Network


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

Hotspot


Dive into the research topics where Yu Jin Jang is active.

Publication


Featured researches published by Yu Jin Jang.


Cell Reports | 2016

Small Molecules Facilitate Single Factor-Mediated Hepatic Reprogramming

Kyung Tae Lim; Seung Chan Lee; Yimeng Gao; Kee Pyo Kim; Guangqi Song; Su Yeon An; Kenjiro Adachi; Yu Jin Jang; Jonghun Kim; Kyoung Jin Oh; Tae Hwan Kwak; Seon In Hwang; Jueng Soo You; Kinarm Ko; Seung Hoi Koo; Amar Deep Sharma; Jong-Hoon Kim; Lijian Hui; Tobias Cantz; Hans R. Schöler; Dong Wook Han

Recent studies have shown that defined factors could lead to the direct conversion of fibroblasts into induced hepatocyte-like cells (iHeps). However, reported conversion efficiencies are very low, and the underlying mechanism of the direct hepatic reprogramming is largely unknown. Here, we report that direct conversion into iHeps is a stepwise transition involving the erasure of somatic memory, mesenchymal-to-epithelial transition, and induction of hepatic cell fate in a sequential manner. Through screening for additional factors that could potentially enhance the conversion kinetics, we have found that c-Myc and Klf4 (CK) dramatically accelerate conversion kinetics, resulting in remarkably improved iHep generation. Furthermore, we identified small molecules that could lead to the robust generation of iHeps without CK. Finally, we show that Hnf1α supported by small molecules is sufficient to efficiently induce direct hepatic reprogramming. This approach might help to fully elucidate the direct conversion process and also facilitate the translation of iHep into the clinic.


ACS Nano | 2016

Nanotopography Promotes Pancreatic Differentiation of Human Embryonic Stem Cells and Induced Pluripotent Stem Cells

Jong Hyun Kim; Hyung Woo Kim; Kyoung Je Cha; Jiyou Han; Yu Jin Jang; Dong Sung Kim; Jong Hoon Kim

Although previous studies suggest that nanotopographical features influence properties and behaviors of stem cells, only a few studies have attempted to derive clinically useful somatic cells from human pluripotent stem cells using nanopatterned surfaces. In the present study, we report that polystyrene nanopore-patterned surfaces significantly promote the pancreatic differentiation of human embryonic and induced pluripotent stem cells. We compared different diameters of nanopores and showed that 200 nm nanopore-patterned surfaces highly upregulated the expression of PDX1, a critical transcription factor for pancreatic development, leading to an approximately 3-fold increase in the percentage of differentiating PDX1(+) pancreatic progenitors compared with control flat surfaces. Furthermore, in the presence of biochemical factors, 200 nm nanopore-patterned surfaces profoundly enhanced the derivation of pancreatic endocrine cells producing insulin, glucagon, or somatostatin. We also demonstrate that nanopore-patterned surface-induced upregulation of PDX1 is associated with downregulation of TAZ, suggesting the potential role of TAZ in nanopore-patterned surface-mediated mechanotransduction. Our study suggests that appropriate cytokine treatments combined with nanotopographical stimulation could be a powerful tool for deriving a high purity of desired cells from human pluripotent stem cells.


Stem Cells and Development | 2013

Engraftment potential of spheroid-forming hepatic endoderm derived from human embryonic stem cells.

Sung Eun Kim; Su Yeon An; Dong Hun Woo; Jiyou Han; Jong Hyun Kim; Yu Jin Jang; Jeong Sang Son; Hyunwon Yang; Yong Pil Cheon; Jong Hoon Kim

Transplantation and drug discovery programs for liver diseases are hampered by the shortage of donor tissue. While recent studies have shown that hepatic cells can be derived from human embryonic stem cells (hESCs), few cases have shown selective enrichment of hESC-derived hepatocytes and their integration into host liver tissues. Here we demonstrate that the dissociation and reaggregation procedure after an endodermal differentiation of hESC produces spheroids mainly consisted of cells showing hepatic phenotypes in vitro and in vivo. A combined treatment with Wnt3a and bone morphogenic protein 4 efficiently differentiated hESCs into definitive endoderm in an adherent culture. Dissociation followed by reaggregation of these cells in a nonadherent condition lead to the isolation of spheroid-forming cells that preferentially expressed early hepatic markers from the adherent cell population. Further differentiation of these spheroid cells in the presence of the hepatocyte growth factor, oncostatin M, and dexamethasone produced a highly enriched population of cells exhibiting characteristics of early hepatocytes, including glycogen storage, indocyanine green uptake, and synthesis of urea and albumin. Furthermore, we show that grafted spheroid cells express hepatic features and attenuate the serum aspartate aminotransferase level in a model of acute liver injury. These data suggest that hepatic progenitor cells can be enriched by the spheroid formation of differentiating hESCs and that these cells have engraftment potential to replace damaged liver tissues.


Gastroenterology | 2017

Milk Fat Globule-EGF Factor 8, Secreted by Mesenchymal Stem Cells, Protects Against Liver Fibrosis in Mice

Su Yeon An; Yu Jin Jang; Hee Joung Lim; Jiyou Han; Jaehun Lee; Gyunggyu Lee; Ji Young Park; Seo Young Park; Ji Hyang Kim; Byung Rok Do; Choongseong Han; Hee Kyung Park; Ok Hee Kim; Myeong Jun Song; Say June Kim; Jong Hoon Kim

BACKGROUND & AIMSnMesenchymal stem cells (MSCs) mediate tissue repair and might be used to prevent or reduce liver fibrosis. However, little is known about the anti-fibrotic factors secreted from MSCs or their mechanisms.nnnMETHODSnUmbilical cord-derived MSCs (UCMSCs) were differentiated into hepatocyte-like cells (hpUCMSCs), medium was collected, andxa0secretome proteins were identified and quantified using nanochip-liquid chromatography/quadrupole time-of-flight mass spectrometry. Liver fibrosis was induced in mice by intraperitoneal injection of thioacetamide or CCl4; some mice were then given injections of secretomes or proteins. Liver tissues were collected and analyzed by histology or polymerase chain reaction array to analyze changes in gene expression patterns. We analyzed the effects of MSC secretomes and potential anti-fibrotic proteins on transforming growth factor β 1 (TGFβ1)-mediated activation of human hepatic stellate cell (HSC) lines (hTert-HSC and LX2) and human primary HSCs. Liver tissues were collected from 16 patients with liver cirrhosis and 16 individuals without cirrhosis (controls) in Korea and analyzed by immunohistochemistry and immunoblots.nnnRESULTSnIn mice with fibrosis, accumulation of extracellular matrix proteins was significantly reduced 3 days after injecting secretomes from UCMSCs, and to a greater extent from hpUCMSCs; numbers of activated HSCs that expressed the myogenic marker α-smooth muscle actin (α-SMA, encoded by ACTA2 [actin, alpha 2, smooth muscle]) were also reduced. Secretomes from UCMSCs, and to a greater extent from hpUCMSCs, reduced liver expression of multiple fibrotic factors, collagens, metalloproteinases, TGFβ, and Smad proteins in the TGFβ signaling pathways. In HSC cell lines and primary HSCs, TGFβ1-stimulated upregulation of α-SMA was significantly inhibited (and SMAD2 phosphorylation reduced) by secretomes from UCMSCs, and to a greater extent from hpUCMSCs. We identified 32 proteins in secretomes of UCMSCs that were more highly concentrated in secretomes from hpUCMSCs and inhibited TGFβ-mediated activation of HSCs. One of these, milk fat globule-EGF factor 8 (MFGE8), was a strong inhibitor of activation of human primary HSCs. We found MFGE8 to down-regulate expression of TGFβ type I receptor by binding to αvβ3 integrin on HSCs and to be secreted by MSCs from umbilical cord, teeth, and bone marrow. In mice, injection of recombinant human MFGE8 had anti-fibrotic effects comparable to those ofxa0the hpUCMSC secretome, reducing extracellular matrix deposition and HSC activation. Co-injection of an antibody against MFGE8 reduced the anti-fibrotic effects of the hpUCMSC secretome in mice. Levels of MFGE8 were reduced in cirrhotic liver tissue from patients compared with controls.nnnCONCLUSIONSnMFGE8 is an anti-fibrotic protein in MSC secretomes that strongly inhibits TGFβ signaling and reduces extracellular matrix deposition and liver fibrosis in mice.


Cell Transplantation | 2015

Pancreatic Islet-Like Three-Dimensional Aggregates Derived From Human Embryonic Stem Cells Ameliorate Hyperglycemia in Streptozotocin-Induced Diabetic Mice.

Joong Hyun Shim; Jong Hyun Kim; Jiyou Han; Su Yeon An; Yu Jin Jang; Jeongsang Son; Dong Hun Woo; Suel Kee Kim; Jong Hoon Kim

We previously reported the in vitro differentiation of human embryonic stem cells (hESCs) into pancreatic endoderm. Here we demonstrate that islet-like three-dimensional (3D) aggregates can be derived from the pancreatic endoderm by optimizing our previous protocol. Sequential treatment with Wnt3a, activin A, and noggin induced a transient upregulation of T and MixL1, followed by increased expression of endodermal genes, including FOXA2, SOX17, and CXCR4. Subsequent treatment with retinoic acid highly upregulated PDX1 expression. We also show that inhibition of sonic hedgehog signaling by bFGF/activin βB and cotreatment with VEGF and FGF7 produced many 3D cellular clusters that express both SOX17 and PDX1. We found for the first time that proteoglycans and vimentin+ mesenchymal cells were mainly localized in hESC-derived PDX1+ clusters. Importantly, treatment with chlorate, an inhibitor of proteoglycan sulfation, together with inhibition of Notch signaling significantly increased the expression of Neurog3 and NeuroD1, promoting a transition from PDX1+ progenitor cells toward mature pancreatic endocrine cells. Purified dithizone+ 3D aggregates generated by our refined protocol produced pancreatic hormones and released insulin in response to both glucose and pharmacological drugs in vitro. Furthermore, the islet-like 3D aggregates decreased blood glucose levels and continued to exhibit pancreatic features after transplantation into diabetic mice. Generation of islet-like 3D cell aggregates from human pluripotent stem cells may overcome the shortage of cadaveric donor islets for future cases of clinical islet transplantation.


Journal of Biochemistry and Molecular Biology | 2017

Identification of MFGE8 in mesenchymal stem cell secretome as an anti-fibrotic factor in liver fibrosis

Yu Jin Jang; Su Yeon An; Jong Hoon Kim

The beneficial paracrine roles of mesenchymal stem cells (MSCs) in tissue repair have potential in therapeutic strategies against various diseases. However, the key therapeutic factors secreted from MSCs and their exact molecular mechanisms of action remain unclear. In this study, the cell-free secretome of umbilical cord-derived MSCs showed significant anti-fibrotic activity in the mouse models of liver fibrosis. The involved action mechanism was the regulation of hepatic stellate cell activation by direct inhibition of the TGFβ/Smad-signaling. Antagonizing the milk fat globule-EGF factor 8 (MFGE8) activity blocked the anti-fibrotic effects of the MSC secretome in vitro and in vivo. Moreover, MFGE8 was secreted by MSCs from the umbilical cord as well as other tissues, including teeth and bone marrow. Administration of recombinant MFGE8 protein alone had a significant anti-fibrotic effect in two different models of liver fibrosis. Additionally, MFGE8 downregulated TGFβ type I receptor expression by binding to αvβ3 integrin on HSCs. These findings revealed the potential role of MFGE8 in modulating TGFβ-signaling. Thus, MFGE8 could serve as a novel therapeutic agent for liver fibrosis.


Journal of Applied Microbiology | 2012

Cu(II)‐induced molecular and physiological responses in the brown‐rot basidiomycete Polyporales sp. KUC9061

Yu Jin Jang; Hyung-Min Lee; Suck-Ho Lee; Yongho Choi; Byoung Jun Ahn; Gyu-Tae Kim; Joon-Young Kim

A potentially safe disposal method for copper‐containing waste wood is bioremediation using brown‐rot fungi. However, the mechanisms regulating brown‐rot fungi copper tolerance are poorly understood. The objective of this study was to better understand the molecular and physiological changes in Polyporales sp. KUC9061 in response to Cu(II) using GeneFishing technology.


Chemical Communications | 2015

Recent development of biotin conjugation in biological imaging, sensing, and target delivery

Wen Xiu Ren; Jiyou Han; Soojin Uhm; Yu Jin Jang; Chulhun Kang; Jong Hoon Kim; Jong Seung Kim


Toxicological Sciences | 2015

Enhanced Metabolizing Activity of Human ES Cell-Derived Hepatocytes Using a 3D Culture System with Repeated Exposures to Xenobiotics

Jong Hyun Kim; Yu Jin Jang; Su Yeon An; Jeongsang Son; Jaehun Lee; Gyunggyu Lee; Ji Young Park; Han Jin Park; Dong Youn Hwang; Jong Hoon Kim; Jiyou Han


한국발생생물학회 학술발표대회 | 2015

Highly purified hepatocytes derived from human pluripotent stem cells using EpCAM and indomonocarbocyanine

Ji Young Park; Soo Jin Uhm; Jiyou Han; Jong Hyun Kim; Jeong Sang Son; Yu Jin Jang; Jae Hun Lee; Gyunggyu Lee; Jong Seung Kim; Jong Hoon Kim

Collaboration


Dive into the Yu Jin Jang'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge