1
|
He B, Tan HZ, Liu CB, Wu H, He LQ. Synthesis and Antitumor Evaluation of a Novel Class of Chalcone Mannich Base Derivatives. Chem Biol Drug Des 2025; 105:e70079. [PMID: 40047275 DOI: 10.1111/cbdd.70079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/23/2025] [Accepted: 02/18/2025] [Indexed: 03/19/2025]
Abstract
A novel class of chalcone Mannich base derivatives I1-9 and II1-11 was synthesized, which exhibited significant antiproliferation activities in five different cancer cells. The activities of most compounds were superior to those of the positive control drug 5-FU. Moreover, compared with the intermediate chalcone, their water solubility was also significantly enhanced. Among them, the most prospective compound I4 (IC50 = 3.09-5.08 μM for the tested cancer cells) can effectively inhibit the proliferation of A549/DDP cells (IC50 = 4.69 μM). Further mechanistic studies revealed that it can induce apoptosis of A549 and A549/DDP cells by arresting the G2/M phase of the cell cycle. Although the selectivity of compound I4 between tumor cells and normal cells was not obvious, it might be a promising lead compound for lung cancer and is worthy of further investigation.
Collapse
Affiliation(s)
- Bing He
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Hong-Zhou Tan
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Cheng-Bo Liu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Hong Wu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Li-Qin He
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
2
|
Kwon MJ, Raut PK, Jang JH, Chun KS. Isoliquiritigenin Induces Apoptosis via ROS-Mediated Inhibition of p38/mTOR/STAT3 Pathway in Human Melanoma Cells. Biomol Ther (Seoul) 2025; 33:378-387. [PMID: 39933948 PMCID: PMC11893486 DOI: 10.4062/biomolther.2024.118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 02/13/2025] Open
Abstract
Isoliquiritigenin (ISL), a phenolic compound derived from licorice, exhibits various biological activities, including anti-inflammatory, anti-viral, anti-tumor, and antioxidant effects. However, the molecular mechanisms underlying its anti-cancer effects are not well understood in SK-MEL-28 melanoma cells. Melanoma, a highly aggressive and treatment-resistant cancer, remains a significant health challenge. This study investigates the anti-cancer effects of ISL, focusing on identifying reactive oxygen species (ROS)-mediated apoptosis mechanisms on SK-MEL-28 melanoma cells. Our results show that ISL treatment induces apoptosis in SK-MEL-28 cells, as evidenced by the cleavage of caspase-9, -7, -3, and PARP. ISL increased Bax expression, decreased Bcl-2 expression, and promoted cytochrome C release into the cytosol. ISL also reduced the expression of cell cycle markers, including cyclin D1, D3, and survivin. Notably, ISL treatment markedly increased intracellular ROS levels and pretreatment with N-acetyl cysteine, a ROS scavenger, abrogated the ISL-induced inhibition of the p38/mTOR/STAT3 pathway and prevented apoptosis. Moreover, ISL significantly diminished the constitutive phosphorylation of mTOR and STAT3 in SK-MEL-28 cells by blocking the phosphorylation of p38 MAPK, an upstream kinase of mTOR. Pharmacological inhibition of mTOR attenuated the STAT3 signaling, indicating that mTOR acts as an upstream kinase of STAT3 in these cells. Collectively, these findings demonstrate that ISL inhibits SK-MEL-28 cell growth by downregulating cell survival proteins and inducing apoptosis through ROS generation.
Collapse
Affiliation(s)
- Mi Jeong Kwon
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Pawan Kumar Raut
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Jeong-Hoon Jang
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| |
Collapse
|
3
|
Dolkar R, Paudwal G, Singh D, Behera C, Malik SB, Ali SM, Kaur H, Nargotra A, Shankar R, Singh SK, Gupta PN. Mechanistic Approach into 1,2,3-triazoles-based IIIM(S)-RS98 Mediated Apoptosis in Lung Cancer Cells. AAPS J 2025; 27:35. [PMID: 39900819 DOI: 10.1208/s12248-025-01018-9] [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: 08/27/2024] [Accepted: 01/06/2025] [Indexed: 02/05/2025] Open
Abstract
Lung cancer is a major public health problem across the globe, since it is the second most frequent cancer and the leading cause of cancer fatalities. This necessitates careful assessment of current therapies for lung cancer and discovery of novel drug candidates. 1,2,3 triazole compounds have emerged as an important class of prospective chemotherapeutic drugs for the treatment of lung cancer, with promising anti-lung cancer activity shown via a variety of pathways. They may interact with a various enzymes and receptors in cancer cells, causing cell cycle arrest and the activation of apoptosis. The present study aims to investigate the cytotoxic potential of institutional molecule based on 1,2,3 triazole [IIIM(S)-RS98] on multiple cancer cell lines. The compound was found to be most active on A549 cells and displayed the selectivity index as 8.16 in normal cells (e.g. HEK293). The in vitro findings revealed that IIIM(S)-RS98 induced apoptosis, loss of mitochondrial membrane potential, enhanced ROS and nitric oxide levels, and arrest cells in the G1 phase of the cell cycle. It inhibits the cell migration and clonogenic potential of A549 cells. Additionally, the downregulation of PI3K and p-Akt pathway leads to the activation of pro-apoptotic proteins Bax, downregulation of bcl2, activation of caspase 9, cleaved caspase 3, and cleaved parp1 expression and finally contribute towards apoptosis. Furthermore, molecular docking analysis indicated the interactions of IIIM(S)-RS98 with the apoptotic target proteins. The results demonstrated the potential of IIIM(S)-RS98 in the therapy of lung cancer.
Collapse
Affiliation(s)
- Rigzin Dolkar
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Gourav Paudwal
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Davinder Singh
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Chittaranjan Behera
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - Sumera Banoo Malik
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - Syed Mudassir Ali
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - Harjot Kaur
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - Amit Nargotra
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Ravi Shankar
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shashank K Singh
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Prem N Gupta
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
4
|
Xu C, Zhang C, Ganesan K, Qui C, Tang H, Gao F, Liu Q, Wu J, Sui Y, Li P, Zhang J, Chen J. Anti-migratory Properties of Cryoprotective Isoliquiritigenin-zein Phosphatidylcholine Nanoparticles Prevent Triple-negative Breast Cancer through PI3K-mTOR and MMP2/9 Pathways. Curr Med Chem 2025; 32:1770-1788. [PMID: 37936460 DOI: 10.2174/0109298673259973231023110945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/26/2023] [Accepted: 09/15/2023] [Indexed: 11/09/2023]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC), an aggressive type of breast cancer, remains difficult to treat. Isoliquiritigenin (ISL) is a bioactive compound that is insoluble in water and exhibits significant anti-TNBC activity. METHODS We previously prepared oral aqueous ISL@ZLH NPs; however, they were less stable in a freezing environment. Hence, the present study aimed to improve the stability of ISL@ZLH NPs using cryoprotectants that can withstand long storage times and are effective in TNBC treatment by creating an efficient oral drug delivery system. Freeze-dried ISL@ZLH NP powder was prepared by solvent evaporation, followed by the addition of trehalose and sucrose. The freeze-dried ISL@ZLH NP pow was optimized and characterized. The anti-TNBC efficacy and pharmacokinetics of the ISL@ZLH NP-pow were examined in plasma and organs, compared with those of aqueous ISL@ZLH NPs. RESULT The ideal particle size of the ISL@ZLH NP pow was 118 nm, which was not filtered out by the glomerulus and allowed the drug to be delivered to the lesions more effectively. Cellular uptake and biodistribution of the ISL@ZLH NP-pow in vivo and in vitro showed prolonged storage in the organs. In addition, cryopreserved ISL@ZLH NP-treated tumors showed significant anti- proliferative and anti-migratory effects through the downregulation of the PI3K-Akt-mToR and MMP2/9 signaling pathways. CONCLUSION These results suggest that oral ingestion of cryopreserved ISL@ZLH NP has the potential for long-term storage and can be employed as a clinical therapeutic approach to treat TNBC.
Collapse
Affiliation(s)
- Cong Xu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Cheng Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kumar Ganesan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chen Qui
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Department of Breast Oncology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Fei Gao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qingqing Liu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jianming Wu
- State Key Laboratory of Oncology in South China, Department of Breast Oncology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Yue Sui
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Peng Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Jinming Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianping Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
5
|
ZHANG M, CUI Y, YAO Y, GE Y, GAN J, JIN Y, SUN G. [Isoliquiritigenin Modulates the Effect of LINC01503
on Lung Squamous Carcinoma Cells]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:565-578. [PMID: 39318250 PMCID: PMC11425673 DOI: 10.3779/j.issn.1009-3419.2024.102.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND Isoliquiritigenin (ISL) is an important pharmacological constituent of Glycyrrhiza glabra, which possesses a range of physiological and pharmacological activities, as well as significant antitumor activity, and can be used as a potential drug for targeted cancer therapy. LINC01503 is an oncogene, which has been closely associated with the malignant biological processes of many cancers. The aim of this study was to investigate the effects of ISL on the proliferation, apoptosis, invasion and migration of lung squamous carcinoma cells by regulating LINC01503. METHODS Plasma was collected from lung squamous carcinoma patients and healthy individuals treated at Tangshan People's Hospital from January 2021 to December 2022. The expression of LINC01503 in lung squamous carcinoma plasma, tissues and cells was detected by real-time quantitative fluorescence polymerase chain reaction (qRT-PCR). Lung squamous carcinoma cells were treated with different concentrations of ISL for 24 h, and LINC01503 expression was detected by qRT-PCR. The cells were treated in groups: si-NC group, si-LINC01503 group, DMSO (0.1% dimethyl sulfone) group, ISL group, pc DNA3.1(+)-NC group, pc DNA3.1(+)-LINC01503 group, ISL+pc DNA3.1(+)-NC group and ISL+pc DNA3.1(+)- LINC01503 groups. CCK-8 assay, clone formation assay, flow cytometry, Transwell assay and scratch assay were used to explore the effect of LINC01503 on the functional phenotype of lung squamous carcinoma cells. RESULTS Fluorescence in situ hybridization results showed that the average fluorescence intensity of LINC01503 in tissue microarrays of lung squamous carcinoma patients was higher than that in paracancerous tissues (P<0.05). The expression of LINC01503 in the plasma of patients with lung squamous carcinoma was higher than that in the plasma of healthy individuals (P<0.05). Knockdown of LINC01503 inhibited the proliferation, invasion and migration of lung squamous carcinoma cells and promoted apoptosis (P<0.05). ISL inhibited the proliferation, invasion, migration and promoted apoptosis of lung squamous carcinoma cells (P<0.05). Overexpression of LINC01503 followed by intervention with ISL reversed the promotional effect of overexpression of LINC01503 on the proliferation, invasion and migration of lung squamous carcinoma cells as well as the inhibitory effect on apoptosis (P<0.05). CONCLUSIONS LINC01503 was highly expressed in lung squamous carcinoma, and LINC01503 could promote the proliferation, invasion and migration of lung squamous carcinoma cells and inhibit the apoptosis, ISL could inhibit the proliferation, invasion and migration of lung squamous carcinoma cells and promote apoptosis of lung squamous carcinoma cells by regulating the expression of LINC01503.
Collapse
|
6
|
Ji P, Zhao NS, Wu FL, Wei YM, Laba CD, Wujin CM, Hua YL, Yuan ZW, Yao WL. Mechanisms predictive of Tibetan Medicine Sophora moorcroftiana alkaloids for treatment of lung cancer based on the network pharmacology and molecular docking. BMC Complement Med Ther 2024; 24:47. [PMID: 38245694 PMCID: PMC10799429 DOI: 10.1186/s12906-024-04342-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 01/07/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Leguminous Sophora moorcroftiana (SM) is a genuine medicinal material in Tibet. Many research results have reveal the Sophora moorcroftiana alkaloids (SMA), as the main active substance, have a wide range of effects, such as antibacterial, antitumor and antiparasitic effects. However, there are few reports on the inhibition of lung cancer (LC) and its inhibitory mechanism, and the pharmacological mechanism of SMA is still unclear, Therefore, exploring its mechanism of action is of great significance. METHODS The SMA active components were obtained from the literature database. Whereas the corresponding targets were screened from the PubChem and PharmMapper database, UniProt database were conducted the correction and transformation of UniProt ID on the obtained targets. The GeneCards and OMIM databases identified targets associated with LC. Venny tools obtained the intersection targets of SMA and LC. R language and Cytoscape software constructed the visual of SMA - intersection targets - LC disease network. The intersection targets protein-protein interaction (PPI) network were built by the STRING database. The functions and pathways of the common targets of SMA and LC were enriched by gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Subsequently, molecular docking And A549 cells vitro experiment were performed to further validate our finding. RESULTS We obtained six kinds of alkaloids in SM, 635 potential targets for these compounds, and 1,303 genes related to LC. SMA and LC intersection targets was 33, including ALB, CCND1, ESR1, NOTCH1 and AR. GO enrichment indicated that biological process of SMA was mainly involved in the positive regulation of transcription and nitric oxide biosynthetic process, and DNA-templated, etc. Biological functions were mainly involved in transcription factor binding and enzyme binding, etc. Cell components were mainly involved in protein complexes, extracellular exosome, cytoplasm and nuclear chromatin, etc., Which may be associated with its anti-LC effects. KEGG enrichment analysis showed that main pathways involved in the anti-LC effects of SMA, including pathway in cancer, non small-cell lung cancer, p53, PI3K-Akt and FOXO signaling pathways. Molecular docking analyses revealed that the six active compounds had a good binding activity with the main therapeutic targets 2W96, 2CCH and 1O96. Experiments in vitro proved that SMA inhibited the proliferation of LC A549 cells. CONCLUSIONS Results of the present study, we have successfully revealed the SMA compounds had a multi-target and multi-channel regulatory mechanism in treatment LC, These findings provided a solid theoretical reference of SMA in the clinical treatment of LC.
Collapse
Affiliation(s)
- Peng Ji
- College of Veterinary Medicine of Gansu Agricultural University, Lanzhou, 730070, Gansu, China.
| | - Nian-Shou Zhao
- College of Veterinary Medicine of Gansu Agricultural University, Lanzhou, 730070, Gansu, China
| | - Fan-Lin Wu
- College of Veterinary Medicine of Gansu Agricultural University, Lanzhou, 730070, Gansu, China
| | - Yan-Ming Wei
- College of Veterinary Medicine of Gansu Agricultural University, Lanzhou, 730070, Gansu, China.
| | - Ci-Dan Laba
- Institute of Animal Sciences, Tibet Academy of Agricultural Sciences, Tibet Lhasa, 850009, China
| | - Cuo-Mu Wujin
- Institute of Animal Sciences, Tibet Academy of Agricultural Sciences, Tibet Lhasa, 850009, China
| | - Yong-Li Hua
- College of Veterinary Medicine of Gansu Agricultural University, Lanzhou, 730070, Gansu, China
| | - Zi-Wen Yuan
- College of Veterinary Medicine of Gansu Agricultural University, Lanzhou, 730070, Gansu, China
| | - Wan-Ling Yao
- College of Veterinary Medicine of Gansu Agricultural University, Lanzhou, 730070, Gansu, China
| |
Collapse
|
7
|
Sun F, Shen H, Liu Q, Chen Y, Guo W, Du W, Xu C, Wang B, Xing G, Jin Z, Lam JWY, Sun J, Ye R, Kwok RTK, Chen J, Tang BZ. Powerful Synergy of Traditional Chinese Medicine and Aggregation-Induced Emission-Active Photosensitizer in Photodynamic Therapy. ACS NANO 2023; 17:18952-18964. [PMID: 37729494 DOI: 10.1021/acsnano.3c04342] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Breast cancer (BC) remains a significant global health challenge for women despite advancements in early detection and treatment. Isoliquiritigenin (ISL), a compound derived from traditional Chinese medicine, has shown potential as an anti-BC therapy, but its low bioavailability and poor water solubility restrict its effectiveness. In this study, we created theranostic nanoparticles consisting of ISL and a near-infrared (NIR) photosensitizer, TBPI, which displays aggregation-induced emission (AIE), with the goal of providing combined chemo- and photodynamic therapies (PDT) for BC. Initially, we designed an asymmetric organic molecule, TBPI, featuring a rotorlike triphenylamine as the donor and 1-methylpyridinium iodide as the acceptor, which led to the production of reactive oxygen species in mitochondria. We then combined TBPI with ISL and encapsulated them in DSPE-PEG-RGD nanoparticles to produce IT-PEG-RGD nanoparticles, which showed high affinity for BC, better intersystem crossing (ISC) efficiency, and Förster resonance energy transfer (FRET) between TBPI and ISL. In both 4T1 BC cell line and a 4T1 tumor-bearing BC mouse model, the IT-PEG-RGD nanoparticles demonstrated excellent drug delivery, synergistic antitumor effects, enhanced tumor-killing efficacy, and reduced drug dosage and side effects. Furthermore, we exploited the optical properties of TBPI with ISL to reveal the release process and distribution of nanoparticles in cells. This study provides a valuable basis for further exploration of IT-PEG-RGD nanoparticles and their anticancer mechanisms, highlighting the potential of theranostic nanoparticles in BC treatment.
Collapse
Affiliation(s)
- Feiyi Sun
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science & Technology, Hong Kong 999077, China
| | - Hanchen Shen
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science & Technology, Hong Kong 999077, China
| | - Qingqing Liu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Yuyang Chen
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science & Technology, Hong Kong 999077, China
| | - Weihua Guo
- Department of Chemistry, State Key Laboratory of Marine Pollution, City University of Hong Kong, Hong Kong 999077, China
| | - Wutong Du
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science & Technology, Hong Kong 999077, China
| | - Changhuo Xu
- MOE Frontiers Science Center for Precision Oncology, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Bingzhe Wang
- Institute of Applied Physics and Materials Engineering, University of Macau, Macau 999078, China
| | - Guichuan Xing
- Institute of Applied Physics and Materials Engineering, University of Macau, Macau 999078, China
| | - Zhuwei Jin
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science & Technology, Hong Kong 999077, China
| | - Jacky W Y Lam
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science & Technology, Hong Kong 999077, China
| | - Jianwei Sun
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science & Technology, Hong Kong 999077, China
| | - Ruquan Ye
- Department of Chemistry, State Key Laboratory of Marine Pollution, City University of Hong Kong, Hong Kong 999077, China
| | - Ryan T K Kwok
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science & Technology, Hong Kong 999077, China
| | - Jianping Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Ben Zhong Tang
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science & Technology, Hong Kong 999077, China
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| |
Collapse
|
8
|
Kirdeeva Y, Fedorova O, Daks A, Barlev N, Shuvalov O. How Should the Worldwide Knowledge of Traditional Cancer Healing Be Integrated with Herbs and Mushrooms into Modern Molecular Pharmacology? Pharmaceuticals (Basel) 2022; 15:868. [PMID: 35890166 PMCID: PMC9320176 DOI: 10.3390/ph15070868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/04/2022] [Accepted: 07/11/2022] [Indexed: 12/04/2022] Open
Abstract
Traditional herbal medicine (THM) is a "core" from which modern medicine has evolved over time. Besides this, one third of people worldwide have no access to modern medicine and rely only on traditional medicine. To date, drugs of plant origin, or their derivates (paclitaxel, vinblastine, vincristine, vinorelbine, etoposide, camptothecin, topotecan, irinotecan, and omacetaxine), are very important in the therapy of malignancies and they are included in most chemotherapeutic regimes. To date, 391,000 plant and 14,000 mushroom species exist. Their medical and biochemical capabilities have not been studied in detail. In this review, we systematized the information about plants and mushrooms, as well as their active compounds with antitumor properties. Plants and mushrooms are divided based on the regions where they are used in ethnomedicine to treat malignancies. The majority of their active compounds with antineoplastic properties and mechanisms of action are described. Furthermore, on the basis of the available information, we divided them into two priority groups for research and for their potential of use in antitumor therapy. As there are many prerequisites and some examples how THM helps and strengthens modern medicine, finally, we discuss the positive points of THM and the management required to transform and integrate THM into the modern medicine practice.
Collapse
Affiliation(s)
- Yulia Kirdeeva
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (Y.K.); (O.F.); (A.D.)
| | - Olga Fedorova
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (Y.K.); (O.F.); (A.D.)
| | - Alexandra Daks
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (Y.K.); (O.F.); (A.D.)
| | - Nikolai Barlev
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (Y.K.); (O.F.); (A.D.)
- Orekhovich Institute of Biomedical Chemistry, 119435 Moscow, Russia
| | - Oleg Shuvalov
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (Y.K.); (O.F.); (A.D.)
- Orekhovich Institute of Biomedical Chemistry, 119435 Moscow, Russia
| |
Collapse
|
9
|
Mohamed LA, El Bolok AHM, Elgayar SF, Fahmy AN. miRNA-155 as a Novel Target for Isoliquiritigenin to Induce Autophagy in Oral Squamous Cell Carcinoma. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.8278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background and Aim :The most common obstacle facing chemotherapeutic agents is the development of drug resistance to cancer cells by dysregulation of autophagy and apoptosis. Targeting miRNAs by a natural flavonoid such as Isoliquiritigenin (ISL) is a novel strategy to reverse drug resistance. The aim of the present study was to evaluate ISL impacts on apoptosis and autophagy in oral squamous carcinoma cells (OSCC) through the expression levels of related two microRNAs: miRNA-21 and miRNA-155. Materials & Methods: The expression levels of both miRNAs were analysed using quantitative real time PCR and the effect of ISL on apoptosis was evaluated using annexin assay. In addition, the expression of the autophagy marker (ATG7) was measured using immunofluorescence. Results : Our results showed that ISL significantly downregulated both miRNA-21 and miRNA-155 with a fold change of 22.01 and 52.35, respectively. It also induced apoptosis in the cancer cells with high percentage (51.3 %). Moreover, ATG7 was highly expressed after ISL treatment. Conclusion : From this sudy we can conclude that ISL has an apoptotic and autophagic effect on OSCC through the down-regulation of miRNA-21 and miRNA-155, major regulators of PI3K/Akt pathway which can provide novel targets for OSCC therapy.
Collapse
|
10
|
Purnama A, Mardina V, Puspita K, Qanita I, Rizki DR, Hasballah K, Iqbal M, Sarong M. Molecular docking of two cytotoxic compounds from Calotropis gigantea leaves against therapeutic molecular target of pancreatic cancer. NARRA J 2021; 1:e37. [PMID: 38449465 PMCID: PMC10914070 DOI: 10.52225/narraj.v1i2.37] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 07/03/2021] [Indexed: 03/08/2024]
Abstract
The utilization of natural compounds as therapeutic agents to treat pancreatic cancer has recently focused on natural drug research. Calotropis gigantea has long been believed to be a medicinal plant that helps in treating various diseases. The bioactive compounds 9-metoxipinoresinol and isoliquiritigenin isolated from C. gigantea leaves are proven to act as therapeutic agents by inhibiting the cancer cell growth of Panc-1 cells. This study aimed to screen the potential molecular inhibition mechanisms of 9-metoxipinoresinol and isoliquiritigenin against pancreatic cancer development in-silico. We analyzed the activity of the aforementioned two compounds as inhibitors of several proteins that play a role in the growth of pancreatic cancer cells, such as GCNT3, GOT1, c-Met, PPARγ, BUB1, and NF-κβ, through molecular docking investigation. Our data suggested that 9-metoxipinoresinol and isoliquiritigenin were able to have well interaction with the target proteins, in which the predicted affinity energy ranged between -6.8 and 8.7 kcal/mol. The docking scores of 9-metoxipinoresinol and isoliquiritigenin were higher than the standard drug used (gemcitabine). Based on the binding affinity energy, GCNT3 and BUB1 are potentially to be used as target molecules for cancer therapy using 9-metoxipinoresinol and isoliquiritigenin, respectively.
Collapse
Affiliation(s)
- Agnia Purnama
- Department of Chemistry, Universitas Syiah Kuala, Banda Aceh, Aceh, Indonesia
| | - Vivi Mardina
- Department of Biology, Faculty of Engineering, Universitas Samudra, Langsa, Aceh, Indonesia
| | - Kana Puspita
- Department of Chemistry Education, Faculty of Education and Teacher Training, Universitas Syiah Kuala, Banda Aceh, Aceh, Indonesia
| | - Intan Qanita
- School of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, Indonesia
| | - Diva R. Rizki
- School of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, Indonesia
| | - Kartini Hasballah
- Department of Pharmacology, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, Indonesia
| | - Mudassar Iqbal
- Department of Agricultural Chemistry and Biochemistry, Agricultural University, Peshawar, Pakistan
| | - Murniana Sarong
- Department of Chemistry, Universitas Syiah Kuala, Banda Aceh, Aceh, Indonesia
| |
Collapse
|
11
|
Michalkova R, Mirossay L, Gazdova M, Kello M, Mojzis J. Molecular Mechanisms of Antiproliferative Effects of Natural Chalcones. Cancers (Basel) 2021; 13:cancers13112730. [PMID: 34073042 PMCID: PMC8198114 DOI: 10.3390/cancers13112730] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/20/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Despite the important progress in cancer treatment in the past decades, the mortality rates in some types of cancer have not significantly decreased. Therefore, the search for novel anticancer drugs has become a topic of great interest. Chalcones, precursors of flavonoid synthesis in plants, have been documented as natural compounds with pleiotropic biological effects including antiproliferative/anticancer activity. This article focuses on the knowledge on molecular mechanisms of antiproliferative action of chalcones and draws attention to this group of natural compounds that may be of importance in the treatment of cancer disease. Abstract Although great progress has been made in the treatment of cancer, the search for new promising molecules with antitumor activity is still one of the greatest challenges in the fight against cancer due to the increasing number of new cases each year. Chalcones (1,3-diphenyl-2-propen-1-one), the precursors of flavonoid synthesis in higher plants, possess a wide spectrum of biological activities including antimicrobial, anti-inflammatory, antioxidant, and anticancer. A plethora of molecular mechanisms of action have been documented, including induction of apoptosis, autophagy, or other types of cell death, cell cycle changes, and modulation of several signaling pathways associated with cell survival or death. In addition, blockade of several steps of angiogenesis and proteasome inhibition has also been documented. This review summarizes the basic molecular mechanisms related to the antiproliferative effects of chalcones, focusing on research articles from the years January 2015–February 2021.
Collapse
|
12
|
Veeraphan P, Chavasiri W, Muanprasat C, Chatsudthipong V, Yuajit C. A chalcone derivative retards renal cyst enlargement by inhibiting fluid secretion and cell proliferation in an in vitro model of polycystic kidney disease. Clin Exp Nephrol 2021; 25:944-952. [PMID: 34057612 DOI: 10.1007/s10157-021-02080-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 05/17/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Renal bilateral fluid filled-cyst in polycystic kidney disease (PKD) is associated with abnormal epithelial cell proliferation and transepithelial fluid secretion which leads to end-stage renal disease (ESRD). A chalcone derivative, isoliquiritigenin (ISLQ), has been shown to have various pharmacological properties. Since several studies have shown that ISLQ could inhibit CFTR channel activity, it is interesting to see whether it can inhibit renal cyst enlargement. The present study was aimed to determine an inhibitory effect and the mechanism of chalcone derivatives on MDCK cyst progression and Pkd1 mutant cells. METHODS MDCK cyst growth and cyst formation experiments, MTT assay, Ussing chamber experiment, BrdU cell proliferation assay and western blot analysis were performed in this study. RESULTS Among four compounds of chalcone derivatives tested, CHAL-005 (100 µM) was found to inhibit MDCK cyst growth in a dose-dependent manner without cytotoxicity. It inhibited short-circuit current of chloride secretion as well as CFTR protein expression in MDCK cells. CHAL-005 significantly suppressed cell proliferation. In addition, CHAL-005 strongly reduced phosphorylation ERK1/2 and phosphorylation S6 kinase in MDCK and Pkd1 mutant cells. Interestingly, CHAL-005 activated phosphorylation of AMP kinase protein expression in MDCK and Pkd1 mutant cells. CONCLUSION CHAL-005 slowed MDCK cyst progression by inhibiting CFTR expression and reducing ERK1/2 and mTOR/S6K signaling pathways as well as activating AMPK expression. Therefore, a chalcone derivative could represent as a promising drug candidate for polycystic kidney disease intervention.
Collapse
Affiliation(s)
- Peerachat Veeraphan
- Biomedical Science Program, College of Medicine and Public Health, Ubon Ratchathani University, Warin Chamrap, Ubon Ratchathani, 34190, Thailand
| | - Warinthorn Chavasiri
- Center of Excellence in Natural Products Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Phayathai Road, Phayathai, Bangkok, 10330, Thailand
| | - Chatchai Muanprasat
- Faculty of Medicine Ramathibodi Hospital, Chakri Naruebodindra Medical Institute, Mahidol University, Bang Phli, Samut Prakan, 10540, Thailand
| | - Varanuj Chatsudthipong
- Research Center of Transport Proteins for Medical Innovation, Faculty of Science, Mahidol University, Rama VI Road, Rajathevi, Bangkok, 10400, Thailand
| | - Chaowalit Yuajit
- College of Medicine and Public Health, Ubon Ratchathani University, Sathonlamark Road, Warin Chamrap, Ubon Ratchathani, 34190, Thailand.
| |
Collapse
|
13
|
Wang KL, Yu YC, Hsia SM. Perspectives on the Role of Isoliquiritigenin in Cancer. Cancers (Basel) 2021; 13:E115. [PMID: 33401375 PMCID: PMC7795842 DOI: 10.3390/cancers13010115] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 12/20/2022] Open
Abstract
Isoliquiritigenin (2',4',4-trihydroxychalcone, ISL), one of the most important bioactive compounds with a chalcone structure, is derived from licorice root. Licorice is commonly known as Glycyrrhiza, including Glycyrrhiza uralensis, Glycyrrhiza radix, and Glycyrrhiza glabra, which are generally available in common foods and Chinese herbal medicines based on a wide variety of biological functions and pharmacological effects, and its derivative (ISL) is utilized as a food additive and adjunct disease treatment. In this review, we summarized the progress over the last 10 years in the targeted pathways and molecular mechanisms of ISL that are involved in the regulation of the onset and progression of different types of cancers.
Collapse
Affiliation(s)
- Kai-Lee Wang
- Department of Nursing, Ching Kuo Institute of Management and Health, Keelung 20301, Taiwan;
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan
| | - Ying-Chun Yu
- Sex Hormonal Research Center, China Medical University Hospital, Taichung 40403, Taiwan;
- Department of Obstetrics and Gynecology, School of Medicine, China Medical University, Taichung 40403, Taiwan
| | - Shih-Min Hsia
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan
- School of Food and Safety, Taipei Medical University, Taipei 11031, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
| |
Collapse
|
14
|
Song L, Luo Y, Li S, Hong M, Wang Q, Chi X, Yang C. ISL Induces Apoptosis and Autophagy in Hepatocellular Carcinoma via Downregulation of PI3K/AKT/mTOR Pathway in vivo and in vitro. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:4363-4376. [PMID: 33116421 PMCID: PMC7585813 DOI: 10.2147/dddt.s270124] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022]
Abstract
Aims Isoliquiritigenin (ISL), a flavonoid from Glycyrrhiza glabra, has previously been reported to have anti-tumor effects in vivo and in vitro. However, the mechanisms whereby ISL exerts its anticancer effects remain poorly understood in hepatocellular carcinoma (HCC). Purpose In the present study, we investigated the anticancer efficacy and associated mechanisms of ISL in HCC MHCC97-H and SMMC7721 cells. Results We found that ISL inhibited cell viability and proliferation and induced apoptosis in a dose- and time-dependent manner in liver cancer lines. Furthermore, ISL could activate autophagy in HCC cells, and the autophagy inhibitor HCQ enhances ISL-induced apoptosis in HCC cells. Additionally, ISL induced apoptosis and autophagy through inhibition of the PI3K/Akt/mTOR pathway. Most importantly, in a xenograft tumor model in nude mice, data showed that the administration of ISL decreased tumor growth and concurrently promoted the expression of LC3-II and cleaved-caspase-3. Interestingly, we found that ISL inhibits mTOR by docking onto the ATP-binding pocket of mTOR (ie, it competes with ATP). We thus suggest that mTOR is a potential target for ISL inhibition of hepatocellular carcinoma development, which could be of interest for future investigations. Conclusion Taken together, the results reveal that ISL effectively inhibited proliferation and induced apoptosis in HCC through autophagy induction in vivo and in vitro, probably via the PI3K/Akt/mTOR pathway. ISL may be a potential therapeutic agent for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Lei Song
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, People's Republic of China.,The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, People's Republic of China
| | - Yi Luo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, People's Republic of China
| | - Shaoling Li
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, People's Republic of China
| | - Ming Hong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, People's Republic of China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, People's Republic of China
| | - Xiaoling Chi
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, People's Republic of China
| | - Cong Yang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, People's Republic of China
| |
Collapse
|
15
|
Govindammal M, Prasath M. Vibrational spectra, Hirshfeld surface analysis, molecular docking studies of (RS)-N,N-bis(2-chloroethyl)-1,3,2-oxazaphosphinan-2-amine 2-oxide by DFT approach. Heliyon 2020; 6:e04641. [PMID: 32904270 PMCID: PMC7452535 DOI: 10.1016/j.heliyon.2020.e04641] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 06/11/2020] [Accepted: 08/03/2020] [Indexed: 01/06/2023] Open
Abstract
The Cyclophosphamide (CYC) is used as an anti cancer agent. It is chemically known as (RS)-N,N-bis(2-chloroethyl)-1,3,2-oxazaphosphinan-2-amine 2-oxide. The vibrational assignments survey of the CYC was implemented by employing FT-IR and FT-Raman spectroscopic investigation and the results are compared with theoretical features. The optimized geometrical parameters, IR intensity and Raman Activity of the vibrational bands of CYC were determined from the B3LYP functional with 6-311++G (d, p) level of theory. In the current work, quantum chemical calculations were adopted to contemplate the vibrational assignments of CYC and the outcomes are compared with experimental findings. Molecular Electrostatic Potential (MEP) and HOMO-LUMO energies are very effective in the examination of charge transfer and distribution of the molecular structure. The molecular orbital contributions were evaluated by using the Total Density of States (TDOS). The analysis of Natural Bond Orbital (NBO), Mulliken population and Fukui function studies were done. Intermolecular interaction of the title compound was examined through Hirshfeld surface analysis. The evaluation of drug-likeness was accomplished in accordance with Lipinski's Rule of Five and molecular descriptors were utilized to predict the ADMET profiles of the CYC molecule. The recent research studies reports that the structural and bio-activity of the CYC was affirmed by the docking analysis of CYC with protein PI3K/AKT inhibitor, it acts as anti-lung cancer agent.
Collapse
Affiliation(s)
- M. Govindammal
- Department of Physics, Periyar University PG Extension Centre, Dharmapuri, 636701, India
| | - M. Prasath
- Department of Physics, Periyar University PG Extension Centre, Dharmapuri, 636701, India
| |
Collapse
|
16
|
Huang P, Du SX. Puerarin Enhances the Anti-Tumor Effect of Cisplatin on Drug-Resistant A549 Cancer in vivo and in vitro Through Activation of the Wnt Signaling Pathway. Cancer Manag Res 2020; 12:6279-6289. [PMID: 32801873 PMCID: PMC7399457 DOI: 10.2147/cmar.s253327] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/23/2020] [Indexed: 12/24/2022] Open
Abstract
Objective The effect of PUE on enhancing the anti-cancerous efficacy of DDP on drug-resistant A549/DDP cancer and the underlying mechanisms were thoroughly investigated. Materials and Methods The cytotoxicity of PUE, DDP, and PUE + DDP to A549 cells and A549/DDP cells, respectively, is determined by cell apoptosis experiments. Anti-proliferation effect of PUE, DDP, and PUE + DDP on A549 cells and A549/DDP cells is evaluated by the cell cloning assay. Qualitative and quantitative analysis of the levels of PUE, DDP, and PUE + DDP of cell proliferation-related genes and proteins expressions in A549/DDP cells are determined by Western blot assay. The levels of VEGF in A549/DDP cells after different treatment strategies are determined by ELISA assay. Qualitative and quantitative determination of VEGF expression in tumor tissues are done by immunohistochemical staining. Results In vitro cellular experiments revealed that co-incubation of A549/DDP cells with PUE and DDP led to a dramatically decreased cell viability and cell survival rate compared with the cells only treated by DDP. Such a stimulating effect of PUE on DDP was further confirmed in vivo with results shown that the A549/DDP cancer-bearing mice treaded by combination therapy achieved the lowest tumor growth rate and longest survival time. Conclusion Taking these results together, we can draw the conclusion that the PUE enhances the anti-tumor effect of DDP on the drug-resistant A549 cancer in vivo and in vitro through activation of the Wnt signaling pathway.
Collapse
Affiliation(s)
- Ping Huang
- Department of Respiratory, Caoxian People's Hospital, Heze 274400, Shandong, People's Republic of China
| | - Shi-Xia Du
- Department of Respiratory, Caoxian People's Hospital, Heze 274400, Shandong, People's Republic of China
| |
Collapse
|
17
|
Li R, Song Y, Zhou L, Li W, Zhu X. Downregulation of RAGE Inhibits Cell Proliferation and Induces Apoptosis via Regulation of PI3K/AKT Pathway in Cervical Squamous Cell Carcinoma. Onco Targets Ther 2020; 13:2385-2397. [PMID: 32256089 PMCID: PMC7093096 DOI: 10.2147/ott.s240378] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 03/11/2020] [Indexed: 12/15/2022] Open
Abstract
AIM The receptor for advanced glycation endproducts (RAGE) expression has been reported to be implicated with cancer development. In this study, the role of RAGE in the regulation of cervical squamous cancer cell proliferation, apoptosis and the mechanism of RAGE involved in the biological behaviors were explored. METHODS The RAGE expression was overexpressed or downregulated by lentivirus transfection. The effect of RAGE expression on cell proliferation was explored by CCK-8, MTT, and BrdU assay, and the effect of RAGE on tumor development was confirmed by the xenograft mouse model along with the immunohistochemistry stain of proliferating cell nuclear antigen (PCNA). Apoptosis was investigated by flow cytometry and TUNEL assay. Western blotting was performed to investigate the expression of possible proteins, including Bax, Bcl-2, PI3K, p-PI3K, AKT, and p-AKT. RESULTS Overexpression of RAGE promoted proliferation of cervical squamous cancer cell and increased PCNA expression. In the meantime, RAGE overexpression inhibited cell apoptosis along with a decrease of Bax/Bcl-2 ratio, and induction of PI3K/AKT activation. The in vivo results showed that overexpression of RAGE enhanced tumor growth. Conversely, knockdown of RAGE exhibited opposed effects on cervical cancer cells and xenograft mouse model. Furthermore, RAGE inhibitor FPS-ZM1 effectively inhibited SiHa cell viability and PCNA expression, and increased cell apoptosis and Bax/Bcl-2 ratio. Moreover, PI3K inhibitor LY294002 effectively inhibited activation of PI3K and AKT, and further repressed RAGE overexpression-induced cell proliferation and apoptosis inhibition. CONCLUSION RAGE promotes the growth ability of cervical squamous cell carcinoma by inducing PCNA expression and inhibiting cell apoptosis via inactivation of the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Ruyi Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325027, People’s Republic of China
| | - Yizuo Song
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325027, People’s Republic of China
| | - Lulu Zhou
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325027, People’s Republic of China
| | - Weibo Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325027, People’s Republic of China
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325027, People’s Republic of China
| |
Collapse
|
18
|
Lin PH, Chiang YF, Shieh TM, Chen HY, Shih CK, Wang TH, Wang KL, Huang TC, Hong YH, Li SC, Hsia SM. Dietary Compound Isoliquiritigenin, an Antioxidant from Licorice, Suppresses Triple-Negative Breast Tumor Growth via Apoptotic Death Program Activation in Cell and Xenograft Animal Models. Antioxidants (Basel) 2020; 9:228. [PMID: 32164337 PMCID: PMC7139602 DOI: 10.3390/antiox9030228] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/16/2020] [Accepted: 03/07/2020] [Indexed: 02/07/2023] Open
Abstract
Patients with triple-negative breast cancer have few therapeutic strategy options. In this study, we investigated the effect of isoliquiritigenin (ISL) on the proliferation of triple-negative breast cancer cells. We found that treatment with ISL inhibited triple-negative breast cancer cell line (MDA-MB-231) cell growth and increased cytotoxicity. ISL reduced cell cycle progression through the reduction of cyclin D1 protein expression and increased the sub-G1 phase population. The ISL-induced apoptotic cell population was observed by flow cytometry analysis. The expression of Bcl-2 protein was reduced by ISL treatment, whereas the Bax protein level increased; subsequently, the downstream signaling molecules caspase-3 and poly ADP-ribose polymerase (PARP) were activated. Moreover, ISL reduced the expression of total and phosphorylated mammalian target of rapamycin (mTOR), ULK1, and cathepsin B, whereas the expression of autophagic-associated proteins p62, Beclin1, and LC3 was increased. The decreased cathepsin B cause the p62 accumulation to induce caspase-8 mediated apoptosis. In vivo studies further showed that preventive treatment with ISL could inhibit breast cancer growth and induce apoptotic and autophagic-mediated apoptosis cell death. Taken together, ISL exerts an effect on the inhibition of triple-negative MDA-MB-231 breast cancer cell growth through autophagy-mediated apoptosis. Therefore, future studies of ISL as a supplement or alternative therapeutic agent for clinical trials against breast cancer are warranted.
Collapse
Affiliation(s)
- Po-Han Lin
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; (P.-H.L.); (Y.-F.C.); (H.-Y.C.); (C.-K.S.); (S.-C.L.)
| | - Yi-Fen Chiang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; (P.-H.L.); (Y.-F.C.); (H.-Y.C.); (C.-K.S.); (S.-C.L.)
| | - Tzong-Ming Shieh
- School of Dentistry, College of Dentistry, China Medical University, Taichung 40402, Taiwan;
- Department of Dental Hygiene, College of Health Care, China Medical University, Taichung 40402, Taiwan
| | - Hsin-Yuan Chen
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; (P.-H.L.); (Y.-F.C.); (H.-Y.C.); (C.-K.S.); (S.-C.L.)
| | - Chun-Kuang Shih
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; (P.-H.L.); (Y.-F.C.); (H.-Y.C.); (C.-K.S.); (S.-C.L.)
| | - Tong-Hong Wang
- Tissue Bank, Chang Gung Memorial Hospital, Tao-Yuan 33305, Taiwan;
- Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Tao-Yuan 33305, Taiwan
| | - Kai-Lee Wang
- Department of Nursing, Ching Kuo Institute of Management and Health, Keelung City 20301, Taiwan;
| | - Tsui-Chin Huang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
| | - Yong-Han Hong
- Department of Nutrition, I-Shou University, Kaohsiung City 82445, Taiwan;
| | - Sing-Chung Li
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; (P.-H.L.); (Y.-F.C.); (H.-Y.C.); (C.-K.S.); (S.-C.L.)
| | - Shih-Min Hsia
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; (P.-H.L.); (Y.-F.C.); (H.-Y.C.); (C.-K.S.); (S.-C.L.)
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan
- School of Food Safety, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
| |
Collapse
|
19
|
Isoliquiritigenin Inhibits Ovarian Cancer Metastasis by Reversing Epithelial-to-Mesenchymal Transition. Molecules 2019; 24:molecules24203725. [PMID: 31623144 PMCID: PMC6833095 DOI: 10.3390/molecules24203725] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/26/2019] [Accepted: 10/15/2019] [Indexed: 12/11/2022] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) plays a prominent role in cancer metastasis. Isoliquiritigenin (ISL), one of the flavonoids in licorice, has been shown to exhibit anticancer activities in many cancer types through various mechanisms. However, it is unknown whether ISL impacts the EMT process. Here, we show that ISL is able to suppress mesenchymal features of ovarian cancer SKOV3 and OVCAR5 cells, evidenced by an apparent morphological change from a mesenchymal to an epithelial phenotype and reduced levels of mesenchymal markers accompanied by the gain of E-cadherin expression. The suppression of EMT is also supported by the observed decrease in cell migration and in vitro invasion upon ISL treatment. Moreover, we show that ISL effectively blocks the intraperitoneal xenograft development of the SKOV3 cell line and prolonged the survival of tumor-bearing mice. These data suggest that ISL inhibits intraperitoneal ovary tumor development through the suppression of EMT, indicating that ISL may be an effective therapeutic agent against ovarian cancer.
Collapse
|