Network


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

Hotspot


Dive into the research topics where Hye-Myung Ryu is active.

Publication


Featured researches published by Hye-Myung Ryu.


Nephrology Dialysis Transplantation | 2010

Impact of systemic and local peritoneal inflammation on peritoneal solute transport rate in new peritoneal dialysis patients: a 1-year prospective study

Ji-Hyung Cho; In-Kyong Hur; Chan-Duck Kim; Sun-Hee Park; Hye-Myung Ryu; Ju-Min Yook; Ji-Young Choi; Hee-Jung Choi; Hyuk-Joon Choi; Jong-Won Park; Jun-Young Do; Yong-Lim Kim

BACKGROUND The association between peritoneal solute transport rates (PSTRs) and inflammatory markers in patients on peritoneal dialysis (PD) is still under investigation. We aimed to elucidate their relationship during the first year on PD. METHODS We performed a prospective observational study with 187 incident PD patients who were treated with either biocompatible solution (BCS) or conventional solution (CS). Peritoneal dialysate effluent (PDE) and blood samples for the markers and the calculation of mass transfer area coefficient of creatinine (MTAC) were performed at 1, 6 and 12 months after commencing PD. RESULTS Of the 187 enrolled patients, 110 completed a 1-year study protocol. All PDE markers [interleukin-6 (IL-6), transforming growth factor-beta (TGF-beta), TGF-beta-induced gene-h3 (beta ig-h3), vascular endothelial growth factor (VEGF)] except CA125 increased over time, whereas PSTRs, high-sensitivity C-reactive protein (hs-CRP) and serum IL-6 levels did not change. Serum albumin and log PDE appearance rates (ARs) of IL-6, TGF-beta and CA125 predicted MTAC. The Delta value (12-month minus 1-month) of PDE AR of IL-6 was correlated with those of all other PDE markers. Both 12-month IL-6 and Delta IL-6 ARs in PDE were highest in the upper Delta MTAC tertile. PSTRs in the CS group, unlike BCS, had a tendency to increase over time, demonstrating a time-by-group interaction. Solution type and MTAC were not associated with patient and technique survival. CONCLUSIONS The change in PSTR during the first year of PD is related to PDE IL-6 AR, which may represent intraperitoneal inflammation; however, there does not seem to be a close association between PSTR and the degree of systemic inflammation.


American Journal of Nephrology | 2010

Impact of low glucose degradation product bicarbonate/lactate-buffered dialysis solution on the epithelial-mesenchymal transition of peritoneum.

Eun-Joo Oh; Hye-Myung Ryu; Soon-Youn Choi; Ju-Min Yook; Chan-Duck Kim; Sun-Hee Park; Ho-Young Chung; In-San Kim; Mina Yu; Duk-Hee Kang; Yong-Lim Kim

Background: Epithelial-mesenchymal transition (EMT) is important in the development of peritoneal fibrosis. Glucose degradation products (GDPs) may induce EMT in human peritoneal mesothelial cells (HPMCs). Methods: The effects of individual GDPs and GDPs derived from peritoneal dialysis fluid (PDF) in both HPMCs and peritoneal membranes were evaluated. EMT was assessed with α-smooth muscle actin (α-SMA) and E-cadherin. Results: In vitro, α-SMA protein and mRNA levels increased in the presence of the GDPs (formaldehyde, glyoxal, methylglyoxal, and 3-deoxyglucosone), and E-cadherin decreased. Changes in the EMT markers were most prominent after exposure to 3-deoxyglucosone. Changes in both α-SMA and E-cadherin protein levels were less with low (L)-GDP bicarbonate/lactate-buffered PDF compared to high (H)-GDP PDF. In the rat model after 8 weeks’ PDF infusion, the α-SMA/E-cadherin mRNA ratio increased in the H-GDP group compared with the L-GDP group (p < 0.05). The peritoneum in the H-GDP group tended to be thicker (p = 0.052) and had more blood vessels than that in the L-GDP group (p < 0.05). Tissue staining for TGF-β1 decreased in the L-GDP group. Dual-stained cytokeratin and α-SMA-positive myofibroblasts in the submesothelial layer were more prominent in the H-GDP group. Conclusion: GDPs found in PDF induce EMT of HPMCs, which is associated with peritoneal fibrosis and vascularization. Conversely, L-GDP PDF reduces EMT and peritoneal fibrosis.


Transplantation | 2008

Association of G-137C IL-18 promoter polymorphism with acute allograft rejection in renal transplant recipients.

Chan-Duck Kim; Hye-Myung Ryu; Ji-Young Choi; Hee-Jeong Choi; Hyuk-Joon Choi; Ji-Hyung Cho; Sun-Hee Park; Dong Il Won; Yong-Lim Kim

Background. Interleukin 18 (IL-18) is a potent proinflammatory cytokine, which is postulated to play a role in mechanism of renal allograft rejection and strongly induces interferon (IFN)-&ggr; production. The aim of this study was to investigate the association of the G–137C IL-18 promoter polymorphism with acute allograft rejection in renal transplant recipients (RTRs). Methods. A total of 226 RTRs and 148 controls were recruited for association analysis. Genotyping was performed by using a real-time polymerase chain reaction. Patients were separated into two groups, the acute rejection (AR) (n=37) or the no AR group (n=189), depending on their history of AR episodes. IL-18 and IFN-&ggr; serum levels of 73 randomly selected RTRs were measured by ELISA. Results. No significant differences in genotype and allele frequencies were observed between the RTRs and the controls. Significant differences in genotype frequency and allele frequency between the AR and no AR group were observed. The frequency of the –137GG genotype was significantly increased in patients with AR (P=0.015, odds ratio=3.653). Serum levels of IL-18 and IFN-&ggr; were significantly elevated in the AR group with compared with the no AR group. In the AR group, patients with the –137GG genotype had significantly higher IL-18 serum levels compared to other genotypes. Conclusion. These data demonstrate that the –137GG genotype of the IL-18 gene, encoding higher IL-18 production, seems to be associated with AR and may be a useful marker of AR risk in RTRs.


American Journal of Pathology | 2012

Aquaporin 3 expression is up-regulated by TGF-β1 in rat peritoneal mesothelial cells and plays a role in wound healing.

Hye-Myung Ryu; Eun-Joo Oh; Sun-Hee Park; Chan-Duck Kim; Ji-Young Choi; Jang-Hee Cho; In-San Kim; Tae-Hwan Kwon; Ho-Young Chung; Min Yoo; Yong-Lim Kim

Aquaporin 3 (AQP3) is expressed in many tissues including the peritoneum and kidney. In cultured mesothelial cells, glucose up-regulates AQP3, which may be important for water transport through the peritoneal membrane. However, there has been no research into the role of AQP3 in human peritoneal mesothelial cell (HPMC) migration or peritoneal fibrosis. We investigated the effects of transforming growth factor-β1 (TGF-β1) on AQP3 expression in HPMCs. We also investigated the role of AQP3 in the peritoneal wound healing process in rats. Chronic exposure to glucose-containing solution increased peritoneal myofibroblasts, with TGF-β1 and AQP3 expression in a model of long-term peritoneal dialysis. In vitro, TGF-β1 induced AQP3 expression in HPMCs. AQP3 knockdown by small-interfering RNA inhibited TGF-β1-induced AQP3 and α-smooth muscle actin expression and also slowed HPMC migration. AQP3 overexpression induced faster migration of HPMCs. Treatment with an extracellular signal-regulated kinase inhibitor and p38 kinase inhibitor attenuated TGF-β1-induced AQP3 expression in HPMCs. These data suggest that TGF-β1 induces AQP3 and that AQP3 has a critical role in TGF-β-induced HPMC migration. These findings provide evidence of a novel role for AQP3 in peritoneal fibrosis and wound healing. The effect of TGF-β1 on AQP3 expression in HPMCs is mediated, at least in part, by ERK and p38 signaling.


PLOS ONE | 2016

Alpha1-Antitrypsin Attenuates Renal Fibrosis by Inhibiting TGF-β1-Induced Epithelial Mesenchymal Transition.

Jang-Hee Cho; Hye-Myung Ryu; Eun-Joo Oh; Ju-Min Yook; Ji-Sun Ahn; Hee-Yeon Jung; Ji-Young Choi; Sun Hee Park; Yong-Lim Kim; Ihm Soo Kwak; Chan-Duck Kim

Alpha1-antitrypsin (AAT) exerts its anti-inflammatory effect through regulating the activity of serine proteinases. This study evaluated the inhibitory effects of AAT against the transforming growth factor (TGF)-β1 induced epithelial-to-mesenchymal transition (EMT) in unilateral ureter obstruction (UUO) mice and Madin-Darby canine kidney (MDCK) cells. C57BL/6 mice with induced UUO were injected intraperitoneally with AAT (80 mg/Kg) or vehicle for 7 days. MDCK cells were treated with TGF-β1 (2 ng/mL) for 48 hours to induce EMT, and co-treated with AAT (10 mg/mL) to inhibit the EMT. Masson’s trichrome and Sirius red staining was used to estimate the extent of renal fibrosis in UUO mice. The expression of alpha-smooth muscle actin (α-SMA), vimentin, fibronectin, collagen I, and E-cadherin in MDCK cells and kidney tissue were evaluated. Masson’s and Sirius red staining revealed that the area of renal fibrosis was significantly smaller in AAT treated UUO group compared with that of UUO and vehicle treated UUO groups. AAT treatment attenuated upregulation of Smad2/3 phosphorylation in UUO mouse model. Co-treatment of MDCK cells with TGF-β1 and AAT significantly attenuated the changes in the expression of α-SMA, vimentin, fibronectin, collagen I, and E-cadherin. AAT also decreased the phosphorylated Smad3 expression and the phosphorylated Smad3/Smad3 ratio in MDCK cells. AAT treatment inhibited EMT induced by TGF-β1 in MDCK cells and attenuated renal fibrosis in the UUO mouse model. The results of this work suggest that AAT could inhibit the process of EMT through the suppression of TGF-β/Smad3 signaling.


Nephrology Dialysis Transplantation | 2012

Are ex vivo mesothelial cells representative of the in vivo transition from epithelial-to-mesenchymal cells in peritoneal membrane?

Ji-Hyung Cho; Jun-Young Do; Eun-Joo Oh; Hye-Myung Ryu; So-Young Park; San-Ok Kim; Seung-Hyea Hyun; Hye-Jin Seo; Gun-Hyun Kim; Ji-Young Choi; Chan-Duck Kim; Sun-Hee Park; Yong-Lim Kim

BACKGROUND We investigated whether ex vivo mesothelial cells found in peritoneal dialysis (PD) effluents were representative of the in vivo epithelial-to-mesenchymal transition (EMT) in peritoneal membrane. METHODS Thirty-six male Sprague-Dawley rats were equally divided into three groups: Group C (control), no PD; Group D, infused with 4.25% Dianeal and Group P, infused with 4.25% Physioneal. PD infusions (25 mL) were given twice daily for 8 weeks. The in vivo study included morphometric analyses performed on the peritoneal membranes of tissue specimens obtained at the end of the study. The ex vivo study included peritoneal mesothelial cells collected from PD effluent and cultured to confluence. Cells were scored with light microscopy. RESULTS PD for 8 weeks induced significant EMT. The in vivo expression of EMT markers (α-smooth muscle actin:E-cadherin ratio, matrix metalloproteinase-2 and Snail) was higher in Group D than in Group P. However, ex vivo EMT marker expression was similar in cells derived from Groups D and P. A significant correlation was observed among in vivo EMT markers. Moreover, the ex vivo cell score increased with time on PD. However, changes in the ex vivo cell score did not correlated with changes in the in vivo EMT marker expression. Furthermore, we found no correlation between ex vivo and in vivo cells in the expression of EMT markers. CONCLUSIONS In this animal study, ex vivo findings did not reflect the in vivo EMT changes in the peritoneum. It may be necessary to improve the current methodology for ex vivo studies.


Scientific Reports | 2017

Metformin ameliorates the Phenotype Transition of Peritoneal Mesothelial Cells and Peritoneal Fibrosis via a modulation of Oxidative Stress

Hyun-Soo Shin; Jiyeon Ko; Dal-Ah Kim; Eun-Sun Ryu; Hye-Myung Ryu; Sun Hee Park; Yong-Lim Kim; Eok-Soo Oh; Duk-Hee Kang

Phenotype transition of peritoneum is an early mechanism of peritoneal fibrosis. Metformin, 5′-adenosine monophosphate-activated protein kinase (AMPK) activator, has recently received a new attention due to its preventive effect on organ fibrosis and cancer metastasis by inhibiting epithelial-to-mesenchymal transition (EMT). We investigated the effect of metformin on EMT of human peritoneal mesothelial cells (HPMC) and animal model of peritoneal dialysis (PD). TGF-β1-induced EMT in HPMC was ameliorated by metformin. Metformin alleviated NAPDH oxidase- and mitochondria-mediated ROS production with an increase in superoxide dismutase (SOD) activity and SOD2 expression. Metformin inhibited the activation of Smad2/3 and MAPK, GSK-3β phosphorylation, nuclear translocalization of β-catenin and Snail in HPMCs. Effect of metformin on TGF-β1-induced EMT was ameliorated by either AMPK inhibitor or AMPK gene silencing. Another AMPK agonist, 5-amino-1-β-D-ribofuranosyl-imidazole-4-carboxamide partially blocked TGF-β1-induced EMT. In animal model of PD, intraperitoneal metformin decreased the peritoneal thickness and EMT with an increase in ratio of reduced to oxidized glutathione and the expression of SOD whereas it decreased the expression of nitrotyrosine and 8-hydroxy-2′-deoxyguanosine. Therefore, a modulation of AMPK in peritoneum can be a novel tool to prevent peritoneal fibrosis by providing a favorable oxidant/anti-oxidant milieu in peritoneal cavity and ameliorating phenotype transition of peritoneal mesothelial cells.


Biochemical and Biophysical Research Communications | 2017

Hypoxanthine causes endothelial dysfunction through oxidative stress-induced apoptosis

You-Jin Kim; Hye-Myung Ryu; Ji-Young Choi; Jang-Hee Cho; Chan-Duck Kim; Sun-Hee Park; Yong-Lim Kim

Endothelial cell injury and dysfunction caused by reactive oxygen species (ROS) are implicated in the pathogenesis of vascular diseases. ROS are generated and hypoxanthine is degraded by xanthine oxidase. Smoking and alcohol consumption are associated with an increased level of hypoxanthine. We aimed to study the direct role of hypoxanthine in endothelial dysfunction in human umbilical vascular endothelial cells (HUVECs). Hypoxanthine induced cell death and production of ROS. Furthermore, hypoxanthine induced apoptosis through regulation of protein expression related to apoptosis. When cells were pretreated with N-acetylcysteine or a pancaspase inhibitor (Z-VAD-fmk) and stimulated with hypoxanthine, Z-VAD-fmk and N-acetylcysteine prevented hypoxanthine-induced apoptosis by inhibiting the ROS production and caspase pathway. Thus, an increased extracellular concentration of hypoxanthine induces endothelial dysfunction through ROS production and regulates expression of apoptosis-related proteins in HUVECs. These effects are expected to be associated with some vascular diseases.


Journal of Cellular and Molecular Medicine | 2016

Hypoxanthine induces cholesterol accumulation and incites atherosclerosis in apolipoprotein E-deficient mice and cells.

Hye-Myung Ryu; You-Jin Kim; Eun-Joo Oh; Se-Hyun Oh; Ji-Young Choi; Jang-Hee Cho; Chan-Duck Kim; Sun-Hee Park; Yong-Lim Kim

Reactive oxygen species (ROS) generation during purine metabolism is associated with xanthine oxidase and uric acid. However, the direct effect of hypoxanthine on ROS generation and atherosclerosis has not been evaluated. Smoking and heavy drinking are associated with elevated levels of hypoxanthine. In this study, we investigated the role of hypoxanthine on cholesterol synthesis and atherosclerosis development, particularly in apolipoprotein E (APOE)‐deficient mice. The effect of hypoxanthine on the regulation of cholesterol synthesis and atherosclerosis were evaluated in Apoe knockout (KO) mice and cultured HepG2 cells. Hypoxanthine markedly increased serum cholesterol levels and the atherosclerotic plaque area in Apoe KO mice. In HepG2 cells, hypoxanthine increased intracellular ROS production. Hypoxanthine increased cholesterol accumulation and decreased APOE and ATP‐binding cassette transporter A1 (ABCA1) mRNA and protein expression in HepG2 cells. Furthermore, H2O2 also increased cholesterol accumulation and decreased APOE and ABCA1 expression. This effect was partially reversible by treatment with the antioxidant N‐acetyl cysteine and allopurinol. Hypoxanthine and APOE knockdown using APOE‐siRNA synergistically induced cholesterol accumulation and reduced APOE and ABCA1 expression. Hypoxanthine induces cholesterol accumulation in hepatic cells through alterations in enzymes that control lipid transport and induces atherosclerosis in APOE‐deficient cells and mice. These effects are partially mediated through ROS produced in response to hypoxanthine.


Clinica Chimica Acta | 2016

Changes in serum metabolites with the stage of chronic kidney disease: Comparison of diabetes and non-diabetes.

Jueun Lee; Ji-Young Choi; Yong-Kook Kwon; Doohae Lee; Hee-Yeon Jung; Hye-Myung Ryu; Jang-Hee Cho; Do Hyun Ryu; Yong-Lim Kim; Geum-Sook Hwang

BACKGROUND The renal dysfunction of chronic kidney disease (CKD) alters serum metabolite levels, but it is not clear how diabetes mellitus (DM) affects the metabolic changes in CKD. METHODS Serum metabolites from pre-dialysis CKD patients (n=291) with or without DM and from healthy controls (n=56) was measured using nuclear magnetic resonance. RESULTS Initial principal components analysis and partial least squares-discriminant analysis score plots segregated the CKD patients according to CKD stage and separated DM from non-DM patients. In the CKD patients, associations were seen with clinical characteristics, hyperglycemia, altered amino acid metabolism, accumulated uremic toxins, and dyslipidemia. Of interest, diabetes more strongly affected the metabolic signature during early stage CKD. Furthermore, serum metabolite profiles were successfully applied to the PLS regression model to predict the estimated glomerular filtration rate. The R(2) values from the PLS models for CKD patients with DM were higher than those for CKD without DM. CONCLUSIONS Metabolomics is useful clinically for providing a metabolic signature that is associated with the CKD phenotype and diabetes more seriously affects patients with early stage CKD compared to those with advanced CKD.

Collaboration


Dive into the Hye-Myung Ryu's collaboration.

Top Co-Authors

Avatar

Yong-Lim Kim

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Chan-Duck Kim

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Ji-Young Choi

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Sun-Hee Park

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Eun-Joo Oh

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Jang-Hee Cho

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Sun Hee Park

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Soon-Youn Choi

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Ji-Hyung Cho

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Ho-Young Chung

Kyungpook National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge