99 results on '"Wonhee Suh"'
Search Results
2. Hair Growth Regulation by Fibroblast Growth Factor 12 (FGF12)
- Author
-
Jiwon Woo, Wonhee Suh, and Jong-Hyuk Sung
- Subjects
fibroblast growth factor 12 ,hair growth ,anagen induction ,outer root sheath cell ,cell migration ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
The fibroblast growth factor (FGF) family has various biological functions, including cell growth, tissue regeneration, embryonic development, metabolism, and angiogenesis. In the case of hair growth, several members of the FGF family, such as FGF1 and FGF2, are involved in hair growth, while FGF5 has the opposite effect. In this study, the regulation of the hair growth cycle by FGF12 was investigated. To observe its effect, the expression of FGF12 was downregulated in mice and outer root sheath (ORS) by siRNA transfection, while FGF12 overexpression was carried out using FGF12 adenovirus. For the results, FGF12 was primarily expressed in ORS cells with a high expression during the anagen phase of hair follicles. Knockdown of FGF12 delayed telogen-to-anagen transition in mice and decreased the hair length in vibrissae hair follicles. It also inhibited the proliferation and migration of ORS cells. On the contrary, FGF12 overexpression increased the migration of ORS cells. FGF12-overexpressed ORS cells induced the telogen-to-anagen transition in the animal model. In addition, FGF12 overexpression regulated the expression of PDGF-CC, MDK, and HB-EGF, and treatment of these factors exhibited hair growth promotion. Altogether, FGF12 promoted hair growth by inducing the anagen phase of hair follicles, suggesting the potential for hair loss therapy.
- Published
- 2022
- Full Text
- View/download PDF
3. Sustained-Release Microspheres of Rivoceranib for the Treatment of Subfoveal Choroidal Neovascularization
- Author
-
E Seul Kim, Min Sang Lee, Hayoung Jeong, Su Yeon Lim, Doha Kim, Dahwun Kim, Jaeback Jung, Siyan Lyu, Hee Joo Cho, Dong Min Kim, Wonhee Suh, and Ji Hoon Jeong
- Subjects
rivoceranib ,drug repositioning ,microsphere ,subfoveal choroidal neovascularization ,macular degeneration ,Pharmacy and materia medica ,RS1-441 - Abstract
The wet type of age-related macular degeneration (AMD) accompanies the subfoveal choroidal neovascularization (CNV) caused by the abnormal extension or remodeling of blood vessels to the macula and retinal pigment epithelium (RPE). Vascular endothelial growth factor (VEGF) is known to play a crucial role in the pathogenesis of the disease. In this study, we tried to repurpose an investigational anticancer drug, rivoceranib, which is a selective inhibitor of VEGF receptor-2 (VEGFR2), and evaluate the therapeutic potential of the drug for the treatment of wet-type AMD in a laser-induced CNV mouse model using microsphere-based sustained drug release formulations. The PLGA-based rivoceranib microsphere can carry out a sustained delivery of rivoceranib for 50 days. When administered intravitreally, the sustained microsphere formulation of rivoceranib effectively inhibited the formation of subfoveal neovascular lesions in mice.
- Published
- 2021
- Full Text
- View/download PDF
4. A Fully Human Monoclonal Antibody Targeting cKIT Is a Potent Inhibitor of Pathological Choroidal Neovascularization in Mice
- Author
-
Songyi Seo, Koung Li Kim, Yeongju Yeo, Ryul-I Kim, Hayoung Jeong, Jin-Ock Kim, Sun-Hwa Song, Mi-Jin An, Jung-Woong Kim, Hye Kyoung Hong, Min Hee Ham, Se Joon Woo, Jong-Hyuk Sung, Sang Gyu Park, and Wonhee Suh
- Subjects
age-related macular degeneration ,choroidal neovascularization ,cKIT ,fully human monoclonal antibody ,stem cell factor ,Pharmacy and materia medica ,RS1-441 - Abstract
Stem cell factor (SCF) and its receptor, cKIT, are novel regulators of pathological neovascularization in the eye, which suggests that inhibition of SCF/cKIT signaling may be a novel pharmacological strategy for treating neovascular age-related macular degeneration (AMD). This study evaluated the therapeutic potential of a newly developed fully human monoclonal antibody targeting cKIT, NN2101, in a murine model of neovascular AMD. In hypoxic human endothelial cells, NN2101 substantially inhibited the SCF-induced increase in angiogenesis and activation of the cKIT signaling pathway. In a murine model of neovascular AMD, intravitreal injection of NN2101 substantially inhibited the SCF/cKIT-mediated choroidal neovascularization (CNV), with efficacy comparable to aflibercept, a vascular endothelial growth factor inhibitor. A combined intravitreal injection of NN2101 and aflibercept resulted in an additive therapeutic effect on CNV. NN2101 neither caused ocular toxicity nor interfered with the early retinal vascular development in mice. Ocular pharmacokinetic analysis in rabbits indicated that NN2101 demonstrated a pharmacokinetic profile suitable for intravitreal injection. These findings provide the first evidence of the potential use of the anti-cKIT blocking antibody, NN2101, as an alternative or additive therapeutic for the treatment of neovascular AMD.
- Published
- 2021
- Full Text
- View/download PDF
5. Structure-Activity Relationship of Indole-Tethered Pyrimidine Derivatives that Concurrently Inhibit Epidermal Growth Factor Receptor and Other Angiokinases.
- Author
-
Jiho Song, Jakyung Yoo, Ara Kwon, Doran Kim, Hong Khanh Nguyen, Bong-Yong Lee, Wonhee Suh, and Kyung Hoon Min
- Subjects
Medicine ,Science - Abstract
Antiangiogenic agents have been widely investigated in combination with standard chemotherapy or targeted cancer agents for better management of advanced cancers. Therapeutic agents that concurrently inhibit epidermal growth factor receptor and other angiokinases could be useful alternatives to combination therapies for epidermal growth factor receptor-dependent cancers. Here, we report the synthesis of an indole derivative of pazopanib using a bioisosteric replacement strategy, which was designated MKP101. MKP101 inhibited not only the epidermal growth factor receptor with an IC50 value of 43 nM but also inhibited angiokinases as potently as pazopanib. In addition, MKP101 effectively inhibited vascular endothelial growth factor-induced endothelial proliferation, tube formation, migration of human umbilical vein endothelial cells and proliferation of HCC827, an epidermal growth factor receptor-addicted cancer cell line. A docking model of MKP101 and the kinase domain of the epidermal growth factor receptor was generated to predict its binding mode, and validated by synthesizing and evaluating MKP101 derivatives. Additionally, a study of structure-activity relationships of indolylamino or indolyloxy pyrimidine analogues derived from MKP101 demonstrated that selectivity for epidermal growth factor receptor and other angiokinases, especially vascular endothelial growth factor receptor 2 depends on the position of substituents on pyrimidine and the type of link between pyrimidine and the indole moiety. We believe that this study could provide a basis for developing angiokinase inhibitors having high affinity for the epidermal growth factor receptor, from the pyrimidine scaffold.
- Published
- 2015
- Full Text
- View/download PDF
6. Human Cord Blood-Derived Endothelial Progenitor Cells and Their Conditioned Media Exhibit Therapeutic Equivalence for Diabetic Wound Healing
- Author
-
Ji Yeon Kim, Sun-Hwa Song, Koung Li Kim, Jeong-Jae Ko, Ji-Eun Im, Se Won Yie, Young Keun Ahn, Duk-Kyung Kim, and Wonhee Suh Ph.D.
- Subjects
Medicine - Abstract
Transplantation of human cord blood-derived endothelial progenitor cells (EPCs) is reported to contribute to neovascularization in various ischemic diseases. However, the possible beneficial role and underlying mechanisms in diabetes-impaired wound healing have been less well characterized. In this study, EPC transplantation stimulated keratinocyte and fibroblast proliferation substantially as early as 3 days after injury, leading to significantly accelerated wound closure in streptozotocin-induced diabetic nude mice, compared to PBS control. RT-PCR analysis showed that EPCs secreted various wound healing-related growth factors. Among them, keratinocyte growth factor and platelet-derived growth factor were highly expressed in the EPCs and were present at substantial levels in the EPC-injected dermal tissue. Using EPC-conditioned medium (CM), we found that paracrine factors from EPCs directly exerted mitogenic and chemotactic effects on keratinocytes and fibroblasts. Moreover, injection of EPC-CM alone into the same diabetic wound mice promoted wound healing and increased neovascularization to a similar extent as achieved with EPC transplantation. These results indicate that the beneficial effect of EPC transplantation on diabetic wounds was mainly achieved by their direct paracrine action on keratinocytes, fibroblasts, and endothelial cells, rather than through their physical engraftment into host tissues (vasculogenesis). In addition, EPC-CM was shown to be therapeutically equivalent to EPCs, at least for the treatment of diabetic dermal wounds, suggesting that conditioned medium may serve as a novel therapeutic option that is free from allograft-associated immune rejection concern.
- Published
- 2010
- Full Text
- View/download PDF
7. Crosstalk between BMP signaling and KCNK3 in phenotypic switching of pulmonary vascular smooth muscle cells
- Author
-
Yeongju Yeo, Hayoung Jeong, Minju Kim, Yanghee Choi, Koung Li Kim, and Wonhee Suh
- Subjects
General Medicine ,Molecular Biology ,Biochemistry - Published
- 2022
8. Discovery of Novel Small-Molecule Antiangiogenesis Agents to Treat Diabetic Retinopathy
- Author
-
Sang Kook Lee, Sanghee Kim, Kyoungsun Choi, Donghwa Kim, Jaeho Han, Sang Won Choi, Koung Li Kim, Wonhee Suh, Soongyu Choi, S. J. Seo, Jedo Oh, Jae-Hui Been, Wenzhe Jiang, Jihee Cho, and Changmin Park
- Subjects
Cell Survival ,Angiogenesis ,Down-Regulation ,Neovascularization, Physiologic ,Angiogenesis Inhibitors ,Pharmacology ,01 natural sciences ,Angiopoietin-2 ,Small Molecule Libraries ,Mice ,Phosphatidylinositol 3-Kinases ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,Drug Stability ,In vivo ,Drug Discovery ,Human Umbilical Vein Endothelial Cells ,medicine ,Animals ,Humans ,Cytotoxicity ,030304 developmental biology ,0303 health sciences ,Diabetic Retinopathy ,Retinal ,Diabetic retinopathy ,Triazoles ,medicine.disease ,Small molecule ,0104 chemical sciences ,Mice, Inbred C57BL ,Disease Models, Animal ,010404 medicinal & biomolecular chemistry ,Glucose ,Choroidal neovascularization ,chemistry ,Drug Design ,Molecular Medicine ,medicine.symptom ,Signal Transduction ,Retinopathy - Abstract
Diabetic retinopathy is the leading cause of blindness which is associated with excessive angiogenesis. Using the structure of wondonin marine natural products, we previously created a scaffold to develop a novel type of antiangiogenesis agent that possesses minimized cytotoxicity. To overcome its poor pharmaceutical properties, we further modified the structure. A new scaffold was derived in which the stereogenic carbon was changed to nitrogen and the 1,2,3-triazole ring was replaced by an alkyl chain. By comparing the bioactivity versus cytotoxicity, compound 31 was selected, which has improved aqueous solubility and an enhanced selectivity index. Mechanistically, 31 suppressed angiopoietin-2 (ANGPT2) expression induced by high glucose in retinal cells and exhibited in vivo antiangiogenic activity in choroidal neovascularization and oxygen-induced retinopathy mouse models. These results suggest the potential of 31 as a lead to develop antiangiogenic small-molecule drugs to treat diabetic retinopathy and as a chemical tool to elucidate new mechanisms of angiogenesis.
- Published
- 2021
9. FGF12 (Fibroblast Growth Factor 12) Inhibits Vascular Smooth Muscle Cell Remodeling in Pulmonary Arterial Hypertension
- Author
-
Jeong-Min Kim, Eun-Kyung Jo, Yeongju Yeo, Eunhee S. Yi, Wonhee Suh, Koung Li Kim, Sang Gyu Park, S. J. Seo, Jong-Hyuk Sung, and Ryul-I Kim
- Subjects
Male ,0301 basic medicine ,Vascular smooth muscle ,MAP Kinase Signaling System ,Cellular differentiation ,Myocytes, Smooth Muscle ,Mice, Transgenic ,Pulmonary Artery ,Vascular Remodeling ,030204 cardiovascular system & hematology ,Fibroblast growth factor ,Bone morphogenetic protein ,Muscle, Smooth, Vascular ,Rats, Sprague-Dawley ,03 medical and health sciences ,0302 clinical medicine ,Internal Medicine ,medicine ,Animals ,Humans ,Cells, Cultured ,Cell Proliferation ,Pulmonary Arterial Hypertension ,MEF2 Transcription Factors ,Chemistry ,Cell growth ,Wild type ,Hypoxia (medical) ,Cell biology ,Fibroblast Growth Factors ,Mice, Inbred C57BL ,030104 developmental biology ,Bone Morphogenetic Proteins ,Phosphorylation ,medicine.symptom - Abstract
Loss of BMP (bone morphogenic protein) signaling induces a phenotype switch of pulmonary arterial smooth muscle cells (PASMCs), which is the pathological basis of pulmonary vascular remodeling in pulmonary arterial hypertension (PAH). Here, we identified FGF12 (fibroblast growth factor 12) as a novel regulator of the BMP-induced phenotype change in PASMCs and elucidated its role in pulmonary vascular remodeling during PAH development. Using murine models of PAH and lung specimens of patients with PAH, we observed that FGF12 expression was significantly reduced in PASMCs. In human PASMCs, FGF12 expression was increased by canonical BMP signaling. FGF12 knockdown blocked the antiproliferative and prodifferentiation effect of BMP on human PASMCs, suggesting that FGF12 is required for the BMP-mediated acquisition of the quiescent and differentiated PASMC phenotype. Mechanistically, FGF12 regulated the BMP-induced phenotype change by inducing MEF2a (myocyte enhancer factor 2a) phosphorylation via p38MAPK signaling, thereby modulating the expression of MEF2a target genes involved in cell proliferation and differentiation. Furthermore, we observed that TG (transgenic) mice with smooth muscle cell–specific FGF12 overexpression were protected from chronic hypoxia–induced PAH development, pulmonary vascular remodeling, and right ventricular hypertrophy. Consistent with the in vitro data using human PASMCs, FGF12 TG mice showed increased MEF2a phosphorylation and a substantial change in MEF2a target gene expression, compared with the WT (wild type) controls. Overall, our findings demonstrate a novel BMP/FGF12/MEF2a pathway regulating the PASMC phenotype switch and suggest FGF12 as a potential target for the development of therapeutics for ameliorating pulmonary vascular remodeling in PAH.
- Published
- 2020
10. Evogliptin, a dipeptidyl peptidase-4 inhibitor, attenuates pathological retinal angiogenesis by suppressing vascular endothelial growth factor-induced Arf6 activation
- Author
-
Yeongju Yeo, Wonhee Suh, S. J. Seo, Koung Li Kim, Ryul-I Kim, and Mikyung Kim
- Subjects
Male ,Vascular Endothelial Growth Factor A ,0301 basic medicine ,Cell signaling ,Angiogenesis ,Clinical Biochemistry ,Gene Expression ,Angiogenesis Inhibitors ,QD415-436 ,Retinal Neovascularization ,030204 cardiovascular system & hematology ,Pharmacology ,Biochemistry ,Piperazines ,Article ,Pathogenesis ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Evogliptin ,Human Umbilical Vein Endothelial Cells ,medicine ,Animals ,Humans ,Phosphorylation ,Cell adhesion ,Vascular diseases ,Molecular Biology ,Dipeptidyl-Peptidase IV Inhibitors ,ADP-Ribosylation Factors ,Chemistry ,Growth factor signalling ,Adenosine ,Vascular endothelial growth factor ,Disease Models, Animal ,030104 developmental biology ,ADP-Ribosylation Factor 6 ,Medicine ,Molecular Medicine ,Signal transduction ,Signal Transduction ,medicine.drug - Abstract
Dipeptidyl peptidase-4 (DPP-4) inhibitors are used for the treatment of type 2 diabetes mellitus (DM). Recent studies have shown that beyond their effect in lowing glucose, DPP-4 inhibitors mitigate DM-related microvascular complications, such as diabetic retinopathy. However, the mechanism by which pathological retinal neovascularization, a major clinical manifestation of diabetic retinopathy, is inhibited is unclear. This study sought to examine the effects of evogliptin, a potent DPP-4 inhibitor, on pathological retinal neovascularization in mice and elucidate the mechanism by which evogliptin inhibits angiogenesis mediated by vascular endothelial growth factor (VEGF), a key factor in the vascular pathogenesis of proliferative diabetic retinopathy (PDR). In a murine model of PDR, an intravitreal injection of evogliptin significantly suppressed aberrant retinal neovascularization. In human endothelial cells, evogliptin reduced VEGF-induced angiogenesis. Western blot analysis showed that evogliptin inhibited the phosphorylation of signaling molecules associated with VEGF-induced cell adhesion and migration. Moreover, evogliptin substantially inhibited the VEGF-induced activation of adenosine 5′-diphosphate ribosylation factor 6 (Arf6), a small guanosine 5′-triphosphatase (GTPase) that regulates VEGF receptor 2 signal transduction. Direct activation of Arf6 using a chemical inhibitor of Arf-directed GTPase-activating protein completely abrogated the inhibitory effect of evogliptin on VEGF-induced activation of the angiogenic signaling pathway, which suggests that evogliptin suppresses VEGF-induced angiogenesis by blocking Arf6 activation. Our results provide insights into the molecular mechanism of the direct inhibitory effect of the DPP-4 inhibitor evogliptin on pathological retinal neovascularization. In addition to its glucose-lowering effect, the antiangiogenic effect of evogliptin could also render it beneficial for individuals with PDR., Retinal disease: How anti-diabetic drug can help eyes Pathological retinal angiogenesis, the damaging formation of new blood vessels in the retina, which is associated with various diseases including diabetes, could be reduced using the anti-diabetic drug evogliptin to inhibit the effects of a vascular growth factor. Researchers in South Korea led by Wonhee Suh and Koung Li Kim at Chung-Ang University in Seoul investigated the molecular mechanism underlying evogliptin’s effects. In studies using mice and cultured human cells they found that evogliptin inhibited the activation of signaling molecules that mediate the effects of vascular endothelial growth factor. They also identified an enzyme in the signaling pathway that is directly inhibited by evogliptin. The results offer molecular level insights into the additional benefit gained from using evogliptin to treat diabetes, distinct from the drug’s established effects in lowering blood glucose.
- Published
- 2020
11. A Fully Human Monoclonal Antibody Targeting cKIT Is a Potent Inhibitor of Pathological Choroidal Neovascularization in Mice
- Author
-
Jung Woong Kim, Ryul I. Kim, Yeongju Yeo, Min Hee Ham, Wonhee Suh, Se Joon Woo, Sun Hwa Song, Jin Ock Kim, Hye Kyoung Hong, S. J. Seo, Jong Hyuk Sung, Mi Jin An, Sang Gyu Park, Hayoung Jeong, and Koung Li Kim
- Subjects
genetic structures ,medicine.drug_class ,Angiogenesis ,Pharmaceutical Science ,Stem cell factor ,Monoclonal antibody ,choroidal neovascularization ,Article ,Pharmacy and materia medica ,stem cell factor ,Blocking antibody ,medicine ,Receptor ,fully human monoclonal antibody ,age-related macular degeneration ,Aflibercept ,business.industry ,Macular degeneration ,medicine.disease ,eye diseases ,RS1-441 ,Choroidal neovascularization ,Cancer research ,sense organs ,medicine.symptom ,business ,cKIT ,medicine.drug - Abstract
Stem cell factor (SCF) and its receptor, cKIT, are novel regulators of pathological neovascularization in the eye, which suggests that inhibition of SCF/cKIT signaling may be a novel pharmacological strategy for treating neovascular age-related macular degeneration (AMD). This study evaluated the therapeutic potential of a newly developed fully human monoclonal antibody targeting cKIT, NN2101, in a murine model of neovascular AMD. In hypoxic human endothelial cells, NN2101 substantially inhibited the SCF-induced increase in angiogenesis and activation of the cKIT signaling pathway. In a murine model of neovascular AMD, intravitreal injection of NN2101 substantially inhibited the SCF/cKIT-mediated choroidal neovascularization (CNV), with efficacy comparable to aflibercept, a vascular endothelial growth factor inhibitor. A combined intravitreal injection of NN2101 and aflibercept resulted in an additive therapeutic effect on CNV. NN2101 neither caused ocular toxicity nor interfered with the early retinal vascular development in mice. Ocular pharmacokinetic analysis in rabbits indicated that NN2101 demonstrated a pharmacokinetic profile suitable for intravitreal injection. These findings provide the first evidence of the potential use of the anti-cKIT blocking antibody, NN2101, as an alternative or additive therapeutic for the treatment of neovascular AMD.
- Published
- 2021
- Full Text
- View/download PDF
12. SCF (Stem Cell Factor) and cKIT Modulate Pathological Ocular Neovascularization
- Author
-
Won Kim, Sang Gyu Park, Jaetaek Kim, Yeongju Yeo, Jee Taek Kim, Koung Li Kim, S. J. Seo, Jong Hyuk Sung, and Wonhee Suh
- Subjects
Male ,Vascular Endothelial Growth Factor A ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,genetic structures ,Angiogenesis ,Angiogenesis Inhibitors ,Enzyme-Linked Immunosorbent Assay ,Stem cell factor ,Retinal Neovascularization ,Neovascularization ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Retinal Diseases ,medicine ,Animals ,Humans ,Hypoxia ,Cells, Cultured ,Analysis of Variance ,Stem Cell Factor ,business.industry ,Endothelial Cells ,Diabetic retinopathy ,Macular degeneration ,Hypoxia (medical) ,medicine.disease ,Immunohistochemistry ,eye diseases ,Mice, Inbred C57BL ,Endothelial stem cell ,Disease Models, Animal ,Proto-Oncogene Proteins c-kit ,030104 developmental biology ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Catenin ,Female ,sense organs ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,Signal Transduction - Abstract
Objective: Aberrant neovascularization is a leading cause of blindness in several eye diseases, including age-related macular degeneration and proliferative diabetic retinopathy. The identification of key regulators of pathological ocular neovascularization has been a subject of extensive research and great therapeutic interest. Here, we explored the previously unrecognized role of cKIT and its ligand, SCF (stem cell factor), in the pathological ocular neovascularization process. Approach and Results: Compared with normoxia, hypoxia, a crucial driver of neovascularization, caused cKIT to be highly upregulated in endothelial cells, which significantly enhanced the angiogenic response of endothelial cells to SCF. In murine models of pathological ocular neovascularization, such as oxygen-induced retinopathy and laser-induced choroidal neovascularization models, cKIT and SCF expression was significantly increased in ocular tissues, and blockade of cKIT and SCF using cKit mutant mice and anti-SCF neutralizing IgG substantially suppressed pathological ocular neovascularization. Mechanistically, SCF/cKIT signaling induced neovascularization through phosphorylation of glycogen synthase kinase-3β and enhancement of the nuclear translocation of β-catenin and the transcription of β-catenin target genes related to angiogenesis. Inhibition of β-catenin-mediated transcription using chemical inhibitors blocked SCF-induced in vitro angiogenesis in hypoxia, and injection of a β-catenin agonist into cKit mutant mice with oxygen-induced retinopathy significantly enhanced pathological neovascularization in the retina. Conclusions; Our data reveal that SCF and cKIT are promising novel therapeutic targets for treating vision-threatening ocular neovascular diseases.
- Published
- 2019
13. Transforming Growth Factor β Receptor Type I Inhibitor, Galunisertib, Has No Beneficial Effects on Aneurysmal Pathological Changes in Marfan Mice
- Author
-
Min-Seob Kim, Seokran Ham, Koung Li Kim, Jeong-Ho Park, Wonhee Suh, and Eon Sub Park
- Subjects
musculoskeletal diseases ,0301 basic medicine ,Marfan syndrome ,Galunisertib ,macromolecular substances ,Biochemistry ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Thoracic aortic aneurysm ,Drug Discovery ,medicine ,cardiovascular diseases ,Receptor ,Pharmacology ,Kinase ,business.industry ,medicine.disease ,030104 developmental biology ,Losartan ,030220 oncology & carcinogenesis ,Cancer research ,cardiovascular system ,Molecular Medicine ,Phosphorylation ,Original Article ,business ,Transforming growth factor-β receptor I inhibitor ,Transforming growth factor ,medicine.drug - Abstract
Marfan syndrome (MFS), a connective tissue disorder caused by mutations in the fibrillin-1 (Fbn1) gene, has vascular manifestations including aortic aneurysm, dissection, and rupture. Its vascular pathogenesis is assumed to be attributed to increased transforming growth factor β (TGFβ) signaling and blockade of excessive TGFβ signaling has been thought to prevent dissection and aneurysm formation. Here, we investigated whether galunisertib, a potent small-molecule inhibitor of TGFβ receptor I (TβRI), attenuates aneurysmal disease in a murine model of MFS (Fbn1C1039G/+) and compared the impact of galuninsertib on the MFS-related vascular pathogenesis with that of losartan, a prophylactic agent routinely used for patients with MFS. Fbn1C1039G/+ mice were administered galunisertib or losartan for 8 weeks, and their ascending aortas were assessed for histopathological changes and phosphorylation of Smad2 and extracellular signal-regulated kinase 1/2 (Erk1/2). Mice treated with galunisertib or losartan barely exhibited phosphorylated Smad2, suggesting that both drugs effectively blocked overactivated canonical TGFβ signaling in Fbn1C1039G/+ mice. However, galunisertib treatment did not attenuate disrupted medial wall architecture and only partially decreased Erk1/2 phosphorylation, whereas losartan significantly inhibited MFS-associated aortopathy and markedly decreased Erk1/2 phosphorylation in Fbn1C1039G/+ mice. These data unexpectedly revealed that galunisertib, a TβRI inhibitor, showed no benefits in aneurysmal disease in MFS mice although it completely blocked Smad2 phosphorylation. The significant losartan-induced inhibition of both aortic vascular pathogenesis and Smad2 phosphorylation implied that canonical TGFβ signaling might not prominently drive aneurysmal diseases in MFS mice.
- Published
- 2019
14. MD001, a Novel Peroxisome Proliferator-activated Receptor α/γ Agonist, Improves Glucose and Lipid Metabolism
- Author
-
Changjin Lim, Seok Ho Kim, Jong Hyuk Sung, Sang Gyu Park, Wonhee Suh, Hyoungsu Kim, Kyeong Won Lee, Shin Hee Hong, Kyong Soo Park, Kiwon Jung, Young-Joon Park, and Jinhee Kim
- Subjects
Male ,0301 basic medicine ,Agonist ,medicine.medical_specialty ,medicine.drug_class ,Glucose uptake ,Peroxisome proliferator-activated receptor ,lcsh:Medicine ,Carbohydrate metabolism ,Article ,Diabetes Mellitus, Experimental ,Mice ,03 medical and health sciences ,Chalcones ,0302 clinical medicine ,Insulin resistance ,Coumarins ,Internal medicine ,medicine ,Animals ,Humans ,Hypoglycemic Agents ,PPAR alpha ,Receptor ,lcsh:Science ,chemistry.chemical_classification ,Multidisciplinary ,Chemistry ,lcsh:R ,Fatty acid ,Lipid metabolism ,Hep G2 Cells ,Lipid Metabolism ,medicine.disease ,Mice, Inbred C57BL ,PPAR gamma ,Glucose ,HEK293 Cells ,030104 developmental biology ,Endocrinology ,Diabetes Mellitus, Type 2 ,lcsh:Q ,Insulin Resistance ,030217 neurology & neurosurgery - Abstract
Peroxisome proliferator-activated receptor (PPAR)-α/γ dual agonists have been developed to treat metabolic diseases; however, most of them exhibit side effects such as body weight gain and oedema. Therefore, we developed a novel PPARα/γ dual agonist that modulates glucose and lipid metabolism without adverse effects. We synthesised novel compounds composed of coumarine and chalcone, determined their crystal structures, and then examined their binding affinity toward PPARα/γ. We investigated the expression of PPARα and PPARγ target genes by chemicals in HepG2, differentiated 3T3-L1, and C2C12 cells. We examined the effect of chemicals on glucose and lipid metabolism in db/db mice. Only MD001 functions as a PPARα/γ dual agonist in vitro. MD001 increased the transcriptional activity of PPARα and PPARγ, resulting in enhanced expression of genes related to β-oxidation and fatty acid and glucose uptake. MD001 significantly improved blood metabolic parameters, including triglycerides, free fatty acids, and glucose, in db/db mice. In addition, MD001 ameliorated hepatic steatosis by stimulating β-oxidation in vitro and in vivo. Our results demonstrated the beneficial effects of the novel compound MD001 on glucose and lipid metabolism as a PPARα/γ dual agonist. Consequently, MD001 may show potential as a novel drug candidate for the treatment of metabolic disorders.
- Published
- 2019
15. Development of S-Methylmethionine Sulfonium Derivatives and Their Skin-Protective Effect against Ultraviolet Exposure
- Author
-
Wonhee Suh, Jong Hyuk Sung, Dae-Duk Kim, Won Serk Kim, Wang Kyun Kim, Ikyon Kim, Jinu Lee, and Nahyun Choi
- Subjects
0301 basic medicine ,Sulfonium ,S-methylmethionine derivatives ,Proliferation ,Matrix metalloproteinase ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,0404 agricultural biotechnology ,Drug Discovery ,UVA/B protection ,Pharmacology ,chemistry.chemical_classification ,S-Methylmethionine ,Reactive oxygen species ,integumentary system ,Cell growth ,04 agricultural and veterinary sciences ,040401 food science ,HaCaT ,030104 developmental biology ,chemistry ,Odor ,Cell culture ,Molecular Medicine ,Original Article - Abstract
In a previous study, we have demonstrated that S-methylmethionine sulfonium (SMMS) confers wound-healing and photoprotective effects on the skin, suggesting that SMMS can be used as a cosmetic raw material. However, it has an unpleasant odor. Therefore, in the present study, we synthesized odor-free SMMS derivatives by eliminating dimethyl sulfide, which is the cause of the unpleasant odor and identified two derivatives that exhibited skin-protective effects: one derivative comprised (2S,4S)- and (2R,4S)-2-phenylthiazolidine-4-carboxylic acid and the other comprised (2S,4R)-, (2S,4S)-, (2R,4R)-, and (2R,4S)-2-phenyl-1,3-thiazinane-4-carboxylic acid. We performed in vitro proliferation assays using human dermal fibroblasts (hDFs) and an immortalized human keratinocyte cell line (HaCaT). The two SMMS derivatives were shown to increase hDF and HaCaT cell proliferation as well as improve their survival by protecting against ultraviolet exposure. Moreover, the derivatives regulated the expression of collagen type I and MMP mRNAs against ultraviolet exposure in hDFs, suggesting that these derivatives can be developed as cosmetic raw materials.
- Published
- 2018
16. Sustained-Release Microspheres of Rivoceranib for the Treatment of Subfoveal Choroidal Neovascularization
- Author
-
Doha Kim, Ji Hoon Jeong, Dong Min Kim, Siyan Lyu, Hee Joo Cho, E Seul Kim, Jaeback Jung, Su Yeon Lim, Min Sang Lee, Dahwun Kim, Hayoung Jeong, and Wonhee Suh
- Subjects
Drug ,medicine.medical_specialty ,macular degeneration ,genetic structures ,media_common.quotation_subject ,Pharmaceutical Science ,drug repositioning ,rivoceranib ,Article ,Pathogenesis ,chemistry.chemical_compound ,Pharmacy and materia medica ,Ophthalmology ,medicine ,subfoveal choroidal neovascularization ,media_common ,Retinal pigment epithelium ,business.industry ,Macular degeneration ,medicine.disease ,eye diseases ,RS1-441 ,Vascular endothelial growth factor ,Drug repositioning ,PLGA ,medicine.anatomical_structure ,Choroidal neovascularization ,chemistry ,microsphere ,sense organs ,medicine.symptom ,business - Abstract
The wet type of age-related macular degeneration (AMD) accompanies the subfoveal choroidal neovascularization (CNV) caused by the abnormal extension or remodeling of blood vessels to the macula and retinal pigment epithelium (RPE). Vascular endothelial growth factor (VEGF) is known to play a crucial role in the pathogenesis of the disease. In this study, we tried to repurpose an investigational anticancer drug, rivoceranib, which is a selective inhibitor of VEGF receptor-2 (VEGFR2), and evaluate the therapeutic potential of the drug for the treatment of wet-type AMD in a laser-induced CNV mouse model using microsphere-based sustained drug release formulations. The PLGA-based rivoceranib microsphere can carry out a sustained delivery of rivoceranib for 50 days. When administered intravitreally, the sustained microsphere formulation of rivoceranib effectively inhibited the formation of subfoveal neovascular lesions in mice.
- Published
- 2021
17. Src inhibition induces melanogenesis in human G361 cells
- Author
-
Won Serk Kim, Nahyun Choi, Wonhee Suh, Kyung Eun Ku, Sang Ho Oh, and Jong Hyuk Sung
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Cancer Research ,Indoles ,Ultraviolet Rays ,p38 mitogen-activated protein kinases ,p38 ,CREB ,p38 Mitogen-Activated Protein Kinases ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Line, Tumor ,Genetics ,Humans ,Phosphorylation ,RNA, Small Interfering ,G361 cell ,Cyclic AMP Response Element-Binding Protein ,Protein kinase A ,Melanoma ,Molecular Biology ,cyclic adenosine monophosphate response element binding ,Melanins ,Sulfonamides ,Src inhibition ,biology ,Chemistry ,Gene Expression Profiling ,Articles ,melanin ,Cell biology ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,Pyrimidines ,src-Family Kinases ,030104 developmental biology ,Oncology ,SU6656 ,alpha-MSH ,030220 oncology & carcinogenesis ,biology.protein ,Molecular Medicine ,Signal transduction ,Tyrosine kinase ,Biomarkers ,Proto-oncogene tyrosine-protein kinase Src - Abstract
The Src kinase family (SKF) includes non-receptor tyrosine kinases that interact with many cellular cytosolic, nuclear and membrane proteins, and is involved in the progression of cellular transformation and oncogenic activity. However, there is little to no evidence on the effect of SKF or its inhibitors on melanogenesis. Therefore, the present study investigated whether C-terminal Src kinase inhibition can induce melanogenesis and examined the associated signaling pathways and mRNA expression of melanogenic proteins. First, whether stimulators of melanogenesis, such as ultraviolet B and α-melanocyte-stimulating hormone, can dephosphorylate Src protein was evaluated, and the results revealed that SU6656 and PP2 inhibited the phosphorylation of Src in G361 cells. Src inhibition by these chemical inhibitors induced melanogenesis in G361 cells and upregulated the mRNA expression levels of melanogenesis-associated genes encoding microphthalmia-associated transcription factor, tyrosinase-related protein 1 (TRP1), TRP2, and tyrosinase. In addition, Src inhibition by small interfering RNA induced melanogenesis and upregulated the mRNA expression levels of melanogenesis-associated genes. As the p38 mitogen-activated protein kinase (MAPK) and cyclic adenosine monophosphate response element binding (CREB) pathways serve key roles in melanogenesis, the present study further examined whether Src mediates melanogenesis via these pathways. As expected, Src inhibition via SU6656 or PP2 administration induced the phosphorylation of p38 or CREB, as determined by western blotting analysis, and increased the levels of phosphorylated p38 or CREB, as determined by immunofluorescence staining. In addition, the induced pigmentation and melanin content of G361 cells by Src inhibitors was significantly inhibited by p38 or CREB inhibitors. Taken together, these data indicate that Src is associated with melanogenesis, and Src inhibition induces melanogenesis via the MAPK and CREB pathways in G361 cells.
- Published
- 2019
18. Hypoxia induces glucose uptake and metabolism of adipose-derived stem cells
- Author
-
Sun U. Song, Wonhee Suh, Hyoung Sook Park, Bo Kyung Sun, Ji Hye Kim, and Jong Hyuk Sung
- Subjects
Proteomics ,0301 basic medicine ,Cancer Research ,Proteome ,Glucose uptake ,Glucose Transport Proteins, Facilitative ,Carbohydrate metabolism ,Biochemistry ,Gene Expression Regulation, Enzymologic ,03 medical and health sciences ,Downregulation and upregulation ,Genetics ,medicine ,Humans ,Lactic Acid ,Hypoxia ,Molecular Biology ,Glyceraldehyde 3-phosphate dehydrogenase ,biology ,Stem Cells ,Glucose transporter ,Hypoxia (medical) ,Hypoxia-Inducible Factor 1, alpha Subunit ,Cell biology ,Glucose ,030104 developmental biology ,Adipose Tissue ,Oncology ,biology.protein ,Molecular Medicine ,Female ,GLUT1 ,medicine.symptom ,Glycolysis ,GLUT3 - Abstract
It has previously been demonstrated that hypoxia has diverse stimulatory effects on adipose‑derived stem cells (ASCs), however, metabolic responses under hypoxia remain to be elucidated. Thus, the present study aimed to investigate the glucose uptake and metabolism of ASCs under hypoxic conditions, and to identify the underlying molecular mechanisms. ASCs were cultured in 1% oxygen, and experiments were conducted in vitro. As determined by proteomic analysis and western blotting, GAPDH and enolase 1 (ENO1) expression were upregulated under hypoxia. In addition, lactate production was significantly increased, and mRNA levels of glycolytic enzymes, including GAPDH, ENO1, hexokinase 2 (HK2), and lactate dehydrogenase α (LDHα) were upregulated. Hypoxia‑inducible factor 1‑α (HIF‑1α) expression was increased as demonstrated by western blotting, and a pharmacological inhibitor of HIF‑1α significantly attenuated hypoxia‑induced lactate production and expression of glycolytic enzymes. It was also observed that hypoxia significantly increased glucose uptake in ASCs, and glucose transporter (GLUT)1 and GLUT3 expression were upregulated under hypoxia. Pharmacological inhibition of the HIF‑1α signaling pathways also attenuated hypoxia‑induced GLUT1 and GLUT3 expression. These results collectively indicate that hypoxia increases glucose uptake via GLUT1 and GLUT3 upregulation, and induces lactate production of ASCs via GAPDH, ENO1, HK2, and LDHα. Furthermore, HIF‑1α is involved in glucose uptake and metabolism of ASCs.
- Published
- 2016
19. ZNF224, Krüppel like zinc finger protein, induces cell growth and apoptosis-resistance by down-regulation of p21 and p53 via miR-663a
- Author
-
Seho Park, Hong Sook Kim, Tae-Young Roh, Key Hwan Lim, Seokjin Ham, Jong Hyuk Sung, Wonhee Suh, Seung Yong Song, Chae Hyun Lim, Sang Gyu Park, and Jin Gu Cho
- Subjects
0301 basic medicine ,Untranslated region ,p53 ,Cyclin-Dependent Kinase Inhibitor p21 ,Repressor ,Down-Regulation ,Breast Neoplasms ,Biology ,Transfection ,miR-663a ,03 medical and health sciences ,0302 clinical medicine ,Transcription (biology) ,Cell Line, Tumor ,microRNA ,Humans ,Gene ,Cell Proliferation ,Zinc finger ,p21 ,Carcinoma, Ductal, Breast ,Cell Cycle ,apoptosis ,Cell cycle ,Genes, p53 ,Molecular biology ,Repressor Proteins ,MicroRNAs ,030104 developmental biology ,HEK293 Cells ,Oncology ,Apoptosis ,030220 oncology & carcinogenesis ,MCF-7 Cells ,Tumor Suppressor Protein p53 ,ZNF224 ,Research Paper - Abstract
// Jin Gu Cho 1, 2 , Seho Park 3 , Chae Hyun Lim 4 , Hong Sook Kim 2 , Seung Yong Song 5 , Tae-Young Roh 4 , Jong-Hyuk Sung 6 , Wonhee Suh 7 , Seok-Jin Ham 4 , Key-Hwan Lim 2 , Sang Gyu Park 2 1 Department of Biomedical Science, CHA University, Sungnam-si, Gyunggi-do, Korea 2 Laboratory for Tracing of Gene Function, Department of Pharmacy, College of Pharmacy, Ajou University, Suwon, Gyunggi-do, Korea 3 Department of Surgery, Yonsei University College of Medicine, Seoul, Korea 4 Division of Integrative Biosciences & Biotechnology, Pohang University of Science & Technology (POSTECH), Pohang, Gyeongbuk, Korea 5 Department of Plastic and Reconstructive Surgery, Yonsei University College of Medicine, Seoul, Korea 6 Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon, Korea 7 Department of Pharmacy, College of Pharmacy, Chung-Ang University, Seoul, Korea Correspondence to: Sang Gyu Park, e-mail: sgpark@ajou.ac.kr Keywords: ZNF224, miR-663a, p53, p21, apoptosis Received: December 22, 2015 Accepted: March 31, 2016 Published: April 20, 2016 ABSTRACT ZNF224 is a Kruppel-associated box-containing zinc-finger protein which represses gene transcription by interacting with various co-repressors. However, its consensus DNA sequences and target genes are not fully identified. In this study, we identified and characterized consensus DNA sequences containing 5‘-CAGC-3’ recognized by ZNF224 through ChIP-sequencing, which further confirmed by ELISA, SPR, qPCR, and luciferase activity assay. ZNF224 increased miR-663a transcription by binding to miR-663a promoter, which in turn binds to 3’ UTR of p53 and p21 to decrease their expression. miR-663a antagonist abolished ZNF224-mediated suppression of p21 and p53, resulting in the enhanced apoptosis by CPT. The analyses using human breast ductal carcinoma tissues exhibited that the expression of ZNF224 and miR-663a was increased in cancer compared to non-cancer region. Consequently, ZNF224 increases cell survival and decreases apoptosis by decreasing the expression of p53 and p21 via miR-663a as a transcriptional activator. Taken together, we identified and characterized DNA binding element of ZNF224, and its target genes, miR-663a, which provides a novel insight in the down-regulation of p21 and p53 via miR-663a by ZNF224 in breast cancer.
- Published
- 2016
20. Micelle-templated dendritic gold nanoparticles for enhanced cellular delivery of siRNA
- Author
-
Dong Woo Lim, Ji Won Park, Min Sang Lee, Nak Won Kim, Hong Tae Kim, Wonhee Suh, Jung Eun Lee, and Ji Hoon Jeong
- Subjects
Materials science ,Polymers and Plastics ,General Chemical Engineering ,Genetic enhancement ,Organic Chemistry ,Nanoparticle ,Nanochemistry ,Nanotechnology ,Transfection ,Micelle ,Colloidal gold ,Materials Chemistry ,Biophysics ,Copolymer ,Surface charge - Abstract
Successful cellular delivery of synthetic siRNA depends mainly on the capability of a carrier to form a stable complex with siRNA, which can provide efficient protection of the siRNA from enzyme-mediated degradation and improved cellular uptake. However, due to its short length and rigid structure, cellular delivery of siRNA is often not as efficient as that of plasmid DNA using conventional cationic polymer- and lipid-based carriers. Herein, we synthesized a dendritic gold nanoparticle (Au@MC)-based siRNA delivery system, which provides efficient protection of siRNA and improved cellular uptake. The Au@MC can be readily synthesized from a block copolymer micelle template with a dendritic structure. Au@MC can efficiently form a stable complex with the short and rigid siRNA by localizing it in the space between the branches of the Au@MC. The stability and cellular uptake efficiency were significantly influenced by the structural features of Au@MC, such as size, surface charge, and gap width between the branches. A selected Au@MC/siRNA formulation could successfully achieve highly efficient siRNA transfection in the absence and presence of serum proteins without significant cell toxicity, suggesting the formulation as a potential candidate for siRNA-based clinical gene therapy.
- Published
- 2015
21. Dual effects of duplex RNA harboring 5′-terminal triphosphate on gene silencing and RIG-I mediated innate immune response
- Author
-
Si Eun Baek, Yong-Joo Jeong, Wonhee Suh, Kyung Bo Kim, Jungwoo Choe, Yo Han Cho, Soojin Yoon, Dong-Eun Kim, and H.-R. Kim
- Subjects
Small interfering RNA ,RNA-induced transcriptional silencing ,RNA-induced silencing complex ,Biophysics ,RNA-dependent RNA polymerase ,Genome, Viral ,Hepacivirus ,Biology ,Biochemistry ,DEAD-box RNA Helicases ,Small hairpin RNA ,Polyphosphates ,RNA interference ,Cell Line, Tumor ,Humans ,Gene Silencing ,RNA, Small Interfering ,Receptors, Immunologic ,Molecular Biology ,RNA ,Cell Biology ,Molecular biology ,Immunity, Innate ,RNA silencing ,Interferon Type I ,DEAD Box Protein 58 ,Nucleic Acid Conformation ,RNA Interference - Abstract
Duplex RNA harboring the 50-terminal triphosphate RNA is hypothesized to not only execute selective gene silencing via RNA interference, but also induce type I interferon (IFN) through activation of the ret- inoic acid inducible gene I (RIG-I). We evaluated gene silencing efficacy of the shRNA containing 5 0 -tri- phosphate (3p-shRNA) targeting the hepatitis C virus (HCV) RNA genome in hepatic cells. Gene silencing efficacy of the 3p-shRNA was diminished due to the presence of the 5 0 -triphosphate moiety in shRNA, whereas the shRNA counterpart without 5 0-triphosphate (HO-shRNA) showed a strong antiviral activity without significant induction of type I IFN in the cells. 3p-shRNA was observed to be a better acti- vator of the RIG-I signaling than the HO-shRNA with an elevated induction of type I IFN in cells that express RIG-I. Taken together, we suggest that competition for the duplex RNA bearing 5 0 -triphosphate between RIG-I and RNA interference factors may compromise efficacy of selective gene silencing.
- Published
- 2015
22. Apatinib, an Inhibitor of Vascular Endothelial Growth Factor Receptor 2, Suppresses Pathologic Ocular Neovascularization in Mice
- Author
-
Wonhee Suh and Koung Li Kim
- Subjects
Male ,genetic structures ,Pyridines ,Blotting, Western ,Administration, Oral ,Angiogenesis Inhibitors ,Retinal Neovascularization ,Neovascularization ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Cell Movement ,Pregnancy ,medicine ,Animals ,Apatinib ,Fluorescein Angiography ,Phosphorylation ,Fluorescent Antibody Technique, Indirect ,Cells, Cultured ,Cell Proliferation ,Laser Coagulation ,business.industry ,Endothelial Cells ,Retinal Vessels ,Kinase insert domain receptor ,Retinal ,Macular degeneration ,medicine.disease ,Vascular Endothelial Growth Factor Receptor-2 ,eye diseases ,Choroidal Neovascularization ,Vascular endothelial growth factor ,Mice, Inbred C57BL ,Oxygen ,Disease Models, Animal ,Choroidal neovascularization ,chemistry ,Animals, Newborn ,030220 oncology & carcinogenesis ,Intravitreal Injections ,030221 ophthalmology & optometry ,Cancer research ,Female ,sense organs ,medicine.symptom ,business ,Retinopathy - Abstract
Purpose Vascular endothelial growth factor (VEGF) signaling via VEGF receptor 2 (VEGFR2) plays a crucial role in pathologic ocular neovascularization. In this study, we investigated the antiangiogenic effect of apatinib, a pharmacologic inhibitor of VEGFR2 tyrosine kinase, against oxygen-induced retinopathy (OIR) and laser-induced choroidal neovascularization (CNV) in mice. Methods Western blotting and in vitro angiogenesis assays were performed using human retinal microvascular endothelial cells (HRMECs). OIR was induced in neonatal mice by exposure to 75% oxygen from postnatal day (P) 7 to P12 and to room air from P12 to P17. Experimental CNV was induced in mice using laser photocoagulation. Apatinib was intravitreally and orally administered to mice. Neovascularization and phosphorylation of VEGFR2 were evaluated by immunofluorescence staining. Results Apatinib inhibited VEGF-mediated activation of VEGFR2 signaling and substantially reduced VEGF-induced proliferation, migration, and cord formation in HRMECs. A single intravitreal injection of apatinib significantly attenuated retinal or choroidal neovascularization in mice with OIR or laser injury-induced CNV, respectively. Retinal or choroidal tissues of the eyes treated with apatinib exhibited substantially lower phosphorylation of VEGFR2 than those of controls injected with vehicle. Intravitreal injection of apatinib did not cause noticeable ocular toxicity. Moreover, oral administration of apatinib significantly reduced laser-induced CNV in mice. Conclusions Our study demonstrates that apatinib inhibits pathologic ocular neovascularization in mice with OIR or laser-induced CNV. Apatinib may, therefore, be a promising drug for the prevention and treatment of ischemia-induced proliferative retinopathy and neovascular age-related macular degeneration.
- Published
- 2017
23. Apatinib-loaded nanoparticles suppress vascular endothelial growth factor-induced angiogenesis and experimental corneal neovascularization
- Author
-
Danbi Kim, Ji Hoon Jeong, Jung Eun Lee, Sun Hwa Kim, Yeongju Yeo, Wonhee Suh, Koung Li Kim, Hyounkoo Han, Myung Goo Kim, and Hyuncheol Kim
- Subjects
0301 basic medicine ,Vascular Endothelial Growth Factor A ,genetic structures ,Angiogenesis ,Pyridines ,Pharmaceutical Science ,Polyethylene Glycols ,Neovascularization ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,0302 clinical medicine ,apatinib ,corneal neovascularization ,nanoparticle ,vascular endothelialgrowth factor ,International Journal of Nanomedicine ,Drug Discovery ,Apatinib ,Original Research ,Tube formation ,vascular endothelial growth factor ,Neovascularization, Pathologic ,Chemistry ,General Medicine ,Vascular endothelial growth factor ,medicine.symptom ,Biophysics ,Bioengineering ,Biomaterials ,03 medical and health sciences ,In vivo ,medicine ,Human Umbilical Vein Endothelial Cells ,Animals ,Humans ,Serum Albumin ,Organic Chemistry ,Endothelial Cells ,medicine.disease ,Vascular Endothelial Growth Factor Receptor-2 ,In vitro ,eye diseases ,Disease Models, Animal ,030104 developmental biology ,Corneal neovascularization ,030221 ophthalmology & optometry ,Cancer research ,Nanoparticles ,Angiogenesis Inducing Agents ,sense organs - Abstract
Jung Eun Lee,1,* Koung Li Kim,2,* Danbi Kim,2 Yeongju Yeo,2 Hyounkoo Han,3 Myung Goo Kim,1,4 Sun Hwa Kim,4 Hyuncheol Kim,3,5 Ji Hoon Jeong,1,4 Wonhee Suh2 1School of Pharmacy, Sungkyunkwan University, Suwon, 2College of Pharmacy, Chung-Ang University, Seoul, 3Department of Chemical and Biomolecular Engineering, Sogang University, 4Center for Theragnosis Biomedical Research Institute, Korea Institute of Science and Technology (KIST), 5Department of Biomedical Engineering, Sogang University, Seoul, Korea *These authors contributed equally tothis work Abstract: Pathological angiogenesis is one of the major symptoms of severe ocular diseases, including corneal neovascularization. The blockade of vascular endothelial growth factor (VEGF) action has been recognized as an efficient strategy for treating corneal neovascularization. In this study, we aimed to investigate whether nanoparticle-based delivery of apatinib, a novel and selective inhibitor of VEGF receptor 2, inhibits VEGF-mediated angiogenesis and suppresses experimental corneal neovascularization. Water-insoluble apatinib was encapsulated in nanoparticles composed of human serum albumin (HSA)-conjugated polyethylene glycol (PEG). In vitro angiogenesis assays showed that apatinib-loaded HSA-PEG (Apa-HSA-PEG) nanoparticles potently inhibited VEGF-induced tube formation, scratch wounding migration, and proliferation of human endothelial cells. In a rat model of alkali burn injury-induced corneal neovascularization, a subconjunctival injection of Apa-HSA-PEG nanoparticles induced a significant decrease in neovascularization compared to that observed with an injection of free apatinib solution or phosphate-buffered saline. An in vivo distribution study using HSA-PEG nanoparticles loaded with fluorescent hydrophobic model drugs revealed the presence of a substantial number of nanoparticles in the corneal stroma within 24 h after injection. These in vitro and in vivo results demonstrate that apatinib-loaded nanoparticles may be promising for the prevention and treatment of corneal neovascularization-related ocular disorders. Keywords: apatinib, corneal neovascularization, nanoparticle, vascular endothelial growth factor
- Published
- 2017
24. Antiangiogenic effect of dasatinib in murine models of oxygen-induced retinopathy and laser-induced choroidal neovascularization
- Author
-
Songyi, Seo and Wonhee, Suh
- Subjects
Male ,Vascular Endothelial Growth Factor A ,genetic structures ,Dasatinib ,Angiogenesis Inhibitors ,Retinal Neovascularization ,Mice ,Cell Movement ,hemic and lymphatic diseases ,In Situ Nick-End Labeling ,Animals ,Humans ,Fluorescein Angiography ,Phosphorylation ,Protein Kinase Inhibitors ,Cell Proliferation ,Laser Coagulation ,Retinal Vessels ,eye diseases ,Choroidal Neovascularization ,Mice, Inbred C57BL ,Oxygen ,Disease Models, Animal ,Intravitreal Injections ,Female ,sense organs ,Endothelium, Vascular ,Research Article - Abstract
Purpose Vascular endothelial growth factor (VEGF) is a principal mediator of pathological ocular neovascularization, which is the leading cause of blindness in various ocular diseases. As Src, a non-receptor tyrosine kinase, has been implicated as one of the major signaling molecules in VEGF-mediated neovascularization, the present study aimed to investigate whether dasatinib, a potent Src kinase inhibitor, could suppress pathological ocular neovascularization in murine models of oxygen-induced retinopathy (OIR) and choroidal neovascularization (CNV). Methods Tube formation, scratch wounding migration, and cell proliferation assays were performed to measure the inhibitory effect of dasatinib on VEGF-induced angiogenesis in human retinal microvascular endothelial cells. Murine models of OIR and laser-induced CNV were used to assess the preventive effect of an intravitreal injection of dasatinib on pathological neovascularization in the retina and choroid. Neovascularization and Src phosphorylation were evaluated with immunofluorescence staining. Results Dasatinib efficiently inhibited VEGF-induced endothelial proliferation, wounding migration, and tube formation. In mice with OIR and laser injury-induced CNV, eyes treated with a single intravitreal injection of dasatinib exhibited significant decreases in pathological neovascularization compared with that of controls injected with vehicle. The dasatinib-treated OIR mice also showed a decrease in Src phosphorylation in the periretinal tufts. The intravitreal injection of dasatinib did not cause ocular toxicity at the treatment dose administered. Conclusions These results demonstrated that dasatinib suppressed pathological neovascularization in the mouse retina and choroid. Therefore, dasatinib may be indicated for the treatment of ischemia-induced proliferative retinopathy and neovascular age-related macular degeneration.
- Published
- 2017
25. Stem Cell Factor Is a Potent Endothelial Permeability Factor
- Author
-
Kyoungjong Kim, Byung Ho Lee, Sun-Hwa Song, Wonhee Suh, Choun-Ki Joo, Soonboem Kwon, Jun-Sub Choi, Hwa Kyoung Shin, Ji-Eun Im, Jung-Mo Kim, and Ji Yeon Kim
- Subjects
Male ,Time Factors ,Nitric Oxide Synthase Type III ,Vascular permeability ,Stem cell factor ,Biology ,Nitric Oxide ,Transfection ,Diabetes Mellitus, Experimental ,Capillary Permeability ,Adherens junction ,Mice ,chemistry.chemical_compound ,Human Umbilical Vein Endothelial Cells ,Animals ,Humans ,Phosphorylation ,Cells, Cultured ,Mice, Knockout ,Stem Cell Factor ,Diabetic Retinopathy ,Akt/PKB signaling pathway ,Endothelial Cells ,Retinal Vessels ,Adherens Junctions ,Antibodies, Neutralizing ,Cell biology ,Mice, Inbred C57BL ,Vascular endothelial growth factor ,Vascular endothelial growth factor B ,Proto-Oncogene Proteins c-kit ,Vascular endothelial growth factor A ,chemistry ,Vascular endothelial growth factor C ,Intravitreal Injections ,RNA Interference ,Phosphatidylinositol 3-Kinase ,Cardiology and Cardiovascular Medicine ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Objective— Although stem cell factor (SCF) has been shown to play a critical role in hematopoiesis, gametogenesis, and melanogenesis, the function of SCF in the regulation of vascular integrity has not been studied. Approach and Results— We demonstrated that SCF binds to and activates the cKit receptor in endothelial cells, thereby increasing the internalization of vascular endothelial-cadherin and enhancing extravasation of dyes to a similar extent as vascular endothelial growth factor. SCF-mediated cKit activation in endothelial cells enhanced the phosphorylation of endothelial nitric oxide (NO) synthase via the phosphoinositide 3-kinase/Akt signaling pathway and subsequently increased the production of NO. Inhibition of endothelial NO synthase expression and NO synthesis using small interfering RNA knockdown and chemical inhibitors substantially diminished the ability of SCF to increase the internalization of vascular endothelial-cadherin and in vitro endothelial permeability. SCF-induced increase in extravasation of the dyes was abrogated in endothelial NO synthase knockout mice, which indicates that endothelial NO synthase–mediated NO production was responsible for the SCF-induced vascular leakage. Furthermore, we demonstrated that the expression of SCF and cKit was significantly higher in the retina of streptozotocin-injected diabetic mice than in the nondiabetic control animals. Depletion of SCF by intravitreous injection of anti-SCF–neutralizing immunoglobulin G significantly prevented vascular hyperpermeability in the retinas of streptozotocin-injected diabetic mice. Conclusions— Our data reveal that SCF disrupts the endothelial adherens junction and enhances vascular leakage, as well as suggest that anti-SCF/cKit therapy may hold promise as a potential therapy for the treatment of hyperpermeable vascular diseases.
- Published
- 2014
26. Enzyme-catalyzed in situ forming gelatin hydrogels as bioactive wound dressings: effects of fibroblast delivery on wound healing efficacy
- Author
-
Wonhee Suh, Jin Woo Bae, Jin Woo Lee, Ki Dong Park, and Yunki Lee
- Subjects
medicine.medical_specialty ,Materials science ,food.ingredient ,biology ,technology, industry, and agriculture ,Biomedical Engineering ,General Chemistry ,General Medicine ,Gelatin ,Horseradish peroxidase ,Surgery ,Transplantation ,food ,medicine.anatomical_structure ,Tissue engineering ,In vivo ,Self-healing hydrogels ,medicine ,biology.protein ,General Materials Science ,Wound healing ,Fibroblast ,Biomedical engineering - Abstract
In this study, in situ forming gelatin hydrogels via horseradish peroxidase (HRP)-catalyzed cross-linking were developed to serve as bioactive wound dressings with suitable tissue adhesive properties to deliver dermal fibroblasts (DFBs). The DFB-encapsulated gelatin hydrogels with different stiffnesses, GH-soft (1.1 kPa) and GH-hard (6.2 kPa), were prepared by controlling the hydrogen peroxide (H2O2) concentrations. The GH-soft hydrogel was capable of facilitating the proliferation of DFBs and the synthesis of extracellular components, as compared to GH-hard hydrogels. In addition, the subcutaneously injected GH-soft hydrogel with bioluminescent reporter cells provided enhanced cell survival and local retention over 14 days. In vivo transplantation of DFB-encapsulated GH-soft hydrogels accelerated wound contraction, and promoted collagen deposition and neovascularization within the incisions performed on mice skin. Therefore, we expect that HRP-catalyzed in situ forming gelatin hydrogels can be useful for local delivery of cells with high viability in wounds, which holds great promise for advancing wound healing technologies and other tissue engineering applications.
- Published
- 2014
27. Tcea3 Regulates the Vascular Differentiation Potential of Mouse Embryonic Stem Cells
- Author
-
Kyung-Soon Park, Wonhee Suh, Young Joo Cha, Sun-Hee Heo, Seong Kyu Yang, Jihwan Song, and Hee-Jin Ahn
- Subjects
Vascular Endothelial Growth Factor A ,Mesoderm ,animal structures ,Nodal signaling ,Ectoderm ,Biology ,Article ,Mice ,Vasculogenesis ,Genetics ,medicine ,Animals ,Molecular Biology ,Cells, Cultured ,Embryonic Stem Cells ,Oligonucleotide Array Sequence Analysis ,Gene Expression Profiling ,Lineage markers ,Cell Differentiation ,Embryonic stem cell ,Cell biology ,Vascular endothelial growth factor A ,medicine.anatomical_structure ,embryonic structures ,Blood Vessels ,Transcriptional Elongation Factors ,NODAL - Abstract
Tcea3 is present in high concentrations in mouse embryonic stem cells (mESCs) and functions to activate Lefty1, a negative regulator of Nodal signaling. The Nodal pathway has numerous biological activities, including mesoderm induction and patterning in early embryogenesis. Here, we demonstrate that the suppression of Tcea3 in mESCs shifts the cells from pluripotency into enhanced mesoderm development. Vascular endothelial growth factor A (VEGFA) and VEGFC, major transcription factors that regulate vasculogenesis, are activated in Tcea3 knocked down (Tcea3 KD) mESCs. Moreover, differentiating Tcea3 KD mESCs have perturbed gene expression profiles with suppressed ectoderm and activated mesoderm lineage markers. Most early differentiating Tcea3 KD cells expressed Brachyury-T, a mesoderm marker, whereas control cells did not express the gene. Finally, development of chimeric embryos that included Tcea3 KD mESCs was perturbed.
- Published
- 2013
28. Distinct transcriptional profiles of angioblasts derived from human embryonic stem cells
- Author
-
Hyun Ok Kim, Wonpyo Hong, Koung Li Kim, Kiyoung Lee, Woojin Jung, Kyung-Ah Lee, Wonhee Suh, and Sun-Hwa Song
- Subjects
Transcription, Genetic ,Hemangioblasts ,Somatic cell ,Antigens, CD34 ,Biology ,Angioblast ,Mesoderm ,Vasculogenesis ,Cluster Analysis ,Humans ,Mitosis ,Cells, Cultured ,Embryonic Stem Cells ,Microarray analysis techniques ,Gene Expression Profiling ,Endothelial Cells ,Cell Differentiation ,Cell Biology ,Microarray Analysis ,Embryonic stem cell ,Molecular biology ,Cell biology ,Platelet Endothelial Cell Adhesion Molecule-1 ,Gene expression profiling ,Endothelial stem cell ,embryonic structures - Abstract
Identification of differentially expressed genes in angioblasts derived from human embryonic stem cells (hESCs) is of great interest for elucidating the molecular mechanisms underlying human vasculogenesis. The aim of this study was to define hESC-derived angioblasts at the clonal level and to perform comparative transcriptional analysis to characterize their distinct gene expression profiles. In a clonal analysis performed in cell-specific differentiation media, hESC-derived CD34(+)CD31(+) cells were identified as angioblasts in that they exhibited a significantly higher ability to form endothelial cell (EC) and smooth muscle cell (SMC) colonies than CD34(+)CD31(-) and CD34(-) cell populations did. Microarray analysis showed that many genes involved in vascular development and signaling transduction were overexpressed in hESC-derived CD34(+)CD31(+) cells, whereas those related to mitosis, the DNA damage response, and translation were substantially downregulated. In addition, comparative gene expression profiling of hESC-derived CD34(+)CD31(+) cells and human somatic primary vascular cells demonstrated that hESC-derived CD34(+)CD31(+) cells expressed key genes involved in the EC and SMC differentiation processes, which supports the result that hESC-derived CD34(+)CD31(+) cells are bipotent angioblasts. Our results may provide insights into the identity and function of hESC-derived angioblasts and may also facilitate further investigation of the molecular mechanisms regulating human embryonic vasculogenesis.
- Published
- 2013
29. Positive Correlation Between the Dysregulation of Transforming Growth Factor-β1 and Aneurysmal Pathological Changes in Patients With Marfan Syndrome
- Author
-
Jeong-Min Kim, Young-Wook Kim, Koung Li Kim, Sun-Hwa Song, Duk-Kyung Kim, Wonhee Suh, Jee-Ah Kim, Kiick Sung, Yeon-Lim Suh, Shin Yi Jang, Ji Yeon Kim, and Jeong Hoon Yang
- Subjects
Marfan syndrome ,congenital, hereditary, and neonatal diseases and abnormalities ,Pathology ,medicine.medical_specialty ,business.industry ,macromolecular substances ,General Medicine ,medicine.disease ,Pathogenesis ,Aortic aneurysm ,Apoptosis ,cardiovascular system ,Medicine ,Phosphorylation ,Biomarker (medicine) ,cardiovascular diseases ,skin and connective tissue diseases ,Cardiology and Cardiovascular Medicine ,business ,Pathological ,Transforming growth factor - Abstract
Background Our goal was to investigate the correlation between the dysregulation of transforming growth factor-β1 (TGF-β1) and cystic medial degeneration in the aortic aneurysmal tissues of in Marfan syndrome (MFS) patients. Although aortic aneurysm in animal models of MFS is related to the dysregulation of TGF-β, it has yet to be determined whether TGF-β dysregulation correlates with pathogenic aneurysmal characteristics in MFS patients. Methods and results Compared with aortic tissue from normal individuals, the medial layers of aortic tissue from MFS patients exhibited profound cystic medial degeneration and cellular apoptosis. These histopathologic changes positively correlated with the extent of TGF-β1 signaling activation (Smad2 phosphorylation) in aneurysmal aortic tissue. In addition, the level of TGF-β1 expression in peripheral blood and aneurysmal aortic tissues was significantly elevated in MFS patients. A significant positive correlation was observed between the plasma level of active TGF-β1 in MFS patients and the severity of cystic medial degeneration and Smad2 phosphorylation in aneurysmal aortic medial layers. Conclusions We found a strong association between the dysregulation of TGF-β1 and aortic pathogenesis in human MFS patients. This suggests that the plasma concentration of TGF-β1 in MFS patients might be a useful biomarker of the progression of aortic aneurysms.
- Published
- 2013
30. Fibroblast Growth Factor 12 Is a Novel Regulator of Vascular Smooth Muscle Cell Plasticity and Fate
- Author
-
Young-Wook Kim, Wonhee Suh, Jong Hyuk Sung, Sun Hwa Song, Jin Sook Kwon, Eun Kyung Jo, Jee Taek Kim, Sang Gyu Park, Kyungjong Kim, and Sun Sik Bae
- Subjects
0301 basic medicine ,Carotid Artery Diseases ,Male ,Vascular smooth muscle ,Transcription, Genetic ,Cell Plasticity ,Regulator ,Becaplermin ,Fibroblast growth factor ,p38 Mitogen-Activated Protein Kinases ,Muscle, Smooth, Vascular ,Rats, Sprague-Dawley ,Cells, Cultured ,Mice, Knockout ,Cell Differentiation ,Proto-Oncogene Proteins c-sis ,Phenotype ,Cell biology ,cardiovascular system ,RNA Interference ,Cardiology and Cardiovascular Medicine ,Protein Binding ,Signal Transduction ,medicine.medical_specialty ,Genotype ,Carotid Artery, Common ,Myocytes, Smooth Muscle ,Biology ,Transfection ,03 medical and health sciences ,Apolipoproteins E ,Downregulation and upregulation ,Internal medicine ,Neointima ,medicine ,Animals ,Humans ,Cell Lineage ,Embryonic Stem Cells ,Cell Proliferation ,Binding Sites ,Hyperplasia ,Cell growth ,Fibroblast Growth Factors ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Myocardin ,Vasoconstriction ,Phosphatidylinositol 3-Kinase ,5' Untranslated Regions ,Carotid Artery Injuries - Abstract
Objective— Vascular smooth muscle cells (VSMCs) modulate their phenotype between synthetic and contractile states in response to environmental changes; this modulation plays a crucial role in the pathogenesis of restenosis and atherosclerosis. Here, we identified fibroblast growth factor 12 (FGF12) as a novel key regulator of the VSMC phenotype switch. Approach and Results— Using murine models and human specimens, we found that FGF12 was highly expressed in contractile VSMCs of normal vessel walls but was downregulated in synthetic VSMCs from injured and atherosclerotic vessels. In human VSMCs, FGF12 expression was inhibited at the transcriptional level by platelet-derived growth factor-BB. Gain- and loss-of-function experiments showed that FGF12 was both necessary and sufficient for inducing and maintaining the quiescent and contractile phenotypes of VSMCs. FGF12 inhibited cell proliferation through the p53 pathway and upregulated the key factors involved in VSMC lineage differentiation, such as myocardin and serum response factor. Such FGF12-induced phenotypic change was mediated by the p38 MAPK (mitogen-activated protein kinase) pathway. Moreover, FGF12 promoted the differentiation of mouse embryonic stem cells and the transdifferentiation of human dermal fibroblasts into SMC-like cells. Furthermore, adenoviral infection of FGF12 substantially decreased neointima hyperplasia in a rat carotid artery injury model. Conclusions— In general, FGF family members induce a synthetic VSMC phenotype. Interestingly, the present study showed the unanticipated finding that FGF12 belonging to FGF family, strongly induced the quiescent and contractile VSMC phenotypes and directly promoted VSMC lineage differentiation. These novel findings suggested that FGF12 could be a new therapeutic target for treating restenosis and atherosclerosis.
- Published
- 2016
31. Beneficial effects of the Src inhibitor, dasatinib, on breakdown of the blood-retinal barrier
- Author
-
Wonhee Suh and So Ra Kim
- Subjects
0301 basic medicine ,Male ,Vascular Endothelial Growth Factor A ,Blood–retinal barrier ,Dasatinib ,Vascular permeability ,Biology ,Retina ,Streptozocin ,Diabetes Mellitus, Experimental ,Capillary Permeability ,Rats, Sprague-Dawley ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,In vivo ,Antigens, CD ,hemic and lymphatic diseases ,Drug Discovery ,Blood-Retinal Barrier ,medicine ,Animals ,Humans ,Phosphorylation ,Protein Kinase Inhibitors ,Barrier function ,Diabetic Retinopathy ,Organic Chemistry ,Endothelial Cells ,Retinal Vessels ,Retinal ,Cadherins ,Rats ,Vascular endothelial growth factor ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,src-Family Kinases ,chemistry ,030220 oncology & carcinogenesis ,Intravitreal Injections ,Cancer research ,Molecular Medicine ,medicine.drug ,Proto-oncogene tyrosine-protein kinase Src ,Signal Transduction - Abstract
Src kinase signaling is important in the regulation of microvascular barrier function and endothelial hyperpermeability. This study was designed to evaluate the protective effect of dasatinib, a potent Src inhibitor used clinically for the treatment of cancer, against the breakdown of the blood-retinal barrier (BRB) and the retinal vascular leakage caused by vascular endothelial growth factor (VEGF) and diabetes. We examined the effects of dasatinib on VEGF-induced endothelial hyperpermeability and the loss of vascular endothelial (VE)-cadherin, an endothelial junctional protein. Dasatinib inhibited VEGF-induced phosphorylation of Src in human retinal microvascular endothelial cells (HRMECs). In vitro and in vivo vascular permeability assays showed that dasatinib blocked the VEGF-enhanced hyperpermeability of HRMECs and decreased VEGF-mediated retinal vascular leakage in mice. Immunofluorescent staining of VE-cadherin showed that dasatinib abolished the junctional disappearance of VE-cadherin in VEGF-treated HRMECs and murine retinal vasculature. In addition, we examined the protective effect of dasatinib against diabetes-induced retinal vascular leakage in streptozotocin-induced diabetic rats. An intravitreal injection of dasatinib substantially inhibited the development of hyperpermeable retinal vasculature. Our results indicate that dasatinib is a promising agent for the prevention and treatment of diabetes-induced retinal vascular leakage.
- Published
- 2016
32. A new era of disease modeling and drug discovery using induced pluripotent stem cells
- Author
-
Wonhee Suh
- Subjects
0301 basic medicine ,Somatic cell ,Drug discovery ,Organic Chemistry ,Pharmacology toxicology ,Induced Pluripotent Stem Cells ,Disease ,Computational biology ,Pharmacology ,Biology ,Precision medicine ,Cellular Reprogramming ,Models, Biological ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Pharmaceutical Preparations ,Drug Discovery ,Molecular Medicine ,Animals ,Humans ,Precision Medicine ,Induced pluripotent stem cell ,Reprogramming ,030217 neurology & neurosurgery - Abstract
In 2006, Shinya Yamanaka first reported that in vitro reprogramming of somatic cells toward pluripotency was achieved by simple induction of specific transcription factors. Induced pluripotent stem cell (iPSC) technology has since revolutionized the ways in which we explore the mechanisms of human diseases and develop therapeutics. Here, I describe the recent advances in human iPSC-based disease modeling and drug discovery and discuss the current challenges. Additionally, I outline potential future applications of human iPSCs in classifying patients based on their response to drugs in clinical trials and elucidating optimal patient-specific therapeutic strategies, which will contribute to reduced attrition rates and the development of precision medicine.
- Published
- 2016
33. Therapeutic effect of apatinib-loaded nanoparticles on diabetes-inducedretinal vascular leakage
- Author
-
Jung Eun Lee, Wonhee Suh, Ji Hoon Jeong, and Hong Khanh Nguyen
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Materials science ,Pyridines ,Biophysics ,Pharmaceutical Science ,Bioengineering ,Pharmacology ,Diabetes Mellitus, Experimental ,Diabetes Complications ,Biomaterials ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Retinal Diseases ,International Journal of Nanomedicine ,Antigens, CD ,In vivo ,permeability ,retinal vascular endothelial cells ,vascular endothelialgrowth factor ,Blood-Retinal Barrier ,Drug Discovery ,VEGF Signaling Pathway ,medicine ,Animals ,Humans ,Apatinib ,Receptor ,Serum Albumin ,Original Research ,Drug Carriers ,vascular endothelial growth factor ,Organic Chemistry ,Endothelial Cells ,Retinal Vessels ,Retinal ,General Medicine ,Cadherins ,In vitro ,Surgery ,Mice, Inbred C57BL ,Vascular endothelial growth factor ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Paracellular transport ,Intravitreal Injections ,Nanoparticles - Abstract
Ji Hoon Jeong,1,* Hong Khanh Nguyen,2,* Jung Eun Lee,1 Wonhee Suh2 1School of Pharmacy, Sungkyunkwan University, Suwon, 2College of Pharmacy, Chung-Ang University, Seoul, Korea *These authors contributed equally tothis work Abstract: Apatinib, a novel and selective inhibitor of vascular endothelial growth factor (VEGF) receptor 2, has been demonstrated recently to exhibit anticancer efficacy by inhibiting the VEGF signaling pathway. Given the importance of VEGF in retinal vascular leakage, the present study was designed to investigate whether apatinib-loaded polymeric nanoparticles inhibit VEGF-mediated retinal vascular hyperpermeability and block diabetes-induced retinal vascular leakage. For the delivery of water-insoluble apatinib, the drug was encapsulated in nanoparticles composed of human serum albumin (HSA)-conjugated polyethylene glycol (PEG). In vitro paracellular permeability and transendothelial electric resistance assays showed that apatinib-loaded HSA-PEG (Apa-HSA-PEG) nanoparticles significantly inhibited VEGF-induced endothelial hyperpermeability in human retinal microvascular endothelial cells. In addition, they substantially reduced the VEGF-induced junctional loss and internalization of vascular endothelial-cadherin, a major component of endothelial junction complexes. In vivo intravitreal injection of Apa-HSA-PEG nanoparticles in mice blocked VEGF-induced retinal vascular leakage. These in vitro and in vivo data indicated that Apa-HSA-PEG nanoparticles efficiently blocked VEGF-induced breakdown of the blood–retinal barrier. In vivo experiments with streptozotocin-induced diabetic mice showed that an intravitreal injection of Apa-HSA-PEG nanoparticles substantially inhibited diabetes-induced retinal vascular leakage. These results demonstrated, for the first time, that apatinib-loaded nanoparticles may be a promising therapeutic agent for the prevention and treatment of diabetes-induced retinal vascular disorders. Keywords: permeability, retinal vascular endothelial cells, vascular endothelial growth factor
- Published
- 2016
34. Direct comparison of distinct cardiomyogenic induction methodologies in human cardiac-derived c-kit positive progenitor cells
- Author
-
Wonhee Suh, Sang-Mo Kwon, Seok Yun Jung, Sung Hyun Choi, Takayuki Asahara, and Sang Hong Baek
- Subjects
GATA4 ,Biomedical Engineering ,Medicine (miscellaneous) ,Transforming growth factor beta ,Biology ,In vitro ,Cell biology ,Immunology ,biology.protein ,Progenitor cell ,Stem cell ,Transcription factor ,Actin ,Fetal bovine serum - Abstract
Cardiac stem/progenitor cells can be differentiated into cardiomyocytes in vitro using several differentiation methodologies. However, the methodology of cardiomyogenic induction in human c-kit positive progenitor cells (hCPCsc-kit+) was not fully demonstrated. Thus, the purpose of our study was to directly evaluate each cardiomyocyte induction system using hCPCsc-kit+. In this study, cardiomyocyte induction methodologies were divided into the following three groups; treatment with dexamethasone, 5-azacytidine, and co-treatment with 5-azacytidine and Transforming Growth Factor Beta 1 (TGF-β1), using different serum concentrations [2% or 10% fetal bovine serum (FBS)]. GATA4 and Nkx2-5, cardiac-specific transcription factors, were expressed in our hCPCsckit+. However, the GATA4 and Nkx2-5 expressions were significantly decreased in 10% FBS/cardiomyogenic induction system (p 0.05). GATA4 and Nkx2-5 is crucial roles in cardiac development, thus we considered the low serum conditions more affected in our cardiomyogenic induction system. In addition, c-kit expression decreased significantly during cardiomyogenic differentiation. Importantly, we demonstrated that co-treated with 5-azacytidine and TGF-β1 led to an earlier expression pattern of alpha-sarcomeric actin (α-SA), implying that this cardiomyocyte induction system facilitates early cardiomyocyte differentiation of hCPCsc-kit+. Thus, the present study provides a pivotal cardiomyogenic differentiation methodology using hCPCsc-kit+for basic or clinical research.
- Published
- 2012
35. Angiopoietin-1 Gene Therapy Attenuates Hypertension and Target Organ Damage in Nitric Oxide Synthase Inhibited Spontaneously Hypertensive Rats
- Author
-
Koung Li Kim, Wonhee Suh, Gou Young Koh, Jung-Sun Lee, Sun-Hwa Song, and Duk-Kyung Kim
- Subjects
Pathology ,medicine.medical_specialty ,Necrosis ,Endothelium ,Genetic enhancement ,Pharmacology ,Nitric oxide ,chemistry.chemical_compound ,Arteriole ,medicine.artery ,Internal Medicine ,medicine ,Angiopoietin-1 ,Kidney ,biology ,business.industry ,Nitric oxide synthase ,medicine.anatomical_structure ,chemistry ,Hypertension ,biology.protein ,Microvascular Rarefaction ,Original Article ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business - Abstract
BACKGROUND AND OBJECTIVES In our previous study, we found that the gene transfer of a potent derivative of cartilage oligomeric matrix protein Angiopoietin-1 (COMP-Ang-1) substantially prevented hypertension, microvascular rarefaction, and target organ damage in spontaneously hypertensive rats (SHRs). The purpose of the present study was to examine the role of nitric oxide (NO) in the therapeutic effects observed after COMP-Ang-1 gene transfer. MATERIALS AND METHODS To exclude the NO-mediated effects in COMP-Ang-1 gene therapy, the SHRs were treated with an NO synthase (NOS) inhibitor, N(w)-nitro-L-arginine methyl ester (L-NAME) before the electrophoretic gene transfer. RESULTS The pretreatment with L-NAME induced a severe and sustained increase in systolic blood pressure (BP) in a LacZ plasmid transferred control SHR. However, the electrophoretic transfer of a COMP-Ang-1 plasmid instead of LacZ plasmid in L-NAME-pretreated SHRs substantially blocked the development of hypertension without any significant difference in comparison with L-NAME-untreated COMP-Ang-1 plasmid transferred groups. In addition, the COMP-Ang-1 plasmid transfer substantially attenuated microvascular rarefaction and arteriole remodeling in the heart and kidney, which might account for the mild histological alterations observed in the COMP-Ang-1 plasmid transferred group, in contrast to the severe fibrosis and necrosis seen in the LacZ plasmid controls. CONCLUSION These therapeutic outcomes of COMP-Ang-1 gene transfer even in NOS inhibited SHRs suggested that the antihypertensive effect of COMP-Ang-1 was not merely secondary to NO-mediated vasorelaxation, but it may be associated with its ability to protect the vascular endothelium probably via an NO-independent mechanism which serves to attenuate microvascular rarefaction and target organ damage, and also to prevent hypertension by reducing peripheral vascular resistance.
- Published
- 2011
36. Protein-based human iPS cells efficiently generate functional dopamine neurons and can treat a rat model of Parkinson disease
- Author
-
Hyunsu Lee, Yong-Hee Rhee, Wonhee Suh, Chun-Hyung Kim, Sang-Hun Lee, Mi-Yoon Chang, Sang-Hoon Yi, Jae-Won Shim, Kwang-Soo Kim, Ji-Yun Ko, Yong-Sung Lee, Byung-Woo Kim, Robert Lanza, Dohoon Kim, Sukho Lee, Chang-Hwan Park, Hyun-Chul Koh, and A-Young Jo
- Subjects
Dopamine ,Cellular differentiation ,Genetic Vectors ,Induced Pluripotent Stem Cells ,Kruppel-Like Transcription Factors ,Apoptosis ,Biology ,Arginine ,Cell Line ,Proto-Oncogene Proteins c-myc ,Cell therapy ,Kruppel-Like Factor 4 ,Parkinsonian Disorders ,Animals ,Humans ,Cell Lineage ,Induced pluripotent stem cell ,Cellular Senescence ,Neurons ,SOXB1 Transcription Factors ,Lentivirus ,Gene Expression Regulation, Developmental ,Cell Differentiation ,General Medicine ,Cellular Reprogramming ,Genes, p53 ,Embryonic stem cell ,Rats ,Transplantation ,Retroviridae ,Cancer research ,Tumor Suppressor Protein p53 ,Stem cell ,Octamer Transcription Factor-3 ,Cell aging ,Reprogramming - Abstract
Parkinson disease (PD) involves the selective loss of midbrain dopamine (mDA) neurons and is a possible target disease for stem cell-based therapy. Human induced pluripotent stem cells (hiPSCs) are a potentially unlimited source of patient-specific cells for transplantation. However, it is critical to evaluate the safety of hiPSCs generated by different reprogramming methods. Here, we compared multiple hiPSC lines derived by virus- and protein-based reprogramming to human ES cells (hESCs). Neuronal precursor cells (NPCs) and dopamine (DA) neurons delivered from lentivirus-based hiPSCs exhibited residual expression of exogenous reprogramming genes, but those cells derived from retrovirus- and protein-based hiPSCs did not. Furthermore, NPCs derived from virus-based hiPSCs exhibited early senescence and apoptotic cell death during passaging, which was preceded by abrupt induction of p53. In contrast, NPCs derived from hESCs and protein-based hiPSCs were highly expandable without senescence. DA neurons derived from protein-based hiPSCs exhibited gene expression, physiological, and electrophysiological properties similar to those of mDA neurons. Transplantation of these cells into rats with striatal lesions, a model of PD, significantly rescued motor deficits. These data support the clinical potential of protein-based hiPSCs for personalized cell therapy of PD.
- Published
- 2011
37. Activation of Vasculogenic Progenitor Cells by ent-16.ALPHA.,17-Dihydroxy-kauran-19-oic Acid
- Author
-
Yun Mi Jeong, So Hyun Park, Sang Hyun Sung, Jihye Kim, Wonhee Suh, Jong Hyuk Sung, Hyung Min Chung, and Sang Gyu Park
- Subjects
MAPK/ERK pathway ,Cell Survival ,Morpholines ,Pharmaceutical Science ,Asteraceae ,Pharmacology ,Biology ,Phosphatidylinositol 3-Kinases ,chemistry.chemical_compound ,Cell Movement ,Nitriles ,Butadienes ,LY294002 ,Progenitor cell ,Protein Kinase Inhibitors ,Protein kinase B ,Cell Proliferation ,Phosphoinositide-3 Kinase Inhibitors ,Mitogen-Activated Protein Kinase 1 ,Tube formation ,Mitogen-Activated Protein Kinase 3 ,Plant Extracts ,Stem Cells ,General Medicine ,Medicine, Korean Traditional ,In vitro ,Plant Leaves ,Transplantation ,Biochemistry ,chemistry ,Chromones ,Diterpenes ,Signal transduction - Abstract
Vasculogenic progenitor cells (VPCs) circulate in the blood and have the ability to differentiate into endothelial cells that make up the lining of blood vessels. Therefore, VPC transplantation is a new strategy for the treatment of ischemic diseases. Because priming/preconditioning of VPCs before transplantation enhances their regenerative potential, the present study investigated whether ent-16α,17-dihydroxy-kauran-19-oic acid (DHK) isolated from Siegesbeckia pubescens could stimulate/activate VPCs in vitro. Therefore, the effect of DHK (1-100 µM concentration) on the proliferation, migration, and tube forming of VPCs was examined in various systems, and related signaling pathways were identified. DHK treatment significantly increased the proliferation, migration, and tube formation of VPCs in a dose-dependent manner. Phosphorylation of extracellular signal-regulated kinase (ERK)1/2 and Akt was significantly increased by DHK, but chemical inhibitors against ERK1/2 (U0126) and Akt (LY294002) significantly attenuated DHK-enhanced proliferation, migration, and tube formation of VPCs. Collectively, these results indicated that DHK shows promise as a novel VPC primer/activator.
- Published
- 2011
38. The expansion of human ES and iPS cells on porous membranes and proliferating human adipose-derived feeder cells
- Author
-
Tae Hee Lee, Soo-Hong Lee, Sun-Woong Kang, Kwang-Soo Kim, Seung-Taeh Hwang, Dong-Ryul Lee, Suk-Jun Lee, Sung Han Shim, Lakeshia J. Taite, and Wonhee Suh
- Subjects
KOSR ,Homeobox protein NANOG ,Stromal cell ,Surface Properties ,Induced Pluripotent Stem Cells ,Biophysics ,Bioengineering ,Biology ,Biomaterials ,SOX2 ,Humans ,Induced pluripotent stem cell ,Cells, Cultured ,Embryonic Stem Cells ,reproductive and urinary physiology ,Cell Proliferation ,Teratoma ,Membranes, Artificial ,equipment and supplies ,Immunohistochemistry ,Embryonic stem cell ,Molecular biology ,Adipose Tissue ,Mechanics of Materials ,Cell culture ,Karyotyping ,embryonic structures ,Ceramics and Composites ,Stromal Cells ,biological phenomena, cell phenomena, and immunity ,Stem cell ,Porosity - Abstract
For clinical application of human embryonic stem cells (hESCs), it is critical to develop hESC culture techniques that completely exclude the use of animal feeder cells, mitotic inhibition, and enzyme treatments used in conventional hESC culture systems. Toward this goal, we attempted to maintain hESCs and induced pluripotent stem (iPS) cells on porous membranes (PMs) with proliferative human adipose-derived stromal cells (ASCs) seeded on the bottom surface of inverted PMs. This culture condition will ensure that the two cell types are separate from each other, yet retain the ability to interact through the pores of the membrane. We found that hESCs and iPS cells can be maintained stably and mechanically transferred without the need for enzyme treatment. In addition, the pluripotency of hESCs and iPS cells was stably maintained, as evidenced by immunostaining of Oct4, SSEA3/4 and TRA-1-60 as well as RT-PCR analyses of Nanog, Oct4 and Sox2 expression. Furthermore, hESCs cultured on PMs showed a normal karyotype and in vivo teratoma formation containing all three germ layers.
- Published
- 2010
39. Enhanced dermal wound neovascularization by targeted delivery of endothelial progenitor cells using an RGD-g-PLLA scaffold
- Author
-
Se Won Yie, Jeong-Min Kim, Wonhee Suh, Cheong-Rae Roh, Eun-Seok Jeon, Kwideok Park, Sun-Hwa Song, Dong Keun Han, Koung Li Kim, Ho Yun Ki, Ji Yeon Kim, and Duk-Kyung Kim
- Subjects
Male ,Polymers ,Angiogenesis ,Polyesters ,Biophysics ,Mice, Nude ,Neovascularization, Physiologic ,Biocompatible Materials ,Bioengineering ,Biology ,Biomaterials ,Neovascularization ,Mice ,Materials Testing ,Cell Adhesion ,medicine ,Animals ,Regeneration ,Lactic Acid ,Progenitor cell ,Nitrites ,Wound Healing ,Tissue Scaffolds ,Endothelial Cells ,Dermis ,Transplantation ,Endothelial stem cell ,Mechanics of Materials ,embryonic structures ,cardiovascular system ,Ceramics and Composites ,Cancer research ,Stem cell ,medicine.symptom ,Wound healing ,Oligopeptides ,Stem Cell Transplantation ,circulatory and respiratory physiology ,Homing (hematopoietic) ,Biomedical engineering - Abstract
Endothelial progenitor cells (EPCs), endothelial precursors that promote neovascularization in ischemic tissues, have shown the limited vascular regeneration efficacy due to their poor homing into injured sites and low survival, so that a variety of biosynthetic scaffolds have been employed as cell delivery vehicles to overcome the current cell transplantation methods. However, few paralleled studies that directly compare the efficacy of EPCs seeded within synthetic scaffolds to that of EPCs delivered by the conventional transplantation techniques used for EPC therapies have been performed. To address these issues, RGD-g-PLLA biosynthetic scaffold was developed for the targeted EPC delivery and was found to successfully support the in vitro growth and endothelial functions of EPCs. This scaffold also appeared to be good as in vivo targeted delivery carriers of EPCs as it promoted vascular regeneration in a murine dermal wound models. Furthermore, direct comparison with the intradermal EPC injection revealed that the targeted delivery of EPCs by using the RGD-g-PLLA scaffold was superior to their conventional local injection method in terms of the localization and survival/retention of the transplanted EPCs, and their vascular repairing potential. These results suggest that the development of an effective stem cell delivery system may help to maximize the tissue-repairing efficacy with a limited number of stem cells, thereby resolving the limited clinical success of current stem cell therapies that have utilized simple cell injections or infusions.
- Published
- 2009
40. Angiopoietin-1 prevents hypertension and target organ damage through its interaction with endothelial Tie2 receptor
- Author
-
Koung Li Kim, Duk-Kyung Kim, Hak-Zoo Kim, Wonhee Suh, Jung-Sun Lee, Eun-Seok Jeon, Sun-Hwa Song, Yeon-Lim Suh, In-Soon Shin, Gou Young Koh, Jeong-Min Kim, and Jonghoe Byun
- Subjects
Male ,Physiology ,Electrochemotherapy ,Nitric Oxide Synthase Type II ,Blood Pressure ,Rats, Inbred WKY ,chemistry.chemical_compound ,Rats, Inbred SHR ,Endothelial dysfunction ,Receptor ,Kidney ,Gene Transfer Techniques ,Angiopoietin receptor ,Receptor, TIE-2 ,medicine.anatomical_structure ,Hypertension ,cardiovascular system ,Rarefaction ,Disease Progression ,Kidney Diseases ,Cardiology and Cardiovascular Medicine ,medicine.medical_specialty ,Endothelium ,Heart Diseases ,Nitric Oxide Synthase Type III ,Recombinant Fusion Proteins ,Biology ,Nitric oxide ,Physiology (medical) ,Internal medicine ,medicine ,Angiopoietin-1 ,Animals ,Antihypertensive Agents ,Nitrites ,Cartilage oligomeric matrix protein ,Target organ damage ,Original Articles ,Genetic Therapy ,medicine.disease ,Capillaries ,Rats ,Enzyme Activation ,Disease Models, Animal ,Blood pressure ,Endocrinology ,chemistry ,biology.protein ,Endothelium, Vascular - Abstract
Aims The endothelium has emerged recently as a therapeutic target in the treatment of hypertension because endothelial dysfunction and subsequent vascular rarefaction cause target organ damage and further elevate blood pressure (BP). It led us to hypothesize that one of the endothelial survival factors, a potent derivative of angiopoietin-1 (cartilage oligomeric matrix protein, COMP-Ang-1), could be a novel class of antihypertensive agents that maintain endothelial integrity and function, thereby preventing the development of hypertension and target organ damage. Methods and results To study the role of COMP-Ang-1 in preventing hypertension and target organ damage, a COMP-Ang-1 plasmid was electroporated into adductor muscles of 6 weeks old, pre-hypertensive, spontaneously hypertensive rats (SHRs), and the secretion of its expressed protein into the bloodstream was confirmed by western blotting. In comparison with sham and reporter gene transfer, COMP-Ang-1 gene transfer significantly prevented increases in systolic BP and reduced microvascular rarefaction and tissue damage in the heart and kidney. However, overexpression of soluble Tie2 receptor completely abolished these beneficial effects of COMP-Ang-1 gene transfer on SHRs, indicating that expressed COMP-Ang-1 protein has antihypertensive effects in SHRs by binding Tie2 receptors on the vascular endothelium. In particular, COMP-Ang-1 gene-transferred SHRs had significantly higher plasma levels of nitrite than other controls, which was found to be due to that expressed COMP-Ang-1 protein promoted nitrite synthesis by activating endothelial nitric oxide synthase, one of the Tie2 downstream-signalling molecules. Conclusion The present study suggests a new potential of endothelial survival factor, COMP-Ang-1, as an antihypertensive agent that effectively reduces the hypertension-associated cardiovascular and renal damage, as well as prevents the further elevation of BP.
- Published
- 2008
41. Lack of Additional Benefit of Intracoronary Transplantation of Autologous Peripheral Blood Stem Cell in Patients With Acute Myocardial Infarction
- Author
-
Jin-Ho Choi, Hyeon-Cheol Gwon, Wang-Soo Lee, Jin-Oh Choi, Il Rhee, Duk-Kyung Kim, Eun-Seok Jeon, Wonhee Suh, Sang Hoon Lee, Dae-Won Kim, Yeon Hyeon Choe, and Sang-Chol Lee
- Subjects
Adult ,Male ,medicine.medical_specialty ,medicine.medical_treatment ,Myocardial Infarction ,Asymptomatic ,Ventricular Function, Left ,Restenosis ,Internal medicine ,Angioplasty ,Granulocyte Colony-Stimulating Factor ,Humans ,Infusions, Intra-Arterial ,Medicine ,Prospective Studies ,cardiovascular diseases ,Myocardial infarction ,Angioplasty, Balloon, Coronary ,Aged ,Peripheral Blood Stem Cell Transplantation ,Ejection fraction ,business.industry ,Percutaneous coronary intervention ,Stroke Volume ,General Medicine ,Middle Aged ,Hematopoietic Stem Cells ,medicine.disease ,Coronary Vessels ,Surgery ,Survival Rate ,Transplantation ,Treatment Outcome ,surgical procedures, operative ,Acute Disease ,Conventional PCI ,Cardiology ,Female ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business - Abstract
Background Recently the potential of myocardial repair by transplantation of autologous bone marrow stem cells has been suggested. Whether the additional intracoronary transplantation of autologous peripheral blood stem cells (PBSC), which were mobilized by granulocyte-colony-stimulating factor (G-CSF), could safely improve myocardial function in patients with acute myocardial infarction (AMI) was investigated. Methods and Results Seventy-three patients with AMI who had successfully undergone percutaneous coronary intervention (PCI) were enrolled in the present prospective nonrandomized open-labeled study. Ten patients with elective PCI received G-CSF for 4 days followed by intracoronary PBSC transplantation. Thirty-two patients with primary PCI and 31 patients with recent AMI and elective PCI served as controls. The left ventricular (LV) function was evaluated using echocardiography and magnetic resonance imaging. G-CSF and intracoronary transplantation of PBSC did not incur any periprocedural myocardial damage. After 6 months, the LV ejection fraction was significantly improved in the cell therapy group. For 2 years of the follow-up period, there was no adverse clinical events, except one asymptomatic in-stent restenosis. However, comparable improvement of the LV ejection fraction was also identified in the primary PCI and elective PCI control groups. Conclusions In the present study, additional intracoronary infusion of PBSC was safe and feasible for the patients with AMI who had undergone PCI, but did not lead to a significant improvement in LV function compared to standard reperfusion treatment. (Circ J 2007; 71: 486 - 494)
- Published
- 2007
42. Structure-Activity Relationship of Indole-Tethered Pyrimidine Derivatives that Concurrently Inhibit Epidermal Growth Factor Receptor and Other Angiokinases
- Author
-
Kyung Hoon Min, Doran Kim, Jiho Song, Wonhee Suh, Jakyung Yoo, Bong-Yong Lee, Ara Kwon, and Hong Khanh Nguyen
- Subjects
Vascular Endothelial Growth Factor A ,TGF alpha ,Indazoles ,Indoles ,Lung Neoplasms ,lcsh:Medicine ,Angiogenesis Inhibitors ,Biology ,Inhibitory Concentration 50 ,Structure-Activity Relationship ,Growth factor receptor ,Epidermal growth factor ,Carcinoma, Non-Small-Cell Lung ,Human Umbilical Vein Endothelial Cells ,Humans ,Growth factor receptor inhibitor ,Epidermal growth factor receptor ,lcsh:Science ,Protein Kinase Inhibitors ,Cell Proliferation ,Sulfonamides ,Multidisciplinary ,Neovascularization, Pathologic ,integumentary system ,lcsh:R ,ErbB Receptors ,Molecular Docking Simulation ,Vascular endothelial growth factor A ,Pyrimidines ,Biochemistry ,Cancer research ,biology.protein ,lcsh:Q ,A431 cells ,Tyrosine kinase ,Protein Kinases ,Research Article - Abstract
Antiangiogenic agents have been widely investigated in combination with standard chemotherapy or targeted cancer agents for better management of advanced cancers. Therapeutic agents that concurrently inhibit epidermal growth factor receptor and other angiokinases could be useful alternatives to combination therapies for epidermal growth factor receptor-dependent cancers. Here, we report the synthesis of an indole derivative of pazopanib using a bioisosteric replacement strategy, which was designated MKP101. MKP101 inhibited not only the epidermal growth factor receptor with an IC50 value of 43 nM but also inhibited angiokinases as potently as pazopanib. In addition, MKP101 effectively inhibited vascular endothelial growth factor-induced endothelial proliferation, tube formation, migration of human umbilical vein endothelial cells and proliferation of HCC827, an epidermal growth factor receptor-addicted cancer cell line. A docking model of MKP101 and the kinase domain of the epidermal growth factor receptor was generated to predict its binding mode, and validated by synthesizing and evaluating MKP101 derivatives. Additionally, a study of structure-activity relationships of indolylamino or indolyloxy pyrimidine analogues derived from MKP101 demonstrated that selectivity for epidermal growth factor receptor and other angiokinases, especially vascular endothelial growth factor receptor 2 depends on the position of substituents on pyrimidine and the type of link between pyrimidine and the indole moiety. We believe that this study could provide a basis for developing angiokinase inhibitors having high affinity for the epidermal growth factor receptor, from the pyrimidine scaffold.
- Published
- 2015
43. Netrins Promote Developmental and Therapeutic Angiogenesis
- Author
-
Douglas W. Losordo, Gerhardus A H Kock, Tina Thorne, Kirk R. Thomas, Jun Asai, Lise K. Sorensen, Frederic Larrieu-Lahargue, Chi Bin Chien, Marcy Silver, Brent D. Wilson, Arminda Suli, Kye Won Park, Wonhee Suh, Masaaki, Lisa D. Urness, and Dean Y. Li
- Subjects
Vascular Endothelial Growth Factor A ,DNA, Complementary ,Embryo, Nonmammalian ,animal structures ,Angiogenesis ,Neural Conduction ,Neovascularization, Physiologic ,Receptors, Cell Surface ,Article ,Cell Line ,Neovascularization ,Mice ,Diabetic Neuropathies ,Cell Movement ,Ischemia ,Netrin ,medicine ,Animals ,Humans ,Nerve Growth Factors ,Therapeutic angiogenesis ,Muscle, Skeletal ,Zebrafish ,Tube formation ,Multidisciplinary ,biology ,Chemotaxis ,Tumor Suppressor Proteins ,fungi ,Endothelial Cells ,Genetic Therapy ,Netrin-1 ,biology.organism_classification ,Cell biology ,Vascular endothelial growth factor A ,nervous system ,embryonic structures ,Immunology ,Angiogenesis Inducing Agents ,Netrins ,Endothelium, Vascular ,medicine.symptom ,Netrin Receptors ,Diabetic Angiopathies - Abstract
Axonal guidance and vascular patterning share several guidance cues, including proteins in the netrin family. We demonstrate that netrins stimulate proliferation, migration, and tube formation of human endothelial cells in vitro and that this stimulation is independent of known netrin receptors. Suppression of netrin1a messenger RNA in zebrafish inhibits vascular sprouting, implying a proangiogenic role for netrins during vertebrate development. We also show that netrins accelerate neovascularization in an in vivo model of ischemia and that they reverse neuropathy and vasculopathy in a diabetic murine model. We propose that the attractive vascular and neural guidance functions of netrins offer a unique therapeutic potential.
- Published
- 2006
44. A novel chimeric promoter that is highly responsive to hypoxia and metals
- Author
-
Lee Js, Lee Jy, Wonhee Suh, Shin Is, Byun J, Kim Kl, D. K. Kim, Kim Jm, Jeon Es, Lee Ys, and Jang Hs
- Subjects
Genetic enhancement ,Transgene ,DNA, Single-Stranded ,Electrophoretic Mobility Shift Assay ,Enzyme-Linked Immunosorbent Assay ,Biology ,Transfection ,Cell Line ,Mice ,Transcription (biology) ,Cell Line, Tumor ,Gene expression ,Laser-Doppler Flowmetry ,Genetics ,Animals ,Humans ,Inducer ,Luciferase ,Hypoxia ,Promoter Regions, Genetic ,Molecular Biology ,Gene ,Chimera ,Hypoxia-Inducible Factor 1, alpha Subunit ,Molecular biology ,Hindlimb ,DNA-Binding Proteins ,Phosphoglycerate Kinase ,Metals ,Regional Blood Flow ,Cell culture ,Molecular Medicine ,Metallothionein ,Genetic Engineering ,HeLa Cells ,Transcription Factors - Abstract
To develop a potent hypoxia-inducible promoter, we evaluated the usefulness of chimeric combinations of the (Egr-1)-binding site (EBS) from the Egr-1 gene, the metal-response element (MRE) from the metallothionein gene, and the hypoxia-response element (HRE) from the phosphoglycerate kinase 1 gene. In transient transfection assays, combining three copies of HRE (3 x HRE) with either EBS or MRE significantly increased hypoxia responsiveness. When a three-enhancer combination was tested, the EBS-MRE-3 x HRE (E-M-H) gave a hypoxia induction ratio of 69. The expression induced from E-M-H-pGL3 was 2.4-fold higher than that induced from H-pGL3 and even surpassed the expression from a human cytomegalovirus promoter-driven vector. The high inducibility of E-M-H was confirmed by validation studies in different cells and by expressing other cDNAs. Gel shift assays together with functional overexpression studies suggested that increased levels of hypoxia-inducible factor 1alpha, metal transcription factor-1 and Egr-1 may be associated with the high inducibility of the E-M-H chimeric promoter. E-M-H was also induced by hypoxia mimetics such as Co2+ and deferoxamine (DFX) and by hydrogen peroxide. Gene expression from the E-M-H was reversible as shown by the reduced expression of the transgene upon removal of inducers such as hypoxia and DFX. In vivo evaluation of the E-M-H in ischemic muscle revealed that erythropoietin secretion and luciferase and LacZ expression were significantly higher in the E-M-H group than in a control or H group. With its high induction capacity and versatile means of modulation, this novel chimeric promoter should find wide application in the treatment of ischemic diseases and cancer.
- Published
- 2006
45. Monocrotaline-induced pulmonary hypertension correlates with upregulation of connective tissue growth factor expression in the lung
- Author
-
Jonghoe Byun, Koung Li Kim, Jung-Sun Lee, Young-Sam Lee, Hyung-Suk Jang, Jeong-Min Kim, In-Soon Shin, Eun-Seok Jeon, Duk-Kyung Kim, Jeong-a Kim, Jae-Young Lee, Wonhee Suh, and Yeon-Lim Suh
- Subjects
Male ,Pulmonary Metabolism ,Pathology ,medicine.medical_specialty ,Hypertension, Pulmonary ,medicine.medical_treatment ,Clinical Biochemistry ,Connective tissue ,Blood Pressure ,Bronchi ,Pulmonary Artery ,Biology ,Biochemistry ,Immediate-Early Proteins ,Muscle hypertrophy ,Rats, Sprague-Dawley ,Downregulation and upregulation ,Fibrosis ,medicine ,Animals ,Lung ,Molecular Biology ,Monocrotaline ,Reverse Transcriptase Polymerase Chain Reaction ,Growth factor ,Connective Tissue Growth Factor ,Endothelial Cells ,Epithelial Cells ,respiratory system ,medicine.disease ,Pulmonary hypertension ,Rats ,Up-Regulation ,Pulmonary Alveoli ,medicine.anatomical_structure ,Intercellular Signaling Peptides and Proteins ,Molecular Medicine - Abstract
Pulmonary hypertension (PH) is characterized by structural and functional changes in the lung including proliferation of vascular smooth muscle cells (VSMCs) and excessive collagen synthesis. Although connective tissue growth factor (CTGF) is known to promote cell proliferation, migration, adhesion, and extracellular matrix production in various tissues, studies on the role of CTGF in pulmonary hypertension have been limited. Here, we examined CTGF expression in the lung tissues of male Sprague Dawley rats treated with monocrotaline (MCT, 60 microg/kg), a pneumotoxic agent known to induce PH in animals. Establishment of PH was verified by the significantly increased right ventricular systolic pressure and right ventricle/left ventricle weight ratio in the MCT-treated rats. Histological examination of the lung revealed profound muscular hypertrophy in the media of pulmonary artery and arterioles in MCT-treated group. Lung parenchyma, vein, and bronchiole did not appear to be affected. RT-PCR analysis of the lung tissue at 5 weeks indicated significantly increased expression of CTGF in the MCT-treated group. In situ hybridization studies also confirmed abundant CTGF mRNA expression in VSMCs of the arteries and arterioles, clustered pneumocytes, and infiltrated macrophages. Interestingly, CTGF mRNA was not detected in VSMCs of vein or bronchiole. In saline-injected control, basal expression of CTGF was seen in bronchial epithelial cells, alveolar lining cells, and endothelial cells. Taken together, our results suggest that CTGF upregulation in arterial VSMC of the lung might be important in the pathogenesis of pulmonary hypertension. Antagonizing the role of CTGF could thus be one of the potential approaches for the treatment of PH.
- Published
- 2005
46. An Angiogenic, Endothelial-Cell-Targeted Polymeric Gene Carrier
- Author
-
Wonhee Suh, Sung Wan Kim, Lei Yu, and Sang Oh Han
- Subjects
Integrins ,Magnetic Resonance Spectroscopy ,Polymers ,media_common.quotation_subject ,Genetic Vectors ,macromolecular substances ,Gene delivery ,DNA condensation ,Microscopy, Atomic Force ,Transfection ,chemistry.chemical_compound ,Genes, Reporter ,PEG ratio ,Drug Discovery ,Genetics ,Humans ,Receptors, Vitronectin ,Endothelium ,Internalization ,Molecular Biology ,Cells, Cultured ,media_common ,Pharmacology ,Neovascularization, Pathologic ,technology, industry, and agriculture ,Gene Transfer Techniques ,Integrin alphaVbeta3 ,Molecular biology ,Endothelial stem cell ,chemistry ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Biophysics ,Molecular Medicine ,Peptides ,Ethylene glycol ,Oligopeptides ,DNA ,Plasmids - Abstract
Targeting is one of the primary considerations in designing a specific and efficient gene delivery system. Here, an angiogenic endothelial cell-targeted polymeric gene delivery carrier was developed by conjugating an alpha(v)beta3/alpha(v)beta5 integrin-binding RGD peptide, ACDCRGDCFC, into the cationic polymer polyethyleneimine (PEI) via a hydrophilic poly(ethylene glycol) (PEG) spacer. The incorporation of PEG into PEI improved the poor physicochemical properties of PEI-DNA complexes. At a neutral charge ratio, DNA complexes with PEI were polydisperse and substantially aggregated, whereas DNA complexes with PEI-g-1PEG-RGD were homogeneous with 100-200 nm effective diameter. Their surface charge was also significantly reduced due to the charge shielding effect of PEG. However, the extensive grafting of PEI with PEG was shown to inhibit the DNA condensation process, significantly decreasing transfection efficiency. In in vitro transfection experiments with angiogenic endothelial cells, PEI-g-1PEG-RGD showed an approximately fivefold increase in transfection efficiency over PEI, due to an integrin-mediated internalization pathway. PEI-g-1PEG-RGD also exhibited high specificity to angiogenic endothelial cells compared with normal endothelial cells, which was confirmed by in vitro transfection experiments with non-targeting PEI-g-1PEG-RAE in angiostatic endothelial cells.
- Published
- 2002
- Full Text
- View/download PDF
47. Analysis of disease progression-associated gene expression profile in fibrillin-1 mutant mice: new insight into molecular pathogenesis of marfan syndrome
- Author
-
Wonhee Suh, Koung Li Kim, and Chanmi Choi
- Subjects
Marfan syndrome ,musculoskeletal diseases ,Myofibril assembly ,Candidate gene ,Pathology ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,Cytoskeleton organization ,macromolecular substances ,Microarray ,Biochemistry ,Transcriptome ,Drug Discovery ,Gene expression ,medicine ,skin and connective tissue diseases ,Pharmacology ,business.industry ,medicine.disease ,Gene expression profiling ,Molecular Medicine ,Original Article ,mgR mice ,business ,Fibrillin - Abstract
Marfan syndrome (MFS) is a dominantly inherited connective tissue disorder caused by mutations in the gene encoding fibrillin-1 (FBN1) and is characterized by aortic dilatation and dissection, which is the primary cause of death in untreated MFS patients. However, disease progression-associated changes in gene expression in the aortic lesions of MFS patients remained unknown. Using a mouse model of MFS, FBN1 hypomorphic mouse (mgR/mgR), we characterized the aortic gene expression profiles during the progression of the MFS. Homozygous mgR mice exhibited MFS-like phenotypic features, such as fragmentation of elastic fibers throughout the vessel wall and were graded into mgR1-4 based on the pathological severity in aortic walls. Comparative gene expression profiling of WT and four mgR mice using microarrays revealed that the changes in the transcriptome were a direct reflection of the severity of aortic pathological features. Gene ontology analysis showed that genes related to oxidation/reduction, myofibril assembly, cytoskeleton organization, and cell adhesion were differentially expressed in the mgR mice. Further analysis of differentially expressed genes identified several candidate genes whose known roles were suggestive of their involvement in the progressive destruction of aorta during MFS. This study is the first genome-wide analysis of the aortic gene expression profiles associated with the progression of MFS. Our findings provide valuable information regarding the molecular pathogenesis during MFS progression and contribute to the development of new biomarkers as well as improved therapeutic strategies.
- Published
- 2014
48. Anti-JL1 antibody-conjugated poly (l-lysine) for targeted gene delivery to leukemia T cells
- Author
-
Seong Hoe Park, June-Key Chung, Sung Wan Kim, and Wonhee Suh
- Subjects
Antigens, Differentiation, T-Lymphocyte ,Leukemia, T-Cell ,Light ,Antimetabolites ,medicine.drug_class ,Genetic Vectors ,Pharmaceutical Science ,Gene delivery ,Ligands ,Transfection ,Endocytosis ,Monoclonal antibody ,Antigen-Antibody Reactions ,chemistry.chemical_compound ,Antibody Specificity ,Tumor Cells, Cultured ,medicine ,Humans ,Scattering, Radiation ,Polylysine ,Amino Acids ,Ganciclovir ,Electrophoresis, Agar Gel ,Microscopy, Confocal ,biology ,Flow Cytometry ,Ligand (biochemistry) ,Molecular biology ,In vitro ,Biochemistry ,chemistry ,Gene Targeting ,biology.protein ,Antibody ,DNA ,Plasmids - Abstract
We have designed the gene delivery carrier targeted to Molt 4 cells, human leukemia T cells, using monoclonal antibody against leukemia-specific JL1 antigen, anti-JL1 antibody, as a targeting moiety. Anti-JL1 antibody has been proven to bind to JL1 antigen and subsequently be internalized into Molt 4 cells, demonstrating that anti-JL1 antibody has the potential as a targeting ligand for leukemia-specific gene transfer. Anti-JL1 antibody was modified with the heterobifunctional crosslinker, PDPH, at carbohydrate sites and conjugated to thiolated poly- l -lysine (PLL) via disulfide bridges. The composition and antigen binding affinity of antibody–PLL conjugates were analyzed by the amino acid analysis and the flow cytometry, respectively. Antibody–PLL conjugates neutralized pSV-β-galactosidase plasmid DNA at 5:1 weight ratio and condensed into about 200–300-nm complexes. DNA/antibody–PLL complexes were effectively internalized into Molt 4 cells after 4 h incubation at 37°C and showed significantly higher in vitro transfection efficiency than DNA/PLL complexes and DNA/Lipofectin™ formulation due to the targeting effect of receptor-mediated endocytosis induced by anti-JL1 antibody.
- Published
- 2001
49. Lag time data for characterizing the pore pathway of intact and chemically pretreated human epidermal membrane
- Author
-
William I. Higuchi, Abdel-Halim Ghanem, Kendall D. Peck, Hemanshu H Parikh, Samir C. Mehta, Wonhee Suh, and S. Kevin Li
- Subjects
integumentary system ,Chemistry ,Pharmaceutical Science ,Absorption (skin) ,Membrane transport ,Permeation ,Tortuosity ,medicine.anatomical_structure ,Membrane ,Biochemistry ,Permeability (electromagnetism) ,medicine ,Stratum corneum ,Biophysics ,Epidermis - Abstract
This study aimed to gain mechanistic insights into the nature of the pore pathway of fully hydrated human stratum corneum from lag time data obtained using a model polar permeant, urea. Lag times were deduced from transport experiments with human epidermal membranes and with human epidermal membranes after ethanol or chloroform–methanol treatment. A tortuous pore pathway transport model and a `bottleneck' transport model were employed for data analysis, and their appropriateness for the observed data was examined. Important outcomes from the present study with intact and with delipidized stratum corneum were as follows. Long lag times (around 60–800 min) for the transport of urea in human epidermal membranes were generally observed. These results were consistent with an extremely tortuous pore pathway as would be expected if it is associated with the polar/aqueous region of the stratum corneum intercellular lipids (i.e. the bilayers in the intercellular region). The permeability of the stratum corneum increased after ethanol treatment, and, at the same time, the tortuosity decreased but remained relatively high. Chloroform–methanol treatment further increased the permeability and further decreased the tortuosity. Since delipidization by ethanol and chloroform–methanol treatments decreased the tortuosity of the pore pathway, these results suggest that the effectively highly tortuous pathway for polar permeants in stratum corneum may be associated with the polar regions of the intercellular lipids. Untreated skin samples that had high electrical resistance were observed to have longer lag times than those with low resistance; this is consistent with the hypothesis that skin samples of high resistance have less appendage routes or less damage and transport polar permeants predominantly via the tortuous pathways involving the intercellular lipid regions of the stratum corneum. Neither the tortuous pathway transport model alone nor the `bottleneck' transport model alone seems to perfectly represent the experimental data, and a modified model (a hybrid of the two models) has been proposed to be more consistent with the lag time data and the morphology of fully hydrated stratum corneum. The present study has demonstrated the usefulness of lag times obtained with a polar permeant in better understanding the transport mechanisms involved with the pore pathway.
- Published
- 1998
50. Cooperation of Endothelial and Smooth Muscle Cells Derived from Human Induced Pluripotent Stem Cells Enhances Neovascularization in Dermal Wounds
- Author
-
Wonhee Suh, Sun-Hwa Song, Koung Li Kim, and Kyu-Sil Choi
- Subjects
Male ,Cellular differentiation ,Induced Pluripotent Stem Cells ,Myocytes, Smooth Muscle ,Biomedical Engineering ,Mice, Nude ,Neovascularization, Physiologic ,Bioengineering ,Biology ,Biochemistry ,Biomaterials ,Neovascularization ,Mice ,medicine ,Animals ,Humans ,Induced pluripotent stem cell ,Skin ,Tissue Engineering ,Regeneration (biology) ,Endothelial Cells ,Original Articles ,Embryonic stem cell ,Cell biology ,Transplantation ,Immunology ,medicine.symptom ,Wound healing ,Reprogramming - Abstract
Human induced pluripotent stem cells (hiPSCs) are generated through the reprogramming of somatic cells into an embryonic stem cell-like state, such that vascular cells differentiated from hiPSCs might be a suitable autologous cell source for vascular regeneration. The goal of this study was to assess whether cotransplantation of endothelial cells (ECs) and smooth muscle cells (SMCs) differentiated from hiPSCs could promote neovascularization and tissue repair in a murine dermal wound model. hiPSCs were differentiated into ECs and SMCs; the differentiated cells displayed cell-specific surface markers. Compared to primary somatic cells, ECs and SMCs, which were differentiated from hiPSCs, strongly cooperated to enhance in vitro tubular network formation. In vivo gel assays in athymic nude mice showed that the coimplantation of differentiated ECs and SMCs significantly increased vascularization, unlike that observed in the case of implantation of differentiated ECs alone. In a murine full-thickness wound model, when compared with the transplantation of primary somatic cells or phosphate-buffered saline, cotransplantation of differentiated ECs and SMCs markedly enhanced neovascularization in injured tissues and accelerated wound healing. These results demonstrate that cotransplantation of hiPSC-derived ECs and SMCs may be feasible as a new autologous cell therapy for neovascularization and tissue repair.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.