Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters








Publication year range
1.
Proc Natl Acad Sci U S A ; 120(33): e2307287120, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37552759

ABSTRACT

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.


Subject(s)
Histone Deacetylases , Nucleosomes , Histone Deacetylases/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Gene Silencing , Chromatin , Histone Deacetylase 1/genetics
2.
Proc Natl Acad Sci U S A ; 120(25): e2302254120, 2023 06 20.
Article in English | MEDLINE | ID: mdl-37307480

ABSTRACT

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.


Subject(s)
Fetal Hemoglobin , gamma-Globins , Adult , Infant, Newborn , Humans , Genes, Regulator , Transcription Factors , Chromatin , Cytosine , Protein-Arginine N-Methyltransferases , DNA-Binding Proteins
3.
Haematologica ; 104(12): 2361-2371, 2019 12.
Article in English | MEDLINE | ID: mdl-31004025

ABSTRACT

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.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , DNA-Binding Proteins/metabolism , Erythroid Cells/metabolism , Fetal Hemoglobin/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mutation , gamma-Globins/metabolism , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/genetics , Adult , Cells, Cultured , Chromatin Assembly and Disassembly , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Erythroid Cells/cytology , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/antagonists & inhibitors , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , ATP-Binding Cassette Sub-Family B Member 4
5.
Pharmacol Ther ; 184: 98-111, 2018 04.
Article in English | MEDLINE | ID: mdl-29128342

ABSTRACT

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.


Subject(s)
DNA Methylation , DNA-Binding Proteins/drug effects , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Animals , DNA Methylation/drug effects , Humans , Models, Molecular
6.
Methods Mol Biol ; 1698: 259-274, 2018.
Article in English | MEDLINE | ID: mdl-29076096

ABSTRACT

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.


Subject(s)
Cell Differentiation/genetics , Fetal Blood/cytology , Gene Knockdown Techniques , Genetic Vectors/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Lentivirus/genetics , Antigens, CD34/metabolism , Cell Culture Techniques , Cell Separation , Erythroid Cells/cytology , Erythroid Cells/metabolism , Flow Cytometry , Gene Expression , HEK293 Cells , Humans , Transfection
7.
Blood ; 126(12): 1462-72, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26265695

ABSTRACT

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.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Antimetabolites, Antineoplastic/therapeutic use , Autoantigens/genetics , Cytarabine/therapeutic use , Daunorubicin/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Animals , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , Female , Humans , Leukemia, Myeloid, Acute/pathology , Mice, Inbred NOD , Mice, SCID , RNA Interference , Tumor Cells, Cultured
8.
Nucleic Acids Res ; 43(6): 3100-13, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25753662

ABSTRACT

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.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/chemistry , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Amino Acid Sequence , Animals , Cell Line , DNA Methylation , DNA-Binding Proteins/genetics , Epigenesis, Genetic , Gene Knockdown Techniques , HEK293 Cells , Humans , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Kinetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
9.
Transl Res ; 165(1): 115-25, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24880147

ABSTRACT

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.


Subject(s)
Epigenesis, Genetic , Erythroid Cells/metabolism , Fetal Hemoglobin/genetics , Animals , Clinical Trials as Topic , DNA Methylation , Erythropoiesis/genetics , Gene Expression Regulation, Developmental , Gene Silencing , Hemoglobinopathies/genetics , Histones/metabolism , Humans , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , RNA Interference , Transcription Factors/metabolism , Translational Research, Biomedical , beta-Globins/genetics , beta-Thalassemia/genetics , gamma-Globins/genetics
10.
J Biol Chem ; 289(3): 1294-302, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24307175

ABSTRACT

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.


Subject(s)
Avian Proteins/chemistry , CpG Islands/physiology , DNA-Binding Proteins/chemistry , DNA/chemistry , Animals , Avian Proteins/genetics , Avian Proteins/metabolism , Chickens , DNA/genetics , DNA/metabolism , DNA Methylation/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Protein Binding/physiology , Protein Structure, Tertiary
11.
Blood ; 121(17): 3493-501, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23444401

ABSTRACT

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.


Subject(s)
Autoantigens/metabolism , Erythroid Cells/metabolism , Fetal Hemoglobin/genetics , Gene Expression Regulation , Gene Silencing , Hematopoietic Stem Cells/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , gamma-Globins/genetics , Adult , Animals , Autoantigens/genetics , Carrier Proteins/metabolism , Cell Differentiation , Cells, Cultured , Chromatin Immunoprecipitation , Erythroid Cells/cytology , Fetal Hemoglobin/antagonists & inhibitors , Fetal Hemoglobin/metabolism , Hematopoietic Stem Cells/cytology , Humans , Kruppel-Like Transcription Factors/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mice , Mice, Transgenic , Nuclear Proteins/metabolism , RNA, Small Interfering/genetics , Repressor Proteins , gamma-Globins/antagonists & inhibitors , gamma-Globins/metabolism
12.
Mol Cancer Res ; 9(8): 1152-62, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21693597

ABSTRACT

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.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , DNA-Binding Proteins/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival , CpG Islands/genetics , DNA Methylation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , Neoplasm Staging , RNA, Small Interfering , Tumor Cells, Cultured
13.
Nucleic Acids Res ; 39(15): 6741-52, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21531701

ABSTRACT

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.


Subject(s)
Avian Proteins/chemistry , DNA Methylation , DNA-Binding Proteins/chemistry , DNA/chemistry , Animals , Base Sequence , Chickens , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Tertiary
14.
Proc Natl Acad Sci U S A ; 108(18): 7487-92, 2011 May 03.
Article in English | MEDLINE | ID: mdl-21490301

ABSTRACT

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.


Subject(s)
DNA-Binding Proteins/metabolism , Epigenesis, Genetic/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Models, Molecular , Repressor Proteins/metabolism , Blotting, Western , DNA Methylation/genetics , DNA Primers/genetics , Gene Silencing , Humans , Immunoprecipitation , RNA Interference
15.
Cancer ; 117(20): 4772-8, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21455994

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/economics , Black or African American/statistics & numerical data , Health Care Costs , Health Services Accessibility/economics , Insurance, Health , Leukemia, Myeloid, Acute , Medically Uninsured/statistics & numerical data , White People/statistics & numerical data , Adult , Antineoplastic Agents/therapeutic use , Bone Marrow Transplantation/economics , Confounding Factors, Epidemiologic , Ethnicity/statistics & numerical data , Female , Health Services Accessibility/statistics & numerical data , Hospital Costs , Humans , Inpatients , Length of Stay/economics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/economics , Leukemia, Myeloid, Acute/mortality , Logistic Models , Male , Medicaid , Medicare , Middle Aged , Odds Ratio , Proportional Hazards Models , Registries , Retreatment/economics , United States , Virginia/epidemiology
16.
Blood Cells Mol Dis ; 46(3): 212-9, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21296012

ABSTRACT

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.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Gene Silencing , epsilon-Globins/genetics , Animals , Azacitidine/pharmacology , DNA Methylation , Erythroblasts/metabolism , Erythrocytes/metabolism , Female , Gene Order , Hemoglobins, Abnormal/metabolism , Humans , Locus Control Region/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , beta-Globins/metabolism , gamma-Globins/metabolism
17.
Blood ; 117(1): 4-5, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-21212288
18.
Curr Top Dev Biol ; 82: 85-116, 2008.
Article in English | MEDLINE | ID: mdl-18282518

ABSTRACT

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.


Subject(s)
DNA Methylation , Epigenesis, Genetic/genetics , Erythroid Cells/cytology , Erythroid Cells/metabolism , Erythropoiesis/genetics , Gene Expression Regulation, Developmental , Animals , Globins/genetics , Humans
19.
J Immunol ; 177(12): 8612-9, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17142760

ABSTRACT

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.


Subject(s)
Active Transport, Cell Nucleus , HLA-A Antigens/genetics , Interferon-gamma/pharmacology , Karyopherins/physiology , RNA/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Cell Line , Gene Expression Regulation/drug effects , HLA Antigens/genetics , Histocompatibility Antigens Class I/genetics , Humans , Methylation , Exportin 1 Protein , HLA-E Antigens
20.
Blood ; 108(8): 2836-45, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-16778143

ABSTRACT

The chicken embryonic beta-type globin gene, rho, is a member of a small group of vertebrate genes whose developmentally regulated expression is mediated by DNA methylation. Previously, we have shown that a methyl cytosine-binding complex binds to the methylated rho-globin gene in vitro. We have now chromatographically purified and characterized this complex from adult chicken primary erythroid cells. Four components of the MeCP1 transcriptional repression complex were identified: MBD2, RBAP48, HDAC2, and MTA1. These 4 proteins, as well as the zinc-finger protein p66 and the chromatin remodeling factor Mi2, were found to coelute by gel-filtration analysis and pull-down assays. We conclude that these 6 proteins are components of the MeCPC. In adult erythrocytes, significant enrichment for MBD2 is seen at the inactive rho-globin gene by chromatin immunoprecipitation assay, whereas no enrichment is observed at the active beta(A)-globin gene, demonstrating MBD2 binds to the methylated and transcriptionally silent rho-globin gene in vivo. Knock-down of MBD2 resulted in up-regulation of a methylated rho-gene construct in mouse erythroleukemic (MEL)-rho cells. These results represent the first purification of a MeCP1-like complex from a primary cell source and provide support for a role for MBD2 in developmental gene regulation.


Subject(s)
DNA-Binding Proteins/metabolism , Erythroid Cells/metabolism , Animals , Base Sequence , Binding Sites/genetics , Cell Line, Tumor , Chick Embryo , Chickens , CpG Islands , DNA/genetics , DNA/metabolism , DNA Methylation , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Globins/genetics , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/metabolism , Mice , Models, Biological , Molecular Sequence Data , Multiprotein Complexes , RNA Interference , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL