Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cerebellum ; 22(1): 102-119, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35040097

RESUMO

Selective neuronal vulnerability is common to most degenerative disorders, including Niemann-Pick C (NPC), a rare genetic disease with altered intracellular trafficking of cholesterol. Purkinje cell dysfunction and loss are responsible for cerebellar ataxia, which is among the prevailing neurological signs of the NPC disease. In this review, we focus on some questions that are still unresolved. First, we frame the cerebellar vulnerability in the context of the extended postnatal time length by which the development of this structure is completed in mammals. In line with this thought, the much later development of cerebellar symptoms in humans is due to the later development and/or maturation of the cerebellum. Hence, the occurrence of developmental events under a protracted condition of defective intracellular cholesterol mobilization hits the functional maturation of the various cell types generating the ground of increased vulnerability. This is particularly consistent with the high cholesterol demand required for cell proliferation, migration, differentiation, and synapse formation/remodeling. Other major questions we address are why the progression of Purkinje cells loss is always from the anterior to the posterior lobes and why cerebellar defects persist in the mouse model even when genetic manipulations can lead to nearly normal survival.


Assuntos
Doença de Niemann-Pick Tipo C , Masculino , Camundongos , Animais , Humanos , Doença de Niemann-Pick Tipo C/genética , Cerebelo/metabolismo , Células de Purkinje/metabolismo , Neurônios/metabolismo , Colesterol/metabolismo , Mamíferos
2.
J Cell Physiol ; 237(12): 4563-4579, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36322609

RESUMO

The loss of NPC1 or NPC2 function results in cholesterol and sphingolipid dyshomeostasis that impairs developmental trajectories, predisposing the postnatal brain to the appearance of pathological signs, including progressive and stereotyped Purkinje cell loss and microgliosis. Despite increasing evidence reporting the activation of pro-inflammatory microglia as a cardinal event of NPC1 disease progression at symptomatic stages both in patients and preclinical models, how microglia cells respond to altered neurodevelopmental dynamics remains not completely understood. To gain an insight on this issue, we have characterized patterns of microglia activation in the early postnatal cerebellum and young adult olfactory bulb of the hypomorphic Npc1nmf164 mouse model. Previous evidence has shown that both these areas display a number of anomalies affecting neuron and glial cell proliferation and differentiation, which largely anticipate cellular changes and clinical signs, raising our interest on how microglia interplay to these changes. Even so, to separate the contribution of cues provided by the dysfunctional microenvironment we have also studied microglia isolated from mice of increasing ages and cultured in vitro for 1 week. Our findings show that microglia of both cerebellum and olfactory bulb of Npc1nmf164 mice adopt an activated phenotype, characterized by increased cell proliferation, enlarged soma size and de-ramified processes, as well as a robust phagocytic activity, in a time- and space-specific manner. Enhanced phagocytosis associates with a profound remodeling of gene expression signatures towards gene products involved in chemotaxis, cell recognition and engulfment, including Cd68 and Trem2. These early changes in microglia morphology and activities are induced by region-specific developmental anomalies that likely anticipate alterations in neuronal connectivity. As a proof of concept, we show that microglia activation within the granule cell layer and glomerular layer of the olfactory bulb of Npc1nmf164 mice is associated with shortfalls in fine odor discrimination.


Assuntos
Microglia , Doença de Niemann-Pick Tipo C , Percepção Olfatória , Animais , Camundongos , Encéfalo/metabolismo , Modelos Animais de Doenças , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Glicoproteínas de Membrana/metabolismo , Microglia/metabolismo , Proteína C1 de Niemann-Pick/metabolismo , Doença de Niemann-Pick Tipo C/metabolismo , Odorantes , Receptores Imunológicos/metabolismo , Fagócitos/metabolismo
3.
Neurobiol Dis ; 163: 105606, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34974125

RESUMO

Brain-derived neurotrophic factor (BDNF) is one of the most studied neurotrophins in the mammalian brain, essential not only to the development of the central nervous system but also to synaptic plasticity. BDNF is present in various brain areas, but highest levels of expression are seen in the cerebellum and hippocampus. After birth, BDNF acts in the cerebellum as a mitogenic and chemotactic factor, stimulating the cerebellar granule cell precursors to proliferate, migrate and maturate, while in the hippocampus BDNF plays a fundamental role in synaptic transmission and plasticity, representing a key regulator for the long-term potentiation, learning and memory. Furthermore, the expression of BDNF is highly regulated and changes of its expression are associated with both physiological and pathological conditions. The purpose of this review is to provide an overview of the current state of knowledge on the BDNF biology and its neurotrophic role in the proper development and functioning of neurons and synapses in two important brain areas of postnatal neurogenesis, the cerebellum and hippocampus. Dysregulation of BDNF expression and signaling, resulting in alterations in neuronal maturation and plasticity in both systems, is a common hallmark of several neurodevelopmental diseases, such as autism spectrum disorder, suggesting that neuronal malfunction present in these disorders is the result of excessive or reduced of BDNF support. We believe that the more the relevance of the pathophysiological actions of BDNF, and its downstream signals, in early postnatal development will be highlighted, the more likely it is that new neuroprotective therapeutic strategies will be identified in the treatment of various neurodevelopmental disorders.


Assuntos
Transtorno do Espectro Autista/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Cerebelo/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Animais , Humanos , Neurogênese/fisiologia , Sinapses/metabolismo
4.
J Cell Physiol ; 235(4): 3393-3401, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31552693

RESUMO

Activation of Akt/Protein Kinase B (PKB) by phosphatidylinositol-3-kinase (PI3K) controls several cellular functions largely studied in mammalian cells, including preimplantation embryos. We previously showed that early mouse embryos inherit active Akt from oocytes and that the intracellular localization of this enzyme at the two-cell stage depends on the T-cell leukemia/lymphoma 1 oncogenic protein, Tcl1. We have now investigated whether Akt isoforms, namely Akt1, Akt2 and Akt3, exert a specific role in blastomere proliferation during preimplantation embryo development. We show that, in contrast to other Akt family members, Akt2 enters male and female pronuclei of mouse preimplantation embryos at the late one-cell stage and thereafter maintains a nuclear localization during later embryo cleavage stages. Depleting one-cell embryos of single Akt family members by microinjecting Akt isoform-specific antibodies into wild-type zygotes, we observed that: (a) Akt2 is necessary for normal embryo progression through cleavage stages; and (b) the specific nuclear targeting of Akt2 in two-cell embryos depends on Tcl1. Our results indicate that preimplantation mouse embryos have a peculiar regulation of blastomere proliferation based on the activity of the Akt/PKB family member Akt2, which is mediated by the oncogenic protein Tcl1. Both Akt2 and Tcl1 are essential for early blastomere proliferation and embryo development.


Assuntos
Desenvolvimento Embrionário/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas/genética , Animais , Blastocisto/metabolismo , Blastômeros/metabolismo , Proliferação de Células/genética , Embrião de Mamíferos , Feminino , Masculino , Camundongos , Gravidez , Isoformas de Proteínas
5.
Hum Mol Genet ; 27(R2): R119-R129, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29718288

RESUMO

There is growing evidence that the complex clinical manifestations of lysosomal storage diseases (LSDs) are not fully explained by the engorgement of the endosomal-autophagic-lysosomal system. In this review, we explore current knowledge of common pathogenetic mechanisms responsible for the early onset of tissue abnormalities of two LSDs, Mucopolysaccharidosis type II (MPSII) and Niemann-Pick type C (NPC) diseases. In particular, perturbations of the homeostasis of glycosaminoglycans (GAGs) and cholesterol (Chol) in MPSII and NPC diseases, respectively, affect key biological processes, including morphogen signaling. Both GAGs and Chol finely regulate the release, reception and tissue distribution of Shh. Hence, not surprisingly, developmental processes depending on correct Shh signaling have been found altered in both diseases. Besides abnormal signaling, exaggerated activation of microglia and impairment of autophagy and mitophagy occur in both diseases, largely before the appearance of typical pathological signs.


Assuntos
Doenças por Armazenamento dos Lisossomos/fisiopatologia , Lisossomos/patologia , Animais , Autofagia , Colesterol/metabolismo , Endocitose , Endossomos/patologia , Glicosaminoglicanos/metabolismo , Proteínas Hedgehog/fisiologia , Homeostase , Humanos , Doenças por Armazenamento dos Lisossomos/metabolismo , Lisossomos/fisiologia , Mitofagia , Mucopolissacaridose II/patologia , Neuroimunomodulação/imunologia , Neuroimunomodulação/fisiologia , Doença de Niemann-Pick Tipo C/patologia , Via de Sinalização Wnt/fisiologia
6.
J Cell Physiol ; 234(10): 18349-18360, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30912127

RESUMO

Proteins belonging to the TGFß-stimulated clone 22 domain (TSC22D) family display a repertoire of activities, regulating cell proliferation and differentiation. The tumor suppressor activity of the first identified member of the family, TSC22D1 (formerly named TSC-22), has been extensively studied, but afterward a longer isoform encoded by the same gene turned out to play an opposite role. We have previously characterized the role of TSC22D1 and TSC22D4 in cell differentiation using granule neurons (GNs) isolated from the mouse cerebellum. However, the possibility to study the role of these factors in cell proliferation was limited by the fact that GNs readily exit from the cell-cycle and differentiate upon isolation and in vitro culture. To overcome this limitation, we have now exploited DAOY medulloblastoma cells, which are ontogenetically similar to cerebellar GNs and can be efficiently transfected with interfering RNA for gene knockdown purposes. Our findings indicate that TSC22D4-TSC22D1 short isoform heterodimers are involved in the escape from cell proliferation and exit from the cell-cycle, whereas, the TSC22D1 long isoform is required for cell proliferation, acting independently from TSC22D4. We also show that the silencing of specific expression of TSC22D4 or TSC22D1 isoforms affects the cell-cycle progression. These findings add a novel insight on the function of TSC22D proteins, with particular reference to the tumor suppressor activity of the TSC22D1 short isoform, which is re-framed within the context of a functional interplay with TSC22D4 and the mutually exclusive expression with the TSC22D1 long isoform.


Assuntos
Ciclo Celular/fisiologia , Neoplasias Cerebelares/metabolismo , Meduloblastoma/metabolismo , Domínios Proteicos/fisiologia , Isoformas de Proteínas/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Humanos , Camundongos , Neurônios/metabolismo
7.
Neurobiol Dis ; 130: 104531, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31302243

RESUMO

The dyshomeostasis of intracellular cholesterol trafficking is typical of the Niemann-Pick type C (NPC) disease, a fatal inherited lysosomal storage disorder presenting with progressive neurodegeneration and visceral organ involvement. In light of the well-established relevance of cholesterol in regulating the endocannabinoid (eCB) system expression and activity, this study was aimed at elucidating whether NPC disease-related cholesterol dyshomeostasis affects the functional status of the brain eCB system. To this end, we exploited a murine model of NPC deficiency for determining changes in the expression and activity of the major molecular components of the eCB signaling, including cannabinoid type-1 and type-2 (CB1 and CB2) receptors, their ligands, N-arachidonoylethanolamine (AEA) and 2-arachidonoylglycerol (2-AG), along with their main synthesizing/inactivating enzymes. We found a robust alteration of distinct components of the eCB system in various brain regions, including the cortex, hippocampus, striatum and cerebellum, of Npc1-deficient compared to wild-type pre-symptomatic mice. Changes of the eCB component expression and activity differ from one brain structure to another, although 2-AG and AEA are consistently found to decrease and increase in each structure, respectively. The thorough biochemical characterization of the eCB system was accompanied by a behavioral characterization of Npc1-deficient mice using a number of paradigms evaluating anxiety, locomotor activity, spatial learning/memory abilities, and coping response to stressful experience. Our findings provide the first description of an early and region-specific alteration of the brain eCB system in NPC and suggest that defective eCB signaling could contribute at producing and/or worsening the neurological symptoms of this disorder.


Assuntos
Encéfalo/metabolismo , Colesterol/metabolismo , Endocanabinoides/metabolismo , Homeostase/fisiologia , Doença de Niemann-Pick Tipo C/metabolismo , Animais , Modelos Animais de Doenças , Camundongos , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/metabolismo
8.
Hum Mol Genet ; 26(12): 2277-2289, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28379564

RESUMO

The Niemann-Pick type C1 (NPC1) disease is a neurodegenerative lysosomal storage disorder due to mutations in the NPC1 gene, encoding a transmembrane protein related to the Sonic hedgehog (Shh) receptor, Patched, and involved in intracellular trafficking of cholesterol. We have recently found that the proliferation of cerebellar granule neuron precursors is significantly reduced in Npc1-/- mice due to the downregulation of Shh expression. This finding prompted us to analyze the formation of the primary cilium, a non-motile organelle that is specialized for Shh signal transduction and responsible, when defective, for several human genetic disorders. In this study, we show that the expression and subcellular localization of Shh effectors and ciliary proteins are severely disturbed in Npc1-deficient mice. The dysregulation of Shh signaling is associated with a shortening of the primary cilium length and with a reduction of the fraction of ciliated cells in Npc1-deficient mouse brains and the human fibroblasts of NPC1 patients. These defects are prevented by treatment with 2-hydroxypropyl-ß-cyclodextrin, a promising therapy currently under clinical investigation. Our findings indicate that defective Shh signaling is responsible for abnormal morphogenesis of the cerebellum of Npc1-deficient mice and show, for the first time, that the formation of the primary cilium is altered in NPC1 disease.


Assuntos
Cílios/metabolismo , Doença de Niemann-Pick Tipo C/metabolismo , 2-Hidroxipropil-beta-Ciclodextrina , Animais , Proteínas de Transporte/genética , Cerebelo/metabolismo , Colesterol/metabolismo , Fibroblastos/metabolismo , Proteínas Hedgehog/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Neurônios/metabolismo , Doença de Niemann-Pick Tipo C/patologia , Proteínas/genética , Transdução de Sinais , beta-Ciclodextrinas/metabolismo
9.
J Assist Reprod Genet ; 31(4): 463-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24526355

RESUMO

OBJECTIVE: Myo-inositol (myoIns) has a positive role in mammalian development and human reproduction. Since experiments on farming species suggest a similar role in preimplantation development, we evaluated the hypothesis that the inclusion of myoIns in human embryo culture media would produce an increase in embryo quality in IVF cycles, using the mouse embryo assay. METHODS: To determine the effect of myoIns on completion of preimplantation development in vitro, one-cell embryos of the inbred C57BL/6N mouse strain were produced by ICSI, cultured in human fertilization media in the presence of myoIns (myoIns+) or in its absence (myoIns-) and evaluated morphologically. Daily progression through cleavage stages, blastocyst production and expansion and blastomere number at 96 hours post fertilization were assessed. RESULTS: Compared to myoIns- embryos, myoIns+ embryos displayed a faster cleavage rate and by the end of preimplantation development, the majority of myoIns+ blastocysts was expanded and formed by a higher number of blastomeres. CONCLUSION: The presence of myoIns resulted in both an increase in proliferation activity and developmental rate of in vitro cultured early mouse embryos, representing a substantial improvement of culture conditions. These data may identify myoIns as an important supplement for human embryo preimplantation culture.


Assuntos
Blastocisto/efeitos dos fármacos , Desenvolvimento Embrionário/efeitos dos fármacos , Inositol/farmacologia , Animais , Blastocisto/citologia , Proliferação de Células/efeitos dos fármacos , Meios de Cultura , Técnicas de Cultura Embrionária , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Injeções de Esperma Intracitoplásmicas
10.
Lancet ; 390(10104): 1720-1721, 2017 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-28803711
11.
Development ; 137(20): 3393-403, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20826531

RESUMO

Disruption of mouse Prep1, which codes for a homeodomain transcription factor, leads to embryonic lethality during post-implantation stages. Prep1(-/-) embryos stop developing after implantation and before anterior visceral endoderm (AVE) formation. In Prep1(-/-) embryos at E6.5 (onset of gastrulation), the AVE is absent and the proliferating extra-embryonic ectoderm and epiblast, marked by Bmp4 and Oct4, respectively, are reduced in size. At E.7.5, Prep1(-/-) embryos are small and very delayed, showing no evidence of primitive streak or of differentiated embryonic lineages. Bmp4 is expressed residually, while the reduced number of Oct4-positive cells is constant up to E8.5. At E6.5, Prep1(-/-) embryos retain a normal mitotic index but show a major increase in cleaved caspase 3 and TUNEL staining, indicating apoptosis. Therefore, the mouse embryo requires Prep1 when undergoing maximal expansion in cell number. Indeed, the phenotype is partially rescued in a p53(-/-), but not in a p16(-/-), background. Apoptosis is probably due to DNA damage as Atm downregulation exacerbates the phenotype. Despite this early lethal phenotype, Prep1 is not essential for ES cell establishment. A differential embryonic expression pattern underscores the unique function of Prep1 within the Meis-Prep family.


Assuntos
Apoptose/fisiologia , Desenvolvimento Embrionário/fisiologia , Camadas Germinativas/citologia , Proteínas de Homeodomínio/metabolismo , Células-Tronco Pluripotentes/citologia , Animais , Southern Blotting , Primers do DNA/genética , Ensaio de Desvio de Mobilidade Eletroforética , Imunofluorescência , Genótipo , Proteínas de Homeodomínio/genética , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Knockout , Células-Tronco Pluripotentes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Supressora de Tumor p53/metabolismo
12.
Prog Lipid Res ; 91: 101239, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37385352

RESUMO

Dentate gyrus of the hippocampus continuously gives rise to new neurons, namely, adult-born granule cells, which contribute to conferring plasticity to the mature brain throughout life. Within this neurogenic region, the fate and behavior of neural stem cells (NSCs) and their progeny result from a complex balance and integration of a variety of cell-autonomous and cell-to-cell-interaction signals and underlying pathways. Among these structurally and functionally diverse signals, there are endocannabinoids (eCBs), the main brain retrograde messengers. These pleiotropic bioactive lipids can directly and/or indirectly influence adult hippocampal neurogenesis (AHN) by modulating, both positively and negatively, multiple molecular and cellular processes in the hippocampal niche, depending on the cell type or stage of differentiation. Firstly, eCBs act directly as cell-intrinsic factors, cell-autonomously produced by NSCs following their stimulation. Secondly, in many, if not all, niche-associated cells, including some local neuronal and nonneuronal elements, the eCB system indirectly modulates the neurogenesis, linking neuronal and glial activity to regulating distinct stages of AHN. Herein, we discuss the crosstalk of the eCB system with other neurogenesis-relevant signal pathways and speculate how the hippocampus-dependent neurobehavioral effects elicited by (endo)cannabinergic medications are interpretable in light of the key regulatory role that eCBs play on AHN.


Assuntos
Endocanabinoides , Hipocampo , Adulto , Humanos , Hipocampo/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Transdução de Sinais
13.
Biomedicines ; 11(5)2023 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-37238963

RESUMO

Friedreich's ataxia (FRDA) is an autosomal, recessive, inherited neurodegenerative disease caused by the loss of activity of the mitochondrial protein frataxin (FXN), which primarily affects dorsal root ganglia, cerebellum, and spinal cord neurons. The genetic defect consists of the trinucleotide GAA expansion in the first intron of FXN gene, which impedes its transcription. The resulting FXN deficiency perturbs iron homeostasis and metabolism, determining mitochondrial dysfunctions and leading to reduced ATP production, increased reactive oxygen species (ROS) formation, and lipid peroxidation. These alterations are exacerbated by the defective functionality of the nuclear factor erythroid 2-related factor 2 (NRF2), a transcription factor acting as a key mediator of the cellular redox signalling and antioxidant response. Because oxidative stress represents a major pathophysiological contributor to FRDA onset and progression, a great effort has been dedicated to the attempt to restore the NRF2 signalling axis. Despite this, the beneficial effects of antioxidant therapies in clinical trials only partly reflect the promising results obtained in preclinical studies conducted in cell cultures and animal models. For these reasons, in this critical review, we overview the outcomes obtained with the administration of various antioxidant compounds and critically analyse the aspects that may have contributed to the conflicting results of preclinical and clinical studies.

14.
Mech Ageing Dev ; 211: 111802, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36958540

RESUMO

Reactive oxygen species (ROS) is a term that defines a group of unstable compounds derived from exogenous sources or endogenous metabolism. Under physiological conditions, low levels of ROS play a key role in the regulation of signal transduction- or transcription-mediated cellular responses. In contrast, excessive and uncontrolled loading of ROS results in a pathological state known as oxidative stress (OS), a leading contributor to aging and a pivotal factor for the onset and progression of many disorders. Evolution has endowed cells with an antioxidant system involved in stabilizing ROS levels to a specific threshold, maintaining ROS-induced signalling function and limiting negative side effects. In mammals, a great deal of evidence indicates that females defence against ROS is more proficient than males, determining a longer lifespan and lower incidence of most chronic diseases. In this review, we will summarize the most recent sex-related differences in the regulation of redox homeostasis. We will highlight the peculiar aspects of the antioxidant defence in sex-biased diseases whose onset or progression is driven by OS, and we will discuss the molecular, genetic, and evolutionary determinants of female proficiency to cope with ROS.


Assuntos
Antioxidantes , Caracteres Sexuais , Animais , Feminino , Masculino , Antioxidantes/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Oxirredução , Estresse Oxidativo , Homeostase , Mamíferos
15.
Mol Neurobiol ; 60(9): 5395-5410, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37314654

RESUMO

Niemann-Pick type C1 (NPC1) disease is a lysosomal lipid storage disorder due to mutations in the NPC1 gene resulting in the accumulation of cholesterol within the endosomal/lysosomal compartments. The prominent feature of the disorder is the progressive Purkinje cell degeneration leading to ataxia.In a mouse model of NPC1 disease, we have previously demonstrated that impaired Sonic hedgehog signaling causes defective proliferation of granule cells (GCs) and abnormal cerebellar morphogenesis. Studies conducted on cortical and hippocampal neurons indicate a functional interaction between Sonic hedgehog and brain-derived neurotrophic factor (BDNF) expression, leading us to hypothesize that BDNF signaling may be altered in Npc1 mutant mice, contributing to the onset of cerebellar alterations present in NPC1 disease before the appearance of signs of ataxia.We characterized the expression/localization patterns of the BDNF and its receptor, tropomyosin-related kinase B (TrkB), in the early postnatal and young adult cerebellum of the Npc1nmf164 mutant mouse strain.In Npc1nmf164 mice, our results show (i) a reduced expression of cerebellar BDNF and pTrkB in the first 2 weeks postpartum, phases in which most GCs complete the proliferative/migrative program and begin differentiation; (ii) an altered subcellular localization of the pTrkB receptor in GCs, both in vivo and in vitro; (iii) reduced chemotactic response to BDNF in GCs cultured in vitro, associated with impaired internalization of the activated TrkB receptor; (iv) an overall increase in dendritic branching in mature GCs, resulting in impaired differentiation of the cerebellar glomeruli, the major synaptic complex between GCs and mossy fibers.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Ataxia Cerebelar , Feminino , Camundongos , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteínas Hedgehog/metabolismo , Neurônios/metabolismo , Cerebelo/metabolismo , Células de Purkinje/metabolismo , Diferenciação Celular , Ataxia Cerebelar/metabolismo , Colesterol/metabolismo
16.
Cerebellum ; 11(1): 28-40, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20878296

RESUMO

We previously demonstrated that TSC22D4, a protein encoded by the TGF-ß1-activated gene Tsc22d4 (Thg-1pit) and highly expressed in postnatal and adult mouse cerebellum with multiple post-translationally modified protein forms, moves to nucleus when in vitro differentiated cerebellum granule neurons (CGNs) are committed to apoptosis by hyperpolarizing KCl concentrations in the culture medium. We have now studied TSC22D4 cytoplasmic/nuclear localization in CGNs and Purkinje cells: (1) during CGN differentiation/maturation in vivo, (2) during CGN differentiation in vitro, and (3) by in vitro culturing ex vivo cerebellum slices under conditions favoring/inhibiting CGN/Purkinje cell differentiation. We show that TSC22D4 displays both nuclear and cytoplasmic localizations in undifferentiated, early postnatal cerebellum CGNs, irrespectively of CGN proliferation/migration from external to internal granule cell layer, and that it specifically accumulates in the somatodendritic and synaptic compartments when CGNs mature, as indicated by TSC22D4 abundance at the level of adult cerebellum glomeruli and apparent lack in CGN nuclei. These features were also observed in cerebellum slices cultured in vitro under conditions favoring/inhibiting CGN/Purkinje cell differentiation. In vitro TSC22D4 silencing with siRNAs blocked CGN differentiation and inhibited neurite elongation in N1E-115 neuroblastoma cells, pinpointing the relevance of this protein to CGN differentiation.


Assuntos
Córtex Cerebelar/crescimento & desenvolvimento , Córtex Cerebelar/metabolismo , Grânulos Citoplasmáticos/metabolismo , Neurônios/metabolismo , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/genética , Linhagem Celular Tumoral , Córtex Cerebelar/citologia , Inativação Gênica , Masculino , Camundongos , Camundongos Endogâmicos , Neuritos/metabolismo , Neurogênese/genética , Neurônios/citologia , Técnicas de Cultura de Órgãos , Cultura Primária de Células , RNA Interferente Pequeno/farmacologia , Frações Subcelulares/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética
17.
Neurosci Res ; 170: 364-369, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33352204

RESUMO

Previous studies have shown inconsistent results regarding the effect of the Val158Met polymorphism of the catechol-O-methyltransferase (COMT) gene on personality and cognition. Here, nonclinical Caucasian university students of Italian origin were administered the Temperament and Character Inventory-Revised, Tellegen Absorption Scale, Differential Attentional Processes Inventory, and Waterloo-Stanford Group Scale of Hypnotic Susceptibility. We found that the COMT Val158Met polymorphism was significantly associated with the disorderliness facet of novelty seeking (NS4) and that sex was a moderator of this association. Females with the Met/Met genotype showed higher NS4 scores compared to those with the Val/Met and Val/Val genotypes. No significant genotype effect was found for males. Additionally, we failed to find a significant effect of the COMT gene on attention and hypnotic suggestibility measures. These results provide further evidence for a sex-specific influence on the gene-behaviour associations.


Assuntos
Catecol O-Metiltransferase , Comportamento Exploratório , Feminino , Humanos , Masculino , Catecol O-Metiltransferase/genética , Caráter , Genótipo , Polimorfismo de Nucleotídeo Único
18.
Redox Biol ; 38: 101791, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33197769

RESUMO

Ferroptosis is an iron-dependent cell death caused by impaired glutathione metabolism, lipid peroxidation and mitochondrial failure. Emerging evidences report a role for ferroptosis in Friedreich's Ataxia (FRDA), a neurodegenerative disease caused by the decreased expression of the mitochondrial protein frataxin. Nrf2 signalling is implicated in many molecular aspects of ferroptosis, by upstream regulating glutathione homeostasis, mitochondrial function and lipid metabolism. As Nrf2 is down-regulated in FRDA, targeting Nrf2-mediated ferroptosis in FRDA may be an attractive option to counteract neurodegeneration in such disease, thus paving the way to new therapeutic opportunities. In this study, we evaluated ferroptosis hallmarks in frataxin-silenced mouse myoblasts, in hearts of a frataxin Knockin/Knockout (KIKO) mouse model, in skin fibroblasts and blood of patients, particularly focusing on ferroptosis-driven gene expression, mitochondrial impairment and lipid peroxidation. The efficacy of Nrf2 inducers to neutralize ferroptosis has been also evaluated.


Assuntos
Ferroptose , Ataxia de Friedreich , Doenças Neurodegenerativas , Animais , Ataxia de Friedreich/genética , Humanos , Camundongos , Camundongos Knockout , Fator 2 Relacionado a NF-E2/genética
19.
Mol Cell Neurosci ; 40(2): 249-57, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19084601

RESUMO

Thg-1pit (Tsc22d4), a murine gene belonging to the TGF-beta1-stimulated clone 22 domain (TSC22D) family, is expressed in developing and adult cerebellar granule neurons and mature Purkinje cells. We have studied THG-1pit function in primary cultures of mouse cerebellar granule neurons maintained in vitro in the presence of a medium containing 25 mM K+ (differentiating condition) or 5 mM K+ (pro-apoptotic condition), and determined the effect of culture medium, TGF-beta1 and IGF-1 on THG-1pit expression and intracellular localization. Thg-1pit encoded a 42 kDa MW protein and other, higher MW and developmentally-regulated forms. Cell exposure to 5 mM K+ elicited early and/or late waves of Thg-1pit transcription, depending on the presence/absence of TGF-beta1, and caused THG-1pit to massively and transiently move from cytoplasm and neurites to the nucleus. THG-1pit nuclear entrance was concomitant to that of AIF, suggesting that THG-1pit is involved in the induction of granule neuron apoptosis.


Assuntos
Apoptose/fisiologia , Núcleo Celular/metabolismo , Cerebelo/citologia , Neurônios/fisiologia , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/fisiologia , Sobrevivência Celular , Células Cultivadas , Cerebelo/embriologia , Cerebelo/crescimento & desenvolvimento , Humanos , Camundongos , Neurônios/citologia , Potássio/metabolismo , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA