Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
2.
Glia ; 70(1): 50-70, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34519378

RESUMEN

Westernization of dietary habits has led to a progressive reduction in dietary intake of n-3 polyunsaturated fatty acids (n-3 PUFAs). Low maternal intake of n-3 PUFAs has been linked to neurodevelopmental disorders, conditions in which myelination processes are abnormal, leading to defects in brain functional connectivity. Only little is known about the role of n-3 PUFAs in oligodendrocyte physiology and white matter development. Here, we show that lifelong n-3 PUFA deficiency disrupts oligodendrocytes maturation and myelination processes during the postnatal period in mice. This has long-term deleterious consequences on white matter organization and hippocampus-prefrontal functional connectivity in adults, associated with cognitive and emotional disorders. Promoting developmental myelination with clemastine, a first-generation histamine antagonist and enhancer of oligodendrocyte precursor cell differentiation, rescues memory deficits in n-3 PUFA deficient animals. Our findings identify a novel mechanism through which n-3 PUFA deficiency alters brain functions by disrupting oligodendrocyte maturation and brain myelination during the neurodevelopmental period.


Asunto(s)
Ácidos Grasos Omega-3 , Animales , Encéfalo , Ratones , Vaina de Mielina , Neurogénesis , Oligodendroglía
3.
Cannabis Cannabinoid Res ; 6(6): 488-507, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34591647

RESUMEN

Background: Neuroinflammation is a key feature shared by most, if not all, neuropathologies. It involves complex biological processes that act as a protective mechanism to fight against the injurious stimuli, but it can lead to tissue damage if self-perpetuating. In this context, microglia, the main cellular actor of neuroinflammation in the brain, are seen as a double-edged sword. By phagocyting neuronal debris, these cells can not only provide tissue repair but can also contribute to neuronal damage by releasing harmful substances, including inflammatory cytokines. The mechanisms guiding these apparent opposing actions are poorly known. The endocannabinoid system modulates the release of inflammatory factors such as cytokines and could represent a functional link between microglia and neuroinflammatory processes. According to transcriptomic databases and in vitro studies, microglia, the main source of cytokines in pathological conditions, express the cannabinoid type 1 receptor (CB1R). Methods: We thus developed a conditional mouse model of CB1R deletion specifically in microglia, which was subjected to an immune challenge (peripheral lipopolysaccharide injection). Results: Our results reveal that microglial CB1R differentially controls sickness behavior in males and females. Conclusion: These findings add to the comprehension of neuroinflammatory processes and might be of great interest for future studies aimed at developing therapeutic strategies for brain disorders with higher prevalence in men.


Asunto(s)
Cannabinoides , Encefalitis , Animales , Masculino , Ratones , Microglía , Enfermedades Neuroinflamatorias , Receptores de Cannabinoides/genética
4.
Glia ; 67(1): 171-181, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30430665

RESUMEN

Although historically regarded as a homogeneous cell population, astrocytes in different brain regions exhibit differences in their physiological properties, such as gap-junction coupling, glutamate uptake dynamics, and intracellular Ca2+ response. Recent in vivo RNA profiles have further demonstrated the molecular heterogeneity of astrocytes in the adult CNS. Astrocyte heterogeneity exists not only inter-regionally but also intra-regionally. Despite the characteristic laminal organization of cortical layers and multiple sources of radial glia progenitors for (astro)gliogenesis, the molecular profile and functional properties of astroglial subpopulations in the adult cerebral cortex remain essentially undefined. Using two astrocyte reporter mouse lines: eaat2-tdTomato and Bac aldh1l1-eGFP, we identified tdT- eGFP+ , tdTlow eGFP+ , and tdThigh eGFP+ astroglial subpopulations (in an approximate 1:7:2 ratio) within the cortex. The tdT- eGFP+ astrocyte population is selectively localized at layers I-II and exhibits increased resting membrane potential and membrane resistance but reduced functional expression of the potassium channel Kir4.1. We also isolated individual astrocyte subpopulations through fluorescence activated cell sorting (FACS) and examined their transcriptome differences by RNA-seq. We found that the whole-genome transcriptional profiles of tdT- eGFP+ astrocytes are drastically different from that of tdTlow eGFP+ and tdThigh eGFP+ astrocytes. Particularly, elevated levels of several nonastrocyte genes that are typically specific to other glial cells, such as mog, mobp, Iba1, and pdgfrα, are observed in tdT- eGFP+ astrocytes, suggesting a less-specific molecular identity of these astrocytes. Overall, our study has unveiled molecular differences between adult cortical astroglial subpopulations, shedding new light on understanding their unique functions in the adult cortex.


Asunto(s)
Astrocitos/fisiología , Corteza Cerebral/citología , Corteza Cerebral/fisiología , Factores de Edad , Animales , Astrocitos/química , Encéfalo/citología , Encéfalo/fisiología , Corteza Cerebral/química , Femenino , Citometría de Flujo/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Distribución Aleatoria
5.
J Neurosci ; 37(36): 8706-8717, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28821665

RESUMEN

The molecular signature and functional properties of astroglial subtypes in the adult CNS remain largely undefined. By using translational ribosome affinity purification followed by RNA-Seq, we profiled astroglial ribosome-associated (presumably translating) mRNAs in major cortical and subcortical brain regions (cortex, hippocampus, caudate-putamen, nucleus accumbens, thalamus, and hypothalamus) of BAC aldh1l1-translational ribosome affinity purification (TRAP) mice (both sexes). We found that the expression of astroglial translating mRNAs closely follows the dorsoventral axis, especially from cortex/hippocampus to thalamus/hypothalamus posteriorly. This region-specific expression pattern of genes, such as synaptogenic modulator sparc and transcriptional factors (emx2, lhx2, and hopx), was validated by qRT-PCR and immunostaining in brain sections. Interestingly, cortical or subcortical astrocytes selectively promote neurite growth and synaptic activity of neurons only from the same region in mismatched cocultures, exhibiting region-matched astrocyte to neuron communication. Overall, these results generated new molecular signature of astrocyte types in the adult CNS, providing insights into their origin and functional diversity.SIGNIFICANCE STATEMENT We investigated the in vivo molecular and functional heterogeneity of astrocytes inter-regionally from adult brain. Our results showed that the expression pattern of ribosome-associated mRNA profiles in astrocytes closely follows the dorsoventral axis, especially posteriorly from cortex/hippocampus to thalamus/hypothalamus. In line with this, our functional results further demonstrated region-selective roles of cortical and subcortical astrocytes in regulating cortical or subcortical neuronal synaptogenesis and maturation. These in vivo studies provide a previously uncharacterized and important molecular atlas for exploring region-specific astroglial functions.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Regulación de la Expresión Génica , Ratones/metabolismo , Proteínas del Tejido Nervioso/genética , Factores de Transcripción/metabolismo , Animales , Astrocitos/clasificación , Astrocitos/citología , Encéfalo/citología , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos
6.
Front Cell Neurosci ; 11: 140, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28559797

RESUMEN

Hippocampal interneurons release the inhibitory transmitter GABA to regulate excitation, rhythm generation and synaptic plasticity. A subpopulation of GABAergic basket cells co-expresses the GABA/glycine vesicular transporters (VIAAT) and the atypical type III vesicular glutamate transporter (VGLUT3); therefore, these cells have the ability to signal with both GABA and glutamate. GABAergic transmission by basket cells has been extensively characterized but nothing is known about the functional implications of VGLUT3-dependent glutamate released by these cells. Here, using VGLUT3-null mice we observed that the loss of VGLUT3 results in a metaplastic shift in synaptic plasticity at Shaeffer's collaterals - CA1 synapses and an altered theta oscillation. These changes were paralleled by the loss of a VGLUT3-dependent inhibition of GABAergic current in CA1 pyramidal layer. Therefore presynaptic type III metabotropic could be activated by glutamate released from VGLUT3-positive interneurons. This putative presynaptic heterologous feedback mechanism inhibits local GABAergic tone and regulates the hippocampal neuronal network.

7.
J Neurosci ; 36(27): 7079-94, 2016 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-27383586

RESUMEN

UNLABELLED: How the loss of fragile X mental retardation protein (FMRP) in different brain cell types, especially in non-neuron glial cells, induces fragile X syndrome (FXS) phenotypes has just begun to be understood. In the current study, we generated inducible astrocyte-specific Fmr1 conditional knock-out mice (i-astro-Fmr1-cKO) and restoration mice (i-astro-Fmr1-cON) to study the in vivo modulation of FXS synaptic phenotypes by astroglial FMRP. We found that functional expression of glutamate transporter GLT1 is 40% decreased in i-astro-Fmr1-cKO somatosensory cortical astrocytes in vivo, which can be fully rescued by the selective re-expression of FMRP in astrocytes in i-astro-Fmr1-cON mice. Although the selective loss of astroglial FMRP only modestly increases spine density and length in cortical pyramidal neurons, selective re-expression of FMRP in astrocytes significantly attenuates abnormal spine morphology in these neurons of i-astro-Fmr1-cON mice. Moreover, we found that basal protein synthesis levels and immunoreactivity of phosphorylated S6 ribosomal protein (p-s6P) is significantly increased in i-astro-Fmr1-cKO mice, while the enhanced cortical protein synthesis observed in Fmr1 KO mice is mitigated in i-astro-Fmr1-cON mice. Furthermore, ceftriaxone-mediated upregulation of surface GLT1 expression restores functional glutamate uptake and attenuates enhanced neuronal excitability in Fmr1 KO mice. In particular, ceftriaxone significantly decreases the growth rate of abnormally accelerated body weight and completely corrects spine abnormality in Fmr1 KO mice. Together, these results show that the selective loss of astroglial FMRP contributes to cortical synaptic deficits in FXS, presumably through dysregulated astroglial glutamate transporter GLT1 and impaired glutamate uptake. These results suggest the involvement of astrocyte-mediated mechanisms in the pathogenesis of FXS. SIGNIFICANCE STATEMENT: Previous studies to understand how the loss of function of fragile X mental retardation protein (FMRP) causes fragile X syndrome (FXS) have largely focused on neurons; whether the selective loss of astroglial FMRP in vivo alters astrocyte functions and contributes to the pathogenesis of FXS remain essentially unknown. This has become a long-standing unanswered question in the fragile X field, which is also relevant to autism pathogenesis. Our current study generated astrocyte-specific Fmr1 conditional knock-out and restoration mice, and provided compelling evidence that the selective loss of astroglial FMRP contributes to cortical synaptic deficits in FXS, likely through the dysregulated astroglial glutamate transporter GLT1 expression and impaired glutamate uptake. These results demonstrate previously undescribed astrocyte-mediated mechanisms in the pathogenesis of FXS.


Asunto(s)
Astrocitos/metabolismo , Corteza Cerebral/patología , Transportador 2 de Aminoácidos Excitadores/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil , Potenciales de Acción/genética , Factores de Edad , Animales , Animales Recién Nacidos , Astrocitos/ultraestructura , Modelos Animales de Enfermedad , Antagonistas de Estrógenos/farmacología , Transportador 2 de Aminoácidos Excitadores/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/patología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Tamoxifeno/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Neurobiol Dis ; 91: 315-25, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27060558

RESUMEN

Astroglia, the most abundant glial cells in the mammalian central nervous system (CNS), are considered an emerging key player in seizure induction and progression. Although astrocytes undergo reactive gliosis in temporal lobe epilepsy (TLE) with dramatic morphological and molecular changes, specific astrocyte targets/molecular pathways that contribute to the induction and progression of seizure remain largely unknown. By combining translating ribosomal affinity purification (TRAP) with the pilocarpine model of TLE in BAC aldh1l1 TRAP mice, we profiled translating mRNAs from hippocampal or cortical astrocytes at different phases (3days, 30days, and 60days post-pilocarpine injections) of pilocarpine-induced epilepsy models. Our results found that hippocampal (but not cortical) astrocytes undergo early and unique molecular changes at 3days post-pilocarpine injections. These changes indicate a potentially primary pathogenic role of hippocampal astrocytes in seizure induction and progression and provide new insights about the involvement of specific astrocytic pathways/targets in epilepsy. In particular, we validated expression changes of ocrl and aeg1 in pilocarpine models. Follow-up studies on these genes may reveal new roles of hippocampal astrocytes in TLE.


Asunto(s)
Astrocitos/metabolismo , Epilepsia del Lóbulo Temporal/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Animales , Modelos Animales de Enfermedad , Epilepsia del Lóbulo Temporal/patología , Masculino , Ratones , Lóbulo Temporal/metabolismo
9.
Addict Biol ; 20(1): 120-31, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23980619

RESUMEN

We previously showed that maternal deprivation predisposes male rats to anxiety, accompanied with an increase in their opiate consumption. In the present report, we searched for brain epigenetic mechanisms that possibly underlie this increase. For that, we examined the expression of the methyl-CpG-binding protein MeCP2 and of the histone deacetylases HDAC2 and HDAC3, as well as the acetylation status of histone H3 and H4 in mesolimbic structures of adult maternally deprived rats, using immunohistochemistry and Western blot analysis. A long-lasting increase in MeCP2 expression was found throughout the striatum of deprived rats. Enhanced HDAC2 expression and increased nuclear HDAC activity in the nucleus accumbens of deprived rats were associated with lower acetylation levels of histone H3 and H4. Treatment for 3 weeks with the HDAC inhibitor sodium valproate abolished HDAC activation together with the decrease in the acetylation levels of histone H4, and was accompanied with normalized oral morphine consumption. The data indicate that epigenetic mechanisms induced by early adverse environment memorize life experience to trigger greater opiate vulnerability during adult life. They suggest that sodium valproate may lessen vulnerability to opiate intake, particularly in subgroups of individuals subjected to adverse postnatal environments.


Asunto(s)
Histona Desacetilasa 2/genética , Histona Desacetilasas/genética , Privación Materna , Proteína 2 de Unión a Metil-CpG/genética , Trastornos Relacionados con Opioides/genética , Acetilación , Analgésicos Opioides , Animales , Conducta Animal , Epigénesis Genética , Interacción Gen-Ambiente , Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Morfina , Núcleo Accumbens/metabolismo , Ratas , Ácido Valproico/farmacología
10.
Neurobiol Dis ; 71: 305-16, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25158291

RESUMEN

Developmental cortical malformations are associated with a high incidence of drug-resistant epilepsy. The underlying epileptogenic mechanisms, however, are poorly understood. In rodents, cortical malformations can be modeled using neonatal freeze-lesion (FL), which has been shown to cause in vitro cortical hyperexcitability. Here, we investigated the therapeutic potential of gabapentin, a clinically used anticonvulsant and analgesic, in preventing FL-induced in vitro and in vivo hyperexcitability. Gabapentin has been shown to disrupt the interaction of thrombospondin (TSP) with α2δ-1, an auxiliary calcium channel subunit. TSP/α2δ-1 signaling has been shown to drive the formation of excitatory synapses during cortical development and following injury. Gabapentin has been reported to have neuroprotective and anti-epileptogenic effects in other models associated with increased TSP expression and reactive astrocytosis. We found that both TSP and α2δ-1 were transiently upregulated following neonatal FL. We therefore designed a one-week GBP treatment paradigm to block TSP/α2δ-1 signaling during the period of their upregulation. GBP treatment prevented epileptiform activity following FL, as assessed by both glutamate biosensor imaging and field potential recording. GBP also attenuated FL-induced increases in mEPSC frequency at both P7 and 28. Additionally, GBP treated animals had decreased in vivo kainic acid (KA)-induced seizure activity. Taken together these results suggest gabapentin treatment immediately after FL can prevent the formation of a hyperexcitable network and may have therapeutic potential to minimize epileptogenic processes associated with developmental cortical malformations.


Asunto(s)
Aminas/uso terapéutico , Anticonvulsivantes/uso terapéutico , Ácidos Ciclohexanocarboxílicos/uso terapéutico , Epilepsia/tratamiento farmacológico , Epilepsia/etiología , Malformaciones del Desarrollo Cortical/complicaciones , Corteza Somatosensorial/lesiones , Ácido gamma-Aminobutírico/uso terapéutico , Factores de Edad , Animales , Animales Recién Nacidos , Canales de Calcio/metabolismo , Modelos Animales de Enfermedad , Estimulación Eléctrica , Potenciales Evocados/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/toxicidad , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Congelación/efectos adversos , Gabapentina , Proteína Ácida Fibrilar de la Glía , Ácido Glutámico/metabolismo , Técnicas In Vitro , Ácido Kaínico/toxicidad , Malformaciones del Desarrollo Cortical/etiología , Ratones , Ratones Endogámicos C57BL , Neuroimagen , Técnicas de Placa-Clamp , Corteza Somatosensorial/crecimiento & desarrollo , Trombospondinas/metabolismo
11.
J Neurosci ; 34(33): 10950-62, 2014 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-25122895

RESUMEN

Functional maturation of astroglia is characterized by the development of a unique, ramified morphology and the induction of important functional proteins, such as glutamate transporter GLT1. Although pathways regulating the early fate specification of astroglia have been characterized, mechanisms regulating postnatal maturation of astroglia remain essentially unknown. Here we used a new in vivo approach to illustrate and quantitatively analyze developmental arborization of astroglial processes. Our analysis found a particularly high increase in the number of VGluT1(+) neuronal glutamatergic synapses that are ensheathed by processes from individual developing astroglia from postnatal day (P) 14 to P26, when astroglia undergo dramatic postnatal maturation. Subsequent silencing of VGluT1(+) synaptic activity in VGluT1 KO mice significantly reduces astroglial domain growth and the induction of GLT1 in the cortex, but has no effect on astroglia in the hypothalamus, where non-VGluT1(+) synaptic signaling predominates. In particular, electron microscopy analysis showed that the loss of VGluT1(+) synaptic signaling significantly decreases perisynaptic enshealthing of astroglial processes on synapses. To further determine whether synaptically released glutamate mediates VGluT1(+) synaptic signaling, we pharmacologically inhibited and genetically ablated metabotropic glutamate receptors (mGluRs, especially mGluR5) in developing cortical astroglia and found that developmental arborization of astroglial processes and expression of functional proteins, such as GLT1, is significantly decreased. In summary, our genetic analysis provides new in vivo evidence that VGluT1(+) glutamatergic signaling, mediated by the astroglial mGluR5 receptor, regulates the functional maturation of cortical astroglia during development. These results elucidate a new mechanism for regulating the developmental formation of functional neuron-glia synaptic units.


Asunto(s)
Astrocitos/metabolismo , Corteza Cerebral/metabolismo , Neuronas/metabolismo , Transducción de Señal/fisiología , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Animales , Potenciales Postsinápticos Excitadores/fisiología , Ácido Glutámico/metabolismo , Ratones , Ratones Transgénicos , Receptor del Glutamato Metabotropico 5/genética , Receptor del Glutamato Metabotropico 5/metabolismo , Sinapsis/fisiología , Proteína 1 de Transporte Vesicular de Glutamato/genética
12.
J Neuroinflammation ; 11: 86, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24886419

RESUMEN

BACKGROUND: Increasing evidence suggests that inflammation associated with microglial cell activation in the substantia nigra (SN) of patients with Parkinson disease (PD) is not only a consequence of neuronal degeneration, but may actively sustain dopaminergic (DA) cell loss over time. We aimed to study whether the intracellular chaperone heat shock protein 60 (Hsp60) could serve as a signal of CNS injury for activation of microglial cells. METHODS: Hsp60 mRNA expression in the mesencephalon and the striatum of C57/BL6 mice treated with MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) and the Hsp60/TH mRNA ratios in the SN of PD patients and aged-matched subjects were measured. To further investigate a possible link between the neuronal Hsp60 response and PD-related cellular stress, Hsp60 immunoblot analysis and quantification in cell lysates from SH-SY5Y after treatment with 100 µM MPP+ (1-methyl-4-phenylpyridinium) at different time points (6, 12, 24 and 48 hours) compared to control cells were performed. Additional MTT and LDH assay were used. We next addressed the question as to whether Hsp60 influences the survival of TH+ neurons in mesencephalic neuron-glia cultures treated either with MPP+ (1 µM), hHsp60 (10 µg/ml) or a combination of both. Finally, we measured IL-1ß, IL-6, TNF-α and NO-release by ELISA in primary microglial cell cultures following treatment with different hHsp60 preparations. Control cultures were exposed to LPS. RESULTS: In the mesencephalon and striatum of mice treated with MPTP and also in the SN of PD patients, we found that Hsp60 mRNA was up-regulated. MPP+, the active metabolite of MPTP, also caused an increased expression and release of Hsp60 in the human dopaminergic cell line SH-SY5Y. Interestingly, in addition to being toxic to DA neurons in primary mesencephalic cultures, exogenous Hsp60 aggravated the effects of MPP+. Yet, although we demonstrated that Hsp60 specifically binds to microglial cells, it failed to stimulate the production of pro-inflammatory cytokines or NO by these cells. CONCLUSIONS: Overall, our data suggest that Hsp60 is likely to participate in DA cell death in PD but via a mechanism unrelated to cytokine release.


Asunto(s)
Chaperonina 60/metabolismo , Cuerpo Estriado/patología , Neuronas Dopaminérgicas/metabolismo , Intoxicación por MPTP/patología , Mesencéfalo/patología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Animales , Muerte Celular/efectos de los fármacos , Células Cultivadas , Chaperonina 60/genética , Modelos Animales de Enfermedad , Dopaminérgicos/farmacología , L-Lactato Deshidrogenasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Óxido Nítrico/metabolismo , Unión Proteica/efectos de los fármacos , ARN Mensajero/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
13.
J Neurosci ; 34(6): 2331-48, 2014 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-24501372

RESUMEN

Motor neurons are progressively and predominantly degenerated in ALS, which is not only induced by multiple intrinsic pathways but also significantly influenced by the neighboring glial cells. In particular, astrocytes derived from the SOD1 mutant mouse model of ALS or from human familial or sporadic ALS patient brain tissue directly induce motor neuron death in culture; however, the mechanisms of pathological astroglial secretion remain unclear. Here we investigated abnormal calcium homeostasis and altered exocytosis in SOD1G93A astrocytes. We found that purinergic stimulation induces excess calcium release from the ER stores in SOD1G93A astrocytes, which results from the abnormal ER calcium accumulation and is independent of clearance mechanisms. Furthermore, pharmacological studies suggested that store-operated calcium entry (SOCE), a calcium refilling mechanism responsive to ER calcium depletion, is enhanced in SOD1G93A astrocytes. We found that oxidant-induced increased S-glutathionylation and calcium-independent puncta formation of the ER calcium sensor STIM1 underlies the abnormal SOCE response in SOD1G93A astrocytes. Enhanced SOCE contributes to ER calcium overload in SOD1G93A astrocytes and excess calcium release from the ER during ATP stimulation. In addition, ER calcium release induces elevated ATP release from SOD1G93A astrocytes, which can be inhibited by the overexpression of dominant-negative SNARE. Selective inhibition of exocytosis in SOD1G93A astrocytes significantly prevents astrocyte-mediated toxicity to motor neurons and delays disease onset in SOD1G93A mice. Our results characterize a novel mechanism responsible for calcium dysregulation in SOD1G93A astrocytes and provide the first in vivo evidence that astrocyte exocytosis contributes to the pathogenesis of ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Astrocitos/fisiología , Señalización del Calcio/fisiología , Exocitosis/fisiología , Proteínas SNARE/metabolismo , Superóxido Dismutasa/metabolismo , Animales , Astrocitos/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo/métodos , Femenino , Masculino , Ratones , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/metabolismo , Superóxido Dismutasa/toxicidad
14.
J Neurodev Disord ; 5(1): 22, 2013 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-23988237

RESUMEN

Recent studies have implicated potentially significant roles for astrocytes in the pathogenesis of neurodevelopmental disorders. Astrocytes undergo a dramatic maturation process following early differentiation from which typical morphology and important functions are acquired. Despite significant progress in understanding their early differentiation, very little is known about how astrocytes become functionally mature. In addition, whether functional maturation of astrocytes is disrupted in neurodevelopmental disorders and the consequences of this disruption remains essentially unknown. In this review, we discuss our perspectives about how astrocyte developmental maturation is regulated, and how disruption of the astrocyte functional maturation process, especially alterations in their ability to regulate glutamate homeostasis, may alter synaptic physiology and contribute to the pathogenesis of neurodevelopmental disorders.

15.
Sci Rep ; 3: 1393, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23462811

RESUMEN

In mammalians, toll-like receptors (TLR) signal-transduction pathways induce the expression of a variety of immune-response genes, including inflammatory cytokines. It is therefore plausible to assume that TLRs are mediators in glial cells triggering the release of cytokines that ultimately kill DA neurons in the substantia nigra in Parkinson disease (PD). Accordingly, recent data indicate that TLR4 is up-regulated by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment in a mouse model of PD. Here, we wished to evaluate the role of TLR4 in the acute mouse MPTP model of PD: TLR4-deficient mice and wild-type littermates control mice were used for the acute administration way of MPTP or a corresponding volume of saline. We demonstrate that TLR4-deficient mice are less vulnerable to MPTP intoxication than wild-type mice and display a decreased number of Iba1+ and MHC II+ activated microglial cells after MPTP application, suggesting that the TLR4 pathway is involved in experimental PD.


Asunto(s)
Trastornos Parkinsonianos/metabolismo , Receptor Toll-Like 4/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/efectos adversos , Animales , Muerte Celular/genética , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Ácido Homovanílico/metabolismo , Ratones , Ratones Noqueados , Microglía/metabolismo , Trastornos Parkinsonianos/genética , Receptor Toll-Like 4/genética
16.
J Biol Chem ; 288(10): 7105-16, 2013 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-23364798

RESUMEN

Perisynaptic astrocytes express important glutamate transporters, especially excitatory amino acid transporter 2 (EAAT2, rodent analog GLT1) to regulate extracellular glutamate levels and modulate synaptic activation. In this study, we investigated an exciting new pathway, the exosome-mediated transfer of microRNA (in particular, miR-124a), in neuron-to-astrocyte signaling. Exosomes isolated from neuron-conditioned medium contain abundant microRNAs and small RNAs. These exosomes can be directly internalized into astrocytes and increase astrocyte miR-124a and GLT1 protein levels. Direct miR-124a transfection also significantly and selectively increases protein (but not mRNA) expression levels of GLT1 in cultured astrocytes. Consistent with our in vitro findings, intrastriatal injection of specific antisense against miR-124a into adult mice dramatically reduces GLT1 protein expression and glutamate uptake levels in striatum without reducing GLT1 mRNA levels. MiR-124a-mediated regulation of GLT1 expression appears to be indirect and is not mediated by its suppression of the putative GLT1 inhibitory ligand ephrinA3. Moreover, miR-124a is selectively reduced in the spinal cord tissue of end-stage SOD1 G93A mice, the mouse model of ALS. Subsequent exogenous delivery of miR-124a in vivo through stereotaxic injection significantly prevents further pathological loss of GLT1 proteins, as determined by GLT1 immunoreactivity in SOD1 G93A mice. Together, our study characterized a new neuron-to-astrocyte communication pathway and identified miRNAs that modulate GLT1 protein expression in astrocytes in vitro and in vivo.


Asunto(s)
Astrocitos/metabolismo , Transportador 2 de Aminoácidos Excitadores/genética , Exosomas/metabolismo , MicroARNs/genética , Neuronas/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Células Cultivadas , Cuerpo Estriado/citología , Cuerpo Estriado/metabolismo , Endocitosis , Transportador 2 de Aminoácidos Excitadores/metabolismo , Exosomas/ultraestructura , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glutamatos/metabolismo , Células HEK293 , Humanos , Immunoblotting , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Electrónica , Biosíntesis de Proteínas , Transducción de Señal/genética , Médula Espinal/metabolismo , Médula Espinal/patología , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo
17.
Hum Mol Genet ; 22(10): 2041-54, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23396537

RESUMEN

Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by the loss-of-function of fragile X mental retardation protein (FMRP). The loss of FMRP function in neurons abolishes its suppression on mGluR1/5-dependent dendritic protein translation, enhancing mGluR1/5-dependent synaptic plasticity and other disease phenotypes in FXS. In this study, we describe a new activation function of FMRP in regulating protein expression in astroglial cells. We found that astroglial glutamate transporter subtype glutamate transporter 1 (GLT1) and glutamate uptake is significantly reduced in the cortex of fmr1(-/-) mice. Correspondingly, neuronal excitability is also enhanced in acute fmr1(-/-) (but not in fmr1(+/+) control) cortical slices treated with low doses (10 µm) of the GLT1-specific inhibitor dihydrokainate (DHK). Using mismatched astrocyte and neuron co-cultures, we demonstrate that the loss of astroglial (but not neuronal) FMRP particularly reduces neuron-dependent GLT1 expression and glutamate uptake in co-cultures. Interestingly, protein (but not mRNA) expression and the (S)-3,5-dihydroxyphenylglycine-dependent Ca(2+) responses of astroglial mGluR5 receptor are also selectively reduced in fmr1(-/-) astrocytes and brain slices, attenuating neuron-dependent GLT1 expression. Subsequent FMRP immunoprecipitation and QRT-PCR analysis showed that astroglial mGluR5 (but not GLT1) mRNA is associated with FMRP. In summary, our results provide evidence that FMRP positively regulates translational expression of mGluR5 in astroglial cells, and FMRP-dependent down-regulation of mGluR5 underlies GLT1 dysregulation in fmr1(-/-) astrocytes. The dysregulation of GLT1 and reduced glutamate uptake may potentially contribute to enhanced neuronal excitability observed in the mouse model of FXS.


Asunto(s)
Astrocitos/metabolismo , Regulación hacia Abajo , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/metabolismo , Biosíntesis de Proteínas , Receptores de Glutamato Metabotrópico/metabolismo , Animales , Astrocitos/patología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Transportador 2 de Aminoácidos Excitadores/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/patología , Técnicas de Silenciamiento del Gen , Ácido Glutámico/genética , Ácido Glutámico/metabolismo , Humanos , Ácido Kaínico/análogos & derivados , Ácido Kaínico/farmacología , Ratones , Neuronas/metabolismo , Receptor del Glutamato Metabotropico 5 , Receptores de Glutamato Metabotrópico/genética
18.
J Physiol ; 590(20): 5183-98, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22890712

RESUMEN

Neonates respond to hypoxia initially by increasing ventilation, and then by markedly decreasing both ventilation (hypoxic ventilatory decline) and oxygen consumption (hypoxic hypometabolism). This latter process, which vanishes with age, reflects a tight coupling between ventilatory and thermogenic responses to hypoxia. The neurological substrate of hypoxic hypometabolism is unclear, but it is known to be centrally mediated, with a strong involvement of the 5-hydroxytryptamine (5-HT, serotonin) system. To clarify this issue, we investigated the possible role of VGLUT3, the third subtype of vesicular glutamate transporter. VGLUT3 contributes to glutamate signalling by 5-HT neurons, facilitates 5-HT transmission and is expressed in strategic regions for respiratory and thermogenic control. We therefore assumed that VGLUT3 might significantly contribute to the response to hypoxia. To test this possibility, we analysed this response in newborn mice lacking VGLUT3 using anatomical, biochemical, electrophysiological and integrative physiology approaches. We found that the lack of VGLUT3 did not affect the histological organization of brainstem respiratory networks or respiratory activity under basal conditions. However, it impaired respiratory responses to 5-HT and anoxia, showing a marked alteration of central respiratory control. These impairments were associated with altered 5-HT turnover at the brainstem level. Furthermore, under cold conditions, the lack of VGLUT3 disrupted the metabolic rate, body temperature, baseline breathing and the ventilatory response to hypoxia. We conclude that VGLUT3 expression is dispensable under basal conditions but is required for optimal response to hypoxic stress in neonates.


Asunto(s)
Sistemas de Transporte de Aminoácidos Acídicos/fisiología , Hipoxia/fisiopatología , Animales , Animales Recién Nacidos , Tronco Encefálico/anatomía & histología , Tronco Encefálico/fisiología , Ratones , Ratones Transgénicos , Respiración , Serotonina/fisiología , Estrés Fisiológico
19.
Neuropsychopharmacology ; 34(11): 2469-76, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19553915

RESUMEN

Maternal deprivation in rats specifically leads to a vulnerability to opiate dependence. However, the impact of cannabis exposure during adolescence on this opiate vulnerability has not been investigated. Chronic dronabinol (natural delta-9 tetrahydrocannabinol, THC) exposure during postnatal days 35-49 was made in maternal deprived (D) or non-deprived (animal facility rearing, AFR) rats. The effects of dronabinol exposure were studied after 2 weeks of washout on the rewarding effects of morphine measured in the place preference and oral self-administration tests. The preproenkephalin (PPE) mRNA levels and the relative density and functionality of CB1, and mu-opioid receptors were quantified in the striatum and the mesencephalon. Chronic dronabinol exposure in AFR rats induced an increase in sensitivity to morphine conditioning in the place preference paradigm together with a decrease of PPE mRNA levels in the nucleus accumbens and the caudate-putamen nucleus, without any modification for preference to oral morphine consumption. In contrast, dronabinol treatment on D-rats normalized PPE decrease in the striatum, morphine consumption, and suppressed sensitivity to morphine conditioning. CB1 and mu-opioid receptor density and functionality were not changed in the striatum and mesencephalon of all groups of rats. These results indicate THC potency to act as a homeostatic modifier that would worsen the reward effects of morphine on naive animals, but ameliorate the deficits in maternally D-rats. These findings point to the self-medication use of cannabis in subgroups of individuals subjected to adverse postnatal environment.


Asunto(s)
Analgésicos no Narcóticos/farmacología , Analgésicos Opioides/farmacología , Dronabinol/farmacología , Privación Materna , Morfina/farmacología , Trastornos Relacionados con Opioides/tratamiento farmacológico , Envejecimiento , Analgésicos Opioides/administración & dosificación , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Condicionamiento Clásico/efectos de los fármacos , Encefalinas/metabolismo , Masculino , Morfina/administración & dosificación , Trastornos Relacionados con Opioides/fisiopatología , Precursores de Proteínas/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Long-Evans , Receptor Cannabinoide CB1/metabolismo , Receptores Opioides mu/metabolismo , Autoadministración , Percepción Espacial/efectos de los fármacos , Factores de Tiempo
20.
J Neurosci ; 27(10): 2483-92, 2007 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-17344386

RESUMEN

Spinocerebellar ataxia 7 (SCA7) is a neurodegenerative disease caused by a polyglutamine (polyQ) expansion in the ataxin 7 (ATXN7) protein, a member of a multiprotein complex involved in histone acetylation. We have created a conditional Drosophila model of SCA7 in which expression of truncated ATXN7 (ATXN7T) with a pathogenic polyQ expansion is induced in neurons in adult flies. In this model, mutant ATXN7T accumulated in neuronal intranuclear inclusions containing ubiquitin, the 19S proteasome subunit, and HSP70 (heat shock protein 70), as in patients. Aggregation was accompanied by a decrease in locomotion and lifespan but limited neuronal death. Disaggregation of the inclusions, when expression of expanded ATXN7T was stopped, correlated with improved locomotor function and increased lifespan, suggesting that the pathology may respond to treatment. Lifespan was then used as a quantitative marker in a candidate gene approach to validate the interest of the model and to identify generic modulators of polyQ toxicity and specific modifiers of SCA7. Several molecular pathways identified in this focused screen (proteasome function, unfolded protein stress, caspase-dependent apoptosis, and histone acetylation) were further studied in primary neuronal cultures. Sodium butyrate, a histone deacetylase inhibitor, improved the survival time of the neurons. This model is therefore a powerful tool for studying SCA7 and for the development of potential therapies for polyQ diseases.


Asunto(s)
Modelos Animales de Enfermedad , Drosophila , Ataxias Espinocerebelosas/genética , Animales , Animales Modificados Genéticamente , Ataxina-7 , Muerte Celular , Células Cultivadas , Discinesias/genética , Glutamina , Humanos , Cuerpos de Inclusión Intranucleares/ultraestructura , Longevidad , Masculino , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Péptidos/genética , Fenotipo , Ratas , Ataxias Espinocerebelosas/metabolismo , Ataxias Espinocerebelosas/patología , Ataxias Espinocerebelosas/fisiopatología , Treonina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...