1
|
Yang X, Hu R, Yao L, Zhang W, Shi M, Gong J, Yuan X, Li Y, Yan J, Wang Y, Zhang Q, He Z, Hou DX, Fan Z, Zhang H, Chen L, He X, He J, Wu S. The role of uterus mitochondrial function in high-fat diet-related adverse pregnancy outcomes and protection by resveratrol. Food Funct 2024; 15:4852-4861. [PMID: 38573228 DOI: 10.1039/d4fo00671b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
This study elucidates the mechanism of obesity-related adverse pregnancy outcomes and further investigates the effect of resveratrol on reproductive performance in a short- or long-term HFD-induced obese mouse model. Results show that maternal weight had a significant positive correlation with litter mortality in mice. A long-term HFD increased body weight and litter mortality with decreased expression of uterine cytochrome oxidase 4 (COX4), which was recovered by resveratrol in mice. Moreover, HFD decreased the expression of peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), nuclear respiratory factors-1 (Nrf-1), and phosphorylated adenosine 5'-monophosphate (AMP)-activated protein kinase (p-AMPK) and increased the expression of phosphorylated extracellular regulated protein kinases (p-ERK) in the uterus. Resveratrol, a polyphenol that can directly bind to the ERK protein, suppressed the phosphorylation of ERK, increased the expression of p-AMPK, PGC-1α and Nrf-1, and decreased litter mortality in mice.
Collapse
Affiliation(s)
- Xizi Yang
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China.
| | - Ruizhi Hu
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China.
| | - Liping Yao
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China.
| | - Wentao Zhang
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China.
| | - Mingkun Shi
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China.
| | - Jiatai Gong
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China.
| | - Xupeng Yuan
- College of Animal Science and Technology, Hunan Biological and Electromechanical Polytechnic, Changsha 410127, China
| | - Yanli Li
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China.
| | - Jiahao Yan
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China.
| | - Ying Wang
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China.
| | - Qianjin Zhang
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China.
| | - Ziyu He
- Department of Food Science and Biotechnology, Faculty of Agriculture, Kagoshima University, Kagoshima 890-0065, Japan
| | - De-Xing Hou
- Department of Food Science and Biotechnology, Faculty of Agriculture, Kagoshima University, Kagoshima 890-0065, Japan
| | - Zhiyong Fan
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China.
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Liang Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xi He
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China.
| | - Jianhua He
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China.
| | - Shusong Wu
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
2
|
Choi JI, Song WS, Koh DH, Kim EY. In Silico and In Vitro multiple analysis approach for screening naturally derived ligands for red seabream aryl hydrocarbon receptor. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 275:116262. [PMID: 38569320 DOI: 10.1016/j.ecoenv.2024.116262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/20/2024] [Accepted: 03/23/2024] [Indexed: 04/05/2024]
Abstract
The aryl hydrocarbon receptor (AHR) is a key ligand-dependent transcription factor that mediates the toxic effects of compounds such as dioxin. Recently, natural ligands of AHR, including flavonoids, have been attracting physiological and toxicological attention as they have been reported to regulate major biological functions such as inflammation and anti-cancer by reducing the toxic effects of dioxin. Additionally, it is known that natural AHR ligands can accumulate in wildlife tissues, such as fish. However, studies in fish have investigated only a few ligands in experimental fish species, and the AHR response of marine fish to natural AHR ligands of various other structures has not been thoroughly investigated. To explore various natural AHR ligands in marine fish, which make up the most fish, it is necessary to develop new screening methods that consider the specificity of marine fish. In this study, we investigated the response of natural ligands by constructing in vitro and in silico experimental systems using red seabream as a model species. We attempted to develop a new predictive model to screen potential ligands that can induce transcriptional activation of red seabream AHR1 and AHR2 (rsAHR1 and rsAHR2). This was achieved through multiple analyses using in silico/ in vitro data and Tox21 big data. First, we constructed an in vitro reporter gene assay of rsAHR1 and rsAHR2 and measured the response of 10 representatives natural AHR ligands in COS-7 cells. The results showed that FICZ, Genistein, Daidzein, I3C, DIM, Quercetin and Baicalin induced the transcriptional activity of rsAHR1 and rsAHR2, while Resveratrol and Retinol did not induce the transcriptional activity of rsAHR isoforms. Comparing the EC50 values of the respective compounds in rsAHR1 and rsAHR2, FICZ, Genistein, and Daidzein exhibited similar isoform responses, but I3C, Baicalin, DIM and Quercetin show the isoform-specific responses. These results suggest that natural AHR ligands have specific profiling and transcriptional activity for each rsAHR isoform. In silico analysis, we constructed homology models of the ligand binding domains (LBDs) of rsAHR1 and rsAHR2 and calculated the docking energies (U_dock values) of natural ligands with measured in vitro transcriptional activity and dioxins reported in previous studies. The results showed a significant correlation (R2=0.74(rsAHR1), R2=0.83(rsAHR2)) between docking energy and transcriptional activity (EC50) value, suggesting that the homology model of rsAHR1 and rsAHR2 can be utilized to predict the potential transactivation of ligands. To broaden the applicability of the homology model to diverse compound structures and validate the correlation with transcriptional activity, we conducted additional analyses utilizing Tox21 big data. We calculated the docking energy values for 1860 chemicals in both rsAHR1 and rsAHR2, which were tested for transcriptional activation in Tox21 data against human AHR. By comparing the U_dock energy values between 775 active compounds and 1085 inactive compounds, a significant difference (p<0.001) was observed between the U_dock energy values in the two groups, suggesting that the U_dock value can be applied to distinguish the activation of compounds. Furthermore, we observed a significant correlation (R2=0.45) between the AC50 of Tox21 database and U_dock values of human AHR model. In conclusion, we calculated equations to translate the results of an in silico prediction model for ligand screening of rsAHR1 and rsAHR2 transactivation. This ligand screening model can be a powerful tool to quantitatively estimate AHR transactivation of major marine agents to which red seabream may be exposed. The study introduces a new screening approach for potential natural AHR ligands in marine fish, based on homology model-docking energy values of rsAHR1 and rsAHR2, with implications for future agonist development and applications bridging in silico and in vitro data.
Collapse
Affiliation(s)
- Jong-In Choi
- Department of Biology, Kyung Hee University, Seoul, Republic of Korea
| | - Woo-Seon Song
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Republic of Korea
| | - Dong-Hee Koh
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Republic of Korea
| | - Eun-Young Kim
- Department of Biology, Kyung Hee University, Seoul, Republic of Korea; Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Watanabe N, Shinozaki Y, Ogiwara S, Miyagasako R, Sasaki A, Kato J, Suzuki Y, Fukunishi N, Okada Y, Saito T, Iida Y, Higashiseto M, Masuda H, Nagata E, Gotoh K, Amino M, Tsuji T, Morita S, Nakagawa Y, Hirayama N, Inokuchi S. Diphenyl-tetrazol-propanamide Derivatives Act as Dual-Specific Antagonists of Platelet CLEC-2 and Glycoprotein VI. Thromb Haemost 2024; 124:203-222. [PMID: 37967855 DOI: 10.1055/a-2211-5202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
BACKGROUND Platelet C-type lectin-like receptor 2 (CLEC-2) induces platelet activation and aggregation after clustering by its ligand podoplanin (PDPN). PDPN, which is not normally expressed in cells in contact with blood flow, is induced in inflammatory immune cells and some malignant tumor cells, thereby increasing the risk of venous thromboembolism (VTE) and tumor metastasis. Therefore, small-molecule compounds that can interfere with the PDPN-CLEC-2 axis have the potential to become selective antiplatelet agents. METHODS AND RESULTS Using molecular docking analysis of CLEC-2 and a PDPN-CLEC-2 binding-inhibition assay, we identified a group of diphenyl-tetrazol-propanamide derivatives as novel CLEC-2 inhibitors. A total of 12 hit compounds also inhibited PDPN-induced platelet aggregation in humans and mice. Unexpectedly, these compounds also fit the collagen-binding pocket of the glycoprotein VI molecule, thereby inhibiting collagen interaction. These compounds also inhibited collagen-induced platelet aggregation, and one compound ameliorated collagen-induced thrombocytopenia in mice. For clinical use, these compounds will require a degree of chemical modification to decrease albumin binding. CONCLUSION Nonetheless, as dual activation of platelets by collagen and PDPN-positive cells is expected to occur after the rupture of atherosclerotic plaques, these dual antagonists could represent a promising pharmacophore, particularly for arterial thrombosis, in addition to VTE and metastasis.
Collapse
Affiliation(s)
- Nobuo Watanabe
- Department of Emergency and Critical Care Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- Institute of Advanced Biosciences, Tokai University, Hiratsuka, Kanagawa, Japan
| | - Yoshiko Shinozaki
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Sanae Ogiwara
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Riko Miyagasako
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Ayumi Sasaki
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Junko Kato
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Yusuke Suzuki
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Natsuko Fukunishi
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Yoshinori Okada
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Takeshi Saito
- Department of Emergency and Critical Care Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Yumi Iida
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Misaki Higashiseto
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Haruchika Masuda
- Department of Physiology, Tokai University School of Medicine, Shimokasuya, Isehara, Kanagawa, Japan
| | - Eiichiro Nagata
- Department of Neurology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Kazuhito Gotoh
- Department of Laboratory Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Mari Amino
- Department of Emergency and Critical Care Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Tomoatsu Tsuji
- Department of Emergency and Critical Care Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Seiji Morita
- Department of Emergency and Critical Care Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Yoshihide Nakagawa
- Department of Emergency and Critical Care Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Noriaki Hirayama
- Institute of Advanced Biosciences, Tokai University, Hiratsuka, Kanagawa, Japan
- The Institute of Medical Sciences, Tokai University, Isehara, Kanagawa, Japan
| | - Sadaki Inokuchi
- Department of Emergency and Critical Care Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- Institute of Advanced Biosciences, Tokai University, Hiratsuka, Kanagawa, Japan
| |
Collapse
|
4
|
Soufi W, Allali H, Hacene FB, Ghalem S. Molecular Modeling of Brassicaceae Derivatives for Inhibiting Lipoxygenases: A Promising Therapeutic Strategy. Curr Drug Discov Technol 2024; 21:48-e011223224117. [PMID: 39206704 DOI: 10.2174/0115701638269042231122064738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/28/2023] [Accepted: 10/06/2023] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Inflammation plays a crucial role in the body's defense mechanisms, but uncontrolled inflammation can lead to chronic and pathological conditions. This study aimed to identify natural compounds as potential replacements for the synthetic drug Zileuton, known for its side effects. METHOD Utilizing the MOE and Molegro modeling methods, several molecules were evaluated, and three compounds, namely 1-Isothiocyanatopent-4-en-2-ol, 7-Isothiocyanatohept-1-ene, and 5- (Isothiocyanatomethyl)-1,2,3-trimethoxybenzene, exhibited superior inhibitory properties. These compounds consistently demonstrated low energy values, indicating high inhibition potency. Notably, 5-(Isothiocyanatomethyl)-1,2,3-trimethoxybenzene emerged as the most promising candidate among all tested compounds. RESULTS These findings provide valuable insights for the development of alternative anti-inflammatory agents. Further research is required to assess the efficacy and safety profiles of these compounds in clinical settings. CONCLUSION This study represents a significant advancement in the search for innovative therapeutic strategies to manage inflammation-related disorders.
Collapse
Affiliation(s)
- Wassila Soufi
- Laboratory of Natural and Bioactive Substances (LASNABIO), Department of Chemistry, Faculty of Sciences, Abou Bekr Belkaïd University, P.O. Box 119, Tlemcen 13000, Algeria
- Department of Chemistry, Mustapha Stambouli University, P.O. Box 305, Mascara, 29000, Algeria
| | - Hocine Allali
- Department of Chemistry, Faculty of Sciences, Abou Bekr Belkaïd University, P.O. Box 119, Tlemcen 13000, Algeria
| | - Faïza Boukli Hacene
- Laboratory of Natural and Bioactive Substances (LASNABIO), Department of Chemistry, Faculty of Sciences, Abou Bekr Belkaïd University, P.O. Box 119, Tlemcen 13000, Algeria
- Department of Chemistry, Faculty of Sciences, Abou Bekr Belkaïd University, P.O. Box 119, Tlemcen 13000, Algeria
| | - Saïd Ghalem
- Laboratory of Natural and Bioactive Substances (LASNABIO), Department of Chemistry, Faculty of Sciences, Abou Bekr Belkaïd University, P.O. Box 119, Tlemcen 13000, Algeria
- Department of Chemistry, Faculty of Sciences, Abou Bekr Belkaïd University, P.O. Box 119, Tlemcen 13000, Algeria
| |
Collapse
|
5
|
Nguyen HT, Yoshinouchi Y, Hirano M, Nomiyama K, Nakata H, Kim EY, Iwata H. In silico simulations and molecular descriptors to predict in vitro transactivation potencies of Baikal seal estrogen receptors by environmental contaminants. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 265:115495. [PMID: 37748367 DOI: 10.1016/j.ecoenv.2023.115495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/31/2023] [Accepted: 09/16/2023] [Indexed: 09/27/2023]
Abstract
Baikal seals (Pusa sibirica) are vulnerable to high levels of organic pollutants. Here, we evaluated the transactivation potencies of bisphenols (BPs) and hydroxylated polychlorinated biphenyls (OH-PCBs) via the Baikal seal estrogen receptor α and β (bsERα and bsERβ) using in vitro and in silico approaches. In vitro reporter gene assays showed that most BPs and OH-PCBs exhibited estrogenic activity with bsER sub-type-specific potency. Among the BPs tested, bisphenol AF showed the lowest EC50 for both bsERs. 4'-OH-CB50 and 4'-OH-CB30 showed the lowest EC50 among OH-PCBs tested for bsERα and bsERβ, respectively. 4-((4-Isopropoxyphenyl)-sulfonyl)phenol, 4'-OH-CB72, and 4'-OH-CB121 showed weak bsERα-specific transactivation. Only 4-OH-CB107 did not affect both bsERs. In silico docking simulations revealed the binding affinities of these chemicals to bsERs and partially explained the in vitro results. Using the in silico simulations and molecular descriptors as explanatory variables and the in vitro results as objective variables, the quantitative structure-activity relationship (QSAR) models constructed for classification and regression accurately separated bsER-active compounds from non-active compounds and predicted the in vitro bsERα- and bsERβ-transactivation potencies, respectively. The QSAR models also suggested that chemical polarity, van der Waals surface area, bridging atom structure, position of the phenolic-OH group, and ligand interactions with key residues of the ligand binding pocket are critical variables to account for the bsER transactivation potency of the test compounds. We also succeeded in constructing computational models for predicting in vitro transactivation potencies of mouse ERs in the same manner, demonstrating the applicability of our approach independent of species-specific responses.
Collapse
Affiliation(s)
- Hoa Thanh Nguyen
- Center for Marine Environmental Studies, Ehime University, Matsuyama 7908577, Japan
| | - Yuka Yoshinouchi
- Center for Marine Environmental Studies, Ehime University, Matsuyama 7908577, Japan
| | - Masashi Hirano
- Department of Food and Life Science, School of Agriculture, Tokai University, Kumamoto 8612055, Japan
| | - Kei Nomiyama
- Center for Marine Environmental Studies, Ehime University, Matsuyama 7908577, Japan
| | - Haruhiko Nakata
- Faculty of Advanced Science and Technology, Kumamoto University, Kumamoto 8608555, Japan
| | - Eun-Young Kim
- Department of Life and Nanopharmaceutical Science and Department of Biology, Kyung Hee University, Seoul 130701, Republic of Korea
| | - Hisato Iwata
- Center for Marine Environmental Studies, Ehime University, Matsuyama 7908577, Japan.
| |
Collapse
|
6
|
Seto T. General anesthetic binding mode via hydration with weak affinity and molecular discrimination: General anesthetic dissolution in interfacial water of the common binding site of GABA A receptor. Biophys Physicobiol 2023; 20:e200005. [PMID: 38496235 PMCID: PMC10941959 DOI: 10.2142/biophysico.bppb-v20.0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 01/23/2023] [Indexed: 01/25/2023] Open
Abstract
The GABAA receptor (GABAAR) is a target channel for the loss of awareness of general anesthesia. General anesthetic (GA) spans a wide range of chemical structures, such as monatomic molecules, barbital acids, phenols, ethers, and alkanes. GA has a weak binding affinity, and the affinity has a characteristic that correlates with the solubility in olive oil rather than the molecular shape. The GA binding site of GABAAR is common to GAs and exists in the transmembrane domain of the GABAAR intersubunit. In this study, the mechanism of GA binding, which allows binding of various GAs with intersubunit selectivity, was elucidated from the hydration analysis of the binding site. Regardless of the diverse GA chemical structures, a strong correlation was observed between the binding free energy and total dehydration number of the binding process. The GA binding free energy was more involved in the binding dehydration and showed molecular recognition that allowed for the binding of various GA structures via binding site hydration. We regarded the GA substitution for the interfacial water molecule of the binding site as a dissolution into the interfacial hydration layer. The elucidation of the GA binding mechanism mediated by hydration at the GABAAR common binding site provides a rationale for the combined use of anesthetics in medical practice and its combination adjustments via drug interactions.
Collapse
Affiliation(s)
- Tomoyoshi Seto
- Department of Anesthesiology, School of Medicine, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| |
Collapse
|
7
|
El Ashry ESH, Farahat MM, Awad LF, Balbaa M, Yusef H, Badawy ME, Abd Al Moaty MN. New 4-(arylidene)amino-1,2,4-traizole-5-thiol derivatives and their acyclo thioglycosides as α-glucosidase and α-amylase inhibitors: Design, synthesis, and molecular modelling studies. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132733] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
8
|
Unique Mode of Antiviral Action of a Marine Alkaloid against Ebola Virus and SARS-CoV-2. Viruses 2022; 14:v14040816. [PMID: 35458549 PMCID: PMC9028129 DOI: 10.3390/v14040816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/24/2022] [Accepted: 04/02/2022] [Indexed: 11/16/2022] Open
Abstract
Lamellarin α 20-sulfate is a cell-impenetrable marine alkaloid that can suppress infection that is mediated by the envelope glycoprotein of human immunodeficiency virus type 1. We explored the antiviral action and mechanisms of this alkaloid against emerging enveloped RNA viruses that use endocytosis for infection. The alkaloid inhibited the infection of retroviral vectors that had been pseudotyped with the envelope glycoprotein of Ebola virus and SARS-CoV-2. The antiviral effects of lamellarin were independent of the retrovirus Gag-Pol proteins. Interestingly, although heparin and dextran sulfate suppressed the cell attachment of vector particles, lamellarin did not. In silico structural analyses of the trimeric glycoprotein of the Ebola virus disclosed that the principal lamellarin-binding site is confined to a previously unappreciated cavity near the NPC1-binding site and fusion loop, whereas those for heparin and dextran sulfate were dispersed across the attachment and fusion subunits of the glycoproteins. Notably, lamellarin binding to this cavity was augmented under conditions where the pH was 5.0. These results suggest that the final action of the alkaloid against Ebola virus is specific to events following endocytosis, possibly during conformational glycoprotein changes in the acidic environment of endosomes. Our findings highlight the unique biological and physicochemical features of lamellarin α 20-sulfate and should lead to the further use of broadly reactive antivirals to explore the structural mechanisms of virus replication.
Collapse
|
9
|
Synthesis, antibacterial, antioxidant, and molecular docking studies of 6-methylpyrimidin-4(3H)-one and oxo-1,2,4-triazolo[4,3-a]pyrimidine derivatives. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.131551] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
10
|
Koh DH, Song WS, Kim EY. Multi-step structure-activity relationship screening efficiently predicts diverse PPARγ antagonists. CHEMOSPHERE 2022; 286:131540. [PMID: 34346341 DOI: 10.1016/j.chemosphere.2021.131540] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 06/13/2023]
Abstract
In discovering the potential antagonist of peroxisome proliferator-activated receptor gamma (PPARγ), the structure-activity relationship (SAR) is a useful in silico method. However, it is difficult for conventional SAR approaches to predict the activities of antagonists owing to the large structural diversity of antagonistic compounds. This study provides evidence that multi-step SAR screening is applicable for predicting PPARγ antagonists by combining different complementary methodologies. We constructed three models: read-across-like SAR, docking-simulation-interpreting SAR, and deep-learning-based SAR. To provide user-customized prediction results, our multi-step SAR screening model combined the three SAR models in a stepwise manner, which subdivided them according to potential levels of the PPARγ antagonist. The read-across-like SAR, which considered specific antagonist scaffolds, revealed the highest positive predictive value (PPV). The docking-simulation-interpreting SAR, which considered the molecular surface features, revealed high statistics for the PPV and the true-positive rate (TPR). The deep-learning-based SAR showed the highest TPR at the last classification step. This multi-step SAR screening covered the antagonists of high reliability provided by a read-across-like SAR, as well as the antagonists of diverse scaffolds provided by docking-simulation-interpreting SAR and deep-learning-based SAR. Therefore, to predict PPARγ antagonists, multi-step SAR screening could be as a useful tool.
Collapse
Affiliation(s)
- Dong-Hee Koh
- Department of Life and Nanopharmaceutical Science, South Korea
| | - Woo-Seon Song
- Department of Life and Nanopharmaceutical Science, South Korea
| | - Eun-Young Kim
- Department of Life and Nanopharmaceutical Science, South Korea; Department of Biology, Kyung Hee University, Hoegi-Dong, Dongdaemun-Gu, Seoul, 130-701, South Korea.
| |
Collapse
|
11
|
Lohning A, Kidachi Y, Kamiie K, Sasaki K, Ryoyama K, Yamaguchi H. 6-(methylsulfinyl)hexyl isothiocyanate (6-MITC) from Wasabia japonica alleviates inflammatory bowel disease (IBD) by potential inhibition of glycogen synthase kinase 3 beta (GSK-3β). Eur J Med Chem 2021; 216:113250. [PMID: 33691258 DOI: 10.1016/j.ejmech.2021.113250] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/08/2020] [Accepted: 01/25/2021] [Indexed: 02/06/2023]
Abstract
Inflammatory bowel disease (IBD) describes a set of disorders involving alterations to gastrointestinal physiology and mucosal immunity. Unravelling its complex pathophysiology is important since many IBD patients are refractory to or suffer adverse side effects from current treatments. Isothiocyanates (ITCs), such as 6-(methylsulfinyl)hexyl ITC (6-MITC) in Wasabia japonica, have potential anti-inflammatory activity. We aimed to elucidate the pathways through which 6-MITC alleviates inflammation by examining its role in the nuclear factor-kappa B (NF-κB) pathway through inhibition of glycogen synthase kinase 3 beta (GSK-3β) using a chemically induced murine model of IBD, cell-based and in silico techniques. The effects of 6-MITC and two NF-κB inhibitors, sulfasalazine (SS), pyrrolidine dithiolcarbamate (PDTC) were investigated on a dextran sulfate sodium (DSS)-induced murine mouse model of acute and chronic colitis using macroscopic measurements and pro-inflammatory markers. The effect of 6-MITC on NF-κB induction was assessed using a murine macrophage cell line. Complexes of GSK-3β-6-MITC and GSK-3β-ATP were generated in silico to elucidate the mechanism of 6-MITC's direct inhibition of GSK-3β. Changes in pro-inflammatory markers, inducible nitric oxide synthase (iNOS) (increased) and interleukin-6 (IL-6) (decreased) demonstrated that iNOS regulation occurred at the translational level. Intraperitoneal (ip) injection of 6-MITC to the colitis-induced mice ameliorated weight loss whereas oral administration had negligible effect. Fecal blood and colon weight/length ratio parameters improved on treatment with 6-MITC and the other NF-κB inhibitors. Levels of NF-κB decreased upon addition of 6-MITC in vitro while structural studies showed 6-MITC acts competitively to inhibit GSK-3β at the ATP binding site. In this study we demonstrated that 6-MITC inhibits NF-κB signaling via GSK-3β inhibition ameliorating fecal blood, colonic alterations and DSS-induced weight loss indirectly indicating reduced intestinal stress. Taken together these results suggest a role for 6-MITC in the treatment of IBD acting to alleviate inflammation through the GSK-3β/NF-κB pathway. Furthermore, the GSK-3β-6-MITC model can be utilized as a basis for development of novel therapeutics targeting GSK-3β for use in other disorders including cancer.
Collapse
Affiliation(s)
- Anna Lohning
- Faculty of Health Sciences & Medicine, Bond University, Gold Coast, Australia.
| | - Yumi Kidachi
- Department of Pharmacy, Aomori University, 2-3-1 Kobata, Aomori, 030-0943, Japan
| | - Katsuyoshi Kamiie
- Department of Pharmacy, Aomori University, 2-3-1 Kobata, Aomori, 030-0943, Japan
| | - Kazuo Sasaki
- Department of Food and Life Sciences, Toyo University, 1-1-1 Izumino, Itakura, Gunma, 374-0193, Japan
| | - Kazuo Ryoyama
- Department of Pharmacy, Aomori University, 2-3-1 Kobata, Aomori, 030-0943, Japan
| | - Hideaki Yamaguchi
- Department of Applied Biological Chemistry, Meijo University, 1-501 Shiogamaguchi, Tempaku, Nagoya, 468-8502, Japan
| |
Collapse
|
12
|
Ashry ESHE, Elshatanofy MM, Badawy MEI, Kandeel KM, Elhady OM, Abdel-Sayed MA. Synthesis and Evaluation of Antioxidant, Antibacterial, and Target Protein-Molecular Docking of Novel 5-Phenyl-2,4-dihydro-3H-1,2,4-triazole Derivatives Hybridized with 1,2,3-Triazole via the Flexible SCH2-Bonding. RUSS J GEN CHEM+ 2021. [DOI: 10.1134/s1070363220120300] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
13
|
Nandy S, Dey A. Bibenzyls and bisbybenzyls of bryophytic origin as promising source of novel therapeutics: pharmacology, synthesis and structure-activity. Daru 2020; 28:701-734. [PMID: 32803687 PMCID: PMC7429097 DOI: 10.1007/s40199-020-00341-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 03/30/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The amphibian, non-vascular, gametophyte-dominant, bio-indicator class, bryophytes; with their wide ranges of habitat have attained importance due to their promising medicinal attributions and therapeutic role; mostly aided by presence of aromatic bibenzyl and bisbybenzyl class of compounds. Bibenzyls are steroidal ethane derivatives, resembling the structural moiety of bioactive dihydro-stilbenoids or iso-quinoline alkaloids. These stress triggered secondary metabolites are the by-products of the flavonoid biosynthetic pathway. Different classes of bryophytes (Bryophyta, Marchantiophyta and Anthocerotophyta) possess different subtypes of bibenzyls and dimeric bisbibenzyls. Among the liverwort, hornwort and mosses, former one is mostly enriched with bibenzyl type constituents as per the extensive study conducted for phytochemical deposit. Considering macrocyclic and acyclic group of bibenzyls and bisbybenzyls, generally marchantin type compounds are reported vividly for significant biological activity that includes neuro-nephro-cardio-protection besides anti-allergic, anti-microbial, anti-apoptotic and cytotoxic activities studied on in-vitro and in-vivo models or on cell lines. RESULT The critical analysis of reported chemical and pharmaceutical attributions of bibenzyls and bis-bibenzyls yielded detailed report on this compound class along with their application, mode of action, natural source, techniques of synthesis, extraction procedure, isolation and characterization. Further, the structure activity relationship studies and bioactivity of bibenzyls derived from non-bryophytic origin were also summarized. CONCLUSION This review encompasses prospective biological application of botanical reservoir of this primarily ignored, primeval land plant group where recent technical advances has paved the way for qualitative and quantitative isolation and estimation of novel compounds as well as marker components to study their impact on environment, as bio-control agents and as key leads in future drug designing. Graphical abstract.
Collapse
Affiliation(s)
- Samapika Nandy
- Research Scholar, Department of Life Sciences, Presidency University, Kolkata, 700073 India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, 700073 India
| |
Collapse
|
14
|
Hirano M, Toyota K, Ishibashi H, Tominaga N, Sato T, Tatarazako N, Iguchi T. Molecular Insights into Structural and Ligand Binding Features of Methoprene-Tolerant in Daphnids. Chem Res Toxicol 2020; 33:2785-2792. [PMID: 33089992 DOI: 10.1021/acs.chemrestox.0c00179] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Juvenile hormone (JH) is an important endocrine factor regulating many biological activities in arthropods. In daphnids, methoprene-tolerant (Met) belongs to a basic helix-loop-helix/Per-Arnt-Sim (bHLH/PAS) family protein which has recently been confirmed as a JH receptor and can bind and be activated by JHs and JH agonists. Although the activation of the JH signaling pathway causes many physiological effects, the molecular basis for the structural feature and ligand binding properties of Daphnia Met are not fully understood. To study the ligand preference in terms of structural features of Daphnia Met, we built in silico homology models of the PAS-B domain of Daphnia Mets from cladoceran crustaceans, Daphnia pulex and D. magna. Structural comparison of two Daphnia Met PAS-B domain models revealed that the volume in the main cavity of D. magna Met was larger than that of D. pulex Met. Compared with insect Met, Daphnia Met had a less hydrophobic cavity due to polar residues in the core-binding site. Molecular docking simulations of JH and its analogs with Daphnia Met indicated that the interaction energies were correlated with each of the experimental values of in vivo JH activities based on male induction and in vitro Met-mediated transactivation potencies. Furthermore, in silico site-directed mutagenesis supported experimental findings that Thr292 in D. pulex Met and Thr296 in D. magna Met substitution to valine contribute to JH selectivity and differential species response. This study demonstrates that in silico simulations of Daphnia Met and its ligands may be a tool for predicting the ligand profile and cross species sensitivity.
Collapse
Affiliation(s)
- Masashi Hirano
- Department of Biological and Chemical Systems Engineering, National Institute of Technology, Kumamoto College, 2627 Hirayama-shinmachi, Yatsushiro, Kumamoto 866-8501, Japan
| | - Kenji Toyota
- Department of Biological Science, Faculty of Science, Kanagawa University, 2946 Tsuchiya, Hiratsuka, Kanagawa 259-1293, Japan
| | - Hiroshi Ishibashi
- Graduate School of Agriculture, Ehime University, 3-5-7 Tarumi, Matsuyama 790-8566, Japan
| | - Nobuaki Tominaga
- Department of Creative Engineering, National Institute of Technology, Ariake College, 150 Higashi-Hagio, Omuta, Fukuoka 836-8585, Japan
| | - Tomomi Sato
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| | - Norihisa Tatarazako
- Graduate School of Agriculture, Ehime University, 3-5-7 Tarumi, Matsuyama 790-8566, Japan
| | - Taisen Iguchi
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| |
Collapse
|
15
|
Fukushima Y, Kojima A, Mi X, Ding WG, Kitagawa H, Matsuura H. Open-channel blocking action of volatile anaesthetics desflurane and sevoflurane on human voltage-gated K v 1.5 channel. Br J Pharmacol 2020; 177:3811-3827. [PMID: 32436224 DOI: 10.1111/bph.15105] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 04/28/2020] [Accepted: 05/01/2020] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Volatile anaesthetics have been shown to differentially modulate mammalian Shaker-related voltage-gated potassium (Kv 1.x) channels. This study was designed to investigate molecular and cellular mechanisms underlying the modulatory effects of desflurane or sevoflurane on human Kv 1.5 (hKv 1.5) channels. EXPERIMENTAL APPROACH Thirteen single-point mutations were constructed within pore domain of hKv 1.5 channel using site-directed mutagenesis. The effects of desflurane or sevoflurane on heterologously expressed wild-type and mutant hKv 1.5 channels were examined by whole-cell patch-clamp technique. A computer simulation was conducted to predict the docking pose of desflurane or sevoflurane within hKv 1.5 channel. KEY RESULTS Both desflurane and sevoflurane increased hKv 1.5 current at mild depolarizations but decreased it at strong depolarizations, indicating that these anaesthetics produce both stimulatory and inhibitory actions on hKv 1.5 channels. The inhibitory effect of desflurane or sevoflurane on hKv 1.5 channels arose primarily from its open-channel blocking action. The inhibitory action of desflurane or sevoflurane on hKv 1.5 channels was significantly attenuated in T480A, V505A, and I508A mutant channels, compared with wild-type channel. Computational docking simulation predicted that desflurane or sevoflurane resides within the inner cavity of channel pore and has contact with Thr479, Thr480, Val505, and Ile508. CONCLUSION AND IMPLICATIONS Desflurane and sevoflurane exert an open-channel blocking action on hKv 1.5 channels by functionally interacting with specific amino acids located within the channel pore. This study thus identifies a novel molecular basis mediating inhibitory modulation of hKv 1.5 channels by desflurane and sevoflurane.
Collapse
Affiliation(s)
- Yutaka Fukushima
- Department of Physiology, Shiga University of Medical Science, Otsu, Japan.,Department of Anesthesiology, Shiga University of Medical Science, Otsu, Japan
| | - Akiko Kojima
- Department of Anesthesiology, Shiga University of Medical Science, Otsu, Japan
| | - Xinya Mi
- Department of Physiology, Shiga University of Medical Science, Otsu, Japan
| | - Wei-Guang Ding
- Department of Physiology, Shiga University of Medical Science, Otsu, Japan
| | - Hirotoshi Kitagawa
- Department of Anesthesiology, Shiga University of Medical Science, Otsu, Japan
| | - Hiroshi Matsuura
- Department of Physiology, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
16
|
Alexander JAN, Radaeva M, King DT, Chambers HF, Cherkasov A, Chatterjee SS, Strynadka NCJ. Structural analysis of avibactam-mediated activation of the bla and mec divergons in methicillin-resistant Staphylococcus aureus. J Biol Chem 2020; 295:10870-10884. [PMID: 32518158 DOI: 10.1074/jbc.ra120.013029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 06/02/2020] [Indexed: 02/01/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) infections cause significant mortality and morbidity globally. MRSA resistance to β-lactam antibiotics is mediated by two divergons that control levels of a β-lactamase, PC1, and a penicillin-binding protein poorly acylated by β-lactam antibiotics, PBP2a. Expression of genes encoding these proteins is controlled by two integral membrane proteins, BlaR1 and MecR1, which both have an extracellular β-lactam-binding sensor domain. Here, we solved the X-ray crystallographic structures of the BlaR1 and MecR1 sensor domains in complex with avibactam, a diazabicyclooctane β-lactamase inhibitor at 1.6-2.0 Å resolution. Additionally, we show that S. aureus SF8300, a clinically relevant strain from the USA300 clone of MRSA, responds to avibactam by up-regulating the expression of the blaZ and pbp2a antibiotic-resistance genes, encoding PC1 and PBP2a, respectively. The BlaR1-avibactam structure of the carbamoyl-enzyme intermediate revealed that avibactam is bound to the active-site serine in two orientations ∼180° to each other. Although a physiological role of the observed alternative pose remains to be validated, our structural results hint at the presence of a secondary sulfate-binding pocket that could be exploited in the design of future inhibitors of BlaR1/MecR1 sensor domains or the structurally similar class D β-lactamases. The MecR1-avibactam structure adopted a singular avibactam orientation similar to one of the two states observed in the BlaR1-avibactam structure. Given avibactam up-regulates expression of blaZ and pbp2a antibiotic resistance genes, we suggest further consideration and research is needed to explore what effects administering β-lactam-avibactam combinations have on treating MRSA infections.
Collapse
Affiliation(s)
- J Andrew N Alexander
- Department of Biochemistry and Molecular Biology and Centre for Blood Research, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Mariia Radaeva
- Vancouver Prostate Centre, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Dustin T King
- Department of Biochemistry and Molecular Biology and Centre for Blood Research, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Henry F Chambers
- Division of Infectious Disease, Dept. of Medicine, San Francisco General Hospital, San Francisco, California, USA
| | - Artem Cherkasov
- Vancouver Prostate Centre, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Som S Chatterjee
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland and Institute of Marine and Environmental Technology, Baltimore, Maryland, USA ;
| | - Natalie C J Strynadka
- Department of Biochemistry and Molecular Biology and Centre for Blood Research, The University of British Columbia, Vancouver, British Columbia, Canada ;
| |
Collapse
|
17
|
Ye WL, Shen C, Xiong GL, Ding JJ, Lu AP, Hou TJ, Cao DS. Improving Docking-Based Virtual Screening Ability by Integrating Multiple Energy Auxiliary Terms from Molecular Docking Scoring. J Chem Inf Model 2020; 60:4216-4230. [PMID: 32352294 DOI: 10.1021/acs.jcim.9b00977] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Virtual Screening (VS) based on molecular docking is an efficient method used for retrieving novel hit compounds in drug discovery. However, the accuracy of the current docking scoring function (SF) is usually insufficient. In this study, in order to improve the screening power of SF, a novel approach named EAT-Score was proposed by directly utilizing the energy auxiliary terms (EAT) provided by molecular docking scoring through eXtreme Gradient Boosting (XGBoost). Here, EAT specifically refers to the output of the Molecular Operating Environment (MOE) scoring, including the energy scores of five different classical SFs and the Protein-Ligand Interaction Fingerprint (PLIF) terms. The performance of EAT-Score to discriminate actives from decoys was strictly validated on the DUD-E diverse subset by using different performance metrics. The results showed that EAT-Score performed much better than classical SFs in VS, with its AUC values exhibiting an improvement of around 0.3. Meanwhile, EAT-Score could achieve comparable even better prediction performance compared with other state-of-the-art VS methods, such as some machine learning (ML)-based SFs and classical SFs implemented in docking programs, in terms of AUC, LogAUC, or BEDROC. Furthermore, the EAT-Score model can capture important binding pattern information from protein-ligand complexes by Shapley additive explanations (SHAP) analysis, which may be very helpful in interpreting the ligand binding mechanism for a certain target and thereby guiding drug design.
Collapse
Affiliation(s)
- Wen-Ling Ye
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410003, P. R. China
| | - Chao Shen
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Guo-Li Xiong
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410003, P. R. China
| | - Jun-Jie Ding
- Beijing Institute of Pharmaceutical Chemistry, Beijing 102205, P. R. China
| | - Ai-Ping Lu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, P. R. China
| | - Ting-Jun Hou
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Dong-Sheng Cao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410003, P. R. China.,Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, P. R. China
| |
Collapse
|
18
|
Hwang JH, Kannan K, Evans TJ, Iwata H, Kim EY. Assessment of Risks of Dioxins for Aryl Hydrocarbon Receptor-Mediated Effects in Polar Bear ( Ursus maritimus) by in Vitro and in Silico Approaches. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:1770-1781. [PMID: 31841312 DOI: 10.1021/acs.est.9b05941] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Polar bear (Ursus maritimus) populations accumulate dioxins and related compounds (DRCs) at levels that are of health concern. The toxicities of DRCs are primarily mediated via aryl hydrocarbon receptor (AHR) signaling pathway. To evaluate the sensitivity and responses to DRCs in polar bears, we assessed the activation potencies of polar bear-specific AHR (pbAHR) by DRCs through in vitro and in silico approaches. In vitro assays showed that the pbAHR was as sensitive to DRCs as C3H/lpr mouse AHR, which is well-known to be highly sensitive to DRCs. Comparison of pbAHR transactivation potencies indicated that TCDF, 2,3,4,7,8-PeCDF, and BaP exhibited high induction equivalency factors (IEFs). Considering the accumulation levels of DRCs in polar bears, PCB126 was found to be the most active inducer of pbAHR. The in vitro transactivation potencies of ligands of pbAHR showed a significant relationship with in silico ligand docking energies in a pbAHR homology model. The protein ligand interaction fingerprint (PLIF) analysis showed different interaction patterns depending on the ligands. Several amino acids which are highly conserved among mammals may be involved in species-specific responses via backbone interactions with neighboring amino acid residues which are specific to pbAHR. We document high susceptibility of polar bears to DRCs, through a mechanistic approach, for the first time.
Collapse
Affiliation(s)
- Ji-Hee Hwang
- Department of Life and Nanopharmaceutical Science and Department of Biology , Kyung Hee University , Seoul 130-701 , Korea
| | - Kurunthachalam Kannan
- Wadsworth Center , New York State Department of Health, Empire State Plaza , P.O. Box 509, Albany , New York 12201-0509 , United States
| | - Thomas J Evans
- United States Fish and Wildlife Service , Office of Subsistence Management , Anchorage , Alaska 99503 , United States
| | - Hisato Iwata
- Center for Marine Environmental Studies (CMES) , Ehime University , Matsuyama 790-8577 , Japan
| | - Eun-Young Kim
- Department of Life and Nanopharmaceutical Science and Department of Biology , Kyung Hee University , Seoul 130-701 , Korea
| |
Collapse
|
19
|
Nakamura R, Ozeki T, Hirayama N, Sekine A, Yamashita T, Mashimo Y, Mizukawa Y, Shiohara T, Watanabe H, Sueki H, Ogawa K, Asada H, Kaniwa N, Tsukagoshi E, Matsunaga K, Niihara H, Yamaguchi Y, Aihara M, Mushiroda T, Saito Y, Morita E. Association of HLA-A*11:01 with Sulfonamide-Related Severe Cutaneous Adverse Reactions in Japanese Patients. J Invest Dermatol 2020; 140:1659-1662.e6. [PMID: 31981579 DOI: 10.1016/j.jid.2019.12.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 12/03/2019] [Accepted: 12/28/2019] [Indexed: 02/02/2023]
Affiliation(s)
- Ryosuke Nakamura
- Division of Medicinal Safety Science, National Institute of Health Sciences, Kawasaki, Japan
| | - Takeshi Ozeki
- Laboratory for Pharmacogenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Noriaki Hirayama
- Institute of Advanced Biosciences, Tokai University, Kanagawa, Japan
| | - Akihiro Sekine
- Center for Preventive Medical Science, Chiba University, Chiba, Japan
| | - Taiki Yamashita
- Center for Preventive Medical Science, Chiba University, Chiba, Japan
| | - Yoichi Mashimo
- Center for Preventive Medical Science, Chiba University, Chiba, Japan
| | - Yoshiko Mizukawa
- Department of Dermatology, Kyorin University School of Medicine, Tokyo, Japan
| | - Tetsuo Shiohara
- Department of Dermatology, Kyorin University School of Medicine, Tokyo, Japan
| | - Hideaki Watanabe
- Department of Dermatology, Showa University School of Medicine, Tokyo, Japan
| | - Hirohiko Sueki
- Department of Dermatology, Showa University School of Medicine, Tokyo, Japan
| | - Kohei Ogawa
- Department of Dermatology, Nara Medical University, Nara, Japan
| | - Hideo Asada
- Department of Dermatology, Nara Medical University, Nara, Japan
| | - Nahoko Kaniwa
- Division of Medicinal Safety Science, National Institute of Health Sciences, Kawasaki, Japan
| | - Eri Tsukagoshi
- Division of Medicinal Safety Science, National Institute of Health Sciences, Kawasaki, Japan
| | - Kayoko Matsunaga
- Department of Integrative Medical Science for Allergic Disease, Fujita Health University School of Medicine, Aichi, Japan
| | - Hiroyuki Niihara
- Department of Dermatology, Shimane University Faculty of Medicine, Shimane, Japan
| | - Yukie Yamaguchi
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Michiko Aihara
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Taisei Mushiroda
- Laboratory for Pharmacogenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yoshiro Saito
- Division of Medicinal Safety Science, National Institute of Health Sciences, Kawasaki, Japan.
| | - Eishin Morita
- Department of Dermatology, Shimane University Faculty of Medicine, Shimane, Japan
| |
Collapse
|
20
|
Bak SM, Nakata H, Koh DH, Yoo J, Iwata H, Kim EY. In vitro and in silico AHR assays for assessing the risk of heavy oil-derived polycyclic aromatic hydrocarbons in fish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 181:214-223. [PMID: 31195230 DOI: 10.1016/j.ecoenv.2019.06.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/31/2019] [Accepted: 06/02/2019] [Indexed: 06/09/2023]
Abstract
In the aftermath of the Great East Japan Earthquake of March 11, 2011, marine fish in Kesennuma Bay, Japan, have been contaminated with heavy oil containing polycyclic aromatic hydrocarbons (PAHs). To estimate the risk of six PAHs (benzo[α]pyrene, dibenzothiophene, phenanthrene, 2,3,5-trimethylnaphthalene, acenaphthene, and 1-methylphenanthrene), which have been detected at high levels in the tissues of fish from Kesennuma Bay, we attempted to evaluate the effects of these PAHs on the fish aryl hydrocarbon receptor (AHR) signaling pathway. We initially measured PAH concentrations and cytochrome P4501A catalytic activities (EROD: ethoxyresorufin-O-deethylase and MROD: methoxyresorufin-O-demethylase) as markers of AHR activation in greenlings (Hexagrammos otakii) collected from Kesennuma Bay in 2014. The results showed that alkylated PAH concentrations and EROD/MROD activities were higher in sites close to the oil-spilled sites than in the control site, suggesting AHR activation by spilled alkylated PAHs. We then investigated AHR-mediated responses to these PAHs in the in vitro reporter gene assay system where red seabream (Pagrus major) AHR1 (rsAHR1) or rsAHR2 expression plasmids were transiently transfected into COS-7 cells. The in vitro assay showed rsAHR isoform-, PAH-, and dose-dependent transactivation potencies. The relative effective concentrations of benzo[α]pyrene, dibenzothiophene, phenanthrene, 2,3,5-trimethylnaphthalene, acenaphthene, and 1-methylphenanthrene that induce 20% of the maximum benzo[α]pyrene response (REC20-BaP) for rsAHR1 activation were 0.052, 38, 79, 88, 270 nM, and no response, respectively, and those for rsAHR2 activation were 0.0049, 32, 53, 88, 60 nM, and no response, respectively. The results showed that the REC20-BaP values of benzo[α]pyrene for both the rsAHR1 and rsAHR2 isoforms were lower than the concentrations (0.041-0.20 nM) detected in the muscle tissue of fish from Kesennuma Bay, while the REC20-BaP values of other PAHs were higher than their tissue concentrations. In silico rsAHR homology modeling and subsequent ligand docking simulation analyses indicated that the rsAHR activation potencies of PAHs could be predicted from a rsAHR2 model. This study shows that in vitro and in silico rsAHR analyses may be a useful tool for assessing the risks to fish contaminated with PAHs.
Collapse
Affiliation(s)
- Su-Min Bak
- Laboratory of Environmental Toxicology, Center for Marine Environmental Studies, Ehime University, Bunkyo-cho 2-5, Matsuyama, 790-8577, Japan.
| | - Haruhiko Nakata
- Faculty of the Advanced Graduate School of Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto, 860-8555, Japan.
| | - Dong-Hee Koh
- Department of Life and Nanopharmaceutical Science, Kyung Hee University,26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| | - Jean Yoo
- Laboratory of Environmental Toxicology, Center for Marine Environmental Studies, Ehime University, Bunkyo-cho 2-5, Matsuyama, 790-8577, Japan.
| | - Hisato Iwata
- Laboratory of Environmental Toxicology, Center for Marine Environmental Studies, Ehime University, Bunkyo-cho 2-5, Matsuyama, 790-8577, Japan.
| | - Eun-Young Kim
- Department of Life and Nanopharmaceutical Science, Kyung Hee University,26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea; Department of Biology, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|
21
|
Malkhasian AY, Howlin BJ. Automated drug design of kinase inhibitors to treat Chronic Myeloid Leukemia. J Mol Graph Model 2019; 91:52-60. [DOI: 10.1016/j.jmgm.2019.05.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/17/2019] [Accepted: 05/17/2019] [Indexed: 11/25/2022]
|
22
|
Toyama M, Sakakibara N, Takeda M, Okamoto M, Watashi K, Wakita T, Sugiyama M, Mizokami M, Ikeda M, Baba M. Pyrimidotriazine derivatives as selective inhibitors of HBV capsid assembly. Virus Res 2019; 271:197677. [PMID: 31376401 DOI: 10.1016/j.virusres.2019.197677] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 07/31/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023]
Abstract
Chronic hepatitis B virus (HBV) infection is currently treated with nucleoside/nucleotides analogs. They are potent inhibitors of HBV DNA polymerase, which also functions as reverse transcriptase. Although nucleoside/nucleotide analogs efficiently suppress HBV replication in liver cells, they cannot eradicate HBV DNA from liver cells and cure the disease. Therefore, it is still mandatory to identify and develop effective inhibitors that target a step other than reverse transcription in the viral replication cycle. HBV capsid assembly is a critical step for viral replication and an attractive target for inhibition of HBV replication. We conducted in silico screening of compounds expected to bind to the HBV capsid dimer-dimer interaction site. The selected compounds were further examined for their anti-HBV activity in vitro. Among the test compounds, novel pyrimidotriazine derivatives were found to be selective inhibitors of HBV replication in HepG2.2.15.7 cells. Among the compounds, 2-[(2,3-dichlorophenyl)amino]-4-(4-tert-butylphenyl)-8-methyl-4H,9H-pyrimido[1,2-a][1,3,5]triazin-6-one was the most active against HBV replication. Studies on its mechanism of action revealed that the compound interfered with HBV capsid assembly determined by a cell-free capsid assembly system. Thus, the pyrimidotriazine derivatives are considered to be potential leads for novel HBV capsid assembly inhibitors.
Collapse
Affiliation(s)
- Masaaki Toyama
- Division of Antiviral Chemotherapy, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, Japan
| | - Norikazu Sakakibara
- Faculty of Pharmaceutical Sciences at Kagawa Campus, Tokushima Bunri University, Sanuki, Japan
| | - Midori Takeda
- Division of Biological Information Technology, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, Japan
| | - Mika Okamoto
- Division of Antiviral Chemotherapy, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, Japan
| | - Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takaji Wakita
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masaya Sugiyama
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Masashi Mizokami
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Masanori Ikeda
- Division of Biological Information Technology, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, Japan
| | - Masanori Baba
- Division of Antiviral Chemotherapy, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, Japan.
| |
Collapse
|
23
|
Balbaa M, Awad D, Elaal AA, Mahsoub S, Moharram M, Sadek O, Rezki N, Aouad MR, Badawy METI, El Ashry ESH. Action of Thioglycosides of 1,2,4-Triazoles and Imidazoles on the Oxidative Stress and Glycosidases in Mice with Molecular Docking. LETT DRUG DES DISCOV 2019. [DOI: 10.2174/1573413715666181212150955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background:
,2,3-Triazoles and imidazoles are important five-membered heterocyclic
scaffolds due to their extensive biological activities. These products have been an area of growing
interest to many researchers around the world because of their enormous pharmaceutical scope.
Methods:
The in vivo and in vitro enzyme inhibition of some thioglycosides encompassing 1,2,4-
triazole N1, N2, and N3 and/or imidazole moieties N4, N5, and N6. The effect on the antioxidant
enzymes (superoxide dismutase, glutathione S-transferase, glutathione peroxidase and catalase) was
investigated as well as their effect on α-glucosidase and β-glucuronidase. Molecular docking studies
were carried out to investigate the mode of the binding interaction of the compounds with α-
glucosidase and β -glucuronidase. In addition, quantitative structure-activity relationship (QSAR)
investigation was applied to find out the correlation between toxicity and physicochemical properties.
Results:
The decrease of the antioxidant status was revealed by the in vivo effect of the tested compounds.
Furthermore, the in vivo and in vitro inhibitory effects of the tested compounds were clearly
pronounced on α-glucosidase, but not β-glucuronidase. The IC50 and Ki values revealed that the thioglycoside
- based 1,2,4-triazole N3 possesses a high inhibitory action. In addition, the in vitro studies
demonstrated that the whole tested 1,2,4-triazole are potent inhibitors with a Ki magnitude of 10-6
and exhibited a competitive type inhibition. On the other hand, the thioglycosides - based imidazole
ring showed an antioxidant activity and exerted a slight in vivo stimulation of α-glucosidase and β-
glucuronidase. Molecular docking proved that the compounds exhibited binding affinity with the
active sites of α -glucosidase and β-glucuronidase (docking score ranged from -2.320 to -4.370
kcal/mol). Furthermore, QSAR study revealed that the HBD and RB were found to have an overall
significant correlation with the toxicity.
Conclusion:
These data suggest that the inhibition of α-glucosidase is accompanied by an oxidative
stress action.
Collapse
Affiliation(s)
- Mahmoud Balbaa
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Doaa Awad
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Ahmad Abd Elaal
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Shimaa Mahsoub
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Mayssaa Moharram
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Omayma Sadek
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Nadjet Rezki
- Department of Chemistry, Faculty of Science, Taibah University, Al-Madinah AlMunawarah 30002, Saudi Arabia
| | - Mohamed Reda Aouad
- Department of Chemistry, Faculty of Science, Taibah University, Al-Madinah AlMunawarah 30002, Saudi Arabia
| | - Mohamed El-Taher Ibrahim Badawy
- Department of Pesticide Chemistry and Technology, Faculty of Agriculture, 21545-El-Shatby, Alexandria University, Alexandria, Egypt
| | | |
Collapse
|
24
|
Abdelgaleil SAM, Badawy MEI, Mahmoud NF, Marei AESM. Acaricidal activity, biochemical effects and molecular docking of some monoterpenes against two-spotted spider mite (Tetranychus urticae Koch). PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2019; 156:105-115. [PMID: 31027569 DOI: 10.1016/j.pestbp.2019.02.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/09/2019] [Accepted: 02/10/2019] [Indexed: 05/15/2023]
Abstract
Six natural monoterpenes (1,8-cineole, (-)-citronellal, limonene, α-pinene, pulegone and 4-terpineol) showed high acaricidal activity by fumigant and contact actions against adult females of the two-spotted spider mite, Tetranychus urticae Koch. The monoterpenes exhibited varying degrees of acaricidal potency using contact toxicity test after 24 and 48 h of treatment, where the LC50 values were <160 and 45 mg/L, respectively. In fumigation test, of these six monoterpenes, pulegone exhibited the highest toxicity (LC50 = 3.81 mg/L air), while (-)-citronellal had the lowest fumigant toxicity (LC50 = 15.20 mg/L air). All compounds had high inhibitory effect on acetylcholinesterase (AChE) and gama amino butyric acid transaminase (GABA-T) activities. Pulegone was the most AChE inhibitor (IC50 = 8.79 mg/L), while 4-terpineol revealed the lowest inhibitory effect (IC50 = 32.82 mg/L). However, limonene caused the highest inhibition of GABA-T (IC50 = 11.37 mg/L). The molecular docking studies revealed that the compounds displayed different binding interactions with the amino acid residues at the catalytic sites of AChE and GABA-T enzymes. Noncovalent interactions especially van der Waals, hydrogen bonding as well as hydrophobic was found between the compounds and the enzymes. A significant relationship was found between the docking score and the biological activity of monoterpenes compared to the standard acaricide pyridaben. In silico ADMET properties were also performed and displayed potential for the development of good acaricidal candidates.
Collapse
Affiliation(s)
- Samir A M Abdelgaleil
- Department of Pesticide Chemistry and Technology, Faculty of Agriculture, Alexandria University, 21545 El-Shatby, Alexandria, Egypt
| | - Mohamed E I Badawy
- Department of Pesticide Chemistry and Technology, Faculty of Agriculture, Alexandria University, 21545 El-Shatby, Alexandria, Egypt.
| | - Nabila F Mahmoud
- Department of Pesticide Chemistry and Technology, Faculty of Agriculture, Alexandria University, 21545 El-Shatby, Alexandria, Egypt
| | - Abd El-Salam M Marei
- Department of Pesticide Chemistry and Technology, Faculty of Agriculture, Alexandria University, 21545 El-Shatby, Alexandria, Egypt
| |
Collapse
|
25
|
Okada K, Araki M, Sakashita T, Ma B, Kanada R, Yanagitani N, Horiike A, Koike S, Oh-Hara T, Watanabe K, Tamai K, Maemondo M, Nishio M, Ishikawa T, Okuno Y, Fujita N, Katayama R. Prediction of ALK mutations mediating ALK-TKIs resistance and drug re-purposing to overcome the resistance. EBioMedicine 2019; 41:105-119. [PMID: 30662002 PMCID: PMC6441848 DOI: 10.1016/j.ebiom.2019.01.019] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 01/08/2019] [Accepted: 01/08/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Alectinib has shown a greater efficacy to ALK-rearranged non-small-cell lung cancers in first-line setting; however, most patients relapse due to acquired resistance, such as secondary mutations in ALK including I1171N and G1202R. Although ceritinib or lorlatinib was shown to be effective to these resistant mutants, further resistance often emerges due to ALK-compound mutations in relapse patients following the use of ceritinib or lorlatinib. However, the drug for overcoming resistance has not been established yet. METHODS We established lorlatinib-resistant cells harboring ALK-I1171N or -G1202R compound mutations by performing ENU mutagenesis screening or using an in vivo mouse model. We performed drug screening to overcome the lorlatinib-resistant ALK-compound mutations. To evaluate these resistances in silico, we developed a modified computational molecular dynamic simulation (MP-CAFEE). FINDINGS We identified 14 lorlatinib-resistant ALK-compound mutants, including several mutants that were recently discovered in lorlatinib-resistant patients. Some of these compound mutants were found to be sensitive to early generation ALK-TKIs and several BCR-ABL inhibitors. Using our original computational simulation, we succeeded in demonstrating a clear linear correlation between binding free energy and in vitro experimental IC50 value of several ALK-TKIs to single- or compound-mutated EML4-ALK expressing Ba/F3 cells and in recapitulating the tendency of the binding affinity reduction by double mutations found in this study. Computational simulation revealed that ALK-L1256F single mutant conferred resistance to lorlatinib but increased the sensitivity to alectinib. INTERPRETATION We discovered lorlatinib-resistant multiple ALK-compound mutations and an L1256F single mutation as well as the potential therapeutic strategies for these ALK mutations. Our original computational simulation to calculate the binding affinity may be applicable for predicting resistant mutations and for overcoming drug resistance in silico. FUND: This work was mainly supported by MEXT/JSPS KAKENHI Grants and AMED Grants.
Collapse
Affiliation(s)
- Koutaroh Okada
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Mitsugu Araki
- RIKEN Advanced Institute for Computational Science, 7-1-26 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Graduate School of Medicine, Kyoto University, 53 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Takuya Sakashita
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Biao Ma
- Research and Development Group for In Silico Drug Discovery, Pro-Cluster Kobe, Foundation for Biomedical Research and Innovation (FBRI), 6-3-5, Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Ryo Kanada
- RIKEN Compass to Healthy Life Research Complex Program, 6-3-5, Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Noriko Yanagitani
- Department of Thoracic Medical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Atsushi Horiike
- Department of Thoracic Medical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Sumie Koike
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tomoko Oh-Hara
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kana Watanabe
- Department of Respiratory Medicine, Miyagi Cancer Center, Miyagi, Japan
| | - Keiichi Tamai
- Division of Cancer Stem Cell, Miyagi Cancer Center Research Institute, Miyagi, Japan
| | - Makoto Maemondo
- Department of Respiratory Medicine, Miyagi Cancer Center, Miyagi, Japan
| | - Makoto Nishio
- Department of Thoracic Medical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takeshi Ishikawa
- Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Yasushi Okuno
- RIKEN Advanced Institute for Computational Science, 7-1-26 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Graduate School of Medicine, Kyoto University, 53 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Naoya Fujita
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Ryohei Katayama
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan.
| |
Collapse
|
26
|
Isogai H, Hirayama N. <i><b>In silico</b></i><b> analysis of interactions of flucloxacillin and its metabolites with </b><i><b>HLA-B*57:01</b></i><i><b> </b></i>. CHEM-BIO INFORMATICS JOURNAL 2019. [DOI: 10.1273/cbij.19.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Hideto Isogai
- Department of Basic and Molecular Medicine, Tokai University School of Medicine
| | | |
Collapse
|
27
|
Chemical Composition and Antifungal In Vitro and In Silico, Antioxidant, and Anticholinesterase Activities of Extracts and Constituents of Ouratea fieldingiana (DC.) Baill. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:1748487. [PMID: 30524481 PMCID: PMC6247570 DOI: 10.1155/2018/1748487] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 08/02/2018] [Accepted: 08/12/2018] [Indexed: 12/25/2022]
Abstract
Ouratea fieldingiana (Gardner) Engl is popularly used for wound healing. This study describes the main chemical compounds present in extracts of O. fieldingiana and evaluates their biological potential by investigating antifungal, antioxidant, and anticholinesterase activities. The action mechanism of main antifungal compound was investigated by molecular docking using the enzyme sterol 14-α demethylase, CYP51, required for ergosterol biosynthesis. The seeds and leaves were extracted with ethanol in a Soxhlet apparatus and by maceration, respectively. Both extracts were subjected to silica gel column chromatography for isolation of main constituents, followed by purification in sephadex. The structures of compounds were established by 1H and 13C-NMR spectroscopy and identified by comparison with literature data as amentoflavone and kaempferol 3-O-rutinoside, respectively. The antioxidant activities of the extracts were determined by the DPPH and ABTS free radical inhibition methods. In general, the extracts with the highest antioxidant activity corresponded to those with higher content of phenolic compounds and flavonoids. The ethanol extracts and two isolated compounds presented relevant antifungal activity against several Candida strains. The in silico findings revealed that the compound amentoflavone coupled with the CYP450 protein due to the low energy stabilization (-9.39 kcal/mol), indicating a possible mechanism of action by inhibition of the ergosterol biosynthesis of Candida fungi.
Collapse
|
28
|
Araki M, Iwata H, Ma B, Fujita A, Terayama K, Sagae Y, Ono F, Tsuda K, Kamiya N, Okuno Y. Improving the Accuracy of Protein-Ligand Binding Mode Prediction Using a Molecular Dynamics-Based Pocket Generation Approach. J Comput Chem 2018; 39:2679-2689. [DOI: 10.1002/jcc.25715] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 09/19/2018] [Accepted: 09/25/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Mitsugu Araki
- Graduate School of Medicine; Kyoto University; 53 Shogoin-Kawaharacho, Sakyo-ku Kyoto 606-8507 Japan
- RIKEN Advanced Institute for Computational Sciences; 7-1-26 Minatojima-Minamimachi, Chuo-ku Kobe Hyogo 650-0047 Japan
| | - Hiroaki Iwata
- Graduate School of Medicine; Kyoto University; 53 Shogoin-Kawaharacho, Sakyo-ku Kyoto 606-8507 Japan
- Research and Development Group for In Silico Drug Discovery, Pro-Cluster Kobe; Foundation for Biomedical Research and Innovation (FBRI); 6-3-5, Minatojima-Minamimachi Chuo-ku Kobe Hyogo 650-0047 Japan
| | - Biao Ma
- Research and Development Group for In Silico Drug Discovery, Pro-Cluster Kobe; Foundation for Biomedical Research and Innovation (FBRI); 6-3-5, Minatojima-Minamimachi Chuo-ku Kobe Hyogo 650-0047 Japan
| | - Atsuto Fujita
- Research and Development Group for In Silico Drug Discovery, Pro-Cluster Kobe; Foundation for Biomedical Research and Innovation (FBRI); 6-3-5, Minatojima-Minamimachi Chuo-ku Kobe Hyogo 650-0047 Japan
| | - Kei Terayama
- Department of Computational Biology and Medical Sciences; Graduate School of Frontier Sciences, The University of Tokyo; Chiba 277-8561 Japan
| | - Yukari Sagae
- Graduate School of Medicine; Kyoto University; 53 Shogoin-Kawaharacho, Sakyo-ku Kyoto 606-8507 Japan
| | - Fumie Ono
- Graduate School of Medicine; Kyoto University; 53 Shogoin-Kawaharacho, Sakyo-ku Kyoto 606-8507 Japan
| | - Koji Tsuda
- Department of Computational Biology and Medical Sciences; Graduate School of Frontier Sciences, The University of Tokyo; Chiba 277-8561 Japan
| | - Narutoshi Kamiya
- Graduate School of Simulation Studies; University of Hyogo; 7-1-28 Minatojima-Minamimachi, Chuo-ku Kobe Hyogo 650-0047 Japan
| | - Yasushi Okuno
- Graduate School of Medicine; Kyoto University; 53 Shogoin-Kawaharacho, Sakyo-ku Kyoto 606-8507 Japan
- RIKEN Advanced Institute for Computational Sciences; 7-1-26 Minatojima-Minamimachi, Chuo-ku Kobe Hyogo 650-0047 Japan
| |
Collapse
|
29
|
Seto T, Kato M, Koyano K. <b>Molecular Discrimination of Barbital Enantiomer at the Propofol Binding Site of the Human β<sub>3</sub> Homomeric GABA<sub>A</sub> Receptor </b>. CHEM-BIO INFORMATICS JOURNAL 2018. [DOI: 10.1273/cbij.18.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Tomoyoshi Seto
- Department of Anesthesiology, School of Medicine, Shiga University of Medical Science
| | - Minoru Kato
- Graduate School of Life Sciences, Ritsumeikan University
| | - Ken Koyano
- Department of Anesthesiology, School of Medicine, Shiga University of Medical Science
| |
Collapse
|
30
|
Uno Y, Uehara S, Murayama N, Yamazaki H. Cytochrome P450 1A1, 2C9, 2C19, and 3A4 Polymorphisms Account for Interindividual Variability of Toxicological Drug Metabolism in Cynomolgus Macaques. Chem Res Toxicol 2018; 31:1373-1381. [PMID: 30412386 DOI: 10.1021/acs.chemrestox.8b00257] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cytochromes P450 (P450s) and their genetic variants in humans are important drug-metabolizing enzymes partly accounting for interindividual variations in drug metabolism and toxicity. However, these genetic variants in P450s have not been fully investigated in cynomolgus macaques, a nonhuman primate species widely used in toxicological studies. In this study, genetic variants found in cynomolgus CYP1A1, CYP2C9 (formerly CYP2C43), CYP2C19 (CYP2C75), and CYP3A4 (CYP3A8) were assessed on functional importance. Resequencing of CYP1A1 in cynomolgus macaques found 18 nonsynonymous variants, of which M121I and V382I were located in SRSs, domains potentially important for P450 function. By further analyzing these two variants, V382I was significantly associated with lower drug-metabolizing activities in the liver for the heterozygotes than the wild types. Similarly, the heterozygotes or homozygotes of CYP2C9 variants (A82V and H344R) and CYP2C19 variant (A490V) showed significantly lower drug-metabolizing activities in the liver than the wild types. Moreover, the homozygotes of CYP3A4 variant (S437N) showed significantly higher activities than the wild type in the liver. Kinetic analyses using recombinant proteins revealed that CYP2C9 variants (A82V and H344R) showed substantially lower Ks values than the wild type, although CYP1A1 variant (V382I) showed kinetic parameters similar to the wild type. Likewise, CYP2C19 variant (A490V) showed substantially a lower Vmax/ Km value than the wild type, whereas CYP3A4 variant (S437N) showed a higher Vmax/ Km value than the wild type. These results suggest the toxicologically functional importance of CYP2C9 variants (A82V and H344R), CYP2C19 variant (A490V), and CYP3A4 variant (S437N) for hepatic drug metabolism in cynomolgus macaques.
Collapse
Affiliation(s)
- Yasuhiro Uno
- Shin Nippon Biomedical Laboratories, Ltd., Kainan , Wakayama 642-0017 , Japan
| | - Shotaro Uehara
- Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , Machida , Tokyo 194-8543 , Japan
| | - Norie Murayama
- Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , Machida , Tokyo 194-8543 , Japan
| | - Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics , Showa Pharmaceutical University , Machida , Tokyo 194-8543 , Japan
| |
Collapse
|
31
|
Suyama K, Sakai D, Hirayama N, Nakamura Y, Matsushita E, Terayama H, Qu N, Tanaka O, Sakabe K, Watanabe M. Effects of interleukin-17A in nucleus pulposus cells and its small-molecule inhibitors for intervertebral disc disease. J Cell Mol Med 2018; 22:5539-5551. [PMID: 30207057 PMCID: PMC6201370 DOI: 10.1111/jcmm.13828] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/07/2018] [Indexed: 01/05/2023] Open
Abstract
Intervertebral discs (IVD) degeneration, which is caused by ageing or mechanical stress, leads to IVD disease, including back pain and sciatica. The cytokine interleukin (IL)-17A is elevated in NP cells during IVD disease. Here we explored the pharmacotherapeutic potential of IL-17A for the treatment of IVD disease using small-molecule inhibitors that block binding of IL-17A to the IL-17A receptor (IL-17RA). Treatment of NP cells with IL-17A increased expression of cyclooxygenase-2 (COX-2), IL-6, matrix metalloproteinase (MMP)-3 and MMP-13. These increases were suppressed by an IL-17A-neutralizing antibody, and small molecules that were identified as inhibitors by binding to the IL-17A-binding region of IL-17RA. IL-17A signalling also altered sulphated glycosaminoglycan deposition and spheroid colony formation, while treatment with small-molecule inhibitors of IL-17A attenuated this response. Furthermore, mitogen-activated protein kinase pathways were activated by IL-17A stimulation and induced IL-6 and COX-2 expression, while small-molecule inhibitors of IL-17A suppressed their expression. Taken together, these results show that IL-17A is a valid target for IVD disease therapy and that small-molecule inhibitors that inhibit the IL-17A-IL-17RA interaction may be useful for pharmacotherapy of IVD disease.
Collapse
Affiliation(s)
- Kaori Suyama
- Department of Anatomy and Cellular biology, Basic Medical Science, Tokai University School of Medicine, Kanagawa, Japan
| | - Daisuke Sakai
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Kanagawa, Japan
| | - Noriaki Hirayama
- Institute of Advanced Biosciences, Tokai University, Kanagawa, Japan
| | - Yoshihiko Nakamura
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Kanagawa, Japan
| | - Erika Matsushita
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Kanagawa, Japan
| | - Hayato Terayama
- Department of Anatomy and Cellular biology, Basic Medical Science, Tokai University School of Medicine, Kanagawa, Japan
| | - Ning Qu
- Department of Anatomy and Cellular biology, Basic Medical Science, Tokai University School of Medicine, Kanagawa, Japan
| | - Osamu Tanaka
- Department of Anatomy and Cellular biology, Basic Medical Science, Tokai University School of Medicine, Kanagawa, Japan
| | - Kou Sakabe
- Department of Anatomy and Cellular biology, Basic Medical Science, Tokai University School of Medicine, Kanagawa, Japan
| | - Masahiko Watanabe
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Kanagawa, Japan
| |
Collapse
|
32
|
Hori K, Ajioka K, Goda N, Shindo A, Takagishi M, Tenno T, Hiroaki H. Discovery of Potent Disheveled/Dvl Inhibitors Using Virtual Screening Optimized With NMR-Based Docking Performance Index. Front Pharmacol 2018; 9:983. [PMID: 30233369 PMCID: PMC6134994 DOI: 10.3389/fphar.2018.00983] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 08/10/2018] [Indexed: 12/13/2022] Open
Abstract
Most solid tumors have their own cancer stem cells (CSCs), which are resistant to standard chemo-therapies. Recent reports have described that Wnt pathway plays a key role in self-renewal and tumorigenesis of CSCs. Regarding the Wnt/β-catenin pathway, Dvl (mammalian Disheveled) is an attractive target of drug discovery. After analyzing the PDZ domain of human Dvl1 (Dvl1-PDZ) using NMR, we subjected it to preliminary NMR titration studies with 17 potential PDZ-binding molecules including CalBioChem-322338, a commercially available Dvl PDZ domain inhibitor. Next, we performed virtual screening (VS) using the program GOLD with nine parameter sets. Results were evaluated using the NMR-derived docking performance index (NMR-DPI). One parameter set of GOLD docking showing the best NMR-DPI was selected and used for the second VS against 5,135 compounds. The second docking trial identified more than 1,700 compounds that exhibited higher scores than CalBioChem-322338. Subsequent NMR titration experiments with five new candidate molecules (NPL-4001, 4004, 4011, 4012, and 4013), Dvl1-PDZ revealed larger chemical shift changes than those of CalBioChem-322338. Finally, these compounds showed partial proliferation inhibition activity against BT-20, a triple negative breast cancer (TNBC) cell. These compounds are promising Wnt pathway inhibitors that are potentially useful for anti-TNBC therapy.
Collapse
Affiliation(s)
- Kiminori Hori
- Laboratory of Structural Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Kasumi Ajioka
- Department of Biological Science, School of Science, Nagoya University, Nagoya, Japan
| | - Natsuko Goda
- Laboratory of Structural Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Asako Shindo
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Maki Takagishi
- Department of Pathology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Takeshi Tenno
- Laboratory of Structural Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan.,BeCellBar LLC, Business Incubation Center, Nagoya University, Nagoya, Japan
| | - Hidekazu Hiroaki
- Laboratory of Structural Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan.,Department of Biological Science, School of Science, Nagoya University, Nagoya, Japan.,BeCellBar LLC, Business Incubation Center, Nagoya University, Nagoya, Japan
| |
Collapse
|
33
|
Interaction of Nevirapine with the Peptide Binding Groove of HLA-DRB1*01:01 and Its Effect on the Conformation of HLA-Peptide Complex. Int J Mol Sci 2018; 19:ijms19061660. [PMID: 29867033 PMCID: PMC6032195 DOI: 10.3390/ijms19061660] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 01/11/2023] Open
Abstract
Human leukocyte antigen (HLA)-DRB1*01:01 has been shown to be involved in nevirapine-induced hepatic hypersensitivity reactions. In the present study, in silico docking simulations and molecular dynamics simulations were performed to predict the interaction mode of nevirapine with the peptide binding groove of HLA-DRB1*01:01 and its possible effect on the position and orientation of the ligand peptide derived from hemagglutinin (HA). In silico analyses suggested that nevirapine interacts with HLA-DRB1*01:01 around the P4 pocket within the peptide binding groove and the HA peptide stably binds on top of nevirapine at the groove. The analyses also showed that binding of nevirapine at the groove will significantly change the inter-helical distances of the groove. An in vitro competitive assay showed that nevirapine (1000 μM) increases the binding of the HA peptide to HLA-DRB1*01:01 in an allele-specific manner. These results indicate that nevirapine might interact directly with the P4 pocket and modifies its structure, which could change the orientation of loaded peptides and the conformation of HLA-DRB1*01:01; these changes could be distinctively recognized by T-cell receptors. Through this molecular mechanism, nevirapine might stimulate the immune system, resulting in hepatic hypersensitivity reactions.
Collapse
|
34
|
Hirakawa S, Miyawaki T, Hori T, Kajiwara J, Katsuki S, Hirano M, Yoshinouchi Y, Iwata H, Mitoma C, Furue M. Accumulation properties of polychlorinated biphenyl congeners in Yusho patients and prediction of their cytochrome P450-dependent metabolism by in silico analysis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:16455-16463. [PMID: 28639016 PMCID: PMC6301142 DOI: 10.1007/s11356-017-9498-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 06/08/2017] [Indexed: 06/10/2023]
Abstract
In what has become known as the Yusho incident, thousands of people in western Japan were poisoned by the accidental ingestion of rice bran oil contaminated with polychlorinated biphenyls (PCBs) and various dioxins and dioxin-like compounds. In this study, we investigated the accumulation patterns of 69 PCB congeners in the blood of Yusho patients in comparison with those of non-exposed controls. The blood samples were collected at medical check-ups in 2004 and 2005. To compare the patterns of PCB congeners, we calculated the concentration ratio of each congener relative to the 2,2',4,4',5,5'-hexaCB (CB153) concentration. The concentration ratios of tetra- and penta-chlorinated congeners in the blood of Yusho patients were significantly lower than those of controls. To examine the cytochrome P450 (CYP)-dependent metabolic potential of the 2,3',4,4'5-pentaCB (CB118), CB153, and 2,3,3',4,4'5-hexaCB (CB156) congeners, we conducted PCB-CYP (CYP1A1, CYP1A2, CYP2A6, and CYP2B6) docking simulation by in silico analysis. The docking models showed that human CYP1A1, CYP2A6, and CYP2B6 isozymes have the potential to metabolize CB118 and CB153. On the other hand, it was inferred that CB156 is difficult to be metabolized by these four CYP isozymes. These results indicate that CYP1 and CYP2 isozymes may be involved in the characteristic accumulation patterns of PCB congeners in the blood of Yusho patients.
Collapse
Affiliation(s)
- Shusaku Hirakawa
- Fukuoka Institute of Health and Environmental Sciences, 39 Mukaizano, Dazaifu, Fukuoka, 818-0135, Japan.
| | - Takashi Miyawaki
- Fukuoka Institute of Health and Environmental Sciences, 39 Mukaizano, Dazaifu, Fukuoka, 818-0135, Japan
| | - Tsuguhide Hori
- Fukuoka Institute of Health and Environmental Sciences, 39 Mukaizano, Dazaifu, Fukuoka, 818-0135, Japan
| | - Jumboku Kajiwara
- Fukuoka Institute of Health and Environmental Sciences, 39 Mukaizano, Dazaifu, Fukuoka, 818-0135, Japan
| | - Susumu Katsuki
- Fukuoka Institute of Health and Environmental Sciences, 39 Mukaizano, Dazaifu, Fukuoka, 818-0135, Japan
| | - Masashi Hirano
- Kumamoto College, National Institute of Technology, 2627 Hirayamashin-Machi, Yatsushiro, Kumamoto, 866-8501, Japan
| | - Yuka Yoshinouchi
- Center for Marine Environmental Studies (CMES), Ehime University, Bunkyo-cho 2-5, Matsuyama, 790-8577, Japan
| | - Hisato Iwata
- Center for Marine Environmental Studies (CMES), Ehime University, Bunkyo-cho 2-5, Matsuyama, 790-8577, Japan
| | - Chikage Mitoma
- Research and Clinical Center for Yusho and Dioxin, Kyusyu University Hospital, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masutaka Furue
- Research and Clinical Center for Yusho and Dioxin, Kyusyu University Hospital, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
35
|
A secondary RET mutation in the activation loop conferring resistance to vandetanib. Nat Commun 2018; 9:625. [PMID: 29434222 PMCID: PMC5809600 DOI: 10.1038/s41467-018-02994-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 01/08/2018] [Indexed: 01/09/2023] Open
Abstract
Resistance to vandetanib, a type I RET kinase inhibitor, developed in a patient with metastatic lung adenocarcinoma harboring a CCDC6-RET fusion that initially exhibited a response to treatment. The resistant tumor acquired a secondary mutation resulting in a serine-to-phenylalanine substitution at codon 904 in the activation loop of the RET kinase domain. The S904F mutation confers resistance to vandetanib by increasing the ATP affinity and autophosphorylation activity of RET kinase. A reduced interaction with the drug is also observed in vitro for the S904F mutant by thermal shift assay. A crystal structure of the S904F mutant reveals a small hydrophobic core around F904 likely to enhance basal kinase activity by stabilizing an active conformer. Our findings indicate that missense mutations in the activation loop of the kinase domain are able to increase kinase activity and confer drug resistance through allosteric effects. Mechanisms of acquired resistance to RET tyrosine kinase inhibitors in lung cancers are largely unknown. Here, the authors report in a lung adenocarcinoma patient harboring a CCDC6-RET mutation in the RET kinase (S904F) that results in resistance to the kinase inhibitor vandetanib by increasing the ATP affinity and autophosphorylation activity of RET kinase.
Collapse
|
36
|
Katsu K, Suzuki R, Tsuchiya W, Inagaki N, Yamazaki T, Hisano T, Yasui Y, Komori T, Koshio M, Kubota S, Walker AR, Furukawa K, Matsui K. A new buckwheat dihydroflavonol 4-reductase (DFR), with a unique substrate binding structure, has altered substrate specificity. BMC PLANT BIOLOGY 2017; 17:239. [PMID: 29228897 PMCID: PMC5725924 DOI: 10.1186/s12870-017-1200-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 12/01/2017] [Indexed: 05/12/2023]
Abstract
BACKGROUND Dihydroflavonol 4-reductase (DFR) is the key enzyme committed to anthocyanin and proanthocyanidin biosynthesis in the flavonoid biosynthetic pathway. DFR proteins can catalyse mainly the three substrates (dihydrokaempferol, dihydroquercetin, and dihydromyricetin), and show different substrate preferences. Although relationships between the substrate preference and amino acids in the region responsible for substrate specificity have been investigated in several plant species, the molecular basis of the substrate preference of DFR is not yet fully understood. RESULTS By using degenerate primers in a PCR, we isolated two cDNA clones that encoded DFR in buckwheat (Fagopyrum esculentum). Based on sequence similarity, one cDNA clone (FeDFR1a) was identical to the FeDFR in DNA databases (DDBJ/Gen Bank/EMBL). The other cDNA clone, FeDFR2, had a similar sequence to FeDFR1a, but a different exon-intron structure. Linkage analysis in an F2 segregating population showed that the two loci were linked. Unlike common DFR proteins in other plant species, FeDFR2 contained a valine instead of the typical asparagine at the third position and an extra glycine between sites 6 and 7 in the region that determines substrate specificity, and showed less activity against dihydrokaempferol than did FeDFR1a with an asparagine at the third position. Our 3D model suggested that the third residue and its neighbouring residues contribute to substrate specificity. FeDFR1a was expressed in all organs that we investigated, whereas FeDFR2 was preferentially expressed in roots and seeds. CONCLUSIONS We isolated two buckwheat cDNA clones of DFR genes. FeDFR2 has unique structural and functional features that differ from those of previously reported DFRs in other plants. The 3D model suggested that not only the amino acid at the third position but also its neighbouring residues that are involved in the formation of the substrate-binding pocket play important roles in determining substrate preferences. The unique characteristics of FeDFR2 would provide a useful tool for future studies on the substrate specificity and organ-specific expression of DFRs.
Collapse
Affiliation(s)
- Kenjiro Katsu
- National Agriculture and Food Research Organization (NARO), Kyushu Okinawa Agricultural Research Center, Suya 2421, Koshi, Kumamoto, 861-1192 Japan
| | - Rintaro Suzuki
- NARO, Advanced Analysis Center, Kannondai 2-1-2, Tsukuba, Ibaraki, 305-8602 Japan
| | - Wataru Tsuchiya
- NARO, Advanced Analysis Center, Kannondai 2-1-2, Tsukuba, Ibaraki, 305-8602 Japan
| | - Noritoshi Inagaki
- NARO, Advanced Analysis Center, Kannondai 2-1-2, Tsukuba, Ibaraki, 305-8602 Japan
| | - Toshimasa Yamazaki
- NARO, Advanced Analysis Center, Kannondai 2-1-2, Tsukuba, Ibaraki, 305-8602 Japan
| | - Tomomi Hisano
- National Agriculture and Food Research Organization (NARO), Kyushu Okinawa Agricultural Research Center, Suya 2421, Koshi, Kumamoto, 861-1192 Japan
| | - Yasuo Yasui
- Graduate School of Agriculture, Kyoto University, Yoshida-honmachi, Sakyou-ku, Kyoto, 606-8501 Japan
| | - Toshiyuki Komori
- Laboratory of Glycobiology, Department of Bioengineering, Nagaoka University, Kamitomioka 1603-1, Nagaoka, Niigata, 940-2188 Japan
| | - Motoyuki Koshio
- Laboratory of Glycobiology, Department of Bioengineering, Nagaoka University, Kamitomioka 1603-1, Nagaoka, Niigata, 940-2188 Japan
| | - Seiji Kubota
- Laboratory of Glycobiology, Department of Bioengineering, Nagaoka University, Kamitomioka 1603-1, Nagaoka, Niigata, 940-2188 Japan
| | - Amanda R. Walker
- CSIRO Agriculture & Food, Wine Innovation West, Hartley Grove, Urrbrae, SA 5064 Australia
| | - Kiyoshi Furukawa
- Laboratory of Glycobiology, Department of Bioengineering, Nagaoka University, Kamitomioka 1603-1, Nagaoka, Niigata, 940-2188 Japan
| | - Katsuhiro Matsui
- National Agriculture and Food Research Organization (NARO), Kyushu Okinawa Agricultural Research Center, Suya 2421, Koshi, Kumamoto, 861-1192 Japan
- Present address: NARO, Institute of Crop Science, Kannondai 2-1-2, Tsukuba, Ibaraki, 305-8518 Japan
| |
Collapse
|
37
|
Convex-PL: a novel knowledge-based potential for protein-ligand interactions deduced from structural databases using convex optimization. J Comput Aided Mol Des 2017; 31:943-958. [DOI: 10.1007/s10822-017-0068-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 09/08/2017] [Indexed: 12/16/2022]
|
38
|
Tomoike F, Nakagawa N, Fukui K, Yano T, Kuramitsu S, Masui R. Indispensable residue for uridine binding in the uridine-cytidine kinase family. Biochem Biophys Rep 2017; 11:93-98. [PMID: 28955773 PMCID: PMC5614712 DOI: 10.1016/j.bbrep.2017.07.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 06/09/2017] [Accepted: 07/03/2017] [Indexed: 10/31/2022] Open
Abstract
Uridine-cytidine kinase (UCK), including human UCK2, are a family of enzymes that generally phosphorylate both uridine and cytidine. However, UCK of Thermus thermophilus HB8 (ttCK) phosphorylates only cytidine. This cytidine-restricted activity is thought to depend on Tyr93, although the precise mechanism remains unresolved. Exhaustive mutagenesis of Tyr93 in ttCK revealed that the uridine phosphorylation activity was restored only by replacement of Tyr93 with His or Gln. Replacement of His117 in human UCK2, corresponding to residue Tyr93 in ttCK, by Tyr resulted in a loss of uridine phosphorylation activity. These findings indicated that uridine phosphorylation activity commonly depends on a single residue in the UCK family.
Collapse
Affiliation(s)
- Fumiaki Tomoike
- Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
- Research Center for Materials Science, Nagoya University, Furo-Cho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Noriko Nakagawa
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan
| | - Kenji Fukui
- Department of Biochemistry, Osaka Medical College, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan
| | - Takato Yano
- Department of Biochemistry, Osaka Medical College, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan
| | - Seiki Kuramitsu
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan
| | - Ryoji Masui
- Graduate School of Science, Osaka City University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka 558-8585, Japan
| |
Collapse
|
39
|
Yokoyama M, Oka T, Takagi H, Kojima H, Okabe T, Nagano T, Tohya Y, Sato H. A Proposal for a Structural Model of the Feline Calicivirus Protease Bound to the Substrate Peptide under Physiological Conditions. Front Microbiol 2017; 8:1383. [PMID: 28790989 PMCID: PMC5524728 DOI: 10.3389/fmicb.2017.01383] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/10/2017] [Indexed: 11/30/2022] Open
Abstract
Feline calicivirus (FCV) protease functions to cleave viral precursor proteins during productive infection. Previous studies have mapped a protease-coding region and six cleavage sites in viral precursor proteins. However, how the FCV protease interacts with its substrates remains unknown. To gain insights into the interactions, we constructed a molecular model of the FCV protease bound with the octapeptide containing a cleavage site of the capsid precursor protein by homology modeling and docking simulation. The complex model was used to screen for the substrate mimic from a chemical library by pharmacophore-based in silico screening. With this structure-based approach, we identified a compound that has physicochemical features and arrangement of the P3 and P4 sites of the substrate in the protease, is predicted to bind to FCV proteases in a mode similar to that of the authentic substrate, and has the ability to inhibit viral protease activity in vitro and in the cells, and to suppress viral replication in FCV-infected cells. The complex model was further subjected to molecular dynamics simulation to refine the enzyme-substrate interactions in solution. The simulation along with a variation study predicted that the authentic substrate and anti-FCV compound share a highly conserved binding site. These results suggest the validity of our in silico model for elucidating protease-substrate interactions during FCV replication and for developing antivirals.
Collapse
Affiliation(s)
- Masaru Yokoyama
- Pathogen Genomics Center, National Institute of Infectious DiseasesTokyo, Japan
| | - Tomoichiro Oka
- Department of Virology II, National Institute of Infectious DiseasesTokyo, Japan
| | - Hirotaka Takagi
- Division of Biosafety Control and Research, National Institute of Infectious DiseasesTokyo, Japan
| | | | - Takayoshi Okabe
- Drug Discovery Initiative, The University of TokyoTokyo, Japan
| | - Tetsuo Nagano
- Drug Discovery Initiative, The University of TokyoTokyo, Japan
| | - Yukinobu Tohya
- Department of Veterinary Medicine, Nihon UniversityFujisawa, Japan
| | - Hironori Sato
- Pathogen Genomics Center, National Institute of Infectious DiseasesTokyo, Japan
| |
Collapse
|
40
|
Chutiwitoonchai N, Mano T, Kakisaka M, Sato H, Kondoh Y, Osada H, Kotani O, Yokoyama M, Sato H, Aida Y. Inhibition of CRM1-mediated nuclear export of influenza A nucleoprotein and nuclear export protein as a novel target for antiviral drug development. Virology 2017; 507:32-39. [PMID: 28399435 PMCID: PMC7111614 DOI: 10.1016/j.virol.2017.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/31/2017] [Accepted: 04/03/2017] [Indexed: 12/18/2022]
Abstract
An anti-influenza compound, DP2392-E10 based on inhibition of the nuclear export function of the viral nucleoprotein-nuclear export signal 3 (NP-NES3) domain was successfully identified by our previous high-throughput screening system. Here, we demonstrated that DP2392-E10 exerts its antiviral effect by inhibiting replication of a broad range of influenza A subtypes. In regard to the molecular mechanism, we revealed that DP2392-E10 inhibits nuclear export of both viral NP and nuclear export protein (NEP). More specifically, in vitro pull-down assays revealed that DP2392-E10 directly binds cellular CRM1, which mediates nuclear export of NP and NEP. In silico docking suggested that DP2392-E10 binds at a region close to the HEAT9 and HEAT10 domains of CRM1. Together, these results indicate that the CRM1-mediated nuclear export function of influenza virus represents a new potential target for antiviral drug development, and also provide a core structure for a novel class of inhibitors that target this function.
Collapse
Affiliation(s)
| | - Takafumi Mano
- Viral Infectious Disease Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Michinori Kakisaka
- Viral Infectious Disease Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Hirotaka Sato
- Viral Infectious Disease Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Yasumitsu Kondoh
- Chemical Biology Research Group, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Hiroyuki Osada
- Chemical Biology Research Group, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Osamu Kotani
- Laboratory of Viral Genomics, Pathogen Genomics Center, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Masaru Yokoyama
- Laboratory of Viral Genomics, Pathogen Genomics Center, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Hironori Sato
- Laboratory of Viral Genomics, Pathogen Genomics Center, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Yoko Aida
- Viral Infectious Disease Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan.
| |
Collapse
|
41
|
Species Differences in the Binding of Sodium 4-Phenylbutyrate to Serum Albumin. J Pharm Sci 2017; 106:2860-2867. [PMID: 28456727 DOI: 10.1016/j.xphs.2017.04.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 04/13/2017] [Accepted: 04/13/2017] [Indexed: 01/30/2023]
Abstract
Sodium 4-phenylbutyrate (PB) is clinically used as a drug for treating urea cycle disorders. Recent research has shown that PB also has other pharmacologic activities, suggesting that it has the potential for use as a drug for treating other disorders. In the process of drug development, preclinical testing using experimental animals is necessary to verify the efficacy and safety of PB. Although the binding of PB to human albumin has been studied, our knowledge of its binding to albumin from the other animal species is extremely limited. To address this issue, we characterized the binding of PB to albumin from several species (human, bovine, rabbit, and rat). The results indicated that PB interacts with 1 high-affinity site of albumin from these species, which corresponds to site II of human albumin. The affinities of PB to human and bovine albumins were higher than those to rabbit and rat albumin, and that to rabbit albumin was the lowest. Binding and molecular docking studies using structurally related compounds of PB suggested that species differences in the affinity are attributed to differences in the structural feature of the PB-binding sites on albumins (e.g., charge distribution, hydrophobicity, shape, or size).
Collapse
|
42
|
In Silico and In Vitro Analysis of Interaction between Ximelagatran and Human Leukocyte Antigen (HLA)-DRB1*07:01. Int J Mol Sci 2017; 18:ijms18040694. [PMID: 28338626 PMCID: PMC5412280 DOI: 10.3390/ijms18040694] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/21/2017] [Accepted: 03/21/2017] [Indexed: 12/25/2022] Open
Abstract
Idiosyncratic ximelagatran-induced hepatotoxicity has been reported to be associated with human leukocyte antigen (HLA)-DRB1*07:01 and ximelagatran has been reported to inhibit the binding of the ligand peptide to HLA-DRB1*07:01 in vitro. In order to predict the possible interaction modes of ximelagatran with HLA-DR molecules, in silico docking simulations were performed. Molecular dynamics (MD) simulations were also performed to predict the effect of ximelagatran on the binding mode of the ligand peptide to HLA-DRB1*07:01. A series of in silico simulations supported the inhibitory effect of ximelagatran on the binding of the ligand peptide to HLA-DRB1*07:01 in vitro. Furthermore, direct interactions of ximelagatran with HLA-DR molecules were evaluated in vitro, which supported the simulated interaction mode of ximelagatran with HLA-DRB1*07:01. These results indicated that ximelagatran directly interacts with the peptide binding groove of HLA-DRB1*07:01 and competes with the ligand peptide for the binding site, which could alter the immune response and lead to the idiosyncratic ximelagatran-induced hepatotoxicity.
Collapse
|
43
|
Uchibori K, Inase N, Araki M, Kamada M, Sato S, Okuno Y, Fujita N, Katayama R. Brigatinib combined with anti-EGFR antibody overcomes osimertinib resistance in EGFR-mutated non-small-cell lung cancer. Nat Commun 2017; 8:14768. [PMID: 28287083 PMCID: PMC5355811 DOI: 10.1038/ncomms14768] [Citation(s) in RCA: 303] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/30/2017] [Indexed: 12/20/2022] Open
Abstract
Osimertinib has been demonstrated to overcome the epidermal growth factor receptor (EGFR)-T790M, the most relevant acquired resistance to first-generation EGFR-tyrosine kinase inhibitors (EGFR-TKIs). However, the C797S mutation, which impairs the covalent binding between the cysteine residue at position 797 of EGFR and osimertinib, induces resistance to osimertinib. Currently, there are no effective therapeutic strategies to overcome the C797S/T790M/activating-mutation (triple-mutation)-mediated EGFR-TKI resistance. In the present study, we identify brigatinib to be effective against triple-mutation-harbouring cells in vitro and in vivo. Our original computational simulation demonstrates that brigatinib fits into the ATP-binding pocket of triple-mutant EGFR. The structure-activity relationship analysis reveals the key component in brigatinib to inhibit the triple-mutant EGFR. The efficacy of brigatinib is enhanced markedly by combination with anti-EGFR antibody because of the decrease of surface and total EGFR expression. Thus, the combination therapy of brigatinib with anti-EGFR antibody is a powerful candidate to overcome triple-mutant EGFR.
Collapse
Affiliation(s)
- Ken Uchibori
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo 135-8550, Japan
- Department of Respiratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Naohiko Inase
- Department of Respiratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Mitsugu Araki
- RIKEN Advanced Institute for Computational Science, 7-1-26 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Mayumi Kamada
- Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shigeo Sato
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo 135-8550, Japan
| | - Yasushi Okuno
- RIKEN Advanced Institute for Computational Science, 7-1-26 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
- Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Naoya Fujita
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo 135-8550, Japan
| | - Ryohei Katayama
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo 135-8550, Japan
| |
Collapse
|
44
|
Hirayama N. Docking simulations between drugs and HLA molecules associated with idiosyncratic drug toxicity. Drug Metab Pharmacokinet 2017; 32:31-39. [DOI: 10.1016/j.dmpk.2016.10.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 09/10/2016] [Accepted: 10/11/2016] [Indexed: 12/13/2022]
|
45
|
Structural Insights into l-Tryptophan Dehydrogenase from a Photoautotrophic Cyanobacterium, Nostoc punctiforme. Appl Environ Microbiol 2017; 83:AEM.02710-16. [PMID: 27815281 DOI: 10.1128/aem.02710-16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 10/31/2016] [Indexed: 01/08/2023] Open
Abstract
l-Tryptophan dehydrogenase from Nostoc punctiforme NIES-2108 (NpTrpDH), despite exhibiting high amino acid sequence identity (>30%)/homology (>50%) with NAD(P)+-dependent l-Glu/l-Leu/l-Phe/l-Val dehydrogenases, exclusively catalyzes reversible oxidative deamination of l-Trp to 3-indolepyruvate in the presence of NAD+ Here, we determined the crystal structure of the apo form of NpTrpDH. The structure of the NpTrpDH monomer, which exhibited high similarity to that of l-Glu/l-Leu/l-Phe dehydrogenases, consisted of a substrate-binding domain (domain I, residues 3 to 133 and 328 to 343) and an NAD+/NADH-binding domain (domain II, residues 142 to 327) separated by a deep cleft. The apo-NpTrpDH existed in an open conformation, where domains I and II were apart from each other. The subunits dimerized themselves mainly through interactions between amino acid residues around the β-1 strand of each subunit, as was observed in the case of l-Phe dehydrogenase. The binding site for the substrate l-Trp was predicted by a molecular docking simulation and validated by site-directed mutagenesis. Several hydrophobic residues, which were located in the active site of NpTrpDH and possibly interacted with the side chain of the substrate l-Trp, were arranged similarly to that found in l-Leu/l-Phe dehydrogenases but fairly different from that of an l-Glu dehydrogenase. Our crystal structure revealed that Met-40, Ala-69, Ile-74, Ile-110, Leu-288, Ile-289, and Tyr-292 formed a hydrophobic cluster around the active site. The results of the site-directed mutagenesis experiments suggested that the hydrophobic cluster plays critical roles in protein folding, l-Trp recognition, and catalysis. Our results provide critical information for further characterization and engineering of this enzyme. IMPORTANCE In this study, we determined the three-dimensional structure of l-Trp dehydrogenase, analyzed its various site-directed substitution mutants at residues located in the active site, and obtained the following informative results. Several residues in the active site form a hydrophobic cluster, which may be a part of the hydrophobic core essential for protein folding. To our knowledge, there is no previous report demonstrating that a hydrophobic cluster in the active site of any l-amino acid dehydrogenase may have a critical impact on protein folding. Furthermore, our results suggest that this hydrophobic cluster could strictly accommodate l-Trp. These studies show the structural characteristics of l-Trp dehydrogenase and hence would facilitate novel applications of l-Trp dehydrogenase.
Collapse
|
46
|
Araki M, Kamiya N, Sato M, Nakatsui M, Hirokawa T, Okuno Y. The Effect of Conformational Flexibility on Binding Free Energy Estimation between Kinases and Their Inhibitors. J Chem Inf Model 2016; 56:2445-2456. [PMID: 28024406 DOI: 10.1021/acs.jcim.6b00398] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Accurate prediction of binding affinities of drug candidates to their targets remains challenging because of protein flexibility in solution. Conformational flexibility of the ATP-binding site in the CDK2 and ERK2 kinases was identified using molecular dynamics simulations. The binding free energy (ΔG) of twenty-four ATP-competitive inhibitors toward these kinases was assessed using an alchemical free energy perturbation method, MP-CAFEE. However, large calculation errors of 2-3 kcal/mol were observed using this method, where the free energy simulation starts from a single equilibrated conformation. Here, we developed a new ΔG computation method, where the starting structure was set to multiconformations to cover flexibility. The calculation accuracy was successfully improved, especially for larger molecular size compounds, leading to reliable prediction of a broader range of drug candidates. The present study demonstrates that conformational flexibility of interactions between a compound and the glycine-rich loop in the kinases is a key factor in ΔG estimation.
Collapse
Affiliation(s)
- Mitsugu Araki
- RIKEN Advanced Institute for Computational Science, 7-1-26 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Narutoshi Kamiya
- RIKEN Advanced Institute for Computational Science, 7-1-26 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.,Graduate School of Simulation Studies, University of Hyogo , 7-1-28 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Miwa Sato
- Mitsui Knowledge Industry Co., Ltd., 2-5-1 Atago, Minato-ku, Tokyo 105-6215, Japan
| | - Masahiko Nakatsui
- RIKEN Advanced Institute for Computational Science, 7-1-26 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.,Graduate School of Medicine, Kyoto University , 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Takatsugu Hirokawa
- Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST) , 2-4-7 Aomi, Koto-ku, Tokyo 135-0064, Japan.,Division of Biomedical Science, Faculty of Medicine, University of Tsukuba , 1-1-1 Tennodai, Tsukuba-shi, Ibaraki 305-8575, Japan
| | - Yasushi Okuno
- RIKEN Advanced Institute for Computational Science, 7-1-26 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.,Graduate School of Medicine, Kyoto University , 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
47
|
Novel Concepts for Drug Hypersensitivity Based on the Use of Long-Time Scale Molecular Dynamic Simulation. JOURNAL OF PHARMACEUTICS 2016; 2016:9520361. [PMID: 27999707 PMCID: PMC5141551 DOI: 10.1155/2016/9520361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 11/03/2016] [Indexed: 01/11/2023]
Abstract
The discovery that several drug hypersensitivity reactions (DHRs) are associated with specific human leukocyte antigen (HLA) alleles has attracted increasing research interest. However, the underlying mechanisms of these HLA-induced DHRs remain unclear, especially for drug-induced immediate activation of T-cell clones (TCCs). Recently, a novel hypothesis involving partial detachment between self-peptide(s) and the HLA molecule (altered peptide-HLA (pHLA) model) has been proposed to explain these phenomena. In order to clarify this hypothesis, we performed long-timescale molecular dynamics (MD) simulations. We focused on HLA-B⁎57:01-restricted abacavir hypersensitivity reactions (AHRs), one of the most famous DHRs. One of the simulation results showed that this altered-pHLA model might be driven by an increase in the distance not only between HLA and self-peptides but also between the α1 and α2 helices of HLA. Our findings provide novel insights into abacavir-induced immediate activation of TCCs and these findings might also be applied to other DHRs, such as HLA-B⁎58:01-restricted allopurinol hypersensitivity reactions.
Collapse
|
48
|
Okanishi H, Kim K, Fukui K, Yano T, Kuramitsu S, Masui R. Proteome-wide identification of lysine succinylation in thermophilic and mesophilic bacteria. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1865:232-242. [PMID: 27888076 DOI: 10.1016/j.bbapap.2016.11.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 10/13/2016] [Accepted: 11/19/2016] [Indexed: 02/07/2023]
Abstract
Lysine succinylation, one of post-translational acylations conserved from eukaryotes to bacteria, plays regulatory roles in various cellular processes. However, much remains unknown about the general and specific characteristics of lysine succinylation among bacteria, and about its functions different from those of other acylations. In this study, we characterized lysine succinylation, a newly discovered widespread type of lysine acylation in five bacterial species with different characteristics such as optimal growth temperature and cell wall structure. This study is the first to demonstrate that succinylation is general phenomenon occurring not only in mesophiles but also in thermophiles. Mapping of succinylation sites on protein structures revealed that succinylation occurs at many lysine residues important for protein function. Comparison of the succinylation sites in the five bacterial species provides insights regarding common protein regulation mechanisms utilizing lysine succinylation. Many succinylation sites were conserved among five bacteria, especially between Geobacillus kaustophilus and Bacillus subtilis, some of which are functionally important sites. Furthermore, systematic comparison of the succinyl-proteome results and our previous propionyl-proteome results showed that the abundance of these two types of acylations is considerably different among the five bacteria investigated. Many succinylation and propionylation events were detected in G. kaustophilus, whereas Escherichia coli and B. subtilis exhibited high succinylation and low propionylation; low succinylation and high propionylation were identified in Thermus thermophilus, and low succinylation and propionylation were observed in Rhodothermus marinus. Comparison of the characteristics of lysine succinylation and lysine propionylation suggested these two types of acylation play different roles in cellular processes.
Collapse
Affiliation(s)
- Hiroki Okanishi
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan; Division of Biology & Geosciences, Graduate School of Science, Osaka City University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka 558-8585, Japan
| | - Kwang Kim
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan.
| | - Kenji Fukui
- Department of Biochemistry, Faculty of Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan
| | - Takato Yano
- Department of Biochemistry, Faculty of Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan
| | - Seiki Kuramitsu
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan
| | - Ryoji Masui
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan; Division of Biology & Geosciences, Graduate School of Science, Osaka City University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka 558-8585, Japan
| |
Collapse
|
49
|
Development of BCR-ABL degradation inducers via the conjugation of an imatinib derivative and a cIAP1 ligand. Bioorg Med Chem Lett 2016; 26:4865-4869. [DOI: 10.1016/j.bmcl.2016.09.041] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 09/12/2016] [Accepted: 09/15/2016] [Indexed: 11/20/2022]
|
50
|
Kim IS, Hwang JH, Hirano M, Iwata H, Kim EY. In vitro and in silico evaluation of transactivation potencies of avian AHR1 and AHR2 by endogenous ligands: Implications for the physiological role of avian AHR2. Comp Biochem Physiol C Toxicol Pharmacol 2016; 187:1-9. [PMID: 27060260 DOI: 10.1016/j.cbpc.2016.03.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 02/05/2016] [Accepted: 03/29/2016] [Indexed: 02/09/2023]
Abstract
Aryl hydrocarbon receptor (AHR) is well conserved from invertebrates to vertebrates, and it mediates the toxic effects of exogenous ligands, including dioxins. Recent studies reported that AHRs activated by endogenous ligands play critical roles in mammalian physiological homeostasis. Avian species possess at least two AHR isoforms (AHR1 and AHR2), which exhibit species- and isoform-specific transactivation potencies to exogenous ligands, whereas mammals possess a single AHR. To delineate the profiles and roles of endogenous ligands for avian AHR isoforms, we investigated in vitro transactivation potencies of avian AHRs (AHR1 and AHR2 from the jungle crow, Corvus macrorhynchos; common cormorant, Phalacrocorax carbo; and black-footed albatross, Phoebastria nigripes) treated with the endogenous tryptophan metabolites 6-formylindolo [3,2-b] carbazole (FICZ), l-kynurenine (l-Kyn), kynurenic acid (KYNA), and indoxyl sulfate (IS). Furthermore, we analyzed the binding mode of these ligands to each avian AHR isoform by in silico docking simulations. The EC50 of FICZ (0.009-0.032nM) was similar regardless of the species or isoform of AHR. The estimated in silico binding mode of FICZ to AHRs was well conserved in both isoforms. The transactivation potencies of avian AHRs to other tryptophan metabolites were 10(5)-10(7) fold lower than those for FICZ, and EC50 values varied in a species- and isoform-specific manner. This was consistent with poor conservation of the binding mode of l-Kyn, KYNA, and IS predicted in in silico docking simulations. Our results suggest that in avian species, FICZ is the most potent endogenous AHR ligand, and that AHR1 and AHR2 are physiologically functional.
Collapse
Affiliation(s)
- In-Sung Kim
- Department of Biology, Kyung Hee University, Hoegi Dong, Dongdaemun-Gu, Seoul 130-701, Republic of Korea
| | - Ji-Hee Hwang
- Department of Biology, Kyung Hee University, Hoegi Dong, Dongdaemun-Gu, Seoul 130-701, Republic of Korea; Department of Life and Nanopharmaceutical Science, Kyung Hee University, Hoegi Dong, Dongdaemun-Gu, Seoul 130-701, Republic of Korea
| | - Masashi Hirano
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama 790-8577, Japan
| | - Hisato Iwata
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama 790-8577, Japan
| | - Eun-Young Kim
- Department of Biology, Kyung Hee University, Hoegi Dong, Dongdaemun-Gu, Seoul 130-701, Republic of Korea; Department of Life and Nanopharmaceutical Science, Kyung Hee University, Hoegi Dong, Dongdaemun-Gu, Seoul 130-701, Republic of Korea.
| |
Collapse
|