Network


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

Hotspot


Dive into the research topics where Kyung Hee Chang is active.

Publication


Featured researches published by Kyung Hee Chang.


Diabetes | 2007

Ischemic vascular damage can be repaired by healthy, but not diabetic, endothelial progenitor cells.

Sergio Caballero; Nilanjana Sengupta; Aqeela Afzal; Kyung Hee Chang; Sergio Li Calzi; Dennis L Guberski; Timothy S. Kern; Maria B. Grant

Endothelial precursor cells (EPCs) play a key role in vascular repair and maintenance, and their function is impeded in diabetes. We previously demonstrated that EPCs isolated from diabetic patients have a profound inability to migrate in vitro. We asked whether EPCs from normal individuals are better able to repopulate degenerate (acellular) retinal capillaries in chronic (diabetes) and acute (ischemia/reperfusion [I/R] injury and neonatal oxygen-induced retinopathy [OIR]) animal models of ocular vascular damage. Streptozotocin-induced diabetic mice, spontaneously diabetic BBZDR/Wor rats, adult mice with I/R injury, or neonatal mice with OIR were injected within the vitreous or the systemic circulation with fluorescently labeled CD34+ cells from either diabetic patients or age- and sex-matched healthy control subjects. At specific times after administering the cells, the degree of vascular repair of the acellular capillaries was evaluated immunohistologically and quantitated. In all four models, healthy human (hu)CD34+ cells attached and assimilated into vasculature, whereas cells from diabetic donors uniformly were unable to integrate into damaged vasculature. These studies demonstrate that healthy huCD34+ cells can effectively repair injured retina and that there is defective repair of vasculature in patients with diabetes. Defective EPCs may be amenable to pharmacological manipulation and restoration of the cells’ natural robust reparative function.


Proceedings of the National Academy of Sciences of the United States of America | 2007

IGF binding protein-3 regulates hematopoietic stem cell and endothelial precursor cell function during vascular development

Kyung Hee Chang; Tailoi Chan-Ling; E. L. McFarland; Aqeela Afzal; Hao Pan; Louise Baxter; Lynn C. Shaw; Sergio Caballero; Nilanjana Sengupta; Sergio Li Calzi; Sean M. Sullivan; Maria B. Grant

We asked whether the hypoxia-regulated factor, insulin-like growth factor binding protein-3 (IGFBP3), could modulate stem cell factor receptor (c-kit+), stem cell antigen-1 (sca-1+), hematopoietic stem cell (HSC), or CD34+ endothelial precursor cell (EPC) function. Exposure of CD34+ EPCs to IGFBP3 resulted in rapid differentiation into endothelial cells and dose-dependent increases in cell migration and capillary tube formation. IGFBP3-expressing plasmid was injected into the vitreous of neonatal mice undergoing the oxygen-induced retinopathy (OIR) model. In separate studies, GFP-expressing HSCs were transfected with IGFBP3 plasmid and injected into the vitreous of OIR mice. Administering either IGFBP3 plasmid alone or HSCs transfected with the plasmid resulted in a similar reduction in areas of vasoobliteration, protection of the developing vasculature from hyperoxia-induced regression, and reduction in preretinal neovascularization compared to control plasmid or HSCs transfected with control plasmid. In conclusion, IGFBP3 mediates EPC migration, differentiation, and capillary formation in vitro. Targeted expression of IGFBP3 protects the vasculature from damage and promotes proper vascular repair after hyperoxic insult in the OIR model. IGFBP3 expression may represent a physiological adaptation to ischemia and potentially a therapeutic target for treatment of ischemic conditions.


Circulation Research | 2009

Insulin-Like Growth Factor Binding Protein-3 Mediates Vascular Repair by Enhancing Nitric Oxide Generation

Jennifer L. Kielczewski; Yagna P. R. Jarajapu; E. L. McFarland; Jun Cai; Aqeela Afzal; Sergio Li Calzi; Kyung Hee Chang; Todd A. Lydic; Lynn C. Shaw; Julia V. Busik; Jeffrey A. Hughes; Arturo J. Cardounel; Kenneth Wilson; Timothy J. Lyons; Michael E. Boulton; Robert N. Mames; Tailoi Chan-Ling; Maria B. Grant

Rationale: Insulin-like growth factor binding protein (IGFBP)-3 modulates vascular development by regulating endothelial progenitor cell (EPC) behavior, specifically stimulating EPC cell migration. This study was undertaken to investigate the mechanism of IGFBP-3 effects on EPC function and how IGFBP-3 mediates cytoprotection following vascular injury. Objective: To examine the mechanism of IGFBP-3–mediated repair following vascular injury. Methods and Results: We used 2 complementary vascular injury models: laser occlusion of retinal vessels in adult green fluorescent protein (GFP) chimeric mice and oxygen-induced retinopathy in mouse pups. Intravitreal injection of IGFBP-3–expressing plasmid into lasered GFP chimeric mice stimulated homing of EPCs, whereas reversing ischemia induced increases in macrophage infiltration. IGFBP-3 also reduced the retinal ceramide/sphingomyelin ratio that was increased following laser injury. In the OIR model, IGFBP-3 prevented cell death of resident vascular endothelial cells and EPCs, while simultaneously increasing astrocytic ensheathment of vessels. For EPCs to orchestrate repair, these cells must migrate into ischemic tissue. This migratory ability is mediated, in part, by endogenous NO generation. Thus, we asked whether the migratory effects of IGFBP-3 were attributable to stimulation of NO generation. IGFBP-3 increased endothelial NO synthase expression in human EPCs leading to NO generation. IGFBP-3 exposure also led to the redistribution of vasodilator-stimulated phosphoprotein, an NO regulated protein critical for cell migration. IGFBP-3–mediated NO generation required high-density lipoprotein receptor activation and stimulation of phosphatidylinositol 3-kinase/Akt pathway. Conclusion: These studies support consideration of IGFBP-3 as a novel agent to restore the function of injured vasculature and restore NO generation.


Diabetes | 2008

Carbon Monoxide and Nitric Oxide Mediate Cytoskeletal Reorganization in Microvascular Cells via Vasodilator-Stimulated Phosphoprotein Phosphorylation Evidence for Blunted Responsiveness in Diabetes

Sergio Li Calzi; Daniel L. Purich; Kyung Hee Chang; Aqeela Afzal; Takahiko Nakagawa; Julia V. Busik; Anupam Agarwal; Mark S. Segal; Maria B. Grant

OBJECTIVE— We examined the effect of the vasoactive agents carbon monoxide (CO) and nitric oxide (NO) on the phosphorylation and intracellular redistribution of vasodilator-stimulated phosphoprotein (VASP), a critical actin motor protein required for cell migration that also controls vasodilation and platelet aggregation. RESEARCH DESIGN AND METHODS— We examined the effect of donor-released CO and NO in endothelial progenitor cells (EPCs) and platelets from nondiabetic and diabetic subjects and in human microvascular endothelial cells (HMECs) cultured under low (5.5 mmol/l) or high (25 mmol/l) glucose conditions. VASP phosphorylation was evaluated using phosphorylation site-specific antibodies. RESULTS— In control platelets, CO selectively promotes phosphorylation at VASP Ser-157, whereas NO promotes phosphorylation primarily at Ser-157 and also at Ser-239, with maximal responses at 1 min with both agents on Ser-157 and at 15 min on Ser-239 with NO treatment. In diabetic platelets, neither agent resulted in VASP phosphorylation. In nondiabetic EPCs, NO and CO increased phosphorylation at Ser-239 and Ser-157, respectively, but this response was markedly reduced in diabetic EPCs. In endothelial cells cultured under low glucose conditions, both CO and NO induced phosphorylation at Ser-157 and Ser-239; however, this response was completely lost when cells were cultured under high glucose conditions. In control EPCs and in HMECs exposed to low glucose, VASP was redistributed to filopodia-like structures following CO or NO exposure; however, redistribution was dramatically attenuated under high glucose conditions. CONCLUSIONS— Vasoactive gases CO and NO promote cytoskeletal changes through site- and cell type–specific VASP phosphorylation, and in diabetes, blunted responses to these agents may lead to reduced vascular repair and tissue perfusion.


Investigative Ophthalmology & Visual Science | 2010

Paracrine Modulation of CXCR4 by IGF-1 and VEGF: Implications for Choroidal Neovascularization

Nilanjana Sengupta; Aqeela Afzal; Sergio Caballero; Kyung Hee Chang; Lynn C. Shaw; Ji-jing Pang; Vincent C. Bond; Imran Bhutto; Takayuki Baba; Gerard A. Lutty; Maria B. Grant

PURPOSE Modulators of angiogenesis typically work in an orchestrated manner. The authors examined the interaction between insulinlike growth factor (IGF)-1, vascular endothelial growth factor (VEGF), and stromal derived factor (SDF)-1 in vivo and in vitro using angiogenesis models. METHODS The angiogenic effect of SDF-1, alone or in combination with IGF-1 and VEGF, was assessed in human lung microvascular endothelial cells using capillary tube formation and thymidine incorporation. Immunohistochemical analysis for CD31, SDF-1, and CXCR4 was performed on mouse eyes 2 weeks after the initiation of laser rupture of Bruchs membrane, a choroidal neovascularization (CNV) model. CXCR4 antagonist and CXCR4 blocking antibody were tested on inhibition of CNV lesion size in this model. Real-time PCR was used to determine mRNA levels for SDF-1, VEGF, IGF-1, and their cognate receptors in the retinal pigment epithelium/choroid complex of mice that underwent this CNV model. RESULTS IGF-1 and VEGF demonstrated an additive effect on SDF-1-induced in vitro angiogenesis. CXCR4 immunoreactivity was present in both normal and laser-injured mice at the laser burn site and at the ganglion cell layer, the anterior portion of the inner nuclear layer, photoreceptors, and choroidal stroma. SDF-1 was observed in identical locations but was not seen in photoreceptors. mRNA levels for SDF-1, VEGF, and IGF-1 and their receptors were increased after laser injury. CXCR4-neutralizing antibody reduced neovascularization when injected subretinally but not intraperitoneally or intravitreally. CONCLUSIONS The potent proangiogenic factors IGF-1 and VEGF both stimulate SDF-1-induced angiogenesis. Local inhibition of CXCR4 is required for an antiangiogenic effect in CNV lesions.


Microvascular Research | 2009

Labeling of stem cells with monocrystalline iron oxide for tracking and localization by magnetic resonance imaging

Sergio Li Calzi; David Kent; Kyung Hee Chang; Kyle R. Padgett; Aqeela Afzal; Saurav Chandra; Sergio Caballero; Denis English; Wendy Garlington; Paul Hiscott; Carl Sheridan; Maria B. Grant; John R. Forder

Precise localization of exogenously delivered stem cells is critical to our understanding of their reparative response. Our current inability to determine the exact location of small numbers of cells may hinder optimal development of these cells for clinical use. We describe a method using magnetic resonance imaging to track and localize small numbers of stem cells following transplantation. Endothelial progenitor cells (EPC) were labeled with monocrystalline iron oxide nanoparticles (MIONs) which neither adversely altered their viability nor their ability to migrate in vitro and allowed successful detection of limited numbers of these cells in muscle. MION-labeled stem cells were also injected into the vitreous cavity of mice undergoing the model of choroidal neovascularization, laser rupture of Bruchs membrane. Migration of the MION-labeled cells from the injection site towards the laser burns was visualized by MRI. In conclusion, MION labeling of EPC provides a non-invasive means to define the location of small numbers of these cells. Localization of these cells following injection is critical to their optimization for therapy.


Investigative Ophthalmology & Visual Science | 2006

The Effect of a Somatostatin Analog on the Transepithelial Transport of ARPE 19 Cells

W. Garlington; Aqeela Afzal; S. LiCalzi; Yagna P. R. Jarajapu; Kyung Hee Chang; Maria B. Grant; Michael E. Boulton; H.L. Brooks


Investigative Ophthalmology & Visual Science | 2008

IGF Binding Protein-3 (IGFBP3) Affects Endothelial Cell Ensheathment and Apoptosis in Normoxia and in the Oxygen Induced Retinopathy (OIR) Model

E. L. McFarland; Aqeela Afzal; Jennifer L. Kielczewski; Kyung Hee Chang; Tom A. Gardiner; Maria B. Grant; Tailoi Chan-Ling


Investigative Ophthalmology & Visual Science | 2008

IGFBP-3 Acts as a Hematopoietic Stem Cell Homing Factor in an Adult Mouse Ocular Neovascularization Model

Jennifer L. Kielczewski; Aqeela Afzal; Kyung Hee Chang; Lynn C. Shaw; Robert N. Mames; E. L. McFarland; Tailoi Chan-Ling; Jeffrey A. Hughes; Maria B. Grant


Investigative Ophthalmology & Visual Science | 2008

Carbon Monoxide (CO) Mediates Cytoskeletal Rearrangements via Vasp Phosphorylation and Subsequent Redistribution in Vascular Cells

S. LiCalzi; Daniel L. Purich; Kyung Hee Chang; Aqeela Afzal; Anupam Agarwal; Julia V. Busik; Mark S. Segal; Maria B. Grant

Collaboration


Dive into the Kyung Hee Chang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julia V. Busik

Michigan State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge