Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Wulin Aerbajinai is active.

Publication


Featured researches published by Wulin Aerbajinai.


British Journal of Haematology | 2008

The regulation of OLFM4 expression in myeloid precursor cells relies on NF-κB transcription factor

Kyung Chin; Wulin Aerbajinai; Jiangqiong Zhu; Lashawn Drew; Ling Chen; Wenli Liu; Griffin P. Rodgers

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)‐κB 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‐κB 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‐κB, and that this induction is mediated by the ERK1/2 MAPK signaling pathway through PI3K‐driven ROS production.


Journal of Biological Chemistry | 2004

Cloning and Characterization of a Gene Expressed during Terminal Differentiation That Encodes a Novel Inhibitor of Growth

Wulin Aerbajinai; Y. Terry Lee; Urszula Wojda; Valarie A. Barr; Jeffery L. Miller

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.


Journal of Leukocyte Biology | 2011

Glia maturation factor-γ mediates neutrophil chemotaxis

Wulin Aerbajinai; Lunhua Liu; Kyung Chin; Jianqiong Zhu; Carole A. Parent; Griffin P. Rodgers

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.


Blood | 2013

MASL1 induces erythroid differentiation in human erythropoietin-dependent CD34+ cells through the Raf/MEK/ERK pathway

Chutima Kumkhaek; Wulin Aerbajinai; Wenli Liu; Jianqiong Zhu; Naoya Uchida; Roger Kurlander; Matthew M. Hsieh; John F. Tisdale; Griffin P. Rodgers

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.


Blood | 2014

Hydroxyurea-inducible SAR1 gene acts through the Giα/JNK/Jun pathway to regulate γ-globin expression

Jianqiong Zhu; Kyung Chin; Wulin Aerbajinai; Chutima Kumkhaek; Hongzhen Li; Griffin P. Rodgers

Hydroxyurea (HU) is effectively used in the management of β-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 β-globin disorders.


Molecular Biology of the Cell | 2012

Radil controls neutrophil adhesion and motility through β2-integrin activation.

Lunhua Liu; Wulin Aerbajinai; Syed M. Ahmed; Griffin P. Rodgers; Stephane Angers; Carole A. Parent

Various agonists trigger β2-integrin activation in neutrophils, yet the mechanisms that regulate β2-integrin inside-out signaling remain obscure. Radil, a novel Rap downstream effector, is an important adapter in the pathway that links G protein–coupled chemoattractant receptors to adhesion complexes during neutrophil chemotaxis.


Blood | 2011

Recombinant erythroid Kruppel-like factor fused to GATA1 up-regulates delta- and gamma-globin expression in erythroid cells

Jianqiong Zhu; Kyung Chin; Wulin Aerbajinai; Cecelia D. Trainor; Peter Gao; Griffin P. Rodgers

The β-hemoglobinopathies sickle cell disease and β-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, α₂β₂) and HbA2. The lower levels of δ-globin expression compared with β-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 β-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 β-hemoglobinopathies.


Journal of Immunology | 2013

Glia maturation factor-γ negatively modulates TLR4 signaling by facilitating TLR4 endocytic trafficking in macrophages

Wulin Aerbajinai; Kevin Lee; Kyung Chin; Griffin P. Rodgers

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.


Journal of Biological Chemistry | 2016

Glia Maturation Factor-γ Regulates Monocyte Migration through Modulation of β1-Integrin

Wulin Aerbajinai; Lunhua Liu; Jianqiong Zhu; Chutima Kumkhaek; Kyung Chin; Griffin P. Rodgers

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 β1-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 α5β1-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 α5β1-integrin and increased its degradation. Importantly, we demonstrate that GMFG mediates the ubiquitination of β1-integrin through knockdown or overexpression of GMFG. Moreover, GMFG knockdown retarded the efficient recycling of β1-integrin back to the plasma membrane following normal endocytosis of α5β1-integrin, suggesting that the involvement of GMFG in maintaining α5β1-integrin stability may occur in part by preventing ubiquitin-mediated degradation and promoting β1-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 β1-integrin cell surface expression. Knockdown of STXBP4 also substantially inhibited β1-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 α5β1-integrin and facilitating effective β1-integrin recycling back to the plasma membrane.


British Journal of Haematology | 2007

Cl‐IB‐MECA inhibits human erythropoiesis

Natarajan V. Bhanu; Wulin Aerbajinai; Nicole M. Gantt; Edwin K. Jackson; Sung-Ho Goh; Y. Terry Lee; Jeffery L. Miller

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 μmol/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.

Collaboration


Dive into the Wulin Aerbajinai's collaboration.

Top Co-Authors

Avatar

Griffin P. Rodgers

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jianqiong Zhu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kyung Chin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Chutima Kumkhaek

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Wenli Liu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jeffery L. Miller

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Y. Terry Lee

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Hongzhen Li

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Lunhua Liu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Nicole M. Gantt

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge