171 results on '"Nam Doo Kim"'
Search Results
2. p21-activated kinase 4 suppresses fatty acid β-oxidation and ketogenesis by phosphorylating NCoR1
- Author
-
Min Yan Shi, Hwang Chan Yu, Chang Yeob Han, In Hyuk Bang, Ho Sung Park, Kyu Yun Jang, Sangkyu Lee, Jeong Bum Son, Nam Doo Kim, Byung-Hyun Park, and Eun Ju Bae
- Subjects
Science - Abstract
Abstract PPARα corepressor NCoR1 is a key regulator of fatty acid β-oxidation and ketogenesis. However, its regulatory mechanism is largely unknown. Here, we report that oncoprotein p21-activated kinase 4 (PAK4) is an NCoR1 kinase. Specifically, PAK4 phosphorylates NCoR1 at T1619/T2124, resulting in an increase in its nuclear localization and interaction with PPARα, thereby repressing the transcriptional activity of PPARα. We observe impaired ketogenesis and increases in PAK4 protein and NCoR1 phosphorylation levels in liver tissues of high fat diet-fed mice, NAFLD patients, and hepatocellular carcinoma patients. Forced overexpression of PAK4 in mice represses ketogenesis and thereby increases hepatic fat accumulation, whereas genetic ablation or pharmacological inhibition of PAK4 exhibites an opposite phenotype. Interestingly, PAK4 protein levels are significantly suppressed by fasting, largely through either cAMP/PKA- or Sirt1-mediated ubiquitination and proteasome degradation. In this way, our findings provide evidence for a PAK4-NCoR1/PPARα signaling pathway that regulates fatty acid β-oxidation and ketogenesis.
- Published
- 2023
- Full Text
- View/download PDF
3. Author Correction: p21-activated kinase 4 suppresses fatty acid β-oxidation and ketogenesis by phosphorylating NCoR1
- Author
-
Min Yan Shi, Hwang Chan Yu, Chang Yeob Han, In Hyuk Bang, Ho Sung Park, Kyu Yun Jang, Sangkyu Lee, Jeong Bum Son, Nam Doo Kim, Byung-Hyun Park, and Eun Ju Bae
- Subjects
Science - Published
- 2023
- Full Text
- View/download PDF
4. Myricetin Protects Against High Glucose-Induced β-Cell Apoptosis by Attenuating Endoplasmic Reticulum Stress via Inactivation of Cyclin-Dependent Kinase 5
- Author
-
Udayakumar Karunakaran, Suma Elumalai, Jun Sung Moon, Jae-Han Jeon, Nam Doo Kim, Keun-Gyu Park, Kyu Chang Won, Jaechan Leem, and In-Kyu Lee
- Subjects
apoptosis ,cyclin-dependent kinase 5 ,endoplasmic reticulum stress ,insulin-secreting cells ,myricetin ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
BackgroundChronic hyperglycemia has deleterious effects on pancreatic β-cell function and turnover. Recent studies support the view that cyclin-dependent kinase 5 (CDK5) plays a role in β-cell failure under hyperglycemic conditions. However, little is known about how CDK5 impair β-cell function. Myricetin, a natural flavonoid, has therapeutic potential for the treatment of type 2 diabetes mellitus. In this study, we examined the effect of myricetin on high glucose (HG)-induced β-cell apoptosis and explored the relationship between myricetin and CDK5.MethodsTo address this question, we subjected INS-1 cells and isolated rat islets to HG conditions (30 mM) in the presence or absence of myricetin. Docking studies were conducted to validate the interaction between myricetin and CDK5. Gene expression and protein levels of endoplasmic reticulum (ER) stress markers were measured by real-time reverse transcription polymerase chain reaction and Western blot analysis.ResultsActivation of CDK5 in response to HG coupled with the induction of ER stress via the down regulation of sarcoendoplasmic reticulum calcium ATPase 2b (SERCA2b) gene expression and reduced the nuclear accumulation of pancreatic duodenal homeobox 1 (PDX1) leads to β-cell apoptosis. Docking study predicts that myricetin inhibit CDK5 activation by direct binding in the ATP-binding pocket. Myricetin counteracted the decrease in the levels of PDX1 and SERCA2b by HG. Moreover, myricetin attenuated HG-induced apoptosis in INS-1 cells and rat islets and reduce the mitochondrial dysfunction by decreasing reactive oxygen species production and mitochondrial membrane potential (Δψm) loss.ConclusionMyricetin protects the β-cells against HG-induced apoptosis by inhibiting ER stress, possibly through inactivation of CDK5 and consequent upregulation of PDX1 and SERCA2b.
- Published
- 2019
- Full Text
- View/download PDF
5. Nomilin from Yuzu Seed Has In Vitro Antioxidant Activity and Downregulates Melanogenesis in B16F10 Melanoma Cells through the PKA/CREB Signaling Pathway
- Author
-
Moon-Hee Choi, Seung-Hwa Yang, Nam Doo Kim, and Hyun-Jae Shin
- Subjects
yuzu tree ,seed husk ,nomilin ,antioxidant ,antimelanogenic ,tyrosinase inhibitor ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Yuzu (Citrus junos) is a citrus plant native to Asian countries, including Korea, Japan, and China. Yuzu peel and seed contain abundant vitamin C, citric acid, and polyphenols. Although the antioxidative and antimelanogenic activities of other citrus fruits and yuzu extract have been reported, the tyrosinase inhibitory activity of the limonoid aglycone contained in yuzu seed extract is unknown. We separated yuzu seeds into the husk, shell, and meal and evaluated antioxidant activity of each. The limonoid glucoside fraction of the husk identified nomilin, a novel tyrosinase inhibitor. We performed tyrosinase inhibitory activity and noncompetitive inhibition assays and docking studies to determine nomilin binding sites. Furthermore, we evaluated the antioxidative mechanism and antimelanogenic activity of nomilin in B16F10 melanoma cells. The concentration of nomilin that did not show toxicity was
- Published
- 2022
- Full Text
- View/download PDF
6. Acacetin Inhibits the Growth of STAT3-Activated DU145 Prostate Cancer Cells by Directly Binding to Signal Transducer and Activator of Transcription 3 (STAT3)
- Author
-
Sun Yun, Yu-Jin Lee, Jiyeon Choi, Nam Doo Kim, Dong Cho Han, and Byoung-Mog Kwon
- Subjects
antitumor ,acacetin ,STAT3 inhibitor ,natural product ,label-free methods ,Organic chemistry ,QD241-441 - Abstract
Signal transducer and activator of transcription 3 (STAT3) plays a critical role in the formation and growth of human cancer. Therefore, STAT3 is a therapeutic target for cancer drug discovery. Acacetin, a flavone present in various plants, inhibits constitutive and inducible STAT3 activation in STAT3-activated DU145 prostate cancer cells. Acacetin inhibits STAT3 activity by directly binding to STAT3, which we confirmed by a pull-down assay with a biotinylated compound and two level-free methods, namely, a drug affinity responsive target stability (DARTS) experiment and a cellular thermal shift assay (CETSA). Acacetin inhibits STAT3 phosphorylation at the tyrosine 705 residue and nuclear translocation in DU145 cells, which leads to the downregulation of STAT3 target genes. Acacetin then induces apoptosis in a time-dependent manner. Interestingly, acacetin induces the production of reactive oxygen species (ROS) that are not involved in the acacetin-induced inhibition of STAT3 activation because the suppressed p-STAT3 level is not rescued by treatment with GSH or NAC, which are general ROS inhibitors. We also found that acacetin inhibits tumor growth in xenografted nude mice. These results suggest that acacetin, as a STAT3 inhibitor, could be a possible drug candidate for targeting STAT3 for the treatment of cancer in humans.
- Published
- 2021
- Full Text
- View/download PDF
7. Discovery of a novel potent peptide agonist to adiponectin receptor 1.
- Author
-
Sunghwan Kim, Younho Lee, Jun Woo Kim, Young-Jin Son, Min Jung Ma, Jee-Hyun Um, Nam Doo Kim, Sang Hyun Min, Dong Il Kim, and Brian B Kim
- Subjects
Medicine ,Science - Abstract
Activation of adiponectin receptors (AdipoRs) by its natural ligand, adiponectin has been known to be involved in modulating critical metabolic processes such as glucose metabolism and fatty acid oxidation as demonstrated by a number of in vitro and in vivo studies over last two decades. These findings suggest that AdipoRs' agonists could be developed into a potential therapeutic agent for metabolic diseases, such as diabetes mellitus, especially for type II diabetes, a long-term metabolic disorder characterized by high blood sugar, insulin resistance, and relative lack of insulin. Because of limitations in production of biologically active adiponectin, adiponectin-mimetic AdipoRs' agonists have been suggested as alternative ways to expand the opportunity to develop anti-diabetic agents. Based on crystal structure of AdipoR1, we designed AdipoR1's peptide agonists using protein-peptide docking simulation and screened their receptor binding abilities and biological functions via surface plasmon resonance (SPR) and biological analysis. Three candidate peptides, BHD1028, BHD43, and BHD44 were selected and confirmed to activate AdipoR1-mediated signal pathways. In order to enhance the stability and solubility of peptide agonists, candidate peptides were PEGylated. PEGylated BHD1028 exhibited its biological activity at nano-molar concentration and could be a potential therapeutic agent for the treatment of diabetes. Also, SPR and virtual screening techniques utilized in this study may potentially be applied to other peptide-drug screening processes against membrane receptor proteins.
- Published
- 2018
- Full Text
- View/download PDF
8. OCLI-023, a Novel Pyrimidine Compound, Suppresses Osteoclastogenesis In Vitro and Alveolar Bone Resorption In Vivo.
- Author
-
Hye Jung Ihn, Taeho Lee, Ju Ang Kim, Doohyun Lee, Nam Doo Kim, Hong-In Shin, Yong Chul Bae, and Eui Kyun Park
- Subjects
Medicine ,Science - Abstract
An abnormal increase in osteoclast differentiation and activation results in various bone-resorptive diseases, including periodontitis, rheumatoid arthritis, and osteoporosis. Chemical compounds containing pyrimidine ring have been shown to regulate a variety of biological processes. Therefore, in order to identify an antiresorptive agent, we synthesized a series of pyrimidine ring-containing chemical compounds, and found that OCLI-023 suppressed the differentiation and activation of osteoclasts in vitro. OCLI-023 directly inhibited receptor activator of nuclear factor-κB ligand (RANKL)-induced differentiation of bone marrow macrophages into osteoclasts, without a cytotoxic response. OCLI-023 also downregulated the RANKL-induced mRNA expression of osteoclast markers as well as inhibited the formation of actin rings and resorption pits. OCLI-023 attenuated the RANKL-induced activation of c-Jun N-terminal kinase and nuclear factor kappa-light-chain-enhancer of activated B cell signaling pathways. In a mouse model of periodontitis, ligature induced an increase of distance between cementoenamel junction (CEJ) and alveolar bone crest (ABC) in the second molar, and OCLI-023 significantly reduced it. Histological analysis showed ligature-induced increase of osteoclast numbers was also significantly reduced by OCLI-023. These data demonstrated the inhibitory effect of OCLI-023 on osteoclast differentiation and activity of osteoclasts in vitro, as well as on ligature-induced bone loss in vivo, and OCLI-023 can be proposed as a novel anti-resorptive compound.
- Published
- 2017
- Full Text
- View/download PDF
9. Inhibitory Effects of 2N1HIA (2-(3-(2-Fluoro-4-Methoxyphenyl)-6-Oxo-1(6H)-Pyridazinyl)-N-1H-Indol-5-Ylacetamide) on Osteoclast Differentiation via Suppressing Cathepsin K Expression
- Author
-
Sun-Hee Ahn, Zhihao Chen, Jinkyung Lee, Seok-Woo Lee, Sang Hyun Min, Nam Doo Kim, and Tae-Hoon Lee
- Subjects
osteoclast ,cathepsin K ,osteoporosis ,bone resorption ,Organic chemistry ,QD241-441 - Abstract
Osteoclasts are large multinucleated cells which are induced by the regulation of the receptor activator of nuclear factor kappa-Β ligand (RANKL), which is important in bone resorption. Excessive osteoclast differentiation can cause pathologic bone loss and destruction. Numerous studies have targeted molecules inhibiting RANKL signaling or bone resorption activity. In this study, 11 compounds from commercial libraries were examined for their effect on RANKL-induced osteoclast differentiation. Of these compounds, only 2-(3-(2-fluoro-4-methoxyphenyl)-6-oxo-1(6H)-pyridazinyl)-N-1H-indol-5-ylacetamide (2N1HIA) caused a significant decrease in multinucleated tartrate-resistant acid phosphatase (TRAP)-positive cell formation in a dose-dependent manner, without inducing cytotoxicity. The 2N1HIA compound neither affected the expression of osteoclast-specific gene markers such as TRAF6, NFATc1, RANK, OC-STAMP, and DC-STAMP, nor the RANKL signaling pathways, including p38, ERK, JNK, and NF-κB. However, 2N1HIA exhibited a significant impact on the expression levels of CD47 and cathepsin K, the early fusion marker and critical protease for bone resorption, respectively. The activity of matrix metalloprotease-9 (MMP-9) decreased due to 2N1HIA treatment. Accordingly, bone resorption activity and actin ring formation decreased in the presence of 2N1HIA. Taken together, 2N1HIA acts as an inhibitor of osteoclast differentiation by attenuating bone resorption activity and may serve as a potential candidate in preventing and/or treating osteoporosis, or other bone diseases associated with excessive bone resorption.
- Published
- 2018
- Full Text
- View/download PDF
10. Discovery and Development of Anti-HBV Agents and Their Resistance
- Author
-
Baik-Lin Seong, Nam Doo Kim, and Kyun-Hwan Kim
- Subjects
hepatitis B virus ,antiviral agent ,drug development ,drug resistance ,modeling ,rational drug design ,Organic chemistry ,QD241-441 - Abstract
Hepatitis B virus (HBV) infection is a prime cause of liver diseases such as hepatitis, cirrhosis and hepatocellular carcinoma. The current drugs clinically available are nucleot(s)ide analogues that inhibit viral reverse transcriptase activity. Most drugs of this class are reported to have viral resistance with breakthrough. Recent advances in methods for in silico virtual screening of chemical libraries, together with a better understanding of the resistance mechanisms of existing drugs have expedited the discovery and development of novel anti-viral drugs. This review summarizes the current status of knowledge about and viral resistance of HBV drugs, approaches for the development of novel drugs as well as new viral and host targets for future drugs.
- Published
- 2010
- Full Text
- View/download PDF
11. Anti-cancer effect of thiacremonone through down regulation of peroxiredoxin 6.
- Author
-
Miran Jo, Hyung-Mun Yun, Kyung-Ran Park, Mi Hee Park, Dong Hun Lee, Seung Hee Cho, Hwan-Soo Yoo, Yong-Moon Lee, Heon Sang Jeong, Youngsoo Kim, Jae Kyung Jung, Bang Yeon Hwang, Mi Kyeong Lee, Nam Doo Kim, Sang Bae Han, and Jin Tae Hong
- Subjects
Medicine ,Science - Abstract
Thiacremonone (2, 4-dihydroxy-2, 5-dimethyl-thiophene-3-one) is an antioxidant substance as a novel sulfur compound generated from High-Temperature-High-Pressure-treated garlic. Peroxiredoxin 6 (PRDX6) is a member of peroxidases, and has glutathione peroxidase and calcium-independent phospholipase A2 (iPLA2) activities. Several studies have demonstrated that PRDX6 stimulates lung cancer cell growth via an increase of glutathione peroxidase activity. A docking model study and pull down assay showed that thiacremonone completely fits on the active site (cys-47) of glutathione peroxidase of PRDX6 and interacts with PRDX6. Thus, we investigated whether thiacremonone inhibits cell growth by blocking glutathione peroxidase of PRDX6 in the human lung cancer cells, A549 and NCI-H460. Thiacremonone (0-50 μg/ml) inhibited lung cancer cell growth in a concentration dependent manner through induction of apoptotic cell death accompanied by induction of cleaved caspase-3, -8, -9, Bax, p21 and p53, but decrease of xIAP, cIAP and Bcl2 expression. Thiacremonone further inhibited glutathione peroxidase activity in lung cancer cells. However, the cell growth inhibitory effect of thiacremonone was not observed in the lung cancer cells transfected with mutant PRDX6 (C47S) and in the presence of dithiothreitol and glutathione. In an allograft in vivo model, thiacremonone (30 mg/kg) also inhibited tumor growth accompanied with the reduction of PRDX6 expression and glutathione peroxidase activity, but increased expression of cleaved caspase-3, -8, -9, Bax, p21 and p53. These data indicate that thiacremonone inhibits tumor growth via inhibition of glutathione peroxidase activity of PRDX6 through interaction. These data suggest that thiacremonone may have potentially beneficial effects in lung cancer.
- Published
- 2014
- Full Text
- View/download PDF
12. Identification of Selective ERRγ Inverse Agonists
- Author
-
Jina Kim, Chun Young Im, Eun Kyung Yoo, Min Jung Ma, Sang-Bum Kim, Eunmi Hong, Jungwook Chin, Hayoung Hwang, Sungwoo Lee, Nam Doo Kim, Jae-Han Jeon, In-Kyu Lee, Yong Hyun Jeon, Hueng-Sik Choi, Seong Heon Kim, and Sung Jin Cho
- Subjects
estrogen-related receptor gamma ,inverse agonist ,ADMET ,GSK5182 ,Organic chemistry ,QD241-441 - Abstract
GSK5182 (4) is currently one of the lead compounds for the development of estrogen-related receptor gamma (ERRγ) inverse agonists. Here, we report the design, synthesis, pharmacological and in vitro absorption, distribution, metabolism, excretion, toxicity (ADMET) properties of a series of compounds related to 4. Starting from 4, a series of analogs were structurally modified and their ERRγ inverse agonist activity was measured. A key pharmacophore feature of this novel class of ligands is the introduction of a heterocyclic group for A-ring substitution in the core scaffold. Among the tested compounds, several of them are potent ERRγ inverse agonists as determined by binding and functional assays. The most promising compound, 15g, had excellent binding selectivity over related subtypes (IC50 = 0.44, >10, >10, and 10 μM at the ERRγ, ERRα, ERRβ, and ERα subtypes, respectively). Compound 15g also resulted in 95% transcriptional repression at a concentration of 10 μM, while still maintaining an acceptable in vitro ADMET profile. This novel class of ERRγ inverse agonists shows promise in the development of drugs targeting ERRγ-related diseases.
- Published
- 2016
- Full Text
- View/download PDF
13. Inhibition of phospholipase D1 induces immunogenic cell death and potentiates cancer immunotherapy in colorectal cancer
- Author
-
Won Chan, Hwang, Doona, Song, Hyesung, Lee, Changmok, Oh, Seong Hun, Lim, Hyeon Jeong, Bae, Nam Doo, Kim, Gyoonhee, Han, and Do Sik, Min
- Subjects
Clinical Biochemistry ,CD47 Antigen ,Immunogenic Cell Death ,Ligands ,Biochemistry ,Adenosine Triphosphate ,Phospholipase D ,Tumor Microenvironment ,Humans ,Molecular Medicine ,Immunotherapy ,Calreticulin ,Colorectal Neoplasms ,Wnt Signaling Pathway ,Molecular Biology - Abstract
Phospholipase D (PLD) is a potential therapeutic target against cancer. However, the contribution of PLD inhibition to the antitumor response remains unknown. We developed a potent and selective PLD1 inhibitor based on computer-aided drug design. The inhibitor enhanced apoptosis in colorectal cancer (CRC) cells but not in normal colonic cells, and in vitro cardiotoxicity was not observed. The inhibitor downregulated the Wnt/β-catenin signaling pathway and reduced the migration, invasion, and self-renewal capacity of CRC cells. In cancer, therapeutic engagement of immunogenic cell death (ICD) leads to more effective responses by eliciting the antitumor immunity of T cells. The CRC cells treated with the inhibitor showed hallmarks of ICD, including downregulation of “do not eat-me” signals (CD24, CD47, programmed cell death ligand 1 [PD-L1]), upregulation of “eat-me” signal (calreticulin), release of high-mobility group Box 1, and ATP. PLD1 inhibition subsequently enhanced the phagocytosis of cancer cells by macrophages through the surface expression of costimulatory molecules; as a result, the cancer cells were more susceptible to cytotoxic T-cell-mediated killing. Moreover, PLD1 inhibition attenuated colitis-associated CRC and orthotopically injected tumors, probably by controlling multiple pathways, including Wnt signaling, phagocytosis checkpoints, and immune signaling. Furthermore, combination therapy with a PLD1 inhibitor and an anti-PD-L1 antibody further enhanced tumor regression via immune activation in the tumor environment. Collectively, in this study, PLD1 was identified as a critical regulator of the tumor microenvironment in colorectal cancer, suggesting the potential of PLD1 inhibitors for cancer immunotherapy based on ICD and immune activation. PLD1 inhibitors may act as promising immune modulators in antitumor treatment via ICD.
- Published
- 2022
- Full Text
- View/download PDF
14. Abstract P2-13-15: VRN101396, a brain-permeable HER2 inhibitor, shows the anti-tumor activity in preclinical HER2-positive cancer models
- Author
-
Hong-ryul Jung, Sunghwan Kim, Jihye Yoo, Chan mi Park, Somi Lee, Hyoju Lee, Youngyi Lee, Jinhee Park, Kyungah Seo, Dong-Hyuk Seo, Eunhwa Ko, Jung Beom Son, Deakwon Kim, Hwan Geun Choi, and Nam Doo Kim
- Subjects
Cancer Research ,Oncology ,skin and connective tissue diseases - Abstract
Breast cancer (BC) is the second most common cancer associated with brain metastasis (BM) and among patients with breast cancer brain metastasis (BCBM), human epidermal growth factor receptor 2 (HER2)-positive breast cancer is most likely to develop BM. The overall prognosis of HER2-positive BCBM remains dismal with median survival time after BM of less than a year. Drugs like T-DM1, T-DXd and Tucatinib are reported to deliver a progression-free survival benefit in HER2-positive BCBM patients, but there are still unmet needs for a drug that crosses the blood brain barrier more effectively. We develop VRN101396, a brain penetrant, orally available, and irreversible small molecule inhibitor targeting HER2 to offer a new therapeutic option for the treatment of HER2-positive BCBM. VRN101396 potently inhibits catalytic activity of HER2 but spares wild-type EGFR. In HER2-positive breast cancer cell lines BT474 and SK-BR3 and gastric cancer cell line NCI-N87, VRN101396 inhibited the cell proliferation and phosphorylation of HER2 with single- or double-digit nanomolar IC50 values while inhibiting wild type EGFR with an IC50 value of >100 nM. In both subcutaneous and intracranial efficacy mouse models, once-daily oral dose of VRN101396 significantly inhibited tumor growth and achieved tumor regression. In N87 subcutaneous xenograft model, VRN101396 alone showed superior efficacy to the combination of Tucatinib and Trastuzumab. Additionally, in intracranial xenograft model, VRN101396 showed superior efficacy to Tucatinib. Pharmacokinetics of VRN101396 displayed higher brain exposure than tucatinib and lapatinib, indicating better target engagement in the brain. Pharmacodynamic analysis revealed that VRN101396 reduced phosphorylation of HER2, HER3, AKT, and ERK compared to vehicle group after administration up to 24 hours. In conclusion, VRN101396 is a therapeutic candidate for the treatment of HER2-positive BC, BCBM and gastric cancer. Citation Format: Hong-ryul Jung, Sunghwan Kim, Jihye Yoo, Chan mi Park, Somi Lee, Hyoju Lee, Youngyi Lee, Jinhee Park, Kyungah Seo, Dong-Hyuk Seo, Eunhwa Ko, Jung Beom Son, Deakwon Kim, Hwan Geun Choi, Nam Doo Kim. VRN101396, a brain-permeable HER2 inhibitor, shows the anti-tumor activity in preclinical HER2-positive cancer models [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P2-13-15.
- Published
- 2022
- Full Text
- View/download PDF
15. Combination of Type I and Type II MET Tyrosine Kinase Inhibitors as Therapeutic Approach to Prevent Resistance
- Author
-
Magda Bahcall, Luke J. Taus, Taebo Sim, Prafulla C. Gokhale, Jens Köhler, Stephen Wang, Nam Doo Kim, Cloud P. Paweletz, Zihan Wei, Yanan Kuang, Christie J. Lau, Pasi A. Jänne, Pratik R Chopade, Kshiti Dholakia, Sujuan Guo, Fangxin Hong, Jiannan Guo, and Paul Kirschmeier
- Subjects
Cancer Research ,business.industry ,Cell ,Mutant ,High-Throughput Nucleotide Sequencing ,Merestinib ,Mutagenesis (molecular biology technique) ,Drug resistance ,Article ,In vitro ,respiratory tract diseases ,Molecular Docking Simulation ,Mice ,medicine.anatomical_structure ,Oncology ,Drug Resistance, Neoplasm ,In vivo ,medicine ,Cancer research ,Animals ,Humans ,Female ,business ,Protein Kinase Inhibitors ,Tyrosine kinase - Abstract
MET-targeted therapies are clinically effective in MET-amplified and MET exon 14 deletion mutant (METex14) non–small cell lung cancers (NSCLCs), but their efficacy is limited by the development of drug resistance. Structurally distinct MET tyrosine kinase inhibitors (TKIs) (type I/II) have been developed or are under clinical evaluation, which may overcome MET-mediated drug resistance mechanisms. In this study, we assess secondary MET mutations likely to emerge in response to treatment with single-agent or combinations of type I/type II MET TKIs using TPR-MET transformed Ba/F3 cell mutagenesis assays. We found that these inhibitors gave rise to distinct secondary MET mutant profiles. However, a combination of type I/II TKI inhibitors (capmatinib and merestinib) yielded no resistant clones in vitro. The combination of capmatinib/merestinib was evaluated in vivo and led to a significant reduction in tumor outgrowth compared with either MET inhibitor alone. Our findings demonstrate in vitro and in vivo that a simultaneous treatment with a type I and type II MET TKI may be a clinically viable approach to delay and/or diminish the emergence of on target MET-mediated drug-resistance mutations.
- Published
- 2022
- Full Text
- View/download PDF
16. Supplementary Materials and Methods from Combination of Type I and Type II MET Tyrosine Kinase Inhibitors as Therapeutic Approach to Prevent Resistance
- Author
-
Pasi A. Jänne, Stephen Wang, Sujuan Guo, Jiannan Guo, Paul T. Kirschmeier, Jens Köhler, Zihan Wei, Fangxin Hong, Pratik R. Chopade, Prafulla C. Gokhale, Christie J. Lau, Kshiti H. Dholakia, Nam Doo Kim, Taebo Sim, Luke J. Taus, Yanan Kuang, Cloud P. Paweletz, and Magda Bahcall
- Abstract
Application of Bliss independence to test for synergy effect of TKI combination treatment in vivo
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Table S2 from Acquired METD1228V Mutation and Resistance to MET Inhibition in Lung Cancer
- Author
-
Geoffrey R. Oxnard, Pasi A. Jänne, Lynette M. Sholl, Michael T. Jaklitsch, Mark M. Awad, Christine A. Lydon, Nam Doo Kim, Adrian G. Sacher, Yanan Kuang, Ryan S. Alden, Jyoti D. Patel, Cloud P. Paweletz, Taebo Sim, and Magda Bahcall
- Abstract
Supplementary Table S2 shows type I and II MET kinase inhibitors and key trials studying their activity lung cancer populations enriched for MET-dependence.
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Figure S2 from Acquired METD1228V Mutation and Resistance to MET Inhibition in Lung Cancer
- Author
-
Geoffrey R. Oxnard, Pasi A. Jänne, Lynette M. Sholl, Michael T. Jaklitsch, Mark M. Awad, Christine A. Lydon, Nam Doo Kim, Adrian G. Sacher, Yanan Kuang, Ryan S. Alden, Jyoti D. Patel, Cloud P. Paweletz, Taebo Sim, and Magda Bahcall
- Abstract
Supplementary Figure S2 shows expression of D1228V mutation in PC9 cells as confirmed by RT-PCR and cDNA sequencing.
- Published
- 2023
- Full Text
- View/download PDF
19. Data from Combination of Type I and Type II MET Tyrosine Kinase Inhibitors as Therapeutic Approach to Prevent Resistance
- Author
-
Pasi A. Jänne, Stephen Wang, Sujuan Guo, Jiannan Guo, Paul T. Kirschmeier, Jens Köhler, Zihan Wei, Fangxin Hong, Pratik R. Chopade, Prafulla C. Gokhale, Christie J. Lau, Kshiti H. Dholakia, Nam Doo Kim, Taebo Sim, Luke J. Taus, Yanan Kuang, Cloud P. Paweletz, and Magda Bahcall
- Abstract
MET-targeted therapies are clinically effective in MET-amplified and MET exon 14 deletion mutant (METex14) non–small cell lung cancers (NSCLCs), but their efficacy is limited by the development of drug resistance. Structurally distinct MET tyrosine kinase inhibitors (TKIs) (type I/II) have been developed or are under clinical evaluation, which may overcome MET-mediated drug resistance mechanisms. In this study, we assess secondary MET mutations likely to emerge in response to treatment with single-agent or combinations of type I/type II MET TKIs using TPR-MET transformed Ba/F3 cell mutagenesis assays. We found that these inhibitors gave rise to distinct secondary MET mutant profiles. However, a combination of type I/II TKI inhibitors (capmatinib and merestinib) yielded no resistant clones in vitro. The combination of capmatinib/merestinib was evaluated in vivo and led to a significant reduction in tumor outgrowth compared with either MET inhibitor alone. Our findings demonstrate in vitro and in vivo that a simultaneous treatment with a type I and type II MET TKI may be a clinically viable approach to delay and/or diminish the emergence of on target MET-mediated drug-resistance mutations.
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Data from Combination of Type I and Type II MET Tyrosine Kinase Inhibitors as Therapeutic Approach to Prevent Resistance
- Author
-
Pasi A. Jänne, Stephen Wang, Sujuan Guo, Jiannan Guo, Paul T. Kirschmeier, Jens Köhler, Zihan Wei, Fangxin Hong, Pratik R. Chopade, Prafulla C. Gokhale, Christie J. Lau, Kshiti H. Dholakia, Nam Doo Kim, Taebo Sim, Luke J. Taus, Yanan Kuang, Cloud P. Paweletz, and Magda Bahcall
- Abstract
Supplementary Figures S1-3; Supplementary Tables S1-3
- Published
- 2023
- Full Text
- View/download PDF
21. Supplementary Methods from Acquired METD1228V Mutation and Resistance to MET Inhibition in Lung Cancer
- Author
-
Geoffrey R. Oxnard, Pasi A. Jänne, Lynette M. Sholl, Michael T. Jaklitsch, Mark M. Awad, Christine A. Lydon, Nam Doo Kim, Adrian G. Sacher, Yanan Kuang, Ryan S. Alden, Jyoti D. Patel, Cloud P. Paweletz, Taebo Sim, and Magda Bahcall
- Abstract
Supplementary methods, including croplet digital PCR for MET D1228V; molecular docking study; Molecular Dynamics Simulations.
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Figure Legends from Acquired METD1228V Mutation and Resistance to MET Inhibition in Lung Cancer
- Author
-
Geoffrey R. Oxnard, Pasi A. Jänne, Lynette M. Sholl, Michael T. Jaklitsch, Mark M. Awad, Christine A. Lydon, Nam Doo Kim, Adrian G. Sacher, Yanan Kuang, Ryan S. Alden, Jyoti D. Patel, Cloud P. Paweletz, Taebo Sim, and Magda Bahcall
- Abstract
Supplementary Figure Legends.
- Published
- 2023
- Full Text
- View/download PDF
23. Data from Acquired METD1228V Mutation and Resistance to MET Inhibition in Lung Cancer
- Author
-
Geoffrey R. Oxnard, Pasi A. Jänne, Lynette M. Sholl, Michael T. Jaklitsch, Mark M. Awad, Christine A. Lydon, Nam Doo Kim, Adrian G. Sacher, Yanan Kuang, Ryan S. Alden, Jyoti D. Patel, Cloud P. Paweletz, Taebo Sim, and Magda Bahcall
- Abstract
Amplified and/or mutated MET can act as both a primary oncogenic driver and as a promoter of tyrosine kinase inhibitor (TKI) resistance in non–small cell lung cancer (NSCLC). However, the landscape of MET-specific targeting agents remains underdeveloped, and understanding of mechanisms of resistance to MET TKIs is limited. Here, we present a case of a patient with lung adenocarcinoma harboring both a mutation in EGFR and an amplification of MET, who after progression on erlotinib responded dramatically to combined MET and EGFR inhibition with savolitinib and osimertinib. When resistance developed to this combination, a new MET kinase domain mutation, D1228V, was detected. Our in vitro findings demonstrate that METD1228V induces resistance to type I MET TKIs through impaired drug binding, while sensitivity to type II MET TKIs is maintained. Based on these findings, the patient was treated with erlotinib combined with cabozantinib, a type II MET inhibitor, and exhibited a response.Significance: With several structurally distinct MET inhibitors undergoing development for treatment of NSCLC, it is critical to identify mechanism-based therapies for drug resistance. We demonstrate that an acquired METD1228V mutation mediates resistance to type I, but not type II, MET inhibitors, having therapeutic implications for the clinical use of sequential MET inhibitors. Cancer Discov; 6(12); 1334–41. ©2016 AACR.See related commentary by Trusolino, p. 1306.This article is highlighted in the In This Issue feature, p. 1293
- Published
- 2023
- Full Text
- View/download PDF
24. Novel Macrocyclic Peptidomimetics Targeting the Polo-Box Domain of Polo-Like Kinase 1
- Author
-
SeongShick Ryu, Jung-Eun Park, Young Jin Ham, Daniel C. Lim, Nicholas P. Kwiatkowski, Do-Hee Kim, Debabrata Bhunia, Nam Doo Kim, Michael B. Yaffe, Woolim Son, Namkyoung Kim, Tae-Ik Choi, Puspanjali Swain, Cheol-Hee Kim, Jin-Young Lee, Nathanael S. Gray, Kyung S. Lee, and Taebo Sim
- Subjects
Molecular Structure ,Cell Cycle Proteins ,Protein Serine-Threonine Kinases ,Peptides, Cyclic ,Molecular Docking Simulation ,Structure-Activity Relationship ,HEK293 Cells ,Protein Domains ,Proto-Oncogene Proteins ,Drug Discovery ,Animals ,Humans ,Molecular Medicine ,Peptidomimetics ,Protein Kinase Inhibitors ,Zebrafish ,HeLa Cells ,Protein Binding - Abstract
The polo-box domain (PBD) of Plk1 is a promising target for cancer therapeutics. We designed and synthesized novel phosphorylated macrocyclic peptidomimetics targeting PBD based on acyclic phosphopeptide PMQSpTPL. The inhibitory activities of
- Published
- 2022
- Full Text
- View/download PDF
25. Ethyl Gallate Isolated from Castanopsis cuspidata var. sieboldii Branches Inhibits Melanogenesis and Promotes Autophagy in B16F10 Cells
- Author
-
Moon-Hee Choi, Seung-Hwa Yang, Da-Song Kim, Nam-Doo Kim, and Hyun-Jae Shin
- Subjects
autophagy enhancer ,AKT pathway ,Physiology ,ethyl gallate ,Clinical Biochemistry ,Cell Biology ,Castanopsis cuspidata var. sieboldii ,Molecular Biology ,Biochemistry ,antimelanogenesis - Abstract
The Castanopsis cuspidata var. sieboldii (CCS) plant grows predominantly in temperate regions of Asian countries, such as South Korea. Research on CCS has so far concentrated on the nutritional analysis, antioxidant activity, and anti-inflammation properties of its branches. However, the isolation of compounds and structural elucidation of effective single molecules remain unexplored, necessitating further exploration of CCS branches. Therefore, this study demonstrates the antioxidant and antimelanogenic activity of a single substance of ethyl gallate (EG) isolated from CCS branch extracts. Notably, the antimelanogenic (whitening) activity of EG extracted from CCS branches remains unexplored. Tyrosinase inhibition, kinetic enzyme assays, and molecular docking studies were conducted using mushroom tyrosinase in order to examine the antioxidant mechanism and antimelanin activity of EG in B16F10 melanoma cells. Nontoxic EG concentrations were found to be below 5 µg/mL. While EG significantly reduced the levels of whitening-associated proteins, p-CREB, and p-PKA, it dose-dependently inhibited the expression of TYR, TRP-1, TRP-2, and transcription factor (MITF). In addition, EG downregulated melanogenetic gene expression and activated autophagy signals. Therefore, EG extracted from CCS branches could serve as a novel functional cosmetic material with antimelanogenic and autophagy-enhancing activity.
- Published
- 2023
- Full Text
- View/download PDF
26. Sesamin Ameliorates Lipotoxicity and Lipid Accumulation Through the Activation of the Estrogen Receptor Alpha Signaling Pathway
- Author
-
Thi Hoa Pham, Gi Ho Lee, Sun Woo Jin, Seung Yeon Lee, Eun Hee Han, Nam Doo Kim, Chul Yung Choi, Gil-Saeng Jeong, Sang Ki Lee, Hyung Sik Kim, and Hye Gwang Jeong
- Published
- 2023
- Full Text
- View/download PDF
27. Identification of Thiazolo[5,4-b]pyridine Derivatives as c-KIT Inhibitors for Overcoming Imatinib Resistance
- Author
-
Yunju Nam, Chan Kim, Junghee Han, SeongShick Ryu, Hanna Cho, Chiman Song, Nam Doo Kim, Namkyoung Kim, and Taebo Sim
- Subjects
Cancer Research ,Oncology ,c-KIT ,GIST ,GIST-T1 ,HMC1.2 ,imatinib resistance ,thiazolo[5,4-b]pyridine - Abstract
c-KIT is a promising therapeutic target against gastrointestinal stromal tumor (GIST). In order to identify novel c-KIT inhibitors capable of overcoming imatinib resistance, we synthesized 31 novel thiazolo[5,4-b]pyridine derivatives and performed SAR studies. We observed that, among these substances, 6r is capable of inhibiting significantly c-KIT and suppressing substantially proliferation of GIST-T1 cancer cells. It is of note that 6r is potent against a c-KIT V560G/D816V double mutant resistant to imatinib. Compared with sunitinib, 6r possesses higher differential cytotoxicity on c-KIT D816V Ba/F3 cells relative to parental Ba/F3 cells. In addition, kinase panel profiling reveals that 6r has reasonable kinase selectivity. It was found that 6r remarkably attenuates proliferation of cancer cells via blockade of c-KIT downstream signaling, and induction of apoptosis and cell cycle arrest. Furthermore, 6r notably suppresses migration and invasion, as well as anchorage-independent growth of GIST-T1 cells. This study provides useful SAR information for the design of novel c-KIT inhibitors overcoming imatinib-resistance.
- Published
- 2022
- Full Text
- View/download PDF
28. Identification of Thiazolo[5,4
- Author
-
Yunju, Nam, Chan, Kim, Junghee, Han, SeongShick, Ryu, Hanna, Cho, Chiman, Song, Nam Doo, Kim, Namkyoung, Kim, and Taebo, Sim
- Abstract
c-KIT is a promising therapeutic target against gastrointestinal stromal tumor (GIST). In order to identify novel c-KIT inhibitors capable of overcoming imatinib resistance, we synthesized 31 novel thiazolo[5,4
- Published
- 2022
29. Identification of Thieno[3,2-d]pyrimidine Derivatives as Dual Inhibitors of Focal Adhesion Kinase and FMS-like Tyrosine Kinase 3
- Author
-
Chiman Song, SeongShick Ryu, Nam Doo Kim, Nam Hoon Kwon, Taebo Sim, Injae Shin, Eunhye Jeon, Ji Won Lee, Youngji Moon, Young Hoon Kim, Hojong Yoon, Sunghoon Kim, Hwan Geun Choi, and Hanna Cho
- Subjects
Kinase ,Mutant ,Myeloid leukemia ,Drug resistance ,medicine.disease ,Metastasis ,Focal adhesion ,chemistry.chemical_compound ,chemistry ,Drug Discovery ,Fms-Like Tyrosine Kinase 3 ,Cancer research ,medicine ,Molecular Medicine ,Growth inhibition - Abstract
Focal adhesion kinase (FAK) is overexpressed in highly invasive and metastatic cancers. To identify novel FAK inhibitors, we designed and synthesized various thieno[3,2-d]pyrimidine derivatives. An intensive structure-activity relationship (SAR) study led to the identification of 26 as a lead. Moreover, 26, a multitargeted kinase inhibitor, possesses excellent potencies against FLT3 mutants as well as FAK. Gratifyingly, 26 remarkably inhibits recalcitrant FLT3 mutants, including F691L, that cause drug resistance. Importantly, 26 is superior to PF-562271 in terms of apoptosis induction, anchorage-independent growth inhibition, and tumor burden reduction in the MDA-MB-231 xenograft mouse model. Also, 26 causes regression of tumor growth in the MV4-11 xenograft mouse model, indicating that it could be effective against acute myeloid leukemia (AML). Finally, in an orthotopic mouse model using MDA-MB-231, 26 remarkably prevents metastasis of orthotopic tumors to lymph nodes. Taken together, the results indicate that 26 possesses potential therapeutic value against highly invasive cancers and relapsed AML.
- Published
- 2021
- Full Text
- View/download PDF
30. X-ray Crystal Structure-Guided Design and Optimization of 7H-Pyrrolo[2,3-d]pyrimidine-5-carbonitrile Scaffold as a Potent and Orally Active Monopolar Spindle 1 Inhibitor
- Author
-
Seo Kyung Ah, Son Jung Beom, Ikyon Kim, Jun Goo Jee, Nam Doo Kim, Kim Hyunkyung, Haelee Kim, Eunhwa Ko, Hwan Geun Choi, Younho Lee, and Ha Yeon Cho
- Subjects
0303 health sciences ,Scaffold ,Pyrimidine ,Crystal structure ,Tnbc cell ,01 natural sciences ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Orally active ,chemistry ,In vivo ,Drug Discovery ,Cancer research ,Molecular Medicine ,Protein kinase A ,Monopolar spindle ,030304 developmental biology - Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast-cancer subtype associated with poor prognosis and high relapse rates. Monopolar spindle 1 kinase (MPS1) is an apical dual-specificity protein kinase that is over-expressed in TNBC. We herein report a highly selective MPS1 inhibitor based on a 7H-pyrrolo[2,3-d]pyrimidine-5-carbonitrile scaffold. Our lead optimization was guided by key X-ray crystal structure analysis. In vivo evaluation of candidate (9) is shown to effectively mitigate human TNBC cell proliferation.
- Published
- 2021
- Full Text
- View/download PDF
31. Discovery of a selective inhibitor of doublecortin like kinase 1
- Author
-
Jinhua Wang, Senthil Muthaswamy, Kevin M. Haigis, Rita Sulahian, Kenneth D. Westover, Emily J. Poulin, Ryoma Ohi, Sergio Espinosa, Shuning He, Taebo Sim, Lianbo Li, Miljan Kuljanin, Nathanael S. Gray, Yan Liu, Nam Doo Kim, Joseph D. Mancias, Ling Huang, Brian M. Wolpin, Charles Y. Lin, Andrew J. Aguirre, Srivatsan Raghavan, Nora Diéguez-Martínez, Radha L. Kalekar, Jost Vrabic Koren, Zeng Zhiyang, Fleur M. Ferguson, James D Vasta, Behnam Nabet, William C. Hahn, Raymond W.S. Ng, Cesear Corona, Wayne Harshbarger, Alan L. Leggett, Matthew B. Robers, A. Thomas Look, Jose M. Lizcano, and Annan Yang
- Subjects
Male ,Proteomics ,Doublecortin Protein ,Protein Serine-Threonine Kinases ,Biology ,Article ,Transcriptome ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,Doublecortin-Like Kinases ,Downregulation and upregulation ,Cell Movement ,Cell Line, Tumor ,Organoid ,Animals ,Humans ,Protein Kinase Inhibitors ,Molecular Biology ,Zebrafish ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,Molecular Structure ,Kinase ,030302 biochemistry & molecular biology ,Intracellular Signaling Peptides and Proteins ,Phosphoproteomics ,Cell Biology ,Rats ,Molecular Docking Simulation ,Pancreatic Neoplasms ,Gene Expression Regulation ,Protein kinase domain ,Cancer research ,Drug Screening Assays, Antitumor ,Carcinoma, Pancreatic Ductal - Abstract
Doublecortin like kinase 1 (DCLK1) is an understudied kinase that is upregulated in a wide range of cancers, including pancreatic ductal adenocarcinoma (PDAC). However, little is known about its potential as a therapeutic target. We used chemoproteomic profiling and structure-based design to develop a selective, in vivo-compatible chemical probe of the DCLK1 kinase domain, DCLK1-IN-1. We demonstrate activity of DCLK1-IN-1 against clinically relevant patient-derived PDAC organoid models and use a combination of RNA-sequencing, proteomics and phosphoproteomics analysis to reveal that DCLK1 inhibition modulates proteins and pathways associated with cell motility in this context. DCLK1-IN-1 will serve as a versatile tool to investigate DCLK1 biology and establish its role in cancer. A highly selective inhibitor of the DCLK1/2 kinases is used to uncover the consequences of DCLK1 inhibition on viability, phosphosignaling and the transcriptome in patient-derived organoid models of pancreatic ductal adenocarcinoma.
- Published
- 2020
- Full Text
- View/download PDF
32. Identification of Pyridinyltriazine Derivatives as Potent panFGFR Inhibitors against Gatekeeper Mutants for Overcoming Drug Resistance
- Author
-
SeongShick Ryu, Yunju Nam, Namkyoung Kim, Injae Shin, Eunhye Jeon, Younghoon Kim, Nam Doo Kim, and Taebo Sim
- Subjects
Mice ,Cell Line, Tumor ,Drug Discovery ,Drug Resistance ,Molecular Medicine ,Animals ,Humans ,Antineoplastic Agents ,Apoptosis ,Protein Kinase Inhibitors ,Cell Proliferation ,Signal Transduction - Abstract
Although FGFR inhibitors hold promise in treating various cancers, resistance to the FGFR inhibitors caused by acquired secondary mutations has emerged. To discover novel FGFR inhibitors capable of inhibiting FGFR mutations, including gatekeeper mutations, we designed and synthesized several new pyridinyltriazine derivatives. A structure-activity relationship (SAR) study led to the identification of
- Published
- 2022
33. Puerarin attenuates hepatic steatosis via G-protein-coupled estrogen receptor-mediated calcium and SIRT1 signaling pathways
- Author
-
Thi Hoa Pham, Gi Ho Lee, Sun Woo Jin, Seung Yeon Lee, Eun Hee Han, Nam Doo Kim, and Hye Gwang Jeong
- Subjects
Pharmacology ,Mice, Obese ,Hep G2 Cells ,AMP-Activated Protein Kinases ,Lipid Metabolism ,Isoflavones ,Lipids ,Mice ,Liver ,Receptors, Estrogen ,Sirtuin 1 ,GTP-Binding Proteins ,Non-alcoholic Fatty Liver Disease ,Animals ,Humans ,Calcium ,Signal Transduction - Abstract
Puerarin, the major bioactive ingredient isolated from the root of Pueraria lobata (Willd.), attenuates body weight gain and reduces lipid levels in high-fat diet-induced obese mice; however, the underlying mechanism responsible for regulating lipid metabolism remains unclear. This study investigated the molecular mechanism(s) underlying the role of puerarin in regulating lipogenesis and lipolysis in human HepG2 cells. In this study, puerarin strongly inhibited the expression of fatty acid synthase (FASN) and sterol regulatory element binding protein 1c (SREBP-1c). Moreover, puerarin significantly induced the expression of adipose triglyceride lipase (ATGL), which is responsible for triacylglycerol hydrolase activity in cells. Puerarin enhanced 5' AMP-activated protein kinase (AMPK) activity, which is a central regulator of hepatic lipid metabolism. Furthermore, this AMPK activation could be mediated by sirtuin 1 (SIRT1) and calcium signaling pathways involved in G protein-coupled estrogen receptor (GPER) signaling. GPER blockage significantly reversed the effect of puerarin on lipid accumulation and the related signaling pathways. Docking studies showed that puerarin could bind in the GPER in a similar manner as GPER agonist G1. Our results suggest that puerarin can improve hepatic steatosis by activating GPER; it's signaling cascade sequentially induced calcium and SIRT1 signaling pathways. Thus, puerarin may be a potential therapeutic agent for the treatment of non-alcoholic fatty liver disease.
- Published
- 2022
34. Regioselectivity Prediction of CYP1A2-Mediated Phase I Metabolism.
- Author
-
Jihoon Jung, Nam Doo Kim, Su Yeon Kim, Inhee Choi, Kwang-Hwi Cho, Wonseok Oh, Doo Nam Kim, and Kyoung Tai No
- Published
- 2008
- Full Text
- View/download PDF
35. p21-activated kinase 4 inhibition protects against liver ischemia/reperfusion injury: Role of nuclear factor erythroid 2-related factor 2 phosphorylation
- Author
-
Yuancheng Mao, Chang Yeob Han, Lihua Hao, Younho Lee, Jung Beom Son, Hwangeun Choi, Mi Rin Lee, Jae Do Yang, Suk Kyun Hong, Kyung‐Suk Suh, Hee Chul Yu, Nam Doo Kim, Eun Ju Bae, and Byung‐Hyun Park
- Subjects
Mice, Knockout ,Mice ,Hepatology ,Liver ,p21-Activated Kinases ,Ischemia ,NF-E2-Related Factor 2 ,Liver Diseases ,Reperfusion Injury ,Animals ,Humans ,Apoptosis ,Phosphorylation - Abstract
p21-activated kinase 4 (PAK4), an oncogenic protein, has emerged as a promising target for anticancer drug development. Its role in oxidative stress conditions, however, remains elusive. We investigated the effects of PAK4 signaling on hepatic ischemia/reperfusion (I/R) injury.Hepatocyte- and myeloid-specific Pak4 knockout (KO) mice and their littermate controls were subjected to a partial hepatic I/R (HIR) injury. We manipulated the catalytic activity of PAK4, either through genetic engineering (gene knockout, overexpression of wild-type [WT] or dominant-negative kinase) or pharmacological inhibitor, coupled with a readout of nuclear factor erythroid 2-related factor 2 (Nrf2) activity, to test the potential function of PAK4 on HIR injury. PAK4 expression was markedly up-regulated in liver during HIR injury in mice and humans. Deletion of PAK4 in hepatocytes, but not in myeloid cells, ameliorated liver damages, as demonstrated in the decrease in hepatocellular necrosis and inflammatory responses. Conversely, the forced expression of WT PAK4 aggravated the pathological changes. PAK4 directly phosphorylated Nrf2 at T369, and it led to its nuclear export and proteasomal degradation, all of which impaired antioxidant responses in hepatocytes. Nrf2 silencing in liver abolished the protective effects of PAK4 deficiency. A PAK4 inhibitor protected mice from HIR injury.PAK4 phosphorylates Nrf2 and suppresses its transcriptional activity. Genetic or pharmacological suppression of PAK4 alleviates HIR injury. Thus, PAK4 inhibition may represent a promising intervention against I/R-induced liver injury.
- Published
- 2022
36. Acacetin Inhibits the Growth of STAT3-Activated DU145 Prostate Cancer Cells by Directly Binding to Signal Transducer and Activator of Transcription 3 (STAT3)
- Author
-
Nam Doo Kim, Sun Yun, Jiyeon Choi, Byoung-Mog Kwon, Dong Cho Han, and Yu-Jin Lee
- Subjects
Male ,Models, Molecular ,STAT3 Transcription Factor ,natural product ,Pharmaceutical Science ,Gene Expression ,Mice, Nude ,Organic chemistry ,Apoptosis ,Article ,Analytical Chemistry ,chemistry.chemical_compound ,Mice ,QD241-441 ,DU145 ,Downregulation and upregulation ,Cell Line, Tumor ,acacetin ,Drug Discovery ,label-free methods ,Animals ,Humans ,Physical and Theoretical Chemistry ,Tyrosine ,Phosphorylation ,STAT3 ,Cell Proliferation ,antitumor ,Mice, Inbred BALB C ,Acacetin ,biology ,STAT3 inhibitor ,Prostatic Neoplasms ,Protein-Tyrosine Kinases ,Flavones ,Antineoplastic Agents, Phytogenic ,Xenograft Model Antitumor Assays ,Cell biology ,chemistry ,Chemistry (miscellaneous) ,STAT protein ,biology.protein ,Molecular Medicine ,Female ,Reactive Oxygen Species ,Protein Binding - Abstract
Signal transducer and activator of transcription 3 (STAT3) plays a critical role in the formation and growth of human cancer. Therefore, STAT3 is a therapeutic target for cancer drug discovery. Acacetin, a flavone present in various plants, inhibits constitutive and inducible STAT3 activation in STAT3-activated DU145 prostate cancer cells. Acacetin inhibits STAT3 activity by directly binding to STAT3, which we confirmed by a pull-down assay with a biotinylated compound and two level-free methods, namely, a drug affinity responsive target stability (DARTS) experiment and a cellular thermal shift assay (CETSA). Acacetin inhibits STAT3 phosphorylation at the tyrosine 705 residue and nuclear translocation in DU145 cells, which leads to the downregulation of STAT3 target genes. Acacetin then induces apoptosis in a time-dependent manner. Interestingly, acacetin induces the production of reactive oxygen species (ROS) that are not involved in the acacetin-induced inhibition of STAT3 activation because the suppressed p-STAT3 level is not rescued by treatment with GSH or NAC, which are general ROS inhibitors. We also found that acacetin inhibits tumor growth in xenografted nude mice. These results suggest that acacetin, as a STAT3 inhibitor, could be a possible drug candidate for targeting STAT3 for the treatment of cancer in humans.
- Published
- 2021
- Full Text
- View/download PDF
37. Pimozide suppresses cancer cell migration and tumor metastasis through binding to ARPC2, a subunit of the Arp2/3 complex
- Author
-
Yu-Jin Lee, Byoung-Mog Kwon, Jiyeon Choi, Seung-Jin Park, Seon-Young Kim, Dong Cho Han, Cheol-Hee Kim, Nam Doo Kim, and Yae Jin Yoon
- Subjects
actin‐related protein 2/3 complex subunit 2 ,0301 basic medicine ,Cancer Research ,actin‐related protein 2/3 complex ,Arp2/3 complex ,Antineoplastic Agents ,Actin-Related Protein 2-3 Complex ,Focal adhesion ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Pimozide ,Cell Movement ,Cell Line, Tumor ,medicine ,Animals ,Humans ,metastasis ,Neoplasm Invasiveness ,Neoplasm Metastasis ,Gene knockdown ,Binding Sites ,drug repurposing ,biology ,Drug discovery ,Chemistry ,Original Articles ,General Medicine ,Cell biology ,Drug Discovery and Delivery ,030104 developmental biology ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Invadopodia ,Cancer cell ,biology.protein ,Original Article ,medicine.drug - Abstract
ARPC2 is a subunit of the Arp2/3 complex, which is essential for lamellipodia, invadopodia and filopodia, and ARPC2 has been identified as a migrastatic target molecule. To identify ARPC2 inhibitors, we generated an ARPC2 knockout DLD‐1 human colon cancer cell line using the clustered regularly interspaced short palindromic repeats/CRISPR‐associated protein 9 (CRISPR/Cas9) system and explored gene signature‐based strategies, such as a connectivity map (CMap) using the gene expression profiling data of ARPC2 knockout and knockdown cells. From the CMap‐based drug discovery strategy, we identified pimozide (a clinically used antipsychotic drug) as a migrastatic drug and ARPC2 inhibitor. Pimozide inhibited the migration and invasion of various cancer cells. Through drug affinity responsive target stability (DARTS) analysis and cellular thermal shift assay (CETSA), it was confirmed that pimozide directly binds to ARPC2. Pimozide increased the lag phase of Arp2/3 complex‐dependent actin polymerization and inhibited the vinculin‐mediated recruitment of ARPC2 to focal adhesions in cancer cells. To validate the likely binding of pimozide to ARPC2, mutant cells, including ARPC2F225A, ARPC2F247A and ARPC2Y250F cells, were prepared using ARPC2 knockout cells prepared by gene‐editing technology. Pimozide strongly inhibited the migration of mutant cells because the mutated ARPC2 likely has a larger binding pocket than the wild‐type ARPC2. Therefore, pimozide is a potential ARPC2 inhibitor, and ARPC2 is a new molecular target. Taken together, the results of the present study provide new insights into the molecular mechanism and target that are responsible for the antitumor and antimetastatic activity of pimozide., Pimozide is identified as a migrastatic drug and ARPC2 inhibitor from connectivity map‐based drug discovery strategy. Pimozide inhibits migration and invasion in various cancer cell lines, and suppresses metastasis in an in vivo antimetastatic assay. Through drug affinity responsive target stability (DARTS) analysis and cellular thermal shift assay (CETSA), it was confirmed that pimozide directly binds to ARPC2.
- Published
- 2019
- Full Text
- View/download PDF
38. Identification of a Unique Resorcylic Acid Lactone Derivative That Targets Both Lymphangiogenesis and Angiogenesis
- Author
-
Youngsun Han, Nam Doo Kim, Jeong Hun Kim, Hanna Cho, Jin Hyoung Kim, Sandip Sengupta, Taebo Sim, Byung Joo Lee, Hwan Geun Choi, Jiknyeo Kim, and Uttam Dash
- Subjects
Angiogenesis ,Neovascularization, Physiologic ,Apoptosis ,01 natural sciences ,Neovascularization ,Lactones ,Structure-Activity Relationship ,03 medical and health sciences ,Cell Movement ,In vivo ,Cell Line, Tumor ,Drug Discovery ,medicine ,Humans ,Structure–activity relationship ,Lymphangiogenesis ,Naphthyridines ,Protein Kinase Inhibitors ,030304 developmental biology ,chemistry.chemical_classification ,Tube formation ,0303 health sciences ,Chemistry ,Kinase ,Resorcinols ,Vascular Endothelial Growth Factor Receptor-3 ,Vascular Endothelial Growth Factor Receptor-2 ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,fms-Like Tyrosine Kinase 3 ,Biochemistry ,Molecular Medicine ,medicine.symptom ,Lactone - Abstract
We synthesized 11 novel L-783277 derivatives, in which a structure rigidifying phenyl ring is incorporated into the 14-membered chiral resorcylic acid lactone system. The SAR study with these substances demonstrated that 17 possesses excellent kinase selectivity against a panel of 335 kinases in contrast to L-783277 and inhibits VEGFR3, VEGFR2, and FLT3 with single-digit nanomolar IC50 values. Also, we found that 21, a stereoisomer of 17, has excellent potency (IC50 = 9 nM) against VEGFR3 and selectivity over VEGFR2 and FLT3. 17, a potent dual VEGFR3 and VEGFR2 inhibitor, effectively suppresses both lymphangiogenesis and angiogenesis in a 3D-microfluidic tumor lymphangiogenesis assay and in vivo corneal assay while SAR131675 blocks only lymphangiogenesis. In addition, 17 blocks the endothelial tube formation and suppresses proliferation of PHE tumor vascular model. 17 will be a valuable templatefor developing therapeutically active and selective substances that target both lymphangiogenesis and angiogenesis.
- Published
- 2019
- Full Text
- View/download PDF
39. Identification of Novel Resorcinol Amide Derivatives as Potent and Specific Pyruvate Dehydrogenase Kinase (PDHK) Inhibitors
- Author
-
Sungmi Park, Inkyu Lee, Injae Shin, Nam Doo Kim, Mi Jin Kim, Taebo Sim, Eun Kyung Yoo, Hojong Yoon, Kyungseon Cho, and Hanna Cho
- Subjects
Pyruvate dehydrogenase kinase ,Apoptosis ,Molecular Dynamics Simulation ,medicine.disease_cause ,01 natural sciences ,Inhibitory Concentration 50 ,03 medical and health sciences ,Cell Line, Tumor ,Neoplasms ,Drug Discovery ,Cell Adhesion ,medicine ,Humans ,Enzyme Inhibitors ,Phosphorylation ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,Cell growth ,Kinase ,Chemistry ,Pyruvate Dehydrogenase Acetyl-Transferring Kinase ,Resorcinols ,Pyruvate dehydrogenase complex ,Amides ,0104 chemical sciences ,Molecular Docking Simulation ,010404 medicinal & biomolecular chemistry ,Biochemistry ,Cell culture ,Cancer cell ,Molecular Medicine ,Peptides ,Reactive Oxygen Species ,Carcinogenesis ,Hydrophobic and Hydrophilic Interactions - Abstract
Pyruvate dehydrogenase kinases (PDHKs) promote abnormal respiration in cancer cells. Studies with novel resorcinol amide derivatives based on VER-246608 (6) led to the identification of 19n and 19t containing five-membered heteroaromatic rings as unique structural features. These substances possess single-digit nanomolar activities against PDHKs. 19t exhibits higher potencies against PDHK1/2/4 than does 6 and inhibits only PDHKs among 366 kinases. Moreover, 19g, 19l, and 19s were found to be isotype-selective PDHK inhibitors. Molecular dynamics simulations provide a better understanding of how the heteroaromatic rings affect the activities of 19n and 19t on PDHK1/2/3/4. Moreover, 19n possesses a much higher antiproliferative activity against cancer cells than does 6. We demonstrated that the results of PDH assays better correlate with cellular activities than do those of PDHK kinase assays. Furthermore, 19n induces apoptosis of cancer cells via mitochondrial dysfunction, suppresses tumorigenesis, and displays a synergistic effect on satraplatin suppression of cancer cell proliferation.
- Published
- 2019
- Full Text
- View/download PDF
40. An Inhibitor of the Interaction of Survivin with Smac in Mitochondria Promotes Apoptosis
- Author
-
Seong Hyun Park, Sanghyun Park, Nam Doo Kim, Injae Shin, and Insu Shin
- Subjects
Survivin ,Cell ,Apoptosis ,Mitochondrion ,010402 general chemistry ,01 natural sciences ,Biochemistry ,Protein–protein interaction ,Structure-Activity Relationship ,Phenothiazines ,Cell Line, Tumor ,medicine ,Humans ,Protein Interaction Domains and Motifs ,Binding Sites ,biology ,010405 organic chemistry ,Chemistry ,Cytochrome c ,Organic Chemistry ,Cytochromes c ,General Chemistry ,Mitochondria ,0104 chemical sciences ,Cell biology ,Molecular Docking Simulation ,Cytosol ,medicine.anatomical_structure ,Cancer cell ,biology.protein ,biological phenomena, cell phenomena, and immunity ,Oligopeptides - Abstract
Herein we report the first small molecule that disrupts the survivin-Smac interaction taking place in mitochondria. The inhibitor, PZ-6-QN, was identified by initially screening a phenothiazine library using a fluorescence anisotropy assay and then conducting a structure-activity relationship study. Mutagenesis and molecular docking studies suggest that PZ-6-QN binds to survivin similarly to the known Smac peptide, AVPI. The results of the effort also show that PZ-6-QN exhibits good anticancer activity against various cancer cells. Moreover, cell-based mechanistic studies provide evidence for the proposal that PZ-6-QN enters mitochondria to inhibit the survivin-Smac interaction and promotes release of Smac and cytochrome c from mitochondria into the cytosol, a process that induces apoptosis in cancer cells. Overall, the present study suggests that PZ-6-QN can serve as a novel chemical probe for study of processes associated with the mitochondrial survivin-Smac interaction and it will aid the discovery of novel anticancer agents.
- Published
- 2019
- Full Text
- View/download PDF
41. Identification of Thieno[3,2
- Author
-
Hanna, Cho, Injae, Shin, Hojong, Yoon, Eunhye, Jeon, Jiwon, Lee, Younghoon, Kim, SeongShick, Ryu, Chiman, Song, Nam Hoon, Kwon, Youngji, Moon, Sunghoon, Kim, Nam Doo, Kim, Hwan Geun, Choi, and Taebo, Sim
- Subjects
Mice, Inbred BALB C ,Molecular Structure ,Mice, Nude ,Antineoplastic Agents ,Thiophenes ,Xenograft Model Antitumor Assays ,Molecular Docking Simulation ,Structure-Activity Relationship ,Pyrimidines ,fms-Like Tyrosine Kinase 3 ,Cell Line, Tumor ,Focal Adhesion Kinase 1 ,Neoplasms ,Animals ,Humans ,Female ,Drug Screening Assays, Antitumor ,Neoplasm Metastasis ,Phosphorylation ,Protein Kinase Inhibitors - Abstract
Focal adhesion kinase (FAK) is overexpressed in highly invasive and metastatic cancers. To identify novel FAK inhibitors, we designed and synthesized various thieno[3,2
- Published
- 2021
42. A Novel Protein–Protein Interaction between RSK3 and IκBα and a New Binding Inhibitor That Suppresses Breast Cancer Tumorigenesis
- Author
-
Hee Sub Yoon, Ju Yong Hyon, Yeji Yang, Jung Hyun Park, Young Ho Chung, Sung Hoon Choi, Ji Hoon Lee, Jin-Young Min, Nam Doo Kim, Sejin Jung, Eun Hee Han, Jae-Young Kim, and Sung Gil Chi
- Subjects
0301 basic medicine ,Cancer Research ,Programmed cell death ,Cell ,IκBα ,RSK3 (RPS6KA2) ,medicine.disease_cause ,Article ,Ribosomal s6 kinase ,03 medical and health sciences ,0302 clinical medicine ,breast cancer ,medicine ,binding inhibition ,protein-protein interaction (PPI) ,RC254-282 ,cell-based unidentified protein interaction discovery (CUPID) ,biology ,Cell growth ,Kinase ,Chemistry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Apoptosis ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Carcinogenesis - Abstract
Simple Summary Breast cancer is the most common carcinoma and the leading cause of cancer-related death among women worldwide. Many kinases play important roles in the tumorigenesis of various cancers. IκBα phosphorylation is important for the regulation of NF-κB activity and is linked to the regulation of tumorigenesis. However, the kinase signaling network regulating IκBα phosphorylation in the context of cancer is not well understood. Herein, we report that RSK3 (RPS6KA2) is a novel binding partner of IκBα. RSK3 induces IκBα phosphorylation and NF-κB activation. Further, a chemical screening approach identified an inhibitor of RSK3/IκBα binding that impairs RSK3-mediated IκBα phosphorylation and decreases breast cancer cell survival, proliferation, and migration. Abstract Multiple cancer-related biological processes are mediated by protein-protein interactions (PPIs). Through interactions with a variety of factors, members of the ribosomal S6 kinase (RSK) family play roles in cell cycle progression and cell proliferation. In particular, RSK3 contributes to cancer viability, but the underlying mechanisms remain unknown. We performed a kinase library screen to find IκBα PPI binding partners and identified RSK3 as a novel IκBα binding partner using a cell-based distribution assay. In addition, we discovered a new PPI inhibitor using mammalian two-hybrid (MTH) analysis. We assessed the antitumor effects of the new inhibitor using cell proliferation and colony formation assays and monitored the rate of cell death by FACS apoptosis assay. IκBα is phosphorylated by the active form of the RSK3 kinase. A small-molecule inhibitor that targets the RSK3/IκBα complex exhibited antitumor activity in breast cancer cells and increased their rate of apoptosis. RSK3 phosphorylation and RSK3/IκBα complex formation might be functionally important in breast tumorigenesis. The RSK3/IκBα-specific binding inhibitor identified in this study represents a lead compound for the development of new anticancer drugs.
- Published
- 2021
43. X-ray Crystal Structure-Guided Design and Optimization of 7
- Author
-
Younho, Lee, Hyunkyung, Kim, Haelee, Kim, Ha Yeon, Cho, Jun-Goo, Jee, Kyung-Ah, Seo, Jung Beom, Son, Eunhwa, Ko, Hwan Geun, Choi, Nam Doo, Kim, and Ikyon, Kim
- Subjects
Mice, Inbred ICR ,Binding Sites ,Transplantation, Heterologous ,Administration, Oral ,Breast Neoplasms ,Cell Cycle Proteins ,Protein Serine-Threonine Kinases ,Protein-Tyrosine Kinases ,Crystallography, X-Ray ,Molecular Docking Simulation ,Mice ,Structure-Activity Relationship ,Pyrimidines ,Cell Line, Tumor ,Drug Design ,Animals ,Humans ,Female ,Pyrroles ,Protein Kinase Inhibitors ,Half-Life - Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast-cancer subtype associated with poor prognosis and high relapse rates. Monopolar spindle 1 kinase (MPS1) is an apical dual-specificity protein kinase that is over-expressed in TNBC. We herein report a highly selective MPS1 inhibitor based on a 7
- Published
- 2021
44. Novel Quercetin Derivative of 3,7-Dioleylquercetin Shows Less Toxicity and Highly Potent Tyrosinase Inhibition Activity
- Author
-
Hyun-Jae Shin, Da-Song Kim, Kechun Liu, Moon-Hee Choi, Nam Doo Kim, and Seung-Hwa Yang
- Subjects
Antioxidant ,medicine.medical_treatment ,Tyrosinase ,01 natural sciences ,chemistry.chemical_compound ,0302 clinical medicine ,enzyme kinetics ,quercetin derivatives ,heterocyclic compounds ,Biology (General) ,Spectroscopy ,food and beverages ,General Medicine ,Computer Science Applications ,Molecular Docking Simulation ,Chemistry ,Biochemistry ,030220 oncology & carcinogenesis ,Toxicity ,Quercetin ,QH301-705.5 ,Antineoplastic Agents ,Catalysis ,Article ,Inorganic Chemistry ,03 medical and health sciences ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Enzyme kinetics ,RNA, Messenger ,Physical and Theoretical Chemistry ,Molecular Biology ,QD1-999 ,Melanins ,3,7-dioleylquercetin ,skin whitening agent ,010405 organic chemistry ,Organic Chemistry ,fungi ,zebrafish ,In vitro ,0104 chemical sciences ,Kinetics ,stomatognathic diseases ,tyrosinase inhibitor ,chemistry ,Cell culture ,anti-melanogenesis - Abstract
Quercetin is a well-known plant flavonol and antioxidant, however, there has been some debate regarding the efficacy and safety of native quercetin as a skin-whitening agent via tyrosinase inhibition. Several researchers have synthesized quercetin derivatives as low-toxicity antioxidants and whitening agents. However, no suitable quercetin derivatives have been reported to date. In this study, a novel quercetin derivative was synthesized by the SN2 reaction using quercetin and oleyl bromide. The relationship between the structures and activities of quercetin derivatives as anti-melanogenic agents was assessed using in vitro enzyme kinetics, molecular docking, and quenching studies, cell line experiments, and in vivo zebrafish model studies. Novel 3,7-dioleylquercetin (OQ) exhibited a low cytotoxic concentration level at >, 100 µg/mL (125 µM), which is five times less toxic than native quercetin. The inhibition mechanism showed that OQ is a competitive inhibitor, similar to native quercetin. Expression of tyrosinase, tyrosinase-related protein 1 (TRP-1) and tyrosinase-related protein 2 (TRP-2), and microphthalmia-associated transcription factor was inhibited in B16F10 melanoma cell lines. mRNA transcription levels of tyrosinase, TRP-1, and TRP-2 decreased in a dose-dependent manner. Melanin formation was confirmed in the zebrafish model using quercetin derivatives. Therefore, OQ might be a valuable asset for the development of novel skin-whitening agents.
- Published
- 2021
45. Dutomycin Induces Autophagy and Apoptosis by Targeting the Serine Protease Inhibitor SERPINB6
- Author
-
In Ja Ryoo, Mina Jang, Jae-Hyuk Jang, Nak Kyun Soung, Min Jung Kim, Hyemin Seo, Bo Yeon Kim, Hiroyuki Osada, Sung Kyun Ko, Yong Tae Kwon, Seung Min Kim, Jong Seog Ahn, Mincheol Kwon, Nam Doo Kim, Sangkeun Son, Gun-Hee Kim, and Shuta Hara
- Subjects
0301 basic medicine ,Proteases ,Antineoplastic Agents ,Apoptosis ,01 natural sciences ,Biochemistry ,Serine ,03 medical and health sciences ,Neoplasms ,Autophagy ,Animals ,Humans ,Anthracyclines ,Serpins ,Zebrafish ,Cell Proliferation ,Serine protease ,Gene knockdown ,biology ,010405 organic chemistry ,Chemistry ,General Medicine ,Xenograft Model Antitumor Assays ,0104 chemical sciences ,Cell biology ,030104 developmental biology ,biology.protein ,Molecular Medicine ,Target protein ,Serine Proteases ,Intracellular ,HeLa Cells - Abstract
Autophagy plays an important role in maintaining tumor cell progression and survival in response to metabolic stress. Thus, the regulation of autophagy can be used as a strategy for anticancer therapy. Here, we report dutomycin (DTM) as a novel autophagy enhancer that eventually induces apoptosis due to excessive autophagy. Also, human serine protease inhibitor B6 (SERPINB6) was identified as a target protein of DTM, and its novel function which is involved in autophagy was studied for the first time. We show that DTM directly binds SERPINB6 and then activates intracellular serine proteases, resulting in autophagy induction. Inhibitory effects of DTM on the function of SERPINB6 were confirmed through enzyme- and cell-based approaches, and SERPINB6 was validated as a target protein using siRNA-mediated knockdown and an overexpression test. In a zebrafish xenograft model, DTM showed a significant decrease in tumor area. Furthermore, the present findings will be expected to contribute to the expansion of novel basic knowledge about the correlation of cancer and autophagy by promoting active further research on SERPINB6, which was not previously considered the subject of cancer biology.
- Published
- 2021
46. Abstract LB521: Antitumor activity of potent RAF inhibitors in solid tumors with activated RAS-RAF axis
- Author
-
Kwangwoo Hwang, SeoHyun Jo, Jieun Choi, Ga-young Choi, Jiseon Choi, Ji-Hye Kwon, Dong-Guk Shin, Jiyeon Kim, Se-Hyuk Kim, Haelee Kim, Ha Yeon Cho, Jung Beom Son, Nam Doo Kim, Hwan Geun Choi, Daekwon Kim, and Sunghwan Kim
- Subjects
Cancer Research ,Oncology - Abstract
FDA approved three RAF inhibitors for the treatment of tumors containing BRAFV600 mutations, but one of the major drawbacks of these type I RAF inhibitors is to activate MAPK signaling pathway, instead of inhibiting signaling, which is referred to as paradoxical activation. Such undesired paradoxical activation not only leads to renewed tumor growth but also spurs additional cancer growth in non-cancerous wild-type BRAF tissue. Plus, these first-generation RAF inhibitors targeting BRAFV600 mutants are unable to inhibit oncogenic RAF dimers. This has led to the development of type II RAF inhibitors such as belvarafenib and day101 to block the activity of multiple forms of RAF while avoiding paradoxical activation. Two lead-like stage compounds 1 and 2 were specifically designed as type II RAF inhibitors to have activity across RAF isoforms including BRAFV600E, BRAF wild-type, and CRAF, showing higher potency than competitors. Compounds 1 and 2 potently inhibited the growth of BRAFV600E melanoma cells and NRAS or KRAS mutant cancer cells. Compounds 1 and 2 promoted the formation of BRAF/CRAF heterodimers by directly binding to the RAF kinase domain like other type II RAF inhibitors and inhibited phosphorylation of downstream effectors MEK and ERK in a dose-dependent manner in RAS mutant cancer cells, suggesting less paradoxical activation liability. Compound 2 showed superior on-target inhibitory activity for BRAFV600E, BRAF wild-type, and CRAF than belvarafenib through RAF immunoprecipitation (IP) kinase assay. Inhibition of the RAF downstream signaling was also confirmed by quantifying the level of phospho-ERK in NRAS or KRAS mutant cancer cells. In HCT116 (KRASG13D) subcutaneous xenograft model, compound 1 showed tumor growth inhibition efficacy, suggesting a potential to address RAS mutant driven- as well as BRAFV600 mutant driven tumors. Also, combination treatment with MEK inhibitor and/or immune checkpoint inhibitor would further improve the therapeutic activity and expand target indication for unmet medical needs. Citation Format: Kwangwoo Hwang, SeoHyun Jo, Jieun Choi, Ga-young Choi, Jiseon Choi, Ji-Hye Kwon, Dong-Guk Shin, Jiyeon Kim, Se-Hyuk Kim, Haelee Kim, Ha Yeon Cho, Jung Beom Son, Nam Doo Kim, Hwan Geun Choi, Daekwon Kim, Sunghwan Kim. Antitumor activity of potent RAF inhibitors in solid tumors with activated RAS-RAF axis [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr LB521.
- Published
- 2022
- Full Text
- View/download PDF
47. First SAR Study for Overriding NRAS Mutant Driven Acute Myeloid Leukemia
- Author
-
Eunmi Hong, Wooyoung Hur, Nam Doo Kim, Eunhye Ju, Hanna Cho, Hwan Geun Choi, Injae Shin, Taebo Sim, Nathanael S. Gray, Haelee Kim, and Seunghye Choi
- Subjects
Models, Molecular ,0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,Myeloid ,Cell Survival ,Protein Conformation ,Mutant ,Mice, Nude ,Antineoplastic Agents ,Apoptosis ,GTP Phosphohydrolases ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,0302 clinical medicine ,Drug Discovery ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,Protein kinase B ,Molecular Structure ,Chemistry ,Kinase ,Membrane Proteins ,Myeloid leukemia ,medicine.disease ,Xenograft Model Antitumor Assays ,Leukemia, Myeloid, Acute ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Molecular Medicine ,Female ,Signal transduction ,Signal Transduction - Abstract
GNF-7, a multitargeted kinase inhibitor, served as a dual kinase inhibitor of ACK1 and GCK, which provided a novel therapeutic strategy for overriding AML expressing NRAS mutation. This SAR study with GNF-7 derivatives, designed to target NRAS mutant-driven AML, led to identification of the extremely potent inhibitors, 10d, 10g, and 11i, which possess single-digit nanomolar inhibitory activity against both ACK1 and GCK. These substances strongly suppress proliferation of mutant NRAS expressing AML cells via apoptosis and AKT/mTOR signaling blockade. Compound 11i is superior to GNF-7 in terms of kinase inhibitory activity, cellular activity, and differential cytotoxicity. Moreover, 10k possessing a favorable mouse pharmacokinetic profile prolonged life-span of Ba/F3-NRAS-G12D injected mice and significantly delayed tumor growth of OCI-AML3 xenograft model without causing the prominent level of toxicity found with GNF-7. Taken together, this study provides insight into the design of novel ACK1 and GCK dual inhibitors for overriding NRAS mutant-driven AML.
- Published
- 2018
- Full Text
- View/download PDF
48. Structural and Atropisomeric Factors Governing the Selectivity of Pyrimido-benzodiazipinones as Inhibitors of Kinases and Bromodomains
- Author
-
Mingfeng Hao, Fleur M. Ferguson, Dario R. Alessi, Xianming Deng, Michael R. McKeown, Jinwei Zhang, Stephen C. Blacklow, Taebo Sim, Jun Qi, Lingling Dai, Nam Doo Kim, Nathanael S. Gray, Néstor Gómez, Nora Diéguez-Martínez, Amy DiBona, Tatiana Erazo, Nana K. Offei-Addo, Pau Muñoz-Guardiola, James E. Bradner, Paul M.C. Park, Oleg Fedorov, Dennis L. Buckley, Walter Massefski, Jose M. Lizcano, Jinhua Wang, Xiang Xu, Kelly Becht, and Justin M. Roberts
- Subjects
Models, Molecular ,0301 basic medicine ,BRD4 ,Polypharmacology ,Cell Cycle Proteins ,Crystallography, X-Ray ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Biochemistry ,Article ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,Humans ,Structure–activity relationship ,Binding site ,Protein Kinase Inhibitors ,Mitogen-Activated Protein Kinase 7 ,Benzodiazepinones ,Chemistry ,Kinase ,Nuclear Proteins ,General Medicine ,TG101348 ,Bromodomain ,Pyrimidines ,030104 developmental biology ,Docking (molecular) ,Molecular Medicine ,Pharmacophore ,HeLa Cells ,Transcription Factors - Abstract
Bromodomains have been pursued intensively over the past several years as emerging targets for the development of anti-cancer and anti-inflammatory agents. It has recently been shown that some kinase inhibitors are able to potently inhibit the bromodomains of BRD4. The clinical activities of PLK inhibitor BI-2536 and JAK2-FLT3 inhibitor TG101348 have been attributed to this unexpected poly-pharmacology, indicating that dual-kinase/bromodomain activity may be advantageous in a therapeutic context. However, for target validation and biological investigation, a more selective target profile is desired. Here we report that benzo[e]pyrimido-[5,4-b]diazepine-6(11H)-ones, versatile ATP-site directed kinase pharmacophores utilized in the development of inhibitors of multiple kinases including a number of previously reported kinase chemical probes, are also capable of exhibiting potent BRD4-dependent pharmacology. Using a dual kinase-bromodomain inhibitor of the kinase domains of ERK5 and LRRK2, and the bromodomain of BRD4 as a case study, we define the structure-activity relationships required to achieve dual kinase/BRD4 activity as well as how to direct selectivity towards inhibition of either ERK5 or BRD4. This effort resulted in identification of one of the first reported kinase-selective chemical probes for ERK5 (JWG-071), a BET selective inhibitor with 1 μM BRD4 IC(50) (JWG-115), and additional inhibitors with rationally designed polypharmacology (JWG-047, JWG-069). Co-crystallography of seven representative inhibitors with the first bromodomain of BRD4 demonstrate that distinct atropisomeric conformers recognize the kinase ATP-site and the BRD4 acetyl lysine binding site, conformational preferences supported by rigid docking studies.
- Published
- 2018
- Full Text
- View/download PDF
49. 15-Deoxy-Δ12,14-prostaglandin J2 activates PI3K-Akt signaling in human breast cancer cells through covalent modification of the tumor suppressor PTEN at cysteine 136
- Author
-
Sin-Aye Park, Hye-Kyung Na, Young-Ger Suh, Jinyoung Suh, Nam-Doo Kim, Jeong-Hoon Jang, Do-Hee Kim, Jong Min Park, Nam-Jung Kim, Su-Jung Kim, Young-Joon Surh, and Eun-Hee Kim
- Subjects
0301 basic medicine ,Cancer Research ,biology ,Chemistry ,Cancer ,medicine.disease ,Dithiothreitol ,Serine ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,medicine ,Cancer research ,biology.protein ,PTEN ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cysteine - Abstract
15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2), one of the terminal products of cyclooxygenase-2-catalized arachidonic acid metabolism, has been shown to stimulate breast cancer cell proliferation and migration through Akt activation, but the underlying mechanisms remain poorly understood. In the present study, we investigated the effects of 15d-PGJ2 on the activity of PTEN, the inhibitor of the phosphoinositide 3-kinase (PI3K)-Akt axis, in human breast cancer (MCF-7) cells. Since the α,β-unsaturated carbonyl moiety in the cyclopentenone ring of 15d-PGJ2 is electrophilic, we hypothesized that 15d-PGJ2-induced Akt phosphorylation might result from the covalent modification and subsequent inactivation of PTEN that has several critical cysteine residues. When treated to MCF-7 cells, 15d-PGJ2 bound to PTEN, and this was abolished in the presence of the thiol-reducing agent dithiothreitol. A mass spectrometric analysis by using recombinant and endogenous PTEN protein revealed that the cysteine 136 residue (Cys136) of PTEN is covalently modified upon treatment with 15d-PGJ2. Notably, the ability of 15d-PGJ2 to covalently bind to PTEN as well as to induce Akt phosphorylation was abolished in the cells expressing a mutant form of PTEN in which Cys136 was replaced by serine (C136S-PTEN). The present study demonstrates for the first time that electrophilic 15d-PGJ2 directly binds to cysteine 136 of PTEN and provides new insight into PTEN loss in cancer progression associated with chronic inflammation. These observations suggest that 15d-PGJ2 can undergo nucleophilic addition to PTEN, presumably at Cys136, thereby inactivating this tumor suppressor protein with concomitant Akt activation.
- Published
- 2018
- Full Text
- View/download PDF
50. Abstract P234: ORIC-114, an orally bioavailable, irreversible kinase inhibitor, has superior brain penetrant properties and enhanced potency in preclinical studies of HER2-positive breast cancer
- Author
-
Melissa R. Junttila, Jason E. Long, Robert Warne, Sunghwan Kim, Younho Lee, Hwan Kim, Juhee Kang, Jiyoon Seok, Jihye Yoo, Youngyi Lee, Dong-Hyuk Seo Seo, Jung Beom Son, Daekwon Kim, Hwan Geun Choi, Nam Doo Kim, Tatiana Zavorotinskaya, Chelsea Chan, Matthew Panuwat, Jessica Sun, Jae H. Chang, and Lori S. Friedman
- Subjects
Cancer Research ,Oncology - Abstract
Amplification of human epidermal growth factor receptor 2 (HER2) is an oncogenic driver found in approximately 25% of breast cancer. Despite the arsenal of HER2-directed therapies available to patients, more than 50% of patients with HER2 amplification eventually develop central nervous system (CNS) metastases over the course of their disease indicating a clear medical need for brain penetrant therapies in this patient population. ORIC-114 is a brain penetrant, orally bioavailable, irreversible small molecule inhibitor that was designed to target exon20 insertions in epidermal growth factor receptor (EGFR) and HER2. ORIC-114 is highly selective for the EGFR/HER2 family of receptors, reducing the risk for off-target kinase liabilities. In biochemical assays, ORIC-114 displayed low nanomolar potency on HER2. To explore the application of ORIC-114 in the HER2-amplified tumor setting, a cell viability screen was performed against a panel of human breast cancer lines containing both HER2-amplified and non HER2-amplified cell lines. ORIC-114 demonstrated greater than 100-fold enhanced cellular potency on HER2-amplified cancer cell lines relative to non-amplified cancer cell lines. Notably, ORIC-114 cellular EC50s in HER2-amplified breast cancer cell lines were below 30 nM and more potent than both lapatinib and tucatinib, two FDA-approved tyrosine kinase inhibitors for the treatment of HER2-positive breast cancer. ORIC-114 was designed to incorporate physicochemical properties suitable to cross the blood-brain barrier and has exhibited good brain penetration across multiple preclinical species. To further investigate the brain penetrant attributes of ORIC-114 in the context of HER2-positive breast cancer with brain metastases, key features were assessed relative to tucatinib, which has demonstrated clinical efficacy in this setting. In contrast to tucatinib, ORIC-114 displayed minimal impact on efflux transporters as it was only a weak substrate for P-glycoprotein (P-gp) and was not a substrate for breast cancer associated protein (BCRP) in vitro. In addition, ORIC-114 demonstrated superior in vivo brain penetration relative to tucatinib as measured by free brain-to-plasma ratio in mouse. Consequently, ORIC-114 free brain concentrations in rodents were greater than tucatinib, even when the active metabolites of tucatinib were considered. Taken together, these data further establish ORIC-114 as a selective, irreversible, and brain penetrant EGFR/HER2 inhibitor, making it a promising therapeutic candidate for development in patients with HER2-positive tumors including those with CNS metastases. A Clinical Trial Application for ORIC-114 is anticipated in the second half of 2021. Citation Format: Melissa R. Junttila, Jason E. Long, Robert Warne, Sunghwan Kim, Younho Lee, Hwan Kim, Juhee Kang, Jiyoon Seok, Jihye Yoo, Youngyi Lee, Dong-Hyuk Seo Seo, Jung Beom Son, Daekwon Kim, Hwan Geun Choi, Nam Doo Kim, Tatiana Zavorotinskaya, Chelsea Chan, Matthew Panuwat, Jessica Sun, Jae H. Chang, Lori S. Friedman. ORIC-114, an orally bioavailable, irreversible kinase inhibitor, has superior brain penetrant properties and enhanced potency in preclinical studies of HER2-positive breast cancer [abstract]. In: Proceedings of the AACR-NCI-EORTC Virtual International Conference on Molecular Targets and Cancer Therapeutics; 2021 Oct 7-10. Philadelphia (PA): AACR; Mol Cancer Ther 2021;20(12 Suppl):Abstract nr P234.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.