Network


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

Hotspot


Dive into the research topics where Bum Ju Ahn is active.

Publication


Featured researches published by Bum Ju Ahn.


Biochemical and Biophysical Research Communications | 2009

Ninjurin1 is expressed in myeloid cells and mediates endothelium adhesion in the brains of EAE rats

Bum Ju Ahn; Hyo-Jong Lee; Min Wook Shin; Jeong-Hyun Choi; Joo-Won Jeong; Kyu-Won Kim

Ninjurin1 (nerve injury-induced protein, Ninj1) is an adhesion molecule that is essential for cell-to-cell interactions. However, little is known about the function of Ninj1 in the central nervous system (CNS). To address its role in the CNS, we analyzed the expression pattern of Ninj1 in normal rats and in an experimental autoimmune encephalomyelitis (EAE) model. Ninj1 was expressed in three major compartments of brains, meninges, the choroid plexus, and parenchymal perivascular spaces. In the EAE brains, Ninj1 was strongly expressed in myeloid cells (macrophages/monocytes and neutrophils) and partially expressed in endothelial cells (ECs). Furthermore, Ninj1 enhanced adhesion between BV2 cells (murine monocyte lineage microglia) and HBMECs (human brain microvascular endothelial cells). Collectively, our findings suggest that Ninj1 may mediate the entry of myeloid cells into the CNS in normal and EAE brains, and it is a potential therapeutic target for regulating myeloid cell trafficking across the blood-brain barrier (BBB) in CNS immune processes.


Molecules and Cells | 2010

Ninjurin1: a potential adhesion molecule and its role in inflammation and tissue remodeling

Hyo-Jong Lee; Bum Ju Ahn; Min Wook Shin; Jeong-Hyun Choi; Kyu-Won Kim

Nerve injury induced protein 1, Ninj1 (Ninjurin1) is a cell surface protein that is induced by nerve injury and promotes axonal growth in the peripheral nervous system. However, the function of Ninj1 in the vascular system and central nervous system (CNS) is incompletely understood. Here we review recent studies that have shed further light on the role and regulation of Ninj1 in vascular remodeling and inflammation. Increasing evidence suggests that Ninj1 mediates cell communication and enhances the entry, migration, and activity of leukocytes such as monocytes and macrophages in developmental processes and inflammatory responses. Moreover, our recent studies show that Ninj1 regulates close interaction between leukocytes and vascular endothelial cells in vascular remodeling and inflamed CNS. Additionally, Ninj1 enhances the apoptosis-inducing activity of leukocytes and is cleaved by MMPs, resulting in loss of adhesion during tissue remodeling. The collective data described here show that Ninj1 is required for the entry, adhesion, activation, and movement of leukocytes during tissue remodeling and might be a potential therapeutic target to regulate the adhesion and trafficking of leukocytes in inflammation and leukocyte-mediated diseases such as multiple sclerosis, diabetic retinopathy, and neuropathy.


Journal of Biological Chemistry | 2014

Ninjurin1 Deficiency Attenuates Susceptibility of Experimental Autoimmune Encephalomyelitis in Mice

Bum Ju Ahn; Hoang Le; Min Wook Shin; Sung Jin Bae; Eun Lee; Hee-Jun Wee; Jong Ho Cha; Hyo Jong Lee; Hye Shin Lee; Jeong Hun Kim; Chang Yeon Kim; Ji Hae Seo; Eng H. Lo; Mi Ni Lee; Goo Taeg Oh; Guo Nan Yin; Ji Kan Ryu; Jun-Kyu Suh; Kyu-Won Kim

Background: Effect of Ninjurin1 deletion in the experimental autoimmune encephalomyelitis (EAE) mice has not been examined. Results: Ninjurin1 knock-out (KO) mice are resistance to EAE due to a defect of leukocyte recruitment into lesion sites. Conclusion: Ninjurin1 is a potent target molecule for treating inflammatory diseases such as multiple sclerosis. Significance: Our study proved contribution of Ninjurin1 in EAE pathogenesis in vivo and supports the importance of its targeting strategies. Ninjurin1 is a homotypic adhesion molecule that contributes to leukocyte trafficking in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. However, in vivo gene deficiency animal studies have not yet been done. Here, we constructed Ninjurin1 knock-out (KO) mice and investigated the role of Ninjurin1 on leukocyte trafficking under inflammation conditions such as EAE and endotoxin-induced uveitis. Ninjurin1 KO mice attenuated EAE susceptibility by reducing leukocyte recruitment into the injury regions of the spinal cord and showed less adhesion of leukocytes on inflamed retinal vessels in endotoxin-induced uveitis mice. Moreover, the administration of a custom-made antibody (Ab26–37) targeting the Ninjurin1 binding domain ameliorated the EAE symptoms, showing the contribution of its adhesion activity to leukocyte trafficking. In addition, we addressed the transendothelial migration (TEM) activity of bone marrow-derived macrophages and Raw264.7 cells according to the expression level of Ninjurin1. TEM activity was decreased in Ninjurin1 KO bone marrow-derived macrophages and siNinj1 Raw264.7 cells. Consistent with this, GFP-tagged mNinj1-overexpressing Raw264.7 cells increased their TEM activity. Taken together, we have clarified the contribution of Ninjurin1 to leukocyte trafficking in vivo and delineated its direct functions to TEM, emphasizing Ninjurin1 as a beneficial therapeutic target against inflammatory diseases such as multiple sclerosis.


PLOS ONE | 2014

AKAP12 Mediates Barrier Functions of Fibrotic Scars during CNS Repair

Jong Ho Cha; Hee-Jun Wee; Ji Hae Seo; Bum Ju Ahn; Ji Hyeon Park; Jun Mo Yang; Sae Won Lee; Eun Hee Kim; Ok Hee Lee; Ji Hoe Heo; Hyo Jong Lee; Irwin H. Gelman; Ken Arai; Eng H. Lo; Kyu-Won Kim

The repair process after CNS injury shows a well-organized cascade of three distinct stages: inflammation, new tissue formation, and remodeling. In the new tissue formation stage, various cells migrate and form the fibrotic scar surrounding the lesion site. The fibrotic scar is known as an obstacle for axonal regeneration in the remodeling stage. However, the role of the fibrotic scar in the new tissue formation stage remains largely unknown. We found that the number of A-kinase anchoring protein 12 (AKAP12)-positive cells in the fibrotic scar was increased over time, and the cells formed a structure which traps various immune cells. Furthermore, the AKAP12-positive cells strongly express junction proteins which enable the structure to function as a physical barrier. In in vivo validation, AKAP12 knock-out (KO) mice showed leakage from a lesion, resulting from an impaired structure with the loss of the junction complex. Consistently, focal brain injury in the AKAP12 KO mice led to extended inflammation and more severe tissue damage compared to the wild type (WT) mice. Accordingly, our results suggest that AKAP12-positive cells in the fibrotic scar may restrict excessive inflammation, demonstrating certain mechanisms that could underlie the beneficial actions of the fibrotic scar in the new tissue formation stage during the CNS repair process.


PLOS ONE | 2014

Nuclear translocation of hARD1 contributes to proper cell cycle progression.

Ji Hyeon Park; Ji Hae Seo; Hee-Jun Wee; Tam Thuy Lu Vo; Eun Lee; Hoon Choi; Jong Ho Cha; Bum Ju Ahn; Min Wook Shin; Sung Jin Bae; Kyu-Won Kim

Arrest defective 1 (ARD1) is an acetyltransferase that is highly conserved across organisms, from yeasts to humans. The high homology and widespread expression of ARD1 across multiple species and tissues signify that it serves a fundamental role in cells. Human ARD1 (hARD1) has been suggested to be involved in diverse biological processes, and its role in cell proliferation and cancer development has been recently drawing attention. However, the subcellular localization of ARD1 and its relevance to cellular function remain largely unknown. Here, we have demonstrated that hARD1 is imported to the nuclei of proliferating cells, especially during S phase. Nuclear localization signal (NLS)-deleted hARD1 (hARD1ΔN), which can no longer access the nucleus, resulted in cell morphology changes and cellular growth impairment. Notably, hARD1ΔN-expressing cells showed alterations in the cell cycle and the expression levels of cell cycle regulators compared to hARD1 wild-type cells. Furthermore, these effects were rescued when the nuclear import of hARD1 was restored by exogenous NLS. Our results show that hARD1 nuclear translocation mediated by NLS is required for cell cycle progression, thereby contributing to proper cell proliferation.


Nature Communications | 2014

Prompt meningeal reconstruction mediated by oxygen-sensitive AKAP12 scaffolding protein after central nervous system injury

Jong Ho Cha; Hee-Jun Wee; Ji Hae Seo; Bum Ju Ahn; Ji Hyeon Park; Jun Mo Yang; Sae Won Lee; Ok Hee Lee; Hyo Jong Lee; Irwin H. Gelman; Ken Arai; Eng H. Lo; Kyu-Won Kim

The meninges forms a critical epithelial barrier, which protects the central nervous system (CNS), and therefore its prompt reconstruction after CNS injury is essential for reducing neuronal damage. Meningeal cells migrate into the lesion site after undergoing an epithelial-mesenchymal transition (EMT) and repair the impaired meninges. However, the molecular mechanisms of meningeal EMT remain largely undefined. Here we show that TGF-β1 and retinoic acid (RA) released from the meninges, together with oxygen tension, could constitute the mechanism for rapid meningeal reconstruction. AKAP12 is an effector of this mechanism, and its expression in meningeal cells is regulated by integrated upstream signals composed of TGF-β1, RA and oxygen tension. Functionally, AKAP12 modulates meningeal EMT by regulating the TGF-β1-non-Smad-SNAI1 signalling pathway. Collectively, TGF-β1, RA and oxygen tension can modulate the dynamic change in AKAP12 expression, causing prompt meningeal reconstruction after CNS injury by regulating the transition between the epithelial and mesenchymal states of meningeal cells.


Biochemical and Biophysical Research Communications | 2012

The N-terminal ectodomain of Ninjurin1 liberated by MMP9 has chemotactic activity

Bum Ju Ahn; Hoang Le; Min Wook Shin; Sung Jin Bae; Eun Lee; Hee-Jun Wee; Jong Ho Cha; Ji Hyeon Park; Hye Shin Lee; Hyo Jong Lee; Hyunsook Jung; Zee Yong Park; Sang Ho Park; Byung Woo Han; Ji Hae Seo; Eng H. Lo; Kyu-Won Kim

Ninjurin1 is known as an adhesion molecule promoting leukocyte trafficking under inflammatory conditions. However, the posttranslational modifications of Ninjurin1 are poorly understood. Herein, we defined the proteolytic cleavage of Ninjurin1 and its functions. HEK293T cells overexpressing the C- or N-terminus tagging mouse Ninjurin1 plasmid produced additional cleaved forms of Ninjurin1 in the lysates or conditioned media (CM). Two custom-made anti-Ninjurin1 antibodies, Ab(1-15) or Ab(139-152), specific to the N- or C-terminal regions of Ninjurin1 revealed the presence of its shedding fragments in the mouse liver and kidney lysates. Furthermore, Matrix Metalloproteinase (MMP) 9 was responsible for Ninjurin1 cleavage between Leu(56) and Leu(57). Interestingly, the soluble N-terminal Ninjurin1 fragment has structural similarity with well-known chemokines. Indeed, the CM from HEK293T cells overexpressing the GFP-mNinj1 plasmid was able to attract Raw264.7 cells in trans-well assay. Collectively, we suggest that the N-terminal ectodomain of mouse Ninjurin1, which may act as a chemoattractant, is cleaved by MMP9.


Molecular Medicine Reports | 2015

Meteorin is upregulated in reactive astrocytes and functions as a negative feedback effector in reactive gliosis

Hye Shin Lee; Soon Hee Lee; Jong Ho Cha; Ji Hae Seo; Bum Ju Ahn; Kyu-Won Kim

Reactive gliosis is a glial response to a wide range of central nervous system insults, which results in cellular and molecular changes to resting glial cells. Despite its fundamental effect on neuropathologies, the identification and characterization of the molecular mechanisms underlying this process remain to be fully elucidated. The aim of the present study was to analyze the expression profile and functions of the astrocytic neurotrophic factor, meteorin, in the progression of reactive gliosis. A mouse model of photothrombotic ischemia, and a primary astrocyte culture were used in the present study. Reverse transcription quantitative polymerase chain reaction, western blotting and immunofluorescence staining were performed to examine the expression levels of meteorin and reactive gliosis markers. Increased expression levels of meteorin were observed in reactive astrocytes in a photothrombotic ischemia mouse model, as well as in cultured astrocytes, which were stimulated by transforming growth factor-β1. Exogenous treatment of the astrocytes with meteorin did not induce janus kinase-signal transducer and activator of transcription 3 signaling, however, silencing the expression of meteorin in the astrocytes resulted in an upregulation of reactive astrocyte markers, including glial fibrillary acidic protein and S100β, indicating that endogenous meteorin is required for the maintenance of astrocytic homeostasis. These results suggested a novel role for meteorin as a negative feedback effector in reactive gliosis.


International Journal of Oncology | 2015

Autoacetylation regulates differentially the roles of ARD1 variants in tumorigenesis

Ji Hae Seo; Ji Hyeon Park; Eun Lee; Tam Thuy Lu Vo; Hoon Choi; Jae Kyung Jang; Hee-Jun Wee; Bum Ju Ahn; Jong Ho Cha; Min Wook Shin; Kyu-Won Kim

ARD1 is an acetyltransferase with several variants derived from alternative splicing. Among ARD1 variants, mouse ARD1(225) (mARD1(225)), mouse ARD1(235) (mARD1(235)), and human ARD1(235) (hARD1(235)) have been the most extensively characterized and are known to have different biological functions. In the present study, we demonstrated that mARD1(225), mARD1(235), and hARD1(235) have conserved autoacetylation activities, and that they selectively regulate distinct roles of ARD1 variants in tumorigenesis. Using purified recombinants for ARD1 variants, we found that mARD1(225), mARD1(235), and hARD1(235) undergo similar autoacetylation with the target site conserved at the Lys136 residue. Moreover, functional investigations revealed that the role of mARD1(225) autoacetylation is completely distinguishable from that of mARD1(235) and hARD1(235). Under hypoxic conditions, mARD1(225) autoacetylation inhibited tumor angiogenesis by decreasing the stability of hypoxia-inducible factor-1α (HIF-1α). Autoacetylation stimulated the catalytic activity of mARD1(225) to acetylate Lys532 of the oxygen-dependent degradation (ODD) domain of HIF-1α, leading to the proteosomal degradation of HIF-1α. In contrast, autoacetylation of mARD1(235) and hARD1(235) contributed to cellular growth under normoxic conditions by increasing the expression of cyclin D1. Taken together, these data suggest that autoacetylation of ARD1 variants differentially regulates angiogenesis and cell proliferation in an isoform-specific manner.


International Journal of Oncology | 2016

Ninjurin1 regulates lipopolysaccharide-induced inflammation through direct binding.

Min Wook Shin; Sung-Jin Bae; Hee-Jun Wee; Hyo-Jong Lee; Bum Ju Ahn; Hoang Le; Eun Lee; Ran Hee Kim; Hye Shin Lee; Ji Hae Seo; Ji-Hyeon Park; Kyu-Won Kim

Ninjurin1 is a transmembrane protein involved in macrophage migration and adhesion during inflammation. It was recently reported that repression of Ninjurin1 attenuated the lipopolysaccharide (LPS)-induced inflammatory response in macrophages; however, the precise mechanism by which Ninjurin1 modulates LPS-induced inflammation remains poorly understood. In the present study, we found that the interaction between Ninjurin1 and LPS contributed to the LPS-induced inflammatory response. Notably, pull-down assays using lysates from HEK293T cells transfected with human or mouse Ninjurin1 and biotinylated LPS (LPS-biotin) showed that LPS directly bound Ninjurin1. Subsequently, LPS binding assays with various truncated forms of Ninjurin1 protein revealed that amino acids (aa) 81-100 of Ninjurin1 were required for LPS binding. In addition, knockdown experiments using Ninj1 siRNA resulted in decreased nitric oxide (NO) and tumor necrosis factor-α (TNFα) secretion upon LPS treatment in Raw264.7 cells. Collectively, our results suggest that Ninjurin1 regulates the LPS-induced inflammatory response through its direct binding to LPS, thus, identifying Ninjurin1 as a putative target for the treatment of inflammatory diseases, such as sepsis and inflammation-associated carcinogenesis.

Collaboration


Dive into the Bum Ju Ahn's collaboration.

Top Co-Authors

Avatar

Kyu-Won Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Ji Hae Seo

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Hee-Jun Wee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Jong Ho Cha

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Min Wook Shin

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Eun Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hoang Le

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Hye Shin Lee

Seoul National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge