Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(33): e2307287120, 2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37552759

RESUMEN

The nucleosome remodeling and deacetylase (NuRD) complex modifies nucleosome positioning and chromatin compaction to regulate gene expression. The methyl-CpG-binding domain proteins 2 and 3 (MBD2 and MBD3) play a critical role in complex formation; however, the molecular details of how they interact with other NuRD components have yet to be fully elucidated. We previously showed that an intrinsically disordered region (IDR) of MBD2 is necessary and sufficient to bind to the histone deacetylase core of NuRD. Building on that work, we have measured the inherent structural propensity of the MBD2-IDR using solvent and site-specific paramagnetic relaxation enhancement measurements. We then used the AlphaFold2 machine learning software to generate a model of the complex between MBD2 and the histone deacetylase core of NuRD. This model is remarkably consistent with our previous studies, including the current paramagnetic relaxation enhancement data. The latter suggests that the free MBD2-IDR samples conformations similar to the bound structure. We tested this model of the complex extensively by mutating key contact residues and measuring binding using an intracellular bioluminescent resonance energy transfer assay. Furthermore, we identified protein contacts that, when mutated, disrupted gene silencing by NuRD in a cell model of fetal hemoglobin regulation. Hence, this work provides insights into the formation of NuRD and highlights critical binding pockets that may be targeted to block gene silencing for therapy. Importantly, we show that AlphaFold2 can generate a credible model of a large complex that involves an IDR that folds upon binding.


Asunto(s)
Histona Desacetilasas , Nucleosomas , Histona Desacetilasas/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Silenciador del Gen , Cromatina , Histona Desacetilasa 1/genética
2.
Proc Natl Acad Sci U S A ; 120(25): e2302254120, 2023 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-37307480

RESUMEN

During human development, there is a switch in the erythroid compartment at birth that results in silencing of expression of fetal hemoglobin (HbF). Reversal of this silencing has been shown to be effective in overcoming the pathophysiologic defect in sickle cell anemia. Among the many transcription factors and epigenetic effectors that are known to mediate HbF silencing, two of the most potent are BCL11A and MBD2-NuRD. In this report, we present direct evidence that MBD2-NuRD occupies the γ-globin gene promoter in adult erythroid cells and positions a nucleosome there that results in a closed chromatin conformation that prevents binding of the transcriptional activator, NF-Y. We show that the specific isoform, MBD2a, is required for the formation and stable occupancy of this repressor complex that includes BCL11A, MBD2a-NuRD, and the arginine methyltransferase, PRMT5. The methyl cytosine binding preference and the arginine-rich (GR) domain of MBD2a are required for high affinity binding to methylated γ-globin gene proximal promoter DNA sequences. Mutation of the methyl cytosine-binding domain (MBD) of MBD2 results in a variable but consistent loss of γ-globin gene silencing, in support of the importance of promoter methylation. The GR domain of MBD2a is also required for recruitment of PRMT5, which in turn results in placement of the repressive chromatin mark H3K8me2s at the promoter. These findings support a unified model that integrates the respective roles of BCL11A, MBD2a-NuRD, PRMT5, and DNA methylation in HbF silencing.


Asunto(s)
Hemoglobina Fetal , gamma-Globinas , Adulto , Recién Nacido , Humanos , Genes Reguladores , Factores de Transcripción , Cromatina , Citosina , Proteína-Arginina N-Metiltransferasas , Proteínas de Unión al ADN
3.
Haematologica ; 104(12): 2361-2371, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31004025

RESUMEN

As high fetal hemoglobin levels ameliorate the underlying pathophysiological defects in sickle cell anemia and beta (ß)-thalassemia, understanding the mechanisms that enforce silencing of fetal hemoglobin postnatally offers the promise of effective molecular therapy. Depletion of the Nucleosome Remodeling and Deacetylase complex member MBD2 causes a 10-20-fold increase in γ-globin gene expression in adult ß-globin locus yeast artificial chromosome transgenic mice. To determine the effect of MBD2 depletion in human erythroid cells, genome editing technology was utilized to knockout MBD2 in Human Umbilical cord Derived Erythroid Progenitor-2 cells resulting in γ/γ+ß mRNA levels of approximately 50% and approximately 40% fetal hemoglobin by high performance liquid chromatography. In contrast, MBD3 knockout had no appreciable effect on γ-globin expression. Knockdown of MBD2 in primary adult erythroid cells consistently increased γ/γ+ß mRNA ratios by approximately 10-fold resulting in approximately 30-40% γ/γ+ß mRNA levels and a corresponding increase in γ-globin protein. MBD2 exerts its repressive effects through recruitment of the chromatin remodeler CHD4 via a coiled-coil domain, and the histone deacetylase core complex via an intrinsically disordered region. Enforced expression of wild-type MBD2 in MBD2 knockout cells caused a 5-fold decrease in γ-globin mRNA while neither the coiled-coil mutant nor the intrinsically disordered region mutant MBD2 proteins had an inhibitory effect. Co-immunoprecipitation assays showed that the coiled-coil and intrinsically disorder region mutations disrupt complex formation by dissociating the CHD4 and the histone deacetylase core complex components, respectively. These results establish the MBD2 Nucleosome Remodeling and Deacetylase complex as a major silencer of fetal hemoglobin in human erythroid cells and point to the coiled-coil and intrinsically disordered region of MBD2 as potential therapeutic targets.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Eritroides/metabolismo , Hemoglobina Fetal/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Mutación , gamma-Globinas/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Adulto , Células Cultivadas , Ensamble y Desensamble de Cromatina , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Células Eritroides/citología , Humanos , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/antagonistas & inhibidores , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
4.
Blood ; 126(12): 1462-72, 2015 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-26265695

RESUMEN

Chromodomain helicase DNA-binding protein 4 (CHD4) is an ATPase that alters the phasing of nucleosomes on DNA and has recently been implicated in DNA double-stranded break (DSB) repair. Here, we show that depletion of CHD4 in acute myeloid leukemia (AML) blasts induces a global relaxation of chromatin that renders cells more susceptible to DSB formation, while concurrently impeding their repair. Furthermore, CHD4 depletion renders AML blasts more sensitive both in vitro and in vivo to genotoxic agents used in clinical therapy: daunorubicin (DNR) and cytarabine (ara-C). Sensitization to DNR and ara-C is mediated in part by activation of the ataxia-telangiectasia mutated pathway, which is preliminarily activated by a Tip60-dependent mechanism in response to chromatin relaxation and further activated by genotoxic agent-induced DSBs. This sensitization preferentially affects AML cells, as CHD4 depletion in normal CD34(+) hematopoietic progenitors does not increase their susceptibility to DNR or ara-C. Unexpectedly, we found that CHD4 is necessary for maintaining the tumor-forming behavior of AML cells, as CHD4 depletion severely restricted the ability of AML cells to form xenografts in mice and colonies in soft agar. Taken together, these results provide evidence for CHD4 as a novel therapeutic target whose inhibition has the potential to enhance the effectiveness of genotoxic agents used in AML therapy.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Antimetabolitos Antineoplásicos/uso terapéutico , Autoantígenos/genética , Citarabina/uso terapéutico , Daunorrubicina/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Animales , Línea Celular Tumoral , Roturas del ADN de Doble Cadena/efectos de los fármacos , Femenino , Humanos , Leucemia Mieloide Aguda/patología , Ratones Endogámicos NOD , Ratones SCID , Interferencia de ARN , Células Tumorales Cultivadas
5.
Nucleic Acids Res ; 43(6): 3100-13, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25753662

RESUMEN

The MBD2-NuRD (Nucleosome Remodeling and Deacetylase) complex is an epigenetic reader of DNA methylation that regulates genes involved in normal development and neoplastic diseases. To delineate the architecture and functional interactions of the MBD2-NuRD complex, we previously solved the structures of MBD2 bound to methylated DNA and a coiled-coil interaction between MBD2 and p66α that recruits the CHD4 nucleosome remodeling protein to the complex. The work presented here identifies novel structural and functional features of a previously uncharacterized domain of MBD2 (MBD2IDR). Biophysical analyses show that the MBD2IDR is an intrinsically disordered region (IDR). However, despite this inherent disorder, MBD2IDR increases the overall binding affinity of MBD2 for methylated DNA. MBD2IDR also recruits the histone deacetylase core components (RbAp48, HDAC2 and MTA2) of NuRD through a critical contact region requiring two contiguous amino acid residues, Arg(286) and Leu(287). Mutating these residues abrogates interaction of MBD2 with the histone deacetylase core and impairs the ability of MBD2 to repress the methylated tumor suppressor gene PRSS8 in MDA-MB-435 breast cancer cells. These findings expand our knowledge of the multi-dimensional interactions of the MBD2-NuRD complex that govern its function.


Asunto(s)
Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/química , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Metilación de ADN , Proteínas de Unión al ADN/genética , Epigénesis Genética , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/genética , Proteínas Intrínsecamente Desordenadas/metabolismo , Cinética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido
6.
J Biol Chem ; 289(3): 1294-302, 2014 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-24307175

RESUMEN

Although highly homologous to other methylcytosine-binding domain (MBD) proteins, MBD3 does not selectively bind methylated DNA, and thus the functional role of MBD3 remains in question. To explore the structural basis of its binding properties and potential function, we characterized the solution structure and binding distribution of the MBD3 MBD on hydroxymethylated, methylated, and unmethylated DNA. The overall fold of this domain is very similar to other MBDs, yet a key loop involved in DNA binding is more disordered than previously observed. Specific recognition of methylated DNA constrains the structure of this loop and results in large chemical shift changes in NMR spectra. Based on these spectral changes, we show that MBD3 preferentially localizes to methylated and, to a lesser degree, unmethylated cytosine-guanosine dinucleotides (CpGs), yet does not distinguish between hydroxymethylated and unmethylated sites. Measuring residual dipolar couplings for the different bound states clearly shows that the MBD3 structure does not change between methylation-specific and nonspecific binding modes. Furthermore, residual dipolar couplings measured for MBD3 bound to methylated DNA can be described by a linear combination of those for the methylation and nonspecific binding modes, confirming the preferential localization to methylated sites. The highly homologous MBD2 protein shows similar but much stronger localization to methylated as well as unmethylated CpGs. Together, these data establish the structural basis for the relative distribution of MBD2 and MBD3 on genomic DNA and their observed occupancy at active and inactive CpG-rich promoters.


Asunto(s)
Proteínas Aviares/química , Islas de CpG/fisiología , Proteínas de Unión al ADN/química , ADN/química , Animales , Proteínas Aviares/genética , Proteínas Aviares/metabolismo , Pollos , ADN/genética , ADN/metabolismo , Metilación de ADN/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Unión Proteica/fisiología , Estructura Terciaria de Proteína
7.
Blood ; 121(17): 3493-501, 2013 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-23444401

RESUMEN

An understanding of the human fetal to adult hemoglobin switch offers the potential to ameliorate ß-type globin gene disorders such as sickle cell anemia and ß-thalassemia through activation of the fetal γ-globin gene. Chromatin modifying complexes, including MBD2-NuRD and GATA-1/FOG-1/NuRD, play a role in γ-globin gene silencing, and Mi2ß (CHD4) is a critical component of NuRD complexes. We observed that knockdown of Mi2ß relieves γ-globin gene silencing in ß-YAC transgenic murine chemical inducer of dimerization hematopoietic cells and in CD34(+) progenitor-derived human primary adult erythroid cells. We show that independent of MBD2-NuRD and GATA-1/FOG-1/NuRD, Mi2ß binds directly to and positively regulates both the KLF1 and BCL11A genes, which encode transcription factors critical for γ-globin gene silencing during ß-type globin gene switching. Remarkably, <50% knockdown of Mi2ß is sufficient to significantly induce γ-globin gene expression without disrupting erythroid differentiation of primary human CD34(+) progenitors. These results indicate that Mi2ß is a potential target for therapeutic induction of fetal hemoglobin.


Asunto(s)
Autoantígenos/metabolismo , Células Eritroides/metabolismo , Hemoglobina Fetal/genética , Regulación de la Expresión Génica , Silenciador del Gen , Células Madre Hematopoyéticas/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , gamma-Globinas/genética , Adulto , Animales , Autoantígenos/genética , Proteínas Portadoras/metabolismo , Diferenciación Celular , Células Cultivadas , Inmunoprecipitación de Cromatina , Células Eritroides/citología , Hemoglobina Fetal/antagonistas & inhibidores , Hemoglobina Fetal/metabolismo , Células Madre Hematopoyéticas/citología , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Ratones , Ratones Transgénicos , Proteínas Nucleares/metabolismo , ARN Interferente Pequeño/genética , Proteínas Represoras , gamma-Globinas/antagonistas & inhibidores , gamma-Globinas/metabolismo
8.
Proc Natl Acad Sci U S A ; 108(18): 7487-92, 2011 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-21490301

RESUMEN

Nucleosome remodeling complexes comprise several large families of chromatin modifiers that integrate multiple epigenetic control signals to play key roles in cell type-specific transcription regulation. We previously isolated a methyl-binding domain protein 2 (MBD2)-containing nucleosome remodeling and deacetylation (NuRD) complex from primary erythroid cells and showed that MBD2 contributes to DNA methylation-dependent embryonic and fetal ß-type globin gene silencing during development in vivo. Here we present structural and biophysical details of the coiled-coil interaction between MBD2 and p66α, a critical component of the MBD2-NuRD complex. We show that enforced expression of the isolated p66α coiled-coil domain relieves MBD2-mediated globin gene silencing and that the expressed peptide interacts only with a subset of components of the MBD2-NuRD complex that does not include native p66α or Mi-2. These results demonstrate the central importance of the coiled-coil interaction and suggest that MBD2-dependent DNA methylation-driven gene silencing can be disrupted by selectively targeting this coiled-coil complex.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Epigénesis Genética/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Modelos Moleculares , Proteínas Represoras/metabolismo , Western Blotting , Metilación de ADN/genética , Cartilla de ADN/genética , Silenciador del Gen , Humanos , Inmunoprecipitación , Interferencia de ARN
9.
Nucleic Acids Res ; 39(15): 6741-52, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21531701

RESUMEN

The epigenetic code of DNA methylation is interpreted chiefly by methyl cytosine binding domain (MBD) proteins which in turn recruit multiprotein co-repressor complexes. We previously isolated one such complex, MBD2-NuRD, from primary erythroid cells and have shown it contributes to embryonic/fetal ß-type globin gene silencing during development. This complex has been implicated in silencing tumor suppressor genes in a variety of human tumor cell types. Here we present structural details of chicken MBD2 bound to a methylated DNA sequence from the ρ-globin promoter to which it binds in vivo and mediates developmental transcriptional silencing in normal erythroid cells. While previous studies have failed to show sequence specificity for MBD2 outside of the symmetric mCpG, we find that this domain binds in a single orientation on the ρ-globin target DNA sequence. Further, we show that the orientation and affinity depends on guanine immediately following the mCpG dinucleotide. Dynamic analyses show that DNA binding stabilizes the central ß-sheet, while the N- and C-terminal regions of the protein maintain mobility. Taken together, these data lead to a model in which DNA binding stabilizes the MBD2 structure and that binding orientation and affinity is influenced by the DNA sequence surrounding the central mCpG.


Asunto(s)
Proteínas Aviares/química , Metilación de ADN , Proteínas de Unión al ADN/química , ADN/química , Animales , Secuencia de Bases , Pollos , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Estructura Terciaria de Proteína
10.
Cancer ; 117(20): 4772-8, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21455994

RESUMEN

BACKGROUND: African American race and uninsurance are associated with undertreatment and poor survival in solid tumor cancers. This relationship has not been examined in acute myeloid leukemia (AML) where absence of treatment or treatment delays can result in death within weeks or months. Induction followed by consolidation treatment, in contrast, has a high probability for remission or cure. We examined the relationship between race and health insurance and inpatient chemotherapy and survival in AML patients between the ages of 21 and 64 years. We also examined inpatient costs associated with inpatient treatment. METHODS: We used population-based data from the Virginia Cancer Registry and the Virginia Health Information discharge data for patients diagnosed with AML between 1999 and 2006 (n = 523). Adjusted logistic regression was used to measure the relationship between the independent variables and chemotherapy. We used the Cox proportional hazards method to estimate survival. RESULTS: Uninsured patients were more likely to be untreated than their privately insured counterparts (odds ratio, 4.40; 95% confidence interval, 1.85-10.49) and had a higher likelihood of death (hazard ratio, 1.29; 95% confidence interval, 1.02-1.84). Once treatment was adjusted in the survival analyses, differences between insurance groups were not statistically significant. The median 1-year cost of inpatient care following diagnosis for patients who received chemotherapy exceeded $100,000. CONCLUSION: This study addressed the urgency for health insurance that affords access to care. Without treatment, the outcome of AML is death within only a few months; with treatment, the chance for long-term remission or even cure exists.


Asunto(s)
Antineoplásicos/economía , Negro o Afroamericano/estadística & datos numéricos , Costos de la Atención en Salud , Accesibilidad a los Servicios de Salud/economía , Seguro de Salud , Leucemia Mieloide Aguda , Pacientes no Asegurados/estadística & datos numéricos , Población Blanca/estadística & datos numéricos , Adulto , Antineoplásicos/uso terapéutico , Trasplante de Médula Ósea/economía , Factores de Confusión Epidemiológicos , Etnicidad/estadística & datos numéricos , Femenino , Accesibilidad a los Servicios de Salud/estadística & datos numéricos , Costos de Hospital , Humanos , Pacientes Internos , Tiempo de Internación/economía , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/economía , Leucemia Mieloide Aguda/mortalidad , Modelos Logísticos , Masculino , Medicaid , Medicare , Persona de Mediana Edad , Oportunidad Relativa , Modelos de Riesgos Proporcionales , Sistema de Registros , Retratamiento/economía , Estados Unidos , Virginia/epidemiología
11.
Blood Cells Mol Dis ; 46(3): 212-9, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21296012

RESUMEN

During erythroid development, the embryonic ε-globin gene becomes silenced as erythropoiesis shifts from the yolk sac to the fetal liver where γ-globin gene expression predominates. Previous studies have shown that the ε-globin gene is autonomously silenced through promoter proximal cis-acting sequences in adult erythroid cells. We have shown a role for the methylcytosine binding domain protein 2 (MBD2) in the developmental silencing of the avian embryonic ρ-globin and human fetal γ-globin genes. To determine the roles of MBD2 and DNA methylation in human ε-globin gene silencing, transgenic mice containing all sequences extending from the 5' hypersensitive site 5 (HS5) of the ß-globin locus LCR to the human γ-globin gene promoter were generated. These mice show correct developmental expression and autonomous silencing of the transgene. Either the absence of MBD2 or treatment with the DNA methyltransferase inhibitor 5-azacytidine increases ε-globin transgene expression by 15-20 fold in adult mice. Adult mice containing the entire human ß-globin locus also show an increase in expression of both the ε-globin gene transgene and endogenous ε(Y) and ß(H1) genes in the absence of MBD2. These results indicate that the human ε-globin gene is subject to multilayered silencing mediated in part by MBD2.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Globinas épsilon/genética , Animales , Azacitidina/farmacología , Metilación de ADN , Eritroblastos/metabolismo , Eritrocitos/metabolismo , Femenino , Orden Génico , Hemoglobinas Anormales/metabolismo , Humanos , Región de Control de Posición/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Globinas beta/metabolismo , gamma-Globinas/metabolismo
12.
Blood ; 117(1): 4-5, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-21212288
13.
Pharmacol Ther ; 184: 98-111, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29128342

RESUMEN

DNA methylation represents a fundamental epigenetic modification that regulates chromatin architecture and gene transcription. Many diseases, including cancer, show aberrant methylation patterns that contribute to the disease phenotype. DNA methylation inhibitors have been used to block methylation dependent gene silencing to treat hematopoietic neoplasms and to restore expression of developmentally silenced genes. However, these inhibitors disrupt methylation globally and show significant off-target toxicities. As an alternative approach, we have been studying readers of DNA methylation, the 5-methylcytosine binding domain family of proteins, as potential therapeutic targets to restore expression of aberrantly and developmentally methylated and silenced genes. In this review, we discuss the role of DNA methylation in gene regulation and cancer development, the structure and function of the 5-methylcytosine binding domain family of proteins, and the possibility of targeting the complexes these proteins form to treat human disease.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/efectos de los fármacos , Terapia Molecular Dirigida/métodos , Neoplasias/tratamiento farmacológico , Animales , Metilación de ADN/efectos de los fármacos , Humanos , Modelos Moleculares
14.
Methods Mol Biol ; 1698: 259-274, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29076096

RESUMEN

Human umbilical cord blood is a rich source of hematopoietic stem and progenitor cells. CD34+ cells in umbilical cord blood are more primitive than those in peripheral blood or bone marrow, and can proliferate at a high rate and differentiate into multiple cell types. In this protocol, a dependable method is described for the isolation of fetal CD34+ cells from umbilical cord blood and expanding these cells in culture. The cells can then be in vitro differentiated along an erythroid pathway, while simultaneously performing knockdown of a gene of choice. The use of lentiviral vectors that express small hairpin RNA (shRNA) is an efficient method to downregulate genes. Flow cytometric analyses are used to enrich for erythroid cells. Using these methods, one can generate in vitro differentiated cells to use for quantitative reverse transcriptase PCR and other purposes.


Asunto(s)
Diferenciación Celular/genética , Sangre Fetal/citología , Técnicas de Silenciamiento del Gen , Vectores Genéticos/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Lentivirus/genética , Antígenos CD34/metabolismo , Técnicas de Cultivo de Célula , Separación Celular , Células Eritroides/citología , Células Eritroides/metabolismo , Citometría de Flujo , Expresión Génica , Células HEK293 , Humanos , Transfección
15.
Mol Cell Biol ; 24(14): 6194-204, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15226423

RESUMEN

The human major histocompatibility complex (MHC) class Ib gene, HLA-E, codes for the major ligand of the inhibitory receptor NK-G-2A, which is present on most natural killer (NK) cells and some CD8(+) cytotoxic T lymphocytes. We have previously shown that gamma interferon (IFN-gamma) induction of HLA-E gene transcription is mediated through a distinct IFN-gamma-responsive element, the IFN response region (IRR), in all cell types studied. We have now identified and characterized a cell type-restricted enhancer of IFN-gamma-mediated induction of HLA-E gene transcription, designated the upstream interferon response region (UIRR), which is located immediately upstream of the IRR. The UIRR mediates a three- to eightfold enhancement of IFN-gamma induction of HLA-E transcription in some cell lines but not in others, and it functions only in the presence of an adjacent IRR. The UIRR contains a variant GATA binding site (AGATAC) that is critical to both IFN-gamma responsiveness and to the formation of a specific binding complex containing GATA-1 in K562 cell nuclear extracts. The binding of GATA-1 to this site in response to IFN-gamma was confirmed in vivo in a chromatin immunoprecipitation assay. Forced expression of GATA-1 in nonexpressing U937 cells resulted in a four- to fivefold enhancement of the IFN-gamma response from HLA-E promoter constructs containing a wild-type but not a GATA-1 mutant UIRR sequence and increased the IFN-gamma response of the endogenous HLA-E gene. Knockdown of GATA-1 expression in K562 cells resulted in a approximately 4-fold decrease in the IFN-gamma response of the endogenous HLA-E gene, consistent with loss of the increase in IFN-gamma response of HLA-E promoter-driven constructs containing the UIRR in wild-type K562 cells. Coexpression of wild-type and mutant adenovirus E1a proteins that sequester p300/CBP eliminated IFN-gamma-mediated enhancement through the UIRR, but only partially reduced induction through the IRR, implicating p300/CBP binding to Stat-1alpha at the IRR in the recruitment of GATA-1 to mediate the cooperation between the UIRR and IRR. We propose that the GATA-1 transcription factor represents a cell type-restricted mediator of IFN-gamma induction of the HLA-E gene.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Antígenos HLA/genética , Antígenos de Histocompatibilidad Clase I/genética , Interferón gamma/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Secuencia de Bases , Línea Celular , Proteínas de Unión al ADN/genética , Factores de Unión al ADN Específico de las Células Eritroides , Factor de Transcripción GATA1 , Genes Reporteros , Antígenos HLA/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Datos de Secuencia Molecular , Unión Proteica , Elementos de Respuesta , Factores de Transcripción/genética , Transfección , Antígenos HLA-E
16.
Transl Res ; 165(1): 115-25, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24880147

RESUMEN

The developmental regulation of globin gene expression has served as an important model for understanding higher eukaryotic transcriptional control mechanisms. During human erythroid development, there is a sequential switch from expression of the embryonic ε-globin gene to the fetal É£-globin gene in utero, and postpartum the É£-globin gene is silenced, as the ß-globin gene becomes the predominantly expressed locus. Because the expression of normally silenced fetal É£-type globin genes and resultant production of fetal hemoglobin (HbF) in adult erythroid cells can ameliorate the pathophysiological consequences of both abnormal ß-globin chains in sickle cell anemia and deficient ß-globin chain production in ß-thalassemia, understanding the complex mechanisms of this developmental switch has direct translational clinical relevance. Of particular interest for translational research are the factors that mediate silencing of the É£-globin gene in adult stage erythroid cells. In addition to the regulatory roles of transcription factors and their cognate DNA sequence motifs, there has been a growing appreciation of the role of epigenetic signals and their cognate factors in gene regulation, and in particular in gene silencing through chromatin. Much of the information about epigenetic silencing stems from studies of globin gene regulation. As discussed here, the term epigenetics refers to postsynthetic modifications of DNA and chromosomal histone proteins that affect gene expression and can be inherited through somatic cell replication. A full understanding of the molecular mechanisms of epigenetic silencing of HbF expression should facilitate the development of more effective treatment of ß-globin chain hemoglobinopathies.


Asunto(s)
Epigénesis Genética , Células Eritroides/metabolismo , Hemoglobina Fetal/genética , Animales , Ensayos Clínicos como Asunto , Metilación de ADN , Eritropoyesis/genética , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Hemoglobinopatías/genética , Histonas/metabolismo , Humanos , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Interferencia de ARN , Factores de Transcripción/metabolismo , Investigación Biomédica Traslacional , Globinas beta/genética , Talasemia beta/genética , gamma-Globinas/genética
18.
Mol Cancer Res ; 9(8): 1152-62, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21693597

RESUMEN

Methyl cytosine binding domain protein 2 (MBD2) has been shown to bind to and mediate repression of methylated tumor suppressor genes in cancer cells, where repatterning of CpG methylation and associated gene silencing is common. We have investigated the role of MBD2 in breast cancer cell growth and tumor suppressor gene expression. We show that stable short hairpin RNA (shRNA)-mediated knockdown of MBD2 leads to growth suppression of cultured human mammary epithelial cancer lines, SK-BR-3, MDA-MB-231, and MDA-MB-435. The peak antiproliferative occurs only after sustained, stable MBD2 knockdown. Once established, the growth inhibition persists over time and leads to a markedly decreased propensity for aggressive breast cancer cell lines to form in vivo xenograft tumors in Bagg Albino (BALB)/C nu/nu mice. The growth effects of MBD2 knockdown are accompanied by derepression of tumor suppressor genes, including DAPK1 and KLK10. Chromatin immunoprecipitation assays and bisulfite sequencing show MBD2 binding directly to the hyper methylated and CpG-rich promoters of both DAPK1 and KLK10. Remarkably, the promoter CpG island-associated methylation of these genes remained stable despite robust transcriptional activation in MBD2 knockdown cells. Expression of a shRNA-resistant MBD2 protein resulted in restoration of growth and resilencing of the MBD2-dependent tumor suppressor genes. Our data suggest that uncoupling CpG methylation from repressive chromatin remodeling and histone modifications by removing MBD2 is sufficient to initiate and maintain tumor suppressor gene transcription and suppress neoplastic cell growth. These results show a role for MBD2 in cancer progression and provide support for the prospect of targeting MBD2 therapeutically in aggressive breast cancers.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas de Unión al ADN/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Islas de CpG/genética , Metilación de ADN/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos BALB C , Estadificación de Neoplasias , ARN Interferente Pequeño , Células Tumorales Cultivadas
19.
Curr Top Dev Biol ; 82: 85-116, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18282518

RESUMEN

The sequence complexity of the known vertebrate genomes alone is insufficient to account for the diversity between individuals of a species. Although our knowledge of vertebrate biology has evolved substantially with the growing compilation of sequenced genomes, understanding the temporal and spatial regulation of genes remains fundamental to fully exploiting this information. The importance of epigenetic factors in gene regulation was first hypothesized decades ago when biologists posited that methylation of DNA could heritably alter gene expression [Holliday and Pugh, 1975. Science 187(4173), 226-232; Riggs, 1975. Cytogenet. and Cell Genet.14(1), 9-25; Scarano et al., 1967. Proc. Natl. Acad. Sci. USA 57(5), 1394-1400)]. It was subsequently shown that vertebrate DNA methylation, almost exclusively at the 5' position of cytosine in the dinucleotide CpG, played a role in a number of processes including embryonic development, genetic imprinting, cell differentiation, and tumorigenesis. At the time of this writing, a large and growing list of genes is known to exhibit DNA methylation-dependent regulation, and we understand in some detail the mechanisms employed by cells in using methylation as a regulatory modality. In this context, we revisit one of the original systems in which the role of DNA methylation in vertebrate gene regulation during development was described and studied: erythroid cells. We briefly review the recent advances in our understanding of DNA methylation and, in particular, its regulatory role in red blood cells during differentiation and development. We also address DNA methylation as a component of erythroid chromatin architecture, and the interdependence of CpG methylation and histone modification.


Asunto(s)
Metilación de ADN , Epigénesis Genética/genética , Células Eritroides/citología , Células Eritroides/metabolismo , Eritropoyesis/genética , Regulación del Desarrollo de la Expresión Génica , Animales , Globinas/genética , Humanos
20.
J Immunol ; 177(12): 8612-9, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17142760

RESUMEN

IFNs regulate most MHC class I genes by stimulating transcription initiation. As shown previously, IFN-gamma controls HLA-A expression primarily at the posttranscriptional level. We have defined two 8-base sequences in a 39-nucleotide region in the 3'-transcribed region of the HLA-A gene that are required for the posttranscriptional response to IFN-gamma. Stimulation of HLA-A expression by IFN-gamma requires nuclear export of HLA-A mRNA by chromosome maintenance region 1 (CRM-1). Treatment of cells with leptomycin B, a specific inhibitor of CRM-1, completely inhibited IFN-gamma induction of HLA-A. Expression of a truncated, dominant-negative form of the nucleoporin NUP214/CAN, DeltaCAN, that specifically interacts with CRM-1, also prevented IFN-gamma stimulation of HLA-A, providing confirmation of the role of CRM-1. Increased expression of HLA-A induced by IFN-gamma also requires protein methylation, as shown by the fact that treatment of SK-N-MC cells or HeLa cells with the PRMT1 inhibitor 5'-methyl-5'-thioadenosine abolished the cellular response to IFN-gamma. In contrast with HLA-A, IFN-gamma-induced expression of the HLA class Ib gene, HLA-E, was not affected by either 5'-methyl-5'-thioadenosine or leptomycin B. These results provide proof of principle that it is possible to differentially modulate the IFN-gamma-induced expression of the HLA-E and HLA-A genes, whose products often mediate opposing effects on cellular immunity to tumor cells, pathogens, and autoantigens.


Asunto(s)
Transporte Activo de Núcleo Celular , Antígenos HLA-A/genética , Interferón gamma/farmacología , Carioferinas/fisiología , ARN/metabolismo , Receptores Citoplasmáticos y Nucleares/fisiología , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Antígenos HLA/genética , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Metilación , Proteína Exportina 1 , Antígenos HLA-E
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA