurons of the primate thalamus.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Sinapsis/metabolismo , Tálamo/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Núcleos Talámicos Intralaminares/metabolismo , Núcleos Talámicos Intralaminares/ultraestructura , Macaca mulatta , Neuronas/ultraestructura , Trastornos Parkinsonianos/metabolismo , Sinapsis/ultraestructura , Tálamo/ultraestructura , Núcleos Talámicos Ventrales/metabolismo , Núcleos Talámicos Ventrales/ultraestructura
17.
Hear Res ; 343: 14-33, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27473502

RESUMEN

Studies of congenital and early-onset deafness have demonstrated that an absence of peripheral sound-evoked activity in the auditory nerve causes pathological changes in central auditory structures. The aim of this study was to establish whether progressive acquired hearing loss could lead to similar brain changes that would degrade the precision of signal transmission. We used complementary physiologic hearing tests and microscopic techniques to study the combined effect of both magnitude and duration of hearing loss on one of the first auditory synapses in the brain, the endbulb of Held (EB), along with its bushy cell (BC) target in the anteroventral cochlear nucleus. We compared two hearing mouse strains (CBA/Ca and heterozygous shaker-2+/-) against a model of early-onset progressive hearing loss (DBA/2) and a model of congenital deafness (homozygous shaker-2-/-), examining each strain at 1, 3, and 6 months of age. Furthermore, we employed a frequency model of the mouse cochlear nucleus to constrain our analyses to regions most likely to exhibit graded changes in hearing function with time. No significant differences in the gross morphology of EB or BC structure were observed in 1-month-old animals, indicating uninterrupted development. However, in animals with hearing loss, both EBs and BCs exhibited a graded reduction in size that paralleled the hearing loss, with the most severe pathology seen in deaf 6-month-old shaker-2-/- mice. Ultrastructural pathologies associated with hearing loss were less dramatic: minor changes were observed in terminal size but mitochondrial fraction and postsynaptic densities remained relatively stable. These results indicate that acquired progressive hearing loss can have consequences on auditory brain structure, with prolonged loss leading to greater pathologies. Our findings suggest a role for early intervention with assistive devices in order to mitigate long-term pathology and loss of function.


Asunto(s)
Nervio Coclear/ultraestructura , Núcleo Coclear/ultraestructura , Pérdida Auditiva/patología , Audición , Sinapsis/ultraestructura , Estimulación Acústica , Factores de Edad , Animales , Umbral Auditivo , Conducta Animal , Nervio Coclear/fisiopatología , Núcleo Coclear/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Predisposición Genética a la Enfermedad , Audición/genética , Pérdida Auditiva/genética , Pérdida Auditiva/fisiopatología , Pérdida Auditiva/psicología , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Endogámicos DBA , Ratones Noqueados , Microscopía Electrónica de Transmisión , Miosinas/deficiencia , Miosinas/genética , Fenotipo , Índice de Severidad de la Enfermedad , Factores de Tiempo
18.
Hear Res ; 343: 34-49, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27421755

RESUMEN

Auditory efferent neurons reside in the brain and innervate the sensory hair cells of the cochlea to modulate incoming acoustic signals. Two groups of efferents have been described in mouse and this report will focus on the medial olivocochlear (MOC) system. Electrophysiological data suggest the MOC efferents function in selective listening by differentially attenuating auditory nerve fiber activity in quiet and noisy conditions. Because speech understanding in noise is impaired in age-related hearing loss, we asked whether pathologic changes in input to MOC neurons from higher centers could be involved. The present study investigated the anatomical nature of descending projections from the inferior colliculus (IC) to MOCs in 3-month old mice with normal hearing, and in 6-month old mice with normal hearing (CBA/CaH), early onset progressive hearing loss (DBA/2), and congenital deafness (homozygous Shaker-2). Anterograde tracers were injected into the IC and retrograde tracers into the cochlea. Electron microscopic analysis of double-labelled tissue confirmed direct synaptic contact from the IC onto MOCs in all cohorts. These labelled terminals are indicative of excitatory neurotransmission because they contain round synaptic vesicles, exhibit asymmetric membrane specializations, and are co-labelled with antibodies against VGlut2, a glutamate transporter. 3D reconstructions of the terminal fields indicate that in normal hearing mice, descending projections from the IC are arranged tonotopically with low frequencies projecting laterally and progressively higher frequencies projecting more medially. Along the mediolateral axis, the projections of DBA/2 mice with acquired high frequency hearing loss were shifted medially towards expected higher frequency projecting regions. Shaker-2 mice with congenital deafness had a much broader spatial projection, revealing abnormalities in the topography of connections. These data suggest that loss in precision of IC directed MOC activation could contribute to impaired signal detection in noise.


Asunto(s)
Cóclea/inervación , Sordera/fisiopatología , Audición , Colículos Inferiores/fisiopatología , Núcleo Olivar/fisiopatología , Estimulación Acústica , Animales , Vías Auditivas/fisiopatología , Percepción Auditiva , Conducta Animal , Sordera/metabolismo , Sordera/patología , Sordera/psicología , Modelos Animales de Enfermedad , Potenciales Evocados Auditivos del Tronco Encefálico , Predisposición Genética a la Enfermedad , Audición/genética , Colículos Inferiores/metabolismo , Colículos Inferiores/ultraestructura , Ratones Endogámicos CBA , Ratones Endogámicos DBA , Ratones Noqueados , Microscopía Electrónica de Transmisión , Miosinas/deficiencia , Miosinas/genética , Técnicas de Trazados de Vías Neuroanatómicas , Núcleo Olivar/metabolismo , Núcleo Olivar/ultraestructura , Fenotipo , Detección de Señal Psicológica , Sinapsis/ultraestructura , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo
19.
Cereb Cortex ; 27(5): 2820-2830, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27166173

RESUMEN

Deafening elicits a deterioration of learned vocalization, in both humans and songbirds. In songbirds, learned vocal plasticity has been shown to depend on the basal ganglia-cortical circuit, but the underlying cellular basis remains to be clarified. Using confocal imaging and electron microscopy, we examined the effect of deafening on dendritic spines in avian vocal motor cortex, the robust nucleus of the arcopallium (RA), and investigated the role of the basal ganglia circuit in motor cortex plasticity. We found rapid structural changes to RA dendritic spines in response to hearing loss, accompanied by learned song degradation. In particular, the morphological characters of RA spine synaptic contacts between 2 major pathways were altered differently. However, experimental disruption of the basal ganglia circuit, through lesions in song-specialized basal ganglia nucleus Area X, largely prevented both the observed changes to RA dendritic spines and the song deterioration after hearing loss. Our results provide cellular evidence to highlight a key role of the basal ganglia circuit in the motor cortical plasticity that underlies learned vocal plasticity.


Asunto(s)
Vías Auditivas/fisiopatología , Ganglios Basales/fisiología , Sordera/patología , Espinas Dendríticas/fisiología , Corteza Motora/patología , Vocalización Animal , Análisis de Varianza , Animales , Biotina/análogos & derivados , Sordera/etiología , Espinas Dendríticas/ultraestructura , Dextranos , Modelos Animales de Enfermedad , Electrólisis/efectos adversos , Pinzones , Centro Vocal Superior/fisiopatología , Masculino , Microscopía Confocal , Microscopía Electrónica de Transmisión , Corteza Motora/ultraestructura , Sinapsis/patología , Sinapsis/ultraestructura
20.
Mol Neurobiol ; 54(4): 3102-3116, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27039309

RESUMEN

Realgar, a type of mineral drug-containing arsenic, exhibits neurotoxicity. Brain glutathione (GSH) is crucial to protect the nervous system and to resist arsenic toxicity. Therefore, the main aim of this study was to explore the neurotoxic mechanisms of realgar and the protective effects of glycyrrhetinic acid (GA) by observing the effects of GA on the hippocampal GSH biosynthetic pathway after exposure to realgar. Institute of Cancer Research (ICR) mice were randomly divided into five groups: a control group, a GA control group, a realgar alone group, a low-dose GA intervention group, and a high-dose GA intervention group. Cognitive ability was tested using an object recognition task (ORT). The ultrastructures of the hippocampal neurons and synapses were observed. mRNA and protein levels of EAAT1, EAAT2, EAAT3, xCT, Nrf2, HO-1, γ-GCS (GCLC, GCLM), and MRP-1 were measured, as was the cellular localization of EAAT3, xCT, MRP-1, and Nrf2. The levels of GSH in the hippocampus, the levels of glutamate (Glu) and cysteine (Cys) in the extracellular fluid of hippocampal CA1 region, and the levels of active sulfur in the brain were also investigated. The results indicate that realgar lowered hippocampal GSH levels, resulting in ultrastructural changes in hippocampal neurons and synapses and deficiencies in cognitive ability, ultimately inducing neurotoxicity. GA could trigger the expression of Nrf2, HO-1, EAAT1, EAAT2, EAAT3, xCT, MRP-1, GCLC, and GCLM. Additionally, the expression of γ-GT and the supply levels of Glu and Cys increased, ultimately causing a significant increase in hippocampal GSH to alleviate realgar-induced neurotoxicity. In conclusion, the findings from our study indicate that GA can antagonize decreased brain GSH levels induced by realgar and can lessen the neurotoxicity of realgar.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG/metabolismo , Sistema de Transporte de Aminoácidos y+/metabolismo , Arsenicales/farmacología , Glutatión/metabolismo , Ácido Glicirretínico/farmacología , Hipocampo/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Sulfuros/farmacología , Animales , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/metabolismo , Líquido Extracelular/efectos de los fármacos , Líquido Extracelular/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/ultraestructura , Masculino , Ratones , Ratones Endogámicos ICR , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/ultraestructura , ARN Mensajero/genética , ARN Mensajero/metabolismo , Azufre/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Sinapsis/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA