Network


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

Hotspot


Dive into the research topics where Hee Pom Lee is active.

Publication


Featured researches published by Hee Pom Lee.


Scientific Reports | 2016

Piperlongumine inhibits lung tumor growth via inhibition of nuclear factor kappa B signaling pathway

Jie Zheng; Dong Ju Son; Sun Mi Gu; Ju Rang Woo; Young Wan Ham; Hee Pom Lee; Wun-Jae Kim; Jae Kyung Jung; Jin Tae Hong

Piperlongumine has anti-cancer activity in numerous cancer cell lines via various signaling pathways. But there has been no study regarding the mechanisms of PL on the lung cancer yet. Thus, we evaluated the anti-cancer effects and possible mechanisms of PL on non-small cell lung cancer (NSCLC) cells in vivo and in vitro. Our findings showed that PL induced apoptotic cell death and suppressed the DNA binding activity of NF-κB in a concentration dependent manner (0–15 μM) in NSCLC cells. Docking model and pull down assay showed that PL directly binds to the DNA binding site of nuclear factor-κB (NF-κB) p50 subunit, and surface plasmon resonance (SPR) analysis showed that PL binds to p50 concentration-dependently. Moreover, co-treatment of PL with NF-κB inhibitor phenylarsine oxide (0.1 μM) or p50 siRNA (100 nM) augmented PL-induced inhibitory effect on cell growth and activation of Fas and DR4. Notably, co-treatment of PL with p50 mutant plasmid (C62S) partially abolished PL-induced cell growth inhibition and decreased the enhanced expression of Fas and DR4. In xenograft mice model, PL (2.5–5 mg/kg) suppressed tumor growth of NSCLC dose-dependently. Therefore, these results indicated that PL could inhibit lung cancer cell growth via inhibition of NF-κB signaling pathway in vitro and in vivo.


Neuropharmacology | 2017

Inhibitory effect of punicalagin on lipopolysaccharide-induced neuroinflammation, oxidative stress and memory impairment via inhibition of nuclear factor-kappaB.

Youngeun Kim; Chul Ju Hwang; Hee Pom Lee; Chun Sik Kim; Dong Ju Son; Young Wan Ham; Mats Hellström; Sang-Bae Han; Hee Sik Kim; Eun Kyung Park; Jin Tae Hong

&NA; Neuroinflammation is significant in the pathogenesis and development of Alzheimers disease (AD). Previously, we showed lipopolysaccharide (LPS)‐induced neuroinflammation caused memory impairment. We investigated the possible preventive effects of punicalagin (PUN), a component of pomegranate, on memory deficiency caused by LPS, along with the fundamental mechanisms. LPS‐treated cultured astrocytes and microglial BV‐2 cells were investigated for anti‐neuroinflammatory effects of PUN. PUN (1.5 mg/kg) ameliorates LPS (250 &mgr;g/kg daily 7 times)‐induced memory impairment as well as prevents the LPS‐induced expression of inflammatory proteins. In in vitro study, we also found that PUN (1 &mgr;g/ml) inhibited the LPS‐(10, 20 and 50 &mgr;M) induced expression of iNOS and Cox‐2 as well as the production of ROS, NO, TNF‐&agr; and IL‐1&bgr;. PUN also suppress activation of NF‐&kgr;B via inhibition of I&kgr;B degradation as well as p50 and p65 translocation into the nucleus in LPS treated mouse brain and cultured astrocytes and microglial BV‐2 cells. Consistent with the inhibitory effect on neuro inflammation, PUN inhibited LPS‐induced A&bgr;1‐42 generation through down‐regulation of APP and BACE1 expression in in vivo and in vitro study. Moreover, PUN directly binds to NF‐&kgr;B subunit p50 evidenced by a docking model and pull down assay. These results suggest that PUN inhibits LPS‐induced memory impairment via anti‐inflammatory and anti‐amylogenic mechanisms through inhibition of NF‐&kgr;B activation. HighlightsNeuroinflammation and amyloidogenesis are main symptoms of Alzheimers disease.NF‐&kgr;B activation can induce the inflammation and amyloidogenesis pathways.Punicalagin inhibits NF‐&kgr;B activation through direct binding to its subunit P50.Punicalagin reduces LPS‐induced neuroinflammation and amyloidogenesis.Punicalagin is a possible candidate for treating Alzheimers disease.


British Journal of Pharmacology | 2013

4-O-methylhonokiol, a PPARγ agonist, inhibits prostate tumour growth: p21-mediated suppression of NF-κB activity

Nam Jin Lee; Ju Hoon Oh; Jung Ok Ban; Jung Hyun Shim; Hee Pom Lee; Jae Kyung Jung; Byeongwoo Ahn; Do-Young Yoon; Sang-Bae Han; Young Wan Ham; Jin-Tae Hong

The effects of 4‐O‐methylhonokiol (MH), a constituent of Magnolia officinalis, were investigated on human prostate cancer cells and its mechanism of action elucidated.


British Journal of Pharmacology | 2014

Anti-arthritis effects of (E)-2,4-bis(p-hydroxyphenyl)-2-butenal are mediated by inhibition of the STAT3 pathway

Jung Ok Ban; Dae Hwan Kim; Hee Pom Lee; Chul Ju Hwang; Jung Hyun Shim; Dae Joong Kim; Tae Myoung Kim; Heon Sang Jeong; Seong Su Nah; Hanyong Chen; Zigang Dong; Young Wan Ham; Youngsoo Kim; Sang-Bae Han; Jin Tae Hong

Products of Maillard reactions between aminoacids and reducing sugars are known to have anti‐inflammatory properties. Here we have assessed the anti‐arthritis effects of (E)‐2,4‐bis(p‐hydroxyphenyl)‐2‐butenal and its possible mechanisms of action.


Apoptosis | 2014

(E)-2,4-Bis(p-hydroxyphenyl)-2-butenal inhibits tumor growth via suppression of NF-κB and induction of death receptor 6

Jung Ok Ban; Young Suk Jung; Dae Hwan Kim; Kyung Ran Park; Hyung Mun Yun; Nam Jin Lee; Hee Pom Lee; Jeong Hyun Shim; Heon Sang Jeong; Yun Hee Lee; Young Wan Ham; Sang-Bae Han; Jin Tae Hong

The Maillard reaction products are known to be effective in chemoprevention. Here, we focused on the anti-cancer effects of (E)-2,4-bis(p-hydroxyphenyl)-2-butenal on in vitro and in vivo colon cancer. We analysed the anti-cancer activity of (E)-2,4-bis(p-hydroxyphenyl)-2-butenal on colon cancer cells by using cell cycle and apoptosis analysis. To elucidate it’s mechanism, NF-κB DNA binding activity, docking model as well as pull-down assay. Further, a xenograft model of colon cancer was studied to test the in vivo effects of (E)-2,4-bis(p-hydroxyphenyl)-2-butenal. (E)-2,4-Bis(p-hydroxyphenyl)-2-butenal inhibited colon cancer cells (SW620 and HCT116) growth followed by induction of apoptosis in a concentration-dependent manner via down-regulation of NF-κB activity. In docking model as well as pull-down assay, (E)-2,4-bis(p-hydroxyphenyl)-2-butenal directly binds to three amino acid residues of IKKβ, thereby inhibited IKKβ activity in addition to induction of death receptor 6 (DR6) as well as their target apoptotic genes. Finally, (E)-2,4-bis(p-hydroxyphenyl)-2-butenal suppressed anchorage-independent cancer cell growth, and tumor growth in xenograft model accompanied with apoptosis through inhibition of IKKβ/NF-κB activity, and overexpression of DR6. These results suggest that (E)-2,4-bis(p-hydroxyphenyl)-2-butenal inhibits colon cancer cell growth through inhibition of IKKβ/NF-κB activity and induction of DR6 expression.


Oncotarget | 2016

Inhibitory effect of thiacremonone on MPTP-induced dopaminergic neurodegeneration through inhibition of p38 activation

Chul Ju Hwang; Hee Pom Lee; Dong-Young Choi; Heon Sang Jeong; Tae Hoon Kim; Tae Hyung Lee; Young Min Kim; Dae Bong Moon; Sung Sik Park; Sunyoung Kim; Ki-Wan Oh; Dae Yeon Hwang; Sang-Bae Han; Hwa-Jeong Lee; Jin Tae Hong

Neuroinflammation is implicated for dopaminergic neurodegeneration. Sulfur compounds extracted from garlic have been shown to have anti-inflammatory properties. Previously, we have investigated that thiacremonone, a sulfur compound isolated from garlic has anti-inflammatory effects on several inflammatory disease models. To investigate the protective effect of thiacremonone against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced behavioral impairment and dopaminergic neurodegeneration, 8 week old ICR mice were given thiacremonone (10 mg/kg) in drinking water for 1 month and received intraperitoneal injection of MPTP (15 mg/kg, four times with 2 h interval) during the last 7 days of treatment. Our data showed that thiacremonone decreased MPTP-induced behavioral impairments (Rotarod test, Pole test, and Gait test), dopamine depletion and microglia and astrocytes activations as well as neuroinflammation. Higher activation of p38 was found in the substantia nigra and striatum after MPTP injection, but p38 activation was reduced in thiacremonone treated group. In an in vitro study, thiacremonone (1, 2, and 5 μg/ml) effectively decreased MPP+ (0.5 mM)-induced glial activation, inflammatory mediators generation and dopaminergic neurodegeneration in cultured astrocytes and microglial BV-2 cells. Moreover, treatment of p38 MAPK inhibitor SB203580 (10 μM) further inhibited thiacremonone induced reduction of neurodegeneration and neuroinflammation. These results indicated that the anti-inflammatory compound, thiacremonone, inhibited neuroinflammation and dopaminergic neurodegeneration through inhibition of p38 activation.


Oncotarget | 2015

Decreased severity of collagen antibody and lipopolysaccharide-induced arthritis in human IL-32β overexpressed transgenic mice

Mi Hee Park; Do-Young Yoon; Jung Ok Ban; Dae Hwan Kim; Dong-Hun Lee; Sukgil Song; Youngsoo Kim; Sang-Bae Han; Hee Pom Lee; Jin Tae Hong

Interleukin (IL)-32, mainly produced by T-lymphocytes, natural killer cells, epithelial cells, and blood monocytes, is dominantly known as a pro-inflammatory cytokine. However, the role of IL-32 on inflammatory disease has been doubtful according to diverse conflicting results. This study was designed to examine the role of IL-32β on the development of collagen antibody (CAIA) and lipopolysaccharide (LPS)-induced inflammatory arthritis. Our data showed that the paw swelling volume and clinical score were significantly reduced in the CAIA and LPS-treated IL-32β transgenic mice compared with non-transgenic mice. The populations of cytotoxic T, NK and dendritic cells was inhibited and NF-κB and STAT3 activities were significantly lowered in the CAIA and LPS-treated IL-32β transgenic mice. The expression of pro-inflammatory proteins was prevented in the paw tissues of CAIA and LPS-treated IL-32β transgenic mice. In addition, IL-32β altered several cytokine levels in the blood, spleen and paw joint. Our data indicates that IL-32β comprehensively inhibits the inflammation responses in the CAIA and LPS-induced inflammatory arthritis model.


Biomolecules & Therapeutics | 2012

Growth Inhibitory Effect of (E)-2,4-bis(p-hydroxyphenyl)-2-Butenal Diacetate through Induction of Apoptotic Cell Death by Increasing DR3 Expression in Human Lung Cancer Cells.

Ung-Soo Lee; Jung Ok Ban; Eung Tae Yeon; Hee Pom Lee; Venkatareddy Udumula; Young Wan Ham; Jin Tae Hong

The Maillard Reaction Products (MRPs) are chemical compounds which have been known to be effective in chemoprevention. Death receptors (DR) play a central role in directing apoptosis in several cancer cells. In our previous study, we demonstrated that (E)-2,4-bis(p-hydroxyphenyl)-2-butenal, a MRP product, inhibited human colon cancer cell growth by inducing apoptosis via nuclear factor-κB (NF-κB) inactivation and G2/M phase cell cycle arrest. In this study, (E)-2,4-bis(p-hydroxyphenyl)-2-butenal diacetate, a new (E)-2,4-bis(p-hydroxyphenyl)-2-butenal derivative, was synthesized to improve their solubility and stability in water and then evaluated against NCI-H460 and A549 human lung cancer cells. (E)-2,4-bis(p-hydroxyphenyl)-2-butenal diacetate reduced the viability in both cell lines in a time and dose-dependent manner. We also found that (E)-2,4-bis(p-hydroxyphenyl)-2-butenal diacetate increased apoptotic cell death through the upregulation of the expression of death receptor (DR)-3 and DR6 in both lung cancer cell lines. In addition to this, the transfection of DR3 siRNA diminished the growth inhibitory and apoptosis inducing effect of (E)-2,4-bis(p-hydroxyphenyl)-2-butenal diacetate on lung cancer cells, however these effects of (E)-2,4-bis(p-hydroxyphenyl)-2-butenal diacetate was not changed by DR6 siRNA. These results indicated that (E)-2,4-bis(p-hydroxyphenyl)-2-butenal diacetate inhibits human lung cancer cell growth via increasing apoptotic cell death by upregulation of the expression of DR3.


Theranostics | 2017

MMPP Attenuates Non-Small Cell Lung Cancer Growth by Inhibiting the STAT3 DNA-Binding Activity via Direct Binding to the STAT3 DNA-Binding Domain

Dong Ju Son; Jie Zheng; Yu Yeon Jung; Chul Ju Hwang; Hee Pom Lee; Ju Rang Woo; Song Yi Baek; Young Wan Ham; Min Woong Kang; Minho Shong; Gi Ryang Kweon; Min Jong Song; Jae Kyung Jung; Sang-Bae Han; Bo Yeon Kim; Do Young Yoon; Bu Young Choi; Jin Tae Hong

Rationale: Signal transducer and activator of transcription-3 (STAT3) plays a pivotal role in cancer biology. Many small-molecule inhibitors that target STAT3 have been developed as potential anticancer drugs. While designing small-molecule inhibitors that target the SH2 domain of STAT3 remains the leading focus for drug discovery, there has been a growing interest in targeting the DNA-binding domain (DBD) of the protein. Methods: We demonstrated the potential antitumor activity of a novel, small-molecule (E)-2-methoxy-4-(3-(4-methoxyphenyl)prop-1-en-1-yl)phenol (MMPP) that directly binds to the DBD of STAT3, in patient-derived non-small cell lung cancer (NSCLC) xenograft model as well as in NCI-H460 cell xenograft model in nude mice. Results: MMPP effectively inhibited the phosphorylation of STAT3 and its DNA binding activity in vitro and in vivo. It induced G1-phase cell cycle arrest and apoptosis through the regulation of cell cycle- and apoptosis-regulating genes by directly binding to the hydroxyl residue of threonine 456 in the DBD of STAT3. Furthermore, MMPP showed a similar or better antitumor activity than that of docetaxel or cisplatin. Conclusion: MMPP is suggested to be a potential candidate for further development as an anticancer drug that targets the DBD of STAT3.


Scientific Reports | 2016

Novel synthetic (E)-2-methoxy-4-(3-(4-methoxyphenyl) prop-1-en-1-yl) phenol inhibits arthritis by targeting signal transducer and activator of transcription 3

Dong Ju Son; Dae Hwan Kim; Seong-Su Nah; Mi Hee Park; Hee Pom Lee; Sang-Bae Han; Udumula Venkatareddy; Benjamin Gann; Kevin Rodriguez; Scott R. Burt; Young Wan Ham; Yu Yeon Jung; Jin Tae Hong

Rheumatoid arthritis (RA) is a severely debilitating chronic autoimmune disease that leads to long-term joint damage. Signal transducer and activator of transcription 3 (STAT3)-targeted small molecules have shown promise as therapeutic drugs for treating RA. We previously identified (E)-2,4-bis(p-hydroxyphenyl)-2-butenal (BHPB), a tyrosine-fructose Maillard reaction product, as a small molecule with potent anti-inflammatory and anti-arthritic properties, mediated through the inhibition of STAT3 activation. The aim of this study was to develop a novel BHPH derivative with improved anti-arthritic properties and drug-likeness. We designed and synthesised (E)-2-methoxy-4-(3-(4-methoxyphenyl) prop-1-en-1-yl) phenol (MMPP), a novel synthetic BHPB analogue, and investigated its anti-inflammatory and anti-arthritic activities in experimentally-induced RA. We showed that MMPP strongly inhibited pro-inflammatory responses by inhibiting in vitro STAT3 activation and its downstream signalling in murine macrophages and human synoviocytes from patients with RA. Furthermore, we demonstrated that MMPP exhibited potent anti-arthritic activity in a collagen antibody-induced arthritis (CAIA) mouse model in vivo. Collectively, our results suggest that MMPP has great potential for use in the treatment of RA.

Collaboration


Dive into the Hee Pom Lee's collaboration.

Top Co-Authors

Avatar

Jin Tae Hong

Chungbuk National University

View shared research outputs
Top Co-Authors

Avatar

Sang-Bae Han

Chungbuk National University

View shared research outputs
Top Co-Authors

Avatar

Young Wan Ham

Chungbuk National University

View shared research outputs
Top Co-Authors

Avatar

Chul Ju Hwang

Chungbuk National University

View shared research outputs
Top Co-Authors

Avatar

Dong Ju Son

Chungbuk National University

View shared research outputs
Top Co-Authors

Avatar

Jung Ok Ban

Chungbuk National University

View shared research outputs
Top Co-Authors

Avatar

Dae Hwan Kim

Chungbuk National University

View shared research outputs
Top Co-Authors

Avatar

Jae Kyung Jung

Chungbuk National University

View shared research outputs
Top Co-Authors

Avatar

Jie Zheng

Chungbuk National University

View shared research outputs
Top Co-Authors

Avatar

Mi Hee Park

Chungbuk National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge