Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
1.
ACS Chem Neurosci ; 15(6): 1276-1285, 2024 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-38454572

RESUMO

Glutamate, the major excitatory neurotransmitter in the vertebrate brain, exerts its functions through the activation of specific plasma membrane receptors and transporters. Overstimulation of glutamate receptors results in neuronal cell death through a process known as excitotoxicity. A family of sodium-dependent glutamate plasma membrane transporters is responsible for the removal of glutamate from the synaptic cleft, preventing an excitotoxic insult. Glial glutamate transporters carry out more than 90% of the brain glutamate uptake activity and are responsible for glutamate recycling through the GABA/Glutamate/Glutamine shuttle. The aryl hydrocarbon receptor is a ligand-dependent transcription factor that integrates environmental clues through its ability to heterodimerize with different transcription factors. Taking into consideration the fundamental role of glial glutamate transporters in glutamatergic synapses and that these transporters are regulated at the transcriptional, translational, and localization levels in an activity-dependent fashion, in this contribution, we explored the involvement of the aryl hydrocarbon receptor, as a model of environmental integrator, in the regulation of the glial sodium-dependent glutamate/aspartate transporter. Using the model of chick cerebellar Bergmann glia cells, we report herein that the aryl hydrocarbon receptors exert a time-dependent decrease in the transporter mRNA levels and a diminution of its uptake activity. The nuclear factor kappa light chain enhancer of the activated B cell signaling pathway is involved in this regulation. Our results favor the notion of an environmentally dependent regulation of glutamate removal in glial cells and therefore strengthen the notion of the involvement of glial cells in xenobiotic neurotoxic effects.


Assuntos
Ácido Aspártico , Receptores de Hidrocarboneto Arílico , Ácido Aspártico/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Proteínas de Transporte de Glutamato da Membrana Plasmática/metabolismo , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Sódio/metabolismo , Neuroglia/metabolismo , Ácido Glutâmico/metabolismo , Células Cultivadas
2.
Neurotoxicology ; 99: 282-291, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37979659

RESUMO

Rotenone is a pesticide commonly used in agriculture that is associated with the risk of developing Parkinson's disease (PD) by inducing mitochondrial damage. As a protective cell response to different challenges, they activate mitophagy, which involves parkin activity. Parkin is an E3 ubiquitin ligase necessary in the initial steps of mitophagy, and its overexpression protects against parkinsonian effects in different models. Recent studies have reported that the aryl hydrocarbon receptor (AHR), a ligand-dependent transcription factor, induces parkin expression. Kynurenine, an endogenous AHR ligand, promotes neuroprotection in chronic neurodegenerative disorders, such as PD, although its neuroprotective mechanism needs to be fully understood. Therefore, we evaluated whether the overexpression of parkin by AHR activation with kynurenine promotes autophagy and reduces the neurotoxicity induced by rotenone in SH-SY5Y cells differentiated to dopaminergic neurons. SH-SY5Y neurons were treated with rotenone or pretreated with kynurenine or 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), and parkin levels, apoptosis, mitochondrial potential membrane, and autophagy were determined. The results showed that kynurenine and TCDD treatments induced parkin expression in an AHR-dependent manner. Kynurenine pretreatment inhibited rotenone-induced neuronal apoptosis in 17%, and the loss of mitochondrial membrane potential in 30% when compare to rotenone alone, together with a decrease in autophagy. By contrast, although TCDD treatment increased parkin levels, non-neuroprotective effects were observed. The kynurenine protective activity was AHR independent, suggesting that parkin induction might not be related to this effect. On the other hand, kynurenine treatment inhibited alpha amine-3-hydroxy-5-methyl-4-isoxazol propionic acid and N-methyl-D-aspartate receptors, which are well-known excitotoxicity mediators activated by rotenone exposure.


Assuntos
Neuroblastoma , Fármacos Neuroprotetores , Doença de Parkinson , Dibenzodioxinas Policloradas , Humanos , Rotenona , Cinurenina/farmacologia , Receptores de Hidrocarboneto Arílico , Ligantes , Morte Celular , Apoptose , Ubiquitina-Proteína Ligases/metabolismo , Linhagem Celular Tumoral , Fármacos Neuroprotetores/farmacologia
4.
Biomed Res Int ; 2023: 7389508, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36760476

RESUMO

Neurodegenerative processes encompass a large variety of diseases with different pathological patterns and clinical features, such as Alzheimer's and Parkinson's diseases. Exposure to metals has been hypothesized to increase oxidative stress in brain cells leading to cell death and neurodegeneration. Neurotoxicity of metals has been demonstrated by several in vitro and in vivo experimental studies, and most probably, each metal has its specific pathway to trigger cell death. As a result, exposure to essential metals, such as manganese, iron, copper, zinc, and cobalt, and nonessential metals, including lead, aluminum, and cadmium, perturbs metal homeostasis at the cellular and organism levels leading to neurodegeneration. In this contribution, a comprehensive review of the molecular mechanisms by which metals affect microglia physiology and signaling properties is presented. Furthermore, studies that validate the disruption of microglia activation pathways as an essential mechanism of metal toxicity that can contribute to neurodegenerative disease are also presented and discussed.


Assuntos
Doenças Neurodegenerativas , Humanos , Doenças Neurodegenerativas/metabolismo , Microglia/metabolismo , Metais/toxicidade , Ferro/toxicidade , Zinco
7.
Life (Basel) ; 13(1)2022 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-36675972

RESUMO

Spinocerebellar ataxia type 7 (SCA7) is a neurodegenerative disorder characterized by cerebellar ataxia and retinopathy. SCA7 is caused by a CAG expansion in the ATXN7 gene, which results in an extended polyglutamine (polyQ) tract in the encoded protein, the ataxin-7. PolyQ expanded ataxin-7 elicits neurodegeneration in cerebellar Purkinje cells, however, its impact on the SCA7-associated retinopathy remains to be addressed. Since Müller glial cells play an essential role in retinal homeostasis, we generate an inducible model for SCA7, based on the glial Müller MIO-M1 cell line. The SCA7 pathogenesis has been explained by a protein gain-of-function mechanism, however, the contribution of the mutant RNA to the disease cannot be excluded. In this direction, we found nuclear and cytoplasmic foci containing mutant RNA accompanied by subtle alternative splicing defects in MIO-M1 cells. RNA foci were also observed in cells from different lineages, including peripheral mononuclear leukocytes derived from SCA7 patient, suggesting that this molecular mark could be used as a blood biomarker for SCA7. Collectively, our data showed that our glial cell model exhibits the molecular features of SCA7, which makes it a suitable model to study the RNA toxicity mechanisms, as well as to explore therapeutic strategies aiming to alleviate glial dysfunction.

8.
Neurotox Res ; 39(6): 2154-2174, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34677787

RESUMO

The blood-brain barrier is a dynamic structure, collectively referred to as the neurovascular unit. It is responsible for the exchange of blood, oxygen, ions, and other molecules between the peripheral circulation and the brain compartment. It is the main entrance to the central nervous system and as such critical for the maintenance of its homeostasis. Dysfunction of the blood-brain barrier is a characteristic of several neurovascular pathologies. Moreover, physiological changes, environmental factors, nutritional habits, and psychological stress can modulate the tightness of the barrier. In this contribution, we summarize our current understanding of structure and function of this important component of the brain. We also describe the neurological deficits associated with its damage. A special emphasis is placed in the effect of the exposure to xenobiotics and pollutants in the permeability of the barrier. Finally, current protective strategies as well as the culture models to study this fascinating structure are discussed.


Assuntos
Barreira Hematoencefálica/fisiologia , Encéfalo/fisiologia , Animais , Barreira Hematoencefálica/anatomia & histologia , Barreira Hematoencefálica/metabolismo , Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Humanos
9.
Biomed Res Int ; 2021: 1596185, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34660781

RESUMO

Over the years, most of the research concerning manganese exposure was restricted to the toxicity of neuronal cells. Manganese is an essential trace element that in high doses exerts neurotoxic effects. However, in the last two decades, efforts have shifted toward a more comprehensive approach that takes into account the involvement of glial cells in the development of neurotoxicity as a brain insult. Glial cells provide structural, trophic, and metabolic support to neurons. Nevertheless, these cells play an active role in adult neurogenesis, regulation of synaptogenesis, and synaptic plasticity. Disturbances in glial cell function can lead to neurological disorders, including neurodegenerative diseases. This review highlights the pivotal role that glial cells have in manganese-induced neurotoxicity as well as the most sounding mechanisms involved in the development of this phenomenon.


Assuntos
Manganês/toxicidade , Neuroglia/patologia , Neurotoxinas/toxicidade , Animais , Humanos , Modelos Biológicos , Neuroglia/efeitos dos fármacos , Síndromes Neurotóxicas/patologia , Estresse Oxidativo/efeitos dos fármacos
10.
Neuropharmacology ; 192: 108602, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33991564

RESUMO

Glutamate is the major excitatory neurotransmitter in the vertebrate central nervous system. Once released, it binds to specific membrane receptors and transporters activating a wide variety of signal transduction cascades, as well as its removal from the synaptic cleft in order to avoid its extracellular accumulation and the overstimulation of extra-synaptic receptors that might result in neuronal death through a process known as excitotoxicity. Although neurodegenerative diseases are heterogenous in clinical phenotypes and genetic etiologies, a fundamental mechanism involved in neuronal degeneration is excitotoxicity. Glutamate homeostasis is critical for brain physiology and Glutamate transporters are key players in maintaining low extracellular Glutamate levels. Therefore, the characterization of Glutamate transporters has been an active area of glutamatergic research for the last 40 years. Transporter activity its regulated at different levels: transcriptional and translational control, transporter protein trafficking and membrane mobility, and through extensive post-translational modifications. The elucidation of these mechanisms has emerged as an important piece to shape our current understanding of glutamate actions in the nervous system.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/química , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Ácido Glutâmico/metabolismo , Transmissão Sináptica/fisiologia , Sistema X-AG de Transporte de Aminoácidos/genética , Animais , Transportador 1 de Aminoácido Excitatório/química , Transportador 1 de Aminoácido Excitatório/genética , Transportador 1 de Aminoácido Excitatório/metabolismo , Transportador 2 de Aminoácido Excitatório/química , Transportador 2 de Aminoácido Excitatório/genética , Transportador 2 de Aminoácido Excitatório/metabolismo , Humanos , Processamento de Proteína Pós-Traducional/fisiologia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
11.
Biomolecules ; 11(2)2021 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-33530452

RESUMO

Myotonic dystrophy type 1 (DM1), the most frequent inherited muscular dystrophy in adults, is caused by the CTG repeat expansion in the 3'UTR of the DMPK gene. Mutant DMPK RNA accumulates in nuclear foci altering diverse cellular functions including alternative splicing regulation. DM1 is a multisystemic condition, with debilitating central nervous system alterations. Although a defective neuroglia communication has been described as a contributor of the brain pathology in DM1, the specific cellular and molecular events potentially affected in glia cells have not been totally recognized. Thus, to study the effects of DM1 mutation on glial physiology, in this work, we have established an inducible DM1 model derived from the MIO-M1 cell line expressing 648 CUG repeats. This new model recreated the molecular hallmarks of DM1 elicited by a toxic RNA gain-of-function mechanism: accumulation of RNA foci colocalized with MBNL proteins and dysregulation of alternative splicing. By applying a microarray whole-transcriptome approach, we identified several gene changes associated with DM1 mutation in MIO-M1 cells, including the immune mediators CXCL10, CCL5, CXCL8, TNFAIP3, and TNFRSF9, as well as the microRNAs miR-222, miR-448, among others, as potential regulators. A gene ontology enrichment analyses revealed that inflammation and immune response emerged as major cellular deregulated processes in the MIO-M1 DM1 cells. Our findings indicate the involvement of an altered immune response in glia cells, opening new windows for the study of glia as potential contributor of the CNS symptoms in DM1.


Assuntos
Mutação , Distrofia Miotônica/metabolismo , Miotonina Proteína Quinase/genética , Neuroglia/metabolismo , Transcriptoma , Regiões 3' não Traduzidas , Processamento Alternativo , Linhagem Celular , Núcleo Celular/metabolismo , Sistema Nervoso Central/metabolismo , Éxons , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Genótipo , Humanos , Sistema Imunitário , Inflamação , Distrofia Miotônica/genética , Análise de Sequência com Séries de Oligonucleotídeos , RNA/metabolismo , Expansão das Repetições de Trinucleotídeos
13.
Neurotox Res ; 38(3): 765-774, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32734566

RESUMO

Fluoride, a pollutant present in contaminated ground water, oral care products, food, and pesticides, has deleterious effects in the structure and function of the central nervous system. Among the established neurological defects described in the exposed population, a reduced score in intelligence quotient tests in children of contaminated areas has gained attention over the past years. Maternal fluoride exposure during gestation decreases learning and memory abilities that correlate with a significant diminution of glutamate receptors expression. Since the involvement of glia cells in the maintenance and regulation of glutamatergic synapses is well-documented, in this contribution, we characterized the effect of fluoride exposure in the regulation of glia glutamine transporters. To this end, we used the Müller glia cell line, Mio-M1, and through the use of [3H]L-Glutamine uptake experiments and a Western blot approach, we demonstrate here the functional expression of system N of glutamine transporters, SNAT3 and SNAT5, in this model of human retina radial glia cells. Furthermore, these transporters interact with the glutamate transporter excitatory amino acid transporter 1, in an activity-dependent manner. Fluoride treatment reduces glutamine uptake and cell membrane [3H]glutamine surface binding, in good correlation with a decrease in SNAT3 and 5 protein levels. These results demonstrate that glia cells respond to the presence of fluoride reducing glutamine mobilization and by these means decreases glutamate turnover suggesting a disruption of glutamatergic transmission.


Assuntos
Fluoretos/farmacologia , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Neuroglia/efeitos dos fármacos , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Transporte Biológico/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Células Ependimogliais/efeitos dos fármacos , Fluoretos/metabolismo , Humanos , Receptores de Glutamato/metabolismo , Sinapses/metabolismo
14.
Front Neurosci ; 14: 121, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32132895

RESUMO

The nervous system (NS) of invertebrates and vertebrates is composed of two main types of cells: neurons and glia. In both types of organisms, nerve cells have similarities in biochemistry and functionality. The neurons are in charge of the synapse, and the glial cells are in charge of important functions of neuronal and homeostatic modulation. Knowing the mechanisms by which NS cells work is important in the biomedical area for the diagnosis and treatment of neurological disorders. For this reason, cellular and animal models to study the properties and characteristics of the NS are always sought. Marine invertebrates are strategic study models for the biological sciences. The sea slug Aplysia californica and the squid Loligo pealei are two examples of marine key organisms in the neurosciences field. The principal characteristic of marine invertebrates is that they have a simpler NS that consists of few and larger cells, which are well organized and have accessible structures. As well, the close phylogenetic relationship between Chordata and Echinodermata constitutes an additional advantage to use these organisms as a model for the functionality of neuronal cells and their cellular plasticity. Currently, there is great interest in analyzing the signaling processes between neurons and glial cells, both in vertebrates and in invertebrates. However, only few types of glial cells of invertebrates, mostly insects, have been studied, and it is important to consider marine organisms' research. For this reason, the objective of the review is to present an update of the most relevant information that exists around the physiology of marine invertebrate neuronal and glial cells.

15.
Neurotox Res ; 37(2): 366-379, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31292883

RESUMO

Attractive due to an alleged high biocompatibility, silica nanoparticles have been widely used in the field of nanomedicine; however, their proven capacity to induce the synthesis and release of pro-inflammatory cytokines in several cellular models has raised concern about their safety. Glutamate, the main excitatory amino acid transmitter triggers a wide variety of signal transduction cascades that regulate protein synthesis at transcriptional and translational levels. A stimulus-dependent dynamic change in the protein repertoire in neurons and glia cells is the molecular framework of higher brain functions. Within the cerebellum, Bergmann glia cells are the most abundant non-neuronal cells and span the entire molecular layer of the cerebellar cortex, wrapping the synapses in this structure. Taking into consideration the functional role of Bergmann glia in terms of the recycling of glutamate, lactate supply to neurons, and prevention of neurotoxic insults, we decided to investigate the possibility that silica nanoparticles affect Bergmann glia and by these means alter the major excitatory neurotransmitter system in the brain. To this end, we exposed cultured chick cerebellar Bergmann glia cells to silica nanoparticles and measured [35S]-methionine incorporation into newly synthesized polypeptides. Our results demonstrate that exposure of the cultured cells to silica nanoparticles exerts a time- and dose-dependent modulation of protein synthesis. Furthermore, altered patterns of eukaryotic initiation factor 2 alpha and eukaryotic elongation factor 2 phosphorylation were present upon nanoparticle exposure. These results demonstrate that glia cells respond to the presence of this nanomaterial modifying their proteome, presumably in an effort to overcome any plausible neurotoxic effect.


Assuntos
Nanopartículas/efeitos adversos , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Dióxido de Silício/efeitos adversos , Animais , Embrião de Galinha , Relação Dose-Resposta a Droga , Quinase do Fator 2 de Elongação/metabolismo , Fator de Iniciação 2 em Eucariotos/metabolismo , Metionina/metabolismo , Fosforilação , Cultura Primária de Células , Radioisótopos de Enxofre/metabolismo , Fatores de Tempo
16.
Neurochem Res ; 45(6): 1365-1374, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31363896

RESUMO

Glutamate is the major excitatory amino acid neurotransmitter in the vertebrate brain. It exerts its actions through the activation of specific plasma membrane receptors expressed in neurons and glial cells. Overactivation of glutamate receptors results in neuronal death, known as excitotoxicity. A family of sodium-dependent glutamate transporters enriched in glial cells are responsible of the vast majority of the removal of this amino acid form the synaptic cleft. Therefore, a precise and exquisite regulation of these proteins is required not only for a proper glutamatergic transmission but also for the prevention of an excitotoxic insult. Manganese is a trace element essential as a cofactor for several enzymatic systems, although in high concentrations is involved in the disruption of brain glutamate homeostasis. The molecular mechanisms associated to manganese neurotoxicity have been focused on mitochondrial function, although energy depletion severely compromises the glutamate uptake process. In this context, in this contribution we analyze the effect of manganese exposure in glial glutamate transporters function. To this end, we used the well-established model of chick cerebellar Bergmann glia cultures. A time and dose dependent modulation of [3H]-D-aspartate uptake was found. An increase in the transporter catalytic efficiency, most probably linked to a discrete increase in the affinity of the transporter was detected upon manganese exposure. Interestingly, glucose uptake was reduced by this metal. These results favor the notion of a direct effect of manganese on glial cells, this in turn alters their coupling with neurons and might lead to changes in glutamatergic transmission.


Assuntos
Transportador 1 de Aminoácido Excitatório/metabolismo , Manganês/administração & dosagem , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Animais , Ácido Aspártico/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Embrião de Galinha , Relação Dose-Resposta a Droga
17.
Front Neurosci ; 14: 613225, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33488353

RESUMO

Glutamate is the main excitatory amino acid acting at the level of pre and postsynaptic neurons, as well as in glial cells. It is involved in the coordinated modulation of energy metabolism, glutamine synthesis, and ammonia detoxification. The relationship between the functional status of liver and brain has been known for many years. The most widely recognized aspect of this relation is the brain dysfunction caused by acute liver injury that manifests a wide spectrum of neurologic and psychiatric abnormalities. Inflammation, circulating neurotoxins, and impaired neurotransmission have been reported in this pathophysiology. In the present contribution, we report the effect of a hepatotoxic compound like CCl4 on the expression of key proteins involved in glutamate uptake and metabolism as glutamate transporters and glutamine synthetase in mice liver, brain, and cerebellum. Our findings highlight a differential expression pattern of glutamate transporters in cerebellum. A significant Purkinje cells loss, in parallel to an up-regulation of glutamine synthetase, and astrogliosis in the brain have also been noticed. In the intoxicated liver, glutamate transporter 1 expression is up-regulated, in contrast to glutamine synthetase which is reduced in a time-dependent manner. Taken together our results demonstrate that the exposure to an acute CCl4 insult, leads to the disruption of glutamate transporters expression in the liver-brain axis and therefore a severe alteration in glutamate-mediated neurotransmission might be present in the central nervous system.

18.
Aging Cell ; 18(5): e13002, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31305018

RESUMO

The study of Hutchinson-Gilford progeria syndrome (HGPS) has provided important clues to decipher mechanisms underlying aging. Progerin, a mutant lamin A, disrupts nuclear envelope structure/function, with further impairment of multiple processes that culminate in senescence. Here, we demonstrate that the nuclear protein export pathway is exacerbated in HGPS, due to progerin-driven overexpression of CRM1, thereby disturbing nucleocytoplasmic partitioning of CRM1-target proteins. Enhanced nuclear export is central in HGPS, since pharmacological inhibition of CRM1 alleviates all aging hallmarks analyzed, including senescent cellular morphology, lamin B1 downregulation, loss of heterochromatin, nuclear morphology defects, and expanded nucleoli. Exogenous overexpression of CRM1 on the other hand recapitulates the HGPS cellular phenotype in normal fibroblasts. CRM1 levels/activity increases with age in fibroblasts from healthy donors, indicating that altered nuclear export is a common hallmark of pathological and physiological aging. Collectively, our findings provide novel insights into HGPS pathophysiology, identifying CRM1 as potential therapeutic target in HGPS.


Assuntos
Senilidade Prematura/metabolismo , Núcleo Celular/metabolismo , Senescência Celular , Carioferinas/metabolismo , Proteínas Nucleares/metabolismo , Progéria/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transporte Ativo do Núcleo Celular , Senilidade Prematura/patologia , Células Cultivadas , Humanos , Fenótipo , Progéria/patologia , Proteína Exportina 1
19.
ACS Chem Neurosci ; 10(6): 2668-2675, 2019 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-31091406

RESUMO

Glutamate exerts its actions through the activation of membrane receptors expressed in neurons and glia cells. The signaling properties of glutamate transporters have been characterized recently, suggesting a complex array of signaling transactions triggered by presynaptic released glutamate. In the cerebellar molecular layer, glutamatergic synapses are surrounded by Bergmann glia cells, compulsory participants of glutamate turnover and supply to neurons. Since a glutamate-dependent increase in cGMP levels has been described in these cells and the nitric oxide-cGMP signaling cascade increases their glutamate uptake activity, we describe here the Bergmann glia expression of neuronal nitric oxide synthetase. An augmentation of neuronal nitric oxide synthase was found upon glutamate exposure. This effect is mediated by glutamate transporters and is related to an increase in the stability of the enzyme. These results strengthen the notion of a complex regulation of glial glutamate uptake that supports neuronal glutamate signaling.


Assuntos
Cerebelo/metabolismo , Ácido Glutâmico/metabolismo , Neuroglia/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Animais , Células Cultivadas , Embrião de Galinha , Transdução de Sinais/fisiologia
20.
Cell Death Dis ; 10(3): 196, 2019 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-30814495

RESUMO

ß-dystroglycan (ß-DG) is a key component of multiprotein complexes in the plasma membrane and nuclear envelope. In addition, ß-DG undergoes two successive proteolytic cleavages that result in the liberation of its intracellular domain (ICD) into the cytosol and nucleus. However, stimuli-inducing ICD cleavage and the physiological relevance of this proteolytic fragment are largely unknown. In this study we show for the first time that ß-DG ICD is targeted to the nucleolus where it interacts with the nuclear proteins B23 and UBF (central factor of Pol I-mediated rRNA gene transcription) and binds to rDNA promoter regions. Interestingly DG silencing results in reduced B23 and UBF levels and aberrant nucleolar morphology. Furthermore, ß-DG ICD cleavage is induced by different nucleolar stressors, including oxidative stress, acidosis, and UV irradiation, which implies its participation in the response to nucleolar stress. Consistent with this idea, overexpression of ß-DG elicited mislocalization and decreased levels of UBF and suppression of rRNA expression, which in turn provoked altered ribosome profiling and decreased cell growth. Collectively our data reveal that ß-DG ICD acts as negative regulator of rDNA transcription by impeding the transcriptional activity of UBF, as a part of the protective mechanism activated in response to nucleolar stress.


Assuntos
Nucléolo Celular/metabolismo , Distroglicanas/metabolismo , Proteínas Pol1 do Complexo de Iniciação de Transcrição/metabolismo , RNA Ribossômico/biossíntese , Animais , Proliferação de Células/genética , Citoplasma/metabolismo , DNA Ribossômico/genética , DNA Ribossômico/metabolismo , Distroglicanas/antagonistas & inibidores , Distroglicanas/genética , Camundongos , Mioblastos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleofosmina , Estresse Oxidativo , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética , Domínios Proteicos/genética , RNA Ribossômico/genética , Ribossomos/metabolismo , Transcrição Gênica , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA