51
|
A novel quinazoline-based analog induces G2/M cell cycle arrest and apoptosis in human A549 lung cancer cells via a ROS-dependent mechanism. Biochem Biophys Res Commun 2017; 486:314-320. [PMID: 28302490 DOI: 10.1016/j.bbrc.2017.03.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 03/11/2017] [Indexed: 12/16/2022]
Abstract
6-amino-4-(4-phenoxyphenylethylamino)quinazoline (QNZ) is an excellent quinazoline-containing NF-κB inhibitor also acting as a novel anticancer agent. Considering both the medicinal significance of quinazoline scaffold and the tunable functionality of Michael acceptor-centric pharmacophores in the electrophilicity-based prooxidant strategy, we designed a novel QNZ-inspired electrophilic molecule QNZ-A by introducing a Michael acceptor unit at position-6 of quinazoline ring in QNZ. Our results identified QNZ-A as a promising selective cytotoxic agent against A549 cells. QNZ-A, by virtue of its Michael acceptor unit, induced reactive oxygen species (ROS) accumulation associated with collapse of the redox buffering system in A549 cells. This caused up-regulation of p53-inducible p21 and down-regulation of redox sensitive Cdc25C along with Cyclin B1/Cdk1, leading to a G2/M cell cycle arrest and final cell apoptosis. By contrast, QNZ-B, a reduction product of QNZ-A lacking the Michael acceptor unit failed to induce ROS generation and all these cell cycle-related events. In conclusion, this work provided a successful example of designing QNZ-directed anticancer agent by a ROS-promoting strategy and identified QNZ-A as a selective anticancer agent against A549 cells through G2/M cell cycle arrest and apoptosis via a ROS-dependent mechanism.
Collapse
|
52
|
Wang HB, Jin XL, Zheng JF, Wang F, Dai F, Zhou B. Developing piperlongumine-directed glutathione S-transferase inhibitors by an electrophilicity-based strategy. Eur J Med Chem 2017; 126:517-525. [PMID: 27914365 DOI: 10.1016/j.ejmech.2016.11.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 10/29/2016] [Accepted: 11/14/2016] [Indexed: 01/10/2023]
Abstract
We report a case of successful design of glutathione S-transferase (GST) inhibitors via a natural product-inspired and electrophilicity-based strategy. Based on this strategy, a novel piperlongumine analog (PL-13) bearing a para-trifluoromethyl group and an α-chlorine on its aromatic and lactam rings, respectively, surfaced as a promising GST inhibitor, thereby overcoming cisplatin resistance in lung cancer A549 cells.
Collapse
Affiliation(s)
- Hai-Bo Wang
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Xiao-Ling Jin
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Jia-Fang Zheng
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Fu Wang
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Fang Dai
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Bo Zhou
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China.
| |
Collapse
|
53
|
Pan L, Li X, Jin H, Yang X, Qin B. Antifungal activity of umbelliferone derivatives: Synthesis and structure-activity relationships. Microb Pathog 2017; 104:110-115. [PMID: 28089948 DOI: 10.1016/j.micpath.2017.01.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 01/04/2017] [Accepted: 01/11/2017] [Indexed: 11/15/2022]
Abstract
Umbelliferone was an important allelochemical with a wide spectrum bioactivity. In our previous study, C7 hydroxy in the backbone of umbelliferone was identified to be responsible for its phytotoxicity and the targeted modification of the above site could lead to the phytotoxicity loss. In view of this, a series of hydroxycoumarins and C7 O-substituted umbelliferone derivatives were efficiently synthesized to evaluate their antifungal activity against four phytopathogenic fungi. Most of them, as we predicted, exhibited improved fungicidal activity. The phytotoxicity of effective compounds was also assayed by Lactuca sativa to investigate their side effects on plant growth. Compounds 9 and 17 were identified to show strong antifungal activity with low phytotoxicity. A brief investigation on structure-activity relationships revealed that the modification at the C7 hydroxy of umbelliferone could be a promising way to enhance the antifungal activity with decreasing the phytotoxicity.
Collapse
Affiliation(s)
- Le Pan
- Key Laboratory of Chemistry of Northwestern Plant Resources of CAS and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, People's Republic of China; Chemical Engineering College, Xinjiang Agricultural University, Urumqi 830052, People's Republic of China
| | - Xiuzhuang Li
- Key Laboratory of Chemistry of Northwestern Plant Resources of CAS and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, People's Republic of China.
| | - Hui Jin
- Key Laboratory of Chemistry of Northwestern Plant Resources of CAS and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, People's Republic of China
| | - Xiaoyan Yang
- Key Laboratory of Chemistry of Northwestern Plant Resources of CAS and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, People's Republic of China
| | - Bo Qin
- Key Laboratory of Chemistry of Northwestern Plant Resources of CAS and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, People's Republic of China.
| |
Collapse
|
54
|
Zhang B, Zhang J, Peng S, Liu R, Li X, Hou Y, Han X, Fang J. Thioredoxin reductase inhibitors: a patent review. Expert Opin Ther Pat 2016; 27:547-556. [DOI: 10.1080/13543776.2017.1272576] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Baoxin Zhang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Junmin Zhang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Shoujiao Peng
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Ruijuan Liu
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Xinming Li
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Yanan Hou
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Xiao Han
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| |
Collapse
|
55
|
Liang B, Liu Z, Cao Y, Zhu C, Zuo Y, Huang L, Wen G, Shang N, Chen Y, Yue X, Du J, Li B, Zhou B, Bu X. MC37, a new mono-carbonyl curcumin analog, induces G2/M cell cycle arrest and mitochondria-mediated apoptosis in human colorectal cancer cells. Eur J Pharmacol 2016; 796:139-148. [PMID: 28024945 DOI: 10.1016/j.ejphar.2016.12.030] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 12/19/2016] [Accepted: 12/20/2016] [Indexed: 12/11/2022]
Abstract
(E)-1-(3'-fluoro-[1,1'-biphenyl-3-yl)-3-(3-hydroxy-4-methoxyphenyl)prop-2-en-1-one) (MC37), a novel mono-carbonyl curcumin analog, was previously synthesized in our laboratory as a nuclear factor kappa B (NF-κB) inhibitor with excellent cytotoxicity against several cancer cell lines. In this study, our further investigations showed that the potent growth inhibitory activity of MC37 in human colorectal cancer cells was associated with the arrest of cell cycle progression and the induction of apoptosis. As a multi-targeted agent, MC37 inhibited the intracellular microtubule assembly, altered the expression of cyclin-dependent kinase 1 (CDK1), and ultimately induced G2/M cell cycle arrest. Moreover, MC37 collapsed the mitochondrial membrane potential (MMP), increased the Bax/Bcl-2 ratio, activated the caspase-9/3 cascade, and finally led to cancer cells apoptosis, suggesting that the mitochondrial-mediated apoptotic pathway was involved in MC37-induced apoptosis. In conclusion, these observations demonstrated that mono-carbonyl curcumin analogs would serve as multi-targeted lead for promising anti-colorectal cancer agent development.
Collapse
Affiliation(s)
- Baoxia Liang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Ziyi Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Yingnan Cao
- Department of Pharmacology, Xinhua College of Sun Yat-sen University, Guangzhou, PR China
| | - Cuige Zhu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Yinglin Zuo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Lei Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Gesi Wen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Nana Shang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Yu Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Xin Yue
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Jun Du
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Baojian Li
- Shenzhen Davoos tech. Ltd.Co., Room A611, Silver star tech. building, 1301 Guanguang Road, Guanlan, Longhua District, Shenzhen, PR China
| | - Binhua Zhou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, PR China.
| | - Xianzhang Bu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, PR China.
| |
Collapse
|
56
|
Koh D, Jung Y, Kim BS, Ahn S, Lim Y. 1 H and 13 C NMR spectral assignments of naphthalenyl chalcone derivatives. MAGNETIC RESONANCE IN CHEMISTRY : MRC 2016; 54:842-851. [PMID: 27296310 DOI: 10.1002/mrc.4458] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/02/2016] [Accepted: 05/07/2016] [Indexed: 06/06/2023]
Affiliation(s)
- Dongsoo Koh
- Department of Applied Chemistry, Dongduk Women's University, Seoul, 136-714, Korea
| | - Yearam Jung
- Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul, 143-701, Korea
| | - Beom Soo Kim
- Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul, 143-701, Korea
| | - Seunghyun Ahn
- Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul, 143-701, Korea
| | - Yoongho Lim
- Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul, 143-701, Korea
| |
Collapse
|
57
|
Song JX, Sun YR, Peluso I, Zeng Y, Yu X, Lu JH, Xu Z, Wang MZ, Liu LF, Huang YY, Chen LL, Durairajan SSK, Zhang HJ, Zhou B, Zhang HQ, Lu A, Ballabio A, Medina DL, Guo Z, Li M. A novel curcumin analog binds to and activates TFEB in vitro and in vivo independent of MTOR inhibition. Autophagy 2016; 12:1372-1389. [PMID: 27172265 PMCID: PMC4968239 DOI: 10.1080/15548627.2016.1179404] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 04/07/2016] [Accepted: 04/11/2016] [Indexed: 10/21/2022] Open
Abstract
Autophagy dysfunction is a common feature in neurodegenerative disorders characterized by accumulation of toxic protein aggregates. Increasing evidence has demonstrated that activation of TFEB (transcription factor EB), a master regulator of autophagy and lysosomal biogenesis, can ameliorate neurotoxicity and rescue neurodegeneration in animal models. Currently known TFEB activators are mainly inhibitors of MTOR (mechanistic target of rapamycin [serine/threonine kinase]), which, as a master regulator of cell growth and metabolism, is involved in a wide range of biological functions. Thus, the identification of TFEB modulators acting without inhibiting the MTOR pathway would be preferred and probably less deleterious to cells. In this study, a synthesized curcumin derivative termed C1 is identified as a novel MTOR-independent activator of TFEB. Compound C1 specifically binds to TFEB at the N terminus and promotes TFEB nuclear translocation without inhibiting MTOR activity. By activating TFEB, C1 enhances autophagy and lysosome biogenesis in vitro and in vivo. Collectively, compound C1 is an orally effective activator of TFEB and is a potential therapeutic agent for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ju-Xian Song
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
- Mr. & Mrs. Ko Chi Ming Center for Parkinson Disease Research (CPDR), Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Yue-Ru Sun
- Department of Chemistry, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Ivana Peluso
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
| | - Yu Zeng
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
- Mr. & Mrs. Ko Chi Ming Center for Parkinson Disease Research (CPDR), Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Xing Yu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
- Mr. & Mrs. Ko Chi Ming Center for Parkinson Disease Research (CPDR), Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Jia-Hong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Zheng Xu
- Department of Chemistry, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Ming-Zhong Wang
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Liang-Feng Liu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
- Mr. & Mrs. Ko Chi Ming Center for Parkinson Disease Research (CPDR), Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Ying-Yu Huang
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
- Mr. & Mrs. Ko Chi Ming Center for Parkinson Disease Research (CPDR), Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Lei-Lei Chen
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
- Mr. & Mrs. Ko Chi Ming Center for Parkinson Disease Research (CPDR), Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Siva Sundara Kumar Durairajan
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
- Mr. & Mrs. Ko Chi Ming Center for Parkinson Disease Research (CPDR), Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Hong-Jie Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
- Mr. & Mrs. Ko Chi Ming Center for Parkinson Disease Research (CPDR), Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Bo Zhou
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, Gansu, China
| | - Hong-Qi Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
| | - Diego L. Medina
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
| | - Zhihong Guo
- Department of Chemistry, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Min Li
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
- Mr. & Mrs. Ko Chi Ming Center for Parkinson Disease Research (CPDR), Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| |
Collapse
|
58
|
Yan WJ, Wang Q, Yuan CH, Wang F, Ji Y, Dai F, Jin XL, Zhou B. Designing piperlongumine-directed anticancer agents by an electrophilicity-based prooxidant strategy: A mechanistic investigation. Free Radic Biol Med 2016; 97:109-123. [PMID: 27233942 DOI: 10.1016/j.freeradbiomed.2016.05.021] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 05/21/2016] [Accepted: 05/21/2016] [Indexed: 02/05/2023]
Abstract
Piperlongumine (PL), a natural electrophilic alkaloid bearing two α, β-unsaturated imides, is a promising anticancer molecule by targeting the stress response to reactive oxygen species (ROS). Considering that ROS generation depends on electrophilicity of PL, PL-CL was designed as its analog by introducing the α-substituent chlorine on the lactam ring to increase moderately its electrophilicity. In comparison with the parent molecule, this molecule was identified as a stronger ROS (O2(∙-) and H2O2) inducer and cytotoxic agent, and manifested more than 15-fold selectivity toward A549 cells over normal WI-38 cells. Mechanistic study uncovers for the first time that the selenoprotein thioredoxin reductase (TrxR) is one of the targets by which PL-CL promotes the ROS generation. Stronger intracellular TrxR inhibition and higher accumulation of ROS (O2(∙-) and H2O2) are responsible for more effective S-phase arrest and mitochondria-mediated apoptotic induction of A549 cells by PL-CL than PLvia p53-p21-cyclinA/CDK2 and ASK1-JNK/p38 signaling cascade pathways, respectively. This work provides an example of successfully designing PL-directed anticancer agent by an electrophilicity-based prooxidant (ROS-generating agent) strategy and gives added confidence for extending this strategy to other natural products.
Collapse
Affiliation(s)
- Wen-Jing Yan
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Qi Wang
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Cui-Hong Yuan
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Fu Wang
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Yuan Ji
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Fang Dai
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Xiao-Ling Jin
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Bo Zhou
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China.
| |
Collapse
|
59
|
2,2'-Fluorine mono-carbonyl curcumin induce reactive oxygen species-Mediated apoptosis in Human lung cancer NCI-H460 cells. Eur J Pharmacol 2016; 786:161-168. [PMID: 27266668 DOI: 10.1016/j.ejphar.2016.06.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 05/27/2016] [Accepted: 06/03/2016] [Indexed: 12/13/2022]
Abstract
In this paper, we synthesized three fluorine-substituted mono-carbonyl curcumin analogs and evaluated their cytotoxicity against several cancer cells by the MTT assay. The results exhibited that all the three compounds were more active than the leading curcumin. Especially, 2,2'-F mono-carbonyl curcumin, 1a, surfaced as an important lead compound displaying almost 4-fold cytotoxicity relative to curcumin. More importantly, 1a was more stable in (RPMI)-1640 medium and more massive uptake than curcumin, which may be relationship to their cytotoxicity, apoptotic acitivity and reactive oxygen species generation. And then, the generation of reactive oxygen species can disrupt the intracellular redox balance, induce lipid peroxidation, cause the collapse of the mitochondrial membrane potential and ultimately lead to apoptosis. The results not only suggest that 2,2'-F mono-carbonyl curcumin (1a) may cause cancer cells apoptosis through reactive oxygen species-Mediated pathway, but also gives us an important information for design of mono-carbonyl curcumin analog.
Collapse
|
60
|
Kang YF, Qiao HX, Xin LZ, Ge LP. Chain elongation analog of resveratrol as potent cancer chemoprevention agent. J Physiol Biochem 2016; 72:445-52. [PMID: 27160168 DOI: 10.1007/s13105-016-0487-3] [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: 01/26/2016] [Accepted: 04/12/2016] [Indexed: 11/26/2022]
Abstract
Resveratrol is identified as a natural cancer chemoprevention agent. There has been a lot of interest in designing and developing resveratrol analogs with cancer chemoprevention activity superior to that of parent molecule and exploring their action mechanism in the past several decades. In this study, we have synthesized resveratrol analogs of compounds A-C via conjugated chain elongation based on isoprene unit retention strategy. Remarkably, cytotoxic activity analysis results indicated that compound B possesses the best proliferation inhibition activity for NCI-H460 cells in all the test compounds. Intriguingly, compound B displayed a higher cytotoxicity against human non-small cell lung cancer cells (NCI-H460) compared to normal human embryonic lung fibroblasts (MRC-5). Afterward, flow cytometry analysis showed that compound B would induce cell apoptosis. We further researched the action mechanism. When NCI-H460 cells were incubated by compound B for 6 or 9 h, respectively, the intracellular reactive oxygen species (ROS) level was enhanced obviously. With elevation of intracellular ROS level, flow cytometry measurement verified mitochondrial transmembrane potential collapse, which was accompanied by the up-regulation of Bax and down-regulation of Bcl-2. More interestingly, compound B increased the expression of caspase-9 and caspase-3, which induced cell apoptosis. Moreover, compound B arrested cell cycle in G0/G1 phase. These are all to provide useful information for designing resveratrol-based chemoprevention agent and understanding the action mechanism.
Collapse
Affiliation(s)
- Yan-Fei Kang
- College of Laboratory Medicine, Hebei North University, 11 Diamond Street South, Zhangjiakou, 075000, Hebei Province, People's Republic of China.
| | - Hai-Xia Qiao
- College of Laboratory Medicine, Hebei North University, 11 Diamond Street South, Zhangjiakou, 075000, Hebei Province, People's Republic of China
| | - Long-Zuo Xin
- College of Agriculture and Forestry Science and Technology, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Li-Ping Ge
- College of Laboratory Medicine, Hebei North University, 11 Diamond Street South, Zhangjiakou, 075000, Hebei Province, People's Republic of China
| |
Collapse
|
61
|
Qiu P, Man S, Li J, Liu J, Zhang L, Yu P, Gao W. Overdose Intake of Curcumin Initiates the Unbalanced State of Bodies. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:2765-2771. [PMID: 26978516 DOI: 10.1021/acs.jafc.6b00053] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Curcumin is the major active component of turmeric and widely used as a spice and coloring agent in food. However, its safety evaluation has been little investigated. To evaluate the 90-day subchronic toxicity of curcumin in rats, its general observation, clinical biochemistry, pathology, and metabolomics were evaluated. The results showed that curcumin induced liver injury through the generation of the overexpression of reactive oxygen species (ROS) and pro-inflammatory cytokines IL-6 and the decreases of the levels of antioxidant enzyme SOD and detoxified enzyme GST. Meanwhile, for the self-protection of rats, curcumin treatment activated the transcription of Nrf-2 and elevated the expression of HO-1 to reduce tissue damage. Furthermore, curcumin significantly increased key mRNA levels of HK2, PKM2, LDHA, CES, Cpt1, Cpt2, FASN, and ATP5b and decreased levels of GLUT2 and ACC1 to enhance glycolysis and inhibit lipid metabolism and TCA cycle. Therefore, overdose or long-term intake of curcumin could initiate the unbalanced state of bodies through oxidative stress, inflammation, and metabolic disorders, which induces liver injury. Intermittent administration of curcumin is necessary in our daily lives.
Collapse
Affiliation(s)
- Peiyu Qiu
- Key Laboratory of Industrial Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science & Technology , Tianjin 300457, China
- Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology , Tianjin 300457, China
| | - Shuli Man
- Key Laboratory of Industrial Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science & Technology , Tianjin 300457, China
- Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology , Tianjin 300457, China
| | - Jing Li
- Key Laboratory of Industrial Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science & Technology , Tianjin 300457, China
| | - Jing Liu
- Key Laboratory of Industrial Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science & Technology , Tianjin 300457, China
| | - Liming Zhang
- Key Laboratory of Industrial Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science & Technology , Tianjin 300457, China
| | - Peng Yu
- Key Laboratory of Industrial Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science & Technology , Tianjin 300457, China
| | - Wenyuan Gao
- Tianjin Key Laboratory for Modern Drug Delivery and High Efficiency, School of Pharmaceutical Science and Technology, Tianjin University , Tianjin 300072, China
| |
Collapse
|
62
|
Zhang J, Li Y, Duan D, Yao J, Gao K, Fang J. Inhibition of thioredoxin reductase by alantolactone prompts oxidative stress-mediated apoptosis of HeLa cells. Biochem Pharmacol 2016; 102:34-44. [PMID: 26686580 DOI: 10.1016/j.bcp.2015.12.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 12/08/2015] [Indexed: 01/17/2023]
Abstract
The mammalian thioredoxin reductase (TrxR) isoenzymes, TrxR1 in cytosol or nucleus, TrxR2 in mitochondria, and TrxR3 in testis, are essential seleno-flavoenzymes with a conserved penultimate selenocysteine (Sec) residue at the C-terminus, and have attracted increasing interests as potential targets for development of cancer chemotherapeutic agents. The sesquiterpene lactone alantolactone (ATL), an active component from the traditional folk medicine Inula helenium, has been documented possessing multiple pharmacological functions, especially the anticancer activity. However, the underlying mechanism has not been well defined. We reported that ATL inhibits both the recombinant TrxR and the enzyme in the cellular environment. The alpha-methylene-gamma-lactone moiety in ATL and the Sec residue in TrxR are critical for targeting TrxR by ATL. By employing our newly developed pull down assay, we demonstrated the remarkable elevation of the oxidized thioredoxin in HeLa cells after ATL treatment. In addition, ATL elicits accumulation of reactive oxygen species, and eventually induces apoptosis of HeLa cells. Importantly, overexpression of the functional TrxR attenuates the cytotoxicity of ATL, while knockdown of the enzyme sensitizes the cells to ATL treatment. Targeting TrxR thus discloses a novel molecular mechanism underlying the cellular action of ATL, and sheds light in considering the usage of ATL as a potential cancer chemotherapeutic agent.
Collapse
Affiliation(s)
- Junmin Zhang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Ya Li
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Dongzhu Duan
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Juan Yao
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Kun Gao
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
63
|
Liu GY, Sun YZ, Zhou N, Du XM, Yang J, Guo SJ. 3,3'-OH curcumin causes apoptosis in HepG2 cells through ROS-mediated pathway. Eur J Med Chem 2016; 112:157-163. [PMID: 26894841 DOI: 10.1016/j.ejmech.2016.02.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 02/04/2016] [Accepted: 02/05/2016] [Indexed: 01/14/2023]
Abstract
In this paper, we synthesized a series of curcumin analogs and evaluated their cytotoxicity against HepG2 cells. The results exhibited that the hydroxyl group at 3,3'-position play an essential role in enhancing their anti-proliferation activity. More importantly, 3,3'-hydroxy curcumin (1b) caused apoptosis in HepG2 cells with the ROS generation, which may be mainly composed of hydroxyl radicals (HO) and H2O2. The more cytotoxic activity and ROS-generating ability of 1b may be due to the more stable in (RPMI)-1640 medium and more massive uptake than curcumin. Then the generation of ROS can disrupt the intracellular redox balance, induce lipid peroxidation, cause the collapse of the mitochondrial membrane potential and ultimately lead to apoptosis. The results not only suggest that 3,3'-hydroxy curcumin (1b) may cause HepG2 cells apoptosis through ROS-mediated pathway, but also offer an important information for design of curcumin analog.
Collapse
Affiliation(s)
- Guo-Yun Liu
- School of Pharmacy, Liaocheng University, 1 Hunan Street, Liaocheng, Shandong 252000, China
| | - Yong-Zheng Sun
- School of Pharmacy, Liaocheng University, 1 Hunan Street, Liaocheng, Shandong 252000, China
| | - Na Zhou
- School of Pharmacy, Liaocheng University, 1 Hunan Street, Liaocheng, Shandong 252000, China
| | - Xiu-Mei Du
- School of Pharmacy, Liaocheng University, 1 Hunan Street, Liaocheng, Shandong 252000, China
| | - Jie Yang
- School of Pharmacy, Liaocheng University, 1 Hunan Street, Liaocheng, Shandong 252000, China.
| | - Shang-Jing Guo
- School of Pharmacy, Liaocheng University, 1 Hunan Street, Liaocheng, Shandong 252000, China
| |
Collapse
|
64
|
Li Y, Zhang LP, Dai F, Yan WJ, Wang HB, Tu ZS, Zhou B. Hexamethoxylated Monocarbonyl Analogues of Curcumin Cause G2/M Cell Cycle Arrest in NCI-H460 Cells via Michael Acceptor-Dependent Redox Intervention. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:7731-7742. [PMID: 26255837 DOI: 10.1021/acs.jafc.5b02011] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Curcumin, derived from the dietary spice turmeric, holds promise for cancer prevention. This prompts much interest in investigating the action mechanisms of curcumin and its analogues. Two symmetrical hexamethoxy-diarylpentadienones (1 and 2) as cucumin analogues were reported to possess significantly enhanced cytotoxicity compared with the parent molecule. However, the detailed mechanisms remain unclear. In this study, compounds 1 and 2 were identified as the G2/M cell cycle arrest agents to mediate the cytotoxicity toward NCI-H460 cells via Michael acceptor-dependent redox intervention. Compared with curcumin, they could more easily induce a burst of reactive oxygen species (ROS) and collapse of the redox buffering system. One possible reason is that they could more effectively target intracellular TrxR to convert this antioxidant enzyme into a ROS promoter. Additionally, they caused up-regulation of p53 and p21 and down-regulation of redox-sensitive Cdc25C along with cyclin B1/Cdk1 in a Michael acceptor- and ROS-dependent fashion. Interestingly, in comparison with compound 2, compound 1 displayed a relatively weak ability to generate ROS but increased cell cycle arrest activity and cytotoxicity probably due to its Michael acceptor-dependent microtubule-destabilizing effect and greater GST-inhibitory activity, as well as its enhanced cellular uptake. This work provides useful information for understanding Michael acceptor-dependent and redox-mediated cytotoxic mechanisms of curcumin and its active analogues.
Collapse
Affiliation(s)
- Yan Li
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University , Lanzhou, Gansu 730000, China
- Zhejiang Key Laboratory of Alternative Technologies for Fine Chemicals Process, Institute of Applied Chemistry, Shaoxing University , Shaoxing, Zhejiang 312000, China
| | - Li-Ping Zhang
- Gansu Provincial Hosipital , Lanzhou, Gansu 730000, China
| | - Fang Dai
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University , Lanzhou, Gansu 730000, China
| | - Wen-Jing Yan
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University , Lanzhou, Gansu 730000, China
| | - Hai-Bo Wang
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University , Lanzhou, Gansu 730000, China
| | - Zhi-Shan Tu
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University , Lanzhou, Gansu 730000, China
| | - Bo Zhou
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University , Lanzhou, Gansu 730000, China
| |
Collapse
|