Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Intern Med ; 284(6): 568-580, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30027599

RESUMEN

The mammalian hypothalamus contains an astounding heterogeneity of neurons to achieve its role in coordinating central responses to virtually any environmental stressor over the life-span of an individual. Therefore, while core features of intrahypothalamic neuronal modalities and wiring patterns are stable during vertebrate evolution, integration of the hypothalamus into hierarchical brain-wide networks evolved to coordinate its output with emotionality, cognition and conscious decision-making. The advent of single-cell technologies represents a recent milestone in the study of hypothalamic organization by allowing the dissection of cellular heterogeneity and establishing causality between opto- and chemogenetic activity modulation of molecularly-resolved neuronal contingents and specific behaviours. Thus, organizational rules to accumulate an unprecedented variety of hierarchical neuroendocrine command networks into a minimal brain volume are being unravelled. Here, we review recent understanding at nanoscale resolution on how neuronal heterogeneity in the mammalian hypothalamus underpins the diversification of hormonal and synaptic output and keeps those sufficiently labile for continuous adaptation to meet environmental demands. Particular emphasis is directed towards the dissection of neuronal circuitry for aggression and food intake. Mechanistic data encompass cell identities, synaptic connectivity within and outside the hypothalamus to link vegetative and conscious levels of innate behaviours, and context- and circadian rhythm-dependent rules of synaptic neurophysiology to distinguish hypothalamic foci that either tune the body's metabolic set-point or specify behaviours. Consequently, novel insights emerge to explain the evolutionary advantages of non-laminar organization for neuroendocrine circuits coincidently using fast neurotransmitters and neuropeptides. These are then accrued into novel therapeutic principles that meet therapeutic criteria for human metabolic diseases.


Asunto(s)
Hipotálamo/ultraestructura , Animales , Humanos , Hipotálamo/anatomía & histología , Hipotálamo/citología , Hipotálamo/fisiología , Neuronas/fisiología , Neuronas/ultraestructura , Análisis de la Célula Individual
2.
Oncogene ; 36(6): 777-786, 2017 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-27375015

RESUMEN

Although genome alterations driving glioma by fueling cell malignancy have largely been resolved, less is known of the impact of tumor environment on disease progression. Here, we demonstrate functional GABAA receptor-activated currents in human glioblastoma cells and show the existence of a continuous GABA signaling within the tumor cell mass that significantly affects tumor growth and survival expectancy in mouse models. Endogenous GABA released by tumor cells, attenuates proliferation of the glioma cells with enriched expression of stem/progenitor markers and with competence to seed growth of new tumors. Our results suggest that GABA levels rapidly increase in tumors impeding further growth. Thus, shunting chloride ions by a maintained local GABAA receptor activity within glioma cells has a significant impact on tumor development by attenuating proliferation, reducing tumor growth and prolonging survival, a mechanism that may have important impact on therapy resistance and recurrence following tumor resection.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Receptores de GABA-A/metabolismo , Animales , Neoplasias Encefálicas/patología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Glioma/patología , Humanos , Ratones , Transducción de Señal , Células Tumorales Cultivadas
3.
Int J Dev Neurosci ; 24(2-3): 195-201, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16386399

RESUMEN

For the delivery of drugs into the brain, the use of nanoparticles as carriers has been described as a promising approach. Here, we prepared nanoparticles as carriers for the model drugs thioflavin T and thioflavin S that bind fibrillar amyloid beta peptides (Abeta). These polymer colloids are composed of a polystyrene core and a degradable PBCA [poly(butyl-2-cyanoacrylate)] shell with a diameter of 90-100nm as shown by dynamic light scattering. Fluorescence spectrophotometric analysis revealed that encapsulated thioflavin T exhibited significantly stronger fluorescence than the free fluorophore. The enzymatic degradation of core-shell nanoparticles, as required in vivo, was shown after their treatment with porcine liver esterase, a non-specific esterase, in vitro. Shells of nanoparticles were dose-dependently degraded while their polystyrene cores remained intact. In the cortices of 7-14 months old APP/PS1 mice with age-dependent beta-amyloidosis, thioflavins selectively targeted fibrillar Abeta after biodegradation-induced release from their nanoparticulate carriers upon intracerebral injection. Collectively, our data suggest that core-shell nanoparticles with controlled degradation in vivo can become versatile tools to trace and clear Abeta in the brain.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Amiloide/metabolismo , Sistemas de Liberación de Medicamentos , Hipocampo/efectos de los fármacos , Ovillos Neurofibrilares/efectos de los fármacos , Tiazoles/administración & dosificación , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Benzotiazoles , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Hipocampo/metabolismo , Ratones , Ratones Transgénicos , Nanoestructuras , Ovillos Neurofibrilares/metabolismo , Unión Proteica/efectos de los fármacos , Factores de Tiempo
5.
Neuroscience ; 135(2): 595-609, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16129564

RESUMEN

Endocannabinoid signaling, mediated by presynaptic CB1 cannabinoid receptors on neurons, is fundamental for the maintenance of synaptic plasticity by modulating neurotransmitter release from axon terminals. In the rodent basal forebrain, CB1 cannabinoid receptor-like immunoreactivity is only harbored by a subpopulation of cholinergic projection neurons. However, endocannabinoid control of cholinergic output from the substantia innominata, coincident target innervation of cholinergic and CB1 cannabinoid receptor-containing afferents, and cholinergic regulation of endocannabinoid synthesis in the hippocampus suggest a significant cholinergic-endocannabinergic interplay. Given the functional importance of the cholinergic modulation of endocannabinoid signaling, here we studied CB1 cannabinoid receptor distribution in cholinergic basal forebrain territories and their cortical projection areas in a prosimian primate, the gray mouse lemur. Perisomatic CB1 cannabinoid receptor immunoreactivity was unequivocally present in non-cholinergic neurons of the olfactory tubercule, and in cholecystokinin-containing interneurons in layers 2/3 of the neocortex. Significantly, CB1 cannabinoid receptor-like immunoreactivity was localized to cholinergic perikarya in the magnocellular basal nucleus. However, cortical cholinergic terminals lacked detectable CB1 cannabinoid receptor levels. A dichotomy of CB1 cannabinoid receptor distribution in frontal (suprasylvian) and parietotemporal (subsylvian) cortices was apparent. In the frontal cortex, CB1 cannabinoid receptor-containing axons concentrated in layers 2/3 and layer 6, while layer 4 and layer 5 were essentially devoid of CB1 cannabinoid receptor immunoreactivity. In contrast, CB1 cannabinoid receptors decorated axons in all layers of the parietotemporal cortex with peak densities in layer 2 and layer 4. In the hippocampus, CB1 cannabinoid receptor-containing terminals concentrated around pyramidal cell somata and proximal dendrites in the CA1-CA3 areas, and granule cell dendrites in the molecular layer of the dentate gyrus. CB1 cannabinoid receptors frequently localized to inhibitory GABAergic terminals while leaving glutamatergic boutons unlabeled. Aging did not affect either the density or layer-specific distribution of CB1 cannabinoid receptor-immunoreactive processes. We concluded that organizing principles of CB1 cannabinoid receptor-containing neurons and their terminal fields within the basal forebrain are evolutionarily conserved between rodents and prosimian primates. In contrast, the areal expansion and cytoarchitectonic differentiation of neocortical subfields in primates is associated with differential cortical patterning of CB1 cannabinoid receptor-containing subcortical and intracortical afferents.


Asunto(s)
Acetilcolina/metabolismo , Evolución Biológica , Vías Nerviosas/citología , Neuronas/fisiología , Prosencéfalo/citología , Receptor Cannabinoide CB1/fisiología , Animales , Compuestos Azo/metabolismo , Recuento de Células/métodos , Cheirogaleidae , Colina O-Acetiltransferasa/metabolismo , Técnica del Anticuerpo Fluorescente/métodos , Masculino , Ratones , Microscopía Confocal/métodos , Naftalenos , Proteínas de Neurofilamentos/metabolismo , Parvalbúminas/metabolismo , Prosencéfalo/fisiología , Ratas , Ratas Sprague-Dawley , Serotonina/metabolismo , Sincalida/metabolismo , Proteínas de Transporte Vesicular de Glutamato/metabolismo , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo
6.
Br J Pharmacol ; 172(4): 1074-86, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25296982

RESUMEN

BACKGROUND AND PURPOSE: Both cannabinoid CB1 and adenosine A2A receptors (CB1 receptors and A2A receptors) control synaptic transmission at corticostriatal synapses, with great therapeutic importance for neurological and psychiatric disorders. A postsynaptic CB1 -A2A receptor interaction has already been elucidated, but the presynaptic A2A receptor-mediated control of presynaptic neuromodulation by CB1 receptors remains to be defined. Because the corticostriatal terminals provide the major input to the basal ganglia, understanding the interactive nature of converging neuromodulation on them will provide us with novel powerful tools to understand the physiology of corticostriatal synaptic transmission and interpret changes associated with pathological conditions. EXPERIMENTAL APPROACH: Pharmacological manipulation of CB1 and A2A receptors was carried out in brain nerve terminals isolated from rats and mice, using flow synaptometry, immunoprecipitation, radioligand binding, ATP and glutamate release measurement. Whole-cell patch-clamp recordings were made in horizontal corticostriatal slices. KEY RESULTS: Flow synaptometry showed that A2A receptors were extensively co-localized with CB1 receptor-immunopositive corticostriatal terminals and A2A receptors co-immunoprecipitated CB1 receptors in these purified terminals. A2A receptor activation decreased CB1 receptor radioligand binding and decreased the CB1 receptor-mediated inhibition of high-K(+) -evoked glutamate release in corticostriatal terminals. Accordingly, A2A receptor activation prevented CB1 receptor-mediated paired-pulse facilitation and attenuated the CB1 receptor-mediated inhibition of synaptic transmission in glutamatergic synapses of corticostriatal slices. CONCLUSIONS AND IMPLICATIONS: Activation of presynaptic A2A receptors dampened CB1 receptor-mediated inhibition of corticostriatal terminals. This constitutes a thus far unrecognized mechanism to modulate the potent CB1 receptor-mediated presynaptic inhibition, allowing frequency-dependent enhancement of synaptic efficacy at corticostriatal synapses.


Asunto(s)
Ácido Glutámico/metabolismo , Receptor de Adenosina A2A/metabolismo , Receptor Cannabinoide CB1/metabolismo , Receptores Presinapticos/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/fisiología , Masculino , Ratones Noqueados , Ratas Wistar , Sinapsis/metabolismo , Transmisión Sináptica
7.
J Dent Res ; 94(7): 945-54, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25838461

RESUMEN

In organized tissues, the precise geometry and the overall shape are critical for the specialized functions that the cells carry out. Odontoblasts are major matrix-producing cells of the tooth and have also been suggested to participate in sensory transmission. However, refined morphologic data on these important cells are limited, which hampers the analysis and understanding of their cellular functions. We took advantage of fluorescent color-coding genetic tracing to visualize and reconstruct in 3 dimensions single odontoblasts, pulp cells, and their assemblages. Our results show distinct structural features and compartments of odontoblasts at different stages of maturation, with regard to overall cellular shape, formation of the main process, orientation, and matrix deposition. We demonstrate previously unanticipated contacts between the processes of pulp cells and odontoblasts. All reported data are related to mouse incisor tooth. We also show that odontoblasts express TRPM5 and Piezo2 ion channels. Piezo2 is expressed ubiquitously, while TRPM5 is asymmetrically distributed with distinct localization to regions proximal to and within odontoblast processes.


Asunto(s)
Imagenología Tridimensional/métodos , Odontoblastos/citología , Ameloblastos/citología , Ameloblastos/ultraestructura , Animales , Compartimento Celular , Núcleo Celular/ultraestructura , Forma de la Célula , Extensiones de la Superficie Celular/ultraestructura , Pulpa Dental/citología , Pulpa Dental/ultraestructura , Dentina/ultraestructura , Matriz Extracelular/ultraestructura , Técnica del Anticuerpo Fluorescente , Incisivo/citología , Incisivo/ultraestructura , Canales Iónicos/ultraestructura , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/ultraestructura , Ratones , Ratones Transgénicos , Microscopía Electrónica de Rastreo/métodos , Odontoblastos/ultraestructura , Canales Catiónicos TRPM/ultraestructura
8.
Rev Neurosci ; 11(4): 329-82, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11065280

RESUMEN

One of the characteristic neuropathological hallmarks of Alzheimer's disease (AD) is the extracellular accumulation of beta-amyloid peptides (Abeta) in neuritic plaques. Experimental data indicate that different molecular forms of Abeta affect a wide array of neuronal and glial functions and thereby may lead to neuronal death in the nervous system. Whereas the fatal outcome of Abeta overproduction in transgenic cell lines, and of exogenous Abeta administration in numerous neurotoxicity models, is well established, particular facets of a complex molecular cascade by which Abeta attack neural cells are still elusive. In the present review we summarize recent knowledge on mechanisms of Abeta aggregation, its role in Abeta neurotoxicity, and binding of Abeta peptides to putative neuronal and glial receptors. Additionally, an integrative view on the interactions of Ca2+ -mediated excitotoxicity and free radical-induced oxidative stress in Abeta toxicity is provided. Furthermore, we survey advances of pharmacological investigations attempting to prevent and antagonize Abeta toxicity, or to promote neuronal regeneration following Abeta-induced neurotoxic insults. We distinguish two major classes of therapeutic approaches: conventional pharmacotherapy that employs blockade of known receptors, signal transduction pathways, and re-uptake of neurotransmitters, and direct targeting of neurotoxic Abeta by means of beta-sheet breakers, functional anti-Abeta peptides, and antibodies. Although a clinically relevant neuroprotective strategy is not yet available, sequential combination of drug regimens may provide prospects for effective antagonism of late-life Abeta burden and subsequent development of dementia.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/efectos de los fármacos , Péptidos beta-Amiloides/metabolismo , Neuronas/metabolismo , Nootrópicos/farmacología , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/toxicidad , Apoptosis , Canales de Calcio , Señalización del Calcio , Radicales Libres , Humanos , Neurotoxinas/metabolismo , Estrés Oxidativo , Placa Amiloide/efectos de los fármacos
10.
Neuroscience ; 121(4): 983-90, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14580948

RESUMEN

Focal traumatic injury to the cerebral cortex is associated with early activation of the neuronal isoform of nitric oxide synthase (nNOS), where high concentrations of nitric oxide-derived free radicals elicit extensive DNA damage. Subsequent activation of the nuclear repair enzyme poly(ADP-ribose) polymerase (PARP) causes a severe energy deficit leading to the ultimate demise of affected neurons. Little is known about the temporal relationship of nNOS and PARP activation and the neuroprotective efficacy of their selective blockade in traumatic brain injury. To determine the relationship of nNOS and PARP activation, brain injury was induced by cryogenic lesion to the somatosensory cortex applying a pre-cooled cylinder after trephination for 6 s to the intact dura mater. Pre-treatment with 3-bromo-7-nitroindazole (BrNI; 25 mg/kg, i.p.), and pre- or combined pre- and post-treatment with 3-aminobenzamide (AB; 10 mg/kg (i.c.v.) or 10 mg/kg/h (i.p.)) were used to inhibit nNOS and PARP, respectively. Cold lesion-induced changes in the somatosensory cortex and neuroprotection by BrNI and AB were determined using immunocytochemistry and immunodot-blot for detection of poly(ADP-ribose; PAR), the end-product of PARP activation, and the triphenyltetrazolium-chloride assay to assess lesion volume. PAR immunoreactivity reached its peak 30 min post-lesion and was followed by gradual reduction of PAR immunolabeling. BrNI pre-treatment significantly decreased the lesion-induced PAR concentration in damaged cerebral cortex. Pre-treatment by i.c.v. infusion of AB markedly diminished cortical PAR immunoreactivity and significantly reduced the lesion volume 24 h post-injury. In contrast, i.p. AB treatment remained largely ineffective. In conclusion, our data indicate early activation of PARP after cold lesion that is, at least in part, related to nNOS induction and supports the relevance of nNOS and/or PARP inhibition to therapeutic approaches of traumatic brain injury.


Asunto(s)
Benzamidas/farmacología , Lesiones Encefálicas/tratamiento farmacológico , Degeneración Nerviosa/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Poli(ADP-Ribosa) Polimerasas/metabolismo , Animales , Lesiones Encefálicas/enzimología , Lesiones Encefálicas/fisiopatología , Infarto Cerebral/tratamiento farmacológico , Infarto Cerebral/enzimología , Infarto Cerebral/prevención & control , Frío/efectos adversos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Interacciones Farmacológicas/fisiología , Radicales Libres/metabolismo , Indazoles/farmacología , Masculino , Degeneración Nerviosa/enzimología , Degeneración Nerviosa/fisiopatología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo I , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Ratas , Ratas Wistar
11.
Neuroscience ; 108(4): 611-27, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11738498

RESUMEN

Cholinergic neurons of the basal forebrain form one of the neuron populations that are susceptible to excitotoxic injury. Whereas neuropharmacological studies have aimed at rescuing cholinergic neurons from acute excitotoxic attacks, the short-term temporal profile of excitotoxic damage to cholinergic nerve cells remains largely elusive. The effects of N-methyl-D-aspartate (NMDA) infusion on cytochemical markers of cholinergic neurons in rat magnocellular nucleus basalis were therefore determined 4, 24 and 48 h post-lesion. Additionally, the influence of excitotoxic damage on the efficacy of in vivo labelling of cholinergic neurons with carbocyanine 3-192IgG was investigated. Carbocyanine 3-192IgG was unilaterally injected in the lateral ventricle. Twenty-four hours later, NMDA (60 nM/microl) was infused in the right magnocellular nucleus basalis, while control lesions were performed contralaterally. Triple immunofluorescence labelling for carbocyanine 3-192IgG, NMDA receptor 2A and B subunits and choline-acetyltransferase (ChAT) was employed to determine temporal changes in NMDA receptor immunoreactivity on cholinergic neurons. The extent of neuronal degeneration was studied by staining with Fluoro-Jade. Moreover, changes in the numbers of ChAT or p75 low-affinity neurotrophin receptor immunoreactive neurons, and the degree of their co-labelling with carbocyanine 3-192IgG were determined in basal forebrain nuclei. The effects of NMDA-induced lesions on cortical projections of cholinergic nucleus basalis neurons were studied by acetylcholinesterase (AChE) histochemistry. Characteristic signs of cellular damage, as indicated by decreased immunoreactivity for NMDA receptors, ChAT and p75 low-affinity neurotrophin receptors, were already detected at the shortest post-lesion interval investigated. Fluoro-Jade at 4 h post-lesion only labelled the core of the excitotoxic lesion. Longer survival led to enhanced Fluoro-Jade staining, and to the decline of ChAT immunoreactivity reaching a maximum 24 h post-surgery. Significant loss of p75 low-affinity neurotrophin receptor immunoreactivity and of cortical AChE-positive projections only became apparent 48 h post-lesion. Carbocyanine 3-192IgG labelling in the ipsilateral basal forebrain exceeded that of the contralateral hemisphere at all time points investigated and progressively declined in the damaged magnocellular nucleus basalis up to 48 h after NMDA infusion. The present study indicates that excitotoxic lesion-induced alteration of cholinergic neuronal markers is a rapid and gradual process reaching its maximum 24 h post-surgery. Furthermore, in vivo labelling of cholinergic neurons may be applied to indicate neuronal survival under pathological conditions, and enable to follow their degeneration process under a variety of experimental conditions.


Asunto(s)
Núcleo Basal de Meynert/efectos de los fármacos , Núcleo Basal de Meynert/patología , Agonistas de Aminoácidos Excitadores/toxicidad , N-Metilaspartato/toxicidad , Neuronas/química , Acetilcolina/fisiología , Animales , Carbocianinas/farmacocinética , Colina O-Acetiltransferasa/análisis , Colina O-Acetiltransferasa/metabolismo , Fluoresceínas , Colorantes Fluorescentes/farmacocinética , Inmunoglobulina G , Inmunohistoquímica/métodos , Inyecciones Intraventriculares , Masculino , Microscopía Confocal , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/patología , Vías Nerviosas , Neuroglía/metabolismo , Neuronas/enzimología , Neuronas/patología , Neurotoxinas/toxicidad , Compuestos Orgánicos , Ratas , Ratas Wistar , Receptor de Factor de Crecimiento Nervioso/análisis , Receptor de Factor de Crecimiento Nervioso/metabolismo , Receptores de N-Metil-D-Aspartato/análisis , Sensibilidad y Especificidad
12.
Neuroscience ; 108(4): 629-42, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11738499

RESUMEN

Recent evidence indicates that stimulation of postsynaptic 5-HT(1A) receptors abates excitotoxic neuronal death. Here we investigated whether oral post-lesion administration of the 5-HT(1A) receptor agonist (-)-(R)-2-[4-[[(3,4-dihydro-2H-1-benzopyran-2-yl)methyl]amino]butyl]-1,2-benzisothiazol-3(2H)-one 1,1-dioxide monohydrochloride (Repinotan HCl) attenuates N-methyl-D-aspartate (NMDA) excitotoxicity (60 nmol/microl) in the rat magnocellular nucleus basalis. Repinotan HCl (1 mg/kg) was administered from day 1, 2, 3, or 6 post-surgery twice daily for five consecutive days. This delayed drug administration protocol was employed to investigate the initiation period during which 5-HT(1A) receptor agonists may significantly influence ongoing neurodegeneration processes. 8-Hydroxy-2-(di-n-propylamino)tetralin (8-OH-DPAT, 1 mg/kg) served as reference compound. Twenty-four hours after drug delivery a small open-field test, while on day 14 post-surgery a passive avoidance test was performed. Effects of Repinotan HCl treatment on the survival of cholinergic magnocellular nucleus basalis neurons and their cortical projections were determined by quantitative acetylcholinesterase (AChE) and choline-acetyltransferase (ChAT) histochemistry. Moreover, AChE and ChAT activities were biochemically measured both in the cerebral cortex and in the magnocellular nucleus basalis. Repinotan HCl treatment markedly increased spontaneous activities in the small open-field at any time-point investigated. Improved memory performance was only demonstrated when Repinotan HCl was administered from day 1 post-lesion on wards. Repinotan HCl treatment from day 2 and 3 post-lesion on markedly attenuated both histochemical and neurochemical characteristics of NMDA excitotoxicity on cholinergic magnocellular nucleus basalis neurons and on their cortical projections. Whereas the neuroprotective profile of Repinotan HCl was superior to that of 8-OH-DPAT, oral administration of both 5-HT(1A) receptor agonists yielded largely equivalent behavioral recovery after NMDA infusion in the magnocellular nucleus basalis. In conclusion, the present data indicate the potent neuroprotective action of the 5-HT(1A) receptor agonist Repinotan HCl with a peak efficacy of delayed (2-3 day) post-lesion drug treatment in vivo. Post-lesion treatment with 5-HT(1A) receptor agonists may therefore be of significance in the intervention of neuronal damage associated with acute excitotoxic conditions.


Asunto(s)
Apoptosis/efectos de los fármacos , Núcleo Basal de Meynert/patología , Benzopiranos/farmacología , Receptores de Serotonina/metabolismo , Agonistas de Receptores de Serotonina/farmacología , Tiazoles/farmacología , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Acetilcolina/fisiología , Administración Oral , Animales , Núcleo Basal de Meynert/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Colina O-Acetiltransferasa/análisis , Agonistas de Aminoácidos Excitadores/toxicidad , Masculino , Memoria/efectos de los fármacos , N-Metilaspartato/toxicidad , Neuronas/enzimología , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Lóbulo Parietal/patología , Ratas , Ratas Wistar , Receptores de Serotonina 5-HT1
13.
Neuroscience ; 101(1): 101-14, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11068140

RESUMEN

In the present study plastic neural responses to N-methyl-D-aspartate-induced excitotoxic lesions and the neuroprotective effects of the L-type voltage-dependent Ca(2+) channel antagonist nimodipine were investigated in the rat magnocellular nucleus basalis. Assessment of spontaneous behaviour in the elevated plus maze and small open-field paradigms on day 5 and day 14 post-surgery indicated anxiety and persistent hypoactivity of N-methyl-D-aspartate-lesioned rats, as compared with sham-operated controls. Nimodipine administration significantly alleviated the behavioural deficits. Quantitative histochemical analysis of acetylcholinesterase-positive fibre innervation of the somatosensory cortex and determination of the numbers of choline-acetyltransferase-positive proximal fibre branches of cholinergic projection neurons in the magnocellular nucleus basalis demonstrated a severe cholinergic deficit as a consequence of the excitotoxic lesion 14 days post-surgery. Nimodipine pre-treatment significantly attenuated the loss of cortical cholinergic innervation and preserved the functional integrity of cholinergic projection neurons in the magnocellular nucleus basalis. Double-labelling immunocytochemistry demonstrated increased amyloid precursor protein expression in shrinking and presumably apoptotic choline-acetyltransferase-positive neurons, whereas surviving cholinergic nerve cells were devoid of excessive amyloid precursor protein immunoreactivity. Moreover, as a consequence of N-methyl-D-aspartate infusion, rim-like accumulation of amyloid precursor protein-positive astrocytes was visualized in a penumbra-like zone of the excitotoxic injury. Furthermore, abundant sprouting of serotonergic projection fibres invading the damaged magnocellular nucleus basalis subdivision was demonstrated. Pharmacological blockade by the Ca(2+) antagonist nimodipine significantly attenuated both neuronal and glial amyloid precursor protein immunoreactivity and serotonergic fibre sprouting following N-methyl-D-aspartate infusion. The present data characterize plastic endogenous glial and neuronal responses in the magnocellular nucleus basalis model of acute excitotoxic brain damage. The increased amyloid precursor protein expression may indicate effective means of intrinsic neuroprotection, as secreted amyloid precursor protein isoforms are suggested to play a role in neuronal rescue following excitotoxic injury. From a pharmacological point of view, extensive sprouting of serotonergic projections in the damaged magnocellular nucleus basalis may also counteract N-methyl-D-aspartate excitotoxicity via serotonin-induced inhibition of Ca(2+) currents and membrane hyperpolarization. Hence, lesion-induced changes in spontaneous animal behaviour, such as anxiety and novelty-induced hypoactivity, may well be attributed to the considerable re-distribution of serotonergic projections in the basal forebrain. In conclusion, our present data emphasize a role of neuron-glia and neurotransmitter-system interactions in functional recovery after acute excitotoxic brain injury, and the efficacy of L-type Ca(2+) channel blockade by the selective 1,4-dihydropyridine antagonist nimodipine.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Axones/efectos de los fármacos , Núcleo Basal de Meynert/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Nimodipina/farmacología , Serotonina/metabolismo , Animales , Axones/metabolismo , Axones/ultraestructura , Núcleo Basal de Meynert/metabolismo , Canales de Calcio Tipo L/efectos de los fármacos , Canales de Calcio Tipo L/metabolismo , Colina O-Acetiltransferasa/metabolismo , Desnervación/efectos adversos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , N-Metilaspartato/efectos adversos , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/metabolismo , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/fisiología , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/metabolismo , Plasticidad Neuronal/fisiología , Neurotoxinas/efectos adversos , Ratas , Ratas Wistar
14.
Neuroscience ; 110(3): 489-504, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11906788

RESUMEN

Estradiol exerts beneficial effects on neurodegenerative disorders associated with the decline of cognitive performance. The present study was designed to further investigate the effect of 17beta-estradiol on learning and memory, and to evaluate its neuroprotective action on cholinergic cells of the nucleus basalis magnocellularis, a neural substrate of cognitive performance. Female rats were ovariectomized at an age of 6 months. Three weeks later they received injections of either a mid-physiological dose of 17beta-estradiol or vehicle (oil), every other day for 2 weeks. The effect of estradiol on cognitive performance was tested in two associative learning paradigms. In the two-way active shock avoidance task estradiol-replaced animals learned significantly faster, while in the passive shock avoidance test no differences were observed between the experimental groups. Subsequent unilateral infusion of N-methyl-D-aspartate in the nucleus basalis magnocellularis resulted in a significant loss of cholinergic neurons concomitant with the loss of their fibers invading the somatosensory cortex. Estradiol treatment did not affect the total number of choline-acetyltransferase-immunoreactive neurons and their coexpression of the p75 low-affinity neurotrophin receptor either contralateral or ipsilateral to the lesion. In contrast, cholinergic fiber densities in estradiol-treated animals were greater both in the contralateral and ipsilateral somatosensory cortices as was detected by quantitative choline-acetyltransferase and vesicular acetylcholine transporter immunocytochemistry. However, estradiol treatment did not affect the lesion-induced relative percentage loss of cholinergic fibers. A significant decline of synaptophysin immunoreactivity paralleled the cholinergic damage in the somatosensory cortex of oil-treated animals, whereas an almost complete preservation of synaptic density was determined in estradiol-treated rats. Our results indicate that estradiol treatment enhances the cortical cholinergic innervation but has no rescuing effect on cholinergic nerve cells in the basal forebrain against excitotoxic damage. Nevertheless, estradiol may restore or maintain synaptic density in the cerebral cortex following cholinergic fiber loss. This estradiol effect may outweigh the lack of cellular protection on cholinergic cells at the functional level.


Asunto(s)
Núcleo Basal de Meynert/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Fibras Colinérgicas/efectos de los fármacos , Estradiol/farmacología , Proteínas de Transporte de Membrana , Memoria/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Terminales Presinápticos/efectos de los fármacos , Proteínas de Transporte Vesicular , Acetilcolinesterasa/metabolismo , Animales , Reacción de Prevención/efectos de los fármacos , Reacción de Prevención/fisiología , Núcleo Basal de Meynert/metabolismo , Núcleo Basal de Meynert/patología , Proteínas Portadoras/metabolismo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Colina O-Acetiltransferasa/metabolismo , Fibras Colinérgicas/metabolismo , Fibras Colinérgicas/patología , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/fisiopatología , Estradiol/metabolismo , Femenino , Inmunohistoquímica , Memoria/fisiología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/fisiopatología , Neurotoxinas/farmacología , Ovariectomía , Terminales Presinápticos/metabolismo , Terminales Presinápticos/patología , Ratas , Ratas Wistar , Receptor de Factor de Crecimiento Nervioso/metabolismo , Sinaptofisina/metabolismo , Proteínas de Transporte Vesicular de Acetilcolina
15.
J Neuroendocrinol ; 13(9): 749-60, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11578524

RESUMEN

Extensive studies during the past decades provided compelling evidence that glucocorticoids (GCs) have the potential to affect the development, survival and death of neurones. These observations, however, reflect paradoxical features of GCs, as they may be critically involved in both neurodegenerative and neuroprotective processes. Hence, we first address different aspects of the complex role of GCs in neurodegeneration and neuroprotection, such as concentration dependent actions of GCs on neuronal viability, anatomical diversity of GC-mediated mechanisms in the brain and species and strain differences in GC-induced neurodegeneration. Second, the modulatory action of GCs during development and ageing of the central nervous system, as well as the contribution of altered GC balance to the pathogenesis of neurodegenerative disorders is considered. In addition, we survey recent data as to the possible mechanisms underlying the neurodegenerative and neuroprotective actions of GCs. As such, two major aspects will be discerned: (i) GC-dependent offensive events, such as GC-induced inhibition of glucose uptake, increased extracellular glutamate concentration and concomitant elevation of intracellular Ca(2+), decrease in GABAergic signalling and regulation of local GC concentrations by 11 beta-hydroxysteroid dehydrogenases; and (ii) GC-related cellular defence mechanisms, such as decrease in after-hyperpolarization, increased synthesis and release of neurotrophic factors and lipocortin-1, feedback regulation of Ca(2+) currents and induction of antioxidant enzymes. The particular relevance of these mechanisms to the neurodegenerative and neuroprotective effects of GCs in the brain is discussed.


Asunto(s)
Glucocorticoides/fisiología , Neuronas/fisiología , Supervivencia Celular/fisiología , Glucocorticoides/farmacología , Degeneración Nerviosa/fisiopatología , Fármacos Neuroprotectores/farmacología
16.
J Neuroendocrinol ; 9(9): 713-20, 1997 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9355039

RESUMEN

The present study demonstrates the effects of adrenalectomy and subcutaneously administered corticosterone on N-methyl-D-aspartate-induced neurodegeneration in the cholinergic magnocellular basal nucleus of the rat. NMDA was unilaterally injected into the nucleus basalis at different plasma corticosterone concentrations in adrenalectomized rats, in adrenalectomized animals with subcutaneously implanted cholesterol-corticosterone pellets containing 25% or 100% corticosterone, and in sham-adrenalectomized controls. The neurotoxic impact of the NMDA injection in the various experimental groups was assessed by the loss of cholinergic fibers stained with acetylcholinesterase histochemistry in the parietal neocortex. Reactive cortical astrocytes as a result of the treatments were detected by glial fibrillary acidic protein immunohistochemistry. Measurements of the densities of astrocytes and cholinergic fibers at the injected side of the brain were carried out by image analysis. Adrenalectomy significantly potentiated the NMDA-induced neurodegeneration by 50%, while chronic administration of corticosterone significantly attenuated the NMDA-neurotoxicity in a dose-dependent manner. Compared to the ADX group, 25% corticosterone application reduced the NMDA damage by 37%, whereas the 100% corticosterone pellet diminished NMDA neurotoxicity by 75%. Both ADX and ADX + corticosterone implantation enhanced the NMDA-induced GFAP immunoreactivity. The increase of GFAP immunoreactivity was most pronounced in the adrenalectomized rats supplied with the 100% corticosterone pellets. The results demonstrate that corticosterone exerts a potent neuroprotective effect on NMDA-induced neurotoxicity in the magnocellular nucleus basalis. The activated astroglia suggest that astrocytes may contribute to the beneficial effect of corticosterone in the neuroprotective mechanisms against excitotoxic neuronal injury.


Asunto(s)
Adrenalectomía , Fibras Colinérgicas/efectos de los fármacos , Corticosterona/sangre , N-Metilaspartato/toxicidad , Sustancia Innominada/citología , Sustancia Innominada/efectos de los fármacos , Acetilcolinesterasa/análisis , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/enzimología , Muerte Celular/efectos de los fármacos , Corticosterona/farmacología , Proteína Ácida Fibrilar de la Glía/análisis , Inmunohistoquímica , Masculino , Degeneración Nerviosa , Ratas , Ratas Wistar
17.
J Neuroendocrinol ; 12(6): 486-94, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10844576

RESUMEN

The impact of glucocorticoids on beta-amyloid(1-42) (Abeta(1-42)) and NMDA-induced neurodegeneration was investigated in vivo. Abeta(1-42) or NMDA was injected into the cholinergic magnocellular nucleus basalis in adrenalectomized (ADX) rats, ADX rats supplemented with 25%, 100%, 2x100% corticosterone pellets, or sham-ADX controls. Abeta(1-42)- or NMDA-induced damage of cholinergic nucleus basalis neurones was assessed by quantitative acetylcholinesterase histochemistry. Plasma concentrations of corticosterone and cholinergic fibre loss after Abeta(1-42) or NMDA injection showed a clear U-shaped dose-response relationship. ADX and subsequent loss of serum corticosterone potentiated both the Abeta(1-42) and NMDA-induced neurodegeneration. ADX+25% corticosterone resulted in a 10-90 nM plasma corticosterone concentration, which significantly attenuated the Abeta(1-42) and NMDA neurotoxicity. ADX+100% corticosterone (corticosterone concentrations of 110-270 nM) potently decreased both Abeta(1-42)- and NMDA-induced neurotoxic brain damage. In contrast, high corticosterone concentrations of 310-650 nM potentiated Abeta(1-42)- and NMDA-triggered neurodegeneration. In conclusion, chronic low or high corticosterone concentrations increase the vulnerability of cholinergic cells to neurotoxic insult, while slightly elevated corticosterone levels protect against neurotoxic injury. Enhanced neurotoxicity of NMDA in the presence of high concentrations of specific glucocorticoid receptor agonists suggests that the corticosterone effects are mediated by glucocorticoid receptors.


Asunto(s)
Núcleo Basal de Meynert/efectos de los fármacos , Núcleo Basal de Meynert/patología , Corticosterona/administración & dosificación , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/patología , Fármacos Neuroprotectores/administración & dosificación , Péptidos beta-Amiloides/farmacología , Animales , Corticosterona/farmacología , Relación Dosis-Respuesta a Droga , Agonistas de Aminoácidos Excitadores/farmacología , Masculino , N-Metilaspartato/farmacología , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/farmacología , Ratas , Ratas Wistar , Factores de Tiempo
18.
Ann N Y Acad Sci ; 903: 374-86, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10818528

RESUMEN

Alzheimer's disease is the most common type of dementia with a still largely unclear etiopathology. One of the factors that may directly contribute to the development and progression of the disorder is the abundant accumulation of beta-amyloid peptides (A beta) in senile plaques. In the present account we review coherent in vivo experimental evidence that A beta infusion into the rat magnocellular nucleus basalis (MBN) induces abrupt and persistent behavioral dysfunctions, perturbations of sensory information processing, storage, and retrieval. These substantial behavioral changes are due to the loss of cholinergic neurons in the MBN and their ascending projections to the frontoparietal cortex. Both neuroanatomical and neurochemical observations pin-point that infusion of A beta into the rat basal forebrain significantly decreases choline-acetyltransferase and acetylcholinesterase activities and the population of--probably--M2 muscarinic acetylcholine receptors in the cerebral cortex. Neuropharmacological data indicate that A beta toxicity is mediated by an excitotoxic cascade involving blockade of astroglial glutamate uptake, sustained activation of N-methyl-D-aspartate receptors and an overt intracellular Ca2+ influx. These changes are associated with increased nitric oxide synthase activity in cortical target areas that may directly lead to the generation of free radicals. Besides, as microvessels of the neocortex receive direct input from the MBN we assume that the loss of cholinergic innervation and hence that of tonic cholinergic vasoregulation ultimately leads to disturbances of vascular (endothelial) function and nutrient supply that may directly enhance neuronal vulnerability during aging and in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/toxicidad , Núcleo Basal de Meynert/patología , Corteza Cerebral/fisiología , Circulación Cerebrovascular/efectos de los fármacos , Neurotoxinas/toxicidad , Enfermedad de Alzheimer/patología , Animales , Núcleo Basal de Meynert/efectos de los fármacos , Circulación Cerebrovascular/fisiología , Desnervación , Humanos , Ratas
19.
Neuroreport ; 10(8): 1693-8, 1999 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-10501559

RESUMEN

A putative tetrapeptide beta-amyloid (Abeta) antagonist (propionyl-Ile-Ile-Gly-Leu [Pr-IIGL]) based on the [31-34] sequence of Abeta was previously shown to rescue astrocytes from Abeta-induced membrane depolarization and subsequent long-term elevations of the intracellular Ca2+ concentration in vitro. Here we provide in vivo evidence that the Pr-IIGL tetrapeptide effectively attenuates the excitotoxic action of Abeta(1-42) on cholinergic neurons of the rat magnocellular nucleus basalis (MBN). We also demonstrate by means of microdialysis that administration of Pr-IIGL abolished Abeta(1-42)-induced increases in extracellular aspartate and glutamate concentrations in the MBN, which coincide with a significant preservation of cholinergic MBN neurons and their cortical projections. This neuroprotective effect was associated with preserved exploratory behavior in an open-field paradigm, and improved memory retention in a step-through passive avoidance task. Our data presented here indicate for the first time the efficacy of short, modified functional Abeta antagonists in ameliorating Abeta excitotoxicity in vivo.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Núcleo Basal de Meynert/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Oligopéptidos/farmacología , Fragmentos de Péptidos/toxicidad , Acetilcolinesterasa/metabolismo , Péptidos beta-Amiloides/antagonistas & inhibidores , Animales , Ácido Aspártico/metabolismo , Reacción de Prevención/efectos de los fármacos , Núcleo Basal de Meynert/anatomía & histología , Núcleo Basal de Meynert/metabolismo , Conducta Animal/efectos de los fármacos , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , Ácido Glutámico/metabolismo , Histocitoquímica , Masculino , Microdiálisis , Actividad Motora/efectos de los fármacos , Fragmentos de Péptidos/antagonistas & inhibidores , Ratas , Ratas Wistar , Taurina/metabolismo
20.
Neuroreport ; 12(17): 3767-72, 2001 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-11726791

RESUMEN

Borna disease virus (BDV)-induced meningoencephalitis is associated with the dysfunction of the cholinergic system. Temporal development of this cholinergic decline during pre-encephalitic and encephalitic stages of BDV infection remains however elusive. Changes in choline acetyltransferase (ChAT) and acetylcholinesterase (AChE) activities were therefore determined in the cerebral cortex, hippocampus, striatum, amygdala and cholinergic basal forebrain nuclei (ChBFN) of rats infected with BDV. Immunocytochemistry for ChAT and vesicular acetylcholine transporter (VAChT) was employed to identify morphological consequences of BDV infection on cholinergic neurons. Whereas both ChAT and AChE activities changed only slightly under pre-encephalitic conditions, the encephalitic stage was characterized by a significant decrease of ChAT activity in the cerebral cortex, horizontal diagonal band of Broca (hDBB), hippocampus and amygdala concomitant with a marked reduction of AChE activity in the cerebral cortex, hDBB and hippocampus. The striatum and medial septum remained unaffected. ChAT and VAChT immunocytochemistry revealed prominent axonal degeneration in affected cortical and limbic projection areas of ChBFN. In summary, our data indicate progressive deterioration of forebrain cholinergic systems that parallels the progression of BDV encephalitis.


Asunto(s)
Acetilcolina/metabolismo , Enfermedad de Borna/metabolismo , Bornaviridae/patogenicidad , Corteza Cerebral/metabolismo , Fibras Colinérgicas/metabolismo , Encefalitis Viral/metabolismo , Proteínas de Transporte de Membrana , Infecciones por Mononegavirales/metabolismo , Proteínas de Transporte Vesicular , Acetilcolinesterasa/metabolismo , Animales , Enfermedad de Borna/patología , Enfermedad de Borna/fisiopatología , Proteínas Portadoras/metabolismo , Corteza Cerebral/patología , Corteza Cerebral/virología , Colina O-Acetiltransferasa/metabolismo , Fibras Colinérgicas/patología , Fibras Colinérgicas/virología , Progresión de la Enfermedad , Regulación hacia Abajo/inmunología , Encefalitis Viral/patología , Encefalitis Viral/fisiopatología , Inmunohistoquímica , Infecciones por Mononegavirales/patología , Infecciones por Mononegavirales/fisiopatología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Degeneración Nerviosa/virología , Neuronas/metabolismo , Neuronas/patología , Neuronas/virología , Prosencéfalo/metabolismo , Prosencéfalo/patología , Prosencéfalo/virología , Ratas , Ratas Endogámicas Lew , Proteínas de Transporte Vesicular de Acetilcolina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA