1
|
Guo J, Yang L, Dai L, Ma Q, Yan J, Xie Q, Wu Y, Dai H, Zhao Y. Neuroprotective and antidiabetic lanostane-type triterpenoids from the fruiting bodies of Ganoderma theaecolum. Chin J Nat Med 2025; 23:245-256. [PMID: 39986700 DOI: 10.1016/s1875-5364(25)60828-4] [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: 04/04/2024] [Revised: 05/28/2024] [Accepted: 06/17/2024] [Indexed: 02/24/2025]
Abstract
Eight previously undescribed lanostane triterpenoids, including five nortriterpenoids with 26 carbons, ganothenoids A-E (1-5), and three lanostanoids, ganothenoids F-H (6-8), along with 24 known ones (9-32), were isolated from the fruiting bodies of Ganodrma theaecolum. The structures of the novel compounds were elucidated using comprehensive spectroscopic methods, including electronic circular dichroism (ECD) and nuclear magnetic resonance (NMR) calculations. Compounds 1-32 were assessed for their neuroprotective effects against H2O2-induced damage in human neuroblastoma SH-SY5Y cells, as well as their inhibitory activities against protein tyrosine phosphatase 1B (PTP1B) and α-glucosidase. Compound 4 demonstrated the most potent neuroprotective activity against H2O2-induced oxidative stress by suppressing G0/G1 phase cell cycle arrest, reducing reactive oxygen species (ROS) levels, and inhibiting cell apoptosis through modulation of B-cell lymphoma 2 protein (Bcl-2) and Bcl-2 associated X-protein (Bax) protein expression. Compounds 26, 12, and 28 exhibited PTP1B inhibitory activities with IC50 values ranging from 13.92 to 56.94 μmol·L-1, while compound 12 alone displayed significant inhibitory effects on α-glucosidase with an IC50 value of 43.56 μmol·L-1. Additionally, enzyme kinetic analyses and molecular docking simulations were conducted for compounds 26 and 12 with PTP1B and α-glucosidase, respectively.
Collapse
Affiliation(s)
- Jiaocen Guo
- School of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Hainan University, Sanya 572025, China; Key Laboratory of Research and Development of Natural Product from Li Folk Medicine of Hainan Province & National Key Laboratory for Tropical Crop Breeding, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Li Yang
- Key Laboratory of Research and Development of Natural Product from Li Folk Medicine of Hainan Province & National Key Laboratory for Tropical Crop Breeding, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Luting Dai
- Key Laboratory of Research and Development of Natural Product from Li Folk Medicine of Hainan Province & National Key Laboratory for Tropical Crop Breeding, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Qingyun Ma
- Key Laboratory of Research and Development of Natural Product from Li Folk Medicine of Hainan Province & National Key Laboratory for Tropical Crop Breeding, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Jiaoyang Yan
- School of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Hainan University, Sanya 572025, China
| | - Qingyi Xie
- Key Laboratory of Research and Development of Natural Product from Li Folk Medicine of Hainan Province & National Key Laboratory for Tropical Crop Breeding, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Yougen Wu
- School of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Hainan University, Sanya 572025, China.
| | - Haofu Dai
- Key Laboratory of Research and Development of Natural Product from Li Folk Medicine of Hainan Province & National Key Laboratory for Tropical Crop Breeding, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China.
| | - Youxing Zhao
- Key Laboratory of Research and Development of Natural Product from Li Folk Medicine of Hainan Province & National Key Laboratory for Tropical Crop Breeding, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China.
| |
Collapse
|
2
|
Lin L, Liu Y, Tang R, Ding S, Lin H, Li H. Evodiamine: A Extremely Potential Drug Development Candidate of Alkaloids from Evodia rutaecarpa. Int J Nanomedicine 2024; 19:9843-9870. [PMID: 39345907 PMCID: PMC11430234 DOI: 10.2147/ijn.s459510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 07/23/2024] [Indexed: 10/01/2024] Open
Abstract
Evodiamine (EVO) is a tryptamine indole alkaloid and the main active ingredient in Evodia rutaecarpa. In recent years, the antitumor, cardioprotective, anti-inflammatory, and anti-Alzheimer's disease effects of EVO have been reported. EVO exerts antitumor effects by inhibiting tumor cell activity and proliferation, blocking the cell cycle, promoting apoptosis and autophagy, and inhibiting the formation of the tumor microvasculature. However, EVO has poor solubility and low bioavailability. Several derivatives with high antitumor activity have been discovered through the structural optimization of EVO, and new drug delivery systems have been developed to improve the solubility and bioavailability of EVO. Current research found that EVO could have toxic effects, such as hepatotoxicity, nephrotoxicity, and cardiac toxicity. This article reviews the pharmacological activity, derivatives, drug delivery systems, toxicity, and pharmacokinetics of EVO and provides research ideas and references for its further in-depth development and clinical applications.
Collapse
Affiliation(s)
- Longfei Lin
- Institute Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Yuling Liu
- Institute Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Ruying Tang
- Institute Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Shilan Ding
- Institute Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Hongmei Lin
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People's Republic of China
- National Medical Products Administration Key Laboratory for Research Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Hui Li
- Institute Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
- Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, People's Republic of China
| |
Collapse
|
3
|
Chen S, Bi K, Liang H, Wu Z, Huang M, Chen X, Dong G, Sheng C. PROTAC derivatization of natural products for target identification and drug discovery: Design of evodiamine-based PROTACs as novel REXO4 degraders. J Adv Res 2024; 63:219-230. [PMID: 37913903 PMCID: PMC11380026 DOI: 10.1016/j.jare.2023.10.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/18/2023] [Accepted: 10/28/2023] [Indexed: 11/03/2023] Open
Abstract
INTRODUCTION Natural products (NPs) play a crucial role in the development of therapeutic drugs. However, it is still highly challenging to identify the targets of NPs. Besides, NPs usually exert their pharmacological activities via acting on multiple targets or pathways, which also poses great difficulties for the target identification of NPs. OBJECTIVES Inspired by our continuous efforts in designing drug-like protein degraders, this study introduced a successful example for the target identification and drug discovery of natural products evodiamine by employing PROTAC technology. METHODS Taking advantages of proteolysis targeting chimera (PROTAC), herein an integrated strategy combining PROTAC derivatization, quantitative proteomic analysis and binding affinity validation was developed for target identification and drug discovery of antitumor NP evodiamine. RESULTS In this study, both highly potent PROTACs and negative controls were designed for quantitative proteomic analysis. Furthermore, REXO4 was confirmed as a direct target of 3-fluoro-10-hydroxylevodiamine, which induced cell death through ROS. In addition, the PROTAC 13c effectively degraded REXO4 both in vitro and in vivo, leading to potent antitumor activities and reduced toxic side effects. CONCLUSION In summary, we developed an integrated strategy for the target identification and drug discovery of NPs, which was successfully applied to the PROTAC derivatization and target characterization of evodiamine. This proof-of-concept study highlighted the superiority of PROTAC technology in target identification of NPs and accelerated the process of NPs-based drug discovery, exhibiting broad application in NP-based drug development.
Collapse
Affiliation(s)
- Shuqiang Chen
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai 200433, People's Republic of China.
| | - Kaijian Bi
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai 200433, People's Republic of China
| | - Huixin Liang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai 200433, People's Republic of China
| | - Zhe Wu
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai 200433, People's Republic of China
| | - Min Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
| | - Xi Chen
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, People's Republic of China
| | - Guoqiang Dong
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai 200433, People's Republic of China
| | - Chunquan Sheng
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai 200433, People's Republic of China.
| |
Collapse
|
4
|
Wu Z, Chen S, Chen Z, Dong G, Xu D, Sheng C. Design of Evodiamine-Glucose Conjugates with Improved In Vivo Antitumor Activity. J Med Chem 2024; 67:7373-7384. [PMID: 38646851 DOI: 10.1021/acs.jmedchem.4c00221] [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: 04/23/2024]
Abstract
Natural product evodiamine is a multitargeting antitumor lead compound. However, clinical development of evodiamine derivatives was hampered by poor water solubility and limited in vivo antitumor potency. Herein, a series of evodiamine-glucose conjugates were designed by additional targeting glucose transporter-1 (GLUT1). Compared with the lead compound, conjugate 8 exhibited obvious enhancement in water solubility and in vivo antitumor efficacy. Furthermore, the effect of GLUT1 targeting also led to lower cytotoxicity to normal cells. Antitumor mechanism studies manifested that conjugate 8 acted by Top1/Top2 dual inhibition, apoptosis induction, and G2/M cell cycle arrest, which selectively targeted tumor cells with a high expression level of GLUT1. Thus, evodiamine-glucose conjugates showed promising features as potential antitumor agents.
Collapse
Affiliation(s)
- Zhe Wu
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou 213164, People's Republic of China
| | - Shuqiang Chen
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai 200433, People's Republic of China
| | - Zhipeng Chen
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai 200433, People's Republic of China
| | - Guoqiang Dong
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai 200433, People's Republic of China
| | - Defeng Xu
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou 213164, People's Republic of China
| | - Chunquan Sheng
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai 200433, People's Republic of China
| |
Collapse
|
5
|
Gao X, Li HN, Liu PJ, Long XK, Guo XH, Hua HM, Li DH. Synthesis of sinomenine derivatives with potential anti-leukemia activity. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024:1-17. [PMID: 38572941 DOI: 10.1080/10286020.2024.2327524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/04/2024] [Indexed: 04/05/2024]
Abstract
In recent years, with sinomenine hydrochloride as the main ingredient, Qingfengteng had been formulated as various dosage forms for clinical treatment. Subsequent findings confirmed a variety of biological roles for sinomenine. Here, 15 H2S-donating sinomenine derivatives were synthesized. Target hybrids a11 displayed substantial cytotoxic effects on cancer cell lines, particularly against K562 cells, with an IC50 value of 1.36 μM. In-depth studies demonstrated that a11 arrested cell cycle at G1 phase, induced apoptosis via both morphological changes in nucleus and membrane potential collapse in mitochondria. These results indicated a11 exerted an antiproliferative effect through apoptosis induction via mitochondrial pathway.
Collapse
Affiliation(s)
- Xiang Gao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hao-Nan Li
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Peng-Ju Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiao-Kang Long
- Department of Pharmacy, The First Affiliated Hospital of Jishou University, Jishou 416000, China
| | - Xue-Hai Guo
- Huangshi Food and Drug Inspection and Testing Center, Huangshi 435000, China
| | - Hui-Ming Hua
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Da-Hong Li
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
6
|
Li H, Bu L, Sun X, Chu X, Xue Y, Zhang M, Shi J, Liu Y, Guan S, Han X, Wang H. Mechanistic investigation of the ameliorative effect of liquiritin on hypoxia/reoxygenation‑induced cardiomyocyte injury based on network pharmacology and in vitro validation. Exp Ther Med 2024; 27:117. [PMID: 38361515 PMCID: PMC10867724 DOI: 10.3892/etm.2024.12405] [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: 09/10/2023] [Accepted: 12/18/2023] [Indexed: 02/17/2024] Open
Abstract
Liquiritin (LIQ) is a flavonoid known for its cardioprotective properties, extracted from Glycyrrhiza uralensis Fisch. The purpose of the present study was to investigate the protective mechanism of LIQ against hypoxia/reoxygenation (H/R) injury through in vitro experiments, with the goal of enhancing its pharmacological effects. Initially, network pharmacology was employed to explore the targets and mechanisms of LIQ. Subsequently, an in vitro H/R model was established using H9c2 cells. Potential targets for LIQ and myocardial ischemia-reperfusion injury (MIRI) were identified through online databases. The STRING, Cytoscape and DAVID databases were used to extract intersecting targets and mechanisms. In vitro experiments were conducted to validate these findings, assessing cardiac enzymes, oxidative stress indicators, mitochondrial fluorescence, apoptotic fluorescence, inflammation and related protein expression. The network pharmacological analysis revealed that the protective effects of LIQ on MIRI involve oxidative stress, inflammation and apoptosis. The results of in vitro experimental validation demonstrated that LIQ significantly reduced the activities of lactated dehydrogenase and creatine kinase isoenzyme-MB (P<0.05 or 0.01), as well as the level of malondialdehyde (P<0.01). It also inhibited the production of reactive oxygen species (P<0.01), the release of inflammatory factors (P<0.05 or 0.01) and apoptosis (P<0.01). By contrast, the LIQ pre-treatment group exhibited a significant increase in mitochondrial membrane potential level (P<0.05 or 0.01) and the activities of antioxidant enzymes superoxide dismutase, catalase and glutathione peroxidase (P<0.05 or 0.01). Furthermore, LIQ reduced the protein expressions of TNF-α receptor type 1 (TNFR1) and MMP9, along with the level of NF-κB phosphorylation (P<0.05 or 0.01). In conclusion, LIQ mitigated H/R-induced cardiomyocyte injury through mechanisms that may involve antioxidants, anti-apoptotic effects, protection against mitochondrial damage and suppression of inflammatory levels. These effects are achieved via inhibition of the TNFR1/NF-κB/MMP9 pathway.
Collapse
Affiliation(s)
- Haoying Li
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Linlin Bu
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Xiaoqi Sun
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Xi Chu
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Yucong Xue
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Muqing Zhang
- Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050000, P.R. China
| | - Jing Shi
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Yanshuang Liu
- College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
- College of Integrative Medicine, Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Shengjiang Guan
- Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050000, P.R. China
| | - Xue Han
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Hongfang Wang
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| |
Collapse
|
7
|
Wei B, Ma J, Guo H, Wang Y, Guo D, Tang Y. Design, synthesis and bioactivity evaluation of the combination of evodiamine and erlotinib linked by indolequinone. Bioorg Med Chem Lett 2024; 99:129619. [PMID: 38244939 DOI: 10.1016/j.bmcl.2024.129619] [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: 07/04/2023] [Revised: 12/16/2023] [Accepted: 01/14/2024] [Indexed: 01/22/2024]
Abstract
Compared with single-targeted therapy, the design and synthesis of heterozygous molecules is still a significant challenge for the discovery of antitumor drugs. Quinone oxidoreductase-1 (NQO1) is a potential target for selective cancer therapy due to its overexpression in many cancer cells and its unique bioredox properties. Based on the principle of combinatorial drug design, we successfully synthesized a new hybrid molecules 13 with an indolequinone structure. We found that the synthesized compounds exhibited much higher cytotoxicity against the tested cancer cells than free drugs. Further mechanism studies confirmed that compound 13 induced cell apoptosis was achieved by regulating p53-dependent mitochondrial pathway and cell cycle arrest at the G0/G1 phase.
Collapse
Affiliation(s)
- Binbin Wei
- College of Pharmacy, Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, China
| | - Jingjing Ma
- College of Pharmacy, Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, China
| | - Hui Guo
- College of Pharmacy, Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, China.
| | - Yuwei Wang
- College of Pharmacy, Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, China
| | - Dongyan Guo
- College of Pharmacy, Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, China
| | - Yuping Tang
- College of Pharmacy, Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, China.
| |
Collapse
|
8
|
Panthalattu Parambil A, Shamjith S, Kurian J, Kesavan A, Sen AK, Thangaraj PR, Maiti KK, Manheri MK. A dual mode 'turn-on' fluorescence-Raman (SERS) response probe based on a 1 H-pyrrol-3(2 H)-one scaffold for monitoring H 2S levels in biological samples. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:2853-2860. [PMID: 37260380 DOI: 10.1039/d3ay00282a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Discovery of the biological signaling roles of H2S has spurred great interest in developing reliable methods for its accurate detection and quantification. As considerable variation in its levels is seen during pathological conditions such as sepsis, real-time quantification methods have relevance in diagnosis as well. Of various approaches, reaction-based probes which respond through 'off-on' fluorescence emission remain the most studied. Since the intensity of emission is related to the analyte concentration in these measurements, the presence of built-in features which provide an opportunity for internal referencing will be advantageous. In view of this, a dual mode response system that senses H2S through characteristic fluorescence and Raman (SERS) signals based on a 1H-pyrrol-3(2H)-one scaffold was developed and is the main highlight of this report. This probe offers several advantages such as fast response (<1 min), and high selectivity and sensitivity with a detection limit of ∼7 nM. Imaging of H2S in HepG2 cells, making use of the SERS signal from the thiolysis product is also demonstrated.
Collapse
Affiliation(s)
| | - Shanmughan Shamjith
- CSIR-National Institute for Interdisciplinary Science and Technology, CSTD, Organic Chemistry Section, Industrial Estate P.O., Thiruvananthapuram 695019, Kerala, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Jais Kurian
- Department of Chemistry, Indian Institute of Technology Madras, Chennai-600036, Tamil Nadu, India.
| | - Akila Kesavan
- Department of Chemistry, Indian Institute of Technology Madras, Chennai-600036, Tamil Nadu, India.
| | - Ashis K Sen
- Department of Mechanical Engineering, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Paul R Thangaraj
- Department of Cardiothoracic and Transplant Surgery, Apollo Hospitals, Chennai, India
- Adjunct Faculty, Department of Mechanical Engineering, IIT-Madras, Chennai, 600036, India
| | - Kaustabh Kumar Maiti
- CSIR-National Institute for Interdisciplinary Science and Technology, CSTD, Organic Chemistry Section, Industrial Estate P.O., Thiruvananthapuram 695019, Kerala, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Muraleedharan K Manheri
- Department of Chemistry, Indian Institute of Technology Madras, Chennai-600036, Tamil Nadu, India.
| |
Collapse
|
9
|
Wang Z, Xiong Y, Peng Y, Zhang X, Li S, Peng Y, Peng X, Zhuo L, Jiang W. Natural product evodiamine-inspired medicinal chemistry: Anticancer activity, structural optimization and structure-activity relationship. Eur J Med Chem 2023; 247:115031. [PMID: 36549115 DOI: 10.1016/j.ejmech.2022.115031] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/06/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022]
Abstract
It is a well-known phenomenon that natural products can serve as powerful drug leads to generate new molecular entities with novel therapeutic utility. Evodiamine (Evo), a major alkaloid component in traditional Chinese medicine Evodiae Fructus, is considered a desirable lead scaffold as its multifunctional pharmacological properties. Although natural Evo has suboptimal biological activity, poor pharmacokinetics, low water solubility, and chemical instability, medicinal chemists have succeeded in producing synthetic analogs that overshadow the deficiency of Evo in terms of further clinical application. Recently, several reviews on the synthesis, structural modification, mechanism pharmacological actions, structure-activity relationship (SAR) of Evo have been published, while few reviews that incorporates intensive structural basis and extensive SAR are reported. The purpose of this article is to review the structural basis, anti-cancer activities, and mechanisms of Evo and its derivatives. Emphasis will be placed on the optimizing strategies to improve the anticancer activities, such as structural modifications, pharmacophore combination and drug delivery systems. The current review would benefit further structural modifications of Evo to discover novel anticancer drugs.
Collapse
Affiliation(s)
- Zhen Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Yongxia Xiong
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Ying Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xi Zhang
- School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Shuang Li
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yan Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xue Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Linsheng Zhuo
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Weifan Jiang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
10
|
Lin H, Yu Y, Zhu L, Lai N, Zhang L, Guo Y, Lin X, Yang D, Ren N, Zhu Z, Dong Q. Implications of hydrogen sulfide in colorectal cancer: Mechanistic insights and diagnostic and therapeutic strategies. Redox Biol 2023; 59:102601. [PMID: 36630819 PMCID: PMC9841368 DOI: 10.1016/j.redox.2023.102601] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/01/2023] [Accepted: 01/02/2023] [Indexed: 01/09/2023] Open
Abstract
Hydrogen sulfide (H2S) is an important signaling molecule in colorectal cancer (CRC). It is produced in the colon by the catalytic synthesis of the colonocytes' enzymatic systems and the release of intestinal microbes, and is oxidatively metabolized in the colonocytes' mitochondria. Both endogenous H2S in colonic epithelial cells and exogenous H2S in intestinal lumen contribute to the onset and progression of CRC. The up-regulation of endogenous synthetases is thought to be the cause of the elevated H2S levels in CRC cells. Different diagnostic probes and combination therapies, as well as tumor treatment approaches through H2S modulation, have been developed in recent years and have become active area of investigation for the diagnosis and treatment of CRC. In this review, we focus on the specific mechanisms of H2S production and oxidative metabolism as well as the function of H2S in the occurrence, progression, diagnosis, and treatment of CRC. We also discuss the present challenges and provide insights into the future research of this burgeoning field.
Collapse
Affiliation(s)
- Hanchao Lin
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, China; Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, China
| | - Yixin Yu
- College of Materials Science and Engineering, Qingdao University of Science and Technology, China
| | - Le Zhu
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, China
| | - Nannan Lai
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, China
| | - Luming Zhang
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, China
| | - Yu Guo
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, China
| | - Xinxin Lin
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, China
| | - Dongqin Yang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, China.
| | - Ning Ren
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, China; Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, And Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, China.
| | - Zhiling Zhu
- College of Materials Science and Engineering, Qingdao University of Science and Technology, China.
| | - Qiongzhu Dong
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, China.
| |
Collapse
|
11
|
Wang Y, Ma H, Narula A, Liu L, Ahn KS. Molecular targets and anticancer potential of evodiamine. PHYTOCHEMISTRY LETTERS 2022; 52:92-103. [DOI: 10.1016/j.phytol.2022.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
12
|
Lu X, Zhu C, Zhang C, Li X, Yu Z, Zhang Z, Shi X. Design, synthesis and biological evaluation of 3-aryl-7-hydroxy scopoletin derivatives as autophagy activators against tumorigenesis. Eur J Med Chem 2022; 244:114805. [DOI: 10.1016/j.ejmech.2022.114805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022]
|
13
|
Liang H, Wang W, Zhu F, Chen S, Liu D, Sheng C. Discovery of novel bis-evodiamine derivatives with potent antitumor activity. Bioorg Med Chem 2022; 65:116793. [PMID: 35550978 DOI: 10.1016/j.bmc.2022.116793] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/19/2022] [Accepted: 04/30/2022] [Indexed: 11/27/2022]
Abstract
Inspired by antitumor natural product evodiamine, a series of novel bis-evodiamine derivatives were designed and synthesized, which showed potent antitumor activity. In particular, compound 13b effectively inhibited the proliferation and migration of HCT116 cells. Further mechanism studies revealed that compound 13b acted by inducing HCT116 cell apoptosis and arresting the cell cycle at the G2/M phase. Thus, compound 13b represents a promising lead compound for the discovery of novel antitumor agents.
Collapse
Affiliation(s)
- Huixin Liang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China; Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, PR China
| | - Wei Wang
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, PR China
| | - Fugui Zhu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China; Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, PR China
| | - Shuqiang Chen
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, PR China.
| | - Dan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China.
| | - Chunquan Sheng
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China; Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, PR China.
| |
Collapse
|
14
|
Wu H, Li X, He C, Liu Y, Wang C, Yang X, Ma F, Liu J, Xu J. Design, synthesis and biological evaluation of hydrogen sulfide-releasing isochroman-4-one derivatives as new antihypertensive agent candidates. Bioorg Med Chem 2022; 64:116776. [PMID: 35483137 DOI: 10.1016/j.bmc.2022.116776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 04/12/2022] [Accepted: 04/20/2022] [Indexed: 11/27/2022]
Abstract
Cardiovascular diseases are increasingly threating the global human health, hypertension is the most important risk factor for cardiovascular and cerebrovascular diseases. To improve the antihypertensive activity and cardiovascular protective effect of natural product (±)-7,8-dihydroxy-3-methyl-isochroman-4-one [(±)-XJP], a series of novel H2S-releasing isochroman-4-one derivatives were designed and synthesized by coupling hydrogen sulfide (H2S)-releasing donors with the analogs of (±)-XJP. Further, the H2S-releasing assay indicated that some target compounds showed excellent H2S generating ability. Moreover, these novel hybrids exhibited moderate to good in vitro vasodilation efficacy. Among them, the most potent compound exhibited potent in vivo antihypertensive activity with the maximum antihypertensive amplitude about 27%, which was more potent than that of the lead compound (±)-XJP. These results suggested that the hybridization of H2S-donors and (±)-XJP analogs may provide a promising approach for the discovery of novel antihypertensive agents.
Collapse
Affiliation(s)
- Hongyu Wu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China
| | - Xinnan Li
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China
| | - Chen He
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China
| | - Yulin Liu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China; Department of Organic Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China
| | - Chaolei Wang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China
| | - Xue Yang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China
| | - Fenfen Ma
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan Univerisity, Shanghai 201399, PR China
| | - Jie Liu
- Department of Organic Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China.
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China.
| |
Collapse
|
15
|
Liu J, Shi Y, Tian Z, Li F, Hao Z, Wen W, Zhang L, Wang Y, Li Y, Fan Z. Bioactivity-Guided Synthesis Accelerates the Discovery of Evodiamine Derivatives as Potent Insecticide Candidates. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:5197-5206. [PMID: 35435667 DOI: 10.1021/acs.jafc.1c08297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Pests threaten worldwide food security by decreasing crop yields and damaging their quality. Natural product-based molecular design and structural optimization have been one of the most effective ways to innovate pesticides for integrated insect management. To continue our previous studies on the discovery of insecticidal lead, a series of evodiamine derivatives were designed, synthesized, and evaluated for their insecticidal activities. The bioassay results demonstrated that compounds Ian and Iao exhibited 90 and 80% insecticidal activities against Mythimna separata at 2.5 mg/L, respectively, which were superior to evodiamine (10% at 10 mg/L), matrine (45% at 600 mg/L), and rotenone (30% at 200 mg/L). Compounds Ian-Iap showed 90% insecticidal activities against Plutella xylostella at 1.0 mg/L, far more potent than those of evodiamine, matrine, and rotenone. Compound Ian displayed 60% insecticidal activity against Helicoverpa armigera at 5.0 mg/L, while evodiamine, matrine, and rotenone showed very poor activities. The study on the insecticidal mechanism of action by a calcium imaging experiment indicated that the insect ryanodine receptors (RyRs) could be the potential target of Ian. Furthermore, the molecular docking indicated that Ian anchored in the binding site of the RyR of P. xylostella. The above results manifested the potential of evodiamine derivatives as potent insecticide candidates.
Collapse
Affiliation(s)
- Jingbo Liu
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin 300384, P. R. China
| | - Yabing Shi
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin 300384, P. R. China
| | - Zhicheng Tian
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin 300384, P. R. China
| | - Fengyun Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P. R. China
| | - Zesheng Hao
- State Key Laboratory of Elemento-Organic Chemistry, College of Chemistry, Nankai University, Tianjin 300071, P. R. China
| | - Wen Wen
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin 300384, P. R. China
| | - Li Zhang
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin 300384, P. R. China
| | - Yuanhong Wang
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin 300384, P. R. China
| | - Yuxin Li
- State Key Laboratory of Elemento-Organic Chemistry, College of Chemistry, Nankai University, Tianjin 300071, P. R. China
| | - Zhijin Fan
- State Key Laboratory of Elemento-Organic Chemistry, College of Chemistry, Nankai University, Tianjin 300071, P. R. China
- Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, P. R. China
| |
Collapse
|
16
|
Wang W, Li C, Chen Z, Zhang J, Ma L, Tian Y, Ma Y, Guo L, Wang X, Ye J, Wang X. Novel diosgenin-amino acid-benzoic acid mustard trihybrids exert antitumor effects via cell cycle arrest and apoptosis. J Steroid Biochem Mol Biol 2022; 216:106038. [PMID: 34861390 DOI: 10.1016/j.jsbmb.2021.106038] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/16/2021] [Accepted: 11/29/2021] [Indexed: 12/14/2022]
Abstract
In discovering new powerful antitumor agents, two series of novel diosgenin-amino acid-benzoic acid mustard trihybrids (7a-7 g and 12a-12 g) were designed and synthesized. The antiproliferative activities were tested against five human tumor cell lines and one normal cell line using CCK-8 assays. Among the trihybrids, 12e was the most promising compound, which inhibited T24 cells with IC50 value of 6.96 μM, and was stronger than its parent compound diosgenin (IC50 = 32.33 μM). In addition, 12e had weak cytotoxicity on the normal GES-1 cell line (IC50 = 213.74 μM). Moreover, 12e could cause G2/M cell cycle arrest, increase the percentage of apoptosis, induce mitochondrial depolarization, and promote reactive oxygen species generation in T24 cells. Further studies on antitumor mechanism demonstrated that 12e triggered the intrinsic (mitochondrial) and extrinsic (death receptor) apoptotic pathways. More importantly, 12e could inhibit T24 cell proliferation in an in vivo zebrafish xenograft model. Therefore, 12e, as a novel trihybrid with potent cytotoxicity, might be applied as a promising skeleton for antitumor agents, which deserved further optimization.
Collapse
Affiliation(s)
- Wenbao Wang
- Qiqihar Medical University, Qiqihar, 161006 Heilongjiang, PR China; Chinese People's Liberation Army Logistics Support Force No. 967 Hospital, Dalian, 116021, PR China.
| | - Chuan Li
- General Hospital of Northern Theater Command, Shenyang, 110016, PR China
| | - Zhe Chen
- Qiqihar Medical University, Qiqihar, 161006 Heilongjiang, PR China
| | - Jinling Zhang
- Qiqihar Medical University, Qiqihar, 161006 Heilongjiang, PR China
| | - Liwei Ma
- Qiqihar Medical University, Qiqihar, 161006 Heilongjiang, PR China
| | - Yanzhao Tian
- Chinese People's Liberation Army Logistics Support Force No. 967 Hospital, Dalian, 116021, PR China
| | - Yukun Ma
- Qiqihar Medical University, Qiqihar, 161006 Heilongjiang, PR China
| | - Lina Guo
- Qiqihar Medical University, Qiqihar, 161006 Heilongjiang, PR China
| | - Xiaoli Wang
- Qiqihar Medical University, Qiqihar, 161006 Heilongjiang, PR China
| | - Jin Ye
- Qiqihar Medical University, Qiqihar, 161006 Heilongjiang, PR China
| | - Xiaobo Wang
- Chinese People's Liberation Army Logistics Support Force No. 967 Hospital, Dalian, 116021, PR China.
| |
Collapse
|
17
|
Hao X, Deng J, Zhang H, Liang Z, Lei F, Wang Y, Yang X, Wang Z. Design, synthesis and bioactivity evaluation of novel N-phenyl-substituted evodiamine derivatives as potent anti-tumor agents. Bioorg Med Chem 2022; 55:116595. [PMID: 34990980 DOI: 10.1016/j.bmc.2021.116595] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/17/2021] [Accepted: 12/27/2021] [Indexed: 02/09/2023]
Abstract
Natural products are important sources for the development of therapeutic medicine, among which evodia fruit has a wide range of medicinal properties in traditional Chinese medicine. Evodiamine, the main active component of evodia fruit, has various anti-cancer effects and has been proved to be a Topo inhibitor. From our previous attempts of modifying evodiamine, we found that the N14 phenyl substituted derivatives had showed great anti-tumor activity, which prompted us to further explore the novel structures and activities of these compounds. Compound 6f, as a N14 3-fluorinated phenyl substituted evodiamine derivative, showed a certain inhibitory activity against Topo I at 200 μM. By studying its anti-tumor effects in vitro, compound 6f could inhibit proliferation and induce apoptosis, as well as arrest the cell cycle of HGC-27 and HT-29 cell lines at G2/M phase in a concentration-dependent manner. Moreover, compound 6f could inhibit the migration and invasion of HGC-27 cell lines. Meanwhile, compound 6f could induce apoptosis of HGC-27 cells by inhibiting PI3K/AKT pathway. Overall, this work demonstrated that the N14 phenyl-substituted evodiamine derivatives had a good inhibitory effect on tumor cells in vitro, providing a promising strategy for developing potential anticancer agents for the treatment of gastrointestinal tumors.
Collapse
Affiliation(s)
- Xiangyong Hao
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou 730000, China
| | - Jiedan Deng
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Honghua Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China.
| | - Ziyi Liang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Fang Lei
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Yuqing Wang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Xiaoyan Yang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - Zhen Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; School of Pharmacy, Lanzhou University, Lanzhou 730000, China; State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
18
|
Research Advances in Antitumor Mechanism of Evodiamine. J CHEM-NY 2022. [DOI: 10.1155/2022/2784257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Evodiamine is a natural alkaloid extracted from Fructus Evodia. This bioactive alkaloid has been reported to have a wide range of biological activities, including anti-injury, antiobesity, vasodilator, and anti-inflammatory effects. In recent years, it has been found that evodiamine has tumor-suppressive effects on a variety of tumors. There is growing evidence that evodiamine can inhibit the rapid proliferation of tumor cells, induce cell cycle arrest at a certain phase, increase the incidence of apoptosis, promote autophagy, inhibit microangiogenesis and migration, and regulate immunotherapy. Evodiamine can inhibit Wnt/β-catenin, mTOR, NF-κB, PI3K/AKT, JAK-STAT, and other signaling pathways in various cancer cells, and it can significantly downregulate the expression of many tumor markers, such as VEGF and COX-2. These facts partially explain the antitumor mechanism of evodiamine. In this article, the antitumor mechanism of evodiamine was reviewed to provide the basis for its clinical application and therapeutic development in the future.
Collapse
|
19
|
Fan M, Yao L. The Synthesis, Structural Modification and Mode of Anticancer Action of Evodiamine: a review. Recent Pat Anticancer Drug Discov 2021; 17:284-296. [PMID: 34939550 DOI: 10.2174/1574892817666211221165739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/29/2021] [Accepted: 11/12/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Finding novel antitumor reagents from naturally occurring alkaloids is a widely accepted strategy. Evodiamine, a tryptamine indole alkaloid isolated from Evodia rutaecarpa, has a wide range of biological activities, such as antitumor, anti-inflammation, and anti-bacteria. Hence, research works on the structural modification of evodiamine will facilitate the discovery of new antitumor drugs. OBJECTIVE The recent advances in the synthesis of evodiamine, and studies on the drug design, biological activities, and structure-activity-relationships of its derivatives, published in patents and primary literatures, are reviewed in this paper. METHODS The literatures, including patents and follow-up research papers from 2015 to 2020, related to evodiamine is searched in the Scifinder, PubMed, Espacenet, China National Knowledge Infrastructure (CNKI), and Wanfang databases. The key words are evodiamine, synthesis, modification, anticancer, mechanism. RESULTS The synthesis of evodiamine are summarized. Then, structural modifications of evodiamine are described, and the possible modes of actions are discussed. CONCLUSION Evodiamine has a 6/5/6/6/6 ring system, and the structural modifications are focused on ring A, D, E, C5, N-13, and N-14. Some compounds show promising anticancer potentials and warrant further study.
Collapse
Affiliation(s)
- Meixia Fan
- School of Pharmacy, Yantai University, 30 Qingquan Road, Yantai, 264005, Shandong. China
| | - Lei Yao
- School of Pharmacy, Yantai University, 30 Qingquan Road, Yantai, 264005, Shandong. China
| |
Collapse
|
20
|
Xu S, Yao H, Qiu Y, Zhou M, Li D, Wu L, Yang DH, Chen ZS, Xu J. Discovery of Novel Polycyclic Heterocyclic Derivatives from Evodiamine for the Potential Treatment of Triple-Negative Breast Cancer. J Med Chem 2021; 64:17346-17365. [PMID: 34844412 DOI: 10.1021/acs.jmedchem.1c01411] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Evodiamine (Evo) is a quinazolinocarboline alkaloid found in Evodia rutaecarpa and exhibits moderate antiproliferative activity. Herein, we report using a scaffold-hopping approach to identify a series of novel polycyclic heterocyclic derivatives based on Evo as the topoisomerase I (Top1) inhibitor for the treatment of triple-negative breast cancer (TNBC), which is an aggressive subtype of breast cancer with limited treatment options. The most potent compound 7f inhibited cell growth in a human breast carcinoma cell line (MDA-MB-231) with an IC50 value of 0.36 μM. Further studies revealed that Top1 was the target of 7f, which directly induced irreversible Top1-DNA covalent complex formation or induced an oxidative DNA lesion through an indirect mechanism mediated by reactive oxygen species. More importantly, in vivo studies showed that 7f exhibited potent antitumor activity in a TNBC-patient-derived tumor xenograft model. These results suggest that compound 7f deserves further investigation as a promising candidate for the treatment of TNBC.
Collapse
Affiliation(s)
- Shengtao Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Hong Yao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Yangyi Qiu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China.,Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, P. R. China
| | - Manzhen Zhou
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Dahong Li
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China.,Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, P. R. China
| | - Liang Wu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Dong-Hua Yang
- College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, New York 11439, United States
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, New York 11439, United States
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| |
Collapse
|
21
|
Patrykei S, Korobko Y, Ogorodniichuk O, Garazd M, Polishchuk P, Džubák P, Gurská S, Hajdúch M, Lesyk R. Synthesis and evaluation of the anticancer activity of some semisynthetic derivatives of rutaecarpine and evodiamine. SYNTHETIC COMMUN 2021. [DOI: 10.1080/00397911.2021.1919712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
| | | | | | | | - Pavel Polishchuk
- Institute of Molecular and Translational Medicine (IMTM), Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Petr Džubák
- Institute of Molecular and Translational Medicine (IMTM), Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Soňa Gurská
- Institute of Molecular and Translational Medicine (IMTM), Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine (IMTM), Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Roman Lesyk
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
- Department of Public Health, Dietetics and Lifestyle Disorders, Faculty of Medicine, University of Information Technology and Management in Rzeszow, Rzeszow, Poland
| |
Collapse
|
22
|
Bai H, Gu RJ, Chen LY, Qian Y, Yu ML, Xu SL, Xia XF, Liu YC, Zhang HR, Gu YH, Lu SF. Electroacupuncture interventions alleviates myocardial ischemia reperfusion injury through regulating gut microbiota in rats. Microvasc Res 2021; 138:104235. [PMID: 34453991 DOI: 10.1016/j.mvr.2021.104235] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/10/2021] [Accepted: 08/20/2021] [Indexed: 12/21/2022]
Abstract
Electroacupuncture (EA) intervention has a remarkable cardioprotection against myocardial ischemia reperfusion injury (MIRI). Recently, it has been suggested that the gut microbiota plays an important role in regulating the progression and prognosis of MIRI. The purpose of this study was to illustrate the relationship between gut microbiota and cardioprotection of EA on MIRI. We conducted a MIRI model by ligating the left anterior descending coronary artery for 30 min followed by reperfusion in male Sprague Dawley rats, which then received 7 days of EA intervention. Echocardiography was employed to evaluate left ventricular function. Fecal samples were collected for microbial analysis by 16S rDNA high-throughput sequencing. Blood samples and myocardium were collected for inflammatory cytokine detection by enzyme linked immunosorbent assay (ELISA) and Western blot. Hematoxylin & eosin (HE) staining and immunofluorescence of ileum tissue were performed for intestinal damage evaluation. After 7 days of EA intervention, the left ventricular function was improved with significantly increased ejection fraction and fractional shortening. Furthermore, we found that EA intervention reversed the changed gut microbiota induced by MIRI, including Clostridiales, RF39, S24-7, Desulfovibrio, and Allobaculum, improved the impaired gut barrier, reduced the production and circulation of lipopolysaccharide (LPS), inhibited the level of interleukin 6 (IL-6) and interleukin 12 (IL-12) in periphery and decreased the expression of Toll like receptor 4 (TLR4) and IL-6 in myocardium. EA intervention could improve the impaired gut mucosal barrier and reduce the production and circulation of LPS after MIRI through regulating gut microbiota, thus inhibiting the circulation and myocardium inflammation and finally exerted the cardioprotective effect.
Collapse
Affiliation(s)
- Hua Bai
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ren-Jun Gu
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Li-Yao Chen
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yi Qian
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mei-Ling Yu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Sen-Lei Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xue-Feng Xia
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yu-Chen Liu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hong-Ru Zhang
- School of Basic Medical Sciences, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yi-Huang Gu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Sheng-Feng Lu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
23
|
Fan X, Deng J, Shi T, Wen H, Li J, Liang Z, Lei F, Liu D, Zhang H, Liang Y, Hao X, Wang Z. Design, synthesis and bioactivity study of evodiamine derivatives as multifunctional agents for the treatment of hepatocellular carcinoma. Bioorg Chem 2021; 114:105154. [PMID: 34378540 DOI: 10.1016/j.bioorg.2021.105154] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/16/2021] [Accepted: 07/04/2021] [Indexed: 12/20/2022]
Abstract
Topoisomerase has been found extremely high level of expression in hepatocellular carcinoma (HCC) and proven to promote the proliferation and survival of HCC. Cancer-associated fibroblasts (CAFs) as a kind of key reactive stromal cell that abundantly present in the microenvironment of HCC, could enhance the metastatic ability and drug resistance of HCC. Therefore, developing new drugs that address the above conundrums would be of the upmost significant in the fight against HCC. Evodiamine, as a multi-target natural product, has been found to exert various biological activities such as anti-cancer and anti-hepatic fibrosis via blocking topoisomerase, NF-κB, TGF-β/HGF, and Smad2/3. Inspired by these facts, 15 evodiamine derivatives were designed and synthesized for HCC treatment by simultaneously targeting Topo I and CAFs. Most of them displayed preferable anti-HCC activities on three HCC cell lines and low cytotoxicity on one normal hepatic cell. In particular, compound 8 showed the best inhibitory effect on HCC cell lines and a good inhibition on Topo I in vitro. Meanwhile, it also induced obvious G2/M arrest and apoptosis, and significantly decreased the migration and invasion capacity of HCC cells. In addition, compound 8 down-regulated the expression of type I collagen in the activated HSC-T6 cells, and induced the apoptosis of activated HSC-T6 cells. In vivo studies demonstrated that compound 8 markedly decreased the volume and weight of tumor (TGI = 40.53%). In vitro and in vivo studies showed that its effects were superior to those of evodiamine. This preliminary attempt may provide a promising strategy for developing anti-HCC lead compounds taking effect through simultaneous inhibition on Topo I and CAFs.
Collapse
Affiliation(s)
- Xiaohong Fan
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jiedan Deng
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Tao Shi
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China.
| | - Huaixiu Wen
- Key Laboratory of Tibetan Medicine Research, Northwest Plateau Institute of Biology, Chinese Academy of Sciences, Xining 810001, China
| | - Junfang Li
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Ziyi Liang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Fang Lei
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Dan Liu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Honghua Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Yan Liang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Xiangyong Hao
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou 730000, China.
| | - Zhen Wang
- School of Pharmaceutical Science, University of South China, Hengyang 421001, China; School of Pharmacy, Lanzhou University, Lanzhou 730000, China; State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
24
|
Li Y, Ma J, Song Z, Zhao Y, Zhang H, Li Y, Xu J, Guo Y. The Antitumor Activity and Mechanism of a Natural Diterpenoid From Casearia graveolens. Front Oncol 2021; 11:688195. [PMID: 34249737 PMCID: PMC8267910 DOI: 10.3389/fonc.2021.688195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/08/2021] [Indexed: 01/26/2023] Open
Abstract
Casearlucin A, a diterpenoid obtained from Casearia graveolens, has been reported to possess strong cytotoxic activity. However, the in vivo anti-tumor effects and the action mechanism of casearlucin A remain poorly understood. Our study revealed that casearlucin A arrested cell cycle at G0/G1 stage and induced cell apoptosis in cell level. Additionally, casearlucin A inhibited HepG2 cell migration via regulating a few of metastasis-related proteins. Furthermore, it inhibited tumor angiogenesis in zebrafish in vivo. More importantly, casearlucin A significantly inhibited cell proliferation and migration in an in vivo zebrafish xenograft model. Collectively, these results are valuable for the further development and application of casearlucin A as an anticancer agent.
Collapse
Affiliation(s)
- Ying Li
- State Key Laboratory of Medicinal Chemistry Biology, College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, and Drug Discovery Center for Infectious Disease, Nankai University, Tianjin, China
| | - Jun Ma
- State Key Laboratory of Medicinal Chemistry Biology, College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, and Drug Discovery Center for Infectious Disease, Nankai University, Tianjin, China
| | - Ziteng Song
- State Key Laboratory of Medicinal Chemistry Biology, College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, and Drug Discovery Center for Infectious Disease, Nankai University, Tianjin, China
| | - Yinan Zhao
- State Key Laboratory of Medicinal Chemistry Biology, College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, and Drug Discovery Center for Infectious Disease, Nankai University, Tianjin, China
| | - Han Zhang
- State Key Laboratory of Medicinal Chemistry Biology, College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, and Drug Discovery Center for Infectious Disease, Nankai University, Tianjin, China
| | - Yeling Li
- State Key Laboratory of Medicinal Chemistry Biology, College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, and Drug Discovery Center for Infectious Disease, Nankai University, Tianjin, China
| | - Jing Xu
- State Key Laboratory of Medicinal Chemistry Biology, College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, and Drug Discovery Center for Infectious Disease, Nankai University, Tianjin, China
| | - Yuanqiang Guo
- State Key Laboratory of Medicinal Chemistry Biology, College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, and Drug Discovery Center for Infectious Disease, Nankai University, Tianjin, China
| |
Collapse
|
25
|
Zhu Z, Liu Z, Cui J, Huang Y, Chen H, Wu Y, Huang X, Gan C. Apoptosis inducing properties of 3-biotinylate-6-benzimidazole B-nor-cholesterol analogues. Steroids 2021; 169:108822. [PMID: 33722574 DOI: 10.1016/j.steroids.2021.108822] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 01/14/2023]
Abstract
In this work, a series of Biotin-substituted B-nor-cholesteryl benzimidazole compounds were synthesized. The antiproliferativeactivities of these compounds were evaluated in vitro using a series of human cancer cell lines, including HeLa (cervical cancer), SKOV3 (ovarian cancer), T-47D (thymus gland cancer), MCF-7 (human breast cancer) and HEK293T (normal renal epithelial) cells. These compounds displayed distinct antiproliferative activities against the currently tested cancer cells. The apoptotic properties induced by compound 6d were further investigated. Our results showed that compound 6d could induce the apoptosis of SKOV3 cells, blocking the cell growth in S-phase. Western blotting analyses revealed that compound 6d can induce cell apoptosis via the mitochondria-dependent pathway.
Collapse
Affiliation(s)
- Zhiling Zhu
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Zhiping Liu
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Jianguo Cui
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Yanmin Huang
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Hualong Chen
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Yulan Wu
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Xiaotong Huang
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Chunfang Gan
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China.
| |
Collapse
|
26
|
Li H, Fang Y, Yan J, Ren X, Zheng C, Wu B, Wang S, Li Z, Hua H, Wang P, Li D. Small-molecule fluorescent probes for H2S detection: Advances and perspectives. Trends Analyt Chem 2021. [DOI: 10.1016/j.trac.2020.116117] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
27
|
Zheng YG, Zhang WQ, Meng L, Wu XQ, Zhang L, An L, Li CL, Gao CY, Xu L, Liu Y. Design, synthesis and biological evaluation of 4-aniline quinazoline derivatives conjugated with hydrogen sulfide (H2S) donors as potent EGFR inhibitors against L858R resistance mutation. Eur J Med Chem 2020; 202:112522. [DOI: 10.1016/j.ejmech.2020.112522] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 10/24/2022]
|
28
|
Yang GZ, Zhang J, Peng JW, Zhang ZJ, Zhao WB, Wang RX, Ma KY, Li JC, Liu YQ, Zhao ZM, Shang XF. Discovery of luotonin A analogues as potent fungicides and insecticides: Design, synthesis and biological evaluation inspired by natural alkaloid. Eur J Med Chem 2020; 194:112253. [PMID: 32222678 DOI: 10.1016/j.ejmech.2020.112253] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 03/04/2020] [Accepted: 03/17/2020] [Indexed: 11/15/2022]
Abstract
The prevention and control of plant diseases and insect pests is the most crucial issue facing crop protection. To discover novel pesticide candidates with diverse chemical structures from natural products, a series of luotonin A analogues were designed, synthesized and evaluated for their antifungal and insecticidal activities. Most of these compounds exhibited potent activity against Botrytis cinerea, Magnaporthe oryzae and Aphis craccivora. Among them, the antifungal activity of compound 10s against B. cinerea was comparable to azoxystrobin (EC50 = 0.09 mM) and against M. oryzae (EC50 = 0.19 mM) was slightly weaker than that of azoxystrobin (EC50 = 0.17 mM). Compounds 10k and 10o are the most active compounds against A. craccivora having identical mortality value of 42.05% at 50 μg/mL, respectively, which were slightly lower than pymetrozine (51.14%) at the same concentration. Revealed morphological changes of the fungal cell surface by scanning electron microscopy indicated that luotonin A analogues might exert their antifungal activity by destroying fungal cell membrane and cell wall. Furthermore, the results of the in vivo protective and curative activities of the compound 10s against S. sclerotiorum and B. cinerea showed that the curative effect was stronger than its protective effect and the curative effects reached 67.17% and 73.82% at 80 μg/mL respectively. The above results further demonstrated the potential of luotonin A analogues as novel fungicides and insecticides.
Collapse
Affiliation(s)
- Guan-Zhou Yang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Jian Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Jing-Wen Peng
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Zhi-Jun Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Wen-Bin Zhao
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Ren-Xuan Wang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Kun-Yuan Ma
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Jun-Cai Li
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Ying-Qian Liu
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, People's Republic of China.
| | - Zhong-Min Zhao
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Xiao-Fei Shang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, People's Republic of China; Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, People's Republic of China
| |
Collapse
|
29
|
Jiao R, Xu F, Huang X, Li H, Liu W, Cao H, Zang L, Li Z, Hua H, Li D. Antiproliferative chromone derivatives induce K562 cell death through endogenous and exogenous pathways. J Enzyme Inhib Med Chem 2020; 35:759-772. [PMID: 32183548 PMCID: PMC7144234 DOI: 10.1080/14756366.2020.1740696] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A series of furoxan derivatives of chromone were prepared. The antiproliferative activities were tested against five cancer cell lines HepG2, MCF-7, HCT-116, B16, and K562, and two normal human cell lines L-02 and PBMCs. Among them, compound 15a exhibited the most potent antiproliferative activity. It was also found 15a produced more than 8 µM of NO at the peak time of 45 min by Griess assay. Generally, antiproliferative activity is positively related to NO release to some extent. Further in-depth studies on apoptosis-related mechanisms showed that 15a caused S-phase cell cycle arrest in a concentration-dependent manner and induced apoptosis significantly through mitochondria-related pathways. Human apoptosis protein array assay also demonstrated 15a increased the expression levels of pro-apoptotic Bax, Bad, HtrA2 and Trail R2/DR5. The expression of catalase and cell cycle blocker claspin were similarly up-regulated. In balance, 15a induced K562 cells death through both endogenous and exogenous pathways.
Collapse
Affiliation(s)
- Runwei Jiao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, P. R. China
| | - Fanxing Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, P. R. China
| | - Xiaofang Huang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, P. R. China
| | - Haonan Li
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, P. R. China
| | - Weiwei Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, P. R. China
| | - Hao Cao
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, P. R. China
| | - Linghe Zang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, P. R. China
| | - Zhanlin Li
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, P. R. China
| | - Huiming Hua
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, P. R. China
| | - Dahong Li
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, P. R. China
| |
Collapse
|
30
|
Liang Y, Zhang Q, Yang X, Li Y, Zhang X, Li Y, Du Q, Jin DQ, Cui J, Lall N, Tuerhong M, Lee D, Abudukeremu M, Xu J, Shuai L, Guo Y. Diterpenoids from the leaves of Casearia kurzii showing cytotoxic activities. Bioorg Chem 2020; 98:103741. [PMID: 32213364 DOI: 10.1016/j.bioorg.2020.103741] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/27/2020] [Accepted: 03/07/2020] [Indexed: 11/16/2022]
Abstract
A phytochemical investigation to obtain bioactive substances as lead compounds or agents for cancer led to the obtainment of six new and two known clerodane diterpenoids from the leaves of Casearia kurzii. Their structures were elucidated using NMR techniques and electronic circular dichroism (ECD) calculations. The subsequent biological cytotoxicity evaluation of these isolates toward human lung cancer A549, human cervical cancer HeLa, human chronic myeloid leukemia K562, and human hepatocellular carcinoma HepG2 was carried out. The most active compound 4 with an IC50 value of 9.7 μM against HepG2 cells was selected to examine the cytotoxic mechanism, which induced the apoptosis and arrested the HepG2 cell cycle at S stage. The in vivo zebrafish experiments revealed that compound 4 had the property of inhibiting tumor proliferation and migration.
Collapse
Affiliation(s)
- Yue Liang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| | - Qi Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| | - Xueyuan Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| | - Ying Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| | - Xuke Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| | - Yuhao Li
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
| | - Qing Du
- Key Laboratory for Tibet Plateau Phytochemistry of Qinghai Province, College of Pharmacy, Qinghai Nationalities University, Xining 810007, People's Republic of China
| | - Da-Qing Jin
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
| | - Jianlin Cui
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
| | - Namrita Lall
- Department of Plant and Soil Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Muhetaer Tuerhong
- College of Chemistry and Environmental Sciences, Laboratory of Xinjiang Native Medicinal and Edible Plant Resources Chemistry, Kashgar University, Kashgar 844000, People's Republic of China
| | - Dongho Lee
- Department of Biosystems and Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Munira Abudukeremu
- College of Chemistry and Environmental Sciences, Laboratory of Xinjiang Native Medicinal and Edible Plant Resources Chemistry, Kashgar University, Kashgar 844000, People's Republic of China
| | - Jing Xu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China; State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin 541004, People's Republic of China.
| | - Ling Shuai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| | - Yuanqiang Guo
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China.
| |
Collapse
|
31
|
Hydrogen sulfide releasing oridonin derivatives induce apoptosis through extrinsic and intrinsic pathways. Eur J Med Chem 2020; 187:111978. [DOI: 10.1016/j.ejmech.2019.111978] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/12/2019] [Accepted: 12/16/2019] [Indexed: 02/06/2023]
|
32
|
Zhu L, Liao W, Chang H, Liu X, Miao S. A Novel Fluorescent Probe for Detection of Hydrogen Sulfide and Its Bioimaging Applications in Living Cells. ChemistrySelect 2020. [DOI: 10.1002/slct.201903451] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Lei Zhu
- College of Chemistry and Chemical Engineering Hunan University ChangSha 410082 P. R. China
| | - Wenhao Liao
- College of Chemistry and Chemical Engineering Hunan University ChangSha 410082 P. R. China
| | - Haizhen Chang
- College of Chemistry and Chemical Engineering Hunan University ChangSha 410082 P. R. China
| | - Xianjun Liu
- College of Chemistry and Chemical Engineering Hunan University ChangSha 410082 P. R. China
| | - Shaobin Miao
- Department of Chemistry and Physics Augusta University Augusta GA 30912 USA
| |
Collapse
|
33
|
Kalluruttimmal R, Thekke Thattariyil D, Panthalattu Parambil A, Sen AK, Chakkumkumarath L, Manheri MK. Electronically-tuned triarylmethine scaffolds for fast and continuous monitoring of H 2S levels in biological samples. Analyst 2019; 144:4210-4218. [PMID: 31188362 DOI: 10.1039/c9an00522f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
A sensor for the detection and quantification of H2S in biological samples should ideally meet a set of criteria such as fast detection, high sensitivity in the desired concentration range, high selectivity, non-interference from biomolecules like proteins, ease of synthesis, long-term stability and water solubility. Although a number of H2S probes are known, none of them possess all the above attributes that are relevant for practical applications. As part of a program to develop reliable chemical probes for continuous monitoring of this gasotransmitter in the blood plasma of sepsis-prone individuals in post-operative wards, we have looked at the possibility of improving the reactivity and selectivity profile of triarylmethine dyes towards different nucleophiles. After achieving high sensitivity through electronic control, the interference from sulfite, thiosulfate and metabisulfite was addressed by introducing a metal salt-mediated desulfuration step that results in dye regeneration selectively from its H2S adduct. Typically, if the analyte contains only H2S, the loss of absorbance in the first step gets completely reinstated after the second step; absorbance changes in both steps vary linearly with sulfide concentration and either of these two steps can be used for the quantification of H2S with the help of standard plots. In the presence of interfering ions, the first step will show decolourization due to the presence of all of them whereas only the H2S-adduct will undergo desulfuration in the second step which can be used for quantification. The decolourization step is instantaneous while the desulfuration requires only about 50 s, making the entire protocol complete in less than a minute. The methodology optimized here also meets the requirements mentioned above for real-life applications.
Collapse
Affiliation(s)
- Ramshad Kalluruttimmal
- Department of Chemistry, Indian Institute of Technology Madras, Chennai-600036, Tamil Nadu, India.
| | | | | | | | | | | |
Collapse
|
34
|
Li H, Gao X, Huang X, Wang X, Xu S, Uchita T, Gao M, Xu J, Hua H, Li D. Hydrogen sulfide donating ent-kaurane and spirolactone-type 6,7-seco-ent-kaurane derivatives: Design, synthesis and antiproliferative properties. Eur J Med Chem 2019; 178:446-457. [PMID: 31202992 DOI: 10.1016/j.ejmech.2019.06.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 01/15/2023]
Abstract
Motivated by our interest in hydrogen sulfide bio-chemistry and ent-kaurane diterpenoid chemistry, 14 hydrogen sulfide donating derivatives (9, 11a-c, 12a-c, 13, 14, 16a-c and 17a-b) of ent-kaurane and spirolactone-type 6,7-seco-ent-kaurane were designed and synthesized. Four human cancer cell lines (K562, Bel-7402, SGC-7901 and A549) and two normal cell lines (L-02 and PBMC) were selected for antiproliferative assay. Most derivatives showed more potent activities than the lead ent-kaurane oridonin. Among them, compound 12b exhibited the most potent antiproliferative activities, with IC50 values of 1.01, 0.88, 4.36 and 5.21 μM against above human cancer cell lines, respectively. Further apoptosis-related mechanism study indicated that 12b could arrest Bel-7402 cell cycle at G1 phase and induce apoptosis through mitochondria related pathway. Through Western blot assay, 12b was shown to influence the intrinsic pathway by increasing the expression of Bax, cleaved caspase-3, cytochrome c and cleaved PARP, meanwhile suppressing procaspase-3, Bcl-2, Bcl-xL and PARP.
Collapse
Affiliation(s)
- Haonan Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Xiang Gao
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Xiaofang Huang
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Xianhua Wang
- School of Public Health, Qingdao University, 38 Dengzhou Road, Qingdao, 266021, PR China
| | - Shengtao Xu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Takahiro Uchita
- School of Pharmaceutical Sciences, Mukogawa Women's University, 11-68 Koshien, Nishinomiya, 663-8179, Japan
| | - Ming Gao
- School of Pharmaceutical Sciences, Mukogawa Women's University, 11-68 Koshien, Nishinomiya, 663-8179, Japan
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Huiming Hua
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Dahong Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China.
| |
Collapse
|
35
|
Wu P, Chen Y. Evodiamine ameliorates paclitaxel-induced neuropathic pain by inhibiting inflammation and maintaining mitochondrial anti-oxidant functions. Hum Cell 2019; 32:251-259. [PMID: 30701373 DOI: 10.1007/s13577-019-00238-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/14/2019] [Indexed: 02/08/2023]
Abstract
Chemotherapy-induced neuropathic pain (CINP) is a common and debilitating side effect of cancer treatment. Evodiamine, a major effective compound isolated from Evodia rutaecarpa, has been associated with anti-inflammatory and anti-nociceptive effects, an important therapeutic strategy for the treatment of neuropathic pain. However, the effects of evodiamine on CINP remain unknown. Thus, this study aims to investigate the pharmacological potential of evodiamine in attenuating paclitaxel-induced peripheral neuropathy. The results showed that evodiamine enhanced but not reduced the sensitivity of cancer cells to paclitaxel treatment. In a rat model of paclitaxel-induced peripheral neuropathy, evodiamine significantly ameliorated the development of mechanical and thermal hypersensitivity. Moreover, paclitaxel-induced the loss of intraepidermal nerve fibers was markedly inhibited by evodiamine administration. This inhibitory effect was accompanied with the decrease in inflammatory and chemoattractant cytokines level in dorsal root ganglia (DRG), such as interleukin (IL)-1β, IL-6, tumor necrosis factor-α and monocyte chemoattractant protein-1. In addition, evodiamine administration limited paclitaxel-induced elevation of oxidative stress in DRG tissues. The mitochondrial dysfunction evoked by paclitaxel was also remarkably improved in evodiamine-treated rats, evidenced by restoration of peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α), uncoupling protein 2 (UCP2), and superoxide dismutase 2 (SOD2) expression. In in vitro studies, we found that evodiamine prevented paclitaxel-induced the loss of mitochondrial membrane potential and PGC-1α, UCP2 and SOD2 expression in DRG cells. In conclusion, our study demonstrates that evodiamine ameliorates paclitaxel-induced neuropathic pain by inhibiting inflammatory response and maintaining mitochondrial anti-oxidant functions, indicating that evodiamine may be a promising therapeutic agent for CINP treatment.
Collapse
Affiliation(s)
- Peipei Wu
- Department of Anesthesiology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, NO. 29 Xinglong Road, Changzhou, 213000, Jiangsu, China
| | - Yong Chen
- Department of Anesthesiology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, NO. 29 Xinglong Road, Changzhou, 213000, Jiangsu, China.
| |
Collapse
|
36
|
Luo G, Tang Z, Li X, Hou Q, Chen Y, Lao K, Xiang H. 3, 9-di-O-substituted coumestrols incorporating basic amine side chains act as novel apoptosis inducers with improved pharmacological selectivity. Bioorg Chem 2019; 85:140-151. [PMID: 30612080 DOI: 10.1016/j.bioorg.2018.12.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/07/2018] [Accepted: 12/18/2018] [Indexed: 01/11/2023]
Abstract
There is much interest in the use of phytoestrogens such as coumestrol in breast cancer intervention due to their antiestrogenic activity and multiple modes of tumor cell death. However, the clear beneficial effects of naturally occurring estrogen mimetic coumestrol remain controversial due to experimental evidence that it has been shown to stimulate MCF-7 cell proliferation via agonist effect on estrogen receptor at low concentration. Herein, to disconnect the ER interaction and apoptosis-specific mechanism of coumestrol, various 3, 9-di-O-substituted coumestrols (7a-7e) and their furan ring-opened analogs (5a-5e) were synthesized and assessed for antiproliferative properties. Attachment of a dimethylamine-containing side chain to 3-O of coumestrol led to the most promising compound 7e with improved antiproliferative activity (1.7-fold increase) against MCF-7 cells, decreased estrogen activity (>20 times weaker ERα binder) and a novel action to induce apoptosis. Mechanistic studies revealed that 7e is a tubulin polymerization inhibitor, which could arrest cell cycle at G2/M phase and induce apoptosis along with the decrease of mitochondrial membrane potential. In summary, such subtle modifications to the 3, 9-di-hydroxyl groups of coumestrol allow the generation of a novel apoptosis inducer with distinct pharmacological properties, providing an excellent starting point to future development of novel tumor-vascular disrupting agents targeting tubulin.
Collapse
Affiliation(s)
- Guoshun Luo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhengpu Tang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xinyu Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qiangqiang Hou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yu Chen
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Smart Drug Delivery, Ministry of Education School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China
| | - Kejing Lao
- Shaanxi Key Laboratory of Brain Disorders and Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, China
| | - Hua Xiang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
37
|
Zhao TQ, Zhao YD, Liu XY, Li ZH, Wang B, Zhang XH, Cao YQ, Ma LY, Liu HM. Novel 3-(2,6,9-trisubstituted-9H-purine)-8-chalcone derivatives as potent anti-gastric cancer agents: Design, synthesis and structural optimization. Eur J Med Chem 2019; 161:493-505. [PMID: 30388465 DOI: 10.1016/j.ejmech.2018.10.058] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/10/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023]
Abstract
To explore anti-gastric cancer agents with high efficacy and selectivity, we report the design, synthesis and optimization of a novel series of 3-(2,6,9-trisubstituted-9H-purine)-8-chalcone derivatives starting from the compound PCA-15 reported by us previously. Most of the target compounds demonstrated significant antiproliferative effects on MGC803 cancer cell line, and more potent than the positive control (PCA-15 and 5-Fu). Among them, compound 6o was identified to be the most active compound against MGC803 cell line with an IC50 value of 0.84 μM. Additionally, high selectivity was also observed between cancer and normal cells (23.35 μM against GES-1). Further mechanism studies confirmed that compound 6o could inhibit colony formation and migration, induce the apoptosis of MGC803 cells through both the mitochondrial-mediated intrinsic pathway and death receptor-mediated extrinsic pathway, which were evidenced by the up-regulation of Bax, cleaved-caspase 9/3/8, cleaved PARP and down-regulation of Bcl-2. Our systematic studies implied a new scaffold targeting gastric cancer cells for further development of small-molecule compounds with improved potency and selectivity.
Collapse
Affiliation(s)
- Tao-Qian Zhao
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, PR China
| | - Yuan-Di Zhao
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, PR China
| | - Xin-Yang Liu
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, PR China
| | - Zhong-Hua Li
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, PR China
| | - Bo Wang
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, PR China
| | - Xin-Hui Zhang
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, PR China
| | - Ya-Quan Cao
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, PR China
| | - Li-Ying Ma
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, PR China.
| | - Hong-Min Liu
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, PR China.
| |
Collapse
|
38
|
Figliuolo VR, Coutinho-Silva R, Coutinho CMLM. Contribution of sulfate-reducing bacteria to homeostasis disruption during intestinal inflammation. Life Sci 2018; 215:145-151. [PMID: 30414430 DOI: 10.1016/j.lfs.2018.11.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/02/2018] [Accepted: 11/05/2018] [Indexed: 12/26/2022]
Abstract
Alteration in microbial populations and metabolism are key events associated with disruption of intestinal homeostasis and immune tolerance during intestinal inflammation. A substantial imbalance in bacterial populations in the intestine and their relationships with the host have been observed in patients with inflammatory bowel disease (IBD), believed to be part of an intricate mechanism of triggering and progression of intestinal inflammation. Because elevated numbers of sulfate-reducing bacteria (SRB) have been found in the intestines of patients with IBD, the study of their interaction with intestinal cells and their potential involvement in IBD has been the focus of investigation to better understand the intestinal pathology during IBD, as well as to find new ways to treat the disease. SRB not only directly interact with intestinal epithelial cells during intestinal inflammation but may also promote intestinal damage through generation of hydrogen sulfide at high levels. Herein we review the literature to discuss the various aspects of SRB interaction with host intestinal tissue, focusing on their interaction with intestinal epithelial and immune cells during intestinal inflammation.
Collapse
Affiliation(s)
- Vanessa Ribeiro Figliuolo
- Instituto de Biofísica Carlos Chagas Filho - IBCCF, Universidade Federal do Rio de Janeiro, RJ, Brazil; LITEB, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, RJ, Brazil; Department of Pediatrics, University of Arizona, Tucson, AZ, USA
| | - Robson Coutinho-Silva
- Instituto de Biofísica Carlos Chagas Filho - IBCCF, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Claudia Mara Lara Melo Coutinho
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niteroi, RJ, Brazil; LITEB, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
39
|
Guo XX, Li XP, Zhou P, Li DY, Lyu XT, Chen Y, Lyu YW, Tian K, Yuan DZ, Ran JH, Chen DL, Jiang R, Li J. Evodiamine Induces Apoptosis in SMMC-7721 and HepG2 Cells by Suppressing NOD1 Signal Pathway. Int J Mol Sci 2018; 19:ijms19113419. [PMID: 30384473 PMCID: PMC6274686 DOI: 10.3390/ijms19113419] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/25/2018] [Accepted: 10/30/2018] [Indexed: 12/18/2022] Open
Abstract
Hepatocellular cancer (HCC) is a lethal malignancy with poor prognosis and easy recurrence. There are few agents with minor toxic side effects that can be used for treatment of HCC. Evodiamine (Evo), one of the major bioactive components derived from fructus Evodiae, has long been shown to exert anti-hepatocellular carcinoma activity by suppressing activation of nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK). In addition, in the Nucleotide-Binding Oligomerization Domain 1 (NOD1) pathway, NOD1 could initiate NF-κB-dependent and MAPK-dependent gene transcription. Recent experimental studies reported that the NOD1 pathway was related to controlling development of various tumors. Here we hypothesize that Evo exerts anti-hepatocellular carcinoma activity by inhibiting NOD1 to suppress NF-κB and MAPK activation. Therefore, we proved the anti-hepatocellular carcinoma activity of Evo on HCC cells and detected the effect of Evo on the NOD1 pathway. We found that Evo significantly induced cell cycle arrest at the G2/M phase, upregulated P53 and Bcl-2 associated X proteins (Bax) proteins, and downregulated B-cell lymphoma-2 (Bcl-2), cyclinB1, and cdc2 proteins in HCC cells. In addition, Evo reduced levels of NOD1, p-P65, p-ERK, p-p38, and p-JNK, where the level of IκBα of HCC cells increased. Furthermore, NOD1 agonist γ-D-Glu-mDAP (IE-DAP) treatment weakened the effect of Evo on suppression of NF-κB and MAPK activation and cellular proliferation of HCC. In an in vivo subcutaneous xenograft model, Evo also exhibited excellent tumor inhibitory effects via the NOD1 signal pathway. Our results demonstrate that Evo could induce apoptosis remarkably and the inhibitory effect of Evo on HCC cells may be through suppressing the NOD1 signal pathway in vitro and in vivo.
Collapse
Affiliation(s)
- Xing-Xian Guo
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
| | - Xiao-Peng Li
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
| | - Peng Zhou
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
| | - Dan-Yang Li
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
| | - Xiao-Ting Lyu
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
| | - Yi Chen
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
| | - Yan-Wei Lyu
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
| | - Kuan Tian
- Neuroscience Research Center, College of basic medicine, Chongqing Medical University, Chongqing 400016, China.
| | - De-Zhi Yuan
- Neuroscience Research Center, College of basic medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Jian-Hua Ran
- Neuroscience Research Center, College of basic medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Di-Long Chen
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
- Chongqing Three Gorges Medical College, Chongqing 400016, China.
| | - Rong Jiang
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
| | - Jing Li
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
40
|
Antiproliferative Effects of Alkaloid Evodiamine and Its Derivatives. Int J Mol Sci 2018; 19:ijms19113403. [PMID: 30380774 PMCID: PMC6274956 DOI: 10.3390/ijms19113403] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 10/20/2018] [Accepted: 10/24/2018] [Indexed: 12/18/2022] Open
Abstract
Alkaloids, a category of natural products with ring structures and nitrogen atoms, include most U.S. Food and Drug Administration approved plant derived anti-cancer agents. Evodiamine is an alkaloid with attractive multitargeting antiproliferative activity. Its high content in the natural source ensures its adequate supply on the market and guarantees further medicinal study. To the best of our knowledge, there is no systematic review about the antiproliferative effects of evodiamine derivatives. Therefore, in this article the review of the antiproliferative activities of evodiamine will be updated. More importantly, the antiproliferative activities of structurally modified new analogues of evodiamine will be summarized for the first time.
Collapse
|
41
|
Han T, Wang Y, Wang M, Li X, Cheng K, Gao X, Li Z, Bai J, Hua H, Li D. Synthesis of scutellarein derivatives with antiproliferative activity and selectivity through the intrinsic pathway. Eur J Med Chem 2018; 158:493-501. [DOI: 10.1016/j.ejmech.2018.09.047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 08/21/2018] [Accepted: 09/14/2018] [Indexed: 12/23/2022]
|