Network


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

Hotspot


Dive into the research topics where Kyung Chin is active.

Publication


Featured researches published by Kyung Chin.


European Journal of Haematology | 2000

Two-phase liquid culture system models normal human adult erythropoiesis at the molecular level.

Sharon H. Pope; Eitan Fibach; Jun Sun; Kyung Chin; Griffin P. Rodgers

Abstract: We have studied the patterns of expression of various genes during maturation of normal human adult erythroid precursors cultured in a two‐phase liquid culture method. In the first phase, peripheral blood mononuclear cells are cultured for one week in the presence of a combination of growth factors, but not erythropoietin (Epo). In Phase II, Epo is included in the medium. Cell samples were taken throughout phase II, and expression of globins, transcription factors, and cytokine receptors was assayed by RT‐PCR and quantified by phosphor imaging. We have divided phase II into stages: early (days 0–5), intermediate (days 6–10) and late (days 11–15) and measured maximum expression of each gene. During early phase II, γ‐globin, Sp1, and GATA‐2 mRNAs were expressed at their highest levels. As the cells matured during the intermediate period, GATA‐2 levels remained high, and then declined, while the transcription factors GATA‐1, EKLF, NF‐E2, and the Epo receptor (EpoR) reached maximum expression. In late phase II, β‐globin increased and reached its maximum level of expression. This erythroid culture system appears to recapitulate normal adult erythropoiesis at the molecular level, and thus may be a suitable model to examine the molecular basis of severe congenital or acquired disorders oferythropoiesis.


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.


British Journal of Haematology | 2002

Hydroxyurea exerts bi-modal dose-dependent effects on erythropoiesis in human cultured erythroid cells via distinct pathways

Min Wang; Delia C. Tang; Wenli Liu; Kyung Chin; Jianqion G. Zhu; Eitan Fibach; Griffin P. Rodgers

Summary. Hydroxyurea (HU) has been shown to increase the proportion of fetal haemoglobin (HbF) in most sickle cell patients. A low‐dosage regimen increased total haemoglobin (Hb) levels in some thalassaemia intermedia patients by preferentially increasing β‐globin biosynthesis. To further characterize these apparent dose‐dependent effects of HU, we examined erythroid cells exposed to HU (5–100 µmol/l) in two‐phase liquid culture. Low doses (from 5 to 25 µmol/l) increased Hb levels by up to 2·7‐fold, and a high dose (100 µmol/l) increased Hb levels when added at d 3–6 of phase II, with no significant changes in response to HU during the late stage of phase II culture (≥ 9 d). HU exposure during d 0–3 of phase II culture increased the number of erythroid colonies to a maximum of fivefold at 5 µmol/l HU. GATA‐1 mRNA was downregulated at a high dose and GATA‐2 was dose dependently upregulated over a lower dosage range. Treatment with 100 µmol/l HU dramatically upregulated the death receptor DR‐5, caspase 3, as determined by cDNA microarray analysis. In contrast, 10 µmol/l HU modestly upregulated mRNA levels of the early growth response gene. Our results suggest that HU exerts concentration‐dependent effects on HbF production and erythropoiesis and that these two effects are mediated by distinct molecular mechanisms.


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 | 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.


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.


PLOS ONE | 2018

Analyses of genome wide association data, cytokines, and gene expression in African-Americans with benign ethnic neutropenia

Bashira A. Charles; Matthew M. Hsieh; Adebowale Adeyemo; Daniel Shriner; Edward Ramos; Kyung Chin; Kshitij Srivastava; Neil A. Zakai; Mary Cushman; Leslie A. McClure; Virginia J. Howard; Willy A. Flegel; Charles N. Rotimi; Griffin P. Rodgers

Background Benign ethnic neutropenia (BEN) is a hematologic condition associated with people of African ancestry and specific Middle Eastern ethnic groups. Prior genetic association studies in large population showed that rs2814778 in Duffy Antigen Receptor for Chemokines (DARC) gene, specifically DARC null red cell phenotype, was associated with BEN. However, the mechanism of this red cell phenotype leading to low white cell count remained elusive. Methods We conducted an extreme phenotype design genome-wide association study (GWAS), analyzed ~16 million single nucleotide polymorphisms (SNP) in 1,178 African-Americans individuals from the Reasons for Geographic and Racial Differences in Stroke (REGARDS) study and replicated from 819 African-American participants in the Atherosclerosis Risk in Communities (ARIC) study. Conditional analyses on rs2814778 were performed to identify additional association signals on chromosome 1q22. In a separate cohort of healthy individuals with and without BEN, whole genome gene expression from peripheral blood neutrophils were analyzed for DARC. Results We confirmed that rs2814778 in DARC was associated with BEN (p = 4.09×10−53). Conditioning on rs2814778 abolished other significant chromosome 1 associations. Inflammatory cytokines (IL-2, 6, and 10) in participants in the Howard University Family Study (HUFS) and Multi-Ethnic Study in Atherosclerosis (MESA) showed similar levels in individuals homozygous for the rs2814778 allele compared to others, indicating cytokine sink hypothesis played a minor role in leukocyte homeostasis. Gene expression in neutrophils of individuals with and without BEN was also similar except for low DARC expression in BEN, suggesting normal function. BEN neutrophils had slightly activated profiles in leukocyte migration and hematopoietic stem cell mobilization pathways (expression fold change <2). Conclusions These results in humans support the notion of DARC null erythroid progenitors preferentially differentiating to myeloid cells, leading to activated DARC null neutrophils egressing from circulation to the spleen, and causing relative neutropenia. Collectively, these human data sufficiently explained the mechanism DARC null red cell phenotype causing BEN and further provided a biologic basis that BEN is clinically benign.


Molecular Brain Research | 2000

Production and processing of erythropoietin receptor transcripts in brain.

Kyung Chin; Xiaobing Yu; Bojana Beleslin-Cokic; Chun Liu; Kun Shen; Harvey W. Mohrenweiser; Constance Tom Noguchi

Collaboration


Dive into the Kyung Chin's collaboration.

Top Co-Authors

Avatar

Griffin P. Rodgers

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Wulin Aerbajinai

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

Chutima Kumkhaek

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Constance Tom Noguchi

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

Wenli Liu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Delia C. Tang

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

Ling Chen

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge