Network


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

Hotspot


Dive into the research topics where Heon-Seok Park is active.

Publication


Featured researches published by Heon-Seok Park.


PLOS ONE | 2013

Generation of Functional Insulin-Producing Cells from Neonatal Porcine Liver-Derived Cells by PDX1/VP16, BETA2/NeuroD and MafA

Dong-Sik Ham; Juyoung Shin; Ji-Won Kim; Heon-Seok Park; Jae-Hyoung Cho; Kun-Ho Yoon

Surrogate β-cells derived from stem cells are needed to cure type 1 diabetes, and neonatal liver cells may be an attractive alternative to stem cells for the generation of β-cells. In this study, we attempted to generate insulin-producing cells from neonatal porcine liver-derived cells using adenoviruses carrying three genes: pancreatic and duodenal homeobox factor1 (PDX1)/VP16, BETA2/NeuroD and v-maf musculo aponeurotic fibrosarcoma oncogene homolog A (MafA), which are all known to play critical roles in pancreatic development. Isolated neonatal porcine liver-derived cells were sequentially transduced with triple adenoviruses and grown in induction medium containing a high concentration of glucose, epidermal growth factors, nicotinamide and a low concentration of serum following the induction of aggregation for further maturation. We noted that the cells displayed a number of molecular characteristics of pancreatic β-cells, including expressing several transcription factors necessary for β-cell development and function. In addition, these cells synthesized and physiologically secreted insulin. Transplanting these differentiated cells into streptozotocin-induced immunodeficient diabetic mice led to the reversal of hyperglycemia, and more than 18% of the cells in the grafts expressed insulin at 6 weeks after transplantation. These data suggested that neonatal porcine liver-derived cells can be differentiated into functional insulin-producing cells under the culture conditions presented in this report and indicated that neonatal porcine liver-derived cells (NPLCs) might be useful as a potential source of cells for β-cell replacement therapy in efforts to cure type I diabetes.


Journal of Biochemistry and Molecular Biology | 2011

Both sitagliptin analogue & pioglitazone preserve the β-cell proportion in the islets with different mechanism in non-obese and obese diabetic mice

Jin-A Yeom; Eun Sook Kim; Heon-Seok Park; Dong-Sik Ham; Chenglin Sun; Ji-Won Kim; Jae-Hyoung Cho; Kun-Ho Yoon

In this study, the effects of sitagliptin analogue (SITA) or pioglitazone (PIO) treatment on glucose homeostasis and Β-cell dynamics in animal models of type 2 diabetes--Akita and db/db mice were evaluated. After 4-6 weeks of treatment, both SITA and PIO were shown to lower non-fasting glucose levels and reduced glycemic excursion in the intraperitoneal glucose tolerance test. In addition, both drugs preserved normal islet structure and the proportion of Β-cells in the islets. Compared to the controls, SITA treatment induced a higher Β-cell proliferation rate in Akita mice and a lower rate of apoptosis in db/db mice, whereas PIO treatment induced a lower rate of apoptosis in db/db mice and reduced proliferation rates in Akita mice. In conclusion, both SITA and PIO appear to exert some beneficial effects on the islet structure in addition to glycemic control via different mechanisms that involve Β-cell dynamics in Akita and db/db mice. [BMB reports 2011; 44(11): 713-718].


Transplantation | 2016

Long-term Efficacy and Biocompatibility of Encapsulated Islet Transplantation With Chitosan-Coated Alginate Capsules in Mice and Canine Models of Diabetes.

Hae Kyung Yang; Dong-Sik Ham; Heon-Seok Park; Marie Rhee; Young Hye You; Min Jung Kim; Juyoung Shin; Onyou Kim; Gilson Khang; Tae Ho Hong; Ji-Won Kim; Seung Hwan Lee; Jae-Hyoung Cho; Kun-Ho Yoon

Background Clinical application of encapsulated islet transplantation is hindered by low biocompatibility of capsules leading to pericapsular fibrosis and decreased islet viability. To improve biocompatibility, we designed a novel chitosan-coated alginate capsules and compared them to uncoated alginate capsules. Methods Alginate capsules were formed by crosslinking with BaCl2, then they were suspended in chitosan solution for 10 minutes at pH 4.5. Xenogeneic islet transplantation, using encapsulated porcine islets in 1,3-galactosyltransferase knockout mice, and allogeneic islet transplantation, using encapsulated canine islets in beagles, were performed without immunosuppressants. Results The chitosan-alginate capsules showed similar pore size, islet viability, and insulin secretory function compared to alginate capsules, in vitro. Xenogeneic transplantation of chitosan-alginate capsules demonstrated a trend toward superior graft survival (P = 0.07) with significantly less pericapsular fibrosis (cell adhesion score: 3.77 ± 0.41 vs 8.08 ± 0.05; P < 0.001) compared to that of alginate capsules up to 1 year after transplantation. Allogeneic transplantation of chitosan-alginate capsules normalized the blood glucose level up to 1 year with little evidence of pericapsular fibrotic overgrowth on graft explantation. Conclusions The efficacy and biocompatibility of chitosan-alginate capsules were demonstrated in xenogeneic and allogeneic islet transplantations using small and large animal models of diabetes. This capsule might be a potential candidate applicable in the treatment of type 1 diabetes mellitus patients, and further studies in nonhuman primates are required.


Biochemical and Biophysical Research Communications | 2009

Rosiglitazone protects the pancreatic β-cell death induced by cyclosporine A

Ji-Won Kim; Ji-Hun Yang; Heon-Seok Park; Chenglin Sun; Seung Hwan Lee; Jae-Hyoung Cho; Chul Woo Yang; Kun-Ho Yoon

The pathogenesis of post-transplant diabetes mellitus (PTDM) is thought to be partly related to the direct toxic effect of cyclosporine (CsA) on pancreatic beta-cells and the resultant decrease in insulin synthesis and secretion. Although rosiglitazone (Rosi) is an insulin sensitizer, recent data has shown that Rosi also directly protects against beta-cell dysfunction and death. This study was undertaken to clarify the effects of Rosi on CsA-induced beta-cell dysfunction and death. The deterioration in glucose tolerance caused by CsA administration was significantly improved by cotreatment with Rosi. The relative volume and absolute mass of beta-cells were significantly reduced by CsA, whereas combined treatment with Rosi had protective effects. Induction of beta-cell death and increased expression of endoplasmic reticulum (ER) stress markers (CHOP and spliced XBP-1) by CsA were rescued by Rosi. Thus, Rosi signaling directly modulates the ER stress response, promoting beta-cell adaptation and survival. Rosi might be an appropriate drug for preventing and treating CsA-induced PTDM.


Journal of Tissue Engineering and Regenerative Medicine | 2017

Antifibrotic effect of rapamycin containing polyethylene glycol-coated alginate microcapsule in islet xenotransplantation

Heon-Seok Park; Ji-Won Kim; Seung Hwan Lee; Hae Kyung Yang; Dong-Sik Ham; Chenglin Sun; Tae Ho Hong; Gilson Khang; Chung-Gyu Park; Kun-Ho Yoon

Islet microencapsulation is an attractive strategy for the minimization or avoidance of life‐long immunosuppression after transplantation. However, the clinical implementation of this technique is currently limited by incomplete biocompatibility. Thus, the aim of the present study was to demonstrate the improved biocompatibility of rapamycin‐containing polyethylene glycol (Rapa–PEG)‐coating on alginate microcapsules containing xenogeneic islets. The Rapa–PEG‐coating on the alginate layer was observed using scanning electron microscopy (SEM) and the molecular cut‐off weight of the microcapsules was approximately 70 kDa. The viabilities of the alginate‐encapsulated and Rapa–PEG‐coated alginate‐encapsulated islets were lower than the viability of the naked islets just after encapsulation, but these the differences diminished over time in culture dishes. Rapa–PEG‐coating on the alginate capsules effectively decreased the proliferation of macrophage cells compared to the non‐coating and alginate coating of xenogeneic pancreas tissues. Glucose‐stimulated insulin secretion did not significantly differ among the groups prior to transplantation. The random blood glucose levels of diabetic mice significantly improved following the transplantation of alginate‐encapsulated and Rapa–PEG‐coated alginate‐encapsulated islets, but there were no significant differences between these two groups. However, there was a significant decrease in the number of microcapsules with fibrotic cell infiltration in the Rapa–PEG‐coated alginate microcapsule group compared to the alginate microcapsule group. In conclusion, Rapa–PEG‐coating might be an effective technique with which to improve the biocompatibility of microcapsules containing xenogeneic islets. Copyright


Xenotransplantation | 2015

Successful xenotransplantation with re‐aggregated and encapsulated neonatal pig liver cells for treatment of mice with acute liver failure

Dong-Sik Ham; Min-Sang Song; Heon-Seok Park; Marie Rhee; Hae Kyung Yang; Seung Hwan Lee; Ji-Won Kim; Eun-Sun Jung; Kun-Ho Yoon

Hepatocyte transplantation is a promising therapy for acute liver failure. Cell therapy using xenogeneic sources has emerged as an alternative treatment for patients with organ failure due to the shortage of transplantable human organs. The purpose of this study was to improve the survival of mice with acute liver failure by transplanting encapsulated neonatal pig re‐aggregated liver cells (NPRLC).


Transplantation | 2010

Rapamycin suppresses the expansion and differentiation of porcine neonatal pancreas cell clusters.

Chenglin Sun; Dong-Sik Ham; Heon-Seok Park; Ji-Won Kim; Jae-Hyoung Cho; Ki-Ho Song; Ho-Young Son; Kun-Ho Yoon

Background. The role of rapamycin in pancreas stem cells remains to be clearly elucidated. Herein, we evaluated the effects of rapamycin on porcine neonatal pancreas cell clusters (NPCCs), which primarily comprised pancreatic precursors, and attempted to find an intracellular mechanism about the harmful effects of rapamycin. Methods. Porcine NPCCs were treated with rapamycin in a monolayer, and the apoptosis and proliferation were determined via caspase-3 assay and H3-thymidine uptake analysis. The expression of transcription factors was assessed via reverse-transcriptase polymerase chain reaction and Western blotting. For the in vivo study, the porcine NPCCs were transplanted into the kidney subcapsules of normal nude mice and treated with rapamycin. Results. Rapamycin treatment significantly reduced the number of &bgr; cells, glucose-stimulated insulin secretion, and the insulin contents in the monolayer-cultured porcine NPCCs. Furthermore, rapamycin treatment increased the apoptosis and inhibited the proliferation of &bgr; cells in the culture dishes. The expressions of the insulin, pancreatic and duodenal homeobox-1, and NeuroD/Beta2 genes were down-regulated via rapamycin treatment. The expression of insulin-like growth factor-II was significantly down-regulated, but the expression of Foxo1 was simultaneously inversely increased, and the translocation of Foxo1 from the cytoplasm to the nucleus was induced by rapamycin treatment. Moreover, rapamycin treatment induced a marked reduction in the relative volume and absolute mass of &bgr; cells in the porcine NPCCs grafts at 8 weeks after transplantation in the normal nude mice. Conclusions. Here, we demonstrate that rapamycin treatment suppresses the expansion and differentiation of porcine NPCCs, and the alteration of Foxo1 and insulin-like growth factor-II gene expression might be the crucial factors.


Scientific Reports | 2016

Preadipocyte factor 1 induces pancreatic ductal cell differentiation into insulin-producing cells

Marie Rhee; Seung Hwan Lee; Ji-Won Kim; Dong-Sik Ham; Heon-Seok Park; Hae Kyung Yang; Juyoung Shin; Jae-Hyoung Cho; Young-Bum Kim; Byung-Soo Youn; Hei Sook Sul; Kun-Ho Yoon

The preadipocyte factor 1 (Pref-1) is involved in the proliferation and differentiation of various precursor cells. However, the intracellular signaling pathways that control these processes and the role of Pref-1 in the pancreas remain poorly understood. Here, we showed that Pref-1 induces insulin synthesis and secretion via two independent pathways. The overexpression of Pref-1 activated MAPK signaling, which induced nucleocytoplasmic translocation of FOXO1 and PDX1 and led to the differentiation of human pancreatic ductal cells into β-like cells and an increase in insulin synthesis. Concurrently, Pref-1 activated Akt signaling and facilitated insulin secretion. A proteomics analysis identified the Rab43 GTPase-activating protein as a downstream target of Akt. A serial activation of both proteins induced various granular protein syntheses which led to enhanced glucose-stimulated insulin secretion. In a pancreatectomised diabetic animal model, exogenous Pref-1 improved glucose homeostasis by accelerating pancreatic ductal and β-cell regeneration after injury. These data establish a novel role for Pref-1, opening the possibility of applying this molecule to the treatment of diabetes.


Diabetes & Metabolism Journal | 2011

Adenoviruses Expressing PDX-1, BETA2/NeuroD and MafA Induces the Transdifferentiation of Porcine Neonatal Pancreas Cell Clusters and Adult Pig Pancreatic Cells into Beta-Cells

Young-Hye You; Dong-Sik Ham; Heon-Seok Park; Marie Rhee; Ji-Won Kim; Kun-Ho Yoon

Background A limitation in the number of insulin-producing pancreatic beta-cells is a special feature of diabetes. The identification of alternative sources for the induction of insulin-producing surrogate beta-cells is a matter of profound importance. PDX-1/VP16, BETA2/NeuroD, and MafA overexpression have been shown to influence the differentiation and proliferation of pancreatic stem cells. However, few studies have been conducted using adult animal pancreatic stem cells. Methods Adult pig pancreatic cells were prepared from the non-endocrine fraction of adult pig pancreata. Porcine neonatal pancreas cell clusters (NPCCs) were prepared from neonatal pigs aged 1-2 days. The dispersed pancreatic cells were infected with PDX-1/VP16, BETA2/NeuroD, and MafA adenoviruses. After infection, these cells were transplanted under the kidney capsules of normoglycemic nude mice. Results The adenovirus-mediated overexpression of PDX-1, BETA2/NeuroD and MafA induced insulin gene expression in NPCCs, but not in adult pig pancreatic cells. Immunocytochemistry revealed that the number of insulin-positive cells in NPCCs and adult pig pancreatic cells was approximately 2.6- and 1.1-fold greater than those in the green fluorescent protein control group, respectively. At four weeks after transplantation, the relative volume of insulin-positive cells in the grafts increased in the NPCCs, but not in the adult porcine pancreatic cells. Conclusion These data indicate that PDX-1, BETA2/NeuroD, and MafA facilitate the beta-cell differentiation of NPCCs, but not adult pig pancreatic cells. Therefore PDX-1, BETA2/NeuroD, and MafA-induced NPCCs can be considered good sources for the induction of pancreatic beta-cells, and may also have some utility in the treatment of diabetes.


Journal of Korean Medical Science | 2015

Reversal of Hypoglycemia Unawareness with a Single-donor, Marginal Dose Allogeneic Islet Transplantation in Korea: A Case Report.

Hae Kyung Yang; Dong-Sik Ham; Heon-Seok Park; Marie Rhee; Young Hye You; Min Jung Kim; Ji-Won Kim; Seung Hwan Lee; Tae Ho Hong; Byung Gil Choi; Jae-Hyoung Cho; Kun-Ho Yoon

Pancreatic islet transplantation is a physiologically advantageous and minimally invasive procedure for the treatment of type 1 diabetes mellitus. Here, we describe the first reported case of successful allogeneic islet transplantation alone, using single-donor, marginal-dose islets in a Korean patient. A 59-yr-old patient with type 1 diabetes mellitus, who suffered from recurrent severe hypoglycemia, received 4,163 islet equivalents/kg from a single brain-death donor. Isolated islets were infused intraportally without any complications. The immunosuppressive regimen was based on the Edmonton protocol, but the maintenance dosage was reduced because of mucositis and leukopenia. Although insulin independence was not achieved, the patient showed stabilized blood glucose concentration, reduced insulin dosage and reversal of hypoglycemic unawareness, even with marginal dose of islets and reduced immunosuppressant. Islet transplantation may successfully improve endogenous insulin production and glycemic stability in subjects with type 1 diabetes mellitus. Graphical Abstract

Collaboration


Dive into the Heon-Seok Park's collaboration.

Top Co-Authors

Avatar

Ji-Won Kim

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Kun-Ho Yoon

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Dong-Sik Ham

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Jae-Hyoung Cho

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Seung Hwan Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Hae Kyung Yang

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Marie Rhee

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Gilson Khang

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Ho-Young Son

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Juyoung Shin

Catholic University of Korea

View shared research outputs
Researchain Logo
Decentralizing Knowledge