Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Genes Dev ; 30(5): 485-6, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26944674

RESUMO

In this issue of Genes & Development, Ginart and colleagues (pp. 567-578) study a mouse model for Russell-Silver syndrome (RSS) and show that similar cells within one individual can display distinct gene expression patterns because of epigenetic marks that are established stochastically during early development. Their results provide an excellent explanation for phenotypes seen in RSS and other imprinting disorders and especially help us understand how patients with similar or even identical genetic mutations can display distinct disease profiles.


Assuntos
Progressão da Doença , Regulação da Expressão Gênica no Desenvolvimento , Impressão Genômica/genética , Alelos , Animais , Metilação de DNA , Dosagem de Genes , Humanos , Camundongos , Modelos Animais , Mutação
2.
EMBO J ; 38(7)2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30842097

RESUMO

BRD4 is a BET family protein that binds acetylated histones and regulates transcription. BET/BRD4 inhibitors block blood cancer growth and inflammation and serve as a new therapeutic strategy. However, the biological role of BRD4 in normal hematopoiesis and inflammation is not fully understood. Analysis of Brd4 conditional knockout (KO) mice showed that BRD4 is required for hematopoietic stem cell expansion and progenitor development. Nevertheless, BRD4 played limited roles in macrophage development and inflammatory response to LPS ChIP-seq analysis showed that despite its limited importance, BRD4 broadly occupied the macrophage genome and participated in super-enhancer (SE) formation. Although BRD4 is critical for SE formation in cancer, BRD4 was not required for macrophage SEs, as KO macrophages created alternate, BRD4-less SEs that compensated BRD4 loss. This and additional mechanisms led to the retention of inflammatory responses in macrophages. Our results illustrate a context-dependent role of BRD4 and plasticity of epigenetic regulation.


Assuntos
Biomarcadores/análise , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Inflamação/imunologia , Macrófagos Peritoneais/imunologia , Proteínas Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Animais , Células Cultivadas , Perfilação da Expressão Gênica , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
3.
Blood ; 135(25): 2252-2265, 2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32181817

RESUMO

Prolonged or enhanced expression of the proto-oncogene Lmo2 is associated with a severe form of T-cell acute lymphoblastic leukemia (T-ALL), designated early T-cell precursor ALL, which is characterized by the aberrant self-renewal and subsequent oncogenic transformation of immature thymocytes. It has been suggested that Lmo2 exerts these effects by functioning as component of a multi-subunit transcription complex that includes the ubiquitous adapter Ldb1 along with b-HLH and/or GATA family transcription factors; however, direct experimental evidence for this mechanism is lacking. In this study, we investigated the importance of Ldb1 for Lmo2-induced T-ALL by conditional deletion of Ldb1 in thymocytes in an Lmo2 transgenic mouse model of T-ALL. Our results identify a critical requirement for Ldb1 in Lmo2-induced thymocyte self-renewal and thymocyte radiation resistance and for the transition of preleukemic thymocytes to overt T-ALL. Moreover, Ldb1 was also required for acquisition of the aberrant preleukemic ETP gene expression signature in immature Lmo2 transgenic thymocytes. Co-binding of Ldb1 and Lmo2 was detected at the promoters of key upregulated T-ALL driver genes (Hhex, Lyl1, and Nfe2) in preleukemic Lmo2 transgenic thymocytes, and binding of both Ldb1 and Lmo2 at these sites was reduced following Cre-mediated deletion of Ldb1. Together, these results identify a key role for Ldb1, a nonproto-oncogene, in T-ALL and support a model in which Lmo2-induced T-ALL results from failure to downregulate Ldb1/Lmo2-nucleated transcription complexes which normally function to enforce self-renewal in bone marrow hematopoietic progenitors.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Autorrenovação Celular , Proteínas de Ligação a DNA/fisiologia , Proteínas com Domínio LIM/fisiologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Timócitos/citologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Transferência Adotiva , Animais , Antígenos CD/biossíntese , Transformação Celular Neoplásica , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Deleção de Genes , Técnicas de Introdução de Genes , Proteínas com Domínio LIM/deficiência , Proteínas com Domínio LIM/genética , Linfopoese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proto-Oncogene Mas , RNA-Seq , Quimera por Radiação , Tolerância a Radiação , Timócitos/metabolismo , Timócitos/efeitos da radiação , Timócitos/transplante
4.
Genes Dev ; 28(5): 491-501, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24532688

RESUMO

Regulated expression of the H19 long noncoding RNA gene has been well characterized as a paradigm for genomic imprinting, but the H19 RNA's biological function remains largely unclear. H19 is abundantly expressed maternally in embryonic tissues but is strongly repressed after birth, and significant transcription persists only in skeletal muscle. Thus, we examined the role of the H19 RNA in skeletal muscle differentiation and regeneration. Knockdown of H19 RNA in myoblast cells and H19 knockout mouse satellite cells decreases differentiation. H19 exon1 encodes two conserved microRNAs, miR-675-3p and miR-675-5p, both of which are induced during skeletal muscle differentiation. The inhibition of myogenesis by H19 depletion during myoblast differentiation is rescued by exogenous expression of miR-675-3p and miR-675-5p. H19-deficient mice display abnormal skeletal muscle regeneration after injury, which is rectified by reintroduction of miR-675-3p and miR-675-5p. miR-675-3p and miR-675-5p function by directly targeting and down-regulating the anti-differentiation Smad transcription factors critical for the bone morphogenetic protein (BMP) pathway and the DNA replication initiation factor Cdc6. Therefore, the H19 long noncoding RNA has a critical trans-regulatory function in skeletal muscle differentiation and regeneration that is mediated by the microRNAs encoded within H19.


Assuntos
Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/biossíntese , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , RNA Longo não Codificante , Regeneração , Animais , Linhagem Celular , Linhagem Celular Tumoral , Masculino , Camundongos , Camundongos Knockout , MicroRNAs/metabolismo , Músculo Esquelético/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
5.
Hum Mol Genet ; 27(9): 1533-1544, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29452352

RESUMO

Cardiac calsequestrin (Casq2) associates with the ryanodine receptor 2 channel in the junctional sarcoplasmic reticulum to regulate Ca2+ release into the cytoplasm. Patients carrying mutations in CASQ2 display low resting heart rates under basal conditions and stress-induced polymorphic ventricular tachycardia (CPVT). In this study, we generate and characterize novel conditional deletion and conditional rescue mouse models to test the influence of developmental programs on the heart rate and CPVT phenotypes. We also compare the requirements for Casq2 function in the cardiac conduction system (CCS) and in working cardiomyocytes. Our study shows that the CPVT phenotype is dependent upon concurrent loss of Casq2 function in both the CCS and in working cardiomyocytes. Accordingly, restoration of Casq2 in only the CCS prevents CPVT. In addition, occurrence of CPVT is independent of the developmental history of Casq2-deficiency. In contrast, resting heart rate depends upon Casq2 gene activity only in the CCS and upon developmental history. Finally, our data support a model where low basal heart rate is a significant risk factor for CPVT.


Assuntos
Calsequestrina/metabolismo , Taquicardia Ventricular/metabolismo , Tamoxifeno/farmacologia , Animais , Cálcio/metabolismo , Calsequestrina/genética , Feminino , Frequência Cardíaca/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Mutantes , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Taquicardia Ventricular/genética
6.
Gastroenterology ; 155(1): 144-155, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29621481

RESUMO

BACKGROUND & AIMS: Inflammation affects regeneration of the intestinal epithelia; long noncoding RNAs (lncRNAs) regulate cell functions, such as proliferation, differentiation, and migration. We investigated the mechanisms by which the lncRNA H19, imprinted maternally expressed transcript (H19) regulates regeneration of intestinal epithelium using cell cultures and mouse models of inflammation. METHODS: We performed RNA-sequencing transcriptome analyses of intestinal tissues from mice with lipopolysaccharide (LPS)-induced sepsis to identify lncRNAs associated with inflammation; findings were confirmed by quantitative real-time polymerase chain reaction and in situ hybridization analyses of intestinal tissues from mice with sepsis or dextran sulfate sodium (DSS)-induced mucosal wound healing and patients with ulcerative colitis compared to healthy individuals (controls). We screened cytokines for their ability to induce expression of H19 in HT-29 cells and intestinal epithelial cells (IECs), and confirmed findings in crypt epithelial organoids derived from mouse small intestine. IECs were incubated with different signal transduction inhibitors and effects on H19 lncRNA levels were measured. We assessed intestinal epithelial proliferation or regeneration in H19ΔEx1/+ mice given LPS or DSS vs wild-type littermates (control mice). H19 was overexpressed in IECs using lentiviral vectors and cell proliferation was measured. We performed RNA antisense purification, RNA immunoprecipitation, and luciferase reporter assays to study functions of H19 in IECs. RESULTS: In RNA-sequencing transcriptome analysis of lncRNA expression in intestinal tissues from mice, we found that levels of H19 lncRNA changed significantly with LPS exposure. Levels of H19 lncRNA increased in intestinal tissues of patients with ulcerative colitis, mice with LPS-induced and polymicrobial sepsis, or mice with DSS-induced colitis, compared with controls. Increased H19 lncRNA localized to epithelial cells in the intestine, regardless of Lgr5 messenger RNA expression. Exposure of IECs to interleukin 22 (IL22) increased levels of H19 lncRNA with time and dose, which required STAT3 and protein kinase A activity. IL22 induced expression of H19 in mouse intestinal epithelial organoids within 6 hours. Exposure to IL22 increased growth of intestinal epithelial organoids derived from control mice, but not H19ΔEx1/+ mice. Overexpression of H19 in HT-29 cells increased their proliferation. Intestinal mucosa healed more slowly after withdrawal of DSS from H19ΔEx1/+ mice vs control mice. Crypt epithelial cells from H19ΔEx1/+ mice proliferated more slowly than those from control mice after exposure to LPS. H19 lncRNA bound to p53 and microRNAs that inhibit cell proliferation, including microRNA 34a and let-7; H19 lncRNA binding blocked their function, leading to increased expression of genes that promote regeneration of the epithelium. CONCLUSIONS: The level of lncRNA H19 is increased in inflamed intestinal tissues from mice and patients. The inflammatory cytokine IL22 induces expression of H19 in IECs, which is required for intestinal epithelial proliferation and mucosal healing. H19 lncRNA appears to inhibit p53 protein and microRNA 34a and let-7 to promote proliferation of IECs and epithelial regeneration.


Assuntos
Colite Ulcerativa/imunologia , Regulação da Expressão Gênica/imunologia , Interleucinas/imunologia , Mucosa Intestinal/imunologia , RNA Longo não Codificante/genética , Regeneração/fisiologia , Sepse/imunologia , Animais , Estudos de Casos e Controles , Proliferação de Células , Modelos Animais de Doenças , Células Epiteliais , Perfilação da Expressão Gênica , Células HT29 , Humanos , Inflamação , Mucosa Intestinal/fisiologia , Camundongos , RNA Longo não Codificante/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Interleucina 22
7.
Nucleic Acids Res ; 45(22): 12766-12779, 2017 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-29244185

RESUMO

Imprinted genes occur in discrete clusters that are coordinately regulated by shared DNA elements called Imprinting Control Regions. H19 and Igf2 are linked imprinted genes that play critical roles in development. Loss of imprinting (LOI) at the IGF2/H19 locus on the maternal chromosome is associated with the developmental disorder Beckwith Wiedemann Syndrome (BWS) and with several cancers. Here we use comprehensive genetic and genomic analyses to follow muscle development in a mouse model of BWS to dissect the separate and shared roles for misexpression of Igf2 and H19 in the disease phenotype. We show that LOI results in defects in muscle differentiation and hypertrophy and identify primary downstream targets: Igf2 overexpression results in over-activation of MAPK signaling while loss of H19 lncRNA prevents normal down regulation of p53 activity and therefore results in reduced AKT/mTOR signaling. Moreover, we demonstrate instances where H19 and Igf2 misexpression work separately, cooperatively, and antagonistically to establish the developmental phenotype. This study thus identifies new biochemical roles for the H19 lncRNA and underscores that LOI phenotypes are multigenic so that complex interactions will contribute to disease outcomes.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Impressão Genômica , Fator de Crescimento Insulin-Like II/genética , Mutação , RNA Longo não Codificante/genética , Animais , Síndrome de Beckwith-Wiedemann/metabolismo , Diferenciação Celular/genética , Células Cultivadas , Modelos Animais de Doenças , Perfilação da Expressão Gênica/métodos , Humanos , Fator de Crescimento Insulin-Like II/metabolismo , Camundongos , Músculo Esquelético/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , RNA Longo não Codificante/metabolismo , Transdução de Sinais/genética
8.
PLoS Genet ; 12(7): e1006200, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27466807

RESUMO

Polycomb group response elements (PREs) in Drosophila are DNA-elements that recruit Polycomb proteins (PcG) to chromatin and regulate gene expression. PREs are easily recognizable in the Drosophila genome as strong peaks of PcG-protein binding over discrete DNA fragments; many small but statistically significant PcG peaks are also observed in PcG domains. Surprisingly, in vivo deletion of the four characterized strong PREs from the PcG regulated invected-engrailed (inv-en) gene complex did not disrupt the formation of the H3K27me3 domain and did not affect inv-en expression in embryos or larvae suggesting the presence of redundant PcG recruitment mechanism. Further, the 3D-structure of the inv-en domain was only minimally altered by the deletion of the strong PREs. A reporter construct containing a 7.5kb en fragment that contains three weak peaks but no large PcG peaks forms an H3K27me3 domain and is PcG-regulated. Our data suggests a model for the recruitment of PcG-complexes to Drosophila genes via interactions with multiple, weak PREs spread throughout an H3K27me3 domain.


Assuntos
Cromatina/genética , Proteínas de Ligação a DNA/genética , Proteínas do Grupo Polycomb/genética , Elementos de Resposta/genética , Animais , Proteínas de Ligação a DNA/biossíntese , Drosophila melanogaster/genética , Regulação da Expressão Gênica no Desenvolvimento , Genoma de Inseto , Histona Desmetilases/genética , Discos Imaginais/crescimento & desenvolvimento , Discos Imaginais/metabolismo , Larva/genética , Larva/crescimento & desenvolvimento , Proteínas do Grupo Polycomb/biossíntese , Proteínas do Grupo Polycomb/química , Ligação Proteica/genética , Domínios Proteicos/genética
9.
Proc Natl Acad Sci U S A ; 113(14): 3826-31, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-27001825

RESUMO

Polycomb group (PcG) proteins are responsible for maintaining the silenced transcriptional state of many developmentally regulated genes. PcG proteins are organized into multiprotein complexes that are recruited to DNA via cis-acting elements known as "Polycomb response elements" (PREs). In Drosophila, PREs consist of binding sites for many different DNA-binding proteins, some known and others unknown. Identification of these DNA-binding proteins is crucial to understanding the mechanism of PcG recruitment to PREs. We report here the identification of Combgap (Cg), a sequence-specific DNA-binding protein that is involved in recruitment of PcG proteins. Cg can bind directly to PREs via GTGT motifs and colocalizes with the PcG proteins Pleiohomeotic (Pho) and Polyhomeotic (Ph) at the majority of PREs in the genome. In addition, Cg colocalizes with Ph at a number of targets independent of Pho. Loss of Cg leads to decreased recruitment of Ph at only a subset of sites; some of these sites are binding sites for other Polycomb repressive complex 1 (PRC1) components, others are not. Our data suggest that Cg can recruit Ph in the absence of PRC1 and illustrate the diversity and redundancy of PcG protein recruitment mechanisms.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Complexo Repressor Polycomb 1/genética , Proteínas do Grupo Polycomb/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Sítios de Ligação , Diferenciação Celular , DNA/genética , Proteínas de Ligação a DNA/genética , Motivos de Nucleotídeos/genética , Proteínas do Grupo Polycomb/metabolismo
10.
Immunol Cell Biol ; 95(5): 473-477, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28244489

RESUMO

V-(D)-J recombination generates the antigen receptor diversity necessary for immune cell function, while allelic exclusion ensures that each cell expresses a single antigen receptor. V-(D)-J recombination of the Ig, Tcrb, Tcrg and Tcrd antigen receptor genes is ordered and sequential so that only one allele generates a productive rearrangement. The mechanism controlling sequential rearrangement of antigen receptor genes, in particular how only one allele is selected to initiate recombination while at least temporarily leaving the other intact, remains unresolved. Genomic imprinting, a widespread phenomenon wherein maternal or paternal allele inheritance determines allele activity, could represent a regulatory mechanism for controlling sequential V-(D)-J rearrangement. We used strain-specific single-nucleotide polymorphisms within antigen receptor genes to determine if maternal vs paternal inheritance could underlie chromosomal choice for the initiation of recombination. We found no parental chromosomal bias in the initiation of V-(D)-J recombination in T or B cells, eliminating genomic imprinting as a potential regulator for this tightly regulated process.


Assuntos
Cromossomos de Mamíferos/genética , Impressão Genômica , Recombinação V(D)J/genética , Animais , Feminino , Masculino , Camundongos , Mapeamento por Restrição , Análise de Sequência de DNA
11.
Biochem Biophys Res Commun ; 476(3): 121-6, 2016 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-27178213

RESUMO

Transcription of the reciprocally imprinted genes Insulin-like growth factor 2 (Igf2) and H19 is orchestrated by the 2.4-kb H19 Imprinting Control Region (H19ICR) located upstream of H19. Three known functions are associated with the H19ICR: (1) it is a germline differentially methylated region, (2) it is a transcriptional insulator, and (3) it is a transcriptional silencer. The molecular mechanisms of the DMR and insulator functions have been well characterized but the basis for the ICR's silencer function is less well understood. In order to study the role the H19ICR intrinsically plays in gene silencing, we transferred the 2.4-kb H19ICR to a heterologous non-imprinted location on chromosome 5, upstream of the alpha fetoprotein (Afp) promoter. Independent of its orientation, the 2.4-kb H19ICR silences transcription from the paternal Afp promoter. Thus silencing is a function intrinsic to this DNA element. Further, ICR mediated silencing is a developmental process that, unexpectedly, does not occur through DNA methylation at the target promoter.


Assuntos
Metilação de DNA , Inativação Gênica , Impressão Genômica , Regiões Promotoras Genéticas , Animais , Fator de Crescimento Insulin-Like II/genética , Camundongos , RNA Longo não Codificante/genética , Transcrição Gênica , alfa-Fetoproteínas/genética
12.
Nucleic Acids Res ; 41(2): 817-26, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23221643

RESUMO

Developmentally regulated transcription often depends on physical interactions between distal enhancers and their cognate promoters. Recent genomic analyses suggest that promoter-promoter interactions might play a similarly critical role in organizing the genome and establishing cell-type-specific gene expression. The Igf2/H19 locus has been a valuable model for clarifying the role of long-range interactions between cis-regulatory elements. Imprinted expression of the linked, reciprocally imprinted genes is explained by parent-of-origin-specific chromosomal loop structures between the paternal Igf2 or maternal H19 promoters and their shared tissue-specific enhancer elements. Here, we further analyze these loop structures for their composition and their impact on expression of the linked long non-coding RNA, Nctc1. We show that Nctc1 is co-regulated with Igf2 and H19 and physically interacts with the shared muscle enhancer. In fact, all three co-regulated genes have the potential to interact not only with the shared enhancer but also with each other via their enhancer interactions. Furthermore, developmental and genetic analyses indicate functional significance for these promoter-promoter interactions. Altogether, we present a novel mechanism to explain developmental specific imprinting of Nctc1 and provide new information about enhancer mechanisms and about the role of chromatin domains in establishing gene expression patterns.


Assuntos
Elementos Facilitadores Genéticos , Impressão Genômica , Regiões Promotoras Genéticas , RNA Longo não Codificante/genética , Animais , Cromossomos de Mamíferos/química , DNA/química , Loci Gênicos , Fator de Crescimento Insulin-Like II/biossíntese , Fator de Crescimento Insulin-Like II/genética , Camundongos , Camundongos Congênicos , Músculo Esquelético/metabolismo , RNA Polimerase II/metabolismo , RNA Longo não Codificante/biossíntese , Ativação Transcricional
13.
Nucleic Acids Res ; 41(17): 8126-34, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23842673

RESUMO

In eukaryotic cells, gene expression is mediated by enhancer activation of RNA polymerase at distant promoters. Recently, distinctions between enhancers and promoters have been blurred by the discovery that enhancers are associated with RNA polymerase and are sites of RNA synthesis. Here, we present an analysis of the insulin-like growth factor 2/H19 muscle enhancer. This enhancer includes a short conserved core element that is organized into chromatin typical of mammalian enhancers, binds tissue-specific transcription factors and functions on its own in vitro to activate promoter transcription. However, in a chromosomal context, this element is not sufficient to activate distant promoters. Instead, enhancer function also requires transcription in cis of a long non-coding RNA, Nctc1. Thus, the insulin-like growth factor 2/H19 enhancer is an active transcriptional complex whose own transcription is essential to its function.


Assuntos
Elementos Facilitadores Genéticos , Fator de Crescimento Insulin-Like II/genética , RNA Longo não Codificante/genética , Transcrição Gênica , Animais , Células Cultivadas , Cromatina/metabolismo , Epigênese Genética , Camundongos , Mioblastos/metabolismo , Regiões Promotoras Genéticas , RNA Longo não Codificante/biossíntese
14.
Nucleic Acids Res ; 40(16): 7753-65, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22718969

RESUMO

Insulators regulate transcription as they modulate the interactions between enhancers and promoters by organizing the chromatin into distinct domains. To gain better understanding of the nature of chromatin domains defined by insulators, we analyzed the ability of an insulator to interfere in VDJ recombination, a process that is critically dependent on long-range interactions between diverse types of cis-acting DNA elements. A well-established CTCF-dependent transcriptional insulator, H19 imprint control region (H19-ICR), was inserted in the mouse TCRß locus by genetic manipulation. Analysis of the mutant mice demonstrated that the insulator retains its CTCF and position-dependent enhancer-blocking potential in this heterologous context in vivo. Remarkably, the inserted H19-ICR appears to have the ability to modulate cis-DNA interactions between recombination signal sequence elements of the TCRß locus leading to a dramatically altered usage of Vß segments for Vß-to-DßJß recombination in the mutant mice. This reveals a novel ability of CTCF to govern long range cis-DNA interactions other than enhancer-promoter interactions and suggests that CTCF-dependent insulators may play a diverse and complex role in genome organization beyond transcriptional control. Our functional analysis of mutated TCRß locus supports the emerging role of CTCF in governing VDJ recombination.


Assuntos
Rearranjo Gênico da Cadeia beta dos Receptores de Antígenos dos Linfócitos T , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T , Elementos Isolantes , Proteínas Repressoras/metabolismo , Recombinação V(D)J , Animais , Fator de Ligação a CCCTC , Loci Gênicos , Região Variável de Imunoglobulina/genética , Camundongos , Camundongos Congênicos , Regiões Promotoras Genéticas , RNA Longo não Codificante/genética , Timócitos/imunologia
15.
JCI Insight ; 7(3)2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-34990403

RESUMO

Catecholaminergic polymorphic ventricular tachycardia (CPVT) is an arrhythmia syndrome caused by gene mutations that render RYR2 Ca release channels hyperactive, provoking spontaneous Ca release and delayed afterdepolarizations (DADs). What remains unknown is the cellular source of ventricular arrhythmia triggered by DADs: Purkinje cells in the conduction system or ventricular cardiomyocytes in the working myocardium. To answer this question, we used a genetic approach in mice to knock out cardiac calsequestrin either in Purkinje cells or in ventricular cardiomyocytes. Total loss of calsequestrin in the heart causes a severe CPVT phenotype in mice and humans. We found that loss of calsequestrin only in ventricular myocytes produced a full-blown CPVT phenotype, whereas mice with loss of calsequestrin only in Purkinje cells were comparable to WT mice. Subendocardial chemical ablation or restoration of calsequestrin expression in subendocardial cardiomyocytes neighboring Purkinje cells was sufficient to protect against catecholamine-induced arrhythmias. In silico modeling demonstrated that DADs in ventricular myocardium can trigger full action potentials in the Purkinje fiber, but not vice versa. Hence, ectopic beats in CPVT are likely generated at the Purkinje-myocardial junction via a heretofore unrecognized tissue mechanism, whereby DADs in the ventricular myocardium trigger full action potentials in adjacent Purkinje cells.


Assuntos
Calsequestrina/genética , Regulação da Expressão Gênica , Frequência Cardíaca/fisiologia , Células de Purkinje/patologia , RNA/genética , Taquicardia Ventricular/diagnóstico , Animais , Calsequestrina/biossíntese , Linhagem Celular , Modelos Animais de Doenças , Camundongos Knockout , Células de Purkinje/metabolismo , Taquicardia Ventricular/genética , Taquicardia Ventricular/fisiopatologia
16.
Sci Adv ; 8(48): eadd4136, 2022 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-36449618

RESUMO

Cohesin rings interact with DNA and modulate the expression of thousands of genes. NIPBL loads cohesin onto chromosomes, and WAPL takes it off. Haploinsufficiency for NIPBL causes a developmental disorder, Cornelia de Lange syndrome (CdLS), that is modeled by Nipbl+/- mice. Mutations in WAPL have not been shown to cause disease or gene expression changes in mammals. Here, we show dysregulation of >1000 genes in WaplΔ/+ embryonic mouse brain. The patterns of dysregulation are highly similar in Wapl and Nipbl heterozygotes, suggesting that Wapl mutations may also cause human disease. Since WAPL and NIPBL have opposite effects on cohesin's association with DNA, we asked whether decreasing Wapl dosage could correct phenotypes seen in Nipbl+/- mice. Gene expression and embryonic growth are partially corrected, but perinatal lethality is not. Our data are consistent with the view that cohesin dynamics play a key role in regulating gene expression.


Assuntos
Encéfalo , Transcriptoma , Humanos , Feminino , Gravidez , Animais , Camundongos , Fenótipo , Mutação , Heterozigoto , Mamíferos , Proteínas de Ciclo Celular/genética , Proteínas
17.
Pflugers Arch ; 461(1): 45-52, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20978783

RESUMO

Thyroid hormones T3/T4 participate in the fine tuning of development and performance. The formation of thyroid hormones requires the accumulation of I(-) by the electrogenic Na(+)/I(-) symporter, which depends on the electrochemical gradient across the cell membrane and thus on K(+) channel activity. The present paper explored whether Kcnq1, a widely expressed voltage-gated K(+) channel, participates in the regulation of thyroid function. To this end, Kcnq1 expression was determined by RT-PCR, confocal microscopy, and thyroid function analyzed in Kcnq1 deficient mice (Kcnq1 ( -/- )) and their wild-type littermates (Kcnq1 ( +/+ )). Moreover, Kcnq1 abundance and current were determined in the thyroid FRTL-5 cell line. Furthermore, mRNA encoding KCNQ1 and the subunits KCNE1-5 were discovered in human thyroid tissue. According to patch-clamp TSH (10 mUnits/ml) induced a voltage-gated K(+) current in FRTL-5 cells, which was inhibited by the Kcnq inhibitor chromanol (10 µM). Despite a tendency of TSH plasma concentrations to be higher in Kcnq1 ( -/- ) than in Kcnq1 ( +/+ ) mice, the T3 and T4 plasma concentrations were significantly smaller in Kcnq1 ( -/- ) than in Kcnq1 ( +/+ ) mice. Moreover, body temperature was significantly lower in Kcnq1 ( -/- ) than in Kcnq1 ( +/+ ) mice. In conclusion, Kcnq1 is required for proper function of thyroid glands.


Assuntos
Hipotireoidismo/genética , Canal de Potássio KCNQ1/genética , Animais , Temperatura Corporal/efeitos dos fármacos , Temperatura Corporal/fisiologia , Cromanos/farmacologia , Feminino , Humanos , Hipotireoidismo/fisiopatologia , Imuno-Histoquímica , Canal de Potássio KCNQ1/biossíntese , Canal de Potássio KCNQ1/deficiência , Masculino , Camundongos , RNA Mensageiro/metabolismo , Glândula Tireoide/metabolismo , Tireotropina/sangue , Tiroxina/sangue , Tri-Iodotironina/sangue
18.
Elife ; 102021 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-34402430

RESUMO

Maternal loss of imprinting (LOI) at the H19/IGF2 locus results in biallelic IGF2 and reduced H19 expression and is associated with Beckwith--Wiedemann syndrome (BWS). We use mouse models for LOI to understand the relative importance of Igf2 and H19 mis-expression in BWS phenotypes. Here we focus on cardiovascular phenotypes and show that neonatal cardiomegaly is exclusively dependent on increased Igf2. Circulating IGF2 binds cardiomyocyte receptors to hyperactivate mTOR signaling, resulting in cellular hyperplasia and hypertrophy. These Igf2-dependent phenotypes are transient: cardiac size returns to normal once Igf2 expression is suppressed postnatally. However, reduced H19 expression is sufficient to cause progressive heart pathologies including fibrosis and reduced ventricular function. In the heart, H19 expression is primarily in endothelial cells (ECs) and regulates EC differentiation both in vivo and in vitro. Finally, we establish novel mouse models to show that cardiac phenotypes depend on H19 lncRNA interactions with Mirlet7 microRNAs.


Assuntos
Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/patologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Animais , Síndrome de Beckwith-Wiedemann/genética , Síndrome de Beckwith-Wiedemann/metabolismo , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Diferenciação Celular , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fenótipo , Transdução de Sinais
19.
Mol Cell Biol ; 27(9): 3499-510, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17339341

RESUMO

Transcriptional insulators are specialized cis-acting elements that protect promoters from inappropriate activation by distal enhancers. The H19 imprinting control region (ICR) functions as a CTCF-dependent, methylation-sensitive transcriptional insulator. We analyzed several insertional mutations and demonstrate that the ICR can function as a methylation-regulated maternal chromosome-specific insulator in novel chromosomal contexts. We used chromosome conformation capture and chromatin immunoprecipitation assays to investigate the configuration of cis-acting elements at these several insertion sites. By comparing maternal and paternal organizations on wild-type and mutant chromosomes, we hoped to identify mechanisms for ICR insulator function. We found that promoter and enhancer elements invariably associate to form DNA loop domains at transcriptionally active loci. Conversely, active insulators always prevent these promoter-enhancer interactions. Instead, the ICR insulator forms novel loop domains by associating with the blocked promoters and enhancers. We propose that these associations are fundamental to insulator function.


Assuntos
Cromossomos de Mamíferos/genética , Elementos Isolantes/genética , Animais , Fator de Ligação a CCCTC , Imunoprecipitação da Cromatina , Metilação de DNA , Proteínas de Ligação a DNA/genética , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica , Fígado/embriologia , Fígado/metabolismo , Camundongos , Mães , Células Musculares/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas/genética , Proteínas Repressoras/genética , alfa-Fetoproteínas/genética
20.
J Clin Invest ; 116(9): 2510-20, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16932808

RESUMO

Cardiac calsequestrin (Casq2) is thought to be the key sarcoplasmic reticulum (SR) Ca2+ storage protein essential for SR Ca2+ release in mammalian heart. Human CASQ2 mutations are associated with catecholaminergic ventricular tachycardia. However, homozygous mutation carriers presumably lacking functional Casq2 display surprisingly normal cardiac contractility. Here we show that Casq2-null mice are viable and display normal SR Ca2+ release and contractile function under basal conditions. The mice exhibited striking increases in SR volume and near absence of the Casq2-binding proteins triadin-1 and junctin; upregulation of other Ca2+ -binding proteins was not apparent. Exposure to catecholamines in Casq2-null myocytes caused increased diastolic SR Ca2+ leak, resulting in premature spontaneous SR Ca2+ releases and triggered beats. In vivo, Casq2-null mice phenocopied the human arrhythmias. Thus, while the unique molecular and anatomic adaptive response to Casq2 deletion maintains functional SR Ca2+ storage, lack of Casq2 also causes increased diastolic SR Ca2+ leak, rendering Casq2-null mice susceptible to catecholaminergic ventricular arrhythmias.


Assuntos
Cálcio/fisiologia , Calsequestrina/deficiência , Retículo Sarcoplasmático/fisiologia , Taquicardia Ventricular/genética , Alelos , Animais , Sequência de Bases , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Calsequestrina/genética , Cruzamentos Genéticos , Primers do DNA , Diástole , Eletrocardiografia , Éxons , Coração/fisiologia , Homozigoto , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Knockout , Polimorfismo Genético , RNA/genética , RNA/isolamento & purificação
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa