Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Cereb Cortex ; 29(4): 1634-1643, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29668872

RESUMEN

Exaggerated synaptic elimination in the prefrontal cortex (PFC) during adolescence has been suggested to contribute to the neuropathological changes of schizophrenia. Recent data indicate that microglia (MG) sculpt synapses during early postnatal development. However, it is not known if MG contribute to the structural maturation of the PFC, which has a protracted postnatal development. We determined if MG are involved in developmentally specific synapse elimination in the PFC, focusing on adolescence. Layer 5 PFC pyramidal cells (PCs) were intracellularly filled with Lucifer Yellow for dendritic spine measurements in postnatal day (P) 24, P30, P35, P39, and P50 rats. In the contralateral PFC we evaluated if MG engulfed presynaptic (glutamatergic) and postsynaptic (dendritic spines) elements. Dendritic spine density increased from P24 to P35, when spine density peaked. There was a significant increase in MG engulfment of spines at P39 relative to earlier ages; this subsided by P50. MG also phagocytosed presynaptic glutamatergic terminals. These data indicate that MG transiently prune synapses of PFC PCs during adolescence, when the symptoms of schizophrenia typically first appear. An increase in MG-mediated synaptic remodeling of PFC PCs may contribute to the structural changes observed in schizophrenia.


Asunto(s)
Espinas Dendríticas/fisiología , Microglía/fisiología , Corteza Prefrontal/crecimiento & desarrollo , Células Piramidales/fisiología , Sinapsis/fisiología , Animales , Femenino , Masculino , Microglía/citología , Corteza Prefrontal/citología , Células Piramidales/citología , Ratas Sprague-Dawley
2.
Biochim Biophys Acta Proteins Proteom ; 1865(7): 967-977, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28254588

RESUMEN

Imaging mass spectrometry (IMS) is a molecular imaging technology that can measure thousands of biomolecules concurrently without prior tagging, making it particularly suitable for exploratory research. However, the data size and dimensionality often makes thorough extraction of relevant information impractical. To help guide and accelerate IMS data analysis, we recently developed a framework that integrates IMS measurements with anatomical atlases, opening up opportunities for anatomy-driven exploration of IMS data. One example is the automated anatomical interpretation of ion images, where empirically measured ion distributions are automatically decomposed into their underlying anatomical structures. While offering significant potential, IMS-atlas integration has thus far been restricted to the Allen Mouse Brain Atlas (AMBA) and mouse brain samples. Here, we expand the applicability of this framework by extending towards new animal species and a new set of anatomical atlases retrieved from the Scalable Brain Atlas (SBA). Furthermore, as many SBA atlases are based on magnetic resonance imaging (MRI) data, a new registration pipeline was developed that enables direct non-rigid IMS-to-MRI registration. These developments are demonstrated on protein-focused FTICR IMS measurements from coronal brain sections of a Parkinson's disease (PD) rat model. The measurements are integrated with an MRI-based rat brain atlas from the SBA. The new rat-focused IMS-atlas integration is used to perform automated anatomical interpretation and to find differential ions between healthy and diseased tissue. IMS-atlas integration can serve as an important accelerator in IMS data exploration, and with these new developments it can now be applied to a wider variety of animal species and modalities. This article is part of a Special Issue entitled: MALDI Imaging, edited by Dr. Corinna Henkel and Prof. Peter Hoffmann.


Asunto(s)
Encéfalo/anatomía & histología , Imagen por Resonancia Magnética/métodos , Espectrometría de Masas/métodos , Animales , Encéfalo/metabolismo , Encéfalo/patología , Procesamiento de Imagen Asistido por Computador/métodos , Imagenología Tridimensional/métodos , Iones/metabolismo , Masculino , Ratones , Enfermedad de Parkinson/patología , Ratas , Ratas Sprague-Dawley
3.
J Neurosci ; 35(49): 16259-71, 2015 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-26658874

RESUMEN

In the ventral tegmental area (VTA), a subpopulation of dopamine neurons express vesicular glutamate transporter 2 and make glutamatergic connections to nucleus accumbens (NAc) and olfactory tubercle (OT) neurons. However, their glutamatergic connections across the forebrain have not been explored systematically. To visualize dopamine neuron forebrain projections and to enable photostimulation of their axons independent of transmitter status, we virally transfected VTA neurons with channelrhodopsin-2 fused to enhanced yellow fluorescent protein (ChR2-EYFP) and used DAT(IREScre) mice to restrict expression to dopamine neurons. ChR2-EYFP-expressing neurons almost invariably stained for tyrosine hydroxylase, identifying them as dopaminergic. Dopamine neuron axons visualized by ChR2-EYFP fluorescence projected most densely to the striatum, moderately to the amygdala and entorhinal cortex (ERC), sparsely to prefrontal and cingulate cortices, and rarely to the hippocampus. Guided by ChR2-EYFP fluorescence, we recorded systematically from putative principal neurons in target areas and determined the incidence and strength of glutamatergic connections by activating all dopamine neuron terminals impinging on recorded neurons with wide-field photostimulation. This revealed strong glutamatergic connections in the NAc, OT, and ERC; moderate strength connections in the central amygdala; and weak connections in the cingulate cortex. No glutamatergic connections were found in the dorsal striatum, hippocampus, basolateral amygdala, or prefrontal cortex. These results indicate that VTA dopamine neurons elicit widespread, but regionally distinct, glutamatergic signals in the forebrain and begin to define the dopamine neuron excitatory functional connectome. SIGNIFICANCE STATEMENT: Dopamine neurons are important for the control of motivated behavior and are involved in the pathophysiology of several major neuropsychiatric disorders. Recent studies have shown that some ventral midbrain dopamine neurons are capable of glutamate cotransmission. With conditional expression of channelrhodopsin in dopamine neurons, we systematically explored dopamine neuron connections in the forebrain and identified regionally specific dopamine neuron excitatory connections. Establishing that only a subset of forebrain regions receive excitatory connections from dopamine neurons will help to determine the function of dopamine neuron glutamate cotransmission, which likely involves transmission of precise temporal signals and enhancement of the dynamic range of dopamine neuron signals.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Ácido Glutámico/metabolismo , Red Nerviosa/fisiología , Vías Nerviosas/fisiología , Prosencéfalo/citología , Animales , Channelrhodopsins , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neurotransmisores/farmacología , Fosfopiruvato Hidratasa/metabolismo , Prosencéfalo/fisiología , Transducción Genética , Tirosina 3-Monooxigenasa/metabolismo , Área Tegmental Ventral/citología
4.
J Biol Chem ; 289(31): 21205-16, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24939843

RESUMEN

The p75 neurotrophin receptor (p75(NTR)) mediates the death of specific populations of neurons during the development of the nervous system or after cellular injury. The receptor has also been implicated as a contributor to neurodegeneration caused by numerous pathological conditions. Because many of these conditions are associated with increases in reactive oxygen species, we investigated whether p75(NTR) has a role in neurodegeneration in response to oxidative stress. Here we demonstrate that p75(NTR) signaling is activated by 4-hydroxynonenal (HNE), a lipid peroxidation product generated naturally during oxidative stress. Exposure of sympathetic neurons to HNE resulted in neurite degeneration and apoptosis. However, these effects were reduced markedly in neurons from p75(NTR-/-) mice. The neurodegenerative effects of HNE were not associated with production of neurotrophins and were unaffected by pretreatment with a receptor-blocking antibody, suggesting that oxidative stress activates p75(NTR) via a ligand-independent mechanism. Previous studies have established that proteolysis of p75(NTR) by the metalloprotease TNFα-converting enzyme and γ-secretase is necessary for p75(NTR)-mediated apoptotic signaling. Exposure of sympathetic neurons to HNE resulted in metalloprotease- and γ-secretase-dependent cleavage of p75(NTR). Pharmacological blockade of p75(NTR) proteolysis protected sympathetic neurons from HNE-induced neurite degeneration and apoptosis, suggesting that cleavage of p75(NTR) is necessary for oxidant-induced neurodegeneration. In vivo, p75(NTR-/-) mice exhibited resistance to axonal degeneration associated with oxidative injury following administration of the neurotoxin 6-hydroxydopamine. Together, these data suggest a novel mechanism linking oxidative stress to ligand-independent cleavage of p75(NTR), resulting in axonal fragmentation and neuronal death.


Asunto(s)
Apoptosis/fisiología , Axones , Estrés Oxidativo , Receptores de Factor de Crecimiento Nervioso/fisiología , Animales , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso , Ratas , Ratas Sprague-Dawley , Receptores de Factores de Crecimiento , Pruebas del Campo Visual
5.
J Neurochem ; 130(1): 109-14, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24611756

RESUMEN

Both dopamine and glutamate are critically involved in cognitive processes such as working memory. Astrocytes, which express dopamine receptors, are essential elements in the termination of glutamatergic signaling: the astrocytic glutamate transporter GLT-1 is responsible for > 90% of cortical glutamate uptake. The effect of dopamine depletion on glutamate transporters in the prefrontal cortex (PFC) remains unknown. In an effort to determine if astrocytes are a locus of cortical dopamine-glutamate interactions, we examined the effects of chronic dopamine denervation on PFC protein and mRNA levels of glutamate transporters. PFC dopamine denervation elicited a marked increase in GLT-1 protein levels, but had no effect on levels of other glutamate transporters; high-affinity glutamate transport was positively correlated with the extent of dopamine depletion. GLT-1 gene expression was not altered. Our data suggest that dopamine depletion may lead to post-translational modifications that result in increased expression and activity of GLT-1 in PFC astrocytes. The glutamate transporter GLT-1 is expressed by astrocytes, which also express dopamine receptors. Regulation of prefrontal cortical (PFC) GLT-1 potentially offers a novel treatment approach to the cognitive deficits of schizophrenia. Partial PFC dopamine deafferentation increased membrane expression of GLT-1 protein and glutamate uptake, but did not alter levels of the other two neocortical glutamate transporters, GLAST and EAAC1.


Asunto(s)
Astrocitos/metabolismo , Dopamina/metabolismo , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Regulación de la Expresión Génica , Corteza Prefrontal/metabolismo , Animales , Desnervación , Masculino , Unión Proteica/genética , Ratas , Ratas Sprague-Dawley
6.
bioRxiv ; 2023 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-37808655

RESUMEN

Psychedelic drugs like lysergic acid diethylamide (LSD) and psilocybin have emerged as potentially transformative therapeutics for many neuropsychiatric diseases, including depression, anxiety, post-traumatic stress disorder, migraine, and cluster headaches. LSD and psilocybin exert their psychedelic effects via activation of the 5-hydroxytryptamine 2A receptor (HTR2A). Here we provide a suite of engineered mice useful for clarifying the role of HTR2A and HTR2A-expressing neurons in psychedelic drug actions. We first generated Htr2a-EGFP-CT-IRES-CreERT2 mice (CT:C-terminus) to independently identify both HTR2A-EGFP-CT receptors and HTR2A-containing cells thereby providing a detailed anatomical map of HTR2A and identifying cell types that express HTR2A. We also generated a humanized Htr2a mouse line and an additional constitutive Htr2A-Cre mouse line. Psychedelics induced a variety of known behavioral changes in our mice validating their utility for behavioral studies. Finally, electrophysiology studies revealed that extracellular 5-HT elicited a HTR2A-mediated robust increase in firing of genetically-identified pyramidal neurons--consistent with a plasma membrane localization and mode of action. These mouse lines represent invaluable tools for elucidating the molecular, cellular, pharmacological, physiological, behavioral, and other actions of psychedelic drugs in vivo.

7.
J Neurochem ; 120(4): 528-40, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22117778

RESUMEN

Cerebellin precursor protein 1 (Cbln1) is the prototype of a family of secreted neuronal glycoproteins (Cbln1-4) and its genetic elimination results in synaptic alterations in cerebellum (CB) and striatum. In CB, Cbln1 acts as a bi-functional ligand bridging pre-synaptic ß-neurexins on granule cells to post-synaptic Grid2 on Purkinje neurons. Although much is known concerning the action of Cbln1, little is known of the function of its other family members. Here, we show that Cbln1 and Cbln2 have similar binding activities to ß-neurexins and Grid2 and the targeted ectopic expression of Cbln2 to Purkinje cells in transgenic mice rescues the cerebellar deficits in Cbln1-null animals: suggesting that the two proteins have redundant function mediated by their common receptor binding properties. Cbln1 and Cbln2 are also co-expressed in the endolysosomal compartment of the thalamic neurons responsible for the synaptic alterations in striatum of Cbln1-null mice. Therefore, to determine whether the two family members have similar functions, we generated Cbln2-null mice. Cbln2-null mice do not show the synaptic alterations evident in striatum of Cbln1-null mice. Thus, Cbln2 can exhibit functional redundancy with Cbln1 in CB but it does not have the same properties as Cbln1 in thalamic neurons, implying one or both utilize different receptors/mechanisms in this brain region.


Asunto(s)
Proteínas del Tejido Nervioso/fisiología , Precursores de Proteínas/fisiología , Animales , Femenino , Células HEK293 , Humanos , Lisosomas/enzimología , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Unión Proteica/genética , Precursores de Proteínas/biosíntesis , Precursores de Proteínas/deficiencia , Precursores de Proteínas/genética , Células de Purkinje/enzimología , Células de Purkinje/metabolismo
8.
J Neurochem ; 121(5): 717-29, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22220752

RESUMEN

Cerebellin precursor protein (Cbln1) is essential for synapse integrity in cerebellum through assembly into complexes that bridge pre-synaptic ß-neurexins (Nrxn) to post-synaptic GluRδ2. However, GluRδ2 is largely cerebellum-specific, yet Cbln1 and its little studied family members, Cbln2 and Cbln4, are expressed throughout brain. Therefore, we investigated whether additional proteins mediate Cbln family actions. Whereas Cbln1 and Cbln2 bound to GluRδ2 and Nrxns1-3, Cbln4 bound weakly or not at all, suggesting it has distinct binding partners. In a candidate receptor-screening assay, Cbln4 (but not Cbln1 or Cbln2) bound selectively to the netrin receptor, (deleted in colorectal cancer (DCC) in a netrin-displaceable fashion. To determine whether Cbln4 had a netrin-like function, Cbln4-null mice were generated. Cbln4-null mice did not phenocopy netrin-null mice. Cbln1 and Cbln4 were likely co-localized in neurons thought to be responsible for synaptic changes in striatum of Cbln1-null mice. Furthermore, complexes containing Cbln1 and Cbln4 had greatly reduced affinity to DCC but increased affinity to Nrxns, suggesting a functional interaction. However, Cbln4-null mice lacked the striatal synaptic changes seen in Cbln null mice. Thus, Cbln family members interact with multiple receptors/signaling pathways in a subunit composition-dependent manner and have independent functions with Cbln4 potentially involved in the less well-characterized role of netrin/DCC in adult brain.


Asunto(s)
Encéfalo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Precursores de Proteínas/metabolismo , Transducción de Señal/fisiología , Animales , Western Blotting , Ratones , Ratones Noqueados , Neuronas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Nat Methods ; 5(1): 101-8, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18165806

RESUMEN

As large genomic and proteomic datasets are generated from homogenates of various tissues, the need for information on the spatial localization of their encoded products has become more pressing. Matrix-assisted laser desorption-ionization (MALDI) imaging mass spectrometry (IMS) offers investigators the means with which to unambiguously study peptides and proteins with molecular specificity, and to determine their distribution in two and three dimensions. In the past few years, several parameters have been optimized for IMS, including sample preparation, matrix application and instrumental acquisition parameters (Box 1). These developments have resulted in a high degree of reproducibility in mass accuracy and peak intensities (Supplementary Fig. 1 online). Recently, we have optimized our protocol to be able to increase the number of molecular species analyzed by collecting two sets of sections, covering one set of sections with sinapinic acid for optimal detection of proteins and adjacent sections with 2,5-dihydroxybenzoic acid (DHB) matrix for the optimal detection of low-mass species, including peptides. Approximately 1,000 peaks can be observed in each dataset (Fig. 1). Furthermore, the sections are collected at an equal distance, 200 mum instead of 400-500 mum used previously, thus enabling the use of virtual z-stacks and three-dimensional (3D) volume renderings to investigate differential localization patterns in much smaller brain structures such as the substantia nigra and the interpeduncular nucleus. Here we present our optimized step-by-step procedure based on previous work in our laboratory, describing how to make 3D volume reconstructions of MALDI IMS data, as applied to the rat brain.


Asunto(s)
Aumento de la Imagen/métodos , Interpretación de Imagen Asistida por Computador/métodos , Imagenología Tridimensional/métodos , Imagen por Resonancia Magnética/métodos , Espectrometría de Masas/métodos , Mapeo Peptídico/métodos , Integración de Sistemas
10.
Cereb Cortex ; 20(10): 2423-32, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20118184

RESUMEN

Striatal medium spiny neurons (MSNs) receive glutamatergic afferents from the cerebral cortex and dopaminergic inputs from the substantia nigra (SN). Striatal dopamine loss decreases the number of MSN dendritic spines. This loss of spines has been suggested to reflect the removal of tonic dopamine inhibitory control over corticostriatal glutamatergic drive, with increased glutamate release culminating in MSN spine loss. We tested this hypothesis in two ways. We first determined in vivo if decortication reverses or prevents dopamine depletion-induced spine loss by placing motor cortex lesions 4 weeks after, or at the time of, 6-hydroxydopamine lesions of the SN. Animals were sacrificed 4 weeks after cortical lesions. Motor cortex lesions significantly reversed the loss of MSN spines elicited by dopamine denervation; a similar effect was observed in the prevention experiment. We then determined if modulating glutamate release in organotypic cocultures prevented spine loss. Treatment of the cultures with the mGluR2/3 agonist LY379268 to suppress corticostriatal glutamate release completely blocked spine loss in dopamine-denervated cultures. These studies provide the first evidence to show that MSN spine loss associated with parkinsonism can be reversed and point to suppression of corticostriatal glutamate release as a means of slowing progression in Parkinson's disease.


Asunto(s)
Corteza Cerebral/fisiología , Cuerpo Estriado/patología , Dendritas/metabolismo , Dopamina/metabolismo , Ácido Glutámico/metabolismo , Neuronas/ultraestructura , Trastornos Parkinsonianos/patología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Análisis de Varianza , Animales , Dendritas/efectos de los fármacos , Modelos Animales de Enfermedad , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Fluoresceínas , Masculino , Neuronas/metabolismo , Técnicas de Cultivo de Órganos , Compuestos Orgánicos , Oxidopamina , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/fisiopatología , Ratas , Ratas Sprague-Dawley , Tinción con Nitrato de Plata/métodos
11.
Pharmacol Rev ; 60(3): 358-403, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18922967

RESUMEN

Various lines of evidence indicate the presence of progressive pathophysiological processes occurring within the brains of patients with schizophrenia. By modulating chemical neurotransmission, antipsychotic drugs may influence a variety of functions regulating neuronal resilience and viability and have the potential for neuroprotection. This article reviews the current literature describing preclinical and clinical studies that evaluate the efficacy of antipsychotic drugs, their mechanism of action and the potential of first- and second-generation antipsychotic drugs to exert effects on cellular processes that may be neuroprotective in schizophrenia. The evidence to date suggests that although all antipsychotic drugs have the ability to reduce psychotic symptoms via D(2) receptor antagonism, some antipsychotics may differ in other pharmacological properties and their capacities to mitigate and possibly reverse cellular processes that may underlie the pathophysiology of schizophrenia.


Asunto(s)
Antipsicóticos/farmacología , Fármacos Neuroprotectores/farmacología , Esquizofrenia/tratamiento farmacológico , Animales , Antipsicóticos/uso terapéutico , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Antagonistas de los Receptores de Dopamina D2 , Humanos , Plasticidad Neuronal/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Esquizofrenia/fisiopatología
12.
Neurobiol Dis ; 39(3): 283-91, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20423730

RESUMEN

Angelman syndrome (AS) is a neurogenetic disorder caused by loss of maternal UBE3A expression or mutation-induced dysfunction of its protein product, the E3 ubiquitin-protein ligase, UBE3A. In humans and rodents, UBE3A/Ube3a transcript is maternally imprinted in several brain regions, but the distribution of native UBE3A/Ube3a(1) protein expression has not been comprehensively examined. To address this, we systematically evaluated Ube3a expression in the brain and peripheral tissues of wild-type (WT) and Ube3a maternal knockout mice (AS mice). Immunoblot and immunohistochemical analyses revealed a marked loss of Ube3a protein in hippocampus, hypothalamus, olfactory bulb, cerebral cortex, striatum, thalamus, midbrain, and cerebellum in AS mice relative to WT littermates. Also, Ube3a expression in heart and liver of AS mice showed greater than the predicted 50% reduction relative to WT mice. Co-localization studies showed Ube3a expression to be primarily neuronal in all brain regions and present in GABAergic interneurons as well as principal neurons. These findings suggest that neuronal function throughout the brain is compromised in AS.


Asunto(s)
Síndrome de Angelman/metabolismo , Encéfalo/metabolismo , Hígado/metabolismo , Miocardio/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Análisis de Varianza , Síndrome de Angelman/genética , Animales , Western Blotting , Modelos Animales de Enfermedad , Inmunohistoquímica , Ratones , Ratones Noqueados , Neuronas/metabolismo , Distribución Tisular , Ubiquitina-Proteína Ligasas/genética , Ácido gamma-Aminobutírico/metabolismo
13.
Cereb Cortex ; 19(10): 2372-9, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19168664

RESUMEN

The claustrum is a prominent but ill-defined forebrain structure that has been suggested to integrate multisensory information and perhaps transform percepts into consciousness. The claustrum's shape and vague borders have hampered experimental assessment of its functions. We used matrix-assisted laser desorption ionization-imaging mass spectrometry to reveal a novel protein marker, G-protein gamma2 subunit (Gng2), which is enriched in the claustrum but not adjacent structures of the rat forebrain. The spatial pattern of Gng2 expression suggests key differences from commonly held views of the claustrum's structure. Using anatomical methods, we found that the rat claustrum is present only at striatal levels of the telencephalon and does not extend to frontal cortical territories. Moreover, the claustrum is surrounded on all sides by layer VI insular cortex cells in both the rat and primate. Using these defining characteristics of the claustrum, we found that the claustrum projects to cortical but not to subcortical sites. The definition of the claustrum as a cortical site is considered. The identification of a claustrum-specific protein opens the door to selective molecular lesions and the subsequent evaluation of the role of the claustrum in cognition.


Asunto(s)
Ganglios Basales/anatomía & histología , Corteza Cerebral/anatomía & histología , Proteínas de Unión al GTP/metabolismo , Animales , Ganglios Basales/metabolismo , Corteza Cerebral/metabolismo , Cromatografía Líquida de Alta Presión , Lóbulo Frontal/metabolismo , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Masculino , Vías Nerviosas/anatomía & histología , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
14.
Nat Neurosci ; 9(2): 251-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16415865

RESUMEN

Parkinson disease is a common neurodegenerative disorder that leads to difficulty in effectively translating thought into action. Although it is known that dopaminergic neurons that innervate the striatum die in Parkinson disease, it is not clear how this loss leads to symptoms. Recent work has implicated striatopallidal medium spiny neurons (MSNs) in this process, but how and precisely why these neurons change is not clear. Using multiphoton imaging, we show that dopamine depletion leads to a rapid and profound loss of spines and glutamatergic synapses on striatopallidal MSNs but not on neighboring striatonigral MSNs. This loss of connectivity is triggered by a new mechanism-dysregulation of intraspine Cav1.3 L-type Ca(2+) channels. The disconnection of striatopallidal neurons from motor command structures is likely to be a key step in the emergence of pathological activity that is responsible for symptoms in Parkinson disease.


Asunto(s)
Cuerpo Estriado/patología , Espinas Dendríticas/patología , Glutamina/metabolismo , Vías Nerviosas/patología , Enfermedad de Parkinson/fisiopatología , Sinapsis/metabolismo , Animales , Canales de Calcio Tipo L/metabolismo , Cuerpo Estriado/fisiopatología , Cuerpo Estriado/ultraestructura , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Microscopía Inmunoelectrónica , Vías Nerviosas/metabolismo , Técnicas de Cultivo de Órganos , Enfermedad de Parkinson/patología , Técnicas de Placa-Clamp , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sinapsis/ultraestructura
15.
J Neurosci ; 28(41): 10422-33, 2008 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-18842902

RESUMEN

Recent studies suggest that subtype-selective activators of M(1)/M(4) muscarinic acetylcholine receptors (mAChRs) may offer a novel approach for the treatment of psychotic symptoms associated with schizophrenia and Alzheimer's disease. Previously developed muscarinic agonists have provided clinical data in support of this hypothesis, but failed in clinical development because of a lack of true subtype specificity and adverse effects associated with activation of other mAChR subtypes. We now report characterization of a novel highly selective agonist for the M(1) receptor with no agonist activity at any of the other mAChR subtypes, termed TBPB [1-(1'-2-methylbenzyl)-1,4'-bipiperidin-4-yl)-1H-benzo[d]imidazol-2(3H)-one]. Mutagenesis and molecular pharmacology studies revealed that TBPB activates M(1) through an allosteric site rather than the orthosteric acetylcholine binding site, which is likely critical for its unprecedented selectivity. Whole-cell patch-clamp recordings demonstrated that activation of M(1) by TBPB potentiates NMDA receptor currents in hippocampal pyramidal cells but does not alter excitatory or inhibitory synaptic transmission, responses thought to be mediated by M(2) and M(4). TBPB was efficacious in models predictive of antipsychotic-like activity in rats at doses that did not produce catalepsy or peripheral adverse effects of other mAChR agonists. Finally, TBPB had effects on the processing of the amyloid precursor protein toward the non-amyloidogenic pathway and decreased Abeta production in vitro. Together, these data suggest that selective activation of M(1) may provide a novel approach for the treatment of symptoms associated with schizophrenia and Alzheimer's disease.


Asunto(s)
Sitio Alostérico/fisiología , Amiloide/metabolismo , Antipsicóticos/farmacología , Bencimidazoles/farmacología , Piperidinas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Receptor Muscarínico M1/química , Receptor Muscarínico M1/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Bencimidazoles/administración & dosificación , Bencimidazoles/metabolismo , Células CHO , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Conductividad Eléctrica , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Técnicas In Vitro , Masculino , Técnicas de Placa-Clamp , Piperidinas/administración & dosificación , Piperidinas/metabolismo , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Ratas , Ratas Sprague-Dawley , Receptor Muscarínico M1/agonistas , Receptor Muscarínico M1/efectos de los fármacos , Receptores de Dopamina D2/metabolismo , Receptores de N-Metil-D-Aspartato/fisiología , Transmisión Sináptica/efectos de los fármacos , Transfección
16.
Eur J Neurosci ; 30(2): 207-16, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19659923

RESUMEN

Parkinson's Disease (PD) is marked by prominent motor symptoms that reflect striatal dopamine insufficiency. However, non-motor symptoms, including depression, are common in PD. It has been suggested that these changes reflect pathological involvement of non-dopaminergic systems. We examined regional changes in serotonin (5-HT) and norepinephrine (NE) systems in mice treated with two different 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment paradigms, at survival times of 3 or 16 weeks after the last MPTP injection. MPTP caused a decrease in striatal dopamine concentration, the magnitude of which depended on the treatment regimen and survival interval after MPTP treatment. There was significant involvement of other subcortical areas receiving a dopamine innervation, but no consistent changes in 5-HT or NE levels in subcortical sites. In contrast, we observed an enduring decrease in 5-HT and NE concentrations in both the somatosensory cortex and medial prefrontal cortex (PFC). Immunohistochemical studies also revealed a decrease in the density of PFC NE and 5-HT axons. The decrease in the cortical serotonergic innervation preferentially involved the thick beaded but not smooth fine 5-HT axons. Similar changes in the 5-HT innervation of post-mortem samples of the PFC from idiopathic PD cases were seen. Our findings point to a major loss of the 5-HT and NE innervations of the cortex in MPTP-induced parkinsonism, and suggest that loss of the beaded cortical 5-HT innervation is associated with a predisposition to the development of depression in PD.


Asunto(s)
Corteza Cerebral/metabolismo , Norepinefrina/metabolismo , Trastornos Parkinsonianos/metabolismo , Serotonina/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/análogos & derivados , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Anciano , Anciano de 80 o más Años , Animales , Recuento de Células/métodos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Desnervación/métodos , Dopamina/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Trastornos Parkinsonianos/patología
17.
Neuropsychopharmacology ; 33(6): 1276-86, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-17687264

RESUMEN

Dystrophic changes in dendrites of cortical neurons are present in several neuro-psychiatric disorders, including schizophrenia. The mechanisms that account for dendritic changes in the prefrontal cortex (PFC) in schizophrenia are unclear. Cognitive deficits in schizophrenia have been linked to compromised cortical dopamine function, and the density of the PFC dopamine innervation is decreased in schizophrenia. We determined if 6-hydroxydopamine lesions of the ventral tegmental area that disrupt the PFC dopamine innervation cause dystrophic changes in cortical neurons. Three weeks post-operatively we observed a marked decrease in basal dendritic length and spine density of layer V pyramidal cells in the prelimbic cortex; no change was seen in neurons of the motor cortex. We then examined rats in which the PFC dopamine innervation was lesioned and 3 weeks later were started on chronic treatment with an atypical (olanzapine) or typical (haloperidol) antipsychotic drug. Olanzapine but not haloperidol reversed lesion-induced changes in PFC pyramidal cell dendrites. These data suggest that dopamine regulates dendritic structure in PFC neurons. Moreover, the findings are consistent with a decrease in cortical dopaminergic tone contributing to the pathological changes in the cortex of schizophrenia, and suggest that the progressive cortical loss in schizophrenia may be slowed or reversed by treatment with atypical antipsychotic drugs.


Asunto(s)
Antipsicóticos/farmacología , Benzodiazepinas/farmacología , Espinas Dendríticas/efectos de los fármacos , Dopamina/deficiencia , Haloperidol/farmacología , Neuronas/ultraestructura , Corteza Prefrontal , Adrenérgicos/efectos adversos , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Masculino , Olanzapina , Oxidopamina/efectos adversos , Corteza Prefrontal/citología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas , Ratas Sprague-Dawley
18.
Neurosci Lett ; 435(2): 90-4, 2008 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-18358609

RESUMEN

Pericytes are small cells that are apposed to brain and meningeal microvasculature and control capillary contraction, thereby regulating local cerebral perfusion. Pericytes respond to exogenously applied glutamate in vitro and express metabotropic glutamate receptors. However, it is unclear if pericytes have the capacity to release glutamate. We therefore determined whether pericytes express vesicular glutamate transporters (VGLUTs), which are considered to be unambiguous markers of cells that use glutamate as an intercellular signaling molecule. Leptomeningeal and brain microvasculature-associated pericytes of the adult rat, as defined by the presence of NG2 proteoglycan, expressed both VGLUT2- and VGLUT3-immunoreactivity, but did not express VGLUT1. Consistent with the hypothesis that pericytes release glutamate, VGLUT2- and VGLUT3-immunoreactivities appeared to be localized to secretory vesicles. These results suggest that glutamate is released from pericytes of the leptomeninges and brain microvasculature, and demonstrate for the first time the co-localization of VGLUT2 and VGLUT3.


Asunto(s)
Encéfalo/citología , Meninges/citología , Pericitos/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Proteínas de Transporte Vesicular de Glutamato/metabolismo , Animales , Antígenos/metabolismo , Biotina/análogos & derivados , Biotina/metabolismo , Dextranos/metabolismo , Masculino , Proteoglicanos/metabolismo , Ratas , Ratas Sprague-Dawley , Estilbamidinas/metabolismo
20.
J Comp Neurol ; 526(9): 1498-1526, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29524205

RESUMEN

The prefrontal cortex (PFC) is usually defined as the frontal cortical area receiving a mediodorsal thalamic (MD) innervation. Certain areas in the medial wall of the rat frontal area receive a MD innervation. A second frontal area that is the target of MD projections is located dorsal to the rhinal sulcus and often referred to as the orbitofrontal cortex (OFC). Both the medial PFC and OFC are comprised of a large number of cytoarchitectonic regions. We assessed the afferent innervation of the different areas of the OFC, with a focus on projections arising from the mediodorsal thalamic nucleus, the basolateral nucleus of the amygdala, and the midbrain dopamine neurons. Although there are specific inputs to various OFC areas, a simplified organizational scheme could be defined, with the medial areas of the OFC receiving thalamic inputs, the lateral areas of the OFC being the recipient of amygdala afferents, and a central zone that was the target of midbrain dopamine neurons. Anterograde tracer data were consistent with this organization of afferents, and revealed that the OFC inputs from these three subcortical sites were largely spatially segregated. This spatial segregation suggests that the central portion of the OFC (pregenual agranular insular cortex) is the only OFC region that is a prefrontal cortical area, analogous to the prelimbic cortex in the medial prefrontal cortex. These findings highlight the heterogeneity of the OFC, and suggest possible functional attributes of the three different OFC areas.


Asunto(s)
Vías Aferentes/fisiología , Complejo Nuclear Basolateral/citología , Núcleo Talámico Mediodorsal/citología , Mesencéfalo/citología , Corteza Prefrontal/citología , Animales , Complejo Nuclear Basolateral/metabolismo , Monoaminas Biogénicas/metabolismo , Toxina del Cólera/metabolismo , Células HEK293 , Humanos , Masculino , Núcleo Talámico Mediodorsal/metabolismo , Mesencéfalo/metabolismo , Corteza Prefrontal/metabolismo , Ratas , Ratas Sprague-Dawley , Estilbamidinas , Transfección , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA