Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Cell ; 163(1): 246-55, 2015 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-26406379

RESUMO

We present ChromATin, a quantitative high-resolution imaging approach for investigating chromatin organization in complex tissues. This method combines analysis of epigenetic modifications by immunostaining, localization of specific DNA sequences by FISH, and high-resolution segregation of nuclear compartments using array tomography (AT) imaging. We then apply this approach to examine how the genome is organized in the mammalian brain using female Rett syndrome mice, which are a mosaic of normal and Mecp2-null cells. Side-by-side comparisons within the same field reveal distinct heterochromatin territories in wild-type neurons that are altered in Mecp2-null nuclei. Mutant neurons exhibit increased chromatin compaction and a striking redistribution of the H4K20me3 histone modification into pericentromeric heterochromatin, a territory occupied normally by MeCP2. These events are not observed in every neuronal cell type, highlighting ChromATin as a powerful in situ method for examining cell-type-specific differences in chromatin architecture in complex tissues.


Assuntos
Encéfalo/metabolismo , Cromatina/metabolismo , Processamento de Imagem Assistida por Computador/métodos , Síndrome de Rett/metabolismo , Tomografia/métodos , Animais , Núcleo Celular/metabolismo , Feminino , Heterocromatina/metabolismo , Histonas/metabolismo , Hibridização in Situ Fluorescente , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos , Neurônios/metabolismo , Síndrome de Rett/genética
2.
Proc Natl Acad Sci U S A ; 119(33): e2206053119, 2022 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-35939700

RESUMO

Rett syndrome is a neurological disease due to loss-of-function mutations in the transcription factor, Methyl CpG binding protein 2 (MECP2). Because overexpression of endogenous MECP2 also causes disease, we have exploited a targeted RNA-editing approach to repair patient mutations where levels of MECP2 protein will never exceed endogenous levels. Here, we have constructed adeno-associated viruses coexpressing a bioengineered wild-type ADAR2 catalytic domain (Editasewt) and either Mecp2-targeting or nontargeting gfp RNA guides. The viruses are introduced systemically into male mice containing a guanosine to adenosine mutation that eliminates MeCP2 protein and causes classic Rett syndrome in humans. We find that in the mutant mice injected with the Mecp2-targeting virus, the brainstem exhibits the highest RNA-editing frequency compared to other brain regions. The efficiency is sufficient to rescue MeCP2 expression and function in the brainstem of mice expressing the Mecp2-targeting virus. Correspondingly, we find that abnormal Rett-like respiratory patterns are alleviated, and survival is prolonged, compared to mice injected with the control gfp guide virus. The levels of RNA editing among most brain regions corresponds to the distribution of guide RNA rather than Editasewt. Our results provide evidence that a targeted RNA-editing approach can alleviate a hallmark symptom in a mouse model of human disease.


Assuntos
Tronco Encefálico , Proteína 2 de Ligação a Metil-CpG , Edição de RNA , Transtornos Respiratórios , Síndrome de Rett , Animais , Tronco Encefálico/metabolismo , Modelos Animais de Doenças , Humanos , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Mutação , Transtornos Respiratórios/genética , Transtornos Respiratórios/terapia , Síndrome de Rett/genética , Síndrome de Rett/terapia
3.
J Neurosci ; 41(31): 6582-6595, 2021 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-34210779

RESUMO

Early studies in mouse neurodevelopment led to the discovery of the RE1 Silencing Transcription Factor (REST) and its role as a master repressor of neuronal gene expression. Recently, REST was reported to also repress neuronal genes in the human adult brain. These genes were found to be involved in pro-apoptotic pathways; and their repression, associated with increased REST levels during aging, were found to be neuroprotective and conserved across species. However, direct genome-wide REST binding profiles for REST in adult brain have not been identified for any species. Here, we apply this approach to mouse and human hippocampus. We find an expansion of REST binding sites in the human hippocampus that are lacking in both mouse hippocampus and other human non-neuronal cell types. The unique human REST binding sites are associated with genes involved in innate immunity processes and inflammation signaling which, on the basis of histology and recent public transcriptomic analyses, suggest that these new target genes are repressed in glia. We propose that the increases in REST expression in mid-adulthood presage the beginning of brain aging, and that human REST function has evolved to protect the longevity and function of both neurons and glia in human brain.SIGNIFICANCE STATEMENT The RE1 Silencing Transcription Factor (REST) repressor has served historically as a model for gene regulation during mouse neurogenesis. Recent studies of REST have also suggested a conserved role for REST repressor function across lower species during aging. However, direct genome-wide studies for REST have been lacking for human brain. Here, we perform the first genome-wide analysis of REST binding in both human and mouse hippocampus. The majority of REST-occupied genes in human hippocampus are distinct from those in mouse. Further, the REST-associated genes unique to human hippocampus represent a new set related to innate immunity and inflammation, where their gene dysregulation has been implicated in aging-related neuropathology, such as Alzheimer's disease.


Assuntos
Envelhecimento/metabolismo , Hipocampo/metabolismo , Neuroglia/metabolismo , Proteínas Repressoras/metabolismo , Idoso , Envelhecimento/imunologia , Animais , Feminino , Estudo de Associação Genômica Ampla , Hipocampo/imunologia , Humanos , Imunidade Inata/fisiologia , Masculino , Camundongos , Pessoa de Meia-Idade , Neuroglia/imunologia , Neurônios/metabolismo , Proteínas Repressoras/imunologia
4.
Genome Res ; 29(5): 857-869, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30936163

RESUMO

Here we present a comprehensive map of the accessible chromatin landscape of the mouse hippocampus at single-cell resolution. Substantial advances of this work include the optimization of a single-cell combinatorial indexing assay for transposase accessible chromatin (sci-ATAC-seq); a software suite, scitools, for the rapid processing and visualization of single-cell combinatorial indexing data sets; and a valuable resource of hippocampal regulatory networks at single-cell resolution. We used sci-ATAC-seq to produce 2346 high-quality single-cell chromatin accessibility maps with a mean unique read count per cell of 29,201 from both fresh and frozen hippocampi, observing little difference in accessibility patterns between the preparations. By using this data set, we identified eight distinct major clusters of cells representing both neuronal and nonneuronal cell types and characterized the driving regulatory factors and differentially accessible loci that define each cluster. Within pyramidal neurons, we identified four major clusters, including CA1 and CA3 neurons, and three additional subclusters. We then applied a recently described coaccessibility framework, Cicero, which identified 146,818 links between promoters and putative distal regulatory DNA. Identified coaccessibility networks showed cell-type specificity, shedding light on key dynamic loci that reconfigure to specify hippocampal cell lineages. Lastly, we performed an additional sci-ATAC-seq preparation from cultured hippocampal neurons (899 high-quality cells, 43,532 mean unique reads) that revealed substantial alterations in their epigenetic landscape compared with nuclei from hippocampal tissue. This data set and accompanying analysis tools provide a new resource that can guide subsequent studies of the hippocampus.


Assuntos
Cromatina/genética , Hipocampo/metabolismo , Células Piramidais/metabolismo , Animais , Linhagem da Célula/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Cultivadas , Cromatina/metabolismo , Epigenômica/métodos , Camundongos , Plasticidade Neuronal/genética , Células Piramidais/citologia , Análise de Sequência de DNA , Análise de Célula Única/métodos , Transposases/genética , Transposases/metabolismo
5.
Proc Natl Acad Sci U S A ; 114(44): E9395-E9402, 2017 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-29078406

RESUMO

Rett syndrome (RTT) is a debilitating neurological disorder caused by mutations in the gene encoding the transcription factor Methyl CpG Binding Protein 2 (MECP2). A distinct disorder results from MECP2 gene duplication, suggesting that therapeutic approaches must restore close to normal levels of MECP2. Here, we apply the approach of site-directed RNA editing to repair, at the mRNA level, a disease-causing guanosine to adenosine (G > A) mutation in the mouse MeCP2 DNA binding domain. To mediate repair, we exploit the catalytic domain of Adenosine Deaminase Acting on RNA (ADAR2) that deaminates A to inosine (I) residues that are subsequently translated as G. We fuse the ADAR2 domain, tagged with a nuclear localization signal, to an RNA binding peptide from bacteriophage lambda. In cultured neurons from mice that harbor an RTT patient G > A mutation and express engineered ADAR2, along with an appropriate RNA guide to target the enzyme, 72% of Mecp2 mRNA is repaired. Levels of MeCP2 protein are also increased significantly. Importantly, as in wild-type neurons, the repaired MeCP2 protein is enriched in heterochromatic foci, reflecting restoration of normal MeCP2 binding to methylated DNA. This successful use of site-directed RNA editing to repair an endogenous mRNA and restore protein function opens the door to future in vivo applications to treat RTT and other diseases.


Assuntos
Proteína 2 de Ligação a Metil-CpG/genética , Neurônios/fisiologia , RNA/genética , Adenosina Desaminase/genética , Animais , Células Cultivadas , Metilação de DNA/genética , Modelos Animais de Doenças , Humanos , Camundongos , Mutação/genética , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Síndrome de Rett/genética
6.
Proc Natl Acad Sci U S A ; 114(3): E406-E415, 2017 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-28049845

RESUMO

The transcriptional events that lead to the cessation of neural proliferation, and therefore enable the production of proper numbers of differentiated neurons and glia, are still largely uncharacterized. Here, we report that the transcription factor Insulinoma-associated 1 (INSM1) forms complexes with RE1 Silencing Transcription factor (REST) corepressors RCOR1 and RCOR2 in progenitors in embryonic mouse brain. Mice lacking both RCOR1 and RCOR2 in developing brain die perinatally and generate an abnormally high number of neural progenitors at the expense of differentiated neurons and oligodendrocyte precursor cells. In addition, Rcor1/2 deletion detrimentally affects complex morphological processes such as closure of the interganglionic sulcus. We find that INSM1, a transcription factor that induces cell-cycle arrest, is coexpressed with RCOR1/2 in a subset of neural progenitors and forms complexes with RCOR1/2 in embryonic brain. Further, the Insm1-/- mouse phenocopies predominant brain phenotypes of the Rcor1/2 knockout. A large number of genes are concordantly misregulated in both knockout genotypes, and a majority of the down-regulated genes are targets of REST. Rest transcripts are up-regulated in both knockouts, and reducing transcripts to control levels in the Rcor1/2 knockout partially rescues the defect in interganglionic sulcus closure. Our findings indicate that an INSM1/RCOR1/2 complex controls the balance of proliferation and differentiation during brain development.


Assuntos
Encéfalo/fisiologia , Diferenciação Celular/genética , Proliferação de Células/genética , Proteínas Correpressoras/genética , Proteínas de Ligação a DNA/genética , Proteínas do Tecido Nervoso/genética , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Animais , Regulação para Baixo/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/fisiologia , Regulação para Cima/genética
7.
Proc Natl Acad Sci U S A ; 113(3): E378-86, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26715759

RESUMO

Synaptic depression is prominent among synapses, but the underlying mechanisms remain uncertain. Here, we use paired patch clamp recording to study neuromuscular transmission between the caudal primary motor neuron and target skeletal muscle in zebrafish. This synapse has an unusually low number of release sites, all with high probabilities of release in response to low-frequency stimulation. During high-frequency stimulation, the synapse undergoes short-term depression and reaches steady-state levels of transmission that sustain the swimming behavior. To determine the release parameters underlying this steady state, we applied variance analysis. Our analysis revealed two functionally distinct subclasses of release sites differing by over 60-fold in rates of vesicle reloading. A slow reloading class requires seconds to recover and contributes to depression onset but not the steady-state transmission. By contrast, a fast reloading class recovers within tens of milliseconds and is solely responsible for steady-state transmission. Thus, in contrast to most current models that assign levels of steady-state depression to vesicle availability, our findings instead assign this function to nonuniform release site kinetics. The duality of active-site properties accounts for the highly nonlinear dependence of steady-state depression levels on frequency.


Assuntos
Plasticidade Neuronal/fisiologia , Sinapses/fisiologia , Animais , Estimulação Elétrica , Proteínas de Fluorescência Verde/metabolismo , Camundongos Transgênicos , Neurônios Motores/fisiologia , Junção Neuromuscular/fisiologia , Probabilidade , Reprodutibilidade dos Testes , Fatores de Tempo , Peixe-Zebra/fisiologia
8.
Glia ; 66(9): 2024-2039, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30043519

RESUMO

The idea that astrocytes provide support for neurons has a long history, but whether neurons play an instructive role in these processes is poorly understood. To address this question, we co-culture astrocytes with genetically labeled neurons, permitting their separation by flow cytometry, and test whether the presence of neurons influences the astrocyte transcriptome. We find that numerous pathways are regulated in the co-cultured astrocytes, in a time-dependent matter coincident with synaptic maturation. In particular, the induction of glutathione metabolic genes is prominent, resulting in increased glutathione production. We show that the induction of the glutathione pathway is mediated by astrocytic metabotropic glutamate receptors. Using a candidate approach, we identify direct binding of the nuclear factor E2-related factor, NRF2, to several of the induced genes. Blocking nuclear accumulation of astrocytic NRF2 abolishes neuron-induced glutathione gene induction and glutathione production. Our results suggest that astrocyte transcriptional and metabolic profiles are tightly coupled to the activity of neurons, consistent with the model that astrocytes dynamically support healthy brain function.


Assuntos
Astrócitos/metabolismo , Regulação da Expressão Gênica/fisiologia , Glutationa/metabolismo , Neurônios/fisiologia , Animais , Astrócitos/citologia , Células Cultivadas , Técnicas de Cocultura , Hipocampo/citologia , Hipocampo/fisiologia , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , Neurônios/citologia , Receptores de Glutamato Metabotrópico/metabolismo , Transcriptoma
9.
Hum Mol Genet ; 25(9): 1690-702, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26908602

RESUMO

Germline mutations in the X-linked gene, methyl-CpG-binding protein 2 (MECP2), underlie most cases of Rett syndrome (RTT), an autism spectrum disorder affecting approximately one in 10 000 female live births. The disease is characterized in affected girls by a latent appearance of symptoms between 12 and 18 months of age while boys usually die before the age of two. The nature of the latency is not known, but RTT-like phenotypes are recapitulated in mouse models, even when MeCP2 is removed at different postnatal stages, including juvenile and adolescent stages. Unexpectedly, here, we show that within a very brief developmental window, between 10 (adolescent) and 15 (adult) weeks after birth, symptom initiation and progression upon removal of MeCP2 in male mice transitions from 3 to 4 months to only several days, followed by lethality. We further show that this accelerated development of RTT phenotype and lethality occur at the transition to adult stage (15 weeks of age) and persists thereafter. Importantly, within this abbreviated time frame of days, the brain acquires dramatic anatomical, cellular and molecular abnormalities, typical of classical RTT. This study reveals a new postnatal developmental stage, which coincides with full-brain maturation, where the structure/function of the brain is extremely sensitive to levels of MeCP2 and loss of MeCP2 leads to precipitous collapse of the neuronal networks and incompatibility with life within days.


Assuntos
Encéfalo/patologia , Modelos Animais de Doenças , Genes Ligados ao Cromossomo X/genética , Proteína 2 de Ligação a Metil-CpG/fisiologia , Neurônios/patologia , Síndrome de Rett/etiologia , Envelhecimento , Animais , Encéfalo/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Neurônios/metabolismo , Fenótipo , Síndrome de Rett/patologia
10.
Nature ; 475(7357): 497-500, 2011 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-21716289

RESUMO

Rett's syndrome (RTT) is an X-chromosome-linked autism spectrum disorder caused by loss of function of the transcription factor methyl-CpG-binding protein 2 (MeCP2). Although MeCP2 is expressed in most tissues, loss of MeCP2 expression results primarily in neurological symptoms. Earlier studies suggested the idea that RTT is due exclusively to loss of MeCP2 function in neurons. Although defective neurons clearly underlie the aberrant behaviours, we and others showed recently that the loss of MECP2 from glia negatively influences neurons in a non-cell-autonomous fashion. Here we show that in globally MeCP2-deficient mice, re-expression of Mecp2 preferentially in astrocytes significantly improved locomotion and anxiety levels, restored respiratory abnormalities to a normal pattern, and greatly prolonged lifespan compared to globally null mice. Furthermore, restoration of MeCP2 in the mutant astrocytes exerted a non-cell-autonomous positive effect on mutant neurons in vivo, restoring normal dendritic morphology and increasing levels of the excitatory glutamate transporter VGLUT1. Our study shows that glia, like neurons, are integral components of the neuropathology of RTT, and supports the targeting of glia as a strategy for improving the associated symptoms.


Assuntos
Neuroglia/metabolismo , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Animais , Ansiedade/metabolismo , Astrócitos/metabolismo , Comportamento Animal , Progressão da Doença , Feminino , Regulação da Expressão Gênica , Masculino , Proteína 2 de Ligação a Metil-CpG/deficiência , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora , Neuroglia/patologia , Neurônios/metabolismo , Síndrome de Rett/fisiopatologia , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo
11.
Proc Natl Acad Sci U S A ; 111(37): E3929-36, 2014 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-25197063

RESUMO

The repressor element 1 (RE1) silencing transcription factor (REST) in stem cells represses hundreds of genes essential to neuronal function. During neurogenesis, REST is degraded in neural progenitors to promote subsequent elaboration of a mature neuronal phenotype. Prior studies indicate that part of the degradation mechanism involves phosphorylation of two sites in the C terminus of REST that require activity of beta-transducin repeat containing E3 ubiquitin protein ligase, ßTrCP. We identify a proline-directed phosphorylation motif, at serines 861/864 upstream of these sites, which is a substrate for the peptidylprolyl cis/trans isomerase, Pin1, as well as the ERK1/2 kinases. Mutation at S861/864 stabilizes REST, as does inhibition of Pin1 activity. Interestingly, we find that C-terminal domain small phosphatase 1 (CTDSP1), which is recruited by REST to neuronal genes, is present in REST immunocomplexes, dephosphorylates S861/864, and stabilizes REST. Expression of a REST peptide containing S861/864 in neural progenitors inhibits terminal neuronal differentiation. Together with previous work indicating that both REST and CTDSP1 are expressed to high levels in stem cells and down-regulated during neurogenesis, our results suggest that CTDSP1 activity stabilizes REST in stem cells and that ERK-dependent phosphorylation combined with Pin1 activity promotes REST degradation in neural progenitors.


Assuntos
Diferenciação Celular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Proteínas Repressoras/metabolismo , Animais , Galinhas , Cromatina/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Mutação/genética , Peptidilprolil Isomerase de Interação com NIMA , Células PC12 , Peptídeos/metabolismo , Peptidilprolil Isomerase/metabolismo , Fosforilação , Fosfosserina/metabolismo , Ligação Proteica , Estabilidade Proteica , Proteólise , Ratos , Proteínas Contendo Repetições de beta-Transducina/metabolismo , Proteínas ras/metabolismo
12.
Blood ; 123(20): 3175-84, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24652990

RESUMO

The corepressor Rcor1 has been linked biochemically to hematopoiesis, but its function in vivo remains unknown. We show that mice deleted for Rcor1 are profoundly anemic and die in late gestation. Definitive erythroid cells from mutant mice arrest at the transition from proerythroblast to basophilic erythroblast. Remarkably, Rcor1 null erythroid progenitors cultured in vitro form myeloid colonies instead of erythroid colonies. The mutant proerythroblasts also aberrantly express genes of the myeloid lineage as well as genes typical of hematopoietic stem cells (HSCs) and/or progenitor cells. The colony-stimulating factor 2 receptor ß subunit (Csf2rb), which codes for a receptor implicated in myeloid cytokine signaling, is a direct target for both Rcor1 and the transcription repressor Gfi1b in erythroid cells. In the absence of Rcor1, the Csf2rb gene is highly induced, and Rcor1(-/-) progenitors exhibit CSF2-dependent phospho-Stat5 hypersensitivity. Blocking this pathway can partially reduce myeloid colony formation by Rcor1-deficient erythroid progenitors. Thus, Rcor1 promotes erythropoiesis by repressing HSC and/or progenitor genes, as well as the genes and signaling pathways that lead to myeloid cell fate.


Assuntos
Proteínas Correpressoras/metabolismo , Eritropoese , Animais , Células Cultivadas , Proteínas Correpressoras/genética , Subunidade beta Comum dos Receptores de Citocinas/metabolismo , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Eritroblastos/citologia , Eritroblastos/metabolismo , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patologia , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , Células Mieloides/citologia , Receptores de Interleucina-3/metabolismo , Transdução de Sinais
13.
Stem Cells ; 33(11): 3304-14, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26119982

RESUMO

Based on its physical interactions with histone-modifying enzymes, the transcriptional corepressor Rcor1 has been implicated in the epigenetic regulation blood cell development. Previously, we have demonstrated that Rcor1 is essential for the maturation of definitive erythroid cells and fetal survival. To determine the functional role of Rcor1 in steady-state hematopoiesis in the adult, we used a conditional knockout approach. Here, we show that the loss of Rcor1 expression results in the rapid onset of severe anemia due to a complete, cell autonomous block in the maturation of committed erythroid progenitors. By contrast, both the frequency of megakaryocyte progenitors and their capacity to produce platelets were normal. Although the frequency of common lymphoid progenitors and T cells was not altered, B cells were significantly reduced and showed increased apoptosis. However, Rcor1-deficient bone marrow sustained normal levels of B-cells following transplantation, indicating a non-cell autonomous requirement for Rcor1 in B-cell survival. Evaluation of the myelomonocytic lineage revealed an absence of mature neutrophils and a significant increase in the absolute number of monocytic cells. Rcor1-deficient monocytes were less apoptotic and showed ∼100-fold more colony-forming activity than their normal counterparts, but did not give rise to leukemia. Moreover, Rcor1(-/-) monocytes exhibited extensive, cytokine-dependent self-renewal and overexpressed genes associated with hematopoietic stem/progenitor cell expansion including Gata2, Meis1, and Hoxa9. Taken together, these data demonstrate that Rcor1 is essential for the normal differentiation of myeloerythroid progenitors and for appropriately regulating self-renewal activity in the monocyte lineage.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Proteínas Correpressoras/metabolismo , Células Precursoras Eritroides/metabolismo , Neutrófilos/metabolismo , Animais , Células Eritroides/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Monócitos/metabolismo
14.
Proc Natl Acad Sci U S A ; 110(16): 6518-23, 2013 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-23576758

RESUMO

Reactivation of fetal hemoglobin (HbF) in adults ameliorates the severity of the common ß-globin disorders. The transcription factor BCL11A is a critical modulator of hemoglobin switching and HbF silencing, yet the molecular mechanism through which BCL11A coordinates the developmental switch is incompletely understood. Particularly, the identities of BCL11A cooperating protein complexes and their roles in HbF expression and erythroid development remain largely unknown. Here we determine the interacting partner proteins of BCL11A in erythroid cells by a proteomic screen. BCL11A is found within multiprotein complexes consisting of erythroid transcription factors, transcriptional corepressors, and chromatin-modifying enzymes. We show that the lysine-specific demethylase 1 and repressor element-1 silencing transcription factor corepressor 1 (LSD1/CoREST) histone demethylase complex interacts with BCL11A and is required for full developmental silencing of mouse embryonic ß-like globin genes and human γ-globin genes in adult erythroid cells in vivo. In addition, LSD1 is essential for normal erythroid development. Furthermore, the DNA methyltransferase 1 (DNMT1) is identified as a BCL11A-associated protein in the proteomic screen. DNMT1 is required to maintain HbF silencing in primary human adult erythroid cells. DNMT1 haploinsufficiency combined with BCL11A deficiency further enhances γ-globin expression in adult animals. Our findings provide important insights into the mechanistic roles of BCL11A in HbF silencing and clues for therapeutic targeting of BCL11A in ß-hemoglobinopathies.


Assuntos
Proteínas de Transporte/farmacologia , Proteínas Correpressoras/metabolismo , Hemoglobina Fetal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/farmacologia , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Cromatografia Líquida , Células Precursoras Eritroides , Humanos , Camundongos , Proteínas Nucleares/metabolismo , Proteômica , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Repressoras , Espectrometria de Massas em Tandem , Globinas beta/metabolismo
15.
J Neurosci ; 34(50): 16650-61, 2014 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-25505318

RESUMO

The RE1 Silencing Transcription Factor (REST) acts as a governor of the mature neuronal phenotype by repressing a large consortium of neuronal genes in non-neuronal cells. In the developing nervous system, REST is present in progenitors and downregulated at terminal differentiation to promote acquisition of mature neuronal phenotypes. Paradoxically, REST is still detected in some regions of the adult nervous system, but how REST levels are regulated, and whether REST can still repress neuronal genes, is not known. Here, we report that homeostatic levels of REST are maintained in mature peripheral neurons by a constitutive post-transcriptional mechanism. Specifically, using a three-hybrid genetic screen, we identify the RNA binding protein, ZFP36L2, associated previously only with female fertility and hematopoiesis, and show that it regulates REST mRNA stability. Dorsal root ganglia in Zfp36l2 knock-out mice, or wild-type ganglia expressing ZFP36L2 shRNA, show higher steady-state levels of Rest mRNA and protein, and extend thin and disintegrating axons. This phenotype is due, at least in part, to abnormally elevated REST levels in the ganglia because the axonal phenotype is attenuated by acute knockdown of REST in Zfp36l2 KO DRG explants. The higher REST levels result in lower levels of target genes, indicating that REST can still fine-tune gene expression through repression. Thus, REST levels are titrated in mature peripheral neurons, in part through a ZFP36L2-mediated post-transcriptional mechanism, with consequences for axonal integrity.


Assuntos
Axônios/metabolismo , Gânglios Espinais/metabolismo , Proteínas de Ligação a RNA/biossíntese , Proteínas Repressoras/biossíntese , Tristetraprolina/biossíntese , Animais , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Knockout , Células PC12 , Proteínas de Ligação a RNA/genética , Ratos , Proteínas Repressoras/genética , Tristetraprolina/genética
16.
Proc Natl Acad Sci U S A ; 109(43): 17711-6, 2012 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-23045675

RESUMO

Mutations in muscle ACh receptors cause slow-channel syndrome (SCS) and Escobar syndrome, two forms of congenital myasthenia. SCS is a dominant disorder with mutations reported for all receptor subunits except γ. Escobar syndrome is distinct, with mutations located exclusively in γ, and characterized by developmental improvement of muscle function. The zebrafish mutant line, twister, models SCS in terms of a dominant mutation in the α subunit (α(twi)) but shows the behavioral improvement associated with Escobar syndrome. Here, we present a unique electrophysiological study into developmental improvement for a myasthenic syndrome. The embryonic α(twi)ßδγ receptor isoform produces slowly decaying synaptic currents typical of SCS that transit to a much faster decay upon the appearance of adult ε, despite the α(twi) mutation. Thus, the continued expression of α(twi) into adulthood is tolerated because of the ε expression and associated recovery, raising the likelihood of unappreciated myasthenic cases that benefit from the γ-ε switch.


Assuntos
Modelos Animais de Doenças , Síndromes Miastênicas Congênitas/etiologia , Animais , Sequência de Bases , Primers do DNA , Síndromes Miastênicas Congênitas/fisiopatologia , Técnicas de Patch-Clamp , Peixe-Zebra
17.
J Neurosci ; 33(17): 7384-92, 2013 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-23616544

RESUMO

A long-held tenet of neuromuscular transmission is that calcium-dependent neurotransmitter release is mediated by N-type calcium channels in frog but P/Q-type channels in mammals. The N-type assignment in frog is based principally on pharmacological sensitivity to ω-conotoxin GVIA. Our studies show that zebrafish neuromuscular transmission is also sensitive to ω-conotoxin GVIA. However, positional cloning of a mutant line with compromised neuromuscular function identified a mutation in a P/Q- rather than N-type channel. Cloning and heterologous expression of this P/Q-type channel confirmed a block by ω-conotoxin GVIA raising the likelihood that all vertebrates, including frog, use the P/Q-type calcium channel for neuromuscular transmission. In addition, our P/Q defective mutant line offered a means of testing the ability of roscovitine, known to potentiate frog neuromuscular transmission, to mediate behavioral and functional rescue. Acute treatment led to rapid improvement of both, pointing to potential therapeutic benefit for myasthenic disorders involving calcium channel dysfunction.


Assuntos
Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/fisiologia , Junção Neuromuscular/fisiologia , Transmissão Sináptica/fisiologia , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/genética , Canais de Cálcio/fisiologia , Canais de Cálcio Tipo N/genética , Canais de Cálcio Tipo N/fisiologia , Canais de Cálcio Tipo P/genética , Canais de Cálcio Tipo Q/genética , Clonagem Molecular , Células HEK293 , Humanos , Dados de Sequência Molecular , Mutação/fisiologia , Junção Neuromuscular/genética , Transmissão Sináptica/genética , Peixe-Zebra
18.
J Neurosci ; 33(48): 18764-74, 2013 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-24285883

RESUMO

Mutations in the methyl-CpG binding protein 2 gene, Mecp2, affect primarily the brain and lead to a wide range of neuropsychiatric disorders, most commonly Rett syndrome (RTT). Although the neuropathology of RTT is well understood, the cellular and molecular mechanism(s), which lead to the disease initiation and progression, has yet to be elucidated. RTT was initially attributed only to neuronal dysfunction, but our recent studies and those of others show that RTT is not exclusively neuronal but rather also involves interactions between neurons and glia. Importantly, studies have shown that MeCP2-restored astrocytes and microglia are able to attenuate the disease progression in otherwise MeCP2-null mice. Here we show that another type of glia, oligodendrocytes, and their progenitors are also involved in manifestation of specific RTT symptoms. Mice that lost MeCP2 specifically in the oligodendrocyte lineage cells, although overall normal, were more active and developed severe hindlimb clasping phenotypes. Inversely, restoration of MeCP2 in oligodendrocyte lineage cells, in otherwise MeCP2-null mice, although only mildly prolonging their lifespan, significantly improved the locomotor deficits and hindlimb clasping phenotype, both in male and female mice, and fully restored the body weight in male mice. Finally, we found that the level of some myelin-related proteins was impaired in the MeCP2-null mice. Expression of MeCP2 in oligodendrocytes of these mice only partially restored their expression, suggesting that there is a non-cell-autonomous effect by other cell types in the brains on the expression of myelin-related proteins in oligodendrocytes.


Assuntos
Linhagem da Célula/fisiologia , Proteína 2 de Ligação a Metil-CpG/genética , Oligodendroglia/patologia , Síndrome de Rett/patologia , Animais , Astrócitos/fisiologia , Western Blotting , Escuridão , Feminino , Força da Mão/fisiologia , Membro Posterior/fisiologia , Imuno-Histoquímica , Luz , Locomoção/fisiologia , Masculino , Proteína 2 de Ligação a Metil-CpG/fisiologia , Camundongos , Mutação/genética , Mutação/fisiologia , Proteína Básica da Mielina/fisiologia , Fenótipo , Reação em Cadeia da Polimerase
19.
J Neurosci ; 33(34): 13612-20, 2013 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-23966684

RESUMO

De novo mutations in the X-linked gene encoding the transcription factor methyl-CpG binding protein 2 (MECP2) are the most frequent cause of the neurological disorder Rett syndrome (RTT). Hemizygous males usually die of neonatal encephalopathy. Heterozygous females survive into adulthood but exhibit severe symptoms including microcephaly, loss of purposeful hand motions and speech, and motor abnormalities, which appear after a period of apparently normal development. Most studies have focused on male mouse models because of the shorter latency to and severity in symptoms, yet how well these mice mimic the disease in affected females is not clear. Very few therapeutic treatments have been proposed for females, the more gender-appropriate model. Here, we show that self-complementary AAV9, bearing MeCP2 cDNA under control of a fragment of its own promoter (scAAV9/MeCP2), is capable of significantly stabilizing or reversing symptoms when administered systemically into female RTT mice. To our knowledge, this is the first potential gene therapy for females afflicted with RTT.


Assuntos
Comportamento Animal/efeitos dos fármacos , Proteína 2 de Ligação a Metil-CpG/administração & dosagem , Síndrome de Rett/fisiopatologia , Síndrome de Rett/terapia , Animais , Comportamento Animal/fisiologia , Contagem de Células , Dependovirus/fisiologia , Modelos Animais de Doenças , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Proteína 2 de Ligação a Metil-CpG/biossíntese , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Mutação/genética , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/metabolismo , Neurônios/patologia , Fosfopiruvato Hidratase/metabolismo , Pletismografia , Equilíbrio Postural/genética , Equilíbrio Postural/fisiologia , Reconhecimento Psicológico/fisiologia , Respiração , Síndrome de Rett/genética , Síndrome de Rett/patologia , Teste de Desempenho do Rota-Rod
20.
Proc Natl Acad Sci U S A ; 108(40): 16789-94, 2011 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-21921234

RESUMO

Neurogenesis requires mechanisms that coordinate early cell-fate decisions, migration, and terminal differentiation. Here, we show that the transcriptional repressor, repressor element 1 silencing transcription factor (REST), regulates radial migration and the timing of neural progenitor differentiation during neocortical development, and that the regulation is contingent upon differential REST levels. Specifically, a sustained presence of REST blocks migration and greatly delays--but does not prevent--neuronal differentiation, resulting in a subcortical band heterotopia-like phenotype, reminiscent of loss of doublecortin. We further show that doublecortin is a direct gene target of REST, and that its overexpression rescues, at least in part, the aberrant phenotype caused by persistent presence of REST. Our studies support the view that the targeted down-regulation of REST to low levels in neural progenitors, and its subsequent disappearance during neurogenesis, is critical for timing the spatiotemporal transition of neural progenitor cells to neurons.


Assuntos
Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Neocórtex/embriologia , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/fisiologia , Proteínas Repressoras/metabolismo , Animais , Western Blotting , Linhagem Celular , Imunoprecipitação da Cromatina , Proteínas Correpressoras , Primers do DNA/genética , DNA Complementar/genética , Proteínas do Domínio Duplacortina , Eletroporação , Vetores Genéticos , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Microscopia Confocal , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/genética , Neuropeptídeos/metabolismo , Proteínas Repressoras/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa