1
|
Lei X, Wang F, Zhang X, Huang J, Huang Y. The potential mechanisms by which Xiaoyao Powder may exert therapeutic effects on thyroid cancer were examined at various levels. Comput Biol Chem 2025; 117:108412. [PMID: 40056710 DOI: 10.1016/j.compbiolchem.2025.108412] [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: 12/06/2024] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Thyroid cancer (TC) is the most prevalent endocrine malignancy, with a rising incidence necessitating safer treatment strategies to reduce overtreatment and its side effects. Xiaoyao Powder (XYP), a widely used herbal formula, shows promise in treating TC. This study aims to investigate the mechanisms by which XYP may affect TC. METHODS The components of XYP were identified through database retrieval, and targets related to TC were collected to construct a target network for key screening. GEO dataset samples analyzed immune cells and identified significantly differentially expressed core genes (SDECGs). Based on SDECG expression and clustering, samples were classified for comparison. WGCNA was employed to identify gene modules linked to clinical characteristics. ML models screened characteristic genes and constructed a nomogram validated using another GEO dataset. MR methods explored causal relationships between genes and TC. RESULTS The top ten active components of XYP were identified, along with 27 SDECGs that exhibited significant differences in immune cell infiltration between TC patients and normal controls. The nomogram effectively predicted TC risk, validated through ROC curves. Key characteristic genes included SMIM1, PPP1R16A, KIAA1462, DNAJC22, and EFNA5. CONCLUSION XYP may treat TC by regulating SMIM1, PPP1R16A, KIAA1462, DNAJC22, EFNA5, and associated immune pathways; this provides theoretical support for its potential mechanisms.
Collapse
Affiliation(s)
- Xiaoli Lei
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Feifei Wang
- Department of Quality Control, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng, China
| | - Xinying Zhang
- Department of Endocrinology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiaxi Huang
- Department of Pharmacy, Huoqiu County First People's Hospital, Liuan, China
| | - Yanqin Huang
- Department of Endocrinology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
2
|
Liu F, Guo C, Liu X, Gu Z, Zou W, Tang X, Tang J. Luteolin in Inflammatory Bowel Disease and Colorectal Cancer: A Disease Continuum Perspective. Curr Issues Mol Biol 2025; 47:126. [PMID: 39996847 PMCID: PMC11853781 DOI: 10.3390/cimb47020126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic intestinal inflammatory condition that may progress to colorectal cancer (CRC), presenting significant challenges to global health. With shifts in lifestyle, the incidence of both conditions continues to rise, underscoring the urgent need for effective treatments. While traditional therapies can be effective, their high recurrence rates and associated adverse reactions limit their broader application. Luteolin, a flavonoid derived from natural plants, has emerged as a promising focus in both IBD and CRC research due to its multi-target therapeutic potential. This article reviews the molecular mechanisms and signaling pathways through which luteolin regulates immune cell differentiation, mitigates inflammation and oxidative stress, modulates gut microbiota, and restores intestinal mucosal barrier function in IBD. In the context of CRC, luteolin demonstrates significant anti-tumor effects by inhibiting cancer cell proliferation, inducing apoptosis, and suppressing cell migration and invasion. Notably, luteolin has demonstrated significant improvements in IBD symptoms by influencing the differentiation of T cell subsets, decreasing the expression of inflammatory mediators, activating antioxidant pathways, and enhancing the structure of gut microbiota. Furthermore, advancements in formulation technology, such as the use of polymer micelles and responsive nanoparticles, have greatly improved the bioavailability and efficacy of luteolin. However, further investigation is needed to address the bioavailability and potential toxicity of luteolin, particularly in the critical transition from IBD to CRC. This article emphasizes the potential of luteolin in the treatment of IBD and CRC and anticipates its promising prospects for future clinical applications as a natural therapeutic agent.
Collapse
Affiliation(s)
- Fang Liu
- Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China; (F.L.); (C.G.)
- Clinical Medicine College of Integrated Chinese and Western Medicine, North Sichuan Medical College, Nanchong 637100, China; (X.L.); (Z.G.); (W.Z.)
| | - Cui Guo
- Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China; (F.L.); (C.G.)
| | - Xue Liu
- Clinical Medicine College of Integrated Chinese and Western Medicine, North Sichuan Medical College, Nanchong 637100, China; (X.L.); (Z.G.); (W.Z.)
| | - Zhili Gu
- Clinical Medicine College of Integrated Chinese and Western Medicine, North Sichuan Medical College, Nanchong 637100, China; (X.L.); (Z.G.); (W.Z.)
| | - Wenxuan Zou
- Clinical Medicine College of Integrated Chinese and Western Medicine, North Sichuan Medical College, Nanchong 637100, China; (X.L.); (Z.G.); (W.Z.)
| | - Xuegui Tang
- Clinical Medicine College of Integrated Chinese and Western Medicine, North Sichuan Medical College, Nanchong 637100, China; (X.L.); (Z.G.); (W.Z.)
| | - Jianyuan Tang
- Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China; (F.L.); (C.G.)
| |
Collapse
|
3
|
Ren F, Li Y, Luo H, Gao S, Jiang S, Yang J, Rao C, Chen Y, Peng C. Extraction, detection, bioactivity, and product development of luteolin: A review. Heliyon 2024; 10:e41068. [PMID: 39759280 PMCID: PMC11700251 DOI: 10.1016/j.heliyon.2024.e41068] [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: 06/15/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 01/07/2025] Open
Abstract
Luteolin is a kind of natural flavonoid, widely existing in a variety of plants, has been revealed to have a wide range of biological activities. In recent years, the research results of luteolin are abundant. Here we review the latest research results of luteolin in order to provide new ideas for further research and development of luteolin. In this paper, the focus of the search was published between 2010 and 2024 on the extraction and determination of luteolin, biological activities, and the development and application of luteolin products. A comprehensive search using the keyword "luteolin" was conducted in the PubMed, Web of Science and WIPO databases. Through the collection of related literature, this paper summarized a variety of extraction techniques of luteolin, including immersion extraction, solvent extraction, ultrasonic-assisted extraction, supercritical fluid extraction and so on. The determination methods include: thin layer chromatography (TLC), high performance liquid chromatography (HPLC), capillary electrophoresis (CE), electrochemical method (ED) and so on. In addition, the biological activities of luteolin, including antioxidant, anti-inflammatory, anti-tumor, antibacterial, analgesic and so on, were described. And luteolin as the main component of the product is being gradually developed, and has been used in the field of food, medicine and cosmetics. This paper provides a reference for further study of luteolin.
Collapse
Affiliation(s)
- Fajian Ren
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Ying Li
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Hanyuan Luo
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Song Gao
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Shanshan Jiang
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Jian Yang
- Chuan-chu UNITED INTERNATIONAL Engineering Co., LTD, Chengdu, China
| | - Chaolong Rao
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Yan Chen
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Cheng Peng
- State Key Laboratory of Traditional Chinese Medicine Resources in Southwest China, Chengdu, 611137, China
| |
Collapse
|
4
|
Wang J, Li H, Wang Z, Ruan S. Luteolin: A Comprehensive and Visualized Analysis of Research Hotspots and Its Antitumor Mechanisms. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:2377-2401. [PMID: 39686791 DOI: 10.1142/s0192415x24500903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
The aim of this study was to analyze the research hotspots and mechanisms of luteolin in tumor-related fields using bibliometric and bioinformatic approaches to guide future research. We conducted a comprehensive screening of all articles on luteolin and tumors in Web of Science from 2008 to 2023. The extracted words from these publications were visualized using VOSviewer, Scimago Graphica, and CiteSpace. Public databases were used to collect luteolin and tumor-related targets. GO and KEGG analyses of luteolin antitumor-related genes were performed using Metascape. Protein interaction networks were built with Cytoscape and STRING, identifying hub targets of luteolin in antitumor activity. Subsequently, the binding capacity of luteolin to these hub targets was assessed using molecular docking technology. We found that China dominated this field, the Egyptian Knowledge Bank published the most articles, and Molecules had the highest number of related publications. Recently, network pharmacology, target, traditional Chinese medicine, and nanoparticles have become research hotspots in luteolin's antitumor research. A total of 483 overlapping genes between luteolin and tumors were identified, and they were closely associated with the cancer-associated pathways, PI3K-Akt, and MAPK signaling pathways. Luteolin forms a complex network of antitumor-related genes, with TP53, TNF, STAT3, AKT1, JUN, IL6, and SRC playing key roles and showing strong binding affinity to luteolin. Computer technology is becoming increasingly integral to the discipline, and future research will focus on more precise antitumor mechanisms and effective clinical applications.
Collapse
Affiliation(s)
- Jiaxuan Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, P. R. China
| | - Hao Li
- The First Affiliated Hospital of Zhejiang Chinese Medical University, (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, P. R. China
| | - Zhenru Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, P. R. China
| | - Shanming Ruan
- The First Affiliated Hospital of Zhejiang Chinese Medical University, (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, P. R. China
| |
Collapse
|
5
|
Shi L, Zhang X, Mao L, Zhang Y. Anti-neoplastic effect of heterophyllin B on ovarian cancer via the regulation of NRF2/HO-1 in vitro and in vivo. Tissue Cell 2024; 91:102566. [PMID: 39341007 DOI: 10.1016/j.tice.2024.102566] [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: 05/24/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Heterophyllin B (HB) is a cyclic peptide with anti-neoplastic effect on many cancers. However, its effect and mechanism of action in ovarian cancer cells are still unknown. PURPOSE The primary objective of this study was to assess the impact of HB on the proliferation of ovarian cancer (OC) cells and delve into the underlying mechanisms involved. METHODS We performed CCK-8 assays, HE staining, KI67 staining, clonogenic formation assays, Annexin V-FITC/PI staining, tumor invasion assays, and migration assays to detect the effects of HB on cell viability, proliferation, apoptosis, migration, and invasion in ovarian cancer cells. Additionally, real-time fluorescent quantitative PCR (qPCR) and Western blotting were utilized for verification. The expression of NF-E2-related factor 2 (NRF2) and heme oxygenase 1 (HMOX1/HO-1) signaling molecules was detected using qPCR and Western blotting. A specific inducer, Hemin, was used to activate HO-1 and Nrf2 overexpression, in order to verify the pharmacological mechanism of HB on ovarian cancer cells. The binding relationship between HB and NRF2 was investigated through molecular docking. RESULTS HB treatment inhibited the viability of OC cells, meanwhile it showed suppressive effect on the proliferation, migration, and invasion of OC cells, Meanwhile, HB could promote the apoptosis of tumor cells. For the mechanisms, we found that HB treatment could significantly down-regulate the levels of NRF2/HO-1. Consistent with the results of in vitro experiments, administration of HB significantly delayed tumor growth in OVCAR8 xenografted nude mice, and inhibited the expression of Ki67, Nrf2 and HO-1. CONCLUSION This study demonstrated that HB had anti-neoplastic effect on OC by inhibiting Nrf2/HO-1 signaling pathway and may be a potential drug for the treatment of OC.
Collapse
Affiliation(s)
- Linyu Shi
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, No.20, Xisi Road, Nantong, Jiangsu Province 226001, China
| | - Xiaoyu Zhang
- Department of Gastroenterology and Hepatology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Liming Mao
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China; Basic Medical Research Center, School of Medicine, Nantong University, Nantong 226019, China
| | - Yuquan Zhang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, No.20, Xisi Road, Nantong, Jiangsu Province 226001, China.
| |
Collapse
|
6
|
Zhang J, Li C, Li W, Shi Z, Liu Z, Zhou J, Tang J, Ren Z, Qiao Y, Liu D. Mechanism of luteolin against non-small-cell lung cancer: a study based on network pharmacology, molecular docking, molecular dynamics simulation, and in vitro experiments. Front Oncol 2024; 14:1471109. [PMID: 39582546 PMCID: PMC11582065 DOI: 10.3389/fonc.2024.1471109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/21/2024] [Indexed: 11/26/2024] Open
Abstract
Introduction Luteolin, a naturally occurring flavonoid compound, demonstrates promising anti-cancer properties. However, its mechanism against non-small-cell lung cancer (NSCLC) remains unknown. This study employed network pharmacology, molecular docking, molecular dynamics simulation (MDS), and in vitro experiments to investigate the potential mechanisms by which luteolin against NSCLC. Methods Initially, the potential targets of luteolin and NSCLC-related targets were identified from public databases such as TCMSP, GeneCards, OMIM, DrugBank, and TTD. Subsequently, the protein-protein interaction (PPI) network screening and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted. The binding affinity and stability of luteolin with the core targets were assessed using molecular docking and MDS. Finally, the results were validated by in vitro experiments. Results A total of 56 luteolin targets and 2145 NSCLC-related targets were identified. Six core targets, TP53, EGFR, AKT1, TNF, JUN, and CASP3, were screened via the PPI network. The GO and KEGG analyses indicated that luteolin's activity against NSCLC potentially involves PI3K-Akt, NF-kappa B, and other signaling pathways. Molecular docking revealed that luteolin had high binding affinity with the core targets. MDS confirmed the stable interaction between luteolin and key proteins TP53 and AKT1. in vitro, luteolin significantly inhibited the proliferation and migration of A549 cells, while also inducing apoptosis. In addition, luteolin downregulated the expression of p-Akt (Ser473), MDM2, and Bcl-2 but upregulated the expression of p53 and Bax, which was consistent with the effect of LY294002. Conclusion Luteolin had a good anti-NSCLC effect, and the apoptosis-inducing effect might be related to the Akt/MDM2/p53 signaling pathway.
Collapse
Affiliation(s)
- Jihang Zhang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
- Research Center for Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Changling Li
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Wenyi Li
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhenpeng Shi
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhenguo Liu
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Junyu Zhou
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jing Tang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zixuan Ren
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yun Qiao
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Deshan Liu
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
7
|
Guo L, Zhou Y, Ma R. Exploring the anti-gastric cancer mechanism of action of Bidentis Bipinnatae Herba based on network pharmacology, molecular docking, and cellular experimental validation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8681-8690. [PMID: 38822119 DOI: 10.1007/s00210-024-03169-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/16/2024] [Indexed: 06/02/2024]
Abstract
The purpose of this study is to explore the potential molecular mechanism of Bidentis Bipinnatae Herba against gastric cancer by using network pharmacology methods, molecular docking, and cellular experimental validation. Medicinal plants related to gastric cancer were queried through TCMSP, SymMap, and Herb databases. The TCSMP database (drug-likeness ≥ 0.18) was used to retrieve the bioactive constituents. TCSMP, SwissTargetPrediction, and Herb databases were used to retrieve the target genes, and Cytoscape software was used to construct the "active ingredient-target" network. After protein interaction analysis using String 11.0 platform, the hub genes were screened using CytoHubba. The obtained hub genes were uploaded to the cBioPortal for pathway enrichment. The genes involved in gastric cancer-related RTK-RAS pathway were molecularly docked and experimentally validated. Bidentis Bipinnatae Herba was common to TCMSP, SymMap, and Herb databases. A total of nine active ingredients were obtained in Bidentis Bipinnatae Herba, acting on 192 targets. Seven hub genes were obtained from these target genes and enriched in the RTK-RAS pathway in gastric cancer. MAPK1 and EGFR had good molecular docking results with their corresponding chemicals. Cellular experiments showed that the treatment of luteolin, quercetin, and Okanin reduced the expression of EGFR in AGS cells; the treatment of luteolin and quercetin could reduce the expression of MAPK1. Bidentis Bipinnatae Herba contained active components, which may be anti-gastric cancer in a multi-target (MAPK1 and EGFR) manner.
Collapse
Affiliation(s)
- Linglong Guo
- Department of Oncology, Yizheng Hospital of TCM, Yizheng, 211400, China
| | - Yuyi Zhou
- Graduate School, Zhejiang Chinese Medical University, Zhejiang, 310053, China
| | - Rui Ma
- Department of Traditional Chinese Medicine, Nanjing Luhe People's Hospital, No. 9, Jiankang Lane, Xiongzhou Street, Luhe District, Nanjing, 211500, China.
| |
Collapse
|
8
|
Rong S, Dai B, Yang C, Lan Z, Wang L, Xu L, Chen W, Chen J, Wu Z. HNRNPC modulates PKM alternative splicing via m6A methylation, upregulating PKM2 expression to promote aerobic glycolysis in papillary thyroid carcinoma and drive malignant progression. J Transl Med 2024; 22:914. [PMID: 39380010 PMCID: PMC11459990 DOI: 10.1186/s12967-024-05668-9] [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: 03/19/2024] [Accepted: 09/04/2024] [Indexed: 10/10/2024] Open
Abstract
The heterogeneous nuclear ribonucleoprotein C (HNRNPC) plays a crucial role in tumorigenesis, yet its role in papillary thyroid carcinoma (PTC) remains elusive. Herein, we elucidated the function and molecular mechanism of HNRNPC in PTC tumorigenesis and progression. Our study unveiled a significant upregulation of HNRNPC in PTC, and knockdown of HNRNPC markedly inhibited the proliferation, invasion, and metastasis of BCPAP cells. Furthermore, HNRNPC modulated PKM alternative splicing in BCPAP cells primarily through m6A modification. Additionally, by upregulating PKM2 expression, HNRNPC promoted aerobic glycolysis in BCPAP cells, thereby facilitating malignant progression in PTC. In summary, our findings demonstrate that HNRNPC regulates PKM alternative splicing through m6A methylation modification and promotes the proliferation, invasion and metastasis of PTC through glucose metabolism pathways mediated by PKM2. These discoveries provide new biomarkers for screening and diagnosing PTC patients and offer novel therapeutic targets for personalized treatment strategies.
Collapse
Affiliation(s)
- Shikuo Rong
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.
- Department of Thyroid Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China.
| | - Bao Dai
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Chunrong Yang
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Ziteng Lan
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Linhe Wang
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Lei Xu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Weijian Chen
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Jian Chen
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.
| | - Zeyu Wu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
9
|
Wang R, Li X, Xu Y, Li Y, Zhang W, Guo R, Song J. Progress, pharmacokinetics and future perspectives of luteolin modulating signaling pathways to exert anticancer effects: A review. Medicine (Baltimore) 2024; 103:e39398. [PMID: 39183411 PMCID: PMC11346905 DOI: 10.1097/md.0000000000039398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/27/2024] Open
Abstract
Luteolin (3, 4, 5, 7-tetrahydroxyflavone) are natural flavonoids widely found in vegetables, fruits and herbs, with anti-tumor, anti-inflammatory and antioxidant effects, and also play an anti-cancer effect in various cancers such as lung, breast, prostate, and liver cancer, etc. Specifically, the anti-cancer mechanism includes regulation of various signaling pathways to induce apoptosis of tumor cells, inhibition of tumor cell proliferation and metastasis, anti-angiogenesis, regulation of immune function, synergistic anti-cancer drugs and regulation of reactive oxygen species levels of tumor cells. Specific anti-cancer mechanisms include regulation of various signaling pathways to induce apoptosis, inhibition of tumor cell proliferation and metastasis, anti-angiogenesis, reversal of epithelial-mesenchymal transition, regulation of immune function, synergism with anti-cancer drugs and regulation of reactive oxygen species levels in tumor cells. This paper integrates the latest cutting-edge research on luteolin and combines it with the prospect of future clinical applications, aiming to explore the mechanism of luteolin exerting different anticancer effects through the regulation of different signaling pathways, so as to provide a practical theoretical basis for the use of luteolin in clinical treatment and hopefully provide some reference for the future research direction of luteolin.
Collapse
Affiliation(s)
- Rui Wang
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
- Medical School of Nantong University, Nantong, PR China
| | - Xia Li
- Department of General Medicine, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
| | - Yanhan Xu
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
| | - Yangyang Li
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
| | - Weisong Zhang
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
| | - Rongqi Guo
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
| | - Jianxiang Song
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
| |
Collapse
|
10
|
Cai J, Tan X, Hu Q, Pan H, Zhao M, Guo C, Zeng J, Ma X, Zhao Y. Flavonoids and Gastric Cancer Therapy: From Signaling Pathway to Therapeutic Significance. Drug Des Devel Ther 2024; 18:3233-3253. [PMID: 39081701 PMCID: PMC11287762 DOI: 10.2147/dddt.s466470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Gastric cancer (GC) is a prevalent gastrointestinal tumor characterized by high mortality and recurrence rates. Current treatments often have limitations, prompting researchers to explore novel anti-tumor substances and develop new drugs. Flavonoids, natural compounds with diverse biological activities, are gaining increasing attention in this regard. We searched from PubMed, Web of Science, SpringerLink and other databases to find the relevant literature in the last two decades. Using "gastric cancer", "stomach cancers", "flavonoid", "bioflavonoid", "2-Phenyl-Chromene" as keywords, were searched, then analyzed and summarized the mechanism of flavonoids in the treatment of GC. It was revealed that the anti-tumor mechanism of flavonoids involves inhibiting tumor growth, proliferation, invasion, and metastasis, as well as inducing cell death through various processes such as apoptosis, autophagy, ferroptosis, and pyroptosis. Additionally, combining flavonoids with other chemotherapeutic agents like 5-FU and platinum compounds can potentially reduce chemoresistance. Flavonoids have also demonstrated enhanced biological activity when used in combination with other natural products. Consequently, this review proposes innovative perspectives for the development of flavonoids as new anti-GC agents.
Collapse
Affiliation(s)
- Jiaying Cai
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Xiyue Tan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Qichao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Huafeng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Maoyuan Zhao
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Cui Guo
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Jinhao Zeng
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Yanling Zhao
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
11
|
Chen P, Wu HY. Network pharmacology- and molecular docking-based exploration of the molecular mechanism underlying Jianpi Yiwei Recipe treatment of gastric cancer. World J Gastrointest Oncol 2024; 16:2988-2998. [PMID: 39072163 PMCID: PMC11271781 DOI: 10.4251/wjgo.v16.i7.2988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/26/2024] [Accepted: 05/14/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Traditional Chinese medicine (TCM) is widely used as an important complementary and alternative healthcare system for cancer treatment in Asian countries. Network pharmacology, which utilizes various database platforms and computer software to study the interactions between complex drug components in vivo, is particularly useful for studying the pharmacodynamic mechanisms of multi-pathway and multi-target Chinese medicines. AIM To explore the potential targets and function of Jianpi Yiwei Recipe treatment of gastric cancer (GC) through network pharmacology and molecular docking. METHODS Data on the components of Jianpi Yiwei Recipe (Radix Astragali, Radix Codonopsis, Agrimonia eupatoria, Atractylodes macrocephala Koidz., Poria cocos, stir-baked rhizoma dioscoreae, Amomum villosum Lour., fried Fructus Aurantii, pericarpium citri reticulatae, Rhizoma Pinelliae Preparata, and Radix Glycyrrhizae Preparata) were collected and screened by using the TCM systems pharmacology database and analysis platform (TCMSP). Then the targets of these compounds were predicted. GC-related targets were screened using the GeneCards database. Venn diagram was used to identify common targets. An active ingredient-core target interaction network and a protein-protein interaction (PPI) network were built. Moreover, we performed gene ontology (GO) functional annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses on the core targets and validated them by molecular docking. RESULTS TCMSP screening revealed 11 active components and 184 targets, whereas GeneCards found 10118 disease-related targets, with 180 shared targets between them. Topology analysis of the PPI network identified 38 targets, including ATK1, TP53, and tumor necrosis factor, as key targets for the treatment of GC by Jianpi Yiwei Recipe. Quercetin, naringenin, luteolin, etc., may be the main active components of Jianpi Yiwei Recipe. GO enrichment analysis identified 2809, 1218, and 553 functions related to biological process, molecular function, and cellular component, respectively. KEGG pathway enrichment analysis revealed 167 related pathways, mainly involved in cancer, endocrine resistance, and AGE-RAGE signaling in diabetic complication. Validation with molecular docking analysis showed docking of key active components with core targets. CONCLUSION Jianpi Yiwei Recipe plays a therapeutic role in GC through multiple components, targets, and pathways. These findings form a basis for follow-up exploration of Jianpi Yiwei Recipe in the treatment of GC.
Collapse
Affiliation(s)
- Peng Chen
- Traditional Chinese Medicine, The First Teaching Hospital of Tianjin University, Tianjin 300193, China
| | - Huan-Yu Wu
- Traditional Chinese Medicine, The First Teaching Hospital of Tianjin University, Tianjin 300193, China
| |
Collapse
|
12
|
Zhu M, Sun Y, Su Y, Guan W, Wang Y, Han J, Wang S, Yang B, Wang Q, Kuang H. Luteolin: A promising multifunctional natural flavonoid for human diseases. Phytother Res 2024; 38:3417-3443. [PMID: 38666435 DOI: 10.1002/ptr.8217] [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: 01/02/2024] [Revised: 04/06/2024] [Accepted: 04/14/2024] [Indexed: 07/12/2024]
Abstract
Natural products are closely associated with human health. Luteolin (LUT), a flavonoid polyphenolic compound, is widely found in fruits, vegetables, flowers, and herbs. It is noteworthy that LUT exhibits a variety of beneficial pharmacological properties and holds significant potential for clinical applications, particularly in antitumor, anti-convulsion, diabetes control, anti-inflammatory, neuroprotection, anti-oxidation, anti-cardiovascular, and other aspects. The potential mechanism of action has been partially elucidated, including the mediation of NF-κB, toll-like receptor, MAPK, Wnt/β-catenin, PI3K/Akt, AMPK/mTOR, and Nrf-2, among others. The review that aimed to comprehensively consolidate essential information on natural sources, pharmacological effects, therapeutic and preventive potential, as well as potential mechanisms of LUT. The objective is to establish a theoretical basis for the continued development and application of LUT.
Collapse
Affiliation(s)
- Mingtao Zhu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Yanping Sun
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Yang Su
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Wei Guan
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Yu Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Jianwei Han
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Shuang Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Bingyou Yang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Qiuhong Wang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Haixue Kuang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| |
Collapse
|
13
|
Shao G, Liu Y, Lu L, Wang L, Ji G, Xu H. Therapeutic potential of traditional Chinese medicine in the prevention and treatment of digestive inflammatory cancer transformation: Portulaca oleracea L. as a promising drug. JOURNAL OF ETHNOPHARMACOLOGY 2024; 327:117999. [PMID: 38447616 DOI: 10.1016/j.jep.2024.117999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/19/2024] [Accepted: 02/28/2024] [Indexed: 03/08/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine (TCM) has been used for centuries to treat various types of inflammation and tumors of the digestive system. Portulaca oleracea L. (POL), has been used in TCM for thousands of years. The chemical composition of POL is variable and includes flavonoids, alkaloids, terpenoids and organic acids and other classes of natural compounds. Many of these compounds exhibit powerful anti-inflammatory and anti-cancer-transforming effects in the digestive system. AIM OF STUDY In this review, we focus on the potential therapeutic role of POL in NASH, gastritis and colitis and their associated cancers, with a focus on the pharmacological properties and potential mechanisms of action of the main natural active compounds in POL. METHODS The information and data on Portulaca oleracea L. and its main active ingredients were collated from various resources like ethnobotanical textbooks and literature databases such as CNKI, VIP (Chinese literature), PubMed, Science Direct, Elsevier and Google Scholar (English literatures), Wiley, Springer, Tailor and Francis, Scopus, Inflibnet. RESULTS Kaempferol, luteolin, myricetin, quercetin, genistein, EPA, DHA, and melatonin were found to improve NASH and NASH-HCC, while kaempferol, apigenin, luteolin, and quercetin played a therapeutic role in gastritis and gastric cancer. Apigenin, luteolin, myricetin, quercetin, genistein, lupeol, vitamin C and melatonin were found to have therapeutic effects in the treatment of colitis and its associated cancers. The discovery of the beneficial effects of these natural active compounds in POL supports the idea that POL could be a promising novel candidate for the treatment and prevention of inflammation-related cancers of the digestive system. CONCLUSION The discovery of the beneficial effects of these natural active compounds in POL supports the idea that POL could be a promising novel candidate for the treatment and prevention of inflammation-related cancers of the digestive system. However, clinical data describing the mode of action of the naturally active compounds of POL are still lacking. In addition, pharmacokinetic data for POL compounds, such as changes in drug dose and absorption rates, cannot be extrapolated from animal models and need to be measured in patients in clinical trials. On the one hand, a systematic meta-analysis of the existing publications on TCM containing POL still needs to be carried out. On the other hand, studies on the hepatic and renal toxicity of POL are also needed. Additionally, well-designed preclinical and clinical studies to validate the therapeutic effects of TCM need to be performed, thus hopefully providing a basis for the validation of the clinical benefits of POL.
Collapse
Affiliation(s)
- Gaoxuan Shao
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, China
| | - Ying Liu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, China
| | - Lu Lu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, China
| | - Lei Wang
- Department of Hepatology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, China.
| | - Hanchen Xu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, China.
| |
Collapse
|
14
|
Zhang W, Wang Y, Yu H, Jin Z, Yuan Y, Liu L, Zhou J. Exploring the mechanism of Erteng-Sanjie capsule in treating gastric and colorectal cancers via network pharmacology and in-vivo validation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 327:117945. [PMID: 38428659 DOI: 10.1016/j.jep.2024.117945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/26/2024] [Accepted: 02/19/2024] [Indexed: 03/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Erteng-Sanjie capsule (ETSJC) has therapeutic effects against gastric cancer (GC) and colorectal cancer (CRC). However, its underlying pharmacological mechanism remains unclear. AIM OF THE STUDY To explore the pharmacological mechanism of ETSJC against GC and CRC via network pharmacology and in-vivo validation. MATERIALS AND METHODS Data on the ingredients of ETSJC were obtained from the TCMSP and HERB databases. Further, details on the related targets of the active ingredients were collected from the HERB and SwissTargetPrediction databases. The targets in GC and CRC, which were screened from the OMIM, GeneCards, and TTD databases, were uploaded to STRING for a separate protein-protein interaction network analysis. The common targets shared by ETSJC, GC, and CRC were then screened. Cytoscape and STRING were used to construct the networks of herbs-compounds-targets and PPI. Metascape was utilized to analyze the enrichment of the GO and KEGG pathways. Molecular docking was used to validate the potential binding mode between the core ingredients and targets. Finally, the predicted results were verified with animal experiment. RESULTS Eight core ingredients (resveratrol, quercetin, luteolin, baicalein, delphinidin, kaempferol, pinocembrin, and naringenin) and six core targets (TP53, SRC, PIK3R1, AKT1, MAPK3, and STAT3) were filtered via network analysis. The molecular mechanism mainly involved the positive regulation of various processes such as cell migration, protein phosphorylation, and the PI3K-Akt signaling pathway. Molecular docking revealed that the core ingredients could be significantly combined with all core targets. The animal experiment revealed that ETSJC could suppress proliferation and promote apoptosis of both GC and CRC tumor cells by regulating the PI3K/Akt signaling pathway. CONCLUSIONS Multiple targets (TP53, SRC, AKT1, and STAT3) were important in GC and CRC. ETSJC could act on these targets and engage in different pathways against GC and CRC. Simultaneously, inhibiting the PI3K/Akt signaling pathway was a promising therapeutic mechanism for treating GC and CRC.
Collapse
Affiliation(s)
- Wencui Zhang
- Department of Oncology, Shanxi Province Academy of Traditional Chinese Medicine, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China.
| | - Ying Wang
- Department of Oncology, Shanxi Province Academy of Traditional Chinese Medicine, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China.
| | - Han Yu
- Department of Oncology, Shanxi Province Academy of Traditional Chinese Medicine, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China.
| | - Zengcai Jin
- Department of Oncology, Shanxi Province Academy of Traditional Chinese Medicine, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China.
| | - Yuyao Yuan
- Department of Oncology, Shanxi Province Academy of Traditional Chinese Medicine, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China.
| | - Likun Liu
- Department of Oncology, Shanxi Province Academy of Traditional Chinese Medicine, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China.
| | - Jing Zhou
- Department of Oncology, Shanxi Province Academy of Traditional Chinese Medicine, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China.
| |
Collapse
|
15
|
Liu H, Chen Y, Hu Y, Zhang W, Zhang H, Su T, Wang J, Yin Z, Zhao X, Zhou X, Li L, Zou Y, Fu Y, Zhang Y, Song X. Protective effects of an alcoholic extract of Kaempferia galanga L. rhizome on ethanol-induced gastric ulcer in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 325:117845. [PMID: 38307355 DOI: 10.1016/j.jep.2024.117845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 02/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The rhizome of Kaempferia galanga L., a medicinal and edible Plant, was widely distributed in many Asian and African counties. It has been traditionally used to treat gastroenteritis, hypertension, rheumatism and asthma. However, there is a lack of modern pharmacology studies regarding its anti-gastric ulcer activity. AIM OF THE STUDY The objective of this study is to investigate the protective effects of an extract from K. galanga L. rhizome (Kge) and its active components kaempferol and luteolin on ethanol-induced gastric ulcer. MATERIALS AND METHODS The kge was prepared by ultrasonic-assisted extraction, and the contents of kaempferol and luteolin were determined by HPLC. The mice were randomly divided into seven groups: blank control (0.5 % CMC-Na; 0.1 mL/10 g), untreatment (0.5 % CMC-Na; 0.1 mL/10 g), Kge (100, 200 and 400 mg/kg), kaempferol (100 mg/kg) and luteolin (100 mg/kg) groups. The mice were treated intragastrically once daily for 7 days. At 1 h post the last administration, the mice in all groups except the blank control group were intragastrically administrated with anhydrous alcohol (0.1 mL/10 g) once to induce gastric ulcer. Then, fasting was continued for 1 h, followed by sample collection for evaluation by enzyme-linked immunosorbent assay and real-time reverse transcription polymerase chain reaction assay. RESULTS The contents of kaempferol and luteolin in Kge were determined as 3713 μg/g and 2510 μg/g, respectively. Alcohol induced severely damages with edema, inflammatory cell infiltration and bleeding, and the ulcer index was 17.63 %. After pre-treatment with Kge (100, 200 and 400 mg/kg), kaempferol and luteolin, the pathological lesions were obviously alleviated and ulcer indices were reduced to 13.42 %, 11.65 %, 6.54 %, 3.58 % and 3.85 %, respectively. In untreated group, the contents of Ca2+, myeloperoxidase, malondialdehyde, NO, cyclic adenosine monophosphate and histamine were significantly increased, while the contents of hexosamine, superoxide dismutase, glutathione peroxidase, and prostaglandin E2 were significantly decreased; the transcriptional levels of IL-1α, IL-1β, IL-6, calcitonin gene related peptide, substance P, M3 muscarinic acetylcholine receptor, histamine H2 receptor, cholecystokinin 2 receptor and H+/K+ ATPase were significantly increased when compared with the blank control group. After pre-treatment, all of these changes were alleviated, even returned to normal levels. Kge exhibited anti-gastric ulcer activity and the high dose of Kge (400 mg/kg) exhibited comparable activity to that of kaempferol and luteolin. CONCLUSION The study showed that K. galanga L., kaempferol, and luteolin have protective effects against ethanol-induced gastric ulcers. This is achieved by regulating the mucosal barrier, oxidative stress, and gastric regulatory mediators, as well as inhibiting the TRPV1 signaling pathway and gastric acid secretion, ultimately reducing the gastric ulcer index.
Collapse
Affiliation(s)
- Haifeng Liu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yaqin Chen
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yilong Hu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Wenrui Zhang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Hui Zhang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Tianli Su
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Juan Wang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhongqiong Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xinhong Zhao
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xun Zhou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Lixia Li
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yuanfeng Zou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yuping Fu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yingying Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Xu Song
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
16
|
Rauf A, Wilairatana P, Joshi PB, Ahmad Z, Olatunde A, Hafeez N, Hemeg HA, Mubarak MS. Revisiting luteolin: An updated review on its anticancer potential. Heliyon 2024; 10:e26701. [PMID: 38455556 PMCID: PMC10918152 DOI: 10.1016/j.heliyon.2024.e26701] [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: 08/03/2023] [Revised: 02/04/2024] [Accepted: 02/19/2024] [Indexed: 03/09/2024] Open
Abstract
Numerous natural products found in our diet, such as polyphenols and flavonoids, can prevent the progression of cancer. Luteolin, a natural flavone, present in significant amounts in various fruits and vegetables plays a key role as a chemopreventive agent in treating various types of cancer. By inducing apoptosis, initiating cell cycle arrest, and decreasing angiogenesis, metastasis, and cell proliferation, luteolin is used to treat cancer. Its anticancer properties are attributed to its capability to engage with multiple molecular targeted sites and modify various signaling pathways in tumor cells. Luteolin has been shown to slow the spread of cancer in breast, colorectal, lung, prostate, liver, skin, pancreatic, oral, and gastric cancer models. It exhibits antioxidant properties and can be given to patients receiving Doxorubicin (DOX) chemotherapy to prevent the development of unexpected adverse reactions in the lungs and hematopoietic system subjected to DOX. Furthermore, it could be an excellent candidate for synergistic studies to overcome drug resistance in cancer cells. Accordingly, this review covers the recent literature related to the use of luteolin against different types of cancer, along with the mechanisms of action. In addition, the review highlights luteolin as a complementary medicine for preventing and treating cancer.
Collapse
Affiliation(s)
- Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar 23561, Khyber Pakhtunkhwa, Pakistan
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Payal B. Joshi
- Operations and Method Development, Shefali Research Laboratories, Ambernath, (East)-421501, Maharashtra, India
| | - Zubair Ahmad
- Department of Chemistry, University of Swabi, Anbar 23561, Khyber Pakhtunkhwa, Pakistan
| | - Ahmed Olatunde
- Department of Medical Biochemistry, Abubakar Tafawa Balewa University, Bauchi, 740272, Nigeria
| | - Nabia Hafeez
- Center of Biotechnology and Microbiology, University of Peshawar, Peshawar, 25120, KPK, Pakistan
| | - Hassan A. Hemeg
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Al-Medinah, Al-Monawara Postcode, Saudi Arabia
| | | |
Collapse
|
17
|
Fakhri S, Moradi SZ, Faraji F, Kooshki L, Webber K, Bishayee A. Modulation of hypoxia-inducible factor-1 signaling pathways in cancer angiogenesis, invasion, and metastasis by natural compounds: a comprehensive and critical review. Cancer Metastasis Rev 2024; 43:501-574. [PMID: 37792223 DOI: 10.1007/s10555-023-10136-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 09/07/2023] [Indexed: 10/05/2023]
Abstract
Tumor cells employ multiple signaling mediators to escape the hypoxic condition and trigger angiogenesis and metastasis. As a critical orchestrate of tumorigenic conditions, hypoxia-inducible factor-1 (HIF-1) is responsible for stimulating several target genes and dysregulated pathways in tumor invasion and migration. Therefore, targeting HIF-1 pathway and cross-talked mediators seems to be a novel strategy in cancer prevention and treatment. In recent decades, tremendous efforts have been made to develop multi-targeted therapies to modulate several dysregulated pathways in cancer angiogenesis, invasion, and metastasis. In this line, natural compounds have shown a bright future in combating angiogenic and metastatic conditions. Among the natural secondary metabolites, we have evaluated the critical potential of phenolic compounds, terpenes/terpenoids, alkaloids, sulfur compounds, marine- and microbe-derived agents in the attenuation of HIF-1, and interconnected pathways in fighting tumor-associated angiogenesis and invasion. This is the first comprehensive review on natural constituents as potential regulators of HIF-1 and interconnected pathways against cancer angiogenesis and metastasis. This review aims to reshape the previous strategies in cancer prevention and treatment.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Farahnaz Faraji
- Department of Pharmaceutics, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Leila Kooshki
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, 6714415153, Iran
| | - Kassidy Webber
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Boulevard, Bradenton, FL, 34211, USA
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Boulevard, Bradenton, FL, 34211, USA.
| |
Collapse
|
18
|
Almatroodi SA, Almatroudi A, Alharbi HOA, Khan AA, Rahmani AH. Effects and Mechanisms of Luteolin, a Plant-Based Flavonoid, in the Prevention of Cancers via Modulation of Inflammation and Cell Signaling Molecules. Molecules 2024; 29:1093. [PMID: 38474604 DOI: 10.3390/molecules29051093] [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: 01/19/2024] [Revised: 02/18/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Luteolin, a flavonoid, is mainly found in various vegetables and fruits, including carrots, cabbages, onions, parsley, apples, broccoli, and peppers. Extensive research in vivo and in vitro has been performed to explore its role in disease prevention and treatment. Moreover, this compound possesses the ability to combat cancer by modulating cell-signaling pathways across various types of cancer. The studies have confirmed that luteolin can inhibit cancer-cell survival and proliferation, angiogenesis, invasion, metastasis, mTOR/PI3K/Akt, STAT3, Wnt/β-catenin, and cell-cycle arrest, and induce apoptosis. Further, scientific evidence describes that this compound plays a vital role in the up/down-regulation of microRNAs (miRNAs) in cancer therapy. This review aims to outline the anti-cancer mechanisms of this compound and its molecular targets. However, a knowledge gap remains regarding the studies on its safety and efficacy and clinical trials. Therefore, it is essential to conduct more research based on safety, efficacy, and clinical trials to explore the beneficial role of this compound in disease management, including cancer.
Collapse
Affiliation(s)
- Saleh A Almatroodi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Hajed Obaid A Alharbi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Amjad Ali Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| |
Collapse
|
19
|
Torcasio R, Gallo Cantafio ME, Veneziano C, De Marco C, Ganino L, Valentino I, Occhiuzzi MA, Perrotta ID, Mancuso T, Conforti F, Rizzuti B, Martino EA, Gentile M, Neri A, Viglietto G, Grande F, Amodio N. Targeting of mitochondrial fission through natural flavanones elicits anti-myeloma activity. J Transl Med 2024; 22:208. [PMID: 38413989 PMCID: PMC10898065 DOI: 10.1186/s12967-024-05013-0] [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: 01/25/2024] [Accepted: 02/21/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Mitochondrial alterations, often dependent on unbalanced mitochondrial dynamics, feature in the pathobiology of human cancers, including multiple myeloma (MM). Flavanones are natural flavonoids endowed with mitochondrial targeting activities. Herein, we investigated the capability of Hesperetin (Hes) and Naringenin (Nar), two aglycones of Hesperidin and Naringin flavanone glycosides, to selectively target Drp1, a pivotal regulator of mitochondrial dynamics, prompting anti-MM activity. METHODS Molecular docking analyses were performed on the crystallographic structure of Dynamin-1-like protein (Drp1), using Hes and Nar molecular structures. Cell viability and apoptosis were assessed in MM cell lines, or in co-culture systems with primary bone marrow stromal cells, using Cell Titer Glo and Annexin V-7AAD staining, respectively; clonogenicity was determined using methylcellulose colony assays. Transcriptomic analyses were carried out using the Ion AmpliSeq™ platform; mRNA and protein expression levels were determined by quantitative RT-PCR and western blotting, respectively. Mitochondrial architecture was assessed by transmission electron microscopy. Real time measurement of oxygen consumption was performed by high resolution respirometry in living cells. In vivo anti-tumor activity was evaluated in NOD-SCID mice subcutaneously engrafted with MM cells. RESULTS Hes and Nar were found to accommodate within the GTPase binding site of Drp1, and to inhibit Drp1 expression and activity, leading to hyperfused mitochondria with reduced OXPHOS. In vitro, Hes and Nar reduced MM clonogenicity and viability, even in the presence of patient-derived bone marrow stromal cells, triggering ER stress and apoptosis. Interestingly, Hes and Nar rewired MM cell metabolism through the down-regulation of master transcriptional activators (SREBF-1, c-MYC) of lipogenesis genes. An extract of Tacle, a Citrus variety rich in Hesperidin and Naringin, was capable to recapitulate the phenotypic and molecular perturbations of each flavanone, triggering anti-MM activity in vivo. CONCLUSION Hes and Nar inhibit proliferation, rewire the metabolism and induce apoptosis of MM cells via antagonism of the mitochondrial fission driver Drp1. These results provide a framework for the development of natural anti-MM therapeutics targeting aberrant mitochondrial dependencies.
Collapse
Affiliation(s)
- Roberta Torcasio
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, Campus Germaneto, 88100, Catanzaro, Italy
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Cosenza, Italy
| | | | - Claudia Veneziano
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, Campus Germaneto, 88100, Catanzaro, Italy
| | - Carmela De Marco
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, Campus Germaneto, 88100, Catanzaro, Italy
| | - Ludovica Ganino
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, Campus Germaneto, 88100, Catanzaro, Italy
| | - Ilenia Valentino
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, Campus Germaneto, 88100, Catanzaro, Italy
| | - Maria Antonietta Occhiuzzi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, CS, Italy
| | - Ida Daniela Perrotta
- Department of Biology, Ecology and Earth Sciences, Centre for Microscopy and Microanalysis, University of Calabria, Cosenza, Italy
| | - Teresa Mancuso
- Annunziata" Regional Hospital Cosenza, 87100, Cosenza, Italy
| | - Filomena Conforti
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, CS, Italy
| | - Bruno Rizzuti
- SS Rende (CS), Department of Physics, CNR-NANOTEC, University of Calabria, Via Pietro Bucci, 87036, Rende, CS, Italy
- Institute for Biocomputation and Physics of Complex Systems (BIFI), Joint Unit GBsC-CSIC-BIFI, University of Zaragoza, 50018, Saragossa, Spain
| | | | - Massimo Gentile
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, CS, Italy
- Annunziata" Regional Hospital Cosenza, 87100, Cosenza, Italy
| | - Antonino Neri
- Scientific Directorate, IRCCS Di Reggio Emilia, Emilia Romagna, Reggio Emilia, Italy
| | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, Campus Germaneto, 88100, Catanzaro, Italy
| | - Fedora Grande
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, CS, Italy
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, Campus Germaneto, 88100, Catanzaro, Italy.
| |
Collapse
|
20
|
Ren Y, Li G, Li E, Deng K, Lian J, Gao Q, Wang H, Wang X, Wang Z, Shen T, Jiang Z, Li X, Qiu G. Luteolin blocks the ROS/PI3K/AKT pathway to inhibit mesothelial-mesenchymal transition and reduce abdominal adhesions. Eur J Pharmacol 2024; 964:176272. [PMID: 38110140 DOI: 10.1016/j.ejphar.2023.176272] [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: 07/26/2023] [Revised: 11/21/2023] [Accepted: 12/08/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND Postoperative abdominal adhesion (PAA) is a common postoperative complication. Clinically, various methods have been used to prevent the occurrence of PAA, such as drugs and physiotherapy; however, no satisfactory results have been obtained. Luteolin (LUT) is a natural flavonoid that reduces inflammation and acts as an antioxidant. This research aimed to examine the impact and mechanism of LUT in reducing PAA. METHODS C57/BL6 mice were used in vivo experiments. PAA model was established using a brush friction method. Visual scoring and hematoxylin and eosin staining were used to score the severity of adhesions. Network pharmacology was used to infer potential targets and core pathways of LUT. Hydrogen peroxide (H2O2) was used to induce oxidative stress in vitro, while the reactive oxygen species (ROS) assay kit was used to evaluate oxidative stress levels. Western blotting, cell immunofluorescence, and multiple immunofluorescence assays were used to detect α-SMA, vimentin, E-cadherin, collagen I, or AKT phosphorylation level. Scratch assay was used to detect cell migration. RESULTS LUT reduced the degree of PAA in mice. It attenuated H2O2-induced ROS production and reversed mesothelial-mesenchymal transition (MMT) in HMrSV5 cells. Network pharmacology analysis showed that LUT likely exerted anti-adhesion activity by regulating the PI3K-Akt signaling pathway. Phosphorylated Akt levels were significantly reduced in LUT-treated HMrSV5 cells. LUT also significantly reduced the expression of vimentin and collagen I in adherent tissues and upregulated E-cadherin expression. CONCLUSION LUT blocks the ROS/PI3K/AKT pathway, thereby inhibiting MMT and reducing PAA. To this end, LUT has potential in PAA therapy.
Collapse
Affiliation(s)
- Yiwei Ren
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Gan Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Enmeng Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Kai Deng
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Jie Lian
- Department of Pathology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Qi Gao
- Department of General Surgery, Shaanxi Provincial People's Hospital, Xi'an Medical University, 710061 Xi'an, Shaanxi, China
| | - Huijun Wang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Xingjie Wang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Zijun Wang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Tianli Shen
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Zhengdong Jiang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Xuqi Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China; Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| | - Guanglin Qiu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| |
Collapse
|
21
|
Macedo C, Costa PC, Rodrigues F. Bioactive compounds from Actinidia arguta fruit as a new strategy to fight glioblastoma. Food Res Int 2024; 175:113770. [PMID: 38129059 DOI: 10.1016/j.foodres.2023.113770] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/10/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023]
Abstract
In recent years, there has been a significant demand for natural products as a mean of disease prevention or as an alternative to conventional medications. The driving force for this change is the growing recognition of the abundant presence of valuable bioactive compounds in natural products. On recent years Actinia arguta fruit, also known as kiwiberry, has attracted a lot of attention from scientific community due to its richness in bioactive compounds, including phenolic compounds, organic acids, vitamins, carotenoids and fiber. These bioactive compounds contribute to the fruit's diverse outstanding biological activities such as antioxidant, anti-inflammatory, neuroprotective, immunomodulatory, and anti-cancer properties. Due to these properties, the fruit may have the potential to be used in the treatment/prevention of various types of cancer, including glioblastoma. Glioblastoma is the most aggressive form of brain cancer, displaying 90 % of recurrence rate within a span of 2 years. Despite the employment of an aggressive approach, the prognosis remains unfavorable, emphasizing the urgent requirement for the development of new effective treatments. The preclinical evidence suggests that kiwiberry has potential impact on glioblastoma by reducing the cancer self-renewal, modulating the signaling pathways involved in the regulation of the cell phenotype and metabolism, and influencing the consolidation of the tumor microenvironment. Even though, challenges such as the imprecise composition and concentration of bioactive compounds, and its low bioavailability after oral administration may be drawbacks to the development of kiwiberry-based treatments, being urgent to ensure the safety and efficacy of kiwiberry for the prevention and treatment of glioblastoma. This review aims to highlight the potential impact of A. arguta bioactive compounds on glioblastoma, providing novel insights into their applicability as complementary or alternative therapies.
Collapse
Affiliation(s)
- Catarina Macedo
- REQUIMTE/LAQV, ISEP, Polytechnic of Porto, Rua Dr. António Bernardino de Almeida, 4249-015 Porto, Portugal; REQUIMTE/UCIBIO, MedTech-Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Paulo C Costa
- REQUIMTE/UCIBIO, MedTech-Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Francisca Rodrigues
- REQUIMTE/LAQV, ISEP, Polytechnic of Porto, Rua Dr. António Bernardino de Almeida, 4249-015 Porto, Portugal.
| |
Collapse
|
22
|
Matić IZ, Mraković A, Rakočević Z, Stoiljković M, Pavlović VB, Momić T. Anticancer effect of novel luteolin capped gold nanoparticles selectively cytotoxic towards human cervical adenocarcinoma HeLa cells: An in vitro approach. J Trace Elem Med Biol 2023; 80:127286. [PMID: 37634345 DOI: 10.1016/j.jtemb.2023.127286] [Citation(s) in RCA: 4] [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: 04/06/2022] [Revised: 07/26/2023] [Accepted: 08/21/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Although luteolin has been confirmed as potent anticancer agent, its potential application as therapeutic is limited by its water solubility. To overcome this shortcoming nanoparticle technology approach was applied. Owing to their proven low toxicity and the possibility to be easily functionalized gold nanoparticles (AuNP) were the nanosystem of choice used in this study. Novel luteolin capped gold nanoparticles (AuNPL) were synthesized and their anticancer effect towards human cervical adenocarcinoma HeLa cells was investigated in vitro. METHODS AuNPL were synthesized by reducing chloroauric acid by trisodium citrate with subsequent addition of luteoline during synthesis and their physicochemical characterization was done. AuNPL cytotoxicity against HeLa, human malignant melanoma A375, and normal human keratinocytes HaCaT cells was tested by MTT cell survival assay, and their IC50 values were determined. The capability of AuNPL to induce cell cycle arrest and apoptosis in HeLa cells were demonstrated by flow cytometry. The antioxidant activity of AuNPL was assessed by DPPH· and ABTS·+ scavenging assays. Cytoprotective properties of AuNPL towards HaCaT cells were examined by measuring the physiological and H2O2 induced intracellular reactive oxygen species (ROS) levels using flow cytometry. Also, genotoxicity of AuNPL in HaCaT cells was investigated by the single cell alkaline comet assay. RESULTS Spherical AuNPL, stable in aqueous solution up to six months at 4 °C were obtained in the synthesis. The selectivity in the cytotoxic action of AuNPL on HeLa and A375 cancer cells compared with their cytotoxicity on normal keratinocytes HaCaT was observed. AuNPL exerted their cytotoxic activity against HeLa cells through accumulation of the cells in the subG1 phase of the cell cycle, inducing the apoptotic cell death mediated by the activation of caspase-3 - 8, and - 9. AuNPL antioxidative potential was confirmed by DPPH· and ABTS·+ scavenging assays. IC50 concentration of AuNPL exerted cytoprotective effect against HaCaT cells by the significant reduction of the physiological intracellular ROS level. Additionally, AuNPL were shown as more cytoprotective towards HaCaT cells then luteolin due to the more successful elimination of H2O2 induced intracellular ROS. Moreover, nontoxic concentrations of AuNPL did not cause considerable DNA damage of HaCaT cells, indicating low genotoxicity of the nanoparticles. CONCLUSION Synthesized AuNPL showed selective cytotoxic activity against HeLa cells, while being nontoxic and cytoprotective against HaCaT cells. The observed findings encourage further investigation of AuNPL as a promising novel anticancer agent.
Collapse
Affiliation(s)
- Ivana Z Matić
- Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Ana Mraković
- VINČA Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovića Alasa 12-14, 11000 Belgrade, Serbia
| | - Zlatko Rakočević
- VINČA Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovića Alasa 12-14, 11000 Belgrade, Serbia
| | - Milovan Stoiljković
- VINČA Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovića Alasa 12-14, 11000 Belgrade, Serbia
| | - Vladimir B Pavlović
- Faculty of Agriculture, University of Belgrade, P.O. Box 127, 11080 Zemun, Serbia
| | - Tatjana Momić
- VINČA Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovića Alasa 12-14, 11000 Belgrade, Serbia.
| |
Collapse
|
23
|
Proença C, Freitas M, Ribeiro D, Rufino AT, Fernandes E, Ferreira de Oliveira JMP. The role of flavonoids in the regulation of epithelial-mesenchymal transition in cancer: A review on targeting signaling pathways and metastasis. Med Res Rev 2023; 43:1878-1945. [PMID: 37147865 DOI: 10.1002/med.21966] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 03/20/2023] [Accepted: 04/12/2023] [Indexed: 05/07/2023]
Abstract
One of the hallmarks of cancer is metastasis, a process that entails the spread of cancer cells to distant regions in the body, culminating in tumor formation in secondary organs. Importantly, the proinflammatory environment surrounding cancer cells further contributes to cancer cell transformation and extracellular matrix destruction. During metastasis, front-rear polarity and emergence of migratory and invasive features are manifestations of epithelial-mesenchymal transition (EMT). A variety of transcription factors (TFs) are implicated in the execution of EMT, the most prominent belonging to the Snail Family Transcriptional Repressor (SNAI) and Zinc Finger E-Box Binding Homeobox (ZEB) families of TFs. These TFs are regulated by interaction with specific microRNAs (miRNAs), as miR34 and miR200. Among the several secondary metabolites produced in plants, flavonoids constitute a major group of bioactive molecules, with several described effects including antioxidant, antiinflammatory, antidiabetic, antiobesogenic, and anticancer effects. This review scrutinizes the modulatory role of flavonoids on the activity of SNAI/ZEB TFs and on their regulatory miRNAs, miR-34, and miR-200. The modulatory role of flavonoids can attenuate mesenchymal features and stimulate epithelial features, thereby inhibiting and reversing EMT. Moreover, this modulation is concomitant with the attenuation of signaling pathways involved in diverse processes as cell proliferation, cell growth, cell cycle progression, apoptosis inhibition, morphogenesis, cell fate, cell migration, cell polarity, and wound healing. The antimetastatic potential of these versatile compounds is emerging and represents an opportunity for the synthesis of more specific and potent agents.
Collapse
Affiliation(s)
- Carina Proença
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Marisa Freitas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Daniela Ribeiro
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Ana T Rufino
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - José Miguel P Ferreira de Oliveira
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
24
|
Liao Y, Gui Y, Li Q, An J, Wang D. The signaling pathways and targets of natural products from traditional Chinese medicine treating gastric cancer provide new candidate therapeutic strategies. Biochim Biophys Acta Rev Cancer 2023; 1878:188998. [PMID: 37858623 DOI: 10.1016/j.bbcan.2023.188998] [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: 07/24/2023] [Revised: 09/26/2023] [Accepted: 10/08/2023] [Indexed: 10/21/2023]
Abstract
Gastric cancer (GC) is one of the severe malignancies with high incidence and mortality, especially in Eastern Asian countries. Significant advancements have been made in diagnosing and treating GC over the past few decades, resulting in tremendous improvements in patient survival. In recent years, traditional Chinese medicine (TCM) has garnered considerable attention as an alternative therapeutic approach for GC due to its multicomponent and multitarget characteristics. Consequently, natural products found in TCM have attracted researchers' attention, as growing evidence suggests that these natural products can impede GC progression by regulating various biological processes. Nevertheless, their molecular mechanisms are not systematically uncovered. Here, we review the major signaling pathways involved in GC development. Additionally, clinical GC samples were analyzed. Moreover, the anti-GC effects of natural products, their underlying mechanisms and potential targets were summarized. These summaries are intended to facilitate further relevant research, and accelerate the clinical applications of natural products in GC treatment.
Collapse
Affiliation(s)
- Yile Liao
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yu Gui
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Qingzhou Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jun An
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Dong Wang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
25
|
Hao X, Zu M, Ning J, Zhou X, Gong Y, Han X, Meng Q, Li D, Ding S. Antitumor effect of luteolin proven by patient-derived organoids of gastric cancer. Phytother Res 2023; 37:5315-5327. [PMID: 37469042 DOI: 10.1002/ptr.7963] [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: 03/06/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/21/2023]
Abstract
Luteolin (Lut) has been shown to inhibit gastric cancer (GC); however, its efficacy compared to other clinical drugs has not been examined in human samples. This study aimed to elucidate the antitumor activity of Lut in GC patient-derived organoids (PDOs). PDOs were established from GC cancer tissues, and the characterization of tissues and PDOs was performed using whole-exome sequencing. Drug sensitivity tests were performed by treating PDOs with Lut, norcantharidin (NCTD), and carboplatin (CP). RNA sequencing of PDOs was performed to elucidate the antitumor mechanism of Lut, which was further verified in three GC cell lines. Eleven PDOs were successfully constructed, and were highly consistent with the pathophysiology and genetic changes in the corresponding tumors. The IC50s of Lut, NCTD, and CP of PDOs were 27.19, 23.9, and 37.87 μM, respectively. Lut treatment upregulated FOXO3, DUSP1, and CDKN1A expression and downregulated IL1R1 and FGFR4 expression in GC cell lines, which was consistent with the results of PDOs. We demonstrate that Lut exerted stronger antitumor effects than CP, but a similar effect to that of NCTD, which was obtained in an in vitro PDO system. Additionally, Lut exerted varying degrees of antitumor effects against the PDOs, thereby indicating that PDO may be a useful preclinical drug screening tool for personalized treatment.
Collapse
Affiliation(s)
- Xinyu Hao
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing, China
| | - Ming Zu
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing, China
| | - Jing Ning
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing, China
| | - Xin Zhou
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Yueqing Gong
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing, China
| | - Xiurui Han
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing, China
| | - Qiao Meng
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing, China
| | - Dong Li
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Shigang Ding
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing, China
| |
Collapse
|
26
|
Rocchetti MT, Bellanti F, Zadorozhna M, Fiocco D, Mangieri D. Multi-Faceted Role of Luteolin in Cancer Metastasis: EMT, Angiogenesis, ECM Degradation and Apoptosis. Int J Mol Sci 2023; 24:8824. [PMID: 37240168 PMCID: PMC10218870 DOI: 10.3390/ijms24108824] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/09/2023] [Accepted: 05/13/2023] [Indexed: 05/28/2023] Open
Abstract
Luteolin (3',4',5,7-tetrahydroxyflavone), a member of the flavonoid family derived from plants and fruits, shows a wide range of biomedical applications. In fact, due to its anti-inflammatory, antioxidant and immunomodulatory activities, Asian medicine has been using luteolin for centuries to treat several human diseases, including arthritis, rheumatism, hypertension, neurodegenerative disorders and various infections. Of note, luteolin displays many anti-cancer/anti-metastatic properties. Thus, the purpose of this review consists in highlighting the relevant mechanisms by which luteolin inhibits tumor progression in metastasis, i.e., affecting epithelial-mesenchymal transition (EMT), repressing angiogenesis and lysis of extracellular matrix (ECM), as well as inducing apoptosis.
Collapse
Affiliation(s)
- Maria Teresa Rocchetti
- Department of Clinical and Experimental Medicine, University of Foggia, Via Pinto 1, 71122 Foggia, Italy; (M.T.R.); (D.F.)
| | - Francesco Bellanti
- Department of Medical and Surgical Sciences, University of Foggia, Via Pinto 1, 71122 Foggia, Italy;
| | - Mariia Zadorozhna
- Medical Genetics Unit, Department of Molecular Medicine, University of Pavia, Via Forlanini 14, 27100 Pavia, Italy;
| | - Daniela Fiocco
- Department of Clinical and Experimental Medicine, University of Foggia, Via Pinto 1, 71122 Foggia, Italy; (M.T.R.); (D.F.)
| | - Domenica Mangieri
- Department of Clinical and Experimental Medicine, University of Foggia, Via Pinto 1, 71122 Foggia, Italy; (M.T.R.); (D.F.)
| |
Collapse
|
27
|
Zhang C, Zhang Y, Hu X, Zhao Z, Chen Z, Wang X, Zhang Z, Jin H, Zhang J. Luteolin inhibits subretinal fibrosis and epithelial-mesenchymal transition in laser-induced mouse model via suppression of Smad2/3 and YAP signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 116:154865. [PMID: 37201365 DOI: 10.1016/j.phymed.2023.154865] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/13/2023] [Accepted: 05/08/2023] [Indexed: 05/20/2023]
Abstract
BACKGROUND Subretinal fibrosis (SF) accounts for vision loss in patients with neovascular age-related macular degeneration (nAMD) even treated with adequate intravitreal injection of anti-vascular endothelial growth factor (anti-VEGF) drugs. Currently, there is no treatment available to prevent or treat SF caused by nAMD. PURPOSE This study aims to investigate the potential effects of luteolin on SF and epithelial-mesenchymal transition (EMT) as well as the underlying molecular pathways both in vivo and in vitro. METHODS Seven-week-old male C57BL/6J mice were employed to establish laser-induced choroidal neovascularization (CNV) and SF. One day after the laser induction, luteolin was administered intravitreally. SF and CNV were assessed with the immunolabeling of collagen type I (collagen I) and isolectin B4 (IB4), respectively. RPE65 and α-SMA colocalization in the lesions was used to evaluate the extent of EMT in retinal pigment epithelial (RPE) cells by using immunofluorescence. In vitro, luteolin was administered to TGFβ1-treated primary human RPE (phRPE) cells. RT-qPCR, Western blot and immunofluorescence were employed to evaluate the change of EMT-related molecules, epithelial markers, and relevant signaling pathways. The functional changes associated with EMT were investigated using the scratch assay, Transwell migration assay, and collagen gel contraction assay. CCK-8 was used to determine the cell viability of phRPE cells. RESULTS On day 7 and 14 after laser induction in mice, intravitreal injection of luteolin dramatically decreased the immunolabeled sizes of both collagen I and IB4, as well as the amount of colocalized double immunostaining of α-SMA and RPE65 in laser-induced SF lesions. In vitro, TGFβ1-treated phRPE cells demonstrated increased cell migration and contraction capacity, accompanied with considerable overexpression of fibronectin, α-SMA, N-cadherin and vimentin, as well as downregulation of E-cadherin and ZO-1. The above changes were largely inhibited by luteolin co-incubation. Mechanistically, luteolin could evidently decrease the phosphorylation of Smad2/3, whereas increase the phosphorylation of YAP in TGFβ1-treated phRPE cells. CONCLUSION This study demonstrates that luteolin exhibits the anti-fibrotic effect in a laser-induced mouse model by inhibiting EMT of RPE cells via deactivating Smad2/3 and YAP signaling, which provides a potential natural compound for the prevention and treatment of SF and fibrosis-related diseases.
Collapse
Affiliation(s)
- Chaoyang Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Yao Zhang
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Xin Hu
- Department of Ophthalmology, Huaihe Hospital, Henan University, Kaifeng, PR China
| | - Zhenzhen Zhao
- Department of Regenerative Medicine, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, PR China
| | - Ziang Chen
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Xi Wang
- Department of Regenerative Medicine, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, PR China
| | - Zhihua Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Haiying Jin
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, PR China.
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China.
| |
Collapse
|
28
|
Hu Q, Li Z, Li Y, Deng X, Chen Y, Ma X, Zeng J, Zhao Y. Natural products targeting signaling pathways associated with regulated cell death in gastric cancer: Recent advances and perspectives. Phytother Res 2023. [PMID: 37157181 DOI: 10.1002/ptr.7866] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/17/2023] [Accepted: 04/25/2023] [Indexed: 05/10/2023]
Abstract
Gastric cancer (GC) is one of the most serious gastrointestinal malignancies with high morbidity and mortality. The complexity of GC process lies in the multi-phenotypic linkage regulation, in which regulatory cell death (RCD) is the core link, which largely dominates the fate of GC cells and becomes a key determinant of GC development and prognosis. In recent years, increasing evidence has been reported that natural products can prevent and inhibit the development of GC by regulating RCDs, showing great therapeutic potential. In order to further clarify its key regulatory characteristics, this review focused on specific expressions of RCDs, combined with a variety of signaling pathways and their crosstalk characteristics, sorted out the key targets and action rules of natural products targeting RCD. It is highlighted that a variety of core biological pathways and core targets are involved in the decision of GC cell fate, including the PI3K/Akt signaling pathway, MAPK-related signaling pathways, p53 signaling pathway, ER stress, Caspase-8, gasdermin D (GSDMD), and so on. Moreover, natural products target the crosstalk of different RCDs by modulating above signaling pathways. Taken together, these findings suggest that targeting various RCDs in GC with natural products is a promising strategy, providing a reference for further clarifying the molecular mechanism of natural products treating GC, which warrants further investigations in this area.
Collapse
Affiliation(s)
- Qichao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Zhibei Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yubing Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinyu Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuan Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanling Zhao
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
29
|
Chang X, Tamauchi S, Yoshida K, Yoshihara M, Yokoi A, Shimizu Y, Ikeda Y, Yoshikawa N, Kiyono T, Yamamoto Y, Kajiyama H. Downregulating vaccinia-related kinase 1 by luteolin suppresses ovarian cancer cell proliferation by activating the p53 signaling pathway. Gynecol Oncol 2023; 173:31-40. [PMID: 37075494 DOI: 10.1016/j.ygyno.2023.04.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/10/2023] [Accepted: 04/03/2023] [Indexed: 04/21/2023]
Abstract
OBJECTIVES Ovarian cancer constitutes one of the most common causes of cancer-related deaths, and preventing chemotherapy resistance and recurrence in patients with ovarian cancer remains a challenge. Herein, we aimed to identify the effect of luteolin, a novel therapeutic agent targeting vaccinia-related kinase 1 (VRK1), on high-grade serous ovarian cancer (HGSOC). METHODS Phosphokinase array, RNA sequencing, and cell cycle and apoptosis assays were conducted to determine the underlying mechanism of the effect of luteolin on HGSOC cells. The anticancer effects of oral and intraperitoneal luteolin administration were assessed in patient-derived xenograft models via several methods, including the assessment of tumor size and immunohistochemistry of phospho-p53, phosphor-HistoneH3 and cleaved caspase 3. RESULTS Luteolin reduced HGSOC cell proliferation and increased apoptosis and cell cycle arrest at G2/M. Compared with controls, several genes were dysregulated in luteolin-treated cells, and luteolin activated the p53 signaling pathway. The human phosphokinase array revealed distinct p53 upregulation in luteolin-treated cells, as confirmed by p53 phosphorylation at ser15 and ser46 using western blot analysis. In patient-derived xenograft models, oral or intraperitoneal luteolin administration substantially suppressed tumor growth. Moreover, combination treatment involving luteolin and cisplatin inhibited tumor cell proliferation, especially in cisplatin-resistant HGSOC cell lines. CONCLUSIONS Luteolin demonstrated considerable anticancer effect on HGSOC cells, reduced VRK1 expression, and activated the p53 signaling pathway, thereby inducing apoptosis and cell cycle arrest in G2/M and inhibiting cell proliferation. Furthermore, luteolin exhibited a synergistic effect with cisplatin both in vivo and in vitro. Thus, luteolin can be considered a promising cotreatment option for HGSOC.
Collapse
Affiliation(s)
- Xuboya Chang
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Satoshi Tamauchi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Kosuke Yoshida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Institute for Advanced Research, Nagoya University, Nagoya 464-8601, Japan
| | - Masato Yoshihara
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Akira Yokoi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Institute for Advanced Research, Nagoya University, Nagoya 464-8601, Japan
| | - Yusuke Shimizu
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yoshiki Ikeda
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Nobuhisa Yoshikawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Tohru Kiyono
- Project for Prevention of HPV-related Cancer, Exploratory Oncology Research and Clinical Trial Center, Chiba 277-8577, Japan
| | - Yusuke Yamamoto
- Laboratory of Integrative Oncology, National Cancer Center Research Institute, Tokyo 104-0045, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
30
|
Ang HL, Mohan CD, Shanmugam MK, Leong HC, Makvandi P, Rangappa KS, Bishayee A, Kumar AP, Sethi G. Mechanism of epithelial-mesenchymal transition in cancer and its regulation by natural compounds. Med Res Rev 2023. [PMID: 36929669 DOI: 10.1002/med.21948] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 12/19/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is a complex process with a primordial role in cellular transformation whereby an epithelial cell transforms and acquires a mesenchymal phenotype. This transformation plays a pivotal role in tumor progression and self-renewal, and exacerbates resistance to apoptosis and chemotherapy. EMT can be initiated and promoted by deregulated oncogenic signaling pathways, hypoxia, and cells in the tumor microenvironment, resulting in a loss-of-epithelial cell polarity, cell-cell adhesion, and enhanced invasive/migratory properties. Numerous transcriptional regulators, such as Snail, Slug, Twist, and ZEB1/ZEB2 induce EMT through the downregulation of epithelial markers and gain-of-expression of the mesenchymal markers. Additionally, signaling cascades such as Wnt/β-catenin, Notch, Sonic hedgehog, nuclear factor kappa B, receptor tyrosine kinases, PI3K/AKT/mTOR, Hippo, and transforming growth factor-β pathways regulate EMT whereas they are often deregulated in cancers leading to aberrant EMT. Furthermore, noncoding RNAs, tumor-derived exosomes, and epigenetic alterations are also involved in the modulation of EMT. Therefore, the regulation of EMT is a vital strategy to control the aggressive metastatic characteristics of tumor cells. Despite the vast amount of preclinical data on EMT in cancer progression, there is a lack of clinical translation at the therapeutic level. In this review, we have discussed thoroughly the role of the aforementioned transcription factors, noncoding RNAs (microRNAs, long noncoding RNA, circular RNA), signaling pathways, epigenetic modifications, and tumor-derived exosomes in the regulation of EMT in cancers. We have also emphasized the contribution of EMT to drug resistance and possible therapeutic interventions using plant-derived natural products, their semi-synthetic derivatives, and nano-formulations that are described as promising EMT blockers.
Collapse
Affiliation(s)
- Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hin Chong Leong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Pooyan Makvandi
- Istituto Italiano di Tecnologia Centre for Materials Interface, Pontedera, Pisa, Italy
| | | | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
31
|
Therapeutic Potential of Luteolin on Cancer. Vaccines (Basel) 2023; 11:vaccines11030554. [PMID: 36992138 DOI: 10.3390/vaccines11030554] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
Cancer is a global concern, as the rate of incidence is increasing each year. The challenges related to the current chemotherapy drugs, such as the concerns related to toxicity, turn to cancer therapeutic research to discover alternative therapy strategies that are less toxic to normal cells. Among those studies, the use of flavonoids—natural compounds produced by plants as secondary metabolites for cancer therapy—has been a hot topic in cancer treatment. Luteolin, a flavonoid that has been present in many fruits, vegetables, and herbs, has been identified to exhibit numerous biological activities, including anti-inflammatory, antidiabetic, and anticancer properties. The anticancer property of Luteolin has been extensively researched in many cancer types and has been related to its ability to inhibit tumor growth by targeting cellular processes such as apoptosis, angiogenesis, migration, and cell cycle progression. It achieves this by interacting with various signaling pathways and proteins. In the current review, the molecular targets of Luteolin as it exerts its anticancer properties, the combination therapy that includes Luteolin with other flavonoids or chemotherapeutic drugs, and the nanodelivery strategies for Luteolin are described for several cancer types.
Collapse
|
32
|
Zhong YL, Wang PQ, Hao DL, Sui F, Zhang FB, Li B. Traditional Chinese medicine for transformation of gastric precancerous lesions to gastric cancer: A critical review. World J Gastrointest Oncol 2023; 15:36-54. [PMID: 36684050 PMCID: PMC9850768 DOI: 10.4251/wjgo.v15.i1.36] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/06/2022] [Accepted: 12/28/2022] [Indexed: 01/10/2023] Open
Abstract
Gastric cancer (GC) is a common gastrointestinal tumor. Gastric precancerous lesions (GPL) are the last pathological stage before normal gastric mucosa transforms into GC. However, preventing the transformation from GPL to GC remains a challenge. Traditional Chinese medicine (TCM) has been used to treat gastric disease for millennia. A series of TCM formulas and active compounds have shown therapeutic effects in both GC and GPL. This article reviews recent progress on the herbal drugs and pharmacological mechanisms of TCM in preventing the transformation from GPL to GC, especially focusing on anti-inflammatory, anti-angiogenesis, proliferation, and apoptosis. This review may provide a meaningful reference for the prevention of the transformation from GPL to GC using TCM.
Collapse
Affiliation(s)
- Yi-Lin Zhong
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Peng-Qian Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Dan-Li Hao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Feng Sui
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Feng-Bin Zhang
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| | - Bing Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| |
Collapse
|
33
|
Liang Z, Xu Y, Zhang Y, Zhang X, Song J, Qian H, Jin J. Anticancer applications of phytochemicals in gastric cancer: Effects and molecular mechanism. Front Pharmacol 2023; 13:1078090. [PMID: 36712679 PMCID: PMC9877357 DOI: 10.3389/fphar.2022.1078090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
Gastric cancer (GC) is the fourth most common malignant cancer and is a life-threatening disease worldwide. Phytochemicals have been shown to be a rational, safe, non-toxic, and very promising approach to the prevention and treatment of cancer. It has been found that phytochemicals have protective effects against GC through inhibiting cell proliferation, inducing apoptosis and autophagy, suppressing cell invasion and migration, anti-angiogenesis, inhibit Helicobacter pylori infection, regulating the microenvironment. In recent years, the role of phytochemicals in the occurrence, development, drug resistance and prognosis of GC has attracted more and more attention. In order to better understand the relationship between phytochemicals and gastric cancer, we briefly summarize the roles and functions of phytochemicals in GC tumorigenesis, development and prognosis. This review will probably help guide the public to prevent the occurrence and development of GC through phytochemicals, and develop functional foods or drugs for the prevention and treatment of gastric cancer.
Collapse
Affiliation(s)
- Zhaofeng Liang
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Chang Zhou, China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yumeng Xu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yue Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xinyi Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jiajia Song
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Hui Qian
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Chang Zhou, China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jianhua Jin
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Chang Zhou, China
| |
Collapse
|
34
|
Wen SY, Wei BY, Ma JQ, Wang L, Chen YY. Phytochemicals, Biological Activities, Molecular Mechanisms, and Future Prospects of Plantago asiatica L. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:143-173. [PMID: 36545763 DOI: 10.1021/acs.jafc.2c07735] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Plantago asiatica L. has been used as a vegetable and nutritious food in Asia for thousands of years. According to recent phytochemical and pharmacological research, the active compositions of the plant contribute to various health benefits, such as antioxidant, anti-inflammatory, antibacterial, antiviral, and anticancer. This article reviews the 87 components of the plant and their structures, as well as their biological activities and molecular research progress, in detail. This review provides valuable reference material for further study, production, and application of P. asiatica, as well as its components in functional foods and therapeutic agents.
Collapse
Affiliation(s)
- Shi-Yuan Wen
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030000, China
| | - Bing-Yan Wei
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030000, China
| | - Jie-Qiong Ma
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030000, China
| | - Li Wang
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030000, China
| | - Yan-Yan Chen
- School of Medicine, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
35
|
Singh Tuli H, Rath P, Chauhan A, Sak K, Aggarwal D, Choudhary R, Sharma U, Vashishth K, Sharma S, Kumar M, Yadav V, Singh T, Yerer MB, Haque S. Luteolin, a Potent Anticancer Compound: From Chemistry to Cellular Interactions and Synergetic Perspectives. Cancers (Basel) 2022; 14:5373. [PMID: 36358791 PMCID: PMC9658186 DOI: 10.3390/cancers14215373] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/22/2022] [Accepted: 10/25/2022] [Indexed: 08/03/2023] Open
Abstract
Increasing rates of cancer incidence and the toxicity concerns of existing chemotherapeutic agents have intensified the research to explore more alternative routes to combat tumor. Luteolin, a flavone found in numerous fruits, vegetables, and herbs, has exhibited a number of biological activities, such as anticancer and anti-inflammatory. Luteolin inhibits tumor growth by targeting cellular processes such as apoptosis, cell-cycle progression, angiogenesis and migration. Mechanistically, luteolin causes cell death by downregulating Akt, PLK-1, cyclin-B1, cyclin-A, CDC-2, CDK-2, Bcl-2, and Bcl-xL, while upregulating BAX, caspase-3, and p21. It has also been reported to inhibit STAT3 signaling by the suppression of STAT3 activation and enhanced STAT3 protein degradation in various cancer cells. Therefore, extensive studies on the anticancer properties of luteolin reveal its promising role in chemoprevention. The present review describes all the possible cellular interactions of luteolin in cancer, along with its synergistic mode of action and nanodelivery insight.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala 133207, India
| | - Prangya Rath
- Amity Institute of Environmental Sciences, Amity University, Noida 201303, India
| | - Abhishek Chauhan
- Amity Institute of Environmental Toxicology, Safety and Management, Amity University, Noida 201303, India
| | | | - Diwakar Aggarwal
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala 133207, India
| | - Renuka Choudhary
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala 133207, India
| | - Ujjawal Sharma
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bhatinda 151001, India
| | - Kanupriya Vashishth
- Department of Cardiology, Advance Cardiac Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
| | - Sheetu Sharma
- Department of Pharmacovigilace and Clinical Research, Chitkara University, Rajpura 140401, India
| | - Manoj Kumar
- Department of Chemistry, Maharishi Markandeshwar University Sadopur, Ambala 133001, India
| | - Vikas Yadav
- Department of Translational Medicine, Clinical Research Centre, Skåne University Hospital, Lund University, SE-20213 Malmö, Sweden
| | - Tejveer Singh
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, Delhi 110007, India
| | - Mukerrem Betul Yerer
- Department of Pharmacology, Faculty of Pharmacy, Erciyes University, Kayseri 38039, Turkey
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
36
|
Flavones: Six Selected Flavones and Their Related Signaling Pathways That Induce Apoptosis in Cancer. Int J Mol Sci 2022; 23:ijms231810965. [PMID: 36142874 PMCID: PMC9505532 DOI: 10.3390/ijms231810965] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/18/2022] Open
Abstract
Cancer is a horrific disease that, to date, has no cure. It is caused by various factors and takes many lives. Apoptosis is a programmed cell death mechanism and if it does not function correctly in cancer cells, it can lead to severe disease. There are various signaling pathways for regulating apoptosis in cancer cells. Flavonoids are non-artificial natural bioactive compounds that are gaining attention as being capable of for inducing apoptosis in cancer cells. Among these, in this study, we focus on flavones. Flavones are a subclass of the numerous available flavonoids and possess several bioactive functions. Some of the most reported and well-known critical flavones, namely apigenin, acacetin, baicalein, luteolin, tangeretin, and wogonin, are discussed in depth in this review. Our main aim is to investigate the effects of the selected flavones on apoptosis and cell signaling pathways that contribute to death due to various types of cancers.
Collapse
|
37
|
Zheng H, Zhu X, Gong E, Lv Y, Li Y, Cai X. Luteolin suppresses lung cancer progression through targeting the circ_0000190/miR-130a-3p/notch-1 signaling pathway. J Chemother 2022:1-13. [PMID: 35943044 DOI: 10.1080/1120009x.2022.2102303] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Lung cancer is the leading cause of cancer mortality worldwide. Luteolin has been reported to repress the development of lung cancer. And circular RNAs (circRNAs) circ_0000190 was upregulated in lung cancer tissues. This study is designed to explore the roles of luteolin and circ_0000190 in lung cancer progression. Cell viability, colony number, migration, invasion, and apoptosis were detected by Cell Counting Kit-8 (CCK-8), colony formation, transwell, and flow cytometry assays, severally. The lactate dehydrogenase (LDH) release was determined by special kits. Protein levels of B-celllymphoma-2 (Bcl-2) Cleaved-caspase3 (casp3), Bcl-2 related X protein (Bax), Notch-1, hairy enhance of split-1(Hes-1), and vascular endothelium growth factor (VEGF) were determined by western blot assay. Circ_0000190 andmicroRNA-130a-3p (miR-130a-3p) expression were measured by real-time quantitative polymerase chain reaction (RT-qPCR). The binding relationship between circ_0000190 andmiR-130a-3pwas predicted by starbase and then verified by a dual-luciferase reporter and RNA pull-down assays. The biological roles of Luteolin and circ_0000190 on tumor growth of lung cancer were examined by the xenograft tumor model in vivo. Luteolin inhibited cell viability, colony formation, migration, invasion, and promoted apoptosis of lung cancer cells. Moreover, overexpression of circ_0000190 could counteract the suppression role of luteolin on lung cancer development. Andcirc_0000190 directly bound with miR-130a-3p. Luteolin blocked lung cancer cell growth, metastasis, and Notch-1 signaling pathway by modulating the circ_0000190/miR-130a-3pin vitro. Luteolin repressed tumor growth of lung cancer in vivo by regulating circ_0000190. Luteolin dampened the progression of lung cancer partly by regulating circ_0000190/miR-130a-3p, providing an underlying therapeutic target for lung cancer.
Collapse
Affiliation(s)
- Hao Zheng
- Department of Respiratory, The Sixth Affiliated Hospital of Wenzhou Medinine University, Lishui People's Hospital, Lishui City, China
| | - Xiaoyu Zhu
- Department of General Surgery, The Sixth Affiliated Hospital of Wenzhou Medinine University, Lishui People's Hospital, Lishui City, China
| | - Enhui Gong
- Department of Respiratory, The Sixth Affiliated Hospital of Wenzhou Medinine University, Lishui People's Hospital, Lishui City, China
| | - Yuankai Lv
- Department of Respiratory, The Sixth Affiliated Hospital of Wenzhou Medinine University, Lishui People's Hospital, Lishui City, China
| | - Yuling Li
- Department of Respiratory, The Sixth Affiliated Hospital of Wenzhou Medinine University, Lishui People's Hospital, Lishui City, China
| | - Xiaoping Cai
- Department of Respiratory, The Sixth Affiliated Hospital of Wenzhou Medinine University, Lishui People's Hospital, Lishui City, China
| |
Collapse
|
38
|
Li M, Zheng Y, Zhao J, Liu M, Shu X, Li Q, Wang Y, Zhou Y. Polyphenol Mechanisms against Gastric Cancer and Their Interactions with Gut Microbiota: A Review. Curr Oncol 2022; 29:5247-5261. [PMID: 35892986 PMCID: PMC9332243 DOI: 10.3390/curroncol29080417] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 12/14/2022] Open
Abstract
The lack of new drugs and resistance to existing drugs are serious problems in gastric cancer(GC) treatment. The research found polyphenols possess anti-Helicobacter pylori(Hp) and antitumor activities and may be used in the research and development of drugs for cancer prevention and treatment. However, polyphenols are affected by their chemical structures and physical properties, which leads to relatively low bioavailability and bioactivity in vivo. The intestinal flora can improve the absorption, utilization, and biological activity of polyphenols, whereas polyphenol compounds can increase the richness of the intestinal flora, reduce the activity of carcinogenic bacteria, stabilize the proportion of core flora, and maintain homeostasis of the intestinal microenvironment. Our review summarizes the gastrointestinal flora-mediated mechanisms of polyphenol against GC.
Collapse
Affiliation(s)
- Matu Li
- The First Clinical Medical School, Lanzhou University, Lanzhou 730000, China; (M.L.); (J.Z.); (M.L.)
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou 730000, China; (Y.Z.); (X.S.); (Q.L.)
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Ya Zheng
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou 730000, China; (Y.Z.); (X.S.); (Q.L.)
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Jinyu Zhao
- The First Clinical Medical School, Lanzhou University, Lanzhou 730000, China; (M.L.); (J.Z.); (M.L.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Meimei Liu
- The First Clinical Medical School, Lanzhou University, Lanzhou 730000, China; (M.L.); (J.Z.); (M.L.)
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou 730000, China; (Y.Z.); (X.S.); (Q.L.)
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Xiaochuang Shu
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou 730000, China; (Y.Z.); (X.S.); (Q.L.)
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Qiang Li
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou 730000, China; (Y.Z.); (X.S.); (Q.L.)
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Yuping Wang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou 730000, China; (Y.Z.); (X.S.); (Q.L.)
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Yongning Zhou
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou 730000, China; (Y.Z.); (X.S.); (Q.L.)
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
39
|
Zang M, Hou J, Huang Y, Wang J, Ding X, Zhang B, Wang Y, Xuan Y, Zhou Y. Crocetin suppresses angiogenesis and metastasis through inhibiting sonic hedgehog signaling pathway in gastric cancer. Biochem Biophys Res Commun 2021; 576:86-92. [PMID: 34482028 DOI: 10.1016/j.bbrc.2021.08.092] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 08/25/2021] [Accepted: 08/28/2021] [Indexed: 12/14/2022]
Abstract
Gastric cancer (GC) is one of the major causes of cancer-related deaths and chemoresistance is a key obstacle to the treatment of GC, particularly in advanced GC. As an active component of saffron stigma, crocetin has important therapeutic effects on various diseases including tumors. However, the therapeutic potential of crocetin targeting GC is still unclear and the underlying mechanisms are remained to be further explored. In this study, crocetin significantly inhibited angiogenesis in GC, including tubes of HUVECs and vasculogenic mimicry (VM) formation of GC cells. Crocetin also suppressed cell proliferation, migration and invasion. To explore which signaling pathway involving in crocetin, HIF-1α, Notch1, Sonic hedgehog (SHH) and VEGF were examined with crocetin treatment and we found that SHH significantly decreased. Crocetin suppressed SHH signaling with SHH, PTCH2, Sufu and Gli1 protein level decreased in western blot assay. In addition, crocetin suppressed SHH secretion in GC and HUVEC cells. The promoted effects on cell migration induced by secreted SHH were also inhibited by crocetin in GC and HUVEC cell co-culture system. Furthermore, recombinant SHH promoted angiogenesis as well as cell migration and proliferation. However, these promoted effects were reversed by crocetin treatment. These results revealed that crocetin suppressed GC angiogenesis and metastasis through SHH signaling pathway, indicating that crocetin may function as an effective therapeutic drug against GC.
Collapse
Affiliation(s)
- Mingde Zang
- Department of Gastric Cancer Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, PR China
| | - Junyi Hou
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, PR China
| | - Yakai Huang
- Department of Gastric Cancer Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, PR China
| | - Jiangli Wang
- Department of Gastric Cancer Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, PR China
| | - Xusheng Ding
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, PR China
| | - Baogui Zhang
- Affiliated Hospital of Jining Medical University, No. 89 Guhuai Road, Rencheng District, Jining City, 272000, PR China
| | - Yanong Wang
- Department of Gastric Cancer Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, PR China
| | - Yi Xuan
- Department of Gastric Cancer Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, PR China.
| | - Ye Zhou
- Department of Gastric Cancer Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, PR China.
| |
Collapse
|
40
|
Zhang SX, Liu W, Ai B, Sun LL, Chen ZS, Lin LZ. Current Advances and Outlook in Gastric Cancer Chemoresistance: A Review. Recent Pat Anticancer Drug Discov 2021; 17:26-41. [PMID: 34587888 DOI: 10.2174/1574892816666210929165729] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 08/19/2021] [Accepted: 09/20/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Surgical resection of the lesion is the standard primary treatment of gastric cancer. Unfortunately, most patients are already in the advanced stage of the disease when they are diagnosed with gastric cancer. Alternative therapies, such as radiation therapy and chemotherapy, can achieve only very limited benefits. The emergence of cancer drug resistance has always been the major obstacle to the cure of tumors. The main goal of modern cancer pharmacology is to determine the underlying mechanism of anticancer drugs. OBJECTIVE Here, we mainly review the latest research results related to the mechanism of chemotherapy resistance in gastric cancer, the application of natural products in overcoming the chemotherapy resistance of gastric cancer, and the new strategies currently being developed to treat tumors based on immunotherapy and gene therapy. CONCLUSION The emergence of cancer drug resistance is the main obstacle in achieving alleviation and final cure for gastric cancer. Mixed therapies are considered to be a possible way to overcome chemoresistance. Natural products are the main resource for discovering new drugs specific for treating chemoresistance, and further research is needed to clarify the mechanism of natural product activity in patients. .
Collapse
Affiliation(s)
- Sheng-Xiong Zhang
- Guangdong Province Work Injury Rehabilitation Hospital, Guangzhou, 510440. China
| | - Wei Liu
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006. China
| | - Bo Ai
- Huazhong University of Science and Technology, Wuhan, 430030. China
| | - Ling-Ling Sun
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405. China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY 11439, New York. United States
| | - Li-Zhu Lin
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405. China
| |
Collapse
|
41
|
Zhao H, Hu H, Chen B, Xu W, Zhao J, Huang C, Xing Y, Lv H, Nie C, Wang J, He Y, Wang SQ, Chen XB. Overview on the Role of E-Cadherin in Gastric Cancer: Dysregulation and Clinical Implications. Front Mol Biosci 2021; 8:689139. [PMID: 34422902 PMCID: PMC8371966 DOI: 10.3389/fmolb.2021.689139] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/19/2021] [Indexed: 01/04/2023] Open
Abstract
Gastric cancer is the fifth most common cancer and the third most common cause of cancer death all over the world. E-cadherin encoded by human CDH1 gene plays important roles in tumorigenesis as well as in tumor progression, invasion and metastasis. Full-length E-cadhrin tethered on the cell membrane mainly mediates adherens junctions between cells and is involved in maintaining the normal structure of epithelial tissues. After proteolysis, the extracellular fragment of the full-length E-cadhein is released into the extracellular environment and the blood, which is called soluble E-cadherin (sE-cadherin). sE-cadherin promots invasion and metastasis as a paracrine/autocrine signaling molecule in the progression of various types of cancer including gastric cancer. This review mainly summarizes the dysregulation of E-cadherin and the regulatory roles in the progression, invasion, metastasis, and drug-resistance, as well as its clinical applications in diagnosis, prognosis, and therapeutics of gastric cancer.
Collapse
Affiliation(s)
- Huichen Zhao
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Huihui Hu
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Beibei Chen
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, China
| | - Weifeng Xu
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Jing Zhao
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Chen Huang
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Yishu Xing
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Huifang Lv
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Caiyun Nie
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Jianzheng Wang
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Yunduan He
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Sai-Qi Wang
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, China
| | - Xiao-Bing Chen
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
42
|
AL-Ishaq RK, Liskova A, Kubatka P, Büsselberg D. Enzymatic Metabolism of Flavonoids by Gut Microbiota and Its Impact on Gastrointestinal Cancer. Cancers (Basel) 2021; 13:3934. [PMID: 34439088 PMCID: PMC8394324 DOI: 10.3390/cancers13163934] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/18/2021] [Accepted: 08/02/2021] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal (GI) cancer is a prevalent global health disease with a massive burden on health care providers. Internal and external factors such as obesity, smoking, diet (red meat), low socioeconomic status and infection with Helicobacter pylori are the critical risk factors of GI cancers. Flavonoids are natural phenolic compounds found abundantly in fruits and vegetables. Upon ingestion, 90% of flavonoids consumed require further enzymatic metabolism by the gut microbiome to enhance their bioavailability and absorption. Several epidemiological studies reported that consumption of flavonoids and their enzymatic conversion by gut microbes is strongly associated with the reduced risk of GI cancer development. This review summarizes the current knowledge on the enzymatic conversion of flavonoids by the human gut microbiome. It also addresses the underlying anti-GI cancer effects on metabolic pathways such as apoptosis and cellular proliferation. Overall, metabolites produced from flavonoid's enzymatic conversion illustrate anti-GI cancer effects, but the mechanisms of action need further clarification.
Collapse
Affiliation(s)
| | - Alena Liskova
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia;
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia;
| | - Dietrich Büsselberg
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| |
Collapse
|
43
|
Bélair V, Orsat V, Maheux M, Lafrance CP, Brochu M, Lightburn B, Moss R. Permeability of native and digested polyphenols from apple, blueberry and cranberry extracts using PAMPA membrane permeability assays. J Food Compost Anal 2021. [DOI: 10.1016/j.jfca.2021.103945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
44
|
Dai G, Liu G, Zheng D, Song Q. Inhibition of the Notch signaling pathway attenuates progression of cell motility, metastasis, and epithelial-to-mesenchymal transition-like phenomena induced by low concentrations of cisplatin in osteosarcoma. Eur J Pharmacol 2021; 899:174058. [PMID: 33757752 DOI: 10.1016/j.ejphar.2021.174058] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/13/2021] [Accepted: 03/17/2021] [Indexed: 01/27/2023]
Abstract
Although advances in osteosarcoma treatment have been made in recent decades, the survival rate for patients suffering from metastatic disease, especially lung metastasis, remains disappointing. Previous studies have confirmed that epithelial-to-mesenchymal transition (EMT) is associated with tumor metastasis, and several studies have suggested that osteosarcoma cells also exhibit EMT-like characteristics. In addition, Notch signaling is known to be related to the development and progression of human malignancies, including osteosarcoma. However, whether chemotherapy affects the EMT-like events and whether these events are medicated by Notch signaling remain to be elucidated. To address these issues, in the current work, osteosarcoma 143B cells were exposed to sublethal concentrations of the first-line chemotherapeutic agent cisplatin (DDP), which promoted cell migration, in vitro invasion, and in vivo lung metastasis. Furthermore, low concentrations of DDP upregulated mesenchymal phenotype-related genes and proteins and promoted EMT-like properties in osteosarcoma cells. In addition, low concentrations of DDP could activate the Notch receptor and its target genes. Finally, combined treatment of DDP with the Notch signaling pathway inhibitor DAPT, which can effectively downregulate mesenchymal phenotype-related genes and proteins, inhibited cell migration and invasion in vitro, and it decreased pulmonary metastatic nodules in vivo. The results of the current study supported the idea that low concentrations of DDP could induce EMT-like characteristics in osteosarcoma cells and could promote cell mobility in vitro, as well as pulmonary metastasis in vivo. Importantly, however, these biological processes are mediated by the Notch signaling pathway. Blocking the Notch signaling pathway can effectively attenuate the osteosarcoma EMT-like phenotype and its associated migration, invasion, and metastasis.
Collapse
Affiliation(s)
- Guo Dai
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
| | - Gaiwei Liu
- Department of Orthopedics, Jingzhou Central Hospital, Jingzhou, 434000, Hubei, China
| | - Di Zheng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Qi Song
- Department of Trauma Surgery, Wuhan No. 1 Hospital, Wuhan, 430022, Hubei, China
| |
Collapse
|
45
|
Bley N, Hmedat A, Müller S, Rolnik R, Rausch A, Lederer M, Hüttelmaier S. Musashi-1-A Stemness RBP for Cancer Therapy? BIOLOGY 2021; 10:407. [PMID: 34062997 PMCID: PMC8148009 DOI: 10.3390/biology10050407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 04/29/2021] [Accepted: 05/01/2021] [Indexed: 12/12/2022]
Abstract
The RNA-binding protein Musashi-1 (MSI1) promotes stemness during development and cancer. By controlling target mRNA turnover and translation, MSI1 is implicated in the regulation of cancer hallmarks such as cell cycle or Notch signaling. Thereby, the protein enhanced cancer growth and therapy resistance to standard regimes. Due to its specific expression pattern and diverse functions, MSI1 represents an interesting target for cancer therapy in the future. In this review we summarize previous findings on MSI1's implications in developmental processes of other organisms. We revisit MSI1's expression in a set of solid cancers, describe mechanistic details and implications in MSI1 associated cancer hallmark pathways and highlight current research in drug development identifying the first MSI1-directed inhibitors with anti-tumor activity.
Collapse
Affiliation(s)
- Nadine Bley
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
- Core Facility Imaging, Institute for Molecular Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany
| | - Ali Hmedat
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
| | - Simon Müller
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
| | - Robin Rolnik
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
| | - Alexander Rausch
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
- Core Facility Imaging, Institute for Molecular Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany
| | - Marcell Lederer
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
| | - Stefan Hüttelmaier
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
| |
Collapse
|
46
|
Zuo W, Liu N, Zeng Y, Xiao Z, Wu K, Yang F, Li B, Song Q, Xiao Y, Liu Q. Luteolin Ameliorates Experimental Pulmonary Arterial Hypertension via Suppressing Hippo-YAP/PI3K/AKT Signaling Pathway. Front Pharmacol 2021; 12:663551. [PMID: 33935785 PMCID: PMC8082250 DOI: 10.3389/fphar.2021.663551] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/23/2021] [Indexed: 12/17/2022] Open
Abstract
Luteolin is a flavonoid compound with a variety of pharmacological effects. In this study, we explored the effects of luteolin on monocrotaline (MCT) induced rat pulmonary arterial hypertension (PAH) and underlying mechanisms. A rat PAH model was generated through MCT injection. In this model, luteolin improved pulmonary vascular remodeling and right ventricular hypertrophy, meanwhile, luteolin could inhibit the proliferation and migration of pulmonary artery smooth muscle cells induced by platelet-derived growth factor-BB (PDGF-BB) in a dose-dependent manner. Moreover, our results showed that luteolin could downregulate the expression of LATS1 and YAP, decrease YAP nuclear localization, reduce the expression of PI3K, and thereby restrain the phosphorylation of AKT induced by PDGF-BB. In conclusion, luteolin ameliorated experimental PAH, which was at least partly mediated through suppressing HIPPO-YAP/PI3K/AKT signaling pathway. Therefore, luteolin might become a promising candidate for treatment of PAH.
Collapse
Affiliation(s)
- Wanyun Zuo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Hunan, China
| | - Na Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Hunan, China
| | - Yunhong Zeng
- Department of Cardiology, Hunan Children's Hospital, Hunan, China
| | - Zhenghui Xiao
- Department of Cardiology, Hunan Children's Hospital, Hunan, China
| | - Keke Wu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Hunan, China
| | - Fan Yang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Hunan, China
| | - Biao Li
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Hunan, China
| | - Qingqing Song
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Hunan, China
| | - Yunbin Xiao
- Department of Cardiology, Hunan Children's Hospital, Hunan, China
| | - Qiming Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Hunan, China
| |
Collapse
|
47
|
Zhao F, Hong X, Li D, Wei Z, Ci X, Zhang S. Diosmetin induces apoptosis in ovarian cancer cells by activating reactive oxygen species and inhibiting the Nrf2 pathway. Med Oncol 2021; 38:54. [PMID: 33811596 DOI: 10.1007/s12032-021-01501-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/20/2021] [Indexed: 10/21/2022]
Abstract
The fatality rate of ovarian cancer ranks first among gynecological tumors, and the prognosis is poor. Diosmetin (Dio), a natural flavonoid obtained from citrus fruits, has been shown to have anti-tumor effects in lung, liver, and skin cancers. We aimed to investigate the effects of Dio on ovarian cancer A2780 and SKOV3 cells along with the underlying mechanisms. Our data showed that Dio inhibited the proliferation, migration, and invasion of these cells and induced their apoptosis. Moreover, Dio upregulated the levels of Bax and cleaved Caspase-3 and PARP while downregulating the level of Bcl2. Mechanistically, our results revealed that Dio inhibited Nrf2 and induced the production of reactive oxygen species (ROS). The ROS scavenger N-acetyl-L-cysteine (NAC) suppressed the inhibitory effect of Dio on the proliferation of the ovarian cancer cells. Additionally, overexpression of Nrf2 partially suppressed the Dio-induced apoptosis and proliferation inhibition in these cells. These findings indicate that Dio exerts an anti-tumor activity by upregulating ROS levels and inhibiting Nrf2, indicating that Dio is a promising chemotherapeutic candidate for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Feijie Zhao
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Xinmin Road 519, Changchun, Jilin, 130001, China
| | - Xiaoling Hong
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Xinmin Road 519, Changchun, Jilin, 130001, China
| | - Danjie Li
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Xinmin Road 519, Changchun, Jilin, 130001, China
| | - Zhentong Wei
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Xinmin Road 519, Changchun, Jilin, 130001, China
| | - Xinxin Ci
- Institute of Translational Medicine, The First Hospital of Jilin University, Dongminzhu Road 71, Changchun, Jilin, 130001, China.
| | - Songling Zhang
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Xinmin Road 519, Changchun, Jilin, 130001, China.
| |
Collapse
|
48
|
Sun B, Liu Y, He D, Li J, Wang J, Wen W, Hong M. Traditional Chinese medicines and their active ingredients sensitize cancer cells to TRAIL-induced apoptosis. J Zhejiang Univ Sci B 2021; 22:190-203. [PMID: 33719224 DOI: 10.1631/jzus.b2000497] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The rapidly developing resistance of cancers to chemotherapy agents and the severe cytotoxicity of such agents to normal cells are major stumbling blocks in current cancer treatments. Most current chemotherapy agents have significant cytotoxicity, which leads to devastating adverse effects and results in a substandard quality of life, including increased daily morbidity and premature mortality. The death receptor of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can sidestep p53-dependent pathways to induce tumor cell apoptosis without damaging most normal cells. However, various cancer cells can develop resistance to TRAIL-induced apoptosis via different pathways. Therefore, it is critical to find an efficient TRAIL sensitizer to reverse the resistance of tumor cells to TRAIL, and to reinforce TRAIL's ability to induce tumor cell apoptosis. In recent years, traditional Chinese medicines and their active ingredients have shown great potential to trigger apoptotic cell death in TRAIL-resistant cancer cell lines. This review aims to collate information about Chinese medicines that can effectively reverse the resistance of tumor cells to TRAIL and enhance TRAIL's ability to induce apoptosis. We explore the therapeutic potential of TRAIL and provide new ideas for the development of TRAIL therapy and the generation of new anti-cancer drugs for human cancer treatment. This study involved an extensive review of studies obtained from literature searches of electronic databases such as Google Scholar and PubMed. "TRAIL sensitize" and "Chinese medicine" were the search keywords. We then isolated newly published studies on the mechanisms of TRAIL-induced apoptosis. The name of each plant was validated using certified databases such as The Plant List. This study indicates that TRAIL can be combined with different Chinese medicine components through intrinsic or extrinsic pathways to promote cancer cell apoptosis. It also demonstrates that the active ingredients of traditional Chinese medicines enhance the sensitivity of cancer cells to TRAIL-mediated apoptosis. This provides useful information regarding traditional Chinese medicine treatment, the development of TRAIL-based therapies, and the treatment of cancer.
Collapse
Affiliation(s)
- Bingyu Sun
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yongqiang Liu
- Research Center of Chinese Herbal Resource Science and Engineering, Guangzhou University of Traditional Chinese Medicine, Guangzhou 510006, China
| | - Danhua He
- Research Center of Chinese Herbal Resource Science and Engineering, Guangzhou University of Traditional Chinese Medicine, Guangzhou 510006, China
| | - Jinke Li
- Department of Pharmacology & Toxicology, University of Kansas, Lawrence, KS 66105, USA
| | - Jiawei Wang
- Zhongshan People's Hospital, Zhongshan 528400, China
| | - Wulin Wen
- ENT & HN Surgery Department, the Second Affiliated Hospital of Ningxia Medical University, Yinchuan 750000, China.
| | - Ming Hong
- Institute of Advanced Diagnostic and Clinical Medicine, Zhongshan People's Hospital, Guangzhou University & Zhongshan People's Hospital Joint Biomedical Institute, Zhongshan 528400, China. .,Dongguan & Guangzhou University of Chinese Medicine Cooperative Academy of Mathematical Engineering for Chinese Medicine, Dongguan 523000, China.
| |
Collapse
|
49
|
Ganai SA, Sheikh FA, Baba ZA, Mir MA, Mantoo MA, Yatoo MA. Anticancer activity of the plant flavonoid luteolin against preclinical models of various cancers and insights on different signalling mechanisms modulated. Phytother Res 2021; 35:3509-3532. [DOI: 10.1002/ptr.7044] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 01/04/2021] [Accepted: 01/25/2021] [Indexed: 12/13/2022]
Affiliation(s)
- Shabir Ahmad Ganai
- Division of Basic Sciences and Humanities FoA, SKUAST Kashmir Sopore Jammu & Kashmir India
| | - Farooq Ahmad Sheikh
- Division of Genetics and Plant Breeding FoA, SKUAST Kashmir Sopore Jammu & Kashmir India
| | - Zahoor Ahmad Baba
- Division of Basic Sciences and Humanities FoA, SKUAST Kashmir Sopore Jammu & Kashmir India
| | - Mudasir Ahmad Mir
- Department of Microbiology Government Medical College Anantnag Jammu & Kashmir India
| | - Mohd Ayoob Mantoo
- Division of Entomology FoA, SKUAST Kashmir Sopore Jammu & Kashmir India
| | - Manzoor Ahmad Yatoo
- Division of Basic Sciences and Humanities FoA, SKUAST Kashmir Sopore Jammu & Kashmir India
| |
Collapse
|
50
|
Nguyen HT, Couture R, Touaibia M, Martin LJ. Transcriptome modulation following administration of luteolin to bleomycin-etoposide-cisplatin chemotherapy on rat LC540 tumor Leydig cells. Andrologia 2021; 53:e13960. [PMID: 33400304 DOI: 10.1111/and.13960] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/09/2020] [Accepted: 12/22/2020] [Indexed: 01/11/2023] Open
Abstract
Leydig cell tumours represent 1%-3% of all cases of testicular tumours in men. Such tumours respond poorly to radiation or chemotherapy, including bleomycin-etoposide-cisplatin (BEP) combinatorial therapy. In this study, we investigated an alternative approach involving luteolin to improve the efficacy of chemotherapy. LC540 tumour Leydig cells were treated with BEP (bleomycin 40 µg/ml, etoposide 4 µg/ml, cisplatin 8 µg/ml) and/or luteolin 10 µM for comparison with DMSO-treated cells. We performed a transcriptome analysis using RNA-Seq to characterise changes in biological processes and signalling pathways. Treatments of LC540 tumour Leydig cells with luteolin significantly decreased the expression of genes involved in cholesterol biosynthesis, while increasing the expression of genes related to glutathione conjugation (p < .05). Genes being significantly upregulated in response to BEP treatment were involved in the response to toxic substances and transcriptional regulation. Oppositely, genes being significantly downregulated by BEP treatment were enriched for intracellular signal transduction, cell migration, cell adhesion, reproductive system development and cholesterol biosynthesis. BEP chemotherapy proved to be effective in increasing gene expression related to apoptosis of tumour Leydig cells. However, addition of luteolin to BEP treatment had no other effects on biological processes or pathways related to cancer treatment.
Collapse
Affiliation(s)
- Ha Tuyen Nguyen
- Biology Department, Université de Moncton, Moncton, NB, Canada
| | - Roxanne Couture
- Biology Department, Université de Moncton, Moncton, NB, Canada
| | - Mohamed Touaibia
- Chemistry and Biochemistry Department, Université de Moncton, Moncton, NB, Canada
| | - Luc J Martin
- Biology Department, Université de Moncton, Moncton, NB, Canada
| |
Collapse
|