Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 187
Filtrar
1.
Yi Chuan ; 44(9): 783-797, 2022 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-36384955

RESUMO

Human ß-thalassemia is closely associated with aberrant expression of ß-like globin genes. Human ß-like globin genes are organized in the order of 5'-ε-Gγ-Aγ-δ-ß-3' within the ß-globin locus. The expression of ß-like globin genes is regulated by 3'HS1 and five DNase I hypersensitive sites (5'HS5~5'HS1) in a locus control region. The 5'HS2 enhancer transcribes enhancer RNA and regulates the expression of ε-globin, γ-globin and ß-globin. To further study the function of 5'HS2, we detected the local 3D genomic architecture via chromatin conformation capture experiments and used CRISPR/ Cas9-based DNA fragment editing to delete 5'HS2 in human K562 leukaemia cells. In this study, we found that 5'HS2-mediated chromatin interactions were enriched in a topologically associated domain that was bordered by 3'HS1 and 5'HS5. Within this topologically associated domain, 5'HS2 is highly close to the promoter regions of HBE1, HBG2 and HBG1. Upon deletion of the 5'HS2 enhancer, 91 genes were significantly down-regulated with reduced abundance of H3K27ac at their promoter regions. These down-regulated genes are mainly associated with oxygen transport, immune response, cell adhesion, anti-oxidant and thrombosis. These data suggested that many genes associated with functions of erythrocytes were decreased at transcriptional levels upon deletion of the 5'HS2 enhancer.


Assuntos
Elementos Facilitadores Genéticos , Região de Controle de Locus Gênico , Globinas beta , Humanos , Sequência de Bases , Globinas beta/genética , Cromatina/genética , DNA/genética , Células K562 , Região de Controle de Locus Gênico/genética , Deleção de Sequência
2.
Development ; 149(1)2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34982814

RESUMO

Imprinting control region (ICR1) controls the expression of the Igf2 and H19 genes in a parent-of-origin specific manner. Appropriate expression of the Igf2-H19 locus is fundamental for normal fetal development, yet the importance of ICR1 in the placental production of hormones that promote maternal nutrient allocation to the fetus is unknown. To address this, we used a novel mouse model to selectively delete ICR1 in the endocrine junctional zone (Jz) of the mouse placenta (Jz-ΔICR1). The Jz-ΔICR1 mice exhibit increased Igf2 and decreased H19 expression specifically in the Jz. This was accompanied by an expansion of Jz endocrine cell types due to enhanced rates of proliferation and increased expression of pregnancy-specific glycoprotein 23 in the placenta of both fetal sexes. However, changes in the endocrine phenotype of the placenta were related to sexually-dimorphic alterations to the abundance of Igf2 receptors and downstream signalling pathways (Pi3k-Akt and Mapk). There was no effect of Jz-ΔICR1 on the expression of targets of the H19-embedded miR-675 or on fetal weight. Our results demonstrate that ICR1 controls placental endocrine capacity via sex-dependent changes in signalling.


Assuntos
Glândulas Endócrinas/metabolismo , Fator de Crescimento Insulin-Like II/genética , Região de Controle de Locus Gênico , Placenta/metabolismo , RNA Longo não Codificante/genética , Transdução de Sinais , Animais , Feminino , Loci Gênicos , Impressão Genômica , Glicoproteínas/genética , Glicoproteínas/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/metabolismo , Gravidez , Proteínas da Gravidez/genética , Proteínas da Gravidez/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Longo não Codificante/metabolismo
3.
Cell Mol Gastroenterol Hepatol ; 13(4): 1276-1296, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34954189

RESUMO

BACKGROUND & AIMS: Sporadic colorectal cancers arise from initiating mutations in APC, producing oncogenic ß-catenin/TCF-dependent transcriptional reprogramming. Similarly, the tumor suppressor axis regulated by the intestinal epithelial receptor GUCY2C is among the earliest pathways silenced in tumorigenesis. Retention of the receptor, but loss of its paracrine ligands, guanylin and uroguanylin, is an evolutionarily conserved feature of colorectal tumors, arising in the earliest dysplastic lesions. Here, we examined a mechanism of GUCY2C ligand transcriptional silencing by ß-catenin/TCF signaling. METHODS: We performed RNA sequencing analysis of 4 unique conditional human colon cancer cell models of ß-catenin/TCF signaling to map the core Wnt-transcriptional program. We then performed a comparative analysis of orthogonal approaches, including luciferase reporters, chromatin immunoprecipitation sequencing, CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats) knockout, and CRISPR epigenome editing, which were cross-validated with human tissue chromatin immunoprecipitation sequencing datasets, to identify functional gene enhancers mediating GUCY2C ligand loss. RESULTS: RNA sequencing analyses reveal the GUCY2C hormones as 2 of the most sensitive targets of ß-catenin/TCF signaling, reflecting transcriptional repression. The GUCY2C hormones share an insulated genomic locus containing a novel locus control region upstream of the guanylin promoter that mediates the coordinated silencing of both genes. Targeting this region with CRISPR epigenome editing reconstituted GUCY2C ligand expression, overcoming gene inactivation by mutant ß-catenin/TCF signaling. CONCLUSIONS: These studies reveal DNA elements regulating corepression of GUCY2C ligand transcription by ß-catenin/TCF signaling, reflecting a novel pathophysiological step in tumorigenesis. They offer unique genomic strategies that could reestablish hormone expression in the context of canonical oncogenic mutations to reconstitute the GUCY2C axis and oppose transformation.


Assuntos
Neoplasias Colorretais , beta Catenina , Carcinogênese/genética , Cateninas/genética , Cateninas/metabolismo , Neoplasias Colorretais/patologia , Humanos , Ligantes , Região de Controle de Locus Gênico , Receptores de Enterotoxina/genética , Receptores de Enterotoxina/metabolismo , Fatores de Transcrição TCF/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
4.
Int J Mol Sci ; 22(14)2021 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-34299205

RESUMO

Gain and loss of DNA methylation in cells is a dynamic process that tends to achieve an equilibrium. Many factors are involved in maintaining the balance between DNA methylation and demethylation. Previously, it was shown that methyl-DNA protein Kaiso may attract NCoR, SMRT repressive complexes affecting histone modifications. On the other hand, the deficiency of Kaiso resulted in reduced methylation of ICR in H19/Igf2 locus and Oct4 promoter in mouse embryonic fibroblasts. However, nothing is known about how Kaiso influences DNA methylation at the genome level. Here we show that deficiency of Kaiso led to whole-genome hypermethylation, using Kaiso deficient human renal cancer cell line obtained via CRISPR/CAS9 genome editing. However, Kaiso serves to protect genic regions, enhancers, and regions with a low level of histone modifications from demethylation. We detected hypomethylation of binding sites for Oct4 and Nanog in Kaiso deficient cells. Kaiso immunoprecipitated with de novo DNA methyltransferases DNMT3a/3b, but not with maintenance methyltransferase DNMT1. Thus, Kaiso may attract methyltransferases to surrounding regions and modulate genome methylation in renal cancer cells apart from being methyl DNA binding protein.


Assuntos
Metilação de DNA , Impressão Genômica , Fator de Crescimento Insulin-Like II/metabolismo , Região de Controle de Locus Gênico , RNA Longo não Codificante/genética , Fatores de Transcrição/metabolismo , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Edição de Genes , Células HEK293 , Humanos , Fator de Crescimento Insulin-Like II/genética , Regiões Promotoras Genéticas , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , DNA Metiltransferase 3B
5.
Mol Ther ; 29(4): 1625-1638, 2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33515514

RESUMO

Ongoing clinical trials for treatment of beta-globinopathies by gene therapy involve the transfer of the beta-globin gene, which requires integration of three to four copies per genome in most target cells. This high proviral load may increase genome toxicity, potentially limiting the safety of this therapy and relegating its use to total body myeloablation. We hypothesized that introducing an additional hypersensitive site from the locus control region, the complete sequence of the second intron of the beta-globin gene, and the ankyrin insulator may enhance beta-globin expression. We identified a construct, ALS20, that synthesized significantly higher adult hemoglobin levels than those of other constructs currently used in clinical trials. These findings were confirmed in erythroblastic cell lines and in primary cells isolated from sickle cell disease patients. Bone marrow transplantation studies in beta-thalassemia mice revealed that ALS20 was curative at less than one copy per genome. Injection of human CD34+ cells transduced with ALS20 led to safe, long-term, and high polyclonal engraftment in xenograft experiments. Successful treatment of beta-globinopathies with ALS20 could potentially be achieved at less than two copies per genome, minimizing the risk of cytotoxic events and lowering the intensity of myeloablation.


Assuntos
Anemia Falciforme/genética , Transplante de Medula Óssea , Terapia Genética , Globinas beta/genética , Talassemia beta/genética , Anemia Falciforme/sangue , Anemia Falciforme/patologia , Anemia Falciforme/terapia , Animais , Expressão Gênica/genética , Vetores Genéticos/genética , Vetores Genéticos/farmacologia , Hemoglobinas/genética , Xenoenxertos , Humanos , Lentivirus/genética , Região de Controle de Locus Gênico/genética , Camundongos , Transdução Genética , Globinas beta/uso terapêutico , Talassemia beta/sangue , Talassemia beta/patologia , Talassemia beta/terapia
6.
Nucleic Acids Res ; 49(3): 1383-1396, 2021 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-33476375

RESUMO

Super-enhancers (SEs) mediate high transcription levels of target genes. Previous studies have shown that SEs recruit transcription complexes and generate enhancer RNAs (eRNAs). We characterized transcription at the human and murine ß-globin locus control region (LCR) SE. We found that the human LCR is capable of recruiting transcription complexes independently from linked globin genes in transgenic mice. Furthermore, LCR hypersensitive site 2 (HS2) initiates the formation of bidirectional transcripts in transgenic mice and in the endogenous ß-globin gene locus in murine erythroleukemia (MEL) cells. HS2 3'eRNA is relatively unstable and remains in close proximity to the globin gene locus. Reducing the abundance of HS2 3'eRNA leads to a reduction in ß-globin gene transcription and compromises RNA polymerase II (Pol II) recruitment at the promoter. The Integrator complex has been shown to terminate eRNA transcription. We demonstrate that Integrator interacts downstream of LCR HS2. Inducible ablation of Integrator function in MEL or differentiating primary human CD34+ cells causes a decrease in expression of the adult ß-globin gene and accumulation of Pol II and eRNA at the LCR. The data suggest that transcription complexes are assembled at the LCR and transferred to the globin genes by mechanisms that involve Integrator mediated release of Pol II and eRNA from the LCR.


Assuntos
Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , RNA/metabolismo , Transcrição Gênica , Globinas beta/genética , Adulto , Animais , Linhagem Celular Tumoral , Endorribonucleases/genética , Feto , Humanos , Fígado/embriologia , Fígado/metabolismo , Região de Controle de Locus Gênico , Camundongos Transgênicos , RNA/fisiologia , RNA Polimerase II/metabolismo , Globinas beta/biossíntese
7.
Sci Rep ; 10(1): 19634, 2020 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-33184340

RESUMO

Most of the human genome is non-coding. However, some of the non-coding part is transcriptionally active. In humans, the tandemly repeated (TR) pericentromeric non-coding DNA-human satellites 2 and 3 (HS2, HS3)-are transcribed in somatic cells. These transcripts are also found in pre- and post-implantation embryos. The aim of this study was to analyze HS2/HS3 transcription and cellular localization of transcripts in human maturating oocytes. The maternal HS2/HS3 TR transcripts transcribed from both strands were accumulated in the ooplasm in GV-MI oocytes as shown by DNA-RNA FISH (fluorescence in-situ hybridization). The transcripts' content was higher in GV oocytes than in somatic cumulus cells according to real-time PCR. Using bioinformatics analysis, we demonstrated the presence of polyadenylated HS2 and HS3 RNAs in datasets of GV and MII oocyte transcriptomes. The transcripts shared a high degree of homology with HS2, HS3 transcripts previously observed in cancer cells. The HS2/HS3 transcripts were revealed by a combination of FISH and immunocytochemical staining within membraneless RNP structures that contained DEAD-box helicases DDX5 and DDX4. The RNP structures were closely associated with mitochondria, and are therefore similar to membraneless bodies described previously only in oogonia. These membraneless structures may be a site for spatial sequestration of RNAs and proteins in both maturating oocytes and cancer cells.


Assuntos
DNA Satélite/genética , Mitocôndrias/química , Mitocôndrias/metabolismo , Oócitos/citologia , Oócitos/metabolismo , Oogênese , Ribonucleoproteínas/metabolismo , Células do Cúmulo/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Região de Controle de Locus Gênico , Ribonucleoproteínas/química , Transcriptoma
8.
Mol Cell ; 80(5): 845-861.e10, 2020 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-33232656

RESUMO

Locus control region (LCR) functions define cellular identity and have critical roles in diseases such as cancer, although the hierarchy of structural components and associated factors that drive functionality are incompletely understood. Here we show that OCA-B, a B cell-specific coactivator essential for germinal center (GC) formation, forms a ternary complex with the lymphoid-enriched OCT2 and GC-specific MEF2B transcription factors and that this complex occupies and activates an LCR that regulates the BCL6 proto-oncogene and is uniquely required by normal and malignant GC B cells. Mechanistically, through OCA-B-MED1 interactions, this complex is required for Mediator association with the BCL6 promoter. Densely tiled CRISPRi screening indicates that only LCR segments heavily bound by this ternary complex are essential for its function. Our results demonstrate how an intimately linked complex of lineage- and stage-specific factors converges on specific and highly essential enhancer elements to drive the function of a cell-type-defining LCR.


Assuntos
Linfócitos B/imunologia , Centro Germinativo/imunologia , Região de Controle de Locus Gênico/imunologia , Animais , Linfócitos B/citologia , Linhagem Celular Tumoral , Centro Germinativo/citologia , Células HEK293 , Humanos , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/imunologia , Camundongos , Camundongos Knockout , Transportador 2 de Cátion Orgânico/genética , Transportador 2 de Cátion Orgânico/imunologia , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-bcl-6/genética , Proteínas Proto-Oncogênicas c-bcl-6/imunologia , Transativadores/genética , Transativadores/imunologia
9.
J Trauma Acute Care Surg ; 89(1): 173-178, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32118827

RESUMO

BACKGROUND: The genomic landscape of gallbladder disease remains poorly understood. We sought to examine the association between genetic variants and the development of cholecystitis. METHODS: The Biobank of a large multi-institutional health care system was used. All patients with cholecystitis were identified using International Statistical Classification of Diseases, 10th Revision, codes and genotyped across six batches. To control for population stratification, data were restricted to that from individuals of European genomic ancestry using a multidimensional scaling approach. The association between single nucleotide polymorphisms and cholecystitis was evaluated with a mixed linear model-based analysis, controlling for age, sex, and obesity. The threshold for significance was set at 5 × 10. RESULTS: Of 24,635 patients (mean ± SD age, 60.1 ± 16.7 years; 13,022 females [52.9%]), 900 had cholecystitis (mean ± SD age, 65.4 ± 14.3 years; 496 females [55.1%]). After meta-analysis, three single nucleotide polymorphisms on chromosome 5p15 exceeded the threshold for significance (p < 5 × 10). The phenotypic variance of cholecystitis explained by genetics and controlling for sex and obesity was estimated to be 17.9%. CONCLUSION: Using a multi-institutional genomic Biobank, we report that a region on chromosome 5p15 is associated with the development of cholecystitis that can be used to identify patients at risk. LEVEL OF EVIDENCE: Prognostic and epidemiological, Level III.


Assuntos
Colecistite/genética , Cromossomos Humanos Par 5 , Variação Genética , Estudo de Associação Genômica Ampla , Polimorfismo de Nucleotídeo Único , Idoso , Feminino , Predisposição Genética para Doença , Genótipo , Humanos , Região de Controle de Locus Gênico , Aprendizado de Máquina , Masculino , Pessoa de Meia-Idade , Fenótipo
10.
Gene ; 743: 144606, 2020 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-32199948

RESUMO

DNA demethylation and suppression of de novo DNA methylation are activities that maintain an unmethylated state. However, the strength of these two activities at the same locus has not been estimated separately. Furthermore, the association between these two activities and the unmethylated state remains unclear. Octamer-binding transcription factor-binding sequences (OBSs) and CCCTC-binding factor-binding sequences (CBSs) within the mouse H19-imprinted control region (ICR) are involved in the induction of DNA demethylation and maintenance of the unmethylated state in mouse undifferentiated embryonic cell lines. To reveal the association between the two cis-elements and the two unmethylated state maintenance activities in maintaining the unmethylated state of the ICR, we evaluated the altered DNA methylation levels at sites that were initially methylated or unmethylated using a stable transfection-based assay, and estimated the strength of the two unmethylated state maintenance activities separately via a Poisson process model that described the DNA methylation state regulatory process. Although DNA demethylation depending on OBSs affected almost the entire ICR, DNA demethylation depending on CBSs occurred near CBSs, resulting in redundant demethylation of CBS regions. Detailed analysis of the CBS4 region suggested that OBSs were required to induce unmethylated state maintenance activities, and that CBSs-dependent activities contributed, but diminished, during incubation when protection of the CBS4 region by OBSs-dependent activities was absent. Analysis via the Poisson process model indicated that the unmethylated state at the CBS4 region was maintained by OBSs-dependent suppression of de novo DNA methylation rather than DNA demethylation. We propose that the hierarchical regulation of redundant protection of the CBS region via cooperation between the two unmethylated state maintenance activities is a potential function of the ICR that effectively maintains allele-specific methylation status in the same DNA sequence.


Assuntos
Desmetilação do DNA , Metilação de DNA/genética , Impressão Genômica , Região de Controle de Locus Gênico/genética , Animais , Fator de Ligação a CCCTC/metabolismo , Linhagem Celular Tumoral , Camundongos , Fator 3 de Transcrição de Octâmero/metabolismo , RNA Longo não Codificante/genética
11.
Mol Ther ; 28(1): 328-340, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31628051

RESUMO

ß-globin lentiviral vectors (ß-LV) have faced challenges in clinical translation for gene therapy of sickle cell disease (SCD) due to low titer and sub-optimal gene transfer to hematopoietic stem and progenitor cells (HSPCs). To overcome the challenge of preserving efficacious expression while increasing vector performance, we used published genomic and epigenomic data available through ENCODE to redefine enhancer element boundaries of the ß-globin locus control region (LCR) to construct novel ENCODE core sequences. These novel LCR elements were used to design a ß-LV of reduced proviral length, termed CoreGA-AS3-FB, produced at higher titers and possessing superior gene transfer to HSPCs when compared to the full-length parental ß-LV at equal MOI. At low vector copy number, vectors containing the ENCODE core sequences were capable of reversing the sickle phenotype in a mouse model of SCD. These studies provide a ß-LV that will be beneficial for gene therapy of SCD by significantly reducing the cost of vector production and extending the vector supply.


Assuntos
Anemia Falciforme/terapia , Terapia Genética/métodos , Vetores Genéticos , Lentivirus/genética , Região de Controle de Locus Gênico/genética , Transdução Genética/métodos , Globinas beta/genética , Animais , Células da Medula Óssea/metabolismo , Modelos Animais de Doenças , Células HEK293 , Voluntários Saudáveis , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Fenótipo , Transfecção
12.
PLoS Genet ; 15(8): e1008339, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31461456

RESUMO

The NAD+-dependent histone deacetylase Sir2 was originally identified in Saccharomyces cerevisiae as a silencing factor for HML and HMR, the heterochromatic cassettes utilized as donor templates during mating-type switching. MATa cells preferentially switch to MATα using HML as the donor, which is driven by an adjacent cis-acting element called the recombination enhancer (RE). In this study we demonstrate that Sir2 and the condensin complex are recruited to the RE exclusively in MATa cells, specifically to the promoter of a small gene within the right half of the RE known as RDT1. We also provide evidence that the RDT1 promoter functions as a locus control region (LCR) that regulates both transcription and long-range chromatin interactions. Sir2 represses RDT1 transcription until it is removed from the promoter in response to a dsDNA break at the MAT locus induced by HO endonuclease during mating-type switching. Condensin is also recruited to the RDT1 promoter and is displaced upon HO induction, but does not significantly repress RDT1 transcription. Instead condensin appears to promote mating-type donor preference by maintaining proper chromosome III architecture, which is defined by the interaction of HML with the right arm of chromosome III, including MATa and HMR. Remarkably, eliminating Sir2 and condensin recruitment to the RDT1 promoter disrupts this structure and reveals an aberrant interaction between MATa and HMR, consistent with the partially defective donor preference for this mutant. Global condensin subunit depletion also impairs mating-type switching efficiency and donor preference, suggesting that modulation of chromosome architecture plays a significant role in controlling mating-type switching, thus providing a novel model for dissecting condensin function in vivo.


Assuntos
Cromossomos Fúngicos/genética , Genes Fúngicos Tipo Acasalamento/genética , Região de Controle de Locus Gênico/genética , Proteínas Reguladoras de Informação Silenciosa de Saccharomyces cerevisiae/metabolismo , Sirtuína 2/metabolismo , Adenosina Trifosfatases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Loci Gênicos/genética , Complexos Multiproteicos/metabolismo , Regiões Promotoras Genéticas/genética , Recombinação Genética , Saccharomyces cerevisiae , Proteínas Reguladoras de Informação Silenciosa de Saccharomyces cerevisiae/genética , Sirtuína 2/genética
14.
Cell ; 171(1): 103-119.e18, 2017 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-28938112

RESUMO

It is now established that Bcl11b specifies T cell fate. Here, we show that in developing T cells, the Bcl11b enhancer repositioned from the lamina to the nuclear interior. Our search for factors that relocalized the Bcl11b enhancer identified a non-coding RNA named ThymoD (thymocyte differentiation factor). ThymoD-deficient mice displayed a block at the onset of T cell development and developed lymphoid malignancies. We found that ThymoD transcription promoted demethylation at CTCF bound sites and activated cohesin-dependent looping to reposition the Bcl11b enhancer from the lamina to the nuclear interior and to juxtapose the Bcl11b enhancer and promoter into a single-loop domain. These large-scale changes in nuclear architecture were associated with the deposition of activating epigenetic marks across the loop domain, plausibly facilitating phase separation. These data indicate how, during developmental progression and tumor suppression, non-coding transcription orchestrates chromatin folding and compartmentalization to direct with high precision enhancer-promoter communication.


Assuntos
Elementos Facilitadores Genéticos , Regiões Promotoras Genéticas , RNA não Traduzido/genética , Proteínas Repressoras/genética , Linfócitos T/citologia , Proteínas Supressoras de Tumor/genética , Animais , Fator de Ligação a CCCTC , Cromatina/metabolismo , Leucemia/genética , Região de Controle de Locus Gênico , Linfoma/genética , Camundongos , Lâmina Nuclear/metabolismo , Proteínas Repressoras/metabolismo , Linfócitos T/metabolismo , Timo/citologia , Timo/metabolismo , Transcrição Gênica
15.
Nucleic Acids Res ; 45(14): 8255-8268, 2017 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-28520978

RESUMO

Mechanistic studies in erythroid cells indicate that LDB1, as part of a GATA1/TAL1/LMO2 complex, brings erythroid-expressed genes into proximity with enhancers for transcription activation. The role of co-activators in establishing this long-range interaction is poorly understood. Here we tested the contributions of the RNA Pol II pre-initiation complex (PIC), mediator and cohesin to establishment of locus control region (LCR)/ß-globin proximity. CRISPR/Cas9 editing of the ß-globin promoter to eliminate the RNA Pol II PIC by deleting the TATA-box resulted in loss of transcription, but enhancer-promoter interaction was unaffected. Additional deletion of the promoter GATA1 site eliminated LDB1 complex and mediator occupancy and resulted in loss of LCR/ß-globin proximity. To separate the roles of LDB1 and mediator in LCR looping, we expressed a looping-competent but transcription-activation deficient form of LDB1 in LDB1 knock down cells: LCR/ß-globin proximity was restored without mediator core occupancy. Further, Cas9-directed tethering of mutant LDB1 to the ß-globin promoter forced LCR loop formation in the absence of mediator or cohesin occupancy. Moreover, ENCODE data and our chromatin immunoprecipitation results indicate that cohesin is almost completely absent from validated and predicted LDB1-regulated erythroid enhancer-gene pairs. Thus, lineage specific factors largely mediate enhancer-promoter looping in erythroid cells independent of mediator and cohesin.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Elementos Facilitadores Genéticos/genética , Regulação Leucêmica da Expressão Gênica , Proteínas com Domínio LIM/genética , Animais , Sequência de Bases , Western Blotting , Sistemas CRISPR-Cas , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas com Domínio LIM/metabolismo , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/patologia , Região de Controle de Locus Gênico/genética , Camundongos , Regiões Promotoras Genéticas/genética , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Globinas beta/genética , Coesinas
16.
Medicina (B Aires) ; 76(6): 383-389, 2016.
Artigo em Espanhol | MEDLINE | ID: mdl-27959850

RESUMO

Different hemoglobin isoforms are expressed during the embryonic, fetal and postnatal stages. They are formed by combination of polypeptide chains synthesized from the α- and ß-globin gene clusters. Based on the fact that the presence of high hemoglobin F levels is beneficial in both sickle cell disease and severe thalassemic syndromes, a revision of the regulation of the ß-globin cluster expression is proposed, especially regarding the genes encoding the y-globin chains (HBG1 and HBG2). In this review we describe the current knowledge about transcription factors and epigenetic regulators involved in the switches of the ß-globin cluster. It is expected that the consolidation of knowledge in this field will allow finding new therapeutic targets for the treatment of hemoglobinopathies.


Assuntos
Expressão Gênica , Hemoglobinopatias/genética , Família Multigênica/genética , Globinas beta/genética , Regulação da Expressão Gênica , Hemoglobinopatias/terapia , Humanos , Região de Controle de Locus Gênico , alfa-Globinas/genética
17.
Medicina (B.Aires) ; Medicina (B.Aires);76(6): 383-389, dic. 2016. ilus, graf
Artigo em Espanhol | LILACS | ID: biblio-841616

RESUMO

Durante la etapa embrionaria, el desarrollo fetal y la vida posnatal se expresan isoformas funcionalmente distintas de hemoglobina, producto de la combinación de cadenas polipeptídicas sintetizadas a partir de los distintos genes que componen las familias de α- y β-globina. En función de que la presencia de altos niveles de hemoglobina fetal (Hb F) es beneficiosa en síndromes falciformes y talasémicos graves, se plantea revisar las bases de la regulación de la expresión de los genes de la familia de β-globina, en particular los genes que codifican las cadenas de γ-globina (HBG1 y HBG2). En este trabajo se revisan los conocimientos sobre factores de transcripción y reguladores epigenéticos que gobiernan los eventos de encendido y apagado de los genes de la familia de β-globina. Se espera que la consolidación de estos conocimientos permita hallar nuevos blancos terapéuticos para el tratamiento de hemoglobinopatías.


Different hemoglobin isoforms are expressed during the embryonic, fetal and postnatal stages. They are formed by combination of polypeptide chains synthesized from the α- and β- globin gene clusters. Based on the fact that the presence of high hemoglobin F levels is beneficial in both sickle cell disease and severe thalassemic syndromes, a revision of the regulation of the β-globin cluster expression is proposed, especially regarding the genes encoding the γ-globin chains (HBG1 and HBG2). In this review we describe the current knowledge about transcription factors and epigenetic regulators involved in the switches of the β-globin cluster. It is expected that the consolidation of knowledge in this field will allow finding new therapeutic targets for the treatment of hemoglobinopathies.


Assuntos
Humanos , Expressão Gênica , Família Multigênica/genética , Globinas beta/genética , Hemoglobinopatias/genética , Regulação da Expressão Gênica , Região de Controle de Locus Gênico , alfa-Globinas/genética , Hemoglobinopatias/terapia
18.
Immunity ; 45(3): 497-512, 2016 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-27637145

RESUMO

During the humoral immune response, B cells undergo a dramatic change in phenotype to enable antibody affinity maturation in germinal centers (GCs). Using genome-wide chromosomal conformation capture (Hi-C), we found that GC B cells undergo massive reorganization of the genomic architecture that encodes the GC B cell transcriptome. Coordinate expression of genes that specify the GC B cell phenotype-most prominently BCL6-was achieved through a multilayered chromatin reorganization process involving (1) increased promoter connectivity, (2) formation of enhancer networks, (3) 5' to 3' gene looping, and (4) merging of gene neighborhoods that share active epigenetic marks. BCL6 was an anchor point for the formation of GC-specific gene and enhancer loops on chromosome 3. Deletion of a GC-specific, highly interactive locus control region upstream of Bcl6 abrogated GC formation in mice. Thus, large-scale and multi-tiered genomic three-dimensional reorganization is required for coordinate expression of phenotype-driving gene sets that determine the unique characteristics of GC B cells.


Assuntos
Afinidade de Anticorpos/imunologia , Linfócitos B/imunologia , Genoma/imunologia , Centro Germinativo/imunologia , Região de Controle de Locus Gênico/imunologia , Animais , Formação de Anticorpos/imunologia , Cromossomos Humanos Par 3/imunologia , Epigênese Genética/imunologia , Humanos , Imunidade Humoral/imunologia , Camundongos , Regiões Promotoras Genéticas/imunologia , Proteínas Proto-Oncogênicas c-bcl-6/imunologia
19.
Hemoglobin ; 40(4): 236-9, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27492765

RESUMO

In our previous studies on the Iranian ß-thalassemia (ß-thal) patients, we identified an association between the severity of the ß-thal phenotype and the polymorphic palindromic site at the 5' hypersensitive site 4-locus control region (5'HS4-LCR) of the ß-globin gene cluster. Furthermore, a linkage disequilibrium was observed between this region and XmnI-HBG2 in the patient population. Based on this data, it was suggested that the well-recognized phenotype-ameliorating role assigned to positive XmnI could be associated with its linked elements in the LCR. To investigate the functional significance of polymorphisms at the 5'HS4-LCR, we studied its influence on binding of transcription factors. Web-based predictions of transcription factor binding revealed a binding site for runt-related transcription factor 1 (RUNX1), when the allele at the center of the palindrome (TGGGG(A/G)CCCCA) was A but not when it was G. Furthermore, electromobility shift assay (EMSA) presented evidence in support of allele-specific binding of RUNX1 to 5'HS4. Considering that RUNX1 is a well-known regulator of hematopoiesis, these preliminary data suggest the importance of further studies to confirm this interaction and consequently investigate its functional and phenotypical relevance. These studies could help us to understand the molecular mechanism behind the phenotype modifying role of the 5'HS4-LCR polymorphic palindromic region (rs16912979), which has been observed in previous studies.


Assuntos
Alelos , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Região de Controle de Locus Gênico , Sítios de Ligação , Humanos , Sequências Repetidas Invertidas , Fenótipo , Ligação Proteica
20.
Sci Rep ; 6: 24023, 2016 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-27087367

RESUMO

Sexually dimorphic gene expression is commonly found in the liver, and many of these genes are linked to different incidences of liver diseases between sexes. However, the mechanism of sexually dimorphic expression is still not fully understood. In this study, a pCAG-eGFP transgenic mouse strain with a specific transgene integration site in the Akr1A1 locus presented male-biased EGFP expression in the liver, and the expression was activated by testosterone during puberty. The integration of the pCAG-eGFP transgene altered the epigenetic regulation of the adjacent chromatin, including increased binding of STAT5b, a sexually dimorphic expression regulator, and the transformation of DNA methylation from hypermethylation into male-biased hypomethylation. Through this de novo sexually dimorphic expression of the transgene, the Akr1A1(eGFP) mouse provides a useful model to study the mechanisms and the dynamic changes of sexually dimorphic gene expression during either development or pathogenesis of the liver.


Assuntos
Epigênese Genética , Proteínas de Fluorescência Verde/metabolismo , Fígado/metabolismo , Transgenes , Animais , Ilhas de CpG , Citomegalovirus/genética , Metilação de DNA , Estradiol/farmacologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/genética , Região de Controle de Locus Gênico , Masculino , Camundongos Transgênicos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Caracteres Sexuais , Testosterona/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA