Network


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

Hotspot


Dive into the research topics where Ji-Hee Shin is active.

Publication


Featured researches published by Ji-Hee Shin.


Cell Death and Disease | 2016

Human umbilical cord blood-stem cells direct macrophage polarization and block inflammasome activation to alleviate rheumatoid arthritis

Tae-Hoon Shin; Hyung-Sik Kim; Taewook Kang; Byung-Chul Lee; Hwa-Yong Lee; Yoon-Jin Kim; Ji-Hee Shin; Yoojin Seo; Soon Won Choi; Seunghee Lee; Ki-Chul Shin; Kwang-Won Seo; Kyung-Sun Kang

Rheumatoid arthritis (RA) is a long-lasting intractable autoimmune disorder, which has become a substantial public health problem. Despite widespread use of biologic drugs, there have been uncertainties in efficacy and long-term safety. Mesenchymal stem cells (MSCs) have been suggested as a promising alternative for the treatment of RA because of their immunomodulatory properties. However, the precise mechanisms of MSCs on RA-related immune cells are not fully elucidated. The aim of this study was to investigate the therapeutic potential of human umbilical cord blood-derived MSCs (hUCB-MSCs) as a new therapeutic strategy for patients with RA and to explore the mechanisms underlying hUCB-MSC-mediated immunomodulation. Mice with collagen-induced arthritis (CIA) were administered with hUCB-MSCs after the onset of disease, and therapeutic efficacy was assessed. Systemic delivery of hUCB-MSCs significantly ameliorated the severity of CIA to a similar extent observed in the etanercept-treated group. hUCB-MSCs exerted this therapeutic effect by regulating macrophage function. To verify the regulatory effects of hUCB-MSCs on macrophages, macrophages were co-cultured with hUCB-MSCs. The tumor necrosis factor (TNF)-α-mediated activation of cyclooxygenase-2 and TNF-stimulated gene/protein 6 in hUCB-MSCs polarized naive macrophages toward an M2 phenotype. In addition, hUCB-MSCs down-regulated the activation of nucleotide-binding domain and leucine-rich repeat pyrin 3 inflammasome via a paracrine loop of interleukin-1β signaling. These immune-balancing effects of hUCB-MSCs were reproducible in co-culture experiments using peripheral blood mononuclear cells from patients with active RA. hUCB-MSCs can simultaneously regulate multiple cytokine pathways in response to pro-inflammatory cytokines elevated in RA microenvironment, suggesting that treatment with hUCB-MSCs could be an attractive candidate for patients with treatment-refractory RA.


Brain Research | 2012

Region-specific changes in the immunoreactivity of SIRT1 expression in the central nervous system of SOD1G93A transgenic mice as an in vivo model of amyotrophic lateral sclerosis

Jae Chul Lee; Ji-Hee Shin; Byeong-Woo Park; Gwon Sik Kim; Jin Cheon Kim; Kyung-Sun Kang; Choong Ik Cha

SIRT1, which is a mammalian homolog of yeast nicotinamide adenine dinucleotide-dependent deacetylase silent information regulator 2 (SIR2), is the best-characterized SIRT family member. SIRT1 regulates longevity in several model organisms and is involved in several processes in mammalian cells, including cell survival, differentiation, and metabolism. In the present study, we used SOD1(G93A) mutant transgenic mice as an in vivo model of amyotrophic lateral sclerosis (ALS) and performed immunohistochemical studies, RT-PCR and western blotting analysis in order to investigate the changes of SIRT1 immunoreactivity in the central nervous system of these mice. An increased expression of SIRT1 was obvious in the cerebral cortex, hippocampal formation, thalamus and spinal cord of symptomatic SOD1(G93A) transgenic mice. In the cerebral cortex, SIRT1 immunoreactivity was significantly increased in pyramidal cells of SOD1(G93A) transgenic mice. In the hippocampal formation of these mice, SIRT1 immunoreactivity was increased in the pyramidal cells of the CA1-3 areas and in the granule cells of the dentate gyrus. In addition, SIRT1 immunoreactivity was increased in the spinal cord and thalamus of symptomatic SOD1(G93A) transgenic mice. This study, which showed increased SIRT1 in different brain regions of SOD1(G93A) transgenic mice, may provide clues to the understanding of selective neuronal loss in ALS. These findings suggest a role for SIRT1 in the motor functions in ALS but the mechanisms and functional implications of increased SIRT1 require elucidation.


Glia | 2016

Cathepsin S contributes to microglia-mediated olfactory dysfunction through the regulation of Cx3cl1–Cx3cr1 axis in a Niemann–Pick disease type C1 model

Yoojin Seo; Hyung-Sik Kim; Insung Kang; Soon Won Choi; Tae-Hoon Shin; Ji-Hee Shin; Byung-Chul Lee; Jin Young Lee; Jae-Jun Kim; Myung Geun Kook; Kyung-Sun Kang

Microglia can aggravate olfactory dysfunction by mediating neuronal death in the olfactory bulb (OB) of a murine model of Niemann–Pick disease type C1 (NPC1), a fatal neurodegenerative disorder accompanied by lipid trafficking defects. In this study, we focused on the crosstalk between neurons and microglia to elucidate the mechanisms underlying extensive microgliosis in the NPC1‐affected brain. Microglia in the OB of NPC1 mice strongly expressed CX3C chemokine receptor 1 (Cx3cr1), a specific receptor for the neural chemokine C‐X3‐C motif ligand 1 (Cx3cl1). In addition, a high level of Cx3cl1 was detected in NPC1 mouse‐derived CSF due to enhanced catalytic activity of Cathepsin S (Ctss), which is responsible for Cx3cl1 secretion. Notably, nasal delivery of Cx3cl1 neutralizing antibody or Ctss inhibitor could inhibit the Cx3cl1–Cx3cr1 interaction and support neuronal survival through the suppression of microglial activation, leading to an improvement in the olfactory function in NPC1 mice. Relevant in vitro experiments revealed that intracellular cholesterol accumulation could act as a strong inducer of abnormal Ctss activation and, in turn, stimulated the Cx3cl1–Cx3cr1 axis in microglia via p38 mitogen‐activated protein kinase signaling. Our data address the significance of Cx3cl1–Cx3cr1 interaction in the development of microglial neurotoxicity and suggest that Ctss is a key upstream regulator. Therefore, this study contributes to a better understanding of the crosstalk between neurons and microglia in the development of the neurodegeneration and provides a new perspective for the management of olfactory deficits and other microglia‐dependent neuropathies. GLIA 2016;64:2291–2305


Oncotarget | 2017

Human adipose tissue-derived mesenchymal stem cells alleviate atopic dermatitis via regulation of B lymphocyte maturation

Tae-Hoon Shin; Byung-Chul Lee; Soon Won Choi; Ji-Hee Shin; Insung Kang; Jin Young Lee; Jae-Jun Kim; Hong-Ki Lee; Jae-Eon Jung; Yong-Woon Choi; Sunghoon Lee; Jin-Sang Yoon; Jin-Sub Choi; Chi-Seung Lee; Yoojin Seo; Hyung-Sik Kim; Kyung-Sun Kang

Mesenchymal stem cell (MSC) has been applied for the therapy of allergic disorders due to its beneficial immunomodulatory abilities. However, the underlying mechanisms for therapeutic efficacy are reported to be diverse according to the source of cell isolation or the route of administration. We sought to investigate the safety and the efficacy of human adipose tissue-derived MSCs (hAT-MSCs) in mouse atopic dermatitis (AD) model and to determine the distribution of cells after intravenous administration. Murine AD model was established by multiple treatment of Dermatophagoides farinae. AD mice were intravenously infused with hAT-MSCs and monitored for clinical symptoms. The administration of hAT-MSCs reduced the gross and histological signatures of AD, as well as serum IgE level. hAT-MSCs were mostly detected in lung and heart of mice within 3 days after administration and were hardly detectable at 2 weeks. All of mice administered with hAT-MSCs survived until sacrifice and did not demonstrate any adverse events. Co-culture experiments revealed that hAT-MSCs significantly inhibited the proliferation and the maturation of B lymphocytes via cyclooxygenase (COX)-2 signaling. Moreover, mast cell (MC) degranulation was suppressed by hAT-MSC. In conclusion, the intravenous infusion of hAT-MSCs can alleviate AD through the regulation of B cell function.


Aging (Albany NY) | 2016

BMI1 inhibits senescence and enhances the immunomodulatory properties of human mesenchymal stem cells via the direct suppression of MKP-1/DUSP1

Jin Young Lee; Kyung-Rok Yu; Hyung-Sik Kim; Insung Kang; Jae-Jun Kim; Byung-Chul Lee; Soon Won Choi; Ji-Hee Shin; Yoojin Seo; Kyung-Sun Kang

For the application of mesenchymal stem cells (MSCs) as clinical therapeutics, the regulation of cellular aging is important to protect hMSCs from an age-associated decline in their function. In this study, we evaluated the effects of hypoxia on cellular senescence and the immunomodulatory abilities of hUCB-MSCs. Hypoxic-cultured hUCB-MSCs showed enhanced proliferation and had increased immunosuppressive effects on mitogen-induced mononuclear cell proliferation. We found that BMI1, a member of the polycomb repressive complex protein group, showed increased expression in hypoxic-cultured hUCB-MSCs, and the further knock-down of BMI1 in hypoxic cells induced decreased proliferative and immunomodulatory abilities in hUCB-MSCs, along with COX-2/PGE2 down-regulation. Furthermore, the expression of phosphorylated p38 MAP kinase increased in response to the over-expression of BMI1 in normoxic conditions, suggesting that BMI1 regulates the immunomodulatory properties of hUCB-MSCs via p38 MAP kinase-mediated COX-2 expression. More importantly, we identified BMI1 as a direct repressor of MAP kinase phosphatase-1 (MKP-1)/DUSP1, which suppresses p38 MAP kinase activity. In conclusion, our results demonstrate that BMI1 plays a key role in the regulation of the immunomodulatory properties of hUCB-MSCs, and we suggest that these findings might provide a strategy to enhance the functionality of hUCB-MSCs for use in therapeutic applications.


Journal of Veterinary Science | 2017

Inhibition by miR-410 facilitates direct retinal pigment epithelium differentiation of umbilical cord blood-derived mesenchymal stem cells

Soon Won Choi; Jae-Jun Kim; Min-Soo Seo; Sang-Bum Park; Tae-Hoon Shin; Ji-Hee Shin; Yoojin Seo; Hyung-Sik Kim; Kyung-Sun Kang

Retinal pigment epithelium (RPE) is a major component of the eye. This highly specialized cell type facilitates maintenance of the visual system. Because RPE loss induces an irreversible visual impairment, RPE generation techniques have recently been investigated as a potential therapeutic approach to RPE degeneration. The microRNA-based technique is a new strategy for producing RPE cells from adult stem cell sources. Previously, we identified that antisense microRNA-410 (anti-miR-410) induces RPE differentiation from amniotic epithelial stem cells. In this study, we investigated RPE differentiation from umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) via anti-miR-410 treatment. We identified miR-410 as a RPE-relevant microRNA in UCB-MSCs from among 21 putative human RPE-depleted microRNAs. Inhibition of miR-410 induces overexpression of immature and mature RPE-specific factors, including MITF, LRAT, RPE65, Bestrophin, and EMMPRIN. The RPE-induced cells were able to phagocytize microbeads. Results of our microRNA-based strategy demonstrated proof-of-principle for RPE differentiation in UCB-MSCs by using anti-miR-410 treatment without the use of additional factors or exogenous transduction.


Oncotarget | 2016

Direct cell fate conversion of human somatic stem cells into cone and rod photoreceptor-like cells by inhibition of microRNA-203

Soon Won Choi; Ji-Hee Shin; Jae-Jun Kim; Tae-Hoon Shin; Yoojin Seo; Hyung-Sik Kim; Kyung-Sun Kang

Stem cell-based photoreceptor differentiation strategies have been the recent focus of therapies for retinal degenerative diseases. Previous studies utilized embryonic stem (ES) cells and neural retina differentiation cocktails, including DKK1 and Noggin. Here, we show a novel microRNA-mediated strategy of retina differentiation from somatic stem cells, which are potential allogeneic cell sources. Human amniotic epithelial stem cells (AESCs) and umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) treated with a retina differentiation cocktail induced gene expressions of retina development-relevant genes. Furthermore, microRNA-203 (miR-203) is abundantly expressed in human AESCs and human UCB-MSCs. This miR-203 is predicted to target multiple retina development-relevant genes, particularly DKK1, CRX, RORβ, NEUROD1, NRL and THRB. The inhibition of miR-203 induced a retina differentiation of AESCs and UCB-MSCs. Moreover, successive treatments of anti-miR-203 led to the expression of both mature photoreceptor (PR) markers, rhodopsin and opsin. In addition, we determined that CRX, NRL and DKK1 are direct targets of miR-203 using a luciferase assay. Thus, the work presented here suggests that somatic stem cells can potentially differentiate into neural retina cell types when treated with anti-miR-203. They may prove to be a source of both PR subtypes for future allogeneic stem cell-based therapies of non-regenerative retina diseases.


International journal of stem cells | 2017

Direct Conversion of Human Umbilical Cord Blood into Induced Neural Stem Cells with SOX2 and HMGA2

Jae-Jun Kim; Ji-Hee Shin; Kyung-Rok Yu; Byung-Chul Lee; Insung Kang; Jin Young Lee; Da-Hyun Kim; Yoojin Seo; Hyung-Sik Kim; Soon Won Choi; Kyung-Sun Kang

Recent advances have shown the direct reprogramming of mouse and human fibroblasts into induced neural stem cells (iNSCs) without passing through an intermediate pluripotent state. Thus, direct reprogramming strategy possibly provides a safe and homogeneous cellular platform. However, the applications of iNSCs for regenerative medicine are limited by the restricted availability of cell sources. Human umbilical cord blood (hUCB) cells hold great potential in that immunotyped hUCB units can be immediately obtained from public banks. Moreover, hUCB samples do not require invasive procedures during collection or an extensive culture period prior to reprogramming. We recently reported that somatic cells can be directly converted into iNSCs with high efficiency and a short turnaround time. Here, we describe the detailed method for the generation of iNSCs derived from hUCB (hUCB iNSCs) using the lineage-specific transcription factors SOX2 and HMGA2. The protocol for deriving iNSC-like colonies takes 1~2 weeks and establishment of homogenous hUCB iNSCs takes additional 2 weeks. Established hUCB iNSCs are clonally expandable and multipotent producing neurons and glia. Our study provides an accessible method for generating hUCB iNSCs, contributing development of in vitro neuropathological model systems.


Oncotarget | 2017

Generation of patient specific human neural stem cells from Niemann-Pick disease type C patient-derived fibroblasts

Eun-Ah Sung; Kyung-Rok Yu; Ji-Hee Shin; Yoojin Seo; Hyung-Sik Kim; Myung Guen Koog; Insung Kang; Jae-Jun Kim; Byung-Chul Lee; Tae-Hoon Shin; Jin Young Lee; Seunghee Lee; Taewook Kang; Soon Won Choi; Kyung-Sun Kang

Niemann-Pick disease type C (NPC) is a neurodegenerative and lysosomal lipid storage disorder, characterized by the abnormal accumulation of unesterified cholesterol and glycolipids, which is caused by mutations in the NPC1 genes. Here, we report the generation of human induced neural stem cells from NPC patient-derived fibroblasts (NPC-iNSCs) using only two reprogramming factors SOX2 and HMGA2 without going through the pluripotent state. NPC-iNSCs were stably expandable and differentiated into neurons, astrocytes, and oligodendrocytes. However, NPC-iNSCs displayed defects in self-renewal and neuronal differentiation accompanied by cholesterol accumulation, suggesting that NPC-iNSCs retain the main features of NPC. This study revealed that the cholesterol accumulation and the impairments in self-renewal and neuronal differentiation in NPC-iNSCs were significantly improved by valproic acid. Additionally, we demonstrated that the inhibition of cholesterol transportation by U18666A in WT-iNSCs mimicked the impaired self-renewal and neuronal differentiation of NPC-iNSCs, indicating that the regulation of cholesterol homeostasis is a crucial determinant for the neurodegenerative features of NPC. Taken together, these findings suggest that NPC-iNSCs can serve as an unlimited source of neural cells for pathological study or drug screening in a patient specific manner. Furthermore, this direct conversion technology might be extensively applicable for other human neurodegenerative diseases.


Cell Transplantation | 2018

Single-Factor SOX2 Mediates Direct Neural Reprogramming of Human Mesenchymal Stem Cells via Transfection of In Vitro Transcribed mRNA:

Boeun Kim; Soon Won Choi; Ji-Hee Shin; Jae-Jun Kim; Insung Kang; Byung-Chul Lee; Jin Young Lee; Myoung Geun Kook; Kyung-Sun Kang

Neural stem cells (NSCs) are a prominent cell source for understanding neural pathogenesis and for developing therapeutic applications to treat neurodegenerative disease because of their regenerative capacity and multipotency. Recently, a variety of cellular reprogramming technologies have been developed to facilitate in vitro generation of NSCs, called induced NSCs (iNSCs). However, the genetic safety aspects of established virus-based reprogramming methods have been considered, and non-integrating reprogramming methods have been developed. Reprogramming with in vitro transcribed (IVT) mRNA is one of the genetically safe reprogramming methods because exogenous mRNA temporally exists in the cell and is not integrated into the chromosome. Here, we successfully generated expandable iNSCs from human umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) via transfection with IVT mRNA encoding SOX2 (SOX2 mRNA) with properly optimized conditions. We confirmed that generated human UCB-MSC-derived iNSCs (UM-iNSCs) possess characteristics of NSCs, including multipotency and self-renewal capacity. Additionally, we transfected human dermal fibroblasts (HDFs) with SOX2 mRNA. Compared with human embryonic stem cell-derived NSCs, HDFs transfected with SOX2 mRNA exhibited neural reprogramming with similar morphologies and NSC-enriched mRNA levels, but they showed limited proliferation ability. Our results demonstrated that human UCB-MSCs can be used for direct reprogramming into NSCs through transfection with IVT mRNA encoding a single factor, which provides an integration-free reprogramming tool for future therapeutic application.

Collaboration


Dive into the Ji-Hee Shin's collaboration.

Top Co-Authors

Avatar

Kyung-Sun Kang

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Soon Won Choi

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Yoojin Seo

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Hyung-Sik Kim

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Jae-Jun Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Tae-Hoon Shin

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Jin Young Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Insung Kang

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Byung-Chul Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Kyung-Rok Yu

Seoul National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge