Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2440: 253-270, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35218544

RESUMEN

Stimulated emission depletion (STED) microscopy is one of the optical superresolution microscopy (SRM) techniques, more recently also referred to as nanoscopy, that have risen to popularity among biologists during the past decade. These techniques keep pushing the physical boundaries of optical resolution toward the molecular scale. Thereby, they enable biologists to image cellular and tissue structures at a level of almost molecular detail that was previously only achievable using electron microscopy. All the while, they retain the advantages of light microscopy, in particular with regards to sample preparation and flexibility of imaging. Commercially available SRM setups have become more and more available and also increasingly sophisticated, both in terms of optical performance and, importantly, ease of use. Institutional microscopy core facilities now offer widespread access to this type of systems. However, the field has grown so rapidly, and keeps growing, that biologists can be easily overwhelmed by the multitude of available techniques and approaches. From this vast array of SRM modalities, STED stands out in one respect: it is essentially an extension to an advanced confocal microscope. Most experienced users of confocal microscopy will find the transition to STED microscopy relatively easy as compared with some other SRM techniques. This also applies to STED sample preparation. Nonetheless, because resolution in STED microscopy does not only depend on the wavelength of the incident light and the numerical aperture of the objective, but crucially also on the square root of the intensity of the depletion laser and, in general, on the photochemical interaction of the fluorophore with the depletion laser, some additional considerations are necessary in STED sample preparation. Here we describe the single color staining of the somatostatin receptor subtype 2A (SSTR2A) and dual color staining of the trans-Golgi-network protein TGN 38 and the t-SNARE syntaxin-6 for STED in the endocrine cell line AtT20 and STED imaging of the samples, providing the protocols in as general a form as possible. The protocols in this chapter are used in this way in an institutional microscopy core facility.


Asunto(s)
Colorantes Fluorescentes , Rayos Láser , Microscopía Confocal , Microscopía Fluorescente/métodos
2.
Nat Cell Biol ; 23(12): 1271-1286, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34873283

RESUMEN

Mitochondrial-derived vesicles (MDVs) are implicated in diverse physiological processes-for example, mitochondrial quality control-and are linked to various neurodegenerative diseases. However, their specific cargo composition and complex molecular biogenesis are still unknown. Here we report the proteome and lipidome of steady-state TOMM20+ MDVs. We identified 107 high-confidence MDV cargoes, which include all ß-barrel proteins and the TOM import complex. MDV cargoes are delivered as fully assembled complexes to lysosomes, thus representing a selective mitochondrial quality control mechanism for multi-subunit complexes, including the TOM machinery. Moreover, we define key biogenesis steps of phosphatidic acid-enriched MDVs starting with the MIRO1/2-dependent formation of thin membrane protrusions pulled along microtubule filaments, followed by MID49/MID51/MFF-dependent recruitment of the dynamin family GTPase DRP1 and finally DRP1-dependent scission. In summary, we define the function of MDVs in mitochondrial quality control and present a mechanistic model for global GTPase-driven MDV biogenesis.


Asunto(s)
Vesículas Citoplasmáticas/fisiología , Dinaminas/metabolismo , Dinámicas Mitocondriales/fisiología , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Lipidómica , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales/metabolismo , Enfermedades Neurodegenerativas/patología , Factores de Elongación de Péptidos/metabolismo , Ácidos Fosfatidicos/metabolismo , Proteoma/genética , Interferencia de ARN , ARN Interferente Pequeño/genética
3.
J Chem Neuroanat ; 111: 101881, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33160048

RESUMEN

Serotonin (5-HT) is a common neurotransmitter in mammals, playing a central role in the regulation of various processes such as sleep, perception, cognitive and autonomic functions in the nervous system. Previous studies have demonstrated that 5-HT type 3 (5-HT3) receptors are expressed in either or both the substantia nigra (SN) and the dorsal raphe nucleus (DRN) in humans, marmosets, rats and Syrian hamsters. Here, we quantify the distribution of 5-HT3 receptors across these regions in the adult rat. Fluorescent immunohistochemistry was performed on sections of rat brain covering the entire rostro-caudal extent of the SN and DRN with antibodies specific to the 5-HT3A receptor subunit, as well as others targeting the monoaminergic markers tyrosine hydroxylase (TH) and the 5-HT transporter (SERT). The number of 5-HT3A receptor-positive, TH-positive (n = 28,428 ±â€¯888, Gundersen's m = 1 coefficient of error [CE] = 0.05) and SERT-positive (n = 12,852 ±â€¯462, CE = 0.06) cells were estimated in both the SN and the DRN using stereology. We found that 5-HT3A receptor-positive cells are present in the SNr (n = 1250 ±â€¯64, CE = 0.24), but they did not co-localise with TH-positive cells, nor were they present in the SNc. In contrast, no 5-HT3A receptor-positive cells were found in the DRN. These results support the presence of 5-HT3 receptors in the SN, but not in the DRN, and do not support their expression on monoaminergic cells within these two brain areas.


Asunto(s)
Núcleo Dorsal del Rafe/metabolismo , Neuronas/metabolismo , Receptores de Serotonina 5-HT3/metabolismo , Sustancia Negra/metabolismo , Animales , Femenino , Masculino , Ratas , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
4.
Front Neural Circuits ; 14: 21, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32523515

RESUMEN

Neuroendocrine circuits are orchestrated by the pituitary gland in response to hypothalamic hormone-releasing and inhibiting factors to generate an ultradian and/or circadian rhythm of hormone secretion. However, mechanisms that govern this rhythmicity are not fully understood. It has been shown that synaptic transmission in the rodent hypothalamus undergoes cyclical changes in parallel with rhythmic hormone secretion and a growing body of evidence suggests that rapid rewiring of hypothalamic neurons may be the source of these changes. For decades, structural synaptic studies have been utilizing electron microscopy, which provides the resolution suitable for visualizing synapses. However, the small field of view, limited specificity and manual analysis susceptible to bias fuel the search for a more quantitative approach. Here, we apply the fluorescence super-resolution microscopy approach direct Stochastic Optical Reconstruction Microscopy (dSTORM) to quantify and structurally characterize excitatory and inhibitory synapses that contact growth hormone-releasing-hormone (GHRH) neurons during peak and trough values of growth hormone (GH) concentration in mice. This approach relies on a three-color immunofluorescence staining of GHRH and pre- and post-synaptic markers, and a quantitative analysis with a Density-Based Spatial Clustering of Applications with Noise (DBSCAN) algorithm. With this method we confirm our previous findings, using electron microscopy, of increased excitatory synaptic input to GHRH neurons during peak levels of GH. Additionally, we find a shift in synapse numbers during low GH levels, where more inhibitory synaptic inputs are detected. Lastly, we utilize dSTORM to study novel aspects of synaptic structure. We show that more excitatory (but not inhibitory) pre-synaptic clusters associate with excitatory post-synaptic clusters during peaks of GH secretion and that the numbers of post-synaptic clusters increase during high hormone levels. The results presented here provide an opportunity to highlight dSTORM as a valuable quantitative approach to study synaptic structure in the neuroendocrine circuit. Importantly, our analysis of GH circuitry sheds light on the potential mechanism that drives ultradian changes in synaptic transmission and possibly aids in GH pulse generation in mice.


Asunto(s)
Hormona Liberadora de Hormona del Crecimiento/metabolismo , Hormona del Crecimiento/metabolismo , Microscopía/métodos , Sinapsis/metabolismo , Ritmo Ultradiano/fisiología , Animales , Hormona del Crecimiento/análisis , Hormona Liberadora de Hormona del Crecimiento/análisis , Masculino , Ratones , Ratones Endogámicos C57BL , Sinapsis/química
5.
Endocrine ; 69(3): 655-669, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32383089

RESUMEN

BACKGROUND: Somatostatin (SOM) receptor subtype 2 (SSTR2) is the major receptor subtype mediating SOM effects throughout the neuraxis. We previously demonstrated that the non-selective agonist [D-Trp8]-SOM induces intracellular sequestration of SSTR2, whereas this receptor is maintained at the cell surface after treatment with the SSTR2-selective agonist L-779,976 in cells co-expressing SSTR2 and SSTR5. METHODS AND RESULTS: In this study, we knocked-out SSTR5 in AtT20 cells endogenously expressing both SSTR2 and SSTR5 and used immuno-labeling and confocal microscopy to investigate the effect of SSTR5 on regulation of SSTR2 trafficking. Our results indicate that unlike [D-Trp8]-SOM-induced intracellular sequestration, L-779,976 stimulation results in the maintenance of SSTR2 at the cell surface regardless of whether SSTR5 is present or not. We then examined the trafficking pathways of SSTR2 upon stimulation by either agonist. We found that both [D-Trp8]-SOM and L-779,976 induce SSTR2 internalization via transferrin-positive vesicles. However, SSTR2 internalized upon L-779,976 treatment undergoes rapid recycling to the plasma membrane, whereas receptors internalized by [D-Trp8]-SOM recycle slowly after washout of the agonist. Furthermore, [D-Trp8]-SOM stimulation induces degradation of a fraction of internalized SSTR2 whereas L-779,976-dependent, rapid SSTR2 recycling appears to protect internalized SSTR2 from degradation. In addition, Octreotide which has preferential SSTR2 affinity, induced differential effects on both SSTR2 trafficking and degradation. CONCLUSION: Our results indicate that the biased agonistic property of L-779,976 protects against SSTR2 surface depletion by rapidly initiating SSTR2 recycling while SSTR5 does not regulate L-779-976-dependent SSTR2 trafficking.


Asunto(s)
Células Neuroendocrinas , Receptores de Somatostatina , Octreótido , Receptores de Somatostatina/genética , Somatostatina
6.
J Cell Biol ; 219(1)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31825461

RESUMEN

The surfacing of the glucose transporter GLUT4 driven by insulin receptor activation provides the prototypic example of a homeostasis response dependent on mobilization of an intracellular storage compartment. Here, we generalize this concept to a G protein-coupled receptor, somatostatin receptor subtype 2 (SSTR2), in pituitary cells. Following internalization in corticotropes, SSTR2 moves to a juxtanuclear syntaxin-6-positive compartment, where it remains until the corticotropes are stimulated with corticotropin releasing factor (CRF), whereupon SSTR2 exits the compartment on syntaxin-6-positive vesicular/tubular carriers that depend on Rab10 for their fusion with the plasma membrane. As SSTR2 activation antagonizes CRF-mediated hormone release, this storage/resurfacing mechanism may allow for a physiological homeostatic feedback system. In fact, we find that SSTR2 moves from an intracellular compartment to the cell surface in pituitary gland somatotropes, concomitant with increasing levels of serum growth hormone (GH) during natural GH cycles. Our data thus provide a mechanism by which signaling-mediated plasma membrane resurfacing of SSTR2 can fine-tune pituitary hormone release.


Asunto(s)
Corticotrofos/metabolismo , Hormona de Crecimiento Humana/metabolismo , Hipófisis/metabolismo , Proteínas Qa-SNARE/metabolismo , Receptores de Somatostatina/metabolismo , Proteínas de Unión al GTP rab/fisiología , Animales , Hormona Liberadora de Corticotropina , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Hipófisis/citología , Proteínas Qa-SNARE/genética , Receptores de Somatostatina/genética , Transducción de Señal
7.
Neurobiol Dis ; 106: 205-213, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28709992

RESUMEN

Low-voltage fast (LVF) and hypersynchronous (HYP) - onset seizures occur in the EEG obtained with depth electrodes from mesial temporal lobe epilepsy (MTLE) patients and animal models. In epileptic rats analyzed up to approximately two weeks after pilocarpine-induced status epilepticus (SE), these patterns are associated with specific high-frequency oscillation (HFO) content: ripples (80-200Hz) or fast-ripples (250-500Hz) predominate in LVF or HYP seizures, respectively. To establish whether these features change over the course of the disease, we recorded the EEG from the hippocampal CA3 subfield, subiculum, entorhinal cortex and dentate gyrus in two groups of pilocarpine-treated rats: the "early stage group" (n=8) was analyzed from day 3 to 20 post-SE while the "late stage group" (n=7) was studied from day 27 to 53 post-SE. We found that: (i) HYP and LVF seizures prevail in the early and late stage group, respectively; (ii) HYP seizures mainly originate from CA3 in the early stage group only; (iii) LVF seizures in both early and late stage group originate from a diffuse network; (iv) LVF and HYP seizures in the early stage group are mainly associated with ripples and fast ripples, respectively; but (v) fast ripples predominate in the late stage group, regardless of seizure onset pattern. Finally, extensive neuronal loss occurred in the hippocampus of the late stage group. Our results reveal that significant changes in ictogenesis and HFO occurrence, which are associated with the manifestation of severe hippocampal damage, occur over time in this MTLE model.


Asunto(s)
Corteza Entorrinal/fisiopatología , Epilepsia del Lóbulo Temporal/fisiopatología , Hipocampo/fisiopatología , Convulsiones/fisiopatología , Animales , Ondas Encefálicas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Electrocorticografía , Corteza Entorrinal/patología , Epilepsia del Lóbulo Temporal/patología , Hipocampo/patología , Masculino , Pilocarpina , Ratas Sprague-Dawley , Convulsiones/patología , Estado Epiléptico/patología , Estado Epiléptico/fisiopatología , Factores de Tiempo
8.
Cell Metab ; 23(5): 797-810, 2016 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-27166944

RESUMEN

While leptin is a well-known regulator of body fat mass, it remains unclear how circulating leptin is sensed centrally to maintain energy homeostasis. Here we show that genetic and pharmacological ablation of adult NG2-glia (also known as oligodendrocyte precursors), but not microglia, leads to primary leptin resistance and obesity in mice. We reveal that NG2-glia contact the dendritic processes of arcuate nucleus leptin receptor (LepR) neurons in the median eminence (ME) and that these processes degenerate upon NG2-glia elimination, which explains the consequential attenuation of these neurons' molecular and electrical responses to leptin. Our data therefore indicate that LepR dendrites in the ME represent the principal conduits of leptin's anorexigenic action and that NG2-glia are essential for their maintenance. Given that ME-directed X-irradiation confirmed the pharmacological and genetically mediated ablation effects on body weight, our findings provide a rationale for the known obesity risk associated with cranial radiation therapy.


Asunto(s)
Leptina/metabolismo , Eminencia Media/metabolismo , Neuroglía/metabolismo , Envejecimiento/efectos de los fármacos , Envejecimiento/metabolismo , Animales , Anorexia/metabolismo , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Peso Corporal/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citarabina/farmacología , Dendritas/efectos de los fármacos , Dendritas/metabolismo , Fenómenos Electrofisiológicos/efectos de los fármacos , Femenino , Eliminación de Gen , Masculino , Eminencia Media/efectos de los fármacos , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Mitosis/efectos de los fármacos , Obesidad/metabolismo , Obesidad/patología , Fenotipo , Receptores de Leptina/metabolismo , Transducción de Señal/efectos de los fármacos , Aumento de Peso/efectos de los fármacos
9.
Data Brief ; 4: 368-73, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26217818

RESUMEN

We provide a detailed morphometric analysis of eight transmission electron micrographs (TEMs) obtained from the corpus callosum of one cynomolgus macaque. The raw TEM images are included in the article, along with the distributions of the axon caliber and the myelin g-ratio in each image. The distributions are analyzed to determine the relationship between axon caliber and g-ratio, and compared against the aggregate metrics (myelin volume fraction, fiber volume fraction, and the aggregate g-ratio), as defined in the accompanying research article entitled 'In vivo histology of the myelin g-ratio with magnetic resonance imaging' (Stikov et al., NeuroImage, 2015).

10.
Neuroimage ; 118: 397-405, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26004502

RESUMEN

The myelin g-ratio, defined as the ratio between the inner and the outer diameter of the myelin sheath, is a fundamental property of white matter that can be computed from a simple formula relating the myelin volume fraction to the fiber volume fraction or the axon volume fraction. In this paper, a unique combination of magnetization transfer, diffusion imaging and histology is presented, providing a novel method for in vivo magnetic resonance imaging of the axon volume fraction and the myelin g-ratio. Our method was demonstrated in the corpus callosum of one cynomolgus macaque, and applied to obtain full-brain g-ratio maps in one healthy human subject and one multiple sclerosis patient. In the macaque, the g-ratio was relatively constant across the corpus callosum, as measured by both MRI and electron microscopy. In the human subjects, the g-ratio in multiple sclerosis lesions was higher than in normal appearing white matter, which was in turn higher than in healthy white matter. Measuring the g-ratio brings us one step closer to fully characterizing white matter non-invasively, making it possible to perform in vivo histology of the human brain during development, aging, disease and treatment.


Asunto(s)
Axones/ultraestructura , Encéfalo/ultraestructura , Imagen de Difusión por Resonancia Magnética/métodos , Vaina de Mielina/ultraestructura , Adulto , Animales , Cuerpo Calloso/ultraestructura , Humanos , Macaca fascicularis , Fenómenos Magnéticos , Masculino , Ratones Mutantes Neurológicos , Esclerosis Múltiple/patología
11.
J Comp Neurol ; 522(11): 2650-62, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24478034

RESUMEN

The mammalian habenulae consist of medial (MHb) and lateral (LHb) nuclear complexes. Especially the LHb has received much interest because it has been recognized as the potential center of an "anti-reward system." Subnuclear organization and connectivity of the LHb are well known. In contrast, criteria to classify habenular neurons into distinct groups with potentially different biological functions are missing, most likely as a result of the lack of appropriate marker proteins. Actually, a huge amount of data concerning the localization of more than 20,000 mouse protein genes is provided in the Allen Brain Atlas. Unfortunately, the immediate use of this information is prohibited by the fact that the subnuclear organization of the habenular complexes in mouse is not known so far. The present report, therefore, uses topographic, structural, and cytochemical information from the rat to recognize corresponding areas within the mouse habenulae. Taking advantage of the fact that the Klüver-Barrera technique allows simultaneous observation of neuronal cell bodies and myelinated fibers, we were able to correlate subnuclear areas in the mouse habenula to subnuclei, which had been rigorously identified by several criteria in the rat. Our data suggest that the topographic localization of habenular subnuclei is rather similar between mouse and rat and that they may be homologous in these two species. Consequently, our data may allow using the Allen Brain Atlas as a source of basal information, which should be helpful to select candidate molecular markers for functionally different neurons in the mouse and potentially in higher mammalian species.


Asunto(s)
Habénula/anatomía & histología , Ratones/anatomía & histología , Neuronas/citología , Ratas/anatomía & histología , Animales , Atlas como Asunto , Dendritas/metabolismo , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Habénula/metabolismo , Inmunohistoquímica , Masculino , Ratones/metabolismo , Fibras Nerviosas Mielínicas/metabolismo , Proteínas de Neurofilamentos/metabolismo , Neuronas/metabolismo , Neurópilo/metabolismo , Fotomicrografía , Ratas/metabolismo , Receptores de GABA-B/metabolismo , Serotonina/metabolismo , Especificidad de la Especie , Tirosina 3-Monooxigenasa/metabolismo
12.
FASEB J ; 27(9): 3741-52, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23756650

RESUMEN

Management of painful peripheral neuropathies remains challenging, since patients with chronic pain respond poorly to the available pharmacopeia. In recent years, the G-protein-coupled receptor neurotensin (NT) type 2 (NTS2) emerged as an attractive target for treating transitory pain states. To date, however, there is no evidence for its role in the regulation of chronic peripheral neuropathies. Here, we found that NTS2 receptors were largely localized to primary afferent fibers and superficial dorsal horns. Changes in the time course of the gene expression profile of NT, NTS1, and NTS2 were observed over a 28-d period following the sciatic nerve constriction [chronic constriction injury (CCI) model]. We next determined the effects of central delivery of selective-NTS2 agonists to CCI-treated rats on both mechanical allodynia (evoked withdrawal responses) and weight-bearing deficits (discomfort and quality-of-life proxies). The NTS2 analogs JMV431, levocabastine, and ß-lactotensin were all effective in reducing ongoing tactile allodynia in CCI-treated rats. Likewise, amitriptyline, pregabalin, and morphine significantly attenuated CCI-induced mechanical hypersensitivity. NTS2 agonists were also efficient in reversing weight-bearing and postural deficits caused by nerve damage, unlike reference analgesics currently used in the clinic. Thus, NTS2 agonists may offer new treatment avenues for limiting pain associated with peripheral neuropathies and improve functional rehabilitation and well-being.


Asunto(s)
Neuralgia/tratamiento farmacológico , Receptores de Neurotensina/metabolismo , Analgésicos/uso terapéutico , Animales , Western Blotting , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Masculino , Microscopía Electrónica de Transmisión , Neuralgia/inducido químicamente , Oligopéptidos/uso terapéutico , Piperidinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Receptores de Neurotensina/agonistas , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo
13.
Eur J Neurosci ; 36(10): 3388-95, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22925106

RESUMEN

Somatostatin (SST) is a neuropeptide with known inhibitory actions in the hypothalamus, where it inhibits release of growth hormone-releasing hormone (GHRH), while also influencing the sleep-wake cycle. Here we investigated in the rat whether SST neurons might additionally release GABA (gamma-aminobutyric acid) or glutamate in different regions and whether they might contact orexin neurons that play an important role in the maintenance of wakefulness. In dual-immunostained sections viewed by epifluorescence microscopy, we examined if SST varicosities were immunopositive for the vesicular transporter for GABA (VGAT) or glutamate (VGLUT2) in the posterolateral hypothalamus and neighboring arcuate nucleus and median eminence. Of the SST varicosities in the posterolateral hypothalamus, 18% were immunopositive for VGAT, whereas ≤ 1% were immunopositive for VGLUT2. In the arcuate and median eminence, 26 and 64% were VGAT+ and < 3% VGLUT2 + , respectively. In triple-immunostained sections viewed by epifluorescence and confocal microscopy, SST varicosities were seen in contact with orexin somata, and of these varicosities, a significant proportion (23%) contained VGAT along with synaptophysin, the presynaptic marker for small synaptic vesicles, and a similar proportion (25%) abutted puncta that were immunostained for gephyrin, the postsynaptic marker for GABAergic synapses. Our results indicate that a significant proportion of SST varicosities in the hypothalamus have the capacity to release GABA, to form inhibitory synapses upon orexin neurons, and accordingly through their peptide and/or amino acid, to inhibit orexin neurons, as well as GHRH neurons. Thus while regulating GHRH release, SST neurons could serve to attenuate arousal and permit progression through the sleep cycle.


Asunto(s)
Proteínas Transportadoras de GABA en la Membrana Plasmática/análisis , Hipotálamo/química , Péptidos y Proteínas de Señalización Intracelular/análisis , Neuronas/química , Neuropéptidos/análisis , Somatostatina/análisis , Sistema de Transporte de Aminoácidos X-AG/análisis , Animales , Masculino , Orexinas , Ratas , Ratas Sprague-Dawley , Vesículas Sinápticas/química
14.
J Comp Neurol ; 520(5): 889-913, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21953547

RESUMEN

Protein interacting specifically with Tc10, PIST, is a Golgi-associated sorting protein involved in regulating cell-surface targeting of plasma membrane receptors. The present study provides the first comprehensive description of PIST distribution in the mammalian central nervous system and of its subcellular localization by immunocytochemistry. PIST is distributed widely throughout the neuraxis, predominantly associated with neuronal cell bodies and dendrites. In hippocampal neurons, in vitro and in situ, PIST displayed a patchy subcellular distribution in an area surrounding the nucleus and extending into one of the major dendrites. By colocalization with the trans-Golgi marker TGN38, we were able to show that PIST is associated largely but not exclusively with the trans-Golgi network in central neurons. High or moderate to high levels of PIST-like immunoreactivity were found in cortical areas, in particular in layer V of the neocortex. The motor cortex was most strongly labeled. Also, the piriform and insular cortices displayed strong PIST labeling. In the hippocampus, CA2 but not CA1 or CA3 pyramidal cells displayed strong PIST-labeling, extending into their apical dendrites. In the thalamus, ventrolateral and laterodorsal nuclei were most strongly stained, whereas in the hypothalamus the supraoptic nucleus stood out with strong immunoreactivity. Strikingly, in the brainstem all cranial nerve motor nuclei were PIST-positive at varying levels, which is in keeping with the prominent expression of PIST in forebrain motor areas. This selective distribution of PIST suggests that the protein serves distinctive roles in specific neuronal populations, establishing functionally distinct zones, for instance, in the hippocampus.


Asunto(s)
Proteínas Portadoras/metabolismo , Sistema Nervioso Central/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Superficie Celular/metabolismo , Fracciones Subcelulares/metabolismo , Animales , Proteínas Portadoras/química , Células Cultivadas , Sistema Nervioso Central/química , Cobayas , Células HEK293 , Hipocampo/química , Hipocampo/metabolismo , Humanos , Masculino , Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/química , Ratas , Ratas Sprague-Dawley , Fracciones Subcelulares/química , Red trans-Golgi/química , Red trans-Golgi/metabolismo
15.
J Neurosci ; 31(23): 8381-94, 2011 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-21653843

RESUMEN

Netrins are guidance cues involved in neural connectivity. We have shown that the netrin-1 receptor DCC (deleted in colorectal cancer) is involved in the functional organization of the mesocorticolimbic dopamine (DA) system. Adult mice with a heterozygous loss-of-function mutation in dcc exhibit changes in indexes of DA function, including DA-related behaviors. These phenotypes are only observed after puberty, a critical period in the maturation of the mesocortical DA projection. Here, we examined whether dcc heterozygous mice exhibit structural changes in medial prefrontal cortex (mPFC) DA synaptic connectivity, before and after puberty. Stereological counts of tyrosine-hydroxylase (TH)-positive varicosities were increased in the cingulate 1 and prelimbic regions of the pregenual mPFC. dcc heterozygous mice also exhibited alterations in the size, complexity, and dendritic spine density of mPFC layer V pyramidal neuron basilar dendritic arbors. Remarkably, these presynaptic and postsynaptic partner phenotypes were not observed in juvenile mice, suggesting that DCC selectively influences the extensive branching and synaptic differentiation that occurs in the maturing mPFC DA circuit at puberty. Immunolabeling experiments in wild-type mice demonstrated that DCC is segregated to TH-positive fibers innervating the nucleus accumbens, with only scarce DCC labeling in mPFC TH-positive fibers. Netrin had an inverted target expression pattern. Thus, DCC-mediated netrin-1 signaling may influence the formation/maintenance of mesocorticolimbic DA topography. In support of this, we report that dcc heterozygous mice exhibit a twofold increase in the density of mPFC DCC/TH-positive varicosities. Our results implicate DCC-mediated netrin-1 signaling in the establishment of mPFC DA circuitry during puberty.


Asunto(s)
Dopamina/metabolismo , Red Nerviosa/metabolismo , Neuronas/metabolismo , Corteza Prefrontal/metabolismo , Receptores de Superficie Celular/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Análisis de Varianza , Animales , Recuento de Células , Receptor DCC , Espinas Dendríticas/metabolismo , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Receptores de Superficie Celular/genética , Sinapsis/metabolismo , Proteínas Supresoras de Tumor/genética , Tirosina 3-Monooxigenasa/metabolismo
16.
PLoS One ; 5(7): e11463, 2010 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-20628609

RESUMEN

Puberty is a critical period in mesocorticolimbic dopamine (DA) system development, particularly for the medial prefrontal cortex (mPFC) projection which achieves maturity in early adulthood. The guidance cue netrin-1 organizes neuronal networks by attracting or repelling cellular processes through DCC (deleted in colorectal cancer) and UNC-5 homologue (UNC5H) receptors, respectively. We have shown that variations in netrin-1 receptor levels lead to selective reorganization of mPFC DA circuitry, and changes in DA-related behaviors, in transgenic mice and in rats. Significantly, these effects are only observed after puberty, suggesting that netrin-1 mediated effects on DA systems vary across development. Here we report on the normal expression of DCC and UNC5H in the ventral tegmental area (VTA) by DA neurons from embryonic life to adulthood, in both mice and rats. We show a dramatic and enduring pubertal change in the ratio of DCC:UNC5H receptors, reflecting a shift toward predominant UNC5H function. This shift in DCC:UNC5H ratio coincides with the pubertal emergence of UNC5H expression by VTA DA neurons. Although the distribution of DCC and UNC5H by VTA DA neurons changes during puberty, the pattern of netrin-1 immunoreactivity in these cells does not. Together, our findings suggest that DCC:UNC5H ratios in DA neurons at critical periods may have important consequences for the organization and function of mesocorticolimbic DA systems.


Asunto(s)
Dopamina/metabolismo , Neuronas/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Western Blotting , Receptor DCC , Femenino , Regulación del Desarrollo de la Expresión Génica , Genotipo , Inmunohistoquímica , Ratones , Ratones Mutantes , Microscopía Fluorescente , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptores de Netrina , Netrina-1 , Embarazo , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Destete
17.
J Neurosci ; 29(25): 8198-205, 2009 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-19553459

RESUMEN

Growth hormone (GH) secretion in male rats exhibits a 3.3 h ultradian rhythm generated by the reciprocal interaction of GH-releasing hormone (GHRH) and somatostatin (SRIF). SRIF receptor subtypes sst(1) and sst(2) are highly expressed in GHRH neurons of the hypothalamic arcuate nucleus (ARC). We previously demonstrated an ultradian oscillation in binding of SRIF analogs to the ARC in relation to GH peaks and troughs. Here we tested the hypothesis that these ultradian changes in SRIF binding are due to differential plasma membrane targeting of sst(1) receptors in ARC neurons using immunocytochemistry and electron microscopy. We found that 87% of sst(1)-positive ARC neurons also synthesized GHRH. Subcellularly, 80% of sst(1) receptors were located intracellularly and 20% at the plasma membrane regardless of GH status. However, whereas 30% of the cell-surface sst(1) receptors were located perisynaptically or subsynaptically following exposure to high GH secretion, this fraction was increased to 42% following a GH trough period (p = 0.05). Furthermore, the relative abundance of symmetric and asymmetric synapses on sst(1)-positive dendrites also varied significantly, depending on the GH cycle, from approximately equal numbers following GH troughs to 70:30 in favor of symmetric, i.e., inhibitory, inputs after GH peaks (p < 0.02). These findings suggest that postsynaptic localization of sst(1) receptors and synaptic connectivity in the ARC undergo pronounced remodeling in parallel with the GH rhythm. Such synaptic plasticity may be an important mechanism by which sst(1) mediates SRIF's cyclical effects on ARC GHRH neurons to generate the ultradian rhythm of GH secretion.


Asunto(s)
Ciclos de Actividad/fisiología , Núcleo Arqueado del Hipotálamo/metabolismo , Hormona Liberadora de Hormona del Crecimiento/sangre , Hormona del Crecimiento/sangre , Neuronas/metabolismo , Receptores de Somatostatina/metabolismo , Sinapsis/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/ultraestructura , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Masculino , Microscopía Electrónica , Neuronas/ultraestructura , Radioinmunoensayo , Ratas , Ratas Sprague-Dawley , Somatostatina/sangre , Sinapsis/ultraestructura
18.
J Neurochem ; 107(2): 398-417, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18691385

RESUMEN

The netrin-1 receptor Deleted in Colorectal Cancer (DCC) is required for the formation of major axonal projections by embryonic cortical neurons, including the corpus callosum, hippocampal commissure, and cortico-thalamic tracts. The presentation of DCC by axonal growth cones is tightly regulated, but the mechanisms regulating DCC trafficking within neurons are not well understood. Here, we investigated the mechanisms regulating DCC recruitment to the plasma membrane of embryonic cortical neurons. In embryonic spinal commissural neurons, protein kinase A (PKA) activation recruits DCC to the plasma membrane and enhances axon chemoattraction to netrin-1. We demonstrate that PKA activation similarly recruits DCC and increases embryonic cortical neuron axon extension, which, like spinal commissural neurons, respond to netrin-1 as a chemoattractant. We then determined if depolarization might recruit DCC to the plasma membrane. Neither netrin-1 induced axon extension, nor levels of plasma membrane DCC, were altered by depolarizing embryonic spinal commissural neurons with elevated levels of KCl. In contrast, depolarizing embryonic cortical neurons increased the amount of plasma membrane DCC, including at the growth cone, and increased axon outgrowth evoked by netrin-1. Inhibition of PKA, phosphatidylinositol-3-kinase, protein kinase C, or exocytosis blocked the depolarization-induced recruitment of DCC and suppressed axon outgrowth. Inhibiting protein synthesis did not affect DCC recruitment, nor were the distributions of trkB or neural cell adhesion molecule (NCAM) influenced by depolarization, consistent with selective mobilization of DCC. These findings identify a role for membrane depolarization modulating the response of axons to netrin-1 by regulating DCC recruitment to the plasma membrane.


Asunto(s)
Axones/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Corteza Cerebral/citología , Factores de Crecimiento Nervioso/efectos de los fármacos , Neuronas/ultraestructura , Receptores de Superficie Celular/metabolismo , Proteínas Supresoras de Tumor/efectos de los fármacos , Proteínas Supresoras de Tumor/metabolismo , Animales , Axones/fisiología , Axones/efectos de la radiación , Proteína de Unión a CREB/metabolismo , Calcio/metabolismo , Membrana Celular/metabolismo , Membrana Celular/efectos de la radiación , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Colforsina/farmacología , AMP Cíclico/metabolismo , Receptor DCC , Estimulación Eléctrica/métodos , Embrión de Mamíferos , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Ratones , Netrina-1 , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Cloruro de Potasio/farmacología , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Ratas , Receptor trkB/metabolismo , Proteína 2 de Membrana Asociada a Vesículas/metabolismo
19.
J Biol Chem ; 283(33): 22774-86, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18556652

RESUMEN

Scy1-like 1 (Scyl1), a member of the Scy1-like family of catalytically inactive protein kinases, was recently identified as the gene product altered in muscle-deficient mice, which suffer from motor neuron degeneration and cerebellar atrophy. To determine the function of Scyl1, we have now used a mass spectrometry-based screen to search for Scyl1-binding partners and identified components of coatomer I (COPI) coats. The interaction was confirmed in pull-down assays, and Scyl1 co-immunoprecipitates with betaCOP from brain lysates. Interestingly, and unique for a non-transmembrane domain protein, Scyl1 binds COPI coats using a C-terminal RKLD-COO(-) sequence, similar to the KKXX-COO(-) COPI-binding motif found in transmembrane endoplasmic reticulum (ER) proteins. Scyl1 co-localizes with betaCOP and is localized, in an Arf1-independent manner, to the ER-Golgi intermediate compartment and the cis-Golgi, sites of COPI-mediated membrane budding. The localization and binding properties of Scyl1 strongly suggest a function in COPI transport, and inhibitory RNA-mediated knock down of the protein disrupts COPI-mediated retrograde traffic of the KDEL receptor to the ER without affecting anterograde traffic from the ER. Our data demonstrate a function for Scyl1 as an accessory factor in COPI trafficking and suggest for the first time that alterations in the COPI pathway result in neurodegenerative disease.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Proteína Coat de Complejo I/metabolismo , Mutación , Proteínas Quinasas/genética , Degeneraciones Espinocerebelosas/genética , Factores de Transcripción/genética , Animales , Sitios de Unión , Encéfalo/enzimología , Genes Recesivos , Proteínas Quinasas/metabolismo , Ratas , Factores de Transcripción/metabolismo
20.
Endocrinology ; 148(5): 2095-105, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17272399

RESUMEN

The somatostatin [somatotropin release-inhibiting factor (SRIF)] receptor subtypes sst(2A) and sst(5) are frequently coexpressed in SRIF-responsive cells, including endocrine pituitary cells. We previously demonstrated that sst(2A) and sst(5) exhibit different subcellular localizations and regulation of cell surface expression, although they have similar signaling properties. We investigated here whether sst(2A) and sst(5) functionally interact in cells coexpressing the two receptor subtypes. We stimulated both transfected cells stably expressing sst(2A) alone (CHO-sst(2A)) or together with sst(5) (CHO-sst(2A+5)) and the pituitary cell line AtT20, which endogenously expresses the two receptor subtypes, with either the nonselective agonist [D-Trp(8)]-SRIF-14 or the sst(2)-selective agonist L-779,976. In CHO-sst(2A) cells, stimulation with either ligand resulted in the loss of approximately 75% of cell surface SRIF binding sites and massive internalization of sst(2A) receptors. The cells were desensitized to subsequent stimulation with [D-Trp(8)]-SRIF-14, which failed to inhibit forskolin-evoked cAMP accumulation. Similarly, in CHO-sst(2A+5) and AtT20 cells, [D-Trp(8)]-SRIF-14 induced the loss of 60-70% of SRIF binding sites as well as massive sst(2A) endocytosis. By contrast, in cells expressing both sst(2A) and sst(5), selective stimulation of sst(2A) with L-779,976 resulted in only 20-40% loss of cell surface binding and markedly reduced sst(2A) internalization. Consequently, whereas CHO-sst(2A+5) and AtT20 cells stimulated with [D-Trp(8)]-SRIF-14 were desensitized to a second stimulation with the same agonist, cells prestimulated with L-779,976 were not desensitized to subsequent [D-Trp(8)]-SRIF-14 stimulation. These findings indicate that the presence of sst(5) in the same cells modulates trafficking and cell surface regulation of sst(2A) and cellular desensitization to the effects of SRIF.


Asunto(s)
Endocitosis/fisiología , Transporte de Proteínas/fisiología , Receptores de Somatostatina/metabolismo , Amidas/metabolismo , Amidas/farmacología , Animales , Células CHO , Colforsina/farmacología , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Indoles/metabolismo , Indoles/farmacología , Radioisótopos de Yodo , Ratones , Ensayo de Unión Radioligante , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/genética , Transducción de Señal/fisiología , Somatostatina/análogos & derivados , Somatostatina/metabolismo , Somatostatina/farmacología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...