Network


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

Hotspot


Dive into the research topics where Hwan D. Kim is active.

Publication


Featured researches published by Hwan D. Kim.


Drug Delivery and Translational Research | 2016

Riboflavin-induced photo-crosslinking of collagen hydrogel and its application in meniscus tissue engineering

Jiseung Heo; Rachel H. Koh; Whuisu Shim; Hwan D. Kim; Hyun-Gu Yim; Nathaniel S. Hwang

A meniscus tear is a common knee injury, but its regeneration remains a clinical challenge. Recently, collagen-based scaffolds have been applied in meniscus tissue engineering. Despite its prevalence, application of natural collagen scaffold in clinical setting is limited due to its extremely low stiffness and rapid degradation. The purpose of the present study was to increase the mechanical properties and delay degradation rate of a collagen-based scaffold by photo-crosslinking using riboflavin (RF) and UV exposure. RF is a biocompatible vitamin B2 that showed minimal cytotoxicity compared to conventionally utilized photo-initiator. Furthermore, collagen photo-crosslinking with RF improved mechanical properties and delayed enzyme-triggered degradation of collagen scaffolds. RF-induced photo-crosslinked collagen scaffolds encapsulated with fibrochondrocytes resulted in reduced scaffold contraction and enhanced gene expression levels for the collagen II and aggrecan. Additionally, hyaluronic acid (HA) incorporation into photo-crosslinked collagen scaffold showed an increase in its retention. Based on these results, we demonstrate that photo-crosslinked collagen-HA hydrogels can be potentially applied in the scaffold-based meniscus tissue engineering.


Advanced Healthcare Materials | 2016

In Vitro and In Vivo Evaluation of Whitlockite Biocompatibility: Comparative Study with Hydroxyapatite and β-Tricalcium Phosphate

Hae Lin Jang; Guang Bin Zheng; Jungha Park; Hwan D. Kim; Hae-Ri Baek; Hye Kyoung Lee; Keunho Lee; Heung Nam Han; Choon-Ki Lee; Nathaniel S. Hwang; Jae Hyup Lee; Ki Tae Nam

Biomimicking ceramics have been developed to induce efficient recovery of damaged hard tissues. Among them, calcium phosphate-based bioceramics have been the most widely used because of their similar composition with human hard tissue and excellent biocompatibilities. However, the incomplete understanding of entire inorganic phases in natural bone has limited the recreation of complete bone compositions. In this work, broad biomedical evaluation of whitlockite (WH: Ca18Mg2(HPO4)2(PO4)12), which is the secondary inorganic phase in bone, is conducted to better understand human hard tissue and to seek potential application as a biomaterial. Based on the recently developed gram-scale method for synthesizing WH nanoparticles, the properties of WH as a material for cellular scaffolding and bone implants are assessed and compared to those of hydroxyapatite (HAP: Ca10(PO4)6(OH)2) and β-tricalcium phosphate (β-TCP: β-Ca3(PO4)2). WH-reinforced composite scaffolds facilitate bone-specific differentiation compared to HAP-reinforced composite scaffolds. Additionally, WH implants induce similar or better bone regeneration in calvarial defects in a rat model compared to HAP and β-TCP implants, with intermediate resorbability. New findings of the properties of WH that distinguish it from HAP and β-TCP are significant in understanding human hard tissue, mimicking bone tissue at the nanoscale and designing functional bioceramics.


Advanced Healthcare Materials | 2015

Physical Stimuli‐Induced Chondrogenic Differentiation of Mesenchymal Stem Cells Using Magnetic Nanoparticles

Boram Son; Hwan D. Kim; Min Soo Kim; Jeong Ah Kim; Jinkyu Lee; Heungsoo Shin; Nathaniel S. Hwang; Tai Hyun Park

Chondrogenic commitments of mesenchymal stem cells (MSCs) require 3D cellular organization. Furthermore, recent progresses in bioreactor technology have contributed to the development of various biophysical stimulation platforms for efficient cartilage tissue formation. Here, an approach is reported to drive 3D cellular organization and enhance chondrogenic commitment of bone-marrow-derived human mesenchymal stem cells (BM-hMSCs) via magnetic nanoparticle (MNP)-mediated physical stimuli. MNPs isolated from Magnetospirillum sp. AMB-1 are endocytosed by the BM-hMSCs in a highly efficient manner. MNPs-incorporated BM-hMSCs are pelleted and then subjected to static magnetic field and/or magnet-derived shear stress. Magnetic-based stimuli enhance level of sulfated glycosaminoglycan (sGAG) and collagen synthesis, and facilitate the chondrogenic differentiation of BM-hMSCs. In addition, both static magnetic field and magnet-derived shear stress applied for the chondrogenic differentiation of BM-hMSCs do not show increament of hypertrophic differentiation. This MNP-mediated physical stimulation platform demonstrates a promising strategy for efficient cartilage tissue engineering.


International Journal of Biological Macromolecules | 2016

Extracellular matrix-based cryogels for cartilage tissue engineering

Min-Eui Han; Su-Hwan Kim; Hwan D. Kim; Hyun-Gu Yim; Sidi A. Bencherif; Tae-Il Kim; Nathaniel S. Hwang

In this study, we investigated various highly porous extracellular matrix (ECM)-based cryogels for cartilage tissue engineering. For the fabrication of ECM-based cryogels, either methacrylated chondroitin sulfate (MeCS) or methacrylated hyaluronic acid (MeHA) were cross-linked along with poly (ethylene glycol) diacrylates (PEGDA) via free radical polymerization under freezing conditions. This procedure induces ice crystallization (used as a porogen) prior polymer crosslinking in which, after complete cryopolymerization, a thawing process transforms the ice crystals into a unique interconnected macroporous structure within ECM-cryogels. The developed ECM-cryogels exhibited an average macroporosity of 75% and supported the infiltration of chondrocyteds. When rabbit chondrocytes were cultured on ECM-cryogels, MeCS-based cryogels stimulated aggrecan gene expression and GAG accumulation, whereas MeHA-based cryogels stimulated type II collagen gene expression and collagen accumulation. These results demonstrate that design of ECM-based cryogels can play an important role in promoting specific ECM proteins secretion for cartilage tissue engineering.


Advanced Healthcare Materials | 2017

Biomimetic Materials and Fabrication Approaches for Bone Tissue Engineering.

Hwan D. Kim; Sivashanmugam Amirthalingam; Seung Hyun Kim; Seung-Hun Lee; Jayakumar Rangasamy; Nathaniel S. Hwang

Various strategies have been explored to overcome critically sized bone defects via bone tissue engineering approaches that incorporate biomimetic scaffolds. Biomimetic scaffolds may provide a novel platform for phenotypically stable tissue formation and stem cell differentiation. In recent years, osteoinductive and inorganic biomimetic scaffold materials have been optimized to offer an osteo-friendly microenvironment for the osteogenic commitment of stem cells. Furthermore, scaffold structures with a microarchitecture design similar to native bone tissue are necessary for successful bone tissue regeneration. For this reason, various methods for fabricating 3D porous structures have been developed. Innovative techniques, such as 3D printing methods, are currently being utilized for optimal host stem cell infiltration, vascularization, nutrient transfer, and stem cell differentiation. In this progress report, biomimetic materials and fabrication approaches that are currently being utilized for biomimetic scaffold design are reviewed.


ACS Applied Materials & Interfaces | 2017

Chondroitin Sulfate-Based Biomineralizing Surface Hydrogels for Bone Tissue Engineering

Hwan D. Kim; Eunjee Lee; Young-Hyeon An; Seung Hyun Kim; Seung-Hun Lee; Seung Jung Yu; Hae Lin Jang; Ki Tae Nam; Sung Gap Im; Nathaniel S. Hwang

Chondroitin sulfate (CS) is the major component of glycosaminoglycan in connective tissue. In this study, we fabricated methacrylated PEGDA/CS-based hydrogels with varying CS concentration (0, 1, 5, and 10%) and investigated them as biomineralizing three-dimensional scaffolds for charged ion binding and depositions. Due to its negative charge from the sulfate group, CS exhibited an osteogenically favorable microenvironment by binding charged ions such as calcium and phosphate. Particularly, ion binding and distribution within negatively charged hydrogel was dependent on CS concentration. Furthermore, CS dependent biomineralizing microenvironment induced osteogenic differentiation of human tonsil-derived mesenchymal stem cells in vitro. Finally, when we transplanted PEGDA/CS-based hydrogel into a critical sized cranial defect model for 8 weeks, 10% CS hydrogel induced effective bone formation with highest bone mineral density. This PEGDA/CS-based biomineralizing hydrogel platform can be utilized for in situ bone formation in addition to being an investigational tool for in vivo bone mineralization and resorption mechanisms.


Acta Biomaterialia | 2015

Injectable PLGA microspheres encapsulating WKYMVM peptide for neovascularization.

Young Hwan Choi; Soon Chul Heo; Yang Woo Kwon; Hwan D. Kim; Seung Hyun Kim; Il Ho Jang; Jae Ho Kim; Nathaniel S. Hwang

UNLABELLED Formyl peptide receptor-2 (FPR-2) is expressed in various cell types, such as phagocytes, fibroblasts, and endothelial cells. FPR-2 has been reported to play a significant role in inflammation and angiogenic response, and synthetic WKYMVm peptide has been identified as a novel peptide agonist for the FPR-2. In this study, we demonstrate that WKYMVm peptides stimulate the angiogenic potential of outgrowth endothelial cells (OECs). Upon WKYMVm peptide exposure, migration and proliferation of OECs were stimulated. WKYMVm effectively stimulated angiogenesis in tube formation assay and aortic ring assay. Furthermore, we fabricated injectable poly (lactide-co-glycolide) (PLGA) microspheres encapsulating WKYMVm peptides, which showed sustained release of cargo molecule. When WKYMVm peptide encapsulated microspheres were injected into the hind limb ischemia model, a single injection of microspheres was as effective as multiple injections of WKYMVm peptide in restoring blood flow from ischemic injury and promoting capillary growth. These results demonstrate that sustained release of WKYMVm peptide from microspheres in the application to ischemic hind limb extended angiogenic stimulation. STATEMENT OF SIGNIFICANCE Formyl peptide receptor (FPR) has been reported to play an important role in inflammation and angiogenic response. A synthetic WKYMVm peptide has been identified as a novel peptide activating the FPR-2 that is expressed in a various cell types, such as phagocytes, fibroblasts, and endothelial cells. In this manuscript we explored a unique property of high-affinity ligand for formyl peptide receptors-2 (FPR-2) (i.e., WKYMVm). WKYMVm-induced activation of FPR2 has been reported to be crucial in host defense and inflammation by activation of phagocytes, monocytes, and lymphocytes. In this study, highlight the efficacy of WKYMVm peptides role in inducing neovascularization in vivo hind limb ischemia model when the peptide was released from injected PLGA microspheres in sustained manner. Our results demonstrate that sustained release of WKYMVm peptide from microspheres have extended angiogenic stimulation capacity.


International Journal of Biological Macromolecules | 2017

Enzyme-mediated tissue adhesive hydrogels for meniscus repair

Su-Hwan Kim; Young-Hyeon An; Hwan D. Kim; Kyung Min Kim; Sanghyuk Lee; Hyun-Gu Yim; Byung-Gee Kim; Nathaniel S. Hwang

Meniscus tissues have limited regenerative capacity once damaged. The treatment options for the meniscus tissue regeneration have been limited to arthroscopic meniscectomy or surgical interventions. The injectable hydrogels based system would provide an alternative to the conventional meniscus therapy by providing a minimally invasive treatment alternative. Here we utilized enzyme-based approaches to fabricate tissue adhesive hydrogels for meniscus repair. Tyramine (TA) conjugated hyaluronic acid (TA-HA) and gelatin are susceptible to tyrosinase (TYR)-mediated crosslinking in vitro and in vivo. Importantly, mechanical properties and degradation kinetics are modulated by the TA substitution and TYR concentrations. In addition, TYR -mediated crosslinking displayed tissue-adhesive properties. Furthermore, fibrochondrocyte-laden and TYR-crosslinked hydrogels demonstrated strong biocompatibility and resulted in enhancement of cartilage-specific gene expression and matrix synthesis. Overall, this represents a potential application of enzyme-mediated crosslinking hydrogels for meniscus tissue engineering.


Acta Biomaterialia | 2016

High throughput approaches for controlled stem cell differentiation.

Hwan D. Kim; Eunjee Lee; Young Hwan Choi; Young Hyeon An; Rachel H. Koh; Seung Hyun Kim; Nathaniel S. Hwang

UNLABELLED Stem cells have unique ability to undergo self-renewal indefinitely in culture and potential to differentiate into almost all cell types in the human body. However, the developing a method for efficiently differentiating or manipulating these stem cells for therapeutic purposes remains a challenging problem. Pluripotent stem cells, as well as adult stem cells, require biological cues for their proliferation and differentiation. These cues are largely controlled by cell-cell, cell-insoluble factors (such as extracellular matrix), and cell-soluble factors (such as cytokine or growth factors) interactions. In this review, we describe a state of research on various stem cell-based tissue engineering applications and high throughput strategies for developing synthetic or biosynthetic microenvironments to allow efficient commitments in stem cells. STATEMENT OF SIGNIFICANCE Nowadays, pluripotency of stem cells have received much attention to use therapeutic purpose. However, a major difficulty with stem cell therapy is to control its differentiation through desired cells or tissues. In other words, various microenvironment factors are involved during stem cell differentiation, including dimensionality, growth factors, cell junctions, nutritional status, matrix stiffness, matrix composition, mechanical stress, and cell-matrix adhesion. Therefore, researchers have engineered a variety of platforms to enable controlling and monitoring bioactive factors to induce stem cell commitment. In this review, we report on recent advancements in a novel technology based on high-throughput strategies for stem cell-based tissue engineering applications.


Journal of the American Chemical Society | 2018

General and Facile Coating of Single Cells via Mild Reduction

Hyunbum Kim; Kwangsoo Shin; Ok Kyu Park; Daheui Choi; Hwan D. Kim; Seungmin Baik; Soo Hong Lee; Seung Hae Kwon; Kevin J. Yarema; Jinkee Hong; Taeghwan Hyeon; Nathaniel S. Hwang

Cell surface modification has been extensively studied to enhance the efficacy of cell therapy. Still, general accessibility and versatility are remaining challenges to meet the increasing demand for cell-based therapy. Herein, we present a facile and universal cell surface modification method that involves mild reduction of disulfide bonds in cell membrane protein to thiol groups. The reduced cells are successfully coated with biomolecules, polymers, and nanoparticles for an assortment of applications, including rapid cell assembly, in vivo cell monitoring, and localized cell-based drug delivery. No adverse effect on cellular morphology, viability, proliferation, and metabolism is observed. Furthermore, simultaneous coating with polyethylene glycol and dexamethasone-loaded nanoparticles facilitates enhanced cellular activities in mice, overcoming immune rejection.

Collaboration


Dive into the Hwan D. Kim's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eunjee Lee

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Seung-Hun Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Tai Hyun Park

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Boram Son

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Hae Lin Jang

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Hyun-Gu Yim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Jeong Ah Kim

Seoul National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge