Network


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

Hotspot


Dive into the research topics where Seoyeon Bok is active.

Publication


Featured researches published by Seoyeon Bok.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Transcriptional activation of hypoxia-inducible factor-1 (HIF-1) in myeloid cells promotes angiogenesis through VEGF and S100A8.

G-One Ahn; Jun Seita; Hong Bj; You-Me Kim; Seoyeon Bok; Chan-Ju Lee; Kwang S. Kim; Jonghyeob Lee; Nicholas J. Leeper; John P. Cooke; Hyo Jin Kim; Iktae Kim; Irving L. Weissman; Janice M. Brown

Significance Here, we are reporting our findings that hypoxia-inducible factor 1 (HIF-1) activation in monocytes promotes neovascularization in matrigel and improves blood flow in hindlimb ischemia through production of vascular endothelial growth factor and S100A8. We found that HIF-1 regulates S100A8 expression specifically in monocytes isolated from our unique strain of transgenic mice targeting HIF pathways. Emerging evidence indicates that myeloid cells are essential for promoting new blood vessel formation by secreting various angiogenic factors. Given that hypoxia-inducible factor (HIF) is a critical regulator for angiogenesis, we questioned whether HIF in myeloid cells also plays a role in promoting angiogenesis. To address this question, we generated a unique strain of myeloid-specific knockout mice targeting HIF pathways using human S100A8 as a myeloid-specific promoter. We observed that mutant mice where HIF-1 is transcriptionally activated in myeloid cells (by deletion of the von Hippel–Lindau gene) resulted in erythema, enhanced neovascularization in matrigel plugs, and increased production of vascular endothelial growth factor (VEGF) in the bone marrow, all of which were completely abrogated by either genetic or pharmacological inactivation of HIF-1. We further found that monocytes were the major effector producing VEGF and S100A8 proteins driving neovascularization in matrigel. Moreover, by using a mouse model of hindlimb ischemia we observed significantly improved blood flow in mice intramuscularly injected with HIF-1–activated monocytes. This study therefore demonstrates that HIF-1 activation in myeloid cells promotes angiogenesis through VEGF and S100A8 and that this may become an attractive therapeutic strategy to treat diseases with vascular defects.


ACS Nano | 2015

Quantum Dots in an Amphiphilic Polyethyleneimine Derivative Platform for Cellular Labeling, Targeting, Gene Delivery, and Ratiometric Oxygen Sensing.

Joonhyuck Park; Junhwa Lee; Jungheon Kwag; Yeonggyeong Baek; Bumju Kim; Calvin J. Yoon; Seoyeon Bok; So-Hye Cho; Ki Hean Kim; G-One Ahn; Sungjee Kim

Amphiphilic polyethyleneimine derivatives (amPEIs) were synthesized and used to encapsulate dozens of quantum dots (QDs). The QD-amPEI composite was ∼100 nm in hydrodynamic diameter and had the slightly positive outer surface that suited well for cellular internalization. The QD-amPEI showed very efficient QD cellular labeling with the labeled cell fluorescence intensity more than 10 times higher than conventional techniques such as Lipofectamine-assisted QD delivery. QD-amPEI was optimal for maximal intracellular QD delivery by the large QD payload and the rapid endocytosis kinetics. QD-amPEI platform technology was demonstrated for gene delivery, cell-specific labeling, and ratiometric oxygen sensing. Our QD-amPEI platform has two partitions: positive outer surface and hydrophobic inside pocket. The outer positive surface was further exploited for gene delivery and targeting. Co-delivery of QDs and GFP silencing RNAs was successfully demonstrated by assembling siRNAs to the outer surfaces, which showed the transfection efficiency an order of magnitude higher than conventional gene transfections. Hyaluronic acids were tethered onto the QD-amPEI for cell-specific targeted labeling which showed the specific-to-nonspecific signal ratio over 100. The inside hydrophobic compartment was further applied for cohosting oxygen sensing phosphorescence Ru dyes along with QDs. The QD-Ru-amPEI oxygen probe showed accurate and reversible oxygen sensing capability by the ratiometric photoluminescence signals, which was successfully applied to cellular and spheroid models.


Radiation oncology journal | 2016

Tumor hypoxia and reoxygenation: the yin and yang for radiotherapy

Beom-Ju Hong; Jeongwoo Kim; Hoibin Jeong; Seoyeon Bok; Young-Eun Kim; G-One Ahn

Tumor hypoxia, a common feature occurring in nearly all human solid tumors is a major contributing factor for failures of anticancer therapies. Because ionizing radiation depends heavily on the presence of molecular oxygen to produce cytotoxic effect, the negative impact of tumor hypoxia had long been recognized. In this review, we will highlight some of the past attempts to overcome tumor hypoxia including hypoxic radiosensitizers and hypoxia-selective cytotoxin. Although they were (still are) a very clever idea, they lacked clinical efficacy largely because of ‘reoxygenation’ phenomenon occurring in the conventional low dose hyperfractionation radiotherapy prevented proper activation of these compounds. Recent meta-analysis and imaging studies do however indicate that there may be a significant clinical benefit in lowering the locoregional failures by using these compounds. Latest technological advancement in radiotherapy has allowed to deliver high doses of radiation conformally to the tumor volume. Although this technology has brought superb clinical responses for many types of cancer, recent modeling studies have predicted that tumor hypoxia is even more serious because ‘reoxygenation’ is low thereby leaving a large portion of hypoxic tumor cells behind. Wouldn’t it be then reasonable to combine hypoxic radiosensitizers and/or hypoxia-selective cytotoxin with the latest radiotherapy? We will provide some preclinical and clinical evidence to support this idea hoping to revamp an enthusiasm for hypoxic radiosensitizers or hypoxia-selective cytotoxins as an adjunct therapy for radiotherapy.


Blood Research | 2016

Radiation-induced immune responses: mechanisms and therapeutic perspectives

Hoibin Jeong; Seoyeon Bok; Beom-Ju Hong; Hyung-Seok Choi; G-One Ahn

Recent advancement in the radiotherapy technology has allowed conformal delivery of high doses of ionizing radiation precisely to the tumors while sparing large volume of the normal tissues, which have led to better clinical responses. Despite this technological advancement many advanced tumors often recur and they do so within the previously irradiated regions. How could tumors recur after receiving such high ablative doses of radiation? In this review, we outlined how radiation can elicit anti-tumor responses by introducing some of the cytokines that can be induced by ionizing radiation. We then discuss how tumor hypoxia, a major limiting factor responsible for failure of radiotherapy, may also negatively impact the anti-tumor responses. In addition, we highlight how there may be other populations of immune cells including regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs) that can be recruited to tumors interfering with the anti-tumor immunity. Finally, the impact of irradiation on tumor hypoxia and the immune responses according to different radiotherapy regimen is also delineated. It is indeed an exciting time to see that radiotherapy is being combined with immunotherapy in the clinic and we hope that this review can add an excitement to the field.


Scientific Reports | 2016

Moxifloxacin: Clinically compatible contrast agent for multiphoton imaging

Taejun Wang; Won Hyuk Jang; Seunghun Lee; Calvin J. Yoon; Jun Ho Lee; Bumju Kim; Sekyu Hwang; Chun-Pyo Hong; Yeoreum Yoon; Gilgu Lee; Viet-Hoan Le; Seoyeon Bok; G-One Ahn; Jaewook Lee; Yong Song Gho; Euiheon Chung; Sungjee Kim; Myoung Ho Jang; Seung-Jae Myung; Myoung Joon Kim; Peter T. C. So; Ki Hean Kim

Multiphoton microscopy (MPM) is a nonlinear fluorescence microscopic technique widely used for cellular imaging of thick tissues and live animals in biological studies. However, MPM application to human tissues is limited by weak endogenous fluorescence in tissue and cytotoxicity of exogenous probes. Herein, we describe the applications of moxifloxacin, an FDA-approved antibiotic, as a cell-labeling agent for MPM. Moxifloxacin has bright intrinsic multiphoton fluorescence, good tissue penetration and high intracellular concentration. MPM with moxifloxacin was demonstrated in various cell lines, and animal tissues of cornea, skin, small intestine and bladder. Clinical application is promising since imaging based on moxifloxacin labeling could be 10 times faster than imaging based on endogenous fluorescence.


Biomedical Optics Express | 2015

In vivo imaging of activated microglia in a mouse model of focal cerebral ischemia by two-photon microscopy.

Seoyeon Bok; Taejun Wang; Chan-Ju Lee; Seong-Uk Jeon; Young-Eun Kim; Jeongwoo Kim; Beom-Ju Hong; Calvin J. Yoon; Sungjee Kim; Seunghoon Lee; Hak Jae Kim; Il Han Kim; Ki Hean Kim; G-One Ahn

Microglia are brain resident macrophages rapidly responding to various stimuli to exert appropriate inflammatory responses. Although they have recently been exploited as an attractive candidate for imaging neuroinflammation, it is still difficult to visualize them at the cellular and molecular levels. Here we imaged activated microglia by establishing intracranial window chamber (ICW) in a mouse model of focal cerebral ischemia by using two-photon microscopy (TPM), in vivo. Intravenous injection of fluorescent antibodies allowed us to detect significantly elevated levels of Iba-1 and CD68 positive activated microglia in the ipsilateral compared to the contralateral side of the infarct. We further observed that indomethacin, a non-steroidal anti-inflammatory drug significantly attenuated CD68-positive microglial activation in ICW, which was further confirmed by qRT-PCR biochemical analyses. In conclusion, we believe that in vivo TPM imaging of ICW would be a useful tool to screen for therapeutic interventions lowering microglial activation hence neuroinflammation.


Oncotarget | 2017

Hypoxia-inducible factor-1α regulates microglial functions affecting neuronal survival in the acute phase of ischemic stroke in mice

Seoyeon Bok; Young-Eun Kim; Youngsik Woo; Soeun Kim; Suk-Jo Kang; Yoontae Lee; Sang Ki Park; Irving L. Weissman; G-One Ahn

Cells universally adapt to ischemic conditions by turning on a transcription factor hypoxia-inducible factor (HIF), in which its role is known to differ widely across many different types of cells. Given that microglia have been reported as an essential mediator of neuroinflammation in many brain diseases, we examined the role of HIF in microglia in the progression of an acute phase of ischemic stroke by challenging our novel strains of myeloid-specific Hif-1α or Hif-2α knockout (KO) mice created by Cre-loxP system via middle cerebral artery occlusion (MCAO). We observed that Hif-1α but not Hif-2α KO mice exhibited an improved recovery compared to wild-type (WT) mice determined by behavioral tests. Immunostaining analyses revealed that there were increased numbers of both mature and immature neurons while microglia and apoptotic cells were significantly decreased in the dentate gyrus of Hif-1α KO mice following MCAO. By isolating microglia with fluorescence-activated cell sorter, we found that HIF-1α-deficient microglia were impaired in phagocytosis, reactive oxygen species (ROS) production, and tumor necrosis factor-α (TNF-α) secretion. We further observed a significant decrease in the expression of Cd36 and milk fat globule-epidermal growth factor 8 (Mfg-e8) genes, both of which contain hypoxia-responsive element (HRE). Knocking down either of these genes in BV2 microglial cells was sufficient to abrogate HIF-mediated increase in phagocytosis, production of intracellular ROS, or TNF-α secretion. Our results therefore suggest that HIF-1α in microglia is a novel therapeutic target to protect neuronal survival following an acute phase of ischemic stroke.


RSC Advances | 2018

Supramolecular hydrogels encapsulating bioengineered mesenchymal stem cells for ischemic therapy

Byung Woo Hwang; Young-Eun Kim; Mungu Kim; Seulgi Han; Seoyeon Bok; Kyeng Min Park; Annadka Shrinidhi; Ki Su Kim; G-One Ahn; Sei Kwang Hahn

We developed supramolecular hyaluronate (HA) hydrogels to encapsulate genetically engineered mesenchymal stem cells (MSCs) for the treatment of limb ischemia. In vivo angiogenic factors could be produced stably by the bioengineered MSCs (BMSCs) within the supramolecular hydrogels showing effective vascular repair and enhanced blood perfusion.


Cancer Research | 2015

Abstract A13: Hypoxia-inducible factor-1 (HIF-1) in myeloid cells promotes angiogenesis by regulating VEGF and S100A8 production

G-One Ahn; Young-Eun Kim; Beom-Ju Hong; Seoyeon Bok; Chan-Ju Lee; Hak Jae Kim; Il Han Kim; Jun Seita; Irving L. Weissman; J. Martin Brown

Myeloid cells (cells that give rise to monocytes and macrophages) are a critical component in the solid tumor microenvironment, promoting angiogenesis and tumor recurrence to therapies. Recent literature have extensively demonstrated that hypoxia-inducible factor (HIF) is a key transcription factor in myeloid cells in responding to hypoxic stimuli including solid tumors and inflammation by secreting various cytokines and growth factors. Although a number of studies have reported that hypoxia exposure to primary macrophages induces HIF-1 activation and subsequent vascular endothelial growth factor (VEGF) production, there had been no mouse model to demonstrate this phenomenon. To better understand the role of transcriptional activation of HIF in pathological macrophages, we have created a new strain of myeloid-specific knockout (KO) mice targeting HIF pathways using hS100A8 as the myeloid promoter. S100A8 is an intracellular calcium binding protein and its expression has been heavily detected in pathological macrophages of many diseases including solid tumors, inflammatory bowel disease, obesity, and rheumatoid arthritis. Upon generating mice deficient for von Hippel Lindau (pVHL) tumor suppressor, the negative regulator of HIF, in myeloid cells, we observed erythema and increased VEGF expression in the bone marrow lysate. Moreover, these mice exhibited an enhanced angiogenesis in the subcutanouesly implanted matrigel plugs, which was accompanied by increased VEGF-VEGFR2 signaling in matrigel. We further found that these phenotypes were dependent on transcriptional activation of HIF-1 as a pharmacological or genetic inhibition of HIF-1α completely suppressed the phenotypes in mice deficient for pVHL in myeloid cells. Importantly, we found that HIF-1 activation in myeloid cells regulate not only VEGF but also S100A8 production and identified that monocytes were the major effector driving angiogenesis. Together these results suggest that transcriptional activation of HIF-1 in myeloid cells plays a critical role in promoting angiogenesis. We are currently investigating how HIF-1 in myeloid cells regulates tumor microenvironment thereby affecting tumor progression. Citation Format: G-One Ahn, Young-Eun Kim, Beom-Ju Hong, Seoyeon Bok, Chan-Ju Lee, Hak Jae Kim, Il Han Kim, Jun Seita, Irving Weissman, J Martin Brown. Hypoxia-inducible factor-1 (HIF-1) in myeloid cells promotes angiogenesis by regulating VEGF and S100A8 production. [abstract]. In: Abstracts: AACR Special Conference on Cellular Heterogeneity in the Tumor Microenvironment; 2014 Feb 26-Mar 1; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2015;75(1 Suppl):Abstract nr A13. doi:10.1158/1538-7445.CHTME14-A13


International Journal of Radiation Oncology Biology Physics | 2016

Real-time Tumor Oxygenation Changes After Single High-dose Radiation Therapy in Orthotopic and Subcutaneous Lung Cancer in Mice: Clinical Implication for Stereotactic Ablative Radiation Therapy Schedule Optimization

Changhoon Song; Beom-Ju Hong; Seoyeon Bok; Chan-Ju Lee; Young-Eun Kim; Sang-Rok Jeon; Hong-Gyun Wu; Yun-Sang Lee; Gi Jeong Cheon; Jin Chul Paeng; David J. Carlson; Hak Jae Kim; G-One Ahn

Collaboration


Dive into the Seoyeon Bok's collaboration.

Top Co-Authors

Avatar

G-One Ahn

Pohang University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Beom-Ju Hong

Pohang University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Chan-Ju Lee

Pohang University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Hak Jae Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Seong-Uk Jeon

Pohang University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Calvin J. Yoon

Pohang University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Il Han Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Ki Hean Kim

Pohang University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Sungjee Kim

Pohang University of Science and Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge