Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Acta Physiol (Oxf) ; 240(6): e14146, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38606882

RESUMO

AIM: The Repressor Element-1 Silencing Transcription Factor (REST) is an epigenetic master regulator playing a crucial role in the nervous system. In early developmental stages, REST downregulation promotes neuronal differentiation and the acquisition of the neuronal phenotype. In addition, postnatal fluctuations in REST expression contribute to shaping neuronal networks and maintaining network homeostasis. Here we investigate the role of the early postnatal deletion of neuronal REST in the assembly and strength of excitatory and inhibitory synaptic connections. METHODS: We investigated excitatory and inhibitory synaptic transmission by patch-clamp recordings in acute neocortical slices in a conditional knockout mouse model (RestGTi) in which Rest was deleted by delivering PHP.eB adeno-associated viruses encoding CRE recombinase under the control of the human synapsin I promoter in the lateral ventricles of P0-P1 pups. RESULTS: We show that, under physiological conditions, Rest deletion increased the intrinsic excitability of principal cortical neurons in the primary visual cortex and the density and strength of excitatory synaptic connections impinging on them, without affecting inhibitory transmission. Conversely, in the presence of a pathological excitation/inhibition imbalance induced by pentylenetetrazol, Rest deletion prevented the increase in synaptic excitation and decreased seizure severity. CONCLUSION: The data indicate that REST exerts distinct effects on the excitability of cortical circuits depending on whether it acts under physiological conditions or in the presence of pathologic network hyperexcitability. In the former case, REST preserves a correct excitatory/inhibitory balance in cortical circuits, while in the latter REST loses its homeostatic activity and may become pro-epileptogenic.


Assuntos
Córtex Cerebral , Homeostase , Proteínas Repressoras , Animais , Camundongos , Córtex Cerebral/metabolismo , Córtex Cerebral/fisiologia , Homeostase/fisiologia , Camundongos Knockout , Rede Nervosa/fisiologia , Rede Nervosa/metabolismo , Neurônios/metabolismo , Neurônios/fisiologia , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Convulsões/genética , Convulsões/metabolismo , Convulsões/fisiopatologia , Transmissão Sináptica/fisiologia
2.
iScience ; 27(4): 109507, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38591011

RESUMO

Visual cortical plasticity is high during early life, but gradually decreases with development. This is due to the Otx2-driven maturation of intracortical inhibition that parallels the condensation of extracellular matrix components into perineuronal nets mainly around parvalbumin-positive GABAergic neurons. Repressor Element 1 Silencing Transcription (REST) epigenetically controls the expression of a plethora of neuron-specific genes. We demonstrate that the conditional knockout of REST in the primary visual cortex of adult mice induces a shift of ocular dominance after short-term monocular deprivation and promotes the recovery of vision in long-term deprived animals after reverse suture. These phenomena paralleled a reduction of perineuronal net density and increased expression of REST target genes, but not of the homeoprotein Otx2 in the visual cortex contralateral to the deprived eye. This shows that REST regulates adult visual cortical plasticity and is a potential therapeutic target to restore vision in adult amblyopia by enhancing V1 plasticity.

3.
Front Cell Neurosci ; 17: 1267609, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38034589

RESUMO

The repressor element-1 silencing transcription factor/neuron-restrictive silencer factor (REST/NRSF) is an epigenetic master regulator that plays a crucial role during nervous system development and maturation. REST function was originally described during development, where it determines neuronal phenotype. However, recent studies showed that REST participates in several processes in the adult brain, including neuronal plasticity and epileptogenesis. In this regard, the relationships between REST and epilepsy are still controversial and need further investigation. As forebrain excitatory neurons are the common final pathway of seizure susceptibility, we investigated the role of REST in epilepsy by inducing REST conditional knockout (REST-cKO) specifically in excitatory neurons of the hippocampus. To target the excitatory neuronal population, we cloned the calcium/calmodulin-dependent protein kinase IIα minimal promoter upstream of Cre recombinase. After assessing the specificity of the promoter's expression, the transgenes were packaged in an engineered adeno-associated virus able to cross the blood-brain and blood-cerebrospinal fluid barriers and delivered in the lateral ventricles of 2-month-old RESTflox/flox mice to characterize, after 1 month, the cognitive phenotype and the seizure propensity. We show that REST-cKO mice display lower levels of anxiety in the light-dark test with respect to control mice but have unaltered motor, social, and cognitive profiles. The evaluation of the susceptibility to epileptic seizures showed that REST-cKO mice are more resistant to pentylenetetrazole-induced kindling but not to seizures induced by a single administration of the convulsant and show higher survival rates. Overall, these data suggest that the absence of REST in forebrain excitatory neurons decreases seizure susceptibility, pointing to a pro-epileptogenic role of the transcriptional repressor under conditions of pathological excitation/inhibition imbalance.

5.
J Neurochem ; 165(5): 701-721, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36636908

RESUMO

Neuron-restrictive silencer factor/repressor element 1 (RE1)-silencing transcription factor (NRSF/REST) is a transcriptional repressor of a large cluster of neural genes containing RE1 motifs in their promoter region. NRSF/REST is ubiquitously expressed in non-neuronal cells, including astrocytes, while it is down-regulated during neuronal differentiation. While neuronal NRSF/REST homeostatically regulates intrinsic excitability and synaptic transmission, the role of the high NRSF/REST expression levels in the homeostatic functions of astrocytes is poorly understood. Here, we investigated the functional consequences of NRSF/REST deletion in primary cortical astrocytes derived from NRSF/REST conditional knockout mice (KO). We found that NRSF/REST KO astrocyte displayed a markedly reduced activity of inward rectifying K+ channels subtype 4.1 (Kir4.1) underlying spatial K+ buffering that was associated with a decreased expression and activity of the glutamate transporter-1 (GLT-1) responsible for glutamate uptake by astrocytes. The effects of the impaired astrocyte homeostatic functions on neuronal activity were investigated by co-culturing wild-type hippocampal neurons with NRSF/REST KO astrocytes. Interestingly, neurons experienced increased neuronal excitability at high firing rates associated with decrease after hyperpolarization and increased amplitude of excitatory postsynaptic currents. The data indicate that astrocytic NRSF/REST directly participates in neural circuit homeostasis by regulating intrinsic excitability and excitatory transmission and that dysfunctions of NRSF/REST expression in astrocytes may contribute to the pathogenesis of neurological disorders.


Assuntos
Astrócitos , Fatores de Transcrição , Camundongos , Animais , Fatores de Transcrição/genética , Astrócitos/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Regulação da Expressão Gênica
6.
Aging Cell ; 20(10): e13471, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34520100

RESUMO

During aging, brain performances decline. Cellular senescence is one of the aging drivers and a key feature of a variety of human age-related disorders. The transcriptional repressor RE1-silencing transcription factor (REST) has been associated with aging and higher risk of neurodegenerative disorders. However, how REST contributes to the senescence program and functional impairment remains largely unknown. Here, we report that REST is essential to prevent the senescence phenotype in primary mouse neurons. REST deficiency causes failure of autophagy and loss of proteostasis, increased oxidative stress, and higher rate of cell death. Re-establishment of autophagy reverses the main hallmarks of senescence. Our data indicate that REST has a protective role in physiological aging by regulating the autophagic flux and the senescence program in neurons, with implications for neurological disorders associated with aging.


Assuntos
Autofagia/genética , Senescência Celular/genética , Neurônios/metabolismo , Proteínas Repressoras/deficiência , Animais , Humanos , Camundongos , Estresse Oxidativo
7.
Cell Death Dis ; 12(2): 180, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33589593

RESUMO

Neuroinflammation is associated with synapse dysfunction and cognitive decline in patients and animal models. One candidate for translating the inflammatory stress into structural and functional changes in neural networks is the transcriptional repressor RE1-silencing transcription factor (REST) that regulates the expression of a wide cluster of neuron-specific genes during neurogenesis and in mature neurons. To study the cellular and molecular pathways activated under inflammatory conditions mimicking the experimental autoimmune encephalomyelitis (EAE) environment, we analyzed REST activity in neuroblastoma cells and mouse cortical neurons treated with activated T cell or microglia supernatant and distinct pro-inflammatory cytokines. We found that REST is activated by a variety of neuroinflammatory stimuli in both neuroblastoma cells and primary neurons, indicating that a vast transcriptional change is triggered during neuroinflammation. While a dual activation of REST and its dominant-negative splicing isoform REST4 was observed in N2a neuroblastoma cells, primary neurons responded with a pure full-length REST upregulation in the absence of changes in REST4 expression. In both cases, REST upregulation was associated with activation of Wnt signaling and increased nuclear translocation of ß-catenin, a well-known intracellular transduction pathway in neuroinflammation. Among single cytokines, IL-1ß caused a potent and prompt increase in REST transcription and translation in neurons, which promoted a delayed and strong synaptic downscaling specific for excitatory synapses, with decreased frequency and amplitude of spontaneous synaptic currents, decreased density of excitatory synaptic connections, and decreased frequency of action potential-evoked Ca2+ transients. Most important, the IL-1ß effects on excitatory transmission were strictly REST dependent, as conditional deletion of REST completely occluded the effects of IL-1ß activation on synaptic transmission and network excitability. Our results demonstrate that REST upregulation represents a new pathogenic mechanism for the synaptic dysfunctions observed under neuroinflammatory conditions and identify the REST pathway as therapeutic target for EAE and, potentially, for multiple sclerosis.


Assuntos
Córtex Cerebral/metabolismo , Inflamação/metabolismo , Interleucina-1beta/farmacologia , Proteínas Repressoras/metabolismo , Transmissão Sináptica , Animais , Córtex Cerebral/citologia , Técnicas de Cocultura , Meios de Cultivo Condicionados , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/citologia , Microglia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Neurônios/patologia , Proteínas Repressoras/biossíntese , Transmissão Sináptica/efeitos dos fármacos , Linfócitos T/metabolismo , Regulação para Cima
8.
Sci Rep ; 8(1): 8097, 2018 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-29802307

RESUMO

TAR DNA-binding protein 43 (TDP-43) is a key player in neurodegenerative diseases including frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS). Accumulation of TDP-43 is associated with neuronal death in the brain. How increased and disease-causing mutant forms of TDP-43 induce cell death remains unclear. Here we addressed the role of TDP-43 during neural development and show that reduced TDP-43 causes defects in neural stem/progenitor cell proliferation but not cell death. However, overexpression of wild type and TDP-43A315T proteins induce p53-dependent apoptosis of neural stem/progenitors and human induced pluripotent cell (iPS)-derived immature cortical neurons. We show that TDP-43 induces expression of the proapoptotic BH3-only genes Bbc3 and Bax, and that p53 inhibition rescues TDP-43 induced cell death of embryonic mouse, and human cortical neurons, including those derived from TDP-43G298S ALS patient iPS cells. Hence, an increase in wild type and mutant TDP-43 induces p53-dependent cell death in neural progenitors developing neurons and this can be rescued. These findings may have important implications for accumulated or mutant TDP-43 induced neurodegenerative diseases.


Assuntos
Apoptose , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Neurais/citologia , Neurônios/citologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Ciclo Celular , Inibidor de Quinase Dependente de Ciclina p21/genética , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Mutação , Neurogênese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
9.
Oncotarget ; 8(11): 17428-17442, 2017 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-28407697

RESUMO

The Hsp90 family of molecular chaperones includes the cytosolic isoforms Hsp90α and Hsp90ß, and the mitochondrial isoform Trap1. Hsp90α/ß support a large number of client proteins in the cytoplasm and the nucleus whereas Trap1 regulates oxidative phosphorylation in mitochondria. Many of the associated proteins and cellular processes are relevant to cancer, and there is ample pharmacological and genetic evidence to support the idea that Hsp90α/ß and Trap1 are required for tumorigenesis. However, a direct and comparative genetic test in a mouse cancer model has not been done. Here we report the effects of deleting the Hsp90α or Trap1 genes in a mouse model of breast cancer. Neither Hsp90α nor Trap1 are absolutely required for mammary tumor initiation, growth and metastasis induced by the polyoma middle T-antigen as oncogene. However, they do modulate growth and lung metastasis in vivo and cell proliferation, migration and invasion of isolated primary carcinoma cells in vitro. Without Hsp90α, tumor burden and metastasis are reduced, correlating with impaired proliferation, migration and invasion of cells in culture. Without Trap1, the appearance of tumors is initially delayed, and isolated cells are affected similarly to those without Hsp90α. Analysis of expression data of human breast cancers supports the conclusion that this is a valid mouse model highlighting the importance of these molecular chaperones.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Animais , Neoplasias da Mama/patologia , Transformação Celular Neoplásica , Citosol/metabolismo , Modelos Animais de Doenças , Feminino , Immunoblotting , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Isoformas de Proteínas
10.
Nucleic Acids Res ; 45(6): 3102-3115, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-27956497

RESUMO

The dynamic interaction of DNA methylation and transcription factor binding in regulating spatiotemporal gene expression is essential for embryogenesis, but the underlying mechanisms remain understudied. In this study, using mouse models and integration of in vitro and in vivo genetic and epigenetic analyses, we show that the binding of REST (repressor element 1 (RE1) silencing transcription factor; also known as NRSF) to its cognate RE1 sequences is temporally regulated by non-CpG methylation. This process is dependent on DNA methyltransferase 3B (DNMT3B) and leads to suppression of adult cardiac genes in developing hearts. We demonstrate that DNMT3B preferentially mediates non-CpG methylation of REST-targeted genes in the developing heart. Downregulation of DNMT3B results in decreased non-CpG methylation of RE1 sequences, reduced REST occupancy, and consequently release of the transcription suppression during later cardiac development. Together, these findings reveal a critical gene silencing mechanism in developing mammalian hearts that is regulated by the dynamic interaction of DNMT3B-mediated non-CpG methylation and REST binding.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Coração/embriologia , Miocárdio/metabolismo , Proteínas Repressoras/metabolismo , Animais , Sítios de Ligação , Células Cultivadas , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/fisiologia , Metilação de DNA , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/biossíntese , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Camundongos , Camundongos Endogâmicos C57BL , Canais de Potássio/biossíntese , Canais de Potássio/genética , Ligação Proteica , Proteínas Repressoras/fisiologia , DNA Metiltransferase 3B
11.
Mol Cell Biol ; 37(5)2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-27956701

RESUMO

Pf1, also known as Phf12 (plant homeodomain [PHD] zinc finger protein 12), is a member of the PHD zinc finger family of proteins. Pf1 associates with a chromatin-interacting protein complex comprised of MRG15, Sin3B, and histone deacetylase 1 (HDAC1) that functions as a transcriptional modulator. The biological function of Pf1 remains largely elusive. We undertook the generation of Pf1 knockout mice to elucidate its physiological role. We demonstrate that Pf1 is required for mid- to late gestation viability. Pf1 inactivation impairs the proliferative potential of mouse embryonic fibroblasts (MEFs) and is associated with a significant decrease in bromodeoxyuridine incorporation; an increase in senescence-associated ß-galactosidase (SA-ß-Gal) activity, a marker of cellular senescence; and elevated levels of phosphorylated H2AX (γ-H2A.X), a marker associated with DNA double-strand breaks. Analysis of transcripts differentially expressed in wild-type and Pf1-deficient cells revealed the impact of Pf1 in multiple regulatory arms of the ribosome biogenesis pathways. Strikingly, assessment of the morphology of the nucleoli exposed an abnormal nucleolar structure in Pf1-deficient cells. Finally, proteomic analysis of the Pf1-interacting complexes highlighted proteins involved in ribosome biogenesis. Taken together, our data reveal an unsuspected function for the Pf1-associated chromatin complex in the ribosomal biogenesis and senescence pathways.


Assuntos
Nucléolo Celular/metabolismo , Senescência Celular , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Homeodomínio/metabolismo , Células 3T3 , Animais , Proteínas Cromossômicas não Histona/genética , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Ontologia Genética , Proteínas de Homeodomínio/genética , Camundongos , Biogênese de Organelas , Gravidez , Ligação Proteica , Mapas de Interação de Proteínas , Proteômica , Processamento Pós-Transcricional do RNA/genética , RNA Ribossômico/genética , Proteínas Repressoras , Ribossomos/metabolismo
12.
PLoS One ; 11(10): e0164298, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27788151

RESUMO

Sphingolipids and the derived gangliosides have critical functions in spermatogenesis, thus mutations in genes involved in sphingolipid biogenesis are often associated with male infertility. We have generated a transgenic mouse line carrying an insertion in the sphingomyelin synthase gene Sms1, the enzyme which generates sphingomyelin species in the Golgi apparatus. We describe the spermatogenesis defect of Sms1-/- mice, which is characterized by sloughing of spermatocytes and spermatids, causing progressive infertility of male homozygotes. Lipid profiling revealed a reduction in several long chain unsaturated phosphatidylcholins, lysophosphatidylcholins and sphingolipids in the testes of mutants. Multi-Spectral Optoacoustic Tomography indicated blood-testis barrier dysfunction. A supplementary diet of the essential omega-3 docosahexaenoic acid and eicosapentaenoic acid diminished germ cell sloughing from the seminiferous epithelium and restored spermatogenesis and fertility in 50% of previously infertile mutants. Our findings indicate that SMS1 has a wider than anticipated role in testis polyunsaturated fatty acid homeostasis and for male fertility.


Assuntos
Fertilidade , Transferases (Outros Grupos de Fosfato Substituídos)/metabolismo , Envelhecimento/fisiologia , Processamento Alternativo , Animais , Epididimo/efeitos dos fármacos , Epididimo/metabolismo , Ácidos Graxos Ômega-3/biossíntese , Ácidos Graxos Ômega-3/farmacologia , Fertilidade/efeitos dos fármacos , Infertilidade Masculina/enzimologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Mutagênese Insercional , Regiões Promotoras Genéticas/genética , Espermatogênese/efeitos dos fármacos , Testículo/efeitos dos fármacos , Testículo/metabolismo , Transferases (Outros Grupos de Fosfato Substituídos)/genética
13.
Elife ; 5: e09584, 2016 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-26745185

RESUMO

The timely transition from neural progenitor to post-mitotic neuron requires down-regulation and loss of the neuronal transcriptional repressor, REST. Here, we have used mice containing a gene trap in the Rest gene, eliminating transcription from all coding exons, to remove REST prematurely from neural progenitors. We find that catastrophic DNA damage occurs during S-phase of the cell cycle, with long-term consequences including abnormal chromosome separation, apoptosis, and smaller brains. Persistent effects are evident by latent appearance of proneural glioblastoma in adult mice deleted additionally for the tumor suppressor p53 protein (p53). A previous line of mice deleted for REST in progenitors by conventional gene targeting does not exhibit these phenotypes, likely due to a remaining C-terminal peptide that still binds chromatin and recruits co-repressors. Our results suggest that REST-mediated chromatin remodeling is required in neural progenitors for proper S-phase dynamics, as part of its well-established role in repressing neuronal genes until terminal differentiation.


Assuntos
Encéfalo/embriologia , Diferenciação Celular , Neurogênese , Neurônios/fisiologia , Proteínas Repressoras/metabolismo , Células-Tronco/fisiologia , Animais , Ciclo Celular , Técnicas de Silenciamento de Genes , Camundongos
14.
Nat Commun ; 6: 6669, 2015 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-25808990

RESUMO

Receptor families of the innate immune response engage in 'cross-talk' to tailor optimal immune responses against invading pathogens. However, these responses are subject to multiple levels of regulation to keep in check aberrant inflammatory signals. Here, we describe a role for the orphan receptor interleukin-17 receptor D (IL-17RD) in negatively regulating Toll-like receptor (TLR)-induced responses. Deficiency of IL-17RD expression in cells leads to enhanced pro-inflammatory signalling and gene expression in response to TLR stimulation, and Il17rd(-/-) mice are more susceptible to TLR-induced septic shock. We demonstrate that the intracellular Sef/IL-17R (SEFIR) domain of IL-17RD targets TIR adaptor proteins to inhibit TLR downstream signalling thus revealing a paradigm involving cross-regulation of members of the IL-17R and TLR families.


Assuntos
Regulação da Expressão Gênica , Imunidade Inata/imunologia , Fatores Reguladores de Interferon/imunologia , NF-kappa B/imunologia , Receptores de Interleucina/imunologia , Choque Séptico/imunologia , Receptores Toll-Like/imunologia , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Linhagem Celular , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Imunidade Inata/genética , Inflamação , Fatores Reguladores de Interferon/genética , Camundongos , Camundongos Knockout , Estrutura Terciária de Proteína , Receptores de Interleucina/genética , Choque Séptico/genética , Transdução de Sinais , Receptores Toll-Like/metabolismo
15.
Hum Mol Genet ; 24(8): 2247-66, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25552653

RESUMO

Mitochondrial diseases often exhibit tissue-specific pathologies, but this phenomenon is poorly understood. Here we present regulation of mitochondrial translation by the Mitochondrial Translation Optimization Factor 1, MTO1, as a novel player in this scenario. We demonstrate that MTO1 mediates tRNA modification and controls mitochondrial translation rate in a highly tissue-specific manner associated with tissue-specific OXPHOS defects. Activation of mitochondrial proteases, aberrant translation products, as well as defects in OXPHOS complex assembly observed in MTO1 deficient mice further imply that MTO1 impacts translation fidelity. In our mouse model, MTO1-related OXPHOS deficiency can be bypassed by feeding a ketogenic diet. This therapeutic intervention is independent of the MTO1-mediated tRNA modification and involves balancing of mitochondrial and cellular secondary stress responses. Our results thereby establish mammalian MTO1 as a novel factor in the tissue-specific regulation of OXPHOS and fine tuning of mitochondrial translation accuracy.


Assuntos
Proteínas de Transporte/metabolismo , Doenças Mitocondriais/tratamento farmacológico , Doenças Mitocondriais/metabolismo , Fosforilação Oxidativa , Biossíntese de Proteínas , RNA de Transferência/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Transporte/química , Proteínas de Transporte/genética , Dieta Cetogênica , Fibroblastos/metabolismo , Humanos , Camundongos , Mitocôndrias/metabolismo , Doenças Mitocondriais/genética , Proteínas Mitocondriais , Dados de Sequência Molecular , Especificidade de Órgãos , RNA de Transferência/genética , Proteínas de Ligação a RNA , Alinhamento de Sequência
16.
Development ; 142(1): 31-40, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25516968

RESUMO

The maintenance and control of pluripotency is of great interest in stem cell biology. The dual specificity T-box/basic-helix-loop-helix-zipper transcription factor Mga is expressed in the pluripotent cells of the inner cell mass (ICM) and epiblast of the peri-implantation mouse embryo, but its function has not been investigated previously. Here, we use a loss-of-function allele and RNA knockdown to demonstrate that Mga depletion leads to the death of proliferating pluripotent ICM cells in vivo and in vitro, and the death of embryonic stem cells (ESCs) in vitro. Additionally, quiescent pluripotent cells lacking Mga are lost during embryonic diapause. Expression of Odc1, the rate-limiting enzyme in the conversion of ornithine into putrescine in the synthesis of polyamines, is reduced in Mga mutant cells, and the survival of mutant ICM cells as well as ESCs is rescued in culture by the addition of exogenous putrescine. These results suggest a mechanism whereby Mga influences pluripotent cell survival through regulation of the polyamine pool in pluripotent cells of the embryo, whether they are in a proliferative or quiescent state.


Assuntos
Implantação do Embrião , Células-Tronco Pluripotentes/citologia , Fatores de Transcrição/metabolismo , Alelos , Animais , Apoptose , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Massa Celular Interna do Blastocisto/citologia , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Cruzamentos Genéticos , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Marcação de Genes , Genótipo , Camadas Germinativas/citologia , Camadas Germinativas/metabolismo , Masculino , Camundongos , Mutagênese/genética , Mutação/genética , Ornitina Descarboxilase/metabolismo , Células-Tronco Pluripotentes/metabolismo , Poliaminas/metabolismo , Fatores de Transcrição/deficiência
17.
PLoS One ; 9(12): e114918, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25506927

RESUMO

Recently, mutations in the mitochondrial translation optimization factor 1 gene (MTO1) were identified as causative in children with hypertrophic cardiomyopathy, lactic acidosis and respiratory chain defect. Here, we describe an MTO1-deficient mouse model generated by gene trap mutagenesis that mirrors the human phenotype remarkably well. As in patients, the most prominent signs and symptoms were cardiovascular and included bradycardia and cardiomyopathy. In addition, the mutant mice showed a marked worsening of arrhythmias during induction and reversal of anaesthesia. The detailed morphological and biochemical workup of murine hearts indicated that the myocardial damage was due to complex I deficiency and mitochondrial dysfunction. In contrast, neurological examination was largely normal in Mto1-deficient mice. A translational consequence of this mouse model may be to caution against anaesthesia-related cardiac arrhythmias which may be fatal in patients.


Assuntos
Cardiomiopatias/genética , Cardiomiopatias/fisiopatologia , Proteínas de Transporte/genética , Técnicas de Silenciamento de Genes , Coração/fisiopatologia , Miocárdio/patologia , Animais , Cardiomiopatias/patologia , DNA Mitocondrial/genética , Modelos Animais de Doenças , Complexo I de Transporte de Elétrons/genética , Feminino , Dosagem de Genes , Genes Mitocondriais , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas Mitocondriais , Fosforilação Oxidativa , Proteínas de Ligação a RNA
19.
PLoS One ; 9(8): e105478, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25127017

RESUMO

The TAR DNA binding protein (TDP-43) was originally identified as a host cell factor binding to the HIV-1 LTR and thereby suppressing HIV-1 transcription and gene expression (Ou et al., J.Virol. 1995, 69(6):3584). TDP-43 is a global regulator of transcription, can influence RNA metabolism in many different ways and is ubiquitously expressed. Thus, TDP-43 could be a major factor restricting HIV-1 replication at the level of LTR transcription and gene expression. These facts prompted us to revisit the role of TDP-43 for HIV-1 replication. We utilized established HIV-1 cell culture systems as well as primary cell models and performed a comprehensive analysis of TDP-43 function and investigated its putative impact on HIV-1 gene expression. In HIV-1 infected cells TDP-43 was neither degraded nor sequestered from the nucleus. Furthermore, TDP-43 overexpression as well as siRNA mediated knockdown did not affect HIV-1 gene expression and virus production in T cells and macrophages. In summary, our experiments argue against a restricting role of TDP-43 during HIV-1 replication in immune cells.


Assuntos
Proteínas de Ligação a DNA/metabolismo , HIV-1/fisiologia , Replicação Viral , Proteínas de Ligação a DNA/genética , Expressão Gênica , Regulação Viral da Expressão Gênica , Genes Virais , Células HEK293 , Repetição Terminal Longa de HIV , Humanos , Células Jurkat , Transporte Proteico , Ativação Transcricional
20.
Circ Res ; 115(1): 23-31, 2014 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-24748541

RESUMO

RATIONALE: 22q11 deletion syndrome arises from recombination between low-copy repeats on chromosome 22. Typical deletions result in hemizygosity for TBX1 associated with congenital cardiovascular disease. Deletions distal to the typically deleted region result in a similar cardiac phenotype but lack in extracardiac features of the syndrome, suggesting that a second haploinsufficient gene maps to this interval. OBJECTIVE: The transcription factor HIC2 is lost in most distal deletions, as well as in a minority of typical deletions. We used mouse models to test the hypothesis that HIC2 hemizygosity causes congenital heart disease. METHODS AND RESULTS: We created a genetrap mouse allele of Hic2. The genetrap reporter was expressed in the heart throughout the key stages of cardiac morphogenesis. Homozygosity for the genetrap allele was embryonic lethal before embryonic day E10.5, whereas the heterozygous condition exhibited a partially penetrant late lethality. One third of heterozygous embryos had a cardiac phenotype. MRI demonstrated a ventricular septal defect with over-riding aorta. Conditional targeting indicated a requirement for Hic2 within the Nkx2.5+ and Mesp1+ cardiovascular progenitor lineages. Microarray analysis revealed increased expression of Bmp10. CONCLUSIONS: Our results demonstrate a novel role for Hic2 in cardiac development. Hic2 is the first gene within the distal 22q11 interval to have a demonstrated haploinsufficient cardiac phenotype in mice. Together our data suggest that HIC2 haploinsufficiency likely contributes to the cardiac defects seen in distal 22q11 deletion syndrome.


Assuntos
Síndrome da Deleção 22q11/etiologia , Coração/embriologia , Fatores de Transcrição Kruppel-Like/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Síndrome da Deleção 22q11/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Proteínas Morfogenéticas Ósseas/fisiologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Cardiopatias Congênitas/etiologia , Humanos , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Morfogênese , Mutagênese , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Proteínas com Domínio T/genética , Proteínas com Domínio T/fisiologia , Proteínas Supressoras de Tumor/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...