Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Neurosci ; 42(29): 5803-5814, 2022 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-35701158

RESUMEN

Mature protoplasmic astroglia in the mammalian CNS uniquely possess a large number of fine processes that have been considered primary sites to mediate astroglia to neuron synaptic signaling. However, localized mechanisms for regulating interactions between astroglial processes and synapses, especially for regulating the expression of functional surface proteins at these fine processes, are largely unknown. Previously, we showed that the loss of the RNA binding protein FMRP in astroglia disrupts astroglial mGluR5 signaling and reduces expression of the major astroglial glutamate transporter GLT1 and glutamate uptake in the cortex of Fmr1 conditional deletion mice. In the current study, by examining ribosome localization using electron microscopy and identifying mRNAs enriched at cortical astroglial processes using synaptoneurosome/translating ribosome affinity purification and RNA-Seq in WT and FMRP-deficient male mice, our results reveal interesting localization-dependent functional clusters of mRNAs at astroglial processes. We further showed that the lack of FMRP preferentially alters the subcellular localization and expression of process-localized mRNAs. Together, we defined the role of FMRP in altering mRNA localization and expression at astroglial processes at the postnatal development (P30-P40) and provided new candidate mRNAs that are potentially regulated by FMRP in cortical astroglia.SIGNIFICANCE STATEMENT Localized mechanisms for regulating interactions between astroglial processes and synapses, especially for regulating the expression of functional surface proteins at these fine processes, are largely unknown. Previously, we showed that the loss of the RNA binding protein FMRP in astroglia disrupts expression of several astroglial surface proteins, such as mGluR5 and major astroglial glutamate transporter GLT1 in the cortex of FMRP-deficient mice. Our current study examined ribosome localization using electron microscopy and identified mRNAs enriched at cortical astroglial processes in WT and FMRP-deficient mice. These results reveal interesting localization-dependent functional clusters of mRNAs at astroglial processes and demonstrate that the lack of FMRP preferentially alters the subcellular localization and expression of process-localized mRNAs.


Asunto(s)
Astrocitos , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Animales , Astrocitos/metabolismo , Transportador 2 de Aminoácidos Excitadores/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Masculino , Mamíferos , Ratones , Ratones Noqueados , ARN Mensajero/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo , Sinapsis/metabolismo
2.
Glia ; 69(3): 594-608, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32970902

RESUMEN

Fragile X syndrome (FXS) is one of the most common inherited intellectual disability (ID) disorders, in which the loss of FMRP protein induces a range of cellular signaling changes primarily through excess protein synthesis. Although neuron-centered molecular and cellular events underlying FXS have been characterized, how different CNS cell types are involved in typical FXS synaptic signaling changes and behavioral phenotypes is largely unknown. Recent evidence suggests that selective loss of astroglial FMRP is able to dysregulate glutamate uptake, increase spine density, and impair motor-skill learning. Here we investigated the effect of astroglial FMRP on synaptic signaling and FXS-related behavioral and learning phenotypes in astroglial Fmr1 cKO and cON mice in which FMRP expression is selectively diminished or restored in astroglia. We found that selective loss of astroglial FMRP contributes to cortical hyperexcitability by enhancing NMDAR-mediated evoked but not spontaneous miniEPSCs and elongating cortical UP state duration. Selective loss of astroglial FMRP is also sufficient to increase locomotor hyperactivity, significantly diminish social novelty preference, and induce memory acquisition and extinction deficits in astroglial Fmr1 cKO mice. Importantly, re-expression of astroglial FMRP is able to significantly rescue the hyperactivity (evoked NMDAR response, UP state duration, and open field test) and social novelty preference in astroglial Fmr1 cON mice. These results demonstrate a profound role of astroglial FMRP in the evoked synaptic signaling, spontaneously occurring cortical UP states, and FXS-related behavioral and learning phenotypes and provide important new insights in the cell type consideration for the FMRP reactivation strategy.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Síndrome del Cromosoma X Frágil , Animales , Astrocitos , Modelos Animales de Enfermedad , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Ratones , Fenotipo , Receptores de N-Metil-D-Aspartato
3.
Nat Commun ; 10(1): 4136, 2019 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-31515491

RESUMEN

Astroglia play active and diverse roles in modulating neuronal/synaptic functions in the CNS. How these astroglial functions are regulated, especially by neuronal signals, remains largely unknown. Exosomes, a major type of extracellular vesicles (EVs) that originate from endosomal intraluminal vesicles (ILVs), have emerged as a new intercellular communication process. By generating cell-type-specific ILVs/exosome reporter (CD63-GFPf/f) mice and immuno-EM/confocal image analysis, we found that neuronal CD63-GFP+ ILVs are primarily localized in soma and dendrites, but not in axonal terminals in vitro and in vivo. Secreted neuronal exosomes contain a subset of microRNAs (miRs) that is distinct from the miR profile of neurons. These miRs, especially the neuron-specific miR-124-3p, are potentially internalized into astrocytes. MiR-124-3p further up-regulates the predominant glutamate transporter GLT1 by suppressing GLT1-inhibiting miRs. Our findings suggest a previously undescribed neuronal exosomal miR-mediated genetic regulation of astrocyte functions, potentially opening a new frontier in understanding CNS intercellular communication.


Asunto(s)
Astrocitos/metabolismo , Comunicación Celular , Sistema Nervioso Central/metabolismo , Exosomas/metabolismo , Genes Reporteros , Neuronas/metabolismo , Animales , Astrocitos/ultraestructura , Transportador 2 de Aminoácidos Excitadores/metabolismo , Exosomas/ultraestructura , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Modelos Biológicos , Neuronas/ultraestructura
4.
J Cereb Blood Flow Metab ; 39(6): 1122-1137, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-29432698

RESUMEN

Ischemia not only activates cell death pathways but also triggers endogenous protective mechanisms. However, it is largely unknown what is the essence of the endogenous neuroprotective mechanisms induced by preconditioning. In this study we demonstrated that systemic injection of JZL195, a selective inhibitor of eCB clearance enzymes, induces in vivo long-term depression at CA3-CA1 synapses and at PrL-NAc synapses produces neuroprotection. JZL195-elicited long-term depression is blocked by AM281, the antagonist of cannabinoid 1 receptor (CB1R) and is abolished in mice lacking cannabinoid CB1 receptor (CB1R) in astroglial cells, but is conserved in mice lacking CB1R in glutamatergic or GABAergic neurons. Blocking the glutamate NMDA receptor and the synaptic trafficking of glutamate AMPA receptor abolishes both long-term depression and neuroprotection induced by JZL195. Mice lacking CB1R in astroglia show decreased neuronal death following cerebral ischemia. Thus, an acute elevation of extracellular eCB following eCB clearance inhibition results in neuroprotection through long-term depression induction after sequential activation of astroglial CB1R and postsynaptic glutamate receptors.


Asunto(s)
Astrocitos/metabolismo , Cannabinoides/farmacología , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Receptor Cannabinoide CB1/metabolismo , Accidente Cerebrovascular/fisiopatología , Animales , Carbamatos/farmacología , Ratones , Piperazinas/farmacología , Receptores de Glutamato/metabolismo , Roedores
5.
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
6.
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
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.
Exp Neurol ; 267: 115-22, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25779930

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a rapidly progressing neurodegenerative disease in which the majority of upper and lower motor neurons are degenerated. Despite intensive efforts to identify drug targets and develop neuroprotective strategies, effective therapeutics for ALS remains unavailable. The identification and characterization of novel targets and pathways remain crucial in the development of ALS therapeutics. Adenosine is a major neuromodulator that actively regulates synaptic transmission. Interestingly, adenosine levels are significantly elevated in the cerebrospinal fluid (CSF) of progressing human ALS patients. In the current study, we showed that adenosine 2a receptor (A2aR), but not adenosine 1 receptor (A1R), is highly enriched in spinal (motor) neurons. A2aR expression is also selectively increased at the symptomatic onset in the spinal cords of SOD1G93A mice and end-stage human ALS spinal cords. Interestingly, we found that direct adenosine treatment is sufficient to induce embryonic stem cell-derived motor neuron (ESMN) cell death in cultures. Subsequent pharmacological inhibition and partial genetic ablation of A2aR (A2aR(+/-)) significantly protect ESMN from SOD1G93A(+) astrocyte-induced cell death and delay disease progression of SOD1G93A mice. Taken together, our results provide compelling novel evidence that A2aR-mediated adenosine signaling contributes to the selective spinal motor neuron degeneration observed in the SOD1G93A mouse model of ALS.


Asunto(s)
Adenosina/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Receptor de Adenosina A2A/deficiencia , Transducción de Señal/fisiología , Adenosina/uso terapéutico , Antagonistas del Receptor de Adenosina A2/farmacología , Antagonistas del Receptor de Adenosina A2/uso terapéutico , Anciano , Anciano de 80 o más Años , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/genética , Animales , Astrocitos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Complejo Mediador/genética , Complejo Mediador/metabolismo , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/metabolismo , Fuerza Muscular/efectos de los fármacos , Fuerza Muscular/genética , Purinas/farmacología , Purinas/uso terapéutico , Receptor de Adenosina A2A/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Médula Espinal/patología , Superóxido Dismutasa/genética
9.
J Cereb Blood Flow Metab ; 35(7): 1127-36, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25757753

RESUMEN

Functional hyperemia is the regional increase in cerebral blood flow upon increases in neuronal activity which ensures that the metabolic demands of the neurons are met. Hypertension is known to impair the hyperemic response; however, the neurovascular coupling mechanisms by which this cerebrovascular dysfunction occurs have yet to be fully elucidated. To determine whether altered cortical parenchymal arteriole function or astrocyte signaling contribute to blunted neurovascular coupling in hypertension, we measured parenchymal arteriole reactivity and vascular smooth muscle cell Ca(2+) dynamics in cortical brain slices from normotensive Wistar Kyoto (WKY) and spontaneously hypertensive (SHR) rats. We found that vasoconstriction in response to the thromboxane A2 receptor agonist U46619 and basal vascular smooth muscle cell Ca(2+) oscillation frequency were significantly increased in parenchymal arterioles from SHR. In perfused and pressurized parenchymal arterioles, myogenic tone was significantly increased in SHR. Although K(+)-induced parenchymal arteriole dilations were similar in WKY and SHR, metabotropic glutamate receptor activation-induced parenchymal arteriole dilations were enhanced in SHR. Further, neuronal stimulation-evoked parenchymal arteriole dilations were similar in SHR and WKY. Our data indicate that neurovascular coupling is not impaired in SHR, at least at the level of the parenchymal arterioles.


Asunto(s)
Arteriolas/fisiopatología , Astrocitos/patología , Encéfalo/irrigación sanguínea , Hipertensión/fisiopatología , Transducción de Señal , Vasodilatación , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Animales , Arteriolas/efectos de los fármacos , Arteriolas/metabolismo , Astrocitos/metabolismo , Encéfalo/metabolismo , Encéfalo/fisiopatología , Calcio/metabolismo , Hipertensión/metabolismo , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiopatología , Potasio/metabolismo , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptores de Glutamato Metabotrópico/metabolismo , Remodelación Vascular , Vasoconstricción/efectos de los fármacos
10.
J Neurochem ; 131(5): 573-81, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25196144

RESUMEN

Glutamate transport is a critical process in the brain that maintains low extracellular levels of glutamate to allow for efficient neurotransmission and prevent excitotoxicity. Loss of glutamate transport function is implicated in epilepsy, traumatic brain injury, and amyotrophic lateral sclerosis. It remains unclear whether or not glutamate transport can be modulated in these disease conditions to improve outcome. Here, we show that sirtuin (SIRT)4, a mitochondrial sirtuin, is up-regulated in response to treatment with the potent excitotoxin kainic acid. Loss of SIRT4 leads to a more severe reaction to kainic acid and decreased glutamate transporter expression and function in the brain. Together, these results indicate a critical and novel stress response role for SIRT4 in promoting proper glutamate transport capacity and protecting against excitotoxicity.


Asunto(s)
Agonistas de Aminoácidos Excitadores/farmacología , Transportador 2 de Aminoácidos Excitadores/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ácido Glutámico/metabolismo , Ácido Kaínico/farmacología , Proteínas Mitocondriales/deficiencia , Sirtuinas/deficiencia , Animales , Biotinilación , Encéfalo/citología , Encéfalo/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Embrión de Mamíferos , Femenino , Masculino , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/ultraestructura , Convulsiones/inducido químicamente , Convulsiones/patología , Sinaptosomas/efectos de los fármacos , Sinaptosomas/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 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
13.
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.

14.
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
15.
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
16.
J Vis Exp ; (68)2012 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-23093112

RESUMEN

Proper neuron to glia interaction is critical to physiological function of the central nervous system (CNS). This bidirectional communication is sophisticatedly mediated by specific signaling pathways between neuron and glia(1,2) . Identification and characterization of these signaling pathways is essential to the understanding of how neuron to glia interaction shapes CNS physiology. Previously, neuron and glia mixed cultures have been widely utilized for testing and characterizing signaling pathways between neuron and glia. What we have learned from these preparations and other in vivo tools, however, has suggested that mutual signaling between neuron and glia often occurred in specific compartments within neurons (i.e., axon, dendrite, or soma)(3). This makes it important to develop a new culture system that allows separation of neuronal compartments and specifically examines the interaction between glia and neuronal axons/dendrites. In addition, the conventional mixed culture system is not capable of differentiating the soluble factors and direct membrane contact signals between neuron and glia. Furthermore, the large quantity of neurons and glial cells in the conventional co-culture system lacks the resolution necessary to observe the interaction between a single axon and a glial cell. In this study, we describe a novel axon and glia co-culture system with the use of a microfluidic culture platform (MCP). In this co-culture system, neurons and glial cells are cultured in two separate chambers that are connected through multiple central channels. In this microfluidic culture platform, only neuronal processes (especially axons) can enter the glial side through the central channels. In combination with powerful fluorescent protein labeling, this system allows direct examination of signaling pathways between axonal/dendritic and glial interactions, such as axon-mediated transcriptional regulation in glia, glia-mediated receptor trafficking in neuronal terminals, and glia-mediated axon growth. The narrow diameter of the chamber also significantly prohibits the flow of the neuron-enriched medium into the glial chamber, facilitating probing of the direct membrane-protein interaction between axons/dendrites and glial surfaces.


Asunto(s)
Axones/fisiología , Técnicas de Cocultivo/métodos , Técnicas Analíticas Microfluídicas/instrumentación , Técnicas Analíticas Microfluídicas/métodos , Neuroglía/fisiología , Neuronas/fisiología , Animales , Astrocitos/citología , Corteza Cerebral/citología , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Transportador 2 de Aminoácidos Excitadores/genética , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Transgénicos , Neuroglía/citología , Neuronas/citología , Regiones Promotoras Genéticas , Activación Transcripcional
17.
Am J Physiol Cell Physiol ; 299(5): C1068-78, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20844244

RESUMEN

Epoxyeicosatrienoic acids (EETs), synthesized and released by astrocytes in response to glutamate, are known to play a pivotal role in neurovascular coupling. In vascular smooth muscle cells (VSMC), EETs activate large-conductance, Ca(2+)-activated K(+) (BK) channels resulting in hyperpolarization and vasodilation. However, the functional role and mechanism of action for glial-derived EETs are still to be determined. In this study, we evaluated the effect of the synthetic EET analog 11-nonyloxy-undec-8(Z)-enoic acid (NUD-GA) on outward K(+) currents mediated by calcium-activated K(+) channels. Addition of NUD-GA significantly increased intracellular Ca(2+) and outward K(+) currents in perivascular astrocytes. NUD-GA-induced currents were significantly inhibited by BK channel blockers paxilline and tetraethylammonium (TEA) (23.4 ± 2.4%; P < 0.0005). Similarly, NUD-GA-induced currents were also significantly inhibited in the presence of the small-conductance Ca(2+)-activated K(+) channel inhibitor apamin along with a combination of blockers against glutamate receptors (12.8 ± 2.70%; P < 0.05). No changes in outward currents were observed in the presence of the channel blocker for intermediate-conductance K(+) channels TRAM-34. Blockade of the endogenous production of EETs with N-methylsulfonyl-6-(2-propargyloxyphenyl)hexanamide (MS-PPOH) significantly blunted (dl)-1-aminocyclopentane-trans-1,3-dicarboxylic acid (t-ACPD)-induced outward K(+) currents (P < 0.05; n = 6). Both NUD-GA and t-ACPD significantly increased BK channel single open probability; the later was blocked following MS-PPOH incubation. Our data supports the idea that EETs are potent K(+) channel modulators in cortical perivascular astrocytes and further suggest that these metabolites may participate in NVC by modulating the levels of K(+) released at the gliovascular space.


Asunto(s)
Astrocitos/metabolismo , Comunicación Autocrina/fisiología , Ácidos Eicosanoicos/metabolismo , Ácido Glutámico/metabolismo , Canales de Potasio/metabolismo , Transducción de Señal/fisiología , Amidas/metabolismo , Animales , Hidrocarburo de Aril Hidroxilasas/genética , Hidrocarburo de Aril Hidroxilasas/metabolismo , Astrocitos/citología , Calcio/metabolismo , Familia 2 del Citocromo P450 , Ácidos Eicosanoicos/química , Técnicas de Placa-Clamp , Pirazoles/metabolismo , Ratas , Ratas Wistar , Esteroide 16-alfa-Hidroxilasa/genética , Esteroide 16-alfa-Hidroxilasa/metabolismo
18.
J Peripher Nerv Syst ; 13(4): 289-98, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19192069

RESUMEN

The systemic inflammatory response produced by a full-thickness dermal burn injury is associated with a peripheral motor neuropathy. We previously reported that a 20% body surface area (BSA) full-thickness dermal burn in C57BL6 mice produced structural and functional deficits in motor axons at a distance from the burn site. The etiology of the neuropathy, however, is not well characterized. Burn injury leads to an increase in production of a number of proinflammatory mediators, including nitric oxide (NO). We tested the hypothesis that dermal burn-induced motor neuropathy is mediated by increased production of NO. NO synthase (NOS) activity was inhibited following a 20% BSA full-thickness burn by injection of non-specific NOS inhibitor, nitro-L-arginine methyl ester or inducible NOS (iNOS) inhibitors, L-N6-(1-iminoethyl) lysine, and aminoguanidine. NOS inhibitors also prevented the reduction in ventral roots mean axon caliber and the decrease in a motor nerve conduction velocity (MCV) following burn. iNOS knockout mice prevented MCV decrease in the first 3 days post-burn, but iNOS knockout MCV was significantly reduced at 7-14 days post-burn. These results suggest that an increase in NO production generated by systemic inflammatory response pathways after burn injury contributes to the development of structural and functional deficits in peripheral motor axons.


Asunto(s)
Quemaduras/metabolismo , Inflamación/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico/metabolismo , Enfermedades del Sistema Nervioso Periférico/metabolismo , Animales , Quemaduras/complicaciones , Inhibidores Enzimáticos/farmacología , Femenino , Inflamación/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas Motoras/patología , Conducción Nerviosa/efectos de los fármacos , Nitritos/sangre , Enfermedades del Sistema Nervioso Periférico/etiología , Enfermedades del Sistema Nervioso Periférico/patología , Piel/patología , Nervio Tibial/fisiopatología
19.
Glia ; 55(16): 1668-79, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17876807

RESUMEN

The inwardly rectifying potassium channel Kir4.1 is widely expressed by astrocytes throughout the brain. Kir4.1 channels are absent in immature, proliferating glial cells. The progressive expression of Kir4.1 correlates with astrocyte differentiation and is characterized by the establishment of a negative membrane potential (> -70 mV) and an exit from the cell cycle. Despite some correlative evidence, a mechanistic interdependence between Kir4.1 expression, membrane hyperpolarization, and control of cell proliferation has not been demonstrated. To address this question, we used astrocyte-derived tumors (glioma) that lack functional Kir4.1 channels, and generated two glioma cell lines that stably express either AcGFP-tagged Kir4.1 channels or AcGFP vectors only. Kir4.1 expression confers the same K+ conductance to glioma membranes and a similar responsiveness to changes in [K+]o that characterizes differentiated astrocytes. Kir4.1 expression was sufficient to move the resting potential of gliomas from -50 to -80 mV. Importantly, Kir4.1 expression impaired cell growth by shifting a significant number of cells from the G2/M phase into the quiescent G0/G1 stage of the cell cycle. Furthermore, these effects could be nullified entirely if Kir4.1 channels were either pharmacologically inhibited by 100 microM BaCl2 or if cells were chronically depolarized by 20 mM KCl to the membrane voltage of growth competent glioma cells. These studies therefore demonstrate directly that Kir4.1 causes a membrane hyperpolarization that is sufficient to account for the growth attenuation, which in turn induces cell maturation characterized by a shift of the cells from G2/M into G0/G1.


Asunto(s)
Neuroglía/citología , Neuroglía/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Diferenciación Celular , División Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Proliferación Celular , Células Cultivadas , Conductividad Eléctrica , Fase G1 , Fase G2 , Técnicas de Transferencia de Gen , Glioma/metabolismo , Glioma/patología , Ratones , Canales de Potasio de Rectificación Interna/genética , Ratas , Ratas Sprague-Dawley , Fase de Descanso del Ciclo Celular , Distribución Tisular
20.
Plast Reconstr Surg ; 117(1): 152-64, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16404262

RESUMEN

BACKGROUND: A full-thickness 20 percent body surface area burn in mice produces a significant decrease in tibial motor nerve conduction velocity within 6 hours of the burn and in sensory conduction velocity within 7 days. This suggests that cutaneous burn injury produces a systemic response that affects peripheral motor and sensory nerve function at a distance from the burn site. The authors tested the hypothesis that burn wound excision either 30 minutes or 3 hours after burn would prevent neuropathy. METHODS: A 20 percent body surface area third-degree burn was applied to the backs of anesthetized mice using procedures that followed National Institutes of Health guidelines. Motor nerve conduction velocity and sensory conduction velocity were determined in intact, anesthetized mice by percutaneous nerve stimulation. Burn wounds were excised and closed at 30 minutes or 3 hours after burn. Motor nerve conduction velocity and sensory conduction velocity were measured before burn and 1, 3, 7, 14, and 21 days after a burn or sham procedure. The number of circulating neutrophils and serum concentrations of tumor necrosis factor-alpha, nitrite, and electrolytes were also determined in each group. RESULTS: Motor nerve conduction velocity and sensory conduction velocity in the 30-minute excision (n = 10) and sham group (n = 5) were not significantly different. Motor nerve conduction velocity and sensory conduction velocity in the nonexcised group (n = 10) and 3-hour excision group (n = 10) were significantly decreased. Serum tumor necrosis factor-alpha concentration was elevated 6 hours after burn in nonexcised animals (n = 9) and in 3-hour excision mice (n = 8) but was not significantly different in the sham (n = 8) and the 30-minute excision group (n = 7). CONCLUSION: The authors conclude that burn wound excision at 30 minutes but not at 3 hours prevents the nerve conduction deficits measured in mice with 20 percent body surface area burns. The cellular basis of burn-induced neuropathy is unknown, but nitric oxide and tumor necrosis factor alpha-alpha appear to play a role.


Asunto(s)
Quemaduras/fisiopatología , Quemaduras/cirugía , Nervio Tibial/fisiología , Animales , Superficie Corporal , Femenino , Reflejo H/fisiología , Recuento de Leucocitos , Ratones , Ratones Endogámicos C57BL , Conducción Nerviosa , Neutrófilos , Óxido Nítrico/metabolismo , Potasio/sangre , Factores de Tiempo , Factor de Necrosis Tumoral alfa/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...