Network


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

Hotspot


Dive into the research topics where Eun Ju Lee is active.

Publication


Featured researches published by Eun Ju Lee.


Blood | 2010

Induction of pluripotent stem cells from adult somatic cells by protein-based reprogramming without genetic manipulation

Hyun-Jai Cho; Choon-Soo Lee; Yoo-Wook Kwon; Jae Seung Paek; Sun Hee Lee; Jin Hur; Eun Ju Lee; Tae-Young Roh; In-Sun Chu; Sun-Hee Leem; Youngsoo Kim; Hyun-Jae Kang; Young-Bae Park; Hyo-Soo Kim

The concept of reprogramming of somatic cells has opened a new era in regenerative medicine. Transduction of defined factors has successfully achieved pluripotency. However, during the generation process of induced pluripotent stem (iPS) cells, genetic manipulation of certain factors may cause tumorigenicity, which limits further application. We report that that a single transfer of embryonic stem (ES) cell-derived proteins into primarily cultured adult mouse fibroblasts, rather than repeated transfer or prolonged exposure to materials, can achieve full reprogramming up to the pluripotent state without the forced expression of ectopic transgenes. During the process, gene expression and epigenetic status were converted from somatic to ES-equivalent status. We verified that protein-based reprogramming was neither by the contamination of protein donor ES cell nor by DNA/RNA from donor ES cell. Protein-iPS cells were biologically and functionally very similar to ES cells and differentiated into 3 germ layers in vitro. Furthermore, protein-iPS cells possessed in vivo differentiation (well-differentiated teratoma formation) and development (chimeric mice generation and a tetraploid blastocyst complementation) potentials. Our results provide an alternative and safe strategy for the reprogramming of somatic cells that can be used to facilitate pluripotent stem cell-based cell therapy.


Molecular Therapy | 2012

Spherical Bullet Formation via E-cadherin Promotes Therapeutic Potency of Mesenchymal Stem Cells Derived From Human Umbilical Cord Blood for Myocardial Infarction

Eun Ju Lee; Sung Jung Park; Soo Kyoung Kang; Gi-Hwan Kim; Hyun-Jae Kang; Sae-Won Lee; Hong Bae Jeon; Hyo-Soo Kim

The beneficial effects of stem cells in clinical applications to date have been modest, and studies have reported that poor engraftment might be an important reason. As a strategy to overcome such a hurdle, we developed the spheroid three dimensional (3D) bullet as a delivery method for human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) through the maintenance of cell-cell interactions without additional xenofactors, cytokines, or matrix. We made spheroid 3D-bullets from hUCB-MSCs at 24 hours anchorage-deprived suspension culture. To investigate the in vivo therapeutic efficacy of 3D-bullets, we used rat myocardial infarction (MI) model. Transplantation of 3D-bullet was better than that of single cells from monolayer culture or from 3D-bullet in improving left ventricular (LV) contractility [LV ejection fraction (LVEF) or LV fractional shortening (LVFS)] and preventing pathologic LV dilatation [LV end-systolic diameter (LVESD) or LV end-diastolic diameter (LVEDD)] at 8 weeks. In the mechanism study of 3D-bullet formation, we found that calcium-dependent cell-cell interaction was essential and that E-cadherin is a key inducer mediating hUCB-MSC 3D-bullet formation among several calcium-dependent adhesion molecules which were nominated as candidates after cDNA array analysis. In more specific experiments with E-cadherin overexpression using adenoviral vector or with E-cadherin neutralization using blocking antibody, we found that E-cadherin regulates vascular endothelial growth factor (VEGF) secretion via extracellular signal-regulated kinase (ERK)/v-akt murine thymoma viral oncogene homolog1 (AKT) pathways. During formation of spheroid 3D-bullets, activation of E-cadherin in association with cell-cell interaction turns on ERK/AKT signaling pathway that are essential to proliferative and paracrine activity of MSCs leading to the enhanced therapeutic efficacy.


Stem Cells | 2008

Impact of Myocardial Infarct Proteins and Oscillating Pressure on the Differentiation of Mesenchymal Stem Cells : Effect of Acute Myocardial Infarction on Stem Cell Differentiation

Sung-A Chang; Eun Ju Lee; Hyun-Jae Kang; Shu-Ying Zhang; Ji Hyun Kim; Lian Li; Seock-Won Youn; Choon-Soo Lee; Keum-Hyun Kim; Joo-Yun Won; Jong-Woo Sohn; Kyung-Woo Park; Hyun-Jai Cho; Sung-Eun Yang; Won Il Oh; Yoon Sun Yang; Won-Kyung Ho; Young-Bae Park; Hyo-Soo Kim

Stem cell transplantation in acute myocardial infarction (AMI) has emerged as a promising therapeutic option. We evaluated the impact of AMI on mesenchymal stem cell (MSC) differentiation into cardiomyocyte lineage. Cord blood‐derived human MSCs were exposed to in vitro conditions simulating in vivo environments of the beating heart with acute ischemia, as follows: (a) myocardial proteins or serum obtained from sham‐operated rats, and (b) myocardial proteins or serum from AMI rats, with or without application of oscillating pressure. Expression of cardiac‐specific markers on MSCs was greatly induced by the infarcted myocardial proteins, compared with the normal proteins. It was also induced by application of oscillating pressure to MSCs. Treatment of MSCs with infarcted myocardial proteins and oscillating pressure greatly augmented expression of cardiac‐specific genes. Such expression was blocked by inhibitor of transforming growth factor β1 (TGF‐β1) or bone morphogenetic protein‐2 (BMP‐2). In vitro cellular and electrophysiologic experiments showed that these differentiated MSCs expressing cardiomyocyte‐specific markers were able to make a coupling with cardiomyocytes but not to selfbeat. The pathophysiologic significance of in vitro results was confirmed using the rat AMI model. The protein amount of TGF‐β1 and BMP‐2 in myocardium of AMI was significantly higher than that in normal myocardium. When MSCs were transplanted to the heart and analyzed 8 weeks later, they expressed cardiomyocyte‐specific markers, leading to improved cardiac function. These in vitro and in vivo results suggest that infarct‐related biological and physical factors in AMI induce commitment of MSCs to cardiomyocyte‐like cells through TGF‐β/BMP‐2 pathways.


Fertility and Sterility | 2010

Embryonic stem cell-like cells established by culture of adult ovarian cells in mice

Seung Pyo Gong; Seung Tae Lee; Eun Ju Lee; Dae Yong Kim; Gene Lee; Sung Gil Chi; Byung Kyu Ryu; Chae Hyun Lee; Kyung Eun Yum; Ho Joon Lee; Jae Yong Han; Jonathan L. Tilly; Jeong Mook Lim

OBJECTIVEnTo suggest an alternative strategy for deriving histocompatible stems cells without undertaking genetic manipulation.nnnDESIGNnProspective approach using an animal model.nnnSETTINGnStem cell and bioevaluation laboratory, Seoul National University.nnnANIMAL(S)nF1 (C57BL6 X DBA2) and outbred (ICR) mice.nnnINTERVENTION(S)nOvarian stroma cells of less than 40 mum in diameter were subcultured with fibroblast monolayer, and colony-forming cells were characterized.nnnMAIN OUTCOME MEASURE(S)nStemness, genotype, and imprinted gene methylation.nnnRESULT(S)nTwo-lines of colony-forming cells were established, which expressed markers specific for embryonic stem cells (ESC) and formed embryoid bodies and teratomas. Complete matching of microsatellite markers with the cell donor strain confirmed their establishment from ovarian tissue, and identification of both homozygotic and heterozygotic chromosomes raised the possibility of their derivation from parthenogenetic oocytes. However, the use of cells smaller than mature oocytes for primary culture, the difference in imprinted gene methylation compared with parthenogenetic ESCs, and failure to establish the ESC-like cells by primary follicle culture collectively suggested the irrelevancy to gametes.nnnCONCLUSION(S)nCoculture of adult ovarian cells with somatic fibroblasts can yield colony-forming cells having ESC-like activity, which may provide an alternative for establishing autologous stem cells from adults that can be obtained without genetic manipulation.


Molecular Therapy | 2013

Snail as a Potential Target Molecule in Cardiac Fibrosis: Paracrine Action of Endothelial Cells on Fibroblasts Through Snail and CTGF Axis

Sae-Won Lee; Joo-Yun Won; Woo Jean Kim; Jaewon Lee; Kyung-Hee Kim; Seock-Won Youn; Ju-Young Kim; Eun Ju Lee; Yong-Jin Kim; Kyu-Won Kim; Hyo-Soo Kim

Ischemia/reperfusion (I/R) injury to myocardium induces death of cardiomyocytes and destroys the vasculature, leading to cardiac fibrosis that is mainly mediated by the transdifferentiation of fibroblasts to myofibroblasts and the collagen deposition. Snail involvement in fibrosis is well known; however, the contribution of Snail to cardiac fibrosis during I/R injury and its underlying mechanisms have not been defined. We showed that I/R injury to mouse hearts significantly increases the expression of Snail. An in vitro hypoxia/reoxygenation (Hy/Reoxy) experiment showed that the cell source of Snail induction is endothelial cells rather than cardiac fibroblasts (cFibroblasts) or cardiomyoblasts. When Snail was overexpressed in endothelial cells, they underwent endothelial-to-mesenchymal transition (EndMT) but showed very poor capacity for collagen synthesis. Instead, reoxygenation- or Snail overexpression-mediated EndMT-like cells noticeably stimulated transdifferentiation of fibroblasts to myofibroblasts via secretion of connective tissue growth factor (CTGF). The injection of a peroxisome proliferator-activated receptor-γ (PPAR-γ) agonist, a selective Snail inhibitor, remarkably suppressed collagen deposition and cardiac fibrosis in mouse I/R injury, and significantly improved cardiac function and reduced Snail and CTGF expression in vivo. Our findings suggested a new mechanism of cell-to-cell communication between EndMT-like cells and fibroblasts for fibrosis induction and implicated Snail as a potential target molecule in cardiac fibrosis after I/R injury.


Embo Molecular Medicine | 2012

Hypoxic priming of mESCs accelerates vascular-lineage differentiation through HIF1-mediated inverse regulation of Oct4 and VEGF

Sae-Won Lee; Han-Kyul Jeong; Ji Young Lee; Jimin Yang; Eun Ju Lee; Su Yeon Kim; Seock-Won Youn; Jaewon Lee; Woo Jean Kim; Kyu-Won Kim; Jeong Mook Lim; Jong-Wan Park; Young Bae Park; Hyo-Soo Kim

Hypoxic microenvironment plays an important role in determining stem cell fates. However, it is controversial to which direction between self‐renewal and differentiation the hypoxia drives the stem cells. Here, we investigated whether a short exposure to hypoxia (termed ‘hypoxic‐priming’) efficiently directed and promoted mouse embryonic stem cells (mESCs) to differentiate into vascular‐lineage. During spontaneous differentiation of embryoid bodies (EBs), hypoxic region was observed inside EB spheroids even under normoxic conditions. Indeed, hypoxia‐primed EBs more efficiently differentiated into cells of vascular‐lineage, than normoxic EBs did. We found that hypoxia suppressed Oct4 expression via direct binding of HIF‐1 to reverse hypoxia‐responsive elements (rHREs) in the Oct4 promoter. Furthermore, vascular endothelial growth factor (VEGF) was highly upregulated in hypoxia‐primed EBs, which differentiated towards endothelial cells in the absence of exogenous VEGF. Interestingly, this differentiation was abolished by the HIF‐1 or VEGF blocking. In vivo transplantation of hypoxia‐primed EBs into mice ischemic limb elicited enhanced vessel differentiation. Collectively, our findings identify that hypoxia enhanced ESC differentiation by HIF‐1‐mediated inverse regulation of Oct4 and VEGF, which is a novel pathway to promote vascular‐lineage differentiation.


Stem Cells | 2008

Notch Inhibition Promotes Human Embryonic Stem Cell-Derived Cardiac Mesoderm Differentiation

Jiho Jang; Seung Yup Ku; Jung Eun Kim; Kyunghee Choi; Yoon Young Kim; Hee Sun Kim; Sun Kyung Oh; Eun Ju Lee; Hyun-Jai Cho; Young Hwan Song; Sang-Hun Lee; Sukho Lee; Chang Suk Suh; Seok Hyun Kim; Shin Yong Moon; Young Min Choi

The roles of Notch signaling in cardiac differentiation from murine embryonic stem cells have been well documented. We investigated whether Notch signaling plays a similar role in human embryonic stem cells (hESCs). Although, as previously reported, blocking Notch signaling via the addition of γ‐secretase inhibitor (GSI) alone failed to affect hESC differentiation, we found that GSI plus reduced‐volume culture medium (GSI/RVCM) accelerated mesodermal differentiation. GSI/RVCM conditions simultaneously suppressed commitment toward neuroectodermal lineages. Furthermore, sustained inhibition of Notch signaling further enhanced differentiation into cardiac mesoderm. Spontaneous beating activity was typically observed from 12 days after initiation of GSI treatment in RVCM. Moreover, hESC‐derived cardiomyocytes expressed connexin 43 and possessed spontaneous calcium oscillations and cardiomyocyte beats coupled to neonatal rat cardiomyocytes when cocultured. These findings strongly suggest a distinct role for Notch signaling in the induction and specification of hESC‐derived cardiac mesoderm in vitro.


Molecular Therapy | 2012

N-cadherin Determines Individual Variations in the Therapeutic Efficacy of Human Umbilical Cord Blood-derived Mesenchymal Stem Cells in a Rat Model of Myocardial Infarction

Eun Ju Lee; Eue-Keun Choi; Soo Kyoung Kang; Gi-Hwan Kim; Ju Young Park; Hyun-Jae Kang; Sae-Won Lee; Keum-Hyun Kim; Jin Sook Kwon; Ki Hong Lee; Youngkeun Ahn; Ho-Jae Lee; Hyun-Jai Cho; Soo Jin Choi; Won Il Oh; Young-Bae Park; Hyo-Soo Kim

In this study, we established and characterized human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) from four different donors. However, the hUCB-MSCs showed remarkable variations in their therapeutic efficacy for repairing rat infarcted myocardium (including the process of angiogenesis) 8 weeks after transplantation. In addition, we observed that the level of vascular endothelial growth factor (VEGF) is correlated with the therapeutic efficacy of the four hUCB-MSCs. Next, to investigate the practical application of hUCB-MSCs, we searched for surface signature molecules that could serve as indicators of therapeutic efficacy. The gene for N-cadherin was the only cell surface gene that was highly expressed in the most effective hUCB-MSCs, both at the transcriptional and translational levels. We observed downregulation and upregulation of VEGF in response to N-cadherin blocking and N-cadherin overexpression, respectively. Activation of extracellular signal-regulated kinase (ERK), but not protein kinase B, was increased when N-cadherin expression was increased, whereas disruption of N-cadherin-mediated cell-cell contact induced suppression of ERK activation and led to VEGF downregulation. Moreover, by investigating hUCB-MSCs overexpressing N-cadherin or N-cadherin knockdown hUCB-MSCs, we confirmed the in vivo function of N-cadherin. In addition, we observed that DiI-labeled hUCB-MSCs express N-cadherin in the peri-infarct area and interact with cardiomyocytes.


Molecular Therapy | 2012

Secondary Sphere Formation Enhances the Functionality of Cardiac Progenitor Cells

Hyun-Jai Cho; Ho-Jae Lee; Seock-Won Youn; Seok-Jin Koh; Joo-Yun Won; Yeon-Ju Chung; Hyun-Ju Cho; Chang-Hwan Yoon; Sae-Won Lee; Eun Ju Lee; Yoo-Wook Kwon; Hae-Young Lee; Sang-Hun Lee; Won-Kyung Ho; Young-Bae Park; Hyo-Soo Kim

Loss of cardiomyocytes impairs cardiac function after myocardial infarction (MI). Recent studies suggest that cardiac stem/progenitor cells could repair the damaged heart. However, cardiac progenitor cells are difficult to maintain in terms of purity and multipotency when propagated in two-dimensional culture systems. Here, we investigated a new strategy that enhances potency and enriches progenitor cells. We applied the repeated sphere formation strategy (cardiac explant → primary cardiosphere (CS) formation → sphere-derived cells (SDCs) in adherent culture condition → secondary CS formation by three-dimensional culture). Cells in secondary CS showed higher differentiation potentials than SDCs. When transplanted into the infarcted myocardium, secondary CSs engrafted robustly, improved left ventricular (LV) dysfunction, and reduced infarct sizes more than SDCs did. In addition to the cardiovascular differentiation of transplanted secondary CSs, robust vascular endothelial growth factor (VEGF) synthesis and secretion enhanced neovascularization in the infarcted myocardium. Microarray pathway analysis and blocking experiments using E-selectin knock-out hearts, specific chemicals, and small interfering RNAs (siRNAs) for each pathway revealed that E-selectin was indispensable to sphere initiation and ERK/Sp1/VEGF autoparacrine loop was responsible for sphere maturation. These results provide a simple strategy for enhancing cellular potency for cardiac repair. Furthermore, this strategy may be implemented to other types of stem/progenitor cell-based therapy.


Biomaterials | 2012

Regeneration of peripheral nerves by transplanted sphere of human mesenchymal stem cells derived from embryonic stem cells

Eun Ju Lee; Lianji Xu; Gi-Hwan Kim; Soo Kyoung Kang; Sae-Won Lee; Sung-Hye Park; Sukwha Kim; Tae Hyun Choi; Hyo-Soo Kim

In cell therapy, the most important factor for therapeutic efficacy is the stable supply of cells with best engraftment efficiency. To meet this requirement, we have developed a culture strategy such as three-dimensional sphere of human embryonic stem cell-derived mesenchymal stem cells (hESC-MSCs) in serum-free medium. To investigate the in vivo therapeutic efficacy of hESC-MSC spheres in nerve injury model, we transected the sciatic nerve in athymic nude mice and created a 2-mm gap. Transplantation of hESC-MSC as sphere repaired the injured nerve significantly better than transplantation of hESC-MSC as suspended single cells in regard to 1) nerve conduction (sphere; 28.81 ± 3.55 vs. single cells; 18.04 ± 2.10, p < 0.05) and 2) susceptibility of nerve stimulation at low voltage (sphere; 0.38 ± 0.08 vs. single cells; 0.66 ± 0.11, p < 0.05) at 8 weeks. Recovery after sphere transplantation was near-complete when compared with the data of normal control (sphere 28.81 ± 3.55 vs normal 32.62 ± 2.85 in nerve conduction : sphere 0.38 ± 0.08 vs normal 0.36 ± 0.67 in susceptibility of nerve stimulation, no significant difference, respectively). Recovery in function of the injured nerve was well corroborated by the histologic evidence of regenerated nerve. In the mechanistic analysis, the supernatant of sphere-forming hESC-MSC contains hepatocyte growth factor and insulin-like growth factor-binding protein-1 significantly more than the supernatant of the single cells of hESC-MSC has, which might be the key factors for the improved engraftment efficiency and greater regeneration of injured peripheral nerve.

Collaboration


Dive into the Eun Ju Lee's collaboration.

Top Co-Authors

Avatar

Hyo-Soo Kim

Seoul National University Hospital

View shared research outputs
Top Co-Authors

Avatar

Hyun-Jae Kang

Seoul National University Hospital

View shared research outputs
Top Co-Authors

Avatar

Sae-Won Lee

Seoul National University Hospital

View shared research outputs
Top Co-Authors

Avatar

Hyo Soo Kim

Seoul National University Hospital

View shared research outputs
Top Co-Authors

Avatar

Young-Bae Park

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Jeong Mook Lim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Gi-Hwan Kim

Seoul National University Hospital

View shared research outputs
Top Co-Authors

Avatar

Hyun-Jai Cho

Seoul National University Hospital

View shared research outputs
Top Co-Authors

Avatar

Keun Cheon Kim

Seoul National University Hospital

View shared research outputs
Top Co-Authors

Avatar

Seock-Won Youn

Seoul National University Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge