Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 143(4): 564-78, 2010 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-21074048

RESUMEN

Polyglutamylation is a posttranslational modification that generates glutamate side chains on tubulins and other proteins. Although this modification has been shown to be reversible, little is known about the enzymes catalyzing deglutamylation. Here we describe the enzymatic mechanism of protein deglutamylation by members of the cytosolic carboxypeptidase (CCP) family. Three enzymes (CCP1, CCP4, and CCP6) catalyze the shortening of polyglutamate chains and a fourth (CCP5) specifically removes the branching point glutamates. In addition, CCP1, CCP4, and CCP6 also remove gene-encoded glutamates from the carboxyl termini of proteins. Accordingly, we show that these enzymes convert detyrosinated tubulin into Δ2-tubulin and also modify other substrates, including myosin light chain kinase 1. We further analyze Purkinje cell degeneration (pcd) mice that lack functional CCP1 and show that microtubule hyperglutamylation is directly linked to neurodegeneration. Taken together, our results reveal that controlling the length of the polyglutamate side chains on tubulin is critical for neuronal survival.


Asunto(s)
Carboxipeptidasas/metabolismo , Proteínas de Unión al GTP/metabolismo , Degeneración Nerviosa/metabolismo , Ácido Poliglutámico/metabolismo , D-Ala-D-Ala Carboxipeptidasa de Tipo Serina/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Supervivencia Celular , Cerebelo/patología , Humanos , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Bulbo Olfatorio/patología , Alineación de Secuencia , Tubulina (Proteína)/metabolismo
2.
Neurobiol Dis ; 193: 106455, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38408685

RESUMEN

White matter (WM) tract formation and axonal pathfinding are major processes in brain development allowing to establish precise connections between targeted structures. Disruptions in axon pathfinding and connectivity impairments will lead to neural circuitry abnormalities, often associated with various neurodevelopmental disorders (NDDs). Among several neuroimaging methodologies, Diffusion Tensor Imaging (DTI) is a magnetic resonance imaging (MRI) technique that has the advantage of visualizing in 3D the WM tractography of the whole brain non-invasively. DTI is particularly valuable in unpinning structural tract connectivity defects of neural networks in NDDs. In this study, we used 3D DTI to unveil brain-specific tract defects in two mouse models lacking the Nr2f1 gene, which mutations in patients have been proven to cause an emerging NDD, called Bosch-Boonstra-Schaaf Optic Atrophy (BBSOAS). We aimed to investigate the impact of the lack of cortical Nr2f1 function on WM morphometry and tract microstructure quantifications. We found in both mutant mice partial loss of fibers and severe misrouting of the two major cortical commissural tracts, the corpus callosum, and the anterior commissure, as well as the two major hippocampal efferent tracts, the post-commissural fornix, and the ventral hippocampal commissure. DTI tract malformations were supported by 2D histology, 3D fluorescent imaging, and behavioral analyses. We propose that these interhemispheric connectivity impairments are consistent in explaining some cognitive defects described in BBSOAS patients, particularly altered information processing between the two brain hemispheres. Finally, our results highlight 3DDTI as a relevant neuroimaging modality that can provide appropriate morphometric biomarkers for further diagnosis of BBSOAS patients.


Asunto(s)
Atrofia Óptica , Sustancia Blanca , Humanos , Ratones , Animales , Imagen de Difusión Tensora , Sustancia Blanca/diagnóstico por imagen , Sustancia Blanca/patología , Encéfalo , Imagen por Resonancia Magnética , Atrofia Óptica/patología
3.
MAGMA ; 36(4): 577-587, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36695926

RESUMEN

OBJECT: Exploring mouse brains by rapid 3D-Diffusion Tensor Imaging (3D-DTI) of high spatial resolution (HSR) is challenging in vivo. Here we use the super resolution reconstruction (SRR) postprocessing method to demonstrate its performance on Microtubule-Associated-Protein6 Knock-Out (MAP6-KO) mice. MATERIALS AND METHODS: Two spin-echo DTI were acquired (9.4T, CryoProbe RF-coil): (i)-multislice 2D-DTI, (echo-planar integrating reversed-gradient) acquired in vivo in the three orthogonal orientations (360 µm slice-thickness, 120 × 120 µm in-plane resolution, 56 min scan duration); used in SRR software to reconstruct SRR 3D-DTI with HSR in slice-plane (120 × 120 × 120 µm) and (ii)-microscopic 3D-DTI (µ-3D-DTI), (100 × 100 × 100 µm; 8 h 6 min) on fixed-brains ex vivo, that were removed after paramagnetic contrast-agent injection to accelerate scan acquisition using short repetition-times without NMR-signal sensitivity loss. RESULTS: White-matter defects, quantified from both 3D-DTI fiber-tracking were found very similar. Indeed, as expected the fornix and cerebral-peduncle volume losses were - 39% and - 35% in vivo (SRR 3D-DTI) versus - 34% and - 32% ex vivo (µ-3D-DTI), respectively (p<0.001). This finding is robust since the µ-3D-DTI feasibility on MAP6-KO ex vivo was already validated by fluorescent-microscopy of cleared brains. DISCUSSION: First performance of the SRR to generate rapid HSR 3D-DTI of mouse brains in vivo is demonstrated. The method is suitable in neurosciences for longitudinal studies to identify molecular and genetic abnormalities in mouse models that are of growing developments.


Asunto(s)
Imagen de Difusión Tensora , Imagen por Resonancia Magnética , Animales , Ratones , Imagen de Difusión Tensora/métodos , Ratones Noqueados , Imagen por Resonancia Magnética/métodos , Encéfalo/diagnóstico por imagen , Microtúbulos
4.
Mol Cell Neurosci ; 120: 103726, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35367368

RESUMEN

VPS35 is a core component of the retromer complex involved in familial forms of neurodegenerative diseases such as Parkinson's and Alzheimer's disease. In mice, VPS35 is expressed during early brain development. However, previous studies have reported that VPS35 activity is largely dispensable for normal neuronal development and initial elaboration of axonal projections. Here, we evaluated the role of VPS35 in the mouse embryonic brain using two Cre-driver lines that remove Vps35 from the cortex at different prenatal stages. We found that Vps35 mutant mice displayed microcephaly and decreased cortical thickness from the embryonic stages to adulthood. VPS35 also regulates cortical development by affecting a subpopulation of neural progenitor cells and the survival of postmitotic neurons. In addition, we showed that a lack of VPS35 leads to hypoplasia and misrouting of several axonal projections, including the anterior commissure and fornix. Furthermore, VPS35 deficiency impairs the non-autonomous development of thalamocortical axons (TCAs), which show severe disruption of innervation and terminal arborization in the cortex. Together, these data demonstrate that VPS35 plays a greater role in embryonic development of the mammalian brain than it was previously thought.


Asunto(s)
Enfermedades Neurodegenerativas , Proteínas de Transporte Vesicular , Animales , Axones/metabolismo , Mamíferos , Ratones , Enfermedades Neurodegenerativas/metabolismo , Neurogénesis , Neuronas/metabolismo , Proteínas de Transporte Vesicular/metabolismo
5.
Hum Mol Genet ; 28(20): 3391-3405, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31363758

RESUMEN

Reversible detyrosination of tubulin, the building block of microtubules, is crucial for neuronal physiology. Enzymes responsible for detyrosination were recently identified as complexes of vasohibins (VASHs) one or two with small VASH-binding protein (SVBP). Here we report three consanguineous families, each containing multiple individuals with biallelic inactivation of SVBP caused by truncating variants (p.Q28* and p.K13Nfs*18). Affected individuals show brain abnormalities with microcephaly, intellectual disability and delayed gross motor and speech development. Immunoblot testing in cells with pathogenic SVBP variants demonstrated that the encoded proteins were unstable and non-functional, resulting in a complete loss of VASH detyrosination activity. Svbp knockout mice exhibit drastic accumulation of tyrosinated tubulin and a reduction of detyrosinated tubulin in brain tissue. Similar alterations in tubulin tyrosination levels were observed in cultured neurons and associated with defects in axonal differentiation and architecture. Morphological analysis of the Svbp knockout mouse brains by anatomical magnetic resonance imaging showed a broad impact of SVBP loss, with a 7% brain volume decrease, numerous structural defects and a 30% reduction of some white matter tracts. Svbp knockout mice display behavioural defects, including mild hyperactivity, lower anxiety and impaired social behaviour. They do not, however, show prominent memory defects. Thus, SVBP-deficient mice recapitulate several features observed in human patients. Altogether, our data demonstrate that deleterious variants in SVBP cause this neurodevelopmental pathology, by leading to a major change in brain tubulin tyrosination and alteration of microtubule dynamics and neuron physiology.


Asunto(s)
Encéfalo/anomalías , Encéfalo/metabolismo , Proteínas de Ciclo Celular/metabolismo , Neuronas/metabolismo , Tubulina (Proteína)/metabolismo , Animales , Proteínas Portadoras/metabolismo , Disfunción Cognitiva/genética , Disfunción Cognitiva/metabolismo , Femenino , Humanos , Immunoblotting , Imagen por Resonancia Magnética , Ratones , Microcefalia/genética , Microcefalia/metabolismo , Tirosina/metabolismo
6.
Neuroimage ; 96: 133-42, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24704457

RESUMEN

The MAP6 (microtubule-associated protein 6) KO mouse is a microtubule-deficient model of schizophrenia that exhibits severe behavioral disorders that are associated with synaptic plasticity anomalies. These defects are alleviated not only by neuroleptics, which are the gold standard molecules for the treatment of schizophrenia, but also by Epothilone D (Epo D), which is a microtubule-stabilizing molecule. To compare the neuronal transport between MAP6 KO and wild-type mice and to measure the effect of Epo D treatment on neuronal transport in KO mice, MnCl2 was injected in the primary somatosensory cortex. Then, using manganese-enhanced magnetic resonance imaging (MEMRI), we followed the propagation of Mn(2+) through axonal tracts and brain regions that are connected to the somatosensory cortex. In MAP6 KO mice, the measure of the MRI relative signal intensity over 24h revealed that the Mn(2+) transport rate was affected with a stronger effect on long-range and polysynaptic connections than in short-range and monosynaptic tracts. The chronic treatment of MAP6 KO mice with Epo D strongly increased Mn(2+) propagation within both mono- and polysynaptic connections. Our results clearly indicate an in vivo deficit in neuronal Mn(2+) transport in KO MAP6 mice, which might be due to both axonal transport defects and synaptic transmission impairments. Epo D treatment alleviated the axonal transport defects, and this improvement most likely contributes to the positive effect of Epo D on behavioral defects in KO MAP6 mice.


Asunto(s)
Epotilonas/uso terapéutico , Imagen por Resonancia Magnética/métodos , Manganeso/farmacocinética , Proteínas Asociadas a Microtúbulos/metabolismo , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/fisiopatología , Corteza Somatosensorial/fisiopatología , Animales , Medios de Contraste , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Corteza Somatosensorial/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Resultado del Tratamiento , Moduladores de Tubulina/uso terapéutico
7.
Int J Neuropsychopharmacol ; 16(1): 189-98, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22226089

RESUMEN

Hyperforin is one of the main bioactive compounds that underlie the antidepressant actions of the medicinal plant Hypericum perforatum (St. John's wort). However, the effects of a chronic hyperforin treatment on brain cells remains to be fully addressed. The following study was undertaken to further advance our understanding of the biological effects of this plant extract on neurons. Special attention was given to its impact on the brain-derived neurotrophic factor (BDNF) receptor TrkB and on adult hippocampal neurogenesis since they appear central to the mechanisms of action of antidepressants. The consequences of a chronic hyperforin treatment were investigated on cortical neurons in culture and on the brain of adult mice treated for 4 wk with a daily injection (i.p.) of hyperforin (4 mg/kg). Its effects on the expression of the cyclic adenosine monophosphate response element-binding protein (CREB), phospho-CREB (p-CREB), TrkB and phospho-TrkB (p-TrkB) were analysed by Western blot experiments and its impact on adult hippocampal neurogenesis was also investigated. Hyperforin stimulated the expression of TRPC6 channels and TrkB via SKF-96365-sensitive channels controlling a downstream signalling cascade involving Ca(2+), protein kinase A, CREB and p-CREB. In vivo, hyperforin augmented the expression of TrkB in the cortex but not in the hippocampus where hippocampal neurogenesis remained unchanged. In conclusion, this plant extract acts on the cortical BDNF/TrkB pathway leaving adult hippocampal neurogenesis unaffected. This study provides new insights on the neuronal responses controlled by hyperforin. We propose that the cortex is an important brain structure targeted by hyperforin.


Asunto(s)
Antidepresivos/farmacología , Corteza Cerebral/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Floroglucinol/análogos & derivados , Receptor trkB/fisiología , Terpenos/farmacología , Regulación hacia Arriba/fisiología , Factores de Edad , Animales , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Femenino , Hipocampo/citología , Hipocampo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Floroglucinol/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Embarazo , Receptor trkB/biosíntesis , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología
8.
Front Neurosci ; 17: 1172830, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37332879

RESUMEN

Compressed sensing (CS) is widely used to accelerate clinical diffusion MRI acquisitions, but it is not widely used in preclinical settings yet. In this study, we optimized and compared several CS reconstruction methods for diffusion imaging. Different undersampling patterns and two reconstruction approaches were evaluated: conventional CS, based on Berkeley Advanced Reconstruction Toolbox (BART-CS) toolbox, and a new kernel low-rank (KLR)-CS, based on kernel principal component analysis and low-resolution-phase (LRP) maps. 3D CS acquisitions were performed at 9.4T using a 4-element cryocoil on mice (wild type and a MAP6 knockout). Comparison metrics were error and structural similarity index measure (SSIM) on fractional anisotropy (FA) and mean diffusivity (MD), as well as reconstructions of the anterior commissure and fornix. Acceleration factors (AF) up to 6 were considered. In the case of retrospective undersampling, the proposed KLR-CS outperformed BART-CS up to AF = 6 for FA and MD maps and tractography. For instance, for AF = 4, the maximum errors were, respectively, 8.0% for BART-CS and 4.9% for KLR-CS, considering both FA and MD in the corpus callosum. Regarding undersampled acquisitions, these maximum errors became, respectively, 10.5% for BART-CS and 7.0% for KLR-CS. This difference between simulations and acquisitions arose mainly from repetition noise, but also from differences in resonance frequency drift, signal-to-noise ratio, and in reconstruction noise. Despite this increased error, fully sampled and AF = 2 yielded comparable results for FA, MD and tractography, and AF = 4 showed minor faults. Altogether, KLR-CS based on LRP maps seems a robust approach to accelerate preclinical diffusion MRI and thereby limit the effect of the frequency drift.

9.
J Neurochem ; 121(1): 99-114, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22146001

RESUMEN

The microtubule-associated Stable Tubulie Only Polypeptide (STOP; also known as MAP6) protein plays a key role in neuron architecture and synaptic plasticity, the dysfunctions of which are thought to be implicated in the pathophysiology of psychiatric diseases. The deletion of STOP in mice leads to severe disorders reminiscent of several schizophrenia-like symptoms, which are also associated with differential alterations of the serotonergic tone in somas versus terminals. In STOP knockout (KO) compared with wild-type mice, serotonin (5-HT) markers are found to be markedly accumulated in the raphe nuclei and, in contrast, deeply depleted in all serotonergic projection areas. In the present study, we carefully examined whether the 5-HT imbalance would lead to behavioral consequences evocative of mood and/or cognitive disorders. We showed that STOP KO mice exhibited depression-like behavior, associated with a decreased anxiety-status in validated paradigms. In addition, although STOP KO mice had a preserved very short-term memory, they failed to perform well in all other learning and memory tasks. We also showed that STOP KO mice exhibited regional imbalance of the norepinephrine tone as observed for 5-HT. As a consequence, mutant mice were hypersensitive to acute antidepressants with different selectivity. Altogether, these data indicate that the deletion of STOP protein in mice caused deep alterations in mood and cognitive performances and that STOP protein might have a crucial role in the 5-HT and norepinephrine networks development.


Asunto(s)
Trastornos del Conocimiento/genética , Eliminación de Gen , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/genética , Trastornos del Humor/genética , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Animales , Conducta Animal/fisiología , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/psicología , Femenino , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/fisiología , Trastornos del Humor/metabolismo , Trastornos del Humor/psicología , Proteínas del Tejido Nervioso/fisiología
10.
Science ; 377(6613): eabq5011, 2022 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-36137051

RESUMEN

Recent evidence has shown that even mild mutations in the Huntingtin gene that are associated with late-onset Huntington's disease (HD) disrupt various aspects of human neurodevelopment. To determine whether these seemingly subtle early defects affect adult neural function, we investigated neural circuit physiology in newborn HD mice. During the first postnatal week, HD mice have less cortical layer 2/3 excitatory synaptic activity than wild-type mice, express fewer glutamatergic receptors, and show sensorimotor deficits. The circuit self-normalizes in the second postnatal week but the mice nonetheless develop HD. Pharmacologically enhancing glutamatergic transmission during the neonatal period, however, rescues these deficits and preserves sensorimotor function, cognition, and spine and synapse density as well as brain region volume in HD adult mice.


Asunto(s)
Encéfalo , Proteína Huntingtina , Enfermedad de Huntington , Red Nerviosa , Neurogénesis , Sinapsis , Animales , Encéfalo/anomalías , Modelos Animales de Enfermedad , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/embriología , Enfermedad de Huntington/genética , Ratones , Ratones Transgénicos , Red Nerviosa/anomalías , Neurogénesis/genética , Sinapsis/fisiología
11.
Front Mol Neurosci ; 14: 665693, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34025352

RESUMEN

The development and function of the central nervous system rely on the microtubule (MT) and actin cytoskeletons and their respective effectors. Although the structural role of the cytoskeleton has long been acknowledged in neuronal morphology and activity, it was recently recognized to play the role of a signaling platform. Following this recognition, research into Microtubule Associated Proteins (MAPs) diversified. Indeed, historically, structural MAPs-including MAP1B, MAP2, Tau, and MAP6 (also known as STOP);-were identified and described as MT-binding and -stabilizing proteins. Extensive data obtained over the last 20 years indicated that these structural MAPs could also contribute to a variety of other molecular roles. Among multi-role MAPs, MAP6 provides a striking example illustrating the diverse molecular and cellular properties of MAPs and showing how their functional versatility contributes to the central nervous system. In this review, in addition to MAP6's effect on microtubules, we describe its impact on the actin cytoskeleton, on neuroreceptor homeostasis, and its involvement in signaling pathways governing neuron development and maturation. We also discuss its roles in synaptic plasticity, brain connectivity, and cognitive abilities, as well as the potential relationships between the integrated brain functions of MAP6 and its molecular activities. In parallel, the Collapsin Response Mediator Proteins (CRMPs) are presented as examples of how other proteins, not initially identified as MAPs, fall into the broader MAP family. These proteins bind MTs as well as exhibiting molecular and cellular properties very similar to MAP6. Finally, we briefly summarize the multiple similarities between other classical structural MAPs and MAP6 or CRMPs.In summary, this review revisits the molecular properties and the cellular and neuronal roles of the classical MAPs, broadening our definition of what constitutes a MAP.

12.
Elife ; 102021 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-34860155

RESUMEN

Neurodevelopmental axonal pathfinding plays a central role in correct brain wiring and subsequent cognitive abilities. Within the growth cone, various intracellular effectors transduce axonal guidance signals by remodeling the cytoskeleton. Semaphorin-3E (Sema3E) is a guidance cue implicated in development of the fornix, a neuronal tract connecting the hippocampus to the hypothalamus. Microtubule-associated protein 6 (MAP6) has been shown to be involved in the Sema3E growth-promoting signaling pathway. In this study, we identified the collapsin response mediator protein 4 (CRMP4) as a MAP6 partner and a crucial effector in Sema3E growth-promoting activity. CRMP4-KO mice displayed abnormal fornix development reminiscent of that observed in Sema3E-KO mice. CRMP4 was shown to interact with the Sema3E tripartite receptor complex within detergent-resistant membrane (DRM) domains, and DRM domain integrity was required to transduce Sema3E signaling through the Akt/GSK3 pathway. Finally, we showed that the cytoskeleton-binding domain of CRMP4 is required for Sema3E's growth-promoting activity, suggesting that CRMP4 plays a role at the interface between Sema3E receptors, located in DRM domains, and the cytoskeleton network. As the fornix is affected in many psychiatric diseases, such as schizophrenia, our results provide new insights to better understand the neurodevelopmental components of these diseases.


Asunto(s)
Fórnix/crecimiento & desarrollo , Proteínas del Tejido Nervioso/genética , Semaforinas/genética , Transducción de Señal , Animales , Femenino , Fórnix/metabolismo , Masculino , Ratones , Proteínas del Tejido Nervioso/metabolismo , Semaforinas/metabolismo
13.
J Neurochem ; 115(6): 1579-94, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20969568

RESUMEN

The deletion of microtubule-associated protein stable tubule only polypeptide (STOP) leads to neuroanatomical, biochemical and severe behavioral alterations in mice, partly alleviated by antipsychotics. Therefore, STOP knockout (KO) mice have been proposed as a model of some schizophrenia-like symptoms. Preliminary data showed decreased brain serotonin (5-HT) tissue levels in STOP KO mice. As literature data demonstrate various interactions between microtubule-associated proteins and 5-HT, we characterized some features of the serotonergic neurotransmission in STOP KO mice. In the brainstem, mutant mice displayed higher tissue 5-HT levels and in vivo synthesis rate, together with marked increases in 5-HT transporter densities and 5-HT1A autoreceptor levels and electrophysiological sensitivity, without modification of the serotonergic soma number. Conversely, in projection areas, STOP KO mice exhibited lower 5-HT levels and in vivo synthesis rate, associated with severe decreases in 5-HT transporter densities, possibly related to reduced serotonergic terminals. Mutant mice also displayed a deficit of adult hippocampal neurogenesis, probably related to both STOP deletion and 5-HT depletion. Finally, STOP KO mice exhibited a reduced anxiety- and, probably, an increased helpness-status, that could be because of the strong imbalance of the serotonin neurotransmission between somas and terminals. Altogether, these data suggested that STOP deletion elicited peculiar 5-HT disconnectivity.


Asunto(s)
Encéfalo/fisiología , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/genética , Red Nerviosa/fisiología , Serotonina/metabolismo , Animales , Femenino , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Noqueados
14.
Mol Pain ; 6: 96, 2010 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-21172011

RESUMEN

BACKGROUND: Mice deficient for the stable tubule only peptide (STOP) display altered dopaminergic neurotransmission associated with severe behavioural defects including disorganized locomotor activity. Endogenous morphine, which is present in nervous tissues and synthesized from dopamine, may contribute to these behavioral alterations since it is thought to play a role in normal and pathological neurotransmission. RESULTS: In this study, we showed that STOP null brain structures, including cortex, hippocampus, cerebellum and spinal cord, contain high endogenous morphine amounts. The presence of elevated levels of morphine was associated with the presence of a higher density of mu opioid receptor with a higher affinity for morphine in STOP null brains. Interestingly, STOP null mice exhibited significantly lower nociceptive thresholds to thermal and mechanical stimulations. They also had abnormal behavioural responses to the administration of exogenous morphine and naloxone. Low dose of morphine (1 mg/kg, i.p.) produced a significant mechanical antinociception in STOP null mice whereas it has no effect on wild-type mice. High concentration of naloxone (1 mg/kg) was pronociceptive for both mice strain, a lower concentration (0.1 mg/kg) was found to increase the mean mechanical nociceptive threshold only in the case of STOP null mice. CONCLUSIONS: Together, our data show that STOP null mice displayed elevated levels of endogenous morphine, as well as an increase of morphine receptor affinity and density in brain. This was correlated with hypernociception and impaired pharmacological sensitivity to mu opioid receptor ligands.


Asunto(s)
Proteínas Asociadas a Microtúbulos/deficiencia , Morfina/farmacología , Dolor/fisiopatología , Analgésicos Opioides , Animales , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/fisiología , Morfina/análisis , Naloxona/administración & dosificación , Naloxona/farmacología , Antagonistas de Narcóticos , Proteínas del Tejido Nervioso , Alcaloides Opiáceos , Dolor/tratamiento farmacológico , Receptores Opioides mu/análisis , Receptores Opioides mu/metabolismo
15.
J Immunol ; 181(2): 1207-14, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18606674

RESUMEN

The thymus continuously produces T lymphocytes that contribute to the maintenance of the peripheral T cell pool. Since peripheral recirculating T cells represent a very minor population among total thymocytes in normal animals, the relationship between the thymus and secondary lymphoid organs is generally considered unidirectional. Recently, several reports have described the presence of recirculating T cells in the thymus, raising issues regarding their possible function. In this article, we show that the niche for recirculating T cells in the thymus, i.e., their absolute number, is the same in lymphopenic and normal mice. Using a novel combination of TCR-transgenic mice in which the ligand necessary for positive selection of host T cells is only expressed by transferred donor T cells, we show that mature T cells recirculating back to the thymus can mediate positive selection.


Asunto(s)
Linfocitos T/inmunología , Timo/inmunología , Animales , Diferenciación Celular , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Parabiosis , Linfocitos T/metabolismo , Timo/fisiología
16.
Biol Rev Camb Philos Soc ; 95(3): 738-758, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32027773

RESUMEN

The S100 genes encode a conserved group of 21 vertebrate-specific EF-hand calcium-binding proteins. Since their discovery in 1965, S100 proteins have remained enigmatic in terms of their cellular functions. In this review, we summarize the calcium- and zinc-binding properties of the dimeric S100B and S100A1 proteins and highlight data that shed new light on the extracellular and intracellular regulation and functions of S100B. We point out that S100B and S100A1 homodimers are not functionally interchangeable and that in a S100A1/S100B heterodimer, S100A1 acts as a negative regulator for the ability of S100B to bind Zn2+ . The Ca2+ and Zn2+ -dependent interactions of S100B with a wide array of proteins form the basis of its activities and have led to the derivation of some initial rules for S100B recognition of protein targets. However, recent findings have strongly suggested that these rules need to be revisited. Here, we describe a new consensus S100B binding motif present in intracellular and extracellular vertebrate-specific proteins and propose a new model for stable interactions of S100B dimers with full-length target proteins. A chaperone-associated function for intracellular S100B in adaptive cellular stress responses is also discussed. This review may help guide future studies on the functions of S100 proteins in general.


Asunto(s)
Calcio/metabolismo , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Proteínas S100/metabolismo , Zinc/metabolismo , Animales , Humanos , Estructura Molecular , Subunidad beta de la Proteína de Unión al Calcio S100/química , Proteínas S100/química
17.
PLoS One ; 15(7): e0234529, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32673338

RESUMEN

Morphometry characterization is an important procedure in describing neuronal cultures and identifying phenotypic differences. This task usually requires labor-intensive measurements and the classification of numerous neurites from large numbers of neurons in culture. To automate these measurements, we wrote AutoNeuriteJ, an imageJ/Fiji plugin that measures and classifies neurites from a very large number of neurons. We showed that AutoNeuriteJ is able to detect variations of neuritic growth induced by several compounds known to affect the neuronal growth. In these experiments measurement of more than 5000 mouse neurons per conditions was obtained within a few hours. Moreover, by analyzing mouse neurons deficient for the microtubule associated protein 6 (MAP6) and wild type neurons we illustrate that AutoNeuriteJ is capable to detect subtle phenotypic difference in axonal length. Overall the use of AutoNeuriteJ will provide rapid, unbiased and accurate measurement of neuron morphologies.


Asunto(s)
Procesamiento de Imagen Asistido por Computador/métodos , Neuritas/metabolismo , Neuronas/fisiología , Animales , Axones/fisiología , Proliferación Celular , Células Cultivadas , Hipocampo/fisiología , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Neurogénesis/fisiología , Programas Informáticos
18.
Elife ; 92020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32452382

RESUMEN

Studies have suggested that amyloid precursor protein (APP) regulates synaptic homeostasis, but the evidence has not been consistent. In particular, signaling pathways controlling APP transport to the synapse in axons and dendrites remain to be identified. Having previously shown that Huntingtin (HTT), the scaffolding protein involved in Huntington's disease, regulates neuritic transport of APP, we used a microfluidic corticocortical neuronal network-on-a-chip to examine APP transport and localization to the pre- and post-synaptic compartments. We found that HTT, upon phosphorylation by the Ser/Thr kinase Akt, regulates APP transport in axons but not dendrites. Expression of an unphosphorylatable HTT decreased axonal anterograde transport of APP, reduced presynaptic APP levels, and increased synaptic density. Ablating in vivo HTT phosphorylation in APPPS1 mice, which overexpress APP, reduced presynaptic APP levels, restored synapse number and improved learning and memory. The Akt-HTT pathway and axonal transport of APP thus regulate APP presynaptic levels and synapse homeostasis.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteína Huntingtina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sinapsis/metabolismo , Animales , Transporte Axonal , Encéfalo/diagnóstico por imagen , Modelos Animales de Enfermedad , Homeostasis , Imagen por Resonancia Magnética , Masculino , Memoria , Ratones Transgénicos , Técnicas Analíticas Microfluídicas , Prueba del Laberinto Acuático de Morris , Fosforilación
19.
Elife ; 82019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31294694

RESUMEN

Adult neurogenesis in the olfactory bulb (OB) is considered as a competition in which neurons scramble during a critical selection period for integration and survival. Moreover, newborn neurons are thought to replace pre-existing ones that die. Despite indirect evidence supporting this model, systematic in vivo observations are still scarce. We used two-photon in vivo imaging to study neuronal integration and survival. We show that loss of new neurons in the OB after arrival at terminal positions occurs only at low levels. Moreover, long-term observations showed that no substantial cell death occurred at later stages. Neuronal death was induced by standard doses of thymidine analogs, but disappeared when low doses were used. Finally, we demonstrate that the OB grows throughout life. This shows that neuronal selection during OB-neurogenesis does not occur after neurons reached stable positions. Moreover, this suggests that OB neurogenesis does not represent neuronal turnover but lifelong neuronal addition.


Asunto(s)
Neurogénesis , Neuronas/fisiología , Bulbo Olfatorio/crecimiento & desarrollo , Animales , Muerte Celular , Ratones , Modelos Neurológicos
20.
J Neurosci ; 27(17): 4716-24, 2007 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-17460084

RESUMEN

In the germinative zone of the adult rodent brain, neural progenitors migrate into niches delimited by astrocyte processes and differentiate into neuronal precursors. In the present study, we report a modulating role for the scaffolding protein IQGAP1 in neural progenitor migration. We have identified IQGAP1 as a new marker of amplifying neural progenitor and neuronal precursor cells of the subventricular zone (SVZ) and the rostral migratory stream (RMS) in the adult mouse brain. To determine functions for IQGAP1 in neural progenitors, we compared the properties of neural progenitor cells from wild-type and Iqgap1-null mutant mice in vivo and in vitro. The in vivo studies reveal a delay in the transition of de novo neural progenitors into neuronal precursor cells in Iqgap1-null mice. In vitro, we demonstrated that IQGAP1 acts as a downstream effector in the vascular endothelial growth factor (VEGF)-dependent migratory response of neural progenitors that also impacts on their neuronal differentiation. The Rho-family GTPases cdc42/Rac1 and Lis1 are major partners of IQGAP1 in this migratory process. Finally, astrocytes of the neurogenic SVZ and RMS are shown to express VEGF. We propose that VEGF synthesized by astrocytes could be involved in the guidance of neural progenitors to neurogenic niches and that IQGAP1 is an effector of the VEGF-dependent migratory signal.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/fisiología , Movimiento Celular/fisiología , Neuronas/citología , Proteínas Activadoras de ras GTPasa/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Comunicación Celular/fisiología , Diferenciación Celular/fisiología , Movimiento Celular/efectos de los fármacos , Ventrículos Cerebrales/citología , Técnicas In Vitro , Ratones , Ratones Noqueados , Neuropéptidos/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1 , Proteínas Activadoras de ras GTPasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA