Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 294(4): 1363-1379, 2019 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-30523150

RESUMO

The addition of a single ß-d-GlcNAc sugar (O-GlcNAc) by O-GlcNAc-transferase (OGT) and O-GlcNAc removal by O-GlcNAcase (OGA) maintain homeostatic O-GlcNAc levels on cellular proteins. Changes in protein O-GlcNAcylation regulate cellular differentiation and cell fate decisions, but how these changes affect erythropoiesis, an essential process in blood cell formation, remains unclear. Here, we investigated the role of O-GlcNAcylation in erythropoiesis by using G1E-ER4 cells, which carry the erythroid-specific transcription factor GATA-binding protein 1 (GATA-1) fused to the estrogen receptor (GATA-1-ER) and therefore undergo erythropoiesis after ß-estradiol (E2) addition. We observed that during G1E-ER4 differentiation, overall O-GlcNAc levels decrease, and physical interactions of GATA-1 with both OGT and OGA increase. RNA-Seq-based transcriptome analysis of G1E-ER4 cells differentiated in the presence of the OGA inhibitor Thiamet-G (TMG) revealed changes in expression of 433 GATA-1 target genes. ChIP results indicated that the TMG treatment decreases the occupancy of GATA-1, OGT, and OGA at the GATA-binding site of the lysosomal protein transmembrane 5 (Laptm5) gene promoter. TMG also reduced the expression of genes involved in differentiation of NB4 and HL60 human myeloid leukemia cells, suggesting that O-GlcNAcylation is involved in the regulation of hematopoietic differentiation. Sustained treatment of G1E-ER4 cells with TMG before differentiation reduced hemoglobin-positive cells and increased stem/progenitor cell surface markers. Our results show that alterations in O-GlcNAcylation disrupt transcriptional programs controlling erythropoietic lineage commitment, suggesting a role for O-GlcNAcylation in regulating hematopoietic cell fate.


Assuntos
Acetilglucosamina/metabolismo , Diferenciação Celular , Células Eritroides/citologia , Hematopoese , Homeostase , Células Mieloides/citologia , N-Acetilglucosaminiltransferases/metabolismo , Células Cultivadas , Células Eritroides/metabolismo , Fator de Transcrição GATA1/metabolismo , Humanos , Células Mieloides/fisiologia
2.
Hum Mol Genet ; 27(19): 3313-3324, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29931260

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the growth of renal cysts that ultimately destroy kidney function. Mutations in the PKD1 and PKD2 genes cause ADPKD. Their protein products, polycystin-1 (PC1) and polycystin-2 (PC2) have been proposed to form a calcium-permeable receptor-channel complex; however the mechanisms by which they function are almost completely unknown. Most mutations in PKD1 are truncating loss-of-function mutations or affect protein biogenesis, trafficking or stability and reveal very little about the intrinsic biochemical properties or cellular functions of PC1. An ADPKD patient mutation (L4132Δ or ΔL), resulting in a single amino acid deletion in a putative G-protein binding region of the PC1 C-terminal cytosolic tail, was found to significantly decrease PC1-stimulated, G-protein-dependent signaling in transient transfection assays. Pkd1ΔL/ΔL mice were embryo-lethal suggesting that ΔL is a functionally null mutation. Kidney-specific Pkd1ΔL/cond mice were born but developed severe, postnatal cystic disease. PC1ΔL protein expression levels and maturation were comparable to those of wild type PC1, and PC1ΔL protein showed cell surface localization. Expression of PC1ΔL and PC2 complexes in transfected CHO cells failed to support PC2 channel activity, suggesting that the role of PC1 is to activate G-protein signaling to regulate the PC1/PC2 calcium channel.


Assuntos
Proteínas Heterotriméricas de Ligação ao GTP/genética , Rim Policístico Autossômico Dominante/genética , Canais de Cátion TRPP/genética , Animais , Células CHO , Canais de Cálcio/genética , Cílios/genética , Cílios/patologia , Cricetulus , Humanos , Rim/patologia , Camundongos , Mutação , Rim Policístico Autossômico Dominante/patologia , Domínios Proteicos/genética , Transdução de Sinais
3.
Blood Cells Mol Dis ; 79: 102345, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31351219

RESUMO

Pharmacologic induction of fetal hemoglobin (HbF) is an effective strategy for treating sickle cell disease (SCD) by ameliorating disease severity. Hydroxyurea is the only FDA-approved agent that induces HbF, but significant non-responders and requirement for frequent monitoring of blood counts for drug toxicity limit clinical usefulness. Therefore, we investigated a novel prodrug conjugate of butyric acid (BA) and δ-aminolevulinate (ALA) as a potential HbF inducing agent, using erythroid precursors and a preclinical ß-YAC mouse model. We observed significantly increased γ-globin gene transcription and HbF expression mediated by AN-233 in K562 cells. Moreover, AN-233 stimulated mild heme biosynthesis and inhibited expression of heme-regulated eIF2α kinase involved in silencing γ-globin expression. Studies using primary erythroid precursors generated from sickle peripheral blood mononuclear cells verified the ability of AN-233 to induce HbF, increase histone H3 and H4 acetylation levels at the γ-globin promoter and reduce erythroid precursor sickling by 50%. Subsequent drug treatment of ß-YAC transgenic mice confirmed HbF induction in vivo by AN-233 through an increase in the percentage of HbF positive red blood cells and HbF levels measured by flow cytometry. These data support the potential development of AN-233 for the treatment of SCD.


Assuntos
Anemia Falciforme/terapia , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/efeitos dos fármacos , Ácidos Levulínicos/farmacologia , Pró-Fármacos/farmacologia , Animais , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Humanos , Células K562 , Ácidos Levulínicos/uso terapêutico , Camundongos , Camundongos Transgênicos , Ativação Transcricional , gama-Globinas/genética
4.
J Biol Chem ; 292(36): 14940-14962, 2017 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-28739801

RESUMO

Dysfunctional mitochondria and generation of reactive oxygen species (ROS) promote chronic diseases, which have spurred interest in the molecular mechanisms underlying these conditions. Previously, we have demonstrated that disruption of post-translational modification of proteins with ß-linked N-acetylglucosamine (O-GlcNAcylation) via overexpression of the O-GlcNAc-regulating enzymes O-GlcNAc transferase (OGT) or O-GlcNAcase (OGA) impairs mitochondrial function. Here, we report that sustained alterations in O-GlcNAcylation either by pharmacological or genetic manipulation also alter metabolic function. Sustained O-GlcNAc elevation in SH-SY5Y neuroblastoma cells increased OGA expression and reduced cellular respiration and ROS generation. Cells with elevated O-GlcNAc levels had elongated mitochondria and increased mitochondrial membrane potential, and RNA-sequencing analysis indicated transcriptome reprogramming and down-regulation of the NRF2-mediated antioxidant response. Sustained O-GlcNAcylation in mouse brain and liver validated the metabolic phenotypes observed in the cells, and OGT knockdown in the liver elevated ROS levels, impaired respiration, and increased the NRF2 antioxidant response. Moreover, elevated O-GlcNAc levels promoted weight loss and lowered respiration in mice and skewed the mice toward carbohydrate-dependent metabolism as determined by indirect calorimetry. In summary, sustained elevation in O-GlcNAcylation coupled with increased OGA expression reprograms energy metabolism, a finding that has potential implications for the etiology, development, and management of metabolic diseases.


Assuntos
Acetilglucosamina/metabolismo , Metabolismo Energético , Mitocôndrias/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo , Animais , Glicosilação , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , N-Acetilglucosaminiltransferases/deficiência , N-Acetilglucosaminiltransferases/genética , Células Tumorais Cultivadas , beta-N-Acetil-Hexosaminidases/genética
5.
J Biol Chem ; 291(30): 15628-40, 2016 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-27231347

RESUMO

One mode of γ-globin gene silencing involves a GATA-1·FOG-1·Mi2ß repressor complex that binds to the -566 GATA site relative to the (A)γ-globin gene cap site. However, the mechanism of how this repressor complex is assembled at the -566 GATA site is unknown. In this study, we demonstrate that the O-linked N-acetylglucosamine (O-GlcNAc) processing enzymes, O-GlcNAc-transferase (OGT) and O-GlcNAcase (OGA), interact with the (A)γ-globin promoter at the -566 GATA repressor site; however, mutation of the GATA site to GAGA significantly reduces OGT and OGA promoter interactions in ß-globin locus yeast artificial chromosome (ß-YAC) bone marrow cells. When WT ß-YAC bone marrow cells are treated with the OGA inhibitor Thiamet-G, the occupancy of OGT, OGA, and Mi2ß at the (A)γ-globin promoter is increased. In addition, OGT and Mi2ß recruitment is increased at the (A)γ-globin promoter when γ-globin becomes repressed in postconception day E18 human ß-YAC transgenic mouse fetal liver. Furthermore, we show that Mi2ß is modified with O-GlcNAc, and both OGT and OGA interact with Mi2ß, GATA-1, and FOG-1. Taken together, our data suggest that O-GlcNAcylation is a novel mechanism of γ-globin gene regulation mediated by modulating the assembly of the GATA-1·FOG-1·Mi2ß repressor complex at the -566 GATA motif within the promoter.


Assuntos
Inativação Gênica/fisiologia , N-Acetilglucosaminiltransferases/metabolismo , Elementos de Resposta , beta-N-Acetil-Hexosaminidases/metabolismo , gama-Globinas/biossíntese , Animais , Fator de Transcrição GATA1/genética , Fator de Transcrição GATA1/metabolismo , Humanos , Células K562 , Camundongos , Camundongos Transgênicos , N-Acetilglucosaminiltransferases/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , beta-N-Acetil-Hexosaminidases/genética , gama-Globinas/genética
6.
PLoS Genet ; 8(12): e1003155, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23284307

RESUMO

Activation of γ-globin gene expression in adults is known to be therapeutic for sickle cell disease. Thus, it follows that the converse, alleviation of repression, would be equally effective, since the net result would be the same: an increase in fetal hemoglobin. A GATA-1-FOG-1-Mi2 repressor complex was recently demonstrated to be recruited to the -566 GATA motif of the (A)γ-globin gene. We show that Mi2ß is essential for γ-globin gene silencing using Mi2ß conditional knockout ß-YAC transgenic mice. In addition, increased expression of (A)γ-globin was detected in adult blood from ß-YAC transgenic mice containing a T>G HPFH point mutation at the -566 GATA silencer site. ChIP experiments demonstrated that GATA-1 is recruited to this silencer at day E16, followed by recruitment of FOG-1 and Mi2 at day E17 in wild-type ß-YAC transgenic mice. Recruitment of the GATA-1-mediated repressor complex was disrupted by the -566 HPFH mutation at developmental stages when it normally binds. Our data suggest that a temporal repression mechanism is operative in the silencing of γ-globin gene expression and that either a trans-acting Mi2ß knockout deletion mutation or the cis-acting -566 (A)γ-globin HPFH point mutation disrupts establishment of repression, resulting in continued γ-globin gene transcription during adult definitive erythropoiesis.


Assuntos
Eritropoese , Fator de Transcrição GATA1 , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase , Proteínas Nucleares , Fatores de Transcrição , gama-Globinas , Anemia Falciforme/genética , Anemia Falciforme/metabolismo , Animais , Cromossomos Artificiais de Levedura/genética , Cromossomos Artificiais de Levedura/metabolismo , Desenvolvimento Embrionário , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Fator de Transcrição GATA1/genética , Fator de Transcrição GATA1/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Camundongos , Camundongos Transgênicos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Globinas beta/genética , Globinas beta/metabolismo , gama-Globinas/genética , gama-Globinas/metabolismo
7.
Nucleic Acids Res ; 40(22): 11256-69, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23042246

RESUMO

The DNaseI hypersensitive sites (HSs) of the human ß-globin locus control region (LCR) may function as part of an LCR holocomplex within a larger active chromatin hub (ACH). Differential activation of the globin genes during development may be controlled in part by preferential interaction of each gene with specific individual HSs during globin gene switching, a change in conformation of the LCR holocomplex, or both. To distinguish between these possibilities, human ß-globin locus yeast artificial chromosome (ß-YAC) lines were produced in which the ε-globin gene was replaced with a second marked ß-globin gene (ß(m)), coupled to an intact LCR, a 5'HS3 complete deletion (5'ΔHS3) or a 5'HS3 core deletion (5'ΔHS3c). The 5'ΔHS3c mice expressed ß(m)-globin throughout development; γ-globin was co-expressed in the embryonic yolk sac, but not in the fetal liver; and wild-type ß-globin was co-expressed in adult mice. Although the 5'HS3 core was not required for ß(m)-globin expression, previous work showed that the 5'HS3 core is necessary for ε-globin expression during embryonic erythropoiesis. A similar phenotype was observed in 5'HS complete deletion mice, except ß(m)-globin expression was higher during primitive erythropoiesis and γ-globin expression continued into fetal definitive erythropoiesis. These data support a site specificity model of LCR HS-globin gene interaction.


Assuntos
Cromatina , Regulação da Expressão Gênica no Desenvolvimento , Região de Controle de Locus Gênico , Ativação Transcricional , Globinas beta/genética , Animais , Eritropoese/genética , Humanos , Camundongos , Camundongos Transgênicos , Globinas beta/metabolismo
8.
Proc Natl Acad Sci U S A ; 107(39): 16846-51, 2010 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-20837550

RESUMO

Pharmacological treatments designed to reactivate fetal γ-globin can lead to an effective and successful clinical outcome in patients with hemoglobinopathies. However, new approaches remain highly desired because such treatments are not equally effective for all patients, and toxicity issues remain. We have taken a systematic approach to develop an embedded chimeric peptide nucleic acid (PNA) that effectively enters the cell and the nucleus, binds to its target site at the human fetal γ-globin promoter, and reactivates this transcript in adult transgenic mouse bone marrow and human primary peripheral blood cells. In vitro and in vivo DNA-binding assays in conjunction with live-cell imaging have been used to establish and optimize chimeric PNA design parameters that lead to successful gene activation. Our final molecule contains a specific γ-promoter-binding PNA sequence embedded within two amino acid motifs: one leads to efficient cell/nuclear entry, and the other generates transcriptional reactivation of the target. These embedded PNAs overcome previous limitations and are generally applicable to the design of in vivo transcriptional activation reagents that can be directed to any promoter region of interest and are of direct relevance to clinical applications that would benefit from such a need.


Assuntos
Desenho de Fármacos , Expressão Gênica/efeitos dos fármacos , Hemoglobinopatias/tratamento farmacológico , Ácidos Nucleicos Peptídicos/farmacologia , gama-Globinas/genética , Animais , Células da Medula Óssea/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Células Cultivadas , Imunoprecipitação da Cromatina , Hemoglobinopatias/genética , Humanos , Camundongos , Ácidos Nucleicos Peptídicos/química , Ácidos Nucleicos Peptídicos/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos
9.
J Alzheimers Dis ; 92(2): 591-604, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36776072

RESUMO

BACKGROUND: Mitochondria can trigger Alzheimer's disease (AD)-associated molecular phenomena, but how mitochondria impact apolipoprotein E (APOE; apoE) is not well known. OBJECTIVE: Consider whether and how mitochondrial biology influences APOE and apoE biology. METHODS: We measured APOE expression in human SH-SY5Y neuronal cells with different forms of mitochondrial dysfunction including total, chronic mitochondrial DNA (mtDNA) depletion (ρ0 cells); acute, partial mtDNA depletion; and toxin-induced mitochondrial dysfunction. We further assessed intracellular and secreted apoE protein levels in the ρ0 cells and interrogated the impact of transcription factors and stress signaling pathways known to influence APOE expression. RESULTS: SH-SY5Y ρ0 cells exhibited a 65-fold increase in APOE mRNA, an 8-fold increase in secreted apoE protein, and increased intracellular apoE protein. Other models of primary mitochondrial dysfunction including partial mtDNA-depletion, toxin-induced respiratory chain inhibition, and chemical-induced manipulations of the mitochondrial membrane potential similarly increased SH-SY5Y cell APOE mRNA. We explored potential mediators and found in the ρ0 cells knock-down of the C/EBPα and NFE2L2 (Nrf2) transcription factors reduced APOE mRNA. The activity of two mitogen-activated protein kinases, JNK and ERK, also strongly influenced ρ0 cell APOE mRNA levels. CONCLUSION: Primary mitochondrial dysfunction either directly or indirectly activates APOE expression in a neuronal cell model by altering transcription factors and stress signaling pathways. These studies demonstrate mitochondrial biology can influence the biology of the APOE gene and apoE protein, which are implicated in AD.


Assuntos
Doença de Alzheimer , Neuroblastoma , Humanos , Neuroblastoma/metabolismo , Mitocôndrias/metabolismo , DNA Mitocondrial/genética , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Fatores de Transcrição/metabolismo , Doença de Alzheimer/metabolismo , RNA Mensageiro/metabolismo , Biologia , Linhagem Celular Tumoral
10.
Blood ; 114(11): 2315-22, 2009 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-19525479

RESUMO

When successful, human leukocyte antigen (HLA)-matched bone marrow transplantation with reduced-intensity conditioning is a cure for several nonmalignant hematologic disorders that require chronic transfusion, such as sickle cell disease and aplastic anemia. However, there are unusually high bone marrow transplant (BMT) rejection rates in these patients. Rejection correlates with the number of transfusions before bone marrow transplantation, and it has been hypothesized that preimmunization to antigens on transfused blood may prime BMT rejection. Using a novel mouse model of red blood cell (RBC) transfusion and major histocompatibility complex-matched bone marrow transplantation, we report that transfusion of RBC products induced BMT rejection across minor histocompatibility antigen (mHA) barriers. It has been proposed that contaminating leukocytes are responsible for transfusion-induced BMT rejection; however, filter leukoreduction did not prevent rejection in the current studies. Moreover, we generated a novel transgenic mouse with RBC-specific expression of a model mHA and demonstrated that transfusion of RBCs induced a CD8(+) T-cell response. Together, these data suggest that mHAs on RBCs themselves are capable of inducing BMT rejection. Cellular immunization to mHAs is neither monitored nor managed by current transfusion medicine practice; however, the current data suggest that mHAs on RBCs may represent an unappreciated and significant consequence of RBC transfusion.


Assuntos
Transplante de Medula Óssea , Linfócitos T CD8-Positivos/imunologia , Transfusão de Eritrócitos , Rejeição de Enxerto/imunologia , Procedimentos de Redução de Leucócitos , Antígenos de Histocompatibilidade Menor/imunologia , Anemia Aplástica/imunologia , Anemia Aplástica/terapia , Anemia Falciforme/imunologia , Anemia Falciforme/terapia , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Transplante Homólogo
11.
Proc Natl Acad Sci U S A ; 105(36): 13514-9, 2008 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-18757759

RESUMO

Splice-site mutations in the beta-globin gene can lead to aberrant transcripts and decreased functional beta-globin, causing beta-thalassemia. Triplex-forming DNA oligonucleotides (TFOs) and peptide nucleic acids (PNAs) have been shown to stimulate recombination in reporter gene loci in mammalian cells via site-specific binding and creation of altered helical structures that provoke DNA repair. We have designed a series of triplex-forming PNAs that can specifically bind to sequences in the human beta-globin gene. We demonstrate here that these PNAs, when cotransfected with recombinatory donor DNA fragments, can promote single base-pair modification at the start of the second intron of the beta-globin gene, the site of a common thalassemia-associated mutation. This single base pair change was detected by the restoration of proper splicing of transcripts produced from a green fluorescent protein-beta-globin fusion gene. The ability of these PNAs to induce recombination was dependent on dose, sequence, cell-cycle stage, and the presence of a homologous donor DNA molecule. Enhanced recombination, with frequencies up to 0.4%, was observed with use of the lysomotropic agent chloroquine. Finally, we demonstrate that these PNAs were effective in stimulating the modification of the endogenous beta-globin locus in human cells, including primary hematopoietic progenitor cells. This work suggests that PNAs can be effective tools to induce heritable, site-specific modification of disease-related genes in human cells.


Assuntos
Globinas/genética , Ácidos Nucleicos Peptídicos/farmacologia , Sítios de Splice de RNA/genética , Animais , Sequência de Bases , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Genoma/genética , Humanos , Dados de Sequência Molecular , Mutação/genética
12.
Genomics ; 96(5): 303-7, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20691777

RESUMO

KLF1 regulates definitive erythropoiesis of red blood cells by facilitating transcription through high affinity binding to CACCC elements within its erythroid specific target genes including those encoding erythrocyte membrane skeleton (EMS) proteins. Deficiencies of EMS proteins in humans lead to the hemolytic anemia Hereditary Spherocytosis (HS) which includes a subpopulation with no known genetic defect. Here we report that a mutation, E339D, in the second zinc finger domain of KLF1 is responsible for HS in the mouse model Nan. The causative nature of this mutation was verified with an allelic test cross between Nan/+ and heterozygous Klf1(+/-) knockout mice. Homology modeling predicted Nan KLF1 binds CACCC elements more tightly, suggesting that Nan KLF1 is a competitive inhibitor of wild-type KLF1. This is the first association of a KLF1 mutation with a disease state in adult mammals and also presents the possibility of being another causative gene for HS in humans.


Assuntos
Anemia Hemolítica/patologia , Modelos Animais de Doenças , Fatores de Transcrição Kruppel-Like/genética , Mutação/genética , Esferocitose Hereditária/genética , Anemia Hemolítica/genética , Animais , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dedos de Zinco/genética
13.
Cancers (Basel) ; 13(7)2021 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-33916244

RESUMO

O-linked ß-N-acetylglucosamine (O-GlcNAc) is a single sugar post-translational modification (PTM) of intracellular proteins linking nutrient flux through the Hexosamine Biosynthetic Pathway (HBP) to the control of cis-regulatory elements in the genome. Aberrant O-GlcNAcylation is associated with the development, progression, and alterations in gene expression in cancer. O-GlcNAc cycling is defined as the addition and subsequent removal of the modification by O-GlcNAc Transferase (OGT) and O-GlcNAcase (OGA) provides a novel method for cells to regulate various aspects of gene expression, including RNA polymerase function, epigenetic dynamics, and transcription factor activity. We will focus on the complex relationship between phosphorylation and O-GlcNAcylation in the regulation of the RNA Polymerase II (RNAP II) pre-initiation complex and the regulation of the carboxyl-terminal domain of RNAP II via the synchronous actions of OGT, OGA, and kinases. Additionally, we discuss how O-GlcNAcylation of TATA-box binding protein (TBP) alters cellular metabolism. Next, in a non-exhaustive manner, we will discuss the current literature on how O-GlcNAcylation drives gene transcription in cancer through changes in transcription factor or chromatin remodeling complex functions. We conclude with a discussion of the challenges associated with studying O-GlcNAcylation and present several new approaches for studying O-GlcNAc regulated transcription that will advance our understanding of the role of O-GlcNAc in cancer.

14.
Exp Biol Med (Maywood) ; 245(15): 1308-1318, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32715783

RESUMO

IMPACT STATEMENT: Sickle cell disease is an inherited hemoglobin disorder that affects over 100,000 people in the United States causing high morbidity and early mortality. Although new treatments were recently approved by the FDA, only one drug Hydroxyurea induces fetal hemoglobin expression to inhibit sickle hemoglobin polymerization in red blood cells. Our laboratory previously demonstrated the ability of the NRF2 activator, dimethyl fumarate to induce fetal hemoglobin in the sickle cell mouse model. In this study, we investigated molecular mechanisms of γ-globin gene activation by NRF2. We observed the ability of NRF2 to modulate chromatin structure in the human ß-like globin gene locus of ß-YAC transgenic mice during development. Furthermore, an NRF2/TET3 interaction regulates γ-globin gene DNA methylation. These findings provide potential new molecular targets for small molecule drug developed for treating sickle cell disease.


Assuntos
Cromossomos Artificiais de Levedura/metabolismo , Epigênese Genética , Fator 2 Relacionado a NF-E2/metabolismo , gama-Globinas/genética , Animais , Cromatina/metabolismo , DNA/metabolismo , Metilação de DNA/genética , Dioxigenases/metabolismo , Células Eritroides/metabolismo , Eritropoese/genética , Feminino , Loci Gênicos , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Globinas beta/metabolismo
15.
Sci Rep ; 9(1): 19765, 2019 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-31874995

RESUMO

We report the development of episomal vectors for the specific γ-globin transcription activation in its native position by activator Zif-VP64, based on the Scaffold/Matrix Attachment Region (S/MAR) for episomal retention and the ß-globin Replicator, the DNA replication-Initiation Region from the ß-globin locus. Vector Zif-VP64-Ep1 containing transcription cassettes CMV- Zif-VP64 and CMV-eGFP-S/MAR transfected a)K562 cells; b)murine ß-YAC bone marrow cells (BMC); c)human haematopoietic progenitor CD34+ cells, with transfection efficiencies of 46.3 ± 5.2%, 23.0 ± 2.1% and 24.2 ± 2.4% respectively. K562 transfections generated stable cell lines running for 28 weeks with and without selection, with increased levels of γ-globin mRNA by 3.3 ± 0.13, of γ-globin protein by 6.75 ± 3.25 and HbF protein by 2 ± 0.2 fold, while the vector remained episomal and non integrated. In murine ß-YAC BMCs the vector mediated the activation of the silent human γ-globin gene and in CD34+ cells, increased γ-globin mRNA, albeit only transiently. A second vector Zif-VP64-Ep2, with both transcription cassettes carrying promoter SFFV instead of CMV and the addition of ß-globin Replicator, transferred into CD34+ cells, produced CD34+ eGFP+ cells, that generated colonies in colony forming cell cultures. Importantly, these were 100% fluorescent, with 2.11 ± 0.13 fold increased γ-globin mRNA, compared to non-transfected cells. We consider these episomal vectors valid, safer alternatives to viral vectors.


Assuntos
Vetores Genéticos , Células-Tronco Hematopoéticas/metabolismo , Regiões de Interação com a Matriz , Plasmídeos , Regiões Promotoras Genéticas , Globinas beta/biossíntese , Células-Tronco Hematopoéticas/citologia , Humanos , Células K562 , Globinas beta/genética
16.
Sci Rep ; 9(1): 9031, 2019 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-31227746

RESUMO

Development of drugs that allosterically regulate enzyme functions to treat disease is a costly venture. Amino acid susbstitutions that mimic allosteric effectors in vitro will identify therapeutic regulatory targets enhancing the likelihood of developing a disease treatment at a reasonable cost. We demonstrate the potential of this approach utilizing human liver pyruvate kinase (hLPYK) as a model. Inhibition of hLPYK was the first desired outcome of this study. We identified individual point mutations that: 1) mimicked allosteric inhibition by alanine, 2) mimicked inhibition by protein phosphorylation, and 3) prevented binding of fructose-1,6-bisphosphate (Fru-1,6-BP). Our second desired outcome was activation of hLPYK. We identified individual point mutations that: 1) prevented hLPYK from binding alanine, the allosteric inhibitor, 2) prevented inhibitory protein phosphorylation, or 3) mimicked allosteric activation by Fru-1,6-BP. Combining the three activating point mutations produced a constitutively activated enzyme that was unresponsive to regulators. Expression of a mutant hLPYK transgene containing these three mutations in a mouse model was not lethal. Thus, mutational mimics of allosteric effectors will be useful to confirm whether allosteric activation of hLPYK will control glycolytic flux in the diabetic liver to reduce hepatic glucose production and, in turn, reduce or prevent hyperglycemia.


Assuntos
Regulação Alostérica , Edição de Genes , Mutação , Regulação Alostérica/efeitos dos fármacos , Animais , Frutosefosfatos/metabolismo , Humanos , Fígado/enzimologia , Camundongos , Modelos Animais , Piruvato Quinase/genética , Piruvato Quinase/metabolismo
17.
PLoS One ; 14(8): e0217532, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31412036

RESUMO

Transcriptome analyses show a surprisingly large proportion of the mammalian genome is transcribed; much more than can be accounted for by genes and introns alone. Most of this transcription is non-coding in nature and arises from intergenic regions, often overlapping known protein-coding genes in sense or antisense orientation. The functional relevance of this widespread transcription is unknown. Here we characterize a promoter responsible for initiation of an intergenic transcript located approximately 3.3 kb and 10.7 kb upstream of the adult-specific human ß-globin genes. Mutational analyses in ß-YAC transgenic mice show that alteration of intergenic promoter activity results in ablation of H3K4 di- and tri-methylation and H3 hyperacetylation extending over a 30 kb region immediately downstream of the initiation site, containing the adult δ- and ß-globin genes. This results in dramatically decreased expression of the adult genes through position effect variegation in which the vast majority of definitive erythroid cells harbor inactive adult globin genes. In contrast, expression of the neighboring ε- and γ-globin genes is completely normal in embryonic erythroid cells, indicating a developmentally specific variegation of the adult domain. Our results demonstrate a role for intergenic non-coding RNA transcription in the propagation of histone modifications over chromatin domains and epigenetic control of ß-like globin gene transcription during development.


Assuntos
Cromatina/genética , DNA Intergênico/genética , Regulação da Expressão Gênica no Desenvolvimento , Histonas/química , Regiões Promotoras Genéticas , RNA não Traduzido/genética , Globinas beta/genética , Adulto , Animais , Cromossomos Artificiais de Levedura , Células Eritroides/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Transcrição Gênica
18.
Mol Cell Biol ; 25(20): 8765-78, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16199858

RESUMO

To test the role of gene order in globin gene expression, mutant human beta-globin locus yeast artificial chromosome constructs were used, each having one additional globin gene encoding a "marked" transcript (epsilon(m), gamma(m), or beta(m)) integrated at different locations within the locus. When a beta(m)-globin gene was placed between the locus control region (LCR) and the epsilon-globin gene, beta(m)-globin expression dominated primitive and definitive erythropoiesis; only beta(m)-globin mRNA was detected during the fetal and adult definitive stages of erythropoiesis. When an (A)gamma(m)-globin gene was placed at the same location, (A)gamma(m)-globin was expressed during embryonic erythropoiesis and the fetal liver stage of definitive erythropoiesis but was silenced during the adult stage. The downstream wild-type gamma-globin genes were not expressed. When an epsilon(m)-globin gene was placed between the delta- and beta-globin genes, it remained silent during embryonic erythropoiesis; only the LCR-proximal wild-type epsilon-globin gene was expressed. Placement of a beta(m)-globin gene upstream of the (G)gamma-globin gene resulted in expression of beta(m)-globin in embryonic cells and in a significant decrease in expression of the downstream wild-type beta-globin gene. These results indicate that distance from the LCR, an inherent property of spatial gene order, is a major determinant of temporal gene expression during development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Genoma Humano , Globinas/genética , Animais , Cromossomos Artificiais de Levedura/genética , Eritropoese/genética , Feminino , Feto/metabolismo , Ordem dos Genes , Inativação Gênica , Humanos , Região de Controle de Locus Gênico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Gravidez
19.
Mol Cell Biol ; 25(16): 7033-41, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16055715

RESUMO

Deletion of the 234-bp core element of the DNase I hypersensitive site 3 (5'HS3) of the locus control region (LCR) in the context of a human beta-globin locus yeast artificial chromosome (beta-YAC) results in profound effects on globin gene expression in transgenic mice. In contrast, deletion of a 2.3-kb 5'HS3 region, which includes the 234-bp core sequence, has a much milder phenotype. Here we report the effects of these deletions on chromatin structure in the beta-globin locus of adult erythroblasts. The 234-bp 5'HS3 deletion abolished histone acetylation throughout the beta-globin locus; recruitment of RNA polymerase II (pol II) to the LCR and beta-globin gene promoter was reduced to a basal level; and formation of all the 5' DNase I hypersensitive sites of the LCR was disrupted. The 2.3-kb 5'HS3 deletion mildly reduced the level of histone acetylation but did not change the profile across the whole locus; the 5' DNase I hypersensitive sites of the LCR were formed, but to a lesser extent; and recruitment of pol II was reduced, but only marginally. These data support the hypothesis that the LCR forms a specific chromatin structure and acts as a single entity. Based on these results we elaborate on a model of LCR chromatin architecture which accommodates the distinct phenotypes of the 5'HS3 and HS3 core deletions.


Assuntos
Deleção de Genes , Globinas/genética , Região de Controle de Locus Gênico , Acetilação , Animais , Cromatina/química , Cromatina/metabolismo , Imunoprecipitação da Cromatina , DNA/metabolismo , Primers do DNA/química , Proteínas de Ligação a DNA/metabolismo , Desoxirribonuclease I/metabolismo , Eritroblastos/metabolismo , Fatores de Ligação de DNA Eritroide Específicos , Histonas/química , Histonas/metabolismo , Camundongos , Camundongos Transgênicos , Mutação , Fenótipo , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , RNA Polimerase II/metabolismo , Fatores de Transcrição/metabolismo
20.
Methods Mol Biol ; 1698: 37-65, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29076083

RESUMO

Animal models of erythropoiesis have been, and will continue to be, important tools for understanding molecular mechanisms underlying the development of this cell lineage and the pathophysiology associated with various human erythropoietic diseases. In this regard, the mouse is probably the most valuable animal model available to investigators. The physiology and short gestational period of mice make them ideal for studying developmental processes and modeling human diseases. These attributes, coupled with cutting-edge genetic tools such as transgenesis, gene knockouts, conditional gene knockouts, and genome editing, provide a significant resource to the research community to test a plethora of hypotheses. This review summarizes the mouse models available for studying a wide variety of erythroid-related questions, as well as the properties inherent in each one.


Assuntos
Eritropoese , Hemoglobinopatias/genética , Animais , Cromossomos Artificiais Bacterianos , Cromossomos Artificiais de Levedura , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Hemoglobinopatias/metabolismo , Hemoglobinopatias/patologia , Hemoglobinas/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA