Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 4775, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38839750

RESUMO

The metal ion transporter SLC39A8 is associated with physiological traits and diseases, including blood manganese (Mn) levels and inflammatory bowel diseases (IBD). The mechanisms by which SLC39A8 controls Mn homeostasis and epithelial integrity remain elusive. Here, we generate Slc39a8 intestinal epithelial cell-specific-knockout (Slc39a8-IEC KO) mice, which display markedly decreased Mn levels in blood and most organs. Radiotracer studies reveal impaired intestinal absorption of dietary Mn in Slc39a8-IEC KO mice. SLC39A8 is localized to the apical membrane and mediates 54Mn uptake in intestinal organoid monolayer cultures. Unbiased transcriptomic analysis identifies alkaline ceramidase 1 (ACER1), a key enzyme in sphingolipid metabolism, as a potential therapeutic target for SLC39A8-associated IBDs. Importantly, treatment with an ACER1 inhibitor attenuates colitis in Slc39a8-IEC KO mice by remedying barrier dysfunction. Our results highlight the essential roles of SLC39A8 in intestinal Mn absorption and epithelial integrity and offer a therapeutic target for IBD associated with impaired Mn homeostasis.


Assuntos
Ceramidase Alcalina , Proteínas de Transporte de Cátions , Doenças Inflamatórias Intestinais , Mucosa Intestinal , Manganês , Camundongos Knockout , Animais , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Transporte de Cátions/genética , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/patologia , Manganês/metabolismo , Camundongos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Ceramidase Alcalina/metabolismo , Ceramidase Alcalina/genética , Humanos , Camundongos Endogâmicos C57BL , Homeostase , Masculino , Colite/metabolismo , Colite/genética , Colite/patologia , Absorção Intestinal , Células Epiteliais/metabolismo
2.
Nat Commun ; 15(1): 1721, 2024 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-38409226

RESUMO

Quiescence in stem cells is traditionally considered as a state of inactive dormancy or with poised potential. Naive mouse embryonic stem cells (ESCs) can enter quiescence spontaneously or upon inhibition of MYC or fatty acid oxidation, mimicking embryonic diapause in vivo. The molecular underpinning and developmental potential of quiescent ESCs (qESCs) are relatively unexplored. Here we show that qESCs possess an expanded or unrestricted cell fate, capable of generating both embryonic and extraembryonic cell types (e.g., trophoblast stem cells). These cells have a divergent metabolic landscape comparing to the cycling ESCs, with a notable decrease of the one-carbon metabolite S-adenosylmethionine. The metabolic changes are accompanied by a global reduction of H3K27me3, an increase of chromatin accessibility, as well as the de-repression of endogenous retrovirus MERVL and trophoblast master regulators. Depletion of methionine adenosyltransferase Mat2a or deletion of Eed in the polycomb repressive complex 2 results in removal of the developmental constraints towards the extraembryonic lineages. Our findings suggest that quiescent ESCs are not dormant but rather undergo an active transition towards an unrestricted cell fate.


Assuntos
Cromatina , Células-Tronco Embrionárias , Animais , Camundongos , Células-Tronco Embrionárias/metabolismo , Diferenciação Celular , Cromatina/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Complexo Repressor Polycomb 2/metabolismo , S-Adenosilmetionina/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(26): e2121400119, 2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35737834

RESUMO

Deficiencies of the transmembrane iron-transporting protein ferroportin (FPN1) cause the iron misdistribution that underlies ferroportin disease, anemia of inflammation, and several other human diseases and conditions. A small molecule natural product, hinokitiol, was recently shown to serve as a surrogate transmembrane iron transporter that can restore hemoglobinization in zebrafish deficient in other iron transporting proteins and can increase gut iron absorption in FPN1-deficient flatiron mice. However, whether hinokitiol can restore normal iron physiology in FPN1-deficient animals or primary cells from patients and the mechanisms underlying such targeted activities remain unknown. Here, we show that hinokitiol redistributes iron from the liver to red blood cells in flatiron mice, thereby increasing hemoglobin and hematocrit. Mechanistic studies confirm that hinokitiol functions as a surrogate transmembrane iron transporter to release iron trapped within liver macrophages, that hinokitiol-Fe complexes transfer iron to transferrin, and that the resulting transferrin-Fe complexes drive red blood cell maturation in a transferrin-receptor-dependent manner. We also show in FPN1-deficient primary macrophages derived from patients with ferroportin disease that hinokitiol moves labile iron from inside to outside cells and decreases intracellular ferritin levels. The mobilization of nonlabile iron is accompanied by reductions in intracellular ferritin, consistent with the activation of regulated ferritin proteolysis. These findings collectively provide foundational support for the translation of small molecule iron transporters into therapies for human diseases caused by iron misdistribution.


Assuntos
Ferro , Macrófagos , Monoterpenos , Tropolona/análogos & derivados , Animais , Proteínas de Transporte de Cátions/deficiência , Ferritinas/metabolismo , Humanos , Ferro/metabolismo , Macrófagos/metabolismo , Camundongos , Monoterpenos/metabolismo , Transferrina/metabolismo , Tropolona/metabolismo , Peixe-Zebra/metabolismo
4.
J Neurochem ; 160(3): 356-375, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34837396

RESUMO

Neurodegeneration with brain iron accumulation (NBIA) is a clinically and genetically heterogeneous group of neurodegenerative diseases characterized by the abnormal accumulation of brain iron and the progressive degeneration of the nervous system. One of the recently identified subtypes of NBIA is ß-propeller protein-associated neurodegeneration (BPAN). BPAN is caused by de novo mutations in the WDR45/WIPI4 (WD repeat domain 45) gene. WDR45 is one of the four mammalian homologs of yeast Atg18, a regulator of autophagy. WDR45 deficiency in BPAN patients and animal models may result in defects in autophagic flux. However, how WDR45 deficiency leads to brain iron overload remains unclear. To elucidate the role of WDR45, we generated a WDR45-knockout (KO) SH-SY5Y neuroblastoma cell line using CRISPR-Cas9-mediated genome editing. Using these cells, we demonstrated that the non-TF (transferrin)-bound iron pathway dominantly mediated the accumulation of iron. Moreover, the loss of WDR45 led to defects in ferritinophagy, a form of autophagy that degrades the iron storage protein ferritin. We showed that impaired ferritinophagy contributes to iron accumulation in WDR45-KO cells. Iron accumulation was also detected in the mitochondria, which was accompanied by impaired mitochondrial respiration, elevated reactive oxygen species, and increased cell death. Thus, our study links WDR45 to specific iron acquisition pathways and ferritinophagy. Cover Image for this issue: https://doi.org/10.1111/jnc.15388.


Assuntos
Autofagia/genética , Proteínas de Transporte/genética , Sobrecarga de Ferro/genética , Doenças Neurodegenerativas/genética , Química Encefálica/genética , Morte Celular , Linhagem Celular , Técnicas de Inativação de Genes , Humanos , Ferro/metabolismo , Sobrecarga de Ferro/metabolismo , Mitocôndrias/metabolismo , Doenças Neurodegenerativas/metabolismo , Espécies Reativas de Oxigênio , Transferrina/metabolismo
5.
Commun Biol ; 3(1): 278, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32483278

RESUMO

Histone H3 lysine 4 methylation (H3K4me) is extensively regulated by numerous writer and eraser enzymes in mammals. Nine H3K4me enzymes are associated with neurodevelopmental disorders to date, indicating their important roles in the brain. However, interplay among H3K4me enzymes during brain development remains largely unknown. Here, we show functional interactions of a writer-eraser duo, KMT2A and KDM5C, which are responsible for Wiedemann-Steiner Syndrome (WDSTS), and mental retardation X-linked syndromic Claes-Jensen type (MRXSCJ), respectively. Despite opposite enzymatic activities, the two mouse models deficient for either Kmt2a or Kdm5c shared reduced dendritic spines and increased aggression. Double mutation of Kmt2a and Kdm5c clearly reversed dendritic morphology, key behavioral traits including aggression, and partially corrected altered transcriptomes and H3K4me landscapes. Thus, our study uncovers common yet mutually suppressive aspects of the WDSTS and MRXSCJ models and provides a proof of principle for balancing a single writer-eraser pair to ameliorate their associated disorders.


Assuntos
Anormalidades Múltiplas/genética , Agressão , Anormalidades Craniofaciais/genética , Espinhas Dendríticas/metabolismo , Transtornos do Crescimento/genética , Histona Desmetilases/genética , Histona-Lisina N-Metiltransferase/genética , Histonas/metabolismo , Hipertricose/genética , Deficiência Intelectual/genética , Deficiência Intelectual Ligada ao Cromossomo X/genética , Proteína de Leucina Linfoide-Mieloide/genética , Animais , Modelos Animais de Doenças , Histona Desmetilases/deficiência , Histona-Lisina N-Metiltransferase/deficiência , Masculino , Metilação , Camundongos , Proteína de Leucina Linfoide-Mieloide/deficiência
6.
PLoS One ; 14(8): e0219436, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31390367

RESUMO

RAS is the founding member of a superfamily of GTPases and regulates signaling pathways involved in cellular growth control. While recent studies have shown that the activation state of RAS can be controlled by lysine ubiquitylation and acetylation, the existence of lysine methylation of the RAS superfamily GTPases remains unexplored. In contrast to acetylation, methylation does not alter the side chain charge and it has been challenging to deduce its impact on protein structure by conventional amino acid substitutions. Herein, we investigate lysine methylation on RAS and RAS-related GTPases. We developed GoMADScan (Go language-based Modification Associated Database Scanner), a new user-friendly application that scans and extracts posttranslationally modified peptides from databases. The GoMADScan search on PhosphoSitePlus databases identified methylation of conserved lysine residues in the core GTPase domain of RAS superfamily GTPases, including residues corresponding to RAS Lys-5, Lys-16, and Lys-117. To follow up on these observations, we immunoprecipitated endogenous RAS from HEK293T cells, conducted mass spectrometric analysis and found that RAS residues, Lys-5 and Lys-147, undergo dimethylation and monomethylation, respectively. Since mutations of Lys-5 have been found in cancers and RASopathies, we set up molecular dynamics (MD) simulations to assess the putative impact of Lys-5 dimethylation on RAS structure. Results from our MD analyses predict that dimethylation of Lys-5 does not significantly alter RAS conformation, suggesting that Lys-5 methylation may alter existing protein interactions or create a docking site to foster new interactions. Taken together, our findings uncover the existence of lysine methylation as a novel posttranslational modification associated with RAS and the RAS superfamily GTPases, and putative impact of Lys-5 dimethylation on RAS structure.


Assuntos
Mineração de Dados/métodos , GTP Fosfo-Hidrolases/química , GTP Fosfo-Hidrolases/metabolismo , Lisina/metabolismo , Processamento de Proteína Pós-Traducional , Sequência de Aminoácidos , Metilação , Simulação de Dinâmica Molecular , Domínios Proteicos
7.
JCI Insight ; 3(4)2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29467333

RESUMO

CHD7, an ATP-dependent chromatin remodeler, is disrupted in CHARGE syndrome, an autosomal dominant disorder characterized by variably penetrant abnormalities in craniofacial, cardiac, and nervous system tissues. The inner ear is uniquely sensitive to CHD7 levels and is the most commonly affected organ in individuals with CHARGE. Interestingly, upregulation or downregulation of retinoic acid (RA) signaling during embryogenesis also leads to developmental defects similar to those in CHARGE syndrome, suggesting that CHD7 and RA may have common target genes or signaling pathways. Here, we tested three separate potential mechanisms for CHD7 and RA interaction: (a) direct binding of CHD7 with RA receptors, (b) regulation of CHD7 levels by RA, and (c) CHD7 binding and regulation of RA-related genes. We show that CHD7 directly regulates expression of Aldh1a3, the gene encoding the RA synthetic enzyme ALDH1A3 and that loss of Aldh1a3 partially rescues Chd7 mutant mouse inner ear defects. Together, these studies indicate that ALDH1A3 acts with CHD7 in a common genetic pathway to regulate inner ear development, providing insights into how CHD7 and RA regulate gene expression and morphogenesis in the developing embryo.


Assuntos
Aldeído Oxirredutases/metabolismo , Síndrome CHARGE/genética , DNA Helicases/deficiência , Proteínas de Ligação a DNA/deficiência , Regulação da Expressão Gênica no Desenvolvimento , Retinal Desidrogenase/metabolismo , Tretinoína/metabolismo , Aldeído Oxirredutases/genética , Animais , Síndrome CHARGE/patologia , Linhagem Celular Tumoral , DNA Helicases/genética , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Orelha Interna/embriologia , Embrião de Mamíferos , Feminino , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Organogênese/genética , RNA Interferente Pequeno/metabolismo , Retinal Desidrogenase/genética
8.
Mol Cell Neurosci ; 87: 1-3, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29248671

RESUMO

Epigenetic regulation of gene expression is critical during development of the central nervous system. Pathogenic variants in genes encoding epigenetic factors have been found to cause a wide variety of neurodevelopmental disorders including Autism spectrum disorder, intellectual disability, and epilepsy. Cancers affecting neuronal and glial cells in the brain have also been shown to exhibit somatic mutations in epigenetic regulators, suggesting chromatin-based links between regulated and dysregulated cellular proliferation and differentiation. In this special issue, six articles review recent discoveries implicating epigenetic modifiers in normal and disease states affecting the nervous system, and the underlying mechanisms by which these modifiers function. Two articles present new information about roles for chromatin regulators in nervous system development and cancer. Together, these manuscripts provide a concise overview of this rapidly growing field. In this introduction, we briefly summarize themes presented in the issue, and pose questions for ongoing research and discovery.


Assuntos
Encéfalo/crescimento & desenvolvimento , Montagem e Desmontagem da Cromatina/genética , Cromatina/genética , Epigênese Genética/genética , Animais , Transtorno do Espectro Autista/genética , Encéfalo/metabolismo , Epilepsia/genética , Humanos , Neurônios/metabolismo
9.
Neuroscience ; 370: 170-180, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28571721

RESUMO

Potocki-Shaffer Syndrome is a rare neurodevelopmental syndrome associated with microdeletion of a region of Chromosome 11p11.2. Genetic evidence has implicated haploinsufficiency of PHF21A, a gene that encodes a histone-binding protein, as the likely cause of intellectual disability and craniofacial abnormalities in Potocki-Shaffer Syndrome. However, the molecular consequences of reduced PHF21A expression remain elusive. In this study, we analyzed by RNA-Sequencing (RNA-Seq) two patient-derived cell lines with heterozygous loss of PHF21A compared to unaffected individuals and identified 1,885 genes that were commonly misregulated. The patient cells displayed down-regulation of key pathways relevant to learning and memory, including Cyclic Adenosine Monophosphate (cAMP)-signaling pathway genes. We found that PHF21A is required for full induction of a luciferase reporter carrying cAMP-responsive elements (CRE) following stimulation by the cAMP analog, forskolin. Finally, PHF21A-deficient patient-derived cells exhibited a delayed induction of immediate early genes following forskolin stimulation. These results suggest that an impaired response to cAMP signaling might be involved in the pathology of PHF21A deficiency. This article is part of a Special Issue entitled: [SI: Molecules & Cognition].


Assuntos
AMP Cíclico/metabolismo , Histona Desacetilases/deficiência , Linhagem Celular , Deleção Cromossômica , Transtornos Cromossômicos/metabolismo , Cromossomos Humanos Par 11/metabolismo , Colforsina/farmacologia , AMP Cíclico/análogos & derivados , Exostose Múltipla Hereditária/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Histona Desacetilases/genética , Humanos , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , RNA Interferente Pequeno , Transcrição Gênica
10.
Sci Rep ; 7(1): 8801, 2017 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-28821730

RESUMO

Chronic inflammation can be a major driver of the failure of a variety of organs, including chronic kidney disease (CKD). The NLR family pyrin domain-containing 3 (NLRP3) inflammasome has been shown to play a pivotal role in inflammation in a mouse kidney disease model. Nuclear factor erythroid 2-related factor 2 (Nrf2), the master transcription factor for anti-oxidant responses, has also been implicated in inflammasome activation under physiological conditions. However, the mechanism underlying inflammasome activation in CKD remains elusive. Here, we show that the loss of Nrf2 suppresses fibrosis and inflammation in a unilateral ureter obstruction (UUO) model of CKD in mice. We consistently observed decreased expression of inflammation-related genes NLRP3 and IL-1ß in Nrf2-deficient kidneys after UUO. Increased infiltration of M1, but not M2, macrophages appears to mediate the suppression of UUO-induced CKD symptoms. Furthermore, we found that activation of the NLRP3 inflammasome is attenuated in Nrf2-deficient bone marrow-derived macrophages. These results demonstrate that Nrf2-related inflammasome activation can promote CKD symptoms via infiltration of M1 macrophages. Thus, we have identified the Nrf2 pathway as a promising therapeutic target for CKD.


Assuntos
Macrófagos/imunologia , Macrófagos/metabolismo , Fator 2 Relacionado a NF-E2/deficiência , Obstrução Ureteral/etiologia , Obstrução Ureteral/metabolismo , Animais , Modelos Animais de Doenças , Fibrose , Perfilação da Expressão Gênica , Inflamassomos/genética , Inflamassomos/metabolismo , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Rim/metabolismo , Rim/patologia , Ativação de Macrófagos/genética , Ativação de Macrófagos/imunologia , Camundongos , Camundongos Knockout , Obstrução Ureteral/patologia , Obstrução Ureteral/terapia
11.
Epigenomics ; 8(12): 1689-1708, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27855486

RESUMO

Dysregulation of histone methylation has emerged as a major driver of neurodevelopmental disorders including intellectual disabilities and autism spectrum disorders. Histone methyl writer and eraser enzymes generally act within multisubunit complexes rather than in isolation. However, it remains largely elusive how such complexes cooperate to achieve the precise spatiotemporal gene expression in the developing brain. Histone H3K4 methylation (H3K4me) is a chromatin signature associated with active gene-regulatory elements. We review a body of literature that supports a model in which the RAI1-containing H3K4me writer complex counterbalances the LSD1-containing H3K4me eraser complex to ensure normal brain development. This model predicts H3K4me as the nexus of previously unrelated neurodevelopmental disorders.


Assuntos
Encéfalo/metabolismo , Histonas/metabolismo , Anormalidades Múltiplas/genética , Animais , Transtornos Cromossômicos/genética , Duplicação Cromossômica/genética , Ritmo Circadiano/genética , Proteínas Correpressoras/genética , Expressão Gênica , Proteínas de Grupo de Alta Mobilidade/genética , Histona Desmetilases/genética , Histona-Lisina N-Metiltransferase/genética , Humanos , Metilação , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas do Tecido Nervoso/genética , Síndrome de Smith-Magenis/genética , Transativadores , Fatores de Transcrição/genética
12.
J Mol Neurosci ; 60(1): 33-45, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27421841

RESUMO

The X-linked lysine (K)-specific demethylase 5C (KDM5C) gene plays an important role in brain development and behavior. It encodes a histone demethylase that is involved in gene regulation in neuronal differentiation and morphogenesis. When mutated, it causes neuropsychiatric symptoms, such as intellectual disability, delayed language development, epilepsy, and impulsivity. To better understand how the patient mutations affect neuronal development, we expressed KDM5C mutants in Neuro2a cells, a mouse neuroblastoma cell line. Retinoic acid (RA)-induced neurite growth was suppressed by the mutation KDM5C (Y751C) , KDM5C (H514A) , and KDM5C (F642L) , but not KDM5C (D87G) or KDM5C (A388P) . RNA-seq analysis indicated an upregulation of genes important for neuronal development, such as Ntng2, Enah, Gas1, Slit2, and Dscam, in response to the RA treatment in control Neuro2a cells transfected with GFP or wild-type KDM5C. In contrast, in cells transfected with KDM5C (Y751C) , these genes were not upregulated by RA. Ntng2 was downregulated in cells with KDM5C mutations, concordant with the lower levels of H3K4 methylation at its promoter. Moreover, knocking down Ntng2 in control Neuro2a cells led to the phenotype of short neurites similar to that of cells with KDM5C (Y751C) , whereas Ntng2 overexpression in the mutant cells rescued the morphological phenotype. These findings provide new insight into the pathogenesis of phenotypes associated with KDM5C mutations.


Assuntos
Deficiência Intelectual/genética , Mutação , Proteínas do Tecido Nervoso/metabolismo , Crescimento Neuronal , Oxirredutases N-Desmetilantes/genética , Animais , Linhagem Celular Tumoral , Células Cultivadas , Regulação para Baixo , Feminino , Histona Desmetilases , Histonas/metabolismo , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Netrinas , Oxirredutases N-Desmetilantes/metabolismo , Processamento de Proteína Pós-Traducional
13.
Nat Struct Mol Biol ; 18(7): 769-76, 2011 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-21666679

RESUMO

ATR-X (alpha-thalassemia/mental retardation, X-linked) syndrome is a human congenital disorder that causes severe intellectual disabilities. Mutations in the ATRX gene, which encodes an ATP-dependent chromatin-remodeler, are responsible for the syndrome. Approximately 50% of the missense mutations in affected persons are clustered in a cysteine-rich domain termed ADD (ATRX-DNMT3-DNMT3L, ADD(ATRX)), whose function has remained elusive. Here we identify ADD(ATRX) as a previously unknown histone H3-binding module, whose binding is promoted by lysine 9 trimethylation (H3K9me3) but inhibited by lysine 4 trimethylation (H3K4me3). The cocrystal structure of ADD(ATRX) bound to H3(1-15)K9me3 peptide reveals an atypical composite H3K9me3-binding pocket, which is distinct from the conventional trimethyllysine-binding aromatic cage. Notably, H3K9me3-pocket mutants and ATR-X syndrome mutants are defective in both H3K9me3 binding and localization at pericentromeric heterochromatin; thus, we have discovered a unique histone-recognition mechanism underlying the ATR-X etiology.


Assuntos
DNA Helicases/química , Histonas/metabolismo , Deficiência Intelectual/genética , Proteínas Nucleares/química , Sequência de Aminoácidos , Sítios de Ligação , DNA Helicases/genética , DNA Helicases/metabolismo , Heterocromatina/metabolismo , Humanos , Metilação , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Alinhamento de Sequência , Proteína Nuclear Ligada ao X
14.
Nature ; 449(7163): 689-94, 2007 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-17851529

RESUMO

The recent discovery of a large number of histone demethylases suggests a central role for these enzymes in regulating histone methylation dynamics. Histone H3K27 trimethylation (H3K27me3) has been linked to polycomb-group-protein-mediated suppression of Hox genes and animal body patterning, X-chromosome inactivation and possibly maintenance of embryonic stem cell (ESC) identity. An imbalance of H3K27 methylation owing to overexpression of the methylase EZH2 has been implicated in metastatic prostate and aggressive breast cancers. Here we show that the JmjC-domain-containing related proteins UTX and JMJD3 catalyse demethylation of H3K27me3/2. UTX is enriched around the transcription start sites of many HOX genes in primary human fibroblasts, in which HOX genes are differentially expressed, but is selectively excluded from the HOX loci in ESCs, in which HOX genes are largely silent. Consistently, RNA interference inhibition of UTX led to increased H3K27me3 levels at some HOX gene promoters. Importantly, morpholino oligonucleotide inhibition of a zebrafish UTX homologue resulted in mis-regulation of hox genes and a striking posterior developmental defect, which was partially rescued by wild-type, but not by catalytically inactive, human UTX. Taken together, these findings identify a small family of H3K27 demethylases with important, evolutionarily conserved roles in H3K27 methylation regulation and in animal anterior-posterior development.


Assuntos
Padronização Corporal , Histonas/metabolismo , Lisina/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Linhagem Celular , Embrião não Mamífero/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Genes Homeobox/genética , Genoma/genética , Histona Desmetilases , Humanos , Histona Desmetilases com o Domínio Jumonji , Metilação , Camundongos , Proteínas Nucleares/genética , Oxirredutases N-Desmetilantes/genética , Oxirredutases N-Desmetilantes/metabolismo , Transcrição Gênica/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
15.
Cell ; 128(6): 1077-88, 2007 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-17320160

RESUMO

Histone methylation regulates chromatin structure and transcription. The recently identified histone demethylase lysine-specific demethylase 1 (LSD1) is chemically restricted to demethylation of only mono- and di- but not trimethylated histone H3 lysine 4 (H3K4me3). We show that the X-linked mental retardation (XLMR) gene SMCX (JARID1C), which encodes a JmjC-domain protein, reversed H3K4me3 to di- and mono- but not unmethylated products. Other SMCX family members, including SMCY, RBP2, and PLU-1, also demethylated H3K4me3. SMCX bound H3K9me3 via its N-terminal PHD (plant homeodomain) finger, which may help coordinate H3K4 demethylation and H3K9 methylation in transcriptional repression. Significantly, several XLMR-patient point mutations reduced SMCX demethylase activity and binding to H3K9me3 peptides, respectively. Importantly, studies in zebrafish and primary mammalian neurons demonstrated a role for SMCX in neuronal survival and dendritic development and a link to the demethylase activity. Our findings thus identify a family of H3K4me3 demethylases and uncover a critical link between histone modifications and XLMR.


Assuntos
Histonas/metabolismo , Deficiência Intelectual Ligada ao Cromossomo X/genética , Oxirredutases N-Desmetilantes/genética , Proteínas/genética , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , DNA Complementar , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Biblioteca Gênica , Histona Desmetilases , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/química , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Histona Desmetilases com o Domínio Jumonji , Lisina/metabolismo , Metilação , Camundongos , Antígenos de Histocompatibilidade Menor , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Oxirredutases N-Desmetilantes/metabolismo , Proteínas/metabolismo , Proteína 2 de Ligação ao Retinoblastoma , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
16.
FEBS Lett ; 580(13): 3129-35, 2006 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-16684532

RESUMO

BHC80 is a component of BRAF-HDAC complex (BHC) involved in transcriptional repression of neuron-specific genes in non-neuronal cells. However, BHC80 is present in both neuronal and non-neuronal cells. To explore the physiological importance of BHC80 in vivo, and the precise mechanism underlying neuron-specific gene repression by BHC80, we have produced mutant mice lacking Bhc80. The loss of Bhc80 resulted in neonatal lethality without sucking mother's breast milk sufficiently. Although Bhc80-deficient mice showed no developmental defect in the neuronal and non-neuronal tissues, Bhc80 is indispensable for the survival of neonatal pups.


Assuntos
Genes Letais , Histona Desacetilases/fisiologia , Proteínas Proto-Oncogênicas c-raf/fisiologia , Animais , Animais Recém-Nascidos , Apoptose , Astrócitos/citologia , Astrócitos/enzimologia , Movimento Celular , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Pulmão/química , Pulmão/citologia , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Mutantes , Neurônios/citologia , Neurônios/enzimologia , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Proto-Oncogênicas c-raf/genética , Sinapsinas/análise , Sinapsinas/metabolismo
17.
Mol Cell ; 19(6): 857-64, 2005 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-16140033

RESUMO

LSD1 is a recently identified human lysine (K)-specific histone demethylase. LSD1 is associated with HDAC1/2; CoREST, a SANT domain-containing corepressor; and BHC80, a PHD domain-containing protein, among others. We show that CoREST endows LSD1 with the ability to demethylate nucleosomal substrates and that it protects LSD1 from proteasomal degradation in vivo. We find hyperacetylated nucleosomes less susceptible to CoREST/LSD1-mediated demethylation, suggesting that hypoacetylated nucleosomes may be the preferred physiological substrates. This raises the possibility that histone deacetylases and LSD1 may collaborate to generate a repressive chromatin environment. Consistent with this model, TSA treatment results in derepression of LSD1 target genes. While CoREST positively regulates LSD1 function, BHC80 inhibits CoREST/LSD1-mediated demethylation in vitro and may therefore confer negative regulation. Taken together, these findings suggest that LSD1-mediated histone demethylation is regulated dynamically in vivo. This is expected to have profound effects on gene expression under both physiological and pathological conditions.


Assuntos
Regulação da Expressão Gênica , Histonas/metabolismo , Oxirredutases N-Desmetilantes/metabolismo , Estabilidade Enzimática , Células HeLa , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Histona Desmetilases , Humanos , Complexos Multiproteicos , Nucleossomos/metabolismo , Oxirredutases N-Desmetilantes/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , Ligação Proteica , Proteínas Proto-Oncogênicas c-raf/genética , Proteínas Proto-Oncogênicas c-raf/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
18.
Biochem Biophys Res Commun ; 322(2): 601-8, 2004 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-15325272

RESUMO

BRAF-HDAC complex (BHC) has been shown to contain six components, including BHC80, and to mediate REST-dependent transcriptional repression of neuron-specific genes in non-neuronal cells. In this study, we have examined the functional role(s) of BHC80 in mouse tissues and human cultured cells. Two isoforms of mouse BHC80 were predominantly present in the central nervous system and spermatogenic cells. Human cultured cells also contained two isoforms of BHC80. Immunohistochemical analysis showed the presence of mouse BHC80 in the nucleus of neuronal cells in the hippocampus and cerebellum. The C-terminal region of human BHC80 containing PHD zinc-finger domain was capable of binding directly to each of five other components of BHC, and of organizing BHC mediating transcriptional repression. Moreover, two isoforms of human BHC80 were distinguished from each other by reduced binding to HDAC1 and HDAC2, despite the presence of the PHD finger domain. These results suggest that BHC80 presumably serves as a scaffold protein in BHC in neuronal as well as non-neuronal cells. A possible role of BHC80 in spermatogenesis is also suggested.


Assuntos
Regulação da Expressão Gênica/fisiologia , Histona Desacetilases/metabolismo , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Processamento Alternativo/fisiologia , Animais , Encéfalo/metabolismo , Histona Desacetilases/genética , Humanos , Masculino , Camundongos , Isoformas de Proteínas , Proteínas Proto-Oncogênicas c-raf/genética , Espermatogênese/fisiologia , Testículo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA