1
|
Wang S, Li C, Zhang L, Sun B, Cui Y, Sang F. Isolation and biological activity of natural chalcones based on antibacterial mechanism classification. Bioorg Med Chem 2023; 93:117454. [PMID: 37659218 DOI: 10.1016/j.bmc.2023.117454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/07/2023] [Accepted: 08/21/2023] [Indexed: 09/04/2023]
Abstract
Bacterial infection, which is still one of the leading causes of death in humans, poses an enormous threat to the worldwide public health system. Antibiotics are the primary medications used to treat bacterial diseases. Currently, the discovery of antibiotics has reached an impasse, and due to the abuse of antibiotics resulting in bacterial antibiotic resistance, researchers have a critical desire to develop new antibacterial agents in order to combat the deteriorating antibacterial situation. Natural chalcones, the flavonoids consisting of two phenolic rings and a three-carbon α, β-unsaturated carbonyl system, possess a variety of biological and pharmacological properties, including anti-cancer, anti-inflammatory, antibacterial, and so on. Due to their potent antibacterial properties, natural chalcones possess the potential to become a new treatment for infectious diseases that circumvents existing antibiotic resistance. Currently, the majority of research on natural chalcones focuses on their synthesis, biological and pharmacological activities, etc. A few studies have been conducted on their antibacterial activity and mechanism. Therefore, this review focuses on the antibacterial activity and mechanisms of seventeen natural chalcones. Firstly, seventeen natural chalcones have been classified based on differences in antibacterial mechanisms. Secondly, a summary of the isolation and biological activity of seventeen natural chalcones was provided, with a focus on their antibacterial activity. Thirdly, the antibacterial mechanisms of natural chalcones were summarized, including those that act on bacterial cell membranes, biological macromolecules, biofilms, and quorum sensing systems. This review aims to lay the groundwork for the discovery of novel antibacterial agents based on chalcones.
Collapse
Affiliation(s)
- Sinan Wang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, PR China
| | - Chuang Li
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, PR China
| | - Liyan Zhang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, PR China
| | - Bingxia Sun
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, PR China
| | - Yuting Cui
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, PR China.
| | - Feng Sang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, PR China.
| |
Collapse
|
2
|
Ouyang L, Fan Z, He Y, Tan L, Deng G, He Q, He Y, Ouyang T, Li C, Zhang Q, Liu H, Zuo Y. 4-hydroxylonchocarpin and corylifol A: The potential hepatotoxic components of Psoralea corylifolia L. Toxicol Lett 2023; 385:31-41. [PMID: 37598872 DOI: 10.1016/j.toxlet.2023.08.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/10/2023] [Accepted: 08/17/2023] [Indexed: 08/22/2023]
Abstract
Psoralea corylifolia L. (P. corylifolia) has attracted increasing attention because of its potential hepatotoxicity. In this study, we used network analysis (toxic component and hepatotoxic target prediction, proteinprotein interaction, GO enrichment analysis, KEGG pathway analysis, and molecular docking) to predict the components and mechanism of P. corylifolia-induced hepatotoxicity and then selected 4-hydroxylonchocarpin and corylifol A for experimental verification. HepG2 cells were treated with low, medium, and high concentrations of 4-hydroxylonchocarpin or corylifol A. The activities of ALT, AST, and LDH in cell culture media and the MDA level, SOD activity, and GSH level in cell extracts were measured. Moreover, apoptosis, ROS levels, and mitochondrial membrane potential were evaluated. The results showed that the activities of ALT, AST, and LDH in the culture medium increased, and hepatocyte apoptosis increased. The level of MDA increased, and the activity of SOD and level of GSH decreased, and the ROS level increased with 4-hydroxylonchocarpin and corylifol A intervention. Furthermore, the mitochondrial membrane potential decreased in the 4-hydroxylonchocarpin and corylifol A groups. This study suggests that 4-hydroxylonchocarpin and corylifol A cause hepatocyte injury and apoptosis by inducing oxidative stress and mitochondrial dysfunction, suggesting that these compounds may be the potential hepatotoxic components of P. corylifolia.
Collapse
Affiliation(s)
- Linqi Ouyang
- Department of Pharmacy, The First Hospital of Hunan University of Chinese Medicine, Changsha, China; School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Zhiqiang Fan
- Department of Pharmacy, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Yang He
- Department of Pharmacy, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Long Tan
- Department of Pharmacy, People's Hospital of Yizhang County, Chenzhou, China
| | - Guoyan Deng
- Department of Pharmacy, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Qin He
- Department of Pharmacy, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Yiran He
- Department of Pharmacy, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Ting Ouyang
- Department of Pharmacy, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Congjie Li
- Department of Pharmacy, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Qin Zhang
- Department of Pharmacy, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Hongyu Liu
- Department of Pharmacy, The First Hospital of Hunan University of Chinese Medicine, Changsha, China.
| | - Yajie Zuo
- Department of Pharmacy, The First Hospital of Hunan University of Chinese Medicine, Changsha, China.
| |
Collapse
|
3
|
Bavachin and Corylifol A Improve Muscle Atrophy by Enhancing Mitochondria Quality Control in Type 2 Diabetic Mice. Antioxidants (Basel) 2023; 12:antiox12010137. [PMID: 36671000 PMCID: PMC9855061 DOI: 10.3390/antiox12010137] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/28/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023] Open
Abstract
Type 2 diabetes reduces muscle mass and function. Chronic inflammation and mitochondrial dysfunction play critical roles in muscle atrophy pathogenesis. Here, we investigated the effects of bavachin and corylifol A from Psoralea corylifolia L. seeds on muscle atrophy in dexamethasone-treated mice and in db/db mice. Bavachin and corylifol A enhanced muscle strength and muscle mass in dexamethasone-treated mice. In diabetic mice, they enhanced muscle strength and cross-sectional areas. Bavachin and corylifol A suppressed inflammatory cytokine (interleukin-6 and tumor necrosis factor-α) expression levels by downregulating nuclear factor-κB phosphorylation. They decreased the muscle atrophic factor (myostatin, atrogin-1, and muscle RING finger-1) expression levels. They activated the AKT synthetic signaling pathway and induced a switch from fast-type glycolytic fibers (type 2B) to slow-type oxidative fibers (types I and 2A). They increased mitochondrial biogenesis and dynamic factor (optic atrophy-1, mitofusin-1/2, fission, mitochondrial 1, and dynamin 1-like) expression levels via the AMP-activated protein kinase-peroxisome proliferator-activated receptor gamma coactivator 1-alpha signaling pathway. They also improved mitochondrial quality by upregulating the mitophagy factor (p62, parkin, PTEN-induced kinase-1, and BCL2-interacting protein-3) expression levels. Therefore, bavachin and corylifol A exert potential therapeutic effects on muscle atrophy by suppressing inflammation and improving mitochondrial function.
Collapse
|
4
|
Singh A, Patel SK, Kumar P, Das KC, Verma D, Sharma R, Tripathi T, Giri R, Martins N, Garg N. Quercetin acts as a P-gp modulator via impeding signal transduction from nucleotide-binding domain to transmembrane domain. J Biomol Struct Dyn 2020; 40:4507-4515. [PMID: 33306006 DOI: 10.1080/07391102.2020.1858966] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The inherent ability of the cancer cells to resist chemotherapeutic agents is a major challenge in drug discovery. Chemotherapy is one of the most widely used treatment methods for cancer, but due to multidrug resistance (MDR) development in cancer cells, the healing procedure often fails. Various factors impart cancer resistance to cells; among them, P-glycoprotein (P-gp) overexpression is directly linked to MDR in cancer cells. P-gp leads to the efflux of drug molecules to the extracellular space. Several molecules have been reported to inhibit the P-gp activity. Among them, quercetin has revealed a great potential to modulate P-gp activity. However, the mechanistic understanding of quercetin induced modulation is not entirely elucidated. In the present work, we showed that quercetin binds in the interacting region between the transmembrane domain and nucleotide-binding domain out of the three plausible binding sites of P-gp and restrict the conformational change from inward- to outward-facing conformation of P-gp. Due to the absence of the inward-facing structure of human P-gp, we first modeled an inward-facing P-gp structure. Using molecular docking, the interacting residues of P-gp were identified, and the stability and interaction dynamics of the complex were studied using molecular dynamics simulation. Our work reveals the mechanistic understanding of quercetin induced modulation of P-gp and indicates its importance in cancer treatment.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ashutosh Singh
- School of Basic Sciences and BioX Center, Indian Institute of Technology Mandi, VPO Kamand, Mandi, Himachal Pradesh, India
| | - Sandesh Kumar Patel
- School of Basic Sciences and BioX Center, Indian Institute of Technology Mandi, VPO Kamand, Mandi, Himachal Pradesh, India
| | - Prateek Kumar
- School of Basic Sciences and BioX Center, Indian Institute of Technology Mandi, VPO Kamand, Mandi, Himachal Pradesh, India
| | - Kanhu Charan Das
- Department of Biochemistry, North-Eastern Hill University, Umshing, Shillong, Meghalaya, India
| | - Deepanshu Verma
- School of Basic Sciences and BioX Center, Indian Institute of Technology Mandi, VPO Kamand, Mandi, Himachal Pradesh, India
| | - Rohit Sharma
- Department of Rasashastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Timir Tripathi
- Department of Biochemistry, North-Eastern Hill University, Umshing, Shillong, Meghalaya, India
| | - Rajanish Giri
- School of Basic Sciences and BioX Center, Indian Institute of Technology Mandi, VPO Kamand, Mandi, Himachal Pradesh, India
| | - Natália Martins
- Faculty of Medicine, University of Porto, Porto, Portugal.,Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal
| | - Neha Garg
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
5
|
Han Y, Lee H, Li H, Ryu JH. Corylifol A from Psoralea corylifolia L. Enhances Myogenesis and Alleviates Muscle Atrophy. Int J Mol Sci 2020; 21:ijms21051571. [PMID: 32106603 PMCID: PMC7084366 DOI: 10.3390/ijms21051571] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 12/21/2022] Open
Abstract
Inflammatory conditions caused by cancer, chronic diseases or aging can lead to skeletal muscle atrophy. We identified myogenic compounds from Psoralea corylifolia (PC), a medicinal plant that has been used for the treatment of inflammatory and skin diseases. C2C12 mouse skeletal myoblasts were differentiated in the presence of eight compounds isolated from PC to evaluate their myogenic potential. Among them, corylifol A showed the strongest transactivation of MyoD and increased expression of myogenic markers, such as MyoD, myogenin and myosin heavy chain (MHC). Corylifol A increased the number of multinucleated and MHC-expressing myotubes. We also found that the p38 MAPK signaling pathway is essential for the myogenic action of corylifol A. Atrophic condition was induced by treatment with dexamethasone. Corylifol A protected against dexamethasone-induced myotube loss by increasing the proportion of multinucleated MHC-expressing myotubes compared with dexamethasone-damaged myotubes. Corylifol A reduced the expression of muscle-specific ubiquitin-E3 ligases (MAFbx and MuRF1) and myostatin, while activating Akt. These dual effects of corylifol A, inhibition of catabolic and activation of anabolic pathways, protect myotubes against dexamethasone damage. In summary, corylifol A isolated from P. corylifolia alleviates muscle atrophic condition through activating myoblast differentiation and suppressing muscle degradation in atrophic conditions.
Collapse
|
6
|
Flavonoids from the Amazon plant Brosimum acutifolium induce C6 glioma cell line apoptosis by disrupting mitochondrial membrane potential and reducing AKT phosphorylation. Biomed Pharmacother 2019; 113:108728. [DOI: 10.1016/j.biopha.2019.108728] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/21/2019] [Accepted: 02/22/2019] [Indexed: 11/18/2022] Open
|
7
|
Lim J, Nam S, Jeong JH, Kim MJ, Yang Y, Lee MS, Lee HG, Ryu JH, Lim JS. Kazinol U inhibits melanogenesis through the inhibition of tyrosinase-related proteins via AMP kinase activation. Br J Pharmacol 2019; 176:737-750. [PMID: 30579288 DOI: 10.1111/bph.14560] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/23/2018] [Accepted: 11/27/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Kazinol U is a prenylated flavan isolated from an extract of Broussonetia kazinoki Sieb (Moraceae). Kazinol U has shown cytoprotective effects against cytokine-induced apoptotic cell death and induces AMP kinase (AMPK) activation through LKB1 activation. However, kazinol U has not been tested as a regulator of melanogenesis, although bark extract of B. kazinoki has been used as a cosmetic ingredient for skin conditioning. EXPERIMENTAL APPROACH We cultured mouse, human melanoma cells and normal human melanocytes to demonstrate anti-melanogenic effects of kazinol U. A tyrosinase activity assay, Western blot, RT-qPCR and a luciferase reporter gene assay were performed to determine the anti-melanogenic mechanisms of kazinol U. We confirmed its effect on melanogenesis in vivo using zebrafish. KEY RESULTS Kazinol U inhibited the expression and activity of tyrosinase, the rate-limiting enzyme in melanogenesis, and reduced tyrosinase expression and activity in response to cAMP-inducing agents. Kazinol U reduced the expression of other melanogenic enzymes, such as tyrosinase-related protein (Tyrp) 1 and Tyrp2, and down-regulated microphthalmia-associated transcription factor (MITF), the master regulator of the tyrosinase gene family. Moreover, kazinol U induced phosphorylation of AMPK and MAPK proteins, which are MITF inhibitors. It also exhibited anti-melanogenic effects in zebrafish, a recently developed in vivo model. CONCLUSIONS AND IMPLICATIONS Our findings suggest that kazinol U reduces melanogenesis via its inhibitory effect on MITF and its downstream target genes, tyrosinase, Tyrp1 and Tyrp2. This work may provide a basis for the application of kazinol U for the treatment of hyperpigmentation skin disorders.
Collapse
Affiliation(s)
- Jihyun Lim
- Department of Biological Science and Cellular Heterogeneity Research Center, Sookmyung Women's University, Seoul, Republic of Korea
| | - Sorim Nam
- Department of Biological Science and Cellular Heterogeneity Research Center, Sookmyung Women's University, Seoul, Republic of Korea
| | - Ji Hye Jeong
- Research Center for Cell Fate Control and College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Min Jung Kim
- Department of Biological Science and Cellular Heterogeneity Research Center, Sookmyung Women's University, Seoul, Republic of Korea
| | - Young Yang
- Department of Biological Science and Cellular Heterogeneity Research Center, Sookmyung Women's University, Seoul, Republic of Korea
| | - Myeong-Sok Lee
- Department of Biological Science and Cellular Heterogeneity Research Center, Sookmyung Women's University, Seoul, Republic of Korea
| | - Hee Gu Lee
- Medical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Jae-Ha Ryu
- Research Center for Cell Fate Control and College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Jong-Seok Lim
- Department of Biological Science and Cellular Heterogeneity Research Center, Sookmyung Women's University, Seoul, Republic of Korea
| |
Collapse
|
8
|
Lee H, Li H, Kweon M, Choi Y, Kim MJ, Ryu JH. Isobavachalcone from Angelica keiskei Inhibits Adipogenesis and Prevents Lipid Accumulation. Int J Mol Sci 2018; 19:ijms19061693. [PMID: 29882838 PMCID: PMC6032101 DOI: 10.3390/ijms19061693] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/02/2018] [Accepted: 06/04/2018] [Indexed: 12/12/2022] Open
Abstract
We isolated isobavachalcone (IBC) from Angelica keiskei (AK) as an anti-obesity component. IBC dose-dependently inhibited 3T3-L1 adipocyte differentiation by down-regulating adipogenic factors. At the mitotic clonal expansion stage (MCE), IBC caused cell cycle arrest in G0/G1 with decreased expression of cell cycle-regulating proteins. IBC also inhibited autophagic flux by inducing intracellular accumulation of LC3B and SQSTM1/p62 proteins while decreasing expression levels of regulating factors for autophagy initiation. In parallel with the inhibition of adipocyte differentiation, IBC decreased intrahepatic fat deposits and rescued the liver steatosis in high fat cholesterol diet-fed zebrafish. In this study, we found that IBC isolated from AK suppresses mitotic clonal expansion and autophagy flux of adipocytes and also shows anti-obesity activity in a high cholesterol-diet zebrafish model by decreasing intrahepatic fat deposits. These results suggest that IBC could be a leading pharmacological compound for the development of anti-obesity drugs.
Collapse
Affiliation(s)
- Hyejin Lee
- Research Center for Cell Fate Control and College of Pharmacy, Sookmyung Women’s University, 100 Chungparo 47-Gil, Yongsan-Gu, Seoul 04310, Korea; (H.L.); (H.L.); minson-_-@nate.com (M.K.)
| | - Hua Li
- Research Center for Cell Fate Control and College of Pharmacy, Sookmyung Women’s University, 100 Chungparo 47-Gil, Yongsan-Gu, Seoul 04310, Korea; (H.L.); (H.L.); minson-_-@nate.com (M.K.)
| | - Minson Kweon
- Research Center for Cell Fate Control and College of Pharmacy, Sookmyung Women’s University, 100 Chungparo 47-Gil, Yongsan-Gu, Seoul 04310, Korea; (H.L.); (H.L.); minson-_-@nate.com (M.K.)
| | - Youngsook Choi
- Research institute of women’s health, Sookmyung Women’s University, 100 Chungparo 47-Gil, Yongsan-Gu, Seoul 04310, Korea;
| | - Min Jung Kim
- Department of Biological Sciences, Sookmyung Women’s University, 100 Chungparo 47-Gil, Yongsan-Gu, Seoul 04310, Korea;
| | - Jae-Ha Ryu
- Research Center for Cell Fate Control and College of Pharmacy, Sookmyung Women’s University, 100 Chungparo 47-Gil, Yongsan-Gu, Seoul 04310, Korea; (H.L.); (H.L.); minson-_-@nate.com (M.K.)
- Correspondence: ; Tel.: +82-2-710-9568
| |
Collapse
|