Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters











Publication year range
1.
J Leukoc Biol ; 2024 Sep 07.
Article in English | MEDLINE | ID: mdl-39243388

ABSTRACT

Dysfunctional phagocytic clearance of ß-amyloid (Aß) in microglia and peripheral macrophages/monocytes has been implicated in Alzheimer's disease (AD), but the mechanisms underlying this dysfunction are not yet well understood. In this study, we examined the role of glia maturation factor-γ (GMFG), an actin-disassembly protein that is highly expressed in immune cells, in macrophage Aß phagocytosis and in regulating scavenger receptor AI (SR-AI), a cell-surface receptor that has previously been implicated in Aß clearance. GMFG knockdown increased phagocytosis of Aß42 in BMDMs and RAW264.7 murine macrophages, while GMFG overexpression reduced Aß42 uptake in these cells. Blocking with anti-SR-AI antibodies inhibited Aß42 uptake in GMFG-knockdown cells, establishing a role for SR-AI in Aß42 phagocytosis. GMFG knockdown increased SR-AI protein expression under both basal conditions and in response to Aß42 treatment via both the transcriptional and post-transcriptional level in RAW264.7 macrophages. GMFG knockdown modulated Aß42-induced K48-linked and K63-polyubiquitination of SR-AI, the phosphorylation of SR-AI and JNK, suggesting that GMFG plays a role for intracellular signaling in the SR-AI-mediated uptake of Aß. Further, GMFG-knockdown cells displayed increased levels of the transcriptional factor MafB, and silencing of MafB in these cells reduced their SR-AI expression. Finally, GMFG was found to interact with the nuclear pore complex component RanBP2, and silencing of RanBP2 in GMFG-knockdown cells reduced their SR-AI expression. Collectively, these data support the role of GMFG as a novel regulator of SR-AI in macrophage Aß phagocytosis, and may provide insight into therapeutic approaches to potentially slow or prevent the progression of AD.

3.
Blood ; 140(21): 2276-2289, 2022 11 24.
Article in English | MEDLINE | ID: mdl-36399071

ABSTRACT

Sickle cell disease (SCD) and ß-thalassemia are among the most common genetic disorders worldwide, affecting global health and mortality. Hemoglobin A2 (HbA2, α2δ2) is expressed at a low level in adult blood due to the lack of the Kruppel-like factor 1 (KLF1) binding motif in the δ-globin promoter region. However, HbA2 is fully functional as an oxygen transporter, and could be a valid antisickling agent in SCD, as well as a substitute for hemoglobin A in ß-thalassemia. We have previously demonstrated that KLF1-GATA1 fusion protein could interact with the δ-globin promoter and increase δ-globin expression in human primary CD34+ cells. We report the effects of 2 KLF1-GATA1 fusion proteins on hemoglobin expression, as well as SCD phenotypic correction in vitro and in vivo. Forced expression of KLF1-GATA1 fusion protein enhanced δ-globin gene and HbA2 expression, as well as reduced hypoxia-related sickling, in erythroid cells cultured from both human sickle CD34+ cells and SCD mouse hematopoietic stem cells (HSCs). The fusion proteins had no impact on erythroid cell differentiation, proliferation, and enucleation. Transplantation of highly purified SCD mouse HSCs expressing KLF1-GATA1 fusion protein into SCD mice lessened the severity of the anemia, reduced the sickling of red blood cells, improved SCD-related pathological alterations in spleen, kidney, and liver, and restored urine-concentrating ability in recipient mice. Taken together, these results indicate that the use of KLF1-GATA1 fusion constructs may represent a new gene therapy approach for hemoglobinopathies.


Subject(s)
Anemia, Sickle Cell , Recombinant Fusion Proteins , beta-Thalassemia , delta-Globins , Animals , Humans , Mice , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Antigens, CD34/metabolism , beta-Thalassemia/genetics , delta-Globins/genetics , GATA1 Transcription Factor/genetics , Phenotype , Recombinant Fusion Proteins/therapeutic use
4.
Oncogene ; 41(1): 72-82, 2022 01.
Article in English | MEDLINE | ID: mdl-34675408

ABSTRACT

Small intestine adenocarcinoma is a rare intestinal malignancy with distinct clinical, pathological, and molecular characteristics. Recently, a fusion of the intestinal stem-cell marker olfactomedin 4 (OLFM4) and the proto-oncogene RET has been identified in a small intestine adenocarcinoma patient. Here we investigated the biological effects of OLFM4-RET fusion and whether it can initiate tumorigenesis in small intestine. OLFM4 expression was found to be frequently lost or reduced in human small intestine adenocarcinoma, and its downregulation correlated with high tumor grade and advanced tumor stage. Expression of OLFM4-RET fusion-induced cellular transformation in HEK293 cells and blocked RET-induced inhibition of colony growth in HuTu 80 small intestine adenocarcinoma cells. Further, expression of OLFM4-RET activated the RAS-RAF-MAPK and STAT3 cell signaling pathways in both HEK293 cells and HuTu 80 cells. OLFM4-RET expression in HEK293 cells upregulated multiple families of genes related to carcinogenesis, cancer progression, and metastasis. Targeted expression of OLFM4-RET in the small intestine led to the development of hyperplasia, adenoma, or adenocarcinoma in transgenic mice. Our study suggests that OLFM4-RET is an oncogenic driver of small intestine tumorigenesis. Therefore, the small intestine adenocarcinoma patients with OLFM4-RET fusion may benefit from treatment with RET kinase inhibitor.


Subject(s)
Adenocarcinoma/genetics , Intestinal Neoplasms/genetics , Oncogenes/genetics , Proto-Oncogene Proteins c-ret/metabolism , Animals , HEK293 Cells , Humans , Intestinal Neoplasms/pathology , Mice , Signal Transduction , Transfection
6.
Blood Adv ; 3(8): 1211-1225, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30971398

ABSTRACT

In macrophages, cellular iron metabolism status is tightly integrated with macrophage phenotype and associated with mitochondrial function. However, how molecular events regulate mitochondrial activity to integrate regulation of iron metabolism and macrophage phenotype remains unclear. Here, we explored the important role of the actin-regulatory protein glia maturation factor-γ (GMFG) in the regulation of cellular iron metabolism and macrophage phenotype. We found that GMFG was downregulated in murine macrophages by exposure to iron and hydrogen peroxide. GMFG knockdown altered the expression of iron metabolism proteins and increased iron levels in murine macrophages and concomitantly promoted their polarization toward an anti-inflammatory M2 phenotype. GMFG-knockdown macrophages exhibited moderately increased levels of mitochondrial reactive oxygen species (mtROS), which were accompanied by decreased expression of some mitochondrial respiration chain components, including the iron-sulfur cluster assembly scaffold protein ISCU as well as the antioxidant enzymes SOD1 and SOD2. Importantly, treatment of GMFG-knockdown macrophages with the antioxidant N-acetylcysteine reversed the altered expression of iron metabolism proteins and significantly inhibited the enhanced gene expression of M2 macrophage markers, suggesting that mtROS is mechanistically linked to cellular iron metabolism and macrophage phenotype. Finally, GMFG interacted with the mitochondrial membrane ATPase ATAD3A, suggesting that GMFG knockdown-induced mtROS production might be attributed to alteration of mitochondrial function in macrophages. Our findings suggest that GMFG is an important regulator in cellular iron metabolism and macrophage phenotype and could be a novel therapeutic target for modulating macrophage function in immune and metabolic disorders.


Subject(s)
Glia Maturation Factor/metabolism , Iron/metabolism , Macrophages/metabolism , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Animals , Gene Expression Regulation , Gene Knockdown Techniques , Glia Maturation Factor/genetics , Mice , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , RAW 264.7 Cells , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
7.
Endocrinology ; 159(9): 3235-3244, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30052841

ABSTRACT

Glucose-stimulated insulin secretion (GSIS) is essential for blood glucose homeostasis and is impaired in type 2 diabetes mellitus. Understanding the regulatory components of GSIS has clinical implications for diabetes treatment. In this study, we found that olfactomedin 4 (OLFM4) is endogenously expressed in pancreatic islet ß cells and further investigated its potential roles in glucose homeostasis and the pathogenesis of type 2 diabetes using mouse models. Olfm4-deficient mice showed significantly improved glucose tolerance and significantly increased insulin levels after glucose challenge compared with wild-type (WT) mice. GSIS, mitochondrial ATP production, and mitochondrial respiration were all significantly increased in islets isolated from Olfm4-deficient mice compared with those isolated from WT mice. In a high-fat diet (HFD)-induced diabetic mouse model, the increase in insulin levels after glucose challenge was significantly higher in Olfm4-deficient mice compared with WT mice. The impaired glucose tolerance and insulin resistance in HFD-fed mice were improved by loss of Olfm4. Olfm4 was found to be mainly localized in the mitochondria and interacts with GRIM-19 (a gene associated with retinoid-interferon mortality) in Min6 pancreatic ß cells. Collectively, these studies suggest that Olfm4 negatively regulates GSIS. OLFM4 may represent a potential therapeutic target for impaired glucose tolerance and patients with type 2 diabetes.


Subject(s)
Glucose Intolerance/genetics , Glycoproteins/genetics , Insulin Resistance/genetics , Mitochondria/metabolism , Adenosine Triphosphate/metabolism , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2 , Diet, High-Fat , Glucose Intolerance/metabolism , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells , Male , Mice , Mice, Knockout , NADH, NADPH Oxidoreductases/metabolism
8.
J Biol Chem ; 291(16): 8549-64, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26895964

ABSTRACT

Monocyte migration requires the dynamic redistribution of integrins through a regulated endo-exocytosis cycle, but the complex molecular mechanisms underlying this process have not been fully elucidated. Glia maturation factor-γ (GMFG), a novel regulator of the Arp2/3 complex, has been shown to regulate directional migration of neutrophils and T-lymphocytes. In this study, we explored the important role of GMFG in monocyte chemotaxis, adhesion, and ß1-integrin turnover. We found that knockdown of GMFG in monocytes resulted in impaired chemotactic migration toward formyl-Met-Leu-Phe (fMLP) and stromal cell-derived factor 1α (SDF-1α) as well as decreased α5ß1-integrin-mediated chemoattractant-stimulated adhesion. These GMFG knockdown impaired effects could be reversed by cotransfection of GFP-tagged full-length GMFG. GMFG knockdown cells reduced the cell surface and total protein levels of α5ß1-integrin and increased its degradation. Importantly, we demonstrate that GMFG mediates the ubiquitination of ß1-integrin through knockdown or overexpression of GMFG. Moreover, GMFG knockdown retarded the efficient recycling of ß1-integrin back to the plasma membrane following normal endocytosis of α5ß1-integrin, suggesting that the involvement of GMFG in maintaining α5ß1-integrin stability may occur in part by preventing ubiquitin-mediated degradation and promoting ß1-integrin recycling. Furthermore, we observed that GMFG interacted with syntaxin 4 (STX4) and syntaxin-binding protein 4 (STXBP4); however, only knockdown of STXBP4, but not STX4, reduced monocyte migration and decreased ß1-integrin cell surface expression. Knockdown of STXBP4 also substantially inhibited ß1-integrin recycling in human monocytes. These results indicate that the effects of GMFG on monocyte migration and adhesion probably occur through preventing ubiquitin-mediated proteasome degradation of α5ß1-integrin and facilitating effective ß1-integrin recycling back to the plasma membrane.


Subject(s)
Cell Movement/physiology , Glia Maturation Factor/metabolism , Integrin beta1/metabolism , Monocytes/metabolism , Cell Movement/drug effects , Chemokine CXCL12/pharmacology , Gene Knockdown Techniques , Glia Maturation Factor/genetics , Humans , Integrin alpha5beta1/genetics , Integrin alpha5beta1/metabolism , Integrin beta1/genetics , Monocytes/cytology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
9.
Blood ; 124(7): 1146-56, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-24914133

ABSTRACT

Hydroxyurea (HU) is effectively used in the management of ß-hemoglobinopathies by augmenting the production of fetal hemoglobin (HbF). However, the molecular mechanisms underlying HU-mediated HbF regulation remain unclear. We previously reported that overexpression of the HU-induced SAR1 gene closely mimics the known effects of HU on K562 and CD34(+) cells, including γ-globin induction and cell-cycle regulation. Here, we show that HU stimulated nuclear factor-κB interaction with its cognate-binding site on the SAR1 promoter to regulate transcriptional expression of SAR1 in K562 and CD34(+) cells. Silencing SAR1 expression not only significantly lowered both basal and HU-elicited HbF production in K562 and CD34(+) cells, but also significantly reduced HU-mediated S-phase cell-cycle arrest and apoptosis in K562 cells. Inhibition of c-Jun N-terminal kinase (JNK)/Jun phosphorylation and silencing of Giα expression in SAR1-transfected K562 and CD34(+) cells reduced both γ-globin expression and HbF level, indicating that activation of Giα/JNK/Jun proteins is required for SAR1-mediated HbF induction. Furthermore, reciprocal coimmunoprecipitation assays revealed an association between forcibly expressed SAR1 and Giα2 or Giα3 proteins in both K562 and nonerythroid cells. These results indicate that HU induces SAR1, which in turn activates γ-globin expression, predominantly through the Giα/JNK/Jun pathway. Our findings identify SAR1 as an alternative therapeutic target for ß-globin disorders.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Hydroxyurea/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Monomeric GTP-Binding Proteins/metabolism , Proto-Oncogene Proteins c-jun/metabolism , gamma-Globins/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Binding Sites/genetics , Blotting, Western , Cell Cycle Checkpoints/drug effects , Cells, Cultured , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Gene Expression/drug effects , Humans , K562 Cells , Monomeric GTP-Binding Proteins/genetics , NF-kappa B/metabolism , Promoter Regions, Genetic/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , ets-Domain Protein Elk-1/metabolism , gamma-Globins/genetics
10.
J Immunol ; 190(12): 6093-103, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23677465

ABSTRACT

TLR4 signaling must be tightly regulated to provide both effective immune protection and avoid inflammation-induced pathology. Thus, the mechanisms that negatively regulate the TLR4-triggered inflammatory response are of particular importance. Glia maturation factor-γ (GMFG), a novel actin depolymerization factor/cofilin superfamily protein that is expressed in inflammatory cells, has been implicated in mediating neutrophil and T cell migration, but its function in macrophage immune response remains unclear. In the current study, the role of GMFG in the LPS-induced TLR4-signaling pathway was investigated in THP-1 macrophages and human primary macrophages. LPS stimulation of macrophages decreased GMFG mRNA and protein expression. We show that GMFG negatively regulates LPS-induced activation of NF-κB-, MAPK-, and IRF3-signaling pathways and subsequent production of proinflammatory cytokines and type I IFN in human macrophages. We found that endogenous GMFG localized within early and late endosomes. GMFG knockdown delayed LPS-induced TLR4 internalization and caused prolonged TLR4 retention at the early endosome, suggesting that TLR4 transport from early to late endosomes is interrupted, which may contribute to enhanced LPS-induced TLR4 signaling. Taken together, our findings suggest that GMFG functions as a negative regulator of TLR4 signaling by facilitating TLR4 endocytic trafficking in macrophages.


Subject(s)
Glia Maturation Factor/metabolism , Macrophages/metabolism , Signal Transduction/physiology , Toll-Like Receptor 4/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Glia Maturation Factor/immunology , Humans , Immunoblotting , Macrophages/immunology , Microscopy, Confocal , Polymerase Chain Reaction , Protein Transport/physiology , RNA Interference , Toll-Like Receptor 4/immunology
11.
Blood ; 121(16): 3216-27, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23327923

ABSTRACT

Human erythropoiesis is a dynamic and complex multistep process involving differentiation of early erythroid progenitors into enucleated RBCs. The mechanisms underlying erythropoiesis still remain incompletely understood. We previously demonstrated that erythropoietin-stimulated clone-1, which is selectively expressed in normal human erythroid-lineage cells, shares 99.5% identity with malignant fibrous histiocytoma-amplified sequences with leucine-rich tandem repeats 1 (MASL1). In this study, we hypothesized that the MASL1 gene plays a role in erythroid differentiation, and used a human erythroid cell culture system to explore this concept. MASL1 mRNA and protein expression levels were significantly increased during the erythroid differentiation of CD34(+) cells following erythropoietin (EPO) treatment. Conversely, MASL1 knockdown reduced erythroid differentiation in EPO-treated CD34(+) cells. In addition, MASL1 knockdown interrupted the Raf/MEK/ERK signaling pathway in CD34(+) cells. MASL1 mutant-transfected CD34(+) cells also showed decreased erythroid differentiation. Furthermore, inhibition of the SH3 domain of Son of Sevenless, which is an upstream adapter protein in EPO-induced erythroid differentiation, also reduced MASL1 expression and phosphorylation of Raf/MEK/ERK kinases that consequently reduced erythroid differentiation of EPO-induced CD34(+) cells. Importantly, we also demonstrated that MASL1 interacts physically with Raf1. Taken together, our data provide novel insights into MASL1 regulation of erythropoiesis through the Raf/MEK/ERK pathway.


Subject(s)
Antigens, CD34/metabolism , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Erythroid Cells/cytology , Erythropoiesis , Erythropoietin/metabolism , MAP Kinase Signaling System , Oncogene Proteins/metabolism , raf Kinases/metabolism , Cell Cycle Checkpoints , Cell Cycle Proteins/genetics , Cell Proliferation , Cells, Cultured , DNA-Binding Proteins/genetics , Erythrocytes/cytology , Erythrocytes/metabolism , Erythroid Cells/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Gene Knockdown Techniques , Granulocytes/cytology , Granulocytes/metabolism , Humans , Oncogene Proteins/genetics , RNA, Messenger/genetics , Son of Sevenless Proteins/metabolism , Up-Regulation
12.
Mol Biol Cell ; 23(24): 4751-65, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23097489

ABSTRACT

Integrin activation is required to facilitate multiple adhesion-dependent functions of neutrophils, such as chemotaxis, which is critical for inflammatory responses to injury and pathogens. However, little is known about the mechanisms that mediate integrin activation in neutrophils. We show that Radil, a novel Rap1 effector, regulates ß1- and ß2-integrin activation and controls neutrophil chemotaxis. On activation and chemotactic migration of neutrophils, Radil quickly translocates from the cytoplasm to the plasma membrane in a Rap1a-GTP-dependent manner. Cells overexpressing Radil show a substantial increase in cell adhesion, as well as in integrin/focal adhesion kinase (FAK) activation, and exhibit an elongated morphology, with severe tail retraction defects. This phenotype is effectively rescued by treatment with either ß2-integrin inhibitory antibodies or FAK inhibitors. Conversely, knockdown of Radil causes severe inhibition of cell adhesion, ß2-integrin activation, and chemotaxis. Furthermore, we found that inhibition of Rap activity by RapGAP coexpression inhibits Radil-mediated integrin and FAK activation, decreases cell adhesion, and abrogates the long-tail phenotype of Radil cells. Overall, these studies establish that Radil regulates neutrophil adhesion and motility by linking Rap1 to ß2-integrin activation.


Subject(s)
CD18 Antigens/metabolism , Carrier Proteins/metabolism , Cell Movement , Neutrophils/metabolism , Blotting, Western , Carrier Proteins/genetics , Cell Adhesion/drug effects , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Cells, Cultured , Chemotaxis, Leukocyte/drug effects , Flow Cytometry , HEK293 Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Mutation , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/cytology , Protein Transport/drug effects , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/metabolism
13.
J Leukoc Biol ; 90(3): 529-38, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21653232

ABSTRACT

Chemotaxis is fundamental to the directional migration of neutrophils toward endogenous and exogenous chemoattractants. Recent studies have demonstrated that ADF/cofilin superfamily members play important roles in reorganizing the actin cytoskeleton by disassembling actin filaments. GMFG, a novel ADF/cofilin superfamily protein that is expressed in inflammatory cells, has been implicated in regulating actin reorganization in microendothelial cells, but its function in neutrophils remains unclear. Here, we show that GMFG is an important regulator for cell migration and polarity in neutrophils. Knockdown of endogenous GMFG impaired fMLF- and IL-8 (CXCL8)-induced chemotaxis in dHL-60 cells. GMFG knockdown attenuated the formation of lamellipodia at the leading edge of cells exposed to fMLF or CXCL8, as well as the phosphorylation of p38 and PAK1/2 in response to fMLF or CXCL8. Live cell imaging revealed that GMFG was recruited to the leading edge of cells in response to fMLF, as well as CXCL8. Overexpression of GMFG enhanced phosphorylation of p38 but not of PAK1/2 in dHL-60 cells. In addition, we found that GMFG is associated with WAVE2. Taken together, our findings suggest that GMFG is a novel factor in regulating neutrophil chemotaxis by modulating actin cytoskeleton reorganization.


Subject(s)
Cell Movement , Chemotaxis , Glia Maturation Factor/metabolism , Neutrophils/metabolism , Pseudopodia/metabolism , Actins/metabolism , Adult , Blotting, Western , Cell Adhesion , Cell Polarity , Cell Proliferation , Cells, Cultured , Cytoskeleton/metabolism , Fluorescent Antibody Technique , Glia Maturation Factor/antagonists & inhibitors , Glia Maturation Factor/genetics , Humans , Immunoprecipitation , Interleukin-8/pharmacology , Neutrophils/immunology , Phosphorylation , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Blood ; 117(11): 3045-52, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21220744

ABSTRACT

The ß-hemoglobinopathies sickle cell disease and ß-thalassemia are among the most common human genetic disorders worldwide. Hemoglobin A2 (HbA2, α2δ2) and fetal hemoglobin (HbF, α2γ2) both inhibit the polymerization of hemoglobin S, which results in erythrocyte sickling. Expression of erythroid Kruppel-like factor (EKLF) and GATA1 is critical for transitioning hemoglobin from HbF to hemoglobin A (HbA, α2ß2) and HbA2. The lower levels of δ-globin expression compared with ß-globin expression seen in adulthood are likely due to the absence of an EKLF-binding motif in the δ-globin proximal promoter. In an effort to up-regulate δ-globin to increase HbA2 expression, we created a series of EKLF-GATA1 fusion constructs composed of the transactivation domain of EKLF and the DNA-binding domain of GATA1, and then tested their effects on hemoglobin expression. EKLF-GATA1 fusion proteins activated δ-, γ-, and ß-globin promoters in K562 cells, and significantly up-regulated δ- and γ-globin RNA transcript and protein expression in K562 and/or CD34(+) cells. The binding of EKLF-GATA1 fusion proteins at the GATA1 consensus site in the δ-globin promoter was confirmed by chromatin immunoprecipitation assay. Our studies demonstrate that EKLF-GATA1 fusion proteins can enhance δ-globin expression through interaction with the δ-globin promoter, and may represent a new genetic therapeutic approach to ß-hemoglobinopathies.


Subject(s)
Erythroid Cells/metabolism , GATA1 Transcription Factor/metabolism , Kruppel-Like Transcription Factors/metabolism , Recombinant Fusion Proteins/metabolism , Up-Regulation/genetics , delta-Globins/genetics , gamma-Globins/genetics , Antigens, CD34/metabolism , Base Sequence , Genes, Reporter , Humans , K562 Cells , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Protein Binding
15.
Blood ; 114(1): 187-94, 2009 Jul 02.
Article in English | MEDLINE | ID: mdl-19401563

ABSTRACT

Increased fetal hemoglobin expression in adulthood is associated with acute stress erythropoiesis. However, the mechanisms underlying gamma-globin induction during the rapid expansion of adult erythroid progenitor cells have not been fully elucidated. Here, we examined COUP-TFII as a potential repressor of gamma-globin gene after stem cell factor (SCF) stimulation in cultured human adult erythroid progenitor cells. We found that COUP-TFII expression is suppressed by SCF through phosphorylation of serine/threonine phosphatase (PP2A) and correlated well with fetal hemoglobin induction. Furthermore, down-regulation of COUP-TFII expression with small interfering RNA (siRNA) significantly increases the gamma-globin expression during the erythroid maturation. Moreover, SCF-increased expression of NF-YA associated with redox regulator Ref-1 and cellular reducing condition enhances the effect of SCF on gamma-globin expression. Activation of Erk1/2 plays a critical role in SCF modulation of downstream transcriptional factor COUP-TFII, which is involved in the regulation of gamma-globin gene induction. Our data show that SCF stimulates Erk1/2 MAPK signaling pathway, which regulates the downstream repressor COUP-TFII by inhibiting serine/threonine phosphatase 2A activity, and that decreased COUP-TFII expression resulted in gamma-globin reactivation in adult erythropoiesis. These observations provide insight into the molecular pathways that regulate gamma-globin augmentation during stress erythropoiesis.


Subject(s)
COUP Transcription Factor II/metabolism , Stem Cell Factor/pharmacology , gamma-Globins/genetics , Adult , Base Sequence , Binding Sites , CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/metabolism , Cells, Cultured , DNA Primers/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Erythropoiesis/drug effects , Erythropoiesis/genetics , Erythropoiesis/physiology , Humans , MAP Kinase Signaling System/drug effects , Promoter Regions, Genetic , Protein Phosphatase 2/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Stress, Physiological , Transcriptional Activation/drug effects
16.
Br J Haematol ; 143(3): 421-32, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18764868

ABSTRACT

The human olfactomedin 4 gene (OLFM4, also known as hGC-1, GW112) is thought to be a useful marker for early myeloid development. To understand the molecular mechanisms underlying granulocyte colony-stimulating factor (G-CSF)-stimulated OLFM4 expression, we characterized the promoter region of OLFM4. The 35-bp region (-101 to -66) of the proximal promoter regulated reporter gene expression, and mutation of the nuclear factor (NF)-kappaB binding site within the promoter abolished the binding of the transcription factor and the ability to regulate OLFM4 expression. G-CSF increased reactive oxygen species (ROS) production in human CD34(+) cells, which was abrogated by inhibition of phosphatidylinositol 3-kinase (PI3K) or NADPH oxidase. Phosphorylation of ERK1/2 mitogen-activated protein kinase (MAPK) induced by G-CSF inhibited by the antioxidant N-acetyl-L-cysteine (NAC), ERK1/2 inhibitor PD98059, or U0126. However, phosphorylation of signal transducer and activator of transcription (STAT)3 was only partially inhibited by NAC, but not by PD98059 or U0126. Inhibition of the ERK pathway remarkably decreased OLFM4 expression and this inhibition required NF-kappaB transcription factor. Inhibition of ROS or the ERK pathway remarkably decreased G-CSF-induced OLFM4 expression. Our results suggest that G-CSF-induced expression of OLFM4 is regulated by the transcription factor NF-kappaB, and that this induction is mediated by the ERK1/2 MAPK signaling pathway through PI3K-driven ROS production.


Subject(s)
Gene Expression Regulation , Granulocyte Colony-Stimulating Factor/genetics , NF-kappa B/genetics , Transcription Factors/physiology , Base Sequence , Dose-Response Relationship, Drug , Electrophoretic Mobility Shift Assay/methods , Enzyme Activation/drug effects , Genes, Reporter , Granulocyte Colony-Stimulating Factor/biosynthesis , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Sequence Data , NF-kappa B/physiology , Promoter Regions, Genetic , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Signal Transduction , Transcription Factor AP-1/genetics
17.
Biochim Biophys Acta ; 1782(9): 504-10, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18586086

ABSTRACT

In vivo, inhibition of fetal hemoglobin (HbF) expression in humans around the time of birth causes the clinical manifestation of sickle cell and beta-thalassemia syndromes. Inhibition of HbF among cultured cells was recently described by the adenosine derivative molecule named SQ22536. Here, a primary cell culture model was utilized to further explore the inhibition of HbF by adenosine derivative molecules. SQ22536 demonstrated down-regulation of growth and HbF expression among erythroblasts cultured from fetal and adult human blood. The effects upon HbF were noted in a majority of cells, and quantitative PCR analysis demonstrated a transcriptional mechanism. Screening assays demonstrated that two additional molecules named 5'-deoxy adenosine and 2',3'-dideoxy adenosine had effects on HbF comparable to SQ22536. Other adenosine derivative molecules, adenosine receptor binding ligands, and cAMP-signaling regulators failed to inhibit HbF in matched cultures. These results suggest that structurally related ribofuranose-substituted adenosine analogues act through an unknown mechanism to inhibit HbF expression in fetal and adult human erythroblasts.


Subject(s)
Adenosine/pharmacology , Erythroblasts/cytology , Erythroblasts/drug effects , Fetal Hemoglobin/biosynthesis , Adenine/analogs & derivatives , Adenine/chemistry , Adenine/pharmacology , Adenosine/chemistry , Adult , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Colforsin/pharmacology , Cyclic AMP/metabolism , Deoxyadenosines/chemistry , Deoxyadenosines/pharmacology , Dideoxyadenosine/chemistry , Dideoxyadenosine/pharmacology , Erythropoietin/pharmacology , Gene Expression Regulation/drug effects , Gene Silencing/drug effects , Globins/genetics , Globins/metabolism , Humans , Kinetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Stem Cell Factor/pharmacology , Transforming Growth Factor beta/pharmacology
18.
Blood ; 110(8): 2864-71, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17620452

ABSTRACT

Although thalidomide has been shown to improve anemia in some patients with myelodysplastic syndromes and stimulates erythropoietin in patients with multiple myeloma, thalidomide's specific effects on gamma-globin gene expression during erythroid differentiation have not been studied. Here, we investigated the effects of thalidomide on gamma-globin gene expression and the involved signaling pathway using an ex vivo culture system of primary human CD34+ cells. We found that thalidomide induced gamma-globin mRNA expression in a dose-dependent manner, but had no effect on beta-globin expression. We also demonstrated that intracellular reactive oxygen species (ROS) levels were increased by treatment with thalidomide for 48 hours (from day 3 to day 5). Western blot analysis demonstrated that thalidomide activated the p38 mitogen-activated protein kinase (MAPK) signaling pathway in a time- and dose-dependent manner and increased histone H4 acetylation. Pretreatment of cells with the antioxidant enzyme catalase and the intracellular hydroxyl scavenger dimethylthiourea (DMTU) abrogated the thalidomide-induced p38 MAPK activation and histone H4 acetylation. Moreover, pretreatment with catalase and DMTU diminished thalidomide-induced gamma-globin gene expression. These data indicate that thalidomide induces increased expression of the gamma-globin gene via ROS-dependent activation of the p38 MAPK signaling pathway and histone H4 acetylation.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Erythropoiesis/drug effects , Gene Expression/drug effects , Signal Transduction/drug effects , Thalidomide/pharmacology , gamma-Globulins/drug effects , Acetylation , Antigens, CD34/metabolism , Antioxidants/pharmacology , Blotting, Western , Cells, Cultured , Dose-Response Relationship, Drug , Flow Cytometry , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Histones/drug effects , Histones/metabolism , Humans , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , gamma-Globulins/genetics , p38 Mitogen-Activated Protein Kinases/drug effects
19.
Br J Haematol ; 137(3): 233-6, 2007 May.
Article in English | MEDLINE | ID: mdl-17408462

ABSTRACT

Candidate drugs are being sought for the suppression of human erythropoiesis. Cl-IB-MECA [2-chloro-N6-(3-iodobenzyl)-adenosine-5'-N-methyluronamide] is a derivative of adenosine that inhibits the growth of leukaemic cell lines. To determine the effects of Cl-IB-MECA upon erythropoiesis, studies were performed by using an ex vivo culture system of primary human CD34+ cells. Cl-IB-MECA suppressed erythroblast growth and maturation at doses >/=50 mumol/l through a mechanism of cell cycle inhibition and accumulation of cells in the G1/G0 phase. These findings demonstrate that Cl-IB-MECA inhibits human erythropoiesis, and suggest that further consideration of this drug is warranted for patients with erythrocytosis or polycythemia syndromes.


Subject(s)
Adenosine/analogs & derivatives , Antineoplastic Agents/pharmacology , Erythropoiesis/drug effects , Adenosine/pharmacology , Antigens, CD34 , Apoptosis/drug effects , Cell Count/methods , Cell Cycle/drug effects , Cells, Cultured , Culture Media , Erythroblasts/drug effects , Erythropoietin , G1 Phase/drug effects , Humans , Polymerase Chain Reaction/methods , Receptor, Adenosine A3/analysis , Resting Phase, Cell Cycle/drug effects
20.
J Biol Chem ; 279(3): 1916-21, 2004 Jan 16.
Article in English | MEDLINE | ID: mdl-14593112

ABSTRACT

We report here the cloning and initial characterization of a novel growth-related gene (EEG-1) that is located on the short arm of chromosome 12. Two spliced transcripts were cloned from human bone marrow and human erythroid progenitor cells: EEG-1L containing a 4350-nucleotide open reading frame encoding a putative protein of 1077 amino acids including a C1q-like globular domain, and an alternatively spliced transcript lacking exon 5 (EEG-1S) encodes a significantly smaller coding region and no C1q-like domain. Quantitative PCR revealed expression of both EEG-1 transcripts in all analyzed tissues. Plasmids encoding green fluorescent protein-tagged genes (GFP-EEG-1) were transfected into Chinese hamster ovary cells for localization and functional assays. In contrast to the diffuse cellular localization of the GFP control, GFP-EEG-1L was detected throughout the cytoplasm and excluded from the nucleus, and GFP-EEG-1S co-localized with aggregated mitochondria. Transfection of both isoforms was associated with significantly increased levels of apoptosis. Stable transfection assays additionally demonstrated decreased growth in those cells expressing EEG-1 at higher levels. Quantitative PCR analyses of mRNA obtained from differentiating erythroid cells from blood donors were performed to determine the transcriptional pattern of EEG-1 during erythropoiesis. EEG-1 expression was highly regulated with increased expression at the stage of differentiation associated with the onset of global nuclear condensation and reduced cell proliferation. We propose that the regulated expression of EEG-1 is involved in the orchestrated regulation of growth that occurs as erythroblasts shift from a highly proliferative state toward their terminal phase of differentiation.


Subject(s)
Erythroblasts/metabolism , Growth Inhibitors/genetics , Animals , CHO Cells , Cell Differentiation , Cloning, Molecular , Cricetinae , DNA, Complementary/analysis , Erythroblasts/cytology , Growth Inhibitors/analysis , Growth Inhibitors/physiology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL