Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(3)2021 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-33514010

RESUMO

Down syndrome (DS) or trisomy of chromosome 21 (Hsa21) is characterized by impaired hippocampal-dependent learning and memory. These alterations are due to defective neurogenesis and to neuromorphological and functional anomalies of numerous neuronal populations, including hippocampal granular cells (GCs). It has been proposed that the additional gene dose in trisomic cells induces modifications in nuclear compartments and on the chromatin landscape, which could contribute to some DS phenotypes. The Ts65Dn (TS) mouse model of DS carries a triplication of 92 genes orthologous to those found in Hsa21, and shares many phenotypes with DS individuals, including cognitive and neuromorphological alterations. Considering its essential role in hippocampal memory formation, we investigated whether the triplication of this set of Hsa21 orthologous genes in TS mice modifies the nuclear architecture of their GCs. Our results show that the TS mouse presents alterations in the nuclear architecture of its GCs, affecting nuclear compartments involved in transcription and pre-rRNA and pre-mRNA processing. In particular, the GCs of the TS mouse show alterations in the nucleolar fusion pattern and the molecular assembly of Cajal bodies (CBs). Furthermore, hippocampal GCs of TS mice present an epigenetic dysregulation of chromatin that results in an increased heterochromatinization and reduced global transcriptional activity. These nuclear alterations could play an important role in the neuromorphological and/or functional alterations of the hippocampal GCs implicated in the cognitive dysfunction characteristic of TS mice.


Assuntos
Cromatina/genética , Síndrome de Down/genética , Hipocampo/metabolismo , Neurônios/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Nucléolo Celular/genética , Nucléolo Celular/metabolismo , Cognição/fisiologia , Corpos Enovelados/genética , Corpos Enovelados/metabolismo , Modelos Animais de Doenças , Síndrome de Down/patologia , Hipocampo/patologia , Humanos , Memória/fisiologia , Camundongos , Camundongos Transgênicos , Neurogênese/genética , Neurogênese/fisiologia , Neurônios/patologia
2.
Cell Tissue Res ; 381(3): 461-478, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32676861

RESUMO

Spinal muscular atrophy (SMA) is caused by a deletion or mutation of the survival motor neuron 1 (SMN1) gene. Reduced SMN levels lead to motor neuron degeneration and muscular atrophy. SMN protein localizes to the cytoplasm and Cajal bodies. Moreover, in myofibrils from Drosophila and mice, SMN is a sarcomeric protein localized to the Z-disc. Although SMN participates in multiple functions, including the biogenesis of spliceosomal small nuclear ribonucleoproteins, its role in the sarcomere is unclear. Here, we analyzed the sarcomeric organization of SMN in human control and type I SMA skeletal myofibers. In control sarcomeres, we demonstrate that human SMN is localized to the titin-positive M-band and actin-positive I-band, and to SMN-positive granules that flanked the Z-discs. Co-immunoprecipitation assays revealed that SMN interacts with the sarcomeric protein actin, α-actinin, titin, and profilin2. In the type I SMA muscle, SMN levels were reduced, and atrophic (denervated) and hypertrophic (nondenervated) myofibers coexisted. The hypertrophied myofibers, which are potential primary targets of SMN deficiency, exhibited sites of focal or segmental alterations of the actin cytoskeleton, where the SMN immunostaining pattern was altered. Moreover, SMN was relocalized to the Z-disc in overcontracted minisarcomeres from hypertrophic myofibers. We propose that SMN could have an integrating role in the molecular components of the sarcomere. Consequently, low SMN levels might impact the normal sarcomeric architecture, resulting in the disruption of myofibrils found in SMA muscle. This primary effect might be independent of the neurogenic myopathy produced by denervation and contribute to pathophysiology of the SMA myopathy.


Assuntos
Músculo Esquelético/metabolismo , Atrofia Muscular Espinal/metabolismo , Miofibrilas/metabolismo , Sarcômeros/metabolismo , Humanos
3.
Neurobiol Dis ; 127: 312-322, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30905767

RESUMO

The Purkinje cell (PC) degeneration (pcd) mouse harbors a mutation in Agtpbp1 gene that encodes for the cytosolic carboxypeptidase, CCP1. The mutation causes degeneration and death of PCs during the postnatal life, resulting in clinical and pathological manifestation of cerebellar ataxia. Monogenic biallelic damaging variants in the Agtpbp1 gene cause infantile-onset neurodegeneration and cerebellar atrophy, linking loss of functional CCP1 with human neurodegeneration. Although CCP1 plays a key role in the regulation of tubulin stabilization, its loss of function in PCs leads to a severe nuclear phenotype with heterochromatinization and accumulation of DNA damage. Therefore, the pcd mice provides a useful neuronal model to investigate nuclear mechanisms involved in neurodegeneration, particularly the nucleolar stress. In this study, we demonstrated that the Agtpbp1 gene mutation induces a p53-dependent nucleolar stress response in PCs, which is characterized by nucleolar fragmentation, nucleoplasmic and cytoplasmic mislocalization of nucleolin, and dysfunction of both pre-rRNA processing and mRNA translation. RT-qPCR analysis revealed reduction of mature 18S rRNA, with a parallel increase of its intermediate 18S-5'-ETS precursor, that correlates with a reduced expression of Fbl mRNA, which encodes an essential factor for rRNA processing. Moreover, nucleolar alterations were accompanied by a reduction of PTEN mRNA and protein levels, which appears to be related to the chromosome instability and accumulation of DNA damage in degenerating PCs. Our results highlight the essential contribution of nucleolar stress to PC degeneration and also underscore the nucleoplasmic mislocalization of nucleolin as a potential indicator of neurodegenerative processes.


Assuntos
Nucléolo Celular/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Fosfoproteínas/metabolismo , Células de Purkinje/metabolismo , Proteínas de Ligação a RNA/metabolismo , D-Ala-D-Ala Carboxipeptidase Tipo Serina/metabolismo , Animais , Proteínas de Ligação ao GTP/genética , Camundongos , Mutação , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Células de Purkinje/patologia , D-Ala-D-Ala Carboxipeptidase Tipo Serina/genética , Nucleolina
4.
Histochem Cell Biol ; 152(3): 227-237, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31183542

RESUMO

Type I spinal muscular atrophy (SMA) is an autosomal recessive disorder caused by the loss or mutation of the survival motor neuron 1 (SMN1) gene. The reduction in SMN protein levels in SMA leads to the degeneration of motor neurons and muscular atrophy. In this study, we analyzed the nuclear reorganization in human skeletal myofibers from a type I SMA patient carrying a deletion of exons 7 and 8 in the SMN1 gene and two SMN2 gene copies and showing reduced SMN protein levels in the muscle compared with those in control samples. The morphometric analysis of myofiber size revealed the coexistence of atrophic and hypertrophic myofibers in SMA samples. Compared with controls, both nuclear size and the nuclear shape factor were significantly reduced in SMA myonuclei. Nuclear reorganization in SMA myonuclei was characterized by extensive heterochromatinization, the aggregation of splicing factors in large interchromatin granule clusters, and nucleolar alterations with the accumulation of the granular component and a loss of fibrillar center/dense fibrillar component units. These nuclear alterations reflect a severe perturbation of global pre-mRNA transcription and splicing, as well as nucleolar dysfunction, in SMA myofibers. Moreover, the finding of similar nuclear reorganization in both atrophic and hypetrophic myofibers provides additional support that the SMN deficiency in SMA patients may primarily affect the skeletal myofibers.


Assuntos
Núcleo Celular/genética , Músculo Esquelético/patologia , RNA/genética , RNA/metabolismo , Atrofias Musculares Espinais da Infância/genética , Atrofias Musculares Espinais da Infância/patologia , Núcleo Celular/metabolismo , Humanos , Recém-Nascido , Masculino , Músculo Esquelético/metabolismo
5.
Cell Mol Life Sci ; 75(3): 527-546, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28879433

RESUMO

The survival of motor neuron (SMN) protein plays an essential role in the biogenesis of spliceosomal snRNPs and the molecular assembly of Cajal bodies (CBs). Deletion of or mutations in the SMN1 gene cause spinal muscular atrophy (SMA) with degeneration and loss of motor neurons. Reduced SMN levels in SMA lead to deficient snRNP biogenesis with consequent splicing pathology. Here, we demonstrate that SMN is a novel and specific target of the acetyltransferase CBP (CREB-binding protein). Furthermore, we identify lysine (K) 119 as the main acetylation site in SMN. Importantly, SMN acetylation enhances its cytoplasmic localization, causes depletion of CBs, and reduces the accumulation of snRNPs in nuclear speckles. In contrast, the acetylation-deficient SMNK119R mutant promotes formation of CBs and a novel category of promyelocytic leukemia (PML) bodies enriched in this protein. Acetylation increases the half-life of SMN protein, reduces its cytoplasmic diffusion rate and modifies its interactome. Hence, SMN acetylation leads to its dysfunction, which explains the ineffectiveness of HDAC (histone deacetylases) inhibitors in SMA therapy despite their potential to increase SMN levels.


Assuntos
Corpos Enovelados/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Citoplasma/metabolismo , Proteínas do Complexo SMN/metabolismo , Acetilação , Células Cultivadas , Células HEK293 , Humanos , Células MCF-7 , Processamento de Proteína Pós-Traducional , Transporte Proteico
6.
Neurobiol Dis ; 108: 83-99, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28823932

RESUMO

Spinal muscular atrophy (SMA) is caused by a homozygous deletion or mutation in the survival motor neuron 1 (SMN1) gene that leads to reduced levels of SMN protein resulting in degeneration of motor neurons (MNs). The best known functions of SMN is the biogenesis of spliceosomal snRNPs. Linked to this function, Cajal bodies (CBs) are involved in the assembly of spliceosomal (snRNPs) and nucleolar (snoRNPs) ribonucleoproteins required for pre-mRNA and pre-rRNA processing. Recent studies support that the interaction between CBs and nucleoli, which are especially prominent in neurons, is essential for the nucleolar rRNA homeostasis. We use the SMN∆7 murine model of type I SMA to investigate the cellular basis of the dysfunction of RNA metabolism in MNs. SMN deficiency in postnatal MNs produces a depletion of functional CBs and relocalization of coilin, which is a scaffold protein of CBs, in snRNP-free perinucleolar caps or within the nucleolus. Disruption of CBs is the earliest nuclear sign of MN degeneration. We demonstrate that depletion of CBs, with loss of CB-nucleolus interactions, induces a progressive nucleolar dysfunction in ribosome biogenesis. It includes reorganization and loss of nucleolar transcription units, segregation of dense fibrillar and granular components, retention of SUMO-conjugated proteins in intranucleolar bodies and a reactive, compensatory, up-regulation of mature 18S rRNA and genes encoding key nucleolar proteins, such as upstream binding factor, fibrillarin, nucleolin and nucleophosmin. We propose that CB depletion and nucleolar alterations are essential components of the dysfunction of RNA metabolism in SMA.


Assuntos
Nucléolo Celular/metabolismo , Neurônios Motores/metabolismo , Atrofia Muscular Espinal/metabolismo , RNA/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Nucléolo Celular/patologia , Modelos Animais de Doenças , Eletroforese em Gel de Poliacrilamida , Imunofluorescência , Camundongos Transgênicos , Microscopia Confocal , Microscopia Eletrônica , Neurônios Motores/patologia , Atrofia Muscular Espinal/patologia , Proteínas Nucleares/metabolismo , Ribonucleoproteínas/metabolismo , Medula Espinal/metabolismo , Medula Espinal/patologia
7.
J Bone Miner Metab ; 35(2): 150-160, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27038990

RESUMO

Different model systems using osteoblastic cell lines have been developed to help understand the process of bone formation. Here, we report the establishment of two human osteoblastic cell lines obtained from primary cultures upon transduction of immortalizing genes. The resulting cell lines had no major differences to their parental lines in their gene expression profiles. Similar to primary osteoblastic cells, osteocalcin transcription increased following 1,25-dihydroxyvitamin D3 treatment and the immortalized cells formed a mineralized matrix, as detected by Alizarin Red staining. Moreover, these human cell lines responded by upregulating ALPL gene expression after treatment with the demethylating agent 5-aza-2'-deoxycytidine (AzadC), as shown before for primary osteoblasts. We further demonstrate that these cell lines can differentiate in vivo, using a hydroxyapatite/tricalcium phosphate composite as a scaffold, to produce bone matrix. More importantly, we show that these cells respond to demethylating treatment, as shown by the increase in SOST mRNA levels, the gene encoding sclerostin, upon treatment of the recipient mice with AzadC. This also confirms, in vivo, the role of DNA methylation in the regulation of SOST expression previously shown in vitro. Altogether our results show that these immortalized cell lines constitute a particularly useful model system to obtain further insight into bone homeostasis, and particularly into the epigenetic mechanisms regulating sclerostin production.


Assuntos
Linhagem Celular , Epigênese Genética , Osteoblastos/citologia , Osteogênese , Proteínas Adaptadoras de Transdução de Sinal , Animais , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Calcificação Fisiológica , Calcitriol/farmacologia , Diferenciação Celular , Metilação de DNA , Decitabina , Marcadores Genéticos/genética , Humanos , Camundongos , Camundongos Nus , Osteocalcina/genética , Osteocalcina/metabolismo , Alicerces Teciduais
8.
RNA Biol ; 14(6): 712-725, 2017 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-27627892

RESUMO

Cajal is commonly regarded as the father of modern neuroscience in recognition of his fundamental work on the structure of the nervous system. But Cajal also made seminal contributions to the knowledge of nuclear structure in the early 1900s, including the discovery of the "accessory body" later renamed "Cajal body" (CB). This important nuclear structure has emerged as a center for the assembly of ribonucleoproteins (RNPs) required for splicing, ribosome biogenesis and telomere maintenance. The modern era of CB research started in the 1990s with the discovery of coilin, now known as a scaffold protein of CBs, and specific probes for small nuclear RNAs (snRNAs). In this review, we summarize what we have learned in the recent decades concerning CBs in post-mitotic neurons, thereby ruling out dynamic changes in CB functions during the cell cycle. We show that CBs are particularly prominent in neurons, where they frequently associate with the nucleolus. Neuronal CBs are transcription-dependent nuclear organelles. Indeed, their number dynamically accommodates to support the high neuronal demand for splicing and ribosome biogenesis required for sustaining metabolic and bioelectrical activity. Mature neurons have canonical CBs enriched in coilin, survival motor neuron protein and snRNPs. Disruption and loss of neuronal CBs associate with severe neuronal dysfunctions in several neurological disorders such as motor neuron diseases. In particular, CB depletion in motor neurons seems to reflect a perturbation of transcription and splicing in spinal muscular atrophy, the most common genetic cause of infant mortality.


Assuntos
Corpos Enovelados/metabolismo , Neurônios/metabolismo , Animais , Nucléolo Celular/metabolismo , Suscetibilidade a Doenças , Humanos , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Transporte Proteico , Splicing de RNA , RNA Nuclear Pequeno/genética , RNA Nuclear Pequeno/metabolismo , Ribonucleoproteínas Nucleares Pequenas/metabolismo
9.
J Cell Sci ; 127(Pt 5): 939-46, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24413165

RESUMO

Cajal bodies (CBs) are nuclear organelles involved in the maturation of spliceosomal small nuclear ribonucleoproteins (snRNPs). They concentrate coilin, snRNPs and the survival motor neuron protein (SMN). Dysfunction of CB assembly occurs in spinal muscular atrophy (SMA). Here, we demonstrate that SMN is a SUMO1 target that has a small ubiquitin-related modifier (SUMO)-interacting motif (SIM)-like motif in the Tudor domain. The expression of SIM-like mutant constructs abolishes the interaction of SMN with the spliceosomal SmD1 (also known as SNRPD1), severely decreases SMN-coilin interaction and prevents CB assembly. Accordingly, the SMN SIM-like-mediated interactions are important for CB biogenesis and their dysfunction can be involved in SMA pathophysiology.


Assuntos
Corpos Enovelados/metabolismo , Proteínas Nucleares/metabolismo , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo , Proteínas Centrais de snRNP/metabolismo , Sequência de Aminoácidos , Células HEK293 , Humanos , Interações Hidrofóbicas e Hidrofílicas , Células MCF-7 , Dados de Sequência Molecular , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Estrutura Secundária de Proteína , Transporte Proteico , Proteína SUMO-1/metabolismo , Spliceossomos/metabolismo , Sumoilação
10.
Biochim Biophys Acta ; 1842(6): 848-59, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24269586

RESUMO

The dysfunction of the ubiquitin proteasome system has been related to a broad array of neurodegenerative disorders in which the accumulation of misfolded protein aggregates causes proteotoxicity. The ability of proteasome inhibitors to induce cell cycle arrest and apoptosis has emerged as a powerful strategy for cancer therapy. Bortezomib is a proteasome inhibitor used as an antineoplastic drug, although its neurotoxicity frequently causes a severe sensory peripheral neuropathy. In this study we used a rat model of bortezomib treatment to study the nucleolar and Cajal body responses to the proteasome inhibition in sensory ganglion neurons that are major targets of bortezomib-induced neurotoxicity. Treatment with bortezomib induced dose-dependent dissociation of protein synthesis machinery (chromatolysis) and nuclear retention of poly(A) RNA granules resulting in neuronal dysfunction. However, as a compensatory response to the proteotoxic stress, both nucleoli and Cajal bodies exhibited reactive changes. These include an increase in the number and size of nucleoli, strong nucleolar incorporation of the RNA precursor 5'-fluorouridine, and increased expression of both 45S rRNA and genes encoding nucleolar proteins UBF, fibrillarin and B23. Taken together, these findings appear to reflect the activation of the nucleolar transcription in response to proteotoxic stress Furthermore, the number of Cajal bodies, a parameter related to transcriptional activity, increases upon proteasome inhibition. We propose that nucleoli and Cajal bodies are important targets in the signaling pathways that are activated by the proteotoxic stress response to proteasome inhibition. The coordinating activity of these two organelles in the production of snRNA, snoRNA and rRNA may contribute to neuronal survival after proteasome inhibition. This article is part of a Special Issue entitled: Role of the Nucleolus in Human Disease.


Assuntos
Corpos Enovelados/metabolismo , Gânglios Sensitivos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Células Receptoras Sensoriais/metabolismo , Animais , Ácidos Borônicos/administração & dosagem , Bortezomib , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/genética , Nucléolo Celular/metabolismo , Núcleo Celular , Citoplasma/metabolismo , Gânglios Sensitivos/crescimento & desenvolvimento , Humanos , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Pirazinas/administração & dosagem , Ratos , Transdução de Sinais/efeitos dos fármacos
11.
Cell Mol Life Sci ; 71(10): 1961-75, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24061536

RESUMO

Bortezomib is a reversible proteasome inhibitor used as an anticancer drug. However, its clinical use is limited since it causes peripheral neurotoxicity. We have used Sprague-Dawley rats as an animal model to investigate the cellular mechanisms affected by both short-term and chronic bortezomib treatments in sensory ganglia neurons. Proteasome inhibition induces dose-dependent alterations in the architecture, positioning, shape and polarity of the neuronal nucleus. It also produces DNA damage without affecting neuronal survival, and severe disruption of the protein synthesis machinery at the central cytoplasm accompanied by decreased expression of the brain-derived neurotrophic factor. As a compensatory or adaptive survival response against proteotoxic stress caused by bortezomib treatment, sensory neurons preserve basal levels of transcriptional activity, up-regulate the expression of proteasome subunit genes, and generate a new cytoplasmic perinuclear domain for protein synthesis. We propose that proteasome activity is crucial for controlling nuclear architecture, DNA repair and the organization of the protein synthesis machinery in sensory neurons. These neurons are primary targets of bortezomib neurotoxicity, for which reason their dysfunction may contribute to the pathogenesis of the bortezomib-induced peripheral neuropathy in treated patients.


Assuntos
Núcleo Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Gânglios Sensitivos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Animais , Antineoplásicos/farmacologia , Ácidos Borônicos/farmacologia , Bortezomib , Núcleo Celular/fisiologia , Reparo do DNA/efeitos dos fármacos , Gânglios Sensitivos/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Masculino , Microscopia Confocal , Corpos de Nissl/efeitos dos fármacos , Corpos de Nissl/fisiologia , Complexo de Endopeptidases do Proteassoma/química , Complexo de Endopeptidases do Proteassoma/genética , Pirazinas/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
12.
Stem Cells ; 31(6): 1075-85, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23401361

RESUMO

Glioblastoma (GBM) is associated with infiltration of peritumoral (PT) parenchyma by isolated tumor cells that leads to tumor regrowth. Recently, GBM stem-like or initiating cells (GICs) have been identified in the PT area, but whether these GICs have enhanced migratory and invasive capabilities compared with GICs from the tumor mass (TM) is presently unknown. We isolated GICs from the infiltrated PT tissue and the TM of three patients and found that PT cells have an advantage over TM cells in two-dimensional and three-dimensional migration and invasion assays. Interestingly, PT cells display a high plasticity in protrusion formation and cell shape and their migration is insensitive to substrate stiffness, which represent advantages to infiltrate microenvironments of different rigidity. Furthermore, mouse and chicken embryo xenografts revealed that only PT cells showed a dispersed distribution pattern, closely associated to blood vessels. Consistent with cellular plasticity, simultaneous Rac and RhoA activation are required for the enhanced invasive capacity of PT cells. Moreover, Rho GTPase signaling modulators αVß3 and p27 play key roles in GIC invasiveness. Of note, p27 is upregulated in TM cells and inhibits RhoA activity. Gene silencing of p27 increased the invasive capacity of TM GICs. Additionally, ß3 integrin is upregulated in PT cells. Blockade of dimeric integrin αVß3, a Rac activator, reduced the invasive capacity of PT GICs in vitro and abrogated the spreading of PT cells into chicken embryos. Thus, our results describe the invasive features acquired by a unique subpopulation of GICs that infiltrate neighboring tissue.


Assuntos
Neoplasias Encefálicas/patologia , Movimento Celular/fisiologia , Glioblastoma/patologia , Células-Tronco Neoplásicas/patologia , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/genética , Embrião de Galinha , Regulação para Baixo , Feminino , Glioblastoma/genética , Glioblastoma/metabolismo , Xenoenxertos , Humanos , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Células-Tronco Neoplásicas/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas , Regulação para Cima , Proteínas rac de Ligação ao GTP/genética , Proteínas rac de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
13.
Glia ; 61(2): 254-72, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23047288

RESUMO

Purkinje Cell Degeneration (PCD) mice harbor a nna1 gene mutation which leads to an early and rapid degeneration of Purkinje cells (PC) between the third and fourth week of age. This mutation also underlies the death of mitral cells (MC) in the olfactory bulb (OB), but this process is slower and longer than in PC. No clear interpretations supporting the marked differences in these neurodegenerative processes exist. Growing evidence suggests that either beneficial or detrimental effects of gliosis in damaged regions would underlie these divergences. Here, we examined the gliosis occurring during PC and MC death in the PCD mouse. Our results demonstrated different glial reactions in both affected regions. PC disappearance stimulated a severe gliosis characterized by strong morphological changes, enhanced glial proliferation, as well as the release of pro-inflammatory mediators. By contrast, MC degeneration seems to promote a more attenuated glial response in the PCD OB compared with that of the cerebellum. Strikingly, cerebellar oligodendrocytes died by apoptosis in the PCD, whereas bulbar ones were not affected. Interestingly, the level of nna1 mRNA under normal conditions was higher in the cerebellum than in the OB, probably related to a faster neurodegeneration and stronger glial reaction in its absence. The glial responses may thus influence the neurodegenerative course in the cerebellum and OB of the mutant mouse brain, providing harmful and beneficial microenvironments, respectively.


Assuntos
Proteínas de Ligação ao GTP/genética , Mutação/genética , Degeneração Neural/genética , Degeneração Neural/patologia , Neuroglia/fisiologia , Células de Purkinje/patologia , D-Ala-D-Ala Carboxipeptidase Tipo Serina/genética , Fatores Etários , Animais , Animais Recém-Nascidos , Bromodesoxiuridina/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Morte Celular/genética , Proliferação de Células , Cerebelo/patologia , Proteínas de Ligação ao GTP/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/genética , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Microscopia Eletrônica de Transmissão , Proteínas do Tecido Nervoso/metabolismo , Bulbo Olfatório/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Células de Purkinje/ultraestrutura , RNA Mensageiro , D-Ala-D-Ala Carboxipeptidase Tipo Serina/metabolismo
14.
Traffic ; 11(6): 743-55, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20331843

RESUMO

p21(cip1) is a protein with a dual function in oncogenesis depending mainly on its intracellular localization: tumor suppressor in the nucleus and oncogenic in the cytoplasm. After DNA damage, p21(cip1) increases and accumulates in the nucleus to ensure cell cycle arrest. We show here that the nuclear accumulation of p21(cip1) is not only a consequence of its increased levels but to a DNA damage cellular response, which is ataxia telangiectasia and Rad3 related (ATR)/ataxia telangiectasia mutated (ATM) and p53 independent. Furthermore, after DNA damage, p21(cip1) not only accumulates in the nucleoplasm but also in the disrupted nucleolus. Inside the nucleolus, it is found in spherical structures, which are not a protrusion of the nucleoplasm. The steady-state distribution of p21(cip1) in the nucleolus resulted from a highly dynamic equilibrium between nucleoplasmic and nucleolar p21(cip1) and correlated with the inhibition of p21(cip1) nuclear export. Most interestingly, inhibition of ribosomal export after expressing a dominant-negative mutant of nucleophosmin induced p21(cip1) accumulation in the nucleus and the nucleolus in the absence of DNA damage. This proved the existence of a nucleolar export route to the cytoplasm for p21(cip1) in control conditions that would be inhibited upon DNA damage leading to nuclear and nucleolar accumulation of p21(cip1).


Assuntos
Nucléolo Celular/metabolismo , Núcleo Celular/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/química , Dano ao DNA , Ciclo Celular , Linhagem Celular Tumoral , Genes Dominantes , Humanos , Imuno-Histoquímica , Microscopia de Fluorescência/métodos , Mutação , Proteínas Nucleares/química , Nucleofosmina , Fotodegradação , Plasmídeos/metabolismo , Frações Subcelulares/metabolismo
15.
J Biol Chem ; 286(32): 28287-302, 2011 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-21700704

RESUMO

DNA repair protects neurons against spontaneous or disease-associated DNA damage. Dysfunctions of this mechanism underlie a growing list of neurodegenerative disorders. The Purkinje cell (PC) degeneration mutation causes the loss of nna1 expression and is associated with the postnatal degeneration of PCs. This PC degeneration dramatically affects nuclear architecture and provides an excellent model to elucidate the nuclear mechanisms involved in a whole array of neurodegenerative disorders. We used immunocytochemistry for histone variants and components of the DNA damage response, an in situ transcription assay, and in situ hybridization for telomeres to analyze changes in chromatin architecture and function. We demonstrate that the phosphorylation of H2AX, a DNA damage signal, and the trimethylation of the histone H4K20, a repressive mark, in extensive domains of genome are epigenetic hallmarks of chromatin in degenerating PCs. These histone modifications are associated with a large scale reorganization of chromatin, telomere clustering, and heterochromatin-induced gene silencing, all of them key factors in PC degeneration. Furthermore, ataxia telangiectasia mutated and 53BP1, two components of the DNA repair pathway, fail to be concentrated in the damaged chromatin compartments, even though the expression levels of their coding genes were slightly up-regulated. Although the mechanism by which Nna1 loss of function leads to PC neurodegeneration is undefined, the progressive accumulation of DNA damage in chromosome territories irreversibly compromises global gene transcription and seems to trigger PC degeneration and death.


Assuntos
Montagem e Desmontagem da Cromatina , Reparo do DNA , Inativação Gênica , Doenças Neurodegenerativas/metabolismo , Células de Purkinje/metabolismo , Transdução de Sinais , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Dano ao DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Histonas/genética , Histonas/metabolismo , Masculino , Camundongos , Camundongos Mutantes , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Células de Purkinje/patologia , D-Ala-D-Ala Carboxipeptidase Tipo Serina/genética , D-Ala-D-Ala Carboxipeptidase Tipo Serina/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
16.
Neurobiol Dis ; 46(1): 118-29, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22249111

RESUMO

Nuclear speckles are essential nuclear compartments involved in the assembly, delivery and recycling of pre-mRNA processing factors, and in the post-transcriptional processing of pre-mRNAs. Oculopharyngeal muscular dystrophy (OPMD) is caused by a small expansion of the polyalanine tract in the poly(A)-binding protein nuclear 1 (PABPN1). Aggregation of expanded PABPN1 into intranuclear inclusions (INIs) in skeletal muscle fibers is the pathological hallmark of OPMD. In this study what we have analyzed in muscle fibers of OPMD patients and in primary cultures of human myoblasts are the relationships between nuclear speckles and INIs, and the contribution of the former to the biogenesis of the latter. While nuclear speckles concentrate snRNP splicing factors and PABPN1 in control muscle fibers, they are depleted of PABPN1 and appear closely associated with INIs in muscle fibers of OPMD patients. The induction of INI formation in human myoblasts expressing either wild type GFP-PABPN1 or expanded GFP-PABPN1-17ala demonstrates that the initial aggregation of PABPN1 proteins and their subsequent growth in INIs occurs at the edges of the nuclear speckles. Moreover, the growing of INIs gradually depletes PABPN1 proteins and poly(A) RNA from nuclear speckles, although the existence of these nuclear compartments is preserved. Time-lapse experiments in cultured myoblasts confirm nuclear speckles as biogenesis sites of PABPN1 inclusions. Given the functional importance of nuclear speckles in the post-transcriptional processing of pre-mRNAs, the INI-dependent molecular reorganization of these nuclear compartments in muscle fibers may cause a severe dysfunction in nuclear trafficking and processing of polyadenylated mRNAs, thereby contributing to the molecular pathophysiology of OPMD. Our results emphasize the potential importance of nuclear speckles as nuclear targets of neuromuscular disorders.


Assuntos
Fibras Musculares Esqueléticas/patologia , Fibras Musculares Esqueléticas/fisiologia , Distrofia Muscular Oculofaríngea/patologia , Distrofia Muscular Oculofaríngea/fisiopatologia , Proteína I de Ligação a Poli(A)/fisiologia , Idoso de 80 Anos ou mais , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Feminino , Humanos , Pessoa de Meia-Idade , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular Oculofaríngea/genética , Proteína I de Ligação a Poli(A)/genética
17.
Histochem Cell Biol ; 137(5): 657-67, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22302308

RESUMO

Type I spinal muscular atrophy (SMA) is an autosomal recessive disorder caused by loss or mutations of the survival motor neuron 1 (SMN1) gene. The reduction in SMN protein levels in SMA leads to degeneration and death of motor neurons. In this study, we have analyzed the nuclear reorganization of Cajal bodies, PML bodies and nucleoli in type I SMA motor neurons with homozygous deletion of exons 7 and 8 of the SMN1 gene. Western blot analysis is is revealed a marked reduction of SMN levels compared to the control sample. Using a neuronal dissociation procedure to perform a careful immunocytochemical and quantitative analysis of nuclear bodies, we demonstrated a severe decrease in the mean number of Cajal bodies per neuron and in the proportion of motor neurons containing these structures in type I SMA. Moreover, most Cajal bodies fail to recruit SMN and spliceosomal snRNPs, but contain the proteasome activator PA28, a molecular marker associated with the cellular stress response. Neuronal stress in SMA motor neurons also increases PML body number. The existence of chromatolysis and eccentric nuclei in SMA motor neurons correlates with Cajal body disruption and nucleolar relocalization of coil in, a Cajal body marker. Our results indicate that the Cajal body is a pathophysiological target in type I SMA motor neurons. They also suggest the Cajal body-dependent dysfunction of snRNP biogenesis and, therefore, pre-mRNA splicing in these neurons seems to be an essential component for SMA pathogenesis.


Assuntos
Nucléolo Celular/metabolismo , Corpos Enovelados/metabolismo , Neurônios Motores/metabolismo , Proteínas Nucleares/metabolismo , Atrofias Musculares Espinais da Infância/metabolismo , Atrofias Musculares Espinais da Infância/patologia , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo , Nucléolo Celular/química , Corpos Enovelados/química , Humanos , Atrofias Musculares Espinais da Infância/genética , Proteína 1 de Sobrevivência do Neurônio Motor/química , Proteína 1 de Sobrevivência do Neurônio Motor/genética
18.
Antioxidants (Basel) ; 11(12)2022 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-36552646

RESUMO

Oxidative stress (OS) is one of the neuropathological mechanisms responsible for the deficits in cognition and neuronal function in Down syndrome (DS). The Ts65Dn (TS) mouse replicates multiple DS phenotypes including hippocampal-dependent learning and memory deficits and similar brain oxidative status. To better understand the hippocampal oxidative profile in the adult TS mouse, we analyzed cellular OS-associated alterations in hippocampal granule cells (GCs), a neuronal population that plays an important role in memory formation and that is particularly affected in DS. For this purpose, we used biochemical, molecular, immunohistochemical, and electron microscopy techniques. Our results indicate that TS GCs show important OS-associated alterations in the systems essential for neuronal homeostasis: DNA damage response and proteostasis, particularly of the proteasome and lysosomal system. Specifically, TS GCs showed: (i) increased DNA damage, (ii) reorganization of nuclear proteolytic factories accompanied by a decline in proteasome activity and cytoplasmic aggregation of ubiquitinated proteins, (iii) formation of lysosomal-related structures containing lipid droplets of cytotoxic peroxidation products, and (iv) mitochondrial ultrastructural defects. These alterations could be implicated in enhanced cellular senescence, accelerated aging and neurodegeneration, and the early development of Alzheimer's disease neuropathology present in TS mice and the DS population.

19.
Biomolecules ; 12(8)2022 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-35892322

RESUMO

In this work, we performed a methodological comparative analysis to synthesize polyethyleneimine (PEI) nanoparticles using (i) conventional nanoprecipitation (NP), (ii) electrospraying (ES), and (iii) coaxial electrospraying (CA). The nanoparticles transported antisense oligonucleotides (ASOs), either encapsulated (CA nanocomplexes) or electrostatically bound externally (NP and ES nanocomplexes). After synthesis, the PEI/ASO nanoconjugates were functionalized with a muscle-specific RNA aptamer. Using this combinatorial formulation methodology, we obtained nanocomplexes that were further used as nanocarriers for the delivery of RNA therapeutics (ASO), specifically into muscle cells. In particular, we performed a detailed confocal microscopy-based comparative study to analyze the overall transfection efficiency, the cell-to-cell homogeneity, and the mean fluorescence intensity per cell of micron-sized domains enriched with the nanocomplexes. Furthermore, using high-magnification electron microscopy, we were able to describe, in detail, the ultrastructural basis of the cellular uptake and intracellular trafficking of nanocomplexes by the clathrin-independent endocytic pathway. Our results are a clear demonstration that coaxial electrospraying is a promising methodology for the synthesis of therapeutic nanoparticle-based carriers. Some of the principal features that the nanoparticles synthesized by coaxial electrospraying exhibit are efficient RNA-based drug encapsulation, increased nanoparticle surface availability for aptamer functionalization, a high transfection efficiency, and hyperactivation of the endocytosis and early/late endosome route as the main intracellular uptake mechanism.


Assuntos
Nanopartículas , Polietilenoimina , Células Musculares , Nanoconjugados , Nanopartículas/química , Polietilenoimina/química , RNA , Transfecção
20.
Sci Rep ; 12(1): 13408, 2022 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-35927565

RESUMO

Composites of polymer and graphene-based nanomaterials (GBNs) combine easy processing onto porous 3D membrane geometries due to the polymer and cellular differentiation stimuli due to GBNs fillers. Aiming to step forward to the clinical application of polymer/GBNs composites, this study performs a systematic and detailed comparative analysis of the influence of the properties of four different GBNs: (i) graphene oxide obtained from graphite chemically processes (GO); (ii) reduced graphene oxide (rGO); (iii) multilayered graphene produced by mechanical exfoliation method (Gmec); and (iv) low-oxidized graphene via anodic exfoliation (Ganodic); dispersed in polycaprolactone (PCL) porous membranes to induce astrocytic differentiation. PCL/GBN flat membranes were fabricated by phase inversion technique and broadly characterized in morphology and topography, chemical structure, hydrophilicity, protein adsorption, and electrical properties. Cellular assays with rat C6 glioma cells, as model for cell-specific astrocytes, were performed. Remarkably, low GBN loading (0.67 wt%) caused an important difference in the response of the C6 differentiation among PCL/GBN membranes. PCL/rGO and PCL/GO membranes presented the highest biomolecule markers for astrocyte differentiation. Our results pointed to the chemical structural defects in rGO and GO nanomaterials and the protein adsorption mechanisms as the most plausible cause conferring distinctive properties to PCL/GBN membranes for the promotion of astrocytic differentiation. Overall, our systematic comparative study provides generalizable conclusions and new evidences to discern the role of GBNs features for future research on 3D PCL/graphene composite hollow fiber membranes for in vitro neural models.


Assuntos
Grafite , Nanoestruturas , Animais , Grafite/química , Poliésteres/química , Polímeros , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA